US20100173411A1 - Adipose-derived stem cells and lattices - Google Patents

Adipose-derived stem cells and lattices Download PDF

Info

Publication number
US20100173411A1
US20100173411A1 US12/653,940 US65394009A US2010173411A1 US 20100173411 A1 US20100173411 A1 US 20100173411A1 US 65394009 A US65394009 A US 65394009A US 2010173411 A1 US2010173411 A1 US 2010173411A1
Authority
US
United States
Prior art keywords
cell
cells
medium
lipo
derived
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/653,940
Inventor
Adam J. Katz
Ramon Llull
J. William Futrell
Marc H. Hedrick
Prosper Benhaim
Hermann Peter Lorenz
Min Zhu
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.)
Individual
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=32854187&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100173411(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from PCT/US2000/006232 external-priority patent/WO2000053795A1/en
Application filed by Individual filed Critical Individual
Priority to US12/653,940 priority Critical patent/US20100173411A1/en
Publication of US20100173411A1 publication Critical patent/US20100173411A1/en
Priority to US13/226,338 priority patent/US20110318833A1/en
Priority to US13/852,463 priority patent/US20130224858A1/en
Priority to US14/659,308 priority patent/US20150191698A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • 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
    • C12N5/0068General culture methods using substrates
    • 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
    • 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
    • C12N5/0654Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0655Chondrocytes; Cartilage
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0658Skeletal muscle cells, e.g. myocytes, myotubes, myoblasts
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/42Organic phosphate, e.g. beta glycerophosphate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/01Modulators of cAMP or cGMP, e.g. non-hydrolysable analogs, phosphodiesterase inhibitors, cholera toxin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • C12N2502/1305Adipocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells
    • C12N2506/1384Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells from adipose-derived stem cells [ADSC], from adipose stromal stem cells
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • mesenchymal stem cells chiefly obtained from bone marrow, has led to advances in tissue regrowth and differentiation.
  • Such cells are pluripotent cells found in bone marrow and periosteum, and they are capable of differentiating into various mesenchymal or connective tissues.
  • bone-marrow derived stem cells can be induced to develop into myocytes upon exposure to agents such as 5-azacytidine (Wakitani et al., Muscle Nerve, 18(12), 1417-26 (1995)). It has been suggested that such cells are useful for repair of tissues such as cartilage, fat, and bone (see, e.g., U.S. Pat. Nos.
  • a suitable lattice material is secreted extracellular matrix material isolated from tumor cell lines (e.g., Engelbreth-Holm-Swarm tumor secreted matrix—“matrigel”).
  • This material contains type IV collagen and growth factors, and provides an excellent substrate for cell growth (see, e.g., Vukicevic et al., Exp. Cell Res, 202(1), 1-8 (1992)). However, as this material also facilitates the malignant transformation of some cells (see, e.g., Fridman, et al., Int. J. Cancer, 51(5), 740-44 (1992)), it is not suitable for clinical application. While other artificial lattices have been developed, these can prove toxic either to cells or to patients when used in vivo. Accordingly, there remains a need for a lattice material suitable for use as a substrate in culturing and growing populations of cells.
  • the present invention provides adipose-derived stem cells and lattices.
  • the present invention provides a lipo-derived stem cell substantially free of adipocytes and red blood cells and clonal populations of connective tissue stem cells.
  • the cells can be employed, alone or within biologically-compatible compositions, to generate differentiated tissues and structures, both in vivo and in vitro. Additionally, the cells can be expanded and cultured to produce hormones and to provide conditioned culture media for supporting the growth and expansion of other cell populations.
  • the present invention provides a lipo-derived lattice substantially devoid of cells, which includes extracellular matrix material from adipose tissue. The lattice can be used as a substrate to facilitate the growth and differentiation of cells, whether in vivo or in vitro, into anlagen or even mature tissues or structures.
  • the inventive cells and lattice represent a ready source of pluripotent stem cells. Moreover, because the cells can be passaged in culture in an undifferentiated state under culture conditions not requiring prescreened lots of serum, the inventive cells can be maintained with considerably less expense than other types of stem cells.
  • the stem cell is substantially free of other cell types (e.g., adipocytes, red blood cells, other stromal cells, etc.) and extracellular matrix material; more preferably, the stem cell is completely free of such other cell types and matrix material.
  • the inventive cell is derived from the adipose tissue of a primate, and more preferably a higher primate (e.g., a baboon or ape).
  • the inventive cell will be derived from human adipose tissue, using methods such as described herein.
  • inventive cell can be any type of stem cell, for use in tissue engineering, desirably the cell is of mesodermal origin. Typically such cells, when isolated, retain two or more mesodermal or mesenchymal developmental phenotypes (i.e., they are pluripotent).
  • such cells generally have the capacity to develop into mesodermal tissues, such as mature adipose tissue, bone, various tissues of the heart (e.g., pericardium, epicardium, epimyocardium, myocardium, pericardium, valve tissue, etc.), dermal connective tissue, hemangial tissues (e.g., corpuscles, endocardium, vascular epithelium, etc.), muscle tissues (including skeletal muscles, cardiac muscles, smooth muscles, etc.), urogenital tissues (e.g., kidney, pronephros, meta- and meso-nephric ducts, metanephric diverticulum, ureters, renal pelvis, collecting tubules, epithelium of the female reproductive structures (particularly the oviducts, uterus, and vagina)), pleural and peritoneal tissues, viscera, mesodermal glandular tissues (e.g., adrenal cortex tissues), and stromal tissues (e.g., hem
  • the cell can retain potential to develop into mature cells, it also can realize its developmental phenotypic potential by differentiating into an appropriate precursor cell (e.g., a preadipocyte, a premyocyte, a preosteocyte, etc.). Also, depending on the culture conditions, the cells can also exhibit developmental phenotypes such as embryonic, fetal, hematopoetic, neurogenic, or neuralgiagenic developmental phenotypes.
  • an appropriate precursor cell e.g., a preadipocyte, a premyocyte, a preosteocyte, etc.
  • the cells can also exhibit developmental phenotypes such as embryonic, fetal, hematopoetic, neurogenic, or neuralgiagenic developmental phenotypes.
  • the inventive cell can have two or more developmental phenotypes such as adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, splanchogenic, and stromal developmental phenotypes.
  • developmental phenotypes such as adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, splanchogenic, and stromal developmental phenotypes.
  • While such cells can retain two or more of these developmental phenotypes, preferably, such cells have three or more such developmental phenotypes (e.g., four or more mesodermal or mesenchymal developmental phenotypes), and some types of inventive stem cells have a potential to acquire any mesodermal phenotype through the process of differentiation.
  • the inventive stem cell can be obtained from adipose tissue by any suitable method.
  • a first step in any such method requires the isolation of adipose tissue from the source animal.
  • the animal can be alive or dead, so long as adipose stromal cells within the animal are viable.
  • human adipose stromal cells are obtained from living donors, using well-recognized protocols such as surgical or suction lipectomy. Indeed, as liposuction procedures are so common, liposuction effluent is a particularly preferred source from which the inventive cells can be derived.
  • the adipose tissue is processed to separate stem cells from the remainder of the material.
  • the adipose tissue is washed with physiologically-compatible saline solution (e.g., phosphate buffered saline (PBS)) and then vigorously agitated and left to settle, a step that removes loose matter (e.g., damaged tissue, blood, erythrocytes, etc.) from the adipose tissue.
  • physiologically-compatible saline solution e.g., phosphate buffered saline (PBS)
  • PBS phosphate buffered saline
  • the washing and settling steps generally are repeated until the supernatant is relatively clear of debris.
  • the remaining cells generally will be present in lumps of various size, and the protocol proceeds using steps gauged to degrade the gross structure while minimizing damage to the cells themselves.
  • One method of achieving this end is to treat the washed lumps of cells with an enzyme that weakens or destroys bonds between cells (e.g., collagenase, dispase, trypsin, etc.). The amount and duration of such enzymatic treatment will vary, depending on the conditions employed, but the use of such enzymes is generally known in the art.
  • the lumps of cells can be degraded using other treatments, such as mechanical agitation, sonic energy, thermal energy, etc. If degradation is accomplished by enzymatic methods, it is desirable to neutralize the enzyme following a suitable period, to minimize deleterious effects on the cells.
  • the degradation step typically produces a slurry or suspension of aggregated cells (generally liposomes) and a fluid fraction containing generally free stromal cells (e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells).
  • stromal cells e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells.
  • the next stage in the separation process is to separate the aggregated cells from the stromal cells. This can be accomplished by centrifugation, which forces the stromal cells into a pellet covered by a supernatant. The supernatant then can be discarded and the pellet suspended in a physiologically-compatible fluid.
  • the suspended cells typically include erythrocytes, and in most protocols it is desirable to lyse these.
  • erythrocytes Methods for selectively lysing erythrocytes are known in the art, and any suitable protocol can be employed (e.g., incubation in a hyper- or hypotonic medium).
  • any suitable protocol can be employed (e.g., incubation in a hyper- or hypotonic medium).
  • the remaining cells should then be separated from the lysate, for example by filtration or centrifugation.
  • the suspended cells can be washed, re-centrifuged, and resuspended one or more successive times to achieve greater purity.
  • the cells can be separated using a cell sorter or on the basis of cell size and granularity, stem cells being relatively small and agranular.
  • telomerase can also serve as a stem cell-specific marker. They can also be separated immunohistochemically, for example, by panning or using magnetic beads. Any of the steps and procedures for isolating the inventive cells can be performed manually, if desired. Alternatively, the process of isolating such cells can be facilitated through a suitable device, many of which are known in the art (see, e.g., U.S. Pat. No. 5,786,207).
  • the cells can be cultured and, if desired, assayed for number and viability to assess the yield.
  • the cells can be cultured without differentiation using standard cell culture media (e.g., DMEM, typically supplemented with 5-15% (e.g., 10%) serum (e.g., fetal bovine serum, horse serum, etc.).
  • DMEM standard cell culture media
  • serum e.g., fetal bovine serum, horse serum, etc.
  • the cells can be passaged at least five times in such medium without differentiating, while still retaining their developmental phenotype, and more preferably, the cells can be passaged at least 10 times (e.g., at least 15 times or even at least 20 times) without losing developmental phenotype.
  • culturing the cells of the present invention without inducing differentiation can be accomplished without specially screened lots of serum, as is generally the case for mesenchymal stem cells (e.g., derived from marrow).
  • mesenchymal stem cells e.g., derived from marrow.
  • Methods for measuring viability and yield are known in the art (e.g., trypan blue exclusion).
  • the stem cells are further separated by phenotypic identification, to identify those cells that have two or more of the aforementioned developmental phenotypes.
  • the stromal cells are plated at a desired density such as between about 100 cells/cm 2 to about 100,000 cells/cm 2 (such as about 500 cells/cm 2 to about 50,000 cells/cm 2 , or, more particularly, between about 1,000 cells/cm 2 to about 20,000 cells/cm 2 ). If plated at lower densities (e.g., about 300 cells/cm 2 ), the cells can be more easily clonally isolated. For example, after a few days, cells plated at such densities will proliferate into a population.
  • Such cells and populations can be clonally expanded, if desired, using a suitable method for cloning cell populations.
  • a proliferated population of cells can be physically picked and seeded into a separate plate (or the well of a multi-well plate).
  • the cells can be subcloned onto a multi-well plate at a statistical ratio for facilitating placing a single cell into each well (e.g., from about 0.1 to about 1 cell/well or even about 0.25 to about 0.5 cells/well, such as 0.5 cells/well).
  • the cells can be cloned by plating them at low density (e.g., in a petri-dish or other suitable substrate) and isolating them from other cells using devices such as a cloning rings.
  • clones can be obtained by permitting the cells to grow into a monolayer and then shielding one and irradiating the rest of cells within the monolayer. The surviving cell then will grow into a clonal population.
  • a preferred culture condition for cloning stem cells is about 2 ⁇ 3 F 12 medium+20% serum (preferably fetal bovine serum) and about 1 ⁇ 3 standard medium that haw been conditioned with stromal cells (e.g., cells from the stromal vascular fraction of liposuction aspirate), the relative proportions being determined volumetrically).
  • the isolated cells can be cultured to a suitable point when their developmental phenotype can be assessed.
  • the inventive cells have at least two of the aforementioned developmental phenotypes.
  • one or more cells drawn from a given clone can be treated to ascertain whether it possesses such developmental potentials.
  • One type of treatment is to culture the inventive cells in culture media that has been conditioned by exposure to mature cells (pr precursors thereof) of the respective type to be differentiated (e.g., media conditioned by exposure to myocytes can induce myogenic differentiation, media conditioned by exposure to heart valve cells can induce differentiation into heart valve tissue, etc.).
  • defined media for inducing differentiation also can be employed.
  • adipogenic developmental phenotype can be assessed by exposing the cell to a medium that facilitates adipogenesis, e.g., containing a glucocorticoid (e.g., isobutyl-methylxanthine, dexamethasone, hydrocortisone, cortisone, etc.), insulin, a compound which elevates intracellular levels of cAMP (e.g., dibutyryl-cAMP, 8-CPT-cAMP (8-(4)chlorophenylthio)-adenosine 3′,5′ cyclic monophosphate; 8-bromo-cAMP; dioctanoyl-cAMP, forskolin etc.), and/or a compound which inhibits degradation of cAMP (e.g., a phosphodiesterase inhibitor such as methyl isobutylxanthine, theophylline, caffeine, indomethacin, and the like).
  • a glucocorticoid
  • adipogenic differentiation can induce adipogenic differentiation.
  • a medium also can include other agents, such as indomethicin (e.g., about 100 ⁇ M to about 200 ⁇ M), if desired, and preferably the medium is serum free.
  • Osteogenic developmental phenotype can be assessed by exposing the cells to between about 10 ⁇ 7 M and about 10 ⁇ 9 M dexamethasone (e.g., about 1 ⁇ M) in combination with about 10 ⁇ M to about 50 ⁇ M ascorbate-2-phosphate and between about 10 nM and about 50 nM ⁇ -glycerophosphate, and the medium also can include serum (e.g., bovine serum, horse serum, etc.).
  • Myogenic differentiation can be induced by exposing the cells to between about 10 ⁇ M and about 100 ⁇ M hydrocortisone, preferably in a serum-rich medium (e.g., containing between about 10% and about 20% serum (either bovine, horse, or a mixture thereof)).
  • Chondrogenic differentiation can be induced by exposing the cells to between about 1 ⁇ M to about 10 ⁇ M insulin and between about 1 ⁇ M to about 10 ⁇ M transferrin, between about 1 ng/ml and 10 ng/ml transforming growth factor (TGF) ⁇ 1, and between about 10 nM and about 50 nM ascorbate-2-phosphate (50 nM).
  • TGF transforming growth factor
  • the cells are cultured in high density (e.g., at about several million cells/ml or using micromass culture techniques), and also in the presence of low amounts of serum (e.g., from about 1% to about 5%).
  • the cells also can be induced to assume a developmentally more immature phenotype (e.g., a fetal or embryonic phenotype). Such induction is achieved upon exposure of the inventive cell to conditions that mimic those within fetuses and embryos.
  • the inventive cells or populations can be co-cultured with cells isolated from fetuses or embryos, or in the presence of fetal serum.
  • the cells can be induced to differentiate into any of the aforementioned mesodermal lineages by co-culturing them with mature cells of the respective type, or precursors thereof.
  • myogenic differentiation can be induced by culturing the inventive cells with myocytes or precursors, and similar results can be achieved with respect to the other tissue types mentioned herein.
  • Other methods of inducing differentiation are known in the art, and many of them can be employed, as appropriate.
  • the cells After culturing the cells in the differentiating-inducing medium for a suitable time (e.g., several days to a week or more), the cells can be assayed to determine whether, in fact, they have differentiated to acquire physical qualities of a given type of cell.
  • One measurement of differentiation per se is telomere length, undifferentiated stem cells having longer telomeres than differentiated cells; thus the cells can be assayed for the level of telomerase activity.
  • RNA or proteins can be extracted from the cells and assayed (via Northern hybridization, rtPCR, Western blot analysis, etc.) for the presence of markers indicative of the desired phenotype.
  • the cells can be assayed immunohistochemically or stained, using tissue-specific stains.
  • tissue-specific stains e.g., oil red O, safarin red, sudan black, etc.
  • adipose-related factors e.g., type IV collagen, PPAR- ⁇ , adipsin, lipoprotein lipase, etc.
  • ostogenesis can be assessed by staining the cells with bone-specific stains (e.g., alkaline phosphatase, von Kossa, etc.) or probed for the presence of bone-specific markers (e.g., osteocalcin, osteonectin, osteopontin, type I collagen, bone morphogenic proteins, cbfa, etc.).
  • bone-specific markers e.g., osteocalcin, osteonectin, osteopontin, type I collagen, bone morphogenic proteins, cbfa, etc.
  • Myogensis can be assessed by identifying classical morphologic changes (e.g., polynucleated cells, syncitia formation, etc.), or assessed biochemically for the presence of muscle-specific factors (e.g., myo D, myosin heavy chain, NCAM, etc.).
  • Chondrogenesis can be determined by staining the cells using cartallge-specific stains (e.g., alcian blue) or probing the cells for the expression/production of cartilage-specific molecules (e.g., sulfated glycosaminoglycans and proteoglycans (e.g., keratin, chondroitin, etc.) in the medium, type II collagen, etc.).
  • cartilage-specific molecules e.g., sulfated glycosaminoglycans and proteoglycans (e.g., keratin, chondroitin, etc.) in the medium, type II collagen, etc.
  • Other methods of assessing developmental phenotype are known in the art, and any of them is appropriate.
  • the cells can be sorted by size and granularity.
  • the cells can be used to generate monoclonal antibodies, which can then be employed to assess whether they preferentially bind to a given cell type. Correlation of antigenicity can confirm that
  • the cell can be solitary and isolated from other cells, preferably it is within a population of cells, and the invention provides a defined population including the inventive cell.
  • the population is heterogeneous.
  • the population can include support cells for supplying factors to the inventive cells.
  • the inventive stem cells can themselves serve as support cells for culturing other types of cells (such as other types of stem cells, e.g., as neural stem cells (NSC), hematopoetic stem cells (HPC, particularly CD34 + stem cells), embryonic stem cells (ESC) and mixtures thereof), and the population can include such cells.
  • the population is substantially homogeneous, consisting essentially of the inventive lipo-derived stem cells.
  • the inventive cells can be cloned, a substantially homogeneous population containing them can be clonal.
  • the invention also pertains to any defined clonal cell population consisting essentially of mesodermal stem cells, connective tissue stem cell, or mixtures thereof.
  • the cells can be lipo-derived or derived from other mesodermal or connective cell tissues (e.g., bone marrow, muscle, etc.) using methods known in the art. After the isolation, the cells can be expanded clonally as described herein.
  • the inventive cells can be employed for a variety of purposes.
  • the cells can support the growth and expansion of other cell types, and the invention pertains to methods for accomplishing this.
  • the invention pertains to a method of conditioning culture medium using the inventive stem cells and to conditioned medium produced by such a method.
  • the medium becomes conditioned upon exposing a desired culture medium to the cells under conditions sufficient for the cells to condition it.
  • the medium is used to support the growth of the inventive cells, which secrete hormones, cell matrix material, and other factors into the medium. After a suitable period (e.g., one or a few days), the culture medium containing the secreted factors can be separated from the cells and stored for future use.
  • inventive cells and populations can be re-used successively to condition medium, as desired.
  • the cells can remain within the conditioned medium.
  • the invention provides a conditioned medium obtained using this method, which either can contain the inventive cells or be substantially free of the inventive cells, as desired.
  • the conditioned medium can be used to support the growth and expansion of desired cell types, and the invention provides a method of culturing cells (particularly stem cells) using the conditioned medium.
  • the method involves maintaining a desired cell in the conditioned medium under conditions for the cell to remain viable.
  • the cell can be maintained under any suitable condition for culturing them, such as are known in the art.
  • the method permits successive rounds of mitotic division of the cell to form an expanded population.
  • the exact conditions e.g., temperature, CO 2 levels, agitation, presence of antibiotics, etc.
  • the medium it is desirable for the medium to be substantially free of the lipo-derived cells employed to condition the medium as described herein.
  • the inventive lipo-derived cells can express cell-surface mediators of intercellular communication, it often is desirable for the inventive cells and the desired other cells to be co-cultured under conditions in which the two cell types are in contact. This can be achieved, for example, by seeding the cells as a heterogeneous population of cells onto a suitable culture substrate.
  • the inventive lipo-derived cells can first be grown to confluence, which will serve as a substrate for the second desired cells to be cultured within the conditioned medium.
  • the inventive lipo-derived cells can be genetically modified, e.g., to express exogenous genes or to repress the expression of endogenous genes, and the invention provides a method of genetically modifying such cells and populations.
  • the cell is exposed to a gene transfer vector comprising a nucleic acid including a transgene, such that the nucleic acid is introduced into the cell under conditions appropriate for the transgene to be expressed within the cell.
  • the transgene generally is an expression cassette, including a coding polynucleotide operably linked to a suitable promoter.
  • the coding polynucleotide can encode a protein, or it can encode biologically active RNA (e.g., antisense RNA or a ribozyme).
  • the coding polynucleotide can encode a gene conferring resistance to a toxin, a hormone (such as peptide growth hormones, hormone releasing factors, sex hormones, adrenocorticotrophic hormones, cytokines (e.g., interferins, interleukins, lymphokines), etc.), a cell-surface-bound intracellular signaling moiety (e.g., cell adhesion molecules, hormone receptors, etc.), a factor promoting a given lineage of differentiation, etc.
  • a hormone such as peptide growth hormones, hormone releasing factors, sex hormones, adrenocorticotrophic hormones, cytokines (e.g., interferins, interleukins, lymphokines), etc.
  • cytokines
  • the coding polynucleotide is operably linked to a suitable promoter.
  • suitable promoters include prokaryotic promoters and viral promoters (e.g., retroviral ITRs, LTRs, immediate early viral promoters (IEp), such as herpesvirus IEp (e.g., ICP4-IEp and ICP0-IEp), cytomegalovirus (CMV) IEp, and other viral promoters, such as Rous Sarcoma Virus (RSV) promoters, and Murine Leukemia Virus (MLV) promoters).
  • IEp immediate early viral promoters
  • IEp such as herpesvirus IEp (e.g., ICP4-IEp and ICP0-IEp)
  • CMV cytomegalovirus
  • RSV Rous Sarcoma Virus
  • MMV Murine Leukemia Virus
  • promoters are eukaryotic promoters, such as enhancers (e.g., the rabbit ⁇ -globin regulatory elements), constitutively active promoters (e.g., the ⁇ -actin promoter, etc.), signal specific promoters (e.g., inducible promoters such as a promoter responsive to RU486, etc.), and tissue-specific promoters. It is well within the skill of the art to select a promoter suitable for driving gene expression in a predefined cellular context.
  • enhancers e.g., the rabbit ⁇ -globin regulatory elements
  • constitutively active promoters e.g., the ⁇ -actin promoter, etc.
  • signal specific promoters e.g., inducible promoters such as a promoter responsive to RU486, etc.
  • tissue-specific promoters eukaryotic promoters, such as enhancers (e.g., the rabbit ⁇ -globin regulatory elements), constitutively active promoters (e
