US20090162934A1 - Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof - Google Patents

Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof Download PDF

Info

Publication number
US20090162934A1
US20090162934A1 US12/209,813 US20981308A US2009162934A1 US 20090162934 A1 US20090162934 A1 US 20090162934A1 US 20981308 A US20981308 A US 20981308A US 2009162934 A1 US2009162934 A1 US 2009162934A1
Authority
US
United States
Prior art keywords
cells
myoblast
hematopoietic
cell
stromal cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/209,813
Inventor
Jeffrey Martin Gimble
Yuan-Di Chang Halvorsen
William O. Wilkison
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Artecel Sciences Inc
Original Assignee
Artecel Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Artecel Sciences Inc filed Critical Artecel Sciences Inc
Priority to US12/209,813 priority Critical patent/US20090162934A1/en
Publication of US20090162934A1 publication Critical patent/US20090162934A1/en
Priority to US13/085,250 priority patent/US8486700B2/en
Priority to US13/941,213 priority patent/US20130288262A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors
    • 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
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells

Definitions

  • This invention relates to methods and compositions for the differentiation of stromal cells from adipose tissue into hematopoietic supporting stromal cells and myocytes of both the skeletal and smooth muscle types.
  • the neonatal period in human development is characterized by the presence of “stem” cells with the potential to develop along multiple differentiation pathways.
  • the terminal differentiation of these cells is determined by cytokine and hormonal cues which co-ordinate organogenesis and tissue architecture.
  • Murine embryonic stem cells have been isolated and studied extensively in vitro and in vivo. Using exogenous stimuli in vitro, investigators have induced ES cell differentiation along multiple lineage pathways. These include neuronal, B lineage lymphoid, and adipocytes (Dani et al. (1997) J. Cell Sci. 110: 1279; Remoncourt et al. (1998) Mech. Dev. 79:185; O'Shea K S (1999) Anat. Rec. 257:32).
  • the ES cells have been manipulated in vivo by homologous recombination techniques to generate gene specific null or “knock-out” mice (Johnson R S (1989) Science 245:1234). Once ES cell clones lacking a specific gene are isolated, they are transplanted into a fertilized murine zygote. The progeny of this isolated ES cell can develop into any and all murine tissues in a coordinated manner.
  • a stem cell must meet the following criteria: (1) ability of a clonal stem cell population to self-renew; (2) ability of a clonal stem cell population to generate a new, terminally differentiated cell type in vitro; and (3) ability of a clonal stem cell population to replace an absent terminally differentiated cell population when transplanted into an animal depleted of its own natural cells.
  • Multipotential stem cells exist in tissues of the adult organism.
  • the best characterized example of a “stem cell” is the hematopoietic progenitor isolated from the bone marrow and peripheral blood.
  • Seminal studies by Trentin, Till and McCulloch (McCulloch et al. (1996) Proc. Can. Cancer Conf. 6:356-366; Curry et al. (1967) J. Exp. Med. 125:703-720) examined lethally irradiated mice. In the absence of treatment, these animals died because they failed to replenish their circulating blood cells; however, transplantation of bone marrow cells from a syngeneic donor animal would rescue the host animal. The donor cells were responsible for reconstituting all of the circulating blood cells.
  • hematopoietic stem cells remain present in the normal human bone marrow throughout life; they are not limited to the neonatal period.
  • the adult bone marrow microenvironment is the potential source for these hypothetical mesodermal stem cells.
  • Cells isolated from adult marrow are referred to by a variety of names, including stromal cells, stromal stem cells, mesenchymal stem cells (MSCs), mesenchymal fibroblasts, reticular-endothelial cells, and Westen-Bainton cells (Gimble et al. (1996) Bone 19:421-428).
  • MSCs mesenchymal stem cells
  • mesenchymal fibroblasts mesenchymal fibroblasts
  • reticular-endothelial cells reticular-endothelial cells
  • Westen-Bainton cells Westen-Bainton cells
  • the bone marrow has been proposed as a source of stromal stem cells for the regeneration of bone, cartilage, muscle, adipose tissue, and other mesenchymal derived organs.
  • the major limitations to the use of these cells are the difficulty and risk attendant upon bone marrow biopsy procedures and the accompanying loss of memory B cells and hematopoietic stem cells with present harvesting procedures.
  • Adipose stromal cells provide an easily accessible and abundant source of stromal cells which can differentiate along multiple mesenchymal lineages. Methods and compositions are needed for the consistent and quantitative differentiation of adipose derived stromal cells into various cell types including for example hematopoietic stromal cells and skeletal and smooth muscle myocytes.
  • compositions and methods for the differentiation of adipocytes are provided.
  • the present invention provides methods and compositions for consistent and quantitative induction of stromal cells derived from subcutaneous, mammary, gonadal, or omental adipose tissues into the following fully differentiated and functional mesodermal cell lineages: hematopoietic supporting stromal cells, skeletal myocytes, and smooth muscle myocytes (myofibroblasts).
  • compositions include a variety of chemical components which act as mitogens and differentiation inducing agents for the plated stromal cells and yield production of the desired cell type.
  • the mitogens and inducing agents include, but are not limited to, interleukins, flt-3 ligand, stem cell factor, macrophage-colony stimulating factor, granulocyte-monocyte colony stimulating factor, erythropoietin, thrombopoietin, osteoprotegerin ligand, dexamethasone, hydrocortisone, 1,25 dihydroxy vitamin D 3 , 2-mercaptoethanol, glutamine, 5′-azacytadine, amphotericin, transforming growth factor ⁇ and fibroblast growth factor.
  • the invention provides methods for determining the ability of these compositions to direct the differentiation and function of the adipose-derived stromal cells, for the transduction of viral vectors carrying regulatory genes into stromal cells, for the transfection of plasmid vectors carrying regulatory genes into stromal cells, for the tracking and detection of functional proteins encoded by these genes, and for developing biomechanical carriers for the re-introduction of these cells into living organisms.
  • the invention also provides methods and compositions which have utility in drug discovery for compounds and proteins with relevance to a wide spectrum of disease states including, but not limited to, aplastic anemia, muscular dystrophy, radiation poisoning, neuropathic muscular degeneration, urogenital malformations, and gastrointestinal malformations.
  • FIG. 1 Human adipose-derived stromal cells were cultured in 6 well plates until confluent and quiescent in DMEM/F 10 (1:1 vol:vol), 10% fetal bovine serum, penicillin 100 units/ml, streptomycin 100 ⁇ g/ml, and 7.5 mM HEPES pH 7.2. The cells were then incubated in DMEM/F 10 (1:1 vol:vol), 2% fetal bovine serum, penicillin 100 units/ml, streptomycin 100 ⁇ g/ml, and 7.5 mM HEPES pH 7.2 containing 100 ng/ml lipopolysaccharide (LPS).
  • DMEM/F 10 (1:1 vol:vol
  • 10% fetal bovine serum penicillin 100 units/ml
  • streptomycin 100 ⁇ g/ml streptomycin 100 ⁇ g/ml
  • 7.5 mM HEPES pH 7.2 containing 100 ng/ml lipopolysaccharide (LPS).
  • RNA extraction and isolation were harvested immediately (time “0”) or after 4 hours (time “4”) for total RNA extraction and isolation using a phenol/chloroform/acid procedure (Chomczynski & Sacchi (1987) Analytical Biochem 162:156-159). Equal aliquots of total RNA were reverse transcribed and amplified by polymerase chain reaction with the following primer sets specific for the indicated human mRNAs; the actin primers served as a positive control for equal loading between samples:
  • the resulting PCR products were electrophoresed on a 2% agarose gel, stained with ethidium bromide and photographed.
  • cytokines listed in the table were detected using the oligonucleotide primers listed. All cytokines were expressed either in a constitutive or inducible manner.
  • Table 3. Quantitative ELISA (pg/ml) LPS Induction of Adipose-Derived Stromal Cell Secreted Cytokines. The listed cytokines were assayed in conditioned medium from human adipose derived stromal cells induced for 0 to 24 hours with 100 ng/ml of LPS. All cytokines were detected by enzyme linked immunoassay (ELISA) and are expressed as pg/ml of conditioned medium. Those cytokines indicated by an “*” demonstrated significant increases relative to the 0 hour time point within the 24 hour induction period based on one way analysis of variance.
  • the present invention provides methods and compositions for the differentiation and culture of adipose tissue-derived stromal cells into (a) hematopoietic supporting stromal cells, (b) skeletal myocytes, and (c) smooth muscle myocytes (myofibroblasts).
  • Adipose tissue offers a potential alternative to the bone marrow as a source of multipotential stromal stem cells.
  • Adipose tissue is readily accessible and abundant in many individuals. Obesity is a condition of epidemic proportions in the United States, where over 50% of adults exceed the recommended BMI based on their height.
  • Adipocytes can be harvested by liposuction on an outpatient basis. This is a relatively non-invasive procedure with cosmetic effects which are acceptable to the vast majority of patients. It is well documented that adipocytes are a replenishable cell population. Even after surgical removal by liposuction or other procedures, it is common to see a recurrence of adipocytes in an individual over time.
  • adipose tissue contains stromal stem cells which are capable of self-renewal.
  • Pathologic evidence suggests that adipose-derived stromal cells are capable of differentiation along multiple mesenchymal lineages.
  • the most common soft tissue tumor, liposarcomas develop from adipocyte-like cells.
  • Soft tissue tumors of mixed origin are relatively common. These may include elements of adipose tissue, muscle (smooth or skeletal), cartilage, and/or bone. Just as bone forming cells within the bone marrow can differentiate into adipocytes or fat cells, the extramedullary adipocytes are capable of forming bone.
  • subcutaneous adipocytes form bone for unknown reasons (Kaplan (1996) Arch. Dermatol. 132:815-818).
  • Human extramedullary adipose tissue-derived stromal cells represent a stromal stem cell source which can be harvested routinely with minimal risk to the patient. They can be expanded ex vivo, differentiated along unique mesodermal lineage pathways, genetically engineered, and re-introduced into individuals as either an autologous or allogeneic transplantation.
  • This invention presents examples of methods and composition for the isolation, characterization, and differentiation of adult human extramedullary adipose tissue stromal cells along the mesodermal lineages and outlines their use for the treatment of a number of human conditions and diseases.
  • the invention provides a method for differentiating adipose tissue-derived stromal cells into one of three functionally distinct mesodermal cell lineages: hematopoietic supporting cells, myocytes (skeletal), and myocytes (smooth muscle myofibroblasts) comprising: culturing said cells in a composition which comprises a medium (a) capable of supporting the growth of stromal cells and hematopoietic cells in co-culture with factors present capable of inducing stromal expression of hematopoietic growth factors or the addition of exogenous growth factors directly; (b) capable of supporting the growth and differentiation of stromal cells into functional and proliferating skeletal myocytes; and (c) capable of supporting the growth and differentiation of stromal cells into functional and proliferating smooth muscle myocytes or
  • compositions for the differentiation of adipose tissue-derived stromal cells into each of the three different mesodermal derived lineages comprise:
  • adipose tissue-derived stromal cells a medium capable of supporting the growth of the stromal cells, and growth factors and agents capable of inducing stromal cell expression of hematopoietic growth factors or exogenous hematopoietic growth factors or non-peptide factors themselves.
  • adipose tissue-derived stromal cells a medium capable of supporting the growth of the stromal cells, and amounts of 5′ azacytadine and/or amphotericin or other agents sufficient to induce the differentiation of said stromal cells into skeletal muscle myocytes.
  • adipose tissue-derived stromal cells a medium capable of supporting the growth of the stromal cells, and amounts of transforming growth factor ⁇ or other peptide growth factors sufficient to induce the differentiation of said stromal cells into smooth muscle myocytes or myofibroblasts.
  • the methods comprise incubation of isolated adipose tissue-derived stromal cells, plated at densities of about 1,000 to 25,000 cells/cm 2 in medium consisting of the following for each lineage:
  • Adipose stromal cells refers to stromal cells that originate from adipose tissue.
  • adipose is meant any fat tissue.
  • the adipose tissue may be brown or white adipose tissue, derived from subcutaneous, omental/visceral, mammary, gonadal, or other adipose tissue site.
  • the adipose is subcutaneous white adipose tissue.
  • Such cells may comprise a primary cell culture or an immortalized cell line.
  • the adipose tissue may be from any organism having fat tissue.
  • the adipose tissue is mammalian, most preferably the adipose tissue is human.
  • a convenient source of adipose tissue is from liposuction surgery, however, the source of adipose tissue or the method of isolation of adipose tissue is not critical to the invention. If stromal cells are desired for autologous transplantation into a subject, the adipose tissue will be isolated from that subject.
  • Hematopoietic supporting stromal cell refers to stromal cells that are capable of supporting the proliferation and maturation of hematopoietic progenitor, also known as hematopoietic stem cells, derived from bone marrow, spleen, peripheral blood, or umbilical blood, either CD34+ or CD34 ⁇ .
  • Co-cultures of hematopoietic supporting stromal cells with hematopoietic progenitors would result in the production of both adherent and non-adherent populations of hematopoietic blood cells, including but not limited to myeloid (macrophage, neutrophil, osteoclast), erythroid (red blood cells), lymphoid (B-lymphoid, T-lymphoid), and platelet (megakaryocyte), as well as eosinophils, basophils, mast cells and other circulating blood cell types.
  • myeloid microphage, neutrophil, osteoclast
  • erythroid red blood cells
  • lymphoid B-lymphoid, T-lymphoid
  • platelet megakaryocyte
  • hematopoietic cells Growth and differentiation of hematopoietic cells will be determined by assays which include, but are not limited to, those that assess the surface expression of characteristic blood cell lineage specific proteins (such as CD45 for B lineage lymphocytes, T cell receptor for T lineage lymphocytes, Mac-I/LFA11 for macrophages, tartrate resistant acid phosphatase for osteoclasts).
  • assays include, but are not limited to, those that assess the surface expression of characteristic blood cell lineage specific proteins (such as CD45 for B lineage lymphocytes, T cell receptor for T lineage lymphocytes, Mac-I/LFA11 for macrophages, tartrate resistant acid phosphatase for osteoclasts).
  • Proliferation of hematopoietic stem cells will be assessed in vitro by evidence that co-culture derived hematopoietic cells can continue to expand to multiple blood cell lineages when plated onto a fresh hematopoietic supporting stromal cell layer and in vivo based on the ability of co-culture derived hematopoietic cells to repopulate the bone marrow and rescue a lethally irradiated animal host lacking its own blood cells.
  • Myocytes refers to cells that are capable of expressing characteristic biochemical markers of skeletal muscle, including but not limited to the transcription factors myoD and myogenin, skeletal actin, myosin light chain kinase, and myosin heavy chain kinase, characteristic morphologic markers of skeletal muscle, including but not limited to multinucleated complexes and sarcomeres, and able to exhibit contractile function spontaneously or in response to exogenous factors such as acetylcholine.
  • Myocytes smooth muscle, myofibroblasts
  • characteristic biochemical markers of smooth muscle including but not limited to ⁇ -smooth muscle actin, fibronectin, and ⁇ -1 integrin
  • characteristic morphologic markers of smooth muscle including but not limited to the formation of stress fibers in culture, and able to exhibit characteristic smooth muscle functions, including but not limited to the generation of tensile stress on collagen lattices in vitro.
  • Hematopoietic growth factors refers to cytokines, hormones and other protein agents. These may be derived directly from stromal cells in the co-culture system or added to co-cultures at concentrations determined by the investigator and obtained as enriched or purified proteins developed from recombinant or natural sources.
  • cytokines and hormones interleukin 7 for the growth of B lineage lymphocytes; stem cell factor for all hematopoietic lineages; M-CSF for macrophages and osteoclasts; osteoprotegerin ligand for osteoclasts; erythropoietin for erythrocytes; thrombopoietin for platelets and megakaryocytes; interleukin 6 for platelets, megakaryocytes, and B lineage lymphocytes.
  • cytokines and hormones interleukin 7 for the growth of B lineage lymphocytes; stem cell factor for all hematopoietic lineages; M-CSF for macrophages and osteoclasts; osteoprotegerin ligand for osteoclasts; erythropoietin for erythrocytes; thrombopoietin for platelets and megakaryocytes; interleukin 6 for platelets, megakaryocytes, and B lineage lymphocytes.
  • Non-peptide growth factors refers to steroids, retinoids and other chemical compounds or agents which induce the differentiation of blood cell lineages. It is generally recognized that concentrations may vary. Moreover, it is generally recognized that the compounds or agents will be added in amounts sufficient to stimulate differentiation. Generally, however, these will be used at concentrations ranging from about 1 nM to about 100 nM for 1,25 dihydroxy vitamin D 3 , about 1 nM to about 100 nM dexamethasone, about 1 nM to about 100 nM hydrocortisone, about 1 nM to about 100 nM retinoic acid, about 1 nM to about 100 nM 9-cis retinoic acid, or at concentrations to be determined and optimized by the practitioner.
