WO1996023059A1 - Lineage-directed induction of human mesenchymal stem cell differentiation - Google Patents

Lineage-directed induction of human mesenchymal stem cell differentiation Download PDF

Info

Publication number
WO1996023059A1
WO1996023059A1 PCT/US1996/000170 US9600170W WO9623059A1 WO 1996023059 A1 WO1996023059 A1 WO 1996023059A1 US 9600170 W US9600170 W US 9600170W WO 9623059 A1 WO9623059 A1 WO 9623059A1
Authority
WO
WIPO (PCT)
Prior art keywords
factor
cells
culture
cellε
lineage
Prior art date
Application number
PCT/US1996/000170
Other languages
French (fr)
Other versions
WO1996023059A9 (en
Inventor
Scott P. Bruder
Arnold I. Caplan
Stephen E. Haynesworth
Original Assignee
Case Western Reserve University
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
Application filed by Case Western Reserve University filed Critical Case Western Reserve University
Priority to DK96903358T priority Critical patent/DK0805853T3/en
Priority to DE69636979T priority patent/DE69636979T2/en
Priority to EP96903358A priority patent/EP0805853B1/en
Priority to JP52287196A priority patent/JP4454697B2/en
Priority to AU47469/96A priority patent/AU719098B2/en
Priority to CA002211120A priority patent/CA2211120C/en
Publication of WO1996023059A1 publication Critical patent/WO1996023059A1/en
Publication of WO1996023059A9 publication Critical patent/WO1996023059A9/en

Links

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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/203Retinoic acids ; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • 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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2006IL-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0654Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0655Chondrocytes; Cartilage
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0658Skeletal muscle cells, e.g. myocytes, myotubes, myoblasts
    • 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
    • C12N2500/42Organic phosphate, e.g. beta glycerophosphate
    • 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
    • C12N2501/06Anti-neoplasic drugs, anti-retroviral drugs, e.g. azacytidine, cyclophosphamide
    • 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
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2301Interleukin-1 (IL-1)
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones

