US20060223177A1 - Postpartum cells derived from umbilical cord tissue, and methods of making and using the same - Google Patents

Postpartum cells derived from umbilical cord tissue, and methods of making and using the same Download PDF

Info

Publication number
US20060223177A1
US20060223177A1 US11/315,897 US31589705A US2006223177A1 US 20060223177 A1 US20060223177 A1 US 20060223177A1 US 31589705 A US31589705 A US 31589705A US 2006223177 A1 US2006223177 A1 US 2006223177A1
Authority
US
United States
Prior art keywords
cell
cells
protein
alpha
derived
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/315,897
Inventor
Ian Harris
Darin Messina
Anthony Kihm
Agnieszka Seyda
David Colter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DePuy Spine LLC
DePuy Orthopaedics Inc
DePuy Synthes Products Inc
Original Assignee
Ethicon Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/877,012 external-priority patent/US7510873B2/en
Priority to US11/315,897 priority Critical patent/US20060223177A1/en
Application filed by Ethicon Inc filed Critical Ethicon Inc
Assigned to ETHICON, INCORPORATED reassignment ETHICON, INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SEYDA, AGNIESZKA, COLTER, DAVID C., KIHM, ANTHONY J., HARRIS, IAN ROSS, MESSINA, DARIN J.
Publication of US20060223177A1 publication Critical patent/US20060223177A1/en
Priority to US12/946,609 priority patent/US8815587B2/en
Assigned to ADVANCED TECHNOLOGIES AND REGENERATIVE MEDICINE, LLC reassignment ADVANCED TECHNOLOGIES AND REGENERATIVE MEDICINE, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ETHICON, INC.
Assigned to DEPUY ORTHOPAEDICS, INC. reassignment DEPUY ORTHOPAEDICS, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: ADVANCED TECHNOLOGIES AND REGENERATIVE MEDICINE, LLC
Assigned to HAND INNOVATIONS LLC reassignment HAND INNOVATIONS LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DEPUY SPINE, LLC
Assigned to DEPUY SPINE, LLC reassignment DEPUY SPINE, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: DEPUY SPINE, INC.
Assigned to DePuy Synthes Products, LLC reassignment DePuy Synthes Products, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: HAND INNOVATIONS LLC
Assigned to DEPUY SPINE, INC. reassignment DEPUY SPINE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DEPUY ORTHOPAEDICS, INC.
Assigned to DePuy Synthes Products, Inc. reassignment DePuy Synthes Products, Inc. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: DePuy Synthes Products, LLC
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0619Neurons
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/03Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from non-embryonic pluripotent stem cells