  • Amounts of 5′ azacytadine and/or amphotericin sufficient to induce differentiation refers to concentrations of 5′ azacytadine and amphotericin, that when supplied in a medium capable of supporting the growth of stromal cells (e.g. NIH-3T3, C3H 10T1/2, human adipose tissue-derived stromal cells and the like), will induce the differentiation of said stromal cells into skeletal muscle myoblasts and myocytes over a period of about 1 to 6 weeks.
  • Typical use concentrations for 5′azacytadine range from about 1 ⁇ M to about 30 ⁇ M.
  • Typical use concentrations for amphotericin range from about 10 ng/ml to about 100 ng/ml.
  • Optimal concentrations and length of exposure may be determined by the practitioner through the use of known assays for the differentiation of skeletal muscle myoblasts.
  • assays include, but are not limited to, those that assess the morphological or biochemical characteristics associated with skeletal muscle (e.g., formation of multinucleated myotubules, expression of myosin heavy chain, expression of myoD at the protein or RNA level).
  • “Amounts of transforming growth factor ⁇ or other peptide growth factors sufficient to induce differentiation” refers to concentrations of transforming growth factor ⁇ or other peptides, that when supplied to medium capable of supporting the growth of stromal cells (e.g. NIH-3T3, C3H 10T1/2, human adipose tissue-derived stromal cells and the like), will induce the differentiation of said stromal cells into smooth muscle myocytes or myofibroblasts over a period of 1 day to 6 weeks.
  • Typical use concentrations for transforming growth factor ⁇ range from about 20 ng/ml to about 40 ng/ml.
  • Typical use concentrations for fibroblast growth factor range from about 20 ng/ml to about 40 ng/ml.
  • Optimal concentrations and length of exposure may be determined by the practitioner through the use of known assays for the differentiation of smooth muscle myoblasts.
  • assays include, but are not limited to, those that assess the morphological or biochemical characteristics associated with smooth muscle (e.g., expression of a smooth muscle actin and fibronectin, generation of contractile forces when placed in collagen lattices in the presence of thrombin or lysophosphatidic acid).
  • ⁇ MEM alpha modified Minimal Essential Medium
  • RPMI Media 1640 Roswell Park Memorial Institute Media 1640
  • FBS Fetal Bovine Serum
  • a defined medium could be used if the necessary growth factors, cytokines, and hormones in FCS for stromal cell and hematopoietic cell growth are identified and provided at appropriate concentrations in the growth medium.
  • Media useful in the methods of invention may contain one or more compounds of interest, including, but not limited to, antibiotics, compounds that are mitogenic for hematopoietic; stem cells, differentiation inducing for hematopoietic stem cells, and/or mitogenic or differentiative for stromal cells.
  • antibiotics useful in the invention include but are not limited to penicillin and streptomycin. Penicillin is typically used at about 10 units/ml to about 200 units/ml. Streptomycin is typically used at about 10 ⁇ g/ml to 200 ⁇ g/ml.
  • hematopoietic mitogenic factors include but are not limited to stem cell factor and interleukin 3; hematopoietic differentiation inducing factors include but are not limited to 1,25 dihydroxy vitamin D 3 , interleukin 7, and osteoprotegerin ligand; stromal cell mitogens include but are not limited to transforming growth factor ⁇ ; and stromal cell differentiating factors include but are not limited to dexamethasone, hydrocortisone, transforming growth factor ⁇ , and the like.
  • “Adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and growth factors and agents capable of inducing stromal cell expression of hematopoietic growth factors or exogenous hematopoietic growth factors or non-peptide factors themselves” refers to growth factors, both peptide and chemical in composition, which enhance the proliferation and maturation of hematopoietic stem cells in vitro and in vivo. These include, but are not limited to, interleukin 1, interleukin 3, interleukin 6, interleukin.
  • interleukin 11 macrophage colony stimulating factor (M-CSF), granulocyte-monocyte colony stimulating factor (GM-CSF), stem cell factor, flt3 ligand, thrombopoietin, erythropoietin, osteoprotegerin ligand, dexamethasone, hydrocortisone, and 1,25 dihydroxy vitamin D 3 .
  • M-CSF macrophage colony stimulating factor
  • GM-CSF granulocyte-monocyte colony stimulating factor
  • stem cell factor flt3 ligand, thrombopoietin, erythropoietin, osteoprotegerin ligand, dexamethasone, hydrocortisone, and 1,25 dihydroxy vitamin D 3 .
  • the concentrations of these factors and the length of time of exposure will be determined and optimized by the investigators.
  • Interleukins, M-CSF, GM-CSF, Flt 3 ligand and stem cell factor are used at about
  • Thrombopoietin is typically used at concentrations ranging from about 5 pg/ml to about 1 ng/ml. Erythropoietin is used at about 5 units/ml to about 1000 units/ml. Osteoprotegerin ligand is used at about 5 pg/ml to about 1 ng/ml. Dexamethasone, hydrocortisone, and 1,25 dihydroxy vitamin D 3 are at concentrations from about 1 nM to about 100 nM. Optimal concentrations and treatment times will be determined by monitoring the production of specific circulating blood cell lineages in the co-cultures of hematopoietic stem cells and adipose tissue-derived stromal cells.
  • Such assays and indices include, but are not limited to, those that assess the morphological or biochemical characteristics of the cells, such as the expression of cell surface proteins unique to specific blood cell lineages by flow cytometry, immunohistochemistry, and/or immunofluorescent methods, expression of specific mRNAs in the cell population, or by in vivo assessment of the production of hematopoietic stem cells by the co-culture system.
  • “Adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and amounts of azacytadine and/or amphotericin or other agents sufficient to induce the differentiation of said stromal cells into skeletal muscle myocytes” refers to the differentiation inducing agents used to promote expression of skeletal muscle specific gene markers and skeletal muscle function in vitro.
  • the medium comprises fetal bovine serum, antibiotic, L-glutamine, sodium pyruvate, 2-mercaptoethanol, and 5′ azacytadine or amphotericin.
  • Fetal bovine serum can be used at concentrations ranging from 0.5% to 20%.
  • the antibiotic typically used, but not limited to, is penicillin or streptomycin.
  • Penicillin is typically used at concentrations ranging from about 10 units/ml to about 200 units/ml. Streptomycin is typically used in concentrations ranging from about 10 ⁇ g/ml to about 200 ⁇ g/ml. L-glutamine and sodium pyruvate are typically used at 0.5 mM to about 2 mM. 2-mercaptoethanol is typically used at about 10 ⁇ M to about 100 ⁇ M. 5′ azacytadine is typically used at about 1 ⁇ M to about 30 ⁇ M. Amphotericin is typically used at about 10 ng/ml to about 100 ng/ml. The concentrations of these factors and the length of time of exposure will be determined and optimized by the investigators.
  • Optimal concentration and treatment times will be determined by monitoring the morphologic and biochemical markers characteristic of skeletal muscle. These include, but are not limited to, the production of multi-nucleated myotubules in culture and the expression of muscle specific genes and proteins, such as muscle transcription factors (myoD, myogenin), myosin light chain kinase, myosin heavy chain kinase, and skeletal muscle actin.
  • muscle transcription factors myoD, myogenin
  • myosin light chain kinase myosin heavy chain kinase
  • skeletal muscle actin skeletal muscle actin
  • “Adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and amounts of transforming growth factor ⁇ or other peptide growth factors sufficient to induce the differentiation of said stromal cells into smooth muscle myocytes or myofibroblasts” refers to the differentiation inducing conditions used to promote the expression of smooth muscle associated gene markers and proteins and smooth muscle function in vitro. The concentrations of factors and the length of time of exposure will be determined and optimized by the investigators. Optimal concentration and treatment times will be determined by monitoring the morphologic and biochemical markers characteristic of smooth muscle cells. These include, but are not limited to, the generation of tensile forces by the cells when placed in a collagen type I lattice and the expression of smooth muscle specific genes and proteins, such as smooth muscle actin, fibronectin, and laminin.
  • the adipose tissue derived stromal cells are isolated from the adipose tissue of the subject into which the final differentiated cells are to be introduced.
  • the stromal cells may also be isolated from any organism of the same or different species as the subject. Any organism with adipose tissue can be a potential candidate.
  • the organism is mammalian, most preferably the organism is human.
  • the adipose tissue derived stromal cells may be stably or transiently transfected or transduced with a nucleic acid of interest using a plasmid, viral or alternative vector strategy.
  • Nucleic acids of interest include, but are not limited to, those encoding gene products which; (1) enhance the growth, differentiation, maturation and proliferation of hematopoietic cell lineages; examples include osteoprotegerin ligand which induces osteoclast development, interleukin 7, which induces B lineage lymphocyte development, erythropoietin, which induces erythrocyte development, and thrombopoietin, which induces platelet development; (2) enhance the differentiation of skeletal muscle; examples include myoD and myogenin, transcription factors which promote myotubule formation and expression of skeletal muscle specific genes; (3) enhance the growth, differentiation and maturation of smooth muscle cells; examples include transforming growth factor ⁇ , which induces smooth muscle proliferation and extracellular matrix production.
  • the blood cells produced by in vitro co-cultures of hematopoietic stem cells and adipose tissue derived stromal cells can be introduced alone or in combination with the stromal component into subjects subject to anemia or limited blood cell production. These may include, but are not limited to, patients receiving high dose chemotherapy, patients undergoing bone marrow transplantation, patients suffering from aplastic anemia, patients suffering from sickle cell anemia, and other blood dyscrasias.
  • disorders which may be treated with infusion of stem cells include, but are not limited to, diseases resulting from a failure or a dysfunction of normal blood cell production and maturation (i.e., aplastic anemia and hypoproliferative stem cell disorders); neoplastic, malignant diseases in the hematopoietic organs (e.g., leukemia and lymphomas); broad spectrum malignant solid tumors of non-hematopoietic origin; autoimmune conditions; and genetic disorders.
  • Such disorders include, but are not limited to diseases resulting from a failure or dysfunction of normal blood cell production and maturation hyperproliferative stem cell disorders, including aplastic anemia, pancytopenia, agranulocytosis, thrombocytopenia, red cell aplasia, Blackfan-Diamond syndrome, due to drugs, radiation, or infection, idiopathic; hematopoietic malignancies including acute lymphoblastic (lymphocytic) leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, acute malignant myelosclerosis, multiple myeloma, polycythemia vera, agnogenic myelometaplasia, Waldenstrom's macroglobulinemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma; immunosuppression in patients with malignant, solid tumors including malignant melanoma, carcinoma of the stomach, ovarian carcinoma, breast
  • the skeletal muscle cells produced by in vitro manipulation of the adipose tissue derived stromal cells can be introduced alone or in combination with a composition matrix to repair muscle defects secondary to metabolic diseases (muscular dystrophy, myositis), trauma, and disuse atrophy.
  • compositions include, but are not limited to, collagen matrices, poly-lactic polymers, poly-glycolic polymers, alginate, or other solid supports.
  • the smooth muscle cells produced by in vitro manipulation of the adipose tissue derived stromal cells can be introduced alone or in combination with a composition matrix to repair smooth muscle defects.
  • These defects may include, but are not limited to, urinary bladder wall abnormalities due to hereditary malformations in neonates or secondary to trauma or tumor invasion in older individuals, gastrointestinal tract abnormalities due to hereditary malformations in neonates or secondary to trauma or tumor invasion in older individuals, genital tract abnormalities (vaginal) due to hereditary malformations in neonates, secondary to trauma or tumor invasion, or for tissue reconstructive surgeries in transgender operations, or for the development of functional large veins for grafting purposes.
  • Composition matrices may include, but are not limited to, collagen matrices such as swine intestinal submucosa, poly-lactic polymers, poly-glycolic polymers, alginate, or other solid supports.
  • Another object of the invention is to provide methods for the identification and study of compounds that enhance or inhibit the differentiation of adipose tissue derived stromal cells into either hematopoietic supporting stromal cells, skeletal muscle myocytes, or smooth muscle myocytes. Compounds which enhance differentiation.
  • hematopoietic supporting stromal cell function may be of value in the treatment of blood dyscrasias characterized by decreased production of circulating blood cells and improve recovery of patients following high dose chemotherapy;
  • skeletal muscle myocytes may be of value in the treatment of musculoskeletal diseases secondary to hereditary defects or trauma; or
  • smooth muscle myocytes may be of value in the treatment of smooth muscle defects, including those of the urinary bladder (bladder wall), gastrointestinal tract (colon, small intestine), and genital system (vaginal).
  • compounds which inhibit differentiation of may be of value in the treatment of blood dyscrasias characterized by overproduction of circulating blood cells, such as polycythemia vera;
  • skeletal muscle may be of value in the treatment of soft tissue tumors of skeletal muscle origin, such as rhabdomyosarcomas;
  • smooth muscle may be of value in the treatment of soft tissue tumors of smooth muscle origin, such as leiomyosarcomas.
  • Any compound may be tested for its ability to affect the differentiation of adipose tissue derived stromal cells into either hematopoietic supporting stromal cells, skeletal muscle myocytes, or smooth muscle myocytes.
  • Appropriate vehicles compatible with the compound to be tested are known to those skilled in the art and may be found in the current edition of Remington's Pharmaceutical Sciences, the contents of which are incorporated herein by reference.
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 30,000 cells per cm 2 in chamber slides, in 6 well tissue culture plates, or in T25 cm 2 flasks. Cells are maintained in culture for 8 days in DMEM/Ham's F-10 supplemented with 10% fetal bovine serum, penicillin 100 units/ml, streptomycin 100 ⁇ g/ml, and 7.5 mM HEPES pH 7.2.
  • the surface proteins expressed by the stromal cells are determined by immunologic techniques based on immunohistochemistry and/or flow cytometry.
  • immunohistochemical analysis chamber slides are fixed using 95% ethanol/5% glacial acetic acid and incubated with murine monoclonal antibodies detecting human cell surface proteins. After incubation with an enzyme coupled anti-mouse secondary antibody, evidence of protein expression is detected by histochemical reaction.
  • flasks of cells are harvested by trypsin/EDTA digestion and incubated with a fluorescent conjugated murine monoclonal antibody detecting a specific human surface protein. Cells are examined for fluorescent intensity by flow cytometry. The results of these assays are summarized in Table 1.
  • the cytokine expression profile of the adipose-derived stromal cells is determined following induction with lipopolysaccharide (LPS) or endotoxin, an inflammatory agent capable of inducing hematopoietic cytokines in bone marrow stromal cells (Gimble et al. (1989) Blood 74:303-311).
  • LPS lipopolysaccharide
  • endotoxin an inflammatory agent capable of inducing hematopoietic cytokines in bone marrow stromal cells.
  • Confluent and quiescent cultures of cells are exposed to 100 ng/ml LPS for periods of 0 to 24 hours in DMEM medium supplemented with 2% fetal bovine serum, 100 ⁇ g/ml streptomycin, 100 units/ml penicillin, and 7.5 mM HEPES pH 7.2.
  • the conditioned medium from each culture is harvested and stored at ⁇ 80° C.
  • RNA for the cytokines indicated in Table 2 is detected by polymerase chain reaction using the oligonucleotide primer sets listed below the table. A representative set of reactions is demonstrated in FIG. 1 .
  • M-CSF macrophage colony stimulating factor
  • GM-CSF granulocyte/monocyte colony stimulating factor
  • IL-6, 7, 8 interleukin 6, 7, and 8
  • the profile of cytokines expressed by the adipose derived stromal cells is consistent that of bone marrow-derived stromal cells capable of supporting myeloid, lymphoid, and osteoclast proliferation and differentiation in vitro (Pietrangeli et al. (1988) Eur. J. Immunol. 18:863-872; Gimble et al. (1989) Blood 74:303-311; Gimble et al. (1992) J. Cell Biochem 50:73-82; Kelly et al. (1998) Endocrinol. 139:2092-2101).
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm 2 . Stromal cells are established in the cultures for 1 to 3 days prior to the introduction of hematopoietic progenitor cells into the co-culture system. Hematopoietic progenitor cells are isolated from one of the following human tissues: bone marrow, umbilical vein/placental blood, peripheral blood, spleen. Alternatively, murine tissues are used.
  • Murine bone marrow cells are harvested by flushing the marrow cavity of 6 to 10 week old mice with DMEM/10% FCS under sterile conditions.
  • Murine spleen cells are harvested by physical passage through a fine metal screen under sterile conditions.