Definitions

  • the present invention provides methods for directing mesenchymal stem cells cultivated in vitro to differentiate into specific cell lineage pathways prior to, or at the time of, their implantation for the therapeutic treatment of pathologic conditions in humans and other species.
  • MSCs Mesenchymal stem cells
  • the specific differentiation pathway which these cells enter depends upon various influences from mechanical influences and/or endogenous bioactive factors, such as growth factors, cytokines, and/or local microenvironmental conditions established by host tissues.
  • MSCs In prenatal organisms, the differentiation of MSCs into specialized connective tissue cells is well established; for example embryonic chick, mouse or human limb bud mesenchymal cells differentiate into cartilage, bone and other connective tissues (1-5). In addition, a clonal rat fetus calvarial cell line has also been shown to differentiate into muscle, fat, cartilage, and bone (6).
  • the existence of MSCs in post-natal organisms has not been widely studied with the objective of showing the differentiation of post-embryonic cells into several mesoder al phenotypes. The few studies which have been done involve the formation of bone and cartilage by bone marrow cells following their encasement in diffusion chambers and in vivo transplantation (7, 8).
  • bone marrow-derived cells from young rabbits (800-1,000 g) have been shown to form adipocytic and osteogenic cells in vivo (9) and cloned bone marrow stromal cells of post-natal mice were shown to form adipocytes and osteogenic cells (10).
  • cells from chick periosteum have been isolated, expanded in culture, and, under high density conditions in vitro , shown to differentiate into cartilage and bone (11).
  • Rat bone marrow-derived mesenchymal cells have been shown to have the capacity to differentiate into osteoblasts and chondrocytes when implanted in vivo (12, 6).
  • Cells from various marrow sources of post-natal organisms have never been observed to exhibit myogenic properties, with multinuclear appearance being the most easily recognized characteristic in culture.
  • the invention provides a method for effecting the lineage-directed induction of isolated, culture-expanded human mesenchymal stem cells which comprises contacting mesenchymal stem cells with a bioactive factor or combination of factors effective to induce differentiation thereof into a lineage of choice. More particularly, this method is one in which the bioactive factor induces differentiation of such cells into a mesenchymal lineage selected from the group consisting of osteogenic, chondrogenic, tendonogenic, ligamentogenic, myogenic, marrow stro agenic, adipogenic and dermogenic.
  • the cells are contacted ex vivo with one or more bioactive factors in this aspect, thereby providing a method free of any risks that may be associated with in vivo administration of any bioactive factors.
  • the method of the invention further provides administering to an individual in need thereof isolated culture-expanded human mesenchymal stem cells and a bioactive factor effective to induce differentiation of such cells into a lineage of choice.
  • the mesenchymal stem cells and bioactive factor are administered together or they may alternatively be administered separately.
  • this aspect of the method comprises administering the bioactive factor to an individual to whom a preparation comprising isolated autologous human mesenchymal stem cells has been, is being or will be administered.
  • the invention provides a method for inducing the in vivo production of human cytokines in an individual in need thereof which comprises administering to the individual isolated culture-expanded human mesenchymal stem cells and a bioactive factor effective to induce such cells to differentiate into a cytokine-producing mesenchymal lineage descendant in such individual.
  • the mesenchymal stem cells and bioactive factor are administered together or they may alternatively be administered separately.
  • the bioactive factor is a bone morphogenetic protein and the human MSCs are directed into the chondrogenic lineage; the bioactive factor is interleukin 1 and the human MSCs are directed into the ⁇ tromal cell lineage (preferably the interleukin 1 is interleukin la ) ; the bioactive factors are dexamethasone, ascorbic acid-2-pho ⁇ phate and ⁇ - glycerophosphate and the human MSCs are directed into the osteogenic lineage; or the bioactive factor is selected from the group consisting of 5-azacytidine, 5-azadeoxycytidine and analogs of either of them and the human mesenchymal stem cells are directed into the myogenic lineage.
  • compositions comprising isolated, culture-expanded human mesenchymal stem cells and a bioactive factor,- or combination, effective to induce differentiation of such cells into a lineage of choice.
  • the composition further comprises a tissue culture medium.
  • the composition can comprise a medium suitable for administration to an animal particularly a human, in need thereof.
  • This aspect of the invention also provides for specific embodiments using the bioactive factors identified above for lineage induction into the lineages associated therewith as described above.
  • Figure 1 diagrammatically illustrates the mesengenic process by which mesenchymal stem cells differentiate into various lineage pathways.
  • Figure 2 diagrammatically illustrates the osteogenic differentiation pathway.
  • Figure 3 graphically demonstrates the increase in alkaline phosphatase activity as a function of time in cultures, in the initial studies reported in Example 1.
  • Figure 4 shows results from the subsequent studies reported in Example 1.
  • Figure 5 diagrammatically illustrates the chondrogenic differentiation pathway.
  • Figure 6 shows the extent of human mesenchymal stem cell cytokine expression, with and without interleukin-1 stimulation, based on the experiments in Example 4.
  • A Phase contrast micrograph of living culture of MSCs showing the multinucleated cells derived after exposure to 5- aza-CR. This micrograph shows a culture 2 weeks after treatment with 10 ⁇ M 5-aza-CR. Many nuclei (arrows) in the cell can be observed, but striations are not discernible.
  • Figure 8 Immunofluorescence staining for muscle- specific yosin in myotubes derived from rat bone marrow MSCs after exposure to 5-aza-CR. Myosin antibodies do not visualize cross striations, but the antibodies clearly illuminate longitudinal fibers. Scale bar 30 ⁇ m.
  • Figures 9A-9D Myotubes derived from rat bone marrow MSCs 2 weeks [(A) and (B) ] and 5 weeks [(C) and (D) ] after exposure to 5-aza-CR. Phase contrast micrograph [(A) and (C)] and immunofluorescence staining for myosin [(B) and (D)]. (A) and (B), (C) and (D) are the same visual fields. Myotubes 2 weeks after 5-aza-CR exposure are stained with anti-myosin antibody, but those 5 weeks after exposure are not. Scale bar 50 ⁇ M.
  • Figures 10A-10B Micrograph of the 5-aza-CR-treated MSCs containing droplets in their cytoplasm; this culture was stained with Sudan Black.
  • A Clusters of adipocytes (arrows) were observed; scale bar 200 ⁇ M.
  • B Droplets are stained brown to black (arrows), which suggests that these droplets are lipid; scale bar 100 ⁇ M.
  • Figure 11 Phase contrast micrograph of living culture of myogenic cells derived from rat bone marrow MSCs after exposure to 5-aza-CR. Following exposure to 5-aza-CR, these cells were cultured with 4ng/ml bFGF for 10 days. Large myotubes can be seen; scale bar 300 ⁇ m.
  • Figures 12A-12D graphically illustrate the expression of G-CSF, GM-CSF, M-CSF and SCF, respectively, observed in the experiments reported by Example 6.
  • Figures 13A-13C graphically illustrate the expression of LIF, IL-6 and IL-11, respectively observed in the experiments reported by Example 6.
  • Figure 14 graphically illustrates the dose dependent IL- l ⁇ induction of GM-CSF expression observed in the experiments reported by Example 6.
  • the first lies in the ability to direct and accelerate MSC differentiation prior to implantation back into autologous hosts.
  • MSCs which are directed in vitro to become osteogenic cells will synthesize bone matrix at an implant site more rapidly and uniformly than MSCs which must first be recruited into the lineage and then progress through the key differentiation steps.
  • Such an ex vivo treatment also provides for uniform and controlled application of bioactive factors to purified MSCs, leading to uniform lineage commitment and differentiation. In vivo availability of endogenous bioactive factors cannot be as readily assured or controlled.
  • a pretreatment step such as is disclosed herein circumvents this.
  • by pretreating the MSCs prior to implantation potentially harmful side effects associated with systemic or local administration of exogenous bioactive factors are avoided.
  • Another use of this technique lies in the ability to direct tissue regeneration based on the stage of differentiation which the cells are in at the time of implantation. That is, with respect to bone and cartilage, the state of the cells at implantation may control the ultimate tissue type formed. Hypertrophic chondrocyte ⁇ will mineralize their matrix and eventually pave the way for vascular invasion, which finally results in new bone formation. Clearly, MSCs implanted for the purpose of restoring normal hyaline cartilage must not progress down the entire lineage.
  • implants which are designed to repair articular surface defects and the underlying subchondral bone could benefit from a two-component system wherein the cells in the area of the future bone are directed ex vivo to become hypertrophic chondrocytes, while the cells in the area of the future articulating surface are directed only to become chondroblasts.
  • the ex vivo control of differentiation can optimize MSC cell populations for the elaboration of stage-specific cytokines requisite to the needs of the individual.
  • Muscle morphogenesis can similarly be directed to create fast or slow twitch fibers, depending on the indication.
  • the human mesenchymal stem cells isolated and purified as described here can be derived, for example, from bone marrow, blood, dermis or periosteum. When obtained from bone marrow this can be marrow from a number of different sources, including plugs of femoral head cancellous bone pieces, obtained from patients with degenerative joint disease during hip or knee replacement surgery, or from aspirated marrow obtained from normal donors and oncology patients who have marrow harvested for future bone marrow transplantation. The harvested marrow is then prepared for cell culture.
  • the isolation process involves the use of a specially prepared medium that contains agents which allow for not only mesenchymal stem cell growth without differentiation, but also for the direct adherence of only the mesenchymal stem cells to the plastic or glass surface of the culture vessel.
  • Bone marrow is the soft tissue occupying the medullary cavities of long bones, some haversian canals, and spaces between trabeculae of cancellous or spongy bone. Bone marrow is of two types: red, which is found in all bones in early life and in restricted locations in adulthood (i.e. in the spongy bone) and is concerned with the production of blood cells (i.e. hematopoiesis) and hemoglobin (thus, the red color); and yellow, which consists largely of fat cells (thus, the yellow color) and connective tissue.
  • red which is found in all bones in early life and in restricted locations in adulthood (i.e. in the spongy bone) and is concerned with the production of blood cells (i.e. hematopoiesis) and hemoglobin (thus, the red color); and yellow, which consists largely of fat cells (thus, the yellow color) and connective tissue.
  • bone marrow is a complex tissue comprised of hematopoietic cells, including the hematopoietic stem cells, and red and white blood cells and their precursors; and a group of cells including mesenchymal stem cells, fibroblasts, reticulocytes, adipocyte ⁇ , and endothelial cells which contribute to the connective tis ⁇ ue network called " ⁇ troma".
  • Cell ⁇ from the ⁇ troma regulate the differentiation of hematopoietic cell ⁇ through direct interaction via cell ⁇ urface protein ⁇ and the secretion of growth factor ⁇ and are involved in the foundation and ⁇ upport of the bone ⁇ tructure.
  • bone marrow contains "pre-stromal” cells which have the capacity to differentiate into cartilage, bone, and other connective tissue cells.
  • pre-stromal cells which have the capacity to differentiate into cartilage, bone, and other connective tissue cells.
  • pluripotent stromal stem cells or mesenchymal stem cells have the ability to generate into several different types of cell lines (i.e. osteocytes, chondrocytes, adipocyte ⁇ , etc.) upon activation, depending upon the influence of a number of bioactive factors.
  • mesenchymal stem cells are present in the tissue in very minute amounts with a wide variety of other cells (i.e. erythrocyte ⁇ , platelet ⁇ , neutrophil ⁇ , lymphocytes, monocytes, eosinophil ⁇ , basophils, adipocytes, etc.).
  • a process has been developed for isolating and purifying human mesenchymal stem cells from tissue prior to differentiation and then culture expanding the mesenchymal stem cell ⁇ to produce a valuable tool for mu ⁇ culo ⁇ keletal therapy.
  • the objective of such manipulation is to greatly increase the number of mesenchymal stem cells and to utilize these cell ⁇ to redirect and/or reinforce the body's normal reparative capacity.
  • the mesenchymal stem cells are expanded to great numbers and applied to areas of connective tissue damage to enhance or stimulate in vivo growth for regeneration and/or repair, to improve implant adhesion to various prosthetic devices through subsequent activation and differentiation, or enhance hemopoietic cell production, etc.
  • various procedure ⁇ are contemplated for tran ⁇ ferring, immobilizing, and activating the culture- expanded, purified mesenchymal stem cells at the ⁇ ite for repair, implantation, etc., including injecting the cell ⁇ at the site of a skeletal defect, incubating the cells with a prosthesis and implanting the prosthesis, etc.
  • the culture-expanded, mesenchymal stem cells can be utilized for various therapeutic purposes such as to alleviate cellular, molecular, and genetic disorders in a wide number of metabolic bone disea ⁇ es, skeletal dyspla ⁇ ia ⁇ , cartilage defect ⁇ , ligament and tendon injuries and other mu ⁇ culo ⁇ keletal and connective ti ⁇ ue di ⁇ orders.
  • DMEM-LG Dulbecco's Modified Eagle's Medium-Low Glucose
  • the commercial formulation is supplemented with 3700 mg/1 of sodium bicarbonate and 10 ml/1 of lOOx antibiotic- antimycotic containing 10,000 unit ⁇ of penicillin (base), 10,000 ⁇ q of streptomycin (ba ⁇ e) and 25 ⁇ g of amphotericin B/ml utilizing penicillin G ( ⁇ odium salt), ⁇ treptomycin sulfate, and amphotericin B as FUNGIZONE® in 0.85% saline.
  • the medium described above is made up and stored in 90 ml per 100 ml or 450 ml per 500 ml bottles at 4°C until ready to use. For use, 10 ml or 50 ml of fetal bovine serum (from selected lots) i ⁇ added to the bottle ⁇ of media to give a final volume of 10% serum. The medium is warmed to 37°C prior to use.
  • BGJ b medium Gibco, Grand Island, NY
  • 10% fetal bovine serum J.R. Scientific, Woodland, CA, or other suppliers
  • This medium which was also a "Complete Medium” contained factor ⁇ which al ⁇ o ⁇ timulated me ⁇ enchymal ⁇ tem cell growth without differentiation and allowed for the selective attachment through specific protein binding sites, etc. of only the mesenchymal stem cells to the plastic surfaces of Petri dishes.
  • the complete medium can be utilized in a number of different isolation processes depending upon the specific type of initial harvesting processes used in order to prepare the harvested bone marrow for cell culture separation.
  • the marrow was added to the complete medium and vortexed to form a dispersion which was then centrifuged to separate the marrow cells from bone pieces, etc.
  • the marrow cell ⁇ (consi ⁇ ting predominantly of red and white blood cells, and a very minute amount of me ⁇ enchymal stem cell ⁇ , etc.) were then dissociated into single cells by sequentially passing the complete medium containing the marrow cells through syringes fitted with a series of 16, 18, and 20 gauge needles.
  • the single cell su ⁇ pen ⁇ ion (which wa ⁇ made up of approximately 50-100 x 10 6 nucleated cells) wa ⁇ then subsequently plated in 100 mm dishe ⁇ for the purpo ⁇ e of ⁇ electively ⁇ eparating and/or i ⁇ olating the me ⁇ enchymal stem cells from the remaining cells found in the su ⁇ pen ⁇ ion.
  • the marrow stem cells (which contained little or no bone chip ⁇ but a great deal of blood) were added to the complete medium and fractionated with Percoll (Sigma, St. Loui ⁇ , MO) gradient ⁇ more particularly de ⁇ cribed below in Example 1.
  • Percoll Sigma, St. Loui ⁇ , MO
  • the Percoll gradients separated a large percentage of the red blood cells and the mononucleate hematopoietic cells from the low density platelet fraction which contained the marrow-derived mesenchymal stem cells.
  • the platelet fraction which contained approximately 30-50 x 10° cells wa ⁇ made up of an undetermined amount of platelets, 30-50 x 10° nucleated cell ⁇ , and only about 50-500 mesenchymal stem cells depending upon the age of the marrow donor.
  • the low density platelet fraction was then plated in the Petri dish for selective separation based upon cell adherence.
  • the marrow cells obtained from either the cancellous bone or iliac aspirate i.e. the primary cultures
  • the cells were then detached from the culture dishes utilizing a releasing agent such as trypsin with EDTA (ethylene diaminetetra-acetic acid) (0.25% trypsin, lmM EDTA (IX) , Gibco, Grand Island, NY) .
  • a releasing agent such as trypsin with EDTA (ethylene diaminetetra-acetic acid) (0.25% trypsin, lmM EDTA (IX) , Gibco, Grand Island, NY) .
  • trypsin with EDTA ethylene diaminetetra-acetic acid
  • trypsin, lmM EDTA (IX) Gibco, Grand Island, NY
  • the capacity of these undifferentiated cells to enter discrete lineage pathways is referred to as the mesengenic process, and is diagrammatically represented in Figure l.
  • MSCs are recruited to enter specific multi-step lineage pathways which eventually produce functionally differentiated tissues such as bone, cartilage, tendon, muscle, dermis, bone marrow stroma, and other mesenchymal connective tissues.
  • a detailed scheme for the differentiation pathway of bone forming cells is presented in Figure 2.
  • This lineage map implies the existence of individual controlling elements which recruit the MSCs into the osteogenic lineage, promote pre-osteoblast replication, and direct step-wise differentiation all the way to the terminal stage osteocyte. Sub ⁇ tantial work ha ⁇ been reported that supports the view that each step of this complex pathway is controlled by different bioactive factors.
  • progre ⁇ ion of each lineage step is under the control of unique bioactive factors including, but not limited to, the family of bone morphogenetic proteins.
  • Each modulator of the differentiation process may affect the rate of lineage progress ⁇ ion and/or may specifically affect individual step ⁇ along the pathway. That i ⁇ , whether a cell i ⁇ na ⁇ cently committed to a ⁇ pecific lineage, i ⁇ in a bio ⁇ ynthetically active state, or progresses to an end stage phenotype will depend on the variety and timing of bioactive factors in the local environment.
  • the bone and cartilage lineage potentials (i.e. osteo- chondrogenic potential) of fresh and expanded human mesenchymal stem cell ⁇ were determined u ⁇ ing two different in vivo assays in nude mice.
  • One assay involved the subcutaneous implantation of porous calcium phosphate ceramics loaded with cultured mesenchymal stem cell ⁇ ; the other involved peritoneal implantation of diffusion chambers inoculated with cultured mesenchymal stem cells.
  • Whole marrow and Percoll gradient separated aspirate fractions were also analyzed in these in vivo as ⁇ ay ⁇ .
  • Hi ⁇ tological evaluation ⁇ howed bone and cartilage formation in the ceramics implanted with the cultured mesenchymal stem cells derived from the femoral head and the iliac crest. Ceramics loaded with human mesenchymal stem cells at 5xl0 6 cells/ml formed bone within the pores, while ceramic ⁇ loaded with human me ⁇ enchymal ⁇ tem cells at 10x10° cell ⁇ / l formed cartilage within the pores. While whole marrow has now been shown to form bone when placed as a composite graft with ceramics in a subcutaneous site in nude mice, the amount of bone produced i ⁇ ⁇ ub ⁇ tantially le ⁇ than that ⁇ een when culture expanded marrow-derived mesenchymal stem cells are used.
  • culture expanded mesenchymal stem cells have the ability to differentiate into bone or cartilage when incubated as a graft in porous calcium phosphate ceramics.
  • the environmental factors which influence the mesenchymal stem cells to differentiate into bone or cartilage cells appear ⁇ , in part, to be the direct acce ⁇ sibility of the mesenchymal stem cells to growth and nutrient factors supplied by the vasculature in porous calcium phosphate ceramics; cells that are closely as ⁇ ociated with va ⁇ culature differentiate into bone cells while cells that are isolated from vasculature differentiate into cartilage cell ⁇ .
  • the exclu ⁇ ion of vasculature from the pores of ceramics loaded with concentrated human me ⁇ enchymal ⁇ tem cells prevented osteogenic differentiation and provided conditions permis ⁇ ive for chondrogene ⁇ i ⁇ .
  • the i ⁇ olated and culture expanded me ⁇ enchymal ⁇ tem cells can be utilized under certain ⁇ pecific condition ⁇ and/or under the influence of certain factor ⁇ , to differentiate and produce the desired cell phenotype needed for connective ti ⁇ ue repair or regeneration and/or for the implantation of variou ⁇ pro ⁇ thetic device ⁇ .
  • u ⁇ ing porou ⁇ ceramic cube ⁇ filled with culture-expanded human me ⁇ enchymal stem cells, bone formation inside the pore ⁇ of the ceramic ⁇ has been generated after ⁇ ubcutaneous incubations in immunocompatible host ⁇ .
  • rat marrow in a composite graft with porous ceramic was used to fill a segmental defect in the femur of the rat.
  • Factor ⁇ which ⁇ timulate o ⁇ teogenesis (i.e. are osteoinductive) from isolated human mesenchymal stem cells in accordance with the invention are present in several classes of molecules, including the following: bone morphogenic proteins, such as BMP-2 (14) and BMP-3 (15); growth factors, such as basic fibroblast growth factor (bFGF); glucocorticoid ⁇ , ⁇ uch a ⁇ dexamethasone (16); and pro ⁇ taglandin ⁇ , such as prostaglandin El (22).
  • bone morphogenic proteins such as BMP-2 (14) and BMP-3 (15)
  • growth factors such as basic fibroblast growth factor (bFGF); glucocorticoid ⁇ , ⁇ uch a ⁇ dexamethasone (16); and pro ⁇ taglandin ⁇ , such as prostaglandin El (22).
  • ⁇ corbic acid and it ⁇ analog ⁇ are effective adjunct factor ⁇ for advanced differentiation, although alone they do not induce o ⁇ teogenic differentiation.
  • ⁇ uch a ⁇ a ⁇ corbic acid-2- pho ⁇ phate (17) and glycerol phosphates, such as ⁇ - glyceropho ⁇ phate (18) are effective adjunct factor ⁇ for advanced differentiation, although alone they do not induce o ⁇ teogenic differentiation.
  • TGF-3 tran ⁇ forming growth factor- ⁇
  • Inhibin A (20)
  • CSA chondrogenic ⁇ timulatory activity factor
  • bone morphogenic protein ⁇ ⁇ uch a ⁇ BMP-4 (22)
  • collagenous extracellular matrix molecules including type I collagen, particularly a ⁇ a gel (23)
  • vitamin A analogs such as retinoic acid (24).
  • Factors which have stromagenic inductive activity on human MSC ⁇ are al ⁇ o pre ⁇ ent in ⁇ everal classe ⁇ of molecule ⁇ , especially the interleukin ⁇ , such as IL-l ⁇ (25) and IL-2 (26).
  • Factors which have myogenic inductive activity on human MSCs are al ⁇ o present in ⁇ everal cla ⁇ se ⁇ of molecule ⁇ , especially cytidine analogs, such as 5-azacytidine and 5-aza- 2'-deoxycytidine.
  • MSCs me ⁇ enchymal ⁇ tem cell ⁇
  • Human MSCs were harvested and isolated from bone marrow as described above. These cells were culture-expanded in DMEM-LG medium containing preselected 10% fetal bovine serum (Complete Medium). Fresh Complete Medium was replaced every 3-4 days until the cultures were near confluence, at which time the cells were liberated off the plates with trypsin, and reseeded onto new di ⁇ hes at approximately 40% confluence (400,000 cells per 100 mm dish).
  • replated MSCs were allowed to attach overnight, after which the Complete Medium was replaced by a medium composed of DMEM-LG, 10% fetal bovine serum, and either 100 nM dexametha ⁇ one alone, or 100 nM dexcunethasone with 50 ⁇ M ascorbic acid-2-pho ⁇ phate, and 10 mM 8-glyceropho ⁇ phate (O ⁇ teogenic Supplement).
  • O ⁇ teogenic Supplement was replaced every 3 days.
  • Cells were examined daily for morphologic changes. Selected plates were then analyzed for cell ⁇ urface alkaline phosphata ⁇ e (AP) activity, a marker for cell ⁇ which have entered the o ⁇ teogenic lineage.
  • AP alkaline phosphata ⁇ e
  • Control culture ⁇ fed only Complete Medium never developed the ⁇ e mineralized bone nodule ⁇ , and only rarely contained AP po ⁇ itive cell ⁇ .
  • MSCs treated with Osteogenic Supplement uniformly acquired AP activity and synthesized mineralized extracellular matrix nodules throughout the di ⁇ h.
  • the pre ⁇ ence of ascorbic acid- 2-phosphate and 0-glyceropho ⁇ phate in the complete O ⁇ teogenic Supplement further supports extracellular matrix maturation and mineral deposition, respectively.
  • Figure 3 graphically demonstrates the increase in alkaline phosphatase enzyme activity as a function of time in culture. By day 8 and beyond, sub ⁇ tantially more enzyme activity is ob ⁇ erved in cell ⁇ exposed to Osteogenic Supplement ⁇ (OS) than tho ⁇ e cultured with control medium.
  • OS Osteogenic Supplement ⁇
  • MSCs were purified from 3 different patients (ages 26-47), culture expanded (27), and seeded overnight onto 48-well culture plates at 20% confluence in DMEM-LG with 10% FBS from selected lots.
  • Base media for comparison were DMEM-LG, BGJ b , ⁇ MEM, and DMEM/F-12 (1:1).
  • Triplicate cultures for each as ⁇ ay were grown in 10% FBS in the ab ⁇ ence or pre ⁇ ence of "Osteogenic Supplements" (OS) (100 nM dexamethasone, 50 ⁇ M a ⁇ corbic acid-2-pho ⁇ phate, and IOI ⁇ LM / 8-glycerophosphate (28). Media were changed every 3 days.