Definitions

  • This invention relates to mammalian, preferably human, cell therapy, and more particularly to isolated cells derived from postpartum umbilicus, methods of deriving such cells, and methods of their use and growth on various media, including media which are serum-containing or serum-free.
  • umbilical cord blood or simply “cord blood”
  • cord blood a potential source of, for example, hematopoietic cells for use by an individual for whom cord blood has been banked at birth.
  • Such cells would be useful for those individuals, for example, who require therapeutic radiation which may eliminate functional portions of their immune system.
  • the individual's own banked cord blood could be used to reconstitute the lost immune cells, and restore immune function.
  • Stem cells are understood in general terms as cells that 1) have the ability to self-renew for long periods through cell division from a single cell; and 2) have the ability to differentiate into specific cell types given the proper conditions. Accordingly, stem cells are potentially useful in treating a population of individuals, and not merely the person from whose cord blood the cells were initially obtained.
  • cord blood has been considered as a source of hematopoietic progenitor stem cells.
  • Banked (or cryopreserved) cord blood, or stem cells isolated therefrom have been deemed useful for hematopoietic reconstitution, for example in bone marrow and related transplantations. (Boyse et al., U.S. Pat. Nos. 5,004,681 and 5,192,553).
  • Purchio et al. U.S. Pat. No. 5,919,702 have isolated chondrogenic progenitor cells (or prechondrocytes) from Wharton's Jelly. They reported the isolation of cells from human umbilical cord Wharton's Jelly by removing blood and blood vessels and incubating the tissue under conditions purported to allow the prechondrocytes to proliferate. As such, the method did not distinguish the desired cells from the different cell types present in Wharton's Jelly, but rather relied on migration from the tissue or selecting growth conditions favoring prechondrocytes. The prechondrocytes were expanded mitotically after they were established.
  • Cells at passages 2 to 4 were reported as useful to produce cartilage, if triggered by the addition of exogenous growth factors, such as BMP-13 or TGF-beta. Uses of the cells for direct injection or implantation, or use with a hydrogel or tissue matrix, were proposed. However, it was considered important that the cells not exceed about 25% confluence. The cells were not characterized with respect to their biochemical or immunological properties, or with respect to their gene expression.
  • Weiss et al. (U.S. Patent Application Publication US2003/0161818) proposed procedures for isolating pluripotent or lineage-committed cells from mammalian Wharton's Jelly or non-blood umbilical cord matrix sources.
  • the cells isolated were reported to differentiate into hematopoietic, mesenchymal or neuroectodermal lines.
  • the cell lines were not characterized with respect to their identifying properties. Limited characterization was provided with respect to cells after differentiation towards neural lines. Reference was made to Wharton's Jelly-derived bovine and porcine cells that were CD34 ⁇ , CD45 ⁇ .
  • media for culturing cells most contain at least some fetal bovine or calf serum.
  • serum supplements of about 10-20% (v/v).
  • the serum component is often integral to the survival and expansion of cell populations.
  • a myriad of proteins are found in bovine serum, including for example PDGF and FGFs, known growth factors that can have significant influence on cell growth and differentiation on cell populations, including populations of stem and progenitor cells.
  • bovine serum or serum of other species
  • BSE bovine spongiform encephalopathy
  • FIG. 1 Long-term growth potential of umbilical cells isolated under different conditions. Conditions that allowed for more than 5 population doublings were further analyzed for long-term growth potential. Cells were cultured using the indicated media formulations and culture conditions until cells reached senescence. Senescence was determined when cells failed to achieve greater than one population doubling during the study time interval.
  • the invention provides isolated umbilicus-derived cells (UDCs) that are derived from mammalian umbilical cord tissue substantially free of blood.
  • the cells are capable of self-renewal and expansion in culture.
  • the umbilicus-derived cells have the potential to differentiate into cells of other phenotypes.
  • the cells are derived from human umbilicus.
  • the cells have been characterized as to several of their cellular, genetic, immunological, and biochemical properties.
  • the cells have been characterized by their growth properties in culture, by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
  • the postpartum-derived cell is an umbilicus-derived cell. In other embodiments it is a placenta-derived cell. In specific embodiments, the cell has all identifying features of either of cell types UMB 022803 (P7) (ATCC Accession No. PTA-6067); or UMB 022803 (P17) (ATCC Accession No. PTA-6068).
  • cell cultures comprising the isolated umbilicus-derived cells of the invention are provided.
  • the cultures of umbilical cells are free of maternal cells in certain preferred embodiments.
  • isolated umbilicus-derived cells having specific cell surface marker expression profiles are provided, wherein particular cell surface marker proteins are produced.
  • the cells produce one or more of CD10, CD13, CD44, CD73, CD90, CD141, PDGFr-alpha, or HLA-A, B, C.
  • the cells do not produce one or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
  • the cells of the invention have also been characterized according to their expression of a wide variety of genes. Accordingly, another aspect of the invention provides isolated umbilicus-derived cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of genes for one or more of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morph
  • these isolated human umbilicus-derived cells express a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3, wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, an ileac crest bone marrow cell, or placenta-derived cell.
  • the cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes.
  • therapeutic cell cultures comprising the isolated human umbilicus-derived cells.
  • the invention provides isolated human umbilicus-derived cells capable of self-renewal and expansion in culture and which have the potential to differentiate into cells of other phenotypes, wherein the cells do not stimulate allogeneic lymphocytes in a mixed lymphocyte reaction, and expresses PD-L2, but not HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2.
  • the cells do not stimulate allogeneic PBMCs. More preferably they do not stimulate allogeneic lymphocytes, allogeneic T-cells, or na ⁇ ve T-cells, or generate other adverse immunological responses in either matched or unmatched recipients.
  • the cells also can produce vimentin and alpha-smooth muscle actin in certain embodiments.
  • the invention provides isolated human umbilicus-derived cells that secrete one or more of the angiogenic factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1.
  • the cells secrete several or all of the aforementioned molecules.
  • the cells do not secrete one or more of the angiogenic factors SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA. In particular embodiments, they secrete few or none of those molecules.
  • therapeutic cell cultures are provided, the cell cultures comprising the isolated cells as described above for use in treating patients in need of angiogenesis-stimulating trophic factors.
  • Such therapeutic cell cultures are also provided for use in treating a patient in need of neural growth stimulating trophic factors.
  • Methods of deriving cells from non-blood human umbilical tissue are provided.
  • the cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes.
  • the method comprises (a) obtaining human umbilical tissue; (b) removing substantially all of the blood to yield a substantially blood-free umbilical tissue, (c) dissociating the tissue by mechanical or enzymatic treatment, or both, (d) resuspending the tissue in a culture medium, and (e) providing growth conditions which allow for the growth of a human umbilicus-derived cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes.
  • Preferred methods involve enzymatic treatment with, for example, collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided herein.
  • Isolated human umbilicus-derived cells derived by the above method are also provided herein.
  • the cells maintain a consistent normal karyotype notwithstanding repeated passaging in certain embodiments.
  • cultures of human umbilicus-derived cells derived by the above method wherein the cultures are free of maternal cells.
  • cells and cultures of such cells wherein the cells isolated by any of a variety of methods in any of a variety of media, even when cultured under a wide range range of conditions, are capable of maintaining a substantially consistent profile of expressed surface markers, or substantially consistent profile of expression of genes that characterize the cells, e.g. “signature gene profiles” including for example, expression patterns of which are specific to certain stem cells and/or to certain postpartum cells.
  • Co-cultures comprising the cells or cultures of the invention with other mammalian cells are also provided herein.
  • these co-cultures comprise another human cell line whose growth or therapeutic potential, for example, is improved by the presence of the umbilicus-derived cells.
  • Such co-cultures are useful for therapeutic application in vitro or in vivo.
  • compositions comprising an umbilicus-derived cell and another therapeutic agent, factor, or bioactive agent.
  • factors include, but are not limited to, IGF, LIF, PDGF, EGF, FGF, as well as antithrombogenic, antiapoptotic agents, anti-inflammatory agents, immunosuppressive or immunomodulatory agents, and antioxidants.
  • Such therapeutic compositions can further comprise one or more additional cell types in addition to the UDCs and the bioactive component.
  • compositions derived from the cells are provided herein.
  • Cell lysates, soluble cell fractions and membrane-enriched cell fractions are provided herein.
  • Extracellular matrices derived from the cells, for example, comprising basement membranes are also useful and are provided herein.
  • compositions of the invention also include conditioned culture media as provided herein.
  • Such media have first been used to grow the cells or cultures of the invention, which during growth secrete one or more useful products into the medium.
  • Conditioned medium from these novel cells are useful for many purposes, including for example, supporting the growth of other mammalian cells in need of growth factors or trophic factors secreted into the media by the cells and cultures of the invention, and promoting, for example, angiogenesis.
  • Methods are provided of inducing the cells to differentiate along a pathway towards progenitors of various cells, or even into terminally differentiated cells themselves. Such cells have utility for therapeutic treatment of certain conditions, disorders and disease states. Such cells also have utility for diagnostic protocols, such as for use in assays to identify therapeutic agents.
  • the invention also provides methods of utilizing the differentiated umbilicus-derived cells or the progenitors for therapeutic uses, including but not limited to angiogenic application, neuronal applications, soft tissue applications, occular applications, and applications wherein the cells are useful in treatment of heart, kidney, bone, cartilage, pancreas, liver, and other tissues alone or in combination with other therapeutic agents.
  • Kits are also provided herein. Kits useful for the growth, isolation and use of the umbilical-derived cells are provided.
  • the invention provides isolated postpartum-derived cells comprising a “signature gene profile” which distinguishes the PPDCs from other cells of different origin and from other known toti-and pluripotent cells.
  • Preferred signature gene profiles comprise those wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum. Also provide are such cells further comprising the ability to alter the expression of its cell surface markers when grown in medium containing serum relative to that in serum free medium.
  • markers for PDGFreceptor alpha and HLA-ABC can be altered.
  • Also provided are methods for the preparation of therapeutic cells or cultures comprising: isolating cells; initially expanding the cells to a useful number in a serum-containing medium which supports cell expansion but in which the cells produce a quantity of the cell surface marker HLA-ABC; transferring the cells to a medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC; and passaging the cells in the medium in which the cells produce a decreased amount of the HLA-ABC, thereby preparing a therapeutic cell or culture.
  • the medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC is a serum-free medium. The methods are particularly useful for the production of therapeutic cells or cultures for implantation or grafting.
  • serum-free media for the expansion of postpartum-derived cells wherein the media has one or more growth factors added.
  • growth factors presently preferred growth factors to be added are bFGF, EGF, or PDGF.
  • Methods are also provided for isolating and culturing postpartum cells, particularly umbilicus-derived cells, in serum-containing and serum-free media, with or without beta-mercaptoethanol, with or without various growth factors, on surfaces coated with gelatin, or CELLBIND, or other treatments to allow attachment. Methods of culturing the cells also include varied conditions of oxygen, and other growth parameters.
  • the invention provides therapeutic cultures comprising postpartum-derived cells expanded in serum-free medium.
  • Preferred cultures having a signature gene profile wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum.
  • cell culture banks and depositories and the like comprising postpartum-derived cells and cultures of the invention.
  • Stem cells are undifferentiated cells defined by the ability of a single cell both to self-renew, and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute substantially to most, if not all, tissues following injection into blastocysts.
  • Stem cells are classified according to their developmental potential as: (1) totipotent; (2) pluripotent; (3) multipotent; (4) oligopotent; and (5) unipotent.
  • Totipotent cells are able to give rise to all embryonic and extraembryonic cell types.
  • Pluripotent cells are able to give rise to all embryonic cell types.
  • Multipotent cells include those able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem cells (HSC) can produce progeny that include HSC (self-renewal), blood cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood).
  • HSC hematopoietic stem cells
  • Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells; and cells that are unipotent are able to give rise to a single cell lineage (e.g., spermatogenic stem cells).
  • Stem cells are also categorized on the basis of the source from which they may be obtained.
  • An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types.
  • An embryonic stem cell is a pluripotent cell from the inner cell mass of a blastocyst-stage embryo.
  • a fetal stem cell is one that originates from fetal tissues or membranes.
  • a postpartum stem cell is a multipotent or pluripotent cell that originates substantially from extraembryonic tissue available after birth, namely, the placenta and the umbilical cord.
  • Postpartum stem cells may be blood-derived (e.g., as are those obtained from umbilical cord blood) or non-blood-derived (e.g., as obtained from the non-blood tissues of the umbilical cord and placenta).
  • Embryonic tissue is typically defined as tissue originating from the embryo (which in humans refers to the period from fertilization to about six weeks of development. Fetal tissue refers to tissue originating from the fetus, which in humans refers to the period from about six weeks of development to parturition. Extraembryonic tissue is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryonic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta (which itself forms from the chorion and the maternal decidua basalis).
  • Differentiation is the process by which an unspecialized (“uncommitted”) or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example.
  • a differentiated cell is one that has taken on a more specialized (“committed”) position within the lineage of a cell.
  • the term committed, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type.
  • De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell.
  • the lineage of a cell defines the heredity of the cell, i.e. which cells it came from and what cells it can give rise to.
  • the lineage of a cell places the cell within a hereditary scheme of development and differentiation.
  • a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors.
  • stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells.
  • this broad definition of progenitor cell may be used.
  • a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types. This type of progenitor cell is generally not able to self-renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a non-renewing progenitor cell or as an intermediate progenitor or precursor cell.
  • the phrase differentiates into a mesodermal, ectodermal or endodernal lineage refers to a cell that becomes committed to a specific mesodermal, ectodermal or endodermal lineage, respectively.
  • Examples of cells that differentiate into a mesodermal lineage or give rise to specific mesodermal cells include, but are not limited to, cells that are adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoietic, hemangiogenic, myogenic, nephrogenic, urogenitogenic, osteogenic, pericardiogenic, or stromal.
  • Examples of cells that differentiate into ectodermal lineage include, but are not limited to epidermal cells, neurogenic cells, and neurogliagenic cells.
  • Examples of cells that differentiate into endodermal lineage include, but are not limited to, pleurigenic cells, hepatogenic cells, cells that give rise to the lining of the intestine, and cells that give rise to pancreogenic and splanchogenic cells.
  • the cells of the present invention are generally referred to as umbilicus-derived cells (or UDCs). They also may sometimes be referred to more generally herein as postpartum-derived cells or postpartum cells (PPDCs).
  • the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense.
  • the term derived is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a growth medium to expand the population and/or to produce a cell line). The in vitro manipulations of umbilical stem cells and the unique features of the umbilicus-derived cells of the present invention are described in detail below.
  • Cell culture refers generally to cells taken from a living organism and grown under controlled condition (“in culture” or “cultured”).
  • a primary cell culture is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture.
  • Cells are expanded in culture when they are placed in a growth medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells.
  • the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as doubling time.
  • a cell line is a population of cells formed by one or more subcultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been passaged. A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture.
  • the primary culture i.e., the first culture following the isolation of cells from tissue, is designated P0. Following the first subculture, the cells are described as a secondary culture (P1 or passage 1).
  • the cells After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number.
  • the expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and time between passaging.
  • a conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide trophic support to other cells. Such trophic factors include, but are not limited to hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, and granules.
  • the medium containing the cellular factors is the conditioned medium.
  • a trophic factor is defined as a substance that promotes or at least supports, survival, growth, proliferation and/or maturation of a cell, or stimulates increased activity of a cell.
  • senescence also replicative senescence or cellular senescence refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's limit).
  • cellular senescence was first described using fibroblast-like cells, most normal human cell types that can be grown successfully in culture undergo cellular senescence.
  • the in vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide).
  • Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone. Thus, cells made quiescent by removing essential growth factors are able to resume growth and division when the growth factors are re-introduced, and thereafter carry out the same number of doublings as equivalent cells grown continuously. Similarly, when cells are frozen in liquid nitrogen after various numbers of population doublings and then thawed and cultured, they undergo substantially the same number of doublings as cells maintained unfrozen in culture. Senescent cells are not dead or dying cells; they are actually resistant to programmed cell death (apoptosis), and have been maintained in their nondividing state for as long as three years. These cells are very much alive and metabolically active, but they do not divide. The nondividing state of senescent cells has not yet been found to be reversible by any biological, chemical, or viral agent.
  • the term Growth Medium generally refers to a medium sufficient for the culturing of umbilicus-derived cells.
  • one presently preferred medium for the culturing of the cells of the invention herein comprises Dulbecco's Modified Essential Media (also abbreviated DMEM herein).
  • Dulbecco's Modified Essential Media also abbreviated DMEM herein.
  • DMEM-low glucose also DMEM-LG herein
  • the DMEM-low glucose is preferably supplemented with 15% (v/v) fetal bovine serum (e.g.
  • fetal bovine serum defined fetal bovine serum, Hyclone, Logan Utah
  • antibiotics/antimycotics preferably penicillin (100 Units/milliliter), streptomycin (100 milligrams/milliliter), and amphotericin B (0.25 micrograms/milliliter), (Invitrogen, Carlsbad, Calif.)), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis Mo.).
  • different growth media are used, or different supplementations are provided, and these are normally indicated in the text as supplementations to, or deletions from the Growth Medium.
  • the growth medium is not supplemented with serum, and more preferably the medium is not supplemented with any animal-derived protein material. This is presently preferred for example for growing cells for clinical or preclinical uses. Such media lacking serum altogether, and in even more preferably lacking any animal protein supplementation, may be used for both isolation and growth of cells in accordance with the instant invention.
  • standard growth conditions refers to culturing of cells at 37° C., in a standard atmosphere comprising 5% CO 2 . Relative humidity is maintained at about 100%. While foregoing the conditions are useful for culturing, it is to be understood that such conditions are capable of being varied by the skilled artisan who will appreciate the options available in the art for culturing cells, for example, varying the temperature, CO 2 , rotation or agitation, relative humidity, oxygen, growth medium, and the like.
  • ANG2 (or Ang2) for angiopoietin 2
  • BDNF for brain-derived neurotrophic factor
  • DMEM for Dulbecco's Minimal Essential Medium; also used herein as synonymous with Dulbecco's Modified Eagle Medium.
  • Dulbecco's Modified Eagle Medium The skilled artisan will appreciate the composition is the same by either name.
  • DMEM:lg (or DMEM:Lg, DMEM:LG) for DMEM with low glucose
  • EGF for epidermal growth factor
  • FCS fetal calf serum
  • GCP-2 for granulocyte chemotactic protein 2 GCP-2 for granulocyte chemotactic protein 2
  • GFAP for glial fibrillary acidic protein
  • HCAEC for human coronary artery endothelial cells
  • HGF for hepatocyte growth factor
  • HNF-1 alpha for hepatocyte-specific transcription factor
  • HUVEC for human umbilical vein endothelial cells
  • I309 for a chemokine and the ligand for the CCR8 receptor is responsible for chemoattraction of TH2 type T-cells.
  • I309 binds to endothelial cells, stimulates chemotaxis and invasion of these cells, and enhances HUVEC differentiation into capillary-like structures in an in vitro Matrigel assay.
  • 1309 is an inducer of angiogenesis in vivo in both the rabbit cornea and the chick chorioallantoic membrane assay (CAM).
  • KGF for keratinocyte growth factor
  • MIP1beta for macrophage inflammatory protein 1beta
  • MSC for mesenchymal stem cells
  • PDGFbb for platelet derived growth factor
  • PDGFr/alpha for platelet derived growth factor receptor alpha
  • Rantes (or RANTES) for regulated on activation, normal T cell expressed and secreted
  • rhGDF-5 for recombinant human growth and differentiation factor 5;
  • TGFbeta2 for transforming growth factor beta2
  • TGFbeta-3 for transforming growth factor beta-3
  • TIMP1 for tissue inhibitor of matrix metalloproteinase 1
  • VEGF for vascular endothelial growth factor
  • alphaFP for alpha-fetoprotein
  • the invention provides isolated umbilicus-derived cells comprising cells derived from human umbilical cord tissue substantially free of blood.
  • the cells are capable of self-renewal and expansion in culture.
  • the umbilicus-derived cells have the potential to differentiate into cells of other phenotypes.
  • the cells can differentiate into any cell of ectodermal, mesodermal, or endodermal origin.
  • the invention provides, in one of its several aspects cells that are isolated from human umbilical tissues.
  • the cells, as disclosed herein, preferably are not derived from umbilical cord blood. Nor are they endothelial cells derived from, for example, blood vessels. Rather, the cells are derived of the remaining umbilicus tissues.
  • the cells have been characterized as to several of their cellular, genetic, immunological, and biochemical properties.
  • the cells have been well-characterized by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
  • cell cultures comprising the isolated umbilicus-derived cells of the invention are provided.
  • the cultures are free of maternal cells in preferred embodiments herein.
  • cells that have a normal karyotype and those that maintain their karyotype as they are passaged. Most highly preferred are those cells that have and maintain a normal karyotype throughout passaging until at least after senescence.
  • fetal bovine serum fetal bovine serum
  • Preferred media for culturing and expansion of the cells include Growth Medium, defined herein as medium comprising Dulbecco's Modified Essential Media (also abbreviated DMEM herein).
  • DMEM-low glucose also DMEM-LG herein
  • the DMEM-low glucose is most preferably supplemented with 15% (v/v) fetal bovine serum (e.g.
  • antibiotics/antimycotics preferably penicillin at 100 Units/milliliter, streptomycin at 100 micrograms/milliliter, and amphotericin B at 0.25 micrograms/milliliter; (Invitrogen, Carlsbad, Calif.)), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis Mo.).
  • the Growth Medium can be variously supplemented and altered in any of the ways known in the art, and may be optimized for particular reasons. Also preferred for either isolation of cells from postpartum tissue or growth and culturing of the cells are those media which lack any added serum, and those media which are serum-free and animal protein-free. Such media are particularly preferred for uses in isolating or culturing cells for preclinical or clinical use. Also useful are media which are lacking beta-mercaptoethanol as an additive. In addition, the cells are able to grow in many other culture media, including chemically defined media in the absence of added serum. Several such media are exemplified below.
  • the cells can grow under a variety of environmental conditions.
  • the cells can grow under a wide range of atmospheric conditions.
  • Presently preferred are atmospheres which range from about 5% O 2 to about 20% or more O 2 .
  • the cells grow and expand well in Growth Medium under these conditions, typically in the presence of about 5% CO 2 , and the balance of the atmosphere as nitrogen.
  • the skilled artisan will appreciate that the cells may tolerate broader ranges of conditions in different media, and that optimization for specific purposes may be appropriate.
  • tissue culture vessels include plates, flasks, tubes and the like, made of any of the materials known in the art—e.g. plastic, polystyrene, glass. Such vessels, where coated, are coated with any of a variety of compounds as are known in the art. Presently preferred coated vessels comprise a coating with gelatin, laminin, collagen, polyomithine, polylysine, vitronectin, or fibronectin, for example.
  • the isolated cells of the invention also preferably expand in the presence of from about 2% to about 15% serum, preferably Fetal Bovine Serum, and more preferably defined Fetal Bovine Serum.
  • the cells also expand in the presence or absence of beta-mercaptoethanol, and in the presence or absence of added growth factors including one or more of EGF, bFGF, PDGF, VEGF, IGF-I, and LIF.
  • EGF EGF
  • bFGF fetal growth factor
  • PDGF vascular endothelial growth factor
  • VEGF vascular endothelial growth factor
  • IGF-I interleukin-1
  • the cells can also be grown as attachment-dependent cultures on a variety of surfaces.
  • Presently preferred are surfaces treated with gelatin, more prefereably CELLBIND where culture completely free of animal protein are required.
  • Other coatings are compatible with growth of attachment-dependent cultures such as those provided herein.
  • the cells have excellent doubling and expansion potential and are suitable for use in diagnostic and therapeutic applications because their numbers can be scaled up readily.
  • the isolated cells preferably can double sufficiently to generate yields of greater than about 10 14 cells in less than about 80 days in culture when seeded at about 10 3 cells/cm 2 in a suitable medium. More preferred are cells that can double sufficiently to generate greater than about 10 15 cells in less than about 80 days in culture when seeded at about 5,000 cells/cm 2 . Still more preferred is the isolated cell which can double sufficiently to generate greater than about 10 17 cells in less than about 65 days in culture when seeded at about 5,000 cells/cm 2 .
  • the isolated cells of the invention can undergo extensive doublings. Preferably, the cells undergo at least 30 doublings before reaching senescence. More preferably, at least 40 doublings in culture are attainable. Still more preferable are those cells that can achieve more than 40 doublings before becoming senescent.
  • the cells of the invention are preferably able to undergo division over a longer period of time, than are, for example, human mesenchymal cells, or human fibroblasts, cultured under the same conditions.
  • umbilical cells are isolated in the presence of one or more enzyme activities.
  • a broad range of digestive enzymes for use in cell isolation from tissue are known in the art, including enzymes ranging from those considered weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin).
  • enzymes ranging from those considered weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin).
  • mucolytic enzyme activities e.g. deoxyribonucleases and the neutral protease, dispase
  • mucolytic enzyme activities e.g., metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase)
  • enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities.
  • cells that are isolated in the presence of one or more activities of collagenase, hyaluronidase and dispase. More preferred are those cells isolated in the presence of a collagenase from Clostridium histolyticum , and either of the protease activities, dispase and thermolysin. Still more preferred are cells isolated with collagenase and dispase enzyme activities. Also preferred are such cells isolated in the presence of a hyaluronidase activity, in addition to collagenase and dispase activity.
  • isolated umbilicus-derived cells having specific cell surface marker expression profiles are provided.
  • Preferred cells are characterized in either their production or lack of production of one or more cell surface markers selected from CD10, CD13, CD31, CD44, CD45, CD73, CD90, CD117, CD141, PDGFr-alpha, HLA-A, B, C, and HL-DR, DP, DQ. More preferred are cells that are characterized with respect to their production, or lack thereof, of several (e.g., two, four, five, or eight), or even ten of, or all of, the foregoing, to provide a profile of the cell.
  • the cells produce one or more of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, or HLA-A, B, C. More preferred are cells that express several or all of the foregoing markers. Still more preferred are those cell which express each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C.
  • Preferred cells may also be characterized with respect to markers they do not produce, and such information is also useful in forming a characterization or immunological profile of the cell.
  • the cells do not produce one or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry. More preferred are cells that do not produce several or more of the foregoing. Still more preferred are cells for which production of none of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ can be detected by flow cytometry under the conditions described herein.
  • the cells can be shown to produce several or more of the CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C and concomitantly not produce one, or several, or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ as detected by flow cytometry. More highly preferred are cells that produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C and concomitantly not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ as detected by flow cytometry.
  • Such cells are highly characterized with respect to their production of these cell surface proteins.
  • the characterization of the cells with respect to such production remains substantially constant and does not change substantially with variations in isolation procedure, passage, culture conditions, or even the coating or lack thereof on a tissue culture vessel.
  • Another aspect of the invention provides isolated human umbilicus-derived cells, which are characterized, relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, in their expression of genes for: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morph
  • preferred cells have the cell surface marker characterization described above and are further characterized in their relative gene expression.
  • some preferred cells have the cell surface characterization described above and express a gene for one or more of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; or tumor necrosis factor, alpha-induced protein 3. More preferred are those cells that express a gene for several, (e.g., at least two, four, five or more) of each of the foregoing. The cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes. Also provided are therapeutic cell cultures comprising the isolated human umbilicus-derived cells.
  • UDCs comprising the cell surface marker characterization and which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of one or more genes selected from: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetra
  • the invention provides isolated human umbilicus-derived cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of genes for each of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; t
  • Such cells need not be characterized with respect to their cell surface markers, but such cells preferably are capable of self-renewal and expansion in culture and also have the ability to differentiate in culture. Most preferred are cells which are isolated as adherent cells as described below, however, such cells can also be grown after isolation in a spherical form under certain conditions, thus they are not obligatorily adherent.
  • Certain preferred embodiments include cells as described above which also produce vimentin or alpha-smooth muscle actin. More preferred are cells that produce both vimentin and alpha-smooth muscle actin. The production of these proteins appears to distinguish the cells of the instant invention from hematopoietic cells isolated form umbilical cord blood for example.
  • the above gene expression profiles are substantially stable and do not vary with passaging or normal culturing conditions.
  • a profile may vary when cells are grown under conditions which stimulate or induce differentiation into other phenotypes, for example, or the expression of a different set of genes.
  • the invention also provides therapeutic cell cultures comprising the cell having the cell surface production or gene expression profiles, or both, as described above.
  • Cell banks comprising therapeutic cultures are similarly included with in the scope of the invention.
  • a cell bank may include cultured cells of the invention at various passages, as well as cells of the invention which have been induced to differentiate to different phenotypes. Other cells may be successfully banked either separately or in co-culture with the cells of the invention.
  • a complete cell bank may include the banked cells of a wide variety of individuals.
  • banked cells are stored cryopreserved at ⁇ 180° C. for example. Cells are preferably stored at ⁇ 90° C. in some embodiments.
  • the cells of the invention are readily cryopreserved under a variety of conditions such as are known in the art.
  • the cells lack the cell surface molecules required to substantially stimulate allogeneic lymphocytes in a mixed lymphocyte reaction. In more preferred embodiments, the cells lack the surface molecules required to substantially stimulate CD4 + T-cells in in vitro assessments, or in vivo in allogeneic, syngeneic, or autologous recipients. Still more preferred are those cells that do cause not any substantial adverse immunological consequences for in vivo applications.
  • the therapeutic cell cultures lack detectable amounts of at least two, or several, or all of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as determined by flow cytometry. Those lacking all of the foregoing are most preferred.
  • therapeutic cell cultures which further lack detectable amounts of one or both of the immunomodulating proteins HLA-G and CD178, as determined by flow cytometry. Also preferred are therapeutic cell cultures which express detectable amounts of the immuno-modulating protein PD-L2, as determined by flow cytometry. In one embodiment, the therapeutic cell culture does not substantially stimulate a lymphocyte mediated response in vitro, as compared to allogeneic controls in a mixed lymphocyte reaction.
  • the invention provides isolated human umbilicus-derived cells capable of self-renewal and expansion in culture and which have the potential to differentiate into cells of other phenotypes, wherein the cells do not substantially stimulate allogeneic lymphocytes in a mixed lymphocyte reaction. More preferred are cells which do not substantially stimulate CD4 + T cells, and which produce PD-L2, but not one or more of HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2.
  • the cells also can produce vimentin or alpha-smooth muscle actin in certain embodiments. More preferred are cells producing both vimentin and alpha-smooth muscle actin. Still more preferred are those cells not producing any of HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2.
  • Cells of the invention which secrete useful molecules, for example, growth factors, are presently preferred. Such cells have utility not only for the their cellular properties but for their secreted molecules, for example in conditioned medium, or cell-free lysates.
  • the invention provides isolated human umbilicus-derived cells that secrete one or more of the angiogenic factors MCP-1, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, TPO, or TIMP1.
  • the cells secrete all of the foregoing factors.
  • the cells do not secrete one or more of the angiogenic factors SDF-1 alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA. In certain embodiments they secrete none of SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA.
  • the cells of the invention are defined according to a combination of many of the characteristics provided herein.
  • the invention provides isolated umbilicus-derived cells comprising L-valine-requiring cells derived from mammalian postpartum tissue substantially free of blood.
  • the cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes; for example cardiomyocytes, or their progenitors.
  • Cells may be isolated from umbilicus tissue of any mammal of interest by the techniques provided herein. Human cells are presently preferred.
  • the cells can be grown under a wide range of conditions, including a wide variety of culture media, and environmental conditions. The cells can be grown at least from about 35° C.
  • the cells can be grown in chemically-defined media, or in medium with added mammalian serum, for example fetal bovine serum.
  • the cells also tolerate cryopreservation at various stages.
  • Cells can be maintained frozen, or banked at temperatures preferably at or below ⁇ 80° C. for long periods. Other preferred temperatures range from about ⁇ 90° C. to about ⁇ 180° C. or below.
  • Specialized electric freezers can be used, or cells can be stored in the liquid or vapor phases of nitrogen. Tissues can also be banked prior to the isolation of the cells.
  • the cells are capable of growth in atmospheres containing oxygen from about 5% to at least about 20% and comprise at least one of the following characteristics: the cells have the potential for at least about 40 doublings in culture; the cells preferably are adherent, thus attachment and expansion on a coated or uncoated tissue culture vessel is preferred, wherein a coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, polylysine, vitronectin, or fibronectin;
  • the cells preferably produce of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; more preferred are cells which produce each of tissue factor, vimentin, and alpha-smooth muscle actin; production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A, B, C is also preferred.
  • the cells are also characterized in their lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ, as detected by flow cytometry; more preferable cells lack production of all of these surface markers.
  • interleukin 8 reticulon 1
  • chemokine (C-X-C motif) ligand 1 melanoma growth stimulating activity, alpha
  • chemokine (C-X-C motif) ligand 6 granulocyte chemotactic protein 2
  • chemokine (C-X-C motif) ligand 3 and tumor necrosis factor, alpha-induced protein 3.
  • Preferred cells also have expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein);
  • the cells secrete a variety of biochemically-active factors, such as growth factors, chemokines, cytokines and the like.
  • Preferred cells secrete at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1alpha, RANTES, and TIMP1; preferred cells may alternatively be characterized in their lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1beta, 1309, MDC, and VEGF, as detected by ELISA. These and other characteristics are available to identify and characterize the cells, and distinguish the cells of the invention from others known in the art.
  • the cell comprises two or more of the foregoing characteristics. More preferred are those cells comprising, three, four, or five or more of the characteristics. Still more preferred are those isolated postpartum cells comprising six, seven, or eight or more of the characteristics. Still more preferred presently are those cells comprising all nine of the claimed characteristics.
  • tissue factor a cell that produces at least two of tissue factor, vimentin, and alpha-smooth muscle actin. More preferred are those cells producing all three of the proteins tissue factor, vimentin, and alpha-smooth muscle actin.
  • cell markers are subject to vary somewhat under vastly different growth conditions, and that generally herein described are characterizations in Growth Medium, or variations thereof.
  • Postpartum-derived cells that produce of at least one, two, three, or four of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A, B, C are preferred. More preferred are those cells producing five, six, or seven of these cell surface markers. Still more preferred are postpartum cells that can produce all eight of the foregoing cell surface marker proteins.
  • postpartum cells that lack production of at least one, two, three, or four of the proteins CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ, as detected by flow cytometry are presently preferred.
  • Cells lacking production of at least five, six, seven or eight or more of these markers are also preferred. More preferred are cells which lack production of at least nine or ten of the cell surface markers. Most highly preferred are those cells lacking production of all eleven of the foregoing identifying proteins.
  • Presently preferred cells produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C, and do not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
  • postpartum-derived cells express at least one, two or three of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3. More preferred are those cells which express four or five, and still more preferred are cell capable of expressing all six of the foregoing genes.
  • cells which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression for at least one of the genes corresponding to: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (
  • cells that have, relative to human fibroblasts, mesenchymal stem cells, or ileac crest bone marrow cells, reduced expression of at least 5, 10, 15 or 20 genes corresponding to those listed above.
  • cell with reduced expression of at least 25, 30, or 35 of the genes corresponding to the listed sequences are also more preferred.
  • those postpartum-derived cells having expression that is reduced, relative to that of a human fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, of genes corresponding to 35 or more, 40 or more, or even all of the sequences listed.
  • Preferred postpartum-derived cells secrete at least one, two, three or four of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1.
  • Cells which secrete more than five, six, seven or eight of the listed proteins are also useful and preferred.
  • Cells which can secrete at least nine, ten, eleven or more of the factors are more preferred, as are cells which can secrete twelve or more, or even all thirteen of the proteins in the foregoing list.
  • cells can also be characterized by their lack of secretion of factors into the medium.
  • Umbilicus-derived cells that lack secretion of at least one, two, three or four of TGF-beta2, ANG2, PDGFbb, MIP1beta, 1309, MDC, and VEGF, as detected by ELISA, are presently preferred for use.
  • Cells that are characterized in their lack of secretion of five or six of the foregoing proteins are more preferred.
  • Cells which lack secretion of all seven of the factors listed above are also preferred.
  • therapeutic cell cultures are provided, the cell cultures comprising the isolated cells as described above for use in treating patients in need of angiogenesis-stimulating trophic factors.
  • Such therapeutic cell cultures are also provided for use in treating a patient in need of neural growth stimulating trophic factors.
  • the cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes.
  • the method comprises (a) obtaining human umbilical tissue; (b) removing substantially all of blood to yield a substantially blood-free umbilical tissue, (c) dissociating the tissue by mechanical or enzymatic treatment, or both, (d) resuspending the tissue in a culture medium, and (e) providing growth conditions which allow for the growth of a human umbilicus-derived cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes.
  • Tissue can be obtained from any completed pregnancy, term or less than term, whether delivered vaginally, or through other routes, for example surgical Cesarean section. Obtaining tissue from tissue banks is also considered within the scope of the present invention.
  • the tissue is rendered substantially free of blood by any means known in the art.
  • the blood can be physically removed by washing, rinsing, and diluting and the like, before or after bulk blood removal for example by suctioning or draining.
  • Other means of obtaining a tissue substantially free of blood cells might include enzymatic or chemical treatment.
  • Dissociation of the umbilical tissues can be accomplished by any of the various techniques known in the art, including by mechanical disruption, for example, tissue can be aseptically cut with scissors, or a scalpel, or such tissue can be otherwise minced, blended, ground, or homogenized in any manner that is compatible with recovering intact or viable cells from human tissue.
  • the isolation procedure also utilizes an enzymatic digestion process.
  • Many enzymes are known in the art to be useful for the isolation of individual cells from complex tissue matrices to facilitate growth in culture.
  • a broad range of digestive enzymes for use in cell isolation from tissue is available to the skilled artisan. Ranging from weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin), such enzymes are available commercially.
  • a nonexhaustive list of enzymes compatible herewith includes mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and deoxyribonucleases.
  • enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities.
  • collagenases are known to be useful for isolating various cells from tissues.
  • Deoxyribonucleases can digest single-stranded DNA and can minimize cell-clumping during isolation.
  • Enzymes can be used alone or in combination. Serine protease are preferably used in a sequence following the use of other enzymes as they may degrade the other enzymes being used.
  • Serine proteases may be inhibited with alpha 2 microglobulin in serum and therefore the medium used for digestion is preferably serum-free.
  • EDTA and DNase are commonly used and may improve yields or efficiencies.
  • Preferred methods involve enzymatic treatment with for example collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral protease dispase are used in the dissociating step.
  • the skilled artisan will appreciate that many such enzyme treatments are known in the art for isolating cells from various tissue sources. For example, the LIBERASE Blendzyme (Roche) series of enzyme combinations are very useful and may be used in the instant methods. Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources.
  • the skilled artisan is also well-equipped to assess new, or additional enzymes or enzyme combinations for their utility in isolating the cells of the invention.
  • Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer.
  • the tissue is incubated at 37° C. during the enzyme treatment of the dissociation step. Diluting the digest may also improve yields of cells as cells may be trapped within a viscous digest.
  • the cells can be resuspended after the tissue is dissociated into any culture medium as discussed herein above.
  • Cells may be resuspended following a centrifugation step to separate out the cells from tissue or other debris. Resuspension may involve mechanical methods of resuspending, or simply the addition of culture medium to the cells.
  • a preferred temperature is 37° C., however the temperature may range from about 35° C. to 39° C. depending on the other culture conditions and desired use of the cells or culture.
  • Presently preferred factors to be added for growth on serum-free media include one or more of FGF, EGF, IGF, and PDGF. In more preferred embodiments, two, three or all four of the factors are add to serum free or chemically defined media. In other embodiments, LIF is added to serum-free medium to support or improve growth of the cells.
  • Methods to obtain cells that require L-valine require that cells be cultured in the presence of L-valine. After a cell is obtained, its need for L-valine can be tested and confirmed by growing on D-valine containing medium that lacks the L-isomer.
  • Methods are provided wherein the cells can undergo at least 25, 30, 35, or 40 doublings prior to reaching a senescent state.
  • Methods for deriving cells capable of doubling to reach 10 14 cells or more are provided. Preferred are those methods which derive cells that can double sufficiently to produce at least about 10 14 , 10 15 , 10 16 , or 10 17 or more cells when seeded at from about 10 3 to about 10 6 cells/cm 2 in culture. Preferably these cell numbers are produced within 80, 70, or 60 days or less.
  • Isolated human umbilicus-derived cells derived by the above method are also provided herein.
  • the cells maintain a consistent normal karyotype notwithstanding repeated passaging in certain embodiments.
  • cultures of human umbilicus-derived cells derived by the above method wherein the cultures are free of maternal cells.
  • Therapeutic cultures are provided which are obtained by the methods of the invention.
  • the cells of the instant invention may be useful for the growth, maintenance, support or the like, of other useful cells, either directly or indirectly.
  • the cells may be used to make conditioned media, or may be cocultured with another cells of interest, including direct or indirect ex vivo coculture of cells intended to be used therapeutically in an autologous recipient.
  • the cells may be mixed together, while for indirect coculture, the cells may be cultured in compartments separated by semi-permeable membranes which exclude cell migration.
  • polycarbonate, polyester (PET), and collagen-coated polytetrafluoroethylene (PTFE) membranes of about less 3 microns nominal pore size exclude cell migration and are useful for such purposes.
  • Such membranes for culture systems are able to exchange, for example, soluble growth factors between the umbilical-derived cells and another cell type. Examples are known in the art and some are available commercially (e.g. TRANSWELL culture membranes (Corning Inc, Corning N.Y.)).
  • compositions comprising an umbilicus-derived cell and another therapeutic agent or factor.
  • factors include, but are not limited to, IGF, LIF, PDGF, EGF, FGF, as well as antithrombogenic and antiapoptotic agents.
  • Such therapeutic compositions can further comprise one or more additional cell types.
  • Cell lysates include lysates in which substantially all, and more preferably all of the cells have been lysed, for example by mechanical shear, enzymatic treatment, detergent or other chemical treatment, and the like, or combinations thereof.
  • the resulting lysates include all of the components of the cell and have many utilities, for example in supporting or maintaining growth of the cells of the invention or other cells of interest.
  • the cell lysates can serve as starting material for the purification or enrichment of desirable cell products.
  • cell lysates are further separated into at least a soluble cell fraction and a membrane-enriched cell fraction.
  • soluble cell fractions may be particularly useful in minimizing stimulation of allogeneic PBMCs.
  • soluble cell fractions preferably lack the required antigens to stimulate adverse immunological reactions, for example stimulation of allogeneic lymphocytes, allogeneic CD4 + T cells, or even na ⁇ ve CD4 + T cells.
  • Extracellular matrices derived from the cells for example comprising basement membranes are also useful and are provided herein. Such extracellular material is useful for in vitro assays, for example, angiogenesis assays.
  • extracellular matrix compounds are known for use in augmentation and repair such as applications relating to healing of acute and chronic wounds, or in cosmetic and reconstructive surgery.
  • the extracellular matrix material is often useful where collagen-like properties are desirable.
  • Extracellular matrix material is particularly useful in application requiring elasticity or viscosity because the extracellular matrix appears to be able to provide these attributes.
  • Another application of extracellular matrix is in combination with an implantable therapeutic device. Techniques for cellular disruption range from gentle to harsh, the skilled artisan is well-equipped to select the technique for cell disruption based on the end-use of the cell lysate so obtained.
  • Compositions of the invention also include conditioned culture media as provided herein.
  • Such media have first been used to grow the cells or cultures of the invention, which during growth secrete one or more useful products into the medium.
  • Conditioned medium from these novel cells are useful for many purposes, including for example, supporting the growth of other mammalian cells in need of growth factors or trophic factors secreted into the media by the cells and cultures of the invention, and promoting, for example, angiogenesis.
  • Conditioned media are useful for the secreted growth factors they contain, and preferably the conditioned media of the instant invention contain one or more growth factors useful for supporting the growth of another mammalian cell in need of such growth factors.
  • the use of conditioned media is well understood by those of skill in the art. Continuous culture systems for generating conditioned culture medium are contemplated herein, and the cells of the invention are useful for large scale or continuous production of secreted cellular products such as growth factors.
  • the conditioned media of the invention comprise one or more of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1. More preferred are those conditioned media which comprise several, (e.g., at least two, three or four) of the foregoing. Still more preferred are those which contain many (e.g., at least five, six, seven eight or more) of the growth factors listed. Also preferred are conditioned media which comprise additional secreted molecules of interest.
  • Another embodiment provides a mammalian cell culture comprising the conditioned medium and a mammalian cell in need of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1.
  • Culturing in conditioned media provides for indirect methods of supporting, for example, the growth or maintenance of other cells.
  • indirect methods provide ways of expanding populations of mammalian cells, which due to their growth factor requirements, are difficult to expand in culture in the absence of the conditioned medium.
  • Another aspect of the invention provides co-cultures comprising the umbilicus-derived cells and another mammalian cell of any phenotype.
  • These co-cultures can comprise cells of any phenotype and may, for example, provide a way of expanding populations of mammalian cells, which due to their growth factor requirements are difficult to expand. This is a way of using the properties of the umbilicus-derived cells directly to support the growth of other cells.
  • Such co-cultures are also of use in vivo for various applications, particularly where the use of such cells does not generate an undesired immunological response.
  • the presence of the umbilicus-derived cells can encourage establishment and growth of cells, for example into implanted scaffolds, or at surgical repair sites.
  • the co-cultures comprise a human cell line in addition to the umbilicus-derived cell. Still more preferred would be to use the cells in a family member or person closely related to the donor. More preferred yet would be the co-cultures for use in the individual from whom the cells were derived.
  • the cultures of umbilicus-derived cells are useful in promoting angiogenesis. They are also useful in the treatment of soft tissue disease or injury.
  • the therapeutic cell cultures are grown on a scaffold or matrix.
  • the growth on the scaffold or matrix or similar substrate can be in vivo, in vitro, or a combination of these.
  • the substrate can be surgically applied, for example as a repair to damage soft tissue.
  • the substrate can be seeded with UDCs, or pretreated with UDC-conditioned medium, or cell lysates or soluble cell extracts, or other cell fraction, including an extracellular matrix, prior to surgical use.
  • the cells can help other cells to establish on or within the substrate or its interstices.
  • skin or other soft tissue of dermal or subdermal layers can be grown in vitro on such substrates for later use.
  • a population of cells can be established in vitro on the substrate or scaffold and then surgically attached, grafted, or implanted for further growth. Preestablishing a cell population, particularly one that secretes angiogenic factors may lead to much faster healing.
  • Presently preferred for such scaffolds are nonpermanent, bioabsorbable polymeric matrices and the like.
  • scaffolds or matrices are known in the arts of tissue engineering and wound healing for example, and are useful with the cells and methods herein. Examples include, but are not limited to mats, foams, or self assembling peptides.
  • Preferred mats comprise nonwoven fibers. Nonwoven mats may, for example, be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the trade name VICRYL (Ethicon, Inc., Somerville, N.J.).
  • PGA/PLA synthetic absorbable copolymer of glycolic and lactic acids
  • Foams preferred for use herein include porous foams such as those comprising, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, as discussed in U.S. Pat. No. 6,355,699.
  • Porous foams such as those comprising, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, as discussed in U.S. Pat. No. 6,355,699.
  • Self assembling peptides and hydrogels formed therefrom are known in the art, and include RAD 16 hydrogel, exemplified herein. Such materials are frequently used as supports for growth of cells or tissue.
  • the invention provides higher order structures such as implantable tissue structures comprising the cells of the invention, implantable human tissue matrix comprising the cells, human tissues comprising the cells, and human organs comprising these cells.
  • the invention also provides, in another of its several aspects, injectable therapeutic cells comprising isolated human umbilicus-derived cells capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes, wherein the cells do not substantially stimulate allogeneic lymphocytes in a mixed lymphocyte reaction, and produce PD-L2, but not HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2.
  • This injectable therapeutic cell composition is useful as a therapeutic treatment in any application where undifferentiated cells can be recruited to a site through the circulation.
  • the injectable cell is treated to inactivate tissue factor.
  • tissue factor Such treatment includes any treatment capable of inactivating tissue factor without destroying the integrity of the cell. More preferred are treatments which have no effect on viability, doubling time or expansion.
  • the injectable cell is treated with an anti-tissue factor antibody in one presently preferred embodiment.
  • antithrombogenic agents such as antithrombogenic agents, anti-apoptotic agents, and anti-inflammatory agents may be useful and may be administered in sequence with, or coadministered with the cells, individually or in combinations or two or more such compounds or agents.
  • anti-apoptotic agents may be useful to minimize programmed cell death.
  • agents include but are not limited to EPO, EPO derivatives and analogs, and their salts, TPO, IGF-I, IGF-II, hepatocyte growth factor (HGF), and caspase inhibitors.
  • Anti-inflammatory agents include but are not limited to P38 MAP kinase inhibitors, statins, IL-6 and IL-1 inhibitors, Pemirolast, Tranilast, Remicade, Sirolimus, nonsteroidal anti-inflammatory compounds, for example, Tepoxalin, Tolmetin, and Suprofen.
  • bioactive factors or therapeutic agents which can be coadministered with the therapeutic cells of the invention include, for example, antithrombogenic factors, immunosuppressive or immunomodulatory agents, and antioxidants.
  • immunosuppressive and immunomodulatory agents include calcineurin inhibitors, for example cyclosporine, Tacrolimus, mTOR inhibitors such as Sirolimus or Everolimus; anti-proliferatives such as azathioprine and mycophenolate mofetil; corticosteroids for example prednisolone or hydrocortisone; antibodies such as monoclonal anti-IL-2R ⁇ receptor antibodies, Basiliximab, Daclizumab; polyclonal anti-T-cell antibodies such as anti-thymocyte globulin (ATG), anti-lymphocyte globulin (ALG), and the monoclonal anti-T cell antibody OKT3.
  • calcineurin inhibitors for example cyclosporine, Tacrolimus, mTOR inhibitors such as Sirolimus or Ever
  • Antithrombogenic compounds which can be therapeutically provided in conjunction with the cells of the invention include, for example, heparin, heparin derivatives, urokinase, and PPack (dextrophenylalanine proline arginine chloromethylketone); antithrombin compounds, platelet receptor antagonists, anti-thrombin antibodies, anti-platelet receptor antibodies, aspirin, dipyridamole, protamine, hirudin, prostaglandin inhibitors, and platelet inhibitors.
  • Antioxidants are well known in the art of course and any pharmaceutically acceptable antioxidant may be administered in conjunction with the cells of the invention including probucol; vitamins A, C, and E, coenzyme Q-10, glutathione, L cysteine, N-acetylcysteine, or antioxidant derivative, analogs or salts of the foregoing.
  • kits for the growth and maintenance, the isolation and the use of the umbilical-derived cells are provided.
  • the cells, cell lysates, soluble cell fractions, membrane fractions and matrices can conveniently be employed as parts of kits, for example, for a kit for culture or implantation.
  • kits including the UDCs and additional components, including instructions for growth or maintenance, isolation, or use of the cells or cell fractions, together with for example, matrix (e.g., a scaffold) material, hydrating agents (e.g., physiologically-compatible saline solutions, prepared cell culture media), cell culture substrates (e.g., culture dishes, plates, vials, etc.), cell culture media (whether in liquid or dehydrated form), antibiotic compounds, hormones, and the like.
  • Kits for growth can for example include all of the components of the Growth Medium as used herein, including serum, for example fetal bovine serum. While the kit can include any such components, preferably it includes all ingredients necessary for its intended use.
  • the kit also can include cells (typically cryopreserved), which can be seeded into the lattice as described herein.
  • Kits for isolation will contain everything required to practice the isolation methods as provided herein, except for the umbilicus tissue which should be obtained fresh or frozen from a tissue bank at the time of isolation.
  • the surgical equipment for dissociating the tissue, preferred enzymes, or choices of enzymes in stable form are provided, as are the buffers and medium, cell strainers and the like, as required or preferred for the method as disclosed above.
  • Detailed instructions with optional steps and lists of suppliers of optional or alternative materials are also conveniently provided.
  • Control cells can be included for comparison of the cells isolated to, for example the UDC cultures deposited with the ATCC.
  • Kits for utilizing the umbilicus-derived cells preferably contain populations of the cells, or therapeutic compositions comprising the cells, components and products, or fractions or conditioned media derived from the cells as described above.
  • the kits may include one or more cell populations, including at least UDCs and a pharmaceutically acceptable carrier (liquid, semi-solid or solid).
  • the populations in some embodiments are homogenous or even clonal cell lines of UDCs.
  • the kits include other cell lines for use in coculture.
  • Therapeutic application kits preferably include additional bioactive agents as desired for example anithrombogenic agents, anti-inflammatory agents, antiapoptotic agents, and immunosuppressive or immunomodulatory compounds.
  • the kits also optionally may include a means of administering the cells, for example by injection.
  • kits further may include instructions for use of the cells.
  • Kits prepared for field hospital use such as for military use, may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute injuries.
  • Kits for assays and in vitro methods as described herein may contain one or more of (1) UDCs or fractions, components or products of UDCs, (2) reagents for practicing the in vitro method, (3) other cells or cell populations, as appropriate, for example for cocultures and (4) instructions for conducting the in vitro method.
  • isolated postpartum-derived cells comprising a “signature gene profile.”
  • the signature genes are reticulon, oxidized LDL receptor, and IL-8.
  • the cells comprise a profile which includes mRNA from genes for reticulon, oxidized LDL receptor, and IL-8.
  • the profile comprises genes which are present independent of whether the cells are grown in medium containing serum or medium free of serum, and which further distinguish the postpartum-derived cell from other cell that are totipotent or pluripotent.
  • the isolated postpartum-derived cells further comprise the ability to alter their expression of cell surface markers when grown in medium containing serum relative to that in serum free medium.
  • the markers for PDGFreceptor alpha and HLA-ABC are altered. This plasticity of the cells with respect to their cell surface markers allows the cells to be engineered in certain respects to provided full advantage of the cells desirous properties and to minimize any unwanted or deleterious properties relating to cell surface markers. This is particularly useful in engineering or preparing cells for implantation or for use in grafting where adverse immunological reactions and rejection are concerns.
  • the invention provides a method for the preparation of therapeutic cells or cultures comprising: isolating cells; initially expanding the cells to a useful number in a serum-containing medium which supports cell expansion but in which the cells produce a quantity of the cell surface marker HLA-ABC; transferring the cells to a medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC; and passaging the cells in the medium in which the cells produce a decreased amount of the HLA-ABC, thereby preparing a therapeutic cell or culture.
  • the medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC is a serum-free medium. This is useful for the manufacture and commercial application of the cells and therapeutic cultures particularly where required by regulatory agencies and the like. Such methods are beneficial and desirable for the production of therapeutic cells or cultures for implantation and grafting.
  • the invention provides in another aspect, serum-free media for the expansion of postpartum-derived cells.
  • Preferred media have one or more exogenous growth factors added.
  • Presently preferred are media wherein the one or more growth factors added are bFGF, EGF, or PDGF.
  • Even more preferred are media wherein the one or more added growth factors includes bFGF by itself or in combination with other growth factors beneficial for the growth of the cells or for optimizing the expression of certain cell surface markers.
  • the serum-free media of the invention provide support for the expansion for at least 5 passages of the cells with no reduction in the cells viability and no senescence. More preferred are media which support 10, 15, 20 or more passages of the cells, with the populations still capable of more passaging. Even more preferred are media which support 25, 28, 30, 33 and 35 or more passages. Most preferred are media capable of supporting the cells for about 40 or more passages.
  • therapeutic cultures comprising postpartum-derived cells expanded in serum-free medium
  • Such cells have any useful properties and are more readily suited for commercial application and regulatory approval having, for example, little risk of disease transmission related to foreign animal serum or proteins.
  • the therapeutic cultures comprise cells having a signature gene profile wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum.
  • Such cultures may comprise a single cell type or may consist of multiple cell types including PPDCs and other cell types as additional cells present in the co- or mixed culture.
  • cell culture banks comprising the postpartum-derived cells and cultures of the invention.
  • Postpartum umbilicus tissues were obtained upon birth of either a full term or pre-term pregnancy. Cells were harvested from five separate donors of umbilicus tissue. Different methods of cell isolation were tested for their ability to yield cells with: 1) the potential to differentiate into cells with different phenotypes, a characteristic common to stem cells, or 2) the potential to provide critical trophic factors useful for other cells and tissues.
  • Umbilical cell isolation Umbilical cords were obtained from National Disease Research Interchange (NDRI, Philadelphia, Pa.). The tissues were obtained following normal deliveries. The cell isolation protocols were performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of penicillin at 100 Units/milliliter and streptomycin at 100 milligrams/milliliter, and amphotericin B at 0.25 micrograms/milliliter (Invitrogen Carlsbad, Calif.).
  • PBS phosphate buffered saline
  • the tissues were then mechanically dissociated in 150 cm 2 tissue culture plates in the presence of 50 milliliters of medium (DMEM-Low glucose or DMEM-High glucose; Invitrogen), until the tissue was minced into a fine pulp.
  • the chopped tissues were transferred to 50 milliliter conical tubes (approximately 5 grams of tissue per tube).
  • the tissue was then digested in either DMEM-Low glucose medium or DMEM-High glucose medium, each containing 100 Units/milliliter, streptomycin at 100 milligrams/milliliter, and amphotericin B at 0.25 micrograms/milliliter and the digestion enzymes.
  • C:D collagenase and dispase
  • collagenase Sigma, St Louis, Mo.
  • dispase Invitrogen
  • C:D:H collagenase, dispase and hyaluronidase
  • the cell suspension was filtered through a 70-micron nylon BD FALCON Cell Strainer (BD Biosciences, San Jose, Calif.). An additional 5 milliliters rinse comprising Growth Medium was passed through the strainer. The cell suspension was then passed through a 40-micrometer nylon cell strainer (BD Biosciences, San Jose, Calif.) and chased with a rinse of an additional 5 milliliters of Growth Medium.
  • BD FALCON Cell Strainer BD Biosciences, San Jose, Calif.
  • the filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 ⁇ g for 5 minutes. The supernatant was aspirated and the cells were resuspended in 50 milliliters of fresh growth medium. This process was repeated twice more.
  • the cells isolated from umbilical cord tissues were seeded at 5,000 cells/cm 2 onto gelatin-coated T-75 flasks (Corning Inc., Corning, N.Y.) in Growth Medium. After two days, spent medium and unadhered cells were aspirated from the flasks. Adherent cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with Growth Medium and allowed to grow to confluence (about 10 days from passage 0) to passage 1. On subsequent passages (from passage 1 to 2 etc), cells reached sub-confluence (75-85 percent confluence) in 4-5 days. For these subsequent passages, cells were seeded at 5,000 cells/cm 2 . Cells were grown in a humidified incubator with 5 percent carbon dioxide at 37° C.
  • the lysate was centrifuged (10 minutes at 200 ⁇ g).
  • the cell pellet was resuspended in Complete Minimal Essential Medium (Gibco, Carlsbad Calif.) containing 10 percent fetal bovine serum (Hyclone, Logan Utah), 4 millimolar glutamine (Mediatech Herndon, Va.), penicillin at 100 Units per milliliter and streptomycin at 100 micrograms per milliliter (Gibco, Carlsbad, Calif.).
  • the resuspended cells were centrifuged (10 minutes at 200 ⁇ g), the supernatant was aspirated, and the cell pellet was washed in complete medium.
  • T75 flasks Corning, N.Y.
  • T75 laminin-coated flasks T75 laminin-coated flasks
  • T175 fibronectin-coated flasks both Becton Dickinson, Bedford, Mass.
  • the preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.
  • Populations of cells could be isolated from umbilical tissue efficiently using the enzyme combination collagenase (a metalloprotease), dispase (neutral protease) and hyaluronidase (mucolytic enzyme which breaks down hyaluronic acid).
  • LIBERASE which is a blend of collagenase and a neutral protease, may also be used.
  • Blendzyme 3 which is collagenase (4 Wunsch units/gram) and thermolysin (1714 casein Units/gram), was also used together with hyaluronidase to isolate cells. These cells expanded readily over many passages when cultured in growth expansion medium on gelatin coated plastic.
  • Cells were also isolated from residual blood in the cords, but not cord blood. The presence of cells in blood clots washed from the tissue, which adhere and grow under the conditions used, may be due to cells being released during the dissection process.
  • Hayflick L The longevity of cultured human cells. J. Am. Geriatr. Soc. 22(1):1-12, 1974; Hayflick L. The strategy of senescence. Gerontologist 14(1):37-45), 1974).
  • Tissue culture plastic flasks were coated by adding 20 milliliters 2% (w/v) gelatin (Type B: 225 Bloom; Sigma, St Louis, Mo.) to a T75 flask (Corning Inc., Corning, N.Y.) for 20 minutes at room temperature. After removing the gelatin solution, 10 milliliters phosphate-buffered saline (PBS) (Invitrogen, Carlsbad, Calif.) were added and then aspirated.
  • PBS phosphate-buffered saline
  • cells were seeded at 5,000 cells/cm 2 onto gelatin-coated T 75 flasks in 25 milliliters of fresh Growth Medium. Cells were grown in a standard atmosphere (5 percent carbon dioxide (v/v)) at 37° C. The Growth Medium was changed twice per week. When cells reached about 85 percent confluence they were passaged; this process was repeated until the cells reached senescence.
  • v/v percent carbon dioxide
  • Expansion potential of cell banks at low density The expansion potential of cells banked at passage 10 was also tested. A different set of conditions was used. Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, Md.), umbilicus-derived cells, and placenta-derived cells were tested. These cell populations had been banked at passage 10 previously, having been cultured at 5,000 cells/cm 2 at each passage to that point. The effect of cell density on the cell populations following cell thaw at passage 10 was determined. Cells were thawed under standard conditions, counted using Trypan blue staining. Thawed cells were then seeded at 1,000 cells/cm 2 in Growth Medium.
  • umbilicus-derived cells expanded for more than 40 passages generating cell yields of >1E17 cells in 60 days.
  • MSCs and fibroblasts senesced after ⁇ 25 days and ⁇ 60 days, respectively.
  • both adipose-derived and omental cells expanded for almost 60 days they generated total cell yields of 4.5E12 and 4.24E13 respectively.
  • umbilicus-derived cells expanded much better than the other cell types grown under the same conditions (Table 2-1).
  • the current cell expansion conditions of growing isolated umbilicus-derived cells at densities of about 5,000 cells/cm 2 , in Growth Medium on gelatin-coated or uncoated flasks, under standard atmospheric oxygen, are sufficient to generate large numbers of cells at passage 11.
  • the data suggests that the cells can be readily expanded using lower density culture conditions (e.g. 1,000 cells/cm 2 ).
  • Umbilicus-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for growth.
  • culturing umbilicus-derived cells under standard atmospheric conditions is preferred for generating large pools of cells.
  • umbilicus-derived cell expansion can likewise be altered. This strategy may be used to enhance the proliferative and differentiative capacity of these cell populations.
  • Umbilicus-derived cells (P5) and fibroblasts (P9) were seeded at 5,000 cells/cm 2 in gelatin-coated T75 flasks (Corning, Corning, N.Y.). After 24 hours the medium was removed and the cells were washed with phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) to remove residual medium.
  • PBS phosphate buffered saline
  • the medium was replaced with a modified Growth Medium (DMEM with D-valine (special order Gibco), 15% (v/v) dialyzed fetal bovine serum (Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma), penicillin at 50 Units/milliliter and streptomycin at 50 milligrams/milliliter (Gibco)).
  • DMEM modified Growth Medium
  • D-valine special order Gibco
  • fetal bovine serum Hyclone, Logan, Utah
  • betamercaptoethanol Sigma
  • penicillin 50 Units/milliliter
  • streptomycin 50 milligrams/milliliter
  • umbilicus-derived cells Neither the umbilicus-derived cells nor the fibroblast cells seeded in the D-valine-containing medium proliferated, unlike cells seeded in Growth Medium containing dialyzed serum. Fibroblasts cells changed morphologically, increasing in size and changing shape. All of the cells died and eventually detached from the flask surface after four weeks. Thus, it may be concluded that umbilicus-derived cells require L-valine for cell growth and to maintain long-term viability.
  • Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type. Human cells used in cell therapy should have a normal number (46) of chromosomes with normal structure. To identify umbilicus-derived cell lines that are homogeneous and free from cells of non-umbilical tissue origin, karyotypes of cell samples were analyzed.
  • PPDCs from postpartum tissue of a male neonate were cultured in Growth Media.
  • Postpartum tissue from a male neonate (X, Y) was selected to allow distinction between neonatal-derived cells and maternal derived cells (X, X).
  • Cells were seeded at 5,000 cells per square centimeter in Growth Medium in a T25 flask (Corning, Corning, N.Y.) and expanded to 80% confluence. A T25 flask containing cells was filled to the neck with Growth Media. Samples were delivered to a clinical cytogenetics lab by courier (estimated lab to lab transport time is one hour). Chromosome analysis was performed by the Center for Human & Molecular Genetics at the New Jersey Medical School, Newark, N.J.
  • Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two). A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells were counted and analyzed when a heterogeneous karyotype number (four) was identified.
  • Chromosome analysis identified umbilicus-derived PPDCs whose karyotypes appear normal as interpreted by a clinical cytogenetic laboratory.
  • Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.
  • Characterization of cell surface proteins or “markers” by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions. Postpartum cell lines isolated from the umbilicus were characterized by flow cytometry, providing a profile for the identification of these cell lines.
  • Flow Cytometry Analysis was performed with a FACScalibur instrument (Becton Dickinson, San Jose, Calif.).
  • Antibodies to Cell Surface Markers The following antibodies to cell surface markers were used. TABLE 5-1 Antibodies used in characterizing cell surface markers of UDCs.
  • Antibody Manufacture Catalog Number CD10 BD Pharmingen 555375 (San Diego, CA) CD13 BD Pharmingen 555394 CD31 BD Pharmingen 555446 CD34 BD Pharmingen 555821 CD44 BD Pharmingen 555478 CD45RA BD Pharmingen 555489 CD73 BD Pharmingen 550257 CD90 BD Pharmingen 555596 CD117 BD Pharmingen 340529 CD141 BD Pharmingen 559781 PDGFr-alpha BD Pharmingen 556002 HLA-A, B, C BD Pharmingen 555553 HLA-DR, DP, DQ BD Pharmingen 555558 IgG-FITC Sigma (St. Louis, MO) F-6522 IgG-PE Sigma P-4685
  • Umbilicus-derived cells were analyzed at passages 8, 15, and 20.
  • Umbilicus-derived cells cultured on gelatin-coated flasks was compared to umbilicus cultured on uncoated flasks.
  • Umbilicus-Derived Cell Characterization Umbilicus-derived cells analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by the increased values of fluorescence relative to the IgG control (data not shown). These cells were negative for detectable expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values comparable to the IgG control (data not shown). Variations in florescence values of positive curves were accounted for. The mean (i.e. CD13) and range (i.e. CD90) of the positive curves showed some variation, but the curves appear normal, confirming a homogenous population. Both curves individually exhibited values greater than the IgG control.
  • Umbilical cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were within expected ranges. While the means (i.e. CD13) of the positive curves varied all curves individually exhibited values greater than the IgG control.
  • Umbilicus-derived cells isolated from separate donors analyzed by flow cytometry each showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were accounted for. While the mean (i.e. CD10) of the positive curves varied, both curves individually exhibited values greater than the IgG control.
  • Umbilical cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
  • Umbilicus-derived postpartum cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A, B, C and negative for CD31, CD34, CD45, CD117, CD141 and HLA-DR, DP, DQ. This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, and digestion enzymes used in isolation and preparation of the cells.
  • Oligonucleotide arrays were used to compare gene expression profiles of umbilicus- and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.
  • tissue-derived cells Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, Pa.) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 1. Cells were cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C. with 5% CO 2 .
  • Fibroblasts Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, Md.; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines were cultured in DMEM/F12 medium (Invitrogen, Carlsbad, Calif.) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen)). The cells were grown on standard tissue-treated plastic.
  • hMSC Human Mesenchymal Stem Cells
  • Human Ileac Crest Bone Marrow Cells (ICBM). Human ileac crest bone marrow was received from NDRI with patient consent. The marrow was processed according to the method outlined by Ho, et al. (WO03/025149). The marrow was mixed with lysis buffer (155 mM NH 4 Cl, 10 mM KHCO 3 , and 0.1 mM EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500 ⁇ g.
  • lysis buffer 155 mM NH 4 Cl, 10 mM KHCO 3 , and 0.1 mM EDTA, pH 7.2
  • the supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 mM glutamine.
  • the cells were centrifuged again and the cell pellet was resuspended in fresh medium.
  • the viable mononuclear cells were counted using Trypan-blue exclusion (Sigma, St. Louis, Mo.).
  • the mononuclear cells were seeded in tissue-cultured plastic flasks at 5 ⁇ 10 4 cells/cm 2 .
  • the cells were incubated at 37° C. with 5% CO 2 at either standard atmospheric O 2 or at 5% O 2 .
  • Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture. The adherent cells were maintained in culture.
  • the biotin-labeled cRNA was hybridized with Affymetrix GENECHIP HG-U133A oligonucleotide arrays (Affymetrix, Santa Clara, Calif.). The hybridizations and data collection were performed according to the manufacturer's specifications. The hybridization and data collection was performed according to the manufacturer's specifications. Data analyses were performed using “Significance Analysis of Microarrays” (SAM) version 1.21 computer software (Tusher, V. G. et al., 2001 , Proc. Natl. Acad. Sci. USA 98: 5116-5121).
  • SAM Signal Analysis of Microarrays
  • Table 6-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs.
  • the Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types.
  • the Euclidean distance is inversely proportional to similarity between the expression of the 290 genes.
  • TABLE 6-2 The Euclidean Distances for the Cell Pairs.
  • the Euclidean distance was calculated for the cell types using the 290 genes that were expressed differentially between the cell types. Similarity between the cells is inversely proportional to the Euclidean distance.
  • Tables 6-3, 6-4, and 6-5 show the expression of genes increased in placenta-derived cells (Table 6-3), increased in umbilical cord-derived cells (Table 6-4), and reduced in umbilical cord and placenta-derived cells (Table 6-5).
  • Table 6-3 Genes which are specifically increased in expression in the placenta-derived cells as compared to the other cell lines assayed.
  • Tables 6-6, 6-7, and 6-8 show the expression of genes increased in human fibroblasts (Table 6-6), ICBM cells (Table 6-7), and MSCs (Table 6-8). TABLE 6-6 Genes which were increased in expression in fibroblasts as compared to the other cell lines assayed.
  • the present study was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas. The study also included two different lines of dermal fibroblasts, three lines of mesenchymal stem cells, and three lines of ileac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed on a GENECHIP oligonucleotide array that contained oligonucleotide probes for 22,000 genes.
  • transcripts for 290 genes were present in different amounts in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells. Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord.
  • Postpartum-derived cells generally, and umbilical derived cells, in particular, have distinct gene expression profiles, for example, as compared to other human cells, such as the bone marrow-derived cells and fibroblasts tested here.
  • Gene expression profiles of cells derived from the human umbilical cord were compared with those of cells derived from other sources using an Affymetrix GENECHIP.
  • Six “signature” genes were identified: oxidized LDL receptor 1, interleukin-8 (IL-8), renin, reticulon, chemokine receptor ligand 3 (CXC ligand 3), and granulocyte chemotactic protein 2 (GCP-2). These “signature” genes were expressed at relatively high levels in umbilicus-derived cells.
  • Umbilicus-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) were grown in Growth Medium in gelatin-coated T75 flasks.
  • Mesenchymal Stem Cells (MSCs) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, Md.).
  • IL-8 experiments cells were thawed from liquid nitrogen and plated in gelatin-coated flasks at 5,000 cells/cm 2 , grown for 48 hours in Growth Medium and then grown further for 8 hours in 10 milliliters of serum starvation medium [DMEM-low glucose (Gibco, Carlsbad, Calif.), penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter)(Gibco) and 0.1% (w/v) Bovine Serum Albumin (BSA; Sigma, St. Louis, Mo.)].
  • RNA was then extracted and the supernatants were centrifuged at 150 ⁇ g for 5 minutes to remove cellular debris. Supernatants were frozen at ⁇ 80° C. until ELISA analysis.
  • Cell culture for ELISA assay Cells derived from human umbilical cord, as well as human fibroblasts derived from human neonatal foreskin, were cultured in Growth Medium in gelatin-coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen. Cells were thawed and transferred to 15 milliliter centrifuge tubes. After centrifugation at 150 ⁇ g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 milliliters culture medium and counted. Cells were grown in a 75 cm 2 flask containing 15 milliliters of Growth Medium at 375,000 cell/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation medium was collected at the end of incubation, centrifuged at 14,000 ⁇ g for 5 minutes (and stored at ⁇ 20° C.).
  • ELISA assay The amount of IL-8 secreted by the cells into serum starvation medium was analyzed using ELISA assays (R&D Systems, Minneapolis, Minn.). All assays were conducted according to the instructions provided by the manufacturer.
  • RNA was extracted from confluent umbilicus-derived cells and fibroblasts, or for IL-8 expression, from cells treated as described above. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.). RNA was extracted according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.) and subjected to DNase treatment (2.7 Units/sample) (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at ⁇ 80° C. RNA was also extracted from human umbilical cord.
  • Tissue (30 milligrams) was suspended in 700 microliters of buffer RLT containing beta-mercaptoethanol. Samples were mechanically homogenized and the RNA extraction proceeded according to manufacturer's specification. RNA was extracted with 50 microliters of DEPC-treated water and stored at ⁇ 80° C.
  • Genes identified by cDNA microarray as uniquely regulated in postpartum cells were further investigated using real-time and conventional PCR.
  • PCR was performed on cDNA samples using gene expression products sold under the trade name ASSAYS-ON-DEMAND (Applied Biosystems) gene expression products.
  • Oxidized LDL receptor (Hs00234028); renin (Hs00166915); reticulon (Hs00382515); CXC ligand 3 (Hs00171061); GCP-2 (Hs00605742); IL-8 (Hs00174103); and GAPDH were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C.
  • PCR data were analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).
  • PCR Conventional PCR. Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass.) to confirm the results from real-time PCR. PCR was performed using 2 microliters of cDNA solution (1 ⁇ Taq polymerase (trade name AMPLITAQ GOLD) universal mix PCR reaction buffer (Applied Biosystems) and initial denaturation at 94° C. for 5 minutes. Amplification was optimized for each primer set. For IL-8, CXC ligand 3, and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles); for renin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C.
  • ABI PRISM 7700 Perkin Elmer Applied Biosystems, Boston, Mass.
  • anti-human GROalpha-PE (1:100; Becton Dickinson, Franklin Lakes, N.J.
  • anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, Calif.
  • anti-human oxidized LDL receptor 1 ox-LDL R1; 1:100; Santa Cruz Biotech
  • anti-human NOGO-A (1:100; Santa Cruz, Biotech).
  • fluorescence was visualized using an appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution (no 1° control). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Antibody was added to aliquots as per manufactures specifications and the cells were incubated for in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess antibody. Cells requiring a secondary antibody were resuspended in 100 microliters of 3% FBS. Secondary antibody was added as per manufactures specification and the cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess secondary antibody. Washed cells were resuspended in 0.5 milliliter PBS and analyzed by flow cytometry.
  • oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROalpha (555042; BD Pharmingen, Bedford, Mass.), Mouse IgG1 kappa, (P-4685 and M-5284; Sigma), Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.).
  • FACSCalibur Becton Dickinson San Jose, Calif.
  • results of real-time PCR for selected “signature” genes performed on cDNA from cells derived from human umbilical cord, adult and neonatal fibroblasts, and Mesenchymal Stem Cells (MSCs) indicate that both reticulon and oxidized LDL receptor expression were higher in umbilicus-derived cells as compared to other cells.
  • the data obtained from real-time PCR were analyzed by the ⁇ CT (delta delta CT) method and expressed on a logarithmic scale. No significant differences in the expression levels of CXC ligand 3 and GCP-2 were found between postpartum cells and controls.
  • the results of real-time PCR were confirmed by conventional PCR. Sequencing of PCR products further validated these observations. No significant difference in the expression level of CXC ligand 3 was found between postpartum cells and controls using conventional PCR CXC ligand 3 primers listed in Table 7-1.
  • cytokine, IL-8 The expression of the cytokine, IL-8 in postpartum cells was elevated in both Growth Medium-cultured and serum-starved postpartum-derived cells. All real-time PCR data were validated with conventional PCR and by sequencing PCR products.
  • the phenotypes of cells found within human umbilical cord tissue was analyzed by immunohistochemistry.
  • anti-human GROalpha-PE (1:100; Becton Dickinson, Franklin Lakes, N.J.
  • anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, Calif.
  • anti-human oxidized LDL receptor 1 ox-LDL R1; 1:100; Santa Cruz Biotech
  • anti-human NOGO-A (1:100; Santa Cruz Biotech).
  • Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, Calif.) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 microns thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
  • Immunohistochemistry was performed similar to previous studies (e.g., Messina, et al. (2003) Exper. Neurol. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 1 hour to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (CD34, ox-LDL R1), triton was omitted in all steps of the procedure in order to prevent epitope loss.
  • PBS phosphate-buffered saline
  • Triton Triton X-100
  • Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord.
  • vWF and CD34 expression were restricted to blood vessels contained within the cord.
  • CD34+cells were on the innermost layer (lumen side).
  • Vimentin expression was found throughout the matrix and blood vessels of the cord.
  • SMA was limited to the matrix and outer walls of the artery and vein, but not contained with the vessels themselves.
  • CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.
  • Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD34 are produced in cells within human umbilical cord.
  • HGF hepatocyte growth factor
  • MCP-1 monocyte chemotactic protein 1
  • IL-8 interleukin-8
  • KGF keratinocyte growth factor
  • bFGF basic fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • tissue inhibitor of matrix metalloproteinase 1 TGF1, angiopoietin 2 (ANG2), platelet derived growth factor (PDGFbb), thrombopoietin (TPO), heparin-binding epidermal growth factor (HB-EGF), stromal-derived factor 1alpha (SDF-1 alpha)), neurotrophic/neuroprotective activity (brain-derived neurotrophic factor (BDNF) (Cheng et al. (2003) Dev. Biol.
  • TNF heparin-binding epidermal growth factor
  • SDF-1 alpha stromal-derived factor 1alpha
  • BDNF brain-derived neurotrophic factor
  • IL-6 interleukin-6
  • GCP-2 granulocyte chemotactic protein-2
  • TGFbeta2 transforming growth factor beta2
  • chemokine activity microphage inflammatory protein 1alpha (MIP1alpha), macrophage inflammatory protein 1 beta (MIP1beta), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), 1309, thymus and activation-regulated chemokine (TARC), Eotaxin, macrophage-derived chemokine (MDC), IL-8).
  • the medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 Units/milliliter) and streptomycin (50 micrograms/milliliter, Gibco)) for 8 hours.
  • DMEM-low glucose (Gibco) 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 Units/milliliter) and streptomycin (50 micrograms/milliliter, Gibco)
  • Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000 ⁇ g for 5 minutes and stored at ⁇ 20° C.
  • Chemokines (MIP1alpha, MIP1beta, MCP-1, Rantes, I309, TARC, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, TIMP1, ANG2, PDGFbb, TPO, HB-EGF were measured using SearchLight Proteome Arrays (Pierce Biotechnology Inc.).
  • the Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to sixteen proteins per well.
  • the arrays are produced by spotting a 2 ⁇ 2, 3 ⁇ 3, or 4 ⁇ 4 pattern of four to sixteen different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture the chemiluminescent signal generated at each spot within each well of the plate. The signal generated at each spot is proportional to the amount of target protein in the original standard or sample.
  • MCP-1 and IL-6 were secreted by umbilicus-derived PPDCs and dermal fibroblasts (Table 9-1). SDF-1alpha and GCP-2 were secreted by fibroblasts. GCP-2 and IL-8 were secreted by umbilicus-derived PPDCs. TGF-beta2 was not detected from either cell type by ELISA.
  • TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1beta, MCP1, RANTES, I309, TARC, MDC, and IL-8 were secreted from umbilicus-derived PPDCs (Tables 9-2 and 9-3). No Ang2, VEGF, or PDGFbb were detected.
  • Umbilicus-derived cells secreted a number of trophic factors. Some of these trophic factors, such as HGF, bFGF, MCP-1 and IL-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL-6, have important roles in neural regeneration or protection.
  • Postpartum cell lines were evaluated in vitro for their immunological characteristics in an effort to predict the immunological response, if any, these cells would elicit upon in vivo transplantation. Postpartum cell lines were assayed by flow cytometry for the expression of HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2. These proteins are expressed by antigen-presenting cells (APC) and are required for the direct stimulation of na ⁇ ve CD4 + T cells (Abbas & Lichtman, C ELLULAR AND M OLECULAR I MMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171).
  • APC antigen-presenting cells
  • the cell lines were also analyzed by flow cytometry for the expression of HLA-G (Abbas & Lichtman, C ELLULAR AND M OLECULAR I MMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171), CD178 (Coumans, et. al., (1999) Journal of Immunological Methods 224, 185-196), and PD-L2 (Abbas & Lichtman, C ELLULAR AND M OLECULAR I MMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171; Brown, et. al. (2003) The Journal of Immunology 170, 1257-1266).
  • HLA-G Abbas & Lichtman, C ELLULAR AND M OLECULAR I MMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171
  • Brown, et. al. (2003) The Journal of Immunology 170, 1257-1266 The Journal of Immunology 170, 1257-1266.
  • Cell culture Cells were cultured in Growth Media in T75 flasks (Corning, Corning, N.Y.) coated with 2% gelatin (Sigma, St. Louis, Mo.) until confluent.
  • Antibody Staining Cells were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) and detached with Trypsin/EDTA (Gibco, Carlsbad, Mo.). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1 ⁇ 10 7 per milliliter.
  • Antibody (Table 10-1) was added to one hundred microliters of cell suspension as per manufacturer's specifications and incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody.
  • PBMCs Peripheral blood mononuclear cells
  • Stimulator (donor) allogeneic PBMC, autologous PBMC, and postpartum cell lines were treated with mitomycin C.
  • Autologous and mitomycin C-treated stimulator cells were added to responder (recipient) PBMCs and cultured for 4 days. After incubation, [ 3 H]thymidine was added to each sample and cultured for 18 hours. Following harvest of the cells, radiolabeled DNA was extracted, and [ 3 H]-thymidine incorporation was measured using a scintillation counter. Reactions were performed in triplicate using two-cell culture plates with three receivers per plate
  • the stimulation index for the allogeneic donor was calculated as the mean proliferation of the receiver plus mitomycin C-treated allogeneic donor divided by the baseline proliferation of the receiver.
  • the stimulation index of the postpartum cells was calculated as the mean proliferation of the receiver plus mitomycin C-treated postpartum cell line divided by the baseline proliferation of the receiver.
  • umbilical cells were negative for production of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, because the fluorescence values were comparable to the IgG control. This indicates that umbilical cell lines lack the cell surface molecules required to directly stimulate CD4 + T cells.
  • the umbilical cells analyzed by flow cytometry were positive for expression of PD-L2, as reflected in the increase in fluorescence relative to the IgG control.
  • the cells were negative for expression of CD178 and HLA-G, as noted by fluorescence values consistent with the IgG control.
  • Tissue factor a membrane-bound procoagulant glycoprotein
  • Tissue factor also plays an important role in embryonic vessel formation, for example, in the formation of the primitive vascular wall (Brodsky et al. (2002) Exp. Nephrol. 10:299-306).
  • umbilicus-derived PPDCs were evaluated for tissue factor expression and for their ability to initiate plasma clotting.
  • Human Tissue factor Human tissue factor (SIMPLASTIN, Organon Teknika Corporation, Durham, N.C.), was reconstituted with 20 milliliters distilled water. The stock solution was serially diluted (1:2) in eight tubes. Normal human plasma (George King Bio-Medical, Overland Park, Kans.) was thawed at 37° C. in a water bath and then stored in ice before use. 100 microliters phosphate buffered saline (PBS), 10 microliters diluted SIMPLASTIN, 30 microliters 0.1 Molar calcium chloride, and 100 microliters of normal human plasma were added to each well of a 96-well plate. A negative control well did not receive any SIMPLASTIN. The plate was immediately placed in a temperature-controlled microplate reader and absorbance measured at 405 nanometer at 40 second intervals for 30 minutes.
  • PBS phosphate buffered saline
  • SIMPLASTIN 10 microliters diluted SIMPLASTIN
  • 30 microliters 0.1 Molar calcium chloride
  • J-82 and umbilicus-derived cells were grown in Iscove's modified Dulbecco's medium (IMDM; Gibco, Carlsbad, Calif.) containing 10% (v/v) fetal bovine serum (FBS; Hyclone, Logan Utah), 1 millimolar sodium pyruvate (Sigma Chemical, St. Louis, Mo.), 2 millimolar L-Glutamine (Mediatech Herndon, Va.), 1 ⁇ non-essential amino acids (Mediatech Herndon, Va.). At about 70% confluence, cells were transferred at 100,000, 50,000 and 25,000 cells/well to wells of 96-well plate.
  • IMDM Iscove's modified Dulbecco's medium
  • FBS fetal bovine serum
  • FBS fetal bovine serum
  • 1 millimolar sodium pyruvate Sigma Chemical, St. Louis, Mo.
  • 2 millimolar L-Glutamine Mediatech Herndon, Va.
  • 1 ⁇ non-essential amino acids Mediatech Herndon,