  • One of three methods are used to deplete the mixed hematopoietic cell population of its stromal component.
  • the hematopoietic stem cell population from the blood sample will be enriched by magnetic immunobead purification using anti-CD34 antigen according to established techniques.
  • hematopoietic cells will be enriched by passage of the bone marrow or other blood sample over a sterile G-10 Sephadex or nylon wool column; hematopoietic progenitor cells are eluted while stromal cells are retained.
  • hematopoietic cells will be enriched by flow cytometric sorting based on surface protein characteristics. The hematopoietic cells are washed once and the number of nucleated cells is counted using a hematocytometer after erythrocyte lysis with 0.3% acetic acid and trypan blue staining.
  • Hematopoietic cells are introduced into the liquid co-cultures at a ratio of preferably 10 to 100 nucleated hematopoietic cells per stromal cell plated, more preferably 20 to 30 nucleated hematopoietic cells per stromal cell plated, most preferably 25 to 30 nucleated hematopoietic cells per stromal cell plated.
  • Cells are cultured in a medium consisting of DMEM (high glucose), 10% fetal bovine serum supplemented with 10 to 100 nM hydrocortisone or 10% horse serum, 10 to 200 units/ml of penicillin, 10 to 200 ⁇ g of streptomycin at 33° C. in 5% CO 2 .
  • the number of non-adherent cells in the medium is determined by hematocytometer count and/or by flow cytometry.
  • the surface antigen characteristics of the non-adherent cells are documented using routine antibody markers for the major hematopoietic cell lineages. These will include, but are not limited to, Mac-1, Thy 1, Ig Heavy chain, Ter-81 (erythroid marker).
  • the number and character of the non-adherent cell population is determined over a period of up to 10 weeks.
  • the cellular composition of the adherent cell layer is determined by flow cytometric or immunohistochemical methods.
  • the hematopoietic studies are conducted in semi-solid cultures.
  • Cells are prepared as described above, but the hematopoietic progenitor cells are plated in the additional presence of 2.1% methylcellulose. Colony formation is assessed after a 7 to 14 day period for the presence of granulocytes, erythrocytes, macrophages, and monocytes using histologic, morphologic and immunologic criteria.
  • Adipose tissue derived stromal cell/hematopoietic progenitor co-cultures established under liquid culture conditions described in Example 1 are used to assess the ability of this system to maintain the proliferation of the hematopoietic progenitor cells in vitro.
  • Co-cultures are established using human adipose tissue derived stromal cells and murine hematopoietic progenitors. Cultured cells are transduced with a viral vector expressing a traceable protein marker such as green fluorescent protein or beta-galactosidase.
  • co-culture cells are identified by expression of a unique antigen or genetic marker due to their origin; e.g., the expression of human proteins for the stromal cells, and the expression of a transgenic or male gender specific marker for the murine hematopoietic cells.
  • Established co-cultures are harvested by limited incubation with trypsin/EDTA and infused into a lethally irradiated immunodeficient mouse. Animals will be followed over time. After 9 to 14 days, mice are sacrificed and their spleens examined for the appearance of hematopoietic cell islands or splenic colony forming units (CFU-S).
  • mice will be maintained for 14 days or longer and their circulating blood cell count determined by hematologic and flow cytometric assays.
  • the presence of specific markers of the stromal cells and the donor hematopoietic cells is detected with antibody reagents or specific DNA markers on fixed cells, either by flow cytometry or conventional pathologic/histologic methods.
  • the ability of the co-cultured cells to establish CFU-S in the recipient and/or to maintain the proliferation and maturation of donor blood cells in the host after 14 days is evidence of the continued expansion of some hematopoietic progenitors in vitro by the adipose tissue-derived stromal cells.
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm 2 . Stromal cells are established in the cultures for 1 to 3 days prior to the introduction of hematopoietic progenitor cells into the co-culture system. Hematopoietic progenitor cells are isolated from one of the following human tissues: bone marrow, umbilical vein/placental blood, peripheral blood, spleen. Alternatively, murine tissues are used.
  • Murine bone marrow cells are harvested by flushing the marrow cavity of 6 to 10 week old mice with RPMI/10% FCS under sterile conditions.
  • Murine spleen cells are harvested by physical passage through a fine metal screen under sterile conditions.
  • One of three methods are used to deplete the mixed hematopoietic cell population of its stromal component, As a first alternative, the hematopoietic stem cell population from the blood sample will be enriched by magnetic immunobead purification using anti-CD34 antigen according to established techniques.
  • hematopoietic cells will be enriched by passage of the bone marrow or other blood sample over a sterile G-10 Sephadex or nylon wool column; hematopoietic progenitor cells are eluted while stromal. cells are retained.
  • hematopoietic cells will be enriched by flow cytometric sorting. The hematopoietic cells are washed once and the number of nucleated cells is counted using a hematocytometer after erythrocyte lysis with 0.3% acetic acid and trypan blue staining.
  • Hematopoietic cells are introduced into the liquid co-cultures at a ratio of preferably 10 to 100 nucleated hematopoietic cells per stromal cell plated, more preferably 20 to 30 nucleated hematopoietic cells per stromal cell plated, most preferably 25 to 30 nucleated hematopoietic cells per stromal cell plated.
  • Cells are cultured in a medium consisting of RPMI 1640, prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 ⁇ g/ml of streptomycin, 0.5 to 2 mM L-glutamine, 10 to 100 ⁇ M 2-mercaptoethanol at 37° C. in 5% CO 2 .
  • the number of non-adherent cells in the medium is determined by hematocytometer count and/or by flow cytometry.
  • the surface antigen characteristics of the non-adherent cells are documented using routine antibody markers for the major hematopoietic cell lineages. These will include, but are not limited to, Mac-1, Thy 1, Ig Heavy chain, Ter-81 (erythroid marker).
  • the number and character of the non-adherent cell population is determined over a period of up to 10 weeks.
  • the cellular composition of the adherent cell layer is determined by flow cytometric or immunohistochemical methods.
  • the hematopoietic studies are conducted in semi-solid cultures.
  • Cells are prepared as described above but the hematopoietic progenitor cells are plated in the additional presence of 2.1% methylcellulose. Colony formation is assessed after a 7 to 14 day period for the presence of B lineage lymphoid cells as well as granulocytes, erythrocytes, macrophages, and monocytes using histologic, morphologic and immunologic criteria.
  • co-cultures and/or semi-solid cultures are established as described above with the addition of interleukin 7 at concentrations to be determined by the practitioner which enhance the proliferation and maturation of B lineage lymphocytes. The techniques outlined above are used to assess the affect of this growth factor on the hematopoietic support function of the adipose tissue derived stromal cell.
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, Filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm 2 in 24 well plates. Cells are cultured in a medium consisting of DMEM (high glucose), prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 ⁇ g of streptomycin, 0.5 to 2 mM L-glutamine, 0.5 to 2 mM sodium pyruvate, 10 to 100 ⁇ M 2-mercaptoethanol at 37° C. in 5% CO 2 .
  • DMEM high glucose
  • penicillin 10 to 200 ⁇ g
  • streptomycin 0.5 to 2 mM L-glutamine
  • 0.5 to 2 mM sodium pyruvate 10 to 100 ⁇ M 2-mercaptoethanol at
  • the medium is supplemented with either 10 to 100 nM 1,25 dihydroxy vitamin D 3 and or osteoprotegerin ligand at concentrations determined by the practitioner.
  • Stromal cells are established in the cultures for 6 days prior to the introduction of hematopoietic progenitor cells into the co-culture system.
  • Hematopoietic progenitor cells are isolated from one of the following human tissues: bone marrow, umbilical vein/placental blood, peripheral blood, spleen.
  • murine tissues are used.
  • Murine bone marrow cells are harvested by flushing the marrow cavity of 6 to 10 week old mice with DMEM (high glucose)/10% FCS under sterile conditions.
  • Murine spleen cells are harvested by physical passage through a fine metal screen under sterile conditions.
  • One of three methods are used to deplete the mixed hematopoietic cell population of its stromal component.
  • the hematopoietic stem cell population from the blood sample will be enriched by magnetic immunobead purification using anti-CD34 antigen according to established techniques.
  • hematopoietic cells will be enriched by passage of the bone marrow or other blood sample over a sterile G-10 Sephadex or nylon wool column; hematopoietic progenitor cells are eluted while stromal cells are retained.
  • hematopoietic cells will be enriched by flow cytometric sorting based on surface protein characteristics. The hematopoietic cells are washed once and the number of nucleated cells is counted using a hematocytometer after erythrocyte lysis with 0.3% acetic acid and trypan blue staining.
  • Hematopoietic cells are introduced into the liquid co-cultures at a ratio of preferably 10 to 100 nucleated hematopoietic cells per stromal cell plated, more preferably 20 to 30 nucleated hematopoietic cells per stromal cell plated, most preferably 25 to 30 nucleated hematopoietic cells per stromal cell plated.
  • One half of the medium in the co-cultures is replaced every 3 to 4 days.
  • Co-cultures are maintained in the presence of 1,25 dihydroxy vitamin D 3 or osteoprotegerin ligand continuously after the introduction of hematopoietic cells.
  • co-cultures are fixed with 0.5 ml 3.7% (vol:vol) formaldehyde in phosphate buffered saline for 5 minutes, dried for 30 seconds with acetone:ethanol (50:50, vol/vol), and stained for 10 minutes with 10 mM sodium tartrate, 40 mM sodium acetate (pH 5.0), 0.1 mg/ml naphthol AS-MS phosphate (Sigma N-5000), and 0.6 mg/ml fast red violet LB salt (Sigma F-3381). Stained cultures are rinsed in distilled water and stored under 50% glycerol/PBS.
  • the number of tartrate resistant acid phosphatase positive cells per well is assessed under light microscopy based on the red staining of the cytoplasm.
  • TRAP+ cells are, numerically counted and those with 1-2 nuclei are distinguished from those multinucleated cells with >3 nuclei per cell.
  • This assay demonstrates the ability of adipose tissue derived stromal cells to support the differentiation and proliferation of osteoclastogenic precursors in vitro. This culture procedure is able to expand and promote differentiation of osteoclasts. This has potential application to rare clinical conditions such as osteopetrosis characterized by brittle bones where patients fail to produce native osteoclasts.
  • This In vitro method offers a means to expand an individual's own osteoclast progenitors and to promote their differentiation ex vivo.
  • This cell population can be re-infused into the affected individual with potential short-term or long-term benefit.
  • the non-invasive nature of the methodology and the potential to rely exclusively on autologous cells indicates that this procedure could be used repetitively in the treatment of an individual patient.
  • the co-culture models outlined in Examples 1-4 have the potential to be used to facilitate the recovery of bone marrow function in patients receiving high dose chemotherapy, high dose radiation treatment or any other therapeutic modality which compromises blood cell production and bone marrow function.
  • an individual can donate his or her own adipose tissue and blood cells for subsequent autologous transplantation.
  • the individual's own blood cells and stromal co-cultures can be established and expanded.
  • the patients own blood cell/stromal cell co-culture can be re-infused into the patient according to standard transfusion methodologies. This can be done in the absence or presence of exogenous hematopoietic cytokines, either added to the co-cultures or given directly to the patient.
  • This approach may accelerate the rate of blood cell production in the patient, reduce the need for cytokine therapies, and reduce the overall costs and risk of the chemotherapy or other immunocompromising procedure.
  • it is possible to manipulate the stromal cells to enhance production of specific blood cell lineages.
  • Stromal cells transiently expressing interleukin 7, for example, would facilitate the rapid expansion of B lineage lymphoid cells while stromal cells expressing erythropoietin would favor expansion of erythrocytes.
  • the approach is designed primarily for the treatment of nosocomial induced blood cell dyscrasias.
  • large scale ex vivo production of autologous stromal/hematopoietic co-cultures is of potential value for elective and non-elective surgical procedures requiring transfusion intra-operatively and post-operatively.
  • the majority of patients requiring blood transfusion receive blood products donated by others. This presents risk to the recipient of transmission of unrecognized infectious disease from the donor.
  • an individual can expand his/her hematopoietic cell population at a capacity which is no longer limited by the bone marrow cavity volume.
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm 2 in 24 well plates. Cells are cultured in a medium consisting of DMEM (high glucose), prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 ⁇ g of streptomycin, 0.5 to 2 mM L-glutamine, 0.5 to 2 mM sodium pyruvate, 10 to 100 ⁇ M 2-mercaptoethanol at 37° C. in 5% CO 2 .
  • DMEM high glucose
  • penicillin 10 to 200 ⁇ g
  • streptomycin 0.5 to 2 mM L-glutamine
  • 0.5 to 2 mM sodium pyruvate 10 to 100 ⁇ M 2-mercaptoethanol at 37
  • Cells are exposed to 1 to 30 ⁇ M 5′ azacytadine or 10 to 100 ng/ml of amphotericin for periods of 1 to 6 days to assure that cells throughout S phase are continuously exposed to these agents. Following this period, cultures are maintained in the culture medium without azacytadine or amphotericin supplements. Cultures are either continued at the established cell density or sub-cloned by limit dilution methods to select for cell clones capable of expressing characteristic markers of skeletal muscle myoblasts.
  • These cells are selected based on morphologic criteria, specifically, the ability to form multinucleated myotubules in culture; biochemical criteria, specifically, the expression of myosin heavy and light chain kinase, skeletal muscle actin and myosin and the expression of myogenic transcription factors, myoD and/or myogenin.
  • the cells skeletal muscle myoblasts developed in Example 6 can be used for tissue engineering purposes in the treatment of myodystrophies, muscle atrophy, and physical loss of skeletal muscle secondary to surgical procedures for the treatment of cancer or trauma,
  • the ex vivo development of a proliferating population of myoblasts from adipose tissue can be used to supplement and repair skeletal muscle mass in afflicted individuals.
  • Myoblasts can be cultured in biodegradable matrices composed of poly-lactic, poly-glycolic, collagen or other materials to form muscle strands. These can then be implanted to an afflicted site and tethered by suture to existing muscle, tendon or bone.
  • ex vivo expanded myoblasts can be genetically engineered by viral transduction, plasmid transfection, or other means to express novel genes. These cells can be injected directly into existing muscle sites where these novel gene products will now be expressed. This approach has direct application to muscular dystrophy, where patients suffer secondary to a mutation in an important skeletal muscle gene.
  • the engineered stromal cells can convert the muscle into a production site for a deficient circulating protein.
  • adipose tissue derived stromal cells expressing lipoprotein lipase can be used to treat the many patients with mutations in their native lipoprotein lipase gene who are at increased risk of severe cardiovascular disease.
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm 2 in 24 well plates. Cells are cultured in a medium consisting of DMEM (high glucose), prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 ⁇ g of streptomycin, 0.5 to 2 mM L-glutamine, 0.5 to 2 mM sodium pyruvate, at 37° C. in 5% CO 2 .
  • DMEM high glucose
  • penicillin 10 to 200 ⁇ g
  • streptomycin 0.5 to 2 mM L-glutamine
  • 0.5 to 2 mM sodium pyruvate at 37° C. in 5% CO 2 .
  • Cultures are supplemented with transforming growth factor ⁇ and/or fibroblast growth factor at concentrations determined by the practitioner but not to exceed 40 ng/.
  • Cells are maintained in culture as a monolayer or in a 3-dimensional lattice composed of collagen type I or other biodegradable material (alginate, synthetic polymer or other).
  • Cells are characterized based on morphologic, biochemical, and functional criteria for smooth muscle myoblast differentiation; these include, but are not limited to, expression of smooth muscle actin, fibronectin, laminin, and other extracellular matrix proteins, the ability to generate a tensile force as measured by a pressure transducer, and to organize along a line of force in a 3 dimensional lattice.
  • the smooth muscle myoblasts described under Example 8 can be used to treat conditions where smooth muscle function is compromised. For example, over 1000 neonates each year suffer from bladder wall abnormalities. The severity of this disorder is variable but it can be devastating and requires expensive surgical procedures to accomplish an acceptable repair and an approach to normal function. In many cases, the bladder size is too small or the bladder wall is incompletely formed, necessitating the implantation of prosthetic materials as a bladder wall replacement. Methods under investigation include the use of swine intestinal submucosa as a replacement material for the bladder wall. One issue is whether or not the surgically implanted bladder wall will achieve the appropriate physical and mechanical characteristics associated with stretching and retraction. Much of this is mediated by functional smooth muscle cells.