  • OS Steogenic Supplements
  • AP enzyme activity was calculated by incubating live cultures with 5 mM p-nitrophenylphosphate in 50 mM Tris, 150 mM NaCl, pH 9.0 and quantifying the colorimetric reaction by scanning the samples at 405 nm on an ELISA plate reader. AP enzyme activity was expres ⁇ ed a ⁇ nanomoles of product/minute/10 3 cells. The percentage of AP- po ⁇ itive cells in each well was determined from the stained cultures, and the number of mineralized nodules per well were counted.
  • ⁇ MEM+OS would foster even more mineralized foci than DMEM+OS.
  • Difference ⁇ in the media favoring maintenance of the MSC phenotype (DMEM) evidenced by MSC-specific im unostaining, or maximal recruitment and induction into the osteogenic lineage ( ⁇ MEM+OS), noted by the percent AP-positive cell ⁇ and AP activity, are inherently intere ⁇ ting and warrant further examination.
  • the use of various monoclonal and polyclonal antibodies against specific cell and matrix components during this inductive phenomenon are currently underway, and will provide further in ⁇ ight into the molecular nature of the in vitro differentiation process.
  • me ⁇ enchymal progenitor cell ⁇ derived from bone marrow are capable of differentiating into o ⁇ teobla ⁇ t ⁇ .
  • the ⁇ e me ⁇ enchymal ⁇ tem cell ⁇ also give rise to cartilage, tendon, ligament, muscle, and other tissue ⁇ .
  • knowledge of the ⁇ tep ⁇ involved in the commitment and differentiation of MSC ⁇ along the ⁇ e various lineages has been restricted, in part, by the lack of probes specific for cells at various stage ⁇ within the o ⁇ teogenic or other differentiation pathway ⁇ .
  • MSCs were purified from 5 different patients (ages 28-46), culture expanded (29), and grown in DMEM-LG with 10% FBS and "Osteogenic Supplement ⁇ " (100 nM dexametha ⁇ one, 50 ⁇ M a ⁇ corbic acid-2-pho ⁇ phate, and lOmM j8-glycerophosphate (28).
  • FBS FBS
  • "Osteogenic Supplement ⁇ " 100 nM dexametha ⁇ one, 50 ⁇ M a ⁇ corbic acid-2-pho ⁇ phate, and lOmM j8-glycerophosphate (28).
  • the cells were liberated from the plates with 5 mM EGTA. Approximately 4 million 3 and 6 day cells were pooled for each of five weekly immunizations into Balbc/J mice.
  • the pattern ⁇ of reactivity vary; ⁇ ome hybridoma ⁇ upernatant ⁇ react with a large population of cell ⁇ within the o ⁇ teogenic collar and osteoprogenitor-containing perio ⁇ teum, while other ⁇ react with only those cell ⁇ which appear to be actively involved in matrix synthesis. Two hybridoma colonies appear to react with osteogenic cells a ⁇ well a ⁇ hypertrophic chondrocyte ⁇ .
  • the re ⁇ ult ⁇ are ⁇ ummarized in Table 1.
  • Table 1 ⁇ how ⁇ the immunoreactivity of selected hybridoma colonie ⁇ again ⁇ t untreated MSCs, or MSCs cultured with Osteogenic Supplements (OS) for 3 or 6 days. Numbers reflect the relative amount of antibody bound in the ELISA assay described above.
  • MSCs mesenchymal stem cells
  • the additive to Complete Medium which constitutes Chondrogenic Supplement in the example above is only one of the factors known to stimulate chondrogenic cell differentiation or proliferation in vitro .
  • MSCs were grown under conditions which favor MSC proliferation without differentiation using medium consi ⁇ ting of DMEM-LG containing preselected 10% fetal bovine serum (Complete Medium), or conditions which favor expression and differentiation into the marrow stromal phenotype using medium comprising Complete Medium plus 10 U/ml Interleukin-l ⁇ (IL-l ⁇ ) (Stromagenic Supplement (SS)).
  • Conditioned culture media from these tis ⁇ ue culture populations were analyzed for the pre ⁇ ence of cytokine ⁇ u ⁇ ing commercial ⁇ andwich ELISA bioa ⁇ ay ⁇ (R&D Systems) .
  • the cytokine ⁇ that were assayed are those that are known to be secreted by stromal cells and which influence hematopoiesis.
  • the ⁇ e include interleukin-3 (IL-3), interleukin-6 (IL-6), granulocyte colony ⁇ timulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), stem cell factor (SCF), leukemia inhibitory factor (LIF) and transforming growth factor-beta-2 (TGF-32).
  • IL-3 interleukin-3
  • IL-6 interleukin-6
  • G-CSF granulocyte colony ⁇ timulating factor
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • SCF stem cell factor
  • LIF leukemia inhibitory factor
  • TGF-32 transforming growth factor-beta-2
  • Figure 6 illu ⁇ trate ⁇ the cytokine expre ⁇ ion of human MSC ⁇ under the two plating conditions.
  • MSC ⁇ expre ⁇ ed G-CSF, GM-CSF, LIF and SCF at very low levels, but expre ⁇ IL-6 in high abundance.
  • IL-6 in high abundance.
  • MSC ⁇ did not expre ⁇ IL-3 or TGF-jS2 under either of the two culture condition ⁇ .
  • the ⁇ e data ⁇ how that IL-I- ⁇ enhance ⁇ MSC expression of a cytokine profile that ha ⁇ been documented to support differentiation of the hematopoietic ⁇ tem cell and which is characteri ⁇ tic of differentiated marrow ⁇ tromal cells.
  • the purpose of the study de ⁇ cribed in this example was to demonstrate that 5-azacytidine induces mesenchymal stem cells (MSCs) to differentiate along the myogenic lineage.
  • MSCs mesenchymal stem cells
  • 5-azacytidine (5-aza-CR; Sigma Chemical Co., St. Louis, MO), an analogue of cytidine, causes hypomethylation of some cytosine in DNA which may be involved in activating phenotype-specific gene ⁇ .
  • Femora and tibiae of male Fisher rats (Charle ⁇ River, Indianapoli ⁇ , IN) with an average body weight of 100 g were collected and the adherent ⁇ oft ti ⁇ ues were removed.
  • Several isolate ⁇ of marrow cell ⁇ were from 250 g rat ⁇ . Meticulou ⁇ di ⁇ ection of the long bone ⁇ to remove ⁇ oft ti ⁇ ue wa ⁇ done to insure that myogenic precursors were not carried into the marrow preparation. In this regard, myogenic cells were never ob ⁇ erved in untreated MSC culture ⁇ . Both end ⁇ of the bone ⁇ were cut away from the diaphy ⁇ e ⁇ with bone ⁇ ci ⁇ or ⁇ .
  • the bone marrow plug ⁇ were hydrostatically expelled from the bones by insertion of 18-gauge needles fastened to 10 ml syringes filled with Complete Medium consisting of DMEM containing selected lots of 10% fetal calf serum (FCS; IR Scientific Inc., Woodland, CA), 5% horse serum (HS; Hazleton Biologies Inc., Lenexa, KS), and antibiotics (Gibco Laboratories; penicillin G, 100 U/ml; streptomycin, 100 ⁇ g/ml; amphotericin B, 0.25 ⁇ g/ml) .
  • FCS fetal calf serum
  • HS horse serum
  • antibiotics Gibco Laboratories; penicillin G, 100 U/ml; streptomycin, 100 ⁇ g/ml; amphotericin B, 0.25 ⁇ g/ml
  • the needles were inserted into the distal ends of femora and proximal ends of tibias and the marrow plug ⁇ expelled from the oppo ⁇ ite ends.
  • the marrow plugs were disaggregated by sequential passage through 18-gauge, 20-gauge, and 22-gauge needles and these disper ⁇ ed cell ⁇ were centrifuged and re ⁇ u ⁇ pended twice in Complete Medium. After the cell ⁇ were counted in a hemocytometer, 5xl0 7 cells in 7-10 ml of complete medium were introduced into 100 mm petri dishe ⁇ . Three days later, the medium was changed and the non-adherent cells discarded. Medium was completely replaced every 3 days.
  • the twice pa ⁇ aged MSC ⁇ were ⁇ eeded into 35-mm di ⁇ hes at three cell densitie ⁇ , 500, 5,000, and 50,000 cells/dish.
  • the ⁇ e culture ⁇ were treated for 24 hr with Myogenic Medium con ⁇ isting of complete medium containing various concentration ⁇ of 5-aza-CR.
  • the medium was changed to complete medium without added 5-aza-CR and subsequently changed twice a week until the experiment was terminated, 40 days after the treatment.
  • various culture conditions were tested to attempt to optimize the 5-aza-CR effects, especially to optimize myogene ⁇ is.
  • Twice pa ⁇ aged rat bone marrow MSC ⁇ were seeded into 35-mm di ⁇ he ⁇ at 5,000 cell ⁇ /dish and treated with four concentrations (0.1 ⁇ M, 0.3 ⁇ M, 1 ⁇ M and 10 ⁇ M) of 5-aza-2'- deoxycytidine (5-aza-dCR; Sigma Chemical Co.) in the same way as described above for 5-aza-CR.
  • 5-aza-dCR 5-aza-2'- deoxycytidine
  • Sigma Chemical Co. 5-aza-2'- deoxycytidine
  • the living cultures were examined every day with a phase-contrast microscope (Olympus Optical Co., Ltd., Tokyo, Japan), and eventually some of the culture ⁇ were fixed for histology or immunohistochemistry.
  • Muscle cells were first identified morphologically in phase contrast by the presence of multinucleated myotubes, and ⁇ ubsequently immunohistochemically by the pre ⁇ ence of the ⁇ keletal muscle-specific protein, myosin. Contraction of the putative muscle cells was stimulated by a drop of 1 mM acetylcholine (Sigma Chemical Co.) in Tyrode's.
  • cultured cell ⁇ were fixed with -20°C methanol (Fi ⁇ her Scientific Co., Fair Lawn, NJ) for 10 min and incubated with a mouse monoclonal antibody to rat fa ⁇ t twitch ⁇ keletal myo ⁇ in (Sigma Chemical Co.; ascites fluid, 1/400 dilution) in PBS (pho ⁇ phate buffered ⁇ aline, pH7.4) containing 0.1% BSA (bovine serum albumin; Sigma Chemical Co.).
  • PBS pho ⁇ phate buffered ⁇ aline, pH7.4
  • BSA bovine serum albumin
  • the second antibody was biotin-conjugated ⁇ heep anti-mou ⁇ e IgG (Organon Teknika Corp., We ⁇ t Che ⁇ ter, PA; 1/50 dilution) followed by treatment with Texas red-conjugated avidin (Organon Teknika Corp.; 1/4,000 dilution). All incubations were for 30 min at room temperature, each preceded by blocking for 5 min with PBS containing 1% BSA, followed by two 5-min washes in PBS.
  • the cells were mounted in Fluoromount-G (Fisher Biotech, Pitt ⁇ burgh, PA) and ob ⁇ erved with an Olympu ⁇ micro ⁇ cope (BH-2) equipped for fluorescence and photographed with Kodak TMAX 400 film.
  • Fluoromount-G Fluoromount-G (Fisher Biotech, Pitt ⁇ burgh, PA) and ob ⁇ erved with an Olympu ⁇ micro ⁇ cope (BH-2) equipped for fluorescence and photographed with Kodak TMAX 400 film.
  • Second passage rat bone marrow MSCs were plated into 96-well plates at limiting dilution of one cell/well; cell ⁇ were plated in medium con ⁇ isting of 50% Complete Medium and 50% conditioned medium, which wa ⁇ obtained from rat bone marrow cell ⁇ near confluence cultured in Complete Medium for 2 day ⁇ . From a total of 384 well ⁇ , 50 colonie ⁇ were detected; these were subcultured, maintained, and eventually 4 survived. These 4 clones were treated with 5-aza-CR as mentioned above and scored for myogenic or adipocytic morphologies.
  • rat brain fibroblast ⁇ to either 5-aza-CR or 5-aza-CdR.
  • Whole cerebra of brain ⁇ of three male Fisher rats were collected from the inside of the skulls and cut into small pieces with a sharp scalpel. The ⁇ e piece ⁇ were tran ⁇ ferred to a 50-ml conical centrifuge tube, centrifuged at 500 xg for 10 min, re ⁇ u ⁇ pended in 10 ml of Tyrode' ⁇ balanced ⁇ alt ⁇ olution, and homogenized with a loose-fitting Dounce homogenizer.
  • the relea ⁇ ed cells were pa ⁇ ed through a 110- ⁇ m Nitex filter, centrifuged, re ⁇ u ⁇ pended in 10 ml of low gluco ⁇ e DMEM-LG (Gibco Laboratorie ⁇ ) containing 10% FCS, and cultured in three 100-mm culture dishes at 37°C in a C0 2 incubator. The medium was changed twice a week and cells were cultured until the dishes reached confluence.
  • rat brain fibroblasts were seeded into 35-mm dishes at a density of 50,000 cell ⁇ /di ⁇ h and treated with 1 ⁇ M, 3 ⁇ M or 10 ⁇ M 5-aza-CR or 0.1 ⁇ M, 0.3 ⁇ M or 1 ⁇ M 5-aza-CdR in the same way as rat marrow MSCs.
  • the medium wa ⁇ changed to DMEM-LG containing 10% FCS, 5% HS and 50 nM hydrocortisone without added 5-aza-CR or 5-aza-CdR and sub ⁇ equently changed twice a week until the experiment was terminated.
  • Myogenic cells derived from rat bone marrow MSCs were compared with normal fetal rat myogenic cell ⁇ , since a sub ⁇ tantial data ba ⁇ e exi ⁇ t ⁇ for the latter.
  • Mu ⁇ cle cell ⁇ were di ⁇ ociated from the hindlimb mu ⁇ cle ⁇ of 17-day-old Fisher rat fetuses with 0.2% trypsin (Sigma Chemical Co.) in calcium- and magnesium-free Tyrode's for 35 min at 37°C with occasional agitation. After they were passed through a 110- ⁇ m Nitex filter, the concentration of fibroblast ⁇ was reduced by incubating cell suspen ⁇ ion ⁇ for 30 min in Falcon plastic dishe ⁇ , which re ⁇ ult ⁇ in preferential attachment of the fibroblast ⁇ .
  • the number of ⁇ uch multinucleated cell ⁇ increased as isolated colonie ⁇ or grouping ⁇ , and reached a maximum (9 colonie ⁇ in 10 of 35-mm di ⁇ he ⁇ ) 2 weeks after the initial treatment.
  • the number of such cells decreased (6 colonies in 10 of 35-mm dishe ⁇ ) by 5 week ⁇ after treatment; 7 disappeared probably due to their contraction and detachment from the dishe ⁇ and 4 new colonie ⁇ appeared during thi ⁇ time period; a substantial proportion of the multinucleated cells remained for up to 40 day ⁇ after the initial exposure, which was the longest observational period.
  • the contraction of these cells could also be stimulated by placing a drop of an acetylcholine solution onto these cell ⁇ , which i ⁇ a further indication that the ⁇ e cell ⁇ are myogenic.
  • SI* Survival Index
  • 5-aza-2' -deoxycyti ine 5-aza-2' -deoxycyti ine
  • rat bone marrow MSCs were treated with 0.3 ⁇ M, l ⁇ M, and 10 ⁇ M 5-aza-dCR in the same way as 5-aza-CR.
  • concentrations tested 0.3 ⁇ M 5-aza-CdR gave the highest incidence of myogenic conversion, and the observed incidence was much higher than for cells exposed to 10 ⁇ M 5-aza-CR (Table 3) .
  • MSCs were seeded at 200 cells/35mm dish and treated with 5-aza-dCR or 5-aza-CR 24 hr after plating. After 14 day ⁇ , colonie ⁇ containing more than 10 cells were counted, and thi ⁇ number wa ⁇ multiplied by 100% and divided by 200 to generate the percentage.
  • ⁇ econd pa ⁇ age rat bone marrow MSC ⁇ were cloned as described herein.
  • Four clones of indistingui ⁇ hable morphologies were obtained from this procedure and were exposed to 5-aza-CR for 24 hr; for emphasis, no cells in these clones exhibited muscle-like characteristics or positive immunostaining for muscle ⁇ pecific myo ⁇ in prior to exposure to 5-aza-CR.
  • Fir ⁇ t pa ⁇ age rat bone marrow-derived MSC ⁇ were exposed to 10 ⁇ M 5-aza-CR for 24 hr and cloned. From a total of 768 well ⁇ , 136 colonie ⁇ were detected. Of the ⁇ e 136 colonie ⁇ , 7 (5%) exhibited a myogenic phenotype, 27 (20%) exhibited an adipocytic phenotype, and the other colonies lacked morphologies obviously related to discernible phenotypes.
  • Rat brain fibrobla ⁇ t ⁇ were ⁇ eeded into 35-mm dishes at a density of 50,000 cells/dish and treated with 1 ⁇ M, 3 ⁇ M or 10 ⁇ M 5-aza-CR or 0.1 ⁇ M, 0.3 ⁇ M or 1 ⁇ M 5-aza-dCR in the same way as for rat MSCs.
  • Each group had 9 dishe ⁇ and cells were surveyed until 14 days after exposure. At day 7, all di ⁇ he ⁇ reached confluence, except for the group treated with 10 ⁇ M 5-aza-CR.
  • MSC ⁇ were collected from the bone marrow of young (4 week-old, 100 g) and adult (3 month-old, 250 g) donor rat ⁇ and passaged, and the number of colonies of myogenic phenotype after exposure to 5-aza-CR were compared (Table 4).
  • MSCs from young donor rats had more myogenic colonies than those from adult rats.
  • Second passage cultures of young donor MSCs exposed to 5-aza-CR produced more myogenic colonies compared with MSCs from older donors tested in culture ⁇ from the fir ⁇ t to fourth pa ⁇ age.
  • MSCs were cultured in DMEM with 10% FCS, 5% HS and 50 ⁇ M HC, with or without bFGF. The numbers for the incidence of myotubes indicate the total number of phenotypically discernible colonies or groupings observed and the total number of culture dishes examined.
  • MSCs were obtained from young (lOOg) or old (250g) rats.
  • bone marrow-derived MSC ⁇ were plated at 500 cell ⁇ /dish, 5,000 cells/dish, and 50,000 cell ⁇ /di ⁇ h and then exposed to 5-aza-CR.
  • myogenic cells were first observed at 20 days after treatment, with the cells becoming confluent 25 days after treatment; 2 clusters of myogenic cells were observed in 5 dishes 29 days after treatment.
  • myogenic cell ⁇ were first observed at 7 days, with the cell ⁇ becoming confluent 10 day ⁇ after treatment; 3 clu ⁇ ter ⁇ were observed in 4 di ⁇ he ⁇ 14 day ⁇ after treatment.
  • myogenic cells were observed at 6 days, with the cells becoming confluent at 7 days after treatment; 10 clu ⁇ ter ⁇ were ob ⁇ erved in 5 di ⁇ he ⁇ 14 day ⁇ after treatment.
  • the ob ⁇ ervation ⁇ pre ⁇ ented here indicate that rat bone marrow MSCs have the capacity to differentiate into the myogenic lineage in vitro following a brief exposure to 5-aza-CR.
  • the ob ⁇ erved myogenic cell ⁇ exhibited the characteri ⁇ tic multinucleated morphology of myotube ⁇ , contracted ⁇ pontaneou ⁇ ly, contracted when expo ⁇ ed to acetylcholine, and ⁇ tained positively with a monoclonal antibody to skeletal muscle- ⁇ pecific myo ⁇ in, although these myotubes never exhibited apparent striation ⁇ .
  • a clone of rat bone marrow MSCs wa ⁇ converted to both myogenic and adipocytic phenotypes after treatment with 5-aza-CR, which we interpret to mean that non-muscle progenitor cells were converted into the ⁇ e two phenotype ⁇ . Since ⁇ keletal muscle has not been observed in bone marrow, we believe that 5-aza-CR converts the ⁇ e marrow-derived MSC ⁇ into the myogenic cells.
  • the objective of the present study was to further establish the phenotypic characteristic ⁇ of cultured MSC ⁇ through identification of a cytokine expression profile.
  • MSC cytokine expre ⁇ ion under culture conditions that we have previou ⁇ ly reported allow MSC ⁇ to mitotically expand without differentiation (constitutive culture-expansion medium) .
  • IL-l ⁇ which i ⁇ ⁇ ecreted into the marrow microenvironment by a variety of cells during the inflammatory response, has been reported to enhance the bone marrow stroma' ⁇ capacity to ⁇ upport hematopoiesis and thus may play a role in controlling the differentiation and/or expression of bone marrow stromal fibroblasts.
  • Bone marrow wa ⁇ obtained from six human donors, 3 male and 3 female of diverse age ⁇ (Table 6).
  • bone marrow a ⁇ pirates were transferred from their syringes into 50 ml conical tubes containing 25 ml of complete medium con ⁇ i ⁇ ting of Dulbecco' ⁇ Modified Eagle ⁇ Medium ⁇ upplemented with fetal bovine serum (FBS) from selected lots, to a final volume of 10%.
  • the tubes were spun in a Beckman table top centrifuge at 1200 rpm in a GS-6 swing bucket rotor for 5 min to pellet the cells.
  • the layer of fat that forms at the top of the sample ⁇ and the ⁇ upernatant ⁇ were a ⁇ pirated u ⁇ ing a ⁇ erological pipet and di ⁇ carded.
  • the cell pellet ⁇ were resuspended to a volume of 5 ml with Complete Medium and then transferred to the top of preformed gradients of 70% Percoll.
  • the sample ⁇ were loaded into a Sorvall GS-34 fixed angle rotor and centrifuged in a Sorvall High Speed Centrifuge at 460 x g for 15 min.
  • the low den ⁇ ity fraction of approximately 15 ml (pooled den ⁇ ity 1.03 g/ml) wa ⁇ collected from each gradient and tran ⁇ ferred to 50 ml conical tube ⁇ to which were added 30 ml Complete Medium.
  • the tube ⁇ were centrifuged at 1200 rpm to pellet the cell ⁇ .
  • the ⁇ upernatant ⁇ were di ⁇ carded and the cells were resuspended in 20 ml of Complete Medium and counted with a hemocytometer after ly ⁇ ing red blood cells with 4% acetic acid. Cells were adju ⁇ ted to a concentrated of 5xl0 7 cell ⁇ per 7 ml and seeded onto 100-mm culture plates at 7 ml per plate.
  • Marrow-derived MSCs were cultured in Complete Medium at 37°C in a humidified atmo ⁇ phere containing 95% air and 5% C0 2 , with medium change ⁇ every 3-4 day ⁇ .
  • the cell ⁇ were detached with 0.25% trypsin containing 1 mM EDTA (GIBCO) for 5 min at 37°C.
  • the enzymatic activity of trypsin was stopped by adding 1/2 volume of FBS.
  • the cells were counted, split 1:3, and replated in 7 ml of Complete Medium. These first passage cells were allowed to divide for 4-6 days until they became near confluent. Near-confluent first passage cells were trypsinized and replated into the a ⁇ ay format ⁇ a ⁇ described below.
  • cytokine expres ⁇ ion by MSCs were measured using quantitative ELISA.
  • ELISA kits (R&D System ⁇ , Minneapolis MN) with antibody ⁇ pecificitie ⁇ for the following cytokines were purchased; interleukin-3 (IL-3), interleukin-6 (IL-6), interleukin-11 (IL-11), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating activity (M-CSF), stem cell factor (SCF), leukemia inhibitory factor (LIF) and transforming growth factor-beta-2 (TGF- / S-2).
  • IL-3 interleukin-3
  • IL-6 interleukin-6
  • IL-11 interleukin-11
  • G-CSF granulocyte colony stimulating factor
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • M-CSF macrophage colony stimulating activity
  • SCF stem cell factor
  • a ⁇ ay ⁇ were conducted by applying 100 ⁇ l of culture ⁇ upernatant onto the well ⁇ of the ELISA plate followed by proce ⁇ ing the plate ⁇ per manufacturer' ⁇ in ⁇ tructions. Standard curves were generated using standard cytokines supplied with the kit ⁇ and diluted to the appropriate concentrations. In some cases (particularly for the IL-6 as ⁇ ay), the ⁇ upernatant ⁇ had to be diluted ⁇ ub ⁇ tantially to generate low enough ab ⁇ orbance measurements that could be quantified accurately from the standard curves.
  • the cytokine ⁇ expressed in terms of pg/10,000 cell ⁇ in 24 or 48 hour ⁇ , from lowe ⁇ t to highe ⁇ t were: G-CSF, SCF, LIF, M-CSF, IL-11 and IL-6.
  • Three cytokine ⁇ were not detected in the supematants under constitutive culture-expansion conditions: GM-CSF, IL-3 and TGF-02. Large differences were observed in the average cytokine expression of each cytokine in comparison to the average levels of expression of other cytokine ⁇ . At the extremes, he average detectable level of G-CSF expression (10 pg/10,000 cells/24 hours) was over 700 fold lower than the average level of expression of IL-6 (7421 pg/10,000 cells/24 hours) .
  • SS medium increased the expression of several cytokines by MSCs in a concentration dependent manner.
  • Figure 14 illu ⁇ trate ⁇ the 24 hour re ⁇ pon ⁇ e of second passage MSC ⁇ to increa ⁇ ing concentration ⁇ of IL-l ⁇ in term ⁇ of expression of GM-CSF.
  • the ⁇ e data were u ⁇ ed to identify the concentration of IL-l ⁇ to ⁇ upplement to the culture media in the experiment ⁇ described below.
  • 10 U/ml IL-l ⁇ were added to the culture media.
  • IL-l ⁇ Culture medium ⁇ upplemented with 10 U/ml IL-l ⁇ induced ⁇ tatistically significant up-regulation in the expression of G-CSF (P ⁇ .05), M-CSF (p ⁇ 0.02), LIF (p ⁇ 0.02), IL-6 (p ⁇ 0.01) and IL-11 (p ⁇ 0.06) relative to cells cultured in con ⁇ titutive culture-expan ⁇ ion medium.
  • IL-l ⁇ induced the expression of GM-CSF which was not detectable in constitutive culture-expansion medium.
  • IL-l ⁇ had no statistically significant effect on the expression of SCF relative to the level of expression under constitutive culture-expansion medium conditions.
  • the fold increase in response to IL-l ⁇ varied depending on the cytokine.
  • IL-6 (25.1 +/- 13.4 fold increase) wa ⁇ ⁇ timulated to the greate ⁇ t extent, followed by LIF (9.2 + 6.9 fold), M-CSF (5.2 + 1.7 fold) and IL-ll (4.9 + 3.3 fold).
  • LIF 9.2 + 6.9 fold
  • M-CSF 5.2 + 1.7 fold
  • IL-ll 4.9 + 3.3 fold.
  • the average fold increase for G-CSF and GM-CSF were not calculated, since these cytokines were not detected in some or all constitutive culture-expansion cultures.
  • MSCs express a unique profile of cytokines which include G-CSF, M-CSF, SCF, LIF, IL-6 and IL-ll under constitutive culture-expansion conditions. They do not express GM-CSF, IL-3 and TGF-32 under these conditions. OS down-regulates the expression of LIF, IL-6 and IL-ll, while not affecting the expression of the other cytokines expressed under constitutive culture conditions. OS was not observed to up-regulate the expression of any of the cytokines assayed in this study.
  • SS up-regulates the expression of G-CSF, M-CSF, LIF, IL-6 and IL-ll, and induces the expression of GM-CSF which was not detected under constitutive culture-expansion conditions.
  • SS had no effect on SCF expression, and was not observed to down-regulate any of the cytokines assayed in this study.
  • the identity of the cytokine profile should provide clues to determine the role that these cells play in the microenvironment of bone marrow which provides the inductive and regulatory information that ⁇ upport ⁇ hematopoiesis.
  • the alteration ⁇ in thi ⁇ cytokine profile in response to OS and SS identify specific cytokine ⁇ who ⁇ e level ⁇ of expression change as MSCs differentiate or modulate their phenotype in response to regulatory molecules.
  • IL-l ⁇ which is relea ⁇ ed in the marrow microenvironment by a variety of cell type ⁇ during inflammatory response ⁇ , induce ⁇ MSC ⁇ to up-regulate expre ⁇ ion of cytokines that support granulocytic (G-CSF and GM-CSF), monocytic/ osteoclastic (GM-CSF, LIF, M-CSF, IL-6) and megakaryocytic (IL- 11) differentiation.
  • G-CSF and GM-CSF granulocytic
  • GM-CSF monocytic/ osteoclastic
  • IL- 11 megakaryocytic differentiation.
  • IL-l ⁇ ha ⁇ been ⁇ hown to protect bone marrow from radio- and hemo-ablation.
  • the IL-l ⁇ -induced up-regulation of cytokine expression by MSC ⁇ likely plays a role in the mechanism ⁇ of IL-l ⁇ ' ⁇ protective effect ⁇ .
  • Dexametha ⁇ one which induce ⁇ MSCs to differentiate into o ⁇ teobla ⁇ t ⁇ , attenuate ⁇ the expre ⁇ ion of monocytic/osteocla ⁇ tic (LIF, IL-6) and megakaryocytic (IL-ll) ⁇ upportive cytokine ⁇ , and ha ⁇ no effect on the expre ⁇ ion of cytokine ⁇ that ⁇ upport granulocytic progenitor ⁇ (G-CSF, GM-CSF).
  • the three cytokine ⁇ inhibited by dexametha ⁇ one are of interest because each mediates its signal through a receptor that uses gpl30 in its signaling pathway.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Rheumatology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Developmental Biology & Embryology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Materials For Medical Uses (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Methods for in vitro or ex vivo lineage directed induction of isolated, culture expanded human mesenchymal stem cells comprising contacting the mesenchymal stem cells with a bioactive factor effective to induce differentiation thereof into a lineage of choice as well as such compositions including isolated culture expanded human mesenchymal stem cells and bioactive factors effective to induce directed lineage induction are disclosed. Further disclosed is this method which also includes introducing such culturally expanded lineage-induced mesenchymal stem cells into a host from which they have originated for purposes of mesenchymal tissue regeneration or repair.