  • Umbilicus-derived cells were cultured in Growth Medium in gelatin-coated T75 flasks (Corning, Corning, N.Y.). Umbilicus-derived cells at passage 18 were transferred to wells at a density of 50,000 cells/well. Culture medium was removed from each well after centrifugation at 150 ⁇ g for 5 minutes. Cells were suspended in PBS without calcium and magnesium. Cells incubated with anti-tissue factor antibody cells were incubated with 20 micrograms/milliliter CNTO 859 (Centocor, Malvern, Pa.) for 30 minutes. Calcium chloride (30 microliters) was added to each well. The plate was promptly placed in a temperature-controlled microplate reader and absorbance was measured at 405 nanometers at 40 second intervals for 30 minutes.
  • Antibody Staining Cells were washed in PBS and detached from the flask with Trypsin/EDTA (Gibco, Carlsbad, Calif.). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1 ⁇ 10 7 per milliliter. Antibody was added to 100 microliters of cell suspension according to the manufacturer's specifications. The cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS, then centrifuged at 150 ⁇ g for 5 minutes to remove unbound antibody. Cells were resuspended in 100 microliters of 3% FBS and secondary antibody added in accordance with the manufacturer's instructions.
  • Flow Cytometry Analysis was performed with a FACSCalibur instrument (Becton Dickinson, San Jose, Calif.).
  • Flow cytometry analysis revealed that umbilicus-derived postpartum cells are less active in promoting plasma clotting than the J82 cells.
  • a plasma clotting assay demonstrated that the tissue factor present in the umbilicus-derived cells was active, clotting took longer than with the J-82 cells, as evidenced by the longer time to half-maximal absorbance (T1 ⁇ 2 to max; Table 11-1).
  • the T1 ⁇ 2 to max is inversely proportional to the number of J-82 cells.
  • Umbilicus-derived cells decreased the clotting rate as indicated by the T1 ⁇ 2 to max. Clotting was observed with both early (P5) and late (P18) passaged cells.
  • Umbilicus-derived PPDCs produce some tissue factor, but the addition of an antibody against tissue factor can inhibit the clotting activity of the tissue factor.
  • Tissue factor is normally found on cells in a conformation that is inactive, but which is activated by mechanical or chemical (e.g., LPS) stress (Sakariassen et al. (2001) Thromb. Res. 104:149-74; Engstad et al. (2002) Int. Immunopharmacol. 2:1585-97).
  • LPS low-se.g., LPS
  • tissue factor has been associated with angiogenic activity. For this reason, tissue factor activity may be beneficial when umbilicus-derived PPDCs are transplanted in tissue, but should be inhibited when PPDCs are injected intravenously.
  • Cells derived from the postpartum umbilical cord are useful for regenerative therapies.
  • Tissue produced by SCID mice following transplantation of a biodegradable material with and without umbilicus-derived cells was evaluated.
  • the materials evaluated were VNW nonwoven scaffolds, 35/65 PCL/PGA foam, and a self-assembling peptide hydrogel.
  • Umbilicus-derived cells were grown in Growth Medium in gelatin-coated flasks.
  • a nonwoven scaffold was prepared using a traditional needle punching technique as described below.
  • Fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), were obtained from Ethicon, Inc. (Somerville, N.J.). The fibers were filaments of approximately 20 microns in diameter. The fibers were cut and crimped into substantially uniform 2-inch lengths to form 2-inch staple fibers.
  • a dry lay needle-punched nonwoven matrix was prepared utilizing the PGA/PLAstaple fibers, hereinafter “VNW”. The staple fibers were opened and carded on standard nonwoven machinery. The resulting mat was in the form of webbed staple fibers. The webbed staple fibers were needle-punched to form the dry lay needle-punched nonwoven scaffold.
  • the nonwoven scaffold was rinsed in water followed by another incubation in ethanol to remove any residual chemicals or processing aids used during the manufacturing process.
  • Foams composed of 35/65 poly(epsilon-caprolactone)/poly(glycolic acid) (35/65 PCL/PGA) copolymer, were formed by the process of lyophilization, as described in U.S. Pat. No. 6,355,699.
  • a self assembling peptide hydrogel (RAD16 self-assembling peptides (3D Matrix, Cambridge, Mass.)) was obtained as a sterile 1% (w/v) solution in water.
  • the self-assembling peptide solution was mixed 1:1 with 1 ⁇ 10 6 cells in 10% (w/v) sucrose (Sigma, St Louis, Mo.), 10 millimolar HEPES (pH about 7), in Dulbecco's modified medium (DMEM; Gibco) immediately before use.
  • the final concentration of cells in the self-assembling peptide hydrogel was 1 ⁇ 10 6 cells/100 microliters.
  • VNW scaffold +1 ⁇ 10 6 umbilicus-derived cells
  • mice Male mice ( Mus musculus ) (Fox Chase SCID; Harlan Sprague Dawley, Inc., Indianapolis, Ind.), were used at 5 weeks of age. All handling of the SCID mice took place under a hood. Each animal was individually weighed, and anesthetized with an intraperitoneal injection of a mixture of 60 milligram/kilogram KETASET (ketamine hydrochloride) (Aveco Co., Inc., Fort Dodge, Iowa), 10 milligram/kilogram ROMPUN (xylazine) (Mobay Corp., Shawnee, Kans.) and saline.
  • KETASET ketamine hydrochloride
  • ROMPUN xylazine
  • Subcutaneous Implantation Technique Four skin incisions, each approximately 1 centimeter in length, were made on the dorsa of the mice. Two cranial implantation sites, one each to the left and right of the vertebral column, were located transversely over the dorsal lateral thoracic region, about 5 millimeters caudal to the palpated inferior edge of the scapula. Two additional implants, one on each side of the midline, were placed transversely over the gluteal muscle area at the caudal sacro-lumbar level, about 5 millimeters caudal to the palpated iliac crest. Implants were randomly placed in these sites.
  • the skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected in the case of the self-assembling peptide) about 1 centimeter caudal to the incision.
  • the appropriate test material was implanted into the subcutaneous space. The skin incision was closed with metal clips.
  • mice were euthanized by carbon dioxide inhalation at designated intervals.
  • the subcutaneous implants with the overlying skin were excised and frozen for histology.
  • Nonwoven scaffolds seeded with umbilicus-derived cells showed increased matrix deposition and mature blood vessels.
  • pancreatic islets Transplantation of pancreatic islets to the kidney capsule is routinely performed to evaluate transplantation methodologies for the treatment of diabetes (Refaie et al., 1998).
  • other cells may be differentiated into insulin secreting cells capable of blood glucose homeostasis. The suitability of umbilicus-derived cells for this purpose was evaluated.
  • Umbilicus-derived cells (isolate 1, P10) were removed from liquid nitrogen storage and grown in Growth Medium on gelatin (Sigma)-coated T225 (Corning, Corning, N.Y.) flasks until confluent.
  • the culture medium on umbilicus-derived cells was replaced with Ham's F12 medium (Gibco) containing 10 millimolar nicotinamide (Sigma), 25 millimolar glucose (Sigma), 10 nanogram/milliliter EGF (PeproTech, Rocky Hill, N.J.), 20 nanogram/milliliter bFGF (PeproTech) and 15 millimolar GLP-1 (Sigma) and cells were further cultured for 2 weeks.
  • Ham's F12 medium Gibco
  • 10 millimolar nicotinamide Sigma
  • 25 millimolar glucose Sigma
  • 10 nanogram/milliliter EGF PeproTech, Rocky Hill, N.J.
  • 20 nanogram/milliliter bFGF PeproTech
  • 15 millimolar GLP-1 Sigma
  • CD34+ cells were purchased from Cambrex, Walkersville, Md. (lot 1F0174 donor 7956). CD34+ cells were thawed and washed in DMEM medium.
  • the cell suspension was washed twice in DMEM. Cell number and viability was estimated after Trypan blue (Sigma) staining using a hemocytometer. Aliquots of the cell suspension containing ⁇ 300,000 viable cells were centrifuged at 150 ⁇ g and the cells resuspended in approximately 6 microliters of DMEM and drawn into a 20 microliter pipette tip connected to a 1 milliliter syringe. The tip of the pipette tip containing the cells was clamped using a small Ligaclip (Ethicon Endosurgery, Cincinnati Ohio).
  • mice Mus musculus
  • Fox Chase SCID/Male Hard Sprague Dawley, Inc., Indianapolis, Ind.
  • All handling of the SCID mice took place under a hood.
  • the mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligram/kilogram KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligram/kilogram ROMPUN (xylazine, Mobay Corp., Shawnee, Kans.) and saline.
  • KETASET ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa
  • ROMPUN milligram/kilogram
  • a capsule lance (modified glass pipette tip) was used to create a space beneath the kidney capsule into which the cells were introduced.
  • the cells were injected via a syringe with a micropipette tip attached.
  • the pocket was closed by passing an ophthalmic cautery pen (Aaron Medical Industries, St. Russia, Fla.) over the opening (not touching the kidney).
  • the kidney was placed back in the correct anatomical position, and the muscle layer sutured closed.
  • the skin was closed with wound clips.
  • the experimental design comprised one cell transplantation in each mouse; four treatments with n-value of 4 per treatment; and three time-points (1, 14 & 30 days).
  • mice were euthanized by carbon dioxide inhalation at their designated intervals.
  • the kidney implantation sites were excised and frozen for histology.
  • Sections were incubated in anti-human nuclei antibody (Chemicon International, Temecula, Calif.) diluted 1:100 in blocking solution for one hour. Sections were washed with PBS and incubated in florescent labeled goat anti-mouse IgG antibody (Molecular Probes Eugene, Oreg.), diluted 1:200 in blocking solution for 30 minutes in absence of light. Sections were washed in PBS and incubated in 10 micromolar DAPI (Molecular Probes) for five minutes. Sections were washed in PBS and examined by fluorescent microscopy.
  • Tri-Chrome Staining Frozen kidney implantation sites were embedded on edge in O.C.T. Compound (Sakura Inc.). The kidney tissue was trimmed by cryosectioning to yield a five-micron section of the implantation site and adjacent tissue. Yielded sections were fixed in 10% neutral buffered formalin (Richard-Allan Scientific) for 15 minutes. Sections were stained tri-chrome (Poly Scientific) using manufactures methods.
  • the viability of the umbilicus-derived cells was ⁇ 75%; that of the CD34 + cells was 95%.
  • Initial attempts to transplant 1 ⁇ 10 6 viable cells were unsuccessful due to the kidney capsule not being large enough to accommodate the cells.
  • Cells were transplanted within 3 hours of trypsinization.
  • the localization of postpartum cells under the kidney capsule was observed microscopically. There were no apparent differences in the number and distribution of umbilicus-derived cells with or without CD34 + cells at each time point. There was an apparent decrease in cell numbers over time.
  • PPDCs umbilicus-derived postpartum cells
  • Umbilicus-derived PPDCs were isolated and expanded as described in Example 1 and 2.
  • DMEM/F12 medium Invitrogen, Carlsbad, Calif.
  • FBS fetal bovine serum
  • bFGF basic fibroblast growth factor
  • EGF epidermal growth factor
  • penicillin 50 Units/milliliter
  • streptomycin 50 micrograms/milliliter
  • Cells were then cultured for 1-6 hours in an induction medium which was comprised of DMEM/F12 (serum-free) containing 200 millimolar butylated hydroxyanisole, 10 micromolar potassium chloride, 5 milligram/milliliter insulin, 10 micromolar forskolin, 4 micromolar valproic acid, and 2 micromolar hydrocortisone (all chemicals from Sigma, St. Louis, Mo.). Cells were then fixed in cold ( ⁇ 20° C.) 100% methanol and immunocytochemistry was performed (see methods below) to assess human nestin protein expression.
  • DMEM/F12 serum-free
  • PPDCs umbilicus, P11
  • adult human dermal fibroblasts (1F1853, P11) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached.
  • Cells were then trypsinized and seeded at similar density as in (A), but onto (1) 24 well tissue culture-treated plates (TCP, Falcon brand, VWR International), (2) TCP wells+2% (w/v) gelatin adsorbed for 1 hour at room temperature, or (3) TCP wells+20 micrograms/milliliter adsorbed mouse laminin (adsorbed for a minimum of 2 hours at 37° C.; Invitrogen).
  • NPE Neural Progenitor Expansion medium
  • Umbilicus-derived PPDCs P11
  • adult human dermal fibroblasts P11; 1F1853; Cambrex
  • Growth Medium 5,000 cells/cm 2 until sub-confluence (75%) was reached.
  • Cells were then trypsinized and seeded at 2,000 cells/cm 2 , but onto 24 well plates coated with laminin (BD Biosciences, Franklin Lakes, N.J.) in the presence of NPE media supplemented with bFGF (20 nanograms/milliliter; Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter; Peprotech) (whole media composition further referred to as NPE+F+E).
  • Umbilicus-derived PPDCs re thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-ce (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm 2 , onto 24 well laminin-coated plates (BD Biosciences) in the presence of NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter). In addition, some wells contained NPE+F+E+2% FBS or 10% FBS. After four days of “pre-differentiation” conditions, all media were removed and samples were switched to NPE medium supplemented with sonic hedgehog (SHH; 200 nanograms/milliliter; Sigma, St.
  • SHH sonic hedgehog
  • FGF8 100 nanograms/milliliter; Peprotech
  • BDNF 40 nanograms/milliliter; Sigma
  • GDNF 20 nanograms/milliliter; Sigma
  • retinoic acid 1 micromolar; Sigma.
  • cultures were fixed with ice-cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nestin, GFAP, TuJ1, desmin, and alpha-smooth muscle actin expression.
  • Neural Progenitor Co-Culture Protocol Adult rat hippocampal progenitors (062603) were plated as neurospheres or single cells (10,000 cells/well) onto laminin-coated 24 well dishes (BD Biosciences) in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter).
  • Umbilicus-derived PPDCs (P11) were thawed and culture expanded in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter) at 5,000 cells/cm 2 for a period of 48 hours. Cells were then trypsinized and seeded at 2,500 cells/well onto existing cultures of neural progenitors. The existing medium was exchanged for fresh medium. Four days later, cultures were fixed with ice-cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nuclear protein (hNuc, Chemicon) (Table 14-1 above) to identify PPDCs.
  • Immunocytochemistry was performed using the antibodies listed in Table 14-1. Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature.
  • PBS phosphate-buffered saline
  • Triton Triton X-100
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). Positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Umbilicus-derived cell isolates as well as human fibroblasts and rodent neural progenitors (as negative and positive control cell types, respectively), were plated on laminin (neural-promoting)-coated dishes and exposed to 13 different growth conditions (and two control conditions) known to promote differentiation of neural progenitors into neurons and astrocytes.
  • laminin neural-promoting
  • two control conditions two conditions known to promote differentiation of neural progenitors into neurons and astrocytes.
  • two conditions were added to examine the influence of GDF5, and BMP7 on PPDC differentiation.
  • a two-step differentiation approach was taken, where the cells were first placed in neural progenitor expansion conditions for a period of 6 days followed by full differentiation conditions for 7 days. Morphologically, umbilical cells exhibited fundamental changes in cell morphology throughout the time-course of this procedure.
  • Umbilicus-derived cells were plated onto cultures of rat neural progenitors seeded two days earlier in neural expansion conditions (NPE+F+E). While visual confirmation of plated umbilicus proved that these cells were plated as single cells, human-specific nuclear staining (hNuc) 4 days post-plating (6 days total length of experiment) showed that they tended to ball up and avoid contact with the neural progenitors. Furthermore, where umbilicus cells attached, these cells spread out and appeared to be innervated by differentiated neurons that were of rat origin suggesting that the umbilical cells may have differentiated into muscle cells. This observation was based upon morphology under phase contrast microscopy.
  • neural progenitors extended neurites to PPDCs in a way that indicates the progenitors differentiated into neurons and innervated the PPDCs.
  • cultures of neural progenitors and PPDCs had more cells of rat origin and larger amounts of differentiation than control cultures of neural progenitors alone, further indicating that plated PPDCs provided soluble factors and or contact-dependent mechanisms that stimulated neural progenitor survival, proliferation, and/or differentiation.
  • umbilicus-derived cells The ability of umbilicus-derived cells to undergo long-term differentiation into neural lineage cells was evaluated.
  • Umbilicus-derived Cell Thaw and Plating Frozen aliquots of umbilicus-derived cells P11 and P12, previously grown in Growth Medium were thawed and plated at 5,000 cells/cm 2 in T-75 flasks coated with laminin (BD, Franklin Lakes, N.J.) in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine (4 millimolar), penicillin (50 Units/milliliter), and streptomycin (50 micrograms/milliliters), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) media.
  • NPE Neural Progenitor Expansion
  • NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.), herein referred to as NPE+bFGF+EGF.
  • Immunocytochemistry After a period of one month, all flasks were fixed with cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against TuJ1 (BIII Tubulin; 1:500; Sigma, St. Louis, Mo.) and GFAP (glial fibrillary acidic protein; 1:2000; DakoCytomation, Carpinteria, Calif.).
  • TuJ1 BIII Tubulin; 1:500; Sigma, St. Louis, Mo.
  • GFAP glial fibrillary acidic protein; 1:2000; DakoCytomation, Carpinteria, Calif.
  • cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens.
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • PPDCs umbilicus-derived postpartum cells
  • NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.), herein referred to as NPE+bFGF+EGF.
  • tissue minced with a scalpel Following wash, the overlying meninges were removed, and the tissue minced with a scalpel. Minced tissue was collected and trypsin/EDTA (Invitrogen) added as 75% of the total volume. DNAse (100 microliters per 8 milliliters total volume, Sigma, St. Louis, Mo.) was also added. Next, the tissue/medium was sequentially passed through an 18 gauge needle, 20 gauge needle, and finally a 25 gauge needle one time each (all needles from Becton Dickinson, Franklin Lakes, N.J.). The mixture was centrifuged for 3 minutes at 250 ⁇ g. Supernatant was removed, fresh NPE+bFGF+EGF was added and the pellet resuspended.
  • trypsin/EDTA Invitrogen
  • the resultant cell suspension was passed through a 40 micron cell strainer (Becton Dickinson), plated on laminin-coated T-75 flasks (Becton Dickinson) or low cluster 24-well plates (Becton Dickinson), and grown in NPE+bFGF+EGF media until sufficient cell numbers were obtained for the studies outlined.
  • Postpartum-derived cells previously grown in Growth Media were plated at 5,000 cells/transwell insert (sized for 24 well plate) and grown for a period of one week in Growth Media in inserts to achieve confluence.
  • Neural progenitor Plating Neural progenitors, grown as neurospheres or as single cells, were seeded onto laminin-coated 24 well plates at an approximate density of 2,000 cells/well in NPE+bFGF+EGF for a period of one day to promote cellular attachment. One day later, transwell inserts containing postpartum cells were added according to the following scheme:
  • Immunocytochemistry After 7 days in co-culture, all conditions were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the epitopes listed in Table 16-1. Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature.
  • PBS phosphate-buffered saline
  • Triton Triton X-100
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Hippocampal neural progenitor differentiation was examined and quantified. A minimum of 1,000 cells were counted per condition, or if less, the total number of cells observed in that condition. The percentage of cells positive for a given stain was assessed by dividing the number of positive cells by the total number of cells as determined by DAPI (nuclear) staining.
  • Mass Spectrometry Analysis & 2D Gel Electrophoresis In order to identify unique, secreted factors as a result of co-culture, conditioned media samples taken prior to culture fixation were frozen down at ⁇ 80° C. overnight. Samples were then applied to ultrafiltration spin devices (nominal MW cutoff 30 kD). Retentate was subjected to immunoaffinity chromatography (anti-Human-albumin; IgY) (immunoaffinity did not remove albumin from the samples). Filtrate was analyzed by MALDI-TOFF. The eluate was applied to Cibachron Blue affinity chromatography. Samples were analyzed by SDS-PAGE and 2D gel electrophoresis.
  • Umbilical Co-culture Stimulates Adult Neural Progenitor Differentiation. Following culture with umbilicus-derived postpartum cells, co-cultured neural progenitor cells derived from adult rat hippocampus exhibited significant differentiation along all three major lineages in the central nervous system. This effect was clearly observed after five days in co-culture, with numerous cells elaborating complex processes and losing their phase bright features characteristic of dividing progenitor cells. Conversely, neural progenitors grown alone in the absence of bFGF and EGF appeared unhealthy and survival was limited.
  • cultures were stained for markers indicative of undifferentiated stem and progenitor cells (nestin), immature and mature neurons (TuJ1), astrocytes (GFAP), and mature oligodendrocytes (MBP). Differentiation along all three lineages was confirmed while control conditions did not exhibit significant differentiation as evidenced by retention of nestin-positive staining amongst the majority of cells.
  • results presented in this example indicate that the differentiation of adult neural progenitor cells following co-culture with umbilicus-derived postpartum cells is particularly profound. Specifically, a significant percentage of mature oligodendrocytes were generated in co-cultures of umbilical cells. In view of the lack of contact between umbilical cells and the neural progenitors, this result appears to be a function of soluble factors released from the umbilical cells (trophic effect).
  • Angiogenesis or the formation of new vasculature, is necessary for the growth of new tissue. Induction of angiogenesis is an important therapeutic goal in many pathological conditions.
  • the angiogenic activity of umbilicus-derived cells in in vitro assays was examined. A well-established method of assessing angiogenic activity involving seeding endothelial cells onto a culture plate coated with a basement membrane extract (Nicosia and Ottinetti (1990) In Vitro Cell Dev. Biol. 26(2): 119-28), was utilized. Treating endothelial cells on such basement membranes or extracellular matrix material with angiogenic factors will stimulate the cells to form a network that is similar to capillaries.
  • Umbilicus-derived cells Human umbilical cords were received and cells were isolated as previously described (Example 1). Cells were cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C. with 5% CO 2 . Cells used for experiments were between passages 4 and 12.
  • UDCs Actively growing UDCs were trypsinized, counted, and seeded onto 6.5 millimeter diameter tissue culture inserts (COSTAR TRANSWELL, Corning Inc., Corning, N.Y.) at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium with 5% CO 2 at 37° C.
  • COSTAR TRANSWELL Corning Inc., Corning, N.Y.
  • hMSC Human mesenchymal stem cells
  • Actively growing MSCs were trypsinized and counted and seeded onto 6.5 millimeter diameter tissue culture inserts (Corning, Corning, N.Y.) at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium with 5% CO 2 at 37° C.
  • HUVEC Human umbilical vein endothelial cells
  • HUVEC Human umbilical vein endothelial cells
  • Cambrex Walkersville, Md.
  • Cells were grown in separate cultures in either EBM or EGM endothelial cell media (Cambrex).
  • EBM EGM endothelial cell media
  • Cells were grown on standard tissue cultured plastic with 5% CO 2 at 37° C. Cells used in the assay ranged from passages 4 to 10.
  • HCAEC Human coronary artery endothelial cells
  • Angiogenesis Assays on Extracellular Matrix Culture plates were coated with extracellular matrix material according to manufacturer's specifications. Briefly, extracellular matrix material (MATRIGEL, BD Discovery Labware, Bedford, Mass.) was thawed at 4° C. and approximately 250 microliters were distributed onto each well of a chilled 24-well culture plate (Corning). The plate was then incubated at 37° C. for 30 minutes to allow the material to solidify. Actively-growing endothelial cell cultures were trypsinized and counted. Cells were washed twice in Growth Medium supplemented with only 2% FBS by centrifugation, resuspension, and aspiration of the supernatant. Cells were seeded onto the coated wells 20,000 cells per well in approximately 0.5 milliliter Growth Medium supplemented with only 2% (v/v) FBS. Cells were then incubated for approximately 30 minutes to allow the cells to settle.
  • extracellular matrix material MATRIGEL, BD Discovery Labware, Bedford, Mass.
  • Endothelial cell cultures were then treated with either 10 nanomolar human bFGF (Peprotech, Rocky Hill, N.J.) or 10 nanomolar human VEGF (Peprotech, Rocky Hill, N.J.) to serve as positive controls for endothelial cell response.
  • Transwell inserts seeded with postpartum cells were added to appropriate wells with Growth Medium supplemented with only 2% FBS in the insert chamber. Cultures were incubated in 5% CO 2 at 37° C. for approximately 24 hours. The well plate was removed from the incubator, and images of the endothelial cell cultures were collected with an Olympus inverted microscope (Olympus, Melville, N.Y.).
  • HUVEC form structured cell networks.
  • HUVEC cells form limited cell networks in co-culture experiments with hMSC and with 10 nanomolar bFGF. HUVEC cells without any treatment showed very little or no network formation. These results suggest that the umbilicus-derived cells release angiogenic factors that stimulate the HUVEC. Similarly, HCAECs formed cell networks only in co-culture with umbilicus-derived cells.
  • Table 17-1 shows quantities of known angiogenic factors released by UDCs in Growth Medium at atmospheric oxygen conditions.
  • UDCs were seeded onto inserts as described above. The cells were cultured at 37° C. in atmospheric oxygen for 48 hours on the inserts and then switched to a 2% FBS media and returned at 37° C. for 24 hours. Medium was removed, immediately frozen, and stored at ⁇ 80° C., and analyzed by the SearchLight multiplex ELISA assay (Pierce Chemical Company, Rockford, Ill.). Results shown are the averages of duplicate measurements.
  • UDCs do not release detectable levels of platelet-derived growth factor-bb (PDGFbb), heparin-binding epidermal growth factor (HB-EGF), or vascular endothelial growth factor (VEGF).
  • PDGFbb platelet-derived growth factor-bb
  • HB-EGF heparin-binding epidermal growth factor
  • VEGF vascular endothelial growth factor
  • ANG2 angiopoietin 2
  • the umbilicus-derived cells released measurable quantities of tissue inhibitor of metallinoprotease-1 (TIMP-1), thrombopoietin (TPO), and hepatocyte growth factor (HGF).
  • TPO thrombopoietin
  • HGF hepatocyte growth factor
  • KGF keratinocyte growth factor
  • FGF fibroblast growth factor
  • Umbilicus-derived cells were cultured in 24 hours in media with 2% FBS in atmospheric oxygen. Medium was removed and assayed by the SearchLight multiplex ELISA assay (Pierce). Results are the means of a duplicate analysis. Values are concentrations in the media reported in picograms per milliliter of culture media.
  • TIMP1 ANG2 PDGFBB TPO KGF HGF FGF VEGF HBEGF (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) UDCs (P4) 81831.7 ⁇ 9.8 ⁇ 2.0 365.9 14.1 200.2 5.8 ⁇ 4.0 ⁇ 1.2 Media alone ⁇ 9.8 25.1 ⁇ 2.0 ⁇ 6.4 ⁇ 2.0 ⁇ 3.2 ⁇ 5.4 ⁇ 4.0 ⁇ 1.2
  • Table 17-2 shows levels of known angiogenic factors released by UDCs at 5% O 2 .
  • UDCs were seeded onto inserts as described above. The cells were cultured in Growth Medium at 5% oxygen for 48 hours on the inserts and then switched to a 2% FBS medium and returned to 5% O 2 incubation for 24 hours. Medium was removed, immediately frozen, and stored at ⁇ 80° C., and analyzed by the SearchLight multiplex ELISA assay (Pierce Chemical Company, Rockford, Ill.). Results shown are the averages of duplicate measurements. The results show for UDCs are comparable to those under atmospheric oxygen conditions.
  • TIMP1 ANG2 PDGFBB TPO KGF HGF FGF VEGF HBEGF (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) UDCs (P4) 50244.7 ⁇ 9.8 ⁇ 2.0 403.3 10.7 156.8 5.7 ⁇ 4.0 ⁇ 1.2 Media alone ⁇ 9.8 25.1 ⁇ 2.0 ⁇ 6.4 ⁇ 2.0 ⁇ 3.2 ⁇ 5.4 ⁇ 4.0 ⁇ 1.2
  • HNF-1alpha a hepatocyte-specific transcription factor
  • cytoplasmic intermediate filament proteins such as keratin 19 (K19), keratin 8 (K8), and cytokeratin 18 (CK18)
  • keratin 19 K19
  • K8 keratin 8
  • CK18 cytokeratin 18
  • albumin and cytochrome p450 2B6 were selected as markers for hepatocyte differentiation (Schwartz et al. (2002) J. Clin. Invest. 109(10):1291-1302; Okumoto et al. (2003) Biochem. Biophys. Res. Commun. 304(4):691-695; Chargracui et al. (2003) Blood 101(8): 2973-2982).
  • Basic Fibroblast Growth Factor (bFGF), Oncostatin M, Hepatocyte Growth Factor (HGF), Stem Cell Factor (SCF), and Fibroblast Growth Factor 4 (FGF 4) were from PeproTech Inc. (Rocky Hill, N.J.).
  • Platelet Derived Growth Factor BB (PDGFbb) was from R&D Systems (Minneapolis, Minn.).
  • Umbilicus-derived cells P5
  • NHDF Normal Human Dermal Fibroblasts
  • Cells were plated at 22.5 ⁇ 10 3 cells/cm 2 on 1% Matrigel (Becton-Dickinson and Co., Franklin Lakes, N.J.) in serum-free medium (60% (v/v) low glucose DMEM) (DMEM-LG; Gibco, Carlsbad, Calif.), 40% (v/v) MCDB-201 (Sigma, St.
  • Umbilicus-derived cells (P5), neonatal and adult NHDF. Cells were seeded at lower density (22,500 cells/cm 2 ) in 24-well plates coated with gelatin and grown as described above.
  • Umbilicus-derived cells P17
  • Umbilicus-derived cells P15
  • Umbilicus-derived cells P10
  • adult NHDF adult NHDF.
  • Cells were seeded at higher density (50,000 cells/cm 2 ) in 24-well TCP plates and grown in DMEM (Gibco), B27 Supplement (Gibco), penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 20 nanograms/milliliter HGF and/or 10 nanograms/milliliter FGF-4. Cells were grown in these conditions for 4 weeks.
  • Umbilicus-derived cells P4
  • Umbilicus-derived cells P9
  • neonatal and adult NHDF Cells were seeded at a density of 5,000 cells/cm 2 in T25 flasks in Chang C medium (Irvine Scientific, Santa Ana, Calif.) on either fibronectin (PeproTech, Rocky Hill, N.J.) or gelatin (Sigma) and grown for two passages until confluence. Cells were then seeded at 1,000 cells/cm 2 in 24-well TCP plates and grown as described above until they reached about 40-60% confluence.
  • Umbilicus-derived cells (P5) and adult NHDF. Cells were plated in 24-well plates on gelatin in Growth Medium supplemented with either 1 nanogram/milliliter or 10 nanograms/milliliter oncostatin M (Chargracui (2003) Blood 101(8): 2973-2982). Cells were grown in these conditions for 4 weeks.
  • Umbilicus-derived cells (P5) and adult NHDF. Cells were plated in 24-well plates on gelatin in Growth Medium supplemented with 10 nanograms/milliliter bFGF, 10 nanograms/milliliter HGF, 10 nanograms/milliliter SCF. Cells were grown in these conditions for 4 weeks (Okumoto et al. (2003) Biochem. Biophys. Res. Commun. 304(4):691-695.).
  • RNA isolation and quantitative RT-PCR RNA was extracted from umbilicus-derived cells and fibroblasts grown as described in each protocol. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit, Qiagen, Valencia, Calif.) and RNA extracted according to the manufacturer's instructions (RNeasy Mini Kit, Qiagen, Valencia, Calif.) with a 2.7 Units/sample DNase treatment (Sigma). RNA was eluted with 50 microliters DEPC-treated water and stored at ⁇ 80° C.
  • PCR was performed on cDNA samples using ASSAYS-ON-DEMAND gene expression products for albumin (Hs00609411), cytochrome p450 2B6 (Hs00167937), GAPDH (Applied Biosystems, Foster City, Calif.) and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minute. PCR data were analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).
  • Immunofluorescence Cell cultures were fixed with cold (4° C.) 4% (w/v) paraformaldehyde for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the following epitopes: keratin 9 (K19; 1:400; Chemicon, Temecula, Calif.), keratin 19 (K19; 1:400; Chemicon), cytokeratin 18 (CK18; 1:400; Sigma, St. Louis, Mo.), vimentin (1:500; Sigma), desmin (1:150; Sigma), albumin (1:200; Sigma), c-met (1:400; Santa Cruz Biotech, Santa Cruz, Calif.), and HNF-1alpha (1:400; Santa Cruz Biotech).
  • cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100, Sigma) for 30 minutes to access intracellular antigens.
  • PBS phosphate-buffered saline
  • Triton Triton X-100, Sigma
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • umbilicus-derived cells were cultured in Chang C medium.
  • Umbilicus-derived cells (P2) were grown in Chang C medium for 11 days.
  • Umbilicus-derived cells stained negative for cytokeratin 18 and keratin 8 by immunocytochemistry analysis. Samples grown in Growth Medium were negative for both markers.
  • Umbilicus-derived cells were treated as described in method 1 according to a protocol developed by Schwartz et al. (2002) J. Clin. Invest. 109(10):1291-1302.). Both albumin and cytochrome p450 2B6 were detected with ASSAYS-ON-DEMAND primers at levels lower than HepG2 positive control. No clear pattern emerged between conditions applied and gene expression levels, i.e., albumin and cytochrome p450 2B6 expression were each also detected in control samples. Some expression of albumin and cytochrome p450 2B6 was detected with ASSAYS-ON-DEMAND primers, however, the levels were significantly lower than those observed in HepG2 cells.
  • Oncostatin M at low concentration of 1 nanogram/milliliter increased expression levels of cytochrome p450 2B6 in umbilicus-derived cells grown in Growth Medium on gelatin-coated flasks (data not shown).
  • FGF-4 and HGF treatment had little effect and may have reduced the expression of albumin and cytochrome p450 2B6.
  • hepatocyte-specific markers such as albumin and cytochrome p450 2B6 was detected, thereby indicating that the cells underwent some differentiation into hepatocytes.
  • Umbilicus-derived cells were seeded at 200,000 cells per well on 6-well tissue culture-treated plates in Growth Medium.
  • DMEM-Hg Dulbecco's Minimal Essential Medium-high glucose
  • DMEM-Hg fetal bovine serum
  • Hyclone Logan, Utah
  • 0.02 milligrams per milliliter insulin Sigma, St.
  • Oil Red 0 Staining Cells were fixed with 10 percent (v/v) neutral buffered formalin (Richard-Allan Kalamazoo, Mich.). After fixation, the cells were washed in deionized water and incubated for two minutes in propylene glycol (absolute; Poly Scientific, Bay Shore, N.Y.). Propylene glycol was removed by aspiration, and samples were incubated in Oil Red O (Poly Scientific) for one hour. Staining solution was removed by aspiration and stained samples were then incubated in 85 percent (v/v) propylene glycol solution (Poly Scientific) for one minute. Stained samples were washed with two changes of de-ionized water. Stained samples were counter-stained with Mayer's Hematoxylin (Poly Scientific) and examined with light microscopy. Images were taken at magnification of 20 ⁇ .
  • Adipose-derived cells and umbilicus-derived cells were seeded at 200,000 cells/well in 6-well tissue culture-treated plates. Cells were initially seeded in Growth Medium, which was changed to an adipogenic differentiation medium (DMEM-Hg medium; Invitrogen, Carlsbad, Calif.) containing 1 micromolar dexamethasone (Sigma, St. Louis, Mo.), 0.2 millimolar indomethasone (Sigma), 0.01 milligrams per microliter insulin (Sigma), 0.5 millimolar isobutylmethylxanthine (Sigma), 10 percent (v/v) fetal bovine serum (Cat.
  • DMEM-Hg medium adipogenic differentiation medium
  • Adipose differentiation Morphologically MSCs and adipose-derived cells demonstrated lipid formation as early as 5 days in this assay. Large amounts of lipid droplet formation were observed in these cultures by 15 days of culture. Cultures of osteoblasts also deposited large amounts of lipid under these conditions after 10 days in culture and extensively at 15 days. Lipid droplet formation was observed in umbilicus-derived and omental cell cultures after 15 days of culture. Low level lipid droplet formation was observed in the fibroblast cultures after 20 days in adipogenic-inducing conditions.
  • Leptin was not detected by ELISA in umbilicus-derived cell conditioned medium.
  • pancreas contains endocrine cells, organized in islets of Langerhans, that produce insulin, glucagon, somatostatin, and pancreatic polypeptide (PP).
  • the ability of umbilicus-derived cells to differentiate towards cells with an insulin-producing phenotype was tested under eight different induction protocols.
  • Umbilicus-derived cells (various isolates—see below) as well as neonatal or adult Normal Human Dermal Fibroblasts (NHDF) grown in Growth Medium, in gelatin-coated T75 flasks, as well as in different beta-cell promoting differentiation conditions. Flasks were coated with 2% (w/v) gelatin solution (Sigma, St. Louis, Mo.) for 20 minutes at room temperature. Gelatin solution was aspirated off and flasks were washed with PBS. Basic Fibroblast Growth Factor (bFGF), Epidermal Growth Factor (EGF), Transforming Growth Factor alpha (TGFalpha) and Fibroblast Growth Factor 10 (FGF-10) were purchased from PeproTech Inc. (Rocky Hill, N.J.). GLP-1 was purchased from Sigma (St. Louis, Mo.)
  • Adipose-derived cells U.S. Pat. No. 6,555,374
  • omentum-derived cells umbilicus-derived cells, (P15), (P17), (P3), and adult Normal Human Dermal Fibroblasts (NHDF) (P10) were utilized. Cells were maintained under either normal or 5% O 2 conditions.
  • Cells were seeded at low density (5,000 cells/cm 2 ) in gelatin-coated T75 flasks on gelatin and grown in Ham's F12 medium (Clonetics, Santa Rosa, Calif.), 2% (v/v) FBS, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF until confluence.
  • Confluent cells were trypsinized and plated at 50,000 cells/cm 2 in 24-well Tissue Culture Polystyrene (TCPS; BD Biosciences, Bedford, Mass.) plates with or without gelatin or collagen coating.
  • Cells were grown in Ham's F12 medium, 2% FBS, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF and 15 nanomolarGLP-1 (7-37 isoform) for up to 3 weeks.
  • Cells Umbilicus-derived cells, isolate 2 (P17), isolate 1 (P15), isolate 4 (P10) and adult NHDF P10 were utilized. Cells were seeded at 50,000 cells/cm 2 in 24-well TCP plates and grown in DMEM:Ham's F12 (1:1) medium, B-27 supplement (Gibco, Carlsbad, Calif.), 50 units of penicillin per milliliter, 50 milligrams streptomycin per milliliter, 20 nanograms/milliliter EGF, 40 nanograms/milliliter bFGF spherical clusters were generated—usually 4-6 days.
  • the spherical clusters were collected, centrifuged, and replated onto laminin-coated, 24-well plates (BD Biosciences, Bedford, Mass.), and cultured up to 3 weeks in B-27-supplemented medium containing 10 nanomolarGLP-1 (7-37) but no other growth factors (i.e. no bFGF and no EGF).
  • Cells Umbilicus-derived cells, isolate 2 (P17) isolate 1 (P15), isolate 4 (P10), and adult NHDF (P10) were utilized. Cells were seeded at high density (50,000 cells/cm 2 ) in 24-well TCPS plates and grown in DMEM:Ham's F12 (1:1) medium, B-27 supplement, P/S, 20 nanograms/milliliter EGF, 40 nanograms/milliliter bFGF spherical clusters were generated—usually 4-6 days.
  • the spherical clusters were collected, centrifuged, and replated onto laminin-coated, 24-well plates and cultured up to 3 weeks in B-27-supplemented medium containing 10 nanomolarGLP-1 (1-37 isoform) but no other growth factors (i.e. no bFGF and no EGF).
  • Cells Adult NHDF (P15), umbilicus-derived, isolate 1 (P18), isolate 2 (P21), isolate 3 (P5), isolate 3 (P4), were isolated according to the method by Mitchell et al. (2). Cells were seeded at 50,000 cells/cm 2 in 24-well TCPS gelatin-coated plates and grown in DMEM:Ham's F12 (1:1) medium, B-27 supplement, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter FGF-10, and/or 40 nanograms/milliliter TGF alpha for more than two weeks.
  • DMEM Ham's F12 (1:1) medium, B-27 supplement, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter FGF-10, and/or 40 nanograms/milliliter TGF alpha for more than two weeks.
  • Cells Adult NHDF, umbilicus-derived, isolate 1 (P18), isolate 2 (P21) isolate 3 (P5), isolate 3 (P4), were isolated according to the method by Mitchell et al. (2). Cells were seeded at 50,000 cells/cm 2 in 24-well TCPS gelatin-coated plates and grown in EBM-2 medium, 10 nanograms/milliliter FGF-10, and/or 40 nanograms/milliliter TGF alpha for greater than two weeks.
  • Cells Umbilicus-derived, isolate 3 (P2) were utilized. Cells were seeded at 5,000 cells/cm 2 in T75 flasks on gelatin and grown either in Growth Medium or in Ham's F12 medium, 2% FBS, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF until confluence. Confluent cells were trypsinized and plated at 50,000 cells/cm 2 in 24-well TCPS plates, with or without gelatin coating. Three types of basic media were used for up to 3 weeks:
  • Beta I medium Ham's F12 medium, 2% FBS, 10 millimolar nicotinamide, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 25 millimolar glucose;
  • Beta II medium Equal parts of DMEM/Ham's F12 media, 2% FBS, 10 millimolar nicotinamide, 25 millimolar glucose; and
  • EBM Endothelial Cell Basal Medium
  • the following growth factors were added to each of the media: 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF, 10 nanomolar GLP-1 (7-37 isoform).
  • RNA isolation and quantitative RT-PCR RNA was extracted from umbilicus-derived cells and fibroblasts grown as described in each protocol. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini kit, Qiagen, Valencia, Calif.) and RNA extracted according to the manufacturer's instructions (RNeasy Mini kit, Qiagen, Valencia, Calif.) with a 2.7 U/sample DNase treatment (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at ⁇ 80° C.
  • PCR was performed on cDNA samples using ASSAYS-ON-DEMAND gene expression products.
  • PDX-1 Hs00426216
  • pro-insulin Hs00355773
  • Ngn-3 Hs00360700
  • Glut-2 Hs00165775
  • GAPDH Applied Biosystems, Foster City, Calif.
  • TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems, Foster City, Calif.).
  • Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minutes.
  • Table 20-1 contains primer sequences. PCR using these primers was performed as described above. Pancreas total RNA (Ambion, Austin, Tex.) was used as control. PCR data were analyzed according to the ⁇ CT method recommended by Applied Biosystems (1).
  • Immunofluorescence Adult human pancreatic tissue was harvested and immersion fixed in 4% (w/v) paraformaldehyde (Sigma, St. Louis, Mo.) overnight at 4° C. Immunohistochemistry was performed using antibodies directed against the following epitopes: insulin (insulin serum; 1:50; LINCO Research, St. Charles, Mo.), PDX-1 (1:50; Santa Cruz Biotech, Santa Cruz, Calif.), glucagon (1:100; Santa Cruz Biotech), somatostatin (1:100; DAKOCytomation, Carpinteria, Calif.), and cytokeratin 18 (CK18; 1:400; Sigma, St. Louis, Mo.).
  • Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100, Sigma) for 1 hour. Primary antibodies, diluted in blocking solution, were then applied to the samples for a period of 4 hours at room temperature.
  • PBS phosphate-buffered saline
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • pancreas-specific marker was not detected using real-time PCR and the ASSAYS-ON-DEMAND primers, with the exception that low levels of Ngn-3 were detected in cells from protocol 6.
  • the same primers produced positive results with cDNA derived from pancreatic tissue RNA.
  • PCR Real-time PCR for ngn-3 was performed on cDNA samples derived from human umbilical cord grown according to protocol 6. PCR was also performed using ASSAYS-ON-DEMAND Ngn-3 primers (Hs00360700). Human pancreas-derived cDNA was used as control. No other pancreas-specific markers (PDX-1, pro-insulin or Glut-2) were detected with the ASSAYS-ON-DEMAND primers.
  • pancreas-specific markers were detected in tissue derived from a human pancreas using immunofluorescence technique and an array of antibodies (see materials and methods).
  • the expression of pancreas-specific markers e.g. insulin, PDX-1, glucagon, somatostatin, and cytokeratin 18
  • pancreas-specific markers e.g. insulin, PDX-1, glucagon, somatostatin, and cytokeratin 18
  • Cartilage damage and defects lead to approximately 600,000 surgical procedures each year in the United States alone (1).
  • a number of strategies have been developed to treat these conditions but these have had limited success.
  • Cartecel uses autologous chondrocytes that are collected from a patient, expanded in vitro and then implanted into the patient (1). This approach has the disadvantage of collecting healthy cartilage and requiring a second procedure to implant the cultured cells.
  • An alternative possibility is a stem cell-based therapy in which cells are placed at or near the defect site to directly replace the damaged tissue. Cells may be differentiated into chondrocytes prior to the application or progenitor cells that can differentiate in situ may be used. Such transplanted cells would replace the chondrocytes lost in the defect.
  • Candidate cells for this indication should be evaluated for their ability to differentiate into chondrocytes in vitro.
  • a number of protocols have been developed for testing the ability of cells to differentiate and express chondrocyte marker genes.
  • Umbilicus-derived cells were tested for their ability to differentiate into chondrocytes in vitro in two different assay systems: the pellet assay culture system and collagen gel cultures.
  • the pellet culture system has been used successfully with selected lots of human mesenchymal stem cells (MSC). MSC grown in this assay and treated with transforming growth factor-beta3 have been shown to differentiate into chondrocytes (2).
  • the collagen gel system has been used to culture chondrocytes in vitro (3). Chondrocytes grown under these conditions form a cartilage-like structure.
  • Human articular chondrocytes were purchased from Cambrex (Walkersville, Md.) and cultured in the same medium as the postpartum cells. Twenty-four hours before the experiment, the culture medium was changed to a media containing 1% FBS.
  • hMSCs Human mesenchymal stem cells
  • Collagen gel assays Cultured cells were trypsinized to remove from culture plate. Cells were washed with centrifugation twice at 300 ⁇ g for 5 minutes in DMEM without serum and counted. Cells were mixed with the following components at the final concentrations listed.
  • Rat tail collagen (1 milligram/milliliter, BD DiscoveryLabware, Bedford, Mass.), 0.01 Normal NaOH and Chondrocyte cell media DMEM, penicillin (100 Units/milliliter), streptomycin (100 micrograms/milliliter), 2 millimolar L-Glutamine, 1 millimolar Sodium Pyruvate, 0.35 millimolar L-Proline, 100 nanomolar dexamethasone, 0.17 millimolar L-Ascorbic Acid, 1% (v/v) ITS (insulin, transferring, selenium) (All components from Sigma Chemical Company).
  • the cells were gently mixed with the medium the samples were aliquoted into individual wells of a 24 well ultra-low cluster plate (Corning, Corning, N.Y.) at a concentration of either 2 ⁇ 10 5 per well or 5 ⁇ 10 5 per well. Cultures were placed in an incubator and left undisturbed for 24 to 48 hours. Medium was replaced with fresh chondrocyte media supplemented with appropriate growth factor every 24 to 48 hours. Samples were allowed to culture for up to 28 days at which time they were removed and fixed in 10% (v/v) formalin (VWR Scientific, West Chester, Pa.) and processed for histological examination. Samples were stained with Safranin O or hematoxylin/eosin for evaluation.
  • Pellet culture assays Cultured cells were trypsinized to remove from culture plate. Cells were washed with centrifugation twice at 300 ⁇ g for 5 minutes in DMEM without serum and counted. Cells were resuspended in fresh chondrocyte medium (described above) at a concentration of 5 ⁇ 10 5 cells per milliliter. Cells were aliquoted into new polypropylene tubes at 2.5 ⁇ 10 5 cells per tube. The appropriate samples were then treated with TGF-beta3 (10 nanograms/milliliter, Sigma) or GDF-5 (100 nanograms/milliliter; R&D Systems, Minneapolis, Minn.). Cells were then centrifuged at 150 ⁇ g for 3 minutes.
  • Tubes were then transferred to the incubator at and left undisturbed for 24-48 hours in 5% CO 2 at 37° C. Medium was replaced with fresh chondrocyte cell media and growth factor, where appropriate, every 2-3 days. Samples were allowed to culture for up to 28 days at which time they were removed and fixed and stained as described above.
  • Pellets were prepared and cultured and described in Methods. Pellets were grown in medium (Control), or medium supplemented with TGF beta3 (10 nanograms/milliliter) or GDF-5 (100 nanograms/milliliter), that was replaced every 2 to 3 days. Pellets were collected after 21 days of culture and stained by Safranin O to test for the presence of glycosoaminoglycans. The pellets treated with TGFbeta3 and GDF-5 showed some positive Safranin O staining as compared to control cells. The morphology of the umbilical cord cells showed some limited chondrocyte-like morphology.
  • Umbilicus-derived cells partially differentiated into chondrocytes in vitro in the pellet culture and the collagen gel assay systems.
  • the umbilicus-derived cells did show some indications of glycosaminoglycan expression by the cells. Morphology showed limited similarity to cartilage tissue.
  • chondrogenic potential of cells derived from umbilical tissue was performed using in vitro pellet culture based assays. Cells from umbilical cord at early passage (P3) and late passage (P12) were used. The chondrogenic potential of the cells was assessed in pellet culture assays, under chondrogenic induction conditions in medium supplemented with transforming growth factor beta-3 (TGFbeta-3), recombinant human growth and differentiation factor 5 (rhGDF-5), or a combination of both.
  • TGFbeta-3 transforming growth factor beta-3
  • rhGDF-5 recombinant human growth and differentiation factor 5
  • Dulbecco's Modified Essential Media DMEM
  • Penicillin and Streptomycin were obtained from Invitrogen, Carlsbad, Calif.
  • Fetal calf serum FCS
  • FCS Fetal calf serum
  • MSCGM Mesenchymal stem cell growth medium
  • hMSC chondrogenic differentiation bullet kit were obtained from Biowhittaker, Walkersville, Md.
  • TGFbeta-3 was obtained from Oncogene research products, San Diego, Calif.
  • rhGDF-5 was obtained from Biopharm, Heidelberg, Germany (WO9601316 A1, U.S. Pat. No. 5,994,094 A).
  • Human mesenchymal stem cells (Lot# 2F1656) were obtained from Biowhittaker, Walkersville, Md. and were cultured in MSCGM according to manufacturers instructions. This lot has been tested previously, and was shown to be positive in the chondrogenesis assays.
  • Human adult and neonatal fibroblasts were obtained from American Type Culture Collection (ATCC), Manassas, Va. and cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. Postpartum tissue-derived cells, isolated from human umbilical cords as described in previous examples, were utilized. Cells were cultured in Growth Medium in a manner similar to the culture of the fibroblasts. The cell cultures were incubated at 37° C. with 5% CO 2 . Cells used for experiments were at passages 3 and 12.
  • Pellet culture assay For pellet cultures, 0.25 ⁇ 10 6 cells were placed in a 15 milliliter conical tube and centrifuged at 150 ⁇ g for 5 minutes at room temperature to form a spherical pellet according to protocol for chondrogenic assay from Biowhittaker. Pellets were cultured in chondrogenic induction medium containing TGFbeta-3 (10 nanograms/milliliter), rhGDF-5 (500 nanograms/milliliter), or a combination of TGFbeta-3 (10 nanograms/milliliter), and rhGDF-5 (500 nanograms/milliliter) for three weeks. Untreated controls were cultured in growth medium. During culture, pellets were re-fed with fresh medium every other day. Treatment groups included the following:
  • HMSC Human Mesenchymal Stem cells
  • rhGDF-5 Human Mesenchymal Stem cells
  • HMSC Human Mesenchymal Stem cells
  • H Human adult fibroblast cells (HAF)+TGFbeta-3
  • U EP Umbilicus-derived cells early passage
  • U LP Umbilicus-derived cells late passage
  • HMSC Human Mesenchymal Stem cells
  • Umbilicus-derived cells, MSCs and fibroblasts formed cell pellets in chondrogenic induction medium with the different growth factors. The size of the pellets at the end of culture period varied among the different cell types. Pellets formed with the umbilical cells tended to be larger and looser than those formed by MSCs and fibroblasts. Pellets formed with all cell types and cultured in control medium were smaller than pellets cultured in chondrogenic induction medium.
  • Umbilicus-derived cells at late passage did not demonstrate as distinct a chondrogenic potential as did early passage umbilical derived cells. However, this may be due to the fact that chondrogenic induction conditions were optimized for MSCs, not for postpartum derived cells. Some cell condensation was observed with fibroblast, but it was not associated with Safranin O staining.
  • umbilicus-derived cells were treated with 5-azacytidine either alone or in combination with DMSO or chelerythrine chloride and markers of cardiomyocytes measured by real-time PCR.
  • Cryopreserved umbilicus-derived cells were grown in Growth Medium in gelatin-coated flasks. Cells were seeded at 5 ⁇ 10 4 cells/well in 96-well plates in Growth Medium for 24 hours. The medium was changed to 0, 3, 10 and 30 micromolar 5-azacytidine (Sigma, St. Louis, Mo.) alone or with 5 microM chelerythrine chloride (Sigma), 1% (v/v) dimethylsulfoxide (DMSO) (Sigma), or 1 micromolar retinoic acid (Sigma) in MEM-alpha (Sigma), insulin, transferrin, and selenium (ITS; Sigma), 10% (v/v) fetal bovine serum, penicillin and streptomycin.
  • 5 microM chelerythrine chloride Sigma
  • DMSO dimethylsulfoxide
  • ITS selenium
  • 10% (v/v) fetal bovine serum penicillin and streptomycin.
  • the cells were incubated at 37° C., 5% (v/v) O 2 for 48-72 hours.
  • the medium was changed to MEM-alpha, insulin, transferrin, and selenium, 10% (v/v) fetal bovine serum, penicillin (50 Units/milliliter) and streptomycin (50 micrograms/milliliter), and cells incubated at 37° C., 5% (v/v) O 2 for 14 days.
  • RNA extraction and Reverse Transcription were lysed with 150 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, Mo.) according to the manufacturer's instructions (RNeasy 96 kit, Qiagen, Valencia, Calif.) and stored at ⁇ 80° C. Cell lysates were thawed and RNA extracted according to the manufacturer's instructions (RNeasy 96 kit, Qiagen, Valencia, Calif.) with a 2.7 Units/sample DNase treatment (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at ⁇ 80° C.
  • PCR was performed on cDNA samples using ASSAYS-ON-DEMAND gene expression products cardiac myosin (Hs00165276 ml), skeletal myosin (Hs00428600), GATA 4 (Hs00171403 ml), GAPDH (Applied Biosystems, Foster City, Calif.), and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minute. cDNA from heart and skeletal muscle (Ambion, Austin Tex.) were used as controls.
  • Control RNA from cardiac muscle showed expression of cardiac myosin and GATA 4, skeletal muscle RNA showed skeletal myosin and cardiac myosin, but no GATA 4 expression.
  • Umbilicus-derived cells (P12) treated for 48 h with factors and cultured for a further 14 days expressed low levels of GATA 4, but no skeletal myosin or cardiac myosin. Additional samples from umbilicus-derived cells also showed expression of GATA 4.
  • Untreated umbilicus-derived cells constitutively express GATA 4, a nuclear transcription factor in cardiomyocytes, sertoli cells, and hepatocytes.
  • the Charles River Worcester, Mass. test facility is accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care, International (AAALAC) and registered with the United States Department of Agriculture to conduct research in laboratory animals. All the conditions of testing conformed to the Animal Welfare Act (9 CFR) and its amendments. The protocol was reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) at the Test Facility for compliance with regulations prior to study initiation.
  • AALAC Laboratory Animal Care, International
  • IACUC Institutional Animal Care and Use Committee
  • Environmental controls were set to maintain temperatures of 18 to 26° C. (64 to 79° F.) with relative humidity at 30% to 70%. A 12:12 hour light:dark cycle was maintained. Ten or greater air changes per hour were maintained in the animal rooms. Upon receipt and prior to use on the study, the animals were held for a minimum of four days for conditioning according to the Test Facility Vendor Management Program as described in the Test Facility Standard Operating Procedure, Receipt, Conditioning, and Quarantine of Laboratory Animals.
  • Each animal was identified by a unique number indicated by an ear punch. Animals were randomly assigned to groups by a weight-ordered distribution such that individual body weights did not exceed ⁇ 20% of mean weight.
  • mice were anesthetized with sodium pentobarbital (40 milligrams/kilogram) and buprenorphine (0.05 milligrams/kilogram) as a single cocktail given intramuscularly (IM). Following the establishment of anesthesia, animals were intubated using 18 to 16 gauge, 2-inch length angiocaths, or appropriate sized angiocath, and maintained on room air respiration (supplemented with oxygen) and a positive pressure ventilator throughout the surgical procedure. Additional anesthesia was given incrementally as needed. Preoperative antibiotic therapy was also administered, Benzathine/Procaine penicillin G, 40,000 Units/kilogram, IM. Additional antibiotic therapy was administered every 48 hours.
  • Electrode pads were placed around the appropriate paws of the animals to receive a useable ECG signal. Animals were positioned on a heating pad to help maintain body temperature throughout the procedure. A rectal temperature probe was inserted into the animal to monitor body temperature. Ophthalmic ointment was administered to each eye. The surgical sites (thoracic area) were prepared for aseptic surgery by removing any excess fur, and gently wiping the area with sponges soaked in 70% isopropyl alcohol, which was allowed to dry. Iodone (MEDISEPPS, or similar solution) was then applied to the area and allowed to dry. The area was appropriately draped for strict aseptic surgery.
  • Iodone MEDISEPPS, or similar solution
  • the suture placed in the apex was used to ease the heart out of the chest cavity and to place sufficient tension on the heart to allow access to the upper heart and the left anterior descending coronary artery (LAD).
  • LAD left anterior descending coronary artery
  • Another length of 6-0 silk suture was placed into the myocardium so as to surround the LAD. The pressure on the apical suture was released and the heart allowed to return to the interior of the chest cavity.
  • the ligatures around the LAD were tied off to occlude the LAD. This was a permanent occlusion with the suture tied off and the ends trimmed.
  • the surgeon looked for the following indications of successful occlusion: change in color of the area of the heart directly below the ligature to a white/grayish white as a result of the termination of blood flow to the area and a significant change in the ECG corresponding to occlusion of the LAD.
  • Arrhythmias may have developed within the first 10 minutes of the occlusion. The rat was monitored closely during this time period in the event that resuscitation was necessary.
  • Human cells were grown in Growth Medium in gelatin-coated T300 flasks. Cells were washed with phosphate buffered saline (PBS, Gibco, Carlsbad Calif.) and trypsinized using Trypsin/EDTA (Gibco, Carlsbad Calif.). The trypsinization was stopped by adding Growth Medium. The cells were centrifuged at 150 ⁇ g, supernatant removed, and the cell pellet was resuspended in approximately 1 milliliter Growth Medium per million cells. An aliquot of cells was removed and added to Trypan blue (Sigma, St. Louis, Mo.). The viable cell number was estimated using a hemocytometer.
  • PBS phosphate buffered saline
  • Trypsin/EDTA Gibco, Carlsbad Calif.
  • the trypsinization was stopped by adding Growth Medium. The cells were centrifuged at 150 ⁇ g, supernatant removed, and the cell pellet was resuspended in approximately 1 milliliter Growth Medium per million cells
  • the cell suspension was centrifuged and resuspended in 1 milliliter Growth Medium containing 10% (v/v) DMSO (Hybrimax, Sigma, St. Louis, Mo.) per 5 million cells and transferred into Cryovials (Nalgene).
  • the cells were cooled at approximately 1° C./minute overnight in a ⁇ 80° C. freezer using a “Mr. Frosty” freezing container (Nalgene, Rochester, N.Y.). Vials of cells were transferred into liquid nitrogen. Vials were shipped from CBAT, Somerville, N.J. to Charles River, Worcester, Mass. on dry ice and stored at ⁇ 80° C.
  • the upper layers of muscle was sutured using a simple continuous pattern.
  • the skin was closed with 4-0 silk using a horizontal mattress pattern.
  • a 10 milliliter syringe was attached to the intravenous catheter that had been previously placed in the thoracic cavity and the plunger slowly pulled back to withdraw fluids and air from the cavity. At the same time, the catheter was slowly withdrawn from the entry site, thereby allowing the surrounding muscle mass and skin to seal the puncture.
  • the surgical drape was removed and fluids (i.e., lactated Ringers solution, 25 milliliters/kilogram subcutaneously [SC] or intraperitoneally [IP]) were given.
  • fluids i.e., lactated Ringers solution, 25 milliliters/kilogram subcutaneously [SC] or intraperitoneally [IP]
  • ECG echocardiography
  • the left thoracic area was shaved, and warmed, ultrasonic gel was applied to the skin to enhance contact with the transducer. Electrode pads were placed around the appropriate extremities to receive an ECG signal. Echocardiographic images included short axis and long axis views to allow for the determination of ventricular cavity dimensions, contractility, blood flow through vasculature, and wall thickness. These images were saved on optical disk for further analysis.
  • the gel medium was removed from the skin with gauze or paper towel. The rat was removed from the ventilator and placed in a warmed recovery cage until mobile.
  • Analgesics i.e., Buprenorphine, 0.05 milligram/kilogram SC.
  • Analgesics were given twice daily for 4 days post-operatively and thereafter as needed.
  • Visual indications of post-operative pain include lack of normal body postures and movement (e.g., animal remains in hunched position), antipathy, lack of eating/drinking, lack of grooming, and the like.
  • Body weight was recorded for each animal prior to initial treatment, weekly thereafter, and on the day of necropsy. Animals found dead were weighed and necropsied.
  • each rat was anesthetized as was done for surgery.
  • the jugular vein was cannulated.
  • the heart was arrested in diastole with KCl infused via the jugular cannula.
  • the heart was then removed from the thoracic cavity.
  • a limited necropsy was then performed on the heart after which the heart was placed in 10% neutral buffered formalin. The remainder of each carcass was then discarded with no further evaluation.
  • Fractional shortening values were calculated as described by Sahn et al. (1978) Circulation 58:1072-1083.
  • the fractional shortening of the vehicle-treated animals had a significant decrease from 47.7% ⁇ 8.3% at Day 0 to 23.5% ⁇ 30.2% at Day 28 (p ⁇ 0.05).
  • the animals that were treated with umbilicus-derived cells showed small, non-significant differences between the fractional shortening between Day 0 and 28. There were no significant differences between the fractional shortening between the treatment groups at Day 0.
  • the results of the present study suggest that the umbilicus-derived cells have some benefit in reducing the damage of a surgically induced myocardial infarction in rats.
  • the vehicle-treated animals showed a significant reduction in cardiac function from day 0 to day 28, as measured by fractional shortening, while the umbilicus-derived cell-treated animals showed minimal change over the 28-day study.
  • the fibroblast-treated animals showed minimal change but only two animals survived the study.
  • Evaluation of infarct size suggested that there is some modest, but not statistically significant, reduction in the infarct size in the postpartum-derived cell-treated animals as compared to the vehicle controls at Day 28. Taken together, these data support efficacy of the umbilicus-derived cells in reducing damage from a myocardial infarction.
  • RP retinitis pigmentosa
  • the retinal photoreceptors and adjacent retinal pigment epithelium form a functional unit.
  • the Royal College of Surgeons (RCS) rat presents with a tyrosine receptor kinase (Mertk) defect affecting outer segment phagocytosis, leading to photoreceptor cell death (1).
  • Transplantation of retinal pigment epithelial (RPE) cells into the subretinal space of RCS rats was found to limit the progress of photoreceptor loss and preserve visual function (2).
  • RPE retinal pigment epithelial
  • umbilicus-derived cells can be used to promote photoreceptor rescue in an RCS model.
  • dystrophic RCS rats were anesthetized with xylazine-ketamine (1 milligram/kilogram i.p. of the following mixture: 2.5 milliliters xylazine at 20 milligrams/milliliter, 5 milliliters ketamine at 100 milligrams/milliliter, and 0.5 milliliter distilled water) and their heads secured by a nose bar.
  • xylazine-ketamine 1 milligram/kilogram i.p. of the following mixture: 2.5 milliliters xylazine at 20 milligrams/milliliter, 5 milliliters ketamine at 100 milligrams/milliliter, and 0.5 milliliter distilled water
  • Cells devoid of serum were resuspended (2 ⁇ 10 5 cells per injection) in 2 microliters of Leibovitz, L-15 medium (Invitrogen, Carlsbad, Calif.) and transplanted using a fine glass pipette (internal diameter 75-150 micro
  • mice were maintained on oral cyclosporine A (210 milligrams/liter of drinking water; resulting blood concentration: 250-300 micrograms/liter) (Bedford Labs, Bedford, Ohio) from 2 days pre-transplantation until end of the study. Food and water were available ad libitum. Animals were sacrificed at 60 or 90 days postoperatively, with some animals being euthanitized at earlier timepoints for histological assessment of short-term changes associated with cell transplantation.
  • oral cyclosporine A 210 milligrams/liter of drinking water; resulting blood concentration: 250-300 micrograms/liter
  • Food and water were available ad libitum. Animals were sacrificed at 60 or 90 days postoperatively, with some animals being euthanitized at earlier timepoints for histological assessment of short-term changes associated with cell transplantation.
  • ERG recordings Following overnight dark adaptation, animals were prepared for ERG recording under dim red light, as previously described (3). In brief, under anesthesia (with a mixture of 150 milligram/kilogram i.p ketamine, and 10 milligram/kilogram i.p. xylazine) the animal's head was secured with a stereotaxic head holder and the body temperature monitored through a rectal thermometer and maintained at 38° C. using a homeothermic blanket. Pupils were dilated using equal parts of topical 2.5% phenylephrine and 1% tropicamide.
  • Topical anesthesia with 0.75% bupivacaine was used to prevent any corneal reflexes and a drop of 0.9% saline was frequently applied on the cornea to prevent its dehydration and allow electrical contact with the recording electrode (gold wire loop).
  • Amplification (at 1-1,000 Hz bandpass, without notch filtering), stimulus presentation, and data acquisition were provided by the UTAS-3000 system from LKC Technologies (Gaithersburg, Md.). ERGs were recorded at 60 and 90 days of age in the umbilical cell groups and at 60 days only in the fibroblast groups.
  • V max from fitting the data with a Naka-Rushton curve was not used because ERG responses were often erratic at higher luminance levels in dystrophic animals and showed tendencies for depressed responses around 0.4 and 1.4 log candila/m 2 .
  • criterion amplitudes were used: 20 microVolts for a- and b-waves, and 10 microVolts for STR-like responses. The amplitude of the b-wave was measured from the a-wave negative peak up to the b-wave positive apex, and not up to the peak of oscillations, which can exceed the b-wave apex (4).
  • the double flash protocol was used to determine the isolation of rod and cone responses (5).
  • a probe flash was presented 1 second after a conditioning flash, using a specific feature of the UTAS-3000 system (LKC Technologies) with calibrated ganzfeld; assuring complete recharge of the stimulator under the conditions used.
  • the role of the conditioning flash in the procedure was to transiently saturate rods so that they were rendered unresponsive to the probe flash.
  • Response to the probe flash was taken as reflecting cone-driven activity.
  • a rod-driven b-wave was obtained by subtracting the cone-driven response from the mixed response (obtained following presentation of a probe flash alone, i.e. not preceded by any conditioning flash).
  • Physiological retinal sensitivity testing was performed to demonstrate retinal response to dim light. Animals were anesthetized with a recovery dose of urethane at 1.25 grams/kilogram i.p. Physiological assessment in the animals was tested post graft in animals at 90 days by recording multiunit extracellular activity in the superior colliculus to illumination of respective visual receptive fields (6). This procedure was repeated for 20 independent points (spaced 200 millimeters apart, with each step corresponding to approximately 10-150 displacements in the visual field), covering the visual field.
  • Visual thresholds were measured as the increase in intensity over background and maintained at 0.02 candila/m 2 (luminescence unit) [at least 2.6 logarithm units below rod saturation (7)], required for activating units in the superficial 200 microns of the superior colliculus with a spot of light 3° in diameter. Response parameters were compared between transplanted and sham control eyes that received vehicle alone.
  • TAAB embedding resin TAAB Laboratories, Aldemarston, UK. Semi-thin sections were stained with 1% Toluidine Blue in 1% borate buffer and the ultra thin sections were contrasted with uranyl acetate and lead citrate.
  • Umbilicus-derived cell-transplanted animals demonstrated good improvement in all outcome measures tested at 60 days (Table 25-1), a-wave (27 ⁇ 11) versus sham controls (0), mixed b-wave (117 ⁇ 67) versus sham controls (18 ⁇ 13), cone-b-wave (55 ⁇ 25) versus sham controls (28 ⁇ 11), and in rod contribution (49 ⁇ 16%) versus sham controls (6 ⁇ 7%).
  • Efficacy of transplants in preventing visual loss was monitored by assessment of electrophysiological responsiveness in two animals.
  • the threshold sensitivity response to light was used to define the area of visual field rescue in sham-injected control eyes versus eyes transplanted with umbilicus-derived cells.
  • visual thresholds never exceeded 0.5 log candila/m 2 above background.
  • the thresholds are usually in the magnitude of 4 log candila/m 2 units (8).
  • the thresholds were in the order of 2.9-4.9 log candila/m 2 units with an average threshold of 4.0 log candila/m 2 units, in some instances no recording could be attained.
  • the sham-injected rats showed some highly localized functional rescue in the temporal retina.
  • the human umbilicus-derived cell transplanted rats exhibited substantially greater levels of visual preservation with thresholds ranging from 0.8 to 2.1 log candila/m 2 units, with an average threshold of 1.3 log candila/m 2 units.
  • ERG does not provide direct information as to image forming capability.
  • the measurement of collicular threshold sensitivity used in this study provides an indication of relative preservation of visual fields. The importance of this measure is based on a correlation between the amounts of functional rescue and anatomical preservation and that the data collected compares with visual field perimetry testing in humans (9). The transplantation has demonstrated a retardation of the disease process in the test animals.
  • the results presented herein demonstrate clear evidence of functional efficacy of grafting human umbilicus-derived cells into the subretinal space, and that preservation of photoreceptors occurs in the general region in which the grafted cells are located.
  • the chondrogenic potential of cells derived from umbilicus or placenta tissue was evaluated following seeding on bioresorbable growth factor-loaded scaffolds and implantation into SCID mice.
  • Dulbecco's Modified Essential Media DMEM
  • Penicillin and Streptomycin were obtained from Invitrogen, Carlsbad, Calif.
  • Fetal calf serum FCS
  • FCS Fetal calf serum
  • MSCGM Mesenchymal stem cell growth medium
  • TGFbeta-3 was obtained from Oncogene research products, San Diego, Calif.
  • rhGDF-5 was obtained from Biopharm, Heidelberg, Germany (International PCT Publication No. WO96/01316 A1, U.S. Pat. No. 5,994,094A).
  • Chondrocyte growth medium comprised DMEM-High glucose supplemented with 10% fetal calf serum (FCS), 10 millimolar HEPES, 0.1 millimolar nonessential amino acids, 20 micrograms/milliliter L-proline, 50 micrograms/milliliter ascorbic acid, 100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, and 0.25 micrograms/milliliter amphotericin B.
  • Bovine fibrinogen was obtained from Calbiochem.
  • Human mesenchymal stem cells (hMSC, Lot# 2F1656) were obtained from Biowhittaker, Walkersville, Md. and were cultured in MSCGM according to the manufacturer's instructions. This lot was tested in the laboratory previously in in vitro experiments and was shown to be positive in the chondrogenesis assays.
  • Human adult fibroblasts were obtained from American Type Culture Collection (ATCC, Manassas, Va.) and cultured in Growth Medium on gelatin-coated tissue culture plastic flasks.
  • Postpartum-derived cells isolated from human umbilical cords (Lot# 022703Umb) and placenta (Lot# 071003Plac) were prepared as previously described (Example 1). Cells were cultured in Growth Media on gelatin-coated tissue culture plastic flasks. The cell cultures were incubated at 37° C. with 5% CO 2 . Cells used for experiments were at passages 5 (“Low passage”) and 14 (“High passage”).
  • Foams composed of 35/65 Poly(epsilon-caprolactone)(PCL)/Poly(glycolic acid) (PGA) (35/65 PCL/PGA) copolymer, reinforced with Polydioxanone (PDS) mesh (PGA/PCL foam-PDS mesh) were formed by the process of lyophilization, as described in U.S. Pat. No. 6,355,699. The foams were 4 cm ⁇ 5 cm, and 1 mm thick. Foams were sterilized by treatment with ethylene oxide (ETO).
  • ETO ethylene oxide
  • Bovine Cartilage Explants Cartilage explants 5 millimeters in diameter were made from cartilage obtained from young bovine shoulder. Punches (3 millimeters) were excised from the center of the explant and replaced with cells seeded on 3.5 millimeters resorbable scaffold. Scaffolds with cells were retained within the explants using fibrin glue (60 microliters of bovine fibrinogen, 3 milligrams/milliliter). Samples were maintained in chondrocyte growth medium overnight, rinsed in Phosphate Buffered Saline the following day, and implanted into SCID mice.
  • fibrin glue 60 microliters of bovine fibrinogen, 3 milligrams/milliliter
  • SCID mice ( Mus musculus )/Fox Chase SCID/Male), 5 weeks of age, were obtained from Harlan Sprague Dawley, Inc. (Indianapolis, Ind.) and Charles River Laboratories (Portage, Mich.). Animals used in the study were selected without any apparent systematic bias. A tag was placed on each individual animal cage listing the accession number, implantation technique, animal number, species/strain, surgery date, in vivo period, and date of euthanasia. The animals were identified by sequential numbers marked on the ear with an indelible ink marker.
  • mice were tested. Two scaffolds were implanted subcutaneously in each mouse as described below; 42 mice for subcutaneous implantation; 28 treatments with n-value of 3 per treatment.
  • the study corresponds to IACUC Approval Number: Skillman IACUC 01-037. The study lasted six weeks.
  • mice All handling of the SCID mice occurred under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of KETASET® (ketamine hydrochloride [60 milligram/kilogram]), ROMPUN® (xylazine [10 milligram/kilogram]), and saline.
  • KETASET® ketamine hydrochloride [60 milligram/kilogram]
  • ROMPUN® xylazine [10 milligram/kilogram]
  • saline ketamine hydrochloride [60 milligram/kilogram]
  • mice were euthanized by CO 2 inhalation at their designated intervals. Gross observations of the implanted sites were recorded. Samples of the subcutaneous implantation sites with their overlying skin were excised and fixed in 10% buffered formalin. Each implant was bisected into halves, and one half was sent to MPI Research (Mattawan, Mich.) for paraffin embedding, sectioning, and staining with Hematoxylin & Eosin (H&E) and Safranin O (SO).
  • H&E Hematoxylin & Eosin
  • SO Safranin O
  • New cartilage and bone formation was observed in the majority of the samples including growth factor-loaded, cell-seeded scaffolds, cell-seeded control scaffolds, and scaffolds loaded with growth factor alone. The extent of new cartilage and bone formation varied within the treatment and control groups.
  • hMSC-loaded scaffolds also showed new cartilage and bone formation.
  • the extent of new cartilage and bone formation was similar for all the hMSC treatment groups.
  • Human adult fibroblast seeded scaffolds also demonstrated new cartilage and bone formation. Results were similar to those obtained with placenta-derived cells and hMSCs
  • New cartilage formation was observed adjacent to the cartilage explant rings as well as within the scaffolds.
  • New cartilage formation within the scaffolds adjacent to the cartilage rings could be a result of chondrocyte migration.
  • Cartilage formation seen as islands within the scaffolds may be a result of either migration of chondrocytes within the scaffolds, differentiation of seeded cells or differentiation of endogenous mouse progenitor cells. This observation stems from the fact that in control growth factor-loaded scaffolds with no seeded cells, islands of chondrogenic differentiation were observed.
  • New bone formation was observed within the scaffolds independently and also associated with chondrocytes. Bone formation may have arisen from osteoblast differentiation as well as endochondral ossification.
  • results showed that new cartilage and bone formation were observed in growth factor and control scaffolds seeded with placenta- and umbilicus-derived cells.
  • Results with placenta-derived cells were similar to that seen with human mesenchymal stem cells, while the extent of new cartilage like tissue formation was slightly less pronounced in umbilicus-derived cells.
  • Growth factor-loaded scaffolds implanted without cells also demonstrated new cartilage and bone formation. These data indicate that new cartilage formation within the scaffolds may arise from chondrocytes that migrated from the bovine explants, from chondrogenic differentiation of endogenous progenitor cells, and from chondrogenic differentiation of seeded cells.
  • placenta- and umbilicus-derived cells undergo chondrogenic and osteogenic differentiation. These results also suggest that placenta- and umbilicus-derived cells may promote migration of chondrocytes from the cartilage explant into the scaffolds. Abundant new blood vessels were also observed in the scaffolds especially associated with new bone formation.
  • FBS fetal bovine serum
  • Advanced-DMEM (Invitrogen) has been used as a basal medium to optimize serum-free growth of postpartum-derived cells. While serum-free, Advanced-DMEM is not devoid of all animal proteins. Animal protein components contained within Advanced DMEM, such as albumin and insulin, may be replaced with recombinantly-produced alternatives prior to use for making commercialized therapeutic cells or biologics. Postpartum-derived cells (PPDCs) grown from isolation in Advanced-DMEM have been characterized and shown to have reduced expression of PDGFr-alpha and HLA-ABC, relative to that seen in serum-containing media.
  • PPDCs Postpartum-derived cells
  • Cell-based therapies have been or are being developed to treat patients with diseases as broad-ranging as myocardial infarction, stroke, ocular diseases like retinitis pigmentosa and macular degeneration, and diabetes. Bankable quantities of cells are required thus stimulating efforts to rapidly and efficiently isolate and expand such cells.
  • PPDCs were Isolated as in the previous examples.
  • Postpartum-derived cells (umbilicus 022803 (P12)) previously grown in Growth Media were plated at 5,000 cells per cm-squared and weaned of serum over the course of one week by reducing the serum content from 15% to 2%. Cells were then transferred to Growth Medium without serum prior to passage and plating for the short-term MTS assay.
  • a MTS assay was used to colorimetrically quantitate DNA content, an indirect measure of cell number and thus cell proliferation.
  • Umbilicus-derived cells (022803 (P11)) were plated at a density of 1,000 cells/well (96-well plate format; equivalent of 5,000 cells/cm-squared, in 100 microliters of medium) in quadruplicate for each condition tested. Both basal media compositions with additives and complete media formulations not requiring supplementation with amino acids were tested. Refer to Table 27-1 for a list of all conditions (note that all media included penicillin/streptomycin in an effective amount, Invitrogen). After four days, samples were treated according to the manufacturer's instructions (Promega MTS Assay Kit).
  • the 9 best growth conditions plus the control were tested for their ability to support the expansion of cell populations over multiple passages.
  • umbilical-derived cells (022803 (P10)) were thawed from cryopreservation (cells were previously grown in Growth Medium) and plated directly into the experimental conditions (see Table 27-2).
  • Media used under all conditions contained penicillin/streptomycin as in the Growth Medium formulation (see above Examples).
  • Cells were passaged every 3-4 days and passaged/trypsinized equivalently in a base medium used for that growth condition (e.g. Advanced DMEM plus bFGF).
  • Umbilical cord-derived cells (042803 P21) and placenta-derived cells (071503 P10) grown in either Growth Medium or in Advanced DMEM and 10 ng/mL bFGF (Peprotech, Rocky Hill, N.J.)], in a gelatin-coated T225 flask were washed with phosphate buffered saline (PBS; Invitrogen) and trypsinized using 1 mL Trypsin/EDTA (Invitrogen). The trypsinization reaction was diluted by adding 20 mL of Advanced DMEM. Cells were centrifuged at 150 ⁇ g and the supernatant was aspirated off. The cells were washed in advanced DMEM.
  • PBS phosphate buffered saline
  • Trypsin/EDTA Invitrogen
  • Step 1 Wait at 4.0° C.
  • Step 2 1.0° C./min to ⁇ 4.0° C.
  • Step 3 25.0° C./min to ⁇ 40° C.
  • Step 4 10.0° C./min to ⁇ 12.0° C.
  • Step 5 1.0° C./min to ⁇ 40° C.
  • Step 6 10.0° C./min to ⁇ 90° C.
  • Step 7 END
  • Vials of cells were transferred to a nitrogen vapor storage unit (CryoMed 7400, ThermoForma, Marietta, Ohio) for 2 days before thawing rapidly in a 37° C. water bath with gentle swirling until no visible ice crystals could be detected.
  • the cells were added to 10 mL Growth Medium and centrifuged before the cell number and viability was estimated as before. Cells were seeded onto gelatin-coated flasks at 5,000 cells/cm 2 to determine whether the cells were able to attach and proliferate.
  • Cells were also assayed for senescence using the Senescence Cells Histochemical Staining Kit (Sigma, St Louis, Mo.) according to manufacturer's specification. Briefly, cells were seeded in gelatin-coated 6-well plates at 1.5 ⁇ 10 5 cells per well in the Growth Medium. After three days cells were washed once with PBS (Invitrogen, Carlsbad, Calif.) and incubated in the fixative solution for 6-7 minutes at room temperature (RT). Cells were then rinsed 3 times with PBS and 1.5 mL of staining solution was added to each well. Plates were incubated at 37 C in a shaker with gentle rocking until cells stained blue. The percentage of blue (senescent) cells was determined.
  • PBS Invitrogen, Carlsbad, Calif.
  • MTS assay was performed as described above. Similarly, cells were seeded in 8 replicates in 96-well plates at an initial concentration of 1,000 cells per well in the Growth medium (they were not re-seeded in Advanced DMEM).
  • DMEM/MCDB basal media
  • F10 medium basal media
  • Advanced DMEM containing L-glutamine
  • Growth Medium supported umbilical-derived PPC growth over four days in culture. There was a reduction in overall cell number from day 3 to day 4 in UltraCulture medium, and the cells appeared unhealthy.
  • ITS+3 was superior in the expansion of umbilical-derived PPCs over 4 days, relative to the basal medium alone.
  • bFGF the growth factor that stimulated the most expansion based on cell number at 4 days. This effect surpassed those results observed with Growth Medium. Interestingly, adding multiple growth factors did not further stimulate expansion under the conditions tested. Converting the data to population doublings between days 1 and 4, PDGF, EGF and IGF surpassed bFGF. This result may be explained by enhanced cell attachment and growth between time zero and day 1 for cells in bFGF.
  • the best defined medium formulation was Advanced DMEM (Invitrogen) containing high glucose supplemented with 10 ng/ml bFGF. This medium could support both the short and long term expansion of passage 10 umbilical-derived PPDCs previously grown in Growth Medium. Although the cells grew and the populations expanded well, growth was not as substantial as that obtained in Growth Medium with the serum supplementation.
  • Advanced-DMEM Invitrogen
  • Umbilical-derived cells grown from isolation in Advanced-DMEM were characterized and shown to have reduced expression of PDGFr-alpha and HLA-ABC.
  • Postpartum-derived cells grown in both Advanced DMEM and Growth Medium were further characterized using flow cytometry to determine the effect of growth medium on cell surface marker expression.
  • Gene expression profiles were compared with those of other cells including dermal and foreskin fibroblasts, and mesenchymal stem cells (MSCs). Switching media can alter both HLA-ABC and PDGFr expression as determined by flow cytometry.
  • HLA-ABC and PDGFr alpha expression was shown to be altered when postpartum cells were grown in two different media (Growth Medium versus Advanced DMEM supplemented with basic fibroblast growth factor (bFGF)). This finding was interesting and triggered further investigations into whether this phenomenon was due, for example, to the promotion of a different subpopulation of postpartum-derived cells, or whether a specific population of postpartum cells had the ability to modulate the expression of these cell surface markers in different growth media.
  • bFGF basic fibroblast growth factor
  • umbilicus-derived postpartum cells were grown in Growth Medium, Advanced DMEM plus bFGF, or switched between the two, and then analyzed for HLA-ABC and PDGFr expression by flow cytometry.
  • RNA was isolated from cultures of these cells grown under identical conditions, and PCR was performed to determine the expression patterns of genes previously found to be exclusively expressed (versus other cell populations). Differences between media conditions were analyzed, as were differences between other control cell types.
  • Postpartum-derived cells (umbilicus 063004B) grown in Advanced DMEM supplemented with bFGF (as described in Example 27) from isolation were passaged at either P3 or P4, and at that point switched to Growth Medium. Cells were plated at 5,000 cells per cm-squared on standard gelatin coated tissue culture flasks. Flow cytometry was performed on P3 cells for PDGFr alpha, HLA-ABC, and HLA-DRDPDQ, and the respective controls lacking primary antibody (IgG-PE, and IgG-FITC). P4 passaged/media switched cells were grown until passage 8 by passaging every 3 or 4 days, at which time flow cytometry was performed for the cell surface markers.
  • the reciprocal experiment was also performed in which cells grown in Growth Medium were switched to Advanced DMEM supplemented with bFGF (10 ng/ml).
  • Umbilicus-derived (063004B) postpartum cells were grown in Growth Medium from isolation until passage 12.
  • cells were switched to Advanced DMEM+bFGF and grown until passage 14, at which time flow cytometry was performed.
  • umbilicus-derived (090304A) postpartum cells were grown in Growth Medium from isolation until passage 8, and then switched at passage 9 to Advanced DMEM+bFGF. Cultures were maintained until passage 10 at which time flow cytometry was performed.
  • umbilicus-derived (042303) postpartum cells were grown in Growth Medium from isolation until passage 10, at which time they were cryopreserved in Growth Medium supplemented with 10% DMSO (Sigma, St. Louis, Mo.) and placed in a liquid nitrogen container (protocol as described in Example 27). Cells were later thawed and directly plated at 5,000 cells per cm-squared on gelatin coated flasks in Advanced DMEM supplemented with bFGF (10 ng/ml). The cultures were maintained until passage 33, at which time flow cytometry was performed for the cell surface markers of interest.
  • DMSO Sigma, St. Louis, Mo.
  • Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) were grown in Growth Medium in gelatin-coated T75 flasks.
  • Mesenchymal Stem Cells (MSCs, Cambrex, Walkersville, Md.) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex).
  • signature genes oxidized LDL receptor, interleukin-8, renin and reticulon
  • PCR was performed on cDNA samples using Assays-on-DemandTM gene expression products: oxidized LDL receptor (Hs00234028), renin (Hs00166915), reticulon (Hs00382515), IL-8 (Hs00174103) and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 min and 95° C. for 10 min followed by 40 cycles of 95° C. for 15 sec and 60° C. for 1 min.
  • PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass., USA) to confirm the results from real-time PCR.
  • PCR was performed using 2 ⁇ L of cDNA solution, 1 ⁇ AmpliTaq Gold universal mix PCR reaction buffer (Applied Biosystems, Foster City, Calif.) and initial denaturation at 94° C. for 5 minutes.
  • Amplification was optimized for each primer set.
  • IL-8 and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles), for renin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C.
  • Example 27 umbilicus-derived postpartum cells grown in Growth Medium and Advanced DMEM+bFGF from isolation were tested utilizing flow cytometry for a full battery of antibodies to cell surface markers. While the expression of most markers was similar, two markers substantially differed in their expression between the media formulations. Both HLA-ABC and PDGFr-alpha were expressed in Growth Medium, whereas neither was expressed in Advanced DMEM (Tables 27-3 & 27-4 in Example 27).
  • umbilicus-derived postpartum cultures were first grown in one of the two aforementioned media compositions and then switched to the other.
  • cells were grown initially in Advanced DMEM+bFGF and then switched to Growth Medium, cell cultures immediately turned on expression of HLA-ABC in a graded fashion with increasing passage.
  • PDGFreceptor alpha did not appear to turn on, even with extended passage.
  • HLA-ABC expression was retained in the short term (063004B+P14, 090304A+P10), but after several passages, HLA-ABC expression was lost (042303+P33).
  • PDGF receptor alpha expression was lost as early as the second passage in Advanced DMEM+bFGF.
  • results of real-time PCR for selected “signature” genes performed on cDNA from cells derived from human umbilical cord-derived cells grown in two different media, adult fibroblasts, and MSCs indicate that the levels of reticulon and oxidized LDL receptor RNA were higher in umbilical cord-derived cells compared to other cell types.
  • the results also confirmed that the expression pattern of signature genes did not change with medium regardless of whether cells were cultured in Advanced DMEM+10 ng/mL bFGF, or in Growth Medium from isolation, or were subsequently switched from one medium to the other.
  • the results of real-time PCR were confirmed by conventional PCR. Of note, was that bands, while existent, were less intense for oxLDL receptor (063004B P14, 090304A P10). Since conventional PCR is not quantitative, however, potential differences in level of expression could not be discerned here.
  • Umbilicus-derived cells did not express renin, a marker present in placental-derived cells.
  • cytokine, IL-8 The expression of the cytokine, IL-8, in postpartum-derived cells was elevated in both Growth Medium-cultured and Advanced DMEM+10 ng/mL bFGF-cultured umbilical cord-derived cells. This observation was confirmed by conventional PCR.
  • HLA-ABC HLA-ABC
  • PDGF-r alpha HLA-DRDPDQ
  • HLA-ABC and PDGFr alpha expression can be modulated based on changes in the growth medium of the cells. This observation could have significant implications for the rejections of grafts or implantation of therapeutic cells.
  • the HLA-DRDPDQ marker was never expressed regardless of media composition.
  • signature genes such as oxidized LDL receptor and reticulon were unchanged despite the composition of medium used to grow these umbilicus-derived postpartum cells. Because the levels of these genes were detectably higher than in other cell types such as MSCs, these genes in fact constitute a useful signature to determine and distinguish umbilicus-derived postpartum cells. Whether separate subpopulations may exist with in the umbilicus-derived population remains a question, but the consistency of the signature gene expression profile is powerful evidence that any such subpopulations are closely related to each other and more distantly related if at all to other cell types tested.
  • HLA-ABC expression varied suggests that, in fact, HLA-ABC expression can be modulated. Whether this could impact graft rejection is a point of further interest in that a cell population lacking the expression of this marker may be able to engraft better, potentially in the absence of immunosuppressants.
  • PDGFr alpha can be shut off easily and cannot be immediately turned on may indicate the delicate nature and rapid turnover of growth factor receptors and their requirement for very particular growth conditions for their expression. Given a lack of such receptors (or any other growth factor receptor because of, for example, media changes), cell potency may be altered in particular animal model paradigms.
  • umbilicus-derived cells can be cultured in Growth Medium in flasks with gelatin-coated surfaces.
  • This example explored whether two of the components in a preferred cell culture protocol were required. Tested, in particular, were whether (1) beta-mercaptoethanol (BME), as an additive in the standard Growth Medium, was required for growth, and (2) whether gelatin (an animal derived protein) was required as a coating for the cell culture flasks. Additionally, the effectiveness of a synthetic coating to promote attachment, CELLBIND® (Corning, Corning, N.Y.), was tested as an alternative to gelatin or other animal-derived products. CellBIND® is a patented, commercially available flask coating that uses highly reactive plasma to reduce the aromatic groups of the polystyrene surface.
  • Dulbecco's Modified Essential Media DMEM
  • Penicillin and Streptomycin were obtained from Invitrogen (Carlsbad, Calif.).
  • Fetal bovine serum FBS (defined bovine serum) was obtained from Hyclone (Logan, Utah). Beta-mercaptoethanol was obtained from Sigma (St. Louis, Mo.).
  • Umbilicus-derived cells were isolated in two mediums (Growth Medium, and Growth Medium without BME) and in vessels coated with either gelatin or CELLBIND®). Thus, four sets of conditions were tested: (1) Gelatin-Coating/Growth Medium (normal conditions), (2) CELLBIND®-Coating/Growth Medium, (3) Gelatin-Coating/Growth Medium without BME and (4) CelBIND®-Coating/Growth Medium without BME. The cells were serially passaged from isolation until senescence in their designated set of conditions. Population growth kinetics were calculated under each set of conditions to determine the effects of media and surface coating of the vessels, and to assess changes in growth. The expression of a set of cell surface markers was assessed as an initial screen for phenotypic changes.
  • Postpartum cells were generally isolated as described previously herein. The tissue was minced and divided into approximately equal portions. One portion was isolated in standard Growth Medium (DMEM:LG, 15% FBS; beta-mercaptoethanol (1 ⁇ l per 100 mL); 50 microliter/ml penicillin/50 microgram/ml streptomycin (1 mL per 100 mL (10,000 Units per mL)). The other portion was isolated in Growth Medium as described without beta-mercaptoethanol (BME) (Growth Medium w/o BME).
  • DMEM standard Growth Medium
  • BME beta-mercaptoethanol
  • Cells isolated from each portion of the cord tissue were seeded at 3000 cells/cm 2 onto gelatin-coated T225 flasks and CELLBIND-coated T225 flasks (Corning) in the same medium in which they were isolated. Cells were grown at 37° C. in a humidified, 5% CO 2 atmosphere. As cells reached about 85% confluence they were trypsinized and counted. Viable cells were seeded at 5000 cell/cm 2 into new flasks with appropriate medium and surface coating. Cell Yield, population doublings (ln(cell final/cell initial)/ln2) and doubling time (time in culture (h)/population doubling) were determined at each passage. The process was repeated until cells appeared to reach senescence, i.e. the cells appeared or lose their proliferative capabilities, or failed to proliferate further.
  • Cell surface markers were analyzed by flow cytometery essentially as described herein above. Analyses were performed at passages 3, 7 and 15 for cells grown on gelatin in either standard Growth Medium or in Growth Medium without BME, and at passage 7 for cells grown on CellBIND-coatings in either Growth Medium or Growth Medium without BME. In particular, after trypsinization and counting, 500,000 cells were suspended in 100 ⁇ l of Phosphate Buffered Saline with 3% Fetal Bovine Serum per sample. To each sample 20 ⁇ l of the antibody corresponding to the marker being analyzed was added. The cells were incubated at 4° C. for 1 hour.
  • Cells isolated from umbilical cords can be cultured in media with or without BME, and on either gelatin-coated or CELLBIND-coated vessels. The greatest rate of proliferation (0.51 doublings/day) in these experiments was observed with cells cultured in Growth Medium without BME on gelatin coated flasks. Umbilicus-derived cells proliferated at higher rates in Growth Medium without BME as reflected by a greater number of doublings per day; 0.51 doublings per day (Growth Medium without BME/Gelatin) compared to 0.33 doublings per day (Standard Growth Medium/Gelatin). Cells grew on the CellBIND-coated surfaces in medium with or without BME.
  • Umbilicus-derived cells cultured in gelatin-coated vessels in either Growth Medium or in Growth Medium without BME were able to expand for up to about 120 days.
  • Umbilicus-derived cells also grew in either Growth Medium or Growth Medium without BME, on the CELLBIND-coated surface for up to about 103 days.
  • Dulbecco's Modified Essential Media DMEM
  • Phosphate buffered saline Penicillin and Streptomycin
  • dispase were obtained from Invitrogen (Carlsbad, Calif.).
  • Fetal bovine serum FBS (defined bovine serum) was obtained from Hyclone (Logan, Utah).
  • Collagenase, hyaluronidase, and beta-mercaptoethanol were obtained from Sigma (St. Louis, Mo.).
  • Human umbilical cords were obtained from the National Disease Research Interchange (NDRI, Philadelphia, Pa.) following normal deliveries. To remove blood and debris, the cord was washed in Dulbeco's Modified Eagles Medium (DMEM-low glucose) or phosphate buffered saline (PBS). The tissues were then mechanically dissociated in tissue culture plates until the tissue was minced to a fine pulp. The minced tissue was transferred to a 50-milliliter conical tube and digested in an enzyme mixture containing 500 Units/milliliter collagenase, 500 Units/milliliter dispase, and 50 Units/milliliter hyaluronidase.
  • DMEM-low glucose Dulbeco's Modified Eagles Medium
  • PBS phosphate buffered saline
  • test culture medium a specific media formulation as outlined in Table 30-1 below.
  • the conical tubes containing the tissue, medium, and digestion enzymes were incubated at 37° C. in an orbital shaker at 225 rpm for 2 hrs.
  • the tissues were centrifuged at 150 ⁇ g for 5 minutes, and the supernatant was aspirated.
  • the pellet was resuspended in 20 milliliters of test culture media (Table 30-1).
  • the cell suspension was filtered through a 40-micron nylon BD FALCON Cell strainer (BD Biosciences, San Jose, Calif.).
  • the supernatant was again aspirated, and the cells were resuspended in 50 milliliters of fresh test culture medium. The process was repeated twice.
  • the supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh test culture medium.
  • the number of viable cells was determined using a Guava instrument (Guava Technologies, Hayward, Calif.). Cells were then plated at seeding densities of 300 cells/cm 2 , 2000 cells/cm 2 , or 5000 cells/cm 2 on gelatin-coated tissue culture flasks. Cells were cultured in medium and under conditions described in Table 30-1. Growth Media controls were used to determine viability of the isolates for each media type. TABLE 30-1 Media formulations and culture conditions used to isolate umbilical cells from umbilical cord tissue.
  • FACs Analysis Flow cytometry analysis was performed on umbilicus-derived cells grown according the media formulations and culture conditions described in Table 30-1. Cells were expanded to passage four or five in test culture medium on gelatin-coated T225 flasks at 37° C. and 5% carbon dioxide. Adherent cells were washed in PBS and detached with Trypsin/EDTA (Gibco). Cells were harvested, centrifuged and resuspended in 3% (v/v) FBS in PBS at a concentration of 1 ⁇ 10 7 cells/mL. The specific antibody was added to 100 microliters of cell suspension and the mixture was incubated in the dark for 30-45 minutes at 4° C.
  • PCR was performed on cDNA samples using Assays-on-DemandTM gene expression products: renin (Hs00166915), oxidized LDL receptor (Hs00234028), reticulon (Hs00382515), IL-8 (Hs00174103), and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 min and 95° C. for 10 min followed by 40 cycles of 95° C. for 15 sec and 60° C. for 1 min.
  • Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass., USA) to confirm the results from real-time PCR. PCR was performed using 2 ⁇ L of cDNA solution, 1 ⁇ AmpliTaq Gold universal mix PCR reaction buffer (Applied Biosystems, Foster City, Calif.) and initial denaturation at 94° C. for 5 minutes. Amplification was optimized for each primer set. For IL-8 and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles), for renin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C.
  • ELISA Frozen umbilicus-derived cells from different isolation conditions were thawed at passage 4 and seeded onto gelatin coated T75 flasks at 5000 cells/cm 2 each containing 15 milliliters of their respective growth medium (Table 30-1). Cells were cultured for 24 hours at 37° C. in 5% carbon dioxide and atmospheric oxygen. The medium was then changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 Units/milliliter) and streptomycin (50 ug/mL, Gibco)) and further cultured for 8 hours. Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000 ⁇ g for 5 min and stored at ⁇ 20° C.
  • tissue inhibitor of metalloprotease-1 (TIMP1), platelet-derived epithelial growth factor bb (PDGFbb), Keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), Heparin-binding epidermal growth factor (HB-EGF), tissue inhibitor of metalloprotease-2 (TIMP2), monocyte chemotactic protein-1 (MCP1), interleukin-6 (IL6), interleukin-8 (IL8), transforming growth factor alpha (TGFa), brain-derived neurotrophic factor (BDNF), stromal-derived factor 1B (SDF1B), cilliary neurotrophic factor
  • Umbilicus-derived cells were isolated using different media compositions and culture conditions identified in Table 30-1. Some isolated cell populations were cultured under both normoxic and hypoxic (5% O 2 ) atmospheric conditions. Cells were also isolated using low (300 cell s/cm 2 ) or high (2000 cells/cm 2 ) initial seeding density. Ten combinations of growth medium and growth conditions supported the growth of umbilicus-derived cells. These include EGM2, REGM and SMGM2, all under both normoxic and hypoxic atmospheric conditions. Sonic hedgehog medium and KGM, under hypoxia and normoxia respectively, also supported cell growth.
  • Umb032905 isolated in SMGM2 medium under hypoxic conditions, demonstrated the most robust cell expansion potential of greater than 32 population doublings. The population reached senescence at day 74 for cells isolated at low seeding density, and at 64 days for cells isolated at high seeding density. Modest expansion potential was observed in umb052405 isolated in REGM and cultured in normoxic or hypoxic conditions (isolation 3,4). These conditions yielded 18 and 10 population doublings, respectively. TABLE 30-4 Maximal number of cell population doublings achieved.
  • Cell populations generated from different isolation conditions were characterized by flow cytometry. Ten cell lines out of the twenty-two isolation conditions were expanded to passage 4 and cell surface marker profile was determined. Most of the cell isolations showed positive staining for CD10, CD13, CD44, CD49c, CD73, CD90, and HLA-ABC. The cells were negative under the conditions tested for surface expression of CD31, CD34, CD45, CD117, CD141, CD184, and HLA-DR, DP, DQ. Most markers showed a similar expression pattern to cells isolated using the preferred conditions described above in prior examples, as well as to other cell isolations.