  • the expression cassette can include more than one coding polynucleotide, and it can include other elements (e.g., polyadenylation sequences, sequences encoding a membrane-insertion signal or a secretion leader, ribosome entry sequences, transcriptional regulatory elements (e.g., enhancers, silencers, etc.), and the like), as desired.
  • elements e.g., polyadenylation sequences, sequences encoding a membrane-insertion signal or a secretion leader, ribosome entry sequences, transcriptional regulatory elements (e.g., enhancers, silencers, etc.), and the like, as desired.
  • the expression cassette containing the transgene should be incorporated into a genetic vector suitable for delivering the transgene to the cells.
  • any such vector can be so employed to genetically modify the cells (e.g., plasmids, naked DNA, viruses such as adenovirus, adeno-associated virus, herpesviruses, lentiviruses, papillomaviruses, retroviruses, etc.).
  • Any method of constructing the desired expression cassette within such vectors can be employed, many of which are well known in the art (e.g., direct cloning, homologous recombination, etc.).
  • vector will largely determine the method used to introduce the vector into the cells (e.g., by protoplast fusion, calcium-phosphate precipitation, gene gun, electroporation, infection with viral vectors, etc.), which are generally known in the art.
  • the genetically altered cells can be employed as bioreactors for producing the product of the transgene.
  • the genetically modified cells are employed to deliver the transgene and its product to an animal.
  • the cells, once genetically modified can be introduced into the animal under conditions sufficient for the transgene to be expressed in vivo.
  • hormones e.g., cytokines, peptide or other (e.g., monobutyrin) growth factors, etc.
  • the cells of the present invention that secrete hormones can be used in a variety of contexts in vivo and in vitro.
  • such cells can be employed as bioreactors to provide a ready source of a given hormone, and the invention pertains to a method of obtaining hormones from such cells.
  • the cells are cultured, under suitable conditions for them to secrete the hormone into the culture medium.
  • the medium is harvested and processed to isolate the hormone from the medium.
  • Any standard method e.g., gel or affinity chromatography, dialysis, lyophilization, etc.
  • Any standard method e.g., gel or affinity chromatography, dialysis, lyophilization, etc.
  • cells (and populations) of the present invention secreting hormones can be employed as therapeutic agents.
  • such methods involve transferring the cells to desired tissue, either in vitro (e.g., as a graft prior to implantation or engrafting) or in vivo, to animal tissue directly.
  • the cells can be transferred to the desired tissue by any method appropriate, which generally will vary according to the tissue type.
  • cells can be transferred to a graft by bathing the graft (or infusing it) with culture medium continuing the cells.
  • the cells can be seeded onto the desired site within the tissue to establish a population.
  • Cells can be transferred to sites in vivo using devices such as catheters, trocars, cannulae, stents (which can be seeded with the cells), etc.
  • the cell secretes a cytokine or growth hormone such as human growth factor, fibroblast growth factor, nerve growth factor, insulin-like growth factors, hemopoietic stem cell growth factors, members of the fibroblast growth factor family, members of the platelet-derived growth factor family, vascular and endothelial cell growth factors, members of the TGFb family (including bone morphogenic factor), or enzymes specific for congenital disorders (e.g., distrophin).
  • a cytokine or growth hormone such as human growth factor, fibroblast growth factor, nerve growth factor, insulin-like growth factors, hemopoietic stem cell growth factors, members of the fibroblast growth factor family, members of the platelet-derived growth factor family, vascular and endothelial cell growth factors, members of the TGFb family (including bone morphogenic factor), or enzymes
  • the invention provides a method of promoting the closure of a wound within a patient using such cells.
  • the inventive cells secreting the hormone are transferred to the vicinity of a wound under conditions sufficient for the cell to produce the hormone.
  • the presence of the hormone in the vicinity of the wound promotes closure of the wound.
  • the method promotes closure of both external (e.g., surface) and internal wounds.
  • Wounds to which the present inventive method is useful in promoting closure include, but are not limited to, abrasions, avulsions, blowing wounds, burn wounds, contusions, gunshot wounds, incised wounds, open wounds, penetrating wounds, perforating wounds, puncture wounds, seton wounds, stab wounds, surgical wounds, subcutaneous wounds, or tangential wounds.
  • the method need not achieve complete healing or closure of the wound; it is sufficient for the method to promote any degree of wound closure. In this respect, the method can be employed alone or as an adjunct to other methods for healing wounded tissue.
  • the inventive cells secrete an angiogenic hormone (e.g., vascular growth factor, vascular and endothelial cell growth factor, etc.), they (as well as populations containing them) can be employed to induce angiogenesis within tissues.
  • an angiogenic hormone e.g., vascular growth factor, vascular and endothelial cell growth factor, etc.
  • the invention provides a method of promoting neovascularization within tissue using such cells.
  • the cell is introduced the desired tissue under conditions sufficient for the cell to produce the angiogenic hormone.
  • the presence of the hormone within the tissue promotes neovascularization within the tissue.
  • the inventive stem cells have a developmental phenotype, they can be employed in tissue engineering.
  • the invention provides a method of producing animal matter comprising maintaining the inventive cells under conditions sufficient for them to expand and differentiate to form the desired matter.
  • the matter can include mature tissues, or even whole organs, including tissue types into which the inventive cells can differentiate (as set forth herein).
  • tissue types into which the inventive cells can differentiate as set forth herein.
  • tissue types into which the inventive cells can differentiate will comprise adipose, cartilage, heart, dermal connective tissue, blood tissue, muscle, kidney, bone, pleural, splancnic tissues, vascular tissues, and the like. More typically, the matter will comprise combinations of these tissue types (i.e., more than one tissue type).
  • the matter can comprise all or a portion of an animal organ (e.g., a heart, a kidney) or a limb (e.g., a leg, a wing, an arm, a hand, a foot, etc.).
  • an animal organ e.g., a heart, a kidney
  • a limb e.g., a leg, a wing, an arm, a hand, a foot, etc.
  • the cells can divide and differentiate to produce such structures, they can also form anlagen of such structures. At early stages, such anlagen can be cryopreserved for future generation of the desired mature structure or organ.
  • the inventive cells and populations are maintained under conditions suitable for them to expand and divide to form the desired structures. In some applications, this is accomplished by transferring them to an animal (i.e., in vivo) typically at a sight at which the new matter is desired.
  • the invention can facilitate the regeneration of tissues (e.g., bone, muscle, cartilage, tendons, adipose, etc.) within an animal where the cells are implanted into such tissues.
  • the cells can be induced to differentiate and expand into tissues in vitro.
  • the cells are cultured on substrates that facilitate formation into three-dimensional structures conducive for tissue development.
  • the cells can be cultured or seeded onto a bio-compatible lattice, such as one that includes extracellular matrix material, synthetic polymers, cytokines, growth factors, etc.
  • a bio-compatible lattice such as one that includes extracellular matrix material, synthetic polymers, cytokines, growth factors, etc.
  • Such a lattice can be molded into desired shapes for facilitating the development of tissue types.
  • the medium and/or substrate is supplemented with factors (e.g., growth factors, cytokines, extracellular matrix material, etc.) that facilitate the development of appropriate tissue types and structures.
  • factors e.g., growth factors, cytokines, extracellular matrix material, etc.
  • the invention provides a composition including the inventive cells (and populations) and biologically compatible lattice.
  • the lattice is formed from polymeric material, having fibers as a mesh or sponge, typically with spaces on the order of between about 100 ⁇ m and about 300 ⁇ m.
  • Such a structure provides sufficient area on which the cells can grow and proliferate.
  • the lattice is biodegradable over time, so that it will be absorbed into the animal matter as it develops.
  • Suitable polymeric lattices can be formed from monomers such as glycolic acid, lactic acid, propyl fumarate, caprolactone, hyaluronan, hyaluronic acid, and the like.
  • Other lattices can include proteins, polysaccharides, polyhydroxy acids, polyorthoesters, polyanhydrides, polyphosphazenes, or synthetic polymers (particularly biodegradable polymers).
  • a suitable polymer for forming such lattice can include more than one monomers (e.g., combinations of the indicated monomers).
  • the lattice can also include hormones, such as growth factors, cytokines, and morphogens (e.g., retinoic acid, aracadonic acid, etc.), desired extracellular matrix molecules (e.g., fibronectin, laminin, collagen, etc.), or other materials (e.g., DNA, viruses, other cell types, etc.) as desired.
  • hormones such as growth factors, cytokines, and morphogens (e.g., retinoic acid, aracadonic acid, etc.), desired extracellular matrix molecules (e.g., fibronectin, laminin, collagen, etc.), or other materials (e.g., DNA, viruses, other cell types, etc.) as desired.
  • the cells are introduced into the lattice such that they permeate into the interstitial spaces therein.
  • the matrix can be soaked in a solution or suspension containing the cells, or they can be infused or injected into the matrix.
  • a particularly preferred composition is a hydrogel formed by crosslinking of a suspension including the polymer and also having the inventive cells dispersed therein. This method of formation permits the cells to be dispersed throughout the lattice, facilitating more even permeation of the lattice with the cells.
  • composition also can include mature cells of a desired phenotype or precursors thereof, particularly to potentate the induction of the inventive stem cells to differentiate appropriately within the lattice (e.g., as an effect of co-culturing such cells within the lattice).
  • the composition can be employed in any suitable manner to facilitate the growth and generation of the desired tissue types, structures, or anlagen.
  • the composition can be constructed using three-dimensional or stereotactic modeling techniques.
  • a layer or domain within the composition can be populated by cells primed for osteogenic differentiation, and another layer or domain within the composition can be populated with cells primed for myogenic and/or chondrogenic development. Bringing such domains into juxtaposition with each other facilitates the molding and differentiation of complex structures including multiple tissue types (e.g., bone surrounded by muscle, such as found in a limb).
  • the composition can be cultured ex vivo in a bioreactor or incubator, as appropriate.
  • the structure is implanted within the host animal directly at the site in which it is desired to grow the tissue or structure.
  • the composition can be engrafted on a host (typically an animal such as a pig, baboon, etc.), where it will grow and mature until ready for use. Thereafter, the mature structure (or strom) is excised from the host and implanted into the host, as appropriate.
  • Lattices suitable for inclusion into the composition can be derived from any suitable source (e.g., matrigel), and some commercial sources for suitable lattices exist (e.g., suitable of polyglycolic acid can be obtained from sources such as Ethicon, N.J.).
  • suitable lattice can be derived from the acellular portion of adipose tissue—i.e., adipose tissue extracellular matrix matter substantially devoid of cells, and the invention provides such a lipo-derived lattice.
  • such lipo-derived lattice includes proteins such as proteoglycans, glycoproteins, hyaluronins, fibronectins, collagens (type I, type II, type III, type IV, type V, type VI, etc.), and the like, which serve as excellent substrates for cell growth. Additionally, such lipo-derived lattices can include hormones, preferably cytokines and growth factors, for facilitating the growth of cells seeded into the matrix.
  • the lipo-derived matrix can be isolated form adipose tissue similarly as described above, except that it will be present in the acellular fraction.
  • adipose tissue or derivatives thereof e.g., a fraction of the cells following the centrifugation as discussed above
  • the cellular fraction of the adipose tissue is disrupted, for example by treating it with lipases, detergents, proteases, and/or by mechanical or sonic disruption (e.g., using a homogenizer or sonicator).
  • the material is initially identified as a viscous material, but it can be subsequently treated, as desired, depending on the desired end use.
  • the raw matrix material can be treated (e.g., dialyzed or treated with proteases or acids, etc.) to produce a desirable lattice material.
  • the lattice can be prepared in a hydrated form or it can be dried or lyophilized into a substantially anhydrous form or a powder. Thereafter, the powder can be rehydrated for use as a cell culture substrate, for example by suspending it in a suitable cell culture medium.
  • the lipo-derived lattice can be mixed with other suitable lattice materials, such as described above.
  • the invention pertains to compositions including the lipo-derived lattice and cells or populations of cells, such as the inventive lipo-derived cells and other cells as well (particularly other types of stem cells).
  • the cells, populations, lattices, and compositions of the invention can be used in tissue engineering and regeneration.
  • the invention pertains to an implantable structure (i.e., an implant) incorporating any of these inventive features.
  • the exact nature of the implant will vary according to the use to which it is to be put.
  • the implant can be or comprise, as described, mature tissue, or it can include immature tissue or the lattice.
  • one type of implant can be a bone implant, comprising a population of the inventive cells that are undergoing (or are primed for) osteogenic differentiation, optionally seeded within a lattice of a suitable size and dimension, as described above.
  • Such an implant can be injected or engrafted within a host to encourage the generation or regeneration of mature bone tissue within the patient. Similar implants can be used to encourage the growth or regeneration of muscle, fat, cartilage, tendons, etc., within patients. Other types of implants are anlagen (such as described herein), e.g., limb buds, digit buds, developing kidneys, etc, that, once engrafted onto a patient, will mature into the appropriate structures.
  • the lipo-derived lattice can conveniently be employed as part of a cell culture kit.
  • the invention provides a kit including the inventive lipo-derived lattice and one or more other components, such as hydrating agents (e.g., water, physiologically-compatible saline solutions, prepared cell culture media, serum or derivatives thereof, etc.), cell culture substrates (e.g., culture dishes, plates, vials, etc.), cell culture media (whether in liquid or powdered form), antibiotic compounds, hormones, and the like.
  • the kit can include any such ingredients, preferably it includes all ingredients necessary to support the culture and growth of desired cell types upon proper combination.
  • the kit also can include cells (typically frozen), which can be seeded into the lattice as described herein.
  • the lipo-derived lattice can be used as an experimental reagent, such as in developing improved lattices and substrates for tissue growth and differentiation.
  • the lipo-derived lattice also can be employed cosmetically, for example, to hide wrinkles, scars, cutaneous depressions, etc., or for tissue augmentation.
  • the lattice is stylized and packaged in unit dosage form. If desired, it can be admixed with carriers (e.g., solvents such as glycerine or alcohols), perfumes, antibiotics, colorants, and other ingredients commonly employed in cosmetic products.
  • the substrate also can be employed autologously or as an allograft, and it can used as, or included within, ointments or dressings for facilitating wound healing.
  • the lipo-derived cells can also be used as experimental reagents. For example, they can be employed to help discover agents responsible for early events in differentiation.
  • the inventive cells can be exposed to a medium for inducing a particular line of differentiation and then assayed for differential expression of genes (e.g., by random-primed PCR or electrophoresis or protein or RNA, etc.).
  • the invention provides a kit for isolating such reagents from adipose tissues.
  • the kit can include a means for isolating adipose tissue from a patient (e.g., a cannula, a needle, an aspirator, etc.), as well as a means for separating stromal cells (e.g., through methods described herein).
  • the kit can be employed, for example, as a bedside source of stem cells that can then be re-introduced from the same individual as appropriate.
  • the kit can facilitate the isolation of lipo-derived stem cells for implantation in a patient needing regrowth of a desired tissue type, even in the same procedure.
  • the kit can also include a medium for differentiating the cells, such as those set forth herein.
  • the cells can be exposed to the medium to prime them for differentiation within the patient as needed.
  • the kit can be used as a convenient source of stem cells for in vitro manipulation (e.g., cloning or differentiating as described herein).
  • the kit can be employed for isolating a lipo-derived lattice as described herein.
  • This example demonstrates the isolation of a human lipo-derived stem cell substantially free of mature adipocytes.
  • Raw liposuction aspirate was obtained from patients undergoing elective surgery. Prior to the liposuction procedures, the patients were given epinephrine to minimize contamination of the aspirate with blood. The aspirate was strained to separate associated adipose tissue pieces from associated liquid waste. Isolated tissue was rinsed thoroughly with neutral phosphate buffered saline and then enzymatically dissociated with 0.075% w/v collagenase at 37° C. for about 20 minutes under intermittent agitation. Following the digestion, the collagenase was neutralized, and the slurry was centrifuged at about 260 g for about 10 minutes, which produced a multi-layered supernatant and a cellular pellet.
  • the supernatant was removed and retained for further use, and the pellet was resuspended in an erythrocyte-lysing solution and incubated without agitation at about 25° C. for about 10 minutes. Following incubation, the medium was neutralized, and the cells were again centrifuged at about 250 g for about 10 minutes. Following the second centrifugation, the cells were suspended, and assessed for viability (using trypan blue exclusion) and cell number. Thereafter, they were plated at a density of about at about 1 ⁇ 10 6 cells/100 mm dish. They were cultured at 37° C. in DMEM+fetal bovine serum (about 10%) in about 5% CO 2 .
  • telomeres using a commercially available TRAP assay kit
  • HeLa cells and HN-12 cells were used as positive controls.
  • Human foreskin fibroblasts and HN-12 heated cell extracts were used as negative controls.
  • Telomeric products were resolved onto a 12.5% polyacrylamide cells and the signals determined by phosphorimaging. Telemeric ladders representing telomerase activity were observed in the adipose-derived stem cells as well as the positive controls. No ladders were observed in the negative controls.
  • these cells were not identifiable as myocytes, adipocytes, chondrocytes, osteocytes, or blood cells. These results demonstrate that the adipose-derived cells express telomerase activity similar to that previously reported for human stem cells.
  • a population was cultured until near confluence and then exposed to the adipogenic medium for several weeks. The population was examined at two and four weeks after plating by colorimetric assessment of relative opacity following oil red-O staining. Adipogenesis was determined to be underway at two weeks and quite advanced at four weeks (relative opacity of 1 and 5.3, respectively). Bone marrow-derived stem cells were employed as a positive control, and these cells exhibited slightly less adipogenic potential (relative density of 0.7 and 2.8, respectively).
  • a population was cultured until near confluence and then exposed to the osteogenic medium for several weeks. The population was examined at two and four weeks after plating by colorimetric assessment of relative opacity following von Kossa staining. Osteogenesis was determined to be underway at two weeks and quite advanced at four weeks (relative opacity of 1.1 and 7.3, respectively. Bone marrow-derived stem cells were employed as a positive control, and these cells exhibited slightly less osteogenic potential (relative density of 0.2 and 6.6, respectively).
  • a population was cultured until near confluence and then exposed to the myogenic medium for several weeks. The population was examined at one, three, and six weeks after plating by assessment of multinucleated cells and expression of muscle-specific proteins (MyoD and myosin heavy chain).
  • MusoD and myosin heavy chain Human foreskin fibroblasts and skeletal myoblasts were used as controls. Cells expressing MyoD and myosin were found at all time points following exposure to the myogenic medium in the stem cell population, and the proportion of such cells increased at 3 and 6 weeks. Multinucleated cells were observed at 6 weeks. In contrast, the fibroblasts exhibited none of these characteristics at any time points.
  • Lipo-derived stem cells obtained in accordance with Example 1 were cultured in the presence of 5-azacytidine. In contrast with bone marrow-derived stem cells, exposure to this agent did not induce myogenic differentiation (see Wakitani et al., supra).
  • This example demonstrates the generation of a clonal population of human lipo-derived stem cells.
  • Example 1 Cells isolated in accordance with the procedure set forth in Example 1 were plated at about 5,000 cells/100 mm dish and cultured for a few days as indicated in Example 1. After some rounds of cell division, some clones were picked with a cloning ring and transferred to wells in a 48 well plate. These cells were cultured for several weeks, changing the medium twice weekly, until they were about 80% to about 90% confluent (at 37° C. in about 5% CO 2 in 2 ⁇ 3 F 12 medium+20% fetal bovine serum and 1 ⁇ 3 standard medium that was first conditioned by the cells isolated in Example 1, “cloning medium”).
  • each culture was transferred to a 35 mm dish and grown, and then retransferred to a 100 mm dish and grown until close to confluent. Following this, one cell population was frozen, and the remaining populations were plated on 12 well plates, at 1000 cells/well.
  • the cells were cultured for more than 15 passages in cloning medium and monitored for differentiation as indicated in Example 1.
  • the undifferentiated state of each clone remained true after successive rounds of differentiation.
  • This example demonstrates the lipo-derived stem cells can support the culture of other types of stem cells.
  • Human lipo-derived stem cells were passaged onto 96 well plates at a density of about 30,000/well, cultured for one week and then irradiated. Human CD34 + hematopoetic stem cells isolated from umbilical cord blood were then seeded into the wells. Co-cultures were maintained in MyeloCult H5100 media, and cell viability and proliferation were monitored subjectively by microscopic observation. After two weeks of co-culture, the hematopoetic stem cells were evaluated for CD34 expression by flow cytometry.
  • hematopoetic stem cells formed large colonies of rounded cells. Flow analysis revealed that 62% of the cells remained CD34 + . Based on microscopic observations, human adipo-derived stromal cells maintained the survival and supported the growth of human hematopoetic stem cells derived from umbilical cord blood.
  • This example demonstrates that the lipo-derived stem cells can differentiate in vivo.
  • Group A contained lipo-derived stem cells that had been pretreated with osteogenic medium as set forth in Example 1.
  • Group B contained untreated lipo-derived stem cells.
  • Group C contained osteogenic medium but no cells.
  • Group D contained non-osteogenic medium and no cells.
  • six mice were sacrificed at three weeks, and the remaining mice sacrificed at eight weeks following implantation.
  • the cubes were extracted, fixed, decalcified, and sectioned. Each section was analyzed by staining with H&E, Mallory bone stain, and immunostaining for osteocalcin.
  • This example demonstrates the isolation of a lipo-derived lattice substantially devoid of cells.
  • Example 1 In one protocol withheld supernatant from Example 1 was subjected to enzymatic digestion for three days in 0.05% trypsin EDTA/100 U/ml deoxyribonuclease to destroy the cells. Every day the debris was rinsed in saline and fresh enzyme was added. Thereafter the material was rinsed in saline and resuspended in 0.05% collagease and about 0.1% lipase to partially digest the proteins and fat present. This incubation continued for two days.
  • the withheld supernatant from Example 1 was incubated in EDTA to eliminate any epithelial cells.
  • the remaining cells were lysed using a buffer containing 1% NP40, 0.5% sodium deoxycholate, 0.1% SDS, 5 mM EDTA, 0.4M NaCl, 50 mMTris-HCL (pH 8) and protease inhibitors, and 10 ⁇ g/ml each of leupeptin, chymostatin, antipain, and pepstatin A.
  • the tissue was extensively washed in PBS without divalent cations.
  • the term “consisting essentially of” indicates that unlisted ingredients or steps that do not materially affect the basic and novel properties of the invention can be employed in addition to the specifically recited ingredients or steps.
  • terms such as “comprising,” “having,” and “including” indicate that any ingredients or steps can be present in addition to those recited.
  • the term “consisting of” indicates that only the recited ingredients or steps are present, but does not foreclose the possibility that equivalents of the ingredients or steps can substitute for those specifically recited.