Description

LINEAGE-DIRECTED INDUCTION OF HUMAN MESENCHYMAL STEM CELL DIFFERENTIATION
The present invention provides methods for directing mesenchymal stem cells cultivated in vitro to differentiate into specific cell lineage pathways prior to, or at the time of, their implantation for the therapeutic treatment of pathologic conditions in humans and other species.
Mesenchymal stem cells (MSCs) are the formative pluripotent blast or embryonic-like cells found in bone marrow, blood, dermis, and periosteum that are capable of differentiating into specific types of mesenchymal or connective tissues including adipose, osseous, cartilaginous, elastic, muscular, and fibrous connective tissues. The specific differentiation pathway which these cells enter depends upon various influences from mechanical influences and/or endogenous bioactive factors, such as growth factors, cytokines, and/or local microenvironmental conditions established by host tissues. Although these cells are normally present at very low frequencies in bone marrow, a process for isolating, purifying, and mitotically expanding the population of these cells in tissue culture is reported in Caplan et al . U.S. Patent Nos . 5,197,985 and 5,226,914.
In prenatal organisms, the differentiation of MSCs into specialized connective tissue cells is well established; for example embryonic chick, mouse or human limb bud mesenchymal cells differentiate into cartilage, bone and other connective tissues (1-5). In addition, a clonal rat fetus calvarial cell line has also been shown to differentiate into muscle, fat, cartilage, and bone (6). The existence of MSCs in post-natal organisms has not been widely studied with the objective of showing the differentiation of post-embryonic cells into several mesoder al phenotypes. The few studies which have been done involve the formation of bone and cartilage by bone marrow cells following their encasement in diffusion chambers and in vivo transplantation (7, 8). Recently, bone marrow-derived cells from young rabbits (800-1,000 g) have been shown to form adipocytic and osteogenic cells in vivo (9) and cloned bone marrow stromal cells of post-natal mice were shown to form adipocytes and osteogenic cells (10). Likewise, cells from chick periosteum have been isolated, expanded in culture, and, under high density conditions in vitro , shown to differentiate into cartilage and bone (11). Rat bone marrow-derived mesenchymal cells have been shown to have the capacity to differentiate into osteoblasts and chondrocytes when implanted in vivo (12, 6). Cells from various marrow sources of post-natal organisms have never been observed to exhibit myogenic properties, with multinuclear appearance being the most easily recognized characteristic in culture.
In a first aspect, the invention provides a method for effecting the lineage-directed induction of isolated, culture-expanded human mesenchymal stem cells which comprises contacting mesenchymal stem cells with a bioactive factor or combination of factors effective to induce differentiation thereof into a lineage of choice. More particularly, this method is one in which the bioactive factor induces differentiation of such cells into a mesenchymal lineage selected from the group consisting of osteogenic, chondrogenic, tendonogenic, ligamentogenic, myogenic, marrow stro agenic, adipogenic and dermogenic. Preferably, the cells are contacted ex vivo with one or more bioactive factors in this aspect, thereby providing a method free of any risks that may be associated with in vivo administration of any bioactive factors.
In another aspect, the method of the invention further provides administering to an individual in need thereof isolated culture-expanded human mesenchymal stem cells and a bioactive factor effective to induce differentiation of such cells into a lineage of choice. Preferably, the mesenchymal stem cells and bioactive factor are administered together or they may alternatively be administered separately. Particularly, this aspect of the method comprises administering the bioactive factor to an individual to whom a preparation comprising isolated autologous human mesenchymal stem cells has been, is being or will be administered.
In another aspect, the invention provides a method for inducing the in vivo production of human cytokines in an individual in need thereof which comprises administering to the individual isolated culture-expanded human mesenchymal stem cells and a bioactive factor effective to induce such cells to differentiate into a cytokine-producing mesenchymal lineage descendant in such individual. Preferably, the mesenchymal stem cells and bioactive factor are administered together or they may alternatively be administered separately. In specific preferred examples of these aspects, the bioactive factor is a bone morphogenetic protein and the human MSCs are directed into the chondrogenic lineage; the bioactive factor is interleukin 1 and the human MSCs are directed into the εtromal cell lineage (preferably the interleukin 1 is interleukin la ) ; the bioactive factors are dexamethasone, ascorbic acid-2-phoεphate and β- glycerophosphate and the human MSCs are directed into the osteogenic lineage; or the bioactive factor is selected from the group consisting of 5-azacytidine, 5-azadeoxycytidine and analogs of either of them and the human mesenchymal stem cells are directed into the myogenic lineage.
Another aspect of the invention provides a composition comprising isolated, culture-expanded human mesenchymal stem cells and a bioactive factor,- or combination, effective to induce differentiation of such cells into a lineage of choice. Preferably the composition further comprises a tissue culture medium. Alternatively, the composition can comprise a medium suitable for administration to an animal particularly a human, in need thereof. This aspect of the invention also provides for specific embodiments using the bioactive factors identified above for lineage induction into the lineages associated therewith as described above.
Figure 1 diagrammatically illustrates the mesengenic process by which mesenchymal stem cells differentiate into various lineage pathways.
Figure 2 diagrammatically illustrates the osteogenic differentiation pathway.
Figure 3 graphically demonstrates the increase in alkaline phosphatase activity as a function of time in cultures, in the initial studies reported in Example 1. Figure 4 shows results from the subsequent studies reported in Example 1.
Figure 5 diagrammatically illustrates the chondrogenic differentiation pathway.
Figure 6 shows the extent of human mesenchymal stem cell cytokine expression, with and without interleukin-1 stimulation, based on the experiments in Example 4.
Figures 7A and 7B.
(A) Phase contrast micrograph of living culture of MSCs showing the multinucleated cells derived after exposure to 5- aza-CR. This micrograph shows a culture 2 weeks after treatment with 10 μM 5-aza-CR. Many nuclei (arrows) in the cell can be observed, but striations are not discernible.
(B) Phase contrast micrograph of living culture of normal rat fetal muscle cells prepared from the hindlimbs of 17-day-old rat fetuses. As with bone marrow MSC-derived myotubes, no discernible striations are apparent. Scale bar 50 μm.
Figure 8: Immunofluorescence staining for muscle- specific yosin in myotubes derived from rat bone marrow MSCs after exposure to 5-aza-CR. Myosin antibodies do not visualize cross striations, but the antibodies clearly illuminate longitudinal fibers. Scale bar 30 μm.
Figures 9A-9D: Myotubes derived from rat bone marrow MSCs 2 weeks [(A) and (B) ] and 5 weeks [(C) and (D) ] after exposure to 5-aza-CR. Phase contrast micrograph [(A) and (C)] and immunofluorescence staining for myosin [(B) and (D)]. (A) and (B), (C) and (D) are the same visual fields. Myotubes 2 weeks after 5-aza-CR exposure are stained with anti-myosin antibody, but those 5 weeks after exposure are not. Scale bar 50 μM.
Figures 10A-10B: Micrograph of the 5-aza-CR-treated MSCs containing droplets in their cytoplasm; this culture was stained with Sudan Black. (A) Clusters of adipocytes (arrows) were observed; scale bar 200 μM. (B) Droplets are stained brown to black (arrows), which suggests that these droplets are lipid; scale bar 100 μM.
Figure 11: Phase contrast micrograph of living culture of myogenic cells derived from rat bone marrow MSCs after exposure to 5-aza-CR. Following exposure to 5-aza-CR, these cells were cultured with 4ng/ml bFGF for 10 days. Large myotubes can be seen; scale bar 300 μm.
Figures 12A-12D graphically illustrate the expression of G-CSF, GM-CSF, M-CSF and SCF, respectively, observed in the experiments reported by Example 6.
Figures 13A-13C graphically illustrate the expression of LIF, IL-6 and IL-11, respectively observed in the experiments reported by Example 6.
Figure 14 graphically illustrates the dose dependent IL- lα induction of GM-CSF expression observed in the experiments reported by Example 6.
This invention has multiple uses and advantages. The first lies in the ability to direct and accelerate MSC differentiation prior to implantation back into autologous hosts. For example, MSCs which are directed in vitro to become osteogenic cells will synthesize bone matrix at an implant site more rapidly and uniformly than MSCs which must first be recruited into the lineage and then progress through the key differentiation steps. Such an ex vivo treatment also provides for uniform and controlled application of bioactive factors to purified MSCs, leading to uniform lineage commitment and differentiation. In vivo availability of endogenous bioactive factors cannot be as readily assured or controlled. A pretreatment step such as is disclosed herein circumvents this. In addition, by pretreating the MSCs prior to implantation, potentially harmful side effects associated with systemic or local administration of exogenous bioactive factors are avoided. Another use of this technique lies in the ability to direct tissue regeneration based on the stage of differentiation which the cells are in at the time of implantation. That is, with respect to bone and cartilage, the state of the cells at implantation may control the ultimate tissue type formed. Hypertrophic chondrocyteε will mineralize their matrix and eventually pave the way for vascular invasion, which finally results in new bone formation. Clearly, MSCs implanted for the purpose of restoring normal hyaline cartilage must not progress down the entire lineage. However, implants which are designed to repair articular surface defects and the underlying subchondral bone could benefit from a two-component system wherein the cells in the area of the future bone are directed ex vivo to become hypertrophic chondrocytes, while the cells in the area of the future articulating surface are directed only to become chondroblasts. In the area of stromal reconstitution, the ex vivo control of differentiation can optimize MSC cell populations for the elaboration of stage-specific cytokines requisite to the needs of the individual. Muscle morphogenesis can similarly be directed to create fast or slow twitch fibers, depending on the indication. Isolation and Purification of Human Mesenchymal Stem Cells
The human mesenchymal stem cells isolated and purified as described here can be derived, for example, from bone marrow, blood, dermis or periosteum. When obtained from bone marrow this can be marrow from a number of different sources, including plugs of femoral head cancellous bone pieces, obtained from patients with degenerative joint disease during hip or knee replacement surgery, or from aspirated marrow obtained from normal donors and oncology patients who have marrow harvested for future bone marrow transplantation. The harvested marrow is then prepared for cell culture. The isolation process involves the use of a specially prepared medium that contains agents which allow for not only mesenchymal stem cell growth without differentiation, but also for the direct adherence of only the mesenchymal stem cells to the plastic or glass surface of the culture vessel. By creating a medium which allows for the selective attachment of the desired mesenchymal stem cells which were present in the mesenchymal tissue samples in very minute amounts, it then became possible to separate the mesenchymal stem cells from the other cells (i.e. red and white blood cells, other differentiated mesenchymal cells, etc.) present in the mesenchymal tissue of origin.
Bone marrow is the soft tissue occupying the medullary cavities of long bones, some haversian canals, and spaces between trabeculae of cancellous or spongy bone. Bone marrow is of two types: red, which is found in all bones in early life and in restricted locations in adulthood (i.e. in the spongy bone) and is concerned with the production of blood cells (i.e. hematopoiesis) and hemoglobin (thus, the red color); and yellow, which consists largely of fat cells (thus, the yellow color) and connective tissue. As a whole, bone marrow is a complex tissue comprised of hematopoietic cells, including the hematopoietic stem cells, and red and white blood cells and their precursors; and a group of cells including mesenchymal stem cells, fibroblasts, reticulocytes, adipocyteε, and endothelial cells which contribute to the connective tisεue network called "εtroma". Cellε from the εtroma regulate the differentiation of hematopoietic cellε through direct interaction via cell εurface proteinε and the secretion of growth factorε and are involved in the foundation and εupport of the bone εtructure. Studies using animal models have suggested that bone marrow contains "pre-stromal" cells which have the capacity to differentiate into cartilage, bone, and other connective tissue cells. (Beresford, J.N. : Osteogenic Stem Cells and the Stromal System of Bone and Marrow, Clin . Orthop. , 240:270, 1989). Recent evidence indicates that these cells, called pluripotent stromal stem cells or mesenchymal stem cells, have the ability to generate into several different types of cell lines (i.e. osteocytes, chondrocytes, adipocyteε, etc.) upon activation, depending upon the influence of a number of bioactive factors. However, the mesenchymal stem cells are present in the tissue in very minute amounts with a wide variety of other cells (i.e. erythrocyteε, plateletε, neutrophilε, lymphocytes, monocytes, eosinophilε, basophils, adipocytes, etc.).
As a result, a process has been developed for isolating and purifying human mesenchymal stem cells from tissue prior to differentiation and then culture expanding the mesenchymal stem cellε to produce a valuable tool for muεculoεkeletal therapy. The objective of such manipulation is to greatly increase the number of mesenchymal stem cells and to utilize these cellε to redirect and/or reinforce the body's normal reparative capacity. The mesenchymal stem cells are expanded to great numbers and applied to areas of connective tissue damage to enhance or stimulate in vivo growth for regeneration and/or repair, to improve implant adhesion to various prosthetic devices through subsequent activation and differentiation, or enhance hemopoietic cell production, etc.
Along these lines, various procedureε are contemplated for tranεferring, immobilizing, and activating the culture- expanded, purified mesenchymal stem cells at the εite for repair, implantation, etc., including injecting the cellε at the site of a skeletal defect, incubating the cells with a prosthesis and implanting the prosthesis, etc. Thus, by isolating, purifying and greatly expanding the number of cells prior to differentiation and then actively controlling the differentiation process by virtue of their positioning at the site of tissue damage or by pretreating in vitro prior to their transplantation, the culture-expanded, mesenchymal stem cells can be utilized for various therapeutic purposes such as to alleviate cellular, molecular, and genetic disorders in a wide number of metabolic bone diseaεes, skeletal dysplaεiaε, cartilage defectε, ligament and tendon injuries and other muεculoεkeletal and connective tiεεue diεorders.
Several media have been prepared which are particularly well suited to the desired selective attachment and are referred to herein as "Complete Media" when supplemented with serum aε deεcribed below. One such medium is an augmented version of Dulbecco's Modified Eagle's Medium-Low Glucose (DMEM-LG), which is well known and readily commercially available.
The commercial formulation is supplemented with 3700 mg/1 of sodium bicarbonate and 10 ml/1 of lOOx antibiotic- antimycotic containing 10,000 unitε of penicillin (base), 10,000 μq of streptomycin (baεe) and 25 μg of amphotericin B/ml utilizing penicillin G (εodium salt), εtreptomycin sulfate, and amphotericin B as FUNGIZONE® in 0.85% saline.
The medium described above is made up and stored in 90 ml per 100 ml or 450 ml per 500 ml bottles at 4°C until ready to use. For use, 10 ml or 50 ml of fetal bovine serum (from selected lots) iε added to the bottleε of media to give a final volume of 10% serum. The medium is warmed to 37°C prior to use.
In this regard, it was also found that BGJb medium (Gibco, Grand Island, NY) with tested and selected lots of 10% fetal bovine serum (J.R. Scientific, Woodland, CA, or other suppliers) was well suited for use in the invention. This medium, which was also a "Complete Medium", contained factorε which alεo εtimulated meεenchymal εtem cell growth without differentiation and allowed for the selective attachment through specific protein binding sites, etc. of only the mesenchymal stem cells to the plastic surfaces of Petri dishes.
In addition, it was also found that the medium F-12
Nutrient Mixture (Ham) (Gibco, Grand Iεland, NY) exhibited the desired properties for selective mesenchymal stem cell separation.
As indicated above, the complete medium can be utilized in a number of different isolation processes depending upon the specific type of initial harvesting processes used in order to prepare the harvested bone marrow for cell culture separation. In thiε regard, when plugε of cancellouε bone marrow were utilized, the marrow was added to the complete medium and vortexed to form a dispersion which was then centrifuged to separate the marrow cells from bone pieces, etc. The marrow cellε (consiεting predominantly of red and white blood cells, and a very minute amount of meεenchymal stem cellε, etc.) were then dissociated into single cells by sequentially passing the complete medium containing the marrow cells through syringes fitted with a series of 16, 18, and 20 gauge needles. It is believed that the advantage produced through the utilization of the mechanical separation proceεε, aε oppoεed to any enzymatic εeparation process, was that the mechanical process produced little cellular change while an enzymatic process could produce cellular damage particularly to the protein binding sites needed for culture adherence and selective separation, and/or to the protein siteε needed for the production of monoclonal antibodieε specific for said mesenchymal εtem cellε. The single cell suεpenεion (which waε made up of approximately 50-100 x 106 nucleated cells) waε then subsequently plated in 100 mm disheε for the purpoεe of εelectively εeparating and/or iεolating the meεenchymal stem cells from the remaining cells found in the suεpenεion.
When aεpirated marrow was utilized as the source of the human mesenchymal stem cells, the marrow stem cells (which contained little or no bone chipε but a great deal of blood) were added to the complete medium and fractionated with Percoll (Sigma, St. Louiε, MO) gradientε more particularly deεcribed below in Example 1. The Percoll gradients separated a large percentage of the red blood cells and the mononucleate hematopoietic cells from the low density platelet fraction which contained the marrow-derived mesenchymal stem cells. In this regard, the platelet fraction, which contained approximately 30-50 x 10° cells waε made up of an undetermined amount of platelets, 30-50 x 10° nucleated cellε, and only about 50-500 mesenchymal stem cells depending upon the age of the marrow donor. The low density platelet fraction was then plated in the Petri dish for selective separation based upon cell adherence. In this regard, the marrow cells obtained from either the cancellous bone or iliac aspirate (i.e. the primary cultures) were grown in complete medium and allowed to adhere to the surface of the Petri dishes for one to seven days according to the conditions set forth in Example 1 below. Since minimal cell attachment was observed after the third day, three days was chosen as the standard length of time at which the non-adherent cells were removed from the cultures by replacing the original complete medium with fresh complete medium. Subsequent medium changes were performed every four days until the culture dishes became confluent which normally required 14-21 days. This represented a 103-104 fold increase in the number of undifferentiated human mesenchymal stem cells.
The cells were then detached from the culture dishes utilizing a releasing agent such as trypsin with EDTA (ethylene diaminetetra-acetic acid) (0.25% trypsin, lmM EDTA (IX) , Gibco, Grand Island, NY) . The releasing agent was then inactivated and the detached cultured undifferentiated mesenchymal stem cells were washed with complete medium for subsequent use.
The capacity of these undifferentiated cells to enter discrete lineage pathways is referred to as the mesengenic process, and is diagrammatically represented in Figure l. In the mesengenic process, MSCs are recruited to enter specific multi-step lineage pathways which eventually produce functionally differentiated tissues such as bone, cartilage, tendon, muscle, dermis, bone marrow stroma, and other mesenchymal connective tissues. For example, a detailed scheme for the differentiation pathway of bone forming cells is presented in Figure 2. This lineage map implies the existence of individual controlling elements which recruit the MSCs into the osteogenic lineage, promote pre-osteoblast replication, and direct step-wise differentiation all the way to the terminal stage osteocyte. Subεtantial work haε been reported that supports the view that each step of this complex pathway is controlled by different bioactive factors.
A similar lineage diagram haε been developed for chondrocyte differentiation and iε provided in Figure 5. Again, progreεεion of each lineage step is under the control of unique bioactive factors including, but not limited to, the family of bone morphogenetic proteins. Each modulator of the differentiation process, whether in bone, cartilage, muscle, or any other mesenchymal tissue, may affect the rate of lineage progresεion and/or may specifically affect individual stepε along the pathway. That iε, whether a cell iε naεcently committed to a εpecific lineage, iε in a bioεynthetically active state, or progresses to an end stage phenotype will depend on the variety and timing of bioactive factors in the local environment.
The bone and cartilage lineage potentials (i.e. osteo- chondrogenic potential) of fresh and expanded human mesenchymal stem cellε were determined uεing two different in vivo assays in nude mice. One assay involved the subcutaneous implantation of porous calcium phosphate ceramics loaded with cultured mesenchymal stem cellε; the other involved peritoneal implantation of diffusion chambers inoculated with cultured mesenchymal stem cells. Whole marrow and Percoll gradient separated aspirate fractions were also analyzed in these in vivo asεayε. Hiεtological evaluation εhowed bone and cartilage formation in the ceramics implanted with the cultured mesenchymal stem cells derived from the femoral head and the iliac crest. Ceramics loaded with human mesenchymal stem cells at 5xl06 cells/ml formed bone within the pores, while ceramicε loaded with human meεenchymal εtem cells at 10x10° cellε/ l formed cartilage within the pores. While whole marrow has now been shown to form bone when placed as a composite graft with ceramics in a subcutaneous site in nude mice, the amount of bone produced iε εubεtantially leεε than that εeen when culture expanded marrow-derived mesenchymal stem cells are used.
These results indicated that under certain conditions, culture expanded mesenchymal stem cells have the ability to differentiate into bone or cartilage when incubated as a graft in porous calcium phosphate ceramics. The environmental factors which influence the mesenchymal stem cells to differentiate into bone or cartilage cells appearε, in part, to be the direct acceεsibility of the mesenchymal stem cells to growth and nutrient factors supplied by the vasculature in porous calcium phosphate ceramics; cells that are closely asεociated with vaεculature differentiate into bone cells while cells that are isolated from vasculature differentiate into cartilage cellε. The excluεion of vasculature from the pores of ceramics loaded with concentrated human meεenchymal εtem cells prevented osteogenic differentiation and provided conditions permisεive for chondrogeneεiε.
Aε a result, the iεolated and culture expanded meεenchymal εtem cells can be utilized under certain εpecific conditionε and/or under the influence of certain factorε, to differentiate and produce the desired cell phenotype needed for connective tiεεue repair or regeneration and/or for the implantation of variouε proεthetic deviceε. For example, uεing porouε ceramic cubeε filled with culture-expanded human meεenchymal stem cells, bone formation inside the poreε of the ceramicε has been generated after εubcutaneous incubations in immunocompatible hostε. In a recent εtudy (13), rat marrow in a composite graft with porous ceramic was used to fill a segmental defect in the femur of the rat. Bone waε shown to fill the pores of the ceramic and anchor the ceramic-marrow graft to the host bone.
Factorε which εtimulate oεteogenesis (i.e. are osteoinductive) from isolated human mesenchymal stem cells in accordance with the invention are present in several classes of molecules, including the following: bone morphogenic proteins, such as BMP-2 (14) and BMP-3 (15); growth factors, such as basic fibroblast growth factor (bFGF); glucocorticoidε, εuch aε dexamethasone (16); and proεtaglandinε, such as prostaglandin El (22). Further aεcorbic acid and itε analogε, εuch aε aεcorbic acid-2- phoεphate (17) and glycerol phosphates, such as β- glycerophoεphate (18) are effective adjunct factorε for advanced differentiation, although alone they do not induce oεteogenic differentiation.
Factorε which have chondroinductive activity on human MSCs are also present in εeveral claεεeε of molecules, including the following: compounds within the tranεforming growth factor-β (TGF-3) superfa ily, such aε (i) TGF-/S1 (19), (ii) Inhibin A (20), (iii) chondrogenic εtimulatory activity factor (CSA) (21) and (iv) bone morphogenic proteinε, εuch aε BMP-4 (22); collagenous extracellular matrix molecules, including type I collagen, particularly aε a gel (23); and vitamin A analogs, such as retinoic acid (24).
Factors which have stromagenic inductive activity on human MSCε are alεo preεent in εeveral classeε of moleculeε, especially the interleukinε, such as IL-lα (25) and IL-2 (26).
Factors which have myogenic inductive activity on human MSCs are alεo present in εeveral claεseε of moleculeε, especially cytidine analogs, such as 5-azacytidine and 5-aza- 2'-deoxycytidine.
The effect of these modulating factors on human MSCε iε diεclosed here for the first time. This is not represented to be an all-inclusive listing of potentially useful modulatory factors for inducing differentiation into a particular lineage, but illuεtrateε the variety of compoundε which have useful biologic activity for the purpose of promoting the step-wise progression of isolated human mesechymal εtem cell differentiation.
Example 1 Induced Osteogenic Differentiation of MSCs In Vitro
The objective of the experiments described in this example was to demonstrate that meεenchymal εtem cellε (MSCs) were directed along the oεteogenic lineage pathway in vitro by providing appropriate bioactive factorε in the tiεεue culture medium. This set of experiments illustrates just one example of how MSCs can be directed along the osteogenic lineage.
Initial Study
Human MSCs were harvested and isolated from bone marrow as described above. These cells were culture-expanded in DMEM-LG medium containing preselected 10% fetal bovine serum (Complete Medium). Fresh Complete Medium was replaced every 3-4 days until the cultures were near confluence, at which time the cells were liberated off the plates with trypsin, and reseeded onto new diεhes at approximately 40% confluence (400,000 cells per 100 mm dish). These replated MSCs were allowed to attach overnight, after which the Complete Medium was replaced by a medium composed of DMEM-LG, 10% fetal bovine serum, and either 100 nM dexamethaεone alone, or 100 nM dexcunethasone with 50 μM ascorbic acid-2-phoεphate, and 10 mM 8-glycerophoεphate (Oεteogenic Supplement). The Oεteogenic Supplement was replaced every 3 days. Cells were examined daily for morphologic changes. Selected plates were then analyzed for cell εurface alkaline phosphataεe (AP) activity, a marker for cellε which have entered the oεteogenic lineage. It iε theεe cellε which were εubεequently reεponεible for εyntheεizing oεteoid matrix. Standard enzyme hiεtochemiεtry and biochemiεtry reagentε were uεed to demonεtrate activity of thiε cell surface protein. Additional εpecimenε were evaluated for the preεence of mineralized extracellular matrix nodules which correlate with the continued differentiation and phenotypic expreεεion of a mature osteoblaεt population. Silver nitrate precipitation onto calcium phoεphate cryεtalε within the bone nodule was achieved through the standard Von Koεεa staining technique.
The results indicate that after only three days of exposure to dexamethasone, MSCs in culture had already begun expressing alkaline phosphatase on their surface. By day six of culture, approximately 80% of the cellε were AP positive. The grosε organization of the culture diεh had changed from near confluent whorlε of fibroblaεt-like cellε at day 1, to numerous areas of polygonal cells which were piled on top of each other. By day 9, many εmall nodules of birefringent extracellular matrix was asεociated with theεe foci of layered polygonal cells. Theεe areaε were poεitively εtained by the Von Kossa method for mineral. Control cultureε fed only Complete Medium never developed theεe mineralized bone noduleε, and only rarely contained AP poεitive cellε. By contrast, MSCs treated with Osteogenic Supplement uniformly acquired AP activity and synthesized mineralized extracellular matrix nodules throughout the diεh. Although not oεteoinductive themεelveε, the preεence of ascorbic acid- 2-phosphate and 0-glycerophoεphate in the complete Oεteogenic Supplement further supports extracellular matrix maturation and mineral deposition, respectively. Figure 3 graphically demonstrates the increase in alkaline phosphatase enzyme activity as a function of time in culture. By day 8 and beyond, subεtantially more enzyme activity is obεerved in cellε exposed to Osteogenic Supplementε (OS) than thoεe cultured with control medium.
Taken together, these studies demonstrate that MSCs can be rapidly and uniformly stimulated to differentiate along the osteogenic lineage in vitro . Furthermore, not only are the MSCs recruited into the early steps within the lineage, evidenced by AP expresεion, but the MSCε progreεs through the lineage to become mature osteoblastε which secrete and mineralize a bone-like extracellular matrix. Further evidence for this comes from the observation that when chick MSCs are treated with Oεteogenic Supplement, they progreεε through the εtageε of the oεteogenic lineage depicted in Figure 2 aε determined by monoclonal antibody staining againεt εtage-εpecific cell εurface antigenε.
Subsequent Study
Using published techniques, MSCs were purified from 3 different patients (ages 26-47), culture expanded (27), and seeded overnight onto 48-well culture plates at 20% confluence in DMEM-LG with 10% FBS from selected lots. Base media for comparison were DMEM-LG, BGJb, αMEM, and DMEM/F-12 (1:1). Triplicate cultures for each asεay were grown in 10% FBS in the abεence or preεence of "Osteogenic Supplements" (OS) (100 nM dexamethasone, 50 μM aεcorbic acid-2-phoεphate, and IOIΓLM /8-glycerophosphate (28). Media were changed every 3 days. Each set of cultures was asεayed for cell number by the cryεtal violet aεsay, cell surface alkaline phosphatase (AP) by histochemistry and mineralized nodule formation by Von Kossa staining. AP enzyme activity was calculated by incubating live cultures with 5 mM p-nitrophenylphosphate in 50 mM Tris, 150 mM NaCl, pH 9.0 and quantifying the colorimetric reaction by scanning the samples at 405 nm on an ELISA plate reader. AP enzyme activity was expresεed aε nanomoles of product/minute/103 cells. The percentage of AP- poεitive cells in each well was determined from the stained cultures, and the number of mineralized nodules per well were counted. Asεayε were performed every 4 dayε for the 16 day culture period. The paired two-sample t-Teεt waε performed on εelected εampleε. The data in Figure 4 represent one patient, although similar resultε were obtained from all specimens.
MSCs uniformly attached to the plates, assumed their characteristic spindle-εhaped morphology, and proliferated to reach confluence within 8 days. During this period, and particularly beyond, the OS-treated cells developed a cuboidal morphology as their density increaεed, forming multiple layerε. For clarity, only εelected aεpectε of the parameterε deεcribed above are graphically repreεented on Figure 4. All specimens grown in BGJb+OS died within 3 days, while BGJb cultures survived for the duration of the protocol. For this reason, all BGJb data were omitted from the graphs . Highlights of the εtudy demonεtrate εubstantially greater proliferation in αMEM compared to DMEM/F-12 or DMEM alone (i.e., p<0.01 and p<0.05 at day 16). The addition of OS to αMEM cultures inhibited proliferation at days 8 and 12 (p<0.04 and p<0.03), but not by day 16 (p>0.05). αMEM+OS also stimulates a significant proportion of cells to expresε AP on their εurface when compared to MSCε maintained in DMEM (p<0.02 at day 8. p<0.01 at day 16). However, no εignificant difference in the percent of AP cellε iε observed between αMEM with and without OS (p>0.2 at day 8, p>0.05 at day 16). Notably, αMEM+OS induces more AP activity than any other medium throughout the culture period. including αMEM or DMEM (i.e., p<0.004 and p<0.002 at day 16). However, there waε no difference in AP activity between αMEM and DMEM+OS throughout the study period (i.e., p>0.2 at day 16). Of all media teεted, the number of mineralized noduleε by day 16 iε greateεt in DMEM+OS (p<0.02 compared to DMEM).
Theεe investigations demonstrate that purified, culture- expanded human MSCs can be induced into the osteogenic lineage in vitro , thereby eεtabliεhing a model for human oεteoblaεt differentiation. Early in the culture period (day 8) only αMEM+OS induced substantial osteoblastic recruitment of MSCε (>50%), aε noted by AP cell surface εtaining. By day 16, however, all cultureε except DMEM contained >60% AP εtained cellε. In all media εtudied, addition of OS yieldε greater AP activity beyond 4 dayε. Although a large percentage of cellε in moεt media were AP-stained at day 16, the εubstantial differences in the AP activity asεay likely reflect the quantity of enzyme on the cell surface, and therefore, the degree of progresεion into the oεteoblaεtic lineage. At the very leaεt, OS are capable of up-regulating expreεεion of thiε oεteoblaεtic cell surface marker. Interestingly, despite less AP activity, DMEM+OS cells generated more mineralized noduleε than αMEM+OS. This observation may suggeεt that within the 16 day culture period, DMEM+OS εupports further osteogenic differentiation of MSCs than αMEM+OS. It iε possible that, given more time, αMEM+OS would foster even more mineralized foci than DMEM+OS. Differenceε in the media favoring maintenance of the MSC phenotype (DMEM) evidenced by MSC-specific im unostaining, or maximal recruitment and induction into the osteogenic lineage (αMEM+OS), noted by the percent AP-positive cellε and AP activity, are inherently intereεting and warrant further examination. The use of various monoclonal and polyclonal antibodies against specific cell and matrix components during this inductive phenomenon are currently underway, and will provide further inεight into the molecular nature of the in vitro differentiation process.
Figure imgf000024_0001
The Generation of Monoclonal Antibodies Against Human
Osteogenic Cells Reveals gtnt.*γr>nic Bone Formation
In Vivo And Differentiation of Purified Mesenchymal
Stem Cells In Vitro
It has been well-establiεhed that meεenchymal progenitor cellε derived from bone marrow are capable of differentiating into oεteoblaεtε. In addition, theεe meεenchymal εtem cellε (MSCs) also give rise to cartilage, tendon, ligament, muscle, and other tissueε. However, knowledge of the εtepε involved in the commitment and differentiation of MSCε along theεe various lineages has been restricted, in part, by the lack of probes specific for cells at various stageε within the oεteogenic or other differentiation pathwayε. Since monoclonal antibodieε are uεeful probeε for εtudying differentiation, we immunized mice with intact living cell preparationε of human bone marrow-derived MSCε induced into the oεteogenic lineage in vitro . We εcreened hybridoma colonieε against purified MSCε, MSCs undergoing osteogenic differentiation, and frozen εectionε of embryonic human limbs where long bones are developing around the cartilage rudiment. This screening protocol favorε εelection of antibodies which react with MSCε undergoing differentiation in vitro and human oεteogenic cellε in vivo . Uεing thiε approach, we have generated monoclonal antibodieε againεt lineage εtage-specific surface antigens on osteogenic cellε derived from human marrow MSCε.
Uεing published techniques, MSCs were purified from 5 different patients (ages 28-46), culture expanded (29), and grown in DMEM-LG with 10% FBS and "Osteogenic Supplementε" (100 nM dexamethaεone, 50 μM aεcorbic acid-2-phoεphate, and lOmM j8-glycerophosphate (28). At days 3 and 6 of culture, early during alkaline phosphataεe expresεion, and prior to mineralized nodule formation (30), the cells were liberated from the plates with 5 mM EGTA. Approximately 4 million 3 and 6 day cells were pooled for each of five weekly immunizations into Balbc/J mice. Uεing εtandard techniqueε, monoclonal hybridomas were produced, and culture εupernatantε were εcreened by a semiquantitative ELISA againεt purified MSCε, and MSCs cultured for 3 or 6 days with Osteogenic Supplements. Briefly, MSCs were plated on 96-well culture disheε, expoεed to Osteogenic Supplementε, and then reacted with culture εupernatantε followed by goat anti-mouεe IgG conjugated to horεeradish peroxidase. The εecondary antibody waε rinεed, and o-phenylenediamine εubεtrate waε added to the plateε. Primary mouεe monoclonal antibody binding was aεεeεεed by the colorimetric reaction quantified by εcanning the wellε at 490 nm on an ELISA plate reader. Colonieε of intereεt were εelected on the baεiε of differential binding to control MSCε and oεteogenic cellε derived from MSCε. Selected colonieε were further εcreened by immunofluorescence on unfixed frozen sections of human embryonic limbs. Hybridoma colonieε of interest were cloned and further immunocytochemical analyεeε were performed on a variety of normal and experimentally-derived tiεεues from human, rat, rabbit, chick, and bovine sources.
Nearly 10,000 hybridoma colonies were screened by the modified ELISA protocol deεcribed above. Baεed on differential binding to purified MSCε, or MSCε cultured for 3 and 6 dayε with Oεteogenic Supplementε, 224 colonieε were εelected for immunofluoreεcent εcreening against embryonic day 55-60 human limbε. The majority of thoεe 224 colonies either reacted with multiple tissue types present in the developing limb, or were not detected in the developing bone. Thuε far, 9 colonieε have been identified which demonεtrate specific immunoreactivity on cellε of the oεteogenic lineage. The patternε of reactivity vary; εome hybridoma εupernatantε react with a large population of cellε within the oεteogenic collar and osteoprogenitor-containing perioεteum, while otherε react with only those cellε which appear to be actively involved in matrix synthesis. Two hybridoma colonies appear to react with osteogenic cells aε well aε hypertrophic chondrocyteε. The reεultε are εummarized in Table 1.
Table 1
Hybridoma Cell Control MSCε 3 day OS 6 day OS Line culture culture
20E8 0 1 8
13C9 0 1 3
5D9 0 1 2
18H4 0 3 5
18D4 0 2 4
10F1 0 0 2
13B12 0 4 2
Table 1 εhowε the immunoreactivity of selected hybridoma colonieε againεt untreated MSCs, or MSCs cultured with Osteogenic Supplements (OS) for 3 or 6 days. Numbers reflect the relative amount of antibody bound in the ELISA assay described above.
