US20070178590A1 - Pluripotential embryonic stem cells and methods of making same - Google Patents

Pluripotential embryonic stem cells and methods of making same Download PDF

Info

Publication number
US20070178590A1
US20070178590A1 US11/550,713 US55071306A US2007178590A1 US 20070178590 A1 US20070178590 A1 US 20070178590A1 US 55071306 A US55071306 A US 55071306A US 2007178590 A1 US2007178590 A1 US 2007178590A1
Authority
US
United States
Prior art keywords
cells
cell
factor
embryonic stem
stem cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/550,713
Inventor
Brigid Hogan
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
Priority claimed from US07/958,562 external-priority patent/US5453357A/en
Priority claimed from US08/217,921 external-priority patent/US5690926A/en
Application filed by Individual filed Critical Individual
Priority to US11/550,713 priority Critical patent/US20070178590A1/en
Publication of US20070178590A1 publication Critical patent/US20070178590A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0608Germ cells
    • C12N5/0611Primordial germ cells, e.g. embryonic germ cells [EG]
    • 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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • 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
    • C12N2502/99Coculture with; Conditioned medium produced by genetically 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening
    • 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
    • C12N2506/04Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from germ 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

Definitions

  • This invention relates to pluripotential embryonic stem cells and methods and compositions for making pluripotential embryonic stem cells.
  • PGCs Primordial germ cells in the mouse are thought to be derived from a small population of embryonic ectoderm (epiblast) cells set aside at the egg cylinder stage prior to gastrulation (Lawson and Pederson, 1992), or even earlier (Soriano and Jaenisch, 1986).
  • epiblast embryonic ectoderm
  • AP alkaline phosphatase
  • the PGCs migrate from the base of the allantois along the hind gut and reach the genital ridges by 11.5 days p.c. In the genital ridge, PGCs stop dividing at around 13.5 days p.c., and enter either mitotic arrest in the developing testis or meiosis in the ovary. In a few strains of mice, e.g. 129, this normal program can be disrupted if the male genital ridge from an 11.5 to 12.5 days p.c., embryo is grafted to an ectopic site such as the testis or kidney capsule.
  • PDGF is involved in the self renewal growth of O-2A cells (Noble et al., 1988; Raff et al., 1988). After a determined number of cell divisions, 0-2A cells may lose their responsiveness to PDGF and start differentiating into oligodendrocytes. If both PDGF and basic fibroblast growth factor (bFGF) are added in culture, O-2A progenitor cells keep growing without differentiation (Bogler et al., 1990).
  • bFGF basic fibroblast growth factor
  • ES embryonic stem cells
  • mice These murine ES cells were obtained from cultures of early blastocysts. Attempts at isolating ES cells from other animals apparently have failed.
  • mice embryos are expected to be epithelial and to have a very different morphology to mouse ES cells because ungulate embryos normally form an “embryonic disc”. This appears to be the basis of the allegation that the cells which they grow out of pig and cow blastocysts and which have a more epithelial morphology than mouse ES cells are, in fact, ES cells.
  • mouse embryos also develop an epithelial layer of pluripotential embryonic ectoderm or epiblast cells. This layer is called an “egg cylinder” rather than an “embryonic disc”. Therefore, there is apparently no strong embryological reason why the ungulates ES cells should have a different morphology to mouse ES cells.
  • the present invention satisfies this need by demonstrating that, in the presence of bFGF, SF and LIF, PGCs continue to proliferate in culture and give rise to colonies of ES cells. These stem cells can give rise to a wide variety of mature, differentiated cell types both in vitro and when injected into nude mice and when combined with embryos to form a chimera.
  • the present invention provides a non-mouse, including human, pluripotential embryonic stem cell which can:
  • the cells can form chimeras when combined with host embryos and give rise to mature sperm.
  • the invention further provides a method of making a pluripotential embryonic stem cell comprising culturing primordial germ cells, embryonic ectoderm cells and/or germ cell progenitors in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor to primordial germ cells under cell growth conditions, thereby making a pluripotential embryonic stem cell.
  • compositions useful to produce the pluripotent embryonic stem cells and methods of screening associated with the method of making the embryonic stem cell are also provided.
  • FIG. 1 shows the effect of growth factors on murine PGCs in culture.
  • FIG. 2 shows the morphology of primary and secondary cultures of PGCs and their descendants.
  • PGCs from 8.5 d p.c. embryos (A-E, G,H) or 12.5 d p.c. male genital ridges (F) were cultures on SI 4 -m220 cells as described and stained for AP activity.
  • FIG. 3 shows the effect of growth factors on males and female PGCs in culture.
  • Cells were dissociated from either male (squares) or female (circles) genital ridges from 12.5 day p.c. mouse embryos and cultured on SI 4 -m220 feeder cells either alone (empty symbols) or with soluble rSF, LIF and bFGF (filled symbols). Cells ere fixed and the number of AP positive cells counted. The experiment was carried out three times, with duplicate wells.
  • FIG. 4 shows the morphology of undifferentiated PGC derived ES cells and their differentiated derivatives.
  • FIG. 5 shows a photomicrograph of a colony of alkaline phosphatase positive cells derived in culture from an approximately 10.5 week old human embryonic testis. Following dissociation, testis cells were seeded into wells of a 24-well plate containing irradiated feeder cells (SI 4 h220) and growth factors 10 ng/ml bFGF, 60 ng/ml soluble SCF and 10 ng/ml LIF). After 5 days the cells were subculture at a dilution of 1:4 into wells containing a feeder layer of irradiated mouse embryo fibroblasts with the same cocktail of growth factors. After 10 days the cultures were fixed and stained for alkaline phosphatase activity.
  • irradiated feeder cells SI 4 h220
  • FIG. 5A The colony shown here (one of many) closely resembles colonies of alkaline phosphatase positive cells derived from primordial germ cells of the mouse embryo. (See FIG. 2C ). In particular, the human cells associate into tightly packed clusters (see arrow in FIG. 5C ).