Abstract

Cells derived from human umbilical cords are disclosed along with methods for their therapeutic use. Isolation techniques, culture methods and detailed characterization of the cells with respect to their cell surface markers, gene expression, and their secretion of trophic factors are described.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This is a continuation-in-part of U.S. patent application Ser. No. 10/877,012, filed Jun. 25, 2004, which claims benefit of U.S. Provisional Application Ser. No. 60/483,264, filed Jun. 27, 2003, the entire contents of each of which are incorporated by reference herein. This also claims benefit of U.S. Provisional Application No. 60/639,088, filed Dec. 23, 2004, the entirety of which is also herein incorporated by reference.
  • FIELD OF THE INVENTION
  • This invention relates to mammalian, preferably human, cell therapy, and more particularly to isolated cells derived from postpartum umbilicus, methods of deriving such cells, and methods of their use and growth on various media, including media which are serum-containing or serum-free.
  • BACKGROUND OF THE INVENTION
  • As the modern understanding of disease has advanced, the potential utility of cell therapy for improving the prognosis of those afflicted has resulted in increased interest in new sources of human cells useful for therapeutic purposes. One such source of human cells is postpartum tissues, and in particular, the umbilicus or umbilical cord.
  • Recently, attention has focused on the banking of umbilical cord blood (or simply “cord blood”) as a potential source of, for example, hematopoietic cells for use by an individual for whom cord blood has been banked at birth. Such cells would be useful for those individuals, for example, who require therapeutic radiation which may eliminate functional portions of their immune system. Rather than requiring a bone marrow donor carefully matched to avoid rejection, the individual's own banked cord blood could be used to reconstitute the lost immune cells, and restore immune function.
  • Still more recently, there has been interest in obtaining stem cells from cord blood, due to the wider potential therapeutic applications of such cells. Stem cells are understood in general terms as cells that 1) have the ability to self-renew for long periods through cell division from a single cell; and 2) have the ability to differentiate into specific cell types given the proper conditions. Accordingly, stem cells are potentially useful in treating a population of individuals, and not merely the person from whose cord blood the cells were initially obtained.
  • In particular, cord blood has been considered as a source of hematopoietic progenitor stem cells. Banked (or cryopreserved) cord blood, or stem cells isolated therefrom have been deemed useful for hematopoietic reconstitution, for example in bone marrow and related transplantations. (Boyse et al., U.S. Pat. Nos. 5,004,681 and 5,192,553).
  • In addition to cord blood, other sources of therapeutic cells from the human umbilicus have been explored, including cells isolated from the Wharton's Jelly, umbilical vein or artery tissue, and the umbilical matrix itself. Such cells have been largely uncharacterized, or only minimally characterized with respect to their physiological, biochemical, immunological, and genetic properties.
  • For example, Purchio et al. (U.S. Pat. No. 5,919,702) have isolated chondrogenic progenitor cells (or prechondrocytes) from Wharton's Jelly. They reported the isolation of cells from human umbilical cord Wharton's Jelly by removing blood and blood vessels and incubating the tissue under conditions purported to allow the prechondrocytes to proliferate. As such, the method did not distinguish the desired cells from the different cell types present in Wharton's Jelly, but rather relied on migration from the tissue or selecting growth conditions favoring prechondrocytes. The prechondrocytes were expanded mitotically after they were established. Cells at passages 2 to 4 were reported as useful to produce cartilage, if triggered by the addition of exogenous growth factors, such as BMP-13 or TGF-beta. Uses of the cells for direct injection or implantation, or use with a hydrogel or tissue matrix, were proposed. However, it was considered important that the cells not exceed about 25% confluence. The cells were not characterized with respect to their biochemical or immunological properties, or with respect to their gene expression.
  • Weiss et al. (U.S. Patent Application Publication US2003/0161818) proposed procedures for isolating pluripotent or lineage-committed cells from mammalian Wharton's Jelly or non-blood umbilical cord matrix sources. The cells isolated were reported to differentiate into hematopoietic, mesenchymal or neuroectodermal lines. The cell lines were not characterized with respect to their identifying properties. Limited characterization was provided with respect to cells after differentiation towards neural lines. Reference was made to Wharton's Jelly-derived bovine and porcine cells that were CD34, CD45.
  • Weiss et al. also reported investigating transplantation of porcine umbilical cord matrix cells into rat brain. (Exp. Neur. 182: 288-299, 2003). No enzymatic treatment was used in the isolation procedure. They obtained two distinct populations—spherical and flat mesenchymal cells. The cells were genetically modified to express GFP. The cells did not appear to stimulate immune rejection when implanted cross-species.
  • Mitchell et al. (Stem Cells 21:50-60, 2003) reported obtaining Wharton's Jelly matrix cells from porcine umbilical cords. The undifferentiated cells were reported to be positive for telomerase and a subpopulation was also reported positive for c-kit expression, i.e., telomerase+, CD117+. The cells were also reported to produce alpha-smooth muscle actin, indicative of their myofibroblast-like nature. The cells purportedly could differentiate along neural lines in the presence of growth factors. However, both the differentiated and undifferentiated cells were found to express NSE, a marker for neural stem cells. The need for clonal lines and characterization in terms of proliferative capacity, karyotype analysis, and expression of HLA antigens was recognized.
  • Romanov et al. (Stem Cells 21(1): 105-110, 2003) reported a procedure to isolate mesenchymal stem cell (MSC)-like cells from human umbilical cord vein. Their procedure involved treatment of the excised vein tissue with collagenase and required that the enzymatic digestion be short (15 minutes) to obtain the cell population of interest (a subendothelial layer of the vein). In particular, the procedure avoided inclusion of smooth muscle cells (SMCs) and fibroblasts by leaving the deeper layers of the tissue intact, purportedly removing only the outer layers. The resulting “nearly homogenous” cell population was reported to contain approximately 0.5-1% endothelial cells that they also sought to avoid. The cells were reported to be predominantly CD34 and to produce alpha-smooth muscle actin.
  • Because of the diversity of cell populations that are found in umbilical cord matrix, there is a need in the art for methods of isolating defined non-blood cells and populations thereof derived from mammalian umbilical cord; as well as a need for cell lines derived from mammalian umbilicus that are characterized with respect to their biochemistry (e.g. secretion of growth factors), immunology (cell surface markers and potential to stimulate immune responses) and expression of various genes. This need is particularly compelling for cells derived from human umbilicus.
  • With regard to media for culturing cells most contain at least some fetal bovine or calf serum. Generally, commercially available media formulations utilize serum supplements of about 10-20% (v/v). The serum component is often integral to the survival and expansion of cell populations. A myriad of proteins are found in bovine serum, including for example PDGF and FGFs, known growth factors that can have significant influence on cell growth and differentiation on cell populations, including populations of stem and progenitor cells.
  • In spite of the advantages of including bovine serum (or serum of other species), there are, however, a number of disadvantages to supplementing medium with animal serum instead of using chemically-defined or serum-free media when culturing therapeutic cells or producing biologics. Cell and tissue homeostasis occurs under environmental conditions that lack blood-derived serum. Thus, long-term cell exposure to serum and blood-related products simulates a tissue injury paradigm. Further, lot-to-lot variation in composition of serum, including in the stimulatory proteins and any inhibitory substances requires time consuming and expensive pre-testing to ensure each batch meets the standards for therapeutic product development or production. Finally, increased concerns about the transmission of diseases such as bovine spongiform encephalopathy (BSE) from the use of animal-related products may ultimately retard or preclude FDA approval of cell-based therapies developed with foreign serum.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1: Long-term growth potential of umbilical cells isolated under different conditions. Conditions that allowed for more than 5 population doublings were further analyzed for long-term growth potential. Cells were cultured using the indicated media formulations and culture conditions until cells reached senescence. Senescence was determined when cells failed to achieve greater than one population doubling during the study time interval.
  • SUMMARY OF THE INVENTION
  • In a first aspect, the invention provides isolated umbilicus-derived cells (UDCs) that are derived from mammalian umbilical cord tissue substantially free of blood. The cells are capable of self-renewal and expansion in culture. The umbilicus-derived cells have the potential to differentiate into cells of other phenotypes. In preferred embodiments, the cells are derived from human umbilicus.
  • The cells have been characterized as to several of their cellular, genetic, immunological, and biochemical properties. For example, the cells have been characterized by their growth properties in culture, by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
  • In certain embodiments, the postpartum-derived cell is an umbilicus-derived cell. In other embodiments it is a placenta-derived cell. In specific embodiments, the cell has all identifying features of either of cell types UMB 022803 (P7) (ATCC Accession No. PTA-6067); or UMB 022803 (P17) (ATCC Accession No. PTA-6068).
  • In another of its several aspects, cell cultures comprising the isolated umbilicus-derived cells of the invention are provided. The cultures of umbilical cells are free of maternal cells in certain preferred embodiments.
  • Methods of culturing and expanding umbilicus-derived cells and cell cultures and populations comprising them are provided.
  • In another aspect of the invention isolated umbilicus-derived cells having specific cell surface marker expression profiles are provided, wherein particular cell surface marker proteins are produced. In particular, the cells produce one or more of CD10, CD13, CD44, CD73, CD90, CD141, PDGFr-alpha, or HLA-A, B, C. In addition, the cells do not produce one or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
  • The cells of the invention have also been characterized according to their expression of a wide variety of genes. Accordingly, another aspect of the invention provides isolated umbilicus-derived cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of genes for one or more of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus EIB 19 kDa interacting protein 3-like; AE binding protein 1; and cytochrome c oxidase subunit VIIa polypeptide 1 (muscle). In addition, these isolated human umbilicus-derived cells express a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3, wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, an ileac crest bone marrow cell, or placenta-derived cell. The cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes. Also provided are therapeutic cell cultures comprising the isolated human umbilicus-derived cells.
  • In another of its several aspects, the invention provides isolated human umbilicus-derived cells capable of self-renewal and expansion in culture and which have the potential to differentiate into cells of other phenotypes, wherein the cells do not stimulate allogeneic lymphocytes in a mixed lymphocyte reaction, and expresses PD-L2, but not HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2. In some preferred embodiments, the cells do not stimulate allogeneic PBMCs. More preferably they do not stimulate allogeneic lymphocytes, allogeneic T-cells, or naïve T-cells, or generate other adverse immunological responses in either matched or unmatched recipients. The cells also can produce vimentin and alpha-smooth muscle actin in certain embodiments.
  • In another aspect, the invention provides isolated human umbilicus-derived cells that secrete one or more of the angiogenic factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1. In certain embodiments, the cells secrete several or all of the aforementioned molecules. In other embodiments, the cells do not secrete one or more of the angiogenic factors SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA. In particular embodiments, they secrete few or none of those molecules.
  • In another aspect of the invention, therapeutic cell cultures are provided, the cell cultures comprising the isolated cells as described above for use in treating patients in need of angiogenesis-stimulating trophic factors. Such therapeutic cell cultures are also provided for use in treating a patient in need of neural growth stimulating trophic factors.
  • Methods of deriving cells from non-blood human umbilical tissue are provided. The cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes. The method comprises (a) obtaining human umbilical tissue; (b) removing substantially all of the blood to yield a substantially blood-free umbilical tissue, (c) dissociating the tissue by mechanical or enzymatic treatment, or both, (d) resuspending the tissue in a culture medium, and (e) providing growth conditions which allow for the growth of a human umbilicus-derived cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes. Preferred methods involve enzymatic treatment with, for example, collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided herein.
  • Isolated human umbilicus-derived cells derived by the above method are also provided herein. The cells maintain a consistent normal karyotype notwithstanding repeated passaging in certain embodiments. Also provided are cultures of human umbilicus-derived cells derived by the above method, wherein the cultures are free of maternal cells. Further provided are cells and cultures of such cells wherein the cells isolated by any of a variety of methods in any of a variety of media, even when cultured under a wide range range of conditions, are capable of maintaining a substantially consistent profile of expressed surface markers, or substantially consistent profile of expression of genes that characterize the cells, e.g. “signature gene profiles” including for example, expression patterns of which are specific to certain stem cells and/or to certain postpartum cells.
  • Co-cultures comprising the cells or cultures of the invention with other mammalian cells are also provided herein. Preferably these co-cultures comprise another human cell line whose growth or therapeutic potential, for example, is improved by the presence of the umbilicus-derived cells. Such co-cultures are useful for therapeutic application in vitro or in vivo.
  • Also provided herein are therapeutic compositions comprising an umbilicus-derived cell and another therapeutic agent, factor, or bioactive agent. Such factors include, but are not limited to, IGF, LIF, PDGF, EGF, FGF, as well as antithrombogenic, antiapoptotic agents, anti-inflammatory agents, immunosuppressive or immunomodulatory agents, and antioxidants. Such therapeutic compositions can further comprise one or more additional cell types in addition to the UDCs and the bioactive component.
  • In addition to the above, compositions derived from the cells are provided herein. Cell lysates, soluble cell fractions and membrane-enriched cell fractions are provided herein. Extracellular matrices derived from the cells, for example, comprising basement membranes are also useful and are provided herein.
  • Compositions of the invention also include conditioned culture media as provided herein. Such media have first been used to grow the cells or cultures of the invention, which during growth secrete one or more useful products into the medium. Conditioned medium from these novel cells are useful for many purposes, including for example, supporting the growth of other mammalian cells in need of growth factors or trophic factors secreted into the media by the cells and cultures of the invention, and promoting, for example, angiogenesis.
  • Methods are provided of inducing the cells to differentiate along a pathway towards progenitors of various cells, or even into terminally differentiated cells themselves. Such cells have utility for therapeutic treatment of certain conditions, disorders and disease states. Such cells also have utility for diagnostic protocols, such as for use in assays to identify therapeutic agents.
  • The invention also provides methods of utilizing the differentiated umbilicus-derived cells or the progenitors for therapeutic uses, including but not limited to angiogenic application, neuronal applications, soft tissue applications, occular applications, and applications wherein the cells are useful in treatment of heart, kidney, bone, cartilage, pancreas, liver, and other tissues alone or in combination with other therapeutic agents.
  • Kits are also provided herein. Kits useful for the growth, isolation and use of the umbilical-derived cells are provided.
  • In another of it several aspects, the invention provides isolated postpartum-derived cells comprising a “signature gene profile” which distinguishes the PPDCs from other cells of different origin and from other known toti-and pluripotent cells. Preferred signature gene profiles comprise those wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum. Also provide are such cells further comprising the ability to alter the expression of its cell surface markers when grown in medium containing serum relative to that in serum free medium. Of particular interest presently are cells wherein the markers for PDGFreceptor alpha and HLA-ABC can be altered.
  • Also provided are methods for the preparation of therapeutic cells or cultures comprising: isolating cells; initially expanding the cells to a useful number in a serum-containing medium which supports cell expansion but in which the cells produce a quantity of the cell surface marker HLA-ABC; transferring the cells to a medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC; and passaging the cells in the medium in which the cells produce a decreased amount of the HLA-ABC, thereby preparing a therapeutic cell or culture. In presently preferred embodiments, the medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC is a serum-free medium. The methods are particularly useful for the production of therapeutic cells or cultures for implantation or grafting.
  • Also provided in accordance with present invention are serum-free media for the expansion of postpartum-derived cells wherein the media has one or more growth factors added. Presently preferred growth factors to be added are bFGF, EGF, or PDGF.
  • Methods are also provided for isolating and culturing postpartum cells, particularly umbilicus-derived cells, in serum-containing and serum-free media, with or without beta-mercaptoethanol, with or without various growth factors, on surfaces coated with gelatin, or CELLBIND, or other treatments to allow attachment. Methods of culturing the cells also include varied conditions of oxygen, and other growth parameters.
  • In another aspect, the invention provides therapeutic cultures comprising postpartum-derived cells expanded in serum-free medium. Preferred cultures having a signature gene profile wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum.
  • Also provided are cell culture banks and depositories and the like comprising postpartum-derived cells and cultures of the invention.
  • These and further aspects of the invention will be described in greater detail below.
  • DETAILED DESCRIPTION
  • Definitions
  • Various terms used throughout the specification and claims are defined as set forth below.
  • Stem cells are undifferentiated cells defined by the ability of a single cell both to self-renew, and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute substantially to most, if not all, tissues following injection into blastocysts.
  • Stem cells are classified according to their developmental potential as: (1) totipotent; (2) pluripotent; (3) multipotent; (4) oligopotent; and (5) unipotent. Totipotent cells are able to give rise to all embryonic and extraembryonic cell types. Pluripotent cells are able to give rise to all embryonic cell types. Multipotent cells include those able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem cells (HSC) can produce progeny that include HSC (self-renewal), blood cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood). Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells; and cells that are unipotent are able to give rise to a single cell lineage (e.g., spermatogenic stem cells).
  • Stem cells are also categorized on the basis of the source from which they may be obtained. An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types. An embryonic stem cell is a pluripotent cell from the inner cell mass of a blastocyst-stage embryo. A fetal stem cell is one that originates from fetal tissues or membranes. A postpartum stem cell is a multipotent or pluripotent cell that originates substantially from extraembryonic tissue available after birth, namely, the placenta and the umbilical cord. These cells have been found to possess features characteristic of pluripotent stem cells, including rapid proliferation and the potential for differentiation into many cell lineages. Postpartum stem cells may be blood-derived (e.g., as are those obtained from umbilical cord blood) or non-blood-derived (e.g., as obtained from the non-blood tissues of the umbilical cord and placenta).
  • Embryonic tissue is typically defined as tissue originating from the embryo (which in humans refers to the period from fertilization to about six weeks of development. Fetal tissue refers to tissue originating from the fetus, which in humans refers to the period from about six weeks of development to parturition. Extraembryonic tissue is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryonic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta (which itself forms from the chorion and the maternal decidua basalis).
  • Differentiation is the process by which an unspecialized (“uncommitted”) or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example. A differentiated cell is one that has taken on a more specialized (“committed”) position within the lineage of a cell. The term committed, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type. De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell. As used herein, the lineage of a cell defines the heredity of the cell, i.e. which cells it came from and what cells it can give rise to. The lineage of a cell places the cell within a hereditary scheme of development and differentiation.
  • In a broad sense, a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors. By that definition, stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells. When referring to the cells of the present invention, as described in greater detail below, this broad definition of progenitor cell may be used. In a narrower sense, a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types. This type of progenitor cell is generally not able to self-renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a non-renewing progenitor cell or as an intermediate progenitor or precursor cell.
  • As used herein, the phrase differentiates into a mesodermal, ectodermal or endodernal lineage refers to a cell that becomes committed to a specific mesodermal, ectodermal or endodermal lineage, respectively. Examples of cells that differentiate into a mesodermal lineage or give rise to specific mesodermal cells include, but are not limited to, cells that are adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoietic, hemangiogenic, myogenic, nephrogenic, urogenitogenic, osteogenic, pericardiogenic, or stromal. Examples of cells that differentiate into ectodermal lineage include, but are not limited to epidermal cells, neurogenic cells, and neurogliagenic cells. Examples of cells that differentiate into endodermal lineage include, but are not limited to, pleurigenic cells, hepatogenic cells, cells that give rise to the lining of the intestine, and cells that give rise to pancreogenic and splanchogenic cells.
  • The cells of the present invention are generally referred to as umbilicus-derived cells (or UDCs). They also may sometimes be referred to more generally herein as postpartum-derived cells or postpartum cells (PPDCs). In addition, the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense. The term derived is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a growth medium to expand the population and/or to produce a cell line). The in vitro manipulations of umbilical stem cells and the unique features of the umbilicus-derived cells of the present invention are described in detail below.
  • Various terms are used to describe cells in culture. Cell culture refers generally to cells taken from a living organism and grown under controlled condition (“in culture” or “cultured”). A primary cell culture is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture. Cells are expanded in culture when they are placed in a growth medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells. When cells are expanded in culture, the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as doubling time.
  • A cell line is a population of cells formed by one or more subcultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been passaged. A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture. The primary culture, i.e., the first culture following the isolation of cells from tissue, is designated P0. Following the first subculture, the cells are described as a secondary culture (P1 or passage 1). After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number. The expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and time between passaging.
  • A conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide trophic support to other cells. Such trophic factors include, but are not limited to hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, and granules. The medium containing the cellular factors is the conditioned medium.
  • Generally, a trophic factor is defined as a substance that promotes or at least supports, survival, growth, proliferation and/or maturation of a cell, or stimulates increased activity of a cell.
  • When referring to cultured vertebrate cells, the term senescence (also replicative senescence or cellular senescence) refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's limit). Although cellular senescence was first described using fibroblast-like cells, most normal human cell types that can be grown successfully in culture undergo cellular senescence. The in vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide). Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone. Thus, cells made quiescent by removing essential growth factors are able to resume growth and division when the growth factors are re-introduced, and thereafter carry out the same number of doublings as equivalent cells grown continuously. Similarly, when cells are frozen in liquid nitrogen after various numbers of population doublings and then thawed and cultured, they undergo substantially the same number of doublings as cells maintained unfrozen in culture. Senescent cells are not dead or dying cells; they are actually resistant to programmed cell death (apoptosis), and have been maintained in their nondividing state for as long as three years. These cells are very much alive and metabolically active, but they do not divide. The nondividing state of senescent cells has not yet been found to be reversible by any biological, chemical, or viral agent.
  • As used herein, the term Growth Medium generally refers to a medium sufficient for the culturing of umbilicus-derived cells. In particular, one presently preferred medium for the culturing of the cells of the invention herein comprises Dulbecco's Modified Essential Media (also abbreviated DMEM herein). Particularly preferred is DMEM-low glucose (also DMEM-LG herein) (Invitrogen, Carlsbad, Calif.). The DMEM-low glucose is preferably supplemented with 15% (v/v) fetal bovine serum (e.g. defined fetal bovine serum, Hyclone, Logan Utah), antibiotics/antimycotics (preferably penicillin (100 Units/milliliter), streptomycin (100 milligrams/milliliter), and amphotericin B (0.25 micrograms/milliliter), (Invitrogen, Carlsbad, Calif.)), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis Mo.). In some cases different growth media are used, or different supplementations are provided, and these are normally indicated in the text as supplementations to, or deletions from the Growth Medium.
  • In other embodiments, the growth medium is not supplemented with serum, and more preferably the medium is not supplemented with any animal-derived protein material. This is presently preferred for example for growing cells for clinical or preclinical uses. Such media lacking serum altogether, and in even more preferably lacking any animal protein supplementation, may be used for both isolation and growth of cells in accordance with the instant invention.
  • Also relating to the present invention, the term standard growth conditions, as used herein refers to culturing of cells at 37° C., in a standard atmosphere comprising 5% CO2. Relative humidity is maintained at about 100%. While foregoing the conditions are useful for culturing, it is to be understood that such conditions are capable of being varied by the skilled artisan who will appreciate the options available in the art for culturing cells, for example, varying the temperature, CO2, rotation or agitation, relative humidity, oxygen, growth medium, and the like.
  • The following abbreviations are used herein:
  • ANG2 (or Ang2) for angiopoietin 2;
  • APC for antigen-presenting cells;
  • BDNF for brain-derived neurotrophic factor;
  • bFGF for basic fibroblast growth factor;
  • bid (or BID) for “bis in die” (twice per day);
  • BME for beta mercaptoethanol (or 2-mercaptoethanol);
  • BSP for bone sialoprotein;
  • CK18 for cytokeratin 18;
  • CXC ligand 3 for chemokine receptor ligand 3;
  • DAPI for 4′-6-diamidino-2-phenylindole-2-HCl
  • DMEM for Dulbecco's Minimal Essential Medium; also used herein as synonymous with Dulbecco's Modified Eagle Medium. The skilled artisan will appreciate the composition is the same by either name.
  • DMEM:lg (or DMEM:Lg, DMEM:LG) for DMEM with low glucose;
  • EDTA for ethylenediaminetetraacetic acid;
  • EGF for epidermal growth factor;
  • ERG for Electroretmalgram;
  • FACS for fluorescent activated cell sorting;
  • FBS for fetal bovine serum;
  • FCS, fetal calf serum;
  • GCP-2 for granulocyte chemotactic protein 2;
  • GFAP for glial fibrillary acidic protein;
  • HB-EGF for heparin-binding epidermal growth factor;
  • HCAEC for human coronary artery endothelial cells;
  • HGF for hepatocyte growth factor;
  • hMSC for human mesenchymal stem cells;
  • HNF-1 alpha for hepatocyte-specific transcription factor;
  • HUVEC for human umbilical vein endothelial cells;
  • I309 for a chemokine and the ligand for the CCR8 receptor and is responsible for chemoattraction of TH2 type T-cells. I309 binds to endothelial cells, stimulates chemotaxis and invasion of these cells, and enhances HUVEC differentiation into capillary-like structures in an in vitro Matrigel assay. Furthermore, 1309 is an inducer of angiogenesis in vivo in both the rabbit cornea and the chick chorioallantoic membrane assay (CAM).
  • IL-6 for interleukin-6;
  • IL-8 for interleukin-8;
  • K19 for keratin 19;
  • K8 for keratin 8;
  • KGF for keratinocyte growth factor;
  • MCP-1 for monocyte chemotactic protein 1;
  • MDC for macrophage-derived chemokine;
  • MIP1alpha for macrophage inflammatory protein 1alpha;
  • MIP1beta for macrophage inflammatory protein 1beta;
  • MSC for mesenchymal stem cells;
  • NHDF for normal human dermal fibroblasts;
  • NPE for Neural Progenitor Expansion media;
  • PBMC for peripheral blood mononuclear cell;
  • PBS for phosphate buffered saline;
  • PDGFbb for platelet derived growth factor;
  • PDGFr/alpha for platelet derived growth factor receptor alpha;
  • PD-L2 for programmed-death ligand 2;
  • PE for phycoerythrin
  • PO for “per os” (by mouth);
  • PPDC for postpartum-derived cell;
  • Rantes (or RANTES) for regulated on activation, normal T cell expressed and secreted;
  • rhGDF-5 for recombinant human growth and differentiation factor 5;
  • SC for subcutaneously;
  • SDF-1alpha for stromal-derived factor 1alpha;
  • SHH for sonic hedgehog;
  • SOP for standard operating procedure;
  • TARC for thymus and activation-regulated chemokine;
  • TCP for Tissue culture plastic;
  • TGFbeta2 for transforming growth factor beta2;
  • TGFbeta-3 for transforming growth factor beta-3;
  • TIMP1 for tissue inhibitor of matrix metalloproteinase 1;
  • TPO for thrombopoietin;
  • TuJ1 for BIII Tubulin;
  • UDC for umbilicus-derived cell;
  • VEGF for vascular endothelial growth factor;
  • vWF for von Willebrand factor;
  • alphaFP for alpha-fetoprotein;
  • Description
  • Various patents and other publications are cited herein and throughout the specification, each of which is incorporated in its entirety by reference herein.
  • In a first aspect, the invention provides isolated umbilicus-derived cells comprising cells derived from human umbilical cord tissue substantially free of blood. The cells are capable of self-renewal and expansion in culture. The umbilicus-derived cells have the potential to differentiate into cells of other phenotypes. In preferred embodiments, the cells can differentiate into any cell of ectodermal, mesodermal, or endodermal origin. The invention provides, in one of its several aspects cells that are isolated from human umbilical tissues. The cells, as disclosed herein, preferably are not derived from umbilical cord blood. Nor are they endothelial cells derived from, for example, blood vessels. Rather, the cells are derived of the remaining umbilicus tissues.
  • The cells have been characterized as to several of their cellular, genetic, immunological, and biochemical properties. For example, the cells have been well-characterized by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
  • In another of its several aspects, cell cultures comprising the isolated umbilicus-derived cells of the invention are provided. The cultures are free of maternal cells in preferred embodiments herein. Also preferred are cells that have a normal karyotype, and those that maintain their karyotype as they are passaged. Most highly preferred are those cells that have and maintain a normal karyotype throughout passaging until at least after senescence.
  • Methods of culturing and expanding umbilicus-derived cells and cell cultures comprising them are provided. Presently preferred are cells that require no added growth factors, but rather, are capable of expansion in many available culture media, especially those supplemented with for example, fetal bovine serum. Preferred media for culturing and expansion of the cells include Growth Medium, defined herein as medium comprising Dulbecco's Modified Essential Media (also abbreviated DMEM herein). Preferred is DMEM-low glucose (also DMEM-LG herein) (Invitrogen, Carlsbad, Calif.). The DMEM-low glucose is most preferably supplemented with 15% (v/v) fetal bovine serum (e.g. defined fetal bovine serum, Hyclone, Logan Utah), antibiotics/antimycotics (preferably penicillin at 100 Units/milliliter, streptomycin at 100 micrograms/milliliter, and amphotericin B at 0.25 micrograms/milliliter; (Invitrogen, Carlsbad, Calif.)), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis Mo.).
  • The skilled artisan will appreciate that the Growth Medium can be variously supplemented and altered in any of the ways known in the art, and may be optimized for particular reasons. Also preferred for either isolation of cells from postpartum tissue or growth and culturing of the cells are those media which lack any added serum, and those media which are serum-free and animal protein-free. Such media are particularly preferred for uses in isolating or culturing cells for preclinical or clinical use. Also useful are media which are lacking beta-mercaptoethanol as an additive. In addition, the cells are able to grow in many other culture media, including chemically defined media in the absence of added serum. Several such media are exemplified below. In addition to routine culturing and maintenance of the cells, many other media are known in the art for affecting differentiation of such potent cells into specific types of cells or progenitors of specific cells. The skilled artisan will appreciate that these media are useful for many purposes, and are included within the scope of the invention, but they are not necessarily preferred for routine culturing and expansion.
  • In addition to the flexibility of the cells with respect to culturing medium, the cells can grow under a variety of environmental conditions. In particular, the cells can grow under a wide range of atmospheric conditions. Presently preferred are atmospheres which range from about 5% O2 to about 20% or more O2. The cells grow and expand well in Growth Medium under these conditions, typically in the presence of about 5% CO2, and the balance of the atmosphere as nitrogen. The skilled artisan will appreciate that the cells may tolerate broader ranges of conditions in different media, and that optimization for specific purposes may be appropriate.
  • Although the cells have not demonstrated any requirements for specific growth factors, the cells have demonstrated a requirement for L-valine over D-valine. Thus preferred culture media should have the L-isomer of this amino acid present. In addition to the demonstrated flexibility of the cells with respect to their growth requirements, preferred are the isolated cells that can attach and expand on either a coated or an uncoated tissue culture vessel. Tissue culture vessels include plates, flasks, tubes and the like, made of any of the materials known in the art—e.g. plastic, polystyrene, glass. Such vessels, where coated, are coated with any of a variety of compounds as are known in the art. Presently preferred coated vessels comprise a coating with gelatin, laminin, collagen, polyomithine, polylysine, vitronectin, or fibronectin, for example.
  • The isolated cells of the invention also preferably expand in the presence of from about 2% to about 15% serum, preferably Fetal Bovine Serum, and more preferably defined Fetal Bovine Serum. The cells also expand in the presence or absence of beta-mercaptoethanol, and in the presence or absence of added growth factors including one or more of EGF, bFGF, PDGF, VEGF, IGF-I, and LIF. The skilled artisan will appreciate that these flexible growth requirements allow for many options when culturing or working with these cells. In certain embodiments, cells are grown in Advanced DMEM (Gibco) containing any one of the previously mentioned growth factors, particularly bFGF. As described above, while presently Growth Medium is preferred, there is no absolute requirement for serum; growth has been accomplished in serum-free medium.
  • The cells can also be grown as attachment-dependent cultures on a variety of surfaces. Presently preferred are surfaces treated with gelatin, more prefereably CELLBIND where culture completely free of animal protein are required. Other coatings are compatible with growth of attachment-dependent cultures such as those provided herein.
  • In a preferred embodiment, the cells have excellent doubling and expansion potential and are suitable for use in diagnostic and therapeutic applications because their numbers can be scaled up readily. The isolated cells preferably can double sufficiently to generate yields of greater than about 1014 cells in less than about 80 days in culture when seeded at about 103 cells/cm2 in a suitable medium. More preferred are cells that can double sufficiently to generate greater than about 1015 cells in less than about 80 days in culture when seeded at about 5,000 cells/cm2. Still more preferred is the isolated cell which can double sufficiently to generate greater than about 1017 cells in less than about 65 days in culture when seeded at about 5,000 cells/cm2.
  • The isolated cells of the invention can undergo extensive doublings. Preferably, the cells undergo at least 30 doublings before reaching senescence. More preferably, at least 40 doublings in culture are attainable. Still more preferable are those cells that can achieve more than 40 doublings before becoming senescent. The cells of the invention are preferably able to undergo division over a longer period of time, than are, for example, human mesenchymal cells, or human fibroblasts, cultured under the same conditions.
  • In one embodiment, umbilical cells are isolated in the presence of one or more enzyme activities. A broad range of digestive enzymes for use in cell isolation from tissue are known in the art, including enzymes ranging from those considered weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin). Presently preferred are mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and deoxyribonucleases. More preferred are enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities. Presently preferred are cells that are isolated in the presence of one or more activities of collagenase, hyaluronidase and dispase. More preferred are those cells isolated in the presence of a collagenase from Clostridium histolyticum, and either of the protease activities, dispase and thermolysin. Still more preferred are cells isolated with collagenase and dispase enzyme activities. Also preferred are such cells isolated in the presence of a hyaluronidase activity, in addition to collagenase and dispase activity.
  • In another aspect of the invention, isolated umbilicus-derived cells having specific cell surface marker expression profiles are provided. Preferred cells are characterized in either their production or lack of production of one or more cell surface markers selected from CD10, CD13, CD31, CD44, CD45, CD73, CD90, CD117, CD141, PDGFr-alpha, HLA-A, B, C, and HL-DR, DP, DQ. More preferred are cells that are characterized with respect to their production, or lack thereof, of several (e.g., two, four, five, or eight), or even ten of, or all of, the foregoing, to provide a profile of the cell. In preferred embodiments, the cells produce one or more of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, or HLA-A, B, C. More preferred are cells that express several or all of the foregoing markers. Still more preferred are those cell which express each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C.
  • Preferred cells may also be characterized with respect to markers they do not produce, and such information is also useful in forming a characterization or immunological profile of the cell. Preferably, the cells do not produce one or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry. More preferred are cells that do not produce several or more of the foregoing. Still more preferred are cells for which production of none of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ can be detected by flow cytometry under the conditions described herein.
  • In other preferred embodiments, the cells can be shown to produce several or more of the CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C and concomitantly not produce one, or several, or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ as detected by flow cytometry. More highly preferred are cells that produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C and concomitantly not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ as detected by flow cytometry.
  • Such cells are highly characterized with respect to their production of these cell surface proteins. In preferred embodiments, the characterization of the cells with respect to such production remains substantially constant and does not change substantially with variations in isolation procedure, passage, culture conditions, or even the coating or lack thereof on a tissue culture vessel.
  • The cells of the invention have also been characterized according to their expression of a wide variety of genes. Accordingly, another aspect of the invention provides isolated human umbilicus-derived cells, which are characterized, relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, in their expression of genes for: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; as well as interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3.
  • In one embodiment, preferred cells have the cell surface marker characterization described above and are further characterized in their relative gene expression. For example, some preferred cells have the cell surface characterization described above and express a gene for one or more of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; or tumor necrosis factor, alpha-induced protein 3. More preferred are those cells that express a gene for several, (e.g., at least two, four, five or more) of each of the foregoing. The cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes. Also provided are therapeutic cell cultures comprising the isolated human umbilicus-derived cells.
  • Also provided are UDCs comprising the cell surface marker characterization and which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of one or more genes selected from: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa. Preferred are cells that have reduced expression of a gene for several (e.g., at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more, or even all) of the foregoing.
  • Still more highly preferred are those cells having the cell surface marker characterization as described herein above and which also express a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte); chemokine (C-X-C motif) ligand 3; or tumor necrosis factor, alpha-induced protein 3, and which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of one or more genes selected from: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa.
  • In another of its aspects, the invention provides isolated human umbilicus-derived cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression of genes for each of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; and which express a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3, wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, an ileac crest bone marrow cell, or placenta-derived cell. Such cells need not be characterized with respect to their cell surface markers, but such cells preferably are capable of self-renewal and expansion in culture and also have the ability to differentiate in culture. Most preferred are cells which are isolated as adherent cells as described below, however, such cells can also be grown after isolation in a spherical form under certain conditions, thus they are not obligatorily adherent.
  • Certain preferred embodiments include cells as described above which also produce vimentin or alpha-smooth muscle actin. More preferred are cells that produce both vimentin and alpha-smooth muscle actin. The production of these proteins appears to distinguish the cells of the instant invention from hematopoietic cells isolated form umbilical cord blood for example.
  • In preferred embodiments, the above gene expression profiles are substantially stable and do not vary with passaging or normal culturing conditions. Of course, it is understood that such a profile may vary when cells are grown under conditions which stimulate or induce differentiation into other phenotypes, for example, or the expression of a different set of genes.
  • The invention also provides therapeutic cell cultures comprising the cell having the cell surface production or gene expression profiles, or both, as described above. Cell banks comprising therapeutic cultures are similarly included with in the scope of the invention. For example, a cell bank may include cultured cells of the invention at various passages, as well as cells of the invention which have been induced to differentiate to different phenotypes. Other cells may be successfully banked either separately or in co-culture with the cells of the invention. A complete cell bank may include the banked cells of a wide variety of individuals. In preferred embodiments, banked cells are stored cryopreserved at −180° C. for example. Cells are preferably stored at −90° C. in some embodiments. The cells of the invention are readily cryopreserved under a variety of conditions such as are known in the art.
  • In preferred embodiments, the cells lack the cell surface molecules required to substantially stimulate allogeneic lymphocytes in a mixed lymphocyte reaction. In more preferred embodiments, the cells lack the surface molecules required to substantially stimulate CD4+ T-cells in in vitro assessments, or in vivo in allogeneic, syngeneic, or autologous recipients. Still more preferred are those cells that do cause not any substantial adverse immunological consequences for in vivo applications. The therapeutic cell cultures lack detectable amounts of at least two, or several, or all of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as determined by flow cytometry. Those lacking all of the foregoing are most preferred. Also preferred are therapeutic cell cultures which further lack detectable amounts of one or both of the immunomodulating proteins HLA-G and CD178, as determined by flow cytometry. Also preferred are therapeutic cell cultures which express detectable amounts of the immuno-modulating protein PD-L2, as determined by flow cytometry. In one embodiment, the therapeutic cell culture does not substantially stimulate a lymphocyte mediated response in vitro, as compared to allogeneic controls in a mixed lymphocyte reaction.
  • In another of its several aspects, the invention provides isolated human umbilicus-derived cells capable of self-renewal and expansion in culture and which have the potential to differentiate into cells of other phenotypes, wherein the cells do not substantially stimulate allogeneic lymphocytes in a mixed lymphocyte reaction. More preferred are cells which do not substantially stimulate CD4+ T cells, and which produce PD-L2, but not one or more of HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2. The cells also can produce vimentin or alpha-smooth muscle actin in certain embodiments. More preferred are cells producing both vimentin and alpha-smooth muscle actin. Still more preferred are those cells not producing any of HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2.
  • Cells of the invention which secrete useful molecules, for example, growth factors, are presently preferred. Such cells have utility not only for the their cellular properties but for their secreted molecules, for example in conditioned medium, or cell-free lysates. In another aspect, the invention provides isolated human umbilicus-derived cells that secrete one or more of the angiogenic factors MCP-1, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, TPO, or TIMP1. In certain embodiments, the cells secrete all of the foregoing factors. The cells do not secrete one or more of the angiogenic factors SDF-1 alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA. In certain embodiments they secrete none of SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA.
  • In another aspect, the cells of the invention are defined according to a combination of many of the characteristics provided herein. For example, the invention provides isolated umbilicus-derived cells comprising L-valine-requiring cells derived from mammalian postpartum tissue substantially free of blood. The cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes; for example cardiomyocytes, or their progenitors. Cells may be isolated from umbilicus tissue of any mammal of interest by the techniques provided herein. Human cells are presently preferred. The cells can be grown under a wide range of conditions, including a wide variety of culture media, and environmental conditions. The cells can be grown at least from about 35° C. to about 39° C., and possibly a wider range depending on other conditions. The cells can be grown in chemically-defined media, or in medium with added mammalian serum, for example fetal bovine serum. The cells also tolerate cryopreservation at various stages. Cells can be maintained frozen, or banked at temperatures preferably at or below −80° C. for long periods. Other preferred temperatures range from about −90° C. to about −180° C. or below. Specialized electric freezers can be used, or cells can be stored in the liquid or vapor phases of nitrogen. Tissues can also be banked prior to the isolation of the cells. Preferably Good Banking Procedures, such as are known in the art, are followed.
  • The cells are capable of growth in atmospheres containing oxygen from about 5% to at least about 20% and comprise at least one of the following characteristics: the cells have the potential for at least about 40 doublings in culture; the cells preferably are adherent, thus attachment and expansion on a coated or uncoated tissue culture vessel is preferred, wherein a coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, polylysine, vitronectin, or fibronectin;
  • The cells preferably produce of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; more preferred are cells which produce each of tissue factor, vimentin, and alpha-smooth muscle actin; production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A, B, C is also preferred. The cells are also characterized in their lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ, as detected by flow cytometry; more preferable cells lack production of all of these surface markers. Also preferred are cells which express at least one of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3. Preferred cells also have expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; the skilled artisan will appreciate that the expression of a wide variety of genes is conveniently characterized on a gene array, for example on a Affymetrix GENECHIP®.
  • The cells secrete a variety of biochemically-active factors, such as growth factors, chemokines, cytokines and the like. Preferred cells secrete at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1alpha, RANTES, and TIMP1; preferred cells may alternatively be characterized in their lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1beta, 1309, MDC, and VEGF, as detected by ELISA. These and other characteristics are available to identify and characterize the cells, and distinguish the cells of the invention from others known in the art.
  • In preferred embodiments, the cell comprises two or more of the foregoing characteristics. More preferred are those cells comprising, three, four, or five or more of the characteristics. Still more preferred are those isolated postpartum cells comprising six, seven, or eight or more of the characteristics. Still more preferred presently are those cells comprising all nine of the claimed characteristics.
  • Also presently preferred are cells that produce at least two of tissue factor, vimentin, and alpha-smooth muscle actin. More preferred are those cells producing all three of the proteins tissue factor, vimentin, and alpha-smooth muscle actin.
  • The skilled artisan will appreciate that cell markers are subject to vary somewhat under vastly different growth conditions, and that generally herein described are characterizations in Growth Medium, or variations thereof. Postpartum-derived cells that produce of at least one, two, three, or four of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A, B, C are preferred. More preferred are those cells producing five, six, or seven of these cell surface markers. Still more preferred are postpartum cells that can produce all eight of the foregoing cell surface marker proteins.
  • Similarly, postpartum cells that lack production of at least one, two, three, or four of the proteins CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ, as detected by flow cytometry are presently preferred. Cells lacking production of at least five, six, seven or eight or more of these markers are also preferred. More preferred are cells which lack production of at least nine or ten of the cell surface markers. Most highly preferred are those cells lacking production of all eleven of the foregoing identifying proteins.
  • Presently preferred cells produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C, and do not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
  • Presently, it is preferred that postpartum-derived cells express at least one, two or three of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3. More preferred are those cells which express four or five, and still more preferred are cell capable of expressing all six of the foregoing genes.
  • For some embodiments, preferred are cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression for at least one of the genes corresponding to: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa. More preferred are cells that have, relative to human fibroblasts, mesenchymal stem cells, or ileac crest bone marrow cells, reduced expression of at least 5, 10, 15 or 20 genes corresponding to those listed above. Presently more preferred are cell with reduced expression of at least 25, 30, or 35 of the genes corresponding to the listed sequences. Also more preferred are those postpartum-derived cells having expression that is reduced, relative to that of a human fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, of genes corresponding to 35 or more, 40 or more, or even all of the sequences listed.
  • Secretion of certain growth factors and other cellular proteins can make cells of the invention particularly useful. Preferred postpartum-derived cells secrete at least one, two, three or four of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1. Cells which secrete more than five, six, seven or eight of the listed proteins are also useful and preferred. Cells which can secrete at least nine, ten, eleven or more of the factors are more preferred, as are cells which can secrete twelve or more, or even all thirteen of the proteins in the foregoing list.
  • While secretion of such factors is useful, cells can also be characterized by their lack of secretion of factors into the medium. Umbilicus-derived cells that lack secretion of at least one, two, three or four of TGF-beta2, ANG2, PDGFbb, MIP1beta, 1309, MDC, and VEGF, as detected by ELISA, are presently preferred for use. Cells that are characterized in their lack of secretion of five or six of the foregoing proteins are more preferred. Cells which lack secretion of all seven of the factors listed above are also preferred.
  • In another aspect of the invention, therapeutic cell cultures are provided, the cell cultures comprising the isolated cells as described above for use in treating patients in need of angiogenesis-stimulating trophic factors. Such therapeutic cell cultures are also provided for use in treating a patient in need of neural growth stimulating trophic factors.
  • Methods of deriving UDCs from human umbilical tissue are provided. The cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes. The method comprises (a) obtaining human umbilical tissue; (b) removing substantially all of blood to yield a substantially blood-free umbilical tissue, (c) dissociating the tissue by mechanical or enzymatic treatment, or both, (d) resuspending the tissue in a culture medium, and (e) providing growth conditions which allow for the growth of a human umbilicus-derived cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes.
  • Tissue can be obtained from any completed pregnancy, term or less than term, whether delivered vaginally, or through other routes, for example surgical Cesarean section. Obtaining tissue from tissue banks is also considered within the scope of the present invention.
  • The tissue is rendered substantially free of blood by any means known in the art. For example, the blood can be physically removed by washing, rinsing, and diluting and the like, before or after bulk blood removal for example by suctioning or draining. Other means of obtaining a tissue substantially free of blood cells might include enzymatic or chemical treatment.
  • Dissociation of the umbilical tissues can be accomplished by any of the various techniques known in the art, including by mechanical disruption, for example, tissue can be aseptically cut with scissors, or a scalpel, or such tissue can be otherwise minced, blended, ground, or homogenized in any manner that is compatible with recovering intact or viable cells from human tissue.
  • In a presently preferred embodiment, the isolation procedure also utilizes an enzymatic digestion process. Many enzymes are known in the art to be useful for the isolation of individual cells from complex tissue matrices to facilitate growth in culture. As discussed above, a broad range of digestive enzymes for use in cell isolation from tissue is available to the skilled artisan. Ranging from weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin), such enzymes are available commercially. A nonexhaustive list of enzymes compatible herewith includes mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and deoxyribonucleases. Presently preferred are enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities. For example, collagenases are known to be useful for isolating various cells from tissues. Deoxyribonucleases can digest single-stranded DNA and can minimize cell-clumping during isolation. Enzymes can be used alone or in combination. Serine protease are preferably used in a sequence following the use of other enzymes as they may degrade the other enzymes being used. The temperature and time of contact with serine proteases must be monitored. Serine proteases may be inhibited with alpha 2 microglobulin in serum and therefore the medium used for digestion is preferably serum-free. EDTA and DNase are commonly used and may improve yields or efficiencies. Preferred methods involve enzymatic treatment with for example collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral protease dispase are used in the dissociating step. More preferred are those methods which employ digestion in the presence of at least one collagenase from Clostridium histolyticum, and either of the protease activities, dispase and thermolysin. Still more preferred are methods employing digestion with both collagenase and dispase enzyme activities. Also preferred are methods which include digestion with a hyaluronidase activity in addition to collagenase and dispase activities. The skilled artisan will appreciate that many such enzyme treatments are known in the art for isolating cells from various tissue sources. For example, the LIBERASE Blendzyme (Roche) series of enzyme combinations are very useful and may be used in the instant methods. Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources. The skilled artisan is also well-equipped to assess new, or additional enzymes or enzyme combinations for their utility in isolating the cells of the invention. Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer. In other preferred embodiments, the tissue is incubated at 37° C. during the enzyme treatment of the dissociation step. Diluting the digest may also improve yields of cells as cells may be trapped within a viscous digest.
  • While the use of enzyme activities is presently preferred, it is not required for isolation methods as provided herein. Methods based on mechanical separation alone may be successful in isolating the instant cells from the umbilicus as discussed above.
  • The cells can be resuspended after the tissue is dissociated into any culture medium as discussed herein above. Cells may be resuspended following a centrifugation step to separate out the cells from tissue or other debris. Resuspension may involve mechanical methods of resuspending, or simply the addition of culture medium to the cells.
  • Providing the growth conditions allows for a wide range of options as to culture medium, supplements, atmospheric conditions, and relative humidity for the cells. A preferred temperature is 37° C., however the temperature may range from about 35° C. to 39° C. depending on the other culture conditions and desired use of the cells or culture.
  • Presently preferred are methods which provide cells which require no exogenous growth factors, except as are available in the supplemental serum provided with the Growth Medium. Also provided herein are methods of deriving umbilical cells capable of expansion in the absence of particular growth factors. The methods are similar to the method above, however they require that the particular growth factors (for which the cells have no requirement) be absent in the culture medium in which the cells are ultimately resuspended and grown in. In this sense, the method is selective for those cells capable of division in the absence of the particular growth factors. Preferred cells in some embodiments are capable of growth and expansion in chemically-defined growth media with no serum added. In such cases, the cells may require certain growth factors, which can be added to the medium to support and sustain the cells. Presently preferred factors to be added for growth on serum-free media include one or more of FGF, EGF, IGF, and PDGF. In more preferred embodiments, two, three or all four of the factors are add to serum free or chemically defined media. In other embodiments, LIF is added to serum-free medium to support or improve growth of the cells.
  • Also provided are methods wherein the cells can expand in the presence of from about 5% to about 20% oxygen in their atmosphere. Methods to obtain cells that require L-valine require that cells be cultured in the presence of L-valine. After a cell is obtained, its need for L-valine can be tested and confirmed by growing on D-valine containing medium that lacks the L-isomer.
  • Methods are provided wherein the cells can undergo at least 25, 30, 35, or 40 doublings prior to reaching a senescent state. Methods for deriving cells capable of doubling to reach 1014cells or more are provided. Preferred are those methods which derive cells that can double sufficiently to produce at least about 1014, 1015, 1016, or 1017 or more cells when seeded at from about 103 to about 106 cells/cm2 in culture. Preferably these cell numbers are produced within 80, 70, or 60 days or less.
  • Isolated human umbilicus-derived cells derived by the above method are also provided herein. The cells maintain a consistent normal karyotype notwithstanding repeated passaging in certain embodiments. Also provided are cultures of human umbilicus-derived cells derived by the above method, wherein the cultures are free of maternal cells. Therapeutic cultures are provided which are obtained by the methods of the invention.
  • There are many uses for the cells of the instant invention. For example, because of the trophic factors secreted by the cells, they may be useful for the growth, maintenance, support or the like, of other useful cells, either directly or indirectly. For example, the cells may be used to make conditioned media, or may be cocultured with another cells of interest, including direct or indirect ex vivo coculture of cells intended to be used therapeutically in an autologous recipient. For direct coculture, the cells may be mixed together, while for indirect coculture, the cells may be cultured in compartments separated by semi-permeable membranes which exclude cell migration. For example, polycarbonate, polyester (PET), and collagen-coated polytetrafluoroethylene (PTFE) membranes of about less 3 microns nominal pore size exclude cell migration and are useful for such purposes. Such membranes for culture systems are able to exchange, for example, soluble growth factors between the umbilical-derived cells and another cell type. Examples are known in the art and some are available commercially (e.g. TRANSWELL culture membranes (Corning Inc, Corning N.Y.)).
  • Also provided herein are therapeutic compositions comprising an umbilicus-derived cell and another therapeutic agent or factor. Such factors include, but are not limited to, IGF, LIF, PDGF, EGF, FGF, as well as antithrombogenic and antiapoptotic agents. Such therapeutic compositions can further comprise one or more additional cell types.
  • In addition to the above, compositions derived from the cells themselves are provided herein. Cell lysates, soluble cell fractions and membrane-enriched cell fractions are all provided herein. Cell lysates include lysates in which substantially all, and more preferably all of the cells have been lysed, for example by mechanical shear, enzymatic treatment, detergent or other chemical treatment, and the like, or combinations thereof. The resulting lysates include all of the components of the cell and have many utilities, for example in supporting or maintaining growth of the cells of the invention or other cells of interest. In addition the cell lysates can serve as starting material for the purification or enrichment of desirable cell products. In preferred embodiments, cell lysates are further separated into at least a soluble cell fraction and a membrane-enriched cell fraction. Each may be useful for specific purposes. For example, where in vivo uses are contemplated, for example administration by injection, soluble cell fractions may be particularly useful in minimizing stimulation of allogeneic PBMCs. More particularly, soluble cell fractions preferably lack the required antigens to stimulate adverse immunological reactions, for example stimulation of allogeneic lymphocytes, allogeneic CD4+ T cells, or even naïve CD4+ T cells. Extracellular matrices derived from the cells, for example comprising basement membranes are also useful and are provided herein. Such extracellular material is useful for in vitro assays, for example, angiogenesis assays. They also may be useful in vivo as part of a therapeutic regimen. For example, extracellular matrix compounds are known for use in augmentation and repair such as applications relating to healing of acute and chronic wounds, or in cosmetic and reconstructive surgery. The extracellular matrix material is often useful where collagen-like properties are desirable. Extracellular matrix material is particularly useful in application requiring elasticity or viscosity because the extracellular matrix appears to be able to provide these attributes. Another application of extracellular matrix is in combination with an implantable therapeutic device. Techniques for cellular disruption range from gentle to harsh, the skilled artisan is well-equipped to select the technique for cell disruption based on the end-use of the cell lysate so obtained.
  • Compositions of the invention also include conditioned culture media as provided herein. Such media have first been used to grow the cells or cultures of the invention, which during growth secrete one or more useful products into the medium. Conditioned medium from these novel cells are useful for many purposes, including for example, supporting the growth of other mammalian cells in need of growth factors or trophic factors secreted into the media by the cells and cultures of the invention, and promoting, for example, angiogenesis. Conditioned media are useful for the secreted growth factors they contain, and preferably the conditioned media of the instant invention contain one or more growth factors useful for supporting the growth of another mammalian cell in need of such growth factors. The use of conditioned media is well understood by those of skill in the art. Continuous culture systems for generating conditioned culture medium are contemplated herein, and the cells of the invention are useful for large scale or continuous production of secreted cellular products such as growth factors.
  • Preferably, the conditioned media of the invention comprise one or more of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1. More preferred are those conditioned media which comprise several, (e.g., at least two, three or four) of the foregoing. Still more preferred are those which contain many (e.g., at least five, six, seven eight or more) of the growth factors listed. Also preferred are conditioned media which comprise additional secreted molecules of interest.
  • Another embodiment provides a mammalian cell culture comprising the conditioned medium and a mammalian cell in need of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1. Culturing in conditioned media provides for indirect methods of supporting, for example, the growth or maintenance of other cells. For example, such indirect methods provide ways of expanding populations of mammalian cells, which due to their growth factor requirements, are difficult to expand in culture in the absence of the conditioned medium.
  • Another aspect of the invention provides co-cultures comprising the umbilicus-derived cells and another mammalian cell of any phenotype. These co-cultures can comprise cells of any phenotype and may, for example, provide a way of expanding populations of mammalian cells, which due to their growth factor requirements are difficult to expand. This is a way of using the properties of the umbilicus-derived cells directly to support the growth of other cells. Such co-cultures are also of use in vivo for various applications, particularly where the use of such cells does not generate an undesired immunological response. The presence of the umbilicus-derived cells can encourage establishment and growth of cells, for example into implanted scaffolds, or at surgical repair sites. In preferred embodiments, the co-cultures comprise a human cell line in addition to the umbilicus-derived cell. Still more preferred would be to use the cells in a family member or person closely related to the donor. More preferred yet would be the co-cultures for use in the individual from whom the cells were derived.
  • In other aspects of the invention, uses of the cultures are provided. For example, the cultures of umbilicus-derived cells are useful in promoting angiogenesis. They are also useful in the treatment of soft tissue disease or injury. In preferred embodiments, the therapeutic cell cultures are grown on a scaffold or matrix. The growth on the scaffold or matrix or similar substrate can be in vivo, in vitro, or a combination of these. For in vivo application, the substrate can be surgically applied, for example as a repair to damage soft tissue. The substrate can be seeded with UDCs, or pretreated with UDC-conditioned medium, or cell lysates or soluble cell extracts, or other cell fraction, including an extracellular matrix, prior to surgical use. For in vitro applications, the cells can help other cells to establish on or within the substrate or its interstices. For example, skin or other soft tissue of dermal or subdermal layers can be grown in vitro on such substrates for later use. For combination uses, a population of cells can be established in vitro on the substrate or scaffold and then surgically attached, grafted, or implanted for further growth. Preestablishing a cell population, particularly one that secretes angiogenic factors may lead to much faster healing. Presently preferred for such scaffolds are nonpermanent, bioabsorbable polymeric matrices and the like.
  • A variety of scaffolds or matrices are known in the arts of tissue engineering and wound healing for example, and are useful with the cells and methods herein. Examples include, but are not limited to mats, foams, or self assembling peptides. Preferred mats comprise nonwoven fibers. Nonwoven mats may, for example, be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the trade name VICRYL (Ethicon, Inc., Somerville, N.J.). Foams preferred for use herein include porous foams such as those comprising, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, as discussed in U.S. Pat. No. 6,355,699. Self assembling peptides and hydrogels formed therefrom are known in the art, and include RAD 16 hydrogel, exemplified herein. Such materials are frequently used as supports for growth of cells or tissue.
  • Also provided herein are additional uses for the therapeutic cell cultures of the invention that include, but are not limited to, use in the treatment of bone disease or injury, the treatment of pancreatic disease or injury, the treatment of kidney disease or injury, the treatment of neural disease or injury, cardiac disease or injury, and the treatment of hepatic disease or injury.
  • In another of its aspects, the invention provides higher order structures such as implantable tissue structures comprising the cells of the invention, implantable human tissue matrix comprising the cells, human tissues comprising the cells, and human organs comprising these cells.
  • The invention also provides, in another of its several aspects, injectable therapeutic cells comprising isolated human umbilicus-derived cells capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes, wherein the cells do not substantially stimulate allogeneic lymphocytes in a mixed lymphocyte reaction, and produce PD-L2, but not HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2. This injectable therapeutic cell composition is useful as a therapeutic treatment in any application where undifferentiated cells can be recruited to a site through the circulation. In preferred embodiments, the injectable cell is treated to inactivate tissue factor. Such treatment includes any treatment capable of inactivating tissue factor without destroying the integrity of the cell. More preferred are treatments which have no effect on viability, doubling time or expansion. The injectable cell is treated with an anti-tissue factor antibody in one presently preferred embodiment.
  • In conjunction with therapeutic cells, other biologically active molecules, such as antithrombogenic agents, anti-apoptotic agents, and anti-inflammatory agents may be useful and may be administered in sequence with, or coadministered with the cells, individually or in combinations or two or more such compounds or agents. For example, anti-apoptotic agents may be useful to minimize programmed cell death. Such agents include but are not limited to EPO, EPO derivatives and analogs, and their salts, TPO, IGF-I, IGF-II, hepatocyte growth factor (HGF), and caspase inhibitors. Anti-inflammatory agents include but are not limited to P38 MAP kinase inhibitors, statins, IL-6 and IL-1 inhibitors, Pemirolast, Tranilast, Remicade, Sirolimus, nonsteroidal anti-inflammatory compounds, for example, Tepoxalin, Tolmetin, and Suprofen.
  • Other bioactive factors or therapeutic agents which can be coadministered with the therapeutic cells of the invention include, for example, antithrombogenic factors, immunosuppressive or immunomodulatory agents, and antioxidants. Examples of immunosuppressive and immunomodulatory agents include calcineurin inhibitors, for example cyclosporine, Tacrolimus, mTOR inhibitors such as Sirolimus or Everolimus; anti-proliferatives such as azathioprine and mycophenolate mofetil; corticosteroids for example prednisolone or hydrocortisone; antibodies such as monoclonal anti-IL-2Rα receptor antibodies, Basiliximab, Daclizumab; polyclonal anti-T-cell antibodies such as anti-thymocyte globulin (ATG), anti-lymphocyte globulin (ALG), and the monoclonal anti-T cell antibody OKT3. Antithrombogenic compounds which can be therapeutically provided in conjunction with the cells of the invention include, for example, heparin, heparin derivatives, urokinase, and PPack (dextrophenylalanine proline arginine chloromethylketone); antithrombin compounds, platelet receptor antagonists, anti-thrombin antibodies, anti-platelet receptor antibodies, aspirin, dipyridamole, protamine, hirudin, prostaglandin inhibitors, and platelet inhibitors. Antioxidants are well known in the art of course and any pharmaceutically acceptable antioxidant may be administered in conjunction with the cells of the invention including probucol; vitamins A, C, and E, coenzyme Q-10, glutathione, L cysteine, N-acetylcysteine, or antioxidant derivative, analogs or salts of the foregoing.
  • In another aspect of the invention, kits for the growth and maintenance, the isolation and the use of the umbilical-derived cells are provided. The cells, cell lysates, soluble cell fractions, membrane fractions and matrices can conveniently be employed as parts of kits, for example, for a kit for culture or implantation. The invention provides a kit including the UDCs and additional components, including instructions for growth or maintenance, isolation, or use of the cells or cell fractions, together with for example, matrix (e.g., a scaffold) material, hydrating agents (e.g., physiologically-compatible saline solutions, prepared cell culture media), cell culture substrates (e.g., culture dishes, plates, vials, etc.), cell culture media (whether in liquid or dehydrated form), antibiotic compounds, hormones, and the like. Kits for growth can for example include all of the components of the Growth Medium as used herein, including serum, for example fetal bovine serum. While the kit can include any such components, preferably it includes all ingredients necessary for its intended use. If desired, the kit also can include cells (typically cryopreserved), which can be seeded into the lattice as described herein. Kits for isolation will contain everything required to practice the isolation methods as provided herein, except for the umbilicus tissue which should be obtained fresh or frozen from a tissue bank at the time of isolation. The surgical equipment for dissociating the tissue, preferred enzymes, or choices of enzymes in stable form are provided, as are the buffers and medium, cell strainers and the like, as required or preferred for the method as disclosed above. Detailed instructions with optional steps and lists of suppliers of optional or alternative materials are also conveniently provided. Control cells can be included for comparison of the cells isolated to, for example the UDC cultures deposited with the ATCC. Kits for utilizing the umbilicus-derived cells preferably contain populations of the cells, or therapeutic compositions comprising the cells, components and products, or fractions or conditioned media derived from the cells as described above. In some embodiments, the kits may include one or more cell populations, including at least UDCs and a pharmaceutically acceptable carrier (liquid, semi-solid or solid). The populations in some embodiments are homogenous or even clonal cell lines of UDCs. In other embodiments, the kits include other cell lines for use in coculture. Therapeutic application kits preferably include additional bioactive agents as desired for example anithrombogenic agents, anti-inflammatory agents, antiapoptotic agents, and immunosuppressive or immunomodulatory compounds. The kits also optionally may include a means of administering the cells, for example by injection. The kits further may include instructions for use of the cells. Kits prepared for field hospital use, such as for military use, may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute injuries. Kits for assays and in vitro methods as described herein may contain one or more of (1) UDCs or fractions, components or products of UDCs, (2) reagents for practicing the in vitro method, (3) other cells or cell populations, as appropriate, for example for cocultures and (4) instructions for conducting the in vitro method.
  • Also provided herein are isolated postpartum-derived cells comprising a “signature gene profile.” Preferred are cells wherein the signature genes are reticulon, oxidized LDL receptor, and IL-8. In preferred embodiments, the cells comprise a profile which includes mRNA from genes for reticulon, oxidized LDL receptor, and IL-8. Most preferably, the profile comprises genes which are present independent of whether the cells are grown in medium containing serum or medium free of serum, and which further distinguish the postpartum-derived cell from other cell that are totipotent or pluripotent. In certain embodiments, the isolated postpartum-derived cells further comprise the ability to alter their expression of cell surface markers when grown in medium containing serum relative to that in serum free medium. In other embodiments of the isolated postpartum-derived cells, the markers for PDGFreceptor alpha and HLA-ABC are altered. This plasticity of the cells with respect to their cell surface markers allows the cells to be engineered in certain respects to provided full advantage of the cells desirous properties and to minimize any unwanted or deleterious properties relating to cell surface markers. This is particularly useful in engineering or preparing cells for implantation or for use in grafting where adverse immunological reactions and rejection are concerns.
  • In another of its several aspects, the invention provides a method for the preparation of therapeutic cells or cultures comprising: isolating cells; initially expanding the cells to a useful number in a serum-containing medium which supports cell expansion but in which the cells produce a quantity of the cell surface marker HLA-ABC; transferring the cells to a medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC; and passaging the cells in the medium in which the cells produce a decreased amount of the HLA-ABC, thereby preparing a therapeutic cell or culture. In certain embodiments, the medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC is a serum-free medium. This is useful for the manufacture and commercial application of the cells and therapeutic cultures particularly where required by regulatory agencies and the like. Such methods are beneficial and desirable for the production of therapeutic cells or cultures for implantation and grafting.
  • The invention provides in another aspect, serum-free media for the expansion of postpartum-derived cells. Preferred media have one or more exogenous growth factors added. Presently preferred are media wherein the one or more growth factors added are bFGF, EGF, or PDGF. Even more preferred are media wherein the one or more added growth factors includes bFGF by itself or in combination with other growth factors beneficial for the growth of the cells or for optimizing the expression of certain cell surface markers.
  • In preferred embodiments, the serum-free media of the invention provide support for the expansion for at least 5 passages of the cells with no reduction in the cells viability and no senescence. More preferred are media which support 10, 15, 20 or more passages of the cells, with the populations still capable of more passaging. Even more preferred are media which support 25, 28, 30, 33 and 35 or more passages. Most preferred are media capable of supporting the cells for about 40 or more passages.
  • Also provided in conjunction with the present invention are therapeutic cultures comprising postpartum-derived cells expanded in serum-free medium Such cells have any useful properties and are more readily suited for commercial application and regulatory approval having, for example, little risk of disease transmission related to foreign animal serum or proteins. In certain preferred embodiments, the therapeutic cultures comprise cells having a signature gene profile wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum. Such cultures may comprise a single cell type or may consist of multiple cell types including PPDCs and other cell types as additional cells present in the co- or mixed culture.
  • Also provided herein are cell culture banks comprising the postpartum-derived cells and cultures of the invention.
  • The following examples describe several aspects of embodiments of the invention in greater detail. These examples are intended to further illustrate aspects of the invention described herein. These examples should not be construed to limit the aspect so exemplified.
  • EXAMPLE 1 Isolation of Cells from Postpartum Umbilicus Tissues
  • Postpartum umbilicus tissues were obtained upon birth of either a full term or pre-term pregnancy. Cells were harvested from five separate donors of umbilicus tissue. Different methods of cell isolation were tested for their ability to yield cells with: 1) the potential to differentiate into cells with different phenotypes, a characteristic common to stem cells, or 2) the potential to provide critical trophic factors useful for other cells and tissues.
  • Methods & Materials
  • Umbilical cell isolation. Umbilical cords were obtained from National Disease Research Interchange (NDRI, Philadelphia, Pa.). The tissues were obtained following normal deliveries. The cell isolation protocols were performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of penicillin at 100 Units/milliliter and streptomycin at 100 milligrams/milliliter, and amphotericin B at 0.25 micrograms/milliliter (Invitrogen Carlsbad, Calif.). The tissues were then mechanically dissociated in 150 cm2 tissue culture plates in the presence of 50 milliliters of medium (DMEM-Low glucose or DMEM-High glucose; Invitrogen), until the tissue was minced into a fine pulp. The chopped tissues were transferred to 50 milliliter conical tubes (approximately 5 grams of tissue per tube).
  • The tissue was then digested in either DMEM-Low glucose medium or DMEM-High glucose medium, each containing 100 Units/milliliter, streptomycin at 100 milligrams/milliliter, and amphotericin B at 0.25 micrograms/milliliter and the digestion enzymes. In some experiments an enzyme mixture of collagenase and dispase was used (“C:D”) (collagenase (Sigma, St Louis, Mo.), 500 Units/milliliter; and dispase (Invitrogen), 50 Units/milliliter, in DMEM-Low glucose medium). In other experiments a mixture of collagenase, dispase and hyaluronidase (“C:D:H”) was used (C:D:H=collagenase, 500 Units/milliliter; dispase, 50 Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter, in DMEM-Low glucose). The conical tubes containing the tissue, medium and digestion enzymes were incubated at 37° C. in an orbital shaker (Environ, Brooklyn, N.Y.) at 225 rpm for 2 hrs.
  • After digestion, the tissues were centrifuged at 150×g for 5 minutes, the supernatant was aspirated. The pellet was resuspended in 20 milliliters of Growth Medium (DMEM:Low glucose (Invitrogen), 15 percent (v/v) fetal bovine serum (FBS; defined fetal bovine serum; Lot #AND18475; Hyclone, Logan, Utah), 0.001% (v/v) 2-mercaptoethanol (Sigma), penicillin at 100 Units per milliliter, streptomycin at 100 micrograms per milliliter, and amphotericin B at 0.25 micrograms per milliliter; (each from Invitrogen, Carlsbad, Calif.)). The cell suspension was filtered through a 70-micron nylon BD FALCON Cell Strainer (BD Biosciences, San Jose, Calif.). An additional 5 milliliters rinse comprising Growth Medium was passed through the strainer. The cell suspension was then passed through a 40-micrometer nylon cell strainer (BD Biosciences, San Jose, Calif.) and chased with a rinse of an additional 5 milliliters of Growth Medium.
  • The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150×g for 5 minutes. The supernatant was aspirated and the cells were resuspended in 50 milliliters of fresh growth medium. This process was repeated twice more.
  • After the final centrifugation, supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh growth medium. The number of viable cells was determined using Trypan blue staining. Cells were then cultured under standard conditions.
  • The cells isolated from umbilical cord tissues were seeded at 5,000 cells/cm2 onto gelatin-coated T-75 flasks (Corning Inc., Corning, N.Y.) in Growth Medium. After two days, spent medium and unadhered cells were aspirated from the flasks. Adherent cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with Growth Medium and allowed to grow to confluence (about 10 days from passage 0) to passage 1. On subsequent passages (from passage 1 to 2 etc), cells reached sub-confluence (75-85 percent confluence) in 4-5 days. For these subsequent passages, cells were seeded at 5,000 cells/cm2. Cells were grown in a humidified incubator with 5 percent carbon dioxide at 37° C.
  • Cell Isolation using LIBERASE Blendzymes. Cells were isolated from postpartum tissues in DMEM-Low glucose medium with LIBERASE (2.5 milligrams per milliliter, Blendzyme 3; Roche Applied Sciences, Indianapolis, Ind.) and hyaluronidase (5 Units/milliliter, Sigma). Digestion of the tissue and isolation of the cells was as described for other protease digestions above, however, the LIBERASE/hyaluronidase mixture was used instead of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE resulted in the isolation of cell populations from postpartum tissues that expanded readily.
  • Cell isolation using other enzyme combinations. Procedures were compared for isolating cells from the umbilical cord using differing enzyme combinations. Enzymes compared for digestion included: i) collagenase; ii) dispase; iii) hyaluronidase; iv) collagenase:dispase mixture (C:D); v) collagenase:hyaluronidase mixture (C:H); vi) dispase:hyaluronidase mixture (D:H); and vii) collagenase:dispase:hyaluronidase mixture (C:D:H). Differences in cell isolation utilizing these different enzyme digestion conditions were observed (Table 1-1).
  • Isolation of cells from residual blood in the cords. Other attempts were made to isolate pools of cells from umbilical cord by different approaches. In one instance umbilical cord was sliced and washed with growth medium to dislodge the blood clots and gelatinous material. The mixture of blood, gelatinous material and growth medium was collected and centrifuged at 150×g. The pellet was resuspended and seeded onto gelatin coated flasks in growth medium. From these experiments a cell population was isolated that readily expanded.
  • Isolation of cells from cord blood. Cells have also been isolated from cord blood samples attained from NDRI. The isolation protocol used was that of International Patent Application US0229971 by Ho et al. Samples (50 milliliter and 10.5 milliliters, respectively) of umbilical cord blood (NDRI, Philadelphia Pa.) were mixed with lysis buffer (filter-sterilized 155 millimolar ammonium chloride, 10 millimolar potassium bicarbonate, 0.1 millimolar EDTA buffered to pH 7.2 (all components from Sigma, St. Louis, Mo.)). Cells were lysed at a ratio of 1:20 cord blood to lysis buffer. The resulting cell suspension was vortexed for 5 seconds, and incubated for 2 minutes at ambient temperature. The lysate was centrifuged (10 minutes at 200×g). The cell pellet was resuspended in Complete Minimal Essential Medium (Gibco, Carlsbad Calif.) containing 10 percent fetal bovine serum (Hyclone, Logan Utah), 4 millimolar glutamine (Mediatech Herndon, Va.), penicillin at 100 Units per milliliter and streptomycin at 100 micrograms per milliliter (Gibco, Carlsbad, Calif.). The resuspended cells were centrifuged (10 minutes at 200×g), the supernatant was aspirated, and the cell pellet was washed in complete medium. Cells were seeded directly into either T75 flasks (Corning, N.Y.), T75 laminin-coated flasks, or T175 fibronectin-coated flasks (both Becton Dickinson, Bedford, Mass.).
  • Isolation of cells using different enzyme combinations and growth conditions. To determine whether cell populations could be isolated under different conditions and expanded under a variety of conditions immediately after isolation, cells were digested in Growth Medium with or without 0.001 percent (v/v) 2-mercaptoethanol (Sigma, St. Louis, Mo.), using the enzyme combination of C:D:H, according to the procedures provided above. All cells were grown in the presence of penicillin at 100 Units per milliliter and streptomycin at 100 micrograms per milliliter. Under all tested conditions cells attached and expanded well between passage 0 and 1 (Table 1-2). Cells in conditions 5-8 and 13-16 were demonstrated to proliferate well up to 4 passages after seeding at which point they were cryopreserved.
  • Results
  • Cell isolation using different enzyme combinations. The combination of C:D:H, provided the best cell yield following isolation, and generated cells that expanded for many more generations in culture than the other conditions (Table 1-1). An expandable cell population was not attained using collagenase or hyaluronidase alone. No attempt was made to determine if this result is specific to the collagenase that was tested.
    TABLE 1-1
    Isolation of cells from umbilical cord tissue using varying enzyme
    combinations
    Enzyme Digest Cells Isolated Cell Expansion
    Collagenase X X
    Dispase + (>10 h) +
    Hyaluronidase X X
    Collagenase:Dispase ++ (<3 h) ++
    Collagenase:Hyaluronidase ++ (<3 h) +
    Dispase:Hyaluronidase + (>10 h) +
    Collagenase:Dispase:Hyaluronidase +++ (<3 h) +++

    Key:

    + = good,

    ++ = very good,

    +++ = excellent,

    X = no success
  • Isolation of cells using different enzyme combinations and growth conditions. Cells attached and expanded well between passage 0 and 1 under all conditions tested for enzyme digestion and growth (Table 1-2). Cells in experimental conditions 5-8 and 13-16 proliferated well up to 4 passages after seeding, at which point they were cryopreserved. All cells were cryopreserved for further analysis.
    TABLE 1-2
    Isolation and culture expansion of postpartum cells under varying
    conditions:
    Condition Medium 15% FBS BME Gelatin 20% O2 Growth Factors
    1 DMEM-Lg Y Y Y Y N
    2 DMEM-Lg Y Y Y N (5%) N
    3 DMEM-Lg Y Y N Y N
    4 DMEM-Lg Y Y N N (5%) N
    5 DMEM-Lg N (2%) Y N (Laminin) Y EGF/FGF (20 ng/ml)
    6 DMEM-Lg N (2%) Y N (Laminin) N (5%) EGF/FGF (20 ng/ml)
    7 DMEM-Lg N (2%) Y N Y PDGF/VEGF
    (Fibronectin)
    8 DMEM-Lg N (2%) Y N N (5%) PDGF/VEGF
    (Fibronectin)
    9 DMEM-Lg Y N Y Y N
    10 DMEM-Lg Y N Y N (5%) N
    11 DMEM-Lg Y N N Y N
    12 DMEM-Lg Y N N N (5%) N
    13 DMEM-Lg N (2%) N N (Laminin) Y EGF/FGF (20 ng/ml)
    14 DMEM-Lg N (2%) N N (Laminin) N (5%) EGF/FGF (20 ng/ml)
    15 DMEM-Lg N (2%) N N Y PDGF/VEGF
    (Fibronectin)
    16 DMEM-Lg N (2%) N N N (5%) PDGF/VEGF
    (Fibronectin)
  • Isolation of cells from residual blood in the cords. Nucleated cells attached and grew rapidly. These cells were analyzed by flow cytometry and were similar to cells obtained by enzyme digestion.
  • Isolation of cells from cord blood. The preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.
  • Summary. Populations of cells could be isolated from umbilical tissue efficiently using the enzyme combination collagenase (a metalloprotease), dispase (neutral protease) and hyaluronidase (mucolytic enzyme which breaks down hyaluronic acid). LIBERASE, which is a blend of collagenase and a neutral protease, may also be used. Blendzyme 3, which is collagenase (4 Wunsch units/gram) and thermolysin (1714 casein Units/gram), was also used together with hyaluronidase to isolate cells. These cells expanded readily over many passages when cultured in growth expansion medium on gelatin coated plastic.
  • Cells were also isolated from residual blood in the cords, but not cord blood. The presence of cells in blood clots washed from the tissue, which adhere and grow under the conditions used, may be due to cells being released during the dissection process.
  • Reference
    • 1. Ho, Tony, W., et al., WO2003025149 A2 “CELL POPULATIONS WHICH CO-EXPRESS CD49C AND CD90” NEURONYX, INC. Application No. US0229971 US, Filed 20020920, A2 Published 20030327, A3 Published 20031218
    EXAMPLE 2 Growth Characteristics of Umbilicus-Derived Cells
  • The cell expansion potential of umbilicus-derived cells was compared to other populations of isolated stem cells. The process of cell expansion to senescence is referred to as Hayflick's limit (Hayflick L. The longevity of cultured human cells. J. Am. Geriatr. Soc. 22(1):1-12, 1974; Hayflick L. The strategy of senescence. Gerontologist 14(1):37-45), 1974).
  • Materials and Methods
  • Gelatin-coating flasks. Tissue culture plastic flasks were coated by adding 20 milliliters 2% (w/v) gelatin (Type B: 225 Bloom; Sigma, St Louis, Mo.) to a T75 flask (Corning Inc., Corning, N.Y.) for 20 minutes at room temperature. After removing the gelatin solution, 10 milliliters phosphate-buffered saline (PBS) (Invitrogen, Carlsbad, Calif.) were added and then aspirated.
  • Comparison of expansion potential of umbilicus-derived cells with other cell populations. For comparison of growth expansion potential the following cell populations were utilized; i) Mesenchymal stem cells (MSC; Cambrex, Walkersville, Md.); ii) Adipose-derived cells (U.S. Pat. No. 6,555,374 B1; U.S. Patent Application US20040058412); iii) Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, Md.); and iv) Umbilicus-derived cells. Cells were initially seeded at 5,000 cells/cm2 on gelatin-coated T75 flasks in Growth Medium. For subsequent passages, cell cultures were treated as follows. After trypsinization, viable cells were counted after Trypan blue staining. Cell suspension (50 microliters) was combined with Trypan blue (50 microliters, Sigma, St. Louis Mo.). Viable cell numbers were estimated using a hemocytometer.
  • Following counting, cells were seeded at 5,000 cells/cm2 onto gelatin-coated T 75 flasks in 25 milliliters of fresh Growth Medium. Cells were grown in a standard atmosphere (5 percent carbon dioxide (v/v)) at 37° C. The Growth Medium was changed twice per week. When cells reached about 85 percent confluence they were passaged; this process was repeated until the cells reached senescence.
  • At each passage, cells were trypsinized and counted. The viable cell yield, population doublings [ln (cells final/cells initial)/ln2], and doubling time (time in culture/population doubling) were calculated. For the purposes of determining optimal cell expansion, the total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e. expansion factor=cells final/cells initial).
  • Expansion potential of cell banks at low density. The expansion potential of cells banked at passage 10 was also tested. A different set of conditions was used. Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, Md.), umbilicus-derived cells, and placenta-derived cells were tested. These cell populations had been banked at passage 10 previously, having been cultured at 5,000 cells/cm2 at each passage to that point. The effect of cell density on the cell populations following cell thaw at passage 10 was determined. Cells were thawed under standard conditions, counted using Trypan blue staining. Thawed cells were then seeded at 1,000 cells/cm2 in Growth Medium. Cells were grown under standard atmospheric conditions at 37° C. Growth Medium was changed twice a week. Cells were passaged as they reached about 85% confluence. Cells were subsequently passaged until senescence, i.e., until they could not be expanded any further. Cells were trypsinized and counted at each passage. The cell yield, population doubling (ln (cells final/cells initial)/ln2) and doubling time (time in culture)/population doubling). The total cell yield per passage was determined by multiplying total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor=cells final/cells initial).
  • Expansion of umbilicus-derived cells at low density from initial cell seeding. The expansion potential of freshly isolated umbilicus-derived cell cultures under low cell seeding conditions was tested in another experiment. Umbilicus-derived cells were isolated as described in a previous example. Cells were seeded at 1,000 cells/cm2 and passaged as described above until senescence. Cells were grown under standard atmospheric conditions at 37° C. Growth Medium was changed twice per week. Cells were passaged as they reached about 85% confluence. At each passage, cells were trypsinized and counted by Trypan blue staining. The cell yield, population doubling (ln(cell final/cell initial)/ln 2) and doubling time (time in culture/population doubling) were calculated for each passage. The total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e. expansion factor=cell final/cell initial). Cells were grown on gelatin and non-gelatin coated flasks.
  • Expansion of cells in low oxygen culture conditions. It has been demonstrated that low O2 cell culture conditions can improve cell expansion in certain circumstances (Csete, Marie; Doyle, John; Wold, Barbara J.; McKay, Ron; Studer, Lorenz. Low oxygen culturing of central nervous system progenitor cells. US20040005704). In order to determine if cell expansion of umbilicus-derived cells could be improved by altering cell culture conditions, cultures of umbilicus-derived cells were grown in low oxygen conditions. Cells were seeded at 5,000 cells/cm2 in Growth Medium on gelatin coated flasks. Cells were initially cultured under standard atmospheric conditions through passage 5, at which point they were transferred to low oxygen (5% O2) culture conditions.
  • Other growth conditions. In other experiments cells were expanded on non-coated, collagen-coated, fibronectin-coated, laminin-coated and Matrigel-coated plates. Cultures have been demonstrated to expand well on these different matrices.
  • Results
  • Comparison of expansion potential of umbilicus-derived cells with other cell populations. Umbilicus-derived cells expanded for more than 40 passages generating cell yields of >1E17 cells in 60 days. In contrast, MSCs and fibroblasts senesced after <25 days and <60 days, respectively. Although both adipose-derived and omental cells expanded for almost 60 days they generated total cell yields of 4.5E12 and 4.24E13 respectively. Thus, when seeded at 5,000 cells/cm2 under the experimental conditions utilized, umbilicus-derived cells expanded much better than the other cell types grown under the same conditions (Table 2-1).
    TABLE 2-1
    Growth characteristics for different cell populations grown to senescence
    Total Population Yield
    Cell Type Senescence Doublings (Total Cells)
    MSC 24 d 8 4.72E7 
    Adipose- 57 d 24  4.5E12
    derived cell
    Fibroblasts 53 d 26 2.82E13
    Umbilical 65 d 42 6.15E17
  • Expansion of potential of cell banks at low density. Umbilicus-derived and fibroblast cells expanded for greater than 10 passages generating cell yields of >1E11 cells in 60 days (Table 2-2). Under these conditions both the fibroblasts and the umbilicus-derived cell populations senesced after 80 days, completing >50 and >40 population doublings respectively.
    TABLE 2-2
    Growth characteristics for different cell populations using low density
    growth expansion from passage 10 through senescence
    Cell Type Total Population Yield
    (Passage No.) Senescence Doublings (Total Cells)
    Fibroblast (P10) 80 days 43.68 2.59E11
    Umbilical (P10) 80 days 53.6 1.25E14
  • Expansion of cells in low oxygen culture conditions. Cells expanded well under the reduced oxygen conditions, however, culturing under low oxygen conditions does not appear to have a significant effect on cell expansion for postpartum-derived cells. These results are preliminary in the sense that any ultimate conclusions to be made regarding the effect of reduced oxygen would best be drawn from experiments on growing cells in low oxygen from initial isolation. Standard atmospheric conditions have already proven successful for growing sufficient numbers of cells, and low oxygen culture is not required for the growth of postpartum-derived cells.
  • Summary. The current cell expansion conditions of growing isolated umbilicus-derived cells at densities of about 5,000 cells/cm2, in Growth Medium on gelatin-coated or uncoated flasks, under standard atmospheric oxygen, are sufficient to generate large numbers of cells at passage 11. Furthermore, the data suggests that the cells can be readily expanded using lower density culture conditions (e.g. 1,000 cells/cm2). Umbilicus-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for growth. Presently, culturing umbilicus-derived cells under standard atmospheric conditions is preferred for generating large pools of cells. However, when the culture conditions are altered, umbilicus-derived cell expansion can likewise be altered. This strategy may be used to enhance the proliferative and differentiative capacity of these cell populations.
  • Under the conditions utilized, while the expansion potential of MSC and adipose-derived cells is limited, umbilicus-derived cells expand readily to large numbers.
  • References
    • 1) Hayflick L. The longevity of cultured human cells. J Am Geriatr Soc. 22:1-12, 1974.
    • 2) Hayflick L. The strategy of senescence. Gerontologist 14(1):37-45, 1974.
    • 3) United States Patent Application No. 20040058412
    • 4) United States Patent Application No. 20040048372
    • 6) Csete, Marie; Doyle, John; Wold, Barbara J.; McKay, Ron; and Studer, Lorenz. Low oxygen culturing of central nervous system progenitor cells. United States Patent Application No. 20040005704.
    EXAMPLE 3 Growth of Umbilicus-Derived Cells in Medium Containing D-Valine
  • It has been reported that medium containing D-valine instead of the normal L-valine isoform can be used to selectively inhibit the growth of fibroblast-like cells in culture (Hongpaisan, 2000; Sordillo et al., 1988). Experiments were performed to determine whether umbilicus-derived cells could grow in medium containing D-valine.
  • Methods & Materials
  • Umbilicus-derived cells (P5) and fibroblasts (P9) were seeded at 5,000 cells/cm2 in gelatin-coated T75 flasks (Corning, Corning, N.Y.). After 24 hours the medium was removed and the cells were washed with phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) to remove residual medium. The medium was replaced with a modified Growth Medium (DMEM with D-valine (special order Gibco), 15% (v/v) dialyzed fetal bovine serum (Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma), penicillin at 50 Units/milliliter and streptomycin at 50 milligrams/milliliter (Gibco)).
  • Results
  • Neither the umbilicus-derived cells nor the fibroblast cells seeded in the D-valine-containing medium proliferated, unlike cells seeded in Growth Medium containing dialyzed serum. Fibroblasts cells changed morphologically, increasing in size and changing shape. All of the cells died and eventually detached from the flask surface after four weeks. Thus, it may be concluded that umbilicus-derived cells require L-valine for cell growth and to maintain long-term viability.
  • References
    • Hongpaisan J. (2000) Inhibition of proliferation of contaminating fibroblasts by D-valine in cultures of smooth muscle cells from human myometrium. Cell Biol Int. 24:1-7.
    • Sordillo L M, Oliver S P, Akers R M. (1988) Culture of bovine mammary epithelial cells in D-valine modified medium: selective removal of contaminating fibroblasts. Cell Biol Int Rep. 12:355-64.
    EXAMPLE 4 Karyotype Analysis of Umbilicus-Derived PPDCs
  • Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type. Human cells used in cell therapy should have a normal number (46) of chromosomes with normal structure. To identify umbilicus-derived cell lines that are homogeneous and free from cells of non-umbilical tissue origin, karyotypes of cell samples were analyzed.
  • Materials and Methods
  • PPDCs from postpartum tissue of a male neonate were cultured in Growth Media. Postpartum tissue from a male neonate (X, Y) was selected to allow distinction between neonatal-derived cells and maternal derived cells (X, X). Cells were seeded at 5,000 cells per square centimeter in Growth Medium in a T25 flask (Corning, Corning, N.Y.) and expanded to 80% confluence. A T25 flask containing cells was filled to the neck with Growth Media. Samples were delivered to a clinical cytogenetics lab by courier (estimated lab to lab transport time is one hour). Chromosome analysis was performed by the Center for Human & Molecular Genetics at the New Jersey Medical School, Newark, N.J. Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two). A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells were counted and analyzed when a heterogeneous karyotype number (four) was identified.
  • Results
  • All cell samples sent for chromosome analysis were interpreted by as exhibiting a normal appearance. Each of the cell samples was characterized as homogeneous. (Table 4-1).
    TABLE 4-1
    Karyotype results of PPDCs.
    Metaphase
    cells Metaphase Number of ISCN
    Tissue Passage counted cells analyzed karyotypes Karyotype
    Umbilical 23 20 5 2 46, XX
    Umbilical 6 20 5 2 46, XY
    Umbilical 3 20 5 2 46, XX
  • Summary. Chromosome analysis identified umbilicus-derived PPDCs whose karyotypes appear normal as interpreted by a clinical cytogenetic laboratory. Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.
  • EXAMPLE 5 Flow Cytometric Evaluation of Human Umbilicus-Derived Cell Surface Markers
  • Characterization of cell surface proteins or “markers” by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions. Postpartum cell lines isolated from the umbilicus were characterized by flow cytometry, providing a profile for the identification of these cell lines.
  • Materials and Methods
  • Media and Culture Vessels. Cells were cultured in Growth Medium, in plasma-treated T75, T150, and T225 tissue culture flasks (Corning, Corning, N.Y.) until confluent. The growth surfaces of the flasks were coated with gelatin by incubating 2% (w/v) gelatin (Sigma, St. Louis, Mo.) for 20 minutes at room temperature.
  • Antibody Staining. Adherent cells in flasks were washed in phosphate buffered saline (PBS); (Gibco, Carlsbad, Mo.) and detached with Trypsin/EDTA (Gibco). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1×107 per milliliter. In accordance with the manufacture's specifications, antibody to the cell surface marker of interest (see below) was added to 100 microliters of cell suspension and the mixture was incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were resuspended in 500 microliters PBS and analyzed by flow cytometry.
  • Flow Cytometry Analysis. Flow cytometry analysis was performed with a FACScalibur instrument (Becton Dickinson, San Jose, Calif.).
  • Antibodies to Cell Surface Markers. The following antibodies to cell surface markers were used.
    TABLE 5-1
    Antibodies used in characterizing cell surface markers of UDCs.
    Antibody Manufacture Catalog Number
    CD10 BD Pharmingen 555375
    (San Diego, CA)
    CD13 BD Pharmingen 555394
    CD31 BD Pharmingen 555446
    CD34 BD Pharmingen 555821
    CD44 BD Pharmingen 555478
    CD45RA BD Pharmingen 555489
    CD73 BD Pharmingen 550257
    CD90 BD Pharmingen 555596
    CD117 BD Pharmingen 340529
    CD141 BD Pharmingen 559781
    PDGFr-alpha BD Pharmingen 556002
    HLA-A, B, C BD Pharmingen 555553
    HLA-DR, DP, DQ BD Pharmingen 555558
    IgG-FITC Sigma (St. Louis, MO) F-6522
    IgG-PE Sigma P-4685
  • Passage to Passage Comparison. Umbilicus-derived cells were analyzed at passages 8, 15, and 20.
  • Donor to Donor Comparison. To compare differences among donors, umbilical from different donors were compared to each other.
  • Surface Coating Comparison. Umbilicus-derived cells cultured on gelatin-coated flasks was compared to umbilicus cultured on uncoated flasks.
  • Results
  • Umbilicus-Derived Cell Characterization. Umbilicus-derived cells analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by the increased values of fluorescence relative to the IgG control (data not shown). These cells were negative for detectable expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values comparable to the IgG control (data not shown). Variations in florescence values of positive curves were accounted for. The mean (i.e. CD13) and range (i.e. CD90) of the positive curves showed some variation, but the curves appear normal, confirming a homogenous population. Both curves individually exhibited values greater than the IgG control.
  • Passage to Passage Comparison. Umbilical cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were within expected ranges. While the means (i.e. CD13) of the positive curves varied all curves individually exhibited values greater than the IgG control.
  • Donor to Donor Comparison. Umbilicus-derived cells isolated from separate donors analyzed by flow cytometry each showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were accounted for. While the mean (i.e. CD10) of the positive curves varied, both curves individually exhibited values greater than the IgG control.
  • The Effect of Surface Coating with Gelatin. Umbilical cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
  • Summary. Analysis of umbilicus-derived postpartum cells by flow cytometry has established a profile useful to identify these cell lines. Umbilicus-derived postpartum cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A, B, C and negative for CD31, CD34, CD45, CD117, CD141 and HLA-DR, DP, DQ. This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, and digestion enzymes used in isolation and preparation of the cells. Some variation in individual fluorescence value histogram curve means and ranges were observed, but all positive curves under all conditions tested were normal and expressed fluorescence values greater than the IgG control, thus confirming that the cells comprise a homogenous population, which has positive expression of the markers.
  • EXAMPLE 6 Analysis of Cells by Oligonucleotide Array
  • Oligonucleotide arrays were used to compare gene expression profiles of umbilicus- and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.
  • Materials and Methods
  • Isolation and Culture of Cells
  • Postpartum tissue-derived cells. Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, Pa.) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 1. Cells were cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C. with 5% CO2.
  • Fibroblasts. Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, Md.; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines were cultured in DMEM/F12 medium (Invitrogen, Carlsbad, Calif.) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen)). The cells were grown on standard tissue-treated plastic.
  • Human Mesenchymal Stem Cells (hMSC). hMSCs were purchased from Cambrex Incorporated (Walkersville, Md.; Lot numbers 2F1655, 2F1656 and 2F1657) and cultured according to the manufacturer's specifications in MSCGM Media (Cambrex). The cells were grown on standard tissue cultured plastic at 37° C. with 5% CO2.
  • Human Ileac Crest Bone Marrow Cells (ICBM). Human ileac crest bone marrow was received from NDRI with patient consent. The marrow was processed according to the method outlined by Ho, et al. (WO03/025149). The marrow was mixed with lysis buffer (155 mM NH4Cl, 10 mM KHCO3, and 0.1 mM EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500×g. The supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 mM glutamine. The cells were centrifuged again and the cell pellet was resuspended in fresh medium. The viable mononuclear cells were counted using Trypan-blue exclusion (Sigma, St. Louis, Mo.). The mononuclear cells were seeded in tissue-cultured plastic flasks at 5×104 cells/cm2. The cells were incubated at 37° C. with 5% CO2 at either standard atmospheric O2 or at 5% O2. Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture. The adherent cells were maintained in culture.
  • Isolation of mRNA and GENECHIP Analysis. Actively growing cultures of cells were removed from the flasks with a cell scraper in cold phosphate buffered saline (PBS). The cells were centrifuged for 5 minutes at 300×g. The supernatant was removed and the cells were resuspended in fresh PBS and centrifuged again. The supernatant was removed and the cell pellet was immediately frozen and stored at −80° C. Cellular mRNA was extracted and transcribed into cDNA. cDNA was then transcribed into cRNA and biotin-labeled. The biotin-labeled cRNA was hybridized with Affymetrix GENECHIP HG-U133A oligonucleotide arrays (Affymetrix, Santa Clara, Calif.). The hybridizations and data collection were performed according to the manufacturer's specifications. The hybridization and data collection was performed according to the manufacturer's specifications. Data analyses were performed using “Significance Analysis of Microarrays” (SAM) version 1.21 computer software (Tusher, V. G. et al., 2001, Proc. Natl. Acad. Sci. USA 98: 5116-5121). Licenses for the analysis software are available through the Office of Technology Licensing, Stanford University, and more information is available on the World Wide Web at Professor Tibshirani's web site in the Dep't of Statistics, Stanford University (www-stat.stanford.edu/˜tibs/SAM/).
  • Results
  • Fourteen different populations of cells were analyzed in this study. The cells along with passage information, culture substrate, and culture media are listed in Table 6-1.
    TABLE 6-1
    Cells analyzed by the microarray study. The
    cells lines are listed by their identification code along with passage
    at the time of analysis, cell growth substrate, and growth media.
    Cell Population Passage Substrate Media
    Umbilical (022803) 2 Gelatin DMEM, 15% FBS, βME
    Umbilical (042103) 3 Gelatin DMEM, 15% FBS, βME
    Umbilical (071003) 4 Gelatin DMEM, 15% FBS, βME
    Placenta (042203) 12 Gelatin DMEM, 15% FBS, βME
    Placenta (042903) 4 Gelatin DMEM, 15% FBS, βME
    Placenta (071003) 3 Gelatin DMEM, 15% FBS, βME
    ICBM (070203) (5% O2) 3 Plastic MEM 10% FBS
    ICBM (062703) (std O2) 5 Plastic MEM 10% FBS
    ICBM (062703)(5% O2) 5 Plastic MEM 10% FBS
    hMSC (Lot 2F1655) 3 Plastic MSCGM
    hMSC (Lot 2F1656) 3 Plastic MSCGM
    hMSC (Lot 2F1657) 3 Plastic MSCGM
    hFibroblast (9F0844) 9 Plastic DMEM-F12, 10% FBS
    hFibroblast (CCD39SK) 4 Plastic DMEM-F12, 10% FBS
  • The data were evaluated by Principle Component Analysis with SAM software as described above. Analysis revealed 290 genes that were expressed in different relative amounts in the cells tested. This analysis provided relative comparisons between the populations.
  • Table 6-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs. The Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types. The Euclidean distance is inversely proportional to similarity between the expression of the 290 genes.
    TABLE 6-2
    The Euclidean Distances for the Cell Pairs.
    The Euclidean distance was calculated for the cell types using
    the 290 genes that were expressed differentially between the
    cell types. Similarity between the cells is inversely
    proportional to the Euclidean distance.
    Cell Pair Euclidean Distance
    ICBM-hMSC 24.71
    Placenta-umbilical 25.52
    ICBM-Fibroblast 36.44
    Fibroblast-placenta 37.09
    Fibroblast-MSC 39.63
    ICBM-Umbilical 40.15
    Fibroblast-Umbilical 41.59
    MSC-Placenta 42.84
    MSC-Umbilical 46.86
    ICBM-placenta 48.41
  • Tables 6-3, 6-4, and 6-5 show the expression of genes increased in placenta-derived cells (Table 6-3), increased in umbilical cord-derived cells (Table 6-4), and reduced in umbilical cord and placenta-derived cells (Table 6-5).
    TABLE 6-3
    Genes which are specifically increased in expression in the placenta-derived
    cells as compared to the other cell lines assayed.
    Genes Increased in Placenta-Derived Cells
    NCBI Accession
    Probe Set ID Gene Name Number
    209732_at C-type (calcium dependent, carbohydrate-recognition domain) AF070642
    lectin, superfamily member 2 (activation-induced)
    206067_s_at Wilms tumor 1 NM_024426
    207016_s_at aldehyde dehydrogenase 1 family, member A2 AB015228
    206367_at Renin NM_000537
    210004_at oxidized low density lipoprotein (lectin-like) receptor 1 AF035776
    214993_at Homo sapiens, clone IMAGE: 4179671, mRNA, partial cds AF070642
    202178_at protein kinase C, zeta NM_002744
    209780_at hypothetical protein DKFZp564F013 AL136883
    204135_at downregulated in ovarian cancer 1 NM_014890
    213542_at Homo sapiens mRNA; cDNA DKFZp547K1113 (from clone AI246730
    DKFZp547K1113)
  • TABLE 6-4
    Genes which are specifically increased in expression in
    umbilical cord -derived cells as compared to the other cell lines assayed.
    Genes Increased in Umbilicus-Derived Cells
    NCBI
    Accession
    Probe Set ID Gene Name Number
    202859_x_at Interleukin
    8 NM_000584
    211506_s_at Interleukin
    8 AF043337
    210222_s_at reticulon
    1 BC000314
    204470_at chemokine (C—X—C motif) ligand 1 NM_001511
    (melanoma growth stimulating activity
    206336_at chemokine (C—X—C motif) ligand 6 NM_002993
    (granulocyte chemotactic protein 2)
    207850_at Chemokine (C—X—C motif) ligand 3 NM_002090
    203485_at reticulon
    1 NM_021136
    202644_s_at tumor necrosis factor, alpha-induced NM_006290
    protein
    3
  • TABLE 6-5
    Genes which were decreased in expression in the umbilical
    cord and placenta cells as compared to the other cell lines assayed.
    Genes Decreased in Umbilicus- and Placenta-Derived Cells
    Probe Set NCBI Accession
    ID Gene name Number
    210135_s_at short stature homeobox 2 AF022654.1
    205824_at heat shock 27 kDa protein 2 NM_001541.1
    209687_at chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1) U19495.1
    203666_at chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1) NM_000609.1
    212670_at elastin (supravalvular aortic stenosis, Williams-Beuren syndrome) AA479278
    213381_at Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone N91149
    DKFZp586M2022)
    206201_s_at mesenchyme homeobox 2 (growth arrest-specific homeobox) NM_005924.1
    205817_at Sine oculis homeobox homolog 1 (Drosophila) NM_005982.1
    209283_at crystallin, alpha B AF007162.1
    212793_at dishevelled associated activator of morphogenesis 2 BF513244
    213488_at DKFZP586B2420 protein AL050143.1
    209763_at similar to neuralin 1 AL049176
    205200_at Tetranectin (plasminogen binding protein) NM_003278.1
    205743_at src homology three (SH3) and cysteine rich domain NM_003149.1
    200921_s_at B-cell translocation gene 1, anti-proliferative NM_001731.1
    206932_at cholesterol 25-hydroxylase NM_003956.1
    204198_s_at runt-related transcription factor 3 AA541630
    219747_at hypothetical protein FLJ23191 NM_024574.1
    204773_at Interleukin 11 receptor, alpha NM_004512.1
    202465_at Procollagen C-endopeptidase enhancer NM_002593.2
    203706_s_at Frizzled homolog 7 (Drosophila) NM_003507.1
    212736_at hypothetical gene BC008967 BE299456
    214587_at Collagen, type VIII, alpha 1 BE877796
    201645_at Tenascin C (hexabrachion) NM_002160.1
    210239_at iroquois homeobox protein 5 U90304.1
    203903_s_at Hephaestin NM_014799.1
    205816_at integrin, beta 8 NM_002214.1
    203069_at synaptic vesicle glycoprotein 2 NM_014849.1
    213909_at Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744 AU147799
    206315_at cytokine receptor-like factor 1 NM_004750.1
    204401_at potassium intermediate/small conductance calcium-activated channel, NM_002250.1
    subfamily N, member 4
    216331_at integrin, alpha 7 AK022548.1
    209663_s_at integrin, alpha 7 AF072132.1
    213125_at DKFZP586L151 protein AW007573
    202133_at transcriptional co-activator with PDZ-binding motif (TAZ) AA081084
    206511_s_at Sine oculis homeobox homolog 2 (Drosophila) NM_016932.1
    213435_at KIAA1034 protein AB028957.1
    206115_at early growth response 3 NM_004430.1
    213707_s_at distal-less homeobox 5 NM_005221.3
    218181_s_at hypothetical protein FLJ20373 NM_017792.1
    209160_at aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid AB018580.1
    dehydrogenase, type II)
    213905_x_at Biglycan AA845258
    201261_x_at Biglycan BC002416.1
    202132_at transcriptional co-activator with PDZ-binding motif (TAZ) AA081084
    214701_s_at fibronectin 1 AJ276395.1
    213791_at Proenkephalin NM_006211.1
    205422_s_at Integrin, beta-like 1 (with EGF-like repeat domains) NM_004791.1
    214927_at Homo sapiens mRNA full length insert cDNA clone EUROIMAGE AL359052.1
    1968422
    206070_s_at EphA3 AF213459.1
    212805_at KIAA0367 protein AB002365.1
    219789_at natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic AI628360
    peptide receptor C)
    219054_at hypothetical protein FLJ14054 NM_024563.1
    213429_at Homo sapiens mRNA; cDNA DKFZp564B222 (from clone AW025579
    DKFZp564B222)
    204929_s_at vesicle-associated membrane protein 5 (myobrevin) NM_006634.1
    201843_s_at EGF-containing fibulin-like extracellular matrix protein 1 NM_004105.2
    221478_at BCL2/adenovirus E1B 19 kDa interacting protein 3-like AL132665.1
    201792_at AE binding protein 1 NM_001129.2
    204570_at cytochrome c oxidase subunit VIIa polypeptide 1 (muscle) NM_001864.1
    201621_at neuroblastoma, suppression of tumorigenicity 1 NM_005380.1
    202718_at Insulin-like growth factor binding protein 2, 36 kDa NM_000597.1
  • Tables 6-6, 6-7, and 6-8 show the expression of genes increased in human fibroblasts (Table 6-6), ICBM cells (Table 6-7), and MSCs (Table 6-8).
    TABLE 6-6
    Genes which were increased in expression in
    fibroblasts as compared to the other cell lines assayed.
    Genes increased in fibroblasts
    dual specificity phosphatase 2
    KIAA0527 protein
    Homo sapiens cDNA: FLJ23224 fis, clone ADSU02206
    dynein, cytoplasmic, intermediate polypeptide 1
    ankyrin 3, node of Ranvier (ankyrin G)
    inhibin, beta A (activin A, activin AB alpha polypeptide)
    ectonucleotide pyrophosphatase/phosphodiesterase 4 (putative function)
    KIAA1053 protein
    microtubule-associated protein 1A
    zinc finger protein 41
    HSPC019 protein
    Homo sapiens cDNA: FLJ23564 fis, clone LNG10773
    Homo sapiens mRNA; cDNA DKFZp564A072 (from clone
    DKFZp564A072)
    LIM protein (similar to rat protein kinase C-binding enigma)
    inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase
    complex-associated protein
    hypothetical protein FLJ22004
    Human (clone CTG-A4) mRNA sequence
    ESTs, Moderately similar to cytokine receptor-like factor 2; cytokine
    receptor CRL2 precursor [Homo sapiens]
    transforming growth factor, beta 2
    hypothetical protein MGC29643
    antigen identified by monoclonal antibody MRC OX-2
    putative X-linked retinopathy protein
  • TABLE 6-7
    Genes which were increased in expression in
    the ICBM-derived cells as compared to the other cell lines assayed.
    Genes Increased In ICBM Cells
    cardiac ankyrin repeat protein
    MHC class I region ORF
    integrin, alpha 10
    hypothetical protein FLJ22362
    UDP-N-acetyl-alpha-D-galactosamine:polypeptide
    N-acetylgalactosaminyltransferase 3 (GalNAc-T3)
    interferon-induced protein 44
    SRY (sex determining region Y)-box 9 (campomelic dysplasia,
    autosomal sex-reversal)
    keratin associated protein 1-1
    hippocalcin-like 1
    jagged 1 (Alagille syndrome)
    proteoglycan 1, secretory granule
  • TABLE 6-8
    Genes which were increased in expression in
    the MSC cells as compared to the other cell lines assayed.
    Genes Increased In MSC Cells
    interleukin 26
    maltase-glucoamylase (alpha-glucosidase)
    nuclear receptor subfamily 4, group A, member 2
    v-fos FBJ murine osteosarcoma viral oncogene homolog
    hypothetical protein DC42
    nuclear receptor subfamily 4, group A, member 2
    FBJ murine osteosarcoma viral oncogene homolog B
    WNT1 inducible signaling pathway protein 1
    MCF.2 cell line derived transforming sequence
    potassium channel, subfamily K, member 15
    cartilage paired-class homeoprotein 1
    Homo sapiens cDNA FLJ12232 fis, clone MAMMA1001206
    Homo sapiens cDNA FLJ34668 fis, clone LIVER2000775
    jun B proto-oncogene
    B-cell CLL/lymphoma 6 (zinc finger protein 51)
    zinc finger protein 36, C3H type, homolog (mouse)
  • Summary. The present study was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas. The study also included two different lines of dermal fibroblasts, three lines of mesenchymal stem cells, and three lines of ileac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed on a GENECHIP oligonucleotide array that contained oligonucleotide probes for 22,000 genes.
  • The analysis revealed that transcripts for 290 genes were present in different amounts in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells. Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord.
  • The expression of selected genes has been confirmed by PCR, as shown in Example 7. Postpartum-derived cells generally, and umbilical derived cells, in particular, have distinct gene expression profiles, for example, as compared to other human cells, such as the bone marrow-derived cells and fibroblasts tested here.
  • EXAMPLE 7 Cell Markers in Umbilicus-Derived Cells
  • Gene expression profiles of cells derived from the human umbilical cord were compared with those of cells derived from other sources using an Affymetrix GENECHIP. Six “signature” genes were identified: oxidized LDL receptor 1, interleukin-8 (IL-8), renin, reticulon, chemokine receptor ligand 3 (CXC ligand 3), and granulocyte chemotactic protein 2 (GCP-2). These “signature” genes were expressed at relatively high levels in umbilicus-derived cells.
  • The procedures described in this example were conducted to verify the microarray data and compare data for gene and protein expression, as well as to establish a series of reliable assays for detection of unique identifiers for umbilicus-derived cells.
  • Methods & Materials
  • Cells. Umbilicus-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) were grown in Growth Medium in gelatin-coated T75 flasks. Mesenchymal Stem Cells (MSCs) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, Md.).
  • For IL-8 experiments, cells were thawed from liquid nitrogen and plated in gelatin-coated flasks at 5,000 cells/cm2, grown for 48 hours in Growth Medium and then grown further for 8 hours in 10 milliliters of serum starvation medium [DMEM-low glucose (Gibco, Carlsbad, Calif.), penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter)(Gibco) and 0.1% (w/v) Bovine Serum Albumin (BSA; Sigma, St. Louis, Mo.)]. RNA was then extracted and the supernatants were centrifuged at 150×g for 5 minutes to remove cellular debris. Supernatants were frozen at −80° C. until ELISA analysis.
  • Cell culture for ELISA assay. Cells derived from human umbilical cord, as well as human fibroblasts derived from human neonatal foreskin, were cultured in Growth Medium in gelatin-coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen. Cells were thawed and transferred to 15 milliliter centrifuge tubes. After centrifugation at 150×g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 milliliters culture medium and counted. Cells were grown in a 75 cm2 flask containing 15 milliliters of Growth Medium at 375,000 cell/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation medium was collected at the end of incubation, centrifuged at 14,000×g for 5 minutes (and stored at −20° C.).
  • To estimate the number of cells in each flask, 2 milliliters of trypsin/EDTA (Gibco, Carlsbad, Calif.) were added each flask. After cells detached from the flask, trypsin activity was neutralized with 8 milliliters of Growth Medium. Cells were transferred to a 15 milliliter centrifuge tube and centrifuged at 150×g for 5 minutes. Supernatant was removed and 1 milliliter Growth Medium was added to each tube to resuspend the cells. Cell number was determined with a hemocytometer.
  • ELISA assay. The amount of IL-8 secreted by the cells into serum starvation medium was analyzed using ELISA assays (R&D Systems, Minneapolis, Minn.). All assays were conducted according to the instructions provided by the manufacturer.
  • Total RNA isolation. RNA was extracted from confluent umbilicus-derived cells and fibroblasts, or for IL-8 expression, from cells treated as described above. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.). RNA was extracted according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.) and subjected to DNase treatment (2.7 Units/sample) (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at −80° C. RNA was also extracted from human umbilical cord. Tissue (30 milligrams) was suspended in 700 microliters of buffer RLT containing beta-mercaptoethanol. Samples were mechanically homogenized and the RNA extraction proceeded according to manufacturer's specification. RNA was extracted with 50 microliters of DEPC-treated water and stored at −80° C.
  • Reverse transcription. RNA was reverse-transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C. for 10 minutes, 37° C. for 60 minutes, and 95° C. for 10 minutes. Samples were stored at −20° C.
  • Genes identified by cDNA microarray as uniquely regulated in postpartum cells (signature genes—including oxidized LDL receptor, interleukin-8, renin, and reticulon), were further investigated using real-time and conventional PCR.
  • Real-time PCR. PCR was performed on cDNA samples using gene expression products sold under the trade name ASSAYS-ON-DEMAND (Applied Biosystems) gene expression products. Oxidized LDL receptor (Hs00234028); renin (Hs00166915); reticulon (Hs00382515); CXC ligand 3 (Hs00171061); GCP-2 (Hs00605742); IL-8 (Hs00174103); and GAPDH were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes, followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minute. PCR data were analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).
  • Conventional PCR. Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass.) to confirm the results from real-time PCR. PCR was performed using 2 microliters of cDNA solution (1× Taq polymerase (trade name AMPLITAQ GOLD) universal mix PCR reaction buffer (Applied Biosystems) and initial denaturation at 94° C. for 5 minutes. Amplification was optimized for each primer set. For IL-8, CXC ligand 3, and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles); for renin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C. for 30 seconds for 38 cycles); for oxidized LDL receptor and GAPDH (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 33 cycles). Primers used for amplification are listed in Table 7-1. Primer concentration in the final PCR reaction was 1 micromolar except for GAPDH which was 0.5 micromolar. GAPDH primers were the same as for real-time PCR, except that the manufacturer's TaqMan probe was not added to the final PCR reaction. Samples were separated on 2% (w/v) agarose gel and stained with ethidium bromide (Sigma, St. Louis, Mo.). Images were captured on 667 film (Universal Twinpack, VWR International, South Plainfield, N.J.) using a fixed focal-length POLAROID camera (VWR International, South Plainfield, N.J.).
    TABLE 7-1
    Primers used
    Primer name Primers
    Oxidized S: 5′-GAGAAATCCAAAGAGCAAATGG-3′ (SEQ ID NO:1)
    LDL receptor A: 5′-AGAATGGAAAACTGGAATAGG-3′ (SEQ ID NO:2)
    Renin S: 5′-TCTTCGATGCTTCGGATTCC-3′ (SEQ ID NO:3)
    A: 5′-GAATTCTCGGAATCTCTGTTG-3′ (SEQ ID NO:4)
    Reticulon S: 5′-TTACAAGCAGTGCAGAAAACC-3′ (SEQ ID NO:5)
    A: 5′-AGTAAACATTGAAACCACAGCC-3′ (SEQ ID NO:6)
    Interleukin-8 S: 5′-TCTGCAGCTCTGTGTGAAGG-3′ (SEQ ID NO:7)
    A: 5′-CTTCAAAAACTTCTCCACAACC-3′ (SEQ ID NO:8)
    Chemokine S: 5′-CCCACGCCACGCTCTCC-3′ (SEQ ID NO:9)
    (CXC) ligand 3 A: 5′-TCCTGTCAGTTGGTGCTCC-3′ (SEQ ID NO:10)
  • Immunofluorescence. Cells were fixed with cold 4% (w/v) paraformaldehyde (Sigma-Aldrich, St. Louis, Mo.) for 10 minutes at room temperature. Umbilicus-derived cells at passage 0 (P0) (one isolate, directly after isolation) and passage 11 (P11) (two isolates of umbilicus-derived cells), and fibroblasts (P11) were used. Immunocytochemistry was performed using antibodies directed against the following epitopes: vimentin (1:500, Sigma, St. Louis, Mo.), desmin ((Sigma) 1:150; raised against rabbit; or (Chemicon, Temecula, Calif.) 1:300, raised against mouse), alpha-smooth muscle actin (SMA; 1:400; Sigma), cytokeratin 18 (CK18; 1:400; Sigma), von Willebrand Factor (vWF; 1:200; Sigma), and CD34 (human CD34 Class III; 1:100; DAKOCytomation, Carpinteria, Calif.). In addition, the following markers were tested on passage 11 postpartum cells: anti-human GROalpha-PE (1:100; Becton Dickinson, Franklin Lakes, N.J.), anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, Calif.), anti-human oxidized LDL receptor 1 (ox-LDL R1; 1:100; Santa Cruz Biotech), and anti-human NOGO-A (1:100; Santa Cruz, Biotech).
  • Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma, St. Louis, Mo.) for 30 minutes to access intracellular antigens. Where the epitope of interest was located on the cell surface (CD34, ox-LDL R1), Triton X-100 was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL R1, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. The primary antibody solutions were removed and the cultures were washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and/or goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG-FITC (1:150, Santa Cruz Biotech). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using an appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution (no 1° control). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • FACS analysis. Adherent cells in flasks were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) and detached with Trypsin/EDTA (Gibco, Carlsbad, Calif.). Cells were harvested, centrifuged, and resuspended 3% (v/v) FBS in PBS at a concentration of 1×107 cells/milliliter. One hundred microliter aliquots were delivered to conical tubes. Cells stained for intracellular antigens were permeabilized with Perm/Wash buffer (BD Pharmingen, San Diego, Calif.). Antibody was added to aliquots as per manufactures specifications and the cells were incubated for in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess antibody. Cells requiring a secondary antibody were resuspended in 100 microliters of 3% FBS. Secondary antibody was added as per manufactures specification and the cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess secondary antibody. Washed cells were resuspended in 0.5 milliliter PBS and analyzed by flow cytometry. The following antibodies were used: oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROalpha (555042; BD Pharmingen, Bedford, Mass.), Mouse IgG1 kappa, (P-4685 and M-5284; Sigma), Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.). Flow cytometry analysis was performed with FACSCalibur (Becton Dickinson San Jose, Calif.).
  • Results
  • Results of real-time PCR for selected “signature” genes performed on cDNA from cells derived from human umbilical cord, adult and neonatal fibroblasts, and Mesenchymal Stem Cells (MSCs) indicate that both reticulon and oxidized LDL receptor expression were higher in umbilicus-derived cells as compared to other cells. The data obtained from real-time PCR were analyzed by the ΔΔCT (delta delta CT) method and expressed on a logarithmic scale. No significant differences in the expression levels of CXC ligand 3 and GCP-2 were found between postpartum cells and controls. The results of real-time PCR were confirmed by conventional PCR. Sequencing of PCR products further validated these observations. No significant difference in the expression level of CXC ligand 3 was found between postpartum cells and controls using conventional PCR CXC ligand 3 primers listed in Table 7-1.
  • The expression of the cytokine, IL-8 in postpartum cells was elevated in both Growth Medium-cultured and serum-starved postpartum-derived cells. All real-time PCR data were validated with conventional PCR and by sequencing PCR products.
  • After growth in serum-free media, the conditioned media were examined for the presence of IL-8. The greatest amounts of IL-8 were detected in media in which umbilical cells had been grown (Table 7-2). No IL-8 was detected in medium in which human dermal fibroblasts had been grown.
    TABLE 7-2
    IL-8 protein expression measured by ELISA
    Cell type IL-8 secretion
    hFibro ND
    UMBC Isolate 1 2058.42 ± 144.67
    UMBC Isolate 2 2368.86 ± 22.73