These investigations indicate the preεence of human osteogenic lineage stage-εpecific cell surface differentiation markers similar to those detailed for avian osteogenic cells (31). The staining of osteogenic cells in the developing limb supports the view that MSCs cultured with Osteogenic Supplements become "authentic" osteoblaεts in culture. Oεteogenic differentiation in vitro is thuε confirmed by molecular probeε which extend beyond traditional criteria of AP expression and mineralized nodule formation. Correlation of detailed in vitro observations with in vivo analyses of antigen expression will be useful in further studieε of osteogenesiε. Characterization of the εpecific tiεsue culture elements, i.e., bioactive factors, which promote progreεsion of cells through the osteogenic lineage εtepε will be crucial. Identification of osteogenic cell surface, and/or extracellular matrix antigens should provide further insight into bone cell physiology. These and other monoclonal antibodies currently under investigation will prove uεeful in future εtudies of MSC differentiation.
Example 3 Induced Chondrogenic Differentiation of MSCs In Vitro
The objective of the experimentation described in thiε example was to demonstrate that mesenchymal stem cells (MSCs) were directed along the chondrogenic lineage pathway in vitro by providing appropriate bioactive factors in the tissue culture medium. This set of experimentε repreεentε just one example of how MSCε can be directed along the chondrogenic lineage. Human MSCs were harvested and isolated from bone marrow as deεcribed above. Cellε were culture-expanded in DMEM-LG medium containing preεelected 10% fetal bovine serum (Complete Medium). Fresh medium was replaced every 3-4 days until the cultures were near confluence, at which time the cells were liberated off the plates with trypsin, and reεeeded onto new diεhes at approximately 50% confluence (500,000 cells per 100 mm dish). These replated MSCs were allowed to attach overnight, after which the Complete Medium was replaced by DMEM-LG with 10% fetal bovine serum, and 5 mg/ml partially purified Bone Morphogenic Protein (Chondrogenic Supplement), supplied by Dr. Marshall R. Urist. Thiε Chondrogenic Supplement waε replaced every 3 dayε. Cellε were examined daily for morphologic changeε. Selected plates were then analyzed immunohistochemically for CSPG-M, a marker for cells which have entered the chondrogenic lineage. It is these cells which were then actuated for synthesizing the Type II collagen matrix of cartilage. Standard immunohistochemiεtry reagents were used to demonstrate the presence of this extracellular matrix protein. Additional specimens were evaluated for the presence of Toluidine Blue-stained nodules which correlated them with the continued differentiation and phenotypic expresεion of a mature chondrocyte population. Von Koεεa εtaining for the preεence of mineralized nodules of hypertrophic chondrocytes was negative.
The reεults indicated that after only three days of exposure to the Chondrogenic Supplement, MSCε in culture had already begun expressing CSPG-M into their extracellular matrix. The groεε organization of the culture diεh had changed from whorlε of fibroblast-like cells at day 1, to numerous foci of multi-layered round or polygonal cells surrounded by a thin layer of fibroblastic cells resembling a perichondrium. The extracellular matrix of these nodules was εtrongly immunoreactive for Type II collagen. Control cultureε fed only Complete Medium never developed theεe cartilage nodules. Taken together, theεe εtudies demonεtrate that MSCε have been εtimulated to differentiate along the chondrogenic lineage in vitro . Furthermore, not only were the MSCε recruited into the early εtepε within the lineage, evidenced by CSPG-M expreεεion, but the MSCs progresεed along the lineage to become mature chondrocytes which secreted Type II collagen-rich extracellular matrix. Thus far, terminal differentiation of chondrocytes derived from MSCs, evidenced by hypertrophic cellε in a calcified matrix, haε not been obεerved in vitro . Thiε finding reflectε the need for deεigning a Chondrogenic Supplement εpecifically aimed at promoting this terminal differentiation step. Interestingly, Pacifici and his collaborators (32) have devised a medium containing retinoic acid which stimulates terminal differentiation of chick chondrocytes in vitro .
The additive to Complete Medium which constitutes Chondrogenic Supplement in the example above is only one of the factors known to stimulate chondrogenic cell differentiation or proliferation in vitro .
Example 4 Induced Marrow Stromal Cell Differentiation of MSCs in vitro
The purpose of the experimentation described in thiε example waε to demonεtrate that human marrow-derived MSCε were directed along the εtromagenic lineage pathway in vitro by providing appropriate bioactive factorε in the culture medium. Human marrow-derived MSCε were isolated from bone marrow and expanded in culture as described above. In order to demonεtrate the ability of human MSCs to be induced along the marrow stromal cell lineage, specific cytokine expression was measured as a marker of differentiation. MSCs were grown under conditions which favor MSC proliferation without differentiation using medium consiεting of DMEM-LG containing preselected 10% fetal bovine serum (Complete Medium), or conditions which favor expression and differentiation into the marrow stromal phenotype using medium comprising Complete Medium plus 10 U/ml Interleukin-lα (IL-lα) (Stromagenic Supplement (SS)). Conditioned culture media from these tisεue culture populations were analyzed for the preεence of cytokineε uεing commercial εandwich ELISA bioaεεayε (R&D Systems) . The cytokineε that were assayed are those that are known to be secreted by stromal cells and which influence hematopoiesis. Theεe include interleukin-3 (IL-3), interleukin-6 (IL-6), granulocyte colony εtimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), stem cell factor (SCF), leukemia inhibitory factor (LIF) and transforming growth factor-beta-2 (TGF-32). In each case, second passage MSCs were plated onto 35 mm culture plates at a denεity of approximately 30% confluence (30,000 cellε per 35-mm plate). After allowing overnight attachment of the cellε, the culture media were removed, and replaced with either Complete Medium or Complete Medium plus Stromagenic Supplement. Figure 6 illuεtrateε the cytokine expreεεion of human MSCε under the two plating conditions. In the abεence of IL-lα, MSCε expreεεed G-CSF, GM-CSF, LIF and SCF at very low levels, but expreεε IL-6 in high abundance. In compariεon, after 3 dayε of IL-l-α εtimulation, dramatically higher levelε of cytokines were detected for all of the above specieε . MSCε did not expreεε IL-3 or TGF-jS2 under either of the two culture conditionε . Theεe data εhow that IL-I-α enhanceε MSC expression of a cytokine profile that haε been documented to support differentiation of the hematopoietic εtem cell and which is characteriεtic of differentiated marrow εtromal cells.
Example 5 Induced Myogenic Differentiation of MSCs In Vitro
The purpose of the study deεcribed in this example was to demonstrate that 5-azacytidine induces mesenchymal stem cells (MSCs) to differentiate along the myogenic lineage.
The compound, 5-azacytidine (5-aza-CR; Sigma Chemical Co., St. Louis, MO), an analogue of cytidine, causes hypomethylation of some cytosine in DNA which may be involved in activating phenotype-specific geneε. The mouse embryonic cell lines, C3H/10T1/2 C18 and Swiss 3T3, after exposure to 5-aza-CR, have been shown to be converted into 3 different mesodermal cell lineages, myoblast, adipocyte and chondrocyte (33-34). In part, it appears that the mechanism by which 5-aza-CR activates myogenic genes involveε MyoDl (35-36). With the above in mind, we have expoεed rat bone marrow-derived MSCε to 5-aza-CR and have focuεed our analyεiε on their conversion to myogenic phenotypes.
Femora and tibiae of male Fisher rats (Charleε River, Indianapoliε, IN) with an average body weight of 100 g were collected and the adherent εoft tiεεues were removed. Several isolateε of marrow cellε were from 250 g ratε. Meticulouε diεεection of the long boneε to remove εoft tiεεue waε done to insure that myogenic precursors were not carried into the marrow preparation. In this regard, myogenic cells were never obεerved in untreated MSC cultureε. Both endε of the boneε were cut away from the diaphyεeε with bone εciεεorε. The bone marrow plugε were hydrostatically expelled from the bones by insertion of 18-gauge needles fastened to 10 ml syringes filled with Complete Medium consisting of DMEM containing selected lots of 10% fetal calf serum (FCS; IR Scientific Inc., Woodland, CA), 5% horse serum (HS; Hazleton Biologies Inc., Lenexa, KS), and antibiotics (Gibco Laboratories; penicillin G, 100 U/ml; streptomycin, 100 μg/ml; amphotericin B, 0.25 μg/ml) . The needles were inserted into the distal ends of femora and proximal ends of tibias and the marrow plugε expelled from the oppoεite ends. The marrow plugs were disaggregated by sequential passage through 18-gauge, 20-gauge, and 22-gauge needles and these disperεed cellε were centrifuged and reεuεpended twice in Complete Medium. After the cellε were counted in a hemocytometer, 5xl07 cells in 7-10 ml of complete medium were introduced into 100 mm petri disheε. Three days later, the medium was changed and the non-adherent cells discarded. Medium was completely replaced every 3 days. Approximately 10 dayε after seeding, the disheε became nearly confluent and the adherent cellε were releaεed from the diεheε with 0.25% trypεin in ImM εodium EDTA (Gibco Laboratorieε, Grand Iεland, NY), split 1:3, and εeeded onto fresh plates. After these once paεεaged cellε became nearly confluent, they were harvested and used for the experiments described below. We refer to these cells aε rat marrow-derived MSCε. In total, 8 εeparate rat marrow-derived MSC preparationε were uεed in thiε study. The cellε were routinely cultured in Complete Medium at 37°C in a humidified atmoεphere of 5% C02.
The twice paεεaged MSCε were εeeded into 35-mm diεhes at three cell densitieε, 500, 5,000, and 50,000 cells/dish. Beginning 24 hr after εeeding, theεe cultureε were treated for 24 hr with Myogenic Medium conεisting of complete medium containing various concentrationε of 5-aza-CR. After the cultures were washed twice with Tyrode's balanced salt εolution (Sigma Chemical Co.), the medium was changed to complete medium without added 5-aza-CR and subsequently changed twice a week until the experiment was terminated, 40 days after the treatment. As described in detail in the resultε, various culture conditions were tested to attempt to optimize the 5-aza-CR effects, especially to optimize myogeneεis.
Twice paεεaged rat bone marrow MSCε were seeded into 35-mm diεheε at 5,000 cellε/dish and treated with four concentrations (0.1 μM, 0.3 μM, 1 μM and 10 μM) of 5-aza-2'- deoxycytidine (5-aza-dCR; Sigma Chemical Co.) in the same way as described above for 5-aza-CR. At various times during treatment, the morphology of the cultures was observed. The living cultures were examined every day with a phase-contrast microscope (Olympus Optical Co., Ltd., Tokyo, Japan), and eventually some of the cultureε were fixed for histology or immunohistochemistry. Muscle cells were first identified morphologically in phase contrast by the presence of multinucleated myotubes, and εubsequently immunohistochemically by the preεence of the εkeletal muscle-specific protein, myosin. Contraction of the putative muscle cells was stimulated by a drop of 1 mM acetylcholine (Sigma Chemical Co.) in Tyrode's. For immunohistochemistry, cultured cellε were fixed with -20°C methanol (Fiεher Scientific Co., Fair Lawn, NJ) for 10 min and incubated with a mouse monoclonal antibody to rat faεt twitch εkeletal myoεin (Sigma Chemical Co.; ascites fluid, 1/400 dilution) in PBS (phoεphate buffered εaline, pH7.4) containing 0.1% BSA (bovine serum albumin; Sigma Chemical Co.). The second antibody was biotin-conjugated εheep anti-mouεe IgG (Organon Teknika Corp., Weεt Cheεter, PA; 1/50 dilution) followed by treatment with Texas red-conjugated avidin (Organon Teknika Corp.; 1/4,000 dilution). All incubations were for 30 min at room temperature, each preceded by blocking for 5 min with PBS containing 1% BSA, followed by two 5-min washes in PBS. The cells were mounted in Fluoromount-G (Fisher Biotech, Pittεburgh, PA) and obεerved with an Olympuε microεcope (BH-2) equipped for fluorescence and photographed with Kodak TMAX 400 film.
Second passage rat bone marrow MSCs were plated into 96-well plates at limiting dilution of one cell/well; cellε were plated in medium conεisting of 50% Complete Medium and 50% conditioned medium, which waε obtained from rat bone marrow cellε near confluence cultured in Complete Medium for 2 dayε. From a total of 384 wellε, 50 colonieε were detected; these were subcultured, maintained, and eventually 4 survived. These 4 clones were treated with 5-aza-CR as mentioned above and scored for myogenic or adipocytic morphologies.
First passage rat bone marrow cells were exposed to 10 μM 5-aza-CR for 24 hr and plated into 96-well plates at limiting dilution of one cell/well aε above. The number of cloneε exhibiting adipocyte (Sudan Black positive) or myogenic, multinucleated cell morphologies was determined.
To compare the conversion capacity of bone marrow MSCs to various mesodermal phenotypes with that of pure fibroblasts, we exposed rat brain fibroblastε to either 5-aza-CR or 5-aza-CdR. Whole cerebra of brainε of three male Fisher rats were collected from the inside of the skulls and cut into small pieces with a sharp scalpel. Theεe pieceε were tranεferred to a 50-ml conical centrifuge tube, centrifuged at 500 xg for 10 min, reεuεpended in 10 ml of Tyrode'ε balanced εalt εolution, and homogenized with a loose-fitting Dounce homogenizer. The homogenate waε incubated with 0.1% collagenaεe (CLS2, 247 U/mg; Worthington Biochemical Co., Freehold, NJ) at 37°C for 3 hr, during which time it waε vortexed for 30 sec every 30 min. After treatment, the releaεed cells were paεεed through a 110-μm Nitex filter, centrifuged, reεuεpended in 10 ml of low glucoεe DMEM-LG (Gibco Laboratorieε) containing 10% FCS, and cultured in three 100-mm culture dishes at 37°C in a C02 incubator. The medium was changed twice a week and cells were cultured until the dishes reached confluence.
Third passage rat brain fibroblasts were seeded into 35-mm dishes at a density of 50,000 cellε/diεh and treated with 1 μM, 3 μM or 10 μM 5-aza-CR or 0.1 μM, 0.3 μM or 1 μM 5-aza-CdR in the same way as rat marrow MSCs. After 24 hr, the medium waε changed to DMEM-LG containing 10% FCS, 5% HS and 50 nM hydrocortisone without added 5-aza-CR or 5-aza-CdR and subεequently changed twice a week until the experiment was terminated.
Myogenic cells derived from rat bone marrow MSCs were compared with normal fetal rat myogenic cellε, since a subεtantial data baεe exiεtε for the latter. Muεcle cellε were diεεociated from the hindlimb muεcleε of 17-day-old Fisher rat fetuses with 0.2% trypsin (Sigma Chemical Co.) in calcium- and magnesium-free Tyrode's for 35 min at 37°C with occasional agitation. After they were passed through a 110- μm Nitex filter, the concentration of fibroblastε was reduced by incubating cell suspenεionε for 30 min in Falcon plastic disheε, which reεultε in preferential attachment of the fibroblastε. A suspenεion of 5x10s εingle cellε that did not attach to the uncoated dish waε plated in a collagen-coated (1.5 ml of 0.14% gelatin, J.T.Baker Chemical Co., Phillipεberg, NJ) 35-mm plaεtic culture diεh containing 2 ml of 79% DMEM, 10% FCS, 10% HS and 1% non-eεεential amino acids (Gibco Laboratories). Cells were grown at 37°C in a humidified atmosphere of 5% C02.
Cultureε of rat bone marrow-derived MSCε (5,000 cellε/35mm diεh) were exposed to various concentrations of 5-aza-CR (0, 1, 3, 10, 20, and 50 μM) 24 hr after seeding the cells into culture diεhes. The medium containing the 5-aza-CR was removed after the 24-hr exposure period and replaced with medium lacking 5-aza-CR. Seven dayε after this exposure, long multinucleated cells were observed in some of the disheε treated with more than 3 μM 5-aza-CR (Figure 7A); the cells in theεe cultureε were approximately 80% of confluence. The number of εuch multinucleated cellε increased as isolated colonieε or groupingε, and reached a maximum (9 colonieε in 10 of 35-mm diεheε) 2 weeks after the initial treatment. The number of such cells decreased (6 colonies in 10 of 35-mm disheε) by 5 weekε after treatment; 7 disappeared probably due to their contraction and detachment from the disheε and 4 new colonieε appeared during thiε time period; a substantial proportion of the multinucleated cells remained for up to 40 dayε after the initial exposure, which was the longest observational period. The morphology of the multinucleated cellε, obεerved by phase contrast microscopy of living cultures (Figure 7A), waε εimilar to that of rat muεcle in culture. We obεerved no diεcernible εtriationε, aε are routinely observed in embryonic chick myogenic cells in culture, although myotubes derived from myogenic cellε obtained from normal fetal rat limbε alεo did not show εtriationε (Figure 7B). Thus, neither the myotubes derived from MSCε nor those obtained from normal rat embryos exhibit striationε under the conditionε employed in theεe studies. Waves of spontaneouε contractionε or twitching of- some of these multinucleated cellε waε observed when viewing the living cultures. The contraction of these cells could also be stimulated by placing a drop of an acetylcholine solution onto these cellε, which iε a further indication that theεe cellε are myogenic.
To further confirm the identity of theεe multinucleated cellε, antibody to εkeletal muεcle εpecific myoεin waε presented to a fixed preparation of these cultures. Figure 8 shows a myotube stained positively with the anti-myosin antibody; again, cross striationε could not be observed. We alεo εtained yotubeε 2 weekε and 5 weekε after 5-aza-CR treatment with anti- yoεin antibody. Myotubeε 2 weekε after treatment were stained strongly positive (Figure 9A and 9B), although those 5 weeks after treatment were stained weakly (Figure 9C and 9D) .
The effect of 5-aza-CR appeared to be dependent on the concentration presented to MSCs. No myotubes were found in diεheε treated with 0 or 1 μM 5-aza-CR, but in thoεe treated with 3-50 μM 5-aza-CR, myotubes were observed with comparable incidence (Table 2).
TABLE 2
Number of Groupings of Myotubes or Adipocytes
Found Per Culture for MSCs Exposed to Different
Concentrations of 5-aza-CR
Figure imgf000037_0001
Secondary cultures of rat bone marrow cells were plated at 5,000 cells per 35mm dish, treated with the indicated concentration of 5-aza-CR, and observed 14 days after treatment. The numbers for the incidence of myotubes and adipocytes indicate the total number of phenotypically discernible groupings observed and the total number of culture disheε examined.
To measure Survival Index (SI*) in the presence of 5- aza-CR, MSCs were seeded at 200 cells/35mm dish and treated with 5-aza-CR 24 hr after plating. After 14 days, colonieε containing more than 10 cellε were counted, and thiε number waε multiplied by 100% and divided by 200 to generate the percentage.
When cells were treated with higher concentrations of 5-aza-CR, the number of cells on the plate decreased, with 10 μM appearing to be the most effective concentration with regard to the maximum number of myogenic cells and cell survival (plating efficiency of Table 2) . Thus, all subsequent experiments were done with 10 mM 5-aza-CR.
To examine the effect of 5-aza-2' -deoxycyti ine (5-aza-dCR) , a deoxy analogue of 5-aza-CR, rat bone marrow MSCs were treated with 0.3 μM, l μM, and 10 μM 5-aza-dCR in the same way as 5-aza-CR. Of the concentrations tested, 0.3 μM 5-aza-CdR gave the highest incidence of myogenic conversion, and the observed incidence was much higher than for cells exposed to 10 μM 5-aza-CR (Table 3) .
TABLE 3
Number of Groupings of Myotubes Found Per
Culture for MSCs Exposed to Different Concentrations of 5-aza-CdR and 5-aza-CR
Figure imgf000038_0001
* Survival Index
Secondary cultures of rat bone marrow cells were plated at 5,000 cells per 35mm dish, treated with the indicated concentration of 5-aza-dCR or 5-aza-CR, and observed 14 days after treatment. The numbers for the incidence of myotubes indicate the total number of phenotypically discernible groupings observed and the total number of culture dishes examined.
To measure Survival Index in the presence of 5-aza-CdR or 5-aza-CR, MSCs were seeded at 200 cells/35mm dish and treated with 5-aza-dCR or 5-aza-CR 24 hr after plating. After 14 dayε, colonieε containing more than 10 cells were counted, and thiε number waε multiplied by 100% and divided by 200 to generate the percentage.
To eliminate the poεεibility of contamination by εurrounding muεcle-derived myoblaεtε at the time of bone marrow harveεting, εecond paεεage rat bone marrow MSCε were cloned as described herein. Four clones of indistinguiεhable morphologies were obtained from this procedure and were exposed to 5-aza-CR for 24 hr; for emphasis, no cells in these clones exhibited muscle-like characteristics or positive immunostaining for muscle εpecific myoεin prior to exposure to 5-aza-CR. Of 4 clones exposed to 5-aza-CR, one clone exhibited the distinctive morphology of myotubes and adipocytes, which we interpret to indicate that non-muscle cells were converted to or influenced to become myoblasts or adipocyteε .
Firεt paεεage rat bone marrow-derived MSCε were exposed to 10 μM 5-aza-CR for 24 hr and cloned. From a total of 768 wellε, 136 colonieε were detected. Of theεe 136 colonieε, 7 (5%) exhibited a myogenic phenotype, 27 (20%) exhibited an adipocytic phenotype, and the other colonies lacked morphologies obviously related to discernible phenotypes.
To test the effect of 5-aza-CR and 5-aza-dCR on non-MSC preparations, we exposed brain fibroblastε to theεe same reagents . Rat brain fibroblaεtε were εeeded into 35-mm dishes at a density of 50,000 cells/dish and treated with 1 μM, 3 μM or 10 μM 5-aza-CR or 0.1 μM, 0.3 μM or 1 μM 5-aza-dCR in the same way as for rat MSCs. Each group had 9 disheε and cells were surveyed until 14 days after exposure. At day 7, all diεheε reached confluence, except for the group treated with 10 μM 5-aza-CR. No fat cellε nor myotubeε could be found in any diεhes during the period of obεervation. MSCε were collected from the bone marrow of young (4 week-old, 100 g) and adult (3 month-old, 250 g) donor ratε and passaged, and the number of colonies of myogenic phenotype after exposure to 5-aza-CR were compared (Table 4).
TABLE 4
Number of Groupings of Myotubes Per Culture of MSCs Exposed to 5-aza-CR
Figure imgf000040_0001
Secondary cultures of rat bone marrow MSCs were plated at 5,000 cells per 35mm diεh, treated with μM 5-aza-CR and 24 hr later changed to DMEM with different levelε of FCS, HS, or 50 μM HC, and observed 14 dayε after expoεure to 5-aza-CR waε terminated. The numbers for the incidence of myotubes indicate the total number of culture dishes examined.
MSCs from young donor rats had more myogenic colonies than those from adult rats. Second passage cultures of young donor MSCs exposed to 5-aza-CR produced more myogenic colonies compared with MSCs from older donors tested in cultureε from the firεt to fourth paεεage.
A variety of culture conditions were tested to attempt to optimize the expression of the myogenic phenotype of cultured MSCs exposed to 5-aza-CR. Exposed cells were cultured in medium containing various concentrations of FCS, HS, basic fibroblast growth factor (bFGF) and hydrocortisone. Table 4 shows that medium containing 10% FCS, 5% HS and hydrocortisone appeared to be the optimal medium for MSC expression of myogenic properties. Medium containing bFGF seemed to increase the expression of the myogenic phenotype (Table 5), although this may be related to an increase in the number of myoblasts due to myoblast division as opposed to increased conversion from progenitor cells.
TABLE 5
Comparison of 5-aza-CR-Induced Myotubes by Young and Old Rat Bone Marrow MSCs With Each Passage
Figure imgf000041_0001
Cells were cultured in DMEM with 10% FCS, 5% HS and 50 μM HC, with or without bFGF. The numbers for the incidence of myotubes indicate the total number of phenotypically discernible colonies or groupings observed and the total number of culture dishes examined. MSCs were obtained from young (lOOg) or old (250g) rats. In addition, bone marrow-derived MSCε were plated at 500 cellε/dish, 5,000 cells/dish, and 50,000 cellε/diεh and then exposed to 5-aza-CR. At 500 cellε/diεh, myogenic cells were first observed at 20 days after treatment, with the cells becoming confluent 25 days after treatment; 2 clusters of myogenic cells were observed in 5 dishes 29 days after treatment. At 5,000 cells/diεh, myogenic cellε were first observed at 7 days, with the cellε becoming confluent 10 dayε after treatment; 3 cluεterε were observed in 4 diεheε 14 dayε after treatment. At 50,000 cells/dish, myogenic cells were observed at 6 days, with the cells becoming confluent at 7 days after treatment; 10 cluεterε were obεerved in 5 diεheε 14 dayε after treatment.
The obεervationε preεented here indicate that rat bone marrow MSCs have the capacity to differentiate into the myogenic lineage in vitro following a brief exposure to 5-aza-CR. The obεerved myogenic cellε exhibited the characteriεtic multinucleated morphology of myotubeε, contracted εpontaneouεly, contracted when expoεed to acetylcholine, and εtained positively with a monoclonal antibody to skeletal muscle-εpecific myoεin, although these myotubes never exhibited apparent striationε. However, normal rat myoblaεtε collected from fetal rat muεcle did not, in our hands, form obviouεly εtriated myotubes in culture. We have attempted to exclude the posεibility of contamination by committed myogenic cellε by meticulouεly removing attached εoft tissue from the bones at the time of bone marrow harvesting. Importantly, we have never observed myotubes in any culture of rat bone marrow MSCs in hundreds of preparations, except for those exposed to sufficient concentrations of 5-aza-CR. In addition, a clone of rat bone marrow MSCs waε converted to both myogenic and adipocytic phenotypes after treatment with 5-aza-CR, which we interpret to mean that non-muscle progenitor cells were converted into theεe two phenotypeε. Since εkeletal muscle has not been observed in bone marrow, we believe that 5-aza-CR converts theεe marrow-derived MSCε into the myogenic cells.
Cytokine Expression by Human Marrow-Derived
Mesenchymal Stem Cells In Vitro:
Effects of IL-Iα and Dexamethasone
The objective of the present study was to further establish the phenotypic characteristicε of cultured MSCε through identification of a cytokine expression profile. We used commercial ELISAs to identify and measure the levels of expresεion of cytokineε that are known to be important in the regulation of cell diviεion, differentiation or expreεεion of a variety of meεenchymal phenotypeε. We identified MSC cytokine expreεεion under culture conditions that we have previouεly reported allow MSCε to mitotically expand without differentiation (constitutive culture-expansion medium) . In addition, we assayed cytokine expresεion by MSCε in culture medium εupplemented with dexamethaεone or IL-lα. Dexamethaεone haε been reported to induce the differentiation of oεteo-progenitors into oεteoblaεtε. In contraεt, IL-lα, which iε εecreted into the marrow microenvironment by a variety of cells during the inflammatory response, has been reported to enhance the bone marrow stroma'ε capacity to εupport hematopoiesis and thus may play a role in controlling the differentiation and/or expression of bone marrow stromal fibroblasts.
The data from these analyseε show that cultured MSCs express a unique cytokine profile. In addition, dexamethasone and IL-lα alter the MSC cytokine expression profile in different ways. These data add to our understanding of the unique phenotypic profile of MSCs, and also identify macromoleculeε whoεe expression is developmentally regulated aε MSCs differentiate or modulate their phenotype towards the osteogenic lineage or marrow stromal phenotype.
MATERIALS AND METHODS
MSC Isolation and Culture-Expansion
Bone marrow waε obtained from six human donors, 3 male and 3 female of diverse ageε (Table 6).
Table 6 Donor Characteristics
Donor # Donor Age Clin. Cond. Gender
39 NHL*
58 breast cancer
38 myelodysplasia medulloblastoma M
28 Hodgkin's Lymphoma M
47 AML* M
*NHL ≡ non-Hodgkin's lymphoma; AML = acute myelogenouε leukemia
Each donor was in remisεion from cancer and waε undergoing marrow harveεted for future autologouε bone marrow tranεplantation. Approximately 10 ml of unfractionated bone marrow waε obtained from the harveεt and uεed in the aεεayε in thiε εtudy. MSCε were purified and cultured by a modification of previouεly reported methodε. Briefly, bone marrow aεpirates were transferred from their syringes into 50 ml conical tubes containing 25 ml of complete medium conεiεting of Dulbecco'ε Modified Eagleε Medium εupplemented with fetal bovine serum (FBS) from selected lots, to a final volume of 10%. The tubes were spun in a Beckman table top centrifuge at 1200 rpm in a GS-6 swing bucket rotor for 5 min to pellet the cells. The layer of fat that forms at the top of the sampleε and the εupernatantε were aεpirated uεing a εerological pipet and diεcarded. The cell pelletε were resuspended to a volume of 5 ml with Complete Medium and then transferred to the top of preformed gradients of 70% Percoll. The sampleε were loaded into a Sorvall GS-34 fixed angle rotor and centrifuged in a Sorvall High Speed Centrifuge at 460 x g for 15 min. The low denεity fraction of approximately 15 ml (pooled denεity = 1.03 g/ml) waε collected from each gradient and tranεferred to 50 ml conical tubeε to which were added 30 ml Complete Medium. The tubeε were centrifuged at 1200 rpm to pellet the cellε. The εupernatantε were diεcarded and the cells were resuspended in 20 ml of Complete Medium and counted with a hemocytometer after lyεing red blood cells with 4% acetic acid. Cells were adjuεted to a concentrated of 5xl07 cellε per 7 ml and seeded onto 100-mm culture plates at 7 ml per plate.
Culture and Passage of Marrow-derived MSCs
Marrow-derived MSCs were cultured in Complete Medium at 37°C in a humidified atmoεphere containing 95% air and 5% C02, with medium changeε every 3-4 dayε. When primary culture diεheε became near confluent, the cellε were detached with 0.25% trypsin containing 1 mM EDTA (GIBCO) for 5 min at 37°C. The enzymatic activity of trypsin was stopped by adding 1/2 volume of FBS. The cells were counted, split 1:3, and replated in 7 ml of Complete Medium. These first passage cells were allowed to divide for 4-6 days until they became near confluent. Near-confluent first passage cells were trypsinized and replated into the aεεay formatε aε described below.
Quantitative ELISA
Levels of cytokine expresεion by MSCs were measured using quantitative ELISA. ELISA kits (R&D Systemε, Minneapolis MN) with antibody εpecificitieε for the following cytokines were purchased; interleukin-3 (IL-3), interleukin-6 (IL-6), interleukin-11 (IL-11), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating activity (M-CSF), stem cell factor (SCF), leukemia inhibitory factor (LIF) and transforming growth factor-beta-2 (TGF-/S-2). Near-confluent, first passaged MSCs were replated into 35-mm plates at 50,000 cellε per plate and allowed to attach overnight. Culture conditionε were then changed to one of three test conditions: fresh Complete Medium; Complete Medium with Osteogenic Supplement; and Complete Medium with Stromagenic Supplement. Cultures were allowed to incubate in test media for 24 or 48 hourε at which pointε the εupernatantε were collected, flaεh frozen in dry ice-ethanol and εtored at -70°C in a Revco freezer until all of the εampleε were prepared to analyze together. Aεεayε were conducted by applying 100 μl of culture εupernatant onto the wellε of the ELISA plate followed by proceεεing the plateε per manufacturer'ε inεtructions. Standard curves were generated using standard cytokines supplied with the kitε and diluted to the appropriate concentrations. In some cases (particularly for the IL-6 asεay), the εupernatantε had to be diluted εubεtantially to generate low enough abεorbance measurements that could be quantified accurately from the standard curves.
Quantification of Cell Number
RESULTS
Constitutive Culture-Expansion Medium Condition
Detectable levels of εix of the nine aεsayed cytokines were present after 24 hour exposure to constitutive culture-expansion conditions. See Figures 12A-12D and 13A- 13C and εee Tableε 7-10 below). TABLE 7 Detected Cytokine Levels (24 hours.
Figure imgf000047_0001
TABLE 8 Detected Cvtokine Levels (24 hours.
Figure imgf000048_0001
TABLE 9 Detected Cytokine Levels (48 hours.
Figure imgf000049_0001
TABLE 10 Detected Cytokine Levels (48 hours)
Figure imgf000050_0001
The cytokineε expressed in terms of pg/10,000 cellε in 24 or 48 hourε, from loweεt to higheεt were: G-CSF, SCF, LIF, M-CSF, IL-11 and IL-6. Three cytokineε were not detected in the supematants under constitutive culture-expansion conditions: GM-CSF, IL-3 and TGF-02. Large differences were observed in the average cytokine expression of each cytokine in comparison to the average levels of expression of other cytokineε. At the extremes, he average detectable level of G-CSF expression (10 pg/10,000 cells/24 hours) was over 700 fold lower than the average level of expression of IL-6 (7421 pg/10,000 cells/24 hours) .
Osteogenic Supplement Culture Conditions
The addition of Osteogenic Supplements to Complete Medium resulted in no detectable changes in G-CSF, M-CSF and SCF relative to control (Figures 12A-12D and 13A-13B; Tables 7-10) . In contrast, OS medium significantly downregulated the expression of LIF (p<.01), IL-6 (p<.001) and IL-ii (p<.005) relative to the expression of these cytokines under constitutive culture-expansion medium conditions at 24 hours. These levels remained statistically lower than cytokine levels in constitutive culture-expansion medium conditions at 48 hours (Figures 12A-12D and 13A-13C; Tables 7-10) . The amount of OS medium-mediated inhibition varied for the three cytokines; at the 24 hour timepoint the average level of cytokine expression in OS-medium relative to constitutive culture-expansion medium conditions was as follows; LIF expression 55% ± 54%, IL-6 16% ± 9% and IL-ll 1% + 3%. The large standard deviation in the LIF percent change was due primarily to the measurements from one donor (donor #4) where the level of LIF expression was actually higher under OS medium conditions relative to constitutive culture-expansion conditions (Table 7) . For a given donor, the percent inhibition of a cytokine relative to the average absolute level of inhibition of that cytokine, was independent to the percent inhibition of the other two cytokines, relative to their average absolute levels of inhibition (Tables 7-10) . In addition, for each of the cytokineε, the percent inhibition for a given cytokine among the six individuals in the population, was independent of the initial levels of expression under conεtitutive culture-expanεion conditions (Figures 12A-12D and 13A-13C; Tables 7-10).
Stromagenic Supplement Culture Conditions
SS medium increased the expression of several cytokines by MSCs in a concentration dependent manner. Figure 14 illuεtrateε the 24 hour reεponεe of second passage MSCε to increaεing concentrationε of IL-lα in termε of expression of GM-CSF. There is a near linear increaεe in the level of GM-CSF secretion by MSCs, with increasing levels of IL-lα in the culture medium between 0.1-10.0 U/ml. Additional log increases in IL-lα to the culture medium resultε in little additional increaεe in GM-CSF expreεεion. Theεe data were uεed to identify the concentration of IL-lα to εupplement to the culture media in the experimentε described below. For all εubεequent assayε, 10 U/ml IL-lα were added to the culture media.
Culture medium εupplemented with 10 U/ml IL-lα induced εtatistically significant up-regulation in the expression of G-CSF (P<.05), M-CSF (p<0.02), LIF (p<0.02), IL-6 (p<0.01) and IL-11 (p<0.06) relative to cells cultured in conεtitutive culture-expanεion medium. In addition, IL-lα induced the expression of GM-CSF which was not detectable in constitutive culture-expansion medium. In contrast, IL-lα had no statistically significant effect on the expression of SCF relative to the level of expression under constitutive culture-expansion medium conditions. The fold increase in response to IL-lα varied depending on the cytokine. IL-6 (25.1 +/- 13.4 fold increase) waε εtimulated to the greateεt extent, followed by LIF (9.2 + 6.9 fold), M-CSF (5.2 + 1.7 fold) and IL-ll (4.9 + 3.3 fold). The average fold increase for G-CSF and GM-CSF were not calculated, since these cytokines were not detected in some or all constitutive culture-expansion cultures.
DISCUSSION
Our continued analyses of MSCs in this study were aimed at identifying additional phenotypic characteristics, and determining how this phenotype is altered when MSCs are exposed to regulatory molecules that cause differentiation or phenotypic modulation. In this study, we used ELISA assays to characterize the cytokine expression of MSCs under constitutive culture-expansion conditions, and in the presence of OS or SS.
MSCs express a unique profile of cytokines which include G-CSF, M-CSF, SCF, LIF, IL-6 and IL-ll under constitutive culture-expansion conditions. They do not express GM-CSF, IL-3 and TGF-32 under these conditions. OS down-regulates the expression of LIF, IL-6 and IL-ll, while not affecting the expression of the other cytokines expressed under constitutive culture conditions. OS was not observed to up-regulate the expression of any of the cytokines assayed in this study. In contrast, SS up-regulates the expression of G-CSF, M-CSF, LIF, IL-6 and IL-ll, and induces the expression of GM-CSF which was not detected under constitutive culture-expansion conditions. SS had no effect on SCF expression, and was not observed to down-regulate any of the cytokines assayed in this study. Through these data, a unique cytokine expression profile has been generated that can aid in distinguishing MSCs from other mesenchymal phenotypes. The identity of the cytokine profile should provide clues to determine the role that these cells play in the microenvironment of bone marrow which provides the inductive and regulatory information that εupportε hematopoiesis. In addition, the alterationε in thiε cytokine profile in response to OS and SS, identify specific cytokineε whoεe levelε of expression change as MSCs differentiate or modulate their phenotype in response to regulatory molecules.
IL-lα, which is releaεed in the marrow microenvironment by a variety of cell typeε during inflammatory responseε, induceε MSCε to up-regulate expreεεion of cytokines that support granulocytic (G-CSF and GM-CSF), monocytic/ osteoclastic (GM-CSF, LIF, M-CSF, IL-6) and megakaryocytic (IL- 11) differentiation. IL-lα haε been εhown to protect bone marrow from radio- and hemo-ablation. The IL-lα-induced up-regulation of cytokine expression by MSCε likely plays a role in the mechanismε of IL-lα'ε protective effectε.
Dexamethaεone, which induceε MSCs to differentiate into oεteoblaεtε, attenuateε the expreεεion of monocytic/osteoclaεtic (LIF, IL-6) and megakaryocytic (IL-ll) εupportive cytokineε, and haε no effect on the expreεεion of cytokineε that εupport granulocytic progenitorε (G-CSF, GM-CSF). The three cytokineε inhibited by dexamethaεone are of interest because each mediates its signal through a receptor that uses gpl30 in its signaling pathway.
Cited Literature
1. Caplan Al, In: 39th Annual Symposium of the Society for Developmental Biology, ed by S. Subtelney and U Abbott, pp 3768. New York, Alan R Lisε Inc, 1981.
2. Elmer et al. , Teratology, 24:215-223, 1981.
3. Hauεchka SD, Dev Biol, 37:345-368, 1974.
4. Solurεh et al., Dev Biol, 83:9-19, 1981.
5. Swalla et al., Dev Biol, 116:31-38, 1986.
6. Goshima et al., Clin Orthop Rel Res, 269:274-283 , 1991.
7. Aεhton et al., Clin Orthop Rel Res, 151:294-307, 1980.
8. Bruder et al. , Bone Mineral, 11:141-151, 1990.
9. Bennett et al., J Cell Sci , 55:131-139, 1991.
10. Benayahu et al., J Cell Physiol , 140:1-7, 1989.
11. Nakahara et al. , Exp Cell Res, 195:492-503, 1991.
12. Denniε et al. , Cell Transpl, 1:2332, 1991.
13. Ohguεhi, et al., Acta Scandia. , 60:334-339, 1989.
14. Wang et al., Growth Factors, 9:57, 1993.
15. Vukicevic et al., PNAS, S6:8793, 1989.
16. Cheng et al.. Endocrinology, 134:277, 1994.
17. Tenenbaum et al., Calcif. Tissue Int., 34:76, 1982.
18. Bruder et al. , Trans. Ortho. Res. Soc. , 16:58, 1991.
19. Leonard et al., Devi. Biol., 145:99, 1991.
20. Chen et al., Exp. Cell Res., 206:199, 1993.
21. Syfteεtad et al., Differentiation, 29:230, 1985.
22. Chen et al. , Exp. Cell Res., 1P5:509, 1991.
23. Kimura et al., Biomed. Jes. , 5:465, 1984.
24. Langille et al., Differentiation, 40:84, 1989.
25. Ruεsell et al. , Exp. Hematol . , 20:75-79, 1992.
26. Brenner et al. , Brit. J. of Haem. , 77:237-244, 1991.
27. Hayneεworth, et al. , Bone, 13:81-88, 1992.
28. Grigoriadiε, et al. , J. Cell Biol., 105:2139-2151, 1988.
29. Haynesworth, et al.. Bone, 13:69-80, 1992.
30. Bruder & Haynesworth, in preparation.
31. Bruder & Caplan, Dev. Biol., 141:319-329, 1990. 32. Pacifici, et al., Exp. Cell Res., 195:38, 1991.
33. Constantinides et al. , Nature, 267:364-366 , 1977.
34. Taylor et al. , Cell, 17:771-779, 1979.
35. Konieczny et al., Cell, 35:791-800, 1984.
36. Lassar, Cell, 47:649-656, 1986.