  • FIGS. 5A, 5B and 5 C are different magnifications of the same colony.
  • the shadow in FIG. 5A is the edge of the well in which the cells were growing.
  • embryonic ectoderm is used herein. “Embryonic ectoderm” and “epiblast” can be used interchangeable to refer to the same cell type.
  • a “pluripotential embryonic stem cell” as used herein means a cell which can give rise to many differentiated cell types in an embryo or adult, including the germ cells (sperm and eggs). Pluripotent embryonic stem cells are also capable of self-renewal. Thus, these cells not only populate the germ line and give rise to a plurality of terminally differentiated cells which comprise the adult specialized organs, but also are able to regenerate themselves. This cell type is also referred to as an “ES cell” herein.
  • FGF fibroblast growth factor
  • FGF-1 acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • FGF-3 int-2
  • FGF-4 hst/K-FGF
  • FGF-5 FGF-6
  • FGF-7 FGF-8.
  • FGF-1 acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • FGF-3 int-2
  • FGF-4 hst/K-FGF
  • FGF-5 FGF-6
  • FGF-7 FGF-8.
  • Step factor is used herein.
  • SF is also called stem cell factor, mast cell growth factor and c-kit ligand in the art.
  • SF is a transmembrane protein with a cytoplasmic domain and an extracellular domain.
  • Soluble SF refers to a fragment cleaved from the extracellular domain at a specific proteolytic cleavage site.
  • Membrane associated SF refers to both normal SF before it has been cleaved or the SF which has been altered so that proteolytic cleavage cannot take place.
  • SF is well known in the art; see European Patent Publication No. 0 423 980 A1, corresponding to European Application No. 90310889.1.
  • LIF Leukemia Inhibitory Factor
  • FGF, SF and LIF are all proteins and as such certain modifications can be made to the proteins which are silent and do not remove the activity of the proteins as described herein. Such modifications include additions, substitutions and deletions. Methods modifying proteins are well established in the art (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989).
  • This invention provides a non-mouse pluriopotential ES cell which can be maintained on feeder layers for at least 20 passages, and give rise to embryoid bodies and multiple differentiated cell phenotypes in monolayer culture. Only those non-mouse animals which can be induced to form ES cells by the described methods are within the scope of the invention. Given the methods described herein, an ES cell can be made for any animal. However, mammals are preferred since many beneficial uses of mammalian ES cells exist. Mammalian ES cells such as those from rats, rabbits, guinea pigs, goats, pigs, cows, and humans can be obtained. Alternatively, embryos from these animals can be screened for the ability to produce ES cells.
  • the ES cells of this invention can be maintained for at least 20 passages. However, the ES cells can be capable of indefinite maintenance. Typically, after about 10 passages the cells are frozen so that the starting population is not altered by minor chromosomal alterations.
  • the non-mouse ES cells can be genetically manipulated to produce a desired characteristic.
  • the ES cells can be mutated to render a gene non-functional, e.g. the gene associated with cystic fibrosis or an oncogene.
  • functional genes can be inserted to allow for the production of that gene product in an animal, e.g. growth hormones or valuable proteins. Such methods are very well established in the art (Sedivy and Joyner (1992)).
  • the invention also provides a composition comprising:
  • this composition represents the composition after primordial germ cells, embryonic ectoderm or germ cells have become pluripotential ES cells.
  • the pluripotential ES cells can continue to be maintained in this composition or alternatively they can be maintained on a feeder layer.
  • LIF can be added to the feeder layer.
  • compositions comprising an FGF, LIF, membrane associated SF, and soluble SF in amounts to enhance the growth of, and allow the continued proliferation of primordial germ cells and the formation of pluripotent ES cells from the primordial germ cell.
  • This composition need not include primordial germ cells but comprises the various growth factors in amounts that promote the growth, proliferation and formation of pluripotent ES cells.
  • this composition can be sufficient for the establishment of pluripotent ES cells from the embryonic ectoderm cells or germ cells.
  • composition comprising: (a) mammalian primordial germ cells and/or germ cells and/or embryonic ectoderm cells; and (b) a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cells and the formation of pluripotent embryonic stem cells.
  • the compositions of the invention include a feeder layer.
  • feeder layers can either be cells or cell lines cultured for the purpose of culturing pluripotent ES cells.
  • feeder layers can be derived from or provided by the organ or tissue in which the primordial germ cells, embryonic ectoderm cells or germ cells are located, e.g., the gonad.
  • the somatic cells of the tissue or organ in which the desired cells are located are sufficient to provide the appropriate culture environment, a separate feeder layer is not required.
  • the feeder cells could be substituted with extracellular matrix plus bound growth factors. Feeder layers which are representative of those which can be utilized are set forth in the Examples. Naturally, the membrane associated SF can be contained on the cells of such a feeder layer.
  • compositions arise from the fact that FGF, LIF and SF are used either to enhance the growth and proliferation of primordial germ cells and/or embryonic ectoderm cells to become ES cells.
  • Growth and proliferation enhancing amounts can vary depending on the species or strain of the cells, and type of purity of the factors. Generally, 0.5 to 500 ng/ml of each factor within the culture solution is adequate. In a more narrow range, the amount is between 10 to 20 ng/ml for bFGF and LIF and between 10 to 100 ng/ml for SF. Regardless of whether the actual amounts are known, the optimal concentration of each factor can be routinely determined by one skilled in the art. Such determination is performed by titrating the factors individually and in combination until optimal growth is obtained.
  • FGF FGF
  • LIF fibroblast growth factor
  • SF fibroblast growth factor
  • other factors can also be tested to determine their ability to enhance the effect of FGF, LIF and SF on ES cell proliferation.
  • such other factors, or combinations of factors when used to enhance ES cell proliferation can be included within the above compositions.