Abstract

The present invention provides adipose-derived stem cells and lattices. In one aspect, the present invention provides a lipo-derived stem cell substantially free of adipocytes and red blood cells and clonal populations of connective tissue stem cells. The cells can be employed, alone or within biologically-compatible compositions, to generate differentiated tissues and structures, both in vivo and in vitro. Additionally, the cells can be expanded and cultured to produce hormones and to provide conditioned culture media for supporting the growth and expansion of other cell populations. In another aspect, the present invention provides a lipo-derived lattice substantially devoid of cells, which includes extracellular matrix material from adipose tissue. The lattice can be used as a substrate to facilitate the growth and differentiation of cells, whether in vivo or in vitro, into anlagen or even mature tissues or structures.

Description

    BACKGROUND OF THE INVENTION
  • In recent years, the identification of mesenchymal stem cells, chiefly obtained from bone marrow, has led to advances in tissue regrowth and differentiation. Such cells are pluripotent cells found in bone marrow and periosteum, and they are capable of differentiating into various mesenchymal or connective tissues. For example, such bone-marrow derived stem cells can be induced to develop into myocytes upon exposure to agents such as 5-azacytidine (Wakitani et al., Muscle Nerve, 18(12), 1417-26 (1995)). It has been suggested that such cells are useful for repair of tissues such as cartilage, fat, and bone (see, e.g., U.S. Pat. Nos. 5,908,784, 5,906,934, 5,827,740, 5,827,735), and that they also have applications through genetic modification (see, e.g., U.S. Pat. No. 5,591,625). While the identification of such cells has led to advances in tissue regrowth and differentiation, the use of such cells is hampered by several technical hurdles. One drawback to the use of such cells is that they are very rare (representing as few as 1/2,000,000 cells), making any process for obtaining and isolating them difficult and costly. Of course, bone marrow harvest is universally painful to the donor. Moreover, such cells are difficult to culture without inducing differentiation, unless specifically screened sera lots are used, adding further cost and labor to the use of such stem cells. Thus, there is a need for a more readily available source for pluripotent stem cells, particularly cells that can be cultured without the requirement for costly prescreening of culture materials.
  • Other advances in tissue engineering have shown that cells can be grown in specially-defined cultures to produce three-dimensional structures. Spacial definition typically is achieved by using various acellular lattices or matrices to support and guide cell growth and differentiation. While this technique is still in its infancy, experiments in animal models have demonstrated that it is possible to employ various acellular lattice materials to regenerate whole tissues (see, e.g., Probst et al. BJU Int., 85(3), 362-7 (2000)). A suitable lattice material is secreted extracellular matrix material isolated from tumor cell lines (e.g., Engelbreth-Holm-Swarm tumor secreted matrix—“matrigel”). This material contains type IV collagen and growth factors, and provides an excellent substrate for cell growth (see, e.g., Vukicevic et al., Exp. Cell Res, 202(1), 1-8 (1992)). However, as this material also facilitates the malignant transformation of some cells (see, e.g., Fridman, et al., Int. J. Cancer, 51(5), 740-44 (1992)), it is not suitable for clinical application. While other artificial lattices have been developed, these can prove toxic either to cells or to patients when used in vivo. Accordingly, there remains a need for a lattice material suitable for use as a substrate in culturing and growing populations of cells.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention provides adipose-derived stem cells and lattices. In one aspect, the present invention provides a lipo-derived stem cell substantially free of adipocytes and red blood cells and clonal populations of connective tissue stem cells. The cells can be employed, alone or within biologically-compatible compositions, to generate differentiated tissues and structures, both in vivo and in vitro. Additionally, the cells can be expanded and cultured to produce hormones and to provide conditioned culture media for supporting the growth and expansion of other cell populations. In another aspect, the present invention provides a lipo-derived lattice substantially devoid of cells, which includes extracellular matrix material from adipose tissue. The lattice can be used as a substrate to facilitate the growth and differentiation of cells, whether in vivo or in vitro, into anlagen or even mature tissues or structures.
  • Considering how plentiful adipose tissue is, the inventive cells and lattice represent a ready source of pluripotent stem cells. Moreover, because the cells can be passaged in culture in an undifferentiated state under culture conditions not requiring prescreened lots of serum, the inventive cells can be maintained with considerably less expense than other types of stem cells. These and other advantages of the present invention, as well as additional inventive features, will be apparent from the accompanying drawings and in the following detailed description.
  • DETAILED DESCRIPTION OF THE INVENTION
  • One aspect of the invention pertains to a lipo-derived stem cell. Preferably, the stem cell is substantially free of other cell types (e.g., adipocytes, red blood cells, other stromal cells, etc.) and extracellular matrix material; more preferably, the stem cell is completely free of such other cell types and matrix material. Preferably, the inventive cell is derived from the adipose tissue of a primate, and more preferably a higher primate (e.g., a baboon or ape). Typically, the inventive cell will be derived from human adipose tissue, using methods such as described herein.
  • While the inventive cell can be any type of stem cell, for use in tissue engineering, desirably the cell is of mesodermal origin. Typically such cells, when isolated, retain two or more mesodermal or mesenchymal developmental phenotypes (i.e., they are pluripotent). In particular, such cells generally have the capacity to develop into mesodermal tissues, such as mature adipose tissue, bone, various tissues of the heart (e.g., pericardium, epicardium, epimyocardium, myocardium, pericardium, valve tissue, etc.), dermal connective tissue, hemangial tissues (e.g., corpuscles, endocardium, vascular epithelium, etc.), muscle tissues (including skeletal muscles, cardiac muscles, smooth muscles, etc.), urogenital tissues (e.g., kidney, pronephros, meta- and meso-nephric ducts, metanephric diverticulum, ureters, renal pelvis, collecting tubules, epithelium of the female reproductive structures (particularly the oviducts, uterus, and vagina)), pleural and peritoneal tissues, viscera, mesodermal glandular tissues (e.g., adrenal cortex tissues), and stromal tissues (e.g., bone marrow). Of course, inasmuch as the cell can retain potential to develop into mature cells, it also can realize its developmental phenotypic potential by differentiating into an appropriate precursor cell (e.g., a preadipocyte, a premyocyte, a preosteocyte, etc.). Also, depending on the culture conditions, the cells can also exhibit developmental phenotypes such as embryonic, fetal, hematopoetic, neurogenic, or neuralgiagenic developmental phenotypes. In this sense, the inventive cell can have two or more developmental phenotypes such as adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, splanchogenic, and stromal developmental phenotypes. While such cells can retain two or more of these developmental phenotypes, preferably, such cells have three or more such developmental phenotypes (e.g., four or more mesodermal or mesenchymal developmental phenotypes), and some types of inventive stem cells have a potential to acquire any mesodermal phenotype through the process of differentiation.
  • The inventive stem cell can be obtained from adipose tissue by any suitable method. A first step in any such method requires the isolation of adipose tissue from the source animal. The animal can be alive or dead, so long as adipose stromal cells within the animal are viable. Typically, human adipose stromal cells are obtained from living donors, using well-recognized protocols such as surgical or suction lipectomy. Indeed, as liposuction procedures are so common, liposuction effluent is a particularly preferred source from which the inventive cells can be derived.
  • However derived, the adipose tissue is processed to separate stem cells from the remainder of the material. In one protocol, the adipose tissue is washed with physiologically-compatible saline solution (e.g., phosphate buffered saline (PBS)) and then vigorously agitated and left to settle, a step that removes loose matter (e.g., damaged tissue, blood, erythrocytes, etc.) from the adipose tissue. Thus, the washing and settling steps generally are repeated until the supernatant is relatively clear of debris.
  • The remaining cells generally will be present in lumps of various size, and the protocol proceeds using steps gauged to degrade the gross structure while minimizing damage to the cells themselves. One method of achieving this end is to treat the washed lumps of cells with an enzyme that weakens or destroys bonds between cells (e.g., collagenase, dispase, trypsin, etc.). The amount and duration of such enzymatic treatment will vary, depending on the conditions employed, but the use of such enzymes is generally known in the art. Alternatively or in conjunction with such enzymatic treatment, the lumps of cells can be degraded using other treatments, such as mechanical agitation, sonic energy, thermal energy, etc. If degradation is accomplished by enzymatic methods, it is desirable to neutralize the enzyme following a suitable period, to minimize deleterious effects on the cells.
  • The degradation step typically produces a slurry or suspension of aggregated cells (generally liposomes) and a fluid fraction containing generally free stromal cells (e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells). The next stage in the separation process is to separate the aggregated cells from the stromal cells. This can be accomplished by centrifugation, which forces the stromal cells into a pellet covered by a supernatant. The supernatant then can be discarded and the pellet suspended in a physiologically-compatible fluid. Moreover, the suspended cells typically include erythrocytes, and in most protocols it is desirable to lyse these. Methods for selectively lysing erythrocytes are known in the art, and any suitable protocol can be employed (e.g., incubation in a hyper- or hypotonic medium). Of course, if the erythrocytes are lysed, the remaining cells should then be separated from the lysate, for example by filtration or centrifugation. Of course, regardless of whether the erythrocytes are lysed, the suspended cells can be washed, re-centrifuged, and resuspended one or more successive times to achieve greater purity. Alternatively, the cells can be separated using a cell sorter or on the basis of cell size and granularity, stem cells being relatively small and agranular. Expression of telomerase can also serve as a stem cell-specific marker. They can also be separated immunohistochemically, for example, by panning or using magnetic beads. Any of the steps and procedures for isolating the inventive cells can be performed manually, if desired. Alternatively, the process of isolating such cells can be facilitated through a suitable device, many of which are known in the art (see, e.g., U.S. Pat. No. 5,786,207).
  • Following the final isolation and resuspension, the cells can be cultured and, if desired, assayed for number and viability to assess the yield. Desirably the cells can be cultured without differentiation using standard cell culture media (e.g., DMEM, typically supplemented with 5-15% (e.g., 10%) serum (e.g., fetal bovine serum, horse serum, etc.). Preferably, the cells can be passaged at least five times in such medium without differentiating, while still retaining their developmental phenotype, and more preferably, the cells can be passaged at least 10 times (e.g., at least 15 times or even at least 20 times) without losing developmental phenotype. Thus, culturing the cells of the present invention without inducing differentiation can be accomplished without specially screened lots of serum, as is generally the case for mesenchymal stem cells (e.g., derived from marrow). Methods for measuring viability and yield are known in the art (e.g., trypan blue exclusion).
  • Following isolation, the stem cells are further separated by phenotypic identification, to identify those cells that have two or more of the aforementioned developmental phenotypes. Typically, the stromal cells are plated at a desired density such as between about 100 cells/cm2 to about 100,000 cells/cm2 (such as about 500 cells/cm2 to about 50,000 cells/cm2, or, more particularly, between about 1,000 cells/cm2 to about 20,000 cells/cm2). If plated at lower densities (e.g., about 300 cells/cm2), the cells can be more easily clonally isolated. For example, after a few days, cells plated at such densities will proliferate into a population.
  • Such cells and populations can be clonally expanded, if desired, using a suitable method for cloning cell populations. For example, a proliferated population of cells can be physically picked and seeded into a separate plate (or the well of a multi-well plate). Alternatively, the cells can be subcloned onto a multi-well plate at a statistical ratio for facilitating placing a single cell into each well (e.g., from about 0.1 to about 1 cell/well or even about 0.25 to about 0.5 cells/well, such as 0.5 cells/well). Of course, the cells can be cloned by plating them at low density (e.g., in a petri-dish or other suitable substrate) and isolating them from other cells using devices such as a cloning rings. Alternatively, where an irradiation source is available, clones can be obtained by permitting the cells to grow into a monolayer and then shielding one and irradiating the rest of cells within the monolayer. The surviving cell then will grow into a clonal population. While production of a clonal population can be expanded in any suitable culture medium, a preferred culture condition for cloning stem cells (such as the inventive stem cells or other stem cells) is about ⅔ F12 medium+20% serum (preferably fetal bovine serum) and about ⅓ standard medium that haw been conditioned with stromal cells (e.g., cells from the stromal vascular fraction of liposuction aspirate), the relative proportions being determined volumetrically).
  • In any event, whether clonal or not, the isolated cells can be cultured to a suitable point when their developmental phenotype can be assessed. As mentioned, the inventive cells have at least two of the aforementioned developmental phenotypes. Thus, one or more cells drawn from a given clone can be treated to ascertain whether it possesses such developmental potentials. One type of treatment is to culture the inventive cells in culture media that has been conditioned by exposure to mature cells (pr precursors thereof) of the respective type to be differentiated (e.g., media conditioned by exposure to myocytes can induce myogenic differentiation, media conditioned by exposure to heart valve cells can induce differentiation into heart valve tissue, etc.). Of course, defined media for inducing differentiation also can be employed. For example, adipogenic developmental phenotype can be assessed by exposing the cell to a medium that facilitates adipogenesis, e.g., containing a glucocorticoid (e.g., isobutyl-methylxanthine, dexamethasone, hydrocortisone, cortisone, etc.), insulin, a compound which elevates intracellular levels of cAMP (e.g., dibutyryl-cAMP, 8-CPT-cAMP (8-(4)chlorophenylthio)-adenosine 3′,5′ cyclic monophosphate; 8-bromo-cAMP; dioctanoyl-cAMP, forskolin etc.), and/or a compound which inhibits degradation of cAMP (e.g., a phosphodiesterase inhibitor such as methyl isobutylxanthine, theophylline, caffeine, indomethacin, and the like). Thus, exposure of the stem cells to between about 1 μM and about 10 μM insulin in combination with about 10−9 M to about 10−6 M to (e.g., about 1 μM) dexamethasone can induce adipogenic differentiation. Such a medium also can include other agents, such as indomethicin (e.g., about 100 μM to about 200 μM), if desired, and preferably the medium is serum free. Osteogenic developmental phenotype can be assessed by exposing the cells to between about 10−7 M and about 10−9 M dexamethasone (e.g., about 1 μM) in combination with about 10 μM to about 50 μM ascorbate-2-phosphate and between about 10 nM and about 50 nM β-glycerophosphate, and the medium also can include serum (e.g., bovine serum, horse serum, etc.). Myogenic differentiation can be induced by exposing the cells to between about 10 μM and about 100 μM hydrocortisone, preferably in a serum-rich medium (e.g., containing between about 10% and about 20% serum (either bovine, horse, or a mixture thereof)). Chondrogenic differentiation can be induced by exposing the cells to between about 1 μM to about 10 μM insulin and between about 1 μM to about 10 μM transferrin, between about 1 ng/ml and 10 ng/ml transforming growth factor (TGF) β1, and between about 10 nM and about 50 nM ascorbate-2-phosphate (50 nM). For chondrogenic differentiation, preferably the cells are cultured in high density (e.g., at about several million cells/ml or using micromass culture techniques), and also in the presence of low amounts of serum (e.g., from about 1% to about 5%). The cells also can be induced to assume a developmentally more immature phenotype (e.g., a fetal or embryonic phenotype). Such induction is achieved upon exposure of the inventive cell to conditions that mimic those within fetuses and embryos. For example, the inventive cells or populations can be co-cultured with cells isolated from fetuses or embryos, or in the presence of fetal serum. Along these lines, the cells can be induced to differentiate into any of the aforementioned mesodermal lineages by co-culturing them with mature cells of the respective type, or precursors thereof. Thus, for example, myogenic differentiation can be induced by culturing the inventive cells with myocytes or precursors, and similar results can be achieved with respect to the other tissue types mentioned herein. Other methods of inducing differentiation are known in the art, and many of them can be employed, as appropriate.
  • After culturing the cells in the differentiating-inducing medium for a suitable time (e.g., several days to a week or more), the cells can be assayed to determine whether, in fact, they have differentiated to acquire physical qualities of a given type of cell. One measurement of differentiation per se is telomere length, undifferentiated stem cells having longer telomeres than differentiated cells; thus the cells can be assayed for the level of telomerase activity. Alternatively, RNA or proteins can be extracted from the cells and assayed (via Northern hybridization, rtPCR, Western blot analysis, etc.) for the presence of markers indicative of the desired phenotype. Of course, the cells can be assayed immunohistochemically or stained, using tissue-specific stains. Thus, for example, to assess adipogenic differentiation, the cells can be stained with fat-specific stains (e.g., oil red O, safarin red, sudan black, etc.) or probed to assess the presence of adipose-related factors (e.g., type IV collagen, PPAR-γ, adipsin, lipoprotein lipase, etc.). Similarly, ostogenesis can be assessed by staining the cells with bone-specific stains (e.g., alkaline phosphatase, von Kossa, etc.) or probed for the presence of bone-specific markers (e.g., osteocalcin, osteonectin, osteopontin, type I collagen, bone morphogenic proteins, cbfa, etc.). Myogensis can be assessed by identifying classical morphologic changes (e.g., polynucleated cells, syncitia formation, etc.), or assessed biochemically for the presence of muscle-specific factors (e.g., myo D, myosin heavy chain, NCAM, etc.). Chondrogenesis can be determined by staining the cells using cartallge-specific stains (e.g., alcian blue) or probing the cells for the expression/production of cartilage-specific molecules (e.g., sulfated glycosaminoglycans and proteoglycans (e.g., keratin, chondroitin, etc.) in the medium, type II collagen, etc.). Other methods of assessing developmental phenotype are known in the art, and any of them is appropriate. For example, the cells can be sorted by size and granularity. Also, the cells can be used to generate monoclonal antibodies, which can then be employed to assess whether they preferentially bind to a given cell type. Correlation of antigenicity can confirm that the stem cell has differentiated along a given developmental pathway.