    Results of the ELISA assay for interleukin-8 (IL-8) performed on spent media in which had been grown umbilicus-derived cells and human skin fibroblasts.

    Values are presented here are pg/million cells, n = 2, sem.

    ND: Not Detected
  • Cells derived from the human umbilical cord at passage 0 were probed for the expression of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Umbilicus-derived cells were positive for alpha-smooth muscle actin and vimentin, with the staining pattern consistent through passage 11.
  • The expression of GROalpha, GCP-2, oxidized LDL receptor 1 and reticulon in umbilicus-derived cells at passage 11 was investigated by immunocytochemistry.
  • Summary. Concordance between gene expression levels measured by microarray and PCR (both real-time and conventional) has been established for four genes: oxidized LDL receptor 1, renin, reticulon, and IL-8. The expression of these genes was regulated at the mRNA level in postpartum cells. IL-8 was also regulated at the protein level. Differences in expression of GCP-2 and CXC ligand 3 were not confirmed at the mRNA level.
  • Cells derived from the human umbilical cord at passage 0 were probed for the expression of alpha-smooth muscle actin and vimentin, and were positive for both. The staining pattern was preserved through passage 11, suggesting that expression of vimentin and alpha-smooth muscle actin are preserved in cells with passaging, at least in the Growth Medium used.
  • EXAMPLE 8 Immunohistochemical Characterization of Umbilical Cord-Derived Cell Phenotypes
  • The phenotypes of cells found within human umbilical cord tissue was analyzed by immunohistochemistry.
  • Materials & Methods
  • Tissue Preparation. Human umbilical cord tissue was harvested and immersion fixed in 4% (w/v) paraformaldehyde overnight at 4° C. Immunohistochemistry was performed using antibodies directed against the following epitopes (see Table 8-1): vimentin (1:500; Sigma, St. Louis, Mo.), desmin (1:150, raised against rabbit; Sigma; or 1:300, raised against mouse; Chemicon, Temecula, Calif.), alpha-smooth muscle actin (SMA; 1:400; Sigma), cytokeratin 18 (CK18; 1:400; Sigma), von Willebrand Factor (vWF; 1:200; Sigma), and CD34 (human CD34 Class III; 1:100; DAKOCytomation, Carpinteria, Calif.). In addition, the following markers were tested: anti-human GROalpha-PE (1:100; Becton Dickinson, Franklin Lakes, N.J.), anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, Calif.), anti-human oxidized LDL receptor 1 (ox-LDL R1; 1:100; Santa Cruz Biotech), and anti-human NOGO-A (1:100; Santa Cruz Biotech). Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, Calif.) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 microns thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
  • Immunohistochemistry. Immunohistochemistry was performed similar to previous studies (e.g., Messina, et al. (2003) Exper. Neurol. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 1 hour to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (CD34, ox-LDL R1), triton was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL R1, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout the procedure. Primary antibodies, diluted in blocking solution, were then applied to the sections for a period of 4 hours at room temperature. Primary antibody solutions were removed, and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and/or goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG-FITC (1:150; Santa Cruz Biotech). Cultures were washed, and 10 micromolar DAPI (Molecular Probes) was applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epifluorescent microscope (Olympus, Melville, N.Y.). Positive staining was represented by fluorescence signal above control staining. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
    TABLE 8-1
    Summary of Primary Antibodies Used
    Antibody Concentration Vendor
    Vimentin 1:500 Sigma, St. Louis, MO
    Desmin (rb) 1:150 Sigma
    Desmin (m) 1:300 Chemicon, Temecula, CA
    alpha-smooth muscle actin 1:400 Sigma
    (SMA)
    Cytokeratin 18 (CK18) 1:400 Sigma
    von Willebrand factor 1:200 Sigma
    (vWF)
    CD34 III 1:100 DakoCytomation,
    Carpinteria, CA
    GROalpha-PE 1:100 BD, Franklin Lakes, NJ
    GCP-2 1:100 Santa Cruz Biotech
    Ox-LDL R1 1:100 Santa Cruz Biotech
    NOGO-A 1:100 Santa Cruz Biotech

    Results
  • Umbilical Cord Characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord. In particular, vWF and CD34 expression were restricted to blood vessels contained within the cord. CD34+cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery and vein, but not contained with the vessels themselves. CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.
  • Summary. Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD34 are produced in cells within human umbilical cord.
  • EXAMPLE 9 Secretion of Trophic Factors by Umbilicus-Derived Cells
  • The secretion of selected trophic factors from umbilicus-derived PPDCs was measured. Factors were selected that have angiogenic activity (i.e., hepatocyte growth factor (HGF) (Rosen et al. (1997) Ciba Found. Symp. 212:215-26), monocyte chemotactic protein 1 (MCP-1) (Salcedo et al. (2000) Blood 96;34-40), interleukin-8 (IL-8) (L1 et al. (2003) J. Immunol. 170:3369-76), keratinocyte growth factor (KGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) (Hughes et al. (2004) Ann. Thorac. Surg. 77:812-8), tissue inhibitor of matrix metalloproteinase 1 (TIMP1), angiopoietin 2 (ANG2), platelet derived growth factor (PDGFbb), thrombopoietin (TPO), heparin-binding epidermal growth factor (HB-EGF), stromal-derived factor 1alpha (SDF-1 alpha)), neurotrophic/neuroprotective activity (brain-derived neurotrophic factor (BDNF) (Cheng et al. (2003) Dev. Biol. 258;319-33), interleukin-6 (IL-6), granulocyte chemotactic protein-2 (GCP-2), transforming growth factor beta2 (TGFbeta2)), or chemokine activity (macrophage inflammatory protein 1alpha (MIP1alpha), macrophage inflammatory protein 1 beta (MIP1beta), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), 1309, thymus and activation-regulated chemokine (TARC), Eotaxin, macrophage-derived chemokine (MDC), IL-8).
  • Methods & Materials
  • Cell culture. PPDCs derived from umbilical cord, as well as human fibroblasts derived from human neonatal foreskin, were cultured in Growth Medium on gelatin-coated T75 flasks. Cells were cryopreserved at passage 11 and stored in liquid nitrogen. After thawing, Growth Medium was added to the cells, followed by transfer to a 15 milliliter centrifuge tube and centrifugation of the cells at 150×g for 5 minutes. The cell pellet was resuspended in 4 milliliters Growth Medium, and cells were counted. Cells were seeded at 5,000 cells/cm2 in T75 flasks each containing 15 milliliters of Growth Medium, and cultured for 24 hours. The medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 Units/milliliter) and streptomycin (50 micrograms/milliliter, Gibco)) for 8 hours. Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000×g for 5 minutes and stored at −20° C.
  • To estimate the number of cells in each flask, cells were washed with phosphate-buffered saline (PBS) and detached using 2 milliliters trypsin/EDTA (Gibco). Trypsin activity was inhibited by addition of 8 milliliters Growth Medium. Cells were centrifuged at 150×g for 5 minutes. The supernatant was removed, and cells were resuspended in 1 milliliter Growth Medium. Cell number was estimated with a hemocytometer.
  • ELISA assay. Cells were grown at 37° C. in 5% carbon dioxide and atmospheric oxygen. The amount of MCP-1, IL-6, VEGF, SDF-1alpha, GCP-2, IL-8, and TGF-beta2 produced by each cell sample was determined by ELISA (R&D Systems, Minneapolis, Minn.). All assays were performed according to the manufacturer's instructions. Values presented are picograms per milliliter per million cells (n=2, sem).
  • SearchLight Multiplexed ELISA assay. Chemokines (MIP1alpha, MIP1beta, MCP-1, Rantes, I309, TARC, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, TIMP1, ANG2, PDGFbb, TPO, HB-EGF were measured using SearchLight Proteome Arrays (Pierce Biotechnology Inc.). The Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to sixteen proteins per well. The arrays are produced by spotting a 2×2, 3×3, or 4×4 pattern of four to sixteen different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture the chemiluminescent signal generated at each spot within each well of the plate. The signal generated at each spot is proportional to the amount of target protein in the original standard or sample.
  • Results
  • ELISA assay. MCP-1 and IL-6 were secreted by umbilicus-derived PPDCs and dermal fibroblasts (Table 9-1). SDF-1alpha and GCP-2 were secreted by fibroblasts. GCP-2 and IL-8 were secreted by umbilicus-derived PPDCs. TGF-beta2 was not detected from either cell type by ELISA.
    TABLE 9-1
    ELISA Results: Detection of Trophic Factors
    MCP-1 IL-6 VEGF SDF-1α GCP-2 IL-8 TGF-beta2
    Fibroblast  17 ± 1 61 ± 3 29 ± 2 19 ± 1 21 ± 1 ND ND
    Umbilical (022803) 1150 ± 74 4234 ± 289 ND ND 160 ± 11 2058 ± 145 ND
    Umbilical (071003) 2794 ± 84 1356 ± 43  ND ND 2184 ± 98  2369 ± 23  ND

    Key:

    ND: Not Detected.,

    =/− sem
  • SearchLight Multiplexed ELISA assay. TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1beta, MCP1, RANTES, I309, TARC, MDC, and IL-8 were secreted from umbilicus-derived PPDCs (Tables 9-2 and 9-3). No Ang2, VEGF, or PDGFbb were detected.
    TABLE 9-2
    SearchLight Multiplexed ELISA assay results
    TIMP1 ANG2 PDGFbb TPO KGF HGF FGF VEGF HBEGF BDNF
    hFB 19306.3 ND ND  230.5 5.0 ND ND 27.9 1.3 ND
    U1 57718.4 ND ND 1240.0 5.8 559.3 148.7 ND 9.3 165.7
    U3 21850.0 ND ND 1134.5 9.0 195.6  30.8 ND 5.4 388.6

    Key:

    hFB (human fibroblasts),

    U1 (umbilicus-derived PPDC (022803)),

    U3 (umbilicus-derived PPDC (071003)).

    ND: Not Detected.
  • TABLE 9-3
    SearchLight Multiplexed ELISA assay results
    MIP1a MIP1b MCP1 RANTES I309 TARC Eotaxin MDC IL8
    hFB ND ND 39.6 ND ND 0.1 ND ND 204.9
    U1 ND 8.0 1694.2 ND 22.4 37.6 ND 18.9 51930.1
    U3 ND 5.2 2018.7 41.5 11.6 21.4 ND  4.8 10515.9

    Key:

    hFB (human fibroblasts),

    U1 (umbilicus-derived PPDC (022803)),

    U3 (umbilicus-derived PPDC (071003)).