  • compounds and fragments of FGF, LIF and SF which mimic the function of these factors can be used to enhance the growth and proliferation of the cells to become ES cells and are included within the scope of the invention.
  • the factors are essential to the formation of pluripotent ES cells.
  • the amount of the factors utilized is determined by the end result of the pluripotent ES cells.
  • the factors also serve to enhance the growth and allow the continued proliferation of the cells.
  • the factors also appear to help the cells survive.
  • FGF, LIF, and SF can be used to maintain ES cells.
  • the amounts of FGF, LIF and SF are necessary to maintain ES cells can be much less than that required to enhance growth or proliferation to become ES cells.
  • the cells may be maintained on a feeder layer without the addition of growth factors.
  • LIF can be added to enhance maintenance.
  • FGF or LIF from a species different from the source of the ES, primordial germ cell, germ cell or embryonic ectoderm cell can be utilized.
  • all the factors utilized and especially the SF utilized are preferably from the same species as the utilized cell type.
  • FGF, LIF or SF from various species can be routinely screened and selected for efficacy with a cell from a different species. Recombinant fragments of FGF, LIF or SF can also be screened for efficacy as well as organic compounds derived from, for example, chemical libraries.
  • the invention also provides a method of making a pluripotential ES cell comprising administering a growth enhancing amount of FGF, LIF, membrane associated SF, and soluble SF to primordial germ cells and/or embryonic ectoderm cells under cell growth conditions, thereby making a pluripotential ES cell.
  • primordial germ cells and embryonic ectoderm cells can be cultured as a composition in the presence of these factors to produce pluripotent ES cells.
  • the composition typically includes a feeder layer.
  • the invention also provides a method of making a mammalian pluripotential embryonic stem cell comprising culturing a germ cell or a composition from postnatal mammalian testis in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor, thereby making a pluripotential embryonic stem cell from a germ cell.
  • “Germ cells” as used herein means the cells which exist in neonatal or postnatal testis and are the progenitors of gametes. In the testis, theses germ cells represent a small population of stem cells capable of both self-renewal and differentiation into mature spermatogonia.
  • germ cells are the postnatal equivalent to the prenatal primordial germ cells and can include primitive or immature spermatogonia such as type A spermatogonia or any undifferentiated early stage cell that can form a pluripotent embryonic stem cell.
  • mice can be practiced utilizing any animal cell, especially mammal cells including mice, rats, rabbits, guinea pigs, goats, cows, pigs, humans, etc.
  • the ES cell produced by this method is also contemplated.
  • Also provided is a method of screening cells which can be promoted to become an ES cell comprising contacting the cells with FGF, LIF, membrane associated SF, and soluble SF amounts to enhance the growth of and allow proliferation of the cells and determining which cells become ES cells.
  • FGF FGF
  • LIF membrane associated SF
  • soluble SF soluble SF
  • the invention provides ES cells generated from virtually any animal
  • the invention provides a method is using the ES cells to contribute to chimeras in vivo in non-humans comprising injecting the cell into a blastocyst and growing the blastocyst in a foster mother.
  • aggregating the cell with a non-human morula stage embryo and growing the embryo in a foster mother can be used to produce a chimera.
  • Chimeric animals can subsequently be bred to obtain germ line transmission of ES cell traits.
  • the ES cells can be manipulated to produce a desired effect in the chimeric animal. The methods of producing such chimeric animals are well established (Robertson (1987)).
  • the ES cells can be used to derive cells for therapy to treat an abnormal condition.
  • derivatives of human ES cells could be placed in the brain to treat a neurodegenerative disease.
  • ES cells can be used to screen factors to determine which factors produce derivative (more differentiated cells. Many standard means to determine the presence of a more differentiated cell are well known in the art.
  • FGF, SF and LIF have been shown herein to be critical for making ES cells.
  • other members of the respective growth factor family could also be used to make ES cells.
  • members of each family can merely be substituted to determined if the new factor enhances the growth and allows the continued proliferation of PGCs or embryonic ectoderm cells to form ES cells.
  • the new LIF is merely combined with SF and FGF to determine if the new family member enhances the growth and allows the continued proliferation of PGCs or embryonic ectoderm cells.
  • this invention provides the use of family members and a method of screening family members for activity.
  • additional growth factors may be found useful in enhancing the growth and proliferation of PGCs or embryonic ectoderm cells from various animals.
  • This invention provides combining FGF, SF and LIF with other growth factors to obtain or enhance the production of ES cells.
  • a method of screening other growth factors for the ability to promote PGCs and embryonic ectoderm cells to form ES cells is also provided.
  • IL-11, IL-6, CNTF, NGF, IGFII flt3/flk-2 ligand and members of the Bone Morphogenetic Protein family are good screening candidates and can be used to promote ES cell formation.
  • SI/SI 4 cell line derived from a homozygous null SI/SI mouse embryo, and its derivative, SI 4 -m220, which stably expresses only membrane bound murine SF lacking exon 6 encoding the proteolytic cleavage site, were obtained from Dr. David Williams (Howard Hughes Medical Institute, Indiana University Medical School).
  • Other cell lines which produce adequate SF can be substituted for SI/SI 4 , for example mouse or human embryo fibroblasts or cell lines or somatic cell lines from gonads or genital ridges.
  • Combinations of feeder cells can also be utilized. They were maintained in DMEM with 10% calf serum and 50 ug/ml gentamicin.
  • feeder layers For making feeder layers they were irradiated (500 rads) and plated at a density of 2 ⁇ 105 per well of 24-well plates (Falcon) in the same medium, 24 hrs before use. Wells were pre-treated with 1% gelatin. STO cells stably transfected with human LIF and the bacterial neor gene (SLN) were obtained from Dr. Allan Bradley.