Claims

What iε Claimed Iε:
1. A method for effecting the lineage-directed induction of isolated, culture expanded human mesenchymal stem cells which compriseε contacting the meεenchymal stem cells with a bioactive factor effective to induce differentiation thereof into a lineage of choice.
2. The method of claim 1 wherein the bioactive factor induceε differentiation of εuch cellε into a mesenchymal lineage selected from the group consisting of osteogenic, chondrogenic, tendonogenic, ligamentogenic, myogenic, marrow stromagenic, adipogenic and dermogenic.
3. The method of claim 1 wherein the cellε are contacted with the bioactive factor ex vivo .
4 . The method of claim 3 wherein the cellε are contacted with the bioactive factor in a rigid porouε veεεel.
5. The method of claim 4 wherein the rigid porouε veεεel iε a ceramic cube.
6. The method of claim 3 wherein the cells are contacted with the bioactive factor in a culture vesεel.
7. The method of claim 6 wherein the culture veεεel iε formed of a material εelected from the group conεiεting of glaεs and plastic.
8. The method of claim 3 wherein the cells are contacted with the bioactive factor in an injectable liquid.
9. The method of claim 8 wherein the liquid is suitable for intramuscular, intravenous or intraarticular injection.
10. The method of claim 1 which comprises administering to an individual in need thereof a composition comprising isolated, culture-expanded human meεenchymal εtem cellε and a bioactive factor effective to induce differentiation of such cells into a lineage of choice.
11. The method of claim 1 which comprises administering the bioactive factor to an individual to whom a preparation comprising isolated human mesenchymal εtem cellε had been adminiεtered.
12. A method for inducing the in vivo production of human cytokines in an individual in need thereof which comprises administering to the individual isolated culture-expanded human meεenchymal εtem cellε and a bioactive factor effective to induce εuch cellε to differentiate into a cytokine- producing meεenchymal lineage in εuch individual.
13. The method of claim 12 wherein the meεenchymal εtem cellε and bioactive factor are administered together.
14. The method of claim 12 wherein the mesenchymal stem cell and the bioactive factor are administered separately.
15. The method of claim 1 which compriseε oεteogenic lineage induction and the bioactive factor iε an oεteoinductive factor.
16. The method of claim 15 wherein the oεteoinductive factor iε a bone morphogenic protein.
17. The method of claim 16 wherein the bone morphogenic protein iε εelected from the group conεiεting of BMP-2 and BMP-3.
18. The method of claim 15 wherein the osteoinductive factor is a fibroblast growth factor.
19. The method of claim 18 wherein the fibroblaεt growth factor is baεic fibroblaεt growth factor.
20. The method of claim 15 wherein the osteoinductive factor is a glucocorticoid.
21. The method of claim 20 wherein the glucocorticoid is dexamethasone.
22. The method of claim 15 wherein the osteoinductive factor is a prostaglandin.
23. The method of claim 22 wherein the prostaglandin is proεtaglandin El.
24. The method of claim 15 which further compriεeε contacting the iεolated human meεenchymal stem cellε with an adjunct factor for advanced differentiation.
25. The method of claim 24 wherein the adjunct factor iε εelected from the group conεiεting of aεcorbic acid and itε analogs and a glycerophosphate.
26. The method of claim 1 which compriεeε chondrogenic induction and the bioactive factor iε a chondroinductive factor.
27. The method of claim 26 wherein the chondroinductive factor iε a member of the transforming growth factor-/3 superfamily.
28. The method of claim 27 wherein the transforming growth factor-/8 εuperfamily member is a bone morphogenic protein.
29. The method of claim 28 wherein the bone morphogenic protein is BMP-4.
30. The method of claim 27 wherein the transforming growth factor-β εuperfamily member is TGF-31.
31. The method of claim 27 wherein the transforming growth factor-3 superfamily member iε inhibin A.
32. The method of claim 27 wherein the tranεforming growth factor-/3 εuperfamily member iε chondrogenic εtimulating activity factor.
33. The method of claim 26 wherein the chondroinductive factor iε a component of the collagenous extracellular matrix.
34. The method of claim 33 wherein the collagenous extracellular matrix component is collagen I.
35. The method of claim 34 wherein the collagen I iε in the form of a gel.
36. The method of claim 26 wherein the chondroinductive factor iε a vitamin A analog.
37. The method of claim 36 wherein the vitamin A analog iε retinoic acid.
38. The method of claim 1 which compriεeε εtromagenic induction and the bioactive factor is a εtromainductive factor.
39. The method of claim 38 wherein the εtromainductive factor is an interleukin.
40. The method of claim 39 wherein the interleukin iε selected from the group consisting of interleukin-lα and interleukin-2.
41. The method of claim 1 which compriseε myogenic induction and the bioactive factor iε a myoinductive factor.
42. The method of claim 41 wherein the myoinductive factor is a cytidine analog.
43. The method of claim 42 wherein the cytidine analog is selected from the group consiεting of 5-azacytidine and 5- aza-2'-deoxycytidine.
44. A compoεition compriεing iεolated, culture-expanded human meεenchymal εtem cellε and a bioactive factor effective to induce differentiation of εuch cells into a lineage of choice.
45. The composition of claim 14 which further comprises a culture medium.
46. A composition of matter compriεing the compoεition of claim 44 in a pharmaceutically acceptable carrier.
47. The composition of claim 46 wherein the pharmaceutically acceptable carrier is a rigid porous vessel.
48. The compoεition of claim 46 wherein the pharmaceutically acceptable carrier is a gel.
49. The composition of claim 46 wherein the pharmaceutically acceptable carrier is an injectable liquid.
PCT/US1996/000170 1995-01-24 1996-01-05 Lineage-directed induction of human mesenchymal stem cell differentiation WO1996023059A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
DK96903358T DK0805853T3 (en) 1995-01-24 1996-01-05 Progeny-directed induction of differentiation of human mesenchymal stem cells
DE69636979T DE69636979T2 (en) 1995-01-24 1996-01-05 CELL LINES-ORIENTED INDUCTION OF HUMAN MESENCHYMAL STEM CELL DIFFERENTIATION
EP96903358A EP0805853B1 (en) 1995-01-24 1996-01-05 Lineage-directed induction of human mesenchymal stem cell differentiation
JP52287196A JP4454697B2 (en) 1995-01-24 1996-01-05 Lineage-directed induction of human mesenchymal stem cell differentiation
AU47469/96A AU719098B2 (en) 1995-01-24 1996-01-05 Lineage-directed induction of human mesenchymal stem cell differentiation
CA002211120A CA2211120C (en) 1995-01-24 1996-01-05 Lineage-directed induction of human mesenchymal stem cell differentiation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/377,461 US5736396A (en) 1995-01-24 1995-01-24 Lineage-directed induction of human mesenchymal stem cell differentiation
US08/377,461 1995-01-24

Publications (2)

Publication Number Publication Date
WO1996023059A1 true WO1996023059A1 (en) 1996-08-01
WO1996023059A9 WO1996023059A9 (en) 1996-10-03

Family

ID=23489200

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/000170 WO1996023059A1 (en) 1995-01-24 1996-01-05 Lineage-directed induction of human mesenchymal stem cell differentiation