Abstract

The invention relates to methods and compositions for the differentiation of stromal cells from adipose tissue into hematopoietic supporting stromal cells and myocytes of both the skeletal and smooth muscle type. The cells produced by the methods are useful in providing a source of fully differentiated and functional cells for research, transplantation and development of tissue engineering products for the treatment of human diseases and traumatic tissue injury repair.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims the benefit of U.S. Provisional Application No. 60/149,849 filed on Aug. 19, 1999.
  • FIELD OF INVENTION
  • This invention relates to methods and compositions for the differentiation of stromal cells from adipose tissue into hematopoietic supporting stromal cells and myocytes of both the skeletal and smooth muscle types.
  • BACKGROUND OF INVENTION
  • The neonatal period in human development is characterized by the presence of “stem” cells with the potential to develop along multiple differentiation pathways. The terminal differentiation of these cells is determined by cytokine and hormonal cues which co-ordinate organogenesis and tissue architecture. Murine embryonic stem cells have been isolated and studied extensively in vitro and in vivo. Using exogenous stimuli in vitro, investigators have induced ES cell differentiation along multiple lineage pathways. These include neuronal, B lineage lymphoid, and adipocytes (Dani et al. (1997) J. Cell Sci. 110: 1279; Remoncourt et al. (1998) Mech. Dev. 79:185; O'Shea K S (1999) Anat. Rec. 257:32). The ES cells have been manipulated in vivo by homologous recombination techniques to generate gene specific null or “knock-out” mice (Johnson R S (1989) Science 245:1234). Once ES cell clones lacking a specific gene are isolated, they are transplanted into a fertilized murine zygote. The progeny of this isolated ES cell can develop into any and all murine tissues in a coordinated manner.
  • A stem cell must meet the following criteria: (1) ability of a clonal stem cell population to self-renew; (2) ability of a clonal stem cell population to generate a new, terminally differentiated cell type in vitro; and (3) ability of a clonal stem cell population to replace an absent terminally differentiated cell population when transplanted into an animal depleted of its own natural cells.
  • Multipotential stem cells exist in tissues of the adult organism. The best characterized example of a “stem cell” is the hematopoietic progenitor isolated from the bone marrow and peripheral blood. Seminal studies by Trentin, Till and McCulloch (McCulloch et al. (1996) Proc. Can. Cancer Conf. 6:356-366; Curry et al. (1967) J. Exp. Med. 125:703-720) examined lethally irradiated mice. In the absence of treatment, these animals died because they failed to replenish their circulating blood cells; however, transplantation of bone marrow cells from a syngeneic donor animal would rescue the host animal. The donor cells were responsible for reconstituting all of the circulating blood cells. A wealth of elegant studies have gone on to demonstrate that donation of a finite number of undifferentiated hematopoietic stem cells is capable of regenerating each of the eight or more different blood cell lineages in a host. This work has provided the basis for bone marrow transplantation, a widely accepted therapeutic modality for the treatment of cancer and inborn errors of metabolism in man. Thus, hematopoietic stem cells remain present in the normal human bone marrow throughout life; they are not limited to the neonatal period.
  • The recent development of entire organisms from a single donor cell are consistent with this hypothesis. The “Dolly” experiment showed that cells isolated from an ovine mammary gland could develop into a mature sheep (Pennisi & Williams (1997) Science 275:1415-1416). In similar murine studies, cells derived from the corpus luteum of the ovary could develop into a mature mouse (Pennisi (1998) Science 281:495). These studies suggest that stem cells with the ability to differentiate into any and all cell types continue to exist in the adult organism. Thus, “embryonic” stem cells may be retained throughout life.
  • In vitro experiments using cell lines of embryonic origin indicate that a mesodermal stem cell may exist. Work by Taylor and colleagues in the late 1970's demonstrated that murine embryonic fibroblasts such as C3H10T1/2 or 3T3 cells would differentiate along multiple mesodermal lineage pathways following exposure to 1 to 10 μM of 5′-azacytadine (Constantinides et al. (1977) Nature 267:364; Jones & Taylor (1980) Cell 20:85). Within 2 to 4 weeks, isolated clones displayed a morphology consistent with adipocyte, myocyte, chondrocyte or osteoblast differentiation. Biochemical data provided additional support for the identification of each of these lineages. This finding provided the basis for the identification of the master-regulatory transcription factor for skeletal muscle differentiation, myoD (Lassar (1986) Cell 47:649).
  • The adult bone marrow microenvironment is the potential source for these hypothetical mesodermal stem cells. Cells isolated from adult marrow are referred to by a variety of names, including stromal cells, stromal stem cells, mesenchymal stem cells (MSCs), mesenchymal fibroblasts, reticular-endothelial cells, and Westen-Bainton cells (Gimble et al. (1996) Bone 19:421-428). In vitro studies have determined that these cells can differentiate along multiple mesodermal or mesenchymal lineage pathways. These include, but are not limited to, adipocytes (fat cells) (Gimble et al. (1990) Eur. J. Immunol 20:379-386; Pittenger et al. (1999) Science 284:143-147; Nuttall et al. (1998) JBMR 13:371-382; Park et al. (1999) Bone 24:549-554), chondrocytes (cartilage forming cells) (Dennis et al. (1999) JBMR 14:700-709), hematopoietic supporting cells (Gimble et al. (1990) Eur. J. Immunol. 20:379-386), myocytes (skeletal muscle) (Phinney (1999) J. Cell. Biochem. 72:570-585), myocytes (smooth muscle) (Remy-Martin et al. (1999) Exp. Hematol. 27:1782-1795), and osteoblasts (bone forming cells) (Beresford (1989) Clin Orthop Res 240:270-280; Owen (1988) J. Cell. Sci. 10:63-76; Dorheim et al. (1993) J. Cell. Physiol. 154:317-328; Haynesworth et al. (1992) Bone 13:81-88, Kuznetsov et al. (1997) JBMR 12:1335-1347). The bone marrow has been proposed as a source of stromal stem cells for the regeneration of bone, cartilage, muscle, adipose tissue, and other mesenchymal derived organs. The major limitations to the use of these cells are the difficulty and risk attendant upon bone marrow biopsy procedures and the accompanying loss of memory B cells and hematopoietic stem cells with present harvesting procedures.
  • Another viable alternative to the use of bone marrow multipotential stem cells is adipose tissue. Adipose stromal cells provide an easily accessible and abundant source of stromal cells which can differentiate along multiple mesenchymal lineages. Methods and compositions are needed for the consistent and quantitative differentiation of adipose derived stromal cells into various cell types including for example hematopoietic stromal cells and skeletal and smooth muscle myocytes.
  • SUMMARY OF INVENTION
  • Compositions and methods for the differentiation of adipocytes are provided. Generally, the present invention provides methods and compositions for consistent and quantitative induction of stromal cells derived from subcutaneous, mammary, gonadal, or omental adipose tissues into the following fully differentiated and functional mesodermal cell lineages: hematopoietic supporting stromal cells, skeletal myocytes, and smooth muscle myocytes (myofibroblasts).
  • The compositions include a variety of chemical components which act as mitogens and differentiation inducing agents for the plated stromal cells and yield production of the desired cell type. The mitogens and inducing agents include, but are not limited to, interleukins, flt-3 ligand, stem cell factor, macrophage-colony stimulating factor, granulocyte-monocyte colony stimulating factor, erythropoietin, thrombopoietin, osteoprotegerin ligand, dexamethasone, hydrocortisone, 1,25 dihydroxy vitamin D3, 2-mercaptoethanol, glutamine, 5′-azacytadine, amphotericin, transforming growth factor β and fibroblast growth factor.
  • The invention provides methods for determining the ability of these compositions to direct the differentiation and function of the adipose-derived stromal cells, for the transduction of viral vectors carrying regulatory genes into stromal cells, for the transfection of plasmid vectors carrying regulatory genes into stromal cells, for the tracking and detection of functional proteins encoded by these genes, and for developing biomechanical carriers for the re-introduction of these cells into living organisms.
  • The invention also provides methods and compositions which have utility in drug discovery for compounds and proteins with relevance to a wide spectrum of disease states including, but not limited to, aplastic anemia, muscular dystrophy, radiation poisoning, neuropathic muscular degeneration, urogenital malformations, and gastrointestinal malformations.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Human adipose-derived stromal cells were cultured in 6 well plates until confluent and quiescent in DMEM/F 10 (1:1 vol:vol), 10% fetal bovine serum, penicillin 100 units/ml, streptomycin 100 μg/ml, and 7.5 mM HEPES pH 7.2. The cells were then incubated in DMEM/F 10 (1:1 vol:vol), 2% fetal bovine serum, penicillin 100 units/ml, streptomycin 100 μg/ml, and 7.5 mM HEPES pH 7.2 containing 100 ng/ml lipopolysaccharide (LPS). Cells were harvested immediately (time “0”) or after 4 hours (time “4”) for total RNA extraction and isolation using a phenol/chloroform/acid procedure (Chomczynski & Sacchi (1987) Analytical Biochem 162:156-159). Equal aliquots of total RNA were reverse transcribed and amplified by polymerase chain reaction with the following primer sets specific for the indicated human mRNAs; the actin primers served as a positive control for equal loading between samples:
  • Actin Forward 5′ AGTAACAGCCCACGGTGTTC 3′
    Reverse 5′ AGCCTCCGAAAGGAAATTGT 3′
    Interleukin 6 (IL-6) Forward 5′ GTAGCCGCCCCACACAGACAGCC 3′
    Reverse 5′ GCCATCTTTGGAAGGTTCAGG 3′
    Interleukin 8 (JL-8) Forward 5′ TCTGCAGCTCTGTGTGAAGGT 3′
    Reverse 5′ TGAATTCTCAGCCCTCTTCAA 3′
    Granulocyte Colony Forward 5′ AGCTTCCTGCTCAAGTGCTTAGA 3′
    Stimulating Factor (G-CSF) Reverse 5′ TTCTTCCATCTGCTGCCAGATGGT 3′
    Macrophage Colony Forward 5′ TTGGGAGTGGACACCTGCAGTCT 3′
    Stimulating Factor (M-CSF) Reverse 5′ CCTTGGTGAAGCAGCTCTTCAGCC 3′
    Granulocyte/Monocyte Colony Forward 5′ GTCTCCTGAACCTGAGTAGAGACA 3′
    Stimulating Factor (GM-CSF) Reverse 5′ AAGGGGATGACAAGCAGAAAGTCC 3′
    Flt3 Ligand Forward 5′ TGGAGCCCAACAACCTATCTC 3′
    Reverse 5′ GGGCTGAAAGGCACATTTGGT 3′
    Leukemia Inhibitory Forward 5′ AACAACCTCATGAACCAGATCAGGAGC 3′
    Factor (LIF) Reverse 5′ ATCCTTACCCGAGGTGTCAGGGCCGTAGG 3′
  • The resulting PCR products were electrophoresed on a 2% agarose gel, stained with ethidium bromide and photographed.
  • Table 1. Characterization of Adipose Derived Stromal Cell Surface Markers Based on Antibody and PCR Detection. The listed cell surface proteins and genes analyzed in human adipose derived stromal cells is based on immunohistochemical staining, flow cytometry, and/or by polymerase chain reaction. Markers are divided among those expressed (listed as “positive”) and not expressed (listed as “negative”).
    Table 2. Cytokines Expressed by Adipose-Derived Stromal Cells Constitutively or Following Endotoxin (LPS) Induction. The listed cytokines were analyzed in total RNA isolated from human adipose derived stromal cells following induction with 100 ng/ml of lipopolysaccharide. The cytokines listed in the table were detected using the oligonucleotide primers listed. All cytokines were expressed either in a constitutive or inducible manner.
    Table 3. Quantitative ELISA (pg/ml) LPS Induction of Adipose-Derived Stromal Cell Secreted Cytokines. The listed cytokines were assayed in conditioned medium from human adipose derived stromal cells induced for 0 to 24 hours with 100 ng/ml of LPS. All cytokines were detected by enzyme linked immunoassay (ELISA) and are expressed as pg/ml of conditioned medium. Those cytokines indicated by an “*” demonstrated significant increases relative to the 0 hour time point within the 24 hour induction period based on one way analysis of variance.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides methods and compositions for the differentiation and culture of adipose tissue-derived stromal cells into (a) hematopoietic supporting stromal cells, (b) skeletal myocytes, and (c) smooth muscle myocytes (myofibroblasts).
  • Adipose tissue offers a potential alternative to the bone marrow as a source of multipotential stromal stem cells. Adipose tissue is readily accessible and abundant in many individuals. Obesity is a condition of epidemic proportions in the United States, where over 50% of adults exceed the recommended BMI based on their height. Adipocytes can be harvested by liposuction on an outpatient basis. This is a relatively non-invasive procedure with cosmetic effects which are acceptable to the vast majority of patients. It is well documented that adipocytes are a replenishable cell population. Even after surgical removal by liposuction or other procedures, it is common to see a recurrence of adipocytes in an individual over time. This suggests that adipose tissue contains stromal stem cells which are capable of self-renewal. Pathologic evidence suggests that adipose-derived stromal cells are capable of differentiation along multiple mesenchymal lineages. The most common soft tissue tumor, liposarcomas, develop from adipocyte-like cells. Soft tissue tumors of mixed origin are relatively common. These may include elements of adipose tissue, muscle (smooth or skeletal), cartilage, and/or bone. Just as bone forming cells within the bone marrow can differentiate into adipocytes or fat cells, the extramedullary adipocytes are capable of forming bone. In patients with a rare condition known as paroxysmal osseous heteroplasia, subcutaneous adipocytes form bone for unknown reasons (Kaplan (1996) Arch. Dermatol. 132:815-818).
  • Adult human extramedullary adipose tissue-derived stromal cells represent a stromal stem cell source which can be harvested routinely with minimal risk to the patient. They can be expanded ex vivo, differentiated along unique mesodermal lineage pathways, genetically engineered, and re-introduced into individuals as either an autologous or allogeneic transplantation. This invention presents examples of methods and composition for the isolation, characterization, and differentiation of adult human extramedullary adipose tissue stromal cells along the mesodermal lineages and outlines their use for the treatment of a number of human conditions and diseases.
  • The cells produced by the methods of invention are useful in providing a source of fully differentiated and functional cells for research, transplantation, and development of tissue engineering products for the treatment of human disease and traumatic injury repair. Thus, in one aspect, the invention provides a method for differentiating adipose tissue-derived stromal cells into one of three functionally distinct mesodermal cell lineages: hematopoietic supporting cells, myocytes (skeletal), and myocytes (smooth muscle myofibroblasts) comprising: culturing said cells in a composition which comprises a medium (a) capable of supporting the growth of stromal cells and hematopoietic cells in co-culture with factors present capable of inducing stromal expression of hematopoietic growth factors or the addition of exogenous growth factors directly; (b) capable of supporting the growth and differentiation of stromal cells into functional and proliferating skeletal myocytes; and (c) capable of supporting the growth and differentiation of stromal cells into functional and proliferating smooth muscle myocytes or myofibroblasts.
  • In another aspect, the invention provides compositions for the differentiation of adipose tissue-derived stromal cells into each of the three different mesodermal derived lineages. Such compositions comprise:
  • (a) adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and growth factors and agents capable of inducing stromal cell expression of hematopoietic growth factors or exogenous hematopoietic growth factors or non-peptide factors themselves.
  • (b) adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and amounts of 5′ azacytadine and/or amphotericin or other agents sufficient to induce the differentiation of said stromal cells into skeletal muscle myocytes.
  • (c) adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and amounts of transforming growth factor β or other peptide growth factors sufficient to induce the differentiation of said stromal cells into smooth muscle myocytes or myofibroblasts.
  • The methods comprise incubation of isolated adipose tissue-derived stromal cells, plated at densities of about 1,000 to 25,000 cells/cm2 in medium consisting of the following for each lineage:
      • (a) Hematopoietic supporting stromal cell—Glucose, hematopoietic inducing cytokines, including but not limited to, interleukins-1, 3, 6, 7, 11, 12, stem cell factor, flt-3 ligand, macrophage colony stimulating factor, granulocyte-monocyte colony stimulating factor, thrombopoietin, erythropoietin, osteoprotegerin ligand, 1,25 dihydroxy vitamin D3, and 2-mercaptoethanol. The medium may also contain hydrocortisone, dexamethasone, and osteoprotegerin ligand. Cells are maintained at temperatures of 33° C. (for myeloid cells) or 37° C. (for B-lineage lymphoid cells).
      • (b) Myocytes, Skeletal—Glucose, 5′-azacytadine or amphotericin for a limited exposure period with manipulation of fetal bovine serum of concentrations between 0% and 20%. The medium may also include, but is not limited to, an antibiotic (as for example penicillin or streptomycin), glutamine, sodium pyruvate, and 2-mercaptoethanol.
      • (c) Myocytes, Smooth Muscle/Myofibroblasts—Glucose and 10% fetal bovine serum in the presence of a collagen, gelatin, laminin, fibronectin or other susbtratum or 3-dimensional matrix.
  • “Adipose stromal cells” refers to stromal cells that originate from adipose tissue. By “adipose” is meant any fat tissue. The adipose tissue may be brown or white adipose tissue, derived from subcutaneous, omental/visceral, mammary, gonadal, or other adipose tissue site. Preferably, the adipose is subcutaneous white adipose tissue. Such cells may comprise a primary cell culture or an immortalized cell line. The adipose tissue may be from any organism having fat tissue. Preferably, the adipose tissue is mammalian, most preferably the adipose tissue is human. A convenient source of adipose tissue is from liposuction surgery, however, the source of adipose tissue or the method of isolation of adipose tissue is not critical to the invention. If stromal cells are desired for autologous transplantation into a subject, the adipose tissue will be isolated from that subject.
  • “Hematopoietic supporting stromal cell” refers to stromal cells that are capable of supporting the proliferation and maturation of hematopoietic progenitor, also known as hematopoietic stem cells, derived from bone marrow, spleen, peripheral blood, or umbilical blood, either CD34+ or CD34−. Co-cultures of hematopoietic supporting stromal cells with hematopoietic progenitors would result in the production of both adherent and non-adherent populations of hematopoietic blood cells, including but not limited to myeloid (macrophage, neutrophil, osteoclast), erythroid (red blood cells), lymphoid (B-lymphoid, T-lymphoid), and platelet (megakaryocyte), as well as eosinophils, basophils, mast cells and other circulating blood cell types. Growth and differentiation of hematopoietic cells will be determined by assays which include, but are not limited to, those that assess the surface expression of characteristic blood cell lineage specific proteins (such as CD45 for B lineage lymphocytes, T cell receptor for T lineage lymphocytes, Mac-I/LFA11 for macrophages, tartrate resistant acid phosphatase for osteoclasts). Proliferation of hematopoietic stem cells will be assessed in vitro by evidence that co-culture derived hematopoietic cells can continue to expand to multiple blood cell lineages when plated onto a fresh hematopoietic supporting stromal cell layer and in vivo based on the ability of co-culture derived hematopoietic cells to repopulate the bone marrow and rescue a lethally irradiated animal host lacking its own blood cells.
  • “Myocytes (skeletal)” refers to cells that are capable of expressing characteristic biochemical markers of skeletal muscle, including but not limited to the transcription factors myoD and myogenin, skeletal actin, myosin light chain kinase, and myosin heavy chain kinase, characteristic morphologic markers of skeletal muscle, including but not limited to multinucleated complexes and sarcomeres, and able to exhibit contractile function spontaneously or in response to exogenous factors such as acetylcholine.
  • “Myocytes (smooth muscle, myofibroblasts)” refers to cells that are capable of expressing characteristic biochemical markers of smooth muscle, including but not limited to α-smooth muscle actin, fibronectin, and β-1 integrin, characteristic morphologic markers of smooth muscle, including but not limited to the formation of stress fibers in culture, and able to exhibit characteristic smooth muscle functions, including but not limited to the generation of tensile stress on collagen lattices in vitro.
  • “Hematopoietic growth factors” refers to cytokines, hormones and other protein agents. These may be derived directly from stromal cells in the co-culture system or added to co-cultures at concentrations determined by the investigator and obtained as enriched or purified proteins developed from recombinant or natural sources. These will include but are not limited to the following cytokines and hormones: interleukin 7 for the growth of B lineage lymphocytes; stem cell factor for all hematopoietic lineages; M-CSF for macrophages and osteoclasts; osteoprotegerin ligand for osteoclasts; erythropoietin for erythrocytes; thrombopoietin for platelets and megakaryocytes; interleukin 6 for platelets, megakaryocytes, and B lineage lymphocytes. Optimal concentrations and length of treatment may be determined by the practitioner through the use of known assays for the differentiation of each blood cell lineage.