  • Embryos were from ICR females mated with (C57BLxDBA)F1 males. Noon of the day of plug is 0.5 day post coitum (p.c.). The caudal region of 8.5 day p.c. embryos (between the last somite and the base of the allantois) was dissociated into single cells by incubation at 37° C. with 0.05% trypsin, 0.02% EDTA in CA ++ /MG ++ free Dulbecco's phosphate-buffered saline (PBS) for about 10 mins with gentle pipetting. At this stage there are between about 149 and 379 PGCs in each embryo (Mintz and Russell, 1957).
  • PBS Dulbecco's phosphate-buffered saline
  • Cells from the equivalent of 0.5 embryo were seeded into a well containing feeder cells as above and 1 ml of DMEM, 2mM glutamine, 1 mM sodium pyruvate, 100 i.u./ml penicillin and 100 ug/ml streptomycin and 15% fetal bovine serum (PGC culture medium). Finely minced fragments of genital ridges from 1.5 and 12.5 day p.c. embryos were trypsinized as above and plated at a concentration of 0.1 embryo per well.
  • Growth factors were added at the time of seeding, usually at the following concentrations, which were shown to be optimal for PGC proliferation; recombinant human LIF and bFGF (10-20 ng/ml) and soluble rat SF (60 ng/ml). The medium was changed every day.
  • PGC cultures on SI 4 -m220 feeder cells on a chamber slide were washed twice with PBS containing 2% calf serum, 0.1% sodium azide and then incubated with mouse monoclonal antibody SSEA-1 (1:100 dilution) on ice for 30 min. After washing with PBS, cells were incubated for 30 mins with FITC-conjugated Fab′ fragment of goat anti mouse IgG (H+L) (Cappell, 1:5 dilution). After washing in PBS, cells were fixed in 4% paraformaldehyde before staining for AP.
  • mice Approximately 2 ⁇ 10 6 cells from three independent lines were injected subcutaneously into nude mice (three mice per line). After three weeks tumors were fixed in Bouin's fixative, processed for histology and sections stained with haematoxylin and cosin.
  • Membrane associated SF seems to play an important role in PGC proliferation since SI d mouse mutants which make only soluble SF have a reduced number of PGCs in vivo, and membrane associated SF is more effective than soluble SF in supporting PGC growth and survival in culture (Dolci et al., 1991; Matsui et al., 1991).
  • PGCs were cultured on SI 4 -m220 feeder cells which express only membrane associated SF (Matsui et al., 1991; Toksoz et al., 1991).
  • Both LIF and bFGF separately enhance PGC growth on SI 4 -m220 feeder cells, with added soluble rSF.
  • LIF and bFGF are added together, PGC growth is dramatically stimulated and the cells continue to proliferate through to day 6 in culture ( FIG. 1C ). The cells survive until day 8, at which time the feeder layer deteriorates, but they can be trypsinized and subcultured (see below).
  • Pregonadal PGCs are motile in vivo, and when cultured with LIF on a SI 4 -m220 feeder layer they form burst colonies of cells with a flattened and polarized morphology, characteristic of motile cells ( FIG. 2A ).
  • PGCs cultured on a SI 4 -m220 feeder layer with soluble SF and bFGF or with bFGF and LIF form discrete colonies of tightly packed cells. These colonies increase in size over day 6 in culture only when both bFGF and LIF are present ( FIG. 2C ).
  • Such colonies can be subcultured at least ten times and continue to give rise to colonies of similar morphology.
  • these cultures were transferred to feeder layers of STO cells in medium without added factors normally used for blastocyst-derived ES cell culture (Robertson, 1987). Under these conditions they continue to proliferate in an undifferentiated state, for a total of at least 20 passages.
  • transplantable teratocarcinomas can be induced experimentally by grafting genital ridges from 11.5 or 12.5 days p.c. male embryos of the 129 strain to an ectopic site.
  • Genital ridges were trysinized and the cells plated on an SI 4 -m220 feeder layer with soluble SF, LIF and bFGF.
  • the number of PGCs initially declines but increases after 3 days, and by 6 days colonies of densely packed, AP positive cells can be seen ( FIG. 2F ). If cells from male and female 12.5 days p.c. genital ridges are cultured separately, male PGCs increase in number and form colonies. In contrast, only a few female PGCs form colonies ( FIG. 3 ). The differentiation capacity of genital ridge-derived colonies has not so far been tested.
  • the cultures were grown in Dulbecco's modified Eagle's medium (DMEM) (Specialty Media, Lavallette, N.J.) supplemented with 0.01 mM non-essential amino acids (GIBCO), 2 mM glutamine (GIBCO), 50 ,ug/ml gentamycin (Sigma), 15% fetal bovine serum (selected batches, Hyclone) and 0.1 mM 2-mercaptoethanol (Sigma).
  • DMEM Dulbecco's modified Eagle's medium
  • ES lines derived from 8.5 day C57BL/6 embryos produced chimeras which transmitted the C57BL/6 genome through mature sperm (germ line transmission). This was judged by mating male chimeras with albino females and observing the production of pigmented pups. With the ES cell line known as TGC 8.5 10, nine chimeric males were obtained and four produced pigmented pups. With the ES cell line known as TGC 8.5 19 six chimeric males were obtained and one of these produced pigmented pups.
  • ES cells from other mammals can be produced using the methods described above for murine.
  • the mammalian cell of choice is simply substituted for murine and the murine methods are duplicated.
  • the appropriate species specific growth factors e.g. SF
  • Any additional growth factors which can promote the formation of ES cells can be determined by adding the growth factors to FGF, LIF, and SF as described above and monitored for an effect on ES formation.
  • testes were dissected from a 10.5 week human embryo. Younger or older embryos represent alternative sources. The preferred age range is between 8.5 weeks and 22 weeks.