  • While the cell can be solitary and isolated from other cells, preferably it is within a population of cells, and the invention provides a defined population including the inventive cell. In some embodiments, the population is heterogeneous. Thus, for example, the population can include support cells for supplying factors to the inventive cells. Of course, the inventive stem cells can themselves serve as support cells for culturing other types of cells (such as other types of stem cells, e.g., as neural stem cells (NSC), hematopoetic stem cells (HPC, particularly CD34+ stem cells), embryonic stem cells (ESC) and mixtures thereof), and the population can include such cells. In other embodiments, the population is substantially homogeneous, consisting essentially of the inventive lipo-derived stem cells.
  • As the inventive cells can be cloned, a substantially homogeneous population containing them can be clonal. Indeed, the invention also pertains to any defined clonal cell population consisting essentially of mesodermal stem cells, connective tissue stem cell, or mixtures thereof. In this embodiment, the cells can be lipo-derived or derived from other mesodermal or connective cell tissues (e.g., bone marrow, muscle, etc.) using methods known in the art. After the isolation, the cells can be expanded clonally as described herein.
  • The inventive cells (and cell populations) can be employed for a variety of purposes. As mentioned, the cells can support the growth and expansion of other cell types, and the invention pertains to methods for accomplishing this. In one aspect, the invention pertains to a method of conditioning culture medium using the inventive stem cells and to conditioned medium produced by such a method. The medium becomes conditioned upon exposing a desired culture medium to the cells under conditions sufficient for the cells to condition it. Typically, the medium is used to support the growth of the inventive cells, which secrete hormones, cell matrix material, and other factors into the medium. After a suitable period (e.g., one or a few days), the culture medium containing the secreted factors can be separated from the cells and stored for future use. Of course, the inventive cells and populations can be re-used successively to condition medium, as desired. In other applications (e.g., for co-culturing the inventive cells with other cell types), the cells can remain within the conditioned medium. Thus, the invention provides a conditioned medium obtained using this method, which either can contain the inventive cells or be substantially free of the inventive cells, as desired.
  • The conditioned medium can be used to support the growth and expansion of desired cell types, and the invention provides a method of culturing cells (particularly stem cells) using the conditioned medium. The method involves maintaining a desired cell in the conditioned medium under conditions for the cell to remain viable. The cell can be maintained under any suitable condition for culturing them, such as are known in the art. Desirably, the method permits successive rounds of mitotic division of the cell to form an expanded population. The exact conditions (e.g., temperature, CO2 levels, agitation, presence of antibiotics, etc.) will depend on the other constituents of the medium and on the cell type. However, optimizing these parameters are within the ordinary skill in the art. In some embodiments, it is desirable for the medium to be substantially free of the lipo-derived cells employed to condition the medium as described herein. However, in other embodiments, it is desirable for the lipo-derived cells to remain in the conditioned medium and co-cultured with the cells of interest. Indeed, as the inventive lipo-derived cells can express cell-surface mediators of intercellular communication, it often is desirable for the inventive cells and the desired other cells to be co-cultured under conditions in which the two cell types are in contact. This can be achieved, for example, by seeding the cells as a heterogeneous population of cells onto a suitable culture substrate. Alternatively, the inventive lipo-derived cells can first be grown to confluence, which will serve as a substrate for the second desired cells to be cultured within the conditioned medium.
  • In another embodiment, the inventive lipo-derived cells can be genetically modified, e.g., to express exogenous genes or to repress the expression of endogenous genes, and the invention provides a method of genetically modifying such cells and populations. In accordance with this method, the cell is exposed to a gene transfer vector comprising a nucleic acid including a transgene, such that the nucleic acid is introduced into the cell under conditions appropriate for the transgene to be expressed within the cell. The transgene generally is an expression cassette, including a coding polynucleotide operably linked to a suitable promoter. The coding polynucleotide can encode a protein, or it can encode biologically active RNA (e.g., antisense RNA or a ribozyme). Thus, for example, the coding polynucleotide can encode a gene conferring resistance to a toxin, a hormone (such as peptide growth hormones, hormone releasing factors, sex hormones, adrenocorticotrophic hormones, cytokines (e.g., interferins, interleukins, lymphokines), etc.), a cell-surface-bound intracellular signaling moiety (e.g., cell adhesion molecules, hormone receptors, etc.), a factor promoting a given lineage of differentiation, etc. Of course, where it is desired to employ gene transfer technology to deliver a given transgene, its sequence will be known.
  • Within the expression cassette, the coding polynucleotide is operably linked to a suitable promoter. Examples of suitable promoters include prokaryotic promoters and viral promoters (e.g., retroviral ITRs, LTRs, immediate early viral promoters (IEp), such as herpesvirus IEp (e.g., ICP4-IEp and ICP0-IEp), cytomegalovirus (CMV) IEp, and other viral promoters, such as Rous Sarcoma Virus (RSV) promoters, and Murine Leukemia Virus (MLV) promoters). Other suitable promoters are eukaryotic promoters, such as enhancers (e.g., the rabbit β-globin regulatory elements), constitutively active promoters (e.g., the β-actin promoter, etc.), signal specific promoters (e.g., inducible promoters such as a promoter responsive to RU486, etc.), and tissue-specific promoters. It is well within the skill of the art to select a promoter suitable for driving gene expression in a predefined cellular context. The expression cassette can include more than one coding polynucleotide, and it can include other elements (e.g., polyadenylation sequences, sequences encoding a membrane-insertion signal or a secretion leader, ribosome entry sequences, transcriptional regulatory elements (e.g., enhancers, silencers, etc.), and the like), as desired.
  • The expression cassette containing the transgene should be incorporated into a genetic vector suitable for delivering the transgene to the cells. Depending on the desired end application, any such vector can be so employed to genetically modify the cells (e.g., plasmids, naked DNA, viruses such as adenovirus, adeno-associated virus, herpesviruses, lentiviruses, papillomaviruses, retroviruses, etc.). Any method of constructing the desired expression cassette within such vectors can be employed, many of which are well known in the art (e.g., direct cloning, homologous recombination, etc.). Of course, the choice of vector will largely determine the method used to introduce the vector into the cells (e.g., by protoplast fusion, calcium-phosphate precipitation, gene gun, electroporation, infection with viral vectors, etc.), which are generally known in the art.
  • The genetically altered cells can be employed as bioreactors for producing the product of the transgene. In other embodiments, the genetically modified cells are employed to deliver the transgene and its product to an animal. For example, the cells, once genetically modified, can be introduced into the animal under conditions sufficient for the transgene to be expressed in vivo.
  • In addition to serving as useful targets for genetic modification, many cells and populations of the present invention secrete hormones (e.g., cytokines, peptide or other (e.g., monobutyrin) growth factors, etc.). Some of the cells naturally secrete such hormones upon initial isolation, and other cells can be genetically modified to secrete hormones, as discussed herein. The cells of the present invention that secrete hormones can be used in a variety of contexts in vivo and in vitro. For example, such cells can be employed as bioreactors to provide a ready source of a given hormone, and the invention pertains to a method of obtaining hormones from such cells. In accordance with the method, the cells are cultured, under suitable conditions for them to secrete the hormone into the culture medium. After a suitable period of time, and preferably periodically, the medium is harvested and processed to isolate the hormone from the medium. Any standard method (e.g., gel or affinity chromatography, dialysis, lyophilization, etc.) can be used to purify the hormone from the medium, many of which are known in the art.
  • In other embodiments, cells (and populations) of the present invention secreting hormones can be employed as therapeutic agents. Generally, such methods involve transferring the cells to desired tissue, either in vitro (e.g., as a graft prior to implantation or engrafting) or in vivo, to animal tissue directly. The cells can be transferred to the desired tissue by any method appropriate, which generally will vary according to the tissue type. For example, cells can be transferred to a graft by bathing the graft (or infusing it) with culture medium continuing the cells. Alternatively, the cells can be seeded onto the desired site within the tissue to establish a population. Cells can be transferred to sites in vivo using devices such as catheters, trocars, cannulae, stents (which can be seeded with the cells), etc. For these applications, preferably the cell secretes a cytokine or growth hormone such as human growth factor, fibroblast growth factor, nerve growth factor, insulin-like growth factors, hemopoietic stem cell growth factors, members of the fibroblast growth factor family, members of the platelet-derived growth factor family, vascular and endothelial cell growth factors, members of the TGFb family (including bone morphogenic factor), or enzymes specific for congenital disorders (e.g., distrophin).
  • In one application, the invention provides a method of promoting the closure of a wound within a patient using such cells. In accordance with the method, the inventive cells secreting the hormone are transferred to the vicinity of a wound under conditions sufficient for the cell to produce the hormone. The presence of the hormone in the vicinity of the wound promotes closure of the wound. The method promotes closure of both external (e.g., surface) and internal wounds. Wounds to which the present inventive method is useful in promoting closure include, but are not limited to, abrasions, avulsions, blowing wounds, burn wounds, contusions, gunshot wounds, incised wounds, open wounds, penetrating wounds, perforating wounds, puncture wounds, seton wounds, stab wounds, surgical wounds, subcutaneous wounds, or tangential wounds. The method need not achieve complete healing or closure of the wound; it is sufficient for the method to promote any degree of wound closure. In this respect, the method can be employed alone or as an adjunct to other methods for healing wounded tissue.
  • Where the inventive cells secrete an angiogenic hormone (e.g., vascular growth factor, vascular and endothelial cell growth factor, etc.), they (as well as populations containing them) can be employed to induce angiogenesis within tissues. Thus, the invention provides a method of promoting neovascularization within tissue using such cells. In accordance with this method, the cell is introduced the desired tissue under conditions sufficient for the cell to produce the angiogenic hormone. The presence of the hormone within the tissue promotes neovascularization within the tissue.
  • Because the inventive stem cells have a developmental phenotype, they can be employed in tissue engineering. In this regard, the invention provides a method of producing animal matter comprising maintaining the inventive cells under conditions sufficient for them to expand and differentiate to form the desired matter. The matter can include mature tissues, or even whole organs, including tissue types into which the inventive cells can differentiate (as set forth herein). Typically, such matter will comprise adipose, cartilage, heart, dermal connective tissue, blood tissue, muscle, kidney, bone, pleural, splancnic tissues, vascular tissues, and the like. More typically, the matter will comprise combinations of these tissue types (i.e., more than one tissue type). For example, the matter can comprise all or a portion of an animal organ (e.g., a heart, a kidney) or a limb (e.g., a leg, a wing, an arm, a hand, a foot, etc.). Of course, in as much as the cells can divide and differentiate to produce such structures, they can also form anlagen of such structures. At early stages, such anlagen can be cryopreserved for future generation of the desired mature structure or organ.
  • To produce such structures, the inventive cells and populations are maintained under conditions suitable for them to expand and divide to form the desired structures. In some applications, this is accomplished by transferring them to an animal (i.e., in vivo) typically at a sight at which the new matter is desired. Thus, for example, the invention can facilitate the regeneration of tissues (e.g., bone, muscle, cartilage, tendons, adipose, etc.) within an animal where the cells are implanted into such tissues. In other embodiments, and particularly to create anlagen, the cells can be induced to differentiate and expand into tissues in vitro. In such applications, the cells are cultured on substrates that facilitate formation into three-dimensional structures conducive for tissue development. Thus, for example, the cells can be cultured or seeded onto a bio-compatible lattice, such as one that includes extracellular matrix material, synthetic polymers, cytokines, growth factors, etc. Such a lattice can be molded into desired shapes for facilitating the development of tissue types. Also, at least at an early stage during such culturing, the medium and/or substrate is supplemented with factors (e.g., growth factors, cytokines, extracellular matrix material, etc.) that facilitate the development of appropriate tissue types and structures. Indeed, in some embodiments, it is desired to co-culture the cells with mature cells of the respective tissue type, or precursors thereof, or to expose the cells to the respective conditioned medium, as discussed herein.
  • To facilitate the use of the inventive lipo-derived cells and populations for producing such animal matter and tissues, the invention provides a composition including the inventive cells (and populations) and biologically compatible lattice. Typically, the lattice is formed from polymeric material, having fibers as a mesh or sponge, typically with spaces on the order of between about 100 μm and about 300 μm. Such a structure provides sufficient area on which the cells can grow and proliferate. Desirably, the lattice is biodegradable over time, so that it will be absorbed into the animal matter as it develops. Suitable polymeric lattices, thus, can be formed from monomers such as glycolic acid, lactic acid, propyl fumarate, caprolactone, hyaluronan, hyaluronic acid, and the like. Other lattices can include proteins, polysaccharides, polyhydroxy acids, polyorthoesters, polyanhydrides, polyphosphazenes, or synthetic polymers (particularly biodegradable polymers). Of course, a suitable polymer for forming such lattice can include more than one monomers (e.g., combinations of the indicated monomers). Also, the lattice can also include hormones, such as growth factors, cytokines, and morphogens (e.g., retinoic acid, aracadonic acid, etc.), desired extracellular matrix molecules (e.g., fibronectin, laminin, collagen, etc.), or other materials (e.g., DNA, viruses, other cell types, etc.) as desired.
  • To form the composition, the cells are introduced into the lattice such that they permeate into the interstitial spaces therein. For example, the matrix can be soaked in a solution or suspension containing the cells, or they can be infused or injected into the matrix. A particularly preferred composition is a hydrogel formed by crosslinking of a suspension including the polymer and also having the inventive cells dispersed therein. This method of formation permits the cells to be dispersed throughout the lattice, facilitating more even permeation of the lattice with the cells. Of course, the composition also can include mature cells of a desired phenotype or precursors thereof, particularly to potentate the induction of the inventive stem cells to differentiate appropriately within the lattice (e.g., as an effect of co-culturing such cells within the lattice).
  • The composition can be employed in any suitable manner to facilitate the growth and generation of the desired tissue types, structures, or anlagen. For example, the composition can be constructed using three-dimensional or stereotactic modeling techniques. Thus, for example, a layer or domain within the composition can be populated by cells primed for osteogenic differentiation, and another layer or domain within the composition can be populated with cells primed for myogenic and/or chondrogenic development. Bringing such domains into juxtaposition with each other facilitates the molding and differentiation of complex structures including multiple tissue types (e.g., bone surrounded by muscle, such as found in a limb). To direct the growth and differentiation of the desired structure, the composition can be cultured ex vivo in a bioreactor or incubator, as appropriate. In other embodiments, the structure is implanted within the host animal directly at the site in which it is desired to grow the tissue or structure. In still another embodiment, the composition can be engrafted on a host (typically an animal such as a pig, baboon, etc.), where it will grow and mature until ready for use. Thereafter, the mature structure (or anlage) is excised from the host and implanted into the host, as appropriate.
  • Lattices suitable for inclusion into the composition can be derived from any suitable source (e.g., matrigel), and some commercial sources for suitable lattices exist (e.g., suitable of polyglycolic acid can be obtained from sources such as Ethicon, N.J.). Another suitable lattice can be derived from the acellular portion of adipose tissue—i.e., adipose tissue extracellular matrix matter substantially devoid of cells, and the invention provides such a lipo-derived lattice. Typically, such lipo-derived lattice includes proteins such as proteoglycans, glycoproteins, hyaluronins, fibronectins, collagens (type I, type II, type III, type IV, type V, type VI, etc.), and the like, which serve as excellent substrates for cell growth. Additionally, such lipo-derived lattices can include hormones, preferably cytokines and growth factors, for facilitating the growth of cells seeded into the matrix.
  • The lipo-derived matrix can be isolated form adipose tissue similarly as described above, except that it will be present in the acellular fraction. For example, adipose tissue or derivatives thereof (e.g., a fraction of the cells following the centrifugation as discussed above) can be subjected to sonic or thermal energy and/or enzymatic processed to recover the matrix material. Also, desirably the cellular fraction of the adipose tissue is disrupted, for example by treating it with lipases, detergents, proteases, and/or by mechanical or sonic disruption (e.g., using a homogenizer or sonicator). However isolated, the material is initially identified as a viscous material, but it can be subsequently treated, as desired, depending on the desired end use. For example, the raw matrix material can be treated (e.g., dialyzed or treated with proteases or acids, etc.) to produce a desirable lattice material. Thus the lattice can be prepared in a hydrated form or it can be dried or lyophilized into a substantially anhydrous form or a powder. Thereafter, the powder can be rehydrated for use as a cell culture substrate, for example by suspending it in a suitable cell culture medium. In this regard, the lipo-derived lattice can be mixed with other suitable lattice materials, such as described above. Of course, the invention pertains to compositions including the lipo-derived lattice and cells or populations of cells, such as the inventive lipo-derived cells and other cells as well (particularly other types of stem cells).