  • “Non-peptide growth factors” refers to steroids, retinoids and other chemical compounds or agents which induce the differentiation of blood cell lineages. It is generally recognized that concentrations may vary. Moreover, it is generally recognized that the compounds or agents will be added in amounts sufficient to stimulate differentiation. Generally, however, these will be used at concentrations ranging from about 1 nM to about 100 nM for 1,25 dihydroxy vitamin D3, about 1 nM to about 100 nM dexamethasone, about 1 nM to about 100 nM hydrocortisone, about 1 nM to about 100 nM retinoic acid, about 1 nM to about 100 nM 9-cis retinoic acid, or at concentrations to be determined and optimized by the practitioner.
  • Amounts of 5′ azacytadine and/or amphotericin sufficient to induce differentiation refers to concentrations of 5′ azacytadine and amphotericin, that when supplied in a medium capable of supporting the growth of stromal cells (e.g. NIH-3T3, C3H 10T1/2, human adipose tissue-derived stromal cells and the like), will induce the differentiation of said stromal cells into skeletal muscle myoblasts and myocytes over a period of about 1 to 6 weeks. Typical use concentrations for 5′azacytadine range from about 1 μM to about 30 μM. Typical use concentrations for amphotericin range from about 10 ng/ml to about 100 ng/ml. Optimal concentrations and length of exposure may be determined by the practitioner through the use of known assays for the differentiation of skeletal muscle myoblasts. Such assays include, but are not limited to, those that assess the morphological or biochemical characteristics associated with skeletal muscle (e.g., formation of multinucleated myotubules, expression of myosin heavy chain, expression of myoD at the protein or RNA level).
  • “Amounts of transforming growth factor β or other peptide growth factors sufficient to induce differentiation” refers to concentrations of transforming growth factor β or other peptides, that when supplied to medium capable of supporting the growth of stromal cells (e.g. NIH-3T3, C3H 10T1/2, human adipose tissue-derived stromal cells and the like), will induce the differentiation of said stromal cells into smooth muscle myocytes or myofibroblasts over a period of 1 day to 6 weeks. Typical use concentrations for transforming growth factor β range from about 20 ng/ml to about 40 ng/ml. Typical use concentrations for fibroblast growth factor range from about 20 ng/ml to about 40 ng/ml. Optimal concentrations and length of exposure may be determined by the practitioner through the use of known assays for the differentiation of smooth muscle myoblasts. Such assays include, but are not limited to, those that assess the morphological or biochemical characteristics associated with smooth muscle (e.g., expression of a smooth muscle actin and fibronectin, generation of contractile forces when placed in collagen lattices in the presence of thrombin or lysophosphatidic acid).
  • Any medium capable of supporting stromal cells in tissue culture may be used. Media formulations that will support the growth of fibroblasts include, but are not limited to, Dulbecco's Modified Eagle's Medium (DMEM), alpha modified Minimal Essential Medium (αMEM), and Roswell Park Memorial Institute Media 1640 (RPMI Media 1640) and the like. Typically, 0 to 20% Fetal Bovine Serum (FBS) will be added to the above media in order to support the growth of stromal cells and hematopoietic cells. However, a defined medium could be used if the necessary growth factors, cytokines, and hormones in FCS for stromal cell and hematopoietic cell growth are identified and provided at appropriate concentrations in the growth medium.
  • Media useful in the methods of invention may contain one or more compounds of interest, including, but not limited to, antibiotics, compounds that are mitogenic for hematopoietic; stem cells, differentiation inducing for hematopoietic stem cells, and/or mitogenic or differentiative for stromal cells. Example of antibiotics useful in the invention include but are not limited to penicillin and streptomycin. Penicillin is typically used at about 10 units/ml to about 200 units/ml. Streptomycin is typically used at about 10 μg/ml to 200 μg/ml. Examples of hematopoietic mitogenic factors include but are not limited to stem cell factor and interleukin 3; hematopoietic differentiation inducing factors include but are not limited to 1,25 dihydroxy vitamin D3, interleukin 7, and osteoprotegerin ligand; stromal cell mitogens include but are not limited to transforming growth factor β; and stromal cell differentiating factors include but are not limited to dexamethasone, hydrocortisone, transforming growth factor β, and the like.
  • “Adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and growth factors and agents capable of inducing stromal cell expression of hematopoietic growth factors or exogenous hematopoietic growth factors or non-peptide factors themselves” refers to growth factors, both peptide and chemical in composition, which enhance the proliferation and maturation of hematopoietic stem cells in vitro and in vivo. These include, but are not limited to, interleukin 1, interleukin 3, interleukin 6, interleukin. 7, interleukin 11, macrophage colony stimulating factor (M-CSF), granulocyte-monocyte colony stimulating factor (GM-CSF), stem cell factor, flt3 ligand, thrombopoietin, erythropoietin, osteoprotegerin ligand, dexamethasone, hydrocortisone, and 1,25 dihydroxy vitamin D3. The concentrations of these factors and the length of time of exposure will be determined and optimized by the investigators. Interleukins, M-CSF, GM-CSF, Flt 3 ligand and stem cell factor are used at about 5 pg/ml to about 1 ng/ml. Thrombopoietin is typically used at concentrations ranging from about 5 pg/ml to about 1 ng/ml. Erythropoietin is used at about 5 units/ml to about 1000 units/ml. Osteoprotegerin ligand is used at about 5 pg/ml to about 1 ng/ml. Dexamethasone, hydrocortisone, and 1,25 dihydroxy vitamin D3 are at concentrations from about 1 nM to about 100 nM. Optimal concentrations and treatment times will be determined by monitoring the production of specific circulating blood cell lineages in the co-cultures of hematopoietic stem cells and adipose tissue-derived stromal cells. It is generally recognized that these factors will be added in amounts sufficient to stimulate differentiation. Such assays and indices include, but are not limited to, those that assess the morphological or biochemical characteristics of the cells, such as the expression of cell surface proteins unique to specific blood cell lineages by flow cytometry, immunohistochemistry, and/or immunofluorescent methods, expression of specific mRNAs in the cell population, or by in vivo assessment of the production of hematopoietic stem cells by the co-culture system.
  • “Adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and amounts of azacytadine and/or amphotericin or other agents sufficient to induce the differentiation of said stromal cells into skeletal muscle myocytes” refers to the differentiation inducing agents used to promote expression of skeletal muscle specific gene markers and skeletal muscle function in vitro. The medium comprises fetal bovine serum, antibiotic, L-glutamine, sodium pyruvate, 2-mercaptoethanol, and 5′ azacytadine or amphotericin. Fetal bovine serum can be used at concentrations ranging from 0.5% to 20%. The antibiotic typically used, but not limited to, is penicillin or streptomycin. Penicillin is typically used at concentrations ranging from about 10 units/ml to about 200 units/ml. Streptomycin is typically used in concentrations ranging from about 10 μg/ml to about 200 μg/ml. L-glutamine and sodium pyruvate are typically used at 0.5 mM to about 2 mM. 2-mercaptoethanol is typically used at about 10 μM to about 100 μM. 5′ azacytadine is typically used at about 1 μM to about 30 μM. Amphotericin is typically used at about 10 ng/ml to about 100 ng/ml. The concentrations of these factors and the length of time of exposure will be determined and optimized by the investigators. Optimal concentration and treatment times will be determined by monitoring the morphologic and biochemical markers characteristic of skeletal muscle. These include, but are not limited to, the production of multi-nucleated myotubules in culture and the expression of muscle specific genes and proteins, such as muscle transcription factors (myoD, myogenin), myosin light chain kinase, myosin heavy chain kinase, and skeletal muscle actin.
  • “Adipose tissue-derived stromal cells, a medium capable of supporting the growth of the stromal cells, and amounts of transforming growth factor β or other peptide growth factors sufficient to induce the differentiation of said stromal cells into smooth muscle myocytes or myofibroblasts” refers to the differentiation inducing conditions used to promote the expression of smooth muscle associated gene markers and proteins and smooth muscle function in vitro. The concentrations of factors and the length of time of exposure will be determined and optimized by the investigators. Optimal concentration and treatment times will be determined by monitoring the morphologic and biochemical markers characteristic of smooth muscle cells. These include, but are not limited to, the generation of tensile forces by the cells when placed in a collagen type I lattice and the expression of smooth muscle specific genes and proteins, such as smooth muscle actin, fibronectin, and laminin.
  • Preferably, the adipose tissue derived stromal cells are isolated from the adipose tissue of the subject into which the final differentiated cells are to be introduced. However, the stromal cells may also be isolated from any organism of the same or different species as the subject. Any organism with adipose tissue can be a potential candidate. Preferably, the organism is mammalian, most preferably the organism is human.
  • The adipose tissue derived stromal cells may be stably or transiently transfected or transduced with a nucleic acid of interest using a plasmid, viral or alternative vector strategy. Nucleic acids of interest include, but are not limited to, those encoding gene products which; (1) enhance the growth, differentiation, maturation and proliferation of hematopoietic cell lineages; examples include osteoprotegerin ligand which induces osteoclast development, interleukin 7, which induces B lineage lymphocyte development, erythropoietin, which induces erythrocyte development, and thrombopoietin, which induces platelet development; (2) enhance the differentiation of skeletal muscle; examples include myoD and myogenin, transcription factors which promote myotubule formation and expression of skeletal muscle specific genes; (3) enhance the growth, differentiation and maturation of smooth muscle cells; examples include transforming growth factor β, which induces smooth muscle proliferation and extracellular matrix production.
  • The blood cells produced by in vitro co-cultures of hematopoietic stem cells and adipose tissue derived stromal cells can be introduced alone or in combination with the stromal component into subjects subject to anemia or limited blood cell production. These may include, but are not limited to, patients receiving high dose chemotherapy, patients undergoing bone marrow transplantation, patients suffering from aplastic anemia, patients suffering from sickle cell anemia, and other blood dyscrasias.
  • Other disorders which may be treated with infusion of stem cells include, but are not limited to, diseases resulting from a failure or a dysfunction of normal blood cell production and maturation (i.e., aplastic anemia and hypoproliferative stem cell disorders); neoplastic, malignant diseases in the hematopoietic organs (e.g., leukemia and lymphomas); broad spectrum malignant solid tumors of non-hematopoietic origin; autoimmune conditions; and genetic disorders. Such disorders include, but are not limited to diseases resulting from a failure or dysfunction of normal blood cell production and maturation hyperproliferative stem cell disorders, including aplastic anemia, pancytopenia, agranulocytosis, thrombocytopenia, red cell aplasia, Blackfan-Diamond syndrome, due to drugs, radiation, or infection, idiopathic; hematopoietic malignancies including acute lymphoblastic (lymphocytic) leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, acute malignant myelosclerosis, multiple myeloma, polycythemia vera, agnogenic myelometaplasia, Waldenstrom's macroglobulinemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma; immunosuppression in patients with malignant, solid tumors including malignant melanoma, carcinoma of the stomach, ovarian carcinoma, breast carcinoma, small cell lung carcinoma, retinoblastoma, testicular carcinoma, glioblastoma, rhabdomyosarcoma, neuroblastoma, Ewing's sarcoma, lymphoma; autoimmune diseases including rheumatoid arthritis, diabetes type I, chronic hepatitis, multiple sclerosis, systemic lupus erythematosus; genetic (congenital) disorders including anemias, familial aplastic, Fanconi's syndrome, Bloom's syndrome, pure red cell aplasia (PRCA), dyskeratosis congenita, Blackfan-Diamond syndrome, congenital dyserythropoietic syndrome I-IV, Chwachmann-Diamond syndrome, dihydrofolate reductase deficiencies, formamino transferase deficiency, Lesch-Nyhan syndrome, congenital spherocytosis, congenital elliptocytosis, congenital stomatocytosis, congenital Rh null disease, paroxysmal nocturnal hemoglobinuria, G6PD (glucose-6-phosphate dehydrogenase) variants 1, 2, 3, pyruvate kinase deficiency, congenital erythropoietin sensitivity, deficiency, sickle cell disease and trait, thalassemia alpha, beta, gamma, met-hemoglobinemia, congenital disorders of immunity, severe combined immunodeficiency disease (SCID), bare lymphocyte syndrome, ionophore-responsive combined immunodeficiency, combined immunodeficiency with a capping abnormality, nucleoside phosphorylase deficiency, granulocyte actin deficiency, infantile agranulocytosis, Gaucher's disease, adenosine deaminase deficiency, Kostmann's syndrome, reticular dysgenesis, congenital leukocyte dysfunction syndromes; and others such as osteopetrosis, myelosclerosis, acquired hemolytic anemias, acquired immunodeficiencies, infectious disorders causing primary or secondary immunodeficiencies, bacterial infections (e.g., Brucellosis, Listerosis, tuberculosis, leprosy), parasitic infections (e.g., malaria, Leishmaniasis), fungal infections, disorders involving disproportions in lymphoid cell sets and impaired immune functions due to aging, phagocyte disorders, Kostmann's agranulocytosis, chronic granulomatous disease, Chediak-Higachi syndrome, neutrophil actin deficiency, neutrophil membrane GP-180 deficiency, metabolic storage diseases, mucopolysaccharidoses, mucolipidoses, miscellaneous disorders involving immune mechanisms, Wiskott-Aldrich Syndrome, alpha 1-antitrypsin deficiency, etc.
  • The skeletal muscle cells produced by in vitro manipulation of the adipose tissue derived stromal cells can be introduced alone or in combination with a composition matrix to repair muscle defects secondary to metabolic diseases (muscular dystrophy, myositis), trauma, and disuse atrophy. Such compositions include, but are not limited to, collagen matrices, poly-lactic polymers, poly-glycolic polymers, alginate, or other solid supports.
  • The smooth muscle cells produced by in vitro manipulation of the adipose tissue derived stromal cells can be introduced alone or in combination with a composition matrix to repair smooth muscle defects. These defects may include, but are not limited to, urinary bladder wall abnormalities due to hereditary malformations in neonates or secondary to trauma or tumor invasion in older individuals, gastrointestinal tract abnormalities due to hereditary malformations in neonates or secondary to trauma or tumor invasion in older individuals, genital tract abnormalities (vaginal) due to hereditary malformations in neonates, secondary to trauma or tumor invasion, or for tissue reconstructive surgeries in transgender operations, or for the development of functional large veins for grafting purposes. Composition matrices may include, but are not limited to, collagen matrices such as swine intestinal submucosa, poly-lactic polymers, poly-glycolic polymers, alginate, or other solid supports.
  • Another object of the invention is to provide methods for the identification and study of compounds that enhance or inhibit the differentiation of adipose tissue derived stromal cells into either hematopoietic supporting stromal cells, skeletal muscle myocytes, or smooth muscle myocytes. Compounds which enhance differentiation. (a) hematopoietic supporting stromal cell function may be of value in the treatment of blood dyscrasias characterized by decreased production of circulating blood cells and improve recovery of patients following high dose chemotherapy; (b) skeletal muscle myocytes may be of value in the treatment of musculoskeletal diseases secondary to hereditary defects or trauma; or (c) smooth muscle myocytes may be of value in the treatment of smooth muscle defects, including those of the urinary bladder (bladder wall), gastrointestinal tract (colon, small intestine), and genital system (vaginal). Conversely, compounds which inhibit differentiation of (a) hematopoietic supporting stromal cells may be of value in the treatment of blood dyscrasias characterized by overproduction of circulating blood cells, such as polycythemia vera; (b) skeletal muscle may be of value in the treatment of soft tissue tumors of skeletal muscle origin, such as rhabdomyosarcomas; and (c) smooth muscle may be of value in the treatment of soft tissue tumors of smooth muscle origin, such as leiomyosarcomas.
  • Any compound may be tested for its ability to affect the differentiation of adipose tissue derived stromal cells into either hematopoietic supporting stromal cells, skeletal muscle myocytes, or smooth muscle myocytes. Appropriate vehicles compatible with the compound to be tested are known to those skilled in the art and may be found in the current edition of Remington's Pharmaceutical Sciences, the contents of which are incorporated herein by reference.
  • The features and advantages of the present invention will be more clearly understood by reference to the following examples, which are not to be construed as limiting the invention.
  • EXAMPLES Example 1 Expression of Cell Surface Adhesion Molecules and Hematopoietic Cytokines by Adipose Tissue-Derived Stromal Cells In Vitro
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 30,000 cells per cm2 in chamber slides, in 6 well tissue culture plates, or in T25 cm2 flasks. Cells are maintained in culture for 8 days in DMEM/Ham's F-10 supplemented with 10% fetal bovine serum, penicillin 100 units/ml, streptomycin 100 μg/ml, and 7.5 mM HEPES pH 7.2. The surface proteins expressed by the stromal cells are determined by immunologic techniques based on immunohistochemistry and/or flow cytometry. For immunohistochemical analysis, chamber slides are fixed using 95% ethanol/5% glacial acetic acid and incubated with murine monoclonal antibodies detecting human cell surface proteins. After incubation with an enzyme coupled anti-mouse secondary antibody, evidence of protein expression is detected by histochemical reaction. Alternatively, flasks of cells are harvested by trypsin/EDTA digestion and incubated with a fluorescent conjugated murine monoclonal antibody detecting a specific human surface protein. Cells are examined for fluorescent intensity by flow cytometry. The results of these assays are summarized in Table 1. These studies demonstrate that adipose-derived stromal cells express cell surface proteins associated with and essential for hematopoietic support function by bone marrow stromal cells (Miyake et al. (1990) J Exp Med 171:477-488; Miyake et al. (1991) J Exp Med 173:599-607; Miyake et al. (1991) J Cell Biol 114:557-565, 1991; Jacobsen et al. (1992) J Exp Med 176:927-935; Kincade et al. (1993) Curr Top Microbiol Immunol 184:215-222; Hayashi et al. (2000) Leuk Lymphoma 38:265-270) these include VCAM1, CD44, integrin β1, integrin α4,5 (VLA-4, VLA-5), and CD9, among others.
  • The cytokine expression profile of the adipose-derived stromal cells is determined following induction with lipopolysaccharide (LPS) or endotoxin, an inflammatory agent capable of inducing hematopoietic cytokines in bone marrow stromal cells (Gimble et al. (1989) Blood 74:303-311). Confluent and quiescent cultures of cells are exposed to 100 ng/ml LPS for periods of 0 to 24 hours in DMEM medium supplemented with 2% fetal bovine serum, 100 μg/ml streptomycin, 100 units/ml penicillin, and 7.5 mM HEPES pH 7.2. The conditioned medium from each culture is harvested and stored at −80° C. while the total RNA is harvested by the method of Chomczynski and Sacchi (See Anal. Biochem. (1987)162:156-159). The mRNA for the cytokines indicated in Table 2 is detected by polymerase chain reaction using the oligonucleotide primer sets listed below the table. A representative set of reactions is demonstrated in FIG. 1. The following cytokines demonstrated significant LPS inducible expression of immunoreactive protein based on enzyme linked immunoassay: macrophage colony stimulating factor (M-CSF), granulocyte/monocyte colony stimulating factor (GM-CSF), interleukin 6, 7, and 8 (IL-6, 7, 8). The profile of cytokines expressed by the adipose derived stromal cells is consistent that of bone marrow-derived stromal cells capable of supporting myeloid, lymphoid, and osteoclast proliferation and differentiation in vitro (Pietrangeli et al. (1988) Eur. J. Immunol. 18:863-872; Gimble et al. (1989) Blood 74:303-311; Gimble et al. (1992) J. Cell Biochem 50:73-82; Kelly et al. (1998) Endocrinol. 139:2092-2101).
  • Example 2 Establishment of Myelopoietic Co-Cultures with an Adipose Tissue-Derived Stromal Cells Layer In Vitro