    ND: Not Detected.
  • Summary. Umbilicus-derived cells secreted a number of trophic factors. Some of these trophic factors, such as HGF, bFGF, MCP-1 and IL-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL-6, have important roles in neural regeneration or protection.
  • EXAMPLE 10 In Vitro Immunology
  • Postpartum cell lines were evaluated in vitro for their immunological characteristics in an effort to predict the immunological response, if any, these cells would elicit upon in vivo transplantation. Postpartum cell lines were assayed by flow cytometry for the expression of HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2. These proteins are expressed by antigen-presenting cells (APC) and are required for the direct stimulation of naïve CD4+ T cells (Abbas & Lichtman, CELLULAR AND MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171). The cell lines were also analyzed by flow cytometry for the expression of HLA-G (Abbas & Lichtman, CELLULAR AND MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171), CD178 (Coumans, et. al., (1999) Journal of Immunological Methods 224, 185-196), and PD-L2 (Abbas & Lichtman, CELLULAR AND MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171; Brown, et. al. (2003) The Journal of Immunology 170, 1257-1266). The expression of these proteins by cells residing in placental tissues is thought to mediate the immuno-privileged status of placental tissues in utero. To predict the extent to which postpartum umbilicus-derived cell lines elicit an immune response in vivo, the cell lines were tested in a one-way mixed lymphocyte reaction (MLR).
  • Materials and Methods
  • Cell culture. Cells were cultured in Growth Media in T75 flasks (Corning, Corning, N.Y.) coated with 2% gelatin (Sigma, St. Louis, Mo.) until confluent.
  • Antibody Staining. Cells were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) and detached with Trypsin/EDTA (Gibco, Carlsbad, Mo.). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1×107 per milliliter. Antibody (Table 10-1) was added to one hundred microliters of cell suspension as per manufacturer's specifications and incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were re-suspended in five hundred microliters of PBS and analyzed by flow cytometry using a FACSCalibur instrument (Becton Dickinson, San Jose, Calif.).
    TABLE 10-1
    Antibodies
    Catalog
    Antibody Manufacture Number
    HLA-DR, DP, DQ BD Pharmingen (San Diego, CA) 555558
    CD80 BD Pharmingen 557227
    CD86 BD Pharmingen 555665
    B7-H2 BD Pharmingen 552502
    HLA-G Abcam (Cambridgeshire, UK) ab 7904-100
    CD178 Santa Cruz (San Cruz, CA) sc-19681
    PD-L2 BD Pharmingen 557846
    Mouse IgG2alpha Sigma (St. Louis, MO) F-6522
    Mouse IgG1kappa Sigma P-4685
  • Mixed Lymphocyte Reaction. Cryopreserved vials of passage 10 umbilicus-derived PPDCs labeled as cell line “A” were sent on dry ice to CTBR (Senneville, Quebec) to conduct a mixed lymphocyte reaction using CTBR SOP no. CAC-031. Peripheral blood mononuclear cells (PBMCs) were collected from multiple male and female volunteer donors. Six human volunteer blood donors were screened to identify a single allogeneic donor that exhibited a robust proliferation response in a mixed lymphocyte reaction with the other five blood donors. This donor was selected as the allogeneic positive control donor. The remaining five blood donors were selected as recipients. Stimulator (donor) allogeneic PBMC, autologous PBMC, and postpartum cell lines were treated with mitomycin C. Autologous and mitomycin C-treated stimulator cells were added to responder (recipient) PBMCs and cultured for 4 days. After incubation, [3H]thymidine was added to each sample and cultured for 18 hours. Following harvest of the cells, radiolabeled DNA was extracted, and [3H]-thymidine incorporation was measured using a scintillation counter. Reactions were performed in triplicate using two-cell culture plates with three receivers per plate
  • The stimulation index for the allogeneic donor (SIAD) was calculated as the mean proliferation of the receiver plus mitomycin C-treated allogeneic donor divided by the baseline proliferation of the receiver. The stimulation index of the postpartum cells was calculated as the mean proliferation of the receiver plus mitomycin C-treated postpartum cell line divided by the baseline proliferation of the receiver.
  • Results
  • Mixed Lymphocyte Reaction-Umbilicus-Derived Cells. Results are shown below in Tables 10-2 and -3. The average stimulation index ranged from 6.5 (plate 1) to 9 (plate 2) and the allogeneic donor positive controls ranged from 42.75 (plate 1) to 70 (plate 2) (Table 10-3).
    TABLE 10-2
    Mixed Lymphocyte Reaction Data - Cell Line A (Umbilicus)
    DPM for Proliferation Assay
    Analytical Culture Replicates
    number System 1 2 3 Mean SD CV
    Plate ID: Plate 1
    IM04-2478 Proliferation baseline of receiver 1074 406 391 623.7 390.07 62.5
    Control of autostimulation (Mitomycin C treated autologous cells) 672 510 1402 861.3 475.19 55.2
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 43777 48391 38231 43466.3 5087.12 11.7
    MLR with cell line (Mitomycin C treated cell type A) 2914 5622 6109 4881.7 1721.36 35.3
    SI (donor) 70
    SI (cell line) 8
    IM04-2479 Proliferation baseline of receiver 530 508 527 521.7 11.93 2.3
    Control of autostimulation (Mitomycin C treated autologous cells) 701 567 1111 793.0 283.43 35.7
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 25593 24732 22707 24344.0 1481.61 6.1
    MLR with cell line (Mitomycin C treated cell type A) 5086 3932 1497 3505.0 1832.21 52.3
    SI (donor) 47
    SI (cell line) 7
    IM04-2480 Proliferation baseline of receiver 1192 854 1330 1125.3 244.90 21.8
    Control of autostimulation (Mitomycin C treated autologous cells) 2963 993 2197 2051.0 993.08 48.4
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 25416 29721 23757 26298.0 3078.27 11.7
    MLR with cell line (Mitomycin C treated cell type A) 2596 5076 3426 3699.3 1262.39 34.1
    SI (donor) 23
    SI (cell line) 3
    IM04-2481 Proliferation baseline of receiver 695 451 555 567.0 122.44 21.6
    Control of autostimulation (Mitomycin C treated autologous cells) 738 1252 464 818.0 400.04 48.9
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 13177 24885 15444 17835.3 6209.52 34.8
    MLR with cell line (Mitomycin C treated cell type A) 4495 3671 4674 4280.0 534.95 12.5
    SI (donor) 1
    SI (cell line) 8
    Plate ID: Plate 2
    IM04-2482 Proliferation baseline of receiver 432 533 274 413.0 130.54 31.6
    Control of autostimulation (Mitomycin C treated autologous cells) 1459 633 598 896.7 487.31 54.3
    MLR allogenic donor IM04-2477 (Mitomycin C treated 24286 30823 31346 28818.3 3933.82 13.7
    MLR wilh cell line (Mitomycin C treated cell type A) 2762 1502 6723 3662.3 2724.46 74.4
    SI (donor) 70
    SI (cell line) 9
    IM04-2477 Proliferation baseline of receiver 312 419 349 360.0 54.34 15.1
    (allogenic donor) Control of autostimulation (Mitomycin treated autologous cells) 567 604 374 515.0 123.50 24.0
    Cell line type A Proliferotion baseline of receiver 5101 3735 2973 3936.3 1078.19 27.4
    Control of autostimulation (Mitomycin treated autologous cells) 1924 4570 2153 2882.3 1466.04 50.9
  • TABLE 10-3
    Average stimulation index of umbilical cells and an allogeneic donor
    in a mixed lymphocyte reaction with five individual allogeneic
    receivers.
    Average Stimulation Index
    Recipient Umbilicus
    Plate 1 (receivers 1-4) 42.75 6.5
    Plate 2 (receiver 5) 70 9
  • Antigen Presenting Cell Markers Produced by Umbilicus-Derived Cells. The histograms of the flow cytometry analysis show umbilical cells were negative for production of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, because the fluorescence values were comparable to the IgG control. This indicates that umbilical cell lines lack the cell surface molecules required to directly stimulate CD4+ T cells.
  • Immuno-modulating Markers in Umbilicus-Derived Cells. The umbilical cells analyzed by flow cytometry were positive for expression of PD-L2, as reflected in the increase in fluorescence relative to the IgG control. The cells were negative for expression of CD178 and HLA-G, as noted by fluorescence values consistent with the IgG control.
  • Summary. In the mixed lymphocyte reactions conducted with umbilical cell lines the average stimulation index ranged from 6.5 to 9, while that of the allogeneic positive controls ranged from 42.75 to 70. Umbilical cell lines did not express detectable amounts of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as measured by flow cytometry. Umbilical cell lines also did not express the immuno-modulating proteins HLA-G and CD178, but expression of PD-L2 was detected by flow cytometry. Allogeneic donor PBMCs contain antigen-presenting cells expressing HLA-DR, DQ, CD8, CD86, and B7-H2, thereby allowing for the stimulation of allogeneic lymphocytes. The absence on umbilicus-derived cells of antigen-presenting cell surface molecules required for the direct stimulation of naïve CD4+ T cells, as well as the presence of PD-L2, an immunomodulating protein, could account for the low stimulation index exhibited by these cells in a MLR as compared to allogeneic controls.
  • EXAMPLE 11 Plasma Clotting Assay
  • Cells useful for therapy may be injected systemically for certain applications where the cells are able to target the site of action. It is important that injected cells not cause thrombosis, as it may be fatal. Tissue factor, a membrane-bound procoagulant glycoprotein, is the initiator of the extrinsic clotting cascade, which is the predominant coagulation pathway in vivo. Tissue factor also plays an important role in embryonic vessel formation, for example, in the formation of the primitive vascular wall (Brodsky et al. (2002) Exp. Nephrol. 10:299-306). To determine the potential for PPDCs to initiate clotting, umbilicus-derived PPDCs were evaluated for tissue factor expression and for their ability to initiate plasma clotting.
  • Methods & Materials
  • Human Tissue factor. Human tissue factor (SIMPLASTIN, Organon Teknika Corporation, Durham, N.C.), was reconstituted with 20 milliliters distilled water. The stock solution was serially diluted (1:2) in eight tubes. Normal human plasma (George King Bio-Medical, Overland Park, Kans.) was thawed at 37° C. in a water bath and then stored in ice before use. 100 microliters phosphate buffered saline (PBS), 10 microliters diluted SIMPLASTIN, 30 microliters 0.1 Molar calcium chloride, and 100 microliters of normal human plasma were added to each well of a 96-well plate. A negative control well did not receive any SIMPLASTIN. The plate was immediately placed in a temperature-controlled microplate reader and absorbance measured at 405 nanometer at 40 second intervals for 30 minutes.
  • J-82 and umbilicus-derived cells. J-82 cells (ATCC, MD) were grown in Iscove's modified Dulbecco's medium (IMDM; Gibco, Carlsbad, Calif.) containing 10% (v/v) fetal bovine serum (FBS; Hyclone, Logan Utah), 1 millimolar sodium pyruvate (Sigma Chemical, St. Louis, Mo.), 2 millimolar L-Glutamine (Mediatech Herndon, Va.), 1× non-essential amino acids (Mediatech Herndon, Va.). At about 70% confluence, cells were transferred at 100,000, 50,000 and 25,000 cells/well to wells of 96-well plate. Umbilicus-derived cells were cultured in Growth Medium in gelatin-coated T75 flasks (Corning, Corning, N.Y.). Umbilicus-derived cells at passage 18 were transferred to wells at a density of 50,000 cells/well. Culture medium was removed from each well after centrifugation at 150×g for 5 minutes. Cells were suspended in PBS without calcium and magnesium. Cells incubated with anti-tissue factor antibody cells were incubated with 20 micrograms/milliliter CNTO 859 (Centocor, Malvern, Pa.) for 30 minutes. Calcium chloride (30 microliters) was added to each well. The plate was promptly placed in a temperature-controlled microplate reader and absorbance was measured at 405 nanometers at 40 second intervals for 30 minutes.
  • Antibody Staining. Cells were washed in PBS and detached from the flask with Trypsin/EDTA (Gibco, Carlsbad, Calif.). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1×107 per milliliter. Antibody was added to 100 microliters of cell suspension according to the manufacturer's specifications. The cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS, then centrifuged at 150×g for 5 minutes to remove unbound antibody. Cells were resuspended in 100 microliters of 3% FBS and secondary antibody added in accordance with the manufacturer's instructions. Cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound secondary antibody. Washed cells were resuspended in 500 microliters of PBS and analyzed via flow cytometry.
  • Flow Cytometry Analysis. Flow cytometry analysis was performed with a FACSCalibur instrument (Becton Dickinson, San Jose, Calif.).
  • Results
  • Flow cytometry analysis revealed that umbilicus-derived postpartum cells are less active in promoting plasma clotting than the J82 cells. Although a plasma clotting assay demonstrated that the tissue factor present in the umbilicus-derived cells was active, clotting took longer than with the J-82 cells, as evidenced by the longer time to half-maximal absorbance (T½ to max; Table 11-1). The T½ to max is inversely proportional to the number of J-82 cells. Umbilicus-derived cells decreased the clotting rate as indicated by the T½ to max. Clotting was observed with both early (P5) and late (P18) passaged cells. Preincubation of umbilical cells with CNTO 859, an antibody to tissue factor, inhibited the clotting reaction, establishing that tissue factor was responsible for the clotting.
    TABLE 11-1
    The effect of human tissue factor (Simplastin ®) and
    umbilicus-derived cells (Umb) on plasma clotting. The time
    to half maximal absorbance (T½ to max) at the plateau in
    seconds was used as a measurement unit.
    T½ to max
    (seconds)
    Standard
    (Simplastin ® Dilution)
    1:2 61
    1:4 107
    1:8 147
    1:16 174
    1:32 266
    1:64 317
    1:128 378
    0 (negative control) 1188
    J-82 cells
    100,000 122
     50,000 172
     25,000 275
    Umb P5
     50,000 833
    Umb P18
     50,000 443
  • Summary. Umbilicus-derived PPDCs produce some tissue factor, but the addition of an antibody against tissue factor can inhibit the clotting activity of the tissue factor. Tissue factor is normally found on cells in a conformation that is inactive, but which is activated by mechanical or chemical (e.g., LPS) stress (Sakariassen et al. (2001) Thromb. Res. 104:149-74; Engstad et al. (2002) Int. Immunopharmacol. 2:1585-97). Thus, minimization of stress during the preparation process of PPDCs may prevent activation of tissue factor. In addition to the thrombogenic activity, tissue factor has been associated with angiogenic activity. For this reason, tissue factor activity may be beneficial when umbilicus-derived PPDCs are transplanted in tissue, but should be inhibited when PPDCs are injected intravenously.
  • EXAMPLE 12 Transplantation of Umbilicus-Derived Cells
  • Cells derived from the postpartum umbilical cord are useful for regenerative therapies. Tissue produced by SCID mice following transplantation of a biodegradable material with and without umbilicus-derived cells was evaluated. The materials evaluated were VNW nonwoven scaffolds, 35/65 PCL/PGA foam, and a self-assembling peptide hydrogel.
  • Methods & Materials
  • Cell Culture. Umbilicus-derived cells were grown in Growth Medium in gelatin-coated flasks.
  • Matrix Preparation. A nonwoven scaffold was prepared using a traditional needle punching technique as described below. Fibers, comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), were obtained from Ethicon, Inc. (Somerville, N.J.). The fibers were filaments of approximately 20 microns in diameter. The fibers were cut and crimped into substantially uniform 2-inch lengths to form 2-inch staple fibers. A dry lay needle-punched nonwoven matrix was prepared utilizing the PGA/PLAstaple fibers, hereinafter “VNW”. The staple fibers were opened and carded on standard nonwoven machinery. The resulting mat was in the form of webbed staple fibers. The webbed staple fibers were needle-punched to form the dry lay needle-punched nonwoven scaffold. The nonwoven scaffold was rinsed in water followed by another incubation in ethanol to remove any residual chemicals or processing aids used during the manufacturing process.
  • Foams, composed of 35/65 poly(epsilon-caprolactone)/poly(glycolic acid) (35/65 PCL/PGA) copolymer, were formed by the process of lyophilization, as described in U.S. Pat. No. 6,355,699.
  • A self assembling peptide hydrogel (RAD16 self-assembling peptides (3D Matrix, Cambridge, Mass.)) was obtained as a sterile 1% (w/v) solution in water.
  • Sample Preparation. One million viable cells were seeded in 15 microliters Growth Medium onto 5 millimeter diameter, 2.25 millimeter thick VNW scaffolds (64.33 milligrams/cc); or 5 millimeter diameter 35/65 PCL/PGA foam disks. Cells were allowed to attach for two hours before adding more Growth Medium to cover the scaffolds. Cells were grown on scaffolds overnight. Control scaffolds without cells were also incubated in medium.
  • The self-assembling peptide solution was mixed 1:1 with 1×106 cells in 10% (w/v) sucrose (Sigma, St Louis, Mo.), 10 millimolar HEPES (pH about 7), in Dulbecco's modified medium (DMEM; Gibco) immediately before use. The final concentration of cells in the self-assembling peptide hydrogel was 1×106 cells/100 microliters.
  • Test Material (N=4/condition)
  • 1. VNW scaffold+1×106 umbilicus-derived cells
  • 2. 35/65 PCL/PGA foam+1×106 umbilicus-derived cells
  • 3. Self-assembling peptide+1×106 umbilicus-derived cells
  • 4. 35/65 PCL/PGA foam
  • 5. VNW scaffold
  • Animal Preparation. The animals utilized in this study were handled and maintained in accordance with the current requirements of the Animal Welfare Act. Compliance with the above Public Laws were accomplished by adhering to the Animal Welfare regulations (9 C.F.R.) and conforming to the current standards promulgated in the Guide for the Care and Use of Laboratory Animals, 7th edition.
  • Animals: Male mice (Mus musculus) (Fox Chase SCID; Harlan Sprague Dawley, Inc., Indianapolis, Ind.), were used at 5 weeks of age. All handling of the SCID mice took place under a hood. Each animal was individually weighed, and anesthetized with an intraperitoneal injection of a mixture of 60 milligram/kilogram KETASET (ketamine hydrochloride) (Aveco Co., Inc., Fort Dodge, Iowa), 10 milligram/kilogram ROMPUN (xylazine) (Mobay Corp., Shawnee, Kans.) and saline. After induction of anesthesia, the back of the animal from the dorsal cervical area to the dorsal lumbosacral area was clipped free of hair using electric animal clippers. The area was then scrubbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine. Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period.
  • Subcutaneous Implantation Technique. Four skin incisions, each approximately 1 centimeter in length, were made on the dorsa of the mice. Two cranial implantation sites, one each to the left and right of the vertebral column, were located transversely over the dorsal lateral thoracic region, about 5 millimeters caudal to the palpated inferior edge of the scapula. Two additional implants, one on each side of the midline, were placed transversely over the gluteal muscle area at the caudal sacro-lumbar level, about 5 millimeters caudal to the palpated iliac crest. Implants were randomly placed in these sites. The skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected in the case of the self-assembling peptide) about 1 centimeter caudal to the incision. The appropriate test material was implanted into the subcutaneous space. The skin incision was closed with metal clips.
  • Animal Housing. Animals were individually housed in microisolator cages throughout the course of the study within a temperature range of 64° F.-79° F. and relative humidity of 30% to 70%, and maintained on a 12 hour light/12 hour dark (approximately) cycle. The diet consisted of Irradiated Pico Mouse Chow 5058 (Purina Co.) and water provided ad libitum.
  • Mice were euthanized by carbon dioxide inhalation at designated intervals. The subcutaneous implants with the overlying skin were excised and frozen for histology.
  • Histology. Excised skin with implant was fixed with 10% neutral buffered formalin (Richard-Allan Scientific, Kalamazoo, Mich.). Samples with overlying and adjacent tissue were centrally bisected, paraffin-processed, and embedded on cut surface using routine methods. Embedded tissue was sectioned (five-micron sections) on a microtome and stained with hematoxylin and eosin (Poly Scientific, Bay Shore, N.Y.) using routine methods.
  • Results
  • There was minimal ingrowth of tissue into control foams without umbilicus-derived cells implanted subcutaneously in SCID mice after 30 days. In contrast, there was extensive tissue fill into foams implanted with umbilicus-derived cells.
  • There was some tissue ingrowth in VNW scaffolds. Nonwoven scaffolds seeded with umbilicus-derived cells showed increased matrix deposition and mature blood vessels.
  • Summary. Human umbilicus-derived cells were shown to dramatically increase good quality tissue formation in biodegradable scaffolds. Synthetic absorbable nonwoven scaffolds, foam discs (5.0 millimeters diameter×1.0 millimeter thick), or self-assembling peptide hydrogels were seeded with cells derived from human umbilical cord and implanted subcutaneously bilaterally in the dorsal spine region of SCID mice. The umbilicus-derived cells enhanced tissue ingrowth and blood vessel formation on the scaffolds in immune deficient mice, compared to that on scaffolds not seeded with umbilicus-derived cells.
  • EXAMPLE 13 Transplantation of Umbilicus-Derived Cells Under the Kidney Capsule
  • Transplantation of pancreatic islets to the kidney capsule is routinely performed to evaluate transplantation methodologies for the treatment of diabetes (Refaie et al., 1998). In addition to pancreatic islets, other cells may be differentiated into insulin secreting cells capable of blood glucose homeostasis. The suitability of umbilicus-derived cells for this purpose was evaluated.
  • Methods & Materials
  • Cell Culture. Umbilicus-derived cells (isolate 1, P10) were removed from liquid nitrogen storage and grown in Growth Medium on gelatin (Sigma)-coated T225 (Corning, Corning, N.Y.) flasks until confluent.
  • The culture medium on umbilicus-derived cells was replaced with Ham's F12 medium (Gibco) containing 10 millimolar nicotinamide (Sigma), 25 millimolar glucose (Sigma), 10 nanogram/milliliter EGF (PeproTech, Rocky Hill, N.J.), 20 nanogram/milliliter bFGF (PeproTech) and 15 millimolar GLP-1 (Sigma) and cells were further cultured for 2 weeks.
  • Cells from two flasks were washed with phosphate buffered saline (PBS), and a single cell suspension was obtained by using Trypsin/EDTA (Gibco). Cryopreserved GM-CSF mobilized CD34+ cells were purchased from Cambrex, Walkersville, Md. (lot 1F0174 donor 7956). CD34+ cells were thawed and washed in DMEM medium.
  • The cell suspension was washed twice in DMEM. Cell number and viability was estimated after Trypan blue (Sigma) staining using a hemocytometer. Aliquots of the cell suspension containing ˜300,000 viable cells were centrifuged at 150×g and the cells resuspended in approximately 6 microliters of DMEM and drawn into a 20 microliter pipette tip connected to a 1 milliliter syringe. The tip of the pipette tip containing the cells was clamped using a small Ligaclip (Ethicon Endosurgery, Cincinnati Ohio).
  • Animal preparation. Mice (Mus musculus)/Fox Chase SCID/Male (Harlan Sprague Dawley, Inc., Indianapolis, Ind.) were 8 weeks of age. All handling of the SCID mice took place under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligram/kilogram KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligram/kilogram ROMPUN (xylazine, Mobay Corp., Shawnee, Kans.) and saline. After induction of anesthesia, the entire back of the animal from the dorsal cervical area to the dorsal lumbosacral area was clipped free of hair using electric clippers. The area was scrubbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine. Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period. The anesthetized and surgically prepared animal was placed in the desired recumbent position. A transverse incision was made on the left abdominal side approximately 2 centimeters caudal to the rib cage of animal. The kidney was exposed and the capsule pierced with a 26-gauge needle. A capsule lance (modified glass pipette tip) was used to create a space beneath the kidney capsule into which the cells were introduced. The cells were injected via a syringe with a micropipette tip attached. The pocket was closed by passing an ophthalmic cautery pen (Aaron Medical Industries, St. Petersburg, Fla.) over the opening (not touching the kidney). The kidney was placed back in the correct anatomical position, and the muscle layer sutured closed. The skin was closed with wound clips.
  • The experimental design comprised one cell transplantation in each mouse; four treatments with n-value of 4 per treatment; and three time-points (1, 14 & 30 days).
  • Mice were euthanized by carbon dioxide inhalation at their designated intervals. The kidney implantation sites were excised and frozen for histology.
  • Immunohistochemistry. Frozen kidney implantation site were embedded on edge in O.C.T. Compound (Sakura Inc., Torrance, Calif.). The kidney tissue was trimmed by cryosectioning to yield a five-micron section of the implantation site and adjacent tissue. Yielded sections were fixed in freshly prepared 4% paraformaldehyde (EM Sciences Gibbstown, N.J.) in phosphate buffered saline (Gibco) for 15 minutes. Sections were washed in PBS and incubated in 3% goat serum in PBS blocking solution for one hour. Blocking solution was removed by gentle aspiration. Sections were incubated in anti-human nuclei antibody (Chemicon International, Temecula, Calif.) diluted 1:100 in blocking solution for one hour. Sections were washed with PBS and incubated in florescent labeled goat anti-mouse IgG antibody (Molecular Probes Eugene, Oreg.), diluted 1:200 in blocking solution for 30 minutes in absence of light. Sections were washed in PBS and incubated in 10 micromolar DAPI (Molecular Probes) for five minutes. Sections were washed in PBS and examined by fluorescent microscopy.
  • Tri-Chrome Staining. Frozen kidney implantation sites were embedded on edge in O.C.T. Compound (Sakura Inc.). The kidney tissue was trimmed by cryosectioning to yield a five-micron section of the implantation site and adjacent tissue. Yielded sections were fixed in 10% neutral buffered formalin (Richard-Allan Scientific) for 15 minutes. Sections were stained tri-chrome (Poly Scientific) using manufactures methods.
  • Treatments:
  • 1. 3×103 cells from umbilical cord
  • 2. 3×103 cells from umbilical cord+3×103 CD34+ cells
  • Added three animals as control (No cells)
  • Results
  • The viability of the umbilicus-derived cells was ˜75%; that of the CD34+ cells was 95%. Initial attempts to transplant 1×106 viable cells were unsuccessful due to the kidney capsule not being large enough to accommodate the cells. Cells were transplanted within 3 hours of trypsinization. The localization of postpartum cells under the kidney capsule was observed microscopically. There were no apparent differences in the number and distribution of umbilicus-derived cells with or without CD34+ cells at each time point. There was an apparent decrease in cell numbers over time.
  • Staining of cells under the kidney capsule showed the retention of transplanted cells. Human cells were detected using the human nuclear antigen. All cells (human and mouse) were detected using DAPI.
  • Umbilicus-derived cells were microscopically observed 14 and 30 days post-transplantation. Human cells were again stained for human nuclear antigen. TriChrome staining was used to detect the presence of collagen
  • Summary. Transplantation of cells into the renal capsule was successful. The observed reduction in cell number over time in this experiment may be due to a number of factors such as the viability of cells at the time of transplant, innate immunity and insufficient nutrient availability due to vascularization issues. While long-term survival or even growth of the cells in vivo might be useful for certain purposes, it is not required for the cells to be used in many applications, nor do these results reflect the ability of the cells to survive and grow over longer terms.
  • Reference
    • Refaie A., Gabr M. et al., (1998) Experimental islet cell transplantation in rats: Optimization of the transplantation site. Trans. Proc. 30:400-403
    EXAMPLE 14 Short-Term Neural Differentiation of Umbilicus-Derived Cells
  • The ability umbilicus-derived postpartum cells (PPDCs) to differentiate into neural lineage cells was examined.
  • Materials & Methods
  • Isolation and Expansion of Cells. Umbilicus-derived PPDCs were isolated and expanded as described in Example 1 and 2.
  • Modified Woodbury-Black Protocol.
  • (A) This assay was adapted from an assay originally performed to test the neural induction potential of bone marrow stromal cells (1). Umbilicus PPDCs (P4) were thawed and culture expanded in Growth Media at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 6,000 cells per well of a Titretek II glass slide (VWR International, Bristol, Conn.). As controls, mesenchymal stem cells (P3; 1F2155; Cambrex, Walkersville, Md.), osteoblasts (P5; CC2538; Cambrex), omental cells (P6), adipose-derived cells (U.S. Pat. No. 6,555,374 B1) (P6) and neonatal human dermal fibroblasts (P6; CC2509; Cambrex) were also seeded under the same conditions.
  • All cells were initially expanded for 4 days in DMEM/F12 medium (Invitrogen, Carlsbad, Calif.) containing 15% (v/v) fetal bovine serum (FBS; Hyclone, Logan, Utah), basic fibroblast growth factor (bFGF; 20 nanogram/milliliter; Peprotech, Rocky Hill, N.J.), epidermal growth factor (EGF; 20 nanogram/milliliter; Peprotech), penicillin (50 Units/milliliter), and streptomycin (50 micrograms/milliliter (Invitrogen). After 4 days, cells were rinsed in phosphate-buffered saline (PBS; Invitrogen) and were subsequently cultured in DMEM/F12 medium+20% (v/v) FBS+penicillin (50 Units/milliliter)+streptomycin (50 micrograms/milliliter (Invitrogen) for 24 hours. After 24 hours, cells were rinsed with PBS. Cells were then cultured for 1-6 hours in an induction medium which was comprised of DMEM/F12 (serum-free) containing 200 millimolar butylated hydroxyanisole, 10 micromolar potassium chloride, 5 milligram/milliliter insulin, 10 micromolar forskolin, 4 micromolar valproic acid, and 2 micromolar hydrocortisone (all chemicals from Sigma, St. Louis, Mo.). Cells were then fixed in cold (−20° C.) 100% methanol and immunocytochemistry was performed (see methods below) to assess human nestin protein expression.
  • (B) PPDCs (umbilicus, P11) and adult human dermal fibroblasts (1F1853, P11) were thawed and culture expanded in Growth Medium at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at similar density as in (A), but onto (1) 24 well tissue culture-treated plates (TCP, Falcon brand, VWR International), (2) TCP wells+2% (w/v) gelatin adsorbed for 1 hour at room temperature, or (3) TCP wells+20 micrograms/milliliter adsorbed mouse laminin (adsorbed for a minimum of 2 hours at 37° C.; Invitrogen).
  • As in (A) above, cells were initially expanded and media switched at the aforementioned timeframes. One set of cultures was fixed at 5 days and six hours with cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. In the second set of cultures, the medium was removed and switched to Neural Progenitor Expansion medium (NPE) consisting of Neurobasal-A medium (Invitrogen) containing B27 (B27 supplement; Invitrogen), L-glutamine (4 millimolar), penicillin (50 Units/milliliter), and streptomycin (50 micrograms/milliliter (Invitrogen). NPE medium was further supplemented with retinoic acid (RA; 1 micromolar; Sigma). This medium was removed 4 days later and cultures were fixed with cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for nestin, GFAP, and TuJ1 protein expression (see Table 14-1).
    TABLE 14-1
    Summary of Primary Antibodies Used
    Antibody Concentration Vendor
    Rat 401 (nestin) 1:200 Chemicon, Temecula, CA
    Human Nestin 1:100 Chemicon
    TuJ1 (BIII Tubulin) 1:500 Sigma, St. Louis, MO
    GFAP  1:2000 DakoCytomation,
    Carpinteria, CA
    Tyrosine hydroxylase (TH)  1:1000 Chemicon
    GABA 1:400 Chemicon
    Desmin (mouse) 1:300 Chemicon
    Alpha smooth muscle actin 1:400 Sigma
    Human nuclear protein 1:150 Chemicon
    (hNuc)
  • Two Stage Differentiation Protocol. Umbilicus-derived PPDCs (P11), adult human dermal fibroblasts (P11; 1F1853; Cambrex) were thawed and culture expanded in Growth Medium at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm2, but onto 24 well plates coated with laminin (BD Biosciences, Franklin Lakes, N.J.) in the presence of NPE media supplemented with bFGF (20 nanograms/milliliter; Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter; Peprotech) (whole media composition further referred to as NPE+F+E). At the same time, adult rat neural progenitors isolated from hippocampus (P4; (062603)) were also plated onto 24 well lamin-coated plates in NPE+F+E media. All cultures were maintained in such conditions for a period of 6 days (cells were fed once during that time) at which time media were switched to the differentiation conditions listed in Table 14-2 for an additional period of 7 days. Cultures were fixed with ice-cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human or rat nestin, GFAP, and TuJ1 protein expression.
    TABLE 14-2
    Summary of Conditions for Two-Stage Differentiation Protocol
    A B
    COND. # PRE-DIFFERENTIATION 2nd STAGE DIFF
    1 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + SHH (200 ng/ml) + F8 (100 ng/ml)
    2 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + SHH (200 ng/ml) + F8 (100 ng/ml) + RA
    (1 micromolar)
    3 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + RA (1 micromolar)
    4 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + F (20 ng/ml) + E (20 ng/ml)
    5 NPE + F (20 ng/ml) + E (20 ng/ml) Growth Medium
    6 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 1B + rhGDF-5 (20 ng/ml)
    7 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 1B + BMP7 (20 ng/ml)
    8 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 1B + GDNF (20 ng/ml)
    9 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 2B + rhGDF-5 (20 ng/ml)
    10 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 2B + BMP7 (20 ng/ml)
    11 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 2B + GDNF (20 ng/ml)
    12 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 3B + rhGDF-5 (20 ng/ml)
    13 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 3B + BMP7 (20 ng/ml)
    14 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 3B + GDNF (20 ng/ml)
    15 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + rhGDF-5 (20 ng/ml)
    16 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + BMP7 (20 ng/ml)
    17 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + GDNF (20 ng/ml)
  • Multiple Growth Factor Induction Protocol. Umbilicus-derived PPDCs re thawed and culture expanded in Growth Medium at 5,000 cells/cm2 until sub-ce (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm2, onto 24 well laminin-coated plates (BD Biosciences) in the presence of NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter). In addition, some wells contained NPE+F+E+2% FBS or 10% FBS. After four days of “pre-differentiation” conditions, all media were removed and samples were switched to NPE medium supplemented with sonic hedgehog (SHH; 200 nanograms/milliliter; Sigma, St. Louis, Mo.), FGF8 (100 nanograms/milliliter; Peprotech), BDNF (40 nanograms/milliliter; Sigma), GDNF (20 nanograms/milliliter; Sigma), and retinoic acid (1 micromolar; Sigma). Seven days post medium change, cultures were fixed with ice-cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nestin, GFAP, TuJ1, desmin, and alpha-smooth muscle actin expression.
  • Neural Progenitor Co-Culture Protocol. Adult rat hippocampal progenitors (062603) were plated as neurospheres or single cells (10,000 cells/well) onto laminin-coated 24 well dishes (BD Biosciences) in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter).
  • Umbilicus-derived PPDCs (P11) were thawed and culture expanded in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter) at 5,000 cells/cm2 for a period of 48 hours. Cells were then trypsinized and seeded at 2,500 cells/well onto existing cultures of neural progenitors. The existing medium was exchanged for fresh medium. Four days later, cultures were fixed with ice-cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nuclear protein (hNuc, Chemicon) (Table 14-1 above) to identify PPDCs.
  • Immunocytochemistry. Immunocytochemistry was performed using the antibodies listed in Table 14-1. Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Primary antibody solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) was applied for 10 minutes to help visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). Positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Results
  • Woodbury-Black Protocol.
  • (A) Upon incubation in this neural induction composition, all cell types transformed into cells with bipolar morphologies and extended processes. Other larger non-bipolar morphologies were also observed. Furthermore, the induced cell populations stained positively for nestin, a marker of multipotent neural stem and progenitor cells.
  • (B) When repeated on tissue culture plastic (TCP) dishes, nestin expression was not observed unless laminin was pre-adsorbed to the culture surface. To further assess whether nestin-expressing cells could then go on to generate mature neurons, PPDCs and fibroblasts were exposed to NPE+RA (1 micromolar), a medium composition known to induce the differentiation of neural stem and progenitor cells into such cells (2, 3, 4). Cells were stained for TuJ1, a marker for immature and mature neurons, GFAP, a marker of astrocytes, and nestin, a marker indicative of neural progenitors. TuJ1 expression was not turned on, nor were cells with neuronal morphology observed under any of the tested conditions, suggesting that neurons were not generated in the short term. Furthermore, nestin and GFAP expression, which were found in PPDCs and fibroblasts on laminin-coated substrates, were not produced under these conditions.
  • Two Stage Differentiation Results. Umbilicus-derived cell isolates, as well as human fibroblasts and rodent neural progenitors (as negative and positive control cell types, respectively), were plated on laminin (neural-promoting)-coated dishes and exposed to 13 different growth conditions (and two control conditions) known to promote differentiation of neural progenitors into neurons and astrocytes. In addition, two conditions were added to examine the influence of GDF5, and BMP7 on PPDC differentiation. Generally, a two-step differentiation approach was taken, where the cells were first placed in neural progenitor expansion conditions for a period of 6 days followed by full differentiation conditions for 7 days. Morphologically, umbilical cells exhibited fundamental changes in cell morphology throughout the time-course of this procedure. However, in no cases were neuronal or astrocytic-shaped cells observed except for in control, neural progenitor-plated conditions. Immunocytochemistry, negative for human nestin, TuJ1, and GFAP confirmed these morphological observations. Results are summarized in Table 14-3 below.
    TABLE 14-3
    Staining results for human nestin, GFAP, and TuJ1 respectively
    in Two Stage Differentiation Experiment. Note that + means
    that at least a portion (>0%) of the cells were positive for the
    stain indicated. Human nestin: immature neural stem and progenitor
    cells; GFAP: astrocytes; TuJ1: immature and mature neurons.
    Umbilical Neural
    CONDITION Fibroblasts PPDCs Progenitors.
    1 −/−/− −/−/− +/+/+
    2 −/−/− −/−/− +/+/+
    3 −/−/− −/−/− +/+/+
    4 −/−/− −/−/− +/+/+
    5 −/−/− −/−/− +/+/+
    6 −/−/− −/−/− +/+/+
    7 −/−/− −/−/− +/+/+
    8 −/−/− −/−/− +/+/+
    9 −/−/− −/−/− +/+/+
    10 −/−/− −/−/− +/+/+
    11 −/−/− −/−/− +/+/+
    12 −/−/− −/−/− +/+/+
    13 −/−/− −/−/− +/+/+
    14 −/−/− −/−/− +/+/+
    15 −/−/− −/−/− +/+/+
    16 −/−/− −/−/− +/+/+
    17 −/−/− −/−/− +/+/+
  • Multiple Growth Factor Induction Results. Following a one week exposure to a variety of neural differentiation agents, cells were stained for markers indicative of neural progenitors (human nestin), neurons (TuJ1), and astrocytes (GFAP). Cells grown in the first stage in non-serum containing media had different morphologies than those cells in serum containing (2% or 10%) media, indicating potential neural differentiation. Specifically, following a two step procedure of exposing umbilical PPDCs to EGF and bFGF, followed by SHH, FGF8, GDNF, BDNF, and retinoic acid, cells showed long extended processes similar to the morphology of cultured astrocytes. When 2% FBS or 10% FBS were included in the first stage of differentiation, cell number was increased and cell morphology was unchanged from control cultures at high density. Potential neural differentiation was not evidenced by immunocytochemical analysis for human nestin, TuJ1, or GFAP.
  • Neural Progenitor and PPDC Co-culture Procedures. Umbilicus-derived cells were plated onto cultures of rat neural progenitors seeded two days earlier in neural expansion conditions (NPE+F+E). While visual confirmation of plated umbilicus proved that these cells were plated as single cells, human-specific nuclear staining (hNuc) 4 days post-plating (6 days total length of experiment) showed that they tended to ball up and avoid contact with the neural progenitors. Furthermore, where umbilicus cells attached, these cells spread out and appeared to be innervated by differentiated neurons that were of rat origin suggesting that the umbilical cells may have differentiated into muscle cells. This observation was based upon morphology under phase contrast microscopy. Another observation was that typically large cell bodies (larger than neural progenitors) possessed morphologies resembling neural progenitors, with thin processes spanning out in multiple directions. HNuc staining (found in one half of the cell's nucleus) suggested that in some cases these human cells may have fused with rat progenitors and assumed their phenotype. Controls wells containing neural progenitors only had fewer total progenitors and apparent differentiated cells than did co-culture wells containing umbilicus, further indicating that umbilicus-derived cells influenced the differentiation and behavior of neural progenitors either by release of chemokines and cytokines, or by contact-mediated effects.
  • Summary. Multiple protocols were conducted to determine the short term potential of umbilicus-derived PPDCs to differentiate into neural lineage cells. These included phase contrast imaging of morphology in combination with immunocytochemistry for nestin, TuJ1, and GFAP, proteins associated with multipotent neural stem and progenitor cells, immature and mature neurons, and astrocytes, respectively. Evidence was observed to suggest that neural differentiation occurred in certain instances in these short-term protocols.
  • Several notable observations were made in co-cultures of PPDCs with neural progenitors. This approach, using human PPDCs along with a xenogeneic cell type allowed for absolute determination of the origin of each cell in these cultures. First, some cells were observed in these cultures where the cell cytoplasm was enlarged, with neurite-like processes extending away from the cell body, yet only half of the body labeled with hNuc protein. Those cells may be human PPDCs that have differentiated into neural lineage cells or they may be PPDCs that have fused with neural progenitors of rat origin. Second, it appeared that neural progenitors extended neurites to PPDCs in a way that indicates the progenitors differentiated into neurons and innervated the PPDCs. Third, cultures of neural progenitors and PPDCs had more cells of rat origin and larger amounts of differentiation than control cultures of neural progenitors alone, further indicating that plated PPDCs provided soluble factors and or contact-dependent mechanisms that stimulated neural progenitor survival, proliferation, and/or differentiation.
  • References
    • (1) Woodbury, D. et al. (2000). J Neurosci. Research. 61(4): 364-70.
    • (2) Jang, Y. K. et al. (2004). J. Neurosci. Research. 75(4): 573-84.
    • (3) Jones-Villeneuve, E. M. et al. (1983). Mol Cel Biol. 3(12): 2271-9.
    • (4) Mayer-Proschel, M. et al. (1997). Neuron. 19(4): 773-85.
    EXAMPLE 15 Long-Term Neural Differentiation of Umbilicus-Derived Cells
  • The ability of umbilicus-derived cells to undergo long-term differentiation into neural lineage cells was evaluated.
  • Materials & Methods
  • Isolation and Expansion of Postpartum Cells (PPDCs). Umbilicus-derived PPDCs were isolated and expanded as described in Examples 1 and 2.
  • Umbilicus-derived Cell Thaw and Plating. Frozen aliquots of umbilicus-derived cells P11 and P12, previously grown in Growth Medium were thawed and plated at 5,000 cells/cm2 in T-75 flasks coated with laminin (BD, Franklin Lakes, N.J.) in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine (4 millimolar), penicillin (50 Units/milliliter), and streptomycin (50 micrograms/milliliters), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) media. NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.), herein referred to as NPE+bFGF+EGF.
  • Control Cell Plating. In addition, adult human dermal fibroblasts (P11, Cambrex, Walkersville, Md.) and mesenchymal stem cells (P5, Cambrex) were thawed and plated at the same cell seeding density on laminin-coated T-75 flasks in NPE+bFGF+EGF. As a further control, fibroblasts and umbilical cells were grown in Growth Medium for the period specified for all cultures.
  • Cell Expansion. Media from all cultures were replaced with fresh media once a week and cells observed for expansion. In general, each culture was passaged one time over a period of one month because of limited growth in NPE+bFGF+EGF.
  • Immunocytochemistry. After a period of one month, all flasks were fixed with cold (4° C.) 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against TuJ1 (BIII Tubulin; 1:500; Sigma, St. Louis, Mo.) and GFAP (glial fibrillary acidic protein; 1:2000; DakoCytomation, Carpinteria, Calif.). Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Primary antibody solutions were then removed and the cultures were washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Results
  • NPE+bFGF+EGF Media Slows Proliferation of PPDCs and Alters Their Morphology. Immediately following plating, a subset of umbilical cells attached to the culture flasks coated with laminin. This may have been due to cell death as a function of the freeze/thaw process or because of the new growth conditions. Cells that did attach adopted morphologies different than those observed in Growth Media.
  • Upon confluence, cultures were passaged and observed for growth. Very little expansion took place of those cells that survived passage. At this point, very small cells with no spread morphology and with phase-bright characteristics began to appear in cultures of umbilical cells. These areas of the flask were followed over time. From these small cells, bifurcating processes emerged with varicosities along their lengths, features very similar to previously described PSA-NCAM+ neuronal progenitors and TuJ1+ immature neurons derived from brain and spinal cord (1, 2). With time, these cells became more numerous, yet still were only found in clones.
  • Clones of Umbilical Cells Express Neuronal, but not Glial Proteins. Cultures were fixed at one month post-thawing/plating and stained for the neuronal protein TuJ1 and GFAP, an intermediate filament found in astrocytes. While all control cultures grown in Growth Medium and human fibroblasts and MSCs grown in NPE+bFGF+EGF medium were found to be TuJ1−/GFAP-, umbilical cells turned on expression of TuJ1. Expression was observed in cells with and without neuronal-like morphologies. No expression of GFAP was observed in either culture. The percentage of cells expressing TuJ1 with neuronal-like morphologies was less than or equal to 1% of the total population (n=3 umbilical isolates tested).
  • Summary. Methods for generating differentiated neurons (based on TuJ1 expression AND neuronal morphology) from umbilical cells were developed. While expression for TuJ1 was not examined earlier than one month in vitro, it is clear that at least a small population of umbilicus-derived cells can give rise to neurons either through default differentiation or through long-term induction following one month's exposure to a minimal media supplemented with L-glutamine, basic FGF, and EGF.
  • References for Example 15
    • (1) Mayer-Proschel, M. et al. (1997). Neuron. 19(4): 773-85.
    • (2) Yang, H. et al. (2000). PNAS. 97(24): 13366-71.
    EXAMPLE 16 Umbilical Derived Cell Trophic Factors for Neural Progenitor Differentiation
  • The influence of umbilicus-derived postpartum cells (PPDCs) on adult neural stem and progenitor cell survival and differentiation through non-contact dependent (trophic) mechanisms was examined.
  • Materials & Methods
  • Adult Neural Stem and Progenitor Cell Isolation. Fisher 344 adult rats were sacrificed by CO2 asphyxiation followed by cervical dislocation. Whole brains were removed intact using bone rongeurs and hippocampus tissue dissected based on coronal incisions posterior to the motor and somatosensory regions of the brain (1). Tissue was washed in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement; Invitrogen), L-glutamine (4 millimolar; Invitrogen), and penicillin (50 Units/milliliter) and streptomycin (50 micrograms/milliliter) (Invitrogen), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) medium. NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.), herein referred to as NPE+bFGF+EGF.
  • Following wash, the overlying meninges were removed, and the tissue minced with a scalpel. Minced tissue was collected and trypsin/EDTA (Invitrogen) added as 75% of the total volume. DNAse (100 microliters per 8 milliliters total volume, Sigma, St. Louis, Mo.) was also added. Next, the tissue/medium was sequentially passed through an 18 gauge needle, 20 gauge needle, and finally a 25 gauge needle one time each (all needles from Becton Dickinson, Franklin Lakes, N.J.). The mixture was centrifuged for 3 minutes at 250×g. Supernatant was removed, fresh NPE+bFGF+EGF was added and the pellet resuspended. The resultant cell suspension was passed through a 40 micron cell strainer (Becton Dickinson), plated on laminin-coated T-75 flasks (Becton Dickinson) or low cluster 24-well plates (Becton Dickinson), and grown in NPE+bFGF+EGF media until sufficient cell numbers were obtained for the studies outlined.
  • PPDC Cell Plating. Postpartum-derived cells (P12) previously grown in Growth Media were plated at 5,000 cells/transwell insert (sized for 24 well plate) and grown for a period of one week in Growth Media in inserts to achieve confluence.
  • Adult Neural Progenitor Plating. Neural progenitors, grown as neurospheres or as single cells, were seeded onto laminin-coated 24 well plates at an approximate density of 2,000 cells/well in NPE+bFGF+EGF for a period of one day to promote cellular attachment. One day later, transwell inserts containing postpartum cells were added according to the following scheme:
  • (1) Transwell (umbilicus in Growth Media, 200 microliters)+neural progenitors (NPE+bFGF+EGF, 1 milliliter)
  • (2) Transwell (adult human dermal fibroblasts [1F1853; Cambrex, Walkersville, Md.] P12 in Growth Media, 200 microliters)+neural progenitors (NPE+bFGF+EGF, 1 milliliter)
  • (3) Control: neural progenitors alone (NPE+bFGF+EGF, 1 milliliter)
  • (4) Control: neural progenitors alone (NPE only, 1 milliliter)
  • Immunocytochemistry. After 7 days in co-culture, all conditions were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the epitopes listed in Table 16-1. Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
    TABLE 16-1
    Summary of Primary Antibodies Used
    Antibody Concentration Vendor
    Rat 401 (nestin) 1:200 Chemicon, Temecula, CA
    TuJ1 (BIII Tubulin) 1:500 Sigma, St. Louis, MO
    Tyrosine hydroxylase (TH)  1:1000 Chemicon
    GABA 1:400 Chemicon
    GFAP  1:2000 DakoCytomation,
    Carpinteria, CA
    Myelin Basic Protein 1:400 Chemicon
    (MBP)
  • Quantitative Analysis of Neural Progenitor Differentiation. Hippocampal neural progenitor differentiation was examined and quantified. A minimum of 1,000 cells were counted per condition, or if less, the total number of cells observed in that condition. The percentage of cells positive for a given stain was assessed by dividing the number of positive cells by the total number of cells as determined by DAPI (nuclear) staining.
  • Mass Spectrometry Analysis & 2D Gel Electrophoresis. In order to identify unique, secreted factors as a result of co-culture, conditioned media samples taken prior to culture fixation were frozen down at −80° C. overnight. Samples were then applied to ultrafiltration spin devices (nominal MW cutoff 30 kD). Retentate was subjected to immunoaffinity chromatography (anti-Human-albumin; IgY) (immunoaffinity did not remove albumin from the samples). Filtrate was analyzed by MALDI-TOFF. The eluate was applied to Cibachron Blue affinity chromatography. Samples were analyzed by SDS-PAGE and 2D gel electrophoresis.
  • Results
  • Umbilical Co-culture Stimulates Adult Neural Progenitor Differentiation. Following culture with umbilicus-derived postpartum cells, co-cultured neural progenitor cells derived from adult rat hippocampus exhibited significant differentiation along all three major lineages in the central nervous system. This effect was clearly observed after five days in co-culture, with numerous cells elaborating complex processes and losing their phase bright features characteristic of dividing progenitor cells. Conversely, neural progenitors grown alone in the absence of bFGF and EGF appeared unhealthy and survival was limited.
  • After completion of the procedure, cultures were stained for markers indicative of undifferentiated stem and progenitor cells (nestin), immature and mature neurons (TuJ1), astrocytes (GFAP), and mature oligodendrocytes (MBP). Differentiation along all three lineages was confirmed while control conditions did not exhibit significant differentiation as evidenced by retention of nestin-positive staining amongst the majority of cells.
  • The percentage of differentiated neural progenitors following co-culture with umbilicus-derived PPDCs was quantified (Table 16-2). Umbilicus-derived cells significantly enhanced the number of mature oligodendrocytes (MBP) (24.0% vs 0% in both control conditions). Furthermore, co-culture enhanced the number of GFAP+ astrocytes and TuJ1+ neurons in culture (47.2% and 8.7% respectively). These results were confirmed by nestin staining indicating that progenitor status was lost following co-culture (13.4% vs 71.4% in control condition 3).
  • Though differentiation also appeared to be influenced by adult human fibroblasts, such cells were not able to promote the differentiation of mature oligodendrocytes nor were they able to generate an appreciable quantity of neurons. Though not quantified, fibroblasts did, however, appear to enhance the survival of neural progenitors and their progeny similar to findings for umbilicus-derived postpartum cells.
    TABLE 16-2
    Quantification of progenitor differentiation in control
    vs transwell co-culture with umbilicus-derived
    postpartum cells (E = EGF, F = bFGF).
    F + E/Umb F + E/F + E F + E/removed
    Antibody [Cond. 1] [Cond. 3] [Cond. 4]
    TuJ1  8.7%  2.3%  3.6%
    GFAP 47.2% 30.2% 10.9%
    MBP 23.0%   0%   0%
    Nestin 13.4% 71.4% 39.4%
  • Identification of Unique Compounds. Conditioned media from umbilical test conditions along with the appropriate controls (NPE media ±1.7% serum, media from co-culture with fibroblasts) were examined for differences. Potentially unique compounds were identified and excised from their respective 2D gels.
  • Summary. Results presented in this example indicate that the differentiation of adult neural progenitor cells following co-culture with umbilicus-derived postpartum cells is particularly profound. Specifically, a significant percentage of mature oligodendrocytes were generated in co-cultures of umbilical cells. In view of the lack of contact between umbilical cells and the neural progenitors, this result appears to be a function of soluble factors released from the umbilical cells (trophic effect).
  • Several other observations were made. First, there were very few cells in the control condition where EGF and bFGF were removed. Most cells died and on average, there were about 100 cells or fewer per well. Second, it is to be expected that there would be very little differentiation in the control condition where EGF and bFGF was retained in the medium throughout, since this is normally an expansion medium. While approximately 70% of the cells were observed to retain their progenitor status (nestin+), about 30% were GFAP+ (indicative of astrocytes). This may be due to the fact that such significant expansion occurred throughout the course of the procedure that contact between progenitors induced this differentiation. Similar findings have been reported in the literature (2).
  • References for Example 16
    • (1) Paxinos, G. & Watson, C. (1997). The Rat Brain in Stereotaxic Coordinates.
    • (2) Song, H. et al. (2002). Nature. 417(6884): 29-32.
    EXAMPLE 17 The Effect of Trophic Factors on Angiogenesis
  • Angiogenesis, or the formation of new vasculature, is necessary for the growth of new tissue. Induction of angiogenesis is an important therapeutic goal in many pathological conditions. The angiogenic activity of umbilicus-derived cells in in vitro assays was examined. A well-established method of assessing angiogenic activity involving seeding endothelial cells onto a culture plate coated with a basement membrane extract (Nicosia and Ottinetti (1990) In Vitro Cell Dev. Biol. 26(2): 119-28), was utilized. Treating endothelial cells on such basement membranes or extracellular matrix material with angiogenic factors will stimulate the cells to form a network that is similar to capillaries. These types of assays are common in vitro assays for testing stimulators and inhibitors of blood vessel formation (Ito et al. (1996) Int. J. Cancer 67(1): 148-52). The protocols utilized in this example made use of a co-culture system with the umbilicus-derived cells seeded onto culture well inserts. These permeable inserts allow for the passive exchange of media components between the endothelial and the umbilicus-derived cell culture media.
  • Material & Methods
  • Cell Culture.
  • Umbilicus-derived cells. Human umbilical cords were received and cells were isolated as previously described (Example 1). Cells were cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C. with 5% CO2. Cells used for experiments were between passages 4 and 12.
  • Actively growing UDCs were trypsinized, counted, and seeded onto 6.5 millimeter diameter tissue culture inserts (COSTAR TRANSWELL, Corning Inc., Corning, N.Y.) at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium with 5% CO2 at 37° C.
  • Human mesenchymal stem cells (hMSC). hMSCs were purchased from Cambrex (Walkersville, Md.) and cultured in MSCGM (Cambrex). The cultures were incubated with 5% CO2 at 37° C.
  • Actively growing MSCs were trypsinized and counted and seeded onto 6.5 millimeter diameter tissue culture inserts (Corning, Corning, N.Y.) at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium with 5% CO2 at 37° C.
  • Human umbilical vein endothelial cells (HUVEC). HUVEC were obtained from Cambrex (Walkersville, Md.). Cells were grown in separate cultures in either EBM or EGM endothelial cell media (Cambrex). Cells were grown on standard tissue cultured plastic with 5% CO2 at 37° C. Cells used in the assay ranged from passages 4 to 10.
  • Human coronary artery endothelial cells (HCAEC). HCAEC were purchased from Cambrex Incorporated (Walkersville, Md.). These cells were also maintained in separate cultures in either the EBM or EGM media formulations. Cells were grown on standard tissue cultured plastic with 5% CO2 at 37° C. Cells used for experiments ranged from passages 4 to 8.
  • Angiogenesis Assays on Extracellular Matrix. Culture plates were coated with extracellular matrix material according to manufacturer's specifications. Briefly, extracellular matrix material (MATRIGEL, BD Discovery Labware, Bedford, Mass.) was thawed at 4° C. and approximately 250 microliters were distributed onto each well of a chilled 24-well culture plate (Corning). The plate was then incubated at 37° C. for 30 minutes to allow the material to solidify. Actively-growing endothelial cell cultures were trypsinized and counted. Cells were washed twice in Growth Medium supplemented with only 2% FBS by centrifugation, resuspension, and aspiration of the supernatant. Cells were seeded onto the coated wells 20,000 cells per well in approximately 0.5 milliliter Growth Medium supplemented with only 2% (v/v) FBS. Cells were then incubated for approximately 30 minutes to allow the cells to settle.
  • Endothelial cell cultures were then treated with either 10 nanomolar human bFGF (Peprotech, Rocky Hill, N.J.) or 10 nanomolar human VEGF (Peprotech, Rocky Hill, N.J.) to serve as positive controls for endothelial cell response. Transwell inserts seeded with postpartum cells were added to appropriate wells with Growth Medium supplemented with only 2% FBS in the insert chamber. Cultures were incubated in 5% CO2 at 37° C. for approximately 24 hours. The well plate was removed from the incubator, and images of the endothelial cell cultures were collected with an Olympus inverted microscope (Olympus, Melville, N.Y.).
  • Results
  • In a co-culture system with umbilicus-derived cells, HUVEC form structured cell networks. HUVEC cells form limited cell networks in co-culture experiments with hMSC and with 10 nanomolar bFGF. HUVEC cells without any treatment showed very little or no network formation. These results suggest that the umbilicus-derived cells release angiogenic factors that stimulate the HUVEC. Similarly, HCAECs formed cell networks only in co-culture with umbilicus-derived cells.
  • Table 17-1 shows quantities of known angiogenic factors released by UDCs in Growth Medium at atmospheric oxygen conditions. UDCs were seeded onto inserts as described above. The cells were cultured at 37° C. in atmospheric oxygen for 48 hours on the inserts and then switched to a 2% FBS media and returned at 37° C. for 24 hours. Medium was removed, immediately frozen, and stored at −80° C., and analyzed by the SearchLight multiplex ELISA assay (Pierce Chemical Company, Rockford, Ill.). Results shown are the averages of duplicate measurements. The results show that UDCs do not release detectable levels of platelet-derived growth factor-bb (PDGFbb), heparin-binding epidermal growth factor (HB-EGF), or vascular endothelial growth factor (VEGF). The amounts of angiopoietin 2 (ANG2) detected were less than that of the culture medium control with no cells. The umbilicus-derived cells released measurable quantities of tissue inhibitor of metallinoprotease-1 (TIMP-1), thrombopoietin (TPO), and hepatocyte growth factor (HGF). The amounts of keratinocyte growth factor (KGF) and fibroblast growth factor (FGF) were very low and only slightly above those for the control medium.
    TABLE 17-1
    Potential angiogenic factors released from UDCs.
    Umbilicus-derived cells were cultured in 24 hours in media with 2% FBS in
    atmospheric oxygen. Medium was removed and assayed by the SearchLight
    multiplex ELISA assay (Pierce). Results are the means of a duplicate analysis.
    Values are concentrations in the media reported in picograms per milliliter of
    culture media.
    TIMP1 ANG2 PDGFBB TPO KGF HGF FGF VEGF HBEGF
    (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL)
    UDCs (P4) 81831.7 <9.8 <2.0 365.9 14.1 200.2 5.8 <4.0 <1.2
    Media alone <9.8 25.1 <2.0 <6.4 <2.0 <3.2 <5.4 <4.0 <1.2
  • Table 17-2 shows levels of known angiogenic factors released by UDCs at 5% O2. UDCs were seeded onto inserts as described above. The cells were cultured in Growth Medium at 5% oxygen for 48 hours on the inserts and then switched to a 2% FBS medium and returned to 5% O2 incubation for 24 hours. Medium was removed, immediately frozen, and stored at −80° C., and analyzed by the SearchLight multiplex ELISA assay (Pierce Chemical Company, Rockford, Ill.). Results shown are the averages of duplicate measurements. The results show for UDCs are comparable to those under atmospheric oxygen conditions. While there are essentially no changes in the production of ANG2, PDGFBB, FGF, VEGF, and HB-EGF by UDCs, there was a slight increase apparent in production of TIMP1, KGF, and HGF, and a slight decrease apparent in production of TPO. These apparent differences in raw data were not tested for statistical significance.
    TABLE 17-2
    Potential angiogenic6/39 factors released from UDCs. Cells
    were cultured in 24 hours in media with 2% FBS in 5% oxygen. Medium was
    removed and assayed by the SearchLight multiplex ELISA assay (Pierce).
    Results are the means of a duplicate analysis. Values are concentrations in the
    media reported in picograms per milliliter of culture media.
    TIMP1 ANG2 PDGFBB TPO KGF HGF FGF VEGF HBEGF
    (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL)
    UDCs (P4) 50244.7 <9.8 <2.0 403.3 10.7 156.8 5.7 <4.0 <1.2
    Media alone <9.8 25.1 <2.0 <6.4 <2.0 <3.2 <5.4 <4.0 <1.2
  • Summary. The results of the study show that umbilicus-derived cells can stimulate both human umbilical vein and coronary artery endothelial cells to form networks in an in vitro assay of angiogenesis. This effect is similar to that seen with known angiogenic factors in such assay systems. These results suggest that UDCs are useful for stimulating angiogenesis in vivo.
  • EXAMPLE 18 Differentiation of Umbilicus-Derived Cells into Hepatocytes
  • A variety of conditions were examined to determine a suitable combination of basic media and growth factors for the differentiation of umbilicus-derived cells into hepatocytes. HNF-1alpha, a hepatocyte-specific transcription factor, cytoplasmic intermediate filament proteins, such as keratin 19 (K19), keratin 8 (K8), and cytokeratin 18 (CK18), which are markers of epithelial cells and two liver-specific secreted proteins, albumin and cytochrome p450 2B6, were selected as markers for hepatocyte differentiation (Schwartz et al. (2002) J. Clin. Invest. 109(10):1291-1302; Okumoto et al. (2003) Biochem. Biophys. Res. Commun. 304(4):691-695; Chargracui et al. (2003) Blood 101(8): 2973-2982).
  • Methods & Materials
  • Umbilicus-derived cells obtained according to method of Example 1, as well as neonatal or adult Normal Human Dermal Fibroblasts (NHDF), were grown in Growth Medium in gelatin-coated T75 flasks. Basic Fibroblast Growth Factor (bFGF), Oncostatin M, Hepatocyte Growth Factor (HGF), Stem Cell Factor (SCF), and Fibroblast Growth Factor 4 (FGF 4) were from PeproTech Inc. (Rocky Hill, N.J.). Platelet Derived Growth Factor BB (PDGFbb) was from R&D Systems (Minneapolis, Minn.).
  • The following conditions were tested:
  • Method 1
  • Umbilicus-derived cells (P5), neonatal and adult Normal Human Dermal Fibroblasts (NHDF). Cells were plated at 22.5×103 cells/cm2 on 1% Matrigel (Becton-Dickinson and Co., Franklin Lakes, N.J.) in serum-free medium (60% (v/v) low glucose DMEM) (DMEM-LG; Gibco, Carlsbad, Calif.), 40% (v/v) MCDB-201 (Sigma, St. Louis, Mo.), supplemented with 1× insulin/transferrin/selenium, 4.7 micrograms/milliliter linoleic acid, 1 milligram/milliliter bovine serum albumin, 10 nanomolar Dexamethasone, 100 micromolar ascorbic acid phosphate (all from Sigma), 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco), 2% (v/v) FCS (Hyclone Laboratories, Logan, Utah), and 10 nanograms/milliliter each EGF and PDGFbb). After 8-12 hours, medium was removed, cells were washed twice with PBS (Gibco) and cultured in the above-described medium without EGF and PDGFbb but supplemented with 20 nanograms/milliliter HGF and/or 10 nanograms/milliliter FGF-4 (Schwartz et al. (2002) J. Clin. Invest. 109(10):1291-1302).
  • Method 2
  • Umbilicus-derived cells (P5), neonatal and adult NHDF. Cells were seeded at lower density (22,500 cells/cm2) in 24-well plates coated with gelatin and grown as described above.
  • Method 3
  • Umbilicus-derived cells (P17), Umbilicus-derived cells (P15), Umbilicus-derived cells (P10), adult NHDF. Cells were seeded at higher density (50,000 cells/cm2) in 24-well TCP plates and grown in DMEM (Gibco), B27 Supplement (Gibco), penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 20 nanograms/milliliter HGF and/or 10 nanograms/milliliter FGF-4. Cells were grown in these conditions for 4 weeks.
  • Method 4
  • Umbilicus-derived cells (P4), Umbilicus-derived cells (P9), neonatal and adult NHDF. Cells were seeded at a density of 5,000 cells/cm2 in T25 flasks in Chang C medium (Irvine Scientific, Santa Ana, Calif.) on either fibronectin (PeproTech, Rocky Hill, N.J.) or gelatin (Sigma) and grown for two passages until confluence. Cells were then seeded at 1,000 cells/cm2 in 24-well TCP plates and grown as described above until they reached about 40-60% confluence.
  • Method 5
  • Umbilicus-derived cells (P5) and adult NHDF. Cells were plated in 24-well plates on gelatin in Growth Medium supplemented with either 1 nanogram/milliliter or 10 nanograms/milliliter oncostatin M (Chargracui (2003) Blood 101(8): 2973-2982). Cells were grown in these conditions for 4 weeks.
  • Method 6
  • Umbilicus-derived cells (P5) and adult NHDF. Cells were plated in 24-well plates on gelatin in Growth Medium supplemented with 10 nanograms/milliliter bFGF, 10 nanograms/milliliter HGF, 10 nanograms/milliliter SCF. Cells were grown in these conditions for 4 weeks (Okumoto et al. (2003) Biochem. Biophys. Res. Commun. 304(4):691-695.).
  • Total RNA isolation and quantitative RT-PCR. RNA was extracted from umbilicus-derived cells and fibroblasts grown as described in each protocol. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit, Qiagen, Valencia, Calif.) and RNA extracted according to the manufacturer's instructions (RNeasy Mini Kit, Qiagen, Valencia, Calif.) with a 2.7 Units/sample DNase treatment (Sigma). RNA was eluted with 50 microliters DEPC-treated water and stored at −80° C. RNA was reverse transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C. for 10 minutes, 37° C. for 60 minutes, and 95° C. for 10 minutes. Samples were stored at −20° C.
  • Real-time PCR. PCR was performed on cDNA samples using ASSAYS-ON-DEMAND gene expression products for albumin (Hs00609411), cytochrome p450 2B6 (Hs00167937), GAPDH (Applied Biosystems, Foster City, Calif.) and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minute. PCR data were analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).
  • Immunofluorescence. Cell cultures were fixed with cold (4° C.) 4% (w/v) paraformaldehyde for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the following epitopes: keratin 9 (K19; 1:400; Chemicon, Temecula, Calif.), keratin 19 (K19; 1:400; Chemicon), cytokeratin 18 (CK18; 1:400; Sigma, St. Louis, Mo.), vimentin (1:500; Sigma), desmin (1:150; Sigma), albumin (1:200; Sigma), c-met (1:400; Santa Cruz Biotech, Santa Cruz, Calif.), and HNF-1alpha (1:400; Santa Cruz Biotech). In general, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100, Sigma) for 30 minutes to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (e.g. c-met), Triton was omitted in all steps of the procedure in order to prevent epitope loss. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Primary antibody solutions were then removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) for K8, K19, CK18, vimentin, and albumin, goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) for desmin and c-met, or donkey anti-goat IgG-FITC (1:150; Santa Cruz Biotech) for HNF-1alpha staining. Cultures were washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Results
  • In order to determine whether umbilicus-derived cells could express epithelial markers, cells were cultured in Chang C medium. Umbilicus-derived cells (P2) were grown in Chang C medium for 11 days. Umbilicus-derived cells stained negative for cytokeratin 18 and keratin 8 by immunocytochemistry analysis. Samples grown in Growth Medium were negative for both markers.
  • The effect of passage, as well as gelatin and fibronectin substrata was investigated. Cells were grown in Chang C medium for 11 days. RNA and protein expression of epithelial/hepatocyte-specific proteins were analyzed. Immunocytochemistry staining for cytokeratin18, keratin 8, keratin 19, c-met, albumin, desmin, and HNF-1alpha were negative in all conditions. Cells stained positive for vimentin. Expression of both albumin and cytochrome p450 2B6 at levels lower than that of human HepG2 cells was detected with ASSAYS-ON-DEMAND primers. Albumin and cytochrome p450 2B6 expression were also detected in cells grown in Growth Medium.
  • Umbilicus-derived cells were treated as described in method 1 according to a protocol developed by Schwartz et al. (2002) J. Clin. Invest. 109(10):1291-1302.). Both albumin and cytochrome p450 2B6 were detected with ASSAYS-ON-DEMAND primers at levels lower than HepG2 positive control. No clear pattern emerged between conditions applied and gene expression levels, i.e., albumin and cytochrome p450 2B6 expression were each also detected in control samples. Some expression of albumin and cytochrome p450 2B6 was detected with ASSAYS-ON-DEMAND primers, however, the levels were significantly lower than those observed in HepG2 cells.
  • Oncostatin M at low concentration of 1 nanogram/milliliter increased expression levels of cytochrome p450 2B6 in umbilicus-derived cells grown in Growth Medium on gelatin-coated flasks (data not shown). FGF-4 and HGF treatment had little effect and may have reduced the expression of albumin and cytochrome p450 2B6.
  • Summary. Six protocols were tested for their ability to induce differentiation of umbilicus-derived cells to hepatocyte phenotype. Expression of hepatocyte-specific markers, such as albumin and cytochrome p450 2B6 was detected, thereby indicating that the cells underwent some differentiation into hepatocytes.
  • EXAMPLE 19 Adipogenic Differentiation of Umbilicus-Derived Cells
  • Populations of stem cells have been demonstrated to differentiate into an adipogenic phenotype (Janderova et al. (2003) Obes. Res. 11(1):65-74; Zangani et al. (1999) Differentiation 64(2):91-101; Liu et al. (2003) Curr. Mol. Med. 3(4):325-40). The potential of umbilicus-derived cells to differentiate into an adipogenic phenotype was evaluated.
  • Methods & Materials
  • Adipose differentiation. Umbilicus-derived cells (P4) were seeded at 200,000 cells per well on 6-well tissue culture-treated plates in Growth Medium. Mesenchymal stem cells (P3, IF2155), osteoblasts (P5, CC2538; Cambrex, Walkerville, Md.), omental cells (P6) (isolated from omental tissue from NDRI, following protocol used for postpartum-derived cell isolation in Example 1), adipose-derived cells (U.S. Pat. No. 6,555,374 B1) (P6), and fibroblasts (P6, CC2509) (Cambrex, Walkerville, Md.) were also seeded under the same conditions. Prior to initiation of adipogenesis, Mesenchymal Stem Cells were grown in a Mesenchymal Stem Cell Growth Medium Bullet kit (Cambrex, Walkerville, Md.). After 2 days, spent medium was aspirated off and cells were washed with phosphate buffered saline (PBS). Culture medium was then switched to Dulbecco's Minimal Essential Medium-high glucose (DMEM-Hg; Invitrogen, Carlsbad, Calif.) containing 10 percent FBS (v/v, Hyclone, Logan Utah), 0.02 milligrams insulin per milliliter (Sigma, St. Louis, Mo.), and 100 Units penicillin per milliliter, 100 milligrams streptomycin per milliliter, 0.25 micrograms amphotericin B per milliliter; (Invitrogen, Carlsbad, Calif.). Once the cells had reached confluence, spent medium was aspirated. Cells were then cultured in an adipose differentiation medium (DMEM-Hg (Invitrogen, Carlsbad, Calif.), containing 10 percent defined fetal bovine serum ((v/v), Hyclone, Logan, Utah), 0.02 milligrams per milliliter insulin (Sigma, St. Louis, Mo.) and 100 units penicillin per milliliter, 100 micrograms streptomycin per milliliter, 0.25 micrograms amphotericin B per milliliter, 5 micromolar isobutylmethylxanthine (Sigma, St. Louis, Mo.), 100 micromolar dexamethasone (Sigma, St. Louis, Mo.), and 2.5 micromolar indomethacin (Sigma, St. Louis, Mo.) for up to 4 weeks. Cells were stained with Oil-Red-O to determine the presence of lipid droplet formation.
  • Oil Red 0 Staining. Cells were fixed with 10 percent (v/v) neutral buffered formalin (Richard-Allan Kalamazoo, Mich.). After fixation, the cells were washed in deionized water and incubated for two minutes in propylene glycol (absolute; Poly Scientific, Bay Shore, N.Y.). Propylene glycol was removed by aspiration, and samples were incubated in Oil Red O (Poly Scientific) for one hour. Staining solution was removed by aspiration and stained samples were then incubated in 85 percent (v/v) propylene glycol solution (Poly Scientific) for one minute. Stained samples were washed with two changes of de-ionized water. Stained samples were counter-stained with Mayer's Hematoxylin (Poly Scientific) and examined with light microscopy. Images were taken at magnification of 20×.
  • Leptin Assay. Adipose-derived cells and umbilicus-derived cells were seeded at 200,000 cells/well in 6-well tissue culture-treated plates. Cells were initially seeded in Growth Medium, which was changed to an adipogenic differentiation medium (DMEM-Hg medium; Invitrogen, Carlsbad, Calif.) containing 1 micromolar dexamethasone (Sigma, St. Louis, Mo.), 0.2 millimolar indomethasone (Sigma), 0.01 milligrams per microliter insulin (Sigma), 0.5 millimolar isobutylmethylxanthine (Sigma), 10 percent (v/v) fetal bovine serum (Cat. #SH30070.03; Hyclone, Logan, Utah), 100 Units penicillin per milliliter and 100 micrograms streptomycin per milliliters (Gibco)). At the end of the assay, the conditioned medium was collected and leptin levels were measured using an ELISA kit (Quantikine, R&D Systems, Minneapolis, Minn.).
  • Results
  • Adipose differentiation. Morphologically MSCs and adipose-derived cells demonstrated lipid formation as early as 5 days in this assay. Large amounts of lipid droplet formation were observed in these cultures by 15 days of culture. Cultures of osteoblasts also deposited large amounts of lipid under these conditions after 10 days in culture and extensively at 15 days. Lipid droplet formation was observed in umbilicus-derived and omental cell cultures after 15 days of culture. Low level lipid droplet formation was observed in the fibroblast cultures after 20 days in adipogenic-inducing conditions.
  • Leptin. Leptin was not detected by ELISA in umbilicus-derived cell conditioned medium.
  • Summary. While leptin was not detected in umbilicus-derived cells by ELISA following the adipogenic differentiation protocols used, the data clearly demonstrate that umbilicus-derived cells undergo a low level of differentiation to an adipocyte phenotype when compared to cultures of mesenchymal stem cells, adipose-derived cells, or osteoblasts.
  • EXAMPLE 20 Differentiation into Beta Cell Phenotype
  • The pancreas contains endocrine cells, organized in islets of Langerhans, that produce insulin, glucagon, somatostatin, and pancreatic polypeptide (PP). The ability of umbilicus-derived cells to differentiate towards cells with an insulin-producing phenotype was tested under eight different induction protocols.
  • Methods & Materials
  • Umbilicus-derived cells (various isolates—see below) as well as neonatal or adult Normal Human Dermal Fibroblasts (NHDF) grown in Growth Medium, in gelatin-coated T75 flasks, as well as in different beta-cell promoting differentiation conditions. Flasks were coated with 2% (w/v) gelatin solution (Sigma, St. Louis, Mo.) for 20 minutes at room temperature. Gelatin solution was aspirated off and flasks were washed with PBS. Basic Fibroblast Growth Factor (bFGF), Epidermal Growth Factor (EGF), Transforming Growth Factor alpha (TGFalpha) and Fibroblast Growth Factor 10 (FGF-10) were purchased from PeproTech Inc. (Rocky Hill, N.J.). GLP-1 was purchased from Sigma (St. Louis, Mo.)
  • The following protocols were tested:
  • Protocol 1:
  • Cells: Adipose-derived cells (U.S. Pat. No. 6,555,374) and omentum-derived cells, umbilicus-derived cells, (P15), (P17), (P3), and adult Normal Human Dermal Fibroblasts (NHDF) (P10) were utilized. Cells were maintained under either normal or 5% O2 conditions. Cells were seeded at low density (5,000 cells/cm2) in gelatin-coated T75 flasks on gelatin and grown in Ham's F12 medium (Clonetics, Santa Rosa, Calif.), 2% (v/v) FBS, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF until confluence. Confluent cells were trypsinized and plated at 50,000 cells/cm2 in 24-well Tissue Culture Polystyrene (TCPS; BD Biosciences, Bedford, Mass.) plates with or without gelatin or collagen coating. Cells were grown in Ham's F12 medium, 2% FBS, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF and 15 nanomolarGLP-1 (7-37 isoform) for up to 3 weeks.
  • Protocol 2:
  • Cells: Umbilicus-derived cells, isolate 2 (P17), isolate 1 (P15), isolate 4 (P10) and adult NHDF P10 were utilized. Cells were seeded at 50,000 cells/cm2 in 24-well TCP plates and grown in DMEM:Ham's F12 (1:1) medium, B-27 supplement (Gibco, Carlsbad, Calif.), 50 units of penicillin per milliliter, 50 milligrams streptomycin per milliliter, 20 nanograms/milliliter EGF, 40 nanograms/milliliter bFGF spherical clusters were generated—usually 4-6 days. Following that period, the spherical clusters were collected, centrifuged, and replated onto laminin-coated, 24-well plates (BD Biosciences, Bedford, Mass.), and cultured up to 3 weeks in B-27-supplemented medium containing 10 nanomolarGLP-1 (7-37) but no other growth factors (i.e. no bFGF and no EGF).
  • Protocol 3:
  • Cells: Umbilicus-derived cells, isolate 2 (P17) isolate 1 (P15), isolate 4 (P10), and adult NHDF (P10) were utilized. Cells were seeded at high density (50,000 cells/cm2) in 24-well TCPS plates and grown in DMEM:Ham's F12 (1:1) medium, B-27 supplement, P/S, 20 nanograms/milliliter EGF, 40 nanograms/milliliter bFGF spherical clusters were generated—usually 4-6 days. Following that period, the spherical clusters were collected, centrifuged, and replated onto laminin-coated, 24-well plates and cultured up to 3 weeks in B-27-supplemented medium containing 10 nanomolarGLP-1 (1-37 isoform) but no other growth factors (i.e. no bFGF and no EGF).
  • Protocol 4:
  • Cells: Adult NHDF (P15), umbilicus-derived, isolate 1 (P18), isolate 2 (P21), isolate 3 (P5), isolate 3 (P4), were isolated according to the method by Mitchell et al. (2). Cells were seeded at 50,000 cells/cm2 in 24-well TCPS gelatin-coated plates and grown in DMEM:Ham's F12 (1:1) medium, B-27 supplement, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter FGF-10, and/or 40 nanograms/milliliter TGF alpha for more than two weeks.
  • Protocol 5:
  • Cells: Adult NHDF, umbilicus-derived, isolate 1 (P18), isolate 2 (P21) isolate 3 (P5), isolate 3 (P4), were isolated according to the method by Mitchell et al. (2). Cells were seeded at 50,000 cells/cm2 in 24-well TCPS gelatin-coated plates and grown in EBM-2 medium, 10 nanograms/milliliter FGF-10, and/or 40 nanograms/milliliter TGF alpha for greater than two weeks.
  • Protocol 6:
  • Cells: Umbilicus-derived, isolate 3 (P2) were utilized. Cells were seeded at 5,000 cells/cm2 in T75 flasks on gelatin and grown either in Growth Medium or in Ham's F12 medium, 2% FBS, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF until confluence. Confluent cells were trypsinized and plated at 50,000 cells/cm2 in 24-well TCPS plates, with or without gelatin coating. Three types of basic media were used for up to 3 weeks:
  • Beta I medium: Ham's F12 medium, 2% FBS, 10 millimolar nicotinamide, penicillin (50 Units/milliliter), streptomycin (50 micrograms/milliliter), 25 millimolar glucose;
  • Beta II medium: Equal parts of DMEM/Ham's F12 media, 2% FBS, 10 millimolar nicotinamide, 25 millimolar glucose; and
  • Endothelial Cell Basal Medium (EBM), (Clonetics, Santa Rosa, Calif.).
  • The following growth factors were added to each of the media: 10 nanograms/milliliter EGF, 20 nanograms/milliliter bFGF, 10 nanomolar GLP-1 (7-37 isoform).
  • Total RNA isolation and quantitative RT-PCR. RNA was extracted from umbilicus-derived cells and fibroblasts grown as described in each protocol. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini kit, Qiagen, Valencia, Calif.) and RNA extracted according to the manufacturer's instructions (RNeasy Mini kit, Qiagen, Valencia, Calif.) with a 2.7 U/sample DNase treatment (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at −80° C. RNA was reversed transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C. for 10 minutes, 37° C. for 60 minutes and 95° C. for 10 minutes. Samples were stored at −20° C.
  • Real-time PCR. PCR was performed on cDNA samples using ASSAYS-ON-DEMAND gene expression products. PDX-1 (Hs00426216), pro-insulin (Hs00355773), Ngn-3 (Hs00360700) and Glut-2 (Hs00165775) GAPDH (Applied Biosystems, Foster City, Calif.) and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minutes. In addition, another set of primers designed in-house for PDX-1 and Ngn-3 were tested. Table 20-1 contains primer sequences. PCR using these primers was performed as described above. Pancreas total RNA (Ambion, Austin, Tex.) was used as control. PCR data were analyzed according to the ΔΔCT method recommended by Applied Biosystems (1).
    TABLE 20-1
    Primer name Sequence
    PDX-1 Forward 5′-CTGGATTGGCGTTGTTTGTG-3′
    primer (SEQ ID NO:11)
    PDX-1 Reverse 5′-TCCCAAGGTGGAGTGCTGTAG-3′
    primer (SEQ ID NO:12)
    PDX-1-TaqMan 5′-CTGTTGCGCACATCCCTGCCC-3′
    probe (SEQ ID NO:13)
    Ngn-3 Forward 5′-GGCAGTCTGGCTTTCTCAGATT-3′
    primer (SEQ ID NO:14)
    Ngn-3 Reverse 5′-CCCTCTCCCTTACCCTTAGCA-3′
    primer (SEQ ID NO:15)
    Ngn-3 TaqMan 5′-CTGTGAAAGGACCTGTCTGTCGC-3′
    probe (SEQ ID NO:16)
  • Immunofluorescence. Adult human pancreatic tissue was harvested and immersion fixed in 4% (w/v) paraformaldehyde (Sigma, St. Louis, Mo.) overnight at 4° C. Immunohistochemistry was performed using antibodies directed against the following epitopes: insulin (insulin serum; 1:50; LINCO Research, St. Charles, Mo.), PDX-1 (1:50; Santa Cruz Biotech, Santa Cruz, Calif.), glucagon (1:100; Santa Cruz Biotech), somatostatin (1:100; DAKOCytomation, Carpinteria, Calif.), and cytokeratin 18 (CK18; 1:400; Sigma, St. Louis, Mo.). Briefly, fixed specimens were blocked off with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, Calif.) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 microns thick) using a standard cryostat (Leica Microsystems), and mounted onto glass slides for staining.
  • Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100, Sigma) for 1 hour. Primary antibodies, diluted in blocking solution, were then applied to the samples for a period of 4 hours at room temperature. Primary antibodies solutions were removed and samples washed with PBS prior to application of secondary antibody solutions for 1 hour at room temperature containing blocking solution along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, Oreg.) for CK18, goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) for glucagon and somatostatin, goat anti-guinea pig IgG-FITC (1:150; Santa Cruz Biotech) for insulin, or donkey anti-goat IgG-FITC (1:150; Santa Cruz Biotech) for PDX-1 staining. Samples were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N.Y.). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Results
  • For umbilicus-derived cells treated according to protocols 1-6, expression of pancreas-specific marker was not detected using real-time PCR and the ASSAYS-ON-DEMAND primers, with the exception that low levels of Ngn-3 were detected in cells from protocol 6. The same primers produced positive results with cDNA derived from pancreatic tissue RNA.
  • Real-time PCR for ngn-3 was performed on cDNA samples derived from human umbilical cord grown according to protocol 6. PCR was also performed using ASSAYS-ON-DEMAND Ngn-3 primers (Hs00360700). Human pancreas-derived cDNA was used as control. No other pancreas-specific markers (PDX-1, pro-insulin or Glut-2) were detected with the ASSAYS-ON-DEMAND primers.
  • Experimental conditions in Protocols 2 and 6 applied to umbilicus-derived tissues, but not fibroblasts, produced structures resembling the cellular assembly of pancreatic epithelial cells into islets. These structures emerged 3-5 days after the implementation of the protocol. However unlike islets, these structures were negative for pancreatic markers (PDX-1, Ngn3, Glut-2 and pro-insulin) expression (tested by real-time PCR).
  • Pancreas-specific markers were detected in tissue derived from a human pancreas using immunofluorescence technique and an array of antibodies (see materials and methods). The expression of pancreas-specific markers (e.g. insulin, PDX-1, glucagon, somatostatin, and cytokeratin 18) in human pancreatic tissue was readily detectable.
  • Summary. Limited expression of PDX-1 and Ngn-3 has been observed in umbilicus-derived cells treated with a variety of experimental protocols. There were differences in results between in-house designed and commercially available primers. For example, while protocol number 1 gave positive data for PDX-1 and Ngn-3 using in-house designed primers, ASSAYS-ON-DEMAND primers for the same genes produced negative data. The results were not directly verified by immunological techniques. Notwithstanding such differences, expression of several pancreatic markers has been accomplished suggesting the potential of umbilicus-derived cells to differentiate towards the pancreatic phenotypes.
  • References for Example 20
    • 1. User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System
    • 2. Mitchell K E, Weiss M L, Mitchell B M, Martin P, Davis D, Morales L, Helwig B, Beerenstrauch M, Abou-Easa K, Hildreth T, Troyer D, Medicetty S. (2003) Matrix cells from Wharton's jelly form neurons and glia. Stem Cells 21(1):50-60.
    • 3. H. Edlund. (2002) Pancreatic organogenesis—developmental mechanisms and implications for therapy. Nat. Rev. Genet. 3:524-532.
    • 4. S. K. Kim and M. Hebrok. (2001) Intercellular signals regulating pancreas development and function. Genes Dev. 15:111-127.
    • 5. Street C N, Rajotte R V, Korbutt G S. (2003) Stem cells: a promising source of pancreatic islets for transplantation in type 1 diabetes. Curr Top Dev Biol. 58:111-36.
    EXAMPLE 21 Chondrogenic Differentiation of Umbilicus-Derived Cells
  • Cartilage damage and defects lead to approximately 600,000 surgical procedures each year in the United States alone (1). A number of strategies have been developed to treat these conditions but these have had limited success. One approach, Cartecel (Genzyme), uses autologous chondrocytes that are collected from a patient, expanded in vitro and then implanted into the patient (1). This approach has the disadvantage of collecting healthy cartilage and requiring a second procedure to implant the cultured cells. An alternative possibility is a stem cell-based therapy in which cells are placed at or near the defect site to directly replace the damaged tissue. Cells may be differentiated into chondrocytes prior to the application or progenitor cells that can differentiate in situ may be used. Such transplanted cells would replace the chondrocytes lost in the defect.
  • Candidate cells for this indication should be evaluated for their ability to differentiate into chondrocytes in vitro. A number of protocols have been developed for testing the ability of cells to differentiate and express chondrocyte marker genes. Umbilicus-derived cells were tested for their ability to differentiate into chondrocytes in vitro in two different assay systems: the pellet assay culture system and collagen gel cultures. The pellet culture system has been used successfully with selected lots of human mesenchymal stem cells (MSC). MSC grown in this assay and treated with transforming growth factor-beta3 have been shown to differentiate into chondrocytes (2). The collagen gel system has been used to culture chondrocytes in vitro (3). Chondrocytes grown under these conditions form a cartilage-like structure.
  • Materials and Methods
  • Cell Culture. Human umbilical cords were received and umbilicus-derived cells were prepared as described in Example 1 above. Cells were cultured in Growth Medium on gelatin-coated TCP flasks. The cultures were incubated with 5% CO2 at 37° C. Cells used in experiments ranged from passages 4 through 12.
  • Human articular chondrocytes were purchased from Cambrex (Walkersville, Md.) and cultured in the same medium as the postpartum cells. Twenty-four hours before the experiment, the culture medium was changed to a media containing 1% FBS.
  • Human mesenchymal stem cells (hMSCs) were purchased from Cambrex (Walkersville, Md.) and cultured in MSCGM (Cambrex). Cells used for experiments were between passages 2 and 4.
  • Collagen gel assays. Cultured cells were trypsinized to remove from culture plate. Cells were washed with centrifugation twice at 300×g for 5 minutes in DMEM without serum and counted. Cells were mixed with the following components at the final concentrations listed. Rat tail collagen (1 milligram/milliliter, BD DiscoveryLabware, Bedford, Mass.), 0.01 Normal NaOH and Chondrocyte cell media DMEM, penicillin (100 Units/milliliter), streptomycin (100 micrograms/milliliter), 2 millimolar L-Glutamine, 1 millimolar Sodium Pyruvate, 0.35 millimolar L-Proline, 100 nanomolar dexamethasone, 0.17 millimolar L-Ascorbic Acid, 1% (v/v) ITS (insulin, transferring, selenium) (All components from Sigma Chemical Company). The cells were gently mixed with the medium the samples were aliquoted into individual wells of a 24 well ultra-low cluster plate (Corning, Corning, N.Y.) at a concentration of either 2×105 per well or 5×105 per well. Cultures were placed in an incubator and left undisturbed for 24 to 48 hours. Medium was replaced with fresh chondrocyte media supplemented with appropriate growth factor every 24 to 48 hours. Samples were allowed to culture for up to 28 days at which time they were removed and fixed in 10% (v/v) formalin (VWR Scientific, West Chester, Pa.) and processed for histological examination. Samples were stained with Safranin O or hematoxylin/eosin for evaluation.
  • Pellet culture assays. Cultured cells were trypsinized to remove from culture plate. Cells were washed with centrifugation twice at 300×g for 5 minutes in DMEM without serum and counted. Cells were resuspended in fresh chondrocyte medium (described above) at a concentration of 5×105 cells per milliliter. Cells were aliquoted into new polypropylene tubes at 2.5×105 cells per tube. The appropriate samples were then treated with TGF-beta3 (10 nanograms/milliliter, Sigma) or GDF-5 (100 nanograms/milliliter; R&D Systems, Minneapolis, Minn.). Cells were then centrifuged at 150×g for 3 minutes. Tubes were then transferred to the incubator at and left undisturbed for 24-48 hours in 5% CO2 at 37° C. Medium was replaced with fresh chondrocyte cell media and growth factor, where appropriate, every 2-3 days. Samples were allowed to culture for up to 28 days at which time they were removed and fixed and stained as described above.
  • Results
  • Pellets were prepared and cultured and described in Methods. Pellets were grown in medium (Control), or medium supplemented with TGF beta3 (10 nanograms/milliliter) or GDF-5 (100 nanograms/milliliter), that was replaced every 2 to 3 days. Pellets were collected after 21 days of culture and stained by Safranin O to test for the presence of glycosoaminoglycans. The pellets treated with TGFbeta3 and GDF-5 showed some positive Safranin O staining as compared to control cells. The morphology of the umbilical cord cells showed some limited chondrocyte-like morphology.
  • Summary. Umbilicus-derived cells partially differentiated into chondrocytes in vitro in the pellet culture and the collagen gel assay systems. The umbilicus-derived cells did show some indications of glycosaminoglycan expression by the cells. Morphology showed limited similarity to cartilage tissue. These results suggest that conditions can be optimized to stimulate more complete chondrocyte differentiation of the umbilicus-derived cells.
  • References for Example 21
    • 1. U.S. Markets for Current and Emerging Orthopedic Biomaterials Products and Technologies. Medtech Insight L.L.C. 2002
    • 2. Johnstone, B, T. M. Hering, A. I. Caplan, V. M. Goldberg and J. U. Yoo. In Vitro Chondrogenesis of Bone-Marrow-Derived Mesenchymal Stem Cells. 1998. Exp Cell Res 238:265-272.
    • 3. Gosiewska, A., A. Rezania, S. Dhanaraj, M. Vyakarnam, J. Zhou, D. Burtis, L. Brown, W. Kong, M. Zimmerman and J. Geesin. Development of a Three-Dimensional Transmigration Assay for Testing Cell-Polymer Interactions for Tissue Engineering Applications. 2001 Tissue Eng. 7:267-277.
    EXAMPLE 22 Further Evaluation of Chondrogenic Potential of Cells Derived from Umbilical Cord Tissue in an In Vitro Pellet Culture Based Assay
  • Evaluation of the chondrogenic potential of cells derived from umbilical tissue was performed using in vitro pellet culture based assays. Cells from umbilical cord at early passage (P3) and late passage (P12) were used. The chondrogenic potential of the cells was assessed in pellet culture assays, under chondrogenic induction conditions in medium supplemented with transforming growth factor beta-3 (TGFbeta-3), recombinant human growth and differentiation factor 5 (rhGDF-5), or a combination of both.
  • Materials & Methods
  • Reagents. Dulbecco's Modified Essential Media (DMEM), Penicillin and Streptomycin, were obtained from Invitrogen, Carlsbad, Calif. Fetal calf serum (FCS) was obtained from HyClone (Logan, Utah). Mesenchymal stem cell growth medium (MSCGM) and hMSC chondrogenic differentiation bullet kit were obtained from Biowhittaker, Walkersville, Md. TGFbeta-3 was obtained from Oncogene research products, San Diego, Calif. rhGDF-5 was obtained from Biopharm, Heidelberg, Germany (WO9601316 A1, U.S. Pat. No. 5,994,094 A).
  • Cells. Human mesenchymal stem cells (Lot# 2F1656) were obtained from Biowhittaker, Walkersville, Md. and were cultured in MSCGM according to manufacturers instructions. This lot has been tested previously, and was shown to be positive in the chondrogenesis assays. Human adult and neonatal fibroblasts were obtained from American Type Culture Collection (ATCC), Manassas, Va. and cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. Postpartum tissue-derived cells, isolated from human umbilical cords as described in previous examples, were utilized. Cells were cultured in Growth Medium in a manner similar to the culture of the fibroblasts. The cell cultures were incubated at 37° C. with 5% CO2. Cells used for experiments were at passages 3 and 12.
  • Pellet culture assay. For pellet cultures, 0.25×106 cells were placed in a 15 milliliter conical tube and centrifuged at 150×g for 5 minutes at room temperature to form a spherical pellet according to protocol for chondrogenic assay from Biowhittaker. Pellets were cultured in chondrogenic induction medium containing TGFbeta-3 (10 nanograms/milliliter), rhGDF-5 (500 nanograms/milliliter), or a combination of TGFbeta-3 (10 nanograms/milliliter), and rhGDF-5 (500 nanograms/milliliter) for three weeks. Untreated controls were cultured in growth medium. During culture, pellets were re-fed with fresh medium every other day. Treatment groups included the following:
  • Treatment Group
  • A. Umbilicus-derived cells early passage (U EP)+rhGDF-5
  • B. Umbilicus-derived cells late passage (U LP)+rhGDF-5, n=2
  • C. Human Mesenchymal Stem cells (HMSC)+rhGDF-5
  • D. Human adult fibroblast cells (HAF)+rhGDF-5
  • E. Umbilicus-derived cells early passage (U EP)+TGFbeta-3
  • F. Umbilicus-derived cells late passage (U LP)+TGFbeta-3, n=2
  • G. Human Mesenchymal Stem cells (HMSC)+TGFbeta-3
  • H. Human adult fibroblast cells (HAF)+TGFbeta-3
  • I. Umbilicus-derived cells early passage (U EP)+rhGDF-5+TGFbeta-3
  • J. Umbilicus-derived cells late passage (U LP)+rhGDF-5+TGFbeta-3, n=2
  • K. Human Mesenchymal Stem cells (HMSC)+rhGDF-5+TGFbeta-3
  • L. Human adult fibroblast cells (HAF)+rhGDF-5+TGFbeta-3
  • M. Human neonatal fibroblast cells (HNF)+rhGDF-5+TGFbeta-3
  • N. Umbilicus-derived cells early passage (U EP)
  • O. Umbilicus-derived cells late passage (U LP)
  • P. Human Mesenchymal Stem cells (HMSC)
  • Q. Human adult fibroblast cells (HAF)
  • Histology of in vitro samples. At the end of the culture period pellets were fixed in 10% buffered formalin and sent to MPI Research (Mattawan, Mich.) for paraffin embedding, sectioning, and staining with Hematoxylin & Eosin (H&E) and Safranin O (SO) staining.
  • Results
  • Umbilicus-derived cells, MSCs and fibroblasts formed cell pellets in chondrogenic induction medium with the different growth factors. The size of the pellets at the end of culture period varied among the different cell types. Pellets formed with the umbilical cells tended to be larger and looser than those formed by MSCs and fibroblasts. Pellets formed with all cell types and cultured in control medium were smaller than pellets cultured in chondrogenic induction medium.
  • Examination of cross sections of pellets stained with H&E and Safranin O showed that umbilicus-derived cells at early passage had the potential to undergo chondrogenic differentiation. Chondrogenesis as assessed by cell condensation, cell morphology and Safranin O positive staining of matrix was observed in the umbilical cell pellets cultured in chondrogenic induction medium supplemented with TGFbeta-3, rhGDF-5 or both. Chondrogenesis in pellets was similar for TGFbeta-3, rhGDF-5 and the combined treatments. Control pellets cultured in growth medium showed no evidence of chondrogenesis. Chondrogenic potential of the umbilicus-derived cells was marginally lower than that observed with the MSCs obtained from Biowhittaker.
  • Umbilicus-derived cells at late passage did not demonstrate as distinct a chondrogenic potential as did early passage umbilical derived cells. However, this may be due to the fact that chondrogenic induction conditions were optimized for MSCs, not for postpartum derived cells. Some cell condensation was observed with fibroblast, but it was not associated with Safranin O staining.
  • EXAMPLE 23 Differentiation to the Cardiomyocyte Phenotype
  • There is a tremendous need for therapy that will slow the progression of and/or cure heart disease, such as ischemic heart disease and congestive heart failure. Cells that can differentiate into cardiomyocytes that can fully integrate into the patient's cardiac muscle without arrhythmias are highly desirable. Rodent mesenchymal stem cells treated with 5-azacytidine have been shown to express markers of cardiomyocytes (Fukuda et al. (2002) C. R. Biol. 325:1027-38). This has not been shown for adult human stem cells. Additional factors have been used to improve stem cell differentiation including low oxygen (Storch (1990) Biochim. Biophys. Acta 1055:126-9), retinoic acid (Wobus et al. (1997) J. Mol. Cell Cardiol. 29:1525-39), DMSO (Xu et al. (2002) Circ. Res. 91:501-8), and chelerythrine chloride (International PCT Publication No. WO03/025149), which effects the translocation of PKC from the cytosol to plasma membrane and is an inhibitor of PKC activity.
  • In this example, umbilicus-derived cells were treated with 5-azacytidine either alone or in combination with DMSO or chelerythrine chloride and markers of cardiomyocytes measured by real-time PCR.
  • Methods & Materials
  • Cells. Cryopreserved umbilicus-derived cells (P10) were grown in Growth Medium in gelatin-coated flasks. Cells were seeded at 5×104 cells/well in 96-well plates in Growth Medium for 24 hours. The medium was changed to 0, 3, 10 and 30 micromolar 5-azacytidine (Sigma, St. Louis, Mo.) alone or with 5 microM chelerythrine chloride (Sigma), 1% (v/v) dimethylsulfoxide (DMSO) (Sigma), or 1 micromolar retinoic acid (Sigma) in MEM-alpha (Sigma), insulin, transferrin, and selenium (ITS; Sigma), 10% (v/v) fetal bovine serum, penicillin and streptomycin. The cells were incubated at 37° C., 5% (v/v) O2 for 48-72 hours. The medium was changed to MEM-alpha, insulin, transferrin, and selenium, 10% (v/v) fetal bovine serum, penicillin (50 Units/milliliter) and streptomycin (50 micrograms/milliliter), and cells incubated at 37° C., 5% (v/v) O2 for 14 days.
  • RNA extraction and Reverse Transcription. Cells were lysed with 150 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, Mo.) according to the manufacturer's instructions (RNeasy 96 kit, Qiagen, Valencia, Calif.) and stored at −80° C. Cell lysates were thawed and RNA extracted according to the manufacturer's instructions (RNeasy 96 kit, Qiagen, Valencia, Calif.) with a 2.7 Units/sample DNase treatment (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at −80° C. RNA was reverse-transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C. for 10 minutes, 37° C. for 60 minutes and 95° C. for 10 minutes. Samples were stored at −20° C.
  • PCR. PCR was performed on cDNA samples using ASSAYS-ON-DEMAND gene expression products cardiac myosin (Hs00165276 ml), skeletal myosin (Hs00428600), GATA 4 (Hs00171403 ml), GAPDH (Applied Biosystems, Foster City, Calif.), and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems). Thermal cycle conditions were initially 50° C. for 2 minutes and 95° C. for 10 minutes followed by 40 cycles of 95° C. for 15 seconds and 60° C. for 1 minute. cDNA from heart and skeletal muscle (Ambion, Austin Tex.) were used as controls.
  • Results
  • Control RNA from cardiac muscle showed expression of cardiac myosin and GATA 4, skeletal muscle RNA showed skeletal myosin and cardiac myosin, but no GATA 4 expression. Umbilicus-derived cells (P12) treated for 48 h with factors and cultured for a further 14 days expressed low levels of GATA 4, but no skeletal myosin or cardiac myosin. Additional samples from umbilicus-derived cells also showed expression of GATA 4.
  • Summary. Untreated umbilicus-derived cells constitutively express GATA 4, a nuclear transcription factor in cardiomyocytes, sertoli cells, and hepatocytes.
  • EXAMPLE 24 Assessment of Umbilicus-Derived Cells for Cardiovascular Therapy in a Rodent Coronary Ligation Model
  • Animal models of heart failure have facilitated the understanding of the pathophysiology of the disease and have assisted in the development of new treatments for congestive heart failure (CHF). Coronary artery ligation, or the blocking of the vessels that supply the heart tissue, in the rat closely mimics the pathophysiology of acute myocardial infarction in humans and has been used successfully to study pharmacological interventions for CHF. Cell transplantation of human cells into cardiac lesions is a potential viable therapeutic treatment for CHF.
  • The efficacy of intracardiac human umbilicus-derived cell treatment when administered 15 minutes post-coronary artery occlusion was evaluated in a rodent model of myocardial ischemia/infarction.
  • Methods & Materials
  • The Charles River Worcester, Mass. test facility is accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care, International (AAALAC) and registered with the United States Department of Agriculture to conduct research in laboratory animals. All the conditions of testing conformed to the Animal Welfare Act (9 CFR) and its amendments. The protocol was reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) at the Test Facility for compliance with regulations prior to study initiation.
  • The animals having characteristics identified in Table 24-1 were individually housed in micro-isolator cages on autoclaved bedding. The cages conformed to standards set forth in The Guide for the Care and Use of Laboratory Animals.
    TABLE 24-1
    Animal characteristics
    Species: Rattus norvegicus
    Strain: Rnu
    Source: Charles River Laboratories
    Age at Dosing: 6-8 weeks
    Weight at Dosing: ˜200-250 grams
    Number of Males 40 + 10
    (including spares):
  • PURINA Certified Diet (irradiated) was provided to the animals ad libitum. This diet was routinely analyzed by the manufacturer for nutritional components and environmental contaminants. Results of the manufacturer's analyses are on file at the Test Facility.
  • Autoclaved filtered tap water was provided ad libitum. Samples of the filtered water were analyzed for total dissolved solids, hardness, specified microbiological content, and selected environmental contaminants. Results of these analyses are on file at the Test Facility.
  • Environmental controls were set to maintain temperatures of 18 to 26° C. (64 to 79° F.) with relative humidity at 30% to 70%. A 12:12 hour light:dark cycle was maintained. Ten or greater air changes per hour were maintained in the animal rooms. Upon receipt and prior to use on the study, the animals were held for a minimum of four days for conditioning according to the Test Facility Vendor Management Program as described in the Test Facility Standard Operating Procedure, Receipt, Conditioning, and Quarantine of Laboratory Animals.
  • Each animal was identified by a unique number indicated by an ear punch. Animals were randomly assigned to groups by a weight-ordered distribution such that individual body weights did not exceed ±20% of mean weight.
  • The animals were anesthetized with sodium pentobarbital (40 milligrams/kilogram) and buprenorphine (0.05 milligrams/kilogram) as a single cocktail given intramuscularly (IM). Following the establishment of anesthesia, animals were intubated using 18 to 16 gauge, 2-inch length angiocaths, or appropriate sized angiocath, and maintained on room air respiration (supplemented with oxygen) and a positive pressure ventilator throughout the surgical procedure. Additional anesthesia was given incrementally as needed. Preoperative antibiotic therapy was also administered, Benzathine/Procaine penicillin G, 40,000 Units/kilogram, IM. Additional antibiotic therapy was administered every 48 hours.
  • Electrode pads were placed around the appropriate paws of the animals to receive a useable ECG signal. Animals were positioned on a heating pad to help maintain body temperature throughout the procedure. A rectal temperature probe was inserted into the animal to monitor body temperature. Ophthalmic ointment was administered to each eye. The surgical sites (thoracic area) were prepared for aseptic surgery by removing any excess fur, and gently wiping the area with sponges soaked in 70% isopropyl alcohol, which was allowed to dry. Iodone (MEDISEPPS, or similar solution) was then applied to the area and allowed to dry. The area was appropriately draped for strict aseptic surgery.
  • A surgical incision was made on the skin over the fourth intercostal space. Blunt dissection through the muscle layers was used to access the thoracic cavity. A retractor was carefully inserted into the fourth intercostal space and opened to allow access to the interior cavity. The pericardium was carefully opened via gentle teasing with cotton swabs dampened in sterile saline solution. A damp cotton swab was used to gently push the apex of the heart into the opening where a length of 6-0 silk suture was attached into the myocardium for manipulation of the heart. After a pause to allow the heart to recover, the suture placed in the apex was used to ease the heart out of the chest cavity and to place sufficient tension on the heart to allow access to the upper heart and the left anterior descending coronary artery (LAD). Another length of 6-0 silk suture was placed into the myocardium so as to surround the LAD. The pressure on the apical suture was released and the heart allowed to return to the interior of the chest cavity.
  • Once the heart rate and ECG returned to baseline values, the ligatures around the LAD were tied off to occlude the LAD. This was a permanent occlusion with the suture tied off and the ends trimmed. After the ligature was tied, the surgeon looked for the following indications of successful occlusion: change in color of the area of the heart directly below the ligature to a white/grayish white as a result of the termination of blood flow to the area and a significant change in the ECG corresponding to occlusion of the LAD. Arrhythmias may have developed within the first 10 minutes of the occlusion. The rat was monitored closely during this time period in the event that resuscitation was necessary. In the event of severe arrhythmia and failure of the rat to convert to normal sinus rhythm without assistance, aid was rendered via cardiac massage. Approximately 15 minutes following the initiation of the LAD occlusion, the area of left ventricle made ischemic was treated with either vehicle or test article by direct injection into the ischemic myocardium. Treatment consisted of three to ten intramyocardial injections (100 uL/injection) into the ischemic zone of myocardium.
  • Human cells were grown in Growth Medium in gelatin-coated T300 flasks. Cells were washed with phosphate buffered saline (PBS, Gibco, Carlsbad Calif.) and trypsinized using Trypsin/EDTA (Gibco, Carlsbad Calif.). The trypsinization was stopped by adding Growth Medium. The cells were centrifuged at 150×g, supernatant removed, and the cell pellet was resuspended in approximately 1 milliliter Growth Medium per million cells. An aliquot of cells was removed and added to Trypan blue (Sigma, St. Louis, Mo.). The viable cell number was estimated using a hemocytometer. The cell suspension was centrifuged and resuspended in 1 milliliter Growth Medium containing 10% (v/v) DMSO (Hybrimax, Sigma, St. Louis, Mo.) per 5 million cells and transferred into Cryovials (Nalgene). The cells were cooled at approximately 1° C./minute overnight in a −80° C. freezer using a “Mr. Frosty” freezing container (Nalgene, Rochester, N.Y.). Vials of cells were transferred into liquid nitrogen. Vials were shipped from CBAT, Somerville, N.J. to Charles River, Worcester, Mass. on dry ice and stored at −80° C. Approximately 1-2 hours before injection of cells into the animal, a vial of cells was thawed rapidly in a 37° C. water bath. Under aseptic conditions in a BSL2 biosafety cabinet, cells were added to 40 milliliters PBS with magnesium and calcium (Sigma St. Louis, Mo.) and centrifuged at 150×g for 5 minutes before resuspending the cell pellet in 10 milliliters PBS. The cell number and viability was estimated as described above. The cells were centrifuged at 150×g for 5 minutes and resuspended in PBS at a final concentration of 106 viable cells/100 microliters. The cell suspension was loaded into 1 milliliter syringes with a 30G needle and kept on ice. Viability was assessed again up to 5 hours on ice.
  • Following the administration of treatment (Table 24-2) and stabilization of the heart, the surgeon began closing the surgical incision. The retractor was removed. The lungs were over-inflated for 3 to 4 breaths and visually inspected as much as possible to ensure that they were fully re-inflated. This created a negative pressure necessary to prevent pneumothorax post-recovery. To evacuate fluid and excess air from the thoracic cavity after closing the cavity, an intravenous catheter (i.e., 20 gauge, 2 millimeters in length) was placed through the skin and muscle layers so that the tip remains in the thoracic cavity. Care was taken so that the tip did not pierce the lung or heart. The separated ribs and associated muscle was sutured together with appropriate suture. The upper layers of muscle was sutured using a simple continuous pattern. The skin was closed with 4-0 silk using a horizontal mattress pattern. A 10 milliliter syringe was attached to the intravenous catheter that had been previously placed in the thoracic cavity and the plunger slowly pulled back to withdraw fluids and air from the cavity. At the same time, the catheter was slowly withdrawn from the entry site, thereby allowing the surrounding muscle mass and skin to seal the puncture. The surgical drape was removed and fluids (i.e., lactated Ringers solution, 25 milliliters/kilogram subcutaneously [SC] or intraperitoneally [IP]) were given.
    TABLE 24-2
    Treatment regimens
    Dosage Time of
    Gr. No. of Level Dose Conc. Route/Dose Treatment Necropsy
    No. Males Test Article (cells/animal) (cells/mL) Regimen Administration Day
    1 8 Vehicle 0 0 Direct 15 minutes Day 28
    2 8 Umbilical (P10) 1 million 10 million injection(s) into after coronary (±1 Day)
    (B) the ischemic artery ligation
    2 8 Human region of the left
    fibroblasts ventricle of the
    1F1853 (P10) (D) heart, consisting
    of 3 to 10
    intramyocardial
    injections of
    100 μL total.