  • Tissue was rinsed in buffered saline, and incubated in trypsin solution (0.25% trypsin, 1 mM EDTA in Ca ++ /MG ++ fre HEPES buffered saline) for 10 minutes at 37° C. This tissue was dissociated by pipetting and the cells plated into wells of a 24 well tray containing irradiated feeder cells.
  • the feeder cells were SI/SI mouse fibroblasts transfected with human membrane associated Stem Cell Factor (SI 4 h220 cells from Dr. David Williams, HHMI, Indiana State University School of Medicine).
  • An alternative feeder layer would consist of a mixture of mouse or human embryo fibroblasts and SI 4h220 cells, to provide a more coherent layer for long term cell attachment.
  • the culture medium consists of Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum supplemented with 10 ng/ml human bFGF, 60 ng/ml human Stem Cell Factor and 10 ng/ml human LIF.
  • DMEM Dulbecco's modified Eagle's medium
  • the amounts of bFGF can be increased (e.g. 20 ng/ml).
  • cultures were dissociated with trypsin solution as before and seeded into wells containing a feeder layer of irradiated mouse embryo fibroblasts.
  • the medium was supplemented with growth factors daily as above.
  • the addition of growth factors to the culture medium at this stage can be utilized, and a feeder layer of a mixture of mouse of human fibroblast and SI 4 h220 cells can be substituted.
  • Testes are dissected and the tunica removed. The testes are then incubated at 32° C. with mild shaking in buffered saline containing bovine serum albumin and collagenase (final concentration approximately 0.5 mg/ml). When the tissue has dissociated the tubules are allowed to settle out and then washed in saline several times. The collagenase treatment is repeated to remove all the cells surrounding the tubules (Leydig cells and connective tissue). The tubules are then washed and treated with hyaluronidase in buffered saline (final concentration approximately 0.5 mg/ml) at 32° C. until the tubules are free of adherent material.
  • the tubules are washed and placed onto tissue culture dishes coated with Poly-L-lysine.
  • the Sertoli cells attach strongly to the dish and spread out, while the germ cells remain in suspension.
  • the germ cells are collected and plated onto a layer or irradiated feeder cells comprising membrane bound and soluble stem cell factor, LIF and basic FGF as described above.
  • Chimeras utilizing non-murine non-human ES cells can likewise be produced utilizing the methods for murine described above and simply substituting the appropriate non-murine blastocyst for the species of ES utilized.
  • Cultures were trypsinized and seeded into wells # containing SI 4 -m220 feeder cells with either soluble rat SF alone # or soluble rat SF, LIF, and bFGF. Cultures were fixed and AP-positive # cells (day 1) or colonies (days 2 and 5) counted. Numbers are # mean ⁇ SEM from four experiments. Secondary cultures show a reduced # growth factor requirement compared with primary cultures.

Abstract

The present invention provides a non-mouse, including human, pluripotential embryonic stem cell which can: (a) be maintained on feeder layers for at least 20 passages; and (b) give rise to embryoid bodies and multiple differentiated cell phenotypes in monolayer culture. The invention further provides a method of making a pluripotential embryonic stem cell comprising culturing germs cells and germ cell progenitors in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor to primordial germ cells under cell growth conditions, thereby making a pluripotential embryonic stem cell. Also provided are composition useful to produce the pluripotent embryonic stem cells and methods of screening associated with the method of making the embryonic stem cell.

Description

  • This application is a continuation of, and claims the benefit of, application Ser. No. 08/813,829, filed Mar. 6, 1997, which status pending, which is a continuation of Ser. No. 08/217,921, filed Mar. 25, 1994, now U.S. Pat. No. 5,690,926 issued Nov. 25, 1997, which is a continuation-in-part of Ser. No. 07/958,562, filed Oct. 8, 1992, now U.S. Pat. No. 5,453,357, issued Sep. 26, 1995, which applications are hereby incorporated herein by this reference in their entireties.
  • This invention was made with government support under grant number HD25580-04 from the National Institute of Health Child Health and Development. The United States government has certain rights in the invention.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • This invention relates to pluripotential embryonic stem cells and methods and compositions for making pluripotential embryonic stem cells.
  • 2. Background Art
  • Primordial germ cells (PGCS) in the mouse are thought to be derived from a small population of embryonic ectoderm (epiblast) cells set aside at the egg cylinder stage prior to gastrulation (Lawson and Pederson, 1992), or even earlier (Soriano and Jaenisch, 1986). By 7 days post coitum (p.c.) about 100 alkaline phosphatase (AP) positive PGCs can be detected in the extra embryonic mesoderm just posterior to the definitive primitive streak (Ginsberg et al., 1990). These cells continue to proliferate and their number increases rapidly to around 25,000 at 13.5 days p.c. (Mintz and Russell, 1957; Tam and Snow, 1981). At the same time the PGCs migrate from the base of the allantois along the hind gut and reach the genital ridges by 11.5 days p.c. In the genital ridge, PGCs stop dividing at around 13.5 days p.c., and enter either mitotic arrest in the developing testis or meiosis in the ovary. In a few strains of mice, e.g. 129, this normal program can be disrupted if the male genital ridge from an 11.5 to 12.5 days p.c., embryo is grafted to an ectopic site such as the testis or kidney capsule. Under these conditions some PGCs give rise to teratomas and transplantable teratocarcinomas containing pluripotential embryonal carcinoma (EC) stem cells (Stevens and Makensen, 1961; Stevens, 1983; Noguchi and Stevens, 1982).
  • Previous studies have shown that steel factor (SF) and leukemia inhibitory factor (LIF) synergistically promote the survival and in some cases the proliferation of mouse PGCs in culture (Godin et al., 1991; Dolci et al., 1991; Matsui et al., 1991). However, under these conditions, PGCs have a finite proliferative capacity that correlates with their cessation of division in vivo. A similar finite proliferative capacity has been reported for liogodendrocyte-type 2 astrocyte (O-2A) progenitor cells in the rat optic nerve. In this case, PDGF is involved in the self renewal growth of O-2A cells (Noble et al., 1988; Raff et al., 1988). After a determined number of cell divisions, 0-2A cells may lose their responsiveness to PDGF and start differentiating into oligodendrocytes. If both PDGF and basic fibroblast growth factor (bFGF) are added in culture, O-2A progenitor cells keep growing without differentiation (Bogler et al., 1990).