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm2. Stromal cells are established in the cultures for 1 to 3 days prior to the introduction of hematopoietic progenitor cells into the co-culture system. Hematopoietic progenitor cells are isolated from one of the following human tissues: bone marrow, umbilical vein/placental blood, peripheral blood, spleen. Alternatively, murine tissues are used. Murine bone marrow cells are harvested by flushing the marrow cavity of 6 to 10 week old mice with DMEM/10% FCS under sterile conditions. Murine spleen cells are harvested by physical passage through a fine metal screen under sterile conditions. One of three methods are used to deplete the mixed hematopoietic cell population of its stromal component. As a first alternative, the hematopoietic stem cell population from the blood sample will be enriched by magnetic immunobead purification using anti-CD34 antigen according to established techniques. As a second alternative, hematopoietic cells will be enriched by passage of the bone marrow or other blood sample over a sterile G-10 Sephadex or nylon wool column; hematopoietic progenitor cells are eluted while stromal cells are retained. As a third alternative, hematopoietic cells will be enriched by flow cytometric sorting based on surface protein characteristics. The hematopoietic cells are washed once and the number of nucleated cells is counted using a hematocytometer after erythrocyte lysis with 0.3% acetic acid and trypan blue staining. Hematopoietic cells are introduced into the liquid co-cultures at a ratio of preferably 10 to 100 nucleated hematopoietic cells per stromal cell plated, more preferably 20 to 30 nucleated hematopoietic cells per stromal cell plated, most preferably 25 to 30 nucleated hematopoietic cells per stromal cell plated. Cells are cultured in a medium consisting of DMEM (high glucose), 10% fetal bovine serum supplemented with 10 to 100 nM hydrocortisone or 10% horse serum, 10 to 200 units/ml of penicillin, 10 to 200 μg of streptomycin at 33° C. in 5% CO2. One half of the medium in the co-cultures is replaced every 3 to 4 days. The number of non-adherent cells in the medium is determined by hematocytometer count and/or by flow cytometry. The surface antigen characteristics of the non-adherent cells are documented using routine antibody markers for the major hematopoietic cell lineages. These will include, but are not limited to, Mac-1, Thy 1, Ig Heavy chain, Ter-81 (erythroid marker). The number and character of the non-adherent cell population is determined over a period of up to 10 weeks. At the conclusion of the study, the cellular composition of the adherent cell layer is determined by flow cytometric or immunohistochemical methods. As an alternative approach, the hematopoietic studies are conducted in semi-solid cultures. Cells are prepared as described above, but the hematopoietic progenitor cells are plated in the additional presence of 2.1% methylcellulose. Colony formation is assessed after a 7 to 14 day period for the presence of granulocytes, erythrocytes, macrophages, and monocytes using histologic, morphologic and immunologic criteria.
  • Example 3 Ability of Adipose Tissue Derived Stromal Cell/Hematopoietic Progenitor Cell Co-Cultures to Maintain Proliferation of Hematopoietic Progenitors In Vitro
  • Adipose tissue derived stromal cell/hematopoietic progenitor co-cultures established under liquid culture conditions described in Example 1 are used to assess the ability of this system to maintain the proliferation of the hematopoietic progenitor cells in vitro. Co-cultures are established using human adipose tissue derived stromal cells and murine hematopoietic progenitors. Cultured cells are transduced with a viral vector expressing a traceable protein marker such as green fluorescent protein or beta-galactosidase. Alternatively, co-culture cells are identified by expression of a unique antigen or genetic marker due to their origin; e.g., the expression of human proteins for the stromal cells, and the expression of a transgenic or male gender specific marker for the murine hematopoietic cells. Established co-cultures are harvested by limited incubation with trypsin/EDTA and infused into a lethally irradiated immunodeficient mouse. Animals will be followed over time. After 9 to 14 days, mice are sacrificed and their spleens examined for the appearance of hematopoietic cell islands or splenic colony forming units (CFU-S). Alternatively, mice will be maintained for 14 days or longer and their circulating blood cell count determined by hematologic and flow cytometric assays. The presence of specific markers of the stromal cells and the donor hematopoietic cells is detected with antibody reagents or specific DNA markers on fixed cells, either by flow cytometry or conventional pathologic/histologic methods. The ability of the co-cultured cells to establish CFU-S in the recipient and/or to maintain the proliferation and maturation of donor blood cells in the host after 14 days is evidence of the continued expansion of some hematopoietic progenitors in vitro by the adipose tissue-derived stromal cells.
  • Example 4 Establishment of Lymphopoietic Co-Cultures with an Adipose Tissue-Derived Stromal Cells Layer In Vitro
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm2. Stromal cells are established in the cultures for 1 to 3 days prior to the introduction of hematopoietic progenitor cells into the co-culture system. Hematopoietic progenitor cells are isolated from one of the following human tissues: bone marrow, umbilical vein/placental blood, peripheral blood, spleen. Alternatively, murine tissues are used. Murine bone marrow cells are harvested by flushing the marrow cavity of 6 to 10 week old mice with RPMI/10% FCS under sterile conditions. Murine spleen cells are harvested by physical passage through a fine metal screen under sterile conditions. One of three methods are used to deplete the mixed hematopoietic cell population of its stromal component, As a first alternative, the hematopoietic stem cell population from the blood sample will be enriched by magnetic immunobead purification using anti-CD34 antigen according to established techniques. As a second alternative, hematopoietic cells will be enriched by passage of the bone marrow or other blood sample over a sterile G-10 Sephadex or nylon wool column; hematopoietic progenitor cells are eluted while stromal. cells are retained. As a third alternative, hematopoietic cells will be enriched by flow cytometric sorting. The hematopoietic cells are washed once and the number of nucleated cells is counted using a hematocytometer after erythrocyte lysis with 0.3% acetic acid and trypan blue staining. Hematopoietic cells are introduced into the liquid co-cultures at a ratio of preferably 10 to 100 nucleated hematopoietic cells per stromal cell plated, more preferably 20 to 30 nucleated hematopoietic cells per stromal cell plated, most preferably 25 to 30 nucleated hematopoietic cells per stromal cell plated. Cells are cultured in a medium consisting of RPMI 1640, prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 μg/ml of streptomycin, 0.5 to 2 mM L-glutamine, 10 to 100 μM 2-mercaptoethanol at 37° C. in 5% CO2. One half of the medium in the co-cultures is replaced every 3 to 4 days. The number of non-adherent cells in the medium is determined by hematocytometer count and/or by flow cytometry. The surface antigen characteristics of the non-adherent cells are documented using routine antibody markers for the major hematopoietic cell lineages. These will include, but are not limited to, Mac-1, Thy 1, Ig Heavy chain, Ter-81 (erythroid marker). The number and character of the non-adherent cell population is determined over a period of up to 10 weeks. At the conclusion of the study, the cellular composition of the adherent cell layer is determined by flow cytometric or immunohistochemical methods. As an alternative approach, the hematopoietic studies are conducted in semi-solid cultures. Cells are prepared as described above but the hematopoietic progenitor cells are plated in the additional presence of 2.1% methylcellulose. Colony formation is assessed after a 7 to 14 day period for the presence of B lineage lymphoid cells as well as granulocytes, erythrocytes, macrophages, and monocytes using histologic, morphologic and immunologic criteria. Alternatively, co-cultures and/or semi-solid cultures are established as described above with the addition of interleukin 7 at concentrations to be determined by the practitioner which enhance the proliferation and maturation of B lineage lymphocytes. The techniques outlined above are used to assess the affect of this growth factor on the hematopoietic support function of the adipose tissue derived stromal cell.
  • Example 5 Establishment of Osteoclastogenic Co-Cultures with an Adipose Tissue-Derived Stromal Cells Layer In Vitro
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, Filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm2 in 24 well plates. Cells are cultured in a medium consisting of DMEM (high glucose), prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 μg of streptomycin, 0.5 to 2 mM L-glutamine, 0.5 to 2 mM sodium pyruvate, 10 to 100 μM 2-mercaptoethanol at 37° C. in 5% CO2. Three days after the stromal cultures are established, the medium is supplemented with either 10 to 100 nM 1,25 dihydroxy vitamin D3 and or osteoprotegerin ligand at concentrations determined by the practitioner. Stromal cells are established in the cultures for 6 days prior to the introduction of hematopoietic progenitor cells into the co-culture system.
  • Hematopoietic progenitor cells are isolated from one of the following human tissues: bone marrow, umbilical vein/placental blood, peripheral blood, spleen. Alternatively, murine tissues are used. Murine bone marrow cells are harvested by flushing the marrow cavity of 6 to 10 week old mice with DMEM (high glucose)/10% FCS under sterile conditions. Murine spleen cells are harvested by physical passage through a fine metal screen under sterile conditions. One of three methods are used to deplete the mixed hematopoietic cell population of its stromal component. As a first alternative, the hematopoietic stem cell population from the blood sample will be enriched by magnetic immunobead purification using anti-CD34 antigen according to established techniques. As a second alternative, hematopoietic cells will be enriched by passage of the bone marrow or other blood sample over a sterile G-10 Sephadex or nylon wool column; hematopoietic progenitor cells are eluted while stromal cells are retained. As a third alternative, hematopoietic cells will be enriched by flow cytometric sorting based on surface protein characteristics. The hematopoietic cells are washed once and the number of nucleated cells is counted using a hematocytometer after erythrocyte lysis with 0.3% acetic acid and trypan blue staining. Hematopoietic cells are introduced into the liquid co-cultures at a ratio of preferably 10 to 100 nucleated hematopoietic cells per stromal cell plated, more preferably 20 to 30 nucleated hematopoietic cells per stromal cell plated, most preferably 25 to 30 nucleated hematopoietic cells per stromal cell plated. One half of the medium in the co-cultures is replaced every 3 to 4 days. Co-cultures are maintained in the presence of 1,25 dihydroxy vitamin D3 or osteoprotegerin ligand continuously after the introduction of hematopoietic cells.
  • After a period of co-culture of 6 to 9 days, co-cultures are fixed with 0.5 ml 3.7% (vol:vol) formaldehyde in phosphate buffered saline for 5 minutes, dried for 30 seconds with acetone:ethanol (50:50, vol/vol), and stained for 10 minutes with 10 mM sodium tartrate, 40 mM sodium acetate (pH 5.0), 0.1 mg/ml naphthol AS-MS phosphate (Sigma N-5000), and 0.6 mg/ml fast red violet LB salt (Sigma F-3381). Stained cultures are rinsed in distilled water and stored under 50% glycerol/PBS. The number of tartrate resistant acid phosphatase positive cells per well is assessed under light microscopy based on the red staining of the cytoplasm. TRAP+ cells are, numerically counted and those with 1-2 nuclei are distinguished from those multinucleated cells with >3 nuclei per cell. This assay demonstrates the ability of adipose tissue derived stromal cells to support the differentiation and proliferation of osteoclastogenic precursors in vitro. This culture procedure is able to expand and promote differentiation of osteoclasts. This has potential application to rare clinical conditions such as osteopetrosis characterized by brittle bones where patients fail to produce native osteoclasts. This In vitro method offers a means to expand an individual's own osteoclast progenitors and to promote their differentiation ex vivo. This cell population can be re-infused into the affected individual with potential short-term or long-term benefit. The non-invasive nature of the methodology and the potential to rely exclusively on autologous cells indicates that this procedure could be used repetitively in the treatment of an individual patient.
  • Example 6 Use of Adipose Tissue Derived Stromal Cell Supported Ex Vivo Hematopoiesis as a Therapeutic Modality for Bone Marrow Transplant and Hematologically Compromised Patients
  • The co-culture models outlined in Examples 1-4 have the potential to be used to facilitate the recovery of bone marrow function in patients receiving high dose chemotherapy, high dose radiation treatment or any other therapeutic modality which compromises blood cell production and bone marrow function. In advance of any elective procedure, an individual can donate his or her own adipose tissue and blood cells for subsequent autologous transplantation. Prior to immunocompromising procedures, the individual's own blood cells and stromal co-cultures can be established and expanded. Following any immunocompromising procedure, the patients own blood cell/stromal cell co-culture can be re-infused into the patient according to standard transfusion methodologies. This can be done in the absence or presence of exogenous hematopoietic cytokines, either added to the co-cultures or given directly to the patient. This approach may accelerate the rate of blood cell production in the patient, reduce the need for cytokine therapies, and reduce the overall costs and risk of the chemotherapy or other immunocompromising procedure. With the, ex vivo nature of the procedure, it is possible to manipulate the stromal cells to enhance production of specific blood cell lineages. Stromal cells transiently expressing interleukin 7, for example, would facilitate the rapid expansion of B lineage lymphoid cells while stromal cells expressing erythropoietin would favor expansion of erythrocytes.
  • As outlined above, the approach is designed primarily for the treatment of nosocomial induced blood cell dyscrasias. However, large scale ex vivo production of autologous stromal/hematopoietic co-cultures is of potential value for elective and non-elective surgical procedures requiring transfusion intra-operatively and post-operatively. At present, the majority of patients requiring blood transfusion receive blood products donated by others. This presents risk to the recipient of transmission of unrecognized infectious disease from the donor. With the ability to develop blood cell production ex vivo, an individual can expand his/her hematopoietic cell population at a capacity which is no longer limited by the bone marrow cavity volume. Using cell factory tissue culture approaches with recirculating systems, continuous production of blood cells by an adipose tissue derived stromal cell/hematopoietic cell co-culture is feasible. This approach has the advantage of avoiding risks inherent in transfusion of blood from a donor to a recipient; these include the transmission of infectious diseases such as HIV, hepatitis, cytomegalovirus, Jacob/Creukzfeld disease, among others.
  • Example 7 Differentiation of Adipose Tissue Derived Stromal Cells into Skeletal Muscle Myoblasts
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm2 in 24 well plates. Cells are cultured in a medium consisting of DMEM (high glucose), prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 μg of streptomycin, 0.5 to 2 mM L-glutamine, 0.5 to 2 mM sodium pyruvate, 10 to 100 μM 2-mercaptoethanol at 37° C. in 5% CO2. Cells are exposed to 1 to 30 μM 5′ azacytadine or 10 to 100 ng/ml of amphotericin for periods of 1 to 6 days to assure that cells throughout S phase are continuously exposed to these agents. Following this period, cultures are maintained in the culture medium without azacytadine or amphotericin supplements. Cultures are either continued at the established cell density or sub-cloned by limit dilution methods to select for cell clones capable of expressing characteristic markers of skeletal muscle myoblasts. These cells are selected based on morphologic criteria, specifically, the ability to form multinucleated myotubules in culture; biochemical criteria, specifically, the expression of myosin heavy and light chain kinase, skeletal muscle actin and myosin and the expression of myogenic transcription factors, myoD and/or myogenin.
  • Example 8 Application of Skeletal Muscle Myoblasts Differentiated from Adipose Tissue-Derived Stromal Cell
  • The cells skeletal muscle myoblasts developed in Example 6 can be used for tissue engineering purposes in the treatment of myodystrophies, muscle atrophy, and physical loss of skeletal muscle secondary to surgical procedures for the treatment of cancer or trauma, The ex vivo development of a proliferating population of myoblasts from adipose tissue can be used to supplement and repair skeletal muscle mass in afflicted individuals. Myoblasts can be cultured in biodegradable matrices composed of poly-lactic, poly-glycolic, collagen or other materials to form muscle strands. These can then be implanted to an afflicted site and tethered by suture to existing muscle, tendon or bone. Alternatively, ex vivo expanded myoblasts can be genetically engineered by viral transduction, plasmid transfection, or other means to express novel genes. These cells can be injected directly into existing muscle sites where these novel gene products will now be expressed. This approach has direct application to muscular dystrophy, where patients suffer secondary to a mutation in an important skeletal muscle gene. Likewise, the engineered stromal cells can convert the muscle into a production site for a deficient circulating protein. For example, adipose tissue derived stromal cells expressing lipoprotein lipase can be used to treat the many patients with mutations in their native lipoprotein lipase gene who are at increased risk of severe cardiovascular disease.
  • Example 9 Differentiation and Expansion of Smooth Muscle Myoblasts from Adipose Tissue Derived Stromal Cells Ex Vivo
  • Stromal cells are isolated from human subcutaneous adipose tissue according to methods described in “Methods and Compositions for the Differentiation of Human Preadipocytes into Adipocytes” Ser. No. 09/240,029, filed Jan. 29, 1999. These cells are plated at a density of 500 to 20,000 cells per cm2 in 24 well plates. Cells are cultured in a medium consisting of DMEM (high glucose), prescreened 10% fetal bovine serum, 10 to 200 units/ml of penicillin, 10 to 200 μg of streptomycin, 0.5 to 2 mM L-glutamine, 0.5 to 2 mM sodium pyruvate, at 37° C. in 5% CO2. Cultures are supplemented with transforming growth factor β and/or fibroblast growth factor at concentrations determined by the practitioner but not to exceed 40 ng/. Cells are maintained in culture as a monolayer or in a 3-dimensional lattice composed of collagen type I or other biodegradable material (alginate, synthetic polymer or other). Cells are characterized based on morphologic, biochemical, and functional criteria for smooth muscle myoblast differentiation; these include, but are not limited to, expression of smooth muscle actin, fibronectin, laminin, and other extracellular matrix proteins, the ability to generate a tensile force as measured by a pressure transducer, and to organize along a line of force in a 3 dimensional lattice.
  • Example 10 Application of Smooth Muscle Myoblasts Differentiated from Adipose Tissue-Derived Stromal Cells Ex Vivo
  • The smooth muscle myoblasts described under Example 8 can be used to treat conditions where smooth muscle function is compromised. For example, over 1000 neonates each year suffer from bladder wall abnormalities. The severity of this disorder is variable but it can be devastating and requires expensive surgical procedures to accomplish an acceptable repair and an approach to normal function. In many cases, the bladder size is too small or the bladder wall is incompletely formed, necessitating the implantation of prosthetic materials as a bladder wall replacement. Methods under investigation include the use of swine intestinal submucosa as a replacement material for the bladder wall. One issue is whether or not the surgically implanted bladder wall will achieve the appropriate physical and mechanical characteristics associated with stretching and retraction. Much of this is mediated by functional smooth muscle cells. Current methods implant the SIS material without preimplantation of any smooth muscle cells ex vivo. Surgeons rely on the recruitment of fibroblasts and myofibroblasts from adjacent tissues, including the omental adipose tissue. With the availability of adipose tissue derived stromal cells capable of smooth muscle myoblast differentiation, it is possible to pre-incubate SIS material with these cells ex vivo. The introduction of these cells prior to the surgical repair of the bladder wall is expected to accelerate and improve the attainment of appropriate bladder tone. This approach has broad application to all elastic soft tissue organs which rely on smooth muscle cells. These include, but are not limited to, the small intestine, large intestine, vagina, urethra, and venous blood vessels. The adipose tissue derived stromal cells have potential application to the surgical repair of defects in any of these organs.
  • All publications mentioned in the specification are indicative of the level of those skilled in the art to which this invention pertains. Publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity and understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims.