    Gr. = Group;

    No. = Number;

    Conc. = Concentration
  • Immediately after each rat had undergone treatment with test article and the incision was sutured, the animal underwent an echocardiography (ECG) examination. Anesthesia was maintained throughout the completion of the echo examination. Upon the completion of the echo examination, ventilation was discontinued, and the rat was returned to the recovery area to recover in a heated, oxygenated recovery cage.
  • A second echo examination of each surviving animal was completed at the end of the study (approximately 28 days post-treatment), prior to termination. During the second examination, the animals were anesthetized as described previously.
  • For each echo examination, the left thoracic area was shaved, and warmed, ultrasonic gel was applied to the skin to enhance contact with the transducer. Electrode pads were placed around the appropriate extremities to receive an ECG signal. Echocardiographic images included short axis and long axis views to allow for the determination of ventricular cavity dimensions, contractility, blood flow through vasculature, and wall thickness. These images were saved on optical disk for further analysis. After examination, the gel medium was removed from the skin with gauze or paper towel. The rat was removed from the ventilator and placed in a warmed recovery cage until mobile.
  • At the conclusion of the surgical procedures, respiratory ventilation was turned off. The animals were observed for pedal reflex. The rectal probe and ECG electrodes subsequently were removed, and the animal was extubated and placed in a warmed oxygenated recovery cage. After complete recovery from anesthesia, the animals were given buprenorphine (0.05 milligram/kilogram, SC). Observations were made regularly until the animals showed full mobility and an interest in food and water. The animals then were placed in a clean housing cage and returned to the animal housing room. Animals were monitored for surgical incision integrity twice daily post-surgery.
  • Analgesics (i.e., Buprenorphine, 0.05 milligram/kilogram SC.) were given twice daily for 4 days post-operatively and thereafter as needed. Visual indications of post-operative pain include lack of normal body postures and movement (e.g., animal remains in hunched position), antipathy, lack of eating/drinking, lack of grooming, and the like.
  • Body weight was recorded for each animal prior to initial treatment, weekly thereafter, and on the day of necropsy. Animals found dead were weighed and necropsied.
  • In order for the heart to be harvested, each rat was anesthetized as was done for surgery. The jugular vein was cannulated. The heart was arrested in diastole with KCl infused via the jugular cannula. The heart was then removed from the thoracic cavity. A limited necropsy was then performed on the heart after which the heart was placed in 10% neutral buffered formalin. The remainder of each carcass was then discarded with no further evaluation.
  • Hearts of all animals that were found dead or euthanized moribund were placed in 4% paraformaldehyde until evaluated. The remainder of each carcass was then discarded with no further evaluation.
  • Histology and Image Analysis. Fixed tissues sectioned with a stainless steel coronal heart matrix (Harvard Apparatus, Holliston, Mass.) yielded four two-millimeter thick serial tissue sections. Sections were processed and serially embedded in paraffin using routine methods. Five-micron sections were obtained by microtome and stained with Masson's Tri-Chrome for Connective Tissue (Poly Scientific, Bay Shore, N.Y.) using the manufacturer's procedures. Electronic photomicrographs were captured and analyzed using image analysis methods developed by Phase 3 Imaging System (Glen Mills, Pa.). Photomicrographs of the tri-chrome stained sections were colorimetrically analyzed electronically to determine the overall area of the ventricle and free wall and the area of the differential staining.
  • Results
  • There was no loss in the viability of cells over 5 hours in the vehicle when kept on ice. Cells were injected into the infarct with one to three needle entry points and multiple changes in direction of needle orientation.
  • Fractional shortening values were calculated as described by Sahn et al. (1978) Circulation 58:1072-1083. The fractional shortening of the vehicle-treated animals had a significant decrease from 47.7%±8.3% at Day 0 to 23.5%±30.2% at Day 28 (p<0.05). The animals that were treated with umbilicus-derived cells showed small, non-significant differences between the fractional shortening between Day 0 and 28. There were no significant differences between the fractional shortening between the treatment groups at Day 0.
  • Upon termination of the study, hearts were collected and subjected to histological analysis. The hearts were arrested in diastole and fixed. The results were calculated from an algorithm to estimate the percentage of total heart area that comprises the infarct. The infarct size in the vehicle-treated animals was 22.9%±6.7% of heart area, while the infarct size in hearts treated with umbilical cord cells was 12.5%±2.5%, with placenta-derived cells (isolate 2) was 12.9%±3.4%, and with fibroblasts was 19.3%±8.0%. The difference of infarct size of cell-treated animals relative to vehicle-treated animals was not statistically significant based on Student's t-test.
  • Summary. The results of the present study suggest that the umbilicus-derived cells have some benefit in reducing the damage of a surgically induced myocardial infarction in rats. The vehicle-treated animals showed a significant reduction in cardiac function from day 0 to day 28, as measured by fractional shortening, while the umbilicus-derived cell-treated animals showed minimal change over the 28-day study. The fibroblast-treated animals showed minimal change but only two animals survived the study. Evaluation of infarct size suggested that there is some modest, but not statistically significant, reduction in the infarct size in the postpartum-derived cell-treated animals as compared to the vehicle controls at Day 28. Taken together, these data support efficacy of the umbilicus-derived cells in reducing damage from a myocardial infarction.
  • EXAMPLE 25 Use of Umbilicus-derived Cells in the Treatment of Retinitis Pigmentosa
  • Currently no real treatment exists for blinding disorders that stem from the degeneration of cells in the retina. Loss of photoreceptors as a result of apoptosis or secondary degeneration lead to progressive deterioration of vision, and ultimately to blindness. Diseases in which this occurs include age-related macular degeneration (AMD) and retinitis pigmentosa (RP). RP is most commonly associated with a single gene mutation, which contributes to photoreceptor cell death.
  • The retinal photoreceptors and adjacent retinal pigment epithelium form a functional unit. The Royal College of Surgeons (RCS) rat presents with a tyrosine receptor kinase (Mertk) defect affecting outer segment phagocytosis, leading to photoreceptor cell death (1). Transplantation of retinal pigment epithelial (RPE) cells into the subretinal space of RCS rats was found to limit the progress of photoreceptor loss and preserve visual function (2). In this example, it is demonstrated that umbilicus-derived cells can be used to promote photoreceptor rescue in an RCS model.
  • Methods & Materials
  • Cell transplants. Cultures of human adult umbilical and fibroblast cells (passage 10) were expanded for 1 passage. All cells were initially seeded at 5,000 cells/cm2 on gelatin-coated T75 flasks in Growth Medium. For subsequent passages, all cells were treated as follows: After trypsinization, viable cells were counted after trypan blue staining. Briefly, 50 microliters of cell suspension was combined with 50 microliters of 0.04% w/v trypan blue (Sigma, St. Louis Mo.) and the viable cell number, was estimated using a hemocytometer. Cells were trypsinized and washed three times in supplement-free DMEM:Low glucose medium (Invitrogen, Carlsbad, Calif.). Cultures of human umbilical and fibroblast cells at passage 11 were trypsinized and washed twice in Leibovitz's L-15 medium (Invitrogen, Carlsbad, Calif.).
  • For the transplantation procedure, dystrophic RCS rats were anesthetized with xylazine-ketamine (1 milligram/kilogram i.p. of the following mixture: 2.5 milliliters xylazine at 20 milligrams/milliliter, 5 milliliters ketamine at 100 milligrams/milliliter, and 0.5 milliliter distilled water) and their heads secured by a nose bar. Cells devoid of serum were resuspended (2×105 cells per injection) in 2 microliters of Leibovitz, L-15 medium (Invitrogen, Carlsbad, Calif.) and transplanted using a fine glass pipette (internal diameter 75-150 microns) trans-sclerally.
  • Cells were delivered into the dorso-temporal subretinal space of anesthetized 3 week old dystrophic-pigmented RCS rats (total N=10/cell type). Cells were injected unilaterally into the right eye, while the left eye was injected with carrier medium alone (Sham control; Leibovitz's L-15 medium). Viability of residual transplant cells remained at greater than 95% as assessed by trypan blue exclusion at the end of the transplant session. After cell injections were performed, animals were injected with dexamethasone (2 milligram/kilogram) for 10 days post transplantation. For the duration of the study, animals were maintained on oral cyclosporine A (210 milligrams/liter of drinking water; resulting blood concentration: 250-300 micrograms/liter) (Bedford Labs, Bedford, Ohio) from 2 days pre-transplantation until end of the study. Food and water were available ad libitum. Animals were sacrificed at 60 or 90 days postoperatively, with some animals being euthanitized at earlier timepoints for histological assessment of short-term changes associated with cell transplantation.
  • ERG recordings. Following overnight dark adaptation, animals were prepared for ERG recording under dim red light, as previously described (3). In brief, under anesthesia (with a mixture of 150 milligram/kilogram i.p ketamine, and 10 milligram/kilogram i.p. xylazine) the animal's head was secured with a stereotaxic head holder and the body temperature monitored through a rectal thermometer and maintained at 38° C. using a homeothermic blanket. Pupils were dilated using equal parts of topical 2.5% phenylephrine and 1% tropicamide. Topical anesthesia with 0.75% bupivacaine was used to prevent any corneal reflexes and a drop of 0.9% saline was frequently applied on the cornea to prevent its dehydration and allow electrical contact with the recording electrode (gold wire loop). A 25-gauge needle inserted under the scalp, between the two eyes, served as the reference electrode. Amplification (at 1-1,000 Hz bandpass, without notch filtering), stimulus presentation, and data acquisition were provided by the UTAS-3000 system from LKC Technologies (Gaithersburg, Md.). ERGs were recorded at 60 and 90 days of age in the umbilical cell groups and at 60 days only in the fibroblast groups.
  • Mixed a- and b-wave recording. For the quantification of dark-adapted b-waves, recordings consisted of single flash presentations (10 microseconds duration), repeated 3 to 5 times to verify the response reliability and improve the signal-to-noise ratio, if required. Stimuli were presented at six increasing intensities in one log unit steps varying from −3.6 to 1.4 log candila/m2 in luminance. To minimize the potential bleaching of rods, inter-stimulus intervals were increased as the stimulus luminance was elevated from 10 seconds at lowest stimulus intensity to 2 minutes at highest stimulus intensity. The maximum b-wave amplitude was defined as that obtained from the flash intensity series, regardless of the stimulus intensity. The true Vmax from fitting the data with a Naka-Rushton curve was not used because ERG responses were often erratic at higher luminance levels in dystrophic animals and showed tendencies for depressed responses around 0.4 and 1.4 log candila/m2. In order to determine the age at which ERG components were obtained or lost, criterion amplitudes were used: 20 microVolts for a- and b-waves, and 10 microVolts for STR-like responses. The amplitude of the b-wave was measured from the a-wave negative peak up to the b-wave positive apex, and not up to the peak of oscillations, which can exceed the b-wave apex (4).
  • Isolation of rod and cone responses. The double flash protocol was used to determine the isolation of rod and cone responses (5). A probe flash was presented 1 second after a conditioning flash, using a specific feature of the UTAS-3000 system (LKC Technologies) with calibrated ganzfeld; assuring complete recharge of the stimulator under the conditions used. The role of the conditioning flash in the procedure was to transiently saturate rods so that they were rendered unresponsive to the probe flash. Response to the probe flash was taken as reflecting cone-driven activity. A rod-driven b-wave was obtained by subtracting the cone-driven response from the mixed response (obtained following presentation of a probe flash alone, i.e. not preceded by any conditioning flash).
  • Functional Assessment. Physiological retinal sensitivity testing was performed to demonstrate retinal response to dim light. Animals were anesthetized with a recovery dose of urethane at 1.25 grams/kilogram i.p. Physiological assessment in the animals was tested post graft in animals at 90 days by recording multiunit extracellular activity in the superior colliculus to illumination of respective visual receptive fields (6). This procedure was repeated for 20 independent points (spaced 200 millimeters apart, with each step corresponding to approximately 10-150 displacements in the visual field), covering the visual field. Visual thresholds were measured as the increase in intensity over background and maintained at 0.02 candila/m2 (luminescence unit) [at least 2.6 logarithm units below rod saturation (7)], required for activating units in the superficial 200 microns of the superior colliculus with a spot of light 3° in diameter. Response parameters were compared between transplanted and sham control eyes that received vehicle alone.
  • Histology. Animals were sacrificed with an overdose of urethane (12.5 grams/kilogram). The orientation of the eye was maintained by placing a 6.0 suture through the superior rectus muscle prior to enucleation. After making a corneal incision, the eyes were fixed with 2.5% parafomaldehyde, 2.5% glutaraldehyde, 0.01% picric acid in 0.1 M cacodylate buffer (pH 7.4). After fixation, the cornea and lens were removed by cutting around the cilliary body. A small nick was made in the periphery of the dorsal retina prior to removal of the superior rectus to assist in maintaining orientation. The retinas were then post-fixed in 1% osmium tetroxide for 1 hour. After dehydration through a series of alcohols to epoxypropane, the retinas were embedded in TAAB embedding resin (TAAB Laboratories, Aldemarston, UK). Semi-thin sections were stained with 1% Toluidine Blue in 1% borate buffer and the ultra thin sections were contrasted with uranyl acetate and lead citrate.
  • For Nissl staining, sections were stained with 0.75% cresyl violet (Sigma, St. Louis, Mo.) after which they were dehydrated through graded alcohols at 70, 95 and 100% twice, placed in xylene (Sigma, St. Louis, Mo.), rinsed with PBS (pH 7.4) (Invitrogen, Carlsbad, Calif.), coverslipped and mounted with DPX mountant (Sigma, St. Louis, Mo.).
  • Results
  • ERG Recordings. Animals that received umbilicus-derived cell injections exhibited relative preservation of visual response properties 60 and 90 days post-operatively (Table 25-1). The response observed in these animals was greater than that seen with fibroblast or sham treated animals.
  • Umbilicus-derived cell-transplanted animals (n=6) demonstrated good improvement in all outcome measures tested at 60 days (Table 25-1), a-wave (27±11) versus sham controls (0), mixed b-wave (117±67) versus sham controls (18±13), cone-b-wave (55±25) versus sham controls (28±11), and in rod contribution (49±16%) versus sham controls (6±7%). Furthermore, at 90 days, improved responses were measured in two animals tested, with measures including: a-wave (15±7) versus sham controls (0), mixed b-wave (37±15) versus sham controls (0), cone-b-wave (16±11) versus sham controls (7±5), and in rod contribution (58±39%) versus sham controls (0%). These results indicate that visual responsiveness was improved in umbilicus-derived cell transplanted animals with evidence for photoreceptor rescue. Although a diminution in responsiveness to ERG was observed in the 90-day animals tested, their preservation of visual function in comparison to sham-treated controls was good.
  • In contrast to umbilicus-derived cells, fibroblast transplantations showed no improvement in any of the parameters tested.
    TABLE 25-1
    ERG data
    a-wave mixed b-wave cone b-wave % rod contribution
    Group Untreated Treated Untreated Treated Untreated Treated Untreated Treated
    Sham 60d 0 0 7 ± 9 0 23 ± 5 12 ± 16 N/A N/A
    U (n = 6) 0 27 ± 11 18 ± 13 117 ± 67  28 ± 11 55 ± 25 6 ± 7 49 ± 16
    60d
    U (n = 6) 0 15 ± 7  0  37 ± 15  7 ± 5 16 ± 11 0 58 ± 39
    90d

    N.B. Sham = control (medium only),

    U = Umbilicus-derived cell transplant
  • Histology. Following transplantation, there was no histological evidence of an inflammatory reaction and infiltrating immune cells were not observed in Nissl-stained sections in the postpartum cell groups. However, fibroblast implantations resulted in animal death (n=7) and indications of early stage inflammatory responses. Histologically at the 90 day time point in the umbilicus-derived cell transplanted animals anatomical rescue of photoreceptors was clearly demonstrated. The photoreceptors formed a thick layer separated by a gap from the inner nuclear layer, made up of other retinal cells. By comparison, the width of the outer layer in the sham control was, at best, a discontinuous single layer as opposed to around 5 cells thick in the grafted eye. In comparison to a normal animal this is marginally more than half the thickness of photoreceptor cell layers normally observed.
  • Functional Assessment. Efficacy of transplants in preventing visual loss was monitored by assessment of electrophysiological responsiveness in two animals. The threshold sensitivity response to light was used to define the area of visual field rescue in sham-injected control eyes versus eyes transplanted with umbilicus-derived cells. In nondystrophic rats, visual thresholds never exceeded 0.5 log candila/m2 above background. In non-operated dystrophic rats, the thresholds are usually in the magnitude of 4 log candila/m2 units (8). By contrast, in non-operated sham injected dystrophic rats, the thresholds were in the order of 2.9-4.9 log candila/m2 units with an average threshold of 4.0 log candila/m2 units, in some instances no recording could be attained. Thus, the sham-injected rats showed some highly localized functional rescue in the temporal retina. However, the human umbilicus-derived cell transplanted rats exhibited substantially greater levels of visual preservation with thresholds ranging from 0.8 to 2.1 log candila/m2 units, with an average threshold of 1.3 log candila/m2 units.
  • Summary. Transplantation of umbilicus-derived cells into dystrophic RCS rats can preserve photoreceptors. In this degenerative model, one would expect the a-wave to disappear within 30 to 60 days and the b-wave to disappear within 3 months. Thus, the basically retained a-wave indicates that real and normal rod function is preserved. Rod contribution to b-wave suggests abnormal rod function is still possible. The sustained non-rod b-wave is the measure of how much cone function is maintained, which is a real measure of vision. Thus, the level of improvement assessed both physiologically and anatomically following umbilicus-derived cell transplantation is well defined here. ERG measurements provide an assessment of visual function after photoreceptor loss, indicating changes in electrical activity in the retina. However, ERG does not provide direct information as to image forming capability. The measurement of collicular threshold sensitivity used in this study provides an indication of relative preservation of visual fields. The importance of this measure is based on a correlation between the amounts of functional rescue and anatomical preservation and that the data collected compares with visual field perimetry testing in humans (9). The transplantation has demonstrated a retardation of the disease process in the test animals. Thus, the results presented herein demonstrate clear evidence of functional efficacy of grafting human umbilicus-derived cells into the subretinal space, and that preservation of photoreceptors occurs in the general region in which the grafted cells are located.
  • References for Example 25
    • 1. D'Cruz P M, Yasumura D, Weir J, Matthes M T, Abderrahim H, LaVail MM, Vollrath D. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum Mol Genet. 2000 Mar. 1;9(4):645-51.
    • 2. L1 LX, Turner J E. Inherited retinal dystrophy in the RCS rat: prevention of photoreceptor degeneration by pigment epithelial cell transplantation. Exp Eye Res. 1988 December; 47(6):911-7.
    • 3. Sauve, Y, Lu, B and Lund R D. The relationship between full field electroretinogram and perimetry-like visual thresholds in RCS rats during photoreceptor degeneration and rescue by cell transplants. Vision Res. 2004 January; 44(1):9-18.
    • 4. Nusinowitz, S., Ridder, WH 3rd, and Heckonlively, HR. Rod multifocal electroretinograms in mice. Invest Ophthalmol Vis Sci. 1999 November; 40(12):2848-58.
    • 5. Nixon, PJ, Bui, PV, Armitage, JA, and Vingrys A J. The contribution of cone responses to rat electroretinograms. Clin Experiment Ophthalmol. 2001 June; 29(3):193-6.
    • 6. Lund R D, Adamson P, Sauve Y, Keegan D J, Girman S. V, Wang S, Winton H, Kanuga N, Kwan A S, Beauchene L, Zerbib A, Hetherington L, Couraud P O, Coffey P, Greenwood J. Subretinal transplantation of genetically modified human cell lines attenuates loss of visual function in dystrophic rats. Proc Natl Acad Sci USA. 2001 Aug. 14; 98(17):9942-7.
    • 7. Siminoff R, Kruger L. Properties of reptilian cutaneous mechanoreceptors. Exp Neurol. 1968 March;20(3):403-14.
    • 8. Blakema, G. W. and Drager, U. C. 1991. Visual Neuroscience. 6:577-585.
    • 9. Beck R W, Bergstrom T J, Lichter P R. A clinical comparison of visual field testing with a new automated perimeter, the Humphrey Field Analyzer, and the Goldmann perimeter. Ophthalmology. 1985 January; 92(1):77-82.
    EXAMPLE 26 Chondrogenic Potential of Postpartum-Derived Cells on Implantation in SCID Mice
  • The chondrogenic potential of cells derived from umbilicus or placenta tissue was evaluated following seeding on bioresorbable growth factor-loaded scaffolds and implantation into SCID mice.
  • Materials & Methods
  • Reagents. Dulbecco's Modified Essential Media (DMEM), Penicillin and Streptomycin, were obtained from Invitrogen, Carlsbad, Calif. Fetal calf serum (FCS) was obtained from HyClone (Logan, Utah). Mesenchymal stem cell growth medium (MSCGM) was obtained from Biowhittaker, Walkersville, Md. TGFbeta-3 was obtained from Oncogene research products, San Diego, Calif. rhGDF-5 was obtained from Biopharm, Heidelberg, Germany (International PCT Publication No. WO96/01316 A1, U.S. Pat. No. 5,994,094A). Chondrocyte growth medium comprised DMEM-High glucose supplemented with 10% fetal calf serum (FCS), 10 millimolar HEPES, 0.1 millimolar nonessential amino acids, 20 micrograms/milliliter L-proline, 50 micrograms/milliliter ascorbic acid, 100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, and 0.25 micrograms/milliliter amphotericin B. Bovine fibrinogen was obtained from Calbiochem.
  • Cells. Human mesenchymal stem cells (hMSC, Lot# 2F1656) were obtained from Biowhittaker, Walkersville, Md. and were cultured in MSCGM according to the manufacturer's instructions. This lot was tested in the laboratory previously in in vitro experiments and was shown to be positive in the chondrogenesis assays. Human adult fibroblasts were obtained from American Type Culture Collection (ATCC, Manassas, Va.) and cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. Postpartum-derived cells isolated from human umbilical cords (Lot# 022703Umb) and placenta (Lot# 071003Plac) were prepared as previously described (Example 1). Cells were cultured in Growth Media on gelatin-coated tissue culture plastic flasks. The cell cultures were incubated at 37° C. with 5% CO2. Cells used for experiments were at passages 5 (“Low passage”) and 14 (“High passage”).
  • Scaffolds. Foams composed of 35/65 Poly(epsilon-caprolactone)(PCL)/Poly(glycolic acid) (PGA) (35/65 PCL/PGA) copolymer, reinforced with Polydioxanone (PDS) mesh (PGA/PCL foam-PDS mesh) were formed by the process of lyophilization, as described in U.S. Pat. No. 6,355,699. The foams were 4 cm×5 cm, and 1 mm thick. Foams were sterilized by treatment with ethylene oxide (ETO). Punches (3.5 millimeters) made from scaffolds were loaded with either rhGDF-5 (3.4 micrograms/scaffold), TGFbeta-3 (10 nanograms/scaffold), a combination of rhGDF-5 and TGFbeta-3, or control medium, and lyophilized.
  • Cell seeding on scaffolds. Placenta- and umbilicus-derived cells were treated with trypsin, and cell number and viability was determined. 7.5×105 cells were resuspended in 15 microliters of Growth Medium and seeded onto 3.5 millimeter scaffold punches in a cell culture dish. The cell-seeded scaffold was incubated in a cell culture incubator (37° C., 5% CO2) for 2 hours after which they were placed within cartilage explant rings.
  • Bovine Cartilage Explants. Cartilage explants 5 millimeters in diameter were made from cartilage obtained from young bovine shoulder. Punches (3 millimeters) were excised from the center of the explant and replaced with cells seeded on 3.5 millimeters resorbable scaffold. Scaffolds with cells were retained within the explants using fibrin glue (60 microliters of bovine fibrinogen, 3 milligrams/milliliter). Samples were maintained in chondrocyte growth medium overnight, rinsed in Phosphate Buffered Saline the following day, and implanted into SCID mice.
  • Animals. SCID mice ((Mus musculus)/Fox Chase SCID/Male), 5 weeks of age, were obtained from Harlan Sprague Dawley, Inc. (Indianapolis, Ind.) and Charles River Laboratories (Portage, Mich.). Animals used in the study were selected without any apparent systematic bias. A tag was placed on each individual animal cage listing the accession number, implantation technique, animal number, species/strain, surgery date, in vivo period, and date of euthanasia. The animals were identified by sequential numbers marked on the ear with an indelible ink marker.
  • Experimental Design. A total of 42 mice were tested. Two scaffolds were implanted subcutaneously in each mouse as described below; 42 mice for subcutaneous implantation; 28 treatments with n-value of 3 per treatment. The study corresponds to IACUC Approval Number: Skillman IACUC 01-037. The study lasted six weeks.
  • SCID Implantation.
  • A. Body Weights
  • Each animal was weighed prior to being anesthetized and at necropsy.
  • B. Anesthesia and Surgical Preparation:
  • All handling of the SCID mice occurred under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of KETASET® (ketamine hydrochloride [60 milligram/kilogram]), ROMPUN® (xylazine [10 milligram/kilogram]), and saline.
  • After induction of anesthesia, the entire back of the animal from the dorsal cervical area to the dorsal lumbosacral area was clipped free of hair using electric animal clippers. The area was scrubbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine. Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period. The anesthetized and surgically prepared animal was placed in the desired recumbent position.
  • C. Subcutaneous Implantation Technique:
  • An approximate 2-centimeters skin incision was made just lateral to the thoracic spine parallel to the vertebral column. The skin was separated from the underlying connective tissue via blunt dissection. Each SCID mouse received 2 treatments that were placed in subcutaneous pockets created by blunt dissection in each hemithorax through one skin incision (Table 26-1) Tacking sutures of 5-0 ETHIBOND EXCEL (polyester) (Ethicon Inc, Somerville, N.J.) were used to tack the skin to musculature around each scaffold to prevent subcutaneous migration. Scaffolds were implanted for 6 weeks and then harvested. The experimental design is outlined in Table 26-1.
    TABLE 26-1
    Experimental Design: Treatment (N = 3 per treatment)
    A. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, EP, TGFb3
    B. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, EP, rhGDF-5
    C. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, EP, rhGDF-
    5 + TGFb3
    D. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, Ep, control
    E. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, LP, TGFb3
    F. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, LP, rhGDF-5
    G. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, LP, rhGDF-
    5 + TGFb3
    H. 65/35 PGA/PCL Foam + PDS mesh cultured with Placental derived cells, LP, control
    I. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, EP, TGFb3
    J. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, EP, rhGDF-5
    K. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, EP, rhGDF-
    5 + TGFb3
    L. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, EP, control
    M. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, LP, TGFb3
    N. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, LP, rhGDF-5
    O. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, LP, rhGDF-
    5 + TGFb3
    P. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical derived cells, LP, control
    Q. 65/35 PGA/PCL Foam + PDS mesh cultured with hMSC, TGFb3
    R. 65/35 PGA/PCL Foam + PDS mesh cultured with hMSC, rhGDF-5
    S. 65/35 PGA/PCL Foam + PDS mesh cultured with hMSC, rhGDF-5 + TGFb3
    T. 65/35 PGA/PCL Foam + PDS mesh cultured with hMSC, control
    U. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult TGFb3
    V. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult rhGDF-5
    W. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult rhGDF-5 + TGFb3
    X. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult control
    Y. 65/35 PGA/PCL Foam + PDS mesh, TGFb3
    Z. 65/35 PGA/PCL Foam + PDS mesh, rhGDF-5
    AA. 65/35 PGA/PCL Foam + PDS mesh, rhGDF-5 + TGFb3
    BB. 65/35 PGA/PCL Foam + PDS mesh, control
  • D. Necropsy and Histologic Preparation
  • Gross examination was performed on any animals that died during the course of the study or were euthanized in moribund condition. Selected tissues were saved at the discretion of the study director and/or pathologist.
  • Mice were euthanized by CO2 inhalation at their designated intervals. Gross observations of the implanted sites were recorded. Samples of the subcutaneous implantation sites with their overlying skin were excised and fixed in 10% buffered formalin. Each implant was bisected into halves, and one half was sent to MPI Research (Mattawan, Mich.) for paraffin embedding, sectioning, and staining with Hematoxylin & Eosin (H&E) and Safranin O (SO).
  • Results
  • New cartilage and bone formation was observed in the majority of the samples including growth factor-loaded, cell-seeded scaffolds, cell-seeded control scaffolds, and scaffolds loaded with growth factor alone. The extent of new cartilage and bone formation varied within the treatment and control groups.
  • Early and Late passage placenta-derived cell seeded scaffolds showed new cartilage and bone formation within the scaffolds. No obvious differences in new cartilage and bone formation was observed between the different growth factor-loaded, cell-seeded scaffolds and scaffolds seeded with cells alone. Compared to control scaffolds (without growth factors and without cells), it appeared that there was greater extent of new cartilage formation in cell-seeded scaffolds both with and without growth factors and in growth factor-loaded scaffolds alone. New cartilage formation with placenta-derived cell-seeded scaffolds was similar to MSC- and fibroblast-seeded scaffolds.
  • In growth factor-treated and control scaffolds seeded with umbilicus-derived cells at early and late passage, new cartilage and bone formation were observed. The extent of cartilage formation appeared to be less than that seen with placenta-derived cells. No one sample showed extensive cartilage formation as seen with the placenta-derived cells. Bone formation appeared to be higher in scaffolds seeded with umbilicus-derived cells on scaffolds containing both TGFbeta-3 and rhGDF-5.
  • hMSC-loaded scaffolds also showed new cartilage and bone formation. The extent of new cartilage and bone formation was similar for all the hMSC treatment groups. Human adult fibroblast seeded scaffolds also demonstrated new cartilage and bone formation. Results were similar to those obtained with placenta-derived cells and hMSCs
  • In the control group, in which growth factor-loaded scaffolds or scaffold alone were placed in cartilage rings and implanted, new cartilage and bone formation were also observed. Not surprisingly, the extent of new cartilage formation was greater in scaffolds with growth factor than in scaffolds without growth factor. Increased bone formation was present in the control with the combination of the two tested growth factors.
  • New cartilage formation was observed adjacent to the cartilage explant rings as well as within the scaffolds. New cartilage formation within the scaffolds adjacent to the cartilage rings could be a result of chondrocyte migration. Cartilage formation seen as islands within the scaffolds may be a result of either migration of chondrocytes within the scaffolds, differentiation of seeded cells or differentiation of endogenous mouse progenitor cells. This observation stems from the fact that in control growth factor-loaded scaffolds with no seeded cells, islands of chondrogenic differentiation were observed. New bone formation was observed within the scaffolds independently and also associated with chondrocytes. Bone formation may have arisen from osteoblast differentiation as well as endochondral ossification.
  • It is difficult to separate new cartilage and bone formation associated with chondrocytes that migrated versus that from any chondrogenic and osteogenic differentiation of seeded cells that may have occurred. Staining of sections with specific human antibodies may distinguish the contribution of the seeded cells to the observed chondrogenesis and osteogenesis. It is also possible that placenta-derived cells and umbilicus-derived cells stimulated chondrocyte migration.
  • Abundant new blood vessels were observed with the scaffolds loaded with placenta-derived cells and umbilicus-derived cells. Blood vessels were abundant in areas of bone formation. New blood vessels were also observed within the hMSC- and fibroblast-seeded scaffolds associated with new bone formation.
  • Systemic effects of the adjacent scaffold (with growth factor (GF)) on the control scaffolds (no GF, no cells) on promoting new cartilage and bone formation cannot be ruled out. Analysis of new cartilage and bone formation in scaffolds, taking into consideration the scaffolds implanted adjacent to it in SCID mice, showed no clear pattern of systemic effect of growth factor from the adjacent scaffold.
  • Summary. Results showed that new cartilage and bone formation were observed in growth factor and control scaffolds seeded with placenta- and umbilicus-derived cells. Results with placenta-derived cells were similar to that seen with human mesenchymal stem cells, while the extent of new cartilage like tissue formation was slightly less pronounced in umbilicus-derived cells. Growth factor-loaded scaffolds implanted without cells also demonstrated new cartilage and bone formation. These data indicate that new cartilage formation within the scaffolds may arise from chondrocytes that migrated from the bovine explants, from chondrogenic differentiation of endogenous progenitor cells, and from chondrogenic differentiation of seeded cells.
  • These results suggest that placenta- and umbilicus-derived cells undergo chondrogenic and osteogenic differentiation. These results also suggest that placenta- and umbilicus-derived cells may promote migration of chondrocytes from the cartilage explant into the scaffolds. Abundant new blood vessels were also observed in the scaffolds especially associated with new bone formation.
  • EXAMPLE 27 Identification and Development of Serum-Free Media for the Isolation and Expansion of Postpartum-Derived Cells
  • Summary: Currently, the most common approaches to growing primary human cells for long-term culture utilize fetal bovine serum (FBS). FBS significantly stimulates cell growth because of the proteins it contains, making it a preferred substrate or supplement for in vitro cell growth. There are, however, a number of disadvantages to using media containing animal products instead of chemically-defined or at least serum-free media. As with all biological products variation in the protein composition of serum from lot to lot and increasing concerns about transmission of diseases such as bovine spongiform encephalopathy provide significant roadblocks to the commercialization or regulatory approval of cell-related products which contain or are produced with serum components.
  • The development of cell culture media formulations free of serum, yet retaining the ability to support cell population expansion sufficient for commercial applications was investigated. A number of media formulations were tested, using short-term or long-term cell proliferation of postpartum-derived umbilical cells as indices of usefulness.
  • Advanced-DMEM (Invitrogen) has been used as a basal medium to optimize serum-free growth of postpartum-derived cells. While serum-free, Advanced-DMEM is not devoid of all animal proteins. Animal protein components contained within Advanced DMEM, such as albumin and insulin, may be replaced with recombinantly-produced alternatives prior to use for making commercialized therapeutic cells or biologics. Postpartum-derived cells (PPDCs) grown from isolation in Advanced-DMEM have been characterized and shown to have reduced expression of PDGFr-alpha and HLA-ABC, relative to that seen in serum-containing media.
  • Introduction: Cell-based therapies have been or are being developed to treat patients with diseases as broad-ranging as myocardial infarction, stroke, ocular diseases like retinitis pigmentosa and macular degeneration, and diabetes. Bankable quantities of cells are required thus stimulating efforts to rapidly and efficiently isolate and expand such cells.
  • Most media for growing primary cultures of human cells incorporate some fetal bovine or calf serum. Generally, commercially available media formulations require serum supplements of about 10-20% (v/v) as this component aids in the survival and expansion of numerous cell populations. A myriad of proteins are found in the serum of cows including PDGF and FGFs, known growth factors that can have important influences on cell growth and differentiation of stem and progenitor cell populations. The disadvantages of using foreign serum products for human therapeutics is discussed above.
  • To overcome the disadvantages of fetal bovine serum, numerous media formulations with and without a variety of human recombinant growth factors known to aid cell expansion were evaluated. Both the short and long-term expansion profiles of postpartum-derived cells were examined and the results are reported herein. While numerous media formulations provided significant short-term benefit, several formulations were useful in expanding these cells over multiple passages.
  • Materials & Methods
  • PPDC Isolation
  • PPDCs were Isolated as in the previous examples.
  • PPC Cell Plating
  • Postpartum-derived cells (umbilicus 022803 (P12)) previously grown in Growth Media were plated at 5,000 cells per cm-squared and weaned of serum over the course of one week by reducing the serum content from 15% to 2%. Cells were then transferred to Growth Medium without serum prior to passage and plating for the short-term MTS assay.
  • MTS Assay to Assess Short-Term Expansion
  • In order to assess a broad range of growth conditions, a MTS assay was used to colorimetrically quantitate DNA content, an indirect measure of cell number and thus cell proliferation. Umbilicus-derived cells (022803 (P11)) were plated at a density of 1,000 cells/well (96-well plate format; equivalent of 5,000 cells/cm-squared, in 100 microliters of medium) in quadruplicate for each condition tested. Both basal media compositions with additives and complete media formulations not requiring supplementation with amino acids were tested. Refer to Table 27-1 for a list of all conditions (note that all media included penicillin/streptomycin in an effective amount, Invitrogen). After four days, samples were treated according to the manufacturer's instructions (Promega MTS Assay Kit).
  • Briefly, 20 microliters of CellTiter Aqueous One Solution Reagent (Promega) were pipetted into each well. Plates were incubated for 1-2 hours at 37 degrees in a humidified incubator (containing 5% carbon dioxide). Following incubation, 25 microliters of 10% SDS were added to each well to stop the formation of the soluble formazin product produced by cellular reduction of MTS. Absorbance was then recorded using a colorimetric absorbance photometer (Spectramax190, Molecular Devices) at both 490 and 700 nanometers.
  • Standard concentration curves were generated, and linear equations were fitted to the curves and utilized to extrapolate cell number based on the averaged values from the samples run in quadruplicate. Graphs were then generated to pictorially represent the sample values and their respective standard errors.
    TABLE 27-1
    GROWTH
    BASAL MEDIUM SUPPLEMENT FACTOR(S)
    Growth Medium (containing 15% FBS) ITS BFGF (F)
    F10 ITS + 1 PDGF (P)
    DMEM/low glucose ITS + 3 EGF (E)
    DMEM/LG + MCDB201 (40%) SITE IGF-1 (I)
    Advanced DMEM (high glucose) + L- SPITE F + P
    glutamine (4 mM)
    UltraCulture (Cambrex) SPIT F + E
    F + I
    P + E
    P + I

    *each basal medium was matched with every possible supplement and growth factor

    Abbreviations:

    ITS = bovine pancreas-derived insulin, human transferrin, and sodium selenite;

    ITS + 1 = ITS components plus bovine serum albumin and linoleic acid;

    ITS + 3 = ITS + 1 components plus oleic acid;

    SITE = ITS components plus ethanolamine:

    SPITE = SITE components plus pyruvate;

    SPIT = ITS components plus pyruvate.
  • Long-Term Expansion Analysis
  • Of the approximate 150 different growth conditions tested in the MTS assay, the 9 best growth conditions plus the control (Growth Medium) were tested for their ability to support the expansion of cell populations over multiple passages. For this analysis, umbilical-derived cells (022803 (P10)) were thawed from cryopreservation (cells were previously grown in Growth Medium) and plated directly into the experimental conditions (see Table 27-2). Media used under all conditions contained penicillin/streptomycin as in the Growth Medium formulation (see above Examples). Cells were passaged every 3-4 days and passaged/trypsinized equivalently in a base medium used for that growth condition (e.g. Advanced DMEM plus bFGF). Growth curves were generated for conditions that supported the growth of umbilical-derived PPCs over greater than 10 passages. Conditions that did not support growth were not considered for further analysis at this time.
    TABLE 27-2
    Growth
    Medium Composition Supplement Factor(s)
    Growth medium 15% fetal none
    bovine
    serum
    (Hyclone,
    Logan, UT)
    DMEM/LG (Gibco) + MCDB 201 (40%) ITS + 3 F
    DMEM/LG (Gibco) + MCDB 201 (40%) ITS + 3 P
    DMEM/LG (Gibco) + MCDB 201 (40%) ITS + 3 E
    DMEM/LG (Gibco) + MCDB 201 (40%) SITE F
    DMEM/LG (Gibco) + MCDB 201 (40%) SITE P
    DMEM/LG (Gibco) + MCDB 201 (40%) SITE E
    Advanced DMEM (high glucose) (Gibco) + L- None F
    glutamine (4 mM)
    Advanced DMEM (high glucose) (Gibco) + L- None P
    glutamine (4 mM)
    Advanced DMEM (high glucose) (Gibco) + L- None E
    glutamine (4 mM)

    Abbreviations:

    refer to table 1.
  • Flow Cytometry
  • Flow cytometric analysis of the markers used in the characterization of umbilical-derived PPCs (see Examples above) was performed to establish whether growth in Advanced DMEM+L-glutamine+bFGF (10 ng/ml) altered surface marker expression as compared to equivalent cells grown in Growth Medium (containing 15% fetal bovine serum). Results of stained samples were compared to controls with no primary antibody to establish differences between positive staining and background fluorescence levels. For these experiments, umbilical-derived cells from several donors 090304A (P4 and P10), 091504A (P4), 063004B (P4), and 042303 (P33) were used.
  • Cryopreservation Analysis
  • Umbilical cord-derived cells (042803 P21) and placenta-derived cells (071503 P10) grown in either Growth Medium or in Advanced DMEM and 10 ng/mL bFGF (Peprotech, Rocky Hill, N.J.)], in a gelatin-coated T225 flask were washed with phosphate buffered saline (PBS; Invitrogen) and trypsinized using 1 mL Trypsin/EDTA (Invitrogen). The trypsinization reaction was diluted by adding 20 mL of Advanced DMEM. Cells were centrifuged at 150×g and the supernatant was aspirated off. The cells were washed in advanced DMEM. Aliquots of cells, 60 μL, were removed and each was added to 60 μL trypan blue (Sigma). The number of viable cells was estimated using a hemocytometer. Cells were then centrifuged at 150×g, supernatant was removed, and the cell pellet was resuspended in the freezing formulation: advanced DMEM, penicillin/streptomycin (Invitrogen) glutamine (4 mM), 10% DMSO (v/v) (Sigma, St Louis, Mo.), 2% (w/v) bovine serum albumin (fraction V, Sigma, St. Louis, Mo.) and 10 ng/mL bFGF (Peprotech) at 1.0×106 cell/mL. 1 mL aliquots were distributed into cryovials (Nalgene). Cells were frozen within 15 minutes of exposure to the freezing medium in a control-rate freezer (CryoMed Freezer 7452, ThermoForma, Marietta, Ohio) using the following protocol:
    Step 1 Wait at 4.0° C.
    Step
    2 1.0° C./min to −4.0° C.
    Step
    3 25.0° C./min to −40° C.
    Step
    4 10.0° C./min to −12.0° C.
    Step
    5 1.0° C./min to −40° C.
    Step
    6 10.0° C./min to −90° C.
    Step
    7 END
  • Vials of cells were transferred to a nitrogen vapor storage unit (CryoMed 7400, ThermoForma, Marietta, Ohio) for 2 days before thawing rapidly in a 37° C. water bath with gentle swirling until no visible ice crystals could be detected. The cells were added to 10 mL Growth Medium and centrifuged before the cell number and viability was estimated as before. Cells were seeded onto gelatin-coated flasks at 5,000 cells/cm2 to determine whether the cells were able to attach and proliferate.
  • Cells were also assayed for senescence using the Senescence Cells Histochemical Staining Kit (Sigma, St Louis, Mo.) according to manufacturer's specification. Briefly, cells were seeded in gelatin-coated 6-well plates at 1.5×105 cells per well in the Growth Medium. After three days cells were washed once with PBS (Invitrogen, Carlsbad, Calif.) and incubated in the fixative solution for 6-7 minutes at room temperature (RT). Cells were then rinsed 3 times with PBS and 1.5 mL of staining solution was added to each well. Plates were incubated at 37 C in a shaker with gentle rocking until cells stained blue. The percentage of blue (senescent) cells was determined.
  • An MTS assay was performed as described above. Similarly, cells were seeded in 8 replicates in 96-well plates at an initial concentration of 1,000 cells per well in the Growth medium (they were not re-seeded in Advanced DMEM).
  • Results
  • Short Term Expansion of Umbilical PPCs
  • Over 150 different media formulations were compared by MTS assay to examine the short-term (1, 3, and 4 day) cell proliferation of passage 11 umbilical-derived PPCs. Cells were plated at 1,000 cells per well.
  • With respect to the basal media (no additives), DMEM/MCDB, F10 medium, Advanced DMEM (containing L-glutamine) and Growth Medium supported umbilical-derived PPC growth over four days in culture. There was a reduction in overall cell number from day 3 to day 4 in UltraCulture medium, and the cells appeared unhealthy.
  • Analysis of the supplements tested indicated that, ITS+3 was superior in the expansion of umbilical-derived PPCs over 4 days, relative to the basal medium alone.
  • Analysis of the growth factors added to the basal media indicated that bFGF, EGF, and PDGF were all individually useful in expanding umbilical PPCs, with bFGF being the growth factor that stimulated the most expansion based on cell number at 4 days. This effect surpassed those results observed with Growth Medium. Interestingly, adding multiple growth factors did not further stimulate expansion under the conditions tested. Converting the data to population doublings between days 1 and 4, PDGF, EGF and IGF surpassed bFGF. This result may be explained by enhanced cell attachment and growth between time zero and day 1 for cells in bFGF.
  • Long Term Expansion Analysis
  • Given the results of the short-term expansion experiments aimed at narrowing the search for a serum-free alternative, 10 media combinations were selected and umbilical-derived PPCs were expanded from passage 10 (previously grown in Growth Medium) at a seeding density of 5,000 cells per centimeter squared. Aside from the control condition (Growth Medium), the only medium combinations that supported the expansion of umbilical-derived PPCs beyond 10 passages was Advanced DMEM supplemented with L-glutamine (4 mM) and bFGF (10 ng/ml). Advanced DMEM continued to support expansion of umbilical-derived PPCs, albeit at a lower rate than Growth Medium, for at least about 23 passages, with these cells eventually senescing at passage 33.
  • Flow Cytometry Results
  • Both early and late passage umbilical cells grown in supplemented Advanced DMEM (containing L-glutamine and bFGF, 10 ng/ml) were stained for a variety of cell surface markers previously used to characterize PPDCs grown in Growth Medium. Umbilical-derived PPCs grown in Growth Medium were used for comparison to evaluate differences attributable to the medium composition. While the expression of most markers was similar, two markers differed in their expression between media compositions. Both HLA-ABC and PDGFr-alpha were expressed in Growth Medium, but neither was expressed in Advanced DMEM. (Tables 27-3 & 4). This was observed with both early (P4) and late passage (P33) umbilical-derived cells.
    TABLE 27-3
    Medium Growth Medium
    Tissue Umbilical
    Sample ID 090304A
    Passage
    4
    Present/
    Absent Mean Geo Mean
    CD10-PE + 147.44 91.89
    CD13-PE + 479.09 375.84
    CD31-PE 1.66 1.46
    CD34-FITC 1.65 1.50
    CD44-FITC + 285.41 253.22
    CD45-PE 1.70 1.50
    CD73-PE + 296 250.27
    CD90-FITC na na Na
    CD117-PE 1.84 1.58
    CD141-PE 2.64 2.12
    SSEA-4-PE +/− 32.44 11.84
    PD-L2-FITC +/− 10.44 8.30
    PDGFRalpha-PE +/− 10.81 8.76
    HLA-ABC-PE + 152.15 124.92
    HLA-DRDPDQ-FITC 1.86 1.63
    Controls Mean Geo Mean
    IgG-PE 2.40 2.03
    IgG-FITC 1.86 1.66

    *na = not available
  • TABLE 27-4
    Medium AdvDMEM + bFGF
    Tissue Umbilical
    Sample ID 91504
    Passage 4
    Present/
    Absent Mean Geo Mean
    CD10-PE + 27.50 24.49
    CD13-PE + 20.98 16.87
    CD31-PE 3.23 2.73
    CD34-FITC 2.77 2.43
    CD44-FITC + 10.37 8.11
    CD45-PE 3.12 2.67
    CD73-PE + 11.66 9.46
    CD90-FITC + 160.33 136.83
    CD117-PE 3.22 2.69
    CD141-PE 3.29 2.79
    SSEA-4-PE 3.39 2.93
    PD-L2-FITC Na na Na
    PDGFRalpha-PE 2.91 2.50
    HLA-ABC-PE 5.00 4.62
    HLA-DRDPDQ-FITC 2.66 2.34
    Controls Mean Geo Mean
    IgG-PE 3.19 2.73
    IgG-FITC 2.8 2.46

    *na = not available
  • Cryopreservation Results
  • The initial viability of the cells as assessed by trypan blue staining prior to cryopreservation was close to 100%. Viability of cells post thawing and % senescent cells is summarized in Table 27-5.
    TABLE 27-5
    Medium before Recovery
    Cell type freezing rate Senescence
    Placenta-derived Growth medium 94% ˜3%
    Adv DMEM + bFGF 98% <1%
    Umbilical-derived Growth medium 77% ˜7%
    Adv DMEM + bFGF 86% ˜7%
  • There was a slight reduction in cell viability in both placenta-derived and umbilical cord-derived cells grown in the Growth Medium prior to cryopreservation. The viable cells cryopreserved in the Advanced DMEM-based medium divided and produced a confluent monolayer within 3 days. There were no discernable differences in growth rates (as determined from the MTS measurements). The senescence assay revealed <1% and 3% senescent cell population of placenta-derived cells grown in Advanced DMEM and Growth Medium respectively. For umbilical cord-derived cells the percentages of senescent cells were slightly higher, approximately 7%.
  • Discussion and Conclusions
  • The data indicate that in addition to the currently used Growth Medium (containing 15% fetal bovine serum) there are media formulations that support the growth of postpartum-derived cells. Of those studied, the best defined medium formulation was Advanced DMEM (Invitrogen) containing high glucose supplemented with 10 ng/ml bFGF. This medium could support both the short and long term expansion of passage 10 umbilical-derived PPDCs previously grown in Growth Medium. Although the cells grew and the populations expanded well, growth was not as substantial as that obtained in Growth Medium with the serum supplementation.
  • Another unexpected finding was that Advanced DMEM grown cells lacked expression of HLA-ABC, whereas equivalent cells grown in Growth Medium did express this marker. Whether this has implications for influencing (e.g. reducing) graft rejection is unclear and will require animal studies with cells grown in serum-free media. The change in HLA-ABC and PDGFr markers may influence pre-clinical efficacy. Preliminarily, however, it would be beneficial to examine changes in gene expression as a result of cell growth in these two media formulations.
  • Finally, an analysis of the ability of such cells to be cryopreserved suggests that Advanced DMEM media may be of benefit in reducing the number of non-viable cells (compared with Growth Medium). For both umbilicus-derived and placental-derived PPCs, recovery rate was increased, albeit modestly. The seemingly lower recovery rate for umbilical cord-derived cells versus placenta-derived cells could be due to higher passage (21) of the cells which was also reflected in the senescence assay results. Thus, umbilicus-derived cells grown in Growth Medium can be cryopreserved in Advanced DMEM with certain advantages.
  • EXAMPLE 28 Further Identification and Development of Serum-Free Media for the Isolation and Expansion of Postpartum-Derived Cells
  • Summary: The prior example described Advanced-DMEM (Invitrogen) as a useful basal medium to optimize for the serum-free growth of postpartum-derived cells. Umbilical-derived cells grown from isolation in Advanced-DMEM were characterized and shown to have reduced expression of PDGFr-alpha and HLA-ABC. Postpartum-derived cells grown in both Advanced DMEM and Growth Medium were further characterized using flow cytometry to determine the effect of growth medium on cell surface marker expression. Gene expression profiles were compared with those of other cells including dermal and foreskin fibroblasts, and mesenchymal stem cells (MSCs). Switching media can alter both HLA-ABC and PDGFr expression as determined by flow cytometry. The expression patterns for particular “signature genes” (described in Examples herein) was similar in both media. This confirms that umbilical cord-derived postpartum cells are unique cells from isolation that can be propagated in a defined medium without any apparent change in the expression of these “signature” genes. Alteration of cell surface marker expression of HLA-ABC may impact graft or implantation incorporation or rejection.
  • Introduction: HLA-ABC and PDGFr alpha expression was shown to be altered when postpartum cells were grown in two different media (Growth Medium versus Advanced DMEM supplemented with basic fibroblast growth factor (bFGF)). This finding was interesting and triggered further investigations into whether this phenomenon was due, for example, to the promotion of a different subpopulation of postpartum-derived cells, or whether a specific population of postpartum cells had the ability to modulate the expression of these cell surface markers in different growth media.
  • To investigate this question, two sets of experiments were performed. In the first, umbilicus-derived postpartum cells were grown in Growth Medium, Advanced DMEM plus bFGF, or switched between the two, and then analyzed for HLA-ABC and PDGFr expression by flow cytometry. RNA was isolated from cultures of these cells grown under identical conditions, and PCR was performed to determine the expression patterns of genes previously found to be exclusively expressed (versus other cell populations). Differences between media conditions were analyzed, as were differences between other control cell types.
  • The results of these experiments indicate that while the postpartum-derived cells grown in both Growth Medium and the Advanced DMEM Medium express similar patterns of genes, their cell surface marker expression may be altered between the two media. This is important for many reasons including distinguishing various postpartum-derived cells from each other as well from other stem type cells of adult, fetal or neonatal origin. The method described herein for reducing expression of HLA-ABC also opens significant opportunities for producing cells with improved properties for grafts or implantation. Such a method could reduce the chance of graft rejection in the absence of an immunosuppressant like cyclosporin A.
  • Materials & Methods
  • PPC Isolation
  • Cells were isolated as described herein throughout the above Examples.
  • PPC Cell Plating
  • Postpartum-derived cells (umbilicus 063004B) grown in Advanced DMEM supplemented with bFGF (as described in Example 27) from isolation were passaged at either P3 or P4, and at that point switched to Growth Medium. Cells were plated at 5,000 cells per cm-squared on standard gelatin coated tissue culture flasks. Flow cytometry was performed on P3 cells for PDGFr alpha, HLA-ABC, and HLA-DRDPDQ, and the respective controls lacking primary antibody (IgG-PE, and IgG-FITC). P4 passaged/media switched cells were grown until passage 8 by passaging every 3 or 4 days, at which time flow cytometry was performed for the cell surface markers.
  • The reciprocal experiment was also performed in which cells grown in Growth Medium were switched to Advanced DMEM supplemented with bFGF (10 ng/ml). Umbilicus-derived (063004B) postpartum cells were grown in Growth Medium from isolation until passage 12. At passage 13, cells were switched to Advanced DMEM+bFGF and grown until passage 14, at which time flow cytometry was performed. Similarly, umbilicus-derived (090304A) postpartum cells were grown in Growth Medium from isolation until passage 8, and then switched at passage 9 to Advanced DMEM+bFGF. Cultures were maintained until passage 10 at which time flow cytometry was performed. Finally, umbilicus-derived (042303) postpartum cells were grown in Growth Medium from isolation until passage 10, at which time they were cryopreserved in Growth Medium supplemented with 10% DMSO (Sigma, St. Louis, Mo.) and placed in a liquid nitrogen container (protocol as described in Example 27). Cells were later thawed and directly plated at 5,000 cells per cm-squared on gelatin coated flasks in Advanced DMEM supplemented with bFGF (10 ng/ml). The cultures were maintained until passage 33, at which time flow cytometry was performed for the cell surface markers of interest.
  • Flow Cytometry
  • Flow cytometric analysis of the markers of interest was performed to determine whether growth in Advanced DMEM+bFGF (10 ng/ml) altered surface marker expression as compared to equivalent cells grown in Growth Medium (with its 15% fetal bovine serum). Results of stained samples were compared to the appropriate IgG controls to establish differences between positive staining and background fluorescence levels.
  • Control Cell Growth Conditions
  • Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) were grown in Growth Medium in gelatin-coated T75 flasks. Mesenchymal Stem Cells (MSCs, Cambrex, Walkersville, Md.) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex).
  • Total RNA Isolation
  • RNA was extracted from confluent umbilical cord-derived cells grown in different conditions. RNA was also extracted from fibroblasts and MSCs. Cells were lysed with 350 μL buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.) and RNA extracted according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.) with a 2.7 U/sample DNase treatment (Sigma St. Louis, Mo.). RNA was eluted with 50 μL DEPC-treated water and stored at −80° C.
  • Reverse Transcription
  • RNA was reversed transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) using a temperature/time cycle of 25° C. for 10 minutes, 37° C. for 60 minutes, and 95° C. for 10 minutes. Samples were stored at −20° C. Genes, termed “signature genes” (oxidized LDL receptor, interleukin-8, renin and reticulon), were further investigated using real-time and conventional PCR.
  • Real-Time PCR
  • PCR was performed on cDNA samples using Assays-on-Demand™ gene expression products: oxidized LDL receptor (Hs00234028), renin (Hs00166915), reticulon (Hs00382515), IL-8 (Hs00174103) and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 min and 95° C. for 10 min followed by 40 cycles of 95° C. for 15 sec and 60° C. for 1 min.
  • Conventional PCR
  • Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass., USA) to confirm the results from real-time PCR. PCR was performed using 2 μL of cDNA solution, 1× AmpliTaq Gold universal mix PCR reaction buffer (Applied Biosystems, Foster City, Calif.) and initial denaturation at 94° C. for 5 minutes. Amplification was optimized for each primer set. For IL-8, and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles), for renin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C. for 30 seconds for 38 cycles) for oxidized LDL receptor and GAPDH (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 33 cycles). Primers used for amplification are listed in Table 28-1. Primer concentration in the final PCR reaction was 1 μM except for GAPDH which was 0.5 μM. GAPDH primers were the same as real-time PCR, except that the manufacturer's TaqMan probe was not added to the final PCR reaction. Samples were run on 2% (w/v) agarose gel and stained with ethidium bromide (Sigma, St. Louis, Mo.). Images were captured using a 667 Universal Twinpack film (VWR International, South Plainfield, N.J.) using a focal-length Polaroid™ camera (VWR International, South Plainfield, N.J.).
    TABLE 28-1
    Primers used
    Primer name Primers
    Oxidized S: 5′-GAGAAATCCAAAGAGCAAATGG-3′ (SEQ ID NO:1)
    LDL receptor A: 5′-AGAATGGAAAACTGGAATAGG-3′ (SEQ ID NO:2)
    Renin S: 5′-TCTTCGATGCTTCGGATTCC-3′ (SEQ ID NO:3)
    A: 5′-GAATTCTCGGAATCTCTGTTG-3′ (SEQ ID NO:4)
    Reticulon S: 5′-TTACAAGCAGTGCAGAAAACC-3′ (SEQ ID NO:5)
    A: 5′-AGTAAACATTGAAACCACAGCC-3′ (SEQ ID NO:6)
    Interleukin-8 S: 5′-TCTGCAGCTCTGTGTGAAGG-3′ (SEQ ID NO:7)
    A: 5′-CTTCAAAAACTTCTCCACAACC-3′ (SEQ ID NO:8)
  • Results
  • Differences in Cell Surface Marker Expression Between Cell Populations Grown in Advanced DMEM and Growth Medium from Isolation
  • In Example 27, umbilicus-derived postpartum cells grown in Growth Medium and Advanced DMEM+bFGF from isolation were tested utilizing flow cytometry for a full battery of antibodies to cell surface markers. While the expression of most markers was similar, two markers substantially differed in their expression between the media formulations. Both HLA-ABC and PDGFr-alpha were expressed in Growth Medium, whereas neither was expressed in Advanced DMEM (Tables 27-3 & 27-4 in Example 27).
  • Impact of Media Switching on Cell Surface Marker Expression
  • To ascertain whether the media compositions were responsible for altering cell surface marker expression, umbilicus-derived postpartum cultures were first grown in one of the two aforementioned media compositions and then switched to the other. When cells were grown initially in Advanced DMEM+bFGF and then switched to Growth Medium, cell cultures immediately turned on expression of HLA-ABC in a graded fashion with increasing passage. PDGFreceptor alpha, however, did not appear to turn on, even with extended passage.
  • When cells were first grown in Growth Medium and then later switched to Advanced DMEM+bFGF, HLA-ABC expression was retained in the short term (063004B+P14, 090304A+P10), but after several passages, HLA-ABC expression was lost (042303+P33). In comparison to cells grown exclusively in Growth Medium from isolation, PDGF receptor alpha expression was lost as early as the second passage in Advanced DMEM+bFGF.
  • In addition to HLA-ABC and PDGFr alpha, each of these isolates (except 042303+P33) was tested for HLA-DRDPDQ expression. All isolates were negative regardless of medium they were maintained in.
  • PCR Results
  • Results of real-time PCR for selected “signature” genes performed on cDNA from cells derived from human umbilical cord-derived cells grown in two different media, adult fibroblasts, and MSCs indicate that the levels of reticulon and oxidized LDL receptor RNA were higher in umbilical cord-derived cells compared to other cell types. The results also confirmed that the expression pattern of signature genes did not change with medium regardless of whether cells were cultured in Advanced DMEM+10 ng/mL bFGF, or in Growth Medium from isolation, or were subsequently switched from one medium to the other. The results of real-time PCR were confirmed by conventional PCR. Of note, was that bands, while existent, were less intense for oxLDL receptor (063004B P14, 090304A P10). Since conventional PCR is not quantitative, however, potential differences in level of expression could not be discerned here. Umbilicus-derived cells did not express renin, a marker present in placental-derived cells.
  • The expression of the cytokine, IL-8, in postpartum-derived cells was elevated in both Growth Medium-cultured and Advanced DMEM+10 ng/mL bFGF-cultured umbilical cord-derived cells. This observation was confirmed by conventional PCR.
  • Discussion and Conclusions
  • Switching cells between Growth Medium and Advanced DMEM supplemented with bFGF (10 ng/ml) impacts cell surface marker expression of HLA-ABC, and PDGF-r alpha in PPDCs. Under the conditions tested, HLA-DRDPDQ was not detected. HLA-ABC and PDGFr alpha expression can be modulated based on changes in the growth medium of the cells. This observation could have significant implications for the rejections of grafts or implantation of therapeutic cells. The HLA-DRDPDQ marker was never expressed regardless of media composition.
  • The expression of signature genes such as oxidized LDL receptor and reticulon were unchanged despite the composition of medium used to grow these umbilicus-derived postpartum cells. Because the levels of these genes were detectably higher than in other cell types such as MSCs, these genes in fact constitute a useful signature to determine and distinguish umbilicus-derived postpartum cells. Whether separate subpopulations may exist with in the umbilicus-derived population remains a question, but the consistency of the signature gene expression profile is powerful evidence that any such subpopulations are closely related to each other and more distantly related if at all to other cell types tested.
  • The finding that HLA-ABC expression varied suggests that, in fact, HLA-ABC expression can be modulated. Whether this could impact graft rejection is a point of further interest in that a cell population lacking the expression of this marker may be able to engraft better, potentially in the absence of immunosuppressants.
  • The finding that PDGFr alpha can be shut off easily and cannot be immediately turned on may indicate the delicate nature and rapid turnover of growth factor receptors and their requirement for very particular growth conditions for their expression. Given a lack of such receptors (or any other growth factor receptor because of, for example, media changes), cell potency may be altered in particular animal model paradigms.
  • EXAMPLE 29 Growth Characteristics of Umbilicus-Derived Cells in Standard Growth Medium and Growth Medium without BME, and on Gelatin Coated and CELLBIND Coated Flasks
  • As described herein and above, umbilicus-derived cells can be cultured in Growth Medium in flasks with gelatin-coated surfaces. This example explored whether two of the components in a preferred cell culture protocol were required. Tested, in particular, were whether (1) beta-mercaptoethanol (BME), as an additive in the standard Growth Medium, was required for growth, and (2) whether gelatin (an animal derived protein) was required as a coating for the cell culture flasks. Additionally, the effectiveness of a synthetic coating to promote attachment, CELLBIND® (Corning, Corning, N.Y.), was tested as an alternative to gelatin or other animal-derived products. CellBIND® is a patented, commercially available flask coating that uses highly reactive plasma to reduce the aromatic groups of the polystyrene surface.
  • Materials & Methods
  • Reagents: Dulbecco's Modified Essential Media (DMEM), Penicillin and Streptomycin, were obtained from Invitrogen (Carlsbad, Calif.). Fetal bovine serum (FBS) (defined bovine serum) was obtained from Hyclone (Logan, Utah). Beta-mercaptoethanol was obtained from Sigma (St. Louis, Mo.).
  • Experimental Design: Umbilicus-derived cells were isolated in two mediums (Growth Medium, and Growth Medium without BME) and in vessels coated with either gelatin or CELLBIND®). Thus, four sets of conditions were tested: (1) Gelatin-Coating/Growth Medium (normal conditions), (2) CELLBIND®-Coating/Growth Medium, (3) Gelatin-Coating/Growth Medium without BME and (4) CelBIND®-Coating/Growth Medium without BME. The cells were serially passaged from isolation until senescence in their designated set of conditions. Population growth kinetics were calculated under each set of conditions to determine the effects of media and surface coating of the vessels, and to assess changes in growth. The expression of a set of cell surface markers was assessed as an initial screen for phenotypic changes.
  • Cells. A single umbilical cord was obtained through National Disease Research Interchange (Philadelphia, Pa.) with full consent and divided into two pieces of approximately equal size.
  • Isolation Procedures and Media: Postpartum cells were generally isolated as described previously herein. The tissue was minced and divided into approximately equal portions. One portion was isolated in standard Growth Medium (DMEM:LG, 15% FBS; beta-mercaptoethanol (1 μl per 100 mL); 50 microliter/ml penicillin/50 microgram/ml streptomycin (1 mL per 100 mL (10,000 Units per mL)). The other portion was isolated in Growth Medium as described without beta-mercaptoethanol (BME) (Growth Medium w/o BME).
  • Growth of Cells: Cells isolated from each portion of the cord tissue were seeded at 3000 cells/cm2 onto gelatin-coated T225 flasks and CELLBIND-coated T225 flasks (Corning) in the same medium in which they were isolated. Cells were grown at 37° C. in a humidified, 5% CO2 atmosphere. As cells reached about 85% confluence they were trypsinized and counted. Viable cells were seeded at 5000 cell/cm2 into new flasks with appropriate medium and surface coating. Cell Yield, population doublings (ln(cell final/cell initial)/ln2) and doubling time (time in culture (h)/population doubling) were determined at each passage. The process was repeated until cells appeared to reach senescence, i.e. the cells appeared or lose their proliferative capabilities, or failed to proliferate further.
  • Analysis of Cell Surface Markers: Cell surface markers were analyzed by flow cytometery essentially as described herein above. Analyses were performed at passages 3, 7 and 15 for cells grown on gelatin in either standard Growth Medium or in Growth Medium without BME, and at passage 7 for cells grown on CellBIND-coatings in either Growth Medium or Growth Medium without BME. In particular, after trypsinization and counting, 500,000 cells were suspended in 100 μl of Phosphate Buffered Saline with 3% Fetal Bovine Serum per sample. To each sample 20 μl of the antibody corresponding to the marker being analyzed was added. The cells were incubated at 4° C. for 1 hour. After incubation the cells were washed once with 3 ml PBS and resuspended in 0.5 ml PBS. The samples were analyzed using a FACSCalibur (Becton Dickinson), and the results were compiled using the accompanying software (Cell Quest, BD Applications).
  • Results
  • Cells isolated from umbilical cords can be cultured in media with or without BME, and on either gelatin-coated or CELLBIND-coated vessels. The greatest rate of proliferation (0.51 doublings/day) in these experiments was observed with cells cultured in Growth Medium without BME on gelatin coated flasks. Umbilicus-derived cells proliferated at higher rates in Growth Medium without BME as reflected by a greater number of doublings per day; 0.51 doublings per day (Growth Medium without BME/Gelatin) compared to 0.33 doublings per day (Standard Growth Medium/Gelatin). Cells grew on the CellBIND-coated surfaces in medium with or without BME.
  • Umbilicus-derived cells cultured in gelatin-coated vessels in either Growth Medium or in Growth Medium without BME were able to expand for up to about 120 days. Umbilicus-derived cells also grew in either Growth Medium or Growth Medium without BME, on the CELLBIND-coated surface for up to about 103 days.
  • Cell surface markers were analyzed by flow cytometry at passages 3, 7 and 15 for cells grown on gelatin-coated vessels in media with or without BME, and at passage 7 for cells grown on CELLBIND-coated in either media. The results showed no evidence of any phenotypic changes as reflected in the expression pattern of the cell surface markers analyzed. The results are shown below in Table 29-1.
    TABLE 29-1
    Cell surface marker expression for cell populations grown in different
    conditions at passage 7. Results at passages 3 and 15 are similar.
    Passage P7
    Medium No BME Hayflick No BME Hayflick
    Growth Surface Gelatin Gelatin CellBIND ® CellBIND ®
    Markers CD10-PE + + + +
    CD13-PE + + + +
    CD31-PE
    CD34-FITC
    CD44-FITC + + + +
    CD45RA-PE
    CD73-PE + + + +
    CD90-FITC + + + +
    CD117-PE
    CD141-PE
    SSEA-4-PE + + NA +/−
    PD-L2-FITC +/− +/− +/−
    PDGFRalpha-PE +/− −(slight + shift) +/−
    HLA-ABC-PE + + + +
    HLA-DP DQ DR-FITC
  • Summary. The results provide evidence that umbilicus-derived cells can be grown in culture without BME.
  • EXAMPLE 30 Alternate Conditions for the Isolation of Cells from Umbilical Cord Tissue
  • Initial efforts to isolate a population of cells from human umbilical cord tissue were conducted using a method of tissue processing with a basic cell culture media formulation containing serum as described herein above. Here, alternative methods for isolating cells from minced umbilical cord tissue in the presence of several different media formulations and culture conditions are described. Another objective was to determine if alternate isolation conditions allowed for the outgrowth of a new cell type not previously characterized. Umbilicus-derived cells, grown from isolation under these different conditions were characterized as to their: (1) long-term growth potential, (2) cell surface marker phenotype, (3) stem cell-specific gene expression, and (4) trophic factor production. The data demonstrate that umbilicus-derived cells can be isolated using a variety of media formulations and growth conditions.
  • Materials & Methods
  • Reagents. Dulbecco's Modified Essential Media (DMEM), Phosphate buffered saline, Penicillin and Streptomycin, and dispase were obtained from Invitrogen (Carlsbad, Calif.). Fetal bovine serum (FBS) (defined bovine serum) was obtained from Hyclone (Logan, Utah). Collagenase, hyaluronidase, and beta-mercaptoethanol were obtained from Sigma (St. Louis, Mo.).
  • Isolation of cells from human umbilical cords: Human umbilical cords were obtained from the National Disease Research Interchange (NDRI, Philadelphia, Pa.) following normal deliveries. To remove blood and debris, the cord was washed in Dulbeco's Modified Eagles Medium (DMEM-low glucose) or phosphate buffered saline (PBS). The tissues were then mechanically dissociated in tissue culture plates until the tissue was minced to a fine pulp. The minced tissue was transferred to a 50-milliliter conical tube and digested in an enzyme mixture containing 500 Units/milliliter collagenase, 500 Units/milliliter dispase, and 50 Units/milliliter hyaluronidase. The enzyme mixture was combined with a specific media formulation (“test culture medium”) as outlined in Table 30-1 below. The conical tubes containing the tissue, medium, and digestion enzymes were incubated at 37° C. in an orbital shaker at 225 rpm for 2 hrs.
  • After digestion, the tissues were centrifuged at 150×g for 5 minutes, and the supernatant was aspirated. The pellet was resuspended in 20 milliliters of test culture media (Table 30-1). The cell suspension was filtered through a 40-micron nylon BD FALCON Cell strainer (BD Biosciences, San Jose, Calif.). The filtrate was resuspended in medium (total volume=50 milliliters) and centrifuged at 150×g for 5 minutes. The supernatant was again aspirated, and the cells were resuspended in 50 milliliters of fresh test culture medium. The process was repeated twice.
  • After the final centrifugation, the supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh test culture medium. The number of viable cells was determined using a Guava instrument (Guava Technologies, Hayward, Calif.). Cells were then plated at seeding densities of 300 cells/cm2, 2000 cells/cm2, or 5000 cells/cm2 on gelatin-coated tissue culture flasks. Cells were cultured in medium and under conditions described in Table 30-1. Growth Media controls were used to determine viability of the isolates for each media type.
    TABLE 30-1
    Media formulations and culture conditions used to isolate umbilical
    cells from umbilical cord tissue.
    Culture
    Isolation Isolate Media type condition Doublings
    1 Umb042105 EGM2 (endothelial growth medium-2 (Cambrex BioScience, MD)) Normoxia (low >5
    density)
    2 Umb042105 EGM2 Normoxia (high >5
    density)
    3 Umb052405 REGM (Renal epithelial growth medium (Cambrex)) Hypoxia >5
    4 Umb052405 REGM Normoxia >5
    5 Umb032905 SmGM2 (smooth muscle growth medium-2 (Cambrex) Normoxia (low 5
    density)
    6 Umb051705 SMGM2 Hypoxia (high >5
    density)
    7 Umb051705 SMGM2 Hypoxia (low >5
    density)
    8 Umb032905 SMGM2 Normoxia (high 5
    density)
    9 Umb060805 Sonic hedgehog medium (DMEM-Low glucose (Invitrogen), 2 percent Hypoxia 5
    (v/v) fetal bovine serum (FBS; defined fetal bovine serum; Lot
    #AND18475; (Hyclone), penicillin at 100 Units/mL, streptomycin at 100 ug/mL
    (both from Invitrogen), 10 ng/mL recombinant sonic hedgehog
    growth factor (R&D Systems Inc., Minneapolis, MN)).
    10 Umb080305 Keratinocyte growth medium (Gibco) Normoxia 5

    “Hypoxia”, indicates cultures grown under an atmosphere containing 5% oxygen.

    “Normoxia”, indicates cultures were grown in normal atmospheric oxygen conditions.

    “Low density”, indicates that cultures were initially seeded at 300 cells/cm2.

    “High density”, indicates that cultures were initially seeded at 2000 cells/cm2.

    “Doublings” = approximate number of population doublings at passage 4.
  • FACs Analysis: Flow cytometry analysis was performed on umbilicus-derived cells grown according the media formulations and culture conditions described in Table 30-1. Cells were expanded to passage four or five in test culture medium on gelatin-coated T225 flasks at 37° C. and 5% carbon dioxide. Adherent cells were washed in PBS and detached with Trypsin/EDTA (Gibco). Cells were harvested, centrifuged and resuspended in 3% (v/v) FBS in PBS at a concentration of 1×107 cells/mL. The specific antibody was added to 100 microliters of cell suspension and the mixture was incubated in the dark for 30-45 minutes at 4° C. After incubation, cells were washed with PBS and then centrifuged to remove excess antibody. Cells were resuspended in PBS (500 microliters) and analyzed by flow cytometry. Flow cytometry analysis was performed with a FACSCalibur instrument (Becton Dickinson, San Jose, Calif.). Antibodies used are shown in Table 30-2.
    TABLE 30-2
    Antibodies used for cell surface marker
    analysis of umbilical derived cells.
    Antibody Manufacture Catalog No.
    CD10 BD Pharmingen (San Diego, CA) 555375
    CD13 BD Pharmingen 555394
    CD31 BD Pharmingen 555446
    CD34 BD Pharmingen 555821
    CD44 BD Pharmingen 555478
    CD45R BD Pharmingen 555489
    CD49c BD Pharmingen 556025
    CD73 BD Pharmingen 550257
    CD90 BD Pharmingen 555596
    CD117 BD Pharmingen 340529
    CD141 BD Pharmingen 559781
    CD184 BD Pharmingen 555974
    HLA-A, B, C BD Pharmingen 555553
    HLA-DR, DP, DG BD Pharmingen 555558
    IgG-FITC BD Pharmingen 555748
    IgG-PE BD Pharmingen 555749
  • Total RNA isolation: RNA was extracted from confluent umbilical cord derived cells grown in different conditions (RNeasy Mini Kit; Qiagen, Valencia, Calif.). RNA was eluted with 50 μL DEPC-treated water and stored at −80° C.
  • Reverse transcription: RNA was reverse transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C. for 10 minutes, 37° C. for 60 minutes and 95° C. for 10 minutes. Samples were stored at −20° C. The expression of oxidized LDL receptor, interleukin-8 and reticulon were investigated using real-time and conventional PCR.
  • Real-time PCR: PCR was performed on cDNA samples using Assays-on-Demand™ gene expression products: renin (Hs00166915), oxidized LDL receptor (Hs00234028), reticulon (Hs00382515), IL-8 (Hs00174103), and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 min and 95° C. for 10 min followed by 40 cycles of 95° C. for 15 sec and 60° C. for 1 min.
  • Conventional PCR: Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass., USA) to confirm the results from real-time PCR. PCR was performed using 2 μL of cDNA solution, 1× AmpliTaq Gold universal mix PCR reaction buffer (Applied Biosystems, Foster City, Calif.) and initial denaturation at 94° C. for 5 minutes. Amplification was optimized for each primer set. For IL-8 and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles), for renin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C. for 30 seconds for 38 cycles) for oxidized LDL receptor and GAPDH (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 33 cycles). Primers used for amplification are listed in Table 28-3. Primer concentration in the final PCR reaction was 1 μM except for GAPDH which was 0.5 μM. GAPDH primers were the same as real-time PCR, except that the manufacturer's TaqMan probe was not added to the final PCR reaction. Samples were run on 2% (w/v) agarose gel and stained with ethidium bromide (Sigma, St. Louis, Mo.). Images were captured using 667 Universal Twinpack film (VWR International, South Plainfield, N.J.) using a focal-length camera (VWR International, South Plainfield, N.J.).
    TABLE 30-3
    Primers used for PCR
    Primer sequence
    Primer name (F = forward, R = reverse)
    Renin F: 5′-TCTTCGATGCTTCGGATTCC-3′ (SEQ ID NO:3)
    R: 5′-GAATTCTCGGAATCTCTGTTG-3′ (SEQ ID NO:4)
    Oxidized F: 5′-GAGAAATCCAAAGAGCAAATGG-3′ (SEQ ID NO:1)
    LDL receptor R: 5′-AGAATGGAAAACTGGAATAGG-3′ (SEQ ID NO:2)
    Reticulon F: 5′-TTACAAGCAGTGCAGAAAACC-3′ (SEQ ID NO:5)
    R: 5′-AGTAAACATTGAAACCACAGCC-3′ (SEQ ID NO:6)
    Interleukin-8 F: 5′-TCTGCAGCTCTGTGTGAAGG-3′ (SEQ ID NO:7)
    R: 5′-CTTCAAAAACTTCTCCACAACC-3′ (SEQ ID NO:8)
  • ELISA: Frozen umbilicus-derived cells from different isolation conditions were thawed at passage 4 and seeded onto gelatin coated T75 flasks at 5000 cells/cm2 each containing 15 milliliters of their respective growth medium (Table 30-1). Cells were cultured for 24 hours at 37° C. in 5% carbon dioxide and atmospheric oxygen. The medium was then changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 Units/milliliter) and streptomycin (50 ug/mL, Gibco)) and further cultured for 8 hours. Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000×g for 5 min and stored at −20° C.
  • To estimate the number of cells in each flask, cells were washed with PBS and detached using 2 milliliters trypsin/EDTA (Gibco). Cell number was estimated using the Guava instrument (Guava Technologies Hayward, Calif.)). Samples were then assayed for the following factors: tissue inhibitor of metalloprotease-1 (TIMP1), platelet-derived epithelial growth factor bb (PDGFbb), Keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), Heparin-binding epidermal growth factor (HB-EGF), tissue inhibitor of metalloprotease-2 (TIMP2), monocyte chemotactic protein-1 (MCP1), interleukin-6 (IL6), interleukin-8 (IL8), transforming growth factor alpha (TGFa), brain-derived neurotrophic factor (BDNF), stromal-derived factor 1B (SDF1B), cilliary neurotrophic factor (CNTF), basic nerve growth factor (bNGF), neurotrophin-3 (NT3) with the Searchlight Proteome Arrays (Pierce Biotechnology Inc.).
  • Results
  • Isolation of Umbilicus-Derived Cells
  • Umbilicus-derived cells were isolated using different media compositions and culture conditions identified in Table 30-1. Some isolated cell populations were cultured under both normoxic and hypoxic (5% O2) atmospheric conditions. Cells were also isolated using low (300 cell s/cm2) or high (2000 cells/cm2) initial seeding density. Ten combinations of growth medium and growth conditions supported the growth of umbilicus-derived cells. These include EGM2, REGM and SMGM2, all under both normoxic and hypoxic atmospheric conditions. Sonic hedgehog medium and KGM, under hypoxia and normoxia respectively, also supported cell growth.
  • Long-Term Expansion Analysis
  • Media formulations that supported the growth of umbilicus-derived cells were selected, and cells were expanded from passage 4 or 5, at a seeding density of 5,000 cells/cm2 on gelatin-coated plates. Cell populations were continually passaged for several weeks until senescence was reached. Senescence was determined when cells failed to achieve greater than one population doubling the study time interval.
  • Several isolates were tested for long term expansion potential and demonstrated growth beyond passage 4 (Table 30-4). Umb032905, isolated in SMGM2 medium under hypoxic conditions, demonstrated the most robust cell expansion potential of greater than 32 population doublings. The population reached senescence at day 74 for cells isolated at low seeding density, and at 64 days for cells isolated at high seeding density. Modest expansion potential was observed in umb052405 isolated in REGM and cultured in normoxic or hypoxic conditions (isolation 3,4). These conditions yielded 18 and 10 population doublings, respectively.
    TABLE 30-4
    Maximal number of cell population doublings achieved.
    Isolation Days in culture PD Viability (%)
    3 33 18.3 86.5
    4 29 10.2 97
    6 64 31.4 98
    7 74 31.6 96

    “PD”, number of population doublings achieved when cells were cultured from passage 4 to senescence.