  • As discussed above, the cells, populations, lattices, and compositions of the invention can be used in tissue engineering and regeneration. Thus, the invention pertains to an implantable structure (i.e., an implant) incorporating any of these inventive features. The exact nature of the implant will vary according to the use to which it is to be put. The implant can be or comprise, as described, mature tissue, or it can include immature tissue or the lattice. Thus, for example, one type of implant can be a bone implant, comprising a population of the inventive cells that are undergoing (or are primed for) osteogenic differentiation, optionally seeded within a lattice of a suitable size and dimension, as described above. Such an implant can be injected or engrafted within a host to encourage the generation or regeneration of mature bone tissue within the patient. Similar implants can be used to encourage the growth or regeneration of muscle, fat, cartilage, tendons, etc., within patients. Other types of implants are anlagen (such as described herein), e.g., limb buds, digit buds, developing kidneys, etc, that, once engrafted onto a patient, will mature into the appropriate structures.
  • The lipo-derived lattice can conveniently be employed as part of a cell culture kit. Accordingly, the invention provides a kit including the inventive lipo-derived lattice and one or more other components, such as hydrating agents (e.g., water, physiologically-compatible saline solutions, prepared cell culture media, serum or derivatives thereof, etc.), cell culture substrates (e.g., culture dishes, plates, vials, etc.), cell culture media (whether in liquid or powdered form), antibiotic compounds, hormones, and the like. While the kit can include any such ingredients, preferably it includes all ingredients necessary to support the culture and growth of desired cell types upon proper combination. Of course, if desired, the kit also can include cells (typically frozen), which can be seeded into the lattice as described herein.
  • While many aspects of the invention pertain to tissue growth and differentiation, the invention has other applications as well. For example, the lipo-derived lattice can be used as an experimental reagent, such as in developing improved lattices and substrates for tissue growth and differentiation. The lipo-derived lattice also can be employed cosmetically, for example, to hide wrinkles, scars, cutaneous depressions, etc., or for tissue augmentation. For such applications, preferably the lattice is stylized and packaged in unit dosage form. If desired, it can be admixed with carriers (e.g., solvents such as glycerine or alcohols), perfumes, antibiotics, colorants, and other ingredients commonly employed in cosmetic products. The substrate also can be employed autologously or as an allograft, and it can used as, or included within, ointments or dressings for facilitating wound healing. The lipo-derived cells can also be used as experimental reagents. For example, they can be employed to help discover agents responsible for early events in differentiation. For example, the inventive cells can be exposed to a medium for inducing a particular line of differentiation and then assayed for differential expression of genes (e.g., by random-primed PCR or electrophoresis or protein or RNA, etc.).
  • As any of the steps for isolating the inventive stem cells or the lipo-derived lattice, the, the invention provides a kit for isolating such reagents from adipose tissues. The kit can include a means for isolating adipose tissue from a patient (e.g., a cannula, a needle, an aspirator, etc.), as well as a means for separating stromal cells (e.g., through methods described herein). The kit can be employed, for example, as a bedside source of stem cells that can then be re-introduced from the same individual as appropriate. Thus, the kit can facilitate the isolation of lipo-derived stem cells for implantation in a patient needing regrowth of a desired tissue type, even in the same procedure. In this respect, the kit can also include a medium for differentiating the cells, such as those set forth herein. As appropriate, the cells can be exposed to the medium to prime them for differentiation within the patient as needed. Of course, the kit can be used as a convenient source of stem cells for in vitro manipulation (e.g., cloning or differentiating as described herein). In another embodiment, the kit can be employed for isolating a lipo-derived lattice as described herein.
  • EXAMPLES
  • While one of skill in the art is fully able to practice the instant invention upon reading the foregoing detailed description, the following examples will help elucidate some of its features. In particular, they demonstrate the isolation of a human lipo-derived stem cell substantially free of mature adipocytes, the isolation of a clonal population of such cells, the ability of such cells to differentiate in vivo and in vitro, and the capacity of such cells to support the growth of other types of stem cells. The examples also demonstrate the isolation of a lipo-derived lattice substantially free of cells that is capable of serving as a suitable substrate for cell culture. Of course, as these examples are presented for purely illustrative purposes, they should not be used to construe the scope of the invention in a limited manner, but rather should be seen as expanding upon the foregoing description of the invention as a whole.
  • The procedures employed in these examples, such as surgery, cell culture, enzymatic digestion, histology, and molecular analysis of proteins and polynucleotides, are familiar to those of ordinary skill in this art. As such, and in the interest of brevity, experimental details are not recited in detail.
  • Example 1
  • This example demonstrates the isolation of a human lipo-derived stem cell substantially free of mature adipocytes.
  • Raw liposuction aspirate was obtained from patients undergoing elective surgery. Prior to the liposuction procedures, the patients were given epinephrine to minimize contamination of the aspirate with blood. The aspirate was strained to separate associated adipose tissue pieces from associated liquid waste. Isolated tissue was rinsed thoroughly with neutral phosphate buffered saline and then enzymatically dissociated with 0.075% w/v collagenase at 37° C. for about 20 minutes under intermittent agitation. Following the digestion, the collagenase was neutralized, and the slurry was centrifuged at about 260 g for about 10 minutes, which produced a multi-layered supernatant and a cellular pellet. The supernatant was removed and retained for further use, and the pellet was resuspended in an erythrocyte-lysing solution and incubated without agitation at about 25° C. for about 10 minutes. Following incubation, the medium was neutralized, and the cells were again centrifuged at about 250 g for about 10 minutes. Following the second centrifugation, the cells were suspended, and assessed for viability (using trypan blue exclusion) and cell number. Thereafter, they were plated at a density of about at about 1×106 cells/100 mm dish. They were cultured at 37° C. in DMEM+fetal bovine serum (about 10%) in about 5% CO2.
  • The majority of the cells were adherent, small, mononucleic, relatively agranular fibroblast-like cells containing no visible lipid droplets. The majority the cells stained negatively with oil-red O and von Kossa. The cells were also assayed for expression of telomerase (using a commercially available TRAP assay kit), using HeLa cells and HN-12 cells as positive controls. Human foreskin fibroblasts and HN-12 heated cell extracts were used as negative controls. Telomeric products were resolved onto a 12.5% polyacrylamide cells and the signals determined by phosphorimaging. Telemeric ladders representing telomerase activity were observed in the adipose-derived stem cells as well as the positive controls. No ladders were observed in the negative controls.
  • Thus, these cells were not identifiable as myocytes, adipocytes, chondrocytes, osteocytes, or blood cells. These results demonstrate that the adipose-derived cells express telomerase activity similar to that previously reported for human stem cells.
  • Subpopulations of these cells were then exposed to the following media to assess their developmental phenotype:
  • Adipogenesis Osteogenesis Myogenesis Chondrogenesis
    DMEM DMEM DMEM DMEM
    10% FBS 10% FBS 10% FBS 1% FBS
    0.5 mM IBMX 5% horse serum 5% horse serum 6.25 μg/ml insulin
    1 μM dexamethasone 0.1 μM dexamethasone 50 μM hydrocortisone 6.25 μg/ml transferrin
    10 μM insulin 50 μM ascorbate-2-phosphate 1% ABAM 10 ng/ml TGFβ1
    200 μM indomethacin 10 mM β-glycerophosphate 50 nM ascorbate-2-phosphate
    1% ABAM 1% ABAM 1% ABAM
  • A population was cultured at high density in the chondrogenic medium for several weeks. Histological analysis of the tissue culture and paraffin sections was performed with H&E, alcian blue, toludene blue, and Goldner's trichrome staining at 2, 7, and 14 days. Immunohistochemistry was performed using antibodies against chondroitin-4-sulfate and keratin sulfate and type II collagen. Qualitative estimate of matrix staining was also performed. The results indicated that cartilaginous spheroid nodules with a distinct border of perichondral cells formed as early as 48 hours after initial treatment. Untreated control cells exhibited no evidence of chondrogenic differentiation. These results confirm that the stem cells have chondrogenic developmental phenotype.
  • A population was cultured until near confluence and then exposed to the adipogenic medium for several weeks. The population was examined at two and four weeks after plating by colorimetric assessment of relative opacity following oil red-O staining. Adipogenesis was determined to be underway at two weeks and quite advanced at four weeks (relative opacity of 1 and 5.3, respectively). Bone marrow-derived stem cells were employed as a positive control, and these cells exhibited slightly less adipogenic potential (relative density of 0.7 and 2.8, respectively).
  • A population was cultured until near confluence and then exposed to the osteogenic medium for several weeks. The population was examined at two and four weeks after plating by colorimetric assessment of relative opacity following von Kossa staining. Osteogenesis was determined to be underway at two weeks and quite advanced at four weeks (relative opacity of 1.1 and 7.3, respectively. Bone marrow-derived stem cells were employed as a positive control, and these cells exhibited slightly less osteogenic potential (relative density of 0.2 and 6.6, respectively).
  • A population was cultured until near confluence and then exposed to the myogenic medium for several weeks. The population was examined at one, three, and six weeks after plating by assessment of multinucleated cells and expression of muscle-specific proteins (MyoD and myosin heavy chain). Human foreskin fibroblasts and skeletal myoblasts were used as controls. Cells expressing MyoD and myosin were found at all time points following exposure to the myogenic medium in the stem cell population, and the proportion of such cells increased at 3 and 6 weeks. Multinucleated cells were observed at 6 weeks. In contrast, the fibroblasts exhibited none of these characteristics at any time points.
  • These results demonstrate the isolation of a human lipo-derived pluripotent stem cell substantially free of mature adipocytes.
  • Example 2
  • This example demonstrates that lipo-derived stem cells do not differentiate in response to 5-azacytidine.
  • Lipo-derived stem cells obtained in accordance with Example 1 were cultured in the presence of 5-azacytidine. In contrast with bone marrow-derived stem cells, exposure to this agent did not induce myogenic differentiation (see Wakitani et al., supra).
  • Example 3
  • This example demonstrates the generation of a clonal population of human lipo-derived stem cells.
  • Cells isolated in accordance with the procedure set forth in Example 1 were plated at about 5,000 cells/100 mm dish and cultured for a few days as indicated in Example 1. After some rounds of cell division, some clones were picked with a cloning ring and transferred to wells in a 48 well plate. These cells were cultured for several weeks, changing the medium twice weekly, until they were about 80% to about 90% confluent (at 37° C. in about 5% CO2 in ⅔ F12 medium+20% fetal bovine serum and ⅓ standard medium that was first conditioned by the cells isolated in Example 1, “cloning medium”). Thereafter, each culture was transferred to a 35 mm dish and grown, and then retransferred to a 100 mm dish and grown until close to confluent. Following this, one cell population was frozen, and the remaining populations were plated on 12 well plates, at 1000 cells/well.
  • The cells were cultured for more than 15 passages in cloning medium and monitored for differentiation as indicated in Example 1. The undifferentiated state of each clone remained true after successive rounds of differentiation.
  • Populations of the clones then were established and exposed to adipogenic, chondrogenic, myogenic, and osteogenic medium as discussed in Example 1. It was observed that at least one of the clones was able to differentiate into bone, fat, cartilage, and muscle when exposed to the respective media, and most of the clones were able to differentiate into at least three types of tissues. The capacity of the cells to develop into muscle and cartilage further demonstrates the pluripotentiality of these lipo-derived stem cells.
  • These results demonstrate that the lipo-derived stem cells can be maintained in an undifferentiated state for many passages without the requirement for specially pre-screened lots of serum. The results also demonstrate that the cells retain pluripotentiality following such extensive passaging, proving that the cells are indeed stem cells and not merely committed progenitor cells.
  • Example 4
  • This example demonstrates the lipo-derived stem cells can support the culture of other types of stem cells.
  • Human lipo-derived stem cells were passaged onto 96 well plates at a density of about 30,000/well, cultured for one week and then irradiated. Human CD34+ hematopoetic stem cells isolated from umbilical cord blood were then seeded into the wells. Co-cultures were maintained in MyeloCult H5100 media, and cell viability and proliferation were monitored subjectively by microscopic observation. After two weeks of co-culture, the hematopoetic stem cells were evaluated for CD34 expression by flow cytometry.
  • Over a two-week period of co-culture with stromal cells, the hematopoetic stem cells formed large colonies of rounded cells. Flow analysis revealed that 62% of the cells remained CD34+. Based on microscopic observations, human adipo-derived stromal cells maintained the survival and supported the growth of human hematopoetic stem cells derived from umbilical cord blood.
  • These results demonstrate that stromal cells from human subcutaneous adipose tissue are able to support the ex vivo maintenance, growth and differentiation of other stem cells.
  • Example 5
  • This example demonstrates that the lipo-derived stem cells can differentiate in vivo.
  • Four groups (A-D) of 12 athymic mice each were implanted subcutaneously with hydroxyapatite/tricalcium phosphate cubes containing the following: Group A contained lipo-derived stem cells that had been pretreated with osteogenic medium as set forth in Example 1. Group B contained untreated lipo-derived stem cells. Group C contained osteogenic medium but no cells. Group D contained non-osteogenic medium and no cells. Within each group, six mice were sacrificed at three weeks, and the remaining mice sacrificed at eight weeks following implantation. The cubes were extracted, fixed, decalcified, and sectioned. Each section was analyzed by staining with H&E, Mallory bone stain, and immunostaining for osteocalcin.
  • Distinct regions of osteoid-like tissue staining for osteocalcin and Mallory bone staining was observed in sections from groups A and B. Substantially more osteoid tissue was observed in groups A and B than in the other groups (p<0.05 ANOVA), but no significant difference in osteogenesis was observed between groups A and B. Moreover, a qualitative increase in bone growth was noted in both groups A and B between 3 and 8 weeks. These results demonstrate that the lipo-derived stem cells can differentiate in vivo.
  • Example 6
  • This example demonstrates the isolation of a lipo-derived lattice substantially devoid of cells.
  • In one protocol withheld supernatant from Example 1 was subjected to enzymatic digestion for three days in 0.05% trypsin EDTA/100 U/ml deoxyribonuclease to destroy the cells. Every day the debris was rinsed in saline and fresh enzyme was added. Thereafter the material was rinsed in saline and resuspended in 0.05% collagease and about 0.1% lipase to partially digest the proteins and fat present. This incubation continued for two days.
  • In another protocol, the withheld supernatant from Example 1 was incubated in EDTA to eliminate any epithelial cells. The remaining cells were lysed using a buffer containing 1% NP40, 0.5% sodium deoxycholate, 0.1% SDS, 5 mM EDTA, 0.4M NaCl, 50 mMTris-HCL (pH 8) and protease inhibitors, and 10 μg/ml each of leupeptin, chymostatin, antipain, and pepstatin A. Finally, the tissue was extensively washed in PBS without divalent cations.
  • After both preparatory protocols, remaining substance was washed and identified as a gelatinous mass. Microscopic analysis of this material revealed that it contained no cells, and it was composed of high amounts of collagen (likely type IV) and a wide variety of growth factors. Preparations of this material have supported the growth of cells, demonstrating it to be an excellent substrate for tissue culture.
  • INCORPORATION BY REFERENCE
  • All sources (e.g., inventor's certificates, patent applications, patents, printed publications, repository accessions or records, utility models, world-wide web pages, and the like) referred to or cited anywhere in this document or in any drawing, Sequence Listing, or Statement filed concurrently herewith are hereby incorporated into and made part of this specification by such reference thereto.
  • GUIDE TO INTERPRETATION
  • The foregoing is an integrated description of the invention as a whole, not merely of any particular element of facet thereof. The description describes “preferred embodiments” of this invention, including the best mode known to the inventors for carrying it out. Of course, upon reading the foregoing description, variations of those preferred embodiments will become obvious to those of ordinary skill in the art. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law.
  • As used in the foregoing description and in the following claims, singular indicators (e.g., “a” or “one”) include the plural, unless otherwise indicated. Recitation of a range of discontinuous values is intended to serve as a shorthand method of referring individually to each separate value falling within the range, and each separate value is incorporated into the specification as if it were individually listed. Additionally, the following terms are defined as follows:
      • An anlage is a primordial structure that has a capacity to develop into a specific mature structure.
      • A developmental phenotype is the potential of a cell to acquire a particular physical phenotype through the process of differentiation.
      • A hormone is any substance that is secreted by a cell and that causes a phenotypic change in the same or another cell upon contact.
      • A stem cell is a pluripotent cell that has the capacity to differentiate in accordance with at least two discrete developmental pathways.
  • As regards the claims in particular, the term “consisting essentially of” indicates that unlisted ingredients or steps that do not materially affect the basic and novel properties of the invention can be employed in addition to the specifically recited ingredients or steps. In contrast, terms such as “comprising,” “having,” and “including” indicate that any ingredients or steps can be present in addition to those recited. The term “consisting of” indicates that only the recited ingredients or steps are present, but does not foreclose the possibility that equivalents of the ingredients or steps can substitute for those specifically recited.