Claims (20)

1.-70. (canceled)
71. A method for differentiating adipose tissue derived stromal cells into cells that express at least one characteristic of a myoblast, comprising:
a) plating said stromal cells at a density of about 500 to about 20,000 cells per cm2 in chamber slides;
b) maintaining cells in a medium containing Dulbecco's Modified Eagle's Medium (DMEM) or Ham's F-10;
c) supplementing said medium with: (i) 1 to 10% fetal bovine serum (ii) an antibiotic (iii) glutamine (iv) sodium pyruvate (v) transforming growth factor β and/or fibroblast growth factor;
d) maintaining cells as a monolayer or in a 3-dimensional lattice comprising collagen type I, or alginate or other biodegradable material; and
e) characterizing cells for biochemical or functional criteria to establish myoblast differentiation.
72. The method according to claim 71, wherein said antibiotic is penicillin.
73. The method according to claim 71, wherein said antibiotic is streptomycin.
74. The method according to claim 72, wherein said penicillin is present from about 10 units per ml to about 200 units per ml.
75. The method according to claim 73, wherein said streptomycin is present from about 10 μg per ml to about 200 ug per ml.
76. The method according to claim 71, wherein said sodium pyruvate is present from about 0.5 mM to about 2 mM.
77. The method according to claim 71, wherein said transforming growth factor β is present from about 20 ng/ml to about 40 ng/ml.
78. The method according to claim 71, wherein said fibroblast growth factor is present from about 20 ng/ml to about 40 ng/ml.
79. (canceled)
80. The method according to claim 71, wherein the at least one characteristic of a myoblast is the expression of muscle heavy chain myosin kinase.
81. The method according to claim 71, wherein the at least one characteristic of a myoblast is the expression of muscle light chain myosin kinase.
82. The method according to claim 71, wherein the at least one characteristic of a myoblast is the expression of myosin.
83. The method according to claim 71, wherein the at least one characteristic of a myoblast is the expression of skeletal muscle actin.
84. The method according to claim 71, wherein the at least one characteristic of a myoblast is the expression of myogenic transcription factors.
85. The method according to claim 71, wherein the myogenic transcription factor is selected from myoD and myogenin.
86. The method according to claim 71, wherein the at least one characteristic of a myoblast is the expression of smooth muscle actin.
87. The method according to claim 71, wherein the myoblast is a skeletal muscle myoblast.
88. The method according to claim 71, wherein the myoblast is a smooth muscle myoblast.
89. The method according to claim 71, wherein the myoblast is a cardiac muscle myoblast.
US12/209,813 1999-08-19 2008-09-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof Abandoned US20090162934A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/209,813 US20090162934A1 (en) 1999-08-19 2008-09-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US13/085,250 US8486700B2 (en) 1999-08-19 2011-04-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US13/941,213 US20130288262A1 (en) 1999-08-19 2013-07-12 Multiple Mesodermal Lineage Differentiation Potentials for Adipose Tissue-Derived Stromal Cells and Uses Thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US14984999P 1999-08-19 1999-08-19
US09/638,544 US6555374B1 (en) 1999-08-19 2000-08-14 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US10/327,245 US20030166278A1 (en) 1999-08-19 2002-12-21 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US12/209,813 US20090162934A1 (en) 1999-08-19 2008-09-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/327,245 Division US20030166278A1 (en) 1999-08-19 2002-12-21 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/085,250 Continuation US8486700B2 (en) 1999-08-19 2011-04-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof

Publications (1)

Publication Number Publication Date
US20090162934A1 true US20090162934A1 (en) 2009-06-25

Family

ID=22532054

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/638,544 Expired - Lifetime US6555374B1 (en) 1999-08-19 2000-08-14 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US10/327,245 Abandoned US20030166278A1 (en) 1999-08-19 2002-12-21 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US12/209,813 Abandoned US20090162934A1 (en) 1999-08-19 2008-09-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US13/085,250 Expired - Fee Related US8486700B2 (en) 1999-08-19 2011-04-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US13/941,213 Abandoned US20130288262A1 (en) 1999-08-19 2013-07-12 Multiple Mesodermal Lineage Differentiation Potentials for Adipose Tissue-Derived Stromal Cells and Uses Thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/638,544 Expired - Lifetime US6555374B1 (en) 1999-08-19 2000-08-14 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US10/327,245 Abandoned US20030166278A1 (en) 1999-08-19 2002-12-21 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/085,250 Expired - Fee Related US8486700B2 (en) 1999-08-19 2011-04-12 Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US13/941,213 Abandoned US20130288262A1 (en) 1999-08-19 2013-07-12 Multiple Mesodermal Lineage Differentiation Potentials for Adipose Tissue-Derived Stromal Cells and Uses Thereof

Country Status (5)

Country Link
US (5) US6555374B1 (en)
EP (1) EP1077254A3 (en)
JP (1) JP4180228B2 (en)
CN (1) CN1309180A (en)
CA (1) CA2316400C (en)