    “Viability (%)”, percent of cells that were alive at the time of senescence.
  • Surface Marker Phenotype Analysis by Flow Cytometry
  • Cell populations generated from different isolation conditions were characterized by flow cytometry. Ten cell lines out of the twenty-two isolation conditions were expanded to passage 4 and cell surface marker profile was determined. Most of the cell isolations showed positive staining for CD10, CD13, CD44, CD49c, CD73, CD90, and HLA-ABC. The cells were negative under the conditions tested for surface expression of CD31, CD34, CD45, CD117, CD141, CD184, and HLA-DR, DP, DQ. Most markers showed a similar expression pattern to cells isolated using the preferred conditions described above in prior examples, as well as to other cell isolations.
  • RT-PCR Analysis
  • Umbilicus-derived cell isolates were analyzed for the expression of umbilicus-derived cell-specific genes and stem cell-specific genes. The data provided in Tables 30-5 show that cells derived from all isolation conditions expressed reticulon, LDL-R, and IL-8. Table 30-5 shows the results of real-time PCR performed on cDNA from cells derived from human umbilical cords grown in different media formulations. Results are expressed as a ratio of cycle thresholds (CT) between gene of interest and the internal transcript control, GAPDH. The data indicate that expression levels of reticulon, oxidized LDL receptor, and IL-8 were all similar.
    TABLE 30-5
    Results of real time RT-PCR analysis of signature gene and stem
    cell-specific genes. Results are expressed as a CT ratio between
    gene of interest and an internal transcript control (GAPDH). mRNA
    transcripts not expressed by cells (−). nTERA-2 cells serves as
    a positive control for stem cell-specific gene expression.
    Media LDL-
    Isolation type Condition renin reticulon R IL-8
    1 EGM2 Normoxia 0.85 0.73 0.86
    (low density)
    2 EGM2 Normoxia 0.88 0.78 0.90
    (high density)
    3 REGM Hypoxia 1.12 1.05 0.92
    5 SMGM2 Hypoxia 0.79 0.73 0.89
    (low density)
    6 SMGM2 Hypoxia (high 0.94 0.90 1.04
    density
    7 SMGM2 Normoxia 0.78 0.84 0.96
    (low density)
    8 SMGM2 Normoxia 0.74 0.74 0.91
    (high density)
    9 Sonic Hypoxia 1.00 1.05 0.90
    hedgehog
    medium
    10  KGM Normoxia 0.99 1.08 0.89
    nTERA-2 0.63
  • Analysis of Trophic Factor Production
  • The production of different growth factors and cytokines was analyzed for eight isolates (Table 30-6). All umbilicus-derived cell isolation conditions resulted in cell populations that secreted relatively high mounts (>30 μg/mL/1×106 cells) of tissue inhibitor of metalloprotease-1 (TIMP1), basic fibroblast growth factor (FGF), hepatocyte growth factor (HGF), heparin-binding epidermal growth factor (HB-EGF), tissue inhibitor of metalallinoprotease-2 (TIMP2), interleukin 8 (IL8), and stromal derived factor-1b (SDF1b). Undetectable to low amounts (<30 pg/mL/1×106 cells) of platelet-derived epithelial growth factor-bb (PDGFbb), basic nerve growth factor (NGF), and neurotrophin-3 (NT3) were detected from the same samples.
  • Cells isolated with EGM2 medium yielded populations of cells that produced low amounts of many trophic factors including, keratinocyte growth factor (KGF), transforming growth factor-alpha (TGF-alpha), brain-derived neurotrophic factor (BDNF), and cilliary neurotrophic factor (CNTF). However, these cell lines produced higher amounts of interleukin-6 (IL6) compared to other cell lines tested. Cells isolated in SMGM2 under normal atmosphere with low initial seeding density produced very high (4737 pg/mL/1×106 cells) amounts of HGF. These cells also secreted very high amounts of TIMP2 (26,002 pg/mL/1×106 cells) as compared to other isolation conditions. Slightly higher amounts (39.6 pg/mL/1×106 cells) of TGF-alpha were produced by these cells than other cell lines tested.
  • Alternate isolation conditions also provided populations of cells secreting variable amounts of VEGF. Umbilicus-derived cells isolated in REGM under normoxic and hypoxic conditions produced relatively high amounts of VEGF (64.8 pg/mL/1×106 cells and 131.6 pg/mL/1×106 cells, respectively). Other isolation and growth conditions showed comparatively low VEGF secretion.
    TABLE 30-6
    SearchLight Multiplexed ELISA assay analysis of trophic factor production in umbilical
    cells derived from different isolation and growth conditions. High (>30 pg/mL/
    1 × 106 cells) amounts of trophic factor are indicated in light gray boxes
    and low amounts are indicated in white boxes.
    Isolation TIMP1 PDGFBB KGF HGF FGF VEGF HB-EGF TIMP2 MCP1
    C <9.8 <2.0 <2.0 <3.2 <5.4 <4.0 <3.6 23.7 15.1
    1 42421.0 4.0 23.0 664.0 52.0 33.0 103.0 2257.0 299.0
    2 51515.0 3.0 18.0 350.0 66.0 5.0 93.0 1268.0 752.0
    3 371200.0 8.4 10.0 69.7 101.9 131.5 290.4 5700.0 154.7
    4 260100.0 <2.0 41.4 102.6 624.6 64.8 648.0 6192.0 122.4
    5 225321.0 31.0 41.0 4737.0 408.0 29.0 507.0 26002.0 165.0
    6 81300.0 <2.0 31.2 36.5 259.9 20.5 257.6 1336.8 55.5
    7 73656.0 <2.0 25.3 45.4 172.6 18.6 262.6 1210.4 33.5
    8 137349.0 8.0 24.0 376.0 383.0 37.0 366.0 15997.0 710.0
    Isolation IL6 IL8 TGFA BDNF SDF1B CNTF B-NGF NT3
    C <.8 <.8 3.7 8.0 23.4 104.5 3.2 2.8
    1 34.0 213.0 4.0 26.0 61.0 81.0 4.0 4.0
    2 42.0 2690.0 4.0 29.0 77.0 100.0 4.0 5.0
    3 9.5 163.2 12.7 51.7 256.1 <7.8 6.9 <1.6
    4 18.0 2678.4 39.6 100.8 507.6 459.0 28.8 30.6
    5 17.0 770.0 20.0 99.0 260.0 394.0 16.0 13.0
    6 27.4 608.0 <2.4 41.8 200.6 146.7 11.4 13.7
    7 19.3 923.3 12.6 33.5 239.6 237.3 9.7 14.9
    8 30.0 931.0 13.0 69.0 157.0 373.0 13.0 15.0

    “C”, control sample is serum free media alone without conditioning. Results shown are the average of duplicate measurements.
  • Discussion
  • Different media formulations and culture conditions can be used to isolate cells from human umbilical cord tissue. However, these alternate isolation conditions derived cells that appear to be similar to cells isolated using the conditions initially used to isolate and characterize these cells.
  • Based on the surface marker profile, and gene expression analysis, cells isolated using alternate conditions yield the same cell type as those isolated in Growth Medium under the original conditions. In addition, cells obtained from all isolation conditions tested showed similar trophic factor secretion profiles. Alternate isolation conditions may stimulate some cells to produce more or less of one or more particular factors, and this may applications for therapeutic use.
  • Long-term growth potential is an important measurement for assessing the ability of the alternate isolation condition to produce a cell for allogeneic-based cell therapy. Conditions described in Table 30-1 generally yielded cell populations that could be expanded to approximately 5 population doublings.
  • After comparing long-term growth potential, surface marker phenotype, signature and stem cell specific gene expression and trophic factor production, these cell populations appear to be similar to cells isolated using standard conditions.
  • Biological Deposit of Umbilicus-Derived Cells and Cultures
  • Consistent with the detailed description and the written examples provided herein, examples of umbilicus-derived cells of the invention were deposited with the American Type Culture Collection (ATCC, Manassas, Va.) on Jun. 10, 2004, and assigned ATCC Accession Numbers as follows: (1) strain designation UMB 022803 (P7) was assigned Accession No. PTA-6067; and (2) strain designation UMB 022803 (P17) was assigned Accession No. PTA-6068.
  • While the present invention has been particularly shown and described with reference to the presently preferred embodiments, it is understood that the invention is not limited to the embodiments specifically disclosed and exemplified herein. Numerous changes and modifications may be made to the preferred embodiment of the invention, and such changes and modifications may be made without departing from the scope and spirit of the invention as set forth in the appended claims.

Claims (117)

1. An isolated umbilicus-derived cell comprising a cell derived from mammalian umbilical cord tissue substantially free of blood, said cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes.
2. The isolated cell of claim 1 which can expand in the presence of oxygen from about 5% to about 20%.
3. The cell of claim 2 which requires L-valine for growth.
4. The isolated cell of claim 3 which can double sufficiently to generate yields of greater than about 1014 cells in less than about 80 days in culture when seeded at about 103 cells/cm2.
5. The isolated cell of claim 3 which can double sufficiently to generate greater than about 1015 cells in less than about 80 days in culture when seeded at about 5×103 cells/cm2.
6. The isolated cell of claim 5 which can double sufficiently to generate greater than about 1017 cells in less than about 65 days in culture when seeded at about 5×103 cells/cm2.
7. The isolated cell of claim 3 which can undergo at least 40 doublings in culture.
8. The cell of claim 3 which is isolated from human umbilicus.
9. The isolated cell of claim 8 isolated in the presence of one or more enzyme activities comprising metalloproteases, neutral proteases, or mucolytic enzymes.
10. The isolated cell of claim 9 wherein the enzyme activities include at least one collagenase, and one or more of the protease activities, dispase and thermolysin.
11. The isolated cell of claim 9 wherein the enzyme activities are a collagenase from Clostridium histolyticum and dispase.
12. The isolated cell of claim 11 wherein the enzyme activities further include hyaluronidase.
13. The isolated cell of claim 12 which attaches and expands on a coated or uncoated tissue culture vessel, wherein a coated tissue culture vessel comprises a coating with gelatin, laminin, collagen, polyornithine, polylysine, vitronectin, or fibronectin.
14. The isolated cell of claim 13 which expands in the presence of from about 2% to about 15% added serum, in the presence or absence of beta-mercaptoethanol, and in the presence or absence of added growth factors including EGF, FGF, PDGF, VEGF, IGF or LIF.
15. A cell culture comprising the isolated cell of claim 1 which is free of maternal cells.
16. The isolated cell of claim 1 which maintains a normal karyotype as it is passaged.
17. The isolated cell of claim 1 wherein the cell is characterized in its production or lack of production of one or more cell surface markers comprising CD10, CD13, CD31, CD44, CD45, CD73, CD90, CD117, CD141, PDGFr-alpha, HLA-A, B, C, and HL-Dr, DP, DQ.
18. The isolated cell of claim 17 wherein the cell produces one or more of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, or HLA-A, B, C.
19. The isolated cell of claim 18 wherein the cell produces each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C.
20. The isolated cell of claim 17 wherein the cell does not produce one or more of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
21. The isolated cell of claim 20 wherein the cell produces one or more of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, or HLA-A, B, C.
22. The isolated cell of claim 20 wherein the cell does not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
23. The isolated cell of claim 22 wherein the cell produces each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A, B, C.
24. An isolated CD45 umbilicus-derived cell having a cell surface marker profile wherein the cell produces one or more of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, or HLA-A, B, C, and does not express one or more of CD31, CD34, CD117, CD141, or HLA-DR, DP, DQ, as detected by flow cytometry.
25. The isolated cell of claim 24 wherein the cell surface marker expression profile remains substantially unchanged with passage, culture vessel surface coating, or isolation procedure.
26. The isolated cell of claim 25 expressing a gene for one or more of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; or tumor necrosis factor, alpha-induced protein 3.
27. The isolated cell of claim 26 expressing a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3.
28. The isolated cell of claim 27 wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, an ileac crest bone marrow cell, or placenta-derived cell.
29. The isolated cell of claim 25, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, has reduced expression of one or more genes selected from the group consisting of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; and insulin-like growth factor binding protein 2, 36 kDa.
30. The isolated cell of claim 29 expressing a gene for one or more of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; or tumor necrosis factor, alpha-induced protein 3, wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, an ileac crest bone marrow cell, or placenta-derived cell.
31. The isolated cell of claim 29, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, has reduced expression of genes for each of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; and insulin-like growth factor binding protein 2, 36 kDa.
32. The isolated cell of claim 31 expressing a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3, wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell.
33. An isolated human umbilicus-derived cell, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, has reduced expression of genes for each of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; and which expresses a gene for each of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3, wherein the expression is increased relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell.
34. The isolated cell of claim 33 capable of self-renewal and expansion in culture, and having the potential to differentiate into cells of other phenotypes.
35. The isolated cell of claim 34 which produces one or both of vimentin and alpha smooth muscle actin.
36. The isolated cell of claim 35 which produces both vimentin and alpha-smooth muscle actin.
37. The isolated cell of claim 36 wherein the production of vimentin and alpha smooth muscle actin is retained over passaging under growth conditions.
38. A therapeutic cell culture comprising the cell of claim 34.
39. The therapeutic cell culture of claim 38 which does not substantially stimulate allogeneic PBMCs.
40. The therapeutic cell culture of claim 39 which lacks detectable amounts of HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as determined by flow cytometry.
41. The therapeutic cell culture of claim 40 which further lacks detectable amounts of HLA-G and CD178, as determined by flow cytometry.
42. The therapeutic cell culture of claim 41, which produces detectable amounts of PD-L2, as determined by flow cytometry.
43. The therapeutic cell culture of claim 34 which does not substantially stimulate a lymphocyte mediated response in vitro, as compared to allogeneic controls in a mixed lymphocyte reaction.
44. An isolated human umbilicus-derived cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes, wherein the cell does not substantially stimulate naïve CD4+ T cells, and expresses PD-L2, but not HLA-G, CD178, HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, or B7-H2.
45. The isolated cell of claim 44 wherein the cell produces vimentin and alpha-smooth muscle actin.
46. The isolated cell of claim 44 wherein the cell secretes one or more cellular factors.
47. The isolated cell of claim 46 wherein the factors are MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1.
48. The isolated cell of claim 47 wherein the cell secretes each of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, and TIMP1.
49. The isolated cell of claim 44 wherein the cell does not secrete one or more of the cellular factors SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA.
50. The isolated cell of claim 49, wherein the cell secretes one or more of the factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, and TIMP1.
51. The isolated cell of claim 50 wherein the cell secretes each of the factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, and TIMP1.
52. The isolated cell of claim 49 wherein the cell does not secrete any of the factors SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA.
53. The isolated cell of claim 52, wherein the cell secretes one or more of the factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1.
54. The isolated cell of claim 53 wherein the cell secretes each of the factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, and TIMP1.
55. An isolated human umbilicus-derived cell which secretes one or more of the factors MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1, and does not secrete one or more of the factors SDF-1alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA.
56. The isolated cell of claim 55 which secretes each of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, and TIMP1.
57. The isolated cell of claim 50 wherein the cell does not secrete any of the factors SD F-1 alpha, TGF-beta2, ANG2, PDGFbb or VEGF, as detected by ELISA.
58. A therapeutic cell culture comprising the isolated cell of claim 55.
59. The therapeutic cell culture of claim 58 further comprising one or more of a pharmaceutically-acceptable carrier, another cell culture, an antiapoptotic compound, an antithrombogenic compound, an anti-inflammatory compound, an immunosuppressive compound, an immunomodulatory compound, an angiogenic factor, and a neurotrophic factor.
60. A method of deriving, from umbilical tissue, an isolated cell, said cell capable of self-renewal and expansion in culture, and having the potential to differentiate into cells of other phenotypes, the method comprising the steps of:
(a) obtaining umbilical tissue;
(b) removing substantially all of blood to yield a substantially blood-free umbilical tissue,
(c) dissociating the tissue by mechanical or enzymatic treatment, or both,
(d) resuspending the tissue in a culture medium, and
(e) providing growth conditions which allow for the growth of an umbilicus-derived cell capable of self-renewal and expansion in culture and having the potential to differentiate into cells of other phenotypes.
61. The method of claim 60 further comprising the step of selecting adherent cells after from about ten to about 100 hours in culture.
62. The method of claim 61 wherein the umbilicus tissue is from a human.
63. The method of claim 62 wherein the derived cell is capable of to expansion in the absence of added growth factors in medium with from 2% to about 15% added serum.
64. The method of claim 63 wherein the derived cell can expand in the presence of oxygen from about 5% to about 20%.
65. The method of claim 64 wherein the derived cell cannot be maintained in the absence of L-valine.
66. The method of claim 65 wherein the derived cell can undergo at least 40 doublings in culture.
67. The method of claim 65 wherein the derived cell can double sufficiently to generate at least about 1017 cells in less than about 65 days in culture when seeded at about 5×103 cells/cm2.
68. The method of claim 62 wherein the umbilical tissue is obtained after normal or surgically-assisted child birth from a full-term or pre-term pregnancy.
69. The method of claim 60 wherein the dissociation step comprises the use of one or more enzyme activities selected from the group consisting of metalloprotease, hyaluronidase, and neutral protease.
70. The method of claim 69 wherein the enzyme activities are collagenase and dispase.
71. The method of claim 70 wherein the enzyme activities further include hyaluronidase.
72. The method of claim 71 wherein the dissociating step comprises incubating at about 37° C.
73. The method of claim 72 wherein the incubating is for one or more hours.
74. The method of claim 72 where the incubating is for about 2 hours.
75. The method of claim 69 wherein the derived cell attaches and expands on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating with gelatin, laminin, collagen, polyornithine, polylysine, vitronectin, or fibronectin.
76. The method of claim 69 wherein the derived cell expands in the presence of from about 2% to about 15% Fetal Bovine Serum, in the presence or absence of beta-mercaptoethanol, and in the presence or absence of one or more added growth factors including EGF, FGF, PDGF, VEGF, IGF and LIF.
77. The method of claim 60 wherein the removing step comprises removal of free or clotted blood by one or more of washing, suctioning, blotting, centrifugal separation, or enzymatic removal.
78. The method of claim 60 wherein the dissociating step is accomplished aseptically.
79. The method of claim 78 wherein the dissociation step comprises one or more of mincing, blending, homogenizing, or grinding.
80. An isolated human umbilicus-derived cell derived by the method of claim 69.
81. The isolated cell of claim 80 which maintains a normal karyotype with passaging.
82. A therapeutic culture of human umbilicus-derived cells derived by the method of claim 69, wherein the culture is free of maternal cells.
83. A conditioned culture medium generated by the growth of the culture of claim 15 or 82.
84. The conditioned medium of claim 83 comprising one or more of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1.
85. A mammalian cell culture comprising the conditioned medium of claim 84 and a mammalian cell in need of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, or TIMP1.
86. The culture of claim 15 or claim 82 comprising the umbilicus-derived cells and another mammalian cell of any phenotype.
87. The culture of claim of claim 86 comprising a human cell line in addition to the umbilicus-derived cell.
88. A three dimensional matrix comprising the umbilicus-derived cell of claim 1.
89. The three-dimensional matrix of claim 88 wherein the matrix comprises a biocompatible or bioabsorbable polymer.
90. The three-dimensional matrix of claim 89 wherein the matrix comprises PGA/PLA copolymer, PCL/PGA copolymer, or self assembling peptides.
91. An implantable tissue structure comprising the matrix of claim 89.
92. An implantable device comprising the therapeutic cell of claim 82.
93. An implantable human tissue matrix comprising the cell of claim 80.
95. A human tissue comprising the cell of claim 8.
96. A cell lysate derived from the cell of claim 8.
97. A soluble cell fraction derived from the cell of claim 8
98. A membrane-enriched cell fraction derived from the cell of claim 8.
99. An extracellular membrane fraction derived from the cell of claim 8.
100. An injectable therapeutic cell comprising an isolated human umbilicus-derived cell of claim 44.
101. The injectable cell of claim 100 treated to inactivate tissue factor.
102. The injectable cell of claim 101 wherein the treated is with an anti-tissue factor antibody.
103. An isolated postpartum-derived cell comprising an L-valine-requiring cell derived from human postpartum tissue substantially free of blood, said cell capable of self-renewal and expansion in culture and having the potential to differentiate into a cell of cardiomyocyte phenotypes; the cell capable of growth in an atmosphere containing oxygen from about 5% to at least about 20%; wherein said cell comprises at least one of the following characteristics:
potential for at least about 40 doublings in culture;
attachment and expansion on a coated or uncoated tissue culture vessel, wherein a coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin, or fibronectin;
production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin;
production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A, B, C;
lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ, as detected by flow cytometry;
expression of at least one of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3;
expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa;
secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and
lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1beta, 1309, MDC, and VEGF, as detected by ELISA.
104. The isolated postpartum-derived cell of claim 103; wherein said cell comprises each of the following characteristics:
potential for at least about 40 doublings in culture;
attachment and expansion on a coated or uncoated tissue culture vessel, wherein a coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin, or fibronectin;
production of tissue factor, vimentin, and alpha-smooth muscle actin;
production of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A, B, C;
lack of production of each of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ, as detected by flow cytometry;
expression of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3;
expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for each of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3; distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa;
secretion of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and
lack of secretion of each of TGF-beta2, ANG2, PDGFbb, MIP1beta, 1309, MDC, and VEGF, as detected by ELISA.
105. An isolated postpartum-derived cell comprising a signature gene profile wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum.
106. The isolated postpartum-derived cell of claim 105 further comprising the ability to alter its expression of cell surface markers when grown in medium containing serum relative to that in serum free medium.
107. The isolated postpartum-derived cell of claim 106 wherein the markers for PDGFreceptor alpha and HLA-ABC are altered.
108. A method for the preparation of therapeutic cells or cultures comprising:
a) isolating cells;
b) initially expanding the cells to a useful number in a serum-containing medium which supports cell expansion but in which the cells produce a quantity of the cell surface marker HLA-ABC;
c) transferring the cells to a medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC; and
d) passaging the cells in the medium in which the cells produce a decreased amount of the HLA-ABC,
thereby preparing a therapeutic cell or culture.
109. The method of claim 108 wherein the medium in which the cells produce a decreased amount of the cell surface marker HLA-ABC is a serum-free medium.
110. The method of claim 109 for the production of therapeutic cells or cultures for implantation.
111. The method of claim 108 for the production of therapeutic cells or cultures for grafting.
112. A serum-free medium for the expansion of postpartum-derived cells wherein the medium has one or more growth factors added.
113. The serum-free medium of claim 112 wherein the one or more growth factors added are bFGF, EGF, or PDGF.
114. The serum-free medium of claim 113 wherein the one or more added growth factors includes bFGF.
115. The serum free medium of claim 114 which supports expansion for at least 20 passages.
116. A therapeutic culture comprising postpartum-derived cells expanded in serum-free medium.
117. The therapeutic culture of claim 116 comprising cells having a signature gene profile wherein mRNA from genes for reticulon, oxidized LDL receptor, and IL-8 are present independent of whether the cells are grown in medium containing serum or medium free of serum.
118. A cell culture bank comprising the cells of 117.
US11/315,897 2003-06-27 2005-12-22 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same Abandoned US20060223177A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/315,897 US20060223177A1 (en) 2003-06-27 2005-12-22 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US12/946,609 US8815587B2 (en) 2003-06-27 2010-11-15 Postpartum cells derived from umbilical tissue and methods of making and using the same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US48326403P 2003-06-27 2003-06-27
US10/877,012 US7510873B2 (en) 2003-06-27 2004-06-25 Postpartum cells isolated from umbilical cord tissue, and methods of making and using the same
US63908804P 2004-12-23 2004-12-23
US11/315,897 US20060223177A1 (en) 2003-06-27 2005-12-22 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/877,012 Continuation-In-Part US7510873B2 (en) 2003-06-27 2004-06-25 Postpartum cells isolated from umbilical cord tissue, and methods of making and using the same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/946,609 Division US8815587B2 (en) 2003-06-27 2010-11-15 Postpartum cells derived from umbilical tissue and methods of making and using the same

Publications (1)

Publication Number Publication Date
US20060223177A1 true US20060223177A1 (en) 2006-10-05

Family

ID=36615452

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/315,897 Abandoned US20060223177A1 (en) 2003-06-27 2005-12-22 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US12/946,609 Active 2027-02-04 US8815587B2 (en) 2003-06-27 2010-11-15 Postpartum cells derived from umbilical tissue and methods of making and using the same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/946,609 Active 2027-02-04 US8815587B2 (en) 2003-06-27 2010-11-15 Postpartum cells derived from umbilical tissue and methods of making and using the same

Country Status (8)

Country Link
US (2) US20060223177A1 (en)
EP (1) EP1831356B1 (en)
JP (2) JP5340599B2 (en)
AU (1) AU2005322060B2 (en)
CA (1) CA2589041C (en)
ES (1) ES2621847T3 (en)
PL (1) PL1831356T3 (en)
WO (1) WO2006071794A2 (en)

Cited By (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US20050125077A1 (en) * 2003-12-05 2005-06-09 Harmon Alexander M. Viable tissue repair implants and methods of use
US20060069008A1 (en) * 2004-09-28 2006-03-30 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US20060222634A1 (en) * 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US20060233765A1 (en) * 2003-06-27 2006-10-19 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US20060241376A1 (en) * 2003-04-24 2006-10-26 Koninklijke Philips Electronics N.V. Non-invasive left ventricular volume determination
US20070025973A1 (en) * 2005-07-29 2007-02-01 Fitzsimmons Frederick J Reprogramming of adult or neonic stem cells and methods of use
WO2007073552A1 (en) * 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
US20070190034A1 (en) * 2005-10-13 2007-08-16 Casper Paludan Immunomodulation using placental stem cells
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
US20080064098A1 (en) * 2006-06-05 2008-03-13 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers
US20080286267A1 (en) * 2007-04-13 2008-11-20 Sing George L Methods for treating nervous system inquiry and disease
US20090010899A1 (en) * 2005-03-31 2009-01-08 Palladino Linda O Methods related to surgery
US20090074731A1 (en) * 2007-09-13 2009-03-19 Librach Clifford L Method of isolation and use of cells derived from first trimester umbilical cord tissue
WO2009046335A1 (en) * 2007-10-05 2009-04-09 Ethicon, Incorporated Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
WO2009065093A2 (en) * 2007-11-17 2009-05-22 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
US20090275129A1 (en) * 2008-04-30 2009-11-05 Ethicon, Inc. Tissue engineered blood vessels
US20100196910A1 (en) * 2001-07-20 2010-08-05 Technion Research & Development Foundation Ltd. Methods of generating human cardiac cells and tissues and uses thereof
US20100221268A1 (en) * 2007-06-15 2010-09-02 Centro Di Ricerca E. Menni Fondazione Poliambulanza Istituto Ospedaliero T cell immunomodulation by placenta cell preparations
US7807458B2 (en) 2003-01-30 2010-10-05 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Multilineage-inducible cells and uses thereof
US7824701B2 (en) 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
US7875296B2 (en) 2003-11-26 2011-01-25 Depuy Mitek, Inc. Conformable tissue repair implant capable of injection delivery
US7875273B2 (en) 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US20110143429A1 (en) * 2008-04-30 2011-06-16 Iksoo Chun Tissue engineered blood vessels
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US8016867B2 (en) 1999-07-23 2011-09-13 Depuy Mitek, Inc. Graft fixation device and method
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US20120021509A1 (en) * 2009-03-20 2012-01-26 Snu R&Db Foundation Isolating method for umbilical cord blood-derived pluripotent stem cells expressing znf281
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8221741B2 (en) 2007-01-17 2012-07-17 Marshall Vivienne S Methods for modulating inflammatory and/or immune responses
US8221780B2 (en) 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8226715B2 (en) 2003-06-30 2012-07-24 Depuy Mitek, Inc. Scaffold for connective tissue repair
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US20120315246A1 (en) * 2011-06-13 2012-12-13 Rupp Randall G Compositions and methods for modulating inflammatory and/or immune responses
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US8449561B2 (en) 1999-07-23 2013-05-28 Depuy Mitek, Llc Graft fixation device combination
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
WO2013123292A1 (en) * 2012-02-17 2013-08-22 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US20130302890A1 (en) * 2012-05-08 2013-11-14 Stem Cell Reserve, Llc Stem cells & matrix from cord tissue
US8691259B2 (en) 2000-12-21 2014-04-08 Depuy Mitek, Llc Reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US8722034B2 (en) 2009-03-26 2014-05-13 Depuy Synthes Products Llc hUTC as therapy for Alzheimer's disease
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
US8895045B2 (en) 2003-03-07 2014-11-25 Depuy Mitek, Llc Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US8940294B2 (en) 2012-03-02 2015-01-27 Tissuetech, Inc. Methods of isolating and culturing stem cells
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US20160194609A1 (en) * 2015-01-05 2016-07-07 Hygieia Therapeutics Sdn Bhd Isolation, expansion and characterization of wharton's jelly mesenchymal stem cells
US9511171B2 (en) 2002-10-18 2016-12-06 Depuy Mitek, Llc Biocompatible scaffolds with tissue fragments
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US9611513B2 (en) 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
US10413571B2 (en) 2006-10-12 2019-09-17 DePuy Synthes Products, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US10557116B2 (en) 2008-12-19 2020-02-11 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
US10583220B2 (en) 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US10925903B2 (en) 2007-09-13 2021-02-23 Reprobiogen Inc. Use of cells derived from first trimester umbilical cord tissue
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
US11690877B2 (en) 2015-08-12 2023-07-04 Cha Biotech Co., Ltd. Umbilical cord-derived adherent stem cells, preparation method therefor, and use thereof

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2621847T3 (en) 2004-12-23 2017-07-05 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using them
EP2164953A4 (en) 2007-06-18 2010-06-30 Childrens Hosp & Res Ct Oak Method of isolating stem and progenitor cells from placenta
WO2009021333A1 (en) * 2007-08-14 2009-02-19 Tissue Regeneration Therapeutics Inc. Blood vessel extraction from umbilical cord
PL2237789T3 (en) * 2007-12-27 2017-07-31 DePuy Synthes Products, Inc. Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
AU2009327383B2 (en) * 2008-12-19 2014-08-28 DePuy Synthes Products, LLC Regeneration and repair of neural tissue following injury
US8771677B2 (en) 2008-12-29 2014-07-08 Vladimir B Serikov Colony-forming unit cell of human chorion and method to obtain and use thereof
EP2536824B1 (en) * 2010-02-18 2018-04-04 Osiris Therapeutics, Inc. Immunocompatible chorionic membrane products
CN102191229A (en) * 2010-03-16 2011-09-21 侯亚义 Method for rapidly and effectively acquiring umbilical cord mesenchymal stem cell (MSC)
JP2012031127A (en) * 2010-08-03 2012-02-16 Nagoya Univ Composition including umbilical cord matrix stromal cell
CN101948804A (en) * 2010-10-25 2011-01-19 湖南光琇高新生命科技有限公司 Preparation method of umbilical cord mesenchymal stem cells
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
ITTO20111183A1 (en) * 2011-12-21 2013-06-22 Univ Degli Studi Torino CONDITIONAL MEANS OBTAINED FROM PLACENTARY STEM CELLS AND ITS USE IN THE THERAPEUTIC TREATMENT OF PREECLAMPSIA
CN102703385B (en) * 2012-06-26 2014-04-02 亚太干细胞科研中心有限公司 Mesenchymal stem cell nutrient solution
US20140170748A1 (en) * 2012-12-14 2014-06-19 DePuy Synthes Products, LLC Nutrient Enriched Media for hUTC Growth
US10707531B1 (en) 2016-09-27 2020-07-07 New Dominion Enterprises Inc. All-inorganic solvents for electrolytes
US11285177B2 (en) 2018-01-03 2022-03-29 Globus Medical, Inc. Allografts containing viable cells and methods thereof

Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4882162A (en) * 1987-06-26 1989-11-21 Dow Corning Kabushiki Kaisha Artificial skin
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
US5437994A (en) * 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5443950A (en) * 1986-04-18 1995-08-22 Advanced Tissue Sciences, Inc. Three-dimensional cell and tissue culture system
US5580777A (en) * 1987-10-29 1996-12-03 Amrad Corporation Limited Generation of neural precursor cell lines
US5677181A (en) * 1992-10-29 1997-10-14 The Australian National University Angiogenesis inhibitory antibodies
US5693332A (en) * 1995-08-11 1997-12-02 The Regents Of The University Of California Human keratinocytes supported on a hydrophilic membrane and methods of using same to effect wound closure
US5698518A (en) * 1994-03-30 1997-12-16 Oklahoma Medical Research Foundation Method for regulating inflammation and tumor growth with calmodulin, calmodulin analogues or calmodulin antagonists
US5718922A (en) * 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
US5811094A (en) * 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5843781A (en) * 1993-04-28 1998-12-01 The Johns Hopkins University School Of Medicine Implantable prosthetic vascular device having an adherent cell monolayer produced under shear stress
US5902741A (en) * 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US6171610B1 (en) * 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
US6179872B1 (en) * 1998-03-17 2001-01-30 Tissue Engineering Biopolymer matt for use in tissue repair and reconstruction
US6291240B1 (en) * 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
US6323188B1 (en) * 1998-07-01 2001-11-27 Donald L. Weissman Treatment and prevention of cardiovascular diseases, heart attack, and stroke, primary and subsequent, with help of aspirin and certain vitamins
US6331313B1 (en) * 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
US6333029B1 (en) * 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US6372494B1 (en) * 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
US20020150986A1 (en) * 1996-03-15 2002-10-17 Munin Corporation Extracellular matrix signalling molecules
US20020160510A1 (en) * 2001-02-14 2002-10-31 Hariri Robert J. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US20020182241A1 (en) * 2001-01-02 2002-12-05 Borenstein Jeffrey T. Tissue engineering of three-dimensional vascularized using microfabricated polymer assembly technology
US6495645B1 (en) * 1999-01-25 2002-12-17 Terou Okano Acrylamide derivatives and polymers containing said derivatives
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US6511511B1 (en) * 1997-05-30 2003-01-28 Osteobiologics, Inc. Fiber-reinforced, porous, biodegradable implant device
US6592623B1 (en) * 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US20030228693A1 (en) * 2002-03-12 2003-12-11 Fuji Photo Film Co., Ltd. Carrier for cell culture
US20040009566A1 (en) * 2000-07-21 2004-01-15 Teruo Okano Heart muscle-like cell sheet, three-dimensional construct, heart muscle-like tissue and process for producing the same
US20040028657A1 (en) * 2000-07-21 2004-02-12 Teruo Okano Cultured epidermal cell sheet, laminated cultured skin sheet and process for producing the same
US20040136967A1 (en) * 2002-02-25 2004-07-15 Kansas State University Research Foundation Cultures, products and methods using stem cells
US20040137619A1 (en) * 2002-10-24 2004-07-15 Fuji Photo Film Co., Ltd. Method for cell culture
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US20050074435A1 (en) * 2001-12-21 2005-04-07 Robert Casper Cellular compositions and methods of making and using them
US20050148074A1 (en) * 2003-02-11 2005-07-07 Davies John E. Progenitor cells from wharton's jelly of human umbilical cord
US6916655B2 (en) * 2001-11-22 2005-07-12 Nipro Corporation Cultured skin and method of manufacturing the same
US20050249731A1 (en) * 2002-07-16 2005-11-10 Hadi Aslan Methods of implating mesenchymal stem cells for tissue repair and formation
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
US20060171930A1 (en) * 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060233766A1 (en) * 2004-12-23 2006-10-19 Messina Darin J Treatment of Parkinson's disease and related disorders using postpartum derived cells
US20060233765A1 (en) * 2003-06-27 2006-10-19 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US20070141700A1 (en) * 2005-12-19 2007-06-21 Ethicon, Incorporated In vitro expansion of postpartum-derived cells in roller bottles
US20070160588A1 (en) * 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
US20070264713A1 (en) * 2004-09-07 2007-11-15 Rheinische Friedrich-Wilhelms-Universität Scalable Process for Cultivating Undifferentiated Stem Cells in Suspension
US20070264269A1 (en) * 2005-12-16 2007-11-15 Ethicon, Incorporated Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US20070275362A1 (en) * 2000-12-06 2007-11-29 James Edinger Placental stem cell populations
US20080112939A1 (en) * 2006-10-12 2008-05-15 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US20080145934A1 (en) * 2004-09-28 2008-06-19 Ian Ross Harris Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers

Family Cites Families (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3665061A (en) 1969-07-16 1972-05-23 United States Banknote Corp Process for producing collagen sponges
JPS5651747B2 (en) 1973-05-31 1981-12-08
JPS6040439B2 (en) 1978-03-29 1985-09-11 大正製薬株式会社 hydrocortisone derivatives
US4352883A (en) 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4487865A (en) 1983-12-15 1984-12-11 Biomatrix, Inc. Polymeric articles modified with hyaluronate
US5863531A (en) 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US4925667A (en) 1986-05-27 1990-05-15 Qmax Technology Group, Inc. Substrate with particulate cosmetic
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
GB8803697D0 (en) 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
US20030032178A1 (en) 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
JPH06104061B2 (en) 1989-02-10 1994-12-21 花王株式会社 Cell culture support material
US5840580A (en) 1990-05-01 1998-11-24 Becton Dickinson And Company Phenotypic characterization of the hematopoietic stem cell
US5342761A (en) 1990-10-01 1994-08-30 Research Development Foundation Oncofetal gene, gene product and uses therefor
US5506134A (en) 1990-10-22 1996-04-09 Corvas International, Inc. Hypridoma and monoclonal antibody which inhibits blood coagulation tissue factor/factor VIIa complex
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5286632A (en) 1991-01-09 1994-02-15 Jones Douglas H Method for in vivo recombination and mutagenesis
NL9100038A (en) 1991-01-11 1992-08-03 Stamicarbon ENZYME-CATALYZED PREPARATION OF OPTICALLY ACTIVE CARBONIC ACIDS.
EP0583360B1 (en) 1991-05-02 2002-05-22 Yeda Research And Development Co. Ltd. Compositions for the prevention and/or treatment of pathological processes
US6399369B1 (en) 1991-07-08 2002-06-04 Neurospheres Holdings Ltd. Multipotent neural stem cell cDNA libraries
EP0552380A4 (en) 1991-08-08 1995-01-25 Kao Corp Cell culture support, production thereof, and production of cell cluster using same
EP0529751A1 (en) 1991-08-09 1993-03-03 W.R. Grace & Co.-Conn. Cell culture substrate, test material for cell culture and preparations thereof
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young Scar inhibitory factor and use thereof
US5320962A (en) 1992-07-22 1994-06-14 Duke University DNA encoding the human A1 adenosine receptor
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US5670483A (en) 1992-12-28 1997-09-23 Massachusetts Insititute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5955343A (en) 1992-12-28 1999-09-21 Massachusetts Institute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5707643A (en) 1993-02-26 1998-01-13 Santen Pharmaceutical Co., Ltd. Biodegradable scleral plug
IL110589A0 (en) 1993-08-10 1994-11-11 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF- beta family
US6686198B1 (en) 1993-10-14 2004-02-03 President And Fellows Of Harvard College Method of inducing and maintaining neuronal cells
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US5456835A (en) 1993-11-08 1995-10-10 Hemasure, Inc. Device and process for removing free hemoglobin from blood
DE4406073A1 (en) * 1994-02-24 1995-08-31 Univ Ludwigs Albert Process for the production of human, clonogenic fibroblasts, process for gene transfection of fibroblasts and fibroblasts obtained in this way
US5466233A (en) 1994-04-25 1995-11-14 Escalon Ophthalmics, Inc. Tack for intraocular drug delivery and method for inserting and removing same
US6703017B1 (en) 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
US5834308A (en) 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
JPH10505250A (en) 1994-06-06 1998-05-26 ケース ウエスターン リザーブ ユニバーシティ Biomatrix for tissue regeneration
US5725493A (en) 1994-12-12 1998-03-10 Avery; Robert Logan Intravitreal medicine delivery
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5906934A (en) 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5869079A (en) 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
US5641750A (en) 1995-11-29 1997-06-24 Amgen Inc. Methods for treating photoreceptors using glial cell line-derived neurotrophic factor (GDNF) protein product
US6200606B1 (en) 1996-01-16 2001-03-13 Depuy Orthopaedics, Inc. Isolation of precursor cells from hematopoietic and nonhematopoietic tissues and their use in vivo bone and cartilage regeneration
US5842477A (en) 1996-02-21 1998-12-01 Advanced Tissue Sciences, Inc. Method for repairing cartilage
CA2253724A1 (en) 1996-04-26 1997-11-06 Case Western Reserve University Skin regeneration using mesenchymal stem cells
US6787355B1 (en) 1996-08-26 2004-09-07 Mcgill University Multipotent neural stem cells from peripheral tissues and uses thereof
US5919702A (en) * 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
EP0983062A4 (en) 1997-05-21 2002-10-23 Sloan Kettering Inst Cancer Method for increasing the concentration of ascorbic acid in brain tissues of a subject
US5902598A (en) 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
WO1999028444A1 (en) 1997-12-02 1999-06-10 Zen Bio, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US6059968A (en) 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
ES2237089T3 (en) 1998-03-13 2005-07-16 Osiris Therapeutics, Inc. USES FOR NON-AUTOMATIC HUMAN MESENQUIMATE MOTHER CELLS
CA2323439A1 (en) 1998-03-16 1999-09-23 Cytovia, Inc. Dipeptide caspase inhibitors and the use thereof
ES2246085T3 (en) 1998-05-07 2006-02-01 University Of South Florida OSEA MEDULA CELLS AS A SOURCE OF NEURONS USEFUL TO REPAIR THE SPINAL MEDULA AND THE BRAIN.
WO1999061587A1 (en) 1998-05-29 1999-12-02 Osiris Therapeutics, Inc. Human cd45+ and/or fibroblast + mesenchymal stem cells
US20040037818A1 (en) 1998-07-30 2004-02-26 Brand Stephen J. Treatment for diabetes
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
US6284245B1 (en) 1998-08-25 2001-09-04 Diacrin, Inc. Neural retinal cells and retinal pigment epithelium cells and their use in treatment of retinal disorders
US6610540B1 (en) 1998-11-18 2003-08-26 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
WO2000046351A2 (en) 1999-02-04 2000-08-10 Mcgill University Platform for the differentiation of cells
CA2362593A1 (en) 1999-02-10 2000-08-17 Curis, Inc. Pancreatic progenitor cells, methods and uses related thereto
KR100979664B1 (en) 1999-03-10 2010-09-02 유니버시티 오브 피츠버그 오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Adipose-derived stem cells and lattices
JP2002542339A (en) 1999-04-16 2002-12-10 ダブリューエム・マーシュ・ライス・ユニバーシティー Functionalized poly (propylene fumarate) and (propylene fumarate-ethylene glycol) copolymers
US6287340B1 (en) 1999-05-14 2001-09-11 Trustees Of Tufts College Bioengineered anterior cruciate ligament
US6355699B1 (en) 1999-06-30 2002-03-12 Ethicon, Inc. Process for manufacturing biomedical foams
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US6555374B1 (en) 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US20030129745A1 (en) 1999-10-28 2003-07-10 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20020164307A1 (en) 1999-12-06 2002-11-07 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20030082155A1 (en) 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
CA2392615A1 (en) 1999-12-06 2001-06-07 The General Hospital Corporation Pancreatic stem cells and their use in transplantation
WO2001053503A1 (en) 2000-01-18 2001-07-26 Cornell Research Foundation, Inc. Neural progenitor cells from hippocampal tissue and a method for isolating and purifying them
US7544509B2 (en) 2000-01-24 2009-06-09 Mcgill University Method for preparing stem cell preparations
US6610535B1 (en) 2000-02-10 2003-08-26 Es Cell International Pte Ltd. Progenitor cells and methods and uses related thereto
CA2399434C (en) 2000-02-11 2010-10-19 The Schepens Eye Research Institute, Inc. Isolation and transplantation of retinal stem cells
DE60129943T2 (en) 2000-02-11 2008-05-08 Philadelphia Health And Education Corp. DIFFERENTIATION OF BONE MARROW CELLS IN NEURONAL CELLS AND THEIR USES
US7160724B2 (en) 2000-03-09 2007-01-09 University Of South Florida Human cord blood as a source of neural tissue for repair of the brain and spinal cord
DK1264877T3 (en) 2000-03-16 2013-10-28 Cellseed Inc Cell culture support material, method of co-culture of cells and co-cultured cell layer obtained thereby
US6436704B1 (en) 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
US6673606B1 (en) 2000-04-12 2004-01-06 The Children's Hospital Of Philadelphia Therapeutic uses for mesenchymal stromal cells
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
CA2408701A1 (en) 2000-05-12 2001-11-22 University Of Utah Research Foundation Compositions and methods for tissue dedifferentiation and regeneration
US8273570B2 (en) 2000-05-16 2012-09-25 Riken Process of inducing differentiation of embryonic cell to cell expressing neural surface marker using OP9 or PA6 cells
US7049072B2 (en) 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
US6759039B2 (en) 2000-06-30 2004-07-06 Amcyte, Inc. Culturing pancreatic stem cells having a specified, intermediate stage of development
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
AU2001293586A1 (en) 2000-09-29 2002-04-08 Vincent Tropepe Primitive neural stem cells and method for differentiation of stem cells to neural cells
US20020098584A1 (en) 2000-11-06 2002-07-25 Palmer Theo D. Postmortem stem cells
RU2322497C2 (en) 2000-11-30 2008-04-20 Стемрон Инк. Isolated homozygous stem cell (variants), method for its preparing (variants), method for preparing required cell-ancestor, differentiated cell, group of differentiated cells or tissue type
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
EP3299452A1 (en) 2000-12-06 2018-03-28 Anthrogenesis Corporation Method of collecting placental stem cells
US6599323B2 (en) 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US7449180B2 (en) 2001-02-06 2008-11-11 John Kisiday Macroscopic scaffold containing amphiphilic peptides encapsulating cells
EP2316918B1 (en) * 2001-02-14 2015-07-01 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US7202080B2 (en) 2001-03-29 2007-04-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
EP1401282A4 (en) 2001-05-25 2005-03-30 Cythera Inc Stem cell differentiation
JP2004534049A (en) 2001-06-07 2004-11-11 スキンメディカ,インコーポレーテッド Conditioned cell culture media and their uses
AU2002324645C1 (en) 2001-08-08 2008-11-06 Levesque Biosciences, Inc. Compositions and methods for isolation, propagation, and differentiation of human stem cells and uses thereof
US20030211603A1 (en) 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
AU2002313817A1 (en) 2001-08-27 2003-03-10 Advanced Cell Technology, Inc. Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20030104997A1 (en) 2001-09-05 2003-06-05 Black Ira B. Multi-lineage directed induction of bone marrow stromal cell differentiation
CN1195055C (en) 2001-09-06 2005-03-30 周胜利 Method for establishing hematopoietic stem cells bank by extracting hematopoietic cells from placenta tissues
US20050053588A1 (en) 2001-10-18 2005-03-10 Li Yin Conversion of liver stem and progenitor cells to pancreatic functional cells
US7129034B2 (en) 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
CZ2004696A3 (en) 2001-11-09 2005-02-16 Artecel Sciences, Inc. Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
US6712850B2 (en) 2001-11-30 2004-03-30 Ethicon, Inc. Porous tissue scaffolds for the repair and regeneration of dermal tissue
US20030109036A1 (en) 2001-12-06 2003-06-12 The Regents Of The University Of California Method for differentiating islet precursor cells into beta cells
KR101008868B1 (en) 2001-12-07 2011-01-17 제론 코포레이션 Islet cells from human embryonic stem cells
AU2002364558A1 (en) 2001-12-11 2003-06-23 Cytograft Tissue Engineering, Inc. Tissue engineered cellular sheets, methods of making and use thereof
US20030113910A1 (en) 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
US7101546B2 (en) 2001-12-21 2006-09-05 Amcyte, Inc. In situ maturation of cultured pancreatic stem cells having a specified, intermediate stage of development
US7618621B2 (en) 2002-01-14 2009-11-17 The Board Of Trustees Of The University Of Illinois Mammalian multipotent neural stem cells and compositions, methods of preparation and methods of administration thereof
US20030162290A1 (en) 2002-01-25 2003-08-28 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
US7576065B2 (en) 2002-02-15 2009-08-18 Cornell Research Foundation, Inc. Enhancing neurotrophin-induced neurogenesis by endogenous neural progenitor cells by concurrent overexpression of brain derived neurotrophic factor and an inhibitor of a pro-gliogenic bone morphogenetic protein
US20030161818A1 (en) * 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7150990B2 (en) 2002-03-06 2006-12-19 Reprocell, Inc. Self-renewing pluripotent hepatic stem cells
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
JP4136434B2 (en) 2002-04-17 2008-08-20 進 清野 Induction of insulin-producing cells
AU2003239159A1 (en) 2002-04-19 2003-11-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education Placental derived stem cells and uses thereof
US20030228295A1 (en) 2002-04-25 2003-12-11 Svendsen Clive N. Use of human neural stem cells secreting GDNF for treatment of parkinson's and other neurodegenerative diseases
US20040029269A1 (en) 2002-05-07 2004-02-12 Goldman Steven A Promoter-based isolation, purification, expansion, and transplantation of neuronal progenitor cells, oligodendrocyte progenitor cells, or neural stem cells from a population of embryonic stem cells
US20040014662A1 (en) 2002-05-08 2004-01-22 Per Lindquist Modulation of neural stem cells and neural progenitor cells
MXPA04011851A (en) 2002-05-30 2005-03-31 Celgene Corp Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes.
AU2003247514A1 (en) 2002-06-11 2003-12-22 Roy Ogle Meningeal-derived stem cells
US7285415B2 (en) 2002-07-11 2007-10-23 The Regents Of The University Of California Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US7390659B2 (en) 2002-07-16 2008-06-24 The Trustees Of Columbia University In The City Of New York Methods for inducing differentiation of embryonic stem cells and uses thereof
US20040063202A1 (en) 2002-08-28 2004-04-01 Petersen Bryon E. Neurogenesis from hepatic stem cells
US9969977B2 (en) 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
US20040062753A1 (en) 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US20050089513A1 (en) 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
ES2621847T3 (en) 2004-12-23 2017-07-05 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using them
US20070178073A1 (en) 2006-02-01 2007-08-02 Samsung Life Public Welfare Foundation Composition Comprising Separated or Proliferated Cells from Umbilical Cord Blood for Treating Developmental and/or Chronic Lung Disease
NZ612888A (en) 2007-02-12 2015-02-27 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells

Patent Citations (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5443950A (en) * 1986-04-18 1995-08-22 Advanced Tissue Sciences, Inc. Three-dimensional cell and tissue culture system
US5902741A (en) * 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US4882162A (en) * 1987-06-26 1989-11-21 Dow Corning Kabushiki Kaisha Artificial skin
US5580777A (en) * 1987-10-29 1996-12-03 Amrad Corporation Limited Generation of neural precursor cell lines
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
US5437994A (en) * 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5811094A (en) * 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5677181A (en) * 1992-10-29 1997-10-14 The Australian National University Angiogenesis inhibitory antibodies
US5843781A (en) * 1993-04-28 1998-12-01 The Johns Hopkins University School Of Medicine Implantable prosthetic vascular device having an adherent cell monolayer produced under shear stress
US5698518A (en) * 1994-03-30 1997-12-16 Oklahoma Medical Research Foundation Method for regulating inflammation and tumor growth with calmodulin, calmodulin analogues or calmodulin antagonists
US5718922A (en) * 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
US5693332A (en) * 1995-08-11 1997-12-02 The Regents Of The University Of California Human keratinocytes supported on a hydrophilic membrane and methods of using same to effect wound closure
US5693332C1 (en) * 1995-08-11 2001-01-09 Univ California Human keratinocytes supported on a hydrophilic membrane and methods of using same to effect wound closure
US20020150986A1 (en) * 1996-03-15 2002-10-17 Munin Corporation Extracellular matrix signalling molecules
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US6511511B1 (en) * 1997-05-30 2003-01-28 Osteobiologics, Inc. Fiber-reinforced, porous, biodegradable implant device
US6291240B1 (en) * 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
US6179872B1 (en) * 1998-03-17 2001-01-30 Tissue Engineering Biopolymer matt for use in tissue repair and reconstruction
US6171610B1 (en) * 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
US6323188B1 (en) * 1998-07-01 2001-11-27 Donald L. Weissman Treatment and prevention of cardiovascular diseases, heart attack, and stroke, primary and subsequent, with help of aspirin and certain vitamins
US6495645B1 (en) * 1999-01-25 2002-12-17 Terou Okano Acrylamide derivatives and polymers containing said derivatives
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US6372494B1 (en) * 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
US6333029B1 (en) * 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US6534084B1 (en) * 1999-06-30 2003-03-18 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6592623B1 (en) * 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US6331313B1 (en) * 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
US20040009566A1 (en) * 2000-07-21 2004-01-15 Teruo Okano Heart muscle-like cell sheet, three-dimensional construct, heart muscle-like tissue and process for producing the same
US20040028657A1 (en) * 2000-07-21 2004-02-12 Teruo Okano Cultured epidermal cell sheet, laminated cultured skin sheet and process for producing the same
US20070275362A1 (en) * 2000-12-06 2007-11-29 James Edinger Placental stem cell populations
US20020182241A1 (en) * 2001-01-02 2002-12-05 Borenstein Jeffrey T. Tissue engineering of three-dimensional vascularized using microfabricated polymer assembly technology
US20020160510A1 (en) * 2001-02-14 2002-10-31 Hariri Robert J. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US6916655B2 (en) * 2001-11-22 2005-07-12 Nipro Corporation Cultured skin and method of manufacturing the same
US20050074435A1 (en) * 2001-12-21 2005-04-07 Robert Casper Cellular compositions and methods of making and using them
US20040136967A1 (en) * 2002-02-25 2004-07-15 Kansas State University Research Foundation Cultures, products and methods using stem cells
US20030228693A1 (en) * 2002-03-12 2003-12-11 Fuji Photo Film Co., Ltd. Carrier for cell culture
US20050249731A1 (en) * 2002-07-16 2005-11-10 Hadi Aslan Methods of implating mesenchymal stem cells for tissue repair and formation
US20040137619A1 (en) * 2002-10-24 2004-07-15 Fuji Photo Film Co., Ltd. Method for cell culture
US20050148074A1 (en) * 2003-02-11 2005-07-07 Davies John E. Progenitor cells from wharton's jelly of human umbilical cord
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US20060153816A1 (en) * 2003-06-27 2006-07-13 Laura Brown Soft tissue repair and regeneration using postpartum-derived cells and cell products
US20050058631A1 (en) * 2003-06-27 2005-03-17 Kihm Anthony J. Postpartum cells derived from placental tissue, and methods of making and using the same
US20050058629A1 (en) * 2003-06-27 2005-03-17 Harmon Alexander M. Soft tissue repair and regeneration using postpartum-derived cells
US20050054098A1 (en) * 2003-06-27 2005-03-10 Sanjay Mistry Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20050037491A1 (en) * 2003-06-27 2005-02-17 Sanjay Mistry Repair and regeneration of ocular tissue using postpartum-derived cells
US20060153818A1 (en) * 2003-06-27 2006-07-13 Ethicon, Incorporated Cartilage and bone repair and regeneration using postpartum-derived cells
US20060153817A1 (en) * 2003-06-27 2006-07-13 Ethicon, Incorporated Cartilage and bone repair and regeneration using postpartum-derived cells
US20060154366A1 (en) * 2003-06-27 2006-07-13 Laura Brown Treatment of osteochondral diseases using postpartum-derived cells and products thereof
US20060154367A1 (en) * 2003-06-27 2006-07-13 Ethicon, Incorporated Cartilage and bone repair and regeneration using postpartum-derived cells
US20050032209A1 (en) * 2003-06-27 2005-02-10 Messina Darin J. Regeneration and repair of neural tissue using postpartum-derived cells
US20070036767A1 (en) * 2003-06-27 2007-02-15 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20070014771A1 (en) * 2003-06-27 2007-01-18 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20070009494A1 (en) * 2003-06-27 2007-01-11 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20060188983A1 (en) * 2003-06-27 2006-08-24 Ethicon Incorporated Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US20050058630A1 (en) * 2003-06-27 2005-03-17 Harris Ian Ross Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US20060233765A1 (en) * 2003-06-27 2006-10-19 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US20060234376A1 (en) * 2003-06-27 2006-10-19 Ethicon Incorporated Repair and regeneration of ocular tissue using postpartum-derived cells
US20070264713A1 (en) * 2004-09-07 2007-11-15 Rheinische Friedrich-Wilhelms-Universität Scalable Process for Cultivating Undifferentiated Stem Cells in Suspension
US20080145934A1 (en) * 2004-09-28 2008-06-19 Ian Ross Harris Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20060171930A1 (en) * 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060233766A1 (en) * 2004-12-23 2006-10-19 Messina Darin J Treatment of Parkinson's disease and related disorders using postpartum derived cells
US20070264269A1 (en) * 2005-12-16 2007-11-15 Ethicon, Incorporated Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US20070141700A1 (en) * 2005-12-19 2007-06-21 Ethicon, Incorporated In vitro expansion of postpartum-derived cells in roller bottles
US20070160588A1 (en) * 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
US20080112939A1 (en) * 2006-10-12 2008-05-15 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers

Cited By (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8449561B2 (en) 1999-07-23 2013-05-28 Depuy Mitek, Llc Graft fixation device combination
US8016867B2 (en) 1999-07-23 2011-09-13 Depuy Mitek, Inc. Graft fixation device and method
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US8691259B2 (en) 2000-12-21 2014-04-08 Depuy Mitek, Llc Reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US20100196910A1 (en) * 2001-07-20 2010-08-05 Technion Research & Development Foundation Ltd. Methods of generating human cardiac cells and tissues and uses thereof
US8753883B2 (en) 2002-02-13 2014-06-17 Anthrogenesis Corporation Treatment of psoriasis using placental stem cells
US8637066B2 (en) 2002-10-18 2014-01-28 Depuy Mitek, Llc Biocompatible scaffold for ligament or tendon repair
US9511171B2 (en) 2002-10-18 2016-12-06 Depuy Mitek, Llc Biocompatible scaffolds with tissue fragments
US10603408B2 (en) 2002-10-18 2020-03-31 DePuy Synthes Products, Inc. Biocompatible scaffolds with tissue fragments
US7824701B2 (en) 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
US7807458B2 (en) 2003-01-30 2010-10-05 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Multilineage-inducible cells and uses thereof
US8895045B2 (en) 2003-03-07 2014-11-25 Depuy Mitek, Llc Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
US20060241376A1 (en) * 2003-04-24 2006-10-26 Koninklijke Philips Electronics N.V. Non-invasive left ventricular volume determination
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US11000554B2 (en) 2003-06-27 2021-05-11 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US11179422B2 (en) 2003-06-27 2021-11-23 DePuy Synthes Products, Inc. Method of differentiating umbilical cord tissue into a chondrogenic phenotype
US8658152B2 (en) 2003-06-27 2014-02-25 DePuy Synthes Products, LLC Regeneration and repair of neural tissue using postpartum-derived cells
US10039793B2 (en) 2003-06-27 2018-08-07 DePuy Synthes Products, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US10195233B2 (en) 2003-06-27 2019-02-05 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US9717763B2 (en) 2003-06-27 2017-08-01 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US10220059B2 (en) 2003-06-27 2019-03-05 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US9579351B2 (en) 2003-06-27 2017-02-28 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US11191789B2 (en) 2003-06-27 2021-12-07 DePuy Synthes Products, Inc. Cartilage and bone repair and regeneration using postpartum-derived cells
US9504719B2 (en) 2003-06-27 2016-11-29 DePuy Synthes Products, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US10744164B2 (en) 2003-06-27 2020-08-18 DePuy Synthes Products, Inc. Repair and regeneration of ocular tissue using postpartum-derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US9498501B2 (en) 2003-06-27 2016-11-22 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8703121B2 (en) 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US10383898B2 (en) 2003-06-27 2019-08-20 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US9234172B2 (en) 2003-06-27 2016-01-12 DePuy Synthes Products, Inc. Repair and regeneration of ocular tissue using postpartum-derived cells
US8277796B2 (en) 2003-06-27 2012-10-02 Advanced Technologies And Regenerative Medicine, Llc Regeneration and repair of neural tissue using postpartum-derived cells
US10500234B2 (en) 2003-06-27 2019-12-10 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8318483B2 (en) 2003-06-27 2012-11-27 Advanced Technologies And Regenerative Medicine, Llc Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20060233765A1 (en) * 2003-06-27 2006-10-19 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US10758576B2 (en) 2003-06-27 2020-09-01 DePuy Synthes Products, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US8361459B2 (en) 2003-06-27 2013-01-29 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders using postpartum-derived cells
US9211362B2 (en) 2003-06-30 2015-12-15 Depuy Mitek, Llc Scaffold for connective tissue repair
US8226715B2 (en) 2003-06-30 2012-07-24 Depuy Mitek, Inc. Scaffold for connective tissue repair
US10583220B2 (en) 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
US7875296B2 (en) 2003-11-26 2011-01-25 Depuy Mitek, Inc. Conformable tissue repair implant capable of injection delivery
US8496970B2 (en) 2003-11-26 2013-07-30 Depuy Mitek, Llc Conformable tissue repair implant capable of injection delivery
US8137702B2 (en) 2003-11-26 2012-03-20 Depuy Mitek, Inc. Conformable tissue repair implant capable of injection delivery
US7901461B2 (en) * 2003-12-05 2011-03-08 Ethicon, Inc. Viable tissue repair implants and methods of use
US8641775B2 (en) 2003-12-05 2014-02-04 Depuy Mitek, Llc Viable tissue repair implants and methods of use
US20050125077A1 (en) * 2003-12-05 2005-06-09 Harmon Alexander M. Viable tissue repair implants and methods of use
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
US8221780B2 (en) 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
US20090149371A1 (en) * 2004-09-28 2009-06-11 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US20060069008A1 (en) * 2004-09-28 2006-03-30 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US8846394B2 (en) 2004-09-28 2014-09-30 Ethicon, Inc. Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US7875273B2 (en) 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US8278095B2 (en) 2005-03-31 2012-10-02 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US8709402B2 (en) 2005-03-31 2014-04-29 Stemnion, Inc. Amnion-derived cells, methods of making and uses thereof
US8153430B2 (en) 2005-03-31 2012-04-10 Stemnion, Inc. Methods related to surgery
US8685390B2 (en) 2005-03-31 2014-04-01 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US8741646B2 (en) 2005-03-31 2014-06-03 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US20060222634A1 (en) * 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US20090075381A1 (en) * 2005-03-31 2009-03-19 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US20090010899A1 (en) * 2005-03-31 2009-01-08 Palladino Linda O Methods related to surgery
US8877181B2 (en) 2005-03-31 2014-11-04 Stemnion, Inc. Amnion-derived cells, methods of making and uses thereof
WO2007016245A2 (en) * 2005-07-29 2007-02-08 Vivicells International, Llc Reprogramming of adult or neonic stem cells and methods of use
US20070025973A1 (en) * 2005-07-29 2007-02-01 Fitzsimmons Frederick J Reprogramming of adult or neonic stem cells and methods of use
WO2007016245A3 (en) * 2005-07-29 2007-11-22 Vivicells International Llc Reprogramming of adult or neonic stem cells and methods of use
US7682803B2 (en) 2005-10-13 2010-03-23 Anthrogenesis Corporation Immunomodulation using placental stem cells
US20070190034A1 (en) * 2005-10-13 2007-08-16 Casper Paludan Immunomodulation using placental stem cells
US9539288B2 (en) 2005-10-13 2017-01-10 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8895256B2 (en) 2005-10-13 2014-11-25 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8216566B2 (en) 2005-10-13 2012-07-10 Anthrogenesis Corporation Treatment of multiple sclerosis using placental stem cells
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US8741638B2 (en) 2005-12-19 2014-06-03 DePuy Synthes Products, LLC In vitro expansion of postpartum-derived cells in roller bottles
WO2007073552A1 (en) * 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
US9585918B2 (en) 2005-12-28 2017-03-07 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US8591883B2 (en) 2005-12-29 2013-11-26 Anthrogenesis Corporation Placental stem cell populations
US8691217B2 (en) 2005-12-29 2014-04-08 Anthrogenesis Corporation Placental stem cell populations
US8202703B2 (en) 2005-12-29 2012-06-19 Anthrogenesis Corporation Placental stem cell populations
US9078898B2 (en) 2005-12-29 2015-07-14 Anthrogenesis Corporation Placental stem cell populations
US10383897B2 (en) 2005-12-29 2019-08-20 Celularity, Inc. Placental stem cell populations
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
US20080064098A1 (en) * 2006-06-05 2008-03-13 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US10413571B2 (en) 2006-10-12 2019-09-17 DePuy Synthes Products, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers
US9102915B2 (en) 2006-11-13 2015-08-11 DePuy Synthes Products, Inc. In vitro expansion of postpartum-derived cells using microcarriers
US9138443B2 (en) 2007-01-17 2015-09-22 Stemnion, Inc. Methods for modulating immune responses
US9173908B2 (en) 2007-01-17 2015-11-03 Stemnion, Inc. Methods for modulating immune responses
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
US8221741B2 (en) 2007-01-17 2012-07-17 Marshall Vivienne S Methods for modulating inflammatory and/or immune responses
US8916146B2 (en) 2007-02-12 2014-12-23 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US20080286267A1 (en) * 2007-04-13 2008-11-20 Sing George L Methods for treating nervous system inquiry and disease
US8197804B2 (en) 2007-04-13 2012-06-12 Stemnion, Inc. Methods for treating nervous system injury and disease
US10286015B2 (en) 2007-04-13 2019-05-14 Noveome Biotherapeutics, Inc. Methods for treating traumatic brain injury with amnion-derived cellular cytokine solution (ACCS) or amnion-derived multipotent progenitor (AMP) cells
US20100221268A1 (en) * 2007-06-15 2010-09-02 Centro Di Ricerca E. Menni Fondazione Poliambulanza Istituto Ospedaliero T cell immunomodulation by placenta cell preparations
US8524283B2 (en) 2007-06-15 2013-09-03 Centro Di Ricerca E. Menni Fondazione Poliambulanza Istituto Ospedaliero T cell immunomodulation by placenta cell preparations
US20090074731A1 (en) * 2007-09-13 2009-03-19 Librach Clifford L Method of isolation and use of cells derived from first trimester umbilical cord tissue
US10925903B2 (en) 2007-09-13 2021-02-23 Reprobiogen Inc. Use of cells derived from first trimester umbilical cord tissue
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US9216200B2 (en) 2007-09-28 2015-12-22 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
WO2009046335A1 (en) * 2007-10-05 2009-04-09 Ethicon, Incorporated Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8034329B2 (en) 2007-10-05 2011-10-11 Advanced Technologies And Regenerative Medicine, Llc Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
WO2009065093A2 (en) * 2007-11-17 2009-05-22 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
US20110020291A1 (en) * 2007-11-17 2011-01-27 Debrabrata Banerjee Use of stem cells for wound healing
WO2009065093A3 (en) * 2007-11-17 2009-12-30 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
US8574897B2 (en) 2007-12-20 2013-11-05 DePuy Synthes Products, LLC Methods for sterilizing materials containing biologically active agents
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US9290742B2 (en) 2008-04-30 2016-03-22 Cordis Corporation Tissue engineered blood vessel
US20090275129A1 (en) * 2008-04-30 2009-11-05 Ethicon, Inc. Tissue engineered blood vessels
US20110143429A1 (en) * 2008-04-30 2011-06-16 Iksoo Chun Tissue engineered blood vessels
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US9198938B2 (en) 2008-11-19 2015-12-01 Antrhogenesis Corporation Amnion derived adherent cells
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
US10557116B2 (en) 2008-12-19 2020-02-11 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
US20120021509A1 (en) * 2009-03-20 2012-01-26 Snu R&Db Foundation Isolating method for umbilical cord blood-derived pluripotent stem cells expressing znf281
US10584312B2 (en) * 2009-03-20 2020-03-10 Snu R&Db Foundation Isolating method for umbilical cord blood-derived pluripotent stem cells expressing ZNF281
US9943552B2 (en) 2009-03-26 2018-04-17 DePuy Synthes Products, Inc. hUTC as therapy for Alzheimer's disease
US8722034B2 (en) 2009-03-26 2014-05-13 Depuy Synthes Products Llc hUTC as therapy for Alzheimer's disease
US8647617B2 (en) 2009-07-13 2014-02-11 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US9464274B2 (en) 2010-07-13 2016-10-11 Anthrogenesis Corporation Methods of generating natural killer cells
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US11090339B2 (en) 2011-06-01 2021-08-17 Celularity Inc. Treatment of pain using placental stem cells
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US20120315246A1 (en) * 2011-06-13 2012-12-13 Rupp Randall G Compositions and methods for modulating inflammatory and/or immune responses
US10724105B2 (en) 2011-12-23 2020-07-28 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue-derived cells
US9611513B2 (en) 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
AU2018214122C1 (en) * 2012-02-17 2021-08-19 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
US10758572B2 (en) 2012-02-17 2020-09-01 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
AU2018214122B2 (en) * 2012-02-17 2021-04-01 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
AU2013221458B2 (en) * 2012-02-17 2018-08-30 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
WO2013123292A1 (en) * 2012-02-17 2013-08-22 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
US8940294B2 (en) 2012-03-02 2015-01-27 Tissuetech, Inc. Methods of isolating and culturing stem cells
US20130302890A1 (en) * 2012-05-08 2013-11-14 Stem Cell Reserve, Llc Stem cells & matrix from cord tissue
US9670457B2 (en) * 2012-05-08 2017-06-06 Stem Cell Reserve Lp Stem cells and matrix from cord tissue
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US10542743B2 (en) * 2015-01-05 2020-01-28 Hygieia Therapeutics Sdn Bhd Isolation, expansion and characterization of wharton's jelly mesenchymal stem cells
US20160194609A1 (en) * 2015-01-05 2016-07-07 Hygieia Therapeutics Sdn Bhd Isolation, expansion and characterization of wharton's jelly mesenchymal stem cells
US11690877B2 (en) 2015-08-12 2023-07-04 Cha Biotech Co., Ltd. Umbilical cord-derived adherent stem cells, preparation method therefor, and use thereof
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation

Also Published As

Publication number Publication date
AU2005322060A1 (en) 2006-07-06
US20120315251A1 (en) 2012-12-13
AU2005322060B2 (en) 2011-11-17
PL1831356T3 (en) 2017-07-31
EP1831356A2 (en) 2007-09-12
WO2006071794A2 (en) 2006-07-06
JP2013172725A (en) 2013-09-05
JP2008525039A (en) 2008-07-17
CA2589041A1 (en) 2006-07-06
ES2621847T3 (en) 2017-07-05
CA2589041C (en) 2019-08-20
JP5340599B2 (en) 2013-11-13
US8815587B2 (en) 2014-08-26
WO2006071794A3 (en) 2007-01-25
EP1831356B1 (en) 2017-01-25

Similar Documents

Publication Publication Date Title
US10500234B2 (en) Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8815587B2 (en) Postpartum cells derived from umbilical tissue and methods of making and using the same
US20060171930A1 (en) Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060166361A1 (en) Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
AU2012200947B2 (en) Postpartum cells derived from umbilical cord tissue, and methods of making and using the same

Legal Events

Date Code Title Description
AS Assignment

Owner name: ETHICON, INCORPORATED, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HARRIS, IAN ROSS;MESSINA, DARIN J.;KIHM, ANTHONY J.;AND OTHERS;REEL/FRAME:017753/0732;SIGNING DATES FROM 20060317 TO 20060323

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: ADVANCED TECHNOLOGIES AND REGENERATIVE MEDICINE, L

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ETHICON, INC.;REEL/FRAME:027805/0484

Effective date: 20120305

AS Assignment

Owner name: DEPUY SPINE, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DEPUY ORTHOPAEDICS, INC.;REEL/FRAME:032393/0100

Effective date: 20121230

Owner name: DEPUY SPINE, LLC, MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:DEPUY SPINE, INC.;REEL/FRAME:032422/0444

Effective date: 20121230

Owner name: HAND INNOVATIONS LLC, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DEPUY SPINE, LLC;REEL/FRAME:032393/0296

Effective date: 20121230

Owner name: DEPUY SYNTHES PRODUCTS, LLC, MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:HAND INNOVATIONS LLC;REEL/FRAME:032422/0493

Effective date: 20121231

Owner name: DEPUY ORTHOPAEDICS, INC., INDIANA

Free format text: MERGER;ASSIGNOR:ADVANCED TECHNOLOGIES AND REGENERATIVE MEDICINE, LLC;REEL/FRAME:032392/0109

Effective date: 20121230

AS Assignment

Owner name: DEPUY SYNTHES PRODUCTS, INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:DEPUY SYNTHES PRODUCTS, LLC;REEL/FRAME:037156/0304

Effective date: 20141229