Claims (79)

1. A mammalian lipo-derived stem cell substantially free of mature adipocytes.
2. The cell of claim 1, which can be cultured in DMEM+about 10% fetal bovine serum for at least 15 passages without differentiating.
3. The cell of claim 2, which has two or more developmental phenotypes selected from the group of developmental phenotypes consisting of adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, splanchogenic, and stromal developmental phenotypes.
4. The cell of any of claims 1-3, which is human.
5. The cell of any of claims 1-4, which is genetically modified.
6. The cell of any of claims 1-5, which has a cell-surface bound intercellular signaling moiety.
7. The cell of any of claims 1-5, which secretes a hormone.
8. The cell of claim 7, wherein the hormone is selected from the group of hormones consisting of cytokines and growth factors.
9. A defined cell population comprising a cell of any of claims 1-8.
10. The defined cell population of claim 9, which is heterogeneous.
11. The defined cell population of claim 9 or 10, further compressing a stem cell selected from the group of cells consisting of neural stem cells (NSC), hematopoetic stem cells (HPC), embryonic stem cells (ESC) and mixtures thereof.
12. The defined cell population of claim 9, which consists essentially of cells according to any of claims 1-8.
13. The defined cell population of claim 9 or 12, which is substantially homogenous.
14. The defined cell population of claim 13, which is clonal.
15. A defined cell population consisting essentially of mesodermal stem cells (MHC), connective tissue stem cell (CTSC), or mixtures thereof, wherein the population is clonal.
16. The population of claim 15, wherein the stem cells have two or more developmental phenotypes selected from the group of developmental phenotypes consisting of adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, splanchogenic, and stromal developmental phenotypes.
17. A lipo-derived lattice comprising adipose tissue extracellular matrix matter substantially devoid of cells.
18. The lipo-derived lattice of claim 17, comprising a human protein, proteoglycan, glycoprotein, hyaluronin, or fibronectin molecule.
19. The lipo-derived lattice of claim 17 or 18, comprising a collagen selected from the group of collagens consisting of type I, type II, type III, type IV, type V, type VI collagen.
20. The lipo-derived lattice of any of claims 17-19, comprising a hormone.
21. The lipo-derived lattice of claim 20, wherein the hormone is selected from the group of hormones consisting of cytokines and growth factors.
22. The lipo-derived lattice of any of claims 17-21, which is substantially anhydrous.
23. The lipo-derived lattice of any of claims 17-22, which is lyophilized.
24. The lipo-derived lattice of any of claims 17-21, which is hydrated.
25. A kit comprising the lipo-derived lattice of any of claims 17-24 and one or more components selected from the group of components consisting of hydrating agents, cell culture substrates, cell culture media, antibiotic compounds, and hormones.
26. A composition comprising a cell and the lipo-derived lattice of any of claims 17-24.
27. A composition comprising the cell of any of claims 1-8 and a biologically compatible lattice.
28. A composition comprising the population of any of claims 9-16 and a biologically compatible lattice.
29. The composition of claim 27 or 28, wherein the lattice comprises polymeric material.
30. The composition of claim 29, wherein the polymeric material is formed of polymer fibers as a mesh or sponge.
31. The composition of claim 29 or 30, wherein the polymeric material comprises monomers selected from the group of monomers consisting of glycolic acid, lactic acid, propyl fumarate, caprolactone, hyaluronan, hyaluronic acid and combinations thereof.
32. The composition of any of claims 29-31, wherein the polymeric material comprises proteins, polysaccharides, polyhydroxy acids, polyorthoesters, polyanhydrides, polyphosphazenes, synthetic polymers or combinations thereof.
33. The composition of any of claims 29-32, wherein the polymeric material is a hydrogel formed by crosslinking of a polymer suspension having the cells dispersed therein.
34. The composition of any of claims 29-33, wherein the lattice further comprises a hormone selected from the group of hormones consisting of cytokines and growth factors.
35. The composition of any of claims 29-34, wherein the lattice is the lipo-derived lattice of any of claims 17-24.
36. A method of obtaining a genetically-modified cell comprising exposing the cell of any of claims 1-8 to a gene transfer vector comprising a nucleic acid including a transgene, whereby the nucleic acid is introduced into the cell under conditions whereby the transgene is expressed within the cell.
37. The method of claim 36, wherein the transgene encodes a protein conferring resistance to a toxin.
38. A method of delivering a transgene to an animal comprising (a) obtaining a genetically-modified cell in accordance with claim 36 or 37 and (b) introducing the cell into the animal, such that the transgene is expressed in vivo.
39. A method of differentiating the cell of any of claims 1-8 comprising culturing the cell in a morphogenic medium under conditions sufficient for the cell to differentiate.
40. The method of claim 39, wherein the medium is an adipogenic, chondrogenic, cardiogenic, dermatogenic, embryonic, fetal, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, and splanchogenic, or stromogenic media.
41. The method of claim 39 or 40, wherein the morphogenic medium is an adipogenic medium and the cell is monitored to identify adipogenic differentiation.
42. The method of claim 39 or 40, wherein the morphogenic medium is a chondrogenic medium and the cell is monitored to identify chondrogenic differentiation.
43. The method of claim 39 or 40, wherein the morphogenic medium is an embryonic or fetal medium and the cell is monitored to identify embryonic or fetal phenotype.
44. The method of claim 39 or 40, wherein the morphogenic medium is a myogenic medium and the cell is monitored to identify myogenic differentiation.
45. The method of claim 39 or 40, wherein the morphogenic medium is an osteogenic medium and the cell is monitored to identify osteogenic differentiation.
46. The method of claim 39 or 40, wherein the morphogenic medium is a stromal medium and the cell is monitored to identify stromal or hematopoetic differentiation.
47. The method of any of claims 39-46, wherein the cell differentiates in vitro.
48. The method of any of claims 39-46, wherein the cell differentiates in vivo.
49. A method of producing hormones, comprising (a) culturing the cell of claim 7 or 8 within a medium under conditions sufficient for the cell to secrete the hormone into the medium and (b) isolating the hormone from the medium.
50. A method of promoting the closure of a wound within a patient comprising introducing the cell of claim 7 or 8 into the vicinity of a wound under conditions sufficient for the cell to produce the hormone, whereby the presence of the hormone promotes closure of the wound.
51. A method of promoting neovascularization within tissue, comprising introducing the cell of claim 7 or 8 into the tissue under conditions sufficient for the cell to produce the hormone, whereby the presence of the hormone promotes neovascularization within the tissue.
52. The method of claim 51, wherein the tissue is within an animal.
53. The method of claim 51 or 52, wherein the tissue is a graft.
54. The method of any of claims 49-53, wherein the hormone is a growth factor selected from the group of growth factor consisting of human growth factor, nerve growth factor, vascular and endothelial cell growth factor, and members of the TGFβ superfamily.
55. A method of conditioning culture medium comprising exposing a cell culture medium to the cell of any of claims 1-7 under conditions sufficient for the cell to condition the medium.
56. The method of claim 55, wherein the medium is separated from the cell after it has been conditioned.
57. The method of any of claims 36-56, wherein the cell is within a population of any of claims 9-16.
58. A conditioned culture medium produced in accordance with the method of claim 55 or 56.
59. The conditioned culture medium of claim 58, which is substantially free of a cell of any of claims 1-7.
60. A method of culturing a stem cell comprising maintaining a stem cell in the conditioned medium of claim 58 or 59 under conditions for the stem cell to remain viable.
61. The method of claim 60, which further comprises permitting successive rounds of mitotic division of the stem cell to form an expanded population of stem cells.
62. The method of claim 60 or 61, wherein the medium is substantially free of the lipo-derived cells of any of claims 1-7.
63. The method of any of claims 60-62, wherein the medium contains lipo-derived cells of any of claims 1-7.
64. The method of claim 63, wherein a stem cell and a lipo-derived cell are in contact.
65. The method of any of claims 60-64, wherein a stem cell is a hemopoietic stem cell.
66. A method of producing animal matter comprising maintaining the composition of any of claims 18-26 under conditions sufficient for the cells within the composition to expand and differentiate to form the matter.
67. The method of claim 66, wherein the matter comprises a tissue type selected from the group of tissues consisting of adipose, cartilage, heart, dermal connective tissue, blood tissue, muscle, kidney, bone, pleural, and splancnic tissues, and combinations thereof.
68. The method of claim 66 or 67, wherein the matter comprises more than one tissue type.
69. The method of any of claims 66-68, wherein the matter comprises at least a portion of an animal organ.
70. The method of claim 66-68, wherein the matter comprises at least a portion of an animal limb.
71. The method of any of claims 66-70, wherein the composition is maintained in vitro.
72. The method of any of claims 66-70, wherein the composition is introduced into an animal and maintained in vivo.
73. An implant comprising the cell of any of claims 1-7.
74. An implant comprising the population of any of 8-13.
75. An implant comprising the lipo-derived lattice of any of claims 14-16.
76. An implant comprising the composition of any of claims 17-26.
77. A kit for isolating stem cells from adipose tissues comprising a means for isolating adipose tissue from a patient and a means for separating stem cells from the remainder of the adipose tissue.
78. The kit of claim 77, further comprising a medium for differentiating the stem cells.
79. The kit of claim 78, wherein the medium is selected from the group of media consisting of adipogenic, chondrogenic, cardiogenic, dermatogenic, embryonic, fetal, hematopoetic, hemangiogenic, myogenic, nephrogenic, neurogenic, neuralgiagenic, urogenitogenic, osteogenic, pericardiogenic, peritoneogenic, pleurogenic, and splanchogenic, and stromogenic media.
US12/653,940 1999-03-10 2009-12-18 Adipose-derived stem cells and lattices Abandoned US20100173411A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/653,940 US20100173411A1 (en) 1999-03-10 2009-12-18 Adipose-derived stem cells and lattices
US13/226,338 US20110318833A1 (en) 1999-03-10 2011-09-06 Adipose-derived stem cells and laticces
US13/852,463 US20130224858A1 (en) 1999-03-10 2013-03-28 Adipose-derived stem cells and lattices
US14/659,308 US20150191698A1 (en) 1999-03-10 2015-03-16 Adipose-derived stem cells and lattices

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US12371199P 1999-03-10 1999-03-10
US16246299P 1999-10-29 1999-10-29
PCT/US2000/006232 WO2000053795A1 (en) 1999-03-10 2000-03-10 Adipose-derived stem cells and lattices
US09/936,665 US6777231B1 (en) 1999-03-10 2000-03-10 Adipose-derived stem cells and lattices
US10/797,371 US20040171146A1 (en) 1999-03-10 2004-03-09 Adipose-derived stem cells and lattices
US12/653,940 US20100173411A1 (en) 1999-03-10 2009-12-18 Adipose-derived stem cells and lattices

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/797,371 Division US20040171146A1 (en) 1999-03-10 2004-03-09 Adipose-derived stem cells and lattices

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/226,338 Continuation US20110318833A1 (en) 1999-03-10 2011-09-06 Adipose-derived stem cells and laticces

Publications (1)

Publication Number Publication Date
US20100173411A1 true US20100173411A1 (en) 2010-07-08

Family

ID=32854187

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/936,665 Expired - Lifetime US6777231B1 (en) 1999-03-10 2000-03-10 Adipose-derived stem cells and lattices
US10/797,371 Abandoned US20040171146A1 (en) 1999-03-10 2004-03-09 Adipose-derived stem cells and lattices
US12/653,940 Abandoned US20100173411A1 (en) 1999-03-10 2009-12-18 Adipose-derived stem cells and lattices
US13/226,338 Abandoned US20110318833A1 (en) 1999-03-10 2011-09-06 Adipose-derived stem cells and laticces
US13/852,463 Abandoned US20130224858A1 (en) 1999-03-10 2013-03-28 Adipose-derived stem cells and lattices
US14/659,308 Abandoned US20150191698A1 (en) 1999-03-10 2015-03-16 Adipose-derived stem cells and lattices

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/936,665 Expired - Lifetime US6777231B1 (en) 1999-03-10 2000-03-10 Adipose-derived stem cells and lattices
US10/797,371 Abandoned US20040171146A1 (en) 1999-03-10 2004-03-09 Adipose-derived stem cells and lattices