  • Since pluripotential embryonic stem cells (ES can give rise to virtually any mature cell type they are of great value for uses such as creating genetically manipulated animals. However, according to the published scientific literature, it has previously been possible only to obtain ES cells from mice. These murine ES cells were obtained from cultures of early blastocysts. Attempts at isolating ES cells from other animals apparently have failed. One patent publication, Evans et al., published Apr. 5, 1990 under PCT Publication WO 90/03432, claims that pluripotential ES cells can be obtained from ungulate blastocysts in vitro. The application claims that these cells are expected to be epithelial and to have a very different morphology to mouse ES cells because ungulate embryos normally form an “embryonic disc”. This appears to be the basis of the allegation that the cells which they grow out of pig and cow blastocysts and which have a more epithelial morphology than mouse ES cells are, in fact, ES cells. However, mouse embryos also develop an epithelial layer of pluripotential embryonic ectoderm or epiblast cells. This layer is called an “egg cylinder” rather than an “embryonic disc”. Therefore, there is apparently no strong embryological reason why the ungulates ES cells should have a different morphology to mouse ES cells. In addition, the evidence presented in the Evans application for the differentiation of the cow and pig putative ES cell lines into differentiated cell types in monolayer culture, in embryoid bodies and in tumors, is not convincing. Therefore, there is a great need to produce and maintain ES cells from a variety of different animals.
  • The present invention satisfies this need by demonstrating that, in the presence of bFGF, SF and LIF, PGCs continue to proliferate in culture and give rise to colonies of ES cells. These stem cells can give rise to a wide variety of mature, differentiated cell types both in vitro and when injected into nude mice and when combined with embryos to form a chimera.
  • SUMMARY OF THE INVENTION
  • The present invention provides a non-mouse, including human, pluripotential embryonic stem cell which can:
  • (a) be maintained on feeder layers for at least 20 passages; and
  • (b) give rise to embryoid bodies and multiple differentiated cell phenotypes in monolayer culture.
  • In addition, in non-humans, the cells can form chimeras when combined with host embryos and give rise to mature sperm.
  • The invention further provides a method of making a pluripotential embryonic stem cell comprising culturing primordial germ cells, embryonic ectoderm cells and/or germ cell progenitors in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor to primordial germ cells under cell growth conditions, thereby making a pluripotential embryonic stem cell.
  • Also provided are compositions useful to produce the pluripotent embryonic stem cells and methods of screening associated with the method of making the embryonic stem cell.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the effect of growth factors on murine PGCs in culture.
  • (A) PGCs from 8.5 day p.pc. embryos were seeded into wells containing SI/SI4 feeder cells either along (open circles) or with soluble rSF (closed circles), soluble rSF and LIF (closed squares), or soluble rSF, LIF and bFGF (closed triangles). Cultures were fixed and the number of AP positive cells counted. (B) As in (A) except that cells were cultured without added factors (open circles), with soluble rSF (closed circles), with bFGF (closed triangles) or with soluble rSF and bFGF (open triangles).
  • (C) As in (A) except that cells were cultured on SI4-m220 cells either along (open circles) or with soluble rSF (closed circles), soluble rSF and LIF (closed squares), soluble rSF and bFGF (open triangles) and soluble rSF, LIF and bFGF (closed triangles).
  • Each experiment was carried out with duplicate wells and numbers are the means+s.e.m. of three separate experiments.
  • FIG. 2 shows the morphology of primary and secondary cultures of PGCs and their descendants. PGCs from 8.5 d p.c. embryos (A-E, G,H) or 12.5 d p.c. male genital ridges (F) were cultures on SI4-m220 cells as described and stained for AP activity.
  • (A) Primary culture after 4 days in the presence of LIF. Note that the AP positive cells are scattered among the feeder cells.
  • (B) Primary culture after 4 days in the presence of soluble rSF, LIF and bFGF. Note that the AP positive cells now form tight clumps.
  • (C) As for B, but after 6 days in culture.
  • (D) Secondary culture after 6 days in the presence of soluble rSF, LIF, and bFGF. In this colony all the cells are AP positive.
  • (E) As for D except that cells at the edges of the colony are AP negative.
  • (F) PGCs from 12.5 day p.c. male genital ridge were cultured for 6 days in the presence of soluble rSF, LIF and bFGF. Colonies of tightly packed AP positive cells are present.
  • (G) Colony of ES-like cells in a secondary culture with soluble SF, LIF and bFGF stained with SSEA-1 monoclonal antibody and for AP activity. Phase contrast microsopy.
  • (H) The same colony as in G viewed by fluorescence microscopy. AP positive cells also express SSEA-1.
  • (I) Colony grown under same conditions as (G) but stained without primary antibody Scale bars =200 μm.
  • FIG. 3 shows the effect of growth factors on males and female PGCs in culture. Cells were dissociated from either male (squares) or female (circles) genital ridges from 12.5 day p.c. mouse embryos and cultured on SI4-m220 feeder cells either alone (empty symbols) or with soluble rSF, LIF and bFGF (filled symbols). Cells ere fixed and the number of AP positive cells counted. The experiment was carried out three times, with duplicate wells.
  • FIG. 4 shows the morphology of undifferentiated PGC derived ES cells and their differentiated derivatives.
  • (A) Colony of densely packed ES-like cells obtained from PGCs of an 8.5 day p.c. embryo grown on SI4-m220 cells in the presence of soluble rSF, LIF, and bFGF for 6 days. Scal bar=100 μm.