Country Status (11)

Country Link
US (2) US5736396A (en)
EP (2) EP1717310A1 (en)
JP (2) JP4454697B2 (en)
AT (1) ATE357508T1 (en)
AU (1) AU719098B2 (en)
CA (1) CA2211120C (en)
DE (1) DE69636979T2 (en)
DK (1) DK0805853T3 (en)
ES (1) ES2285710T3 (en)
PT (1) PT805853E (en)
WO (1) WO1996023059A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0868505A1 (en) * 1995-11-16 1998-10-07 Case Western Reserve University In vitro chondrogenic induction of human mesenchymal stem cells
EP0948255A1 (en) * 1996-12-06 1999-10-13 Osiris Therapeutics, Inc. Improved chondrogenic differentiation of human mesenchymal stem cells
EP1059929A2 (en) * 1998-02-24 2000-12-20 MCP Hahnemann University Isolated stromal cells for use in the treatment of diseases of the central nervous system
WO2001048150A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Cells capable of differentiating into heart muscle cells
JP2002521493A (en) * 1998-07-31 2002-07-16 ジェンザイム コーポレーション Improvement of cardiac function by mesenchymal stem cell transplantation
WO2006060779A2 (en) * 2004-12-03 2006-06-08 Case Western Reserve University Novel methods, compositions and devices for inducing neovascularization
US8048673B2 (en) 1999-08-19 2011-11-01 Artecel Sciences, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US8465733B2 (en) 2007-11-02 2013-06-18 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition containing human mesenchymal stem cell
WO2020086687A1 (en) * 2018-10-23 2020-04-30 The Johns Hopkins University Partioning of adult mesenchymal stem cells

Families Citing this family (319)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US6974571B2 (en) * 1995-03-28 2005-12-13 Thomas Jefferson University Isolated stromal cells and methods of using the same
US6787355B1 (en) * 1996-08-26 2004-09-07 Mcgill University Multipotent neural stem cells from peripheral tissues and uses thereof
US5964805A (en) * 1997-02-12 1999-10-12 Stone; Kevin R. Method and paste for articular cartilage transplantation
WO1998025634A1 (en) * 1996-12-10 1998-06-18 Hadasit Medical Research Services & Development Company Ltd. Serum-derived factor inducing cell differentiation and medical uses thereof
AU737641B2 (en) * 1997-03-07 2001-08-23 Chugai Seiyaku Kabushiki Kaisha Novel cell lines and screening methods using said cell lines
AU7481098A (en) * 1997-05-13 1998-12-08 Case Western Reserve University Osteoarthritis cartilage regeneration using human mesenchymal stem ce lls
US6110209A (en) * 1997-08-07 2000-08-29 Stone; Kevin R. Method and paste for articular cartilage transplantation
US6835377B2 (en) 1998-05-13 2004-12-28 Osiris Therapeutics, Inc. Osteoarthritis cartilage regeneration
US6387367B1 (en) * 1998-05-29 2002-05-14 Osiris Therapeutics, Inc. Human mesenchymal stem cells
JP2002521025A (en) * 1998-07-24 2002-07-16 ザ カーネギー インスチチューション オブ ワシントン Methods for germline stem cell maintenance and expansion using members of the TGF-β family of growth factors.
JP2003523166A (en) * 1998-09-29 2003-08-05 ガミダ セル リミテッド Methods for controlling proliferation and differentiation of stem and progenitor cells
EP1137421A1 (en) * 1998-11-13 2001-10-04 Osiris Therapeutics, Inc. Uses of fibroblasts or supernatants from fibroblasts for the suppression of immune responses in transplantation
US7001746B1 (en) 1999-01-29 2006-02-21 Artecel Sciences, Inc. Methods and compositions for the differentiation of human preadipocytes into adipocytes
US6197061B1 (en) * 1999-03-01 2001-03-06 Koichi Masuda In vitro production of transplantable cartilage tissue cohesive cartilage produced thereby, and method for the surgical repair of cartilage damage
US6777231B1 (en) 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
US20030082152A1 (en) * 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
US20050076396A1 (en) * 1999-03-10 2005-04-07 Katz Adam J. Adipose-derived stem cells and lattices
KR100979664B1 (en) * 1999-03-10 2010-09-02 유니버시티 오브 피츠버그 오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Adipose-derived stem cells and lattices
IL147990A0 (en) 1999-08-05 2002-09-12 Mcl Llc Multipotent adult stem cells and methods for isolation
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
AU7500400A (en) * 1999-09-30 2001-04-30 Mcgill University Autologous marrow stem cell (msc) transplantation for myocardial regeneration
JP2003513648A (en) 1999-10-29 2003-04-15 フイラデルフイア・ヘルス・アンド・エデユケーシヨン・コーポレーシヨン Isolation and expansion of human bone marrow stromal cells
CA2395271A1 (en) * 1999-12-28 2001-07-05 Isotis N.V. Cell culture medium containing growth factors and l-glutamine
DE60132429T2 (en) * 2000-02-26 2009-01-08 Artecel, Inc. PLURIPOTENTS FROM FAT-FROZEN-GENERIC STROMA CELLS GENERATED STRAIN CELLS AND THEIR USE
EP1918366A1 (en) 2000-02-26 2008-05-07 Artecel, Inc. Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof
US7582292B2 (en) 2000-02-26 2009-09-01 Artecel, Inc. Adipose tissue derived stromal cells for the treatment of neurological disorders
US7182781B1 (en) 2000-03-02 2007-02-27 Regeneration Technologies, Inc. Cervical tapered dowel
JP5090604B2 (en) * 2000-04-25 2012-12-05 オシリス セラピューティクス,インコーポレイテッド Use of mesenchymal stem cells for the preparation of pharmaceutical compositions for joint repair in animals
DE60120151D1 (en) * 2000-04-28 2006-07-06 Childrens Medical Center ISOLATION OF MESENCHYMAL STEM CELLS AND THEIR USE
US7442390B2 (en) * 2000-06-05 2008-10-28 University Of South Florida Method for enhancing engraftment of cells using mesenchymal progenitor cells
US6936281B2 (en) * 2001-03-21 2005-08-30 University Of South Florida Human mesenchymal progenitor cell
WO2001094541A2 (en) * 2000-06-05 2001-12-13 University Of South Florida Human mesenchymal progenitor cell
US7303769B2 (en) * 2000-06-05 2007-12-04 University Of South Florida Method for purifying pluri-differentiated mesenchymal progenitor 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
US20020012905A1 (en) 2000-06-14 2002-01-31 Snodgrass H. Ralph Toxicity typing using liver stem cells
AU2001286173B2 (en) * 2000-08-01 2007-10-25 Yissum Research Development Company Directed differentiation of embryonic cells
US8044259B2 (en) 2000-08-03 2011-10-25 The Regents Of The University Of Michigan Determining the capability of a test compound to affect solid tumor stem cells
US6984522B2 (en) * 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US20020045260A1 (en) * 2000-10-17 2002-04-18 Shih-Chieh Hung Method of isolating mesenchymal stem cells
WO2002044343A2 (en) * 2000-11-22 2002-06-06 Geron Corporation Tolerizing allografts of pluripotent stem cells
US6576464B2 (en) 2000-11-27 2003-06-10 Geron Corporation Methods for providing differentiated stem cells
US6921665B2 (en) 2000-11-27 2005-07-26 Roslin Institute (Edinburgh) Selective antibody targeting of undifferentiated stem cells
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
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
KR100915482B1 (en) * 2000-12-06 2009-09-03 하리리 로버트 제이 Method of collecting placental stem cells
US20020136709A1 (en) * 2000-12-12 2002-09-26 Nucleus Remodeling, Inc. In vitro-derived adult pluripotent stem cells and uses therefor
EP1362095B1 (en) * 2001-02-14 2015-05-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
NZ528035A (en) * 2001-02-14 2005-07-29 Robert J Hariri Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
EP1367899A4 (en) * 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US20040115238A1 (en) * 2001-02-21 2004-06-17 Laurencin Cato T. Muscle-polymer constructs for bone tissue engineering
EP1379869A4 (en) * 2001-03-15 2004-08-11 Xiao Yong Fu Method for therapeutically treating a clinically recognized form of cardiopathology in a living mammal
WO2002078449A2 (en) * 2001-04-02 2002-10-10 Advanced Cell Technology, Inc. Method for facilitating the production of differentiated cell types and tissues from embryonic and adult pluripotent and multipotent cells
EP1393067A4 (en) * 2001-04-13 2005-01-05 Anterogen Co Ltd Encapsulated cell indicator system
IL158368A0 (en) * 2001-04-13 2004-05-12 Anterogen Co Ltd Methods and reagents for cell transplantation
KR100449141B1 (en) * 2001-04-19 2004-09-21 (주)라이프코드 Method for differentiating a mesenchymal stem cell into neural cells
US20040137612A1 (en) * 2001-04-24 2004-07-15 Dolores Baksh Progenitor cell populations , expansions thereof, and growth of non-hematopoietic cell types and tissues therefrom
US20030049236A1 (en) * 2001-07-27 2003-03-13 Arhus Amt Immortalized stem cells
JP2003052360A (en) * 2001-08-20 2003-02-25 Japan Science & Technology Corp Method for culturing mesenchymal stem cell using extracellular substrate of basement membrane cell
JP2005500847A (en) * 2001-08-24 2005-01-13 アドバンスド セル テクノロジー、インコーポレイテッド Screening assay for identification of differentiation inducers and preparation of differentiated cells for cell therapy
JP2005506860A (en) 2001-09-15 2005-03-10 ラツシユ−プレスビテリアン−セント・リユークズ・メデイカル・センター Layered cartilage tissue and engineering method thereof
US20040120933A9 (en) * 2001-09-17 2004-06-24 Rudnicki Michael A. Pax-encoding vector and use thereof
US7384784B2 (en) * 2001-09-17 2008-06-10 Ottawa Health Research Institute Pax-encoding vector and use thereof
US9969980B2 (en) * 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
US20030165473A1 (en) * 2001-11-09 2003-09-04 Rush-Presbyterian-St. Luke's Medical Center Engineered intervertebral disc tissue
US7771716B2 (en) 2001-12-07 2010-08-10 Cytori Therapeutics, Inc. Methods of using regenerative cells in the treatment of musculoskeletal disorders
US7514075B2 (en) * 2001-12-07 2009-04-07 Cytori Therapeutics, Inc. Systems and methods for separating and concentrating adipose derived stem cells from tissue
US20050008626A1 (en) * 2001-12-07 2005-01-13 Fraser John K. Methods of using adipose tissue-derived cells in the treatment of cardiovascular conditions
US7585670B2 (en) 2001-12-07 2009-09-08 Cytori Therapeutics, Inc. Automated methods for isolating and using clinically safe adipose derived regenerative cells
US8105580B2 (en) 2001-12-07 2012-01-31 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to promote wound healing
US9597395B2 (en) 2001-12-07 2017-03-21 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in the treatment of cardiovascular conditions
US7651684B2 (en) 2001-12-07 2010-01-26 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in augmenting autologous fat transfer
CN1630526B (en) 2001-12-07 2010-05-05 马克罗珀尔生物外科公司 Systems and methods for treating patients with processed lipoaspirate cells
US8404229B2 (en) 2001-12-07 2013-03-26 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to treat acute tubular necrosis
WO2006075986A1 (en) * 2005-01-10 2006-07-20 Macropore Biosurgery, Inc. Devices and methods for monitoring, managing, and servicing medical devices
US7595043B2 (en) * 2001-12-07 2009-09-29 Cytori Therapeutics, Inc. Method for processing and using adipose-derived stem cells
US20060204556A1 (en) * 2001-12-07 2006-09-14 Cytori Therapeutics, Inc. Cell-loaded prostheses for regenerative intraluminal applications
US20050095228A1 (en) 2001-12-07 2005-05-05 Fraser John K. Methods of using regenerative cells in the treatment of peripheral vascular disease and related disorders
JP2005511754A (en) * 2001-12-07 2005-04-28 リージェンツ オブ ザ ユニバーシティ オブ ミシガン Predictive identification and characterization of breast cancer stem cells
JP2005518828A (en) * 2001-12-20 2005-06-30 マクロポア インコーポレイテッド System and method for treating a patient using a collagen-rich substance extracted from adipose tissue
IL152904A0 (en) * 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
EP1465982A4 (en) * 2002-01-25 2006-06-07 Gamida Cell Ltd Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
CA2477411A1 (en) * 2002-03-02 2003-09-12 Board Of Regents, The University Of Texas Local production and/or delivery of anti-cancer agents by stromal cell precursors
US20040091936A1 (en) * 2002-05-24 2004-05-13 Michael West Bank of stem cells for producing cells for transplantation having HLA antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
US9321992B2 (en) * 2002-06-14 2016-04-26 Case Western Reserve University Cell targeting methods and compositions
WO2004016779A1 (en) * 2002-08-17 2004-02-26 Hae-Young Suh A method for transdifferentiating mesenchymal stem cells into neuronal cells
KR100495532B1 (en) * 2002-09-18 2005-06-14 에프씨비파미셀 주식회사 Method of differentiating mesenchymal stem cell into neural cell
US9969977B2 (en) * 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
WO2004035132A2 (en) 2002-10-15 2004-04-29 Irm Llc Compositions and methods for inducing osteogenesis
US20050054097A1 (en) * 2002-11-17 2005-03-10 Tony Peled EX-VIVO expansion of hematopoietic system cell populations in mononuclear cell cultures
US20040101959A1 (en) * 2002-11-21 2004-05-27 Olga Marko Treatment of tissue with undifferentiated mesenchymal cells
EP1571910A4 (en) 2002-11-26 2009-10-28 Anthrogenesis Corp Cytotherapeutics, cytotherapeutic units and methods for treatments using them
EP1428889A1 (en) * 2002-12-10 2004-06-16 Epigenomics AG Method for monitoring the transition of a cell from one state into another
KR100484550B1 (en) * 2003-01-23 2005-04-22 (주)안트로젠 Method of preparing cell for transplantation
WO2004076652A1 (en) * 2003-02-25 2004-09-10 Tokai University Medium for stem cells to be used in intervertebral disk generation and regeneration of intervertebral disk using stem cells
AU2004217699B2 (en) * 2003-03-07 2008-07-03 Gamida-Cell Ltd. Expansion of renewable stem cell populations using modulators of PI 3-kinase
ATE482725T1 (en) * 2003-04-01 2010-10-15 Us Dept Of Veteran S Affaires STEM CELL, PROGRESSOR CELL OR TARGET CELL BASED TREATMENT OF MULTI-ORGAN FAILURE.
US7067123B2 (en) 2003-04-29 2006-06-27 Musculoskeletal Transplant Foundation Glue for cartilage repair
US7901457B2 (en) * 2003-05-16 2011-03-08 Musculoskeletal Transplant Foundation Cartilage allograft plug
CA2528248A1 (en) * 2003-06-06 2004-12-16 The Hospital For Sick Children Neural crest stem cells and uses thereof
US20060019256A1 (en) * 2003-06-09 2006-01-26 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
WO2005038012A2 (en) 2003-06-27 2005-04-28 Ethicon Incorporated Cartilage and bone repair and regeneration using postpartum-derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived 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
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
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
US20070275462A1 (en) * 2003-10-02 2007-11-29 Hall Christopher L Human Prostate Cancer Cell Factor(s) that Induce Stem Cell Commitment and Osteogenesis
US7635591B2 (en) * 2003-10-29 2009-12-22 Fcb Pharmicell Co., Ltd. Method for differentiating mesenchymal stem cell into neural cell and pharmaceutical composition containing the neural cell for neurodegenerative disease
WO2005047524A2 (en) * 2003-11-10 2005-05-26 The Scripps Research Institute Compositions and methods for inducing cell dedifferentiation
WO2005054448A1 (en) * 2003-12-04 2005-06-16 Guhathakurta Dr Soma A novel process and method for deriving cardiomyocyte precursors from bone-marrow stem cells
ES2396689T3 (en) 2003-12-11 2013-02-25 Isto Technologies Inc. Particle Cartilage System
US8147806B2 (en) 2004-01-16 2012-04-03 Carnegie Mellon University Cellular labeling for nuclear magnetic resonance techniques
WO2005074633A2 (en) * 2004-02-03 2005-08-18 The Regents Of The University Of Michigan Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
CN101080486B (en) * 2004-04-23 2012-05-16 佰欧益股份有限公司 Multi-lineage progenitor cells
US20050288796A1 (en) * 2004-06-23 2005-12-29 Hani Awad Native soft tissue matrix for therapeutic applications
US8513011B2 (en) * 2004-08-26 2013-08-20 Biotech Research Ventures Pte Limited Methods and compositions for culturing cardiomyocyte-like cells
EP1799812A4 (en) * 2004-09-16 2009-09-09 Gamida Cell Ltd Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
US7837740B2 (en) * 2007-01-24 2010-11-23 Musculoskeletal Transplant Foundation Two piece cancellous construct for cartilage repair
JP5500773B2 (en) 2004-11-11 2014-05-21 ザ ジェネラル ホスピタル コーポレーション Activation of parathyroid hormone receptor and proliferation of stem and progenitor cells
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
PL1831356T3 (en) 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
KR100833612B1 (en) 2004-12-29 2008-05-30 에프씨비파미셀 주식회사 Pharmaceutical composition containing the neural cell differentated from mesenchymal stem cell for neurodegenerative disease
CA2512667A1 (en) * 2005-01-07 2006-07-07 Takahiro Ochiya Human hepatocyte-like cells and uses thereof
JP2006289062A (en) * 2005-03-18 2006-10-26 Pentax Corp Bone filling material using cartilage cell having hypertrophy ability and scaffold
EP2399991B1 (en) * 2005-04-12 2017-09-27 Mesoblast, Inc. Isolation of adult multipotential cells by tissue non-specific alkaline phosphatase
US20060252073A1 (en) * 2005-04-18 2006-11-09 Regents Of The University Of Michigan Compositions and methods for the treatment of cancer
CA2608048A1 (en) * 2005-05-10 2006-11-16 United States Of America Department Of Veteran's Affairs Therapy of kidney diseases and multiorgan failure with mesenchymal stem cells and mesenchymal stem cell conditioned media
JP2008545703A (en) 2005-05-27 2008-12-18 バイアセル インコーポレーティッド Treatment of ischemia using stem cells
WO2006135886A2 (en) * 2005-06-13 2006-12-21 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
CA2612021A1 (en) * 2005-06-13 2006-12-28 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US7815926B2 (en) * 2005-07-11 2010-10-19 Musculoskeletal Transplant Foundation Implant for articular cartilage repair
US8440460B2 (en) * 2005-07-27 2013-05-14 The Board Of Regents Of The University Of Texas System Methods for transdifferentiating cells
BRPI0614470A2 (en) * 2005-08-01 2011-03-29 Nupotential Inc production of reprogrammed cells with restored potential
JP5292533B2 (en) 2005-08-26 2013-09-18 ジンマー・インコーポレイテッド Implant and joint disease treatment, replacement and treatment methods
CA2623106C (en) 2005-09-19 2013-12-24 Histogenics Corporation Cell-support matrix having narrowly defined uniformly vertically and non-randomly organized porosity and pore density and a method for preparation thereof
ES2452595T3 (en) 2005-10-13 2014-04-02 Anthrogenesis Corporation Immunomodulation using placental stem cells
ES2618785T3 (en) 2005-10-31 2017-06-22 Oncomed Pharmaceuticals, Inc. Compositions and methods for treating cancer based on human FZD receptors
EP1945754B1 (en) * 2005-10-31 2014-07-23 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US7723477B2 (en) 2005-10-31 2010-05-25 Oncomed Pharmaceuticals, Inc. Compositions and methods for inhibiting Wnt-dependent solid tumor cell growth
US20070104692A1 (en) * 2005-11-07 2007-05-10 Quijano Rodolfo C Breast tissue regeneration
US20070104693A1 (en) * 2005-11-07 2007-05-10 Quijano Rodolfo C Breast augmentation system
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
US9155762B2 (en) * 2005-12-08 2015-10-13 University Of Louisville Research Foundation, Inc. Uses and isolation of stem cells from bone marrow
CN101573441A (en) * 2005-12-08 2009-11-04 路易斯维尔大学研究基金会有限公司 Very small embryonic-like (vsel) stem cells and methods of isolating and using the same
WO2007070870A1 (en) 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
EP1976975B1 (en) * 2005-12-19 2012-08-01 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
PL2471904T3 (en) * 2005-12-29 2019-08-30 Celularity, Inc. Placental stem cell populations
US8455250B2 (en) 2005-12-29 2013-06-04 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
CA2640185A1 (en) * 2006-01-24 2007-08-02 Christopher J. Centeno Mesenchymal stem cell isolation and transplantation method and system to be used in a clinical setting
PT1989295T (en) * 2006-02-16 2020-10-29 Univ Bruxelles A method for osteogenic differentiation of bone marrow stem cells (bmsc) and uses thereof
HUE028796T2 (en) 2006-03-23 2017-01-30 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
WO2007120911A2 (en) * 2006-04-14 2007-10-25 Carnegie Mellon University Cellular labeling and quantification for nuclear magnetic resonance techniques
WO2008054509A2 (en) * 2006-04-14 2008-05-08 Celsense, Inc. Methods for assessing cell labeling
EP2019858B1 (en) * 2006-04-17 2012-06-13 BioE LLC Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
WO2007139551A1 (en) * 2006-05-30 2007-12-06 Cytori Therapeutics, Inc. Systems and methods for manipulation of regenerative cells from adipose tissue
DK2024493T3 (en) * 2006-05-31 2014-10-06 Cellect Biotechnology Ltd PROCEDURES FOR SELECTION OF STEM CELLS AND USE THEREOF
ZA200810412B (en) * 2006-06-09 2010-03-31 Anthrogenesis Corp Placental niche and use thereof to culture stem cells
EP2489728A1 (en) 2006-06-15 2012-08-22 Neostem, Inc Processing procedure for peripheral blood stem cells
US20080003681A1 (en) * 2006-06-28 2008-01-03 Mahalaxmi Gita Bangera Methods for altering cellular susceptibility to infection
US20100015104A1 (en) * 2006-07-26 2010-01-21 Cytori Therapeutics, Inc Generation of adipose tissue and adipocytes
US7993918B2 (en) * 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
CA2850793A1 (en) * 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8163549B2 (en) 2006-12-20 2012-04-24 Zimmer Orthobiologics, Inc. Method of obtaining viable small tissue particles and use for tissue repair
US20080154233A1 (en) * 2006-12-20 2008-06-26 Zimmer Orthobiologics, Inc. Apparatus for delivering a biocompatible material to a surgical site and method of using same
US8148147B2 (en) * 2007-01-24 2012-04-03 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing pancreatic cancer
AU2008216749B2 (en) * 2007-02-12 2014-03-13 Celularity Inc. Treatment of inflammatory diseases using placental stem cells
US8435551B2 (en) 2007-03-06 2013-05-07 Musculoskeletal Transplant Foundation Cancellous construct with support ring for repair of osteochondral defects
US7632340B2 (en) * 2007-03-07 2009-12-15 Hamilton Beach Brands, Inc. Air purifier for removing particles or contaminants from air
WO2008128075A1 (en) 2007-04-12 2008-10-23 Isto Technologies, Inc. Compositions and methods for tissue repair
TWM322542U (en) * 2007-05-23 2007-11-21 Universal Scient Ind Co Ltd Testing machine
US9511076B2 (en) * 2008-05-30 2016-12-06 Clarion Research Group Formulations and methods for recovery from dental surgery
US8124130B1 (en) * 2007-05-30 2012-02-28 James Louis Rutkowski Formulations and methods for recovery from dental surgery
US20120195938A1 (en) * 2007-05-30 2012-08-02 James Louis Rutkowski Formulations and methods for recovery from dental surgery
WO2008148218A1 (en) * 2007-06-06 2008-12-11 The Hospital For Sick Children Skin-derived precursor cells and uses thereof
WO2008156728A1 (en) * 2007-06-15 2008-12-24 Neuronyx, Inc. Treatment of diseases and disorders using self-renewing colony forming cells cultured and expanded in vitro
WO2008156659A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US9095562B2 (en) * 2007-07-05 2015-08-04 Regenerative Sciences, Inc. Methods and compositions for optimized expansion and implantation of mesenchymal stem cells
JP5645658B2 (en) * 2007-07-10 2014-12-24 カーネギー メロン ユニバーシティー Compositions and methods for producing cell labels for nuclear magnetic resonance technology
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
CN101802174B (en) 2007-09-11 2013-06-05 北海道公立大学法人札幌医科大学 Cell growth method and pharmaceutical preparation for tissue repair and regeneration
KR20100075924A (en) 2007-09-19 2010-07-05 플루리스템 리미티드 Adherent cells from adipose or placenta tissues and use thereof in therapy
KR20220122774A (en) * 2007-09-26 2022-09-02 셀룰래리티 인코포레이티드 Angiogenic cells from human placental perfusate
EP2783692B1 (en) * 2007-09-28 2019-01-02 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
WO2009046346A2 (en) 2007-10-04 2009-04-09 Medistem Laboratories, Inc. Stem cell therapy for weight loss
UA99152C2 (en) 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8431162B2 (en) * 2007-10-30 2013-04-30 University Of Louisville Research Foundation, Inc. Subpopulations of bone marrow-derived adherent stem cells and methods of use therefor
EP2214497A4 (en) 2007-10-30 2010-11-24 Univ Louisville Res Found Uses and isolation of very small embryonic-like (vsel) stem cells
US20110200642A1 (en) * 2007-12-19 2011-08-18 Regenerative Sciences, Llc Compositions and Methods to Promote Implantation and Engrafment of Stem Cells
US8236538B2 (en) * 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US20100278788A1 (en) * 2008-01-11 2010-11-04 Bone Therapeutics, S.A. Osteogenic Differentiation Of Bone Marrow Stem Cells And Mesenchymal Stem Cells Using A Combination Of Growth Factors
US10046011B2 (en) 2008-01-31 2018-08-14 Rutgers, The State University Of New Jersey Compositions for inducing or suppressing an immune response
US8685728B2 (en) * 2008-01-31 2014-04-01 Rutgers The State University Of New Jersey Kit containing stem cells and cytokines for use in attenuating immune responses
WO2009111069A1 (en) 2008-03-05 2009-09-11 Musculoskeletal Transplant Foundation Cancellous constructs, cartilage particles and combinations of cancellous constructs and cartilage particles
CN102026546A (en) * 2008-03-14 2011-04-20 再生科学有限责任公司 Compositions and methods for cartilage repair
EP2282746B8 (en) * 2008-03-27 2015-12-09 Caladrius Biosciences, Inc. Compositions and methods using stem cells in cutaneous wound healing
CA2723171C (en) * 2008-05-02 2018-03-27 Celsense, Inc. Emulsion compositions and methods for nuclear magnetic resonance and other imaging
CA2722625A1 (en) 2008-05-07 2009-11-12 Bone Therapeutics S.A. Novel mesenchymal stem cells and bone-forming cells
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
WO2009144720A1 (en) 2008-05-27 2009-12-03 Pluristem Ltd. Methods of treating inflammatory colon diseases
WO2010021993A1 (en) 2008-08-19 2010-02-25 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in the treatment of the lymphatic system and malignant disease
MX2011001990A (en) 2008-08-20 2011-03-29 Anthrogenesis Corp Improved cell composition and methods of making the same.
CN105796602A (en) * 2008-08-20 2016-07-27 人类起源公司 Treatment of stroke using isolated placental cells
CN102176919A (en) 2008-08-22 2011-09-07 人类起源公司 Methods and compositions for treatment of bone defects with placental cell populations
WO2010026575A2 (en) 2008-09-02 2010-03-11 Pluristem Ltd. Adherent cells from placenta tissue and use thereof in therapy
PT2331136T (en) 2008-09-26 2018-03-27 Oncomed Pharm Inc Frizzled-binding agents and uses thereof
US8748177B2 (en) 2008-09-30 2014-06-10 The Hospital For Sick Children Compositions for proliferation of cells and related methods
EP2182055A1 (en) 2008-10-14 2010-05-05 Heinrich-Heine-Universität Düsseldorf Human cord blood derived unrestricted somatic stem cells (USSC)
KR20110086176A (en) * 2008-11-19 2011-07-27 안트로제네시스 코포레이션 Amnion derived adherent cells
US9192695B2 (en) * 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
MX2011005230A (en) * 2008-11-21 2011-06-16 Anthrogenesis Corp Treatment of diseases, disorders or conditions of the lung using placental cells.
KR20120006481A (en) 2008-12-05 2012-01-18 리제너러티브 사이언시즈, 엘엘씨 Methods and compositions to facilitate repair of avascular tissue
US20100168022A1 (en) * 2008-12-11 2010-07-01 Centeno Christopher J Use of In-Vitro Culture to Design or Test Personalized Treatment Regimens
PL2379088T3 (en) 2008-12-19 2018-07-31 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
US10179900B2 (en) * 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
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
AU2010229651B2 (en) 2009-03-26 2014-05-08 Advanced Technologies And Regenerative Medicine, Llc Human umbilical cord tissue cells as therapy for Alzheimer' s disease
WO2010111659A1 (en) 2009-03-27 2010-09-30 Wyeth Llc Tumor-initiating cells and methods for using same
WO2010127310A1 (en) * 2009-05-01 2010-11-04 Cytori Therapeutics, Inc. Systems, methods and compositions for optimizing tissue and cell enriched grafts
CA2767014C (en) 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
AU2010276201B2 (en) 2009-07-21 2013-10-17 Abt Holding Company Use of stem cells to reduce leukocyte extravasation
SG10201404281YA (en) 2009-07-21 2014-09-26 Abt Holding Co Use of stem cells to reduce leukocyte extravasation
US20110054929A1 (en) * 2009-09-01 2011-03-03 Cell Solutions Colorado Llc Stem Cell Marketplace
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
WO2011060135A1 (en) 2009-11-12 2011-05-19 Vbi Technologies, Llc Subpopulations of spore-like cells and uses thereof
US20140286911A1 (en) 2013-03-15 2014-09-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
US9044606B2 (en) 2010-01-22 2015-06-02 Ethicon Endo-Surgery, Inc. Methods and devices for activating brown adipose tissue using electrical energy
US8476227B2 (en) 2010-01-22 2013-07-02 Ethicon Endo-Surgery, Inc. Methods of activating a melanocortin-4 receptor pathway in obese subjects
ES2646750T3 (en) * 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
AU2011220721B2 (en) 2010-02-25 2015-02-05 Abt Holding Company Modulation of macrophage activation
BR112012022946A2 (en) 2010-03-12 2017-02-07 Daiichi Sankyo Co Ltd method for proliferating cardiomyocytes using micro-rna
KR20130043102A (en) 2010-04-01 2013-04-29 온코메드 파마슈티칼스, 인크. Frizzled-binding agents and uses thereof
SI2556145T1 (en) 2010-04-07 2017-01-31 Anthrogenesis Corporation Angiogenesis using placental stem cells
KR20130092394A (en) 2010-04-08 2013-08-20 안트로제네시스 코포레이션 Treatment of sarcoidosis using placental stem cells
CA2796251C (en) 2010-04-13 2019-05-14 Cellular Dynamics International, Inc. Hepatocyte production by forward programming
WO2011133658A1 (en) 2010-04-22 2011-10-27 Boston Medical Center Corporation Compositions and methods for targeting and delivering therapeutics into cells
US8455254B2 (en) * 2010-04-22 2013-06-04 Taipei Medical University Method of accelerating osteogenic differentiation and composition thereof
EP2582793B1 (en) 2010-06-15 2017-09-06 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
JP5897002B2 (en) 2010-07-07 2016-04-13 セルラー ダイナミクス インターナショナル, インコーポレイテッド Endothelial cell production by programming
US9296984B2 (en) 2010-07-09 2016-03-29 The Gid Group, Inc. Tissue processing apparatus and method for processing adipose tissue
US9260697B2 (en) 2010-07-09 2016-02-16 The Gid Group, Inc. Apparatus and methods relating to collecting and processing human biological material containing adipose
WO2013106655A1 (en) 2012-01-11 2013-07-18 The Gid Group, Inc. Method for processing adipose tissue and processing apparatus
US9206387B2 (en) 2010-07-09 2015-12-08 The Gid Group, Inc. Method and apparatus for processing adipose tissue
NZ605505A (en) 2010-07-13 2015-02-27 Anthrogenesis Corp Methods of generating natural killer cells
EP2625264B1 (en) 2010-10-08 2022-12-07 Terumo BCT, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
US9381219B2 (en) 2010-12-29 2016-07-05 Ethicon Endo-Surgery, Inc. Brown adipocyte modification
AR093183A1 (en) 2010-12-31 2015-05-27 Anthrogenesis Corp INCREASE IN THE POWER OF PLACENTA MOTHER CELLS USING MODULATING RNA MOLECULES
SG192124A1 (en) 2011-01-25 2013-08-30 Univ Catholique Louvain Compositions and methods for cell transplantation
WO2012109208A2 (en) 2011-02-08 2012-08-16 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
EP3260533B1 (en) 2011-03-22 2019-09-11 Pluristem Ltd. Methods for treating radiation or chemical injury
EP3443968A1 (en) 2011-06-01 2019-02-20 Celularity, Inc. Treatment of pain using placental stem cells
DK2718416T3 (en) 2011-06-06 2020-02-24 ReGenesys BVBA Expansion of stem cells in hollow fiber bioreactors
ES2803499T3 (en) 2011-06-29 2021-01-27 Biorestorative Therapies Inc Brown Fat Cell Compositions and Methods
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
JP2014530065A (en) 2011-10-11 2014-11-17 ボーン セラピューティクスソシエテ アノニム Use of growth differentiation factor 8 (GDF-8)
KR101981450B1 (en) 2011-12-23 2019-08-28 디퍼이 신테스 프로덕츠, 인코포레이티드 Detection of human umbilical cord tissue-derived cells
WO2013105098A1 (en) 2012-01-15 2013-07-18 Yeda Research And Development Co. Ltd. Induction of dedifferentiation of mesenchymal stromal cells
JP6034406B2 (en) 2012-01-25 2016-11-30 ユニヴェルシテ カソリック ド ルーヴァンUniversite Catholique De Louvain Compositions and methods for cell transplantation
CA2863795A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
US9434926B1 (en) * 2012-02-23 2016-09-06 University Of South Florida Graphene hydrogel and methods of using the same
US20150258148A1 (en) * 2012-06-06 2015-09-17 University Of Central Florida Research Foundation, Inc. Compositions, Methods and Systems for Cellular Differentiation from Stem Cells
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
EP3075841B1 (en) 2012-09-06 2021-03-10 GID BIO, Inc. Tissue processing apparatus and method for processing adipose tissue
BE1021755B1 (en) 2012-09-26 2016-01-15 Bone Therapeutics S.A. FORMULATIONS COMPRISING SOLVENT / DETERGENT-TREATED PLASMA (PLASMA S / D) AND USES THEREOF
AU2013334790A1 (en) 2012-10-23 2015-04-30 Oncomed Pharmaceuticals, Inc. Methods of treating neuroendocrine tumors using Wnt pathway-binding agents
US20140178343A1 (en) 2012-12-21 2014-06-26 Jian Q. Yao Supports and methods for promoting integration of cartilage tissue explants
JP2016510411A (en) 2013-02-04 2016-04-07 オンコメッド ファーマシューティカルズ インコーポレイテッド Methods and monitoring of treatment with WNT pathway inhibitors
AU2014215458A1 (en) 2013-02-05 2015-08-13 Anthrogenesis Corporation Natural killer cells from placenta
WO2014128634A1 (en) 2013-02-20 2014-08-28 Pluristem Ltd. Gene and protein expression properties of adherent stromal cells cultured in 3d
WO2014130770A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
US9168300B2 (en) 2013-03-14 2015-10-27 Oncomed Pharmaceuticals, Inc. MET-binding agents and uses thereof
AU2014248167B2 (en) 2013-04-03 2019-10-10 FUJIFILM Cellular Dynamics, Inc. Methods and compositions for culturing endoderm progenitor cells in suspension
HUE052516T2 (en) 2013-04-12 2021-05-28 Houston Methodist Hospital Improving organs for transplantation
EP2986714B1 (en) 2013-04-19 2021-01-06 Biorestorative Therapies, Inc. Human brown adipose derived stem cells and uses
AU2014266935B2 (en) 2013-04-30 2020-06-11 Katholieke Universiteit Leuven Cell therapy for myelodysplastic syndromes
EP3038629B1 (en) 2013-09-05 2020-11-18 GID BIO, Inc. Method for processing adipose tissue
WO2015073913A1 (en) 2013-11-16 2015-05-21 Terumo Bct, Inc. Expanding cells in a bioreactor
US11008547B2 (en) 2014-03-25 2021-05-18 Terumo Bct, Inc. Passive replacement of media
WO2015164228A1 (en) 2014-04-21 2015-10-29 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
US10851344B2 (en) 2014-08-07 2020-12-01 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Reversible stencils for fabricating micro-tissues
WO2016049156A1 (en) 2014-09-23 2016-03-31 Case Western Reserve University Compositions and methods for treating lung remodeling diseases
US20160090569A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled Feed
US10077420B2 (en) 2014-12-02 2018-09-18 Histogenics Corporation Cell and tissue culture container
US10080884B2 (en) 2014-12-29 2018-09-25 Ethicon Llc Methods and devices for activating brown adipose tissue using electrical energy
US10092738B2 (en) 2014-12-29 2018-10-09 Ethicon Llc Methods and devices for inhibiting nerves when activating brown adipose tissue
JP6450478B2 (en) 2015-04-23 2019-01-09 ボーン セラピューティクス エス.エー. In vitro storage of therapeutic cells
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11312940B2 (en) 2015-08-31 2022-04-26 University Of Louisville Research Foundation, Inc. Progenitor cells and methods for preparing and using the same
WO2017051421A1 (en) 2015-09-24 2017-03-30 Cellect Biotherapeutics Ltd. Methods for propagating mesenchymal stem cells (msc) for use in transplantation
WO2017143071A1 (en) 2016-02-18 2017-08-24 The Regents Of The University Of California Methods and compositions for gene editing in stem cells
US9918994B2 (en) 2016-03-04 2018-03-20 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for muscle regeneration using prostaglandin E2
EP3423568A4 (en) 2016-03-04 2019-11-13 University Of Louisville Research Foundation, Inc. Methods and compositions for ex vivo expansion of very small embryonic-like stem cells (vsels)
US20200299649A1 (en) 2016-03-29 2020-09-24 Smsbiotech, Inc. Compositions and methods for using small mobile stem cells
EP3464565A4 (en) 2016-05-25 2020-01-01 Terumo BCT, Inc. Cell expansion
US20200325449A1 (en) 2016-06-02 2020-10-15 The Cleveland Clinic Foundation Complement inhibition for improving cell viability
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
JP2019526342A (en) 2016-08-30 2019-09-19 ライフセル コーポレーションLifeCell Corporation System and method for controlling medical devices
USD851777S1 (en) 2017-01-30 2019-06-18 Lifecell Corporation Canister-type device for tissue processing
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
WO2018227134A1 (en) 2017-06-09 2018-12-13 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for preventing or treating muscle conditions
WO2019018002A1 (en) 2017-07-18 2019-01-24 The Gid Group, Inc. Adipose tissue digestion system and tissue processing method
KR102185697B1 (en) 2017-10-20 2020-12-03 본 테라퓨틱스 소시에테아노님 Mesenchymal stem cell differentiation method
US20210285009A1 (en) 2018-07-13 2021-09-16 The Regents Of The University Of California Retrotransposon-based delivery vehicle and methods of use thereof
SG11202102874UA (en) 2018-09-25 2021-04-29 Bone Therapeutics Sa Methods and uses for determining osteogenic potential of in vitro differentiated cells
BR112021005697A2 (en) 2018-09-25 2021-06-22 Bone Therapeutics Sa methods to obtain cells derived from mesenchymal stem cells (msc) of chondro-osteoblastic lineage from msc, populations of cells derived from msc of chondro-osteoblastic lineage and pharmaceutical formulation
AR118008A1 (en) 2019-02-07 2021-09-08 Vitricell Sa COMPOSITIONS FOR CRYOPRESERVATION OF A BIOLOGICAL MATERIAL
GB2595606B (en) 2019-03-07 2022-09-21 Univ California CRISPR-Cas effector polypeptides and methods of use thereof
CN113825498A (en) 2019-05-13 2021-12-21 骨治疗公司 Improved lyophilized formulations comprising hyaluronic acid and plasma proteins and uses thereof
WO2021007180A1 (en) 2019-07-05 2021-01-14 Case Western Reserve University Priming media and methods for stem cell culture and therapy
EP3889255A1 (en) 2020-03-30 2021-10-06 Rijksuniversiteit Groningen Small molecule inhibitors of rna guided endonucleases