Families Citing this family (114)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2312847C (en) * 1997-12-02 2014-09-30 Zen Bio, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US6777231B1 (en) * 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
US20050076396A1 (en) * 1999-03-10 2005-04-07 Katz Adam J. Adipose-derived stem cells and lattices
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
KR100968165B1 (en) * 1999-03-10 2010-07-06 더 리전츠 오브 더 유니버시티 오브 캘리포니아 Adipose-derived stem cells and lattices
US20030082152A1 (en) * 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
AU3869501A (en) * 2000-02-26 2001-09-03 Artecel Sciences Inc Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof
FR2819265B1 (en) * 2001-01-10 2004-01-02 Centre Nat Rech Scient EXTRAMEDULAR ADIPOSE TISSUE CELLS AND THEIR APPLICATIONS IN THE RECONSTRUCTION OF HEMATOPOIETIC LINES
US6905827B2 (en) 2001-06-08 2005-06-14 Expression Diagnostics, Inc. Methods and compositions for diagnosing or monitoring auto immune and chronic inflammatory diseases
US7235358B2 (en) 2001-06-08 2007-06-26 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
US7026121B1 (en) 2001-06-08 2006-04-11 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
WO2003008592A1 (en) * 2001-07-19 2003-01-30 Yasuhiko Tabata Polyfunctional stem cells originating in adipose tissue
EP1453954A4 (en) * 2001-11-09 2004-12-15 Artecel Sciences Inc Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
JP2005508393A (en) * 2001-11-09 2005-03-31 アーテセル・サイエンシズ・インコーポレーテツド Methods and compositions for the use of stromal cells to support embryonic and adult stem cells
US7651684B2 (en) 2001-12-07 2010-01-26 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in augmenting autologous fat transfer
US8404229B2 (en) 2001-12-07 2013-03-26 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to treat acute tubular necrosis
US20050048035A1 (en) 2001-12-07 2005-03-03 Fraser John K. Methods of using regenerative cells in the treatment of stroke and related diseases and disorders
US7585670B2 (en) 2001-12-07 2009-09-08 Cytori Therapeutics, Inc. Automated methods for isolating and using clinically safe adipose derived regenerative cells
US7595043B2 (en) 2001-12-07 2009-09-29 Cytori Therapeutics, Inc. Method for processing and using adipose-derived stem cells
EP2305276A3 (en) 2001-12-07 2011-09-21 Cytori Therapeutics, Inc. Processed lipoaspirate cells for use in therapy
US7771716B2 (en) 2001-12-07 2010-08-10 Cytori Therapeutics, Inc. Methods of using regenerative cells in the treatment of musculoskeletal disorders
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
US7514075B2 (en) 2001-12-07 2009-04-07 Cytori Therapeutics, Inc. Systems and methods for separating and concentrating adipose derived stem cells from tissue
US9597395B2 (en) 2001-12-07 2017-03-21 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in the treatment of cardiovascular conditions
ES2431518T3 (en) 2002-02-06 2013-11-26 Sanbio Inc. Differentiation / induction method of bone marrow stromal cells in nerve cells by Notch gene transfer
AU2003220424A1 (en) * 2002-03-19 2003-10-08 Advanced Research And Technology Transfer Adipose stromal stem cells for tissue and vascular modification
US7048922B2 (en) * 2002-05-29 2006-05-23 Demao Yang Stimulation of hematopoiesis by ex vivo activated immune cells
US7332158B2 (en) 2002-05-29 2008-02-19 Demao Yang Compositions and treatments for myelosuppression by ex vivo activated immune cells
US7892745B2 (en) 2003-04-24 2011-02-22 Xdx, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders 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
US8491883B2 (en) * 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical 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
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US8703121B2 (en) * 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
EP2450433A3 (en) * 2003-10-08 2012-08-08 Vet-Stem Inc Methods of preparing and using stem cell compositions and kits comprising the same
CA2550961A1 (en) 2003-12-25 2005-07-14 Kanazawa University Technology Licensing Organization Ltd. Induction of myocardial cell with the use of mammalian bone marrow cell or cord blood-origin cell and fat tissue
EP1550715A1 (en) * 2003-12-30 2005-07-06 Bionethos Holding Gmbh Method for the regeneration of tissue
RU2392314C2 (en) 2003-12-30 2010-06-20 Аугустинус БАДЕР Tissue regeneration method
JPWO2005093083A1 (en) * 2004-03-29 2008-02-14 和久 前田 Disease prediction method and use method thereof
JP2008505650A (en) * 2004-07-12 2008-02-28 ソリン・グループ・イタリア・ソシエタ・ア・レスポンサビリタ・リミタータ Apparatus and method for growing human cells
US20060045872A1 (en) 2004-08-25 2006-03-02 Universidad Autonoma De Madrid Ciudad Universitaria de Cantoblanco Use of adipose tissue-derived stromal stem cells in treating fistula
WO2006029184A2 (en) 2004-09-08 2006-03-16 Expression Diagnostics, Inc. Genes useful for diagnosing and monitoring inflammation related disorders
ES2490610T3 (en) * 2004-12-08 2014-09-04 Shire Regenerative Medicine, Inc. Materials and methods for minimally invasive administration of a fluid composition containing cells
WO2006083394A2 (en) * 2004-12-21 2006-08-10 Ethicon, Inc. Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
US20060171930A1 (en) * 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
JP5340599B2 (en) 2004-12-23 2013-11-13 エシコン・インコーポレイテッド Umbilical tissue-derived postpartum cells and methods for producing and using the same
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
ES2436401T3 (en) 2005-03-07 2013-12-30 Sanbio, Inc. Use of neuronal precursor cells for the treatment of lesions of the central nervous system
US20080187518A1 (en) * 2005-05-25 2008-08-07 University Of Virginia Patent Foundation Production of Osteoclasts from Adipose Tissues
US7531355B2 (en) * 2005-07-29 2009-05-12 The Regents Of The University Of California Methods and compositions for smooth muscle reconstruction
US20070027543A1 (en) * 2005-08-01 2007-02-01 Gimble Jeffrey M Use of adipose tissue-derived stromal cells in spinal fusion
US20070212336A1 (en) * 2005-10-31 2007-09-13 Fulkerson Loren D Methods for harvesting and storing autologous stem cells including blood derived hematopoietic stem cells and adipose derived mesenchymal stem cells
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
US9175261B2 (en) * 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
ES2391034T3 (en) * 2005-12-19 2012-11-20 Ethicon, Inc. In vitro expansion of postpartum derived cells in rotary bottles
US20070160588A1 (en) * 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US20070207504A1 (en) * 2006-03-06 2007-09-06 The Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Proteomic methods for the identification of differentiated adipose cells and adipose derived adult stem cells
ES2537641T3 (en) 2006-03-23 2015-06-10 Pluristem Ltd. Methods of cell expansion and uses of cells and conditioned media produced in this way for therapy
EP2019859A4 (en) * 2006-04-25 2009-11-25 Univ Louisiana State Adipose derived adult stem cells in hepatic regeneration
CN101490246B (en) * 2006-05-17 2014-02-19 同源治疗公司 Isolation and purification of hematopoietic stem cells from post-liposuction lipoaspirates
FR2901136B1 (en) * 2006-05-18 2010-10-01 Centre Nat Rech Scient USE OF CELLS DERIVED FROM ADIPOSE TISSUE FOR THE PREPARATION OF AN ANTI-TUMOR DRUG
US7993832B2 (en) 2006-08-14 2011-08-09 Xdx, Inc. Methods and compositions for diagnosing and monitoring the status of transplant rejection and immune disorders
WO2008140484A2 (en) 2006-11-09 2008-11-20 Xdx, Inc. Methods for diagnosing and monitoring the status of systemic lupus erythematosus
EP2607477B1 (en) 2007-05-03 2020-09-23 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US8574567B2 (en) * 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
US8273091B2 (en) * 2007-10-04 2012-09-25 Ebi, Llc Alignment device for locking nail
CN102036688B (en) * 2007-10-05 2014-07-02 伊西康公司 Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
WO2009076548A1 (en) * 2007-12-13 2009-06-18 Cytori Therapeutics, Inc. Methods of inhibiting tumor development using adipose-derived regenerative cells
US8236538B2 (en) * 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
CN102282250A (en) 2008-05-22 2011-12-14 维斯塔治疗公司 Method of differentiating mammalian progenitor cells into insulin producing pancreatic islet cells
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
EP2321407A4 (en) * 2008-09-11 2012-07-18 Univ Florida System and method for producing t cells
US8446586B2 (en) * 2008-10-15 2013-05-21 Allan Yang Wu Method and apparatus for increasing adipose vascular fraction
US9192695B2 (en) * 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
US20130302283A1 (en) 2012-05-14 2013-11-14 Advanced Technologies And Regenerative Medicine, Llc hUTC MODULATION OF PRO-INFLAMMATORY MEDIATORS 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
AU2009327384B2 (en) 2008-12-19 2014-07-10 DePuy Synthes Products, LLC Treatment of lung and pulmonary diseases and disorders
DK2379087T3 (en) 2008-12-19 2014-11-10 Depuy Synthes Products Llc Cells derived from umbilical cord tissue for the treatment of neuropathic pain and spasticity
AU2009327383B2 (en) * 2008-12-19 2014-08-28 DePuy Synthes Products, LLC Regeneration and repair of neural tissue following injury
EP2411504B1 (en) 2009-03-26 2017-05-10 DePuy Synthes Products, Inc. Human umbilical cord tissue cells as therapy for alzheimer's disease
EP2424463B1 (en) 2009-05-01 2016-12-28 Bimini Technologies LLC Systems, methods and compositions for optimizing tissue and cell enriched grafts
US20110027239A1 (en) * 2009-07-29 2011-02-03 Tissue Genesis, Inc. Adipose-derived stromal cells (asc) as delivery tool for treatment of cancer
WO2013106655A1 (en) 2012-01-11 2013-07-18 The Gid Group, Inc. Method for processing adipose tissue and processing apparatus
US9296984B2 (en) 2010-07-09 2016-03-29 The Gid Group, Inc. Tissue processing apparatus and method for processing adipose tissue
WO2014039697A1 (en) 2012-09-06 2014-03-13 The Gid Group, Inc. Tissue processing apparatus and method for processing adipose tissue
US9206387B2 (en) 2010-07-09 2015-12-08 The Gid Group, Inc. Method and apparatus for processing adipose tissue
EP3366327B1 (en) 2010-07-09 2022-09-21 GID BIO, Inc. Apparatus and methods relating to collecting and processing human biological material containing adipose
US9725689B2 (en) 2010-10-08 2017-08-08 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
CN103443272A (en) 2010-12-27 2013-12-11 智能细胞生物科学有限公司 Ultrasonic cavitation derived stromal or mesenchymal vascular extracts and cells derived therefrom obtained from adipose tissue and use thereof
KR101835917B1 (en) 2011-03-22 2018-03-07 플루리스템 리미티드 Methods for treating radiation or chemical injury
US9611513B2 (en) 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
US9650608B2 (en) 2013-02-22 2017-05-16 Medivet America, Llc Activating adipose-derived stem cells for transplantation
WO2014150784A1 (en) 2013-03-15 2014-09-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
EP3038629B1 (en) 2013-09-05 2020-11-18 GID BIO, Inc. Method for processing adipose tissue
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
EP3122866B1 (en) 2014-03-25 2019-11-20 Terumo BCT, Inc. Passive replacement of media
EP3198006B1 (en) 2014-09-26 2021-03-24 Terumo BCT, Inc. Scheduled feed
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
GB201604304D0 (en) 2016-03-14 2016-04-27 Tigenix S A U Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in crohn's disease
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
MA45623A (en) 2016-07-05 2019-05-15 Janssen Biotech Inc TREATMENT OF RETINAL VASCULAR DISEASE USING PROGENITOR CELLS
AU2017319140B2 (en) 2016-08-30 2022-11-17 Lifecell Corporation Systems and methods for medical device control
USD851777S1 (en) 2017-01-30 2019-06-18 Lifecell Corporation Canister-type device for tissue processing
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
WO2019018002A1 (en) 2017-07-18 2019-01-24 The Gid Group, Inc. Adipose tissue digestion system and tissue processing method
EP3778875A1 (en) * 2019-08-14 2021-02-17 MaxiVax SA Immortalized myoblast cell lines and uses thereof
CN111518752B (en) * 2020-04-07 2021-12-10 北京大学 Application of vitamin D3 and analogues thereof in promoting differentiation of human skin fibroblasts into adipocytes

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807999A (en) * 1993-02-01 1998-09-15 Mt. Sinai School Of Medicine Of The City University Of New York Monoclonal antibody that distinguishes between phosphorylated and nonphosphorylated histone H1 and uses therefor
US5914234A (en) * 1994-07-08 1999-06-22 The Johns Hopkins University School Of Medicine Methods of detecting growth differentiation factor-11
US20100204104A1 (en) * 2007-06-15 2010-08-12 Huawei Qiu Fusion proteins containing two tgf-beta binding domains of tgf-beta type ii receptor

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4485096A (en) * 1982-02-26 1984-11-27 Massachusetts Institute Of Technology Tissue-equivalent and method for preparation thereof
US4628027A (en) * 1982-05-19 1986-12-09 Molecular Engineering Associates, Ltd. Vitro diagnostic methods using monoclonal antibodies against connective tissue proteins
US5863531A (en) 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US5763266A (en) * 1989-06-15 1998-06-09 The Regents Of The University Of Michigan Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5139481A (en) * 1991-08-07 1992-08-18 The General Hospital Corporation Treatment for type II diabetes
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young Scar inhibitory factor and use thereof
US5275826A (en) * 1992-11-13 1994-01-04 Purdue Research Foundation Fluidized intestinal submucosa and its use as an injectable tissue graft
GB9312819D0 (en) * 1993-06-22 1993-08-04 Univ Mcgill Acylation stimulating protein
US5599703A (en) * 1993-10-28 1997-02-04 The United States Of America As Represented By The Secretary Of The Navy In vitro amplification/expansion of CD34+ stem and progenitor cells
US6103522A (en) 1994-07-20 2000-08-15 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoiesis
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US7008634B2 (en) * 1995-03-03 2006-03-07 Massachusetts Institute Of Technology Cell growth substrates with tethered cell growth effector molecules
US6653134B2 (en) 1995-03-28 2003-11-25 Cp Hahnemann University Isolated stromal cells for use in the treatment of diseases of the central nervous system
AU713280B2 (en) 1995-11-16 1999-11-25 Case Western Reserve University In vitro chondrogenic induction of human mesenchymal stem cells
US6200606B1 (en) 1996-01-16 2001-03-13 Depuy Orthopaedics, Inc. Isolation of precursor cells from hematopoietic and nonhematopoietic tissues and their use in vivo bone and cartilage regeneration
AU5436998A (en) 1996-11-15 1998-06-03 Osiris Therapeutics, Inc. MSC-megakaryocyte precursor composition and method of isolating MSCs asso ciated with isolated megakaryocytes by isolating megakaryocytes
AU8243798A (en) 1997-07-16 1999-02-10 Kirin Beer Kabushiki Kaisha Agm-derived stroma cells
CA2312847C (en) * 1997-12-02 2014-09-30 Zen Bio, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US6100047A (en) 1999-04-07 2000-08-08 Zen Bio, Inc. Modulation of the sulfonylurea receptor and calcium in adipocytes for treatment of obesity/diabetes
DE69936720T2 (en) 1998-05-29 2008-04-30 Osiris Therapeutics, Inc. HUMAN CD45 + AND / OR FIBROBLASTES + MESENCHYMAL STEM CELLS
CA2329519A1 (en) 1998-06-08 1999-12-16 Osiris Therapeutics, Inc. In vitro maintenance of hematopoietic stem cells
US6113574A (en) 1998-07-27 2000-09-05 Spinello; Ronald P. Anesthetic injection apparatus and methods
US6153432A (en) 1999-01-29 2000-11-28 Zen-Bio, Inc Methods for the differentiation of human preadipocytes into adipocytes
KR100968165B1 (en) 1999-03-10 2010-07-06 더 리전츠 오브 더 유니버시티 오브 캘리포니아 Adipose-derived stem cells and lattices
US6316247B1 (en) 1999-06-15 2001-11-13 University Of Pittsburgh System and method for refining liposuctioned adipose tissue

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807999A (en) * 1993-02-01 1998-09-15 Mt. Sinai School Of Medicine Of The City University Of New York Monoclonal antibody that distinguishes between phosphorylated and nonphosphorylated histone H1 and uses therefor
US5914234A (en) * 1994-07-08 1999-06-22 The Johns Hopkins University School Of Medicine Methods of detecting growth differentiation factor-11
US20100204104A1 (en) * 2007-06-15 2010-08-12 Huawei Qiu Fusion proteins containing two tgf-beta binding domains of tgf-beta type ii receptor

Also Published As

Publication number Publication date
CN1309180A (en) 2001-08-22
US8486700B2 (en) 2013-07-16
US20130288262A1 (en) 2013-10-31
EP1077254A3 (en) 2002-02-06
US6555374B1 (en) 2003-04-29
EP1077254A2 (en) 2001-02-21
US20110250590A1 (en) 2011-10-13
CA2316400A1 (en) 2001-02-19
JP2001103963A (en) 2001-04-17
CA2316400C (en) 2014-06-03
JP4180228B2 (en) 2008-11-12
US20030166278A1 (en) 2003-09-04

Similar Documents

Publication Publication Date Title
US8486700B2 (en) Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US7078230B2 (en) Adipose tissue-derived stromal cell that expresses characteristics of a neuronal cell
US9255249B2 (en) Isolation and purification of hematopoietic stem cells from post-liposuction lipoaspirates
Deasy et al. Long-term self-renewal of postnatal muscle-derived stem cells
JP6764912B2 (en) Cell population with immunomodulatory activity, its preparation method and use
US9415072B2 (en) Expansion of haemopoietic precursors
AU2001238695A1 (en) Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof
KR20140127209A (en) Mesenchymal stromal cells and uses related thereto
US20110182866A1 (en) Isolation of stem cell precursors and expansion in non-adherent conditions
Leijs et al. Encapsulation of allogeneic mesenchymal stem cells in alginate extends local presence and therapeutic function
EP3336176A1 (en) Improved umbilical cord-derived adhesive stem cells, preparation method therefor, and use thereof
EP1918366A1 (en) Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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