  • (B) Simple embryoid bodies with an outer layer of endoderm (arrows) obtained after culturing PGC-derived ES cells for 4 days in suspension.
  • (C) Section of a teratoma obtained by injecting ES-like cells derived from PGCs of an 8.5 day p.c. embryo into a nude mouse. The region shown here contains neural tissue and pigmented epithelium. Scale bar=200 μm.
  • (D) Region of the same tumor as in (C) showing a dermoid cyst and secretory epithelium.
  • (E) Region of the same tumor as in C and D, showing bone and cartilage. The differentiated tissues shown in C-E were seen in addition to other tissue types in multiple tumors from all three lines tested.
  • FIG. 5 shows a photomicrograph of a colony of alkaline phosphatase positive cells derived in culture from an approximately 10.5 week old human embryonic testis. Following dissociation, testis cells were seeded into wells of a 24-well plate containing irradiated feeder cells (SI4 h220) and growth factors 10 ng/ml bFGF, 60 ng/ml soluble SCF and 10 ng/ml LIF). After 5 days the cells were subculture at a dilution of 1:4 into wells containing a feeder layer of irradiated mouse embryo fibroblasts with the same cocktail of growth factors. After 10 days the cultures were fixed and stained for alkaline phosphatase activity. The colony shown here (one of many) closely resembles colonies of alkaline phosphatase positive cells derived from primordial germ cells of the mouse embryo. (See FIG. 2C). In particular, the human cells associate into tightly packed clusters (see arrow in FIG. 5C). FIGS. 5A, 5B and 5C are different magnifications of the same colony. The shadow in FIG. 5A is the edge of the well in which the cells were growing.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The term “embryonic ectoderm” is used herein. “Embryonic ectoderm” and “epiblast” can be used interchangeable to refer to the same cell type.
  • A “pluripotential embryonic stem cell” as used herein means a cell which can give rise to many differentiated cell types in an embryo or adult, including the germ cells (sperm and eggs). Pluripotent embryonic stem cells are also capable of self-renewal. Thus, these cells not only populate the germ line and give rise to a plurality of terminally differentiated cells which comprise the adult specialized organs, but also are able to regenerate themselves. This cell type is also referred to as an “ES cell” herein.
  • A “fibroblast growth factor” (FGF) as used herein means any suitable FGF. There are presently seven known FGFs (Yamaguchi et al. (1992)). These FGFs include FGF-1 (acidic fibroblast growth factor), FGF-2 (basic fibroblast growth factor), FGF-3 (int-2), FGF-4 (hst/K-FGF), FGF-5, FGF-6, FGF-7 and FGF-8. Each of the suitable factors can be utilized directly in the methods taught herein to produce or maintain ES cells. Each of the FGF can be screened in the methods described herein to determine if the FGF is suitable to enhance the growth of or allow continued proliferation of ES cells or their progenitors. Various examples of FGF and methods of producing an FGF are well known; see, for example, U.S. Pat. Nos. 4,994,559; 4,956,455; 4,785,079; 4,444,760; 5,026,839; 5,136,025; 5,126,323; and 5,155,214.
  • “Steel factor” (SF) is used herein. SF is also called stem cell factor, mast cell growth factor and c-kit ligand in the art. SF is a transmembrane protein with a cytoplasmic domain and an extracellular domain. Soluble SF refers to a fragment cleaved from the extracellular domain at a specific proteolytic cleavage site. Membrane associated SF refers to both normal SF before it has been cleaved or the SF which has been altered so that proteolytic cleavage cannot take place. SF is well known in the art; see European Patent Publication No. 0 423 980 A1, corresponding to European Application No. 90310889.1.
  • “Leukemia Inhibitory Factor” (LIF) is also used herein. LIF is also known as DIA or differentiation inhibiting activity. LIF and uses of LIF are also well known; see for example U.S. Pat. Nos. 5,187,077 and 5,166,065.
  • It should be recognized that FGF, SF and LIF are all proteins and as such certain modifications can be made to the proteins which are silent and do not remove the activity of the proteins as described herein. Such modifications include additions, substitutions and deletions. Methods modifying proteins are well established in the art (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989).
  • This invention provides a non-mouse pluriopotential ES cell which can be maintained on feeder layers for at least 20 passages, and give rise to embryoid bodies and multiple differentiated cell phenotypes in monolayer culture. Only those non-mouse animals which can be induced to form ES cells by the described methods are within the scope of the invention. Given the methods described herein, an ES cell can be made for any animal. However, mammals are preferred since many beneficial uses of mammalian ES cells exist. Mammalian ES cells such as those from rats, rabbits, guinea pigs, goats, pigs, cows, and humans can be obtained. Alternatively, embryos from these animals can be screened for the ability to produce ES cells.
  • The ES cells of this invention can be maintained for at least 20 passages. However, the ES cells can be capable of indefinite maintenance. Typically, after about 10 passages the cells are frozen so that the starting population is not altered by minor chromosomal alterations.
  • Once the non-mouse ES cells are established, they can be genetically manipulated to produce a desired characteristic. For example, the ES cells can be mutated to render a gene non-functional, e.g. the gene associated with cystic fibrosis or an oncogene. Alternatively, functional genes can be inserted to allow for the production of that gene product in an animal, e.g. growth hormones or valuable proteins. Such methods are very well established in the art (Sedivy and Joyner (1992)).
  • The invention also provides a composition comprising:
  • (a) pluripotential ES cells; and
  • (b) an FGF, LIF, membrane associated SF, and soluble SF in amounts to enhance the growth of and allow the continued proliferation of the cell. Thus, this composition represents the composition after primordial germ cells, embryonic ectoderm or germ cells have become pluripotential ES cells. The pluripotential ES cells can continue to be maintained in this composition or alternatively they can be maintained on a feeder layer. Optimally, LIF can be added to the feeder layer.