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5226914A (en) * 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL68218A (en) * 1983-03-23 1985-12-31 Univ Ramot Compositions for cartilage repair comprising embryonal chondrocytes
US4904259A (en) * 1988-04-29 1990-02-27 Samuel Itay Compositions and methods for repair of cartilage and bone
US5612211A (en) * 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
US5733542A (en) * 1990-11-16 1998-03-31 Haynesworth; Stephen E. Enhancing bone marrow engraftment using MSCS
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
EP1361267A3 (en) * 1991-06-18 2003-11-26 CAPLAN, Arnold I. Mesenchymal stem cells and their use
US5266914A (en) * 1992-06-15 1993-11-30 The Herman Schmidt Company Magnetic chuck assembly
US5591625A (en) * 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
EP0852463B1 (en) * 1995-06-06 2008-09-03 Case Western Reserve University Myogenic differentiation of human mesenchymal stem cells
PT868505E (en) * 1995-11-16 2005-06-30 Univ Case Western Reserve IN VITRO CONDROGENIC INDUCTION OF HUMAN MESENCHINEAL STAMINA CELLS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5226914A (en) * 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0868505A4 (en) * 1995-11-16 2002-07-17 Univ Case Western Reserve In vitro chondrogenic induction of human mesenchymal stem cells
EP0868505A1 (en) * 1995-11-16 1998-10-07 Case Western Reserve University In vitro chondrogenic induction of human mesenchymal stem cells
EP0948255A4 (en) * 1996-12-06 2002-10-23 Osiris Therapeutics Inc Improved chondrogenic differentiation of human mesenchymal stem cells
JP2001512304A (en) * 1996-12-06 2001-08-21 オシリス セラピューティクス,インコーポレイテッド Improved chondrogenic differentiation of human mesenchymal stem cells
EP0948255A1 (en) * 1996-12-06 1999-10-13 Osiris Therapeutics, Inc. Improved chondrogenic differentiation of human mesenchymal stem cells
EP1815862A1 (en) * 1998-02-24 2007-08-08 MCP Hahnemann University Isolated stromal cells for use in the tretment of diseases of the central nervous system
EP1059929A4 (en) * 1998-02-24 2002-07-17 Univ Mcp Hahnemann Isolated stromal cells for use in the treatment of diseases of the central nervous system
EP1059929A2 (en) * 1998-02-24 2000-12-20 MCP Hahnemann University Isolated stromal cells for use in the treatment of diseases of the central nervous system
JP2002521493A (en) * 1998-07-31 2002-07-16 ジェンザイム コーポレーション Improvement of cardiac function by mesenchymal stem cell transplantation
US8048673B2 (en) 1999-08-19 2011-11-01 Artecel Sciences, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US8911994B2 (en) 1999-08-19 2014-12-16 Artecel Sciences Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
WO2001048150A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Cells capable of differentiating into heart muscle cells
WO2006060779A3 (en) * 2004-12-03 2006-09-14 Univ Case Western Reserve Novel methods, compositions and devices for inducing neovascularization
WO2006060779A2 (en) * 2004-12-03 2006-06-08 Case Western Reserve University Novel methods, compositions and devices for inducing neovascularization
US8465733B2 (en) 2007-11-02 2013-06-18 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition containing human mesenchymal stem cell
WO2020086687A1 (en) * 2018-10-23 2020-04-30 The Johns Hopkins University Partioning of adult mesenchymal stem cells

Also Published As

Publication number Publication date
CA2211120A1 (en) 1996-08-01
JP2010012331A (en) 2010-01-21
EP0805853A4 (en) 1999-12-22
AU719098B2 (en) 2000-05-04
US5736396A (en) 1998-04-07
MX9705612A (en) 1998-10-31
DE69636979T2 (en) 2007-12-20
EP0805853A1 (en) 1997-11-12
ATE357508T1 (en) 2007-04-15
PT805853E (en) 2007-06-27
EP1717310A1 (en) 2006-11-02
ES2285710T3 (en) 2007-11-16
EP0805853B1 (en) 2007-03-21
US5942225A (en) 1999-08-24
JP4454697B2 (en) 2010-04-21
AU4746996A (en) 1996-08-14
JPH10512756A (en) 1998-12-08
CA2211120C (en) 2001-04-03
JP5173982B2 (en) 2013-04-03
DK0805853T3 (en) 2007-07-23
DE69636979D1 (en) 2007-05-03

Similar Documents

Publication Publication Date Title
US5736396A (en) Lineage-directed induction of human mesenchymal stem cell differentiation
WO1996023059A9 (en) Lineage-directed induction of human mesenchymal stem cell differentiation
Haynesworth et al. Cell-based tissue engineering therapies: the influence of whole body physiology.
US7932084B2 (en) Methods and compositions for growing adipose stem cells
AU731468B2 (en) Regeneration and augmentation of bone using mesenchymal stem cells
US20080213235A1 (en) Adipose Tissue Stem Cells, Perivascular Cells and Pericytes
US6761887B1 (en) Alginate layer system for chondrogenic differentiation of human mesenchymal stem cells
US20100111897A1 (en) Methods and Compositions Useful for Diabetic Wound Healing
KR20070085288A (en) Method of enhancing proliferation and/or survival of mesenchymal precursor cells (mpc)
KR20050032020A (en) Adipose-derived stem cells and lattices
BRPI0713572A2 (en) serum free media and their uses for chondrocyte expansion
US20080187518A1 (en) Production of Osteoclasts from Adipose Tissues
US20150164951A1 (en) Human Bone-Forming Cells in the Treatment of Conditions and Bone Diseases Associated with Immunodeficiency or Immunosuppression
JP2002507383A (en) Methods for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US20200032212A1 (en) Human Bone-Forming Cells in the Treatment of Inflammatory Rheumatic Diseases
MXPA97005612A (en) Induction directed by lineage of the differentiation of human mesenquimatosas cells of ori
Koller et al. Primary mesenchymal cells
MÄNTYMAA The effects of hypoxic conditions on the chondrogenic differentiation of adipose stem cells
Dorman In Vitro Effects of BMP-2, BMP-7, AND BMP-13 on Differentiated and Undifferentiated Mouse Mesenchymal Stem Cells
Ishizeki et al. Effects of elcatonin on matrix calcification of Meckel's cartilage in vitro

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP MX

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

COP Corrected version of pamphlet

Free format text: PAGES 1/14-14/14,DRAWINGS,REPLACED BY NEW PAGES 1/15-15/15

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2211120

Country of ref document: CA

Ref country code: CA

Ref document number: 2211120

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1996 522871

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: PA/a/1997/005612

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1996903358

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996903358

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1996903358

Country of ref document: EP