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/226,338 Abandoned US20110318833A1 (en) 1999-03-10 2011-09-06 Adipose-derived stem cells and laticces
US13/852,463 Abandoned US20130224858A1 (en) 1999-03-10 2013-03-28 Adipose-derived stem cells and lattices
US14/659,308 Abandoned US20150191698A1 (en) 1999-03-10 2015-03-16 Adipose-derived stem cells and lattices

Country Status (1)

Country Link
US (6) US6777231B1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10092600B2 (en) 2013-07-30 2018-10-09 Musculoskeletal Transplant Foundation Method of preparing an adipose tissue derived matrix
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same

Families Citing this family (221)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030082152A1 (en) * 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
US6777231B1 (en) * 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
KR100979664B1 (en) * 1999-03-10 2010-09-02 유니버시티 오브 피츠버그 오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Adipose-derived stem cells and lattices
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US8609412B2 (en) * 1999-08-05 2013-12-17 Regents Of The University Of Minnesota Mapc generation of lung tissue
JP5398941B2 (en) * 1999-08-05 2014-01-29 エイビーティー ホールディング カンパニー Pluripotent adult stem cell and method for isolating the same
US8252280B1 (en) 1999-08-05 2012-08-28 Regents Of The University Of Minnesota MAPC generation of muscle
US8075881B2 (en) * 1999-08-05 2011-12-13 Regents Of The University Of Minnesota Use of multipotent adult stem cells in treatment of myocardial infarction and congestive heart failure
US7015037B1 (en) * 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US7927587B2 (en) * 1999-08-05 2011-04-19 Regents Of The University Of Minnesota MAPC administration for the treatment of lysosomal storage disorders
US10638734B2 (en) 2004-01-05 2020-05-05 Abt Holding Company Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
EP1367899A4 (en) * 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US6702744B2 (en) 2001-06-20 2004-03-09 Advanced Cardiovascular Systems, Inc. Agents that stimulate therapeutic angiogenesis and techniques and devices that enable their delivery
DE10144326B4 (en) * 2001-09-10 2005-09-22 Siemens Ag Method and system for monitoring a tire air pressure
US20030054331A1 (en) * 2001-09-14 2003-03-20 Stemsource, Inc. Preservation of non embryonic cells from non hematopoietic tissues
US8608661B1 (en) 2001-11-30 2013-12-17 Advanced Cardiovascular Systems, Inc. Method for intravascular delivery of a treatment agent beyond a blood vessel wall
US20050008626A1 (en) * 2001-12-07 2005-01-13 Fraser John K. Methods of using adipose tissue-derived cells in the treatment of cardiovascular conditions
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
US8404229B2 (en) * 2001-12-07 2013-03-26 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to treat acute tubular necrosis
US20060204556A1 (en) * 2001-12-07 2006-09-14 Cytori Therapeutics, Inc. Cell-loaded prostheses for regenerative intraluminal applications
US7651684B2 (en) * 2001-12-07 2010-01-26 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in augmenting autologous fat transfer
US7595043B2 (en) * 2001-12-07 2009-09-29 Cytori Therapeutics, Inc. Method for processing and using adipose-derived stem cells
US9597395B2 (en) 2001-12-07 2017-03-21 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in the treatment of cardiovascular conditions
CA2469370C (en) * 2001-12-07 2014-07-08 Macropore Biosurgery, Inc. Adipose-derived cell processing unit
US7514075B2 (en) * 2001-12-07 2009-04-07 Cytori Therapeutics, Inc. Systems and methods for separating and concentrating adipose derived stem cells from tissue
US8105580B2 (en) 2001-12-07 2012-01-31 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to promote wound healing
US7585670B2 (en) 2001-12-07 2009-09-08 Cytori Therapeutics, Inc. Automated methods for isolating and using clinically safe adipose derived regenerative cells
US7771716B2 (en) 2001-12-07 2010-08-10 Cytori Therapeutics, Inc. Methods of using regenerative cells in the treatment of musculoskeletal disorders
WO2006075986A1 (en) * 2005-01-10 2006-07-20 Macropore Biosurgery, Inc. Devices and methods for monitoring, managing, and servicing medical devices
CN1620306A (en) * 2001-12-20 2005-05-25 马克罗珀尔公司 Systems and methods for treating patients with collagen-rich material extracted from adipose tissue
WO2003080801A2 (en) * 2002-03-19 2003-10-02 Advanced Research & Technology Transfer Adipose stromal stem cells for tissue and vascular modification
US7992725B2 (en) * 2002-05-03 2011-08-09 Biomet Biologics, Llc Buoy suspension fractionation system
US7832566B2 (en) 2002-05-24 2010-11-16 Biomet Biologics, Llc Method and apparatus for separating and concentrating a component from a multi-component material including macroparticles
US20030205538A1 (en) 2002-05-03 2003-11-06 Randel Dorian Methods and apparatus for isolating platelets from blood
AU2003249642A1 (en) 2002-05-24 2003-12-12 Biomet Manufacturing Corp. Apparatus and method for separating and concentrating fluids containing multiple components
US7845499B2 (en) 2002-05-24 2010-12-07 Biomet Biologics, Llc Apparatus and method for separating and concentrating fluids containing multiple components
US20060278588A1 (en) 2002-05-24 2006-12-14 Woodell-May Jennifer E Apparatus and method for separating and concentrating fluids containing multiple components
US7361368B2 (en) 2002-06-28 2008-04-22 Advanced Cardiovascular Systems, Inc. Device and method for combining a treatment agent and a gel
US20040235121A1 (en) * 2002-08-29 2004-11-25 Integrigen, Inc. High copy number plasmids and their derivatives
WO2004050859A2 (en) 2002-11-27 2004-06-17 Regents Of The University Of Minnesota Homologous recombination in multipotent adult progenitor cells
US20040193274A1 (en) * 2003-03-28 2004-09-30 Trieu Hai H. Materials and methods for augmenting and/or repairing intervertebral discs
US8383158B2 (en) 2003-04-15 2013-02-26 Abbott Cardiovascular Systems Inc. Methods and compositions to treat myocardial conditions
US8821473B2 (en) 2003-04-15 2014-09-02 Abbott Cardiovascular Systems Inc. Methods and compositions to treat myocardial conditions
US8038991B1 (en) 2003-04-15 2011-10-18 Abbott Cardiovascular Systems Inc. High-viscosity hyaluronic acid compositions to treat myocardial conditions
US20040213767A1 (en) * 2003-04-23 2004-10-28 Marc Hendriks Methods for using adipose-derived cells for healing of aortic aneurysmal tissue
US7387645B2 (en) * 2003-04-25 2008-06-17 Medtronic Vascular, Inc. Cellular therapy to heal vascular tissue
AU2004260638A1 (en) * 2003-06-18 2005-02-10 Macropore Biosurgery Inc. Methods of using adipose tissue-derived cells in augmenting autologous fat transfer
WO2005003320A2 (en) * 2003-07-02 2005-01-13 Regents Of The University Of Minnesota Neuronal differentiation of stem cells
US7744869B2 (en) * 2003-08-20 2010-06-29 Ebi, Llc Methods of treatment using electromagnetic field stimulated mesenchymal stem cells
US20050084962A1 (en) * 2003-08-20 2005-04-21 Bruce Simon Methods of treatment using electromagnetic field stimulated stem cells
US9074190B2 (en) * 2003-10-07 2015-07-07 Biomaster, Inc. Cell differentiation of adipose-derived precursor cells
NZ597965A (en) * 2003-10-08 2013-09-27 Vet Stem Inc Methods of preparing and using stem cell compositions and kits comprising the same
WO2005042730A2 (en) * 2003-11-04 2005-05-12 Biomaster, Inc. Method and system for preparing stem cells from fat tissue
AU2004296848A1 (en) 2003-12-04 2005-06-23 Regents Of The University Of Minnesota Compositions and methods for the treatment of lysosomal storage disorders
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
CN101080486B (en) * 2004-04-23 2012-05-16 佰欧益股份有限公司 Multi-lineage progenitor cells
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
US20060051865A1 (en) * 2004-08-31 2006-03-09 Higgins Joel C Systems and methods for isolating stromal cells from adipose tissue and uses thereof
WO2006062989A1 (en) * 2004-12-07 2006-06-15 Bacterin International, Inc. Three-dimensional cell culsture system
US8119398B2 (en) * 2004-12-30 2012-02-21 Primegen Biotech Llc Adipose-derived stem cells for tissue regeneration and wound healing
US7708152B2 (en) 2005-02-07 2010-05-04 Hanuman Llc Method and apparatus for preparing platelet rich plasma and concentrates thereof
EP2666494B1 (en) 2005-02-07 2018-01-17 Hanuman LLC Platelet rich plasma concentrate apparatus and method
US7866485B2 (en) 2005-02-07 2011-01-11 Hanuman, Llc Apparatus and method for preparing platelet rich plasma and concentrates thereof
WO2006086639A1 (en) * 2005-02-10 2006-08-17 Regents Of The University Of Minnesota Vascular/lymphatic endothelial cells
WO2007021321A2 (en) * 2005-04-01 2007-02-22 Wake Forest University Health Sciences Transcriptional profiling of stem cells and their multilineage differentiation
US8303972B2 (en) 2005-04-19 2012-11-06 Advanced Cardiovascular Systems, Inc. Hydrogel bioscaffoldings and biomedical device coatings
US8828433B2 (en) 2005-04-19 2014-09-09 Advanced Cardiovascular Systems, Inc. Hydrogel bioscaffoldings and biomedical device coatings
US9539410B2 (en) 2005-04-19 2017-01-10 Abbott Cardiovascular Systems Inc. Methods and compositions for treating post-cardial infarction damage
US8187621B2 (en) 2005-04-19 2012-05-29 Advanced Cardiovascular Systems, Inc. Methods and compositions for treating post-myocardial infarction damage
US20080125745A1 (en) 2005-04-19 2008-05-29 Shubhayu Basu Methods and compositions for treating post-cardial infarction damage
AU2005331559B2 (en) * 2005-05-05 2012-04-19 Regents Of The University Of Minnesota Use of NK cell inhibition to facilitate persistence of engrafted MHC-I negative cells
US20080311084A1 (en) * 2005-05-05 2008-12-18 Verfaillie Catherine M Mapc Engraftment in the Hematopoietic System
US20080187518A1 (en) * 2005-05-25 2008-08-07 University Of Virginia Patent Foundation Production of Osteoclasts from Adipose Tissues
US20070122392A1 (en) * 2005-06-22 2007-05-31 Sharon Gerecht-Nir Propagation of undifferentiated embryonic stem cells in hyaluronic acid hydrogel
WO2007009981A1 (en) * 2005-07-15 2007-01-25 Medizinische Hochschule Hannover Medium and process for tissue cultivation
US7531355B2 (en) * 2005-07-29 2009-05-12 The Regents Of The University Of California Methods and compositions for smooth muscle reconstruction
US20080194024A1 (en) * 2005-07-29 2008-08-14 Mays Robert W Culture of Non-Embryonic Cells at High Cell Density
US20070027543A1 (en) * 2005-08-01 2007-02-01 Gimble Jeffrey M Use of adipose tissue-derived stromal cells in spinal fusion
EP1924606A4 (en) 2005-08-25 2010-01-13 Repair Technologies Inc Devices, compositions and methods for the protection and repair of cells and tissues
AU2006287306A1 (en) * 2005-09-09 2007-03-15 Duke University Tissue engineering methods and compositions
CA2623353C (en) 2005-09-23 2017-02-21 Cellerix, S.L. Cell populations having immunoregulatory activity, method for isolation and uses
US7989205B2 (en) * 2005-10-06 2011-08-02 American Cryostem Corporation Cell culture media, kits and methods of use
EP1941032A2 (en) * 2005-10-14 2008-07-09 Regents Of The University Of Minnesota Differentiation of non-embryonic stem cells to cells having a pancreatic phenotype
US20070105769A1 (en) * 2005-11-07 2007-05-10 Ebi, L.P. Methods of treating tissue defects
KR100679642B1 (en) * 2005-11-16 2007-02-06 주식회사 알앤엘바이오 Multipotent stem cell derived from human adipose tissue and cellular therapeutic agents comprising the same
WO2007081631A2 (en) * 2006-01-04 2007-07-19 Healtheuniverse, Inc. Method of producing insulin-secreting cells from adult stem cells via transfection
EP1974022B1 (en) * 2006-01-27 2015-12-23 Prostemics Co., Ltd. Mass producing method of growth factor using adipose derived adult stem cells
US7520888B2 (en) 2006-02-14 2009-04-21 Warsaw Orthopedic, Inc. Treatment of the vertebral column
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
TW200813225A (en) * 2006-03-06 2008-03-16 Univ Louisiana State Biocompatible scaffolds and adipose-derived stem cells
EP2422803A3 (en) 2006-03-07 2013-03-13 Geeta Shroff Compositions comprising human embryonic stem cells and their derivatives, methods of use, and methods of preparation
US7727763B2 (en) * 2006-04-17 2010-06-01 Bioe, Llc Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US8567609B2 (en) 2006-05-25 2013-10-29 Biomet Biologics, Llc Apparatus and method for separating and concentrating fluids containing multiple components
WO2007139551A1 (en) * 2006-05-30 2007-12-06 Cytori Therapeutics, Inc. Systems and methods for manipulation of regenerative cells from adipose tissue
WO2008013863A2 (en) * 2006-07-26 2008-01-31 Cytori Therapeutics, Inc. Generation of adipose tissue and adipocytes
US7732190B2 (en) 2006-07-31 2010-06-08 Advanced Cardiovascular Systems, Inc. Modified two-component gelation systems, methods of use and methods of manufacture
WO2008018450A1 (en) * 2006-08-08 2008-02-14 National University Corporation Nagoya University Cell preparation containing multipotential stem cells originating in fat tissue
US9242005B1 (en) 2006-08-21 2016-01-26 Abbott Cardiovascular Systems Inc. Pro-healing agent formulation compositions, methods and treatments
WO2008054825A2 (en) * 2006-11-03 2008-05-08 Aastrom Biosciences, Inc. Mixed cell populations for tissue repair and separation technique for cell processing
US8741326B2 (en) 2006-11-17 2014-06-03 Abbott Cardiovascular Systems Inc. Modified two-component gelation systems, methods of use and methods of manufacture
US9005672B2 (en) 2006-11-17 2015-04-14 Abbott Cardiovascular Systems Inc. Methods of modifying myocardial infarction expansion
US8192760B2 (en) 2006-12-04 2012-06-05 Abbott Cardiovascular Systems Inc. Methods and compositions for treating tissue using silk proteins
WO2008109407A2 (en) 2007-03-02 2008-09-12 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Extracellular matrix-derived gels and related methods
US8034014B2 (en) * 2007-03-06 2011-10-11 Biomet Biologics, Llc Angiogenesis initation and growth
US7632340B2 (en) * 2007-03-07 2009-12-15 Hamilton Beach Brands, Inc. Air purifier for removing particles or contaminants from air
US8328024B2 (en) 2007-04-12 2012-12-11 Hanuman, Llc Buoy suspension fractionation system
EP2146794B1 (en) 2007-04-12 2016-10-19 Biomet Biologics, LLC Buoy suspension fractionation system
KR20080103637A (en) 2007-05-25 2008-11-28 주식회사 알앤엘바이오 Composition for treating of ischemic limb disease comprising stem cells derived from adipose tissue
EP2201910B1 (en) * 2007-06-01 2013-04-03 Allergan, Inc. Biological tissue growth support trough induced tensile stress
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
EP2245141B1 (en) * 2008-01-18 2018-03-14 Regents of the University of Minnesota Stem cell aggregates and methods for making and using
US20090192528A1 (en) * 2008-01-29 2009-07-30 Biomet Biologics, Inc. Method and device for hernia repair
WO2009102452A2 (en) * 2008-02-11 2009-08-20 The Johns Hopkins University Compositions and methods for implantation of adipose tissue and adipose tissue products
EP2567692B1 (en) 2008-02-27 2016-04-06 Biomet Biologics, LLC Use of a device for obtaining interleukin-1 receptor antagonist rich solutions
EP2254991B1 (en) 2008-02-29 2018-08-22 Biomet Manufacturing, LLC A system and process for separating a material
WO2009120879A1 (en) * 2008-03-26 2009-10-01 Ams Research Corporation Treatment of pelvic floor disorders with an adipose-derived cell composition
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
US8012077B2 (en) 2008-05-23 2011-09-06 Biomet Biologics, Llc Blood separating device
CA2729576A1 (en) * 2008-07-02 2010-01-07 Allergan, Inc. Compositions and methods for tissue filling and regeneration
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
US20110208162A1 (en) * 2008-10-24 2011-08-25 Indiana University Rsearch and Technology Corporation Methods for preventing aggregation of adipose stromal cells
EP2362899A1 (en) 2008-10-31 2011-09-07 Katholieke Universiteit Leuven Optimized methods for differentiation of cells into cells with hepatocyte and hepatocyte progenitor phenotypes, cells produced by the methods, and methods for using the cells
US20100112696A1 (en) * 2008-11-03 2010-05-06 Baxter International Inc. Apparatus And Methods For Processing Tissue To Release Cells
US9192695B2 (en) * 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
US8309343B2 (en) * 2008-12-01 2012-11-13 Baxter International Inc. Apparatus and method for processing biological material
US8187475B2 (en) 2009-03-06 2012-05-29 Biomet Biologics, Llc Method and apparatus for producing autologous thrombin
KR101109125B1 (en) 2009-03-24 2012-02-15 한국과학기술연구원 Method for the differentiation of stem cells into vascular endothelial cells and induction of angiogenesis using the same
US20100249924A1 (en) * 2009-03-27 2010-09-30 Allergan, Inc. Bioerodible matrix for tissue involvement
US8313954B2 (en) 2009-04-03 2012-11-20 Biomet Biologics, Llc All-in-one means of separating blood components
US9173975B2 (en) 2009-04-24 2015-11-03 Ingeneron, Inc. Reparative cell delivery via hyaluronic acid vehicles
US20120020937A1 (en) 2009-04-28 2012-01-26 Anterogen Co., Ltd. Autologous and allogenic adipose-derived stromal stem cell composition for treating fistulas
JP5917392B2 (en) * 2009-05-01 2016-05-11 ビミニ テクノロジーズ リミテッド ライアビリティ カンパニー Systems, methods and compositions for optimizing tissue and cell-enriched grafts
US9011800B2 (en) 2009-07-16 2015-04-21 Biomet Biologics, Llc Method and apparatus for separating biological materials
DK2456853T3 (en) * 2009-07-21 2021-02-01 Abt Holding Co USE OF STEM CELLS TO REDUCE LEUCOCYT EXTRAVASATION
US8986377B2 (en) 2009-07-21 2015-03-24 Lifecell Corporation Graft materials for surgical breast procedures
SG10201913618WA (en) * 2009-07-21 2020-03-30 Abt Holding Co Use of stem cells to reduce leukocyte extravasation
US8518681B2 (en) * 2009-12-04 2013-08-27 Sound Surgical Technologies Llc Selective lysing of cells using ultrasound
MX2012006759A (en) * 2009-12-17 2012-07-30 Univ Kingston Decellularized adipose tissue.
WO2011087743A2 (en) 2009-12-22 2011-07-21 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Decellularized adipose cell growth scaffold
US20110206647A1 (en) * 2010-02-25 2011-08-25 Abt Holding Company Modulation of Angiogenesis
WO2011106476A1 (en) * 2010-02-25 2011-09-01 Abt Holding Company Modulation of microglia activation
US9597358B2 (en) 2010-03-04 2017-03-21 University of Pittsburgh—of the Commonwealth System of Higher Education Bioengineered human corneal stromal tissue
US8591391B2 (en) 2010-04-12 2013-11-26 Biomet Biologics, Llc Method and apparatus for separating a material
CA3128483A1 (en) 2010-05-12 2011-11-17 Abt Holding Company Modulation of splenocytes in cell therapy
EP2575913B1 (en) 2010-05-25 2018-07-25 Cook Biotech Incorporated Methods, substrates, and systems useful for cell seeding of medical grafts
WO2011158125A2 (en) 2010-06-17 2011-12-22 Katholieke Universiteit Leuven Methods for differentiating cells into hepatic stellate cells and hepatic sinusoidal endothelial cells, cells produced by the methods, and methods for using the cells
FR2962443B1 (en) 2010-07-06 2017-11-17 Basf Beauty Care Solutions France Sas ADIPOSE TISSUE MODEL AND PROCESS FOR PREPARING THE SAME
US9260697B2 (en) 2010-07-09 2016-02-16 The Gid Group, Inc. Apparatus and methods relating to collecting and processing human biological material containing adipose
US9296984B2 (en) 2010-07-09 2016-03-29 The Gid Group, Inc. Tissue processing apparatus and method for processing adipose tissue
WO2013106655A1 (en) 2012-01-11 2013-07-18 The Gid Group, Inc. Method for processing adipose tissue and processing apparatus
US9206387B2 (en) 2010-07-09 2015-12-08 The Gid Group, Inc. Method and apparatus for processing adipose tissue
EP2609191B1 (en) 2010-08-24 2017-11-22 Regents Of The University Of Minnesota Non-static suspension culture of cell aggregates
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
NZ612801A (en) 2010-12-27 2014-12-24 Intellicell Biosciences Inc Ultrasonic cavitation derived stromal or mesenchymal vascular extracts and cells derived therefrom obtained from adipose tissue and use thereof
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
WO2012158910A2 (en) 2011-05-18 2012-11-22 The Regents Of The University Of California Compositions and methods for treating retinal diseases
EP2782535B1 (en) 2011-11-24 2016-10-19 Cook Biotech Incorporated Modifiable medical grafts and related apparatuses
US9512405B2 (en) 2011-12-14 2016-12-06 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Non-enzymatic method for isolating human adipose-derived stromal stem cells
US9162011B2 (en) 2011-12-19 2015-10-20 Allosource Flowable matrix compositions and methods
ITGE20120034A1 (en) * 2012-03-28 2013-09-29 Carlo Tremolada PREPARATION AND METHOD FOR THE PRODUCTION OF A PREPARATION INCLUDING MESENCHIMAL STEM CELLS
KR20150016970A (en) 2012-06-05 2015-02-13 머핀 인코포레이티드 Catheter systems and methods useful for cell therapy
EP2864472A4 (en) * 2012-06-20 2015-12-30 Celula Inc Materials and methods for processing cell populations
AU2013277350B2 (en) * 2012-06-21 2017-06-29 Lifecell Corporation Implantable prosthesis having acellular tissue attachments
US9642956B2 (en) 2012-08-27 2017-05-09 Biomet Biologics, Llc Apparatus and method for separating and concentrating fluids containing multiple components
BR112015004003B1 (en) 2012-09-06 2020-05-19 The Gid Group Inc apparatus for processing human biological material containing fibrous tissue and method for processing adipose tissue
WO2014052593A1 (en) 2012-09-26 2014-04-03 Cook Biotech Incorporated Medical device design, manufacture and testing systems
US9650608B2 (en) 2013-02-22 2017-05-16 Medivet America, Llc Activating adipose-derived stem cells for transplantation
US9867939B2 (en) 2013-03-12 2018-01-16 Allergan, Inc. Adipose tissue combinations, devices, and uses thereof
US20140271589A1 (en) 2013-03-15 2014-09-18 Biomet Biologics, Llc Treatment of collagen defects using protein solutions
US10143725B2 (en) 2013-03-15 2018-12-04 Biomet Biologics, Llc Treatment of pain using protein solutions
US9895418B2 (en) 2013-03-15 2018-02-20 Biomet Biologics, Llc Treatment of peripheral vascular disease using protein solutions
EP2970882B1 (en) 2013-03-15 2018-11-28 AlloSource Cell repopulated collagen matrix for soft tissue repair and regeneration
US9950035B2 (en) 2013-03-15 2018-04-24 Biomet Biologics, Llc Methods and non-immunogenic compositions for treating inflammatory disorders
US10208095B2 (en) 2013-03-15 2019-02-19 Biomet Manufacturing, Llc Methods for making cytokine compositions from tissues using non-centrifugal methods
SG10201914136UA (en) 2013-04-12 2020-03-30 Saverio Lafrancesca Improving organs for transplantation
US20140350516A1 (en) 2013-05-23 2014-11-27 Allergan, Inc. Mechanical syringe accessory
EP3038629B1 (en) 2013-09-05 2020-11-18 GID BIO, Inc. Method for processing adipose tissue
US9248384B2 (en) 2013-10-02 2016-02-02 Allergan, Inc. Fat processing system
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
US10029048B2 (en) 2014-05-13 2018-07-24 Allergan, Inc. High force injection devices
EP3160480B1 (en) 2014-06-30 2020-09-09 TiGenix, S.A.U. Mesenchymal stromal cells for treating rheumatoid arthritis
SG11201610844PA (en) 2014-06-30 2017-01-27 Tigenix S A U Mesenchymal stromal cells for treating sepsis
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
WO2016109947A1 (en) 2015-01-07 2016-07-14 Frontier Bio-Drug Development Limited Employing human adipose-derived stem cells to propagate serum-derived hepatitis c virus and use thereof
SG11201706680SA (en) 2015-03-10 2017-09-28 Allergan Pharmaceuticals Holdings (Ireland) Unlimited Company Multiple needle injector
US9713810B2 (en) 2015-03-30 2017-07-25 Biomet Biologics, Llc Cell washing plunger using centrifugal force
US9743949B2 (en) 2015-04-22 2017-08-29 Medline Industries, Inc. Two-dimensional needle array device and method of use
CN107921069B (en) 2015-05-05 2021-12-14 萨梅尔·斯洛基 Fat-depleted adipose tissue and devices and methods for making same
US9757721B2 (en) 2015-05-11 2017-09-12 Biomet Biologics, Llc Cell washing plunger using centrifugal force
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
EP3337427A1 (en) 2015-08-21 2018-06-27 Lifecell Corporation Breast treatment device
JP7244991B2 (en) 2015-11-24 2023-03-23 ロート製薬株式会社 Liver disease therapeutic agent containing adipose tissue-derived stromal cells and method for producing the same
SG10201914102UA (en) 2016-01-21 2020-03-30 Abt Holding Co Stem cells for wound healing
WO2017144552A1 (en) 2016-02-22 2017-08-31 Centauri Biotech. S.L. Pharmaceutical or veterinary cell compositions comprising mesenchymal stromal cells (mscs) and dimethyl sulfoxide (dmso)
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
KR102232054B1 (en) 2016-04-08 2021-03-26 알레간 인코포레이티드 Suction and injection device
US11629337B2 (en) 2016-05-11 2023-04-18 Animal Cell Therapy—Act Production of a canine beta cell line from an immature pancreas
US20170340434A1 (en) 2016-05-24 2017-11-30 Cook Medical Technologies Llc Device useful for localized therapeutic delivery without flow obstruction
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
JP2019526342A (en) 2016-08-30 2019-09-19 ライフセル コーポレーションLifeCell Corporation System and method for controlling medical devices
ES2832673T3 (en) 2016-08-31 2021-06-10 Lifecell Corp Device for breast treatment
WO2018074381A1 (en) 2016-10-18 2018-04-26 国立大学法人大阪大学 Kit for preparing disease-treating agent, disease-treating agent and method for preparing disease-treating agent
USD851777S1 (en) 2017-01-30 2019-06-18 Lifecell Corporation Canister-type device for tissue processing
JP7082372B2 (en) 2017-02-15 2022-06-08 ロート製薬株式会社 Pulmonary fibrosis treatment agent, PTPRR expression promoter and pulmonary fibrosis treatment kit
CN110177872A (en) 2017-03-03 2019-08-27 日本乐敦制药株式会社 Mescenchymal stem cell and pharmaceutical composition
EP3591037A4 (en) 2017-03-03 2020-11-11 Rohto Pharmaceutical Co., Ltd. Mesenchymal stem cells and therapeutic agent for liver disease
US20210401895A1 (en) 2017-03-30 2021-12-30 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with live biotherapeutics
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
WO2018226640A1 (en) 2017-06-05 2018-12-13 The Regents Of The University Of The University Of California Compositions for treating retinal diseases and methods for making and using them
US20200171092A1 (en) 2017-06-09 2020-06-04 Osaka University Therapeutic agent for dilated cardiomyopathy
US11732233B2 (en) 2017-07-18 2023-08-22 Gid Bio, Inc. Adipose tissue digestion system and tissue processing method
EP3707243A1 (en) 2017-11-08 2020-09-16 Animal Cell Therapy - Act Production of canine pancreatic islets from an immature pancreas
CN111565732A (en) 2018-01-12 2020-08-21 国立大学法人大阪大学 Agent for promoting normal differentiation-maturation of stratified squamous epithelial cell, agent for treating epithelial disease, and method for promoting normal differentiation-maturation of stratified squamous epithelial cell
EP3923718A1 (en) 2019-02-13 2021-12-22 TiGenix, S.A.U. Cryopreservation of stem cells
WO2020174002A1 (en) 2019-02-27 2020-09-03 Tigenix, S.A.U. Improved stem cell populations for allogeneic therapy
US11298220B2 (en) 2019-05-03 2022-04-12 Lifecell Corporation Breast treatment device
WO2021210515A1 (en) 2020-04-13 2021-10-21 国立大学法人東海国立大学機構 Agent for increasing cd25-positive regulatory t cells in kidney
WO2022176735A1 (en) 2021-02-22 2022-08-25 ロート製薬株式会社 Prophylactic and/or therapeutic agent for diabetes

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6555374B1 (en) * 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US6777231B1 (en) * 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
US7470537B2 (en) * 1999-03-10 2008-12-30 Univ California Adipose-derived stem cells and lattices

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4438032A (en) * 1981-01-30 1984-03-20 The Regents Of The University Of California Unique T-lymphocyte line and products derived therefrom
US5510254A (en) * 1986-04-18 1996-04-23 Advanced Tissue Sciences, Inc. Three dimensional cell and tissue culture system
US5336616A (en) * 1990-09-12 1994-08-09 Lifecell Corporation Method for processing and preserving collagen-based tissues for transplantation
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5226914A (en) 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young Scar inhibitory factor and use thereof
US5591625A (en) 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
FR2723315B1 (en) * 1994-08-02 1996-10-25 Cird Galderma METHOD AND COMPOSITION FOR STIMULATING DIFFERENTIATION OF PREADIPOCYTE CELLS AND RELATED THERAPEUTIC TREATMENTS
US5688531A (en) * 1994-12-23 1997-11-18 Ramot University Authority For Applied Research And Industrial Development, Ltd. Method for regulating bone forming cells
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5906934A (en) * 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5765567A (en) * 1995-03-22 1998-06-16 Knowlton; Edward W. Surgical method for breast reconstruction using a tissue flap
JP3781433B2 (en) 1995-11-16 2006-05-31 ケース ウエスターン リザーブ ユニバーシティ In vitro chondrogenesis 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
AU2730497A (en) 1996-04-17 1997-11-07 Case Western Reserve University Cryopreservation and extensive subculturing of human mesenchymal stem cells
EP2105138A3 (en) 1996-04-19 2013-06-05 Osiris Therapeutics, Inc. Regeneration and augmentation of bone using mesenchymal stem cells
WO1997041208A1 (en) 1996-04-26 1997-11-06 Case Western Reserve University Skin regeneration using mesenchymal stem cells
US5827740A (en) * 1996-07-30 1998-10-27 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
ATE306811T1 (en) 1996-12-06 2005-11-15 Osiris Therapeutics Inc IMPROVED CHONDROGENIC DIFFERENTIATION OF HUMAN MESENCHYME STEM CELLS
EP2110431A1 (en) 1997-05-13 2009-10-21 Osiris Therapeutics, Inc. Cartilage regeneration using human mesenchymal stem cells
US6709860B1 (en) * 1997-05-26 2004-03-23 Biovitrum Ab Animal model
US5786207A (en) 1997-05-28 1998-07-28 University Of Pittsburgh Tissue dissociating system and method
US5817050A (en) 1997-05-29 1998-10-06 Klein; Jeffrey A. Liposuction cannula
EP1028737B1 (en) * 1997-07-03 2007-04-04 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
ATE307195T1 (en) 1997-07-14 2005-11-15 Osiris Therapeutics Inc CARDIAC MUSCLE REGENERATION USING MESENCHYMAL STEM CELLS
US6077987A (en) 1997-09-04 2000-06-20 North Shore-Long Island Jewish Research Institute Genetic engineering of cells to enhance healing and tissue regeneration
EP1036163B1 (en) * 1997-12-02 2010-08-18 Artecel Sciences, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
AU2469299A (en) * 1998-01-23 1999-08-09 Cornell Research Foundation Inc. Purified populations of stem cells
US6153432A (en) * 1999-01-29 2000-11-28 Zen-Bio, Inc Methods for the differentiation of human preadipocytes into adipocytes
US20050076396A1 (en) * 1999-03-10 2005-04-07 Katz Adam J. Adipose-derived stem cells and lattices
KR100979664B1 (en) * 1999-03-10 2010-09-02 유니버시티 오브 피츠버그 오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Adipose-derived stem cells and lattices
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US7266457B1 (en) * 1999-05-21 2007-09-04 Hesperos, Llc High throughput functional genomics
US6429013B1 (en) * 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US7078232B2 (en) * 1999-08-19 2006-07-18 Artecel, Inc. Adipose tissue-derived adult stem or stromal cells for the repair of articular cartilage fractures and uses thereof
AU7611500A (en) 1999-09-24 2001-04-24 Abt Holding Company Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US20030161817A1 (en) * 2001-03-28 2003-08-28 Young Henry E. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US7078230B2 (en) * 2000-02-26 2006-07-18 Artecel, Inc. Adipose tissue-derived stromal cell that expresses characteristics of a neuronal cell
AU2001257436B2 (en) * 2000-04-28 2005-07-14 Children's Medical Center Corporation Isolation of mesenchymal stem cells and use 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
WO2008060374A2 (en) * 2006-10-06 2008-05-22 University Of Virginia Patent Foundation Methods and compositions useful for diabetic wound healing

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6777231B1 (en) * 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
US7470537B2 (en) * 1999-03-10 2008-12-30 Univ California Adipose-derived stem cells and lattices
US6555374B1 (en) * 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11305035B2 (en) 2010-05-14 2022-04-19 Musculoskeletal Transplant Foundatiaon Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10092600B2 (en) 2013-07-30 2018-10-09 Musculoskeletal Transplant Foundation Method of preparing an adipose tissue derived matrix
US11779610B2 (en) 2013-07-30 2023-10-10 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for using same
US10596201B2 (en) 2013-07-30 2020-03-24 Musculoskeletal Transplant Foundation Delipidated, decellularized adipose tissue matrix
US11191788B2 (en) 2013-07-30 2021-12-07 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US11596517B2 (en) 2015-05-21 2023-03-07 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11524093B2 (en) 2015-07-24 2022-12-13 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11806443B2 (en) 2015-08-19 2023-11-07 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11938245B2 (en) 2015-08-19 2024-03-26 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same

Also Published As

Publication number Publication date
US6777231B1 (en) 2004-08-17
US20110318833A1 (en) 2011-12-29
US20150191698A1 (en) 2015-07-09
US20130224858A1 (en) 2013-08-29
US20040171146A1 (en) 2004-09-02

Similar Documents

Publication Publication Date Title
US6777231B1 (en) Adipose-derived stem cells and lattices
EP1165830B1 (en) Adipose-derived stem cells and lattices
AU2004231498B2 (en) Materials and methods for augmenting and/or repairing intervertebral discs
Bartsch Jr et al. Propagation, expansion, and multilineage differentiation of human somatic stem cells from dermal progenitors
AU2008202757B2 (en) Adipose-derived stem cells and lattices
US20080152630A1 (en) Method of generation and expansion of tissue-progenitor cells and mature tissue cells from intact bone marrow or intact umbilical cord tissue
US20060134781A1 (en) Three-dimensional cell culture system
Seruya et al. Clonal population of adult stem cells: life span and differentiation potential
EP1282690A1 (en) Isolation of precursor cells and their use for tissue repair
ZA200106886B (en) Adipose-derived stem cells and lattices.
MXPA01008489A (en) Adipose-derived stem cells and lattices
Suenaga et al. Cell condensation and 3-dimensional dynamic environment in a rotation culture upregulates osteogenic differentiation of mesenchymal stromal cells
CN113667635A (en) Xeno-free medium and method for amplifying mesenchymal stem cells using the same

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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