  • Also provided is a composition comprising an FGF, LIF, membrane associated SF, and soluble SF in amounts to enhance the growth of, and allow the continued proliferation of primordial germ cells and the formation of pluripotent ES cells from the primordial germ cell. This composition need not include primordial germ cells but comprises the various growth factors in amounts that promote the growth, proliferation and formation of pluripotent ES cells. Thus, this composition can be sufficient for the establishment of pluripotent ES cells from the embryonic ectoderm cells or germ cells.
  • Also provided is a composition comprising: (a) mammalian primordial germ cells and/or germ cells and/or embryonic ectoderm cells; and (b) a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cells and the formation of pluripotent embryonic stem cells.
  • Typically, the compositions of the invention include a feeder layer. feeder layers can either be cells or cell lines cultured for the purpose of culturing pluripotent ES cells. Alternatively, feeder layers can be derived from or provided by the organ or tissue in which the primordial germ cells, embryonic ectoderm cells or germ cells are located, e.g., the gonad. Thus, if the somatic cells of the tissue or organ in which the desired cells are located are sufficient to provide the appropriate culture environment, a separate feeder layer is not required. Alternatively, the feeder cells could be substituted with extracellular matrix plus bound growth factors. Feeder layers which are representative of those which can be utilized are set forth in the Examples. Naturally, the membrane associated SF can be contained on the cells of such a feeder layer.
  • The compositions arise from the fact that FGF, LIF and SF are used either to enhance the growth and proliferation of primordial germ cells and/or embryonic ectoderm cells to become ES cells. Growth and proliferation enhancing amounts can vary depending on the species or strain of the cells, and type of purity of the factors. Generally, 0.5 to 500 ng/ml of each factor within the culture solution is adequate. In a more narrow range, the amount is between 10 to 20 ng/ml for bFGF and LIF and between 10 to 100 ng/ml for SF. Regardless of whether the actual amounts are known, the optimal concentration of each factor can be routinely determined by one skilled in the art. Such determination is performed by titrating the factors individually and in combination until optimal growth is obtained. Additionally, other factors can also be tested to determine their ability to enhance the effect of FGF, LIF and SF on ES cell proliferation. As described below, such other factors, or combinations of factors when used to enhance ES cell proliferation can be included within the above compositions. Also, compounds and fragments of FGF, LIF and SF which mimic the function of these factors can be used to enhance the growth and proliferation of the cells to become ES cells and are included within the scope of the invention.
  • The factors are essential to the formation of pluripotent ES cells. Thus, the amount of the factors utilized is determined by the end result of the pluripotent ES cells. However, the factors also serve to enhance the growth and allow the continued proliferation of the cells. Relatedly, the factors also appear to help the cells survive.
  • Alternatively, FGF, LIF, and SF can be used to maintain ES cells. The amounts of FGF, LIF and SF are necessary to maintain ES cells can be much less than that required to enhance growth or proliferation to become ES cells. However, the cells may be maintained on a feeder layer without the addition of growth factors. Optimally, LIF can be added to enhance maintenance.
  • In general, FGF or LIF from a species different from the source of the ES, primordial germ cell, germ cell or embryonic ectoderm cell can be utilized. However, all the factors utilized and especially the SF utilized are preferably from the same species as the utilized cell type. However, FGF, LIF or SF from various species can be routinely screened and selected for efficacy with a cell from a different species. Recombinant fragments of FGF, LIF or SF can also be screened for efficacy as well as organic compounds derived from, for example, chemical libraries.
  • The invention also provides a method of making a pluripotential ES cell comprising administering a growth enhancing amount of FGF, LIF, membrane associated SF, and soluble SF to primordial germ cells and/or embryonic ectoderm cells under cell growth conditions, thereby making a pluripotential ES cell. Thus, primordial germ cells and embryonic ectoderm cells can be cultured as a composition in the presence of these factors to produce pluripotent ES cells. As noted above, typically the composition includes a feeder layer.
  • The invention also provides a method of making a mammalian pluripotential embryonic stem cell comprising culturing a germ cell or a composition from postnatal mammalian testis in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor, thereby making a pluripotential embryonic stem cell from a germ cell. “Germ cells” as used herein means the cells which exist in neonatal or postnatal testis and are the progenitors of gametes. In the testis, theses germ cells represent a small population of stem cells capable of both self-renewal and differentiation into mature spermatogonia. Thus, “germ cells” are the postnatal equivalent to the prenatal primordial germ cells and can include primitive or immature spermatogonia such as type A spermatogonia or any undifferentiated early stage cell that can form a pluripotent embryonic stem cell.
  • These methods can be practiced utilizing any animal cell, especially mammal cells including mice, rats, rabbits, guinea pigs, goats, cows, pigs, humans, etc. The ES cell produced by this method is also contemplated.
  • Also provided is a method of screening cells which can be promoted to become an ES cell comprising contacting the cells with FGF, LIF, membrane associated SF, and soluble SF amounts to enhance the growth of and allow proliferation of the cells and determining which cells become ES cells. Utilizing this method, cells other than primordial germ cells, germ cells, and embryonic ectoderm cells can be selected as a source of ES cells.
  • Since the invention provides ES cells generated from virtually any animal, the invention provides a method is using the ES cells to contribute to chimeras in vivo in non-humans comprising injecting the cell into a blastocyst and growing the blastocyst in a foster mother. Alternatively, aggregating the cell with a non-human morula stage embryo and growing the embryo in a foster mother can be used to produce a chimera. Chimeric animals can subsequently be bred to obtain germ line transmission of ES cell traits. As discussed above, the ES cells can be manipulated to produce a desired effect in the chimeric animal. The methods of producing such chimeric animals are well established (Robertson (1987)).
  • Alternatively, the ES cells can be used to derive cells for therapy to treat an abnormal condition. For example, derivatives of human ES cells could be placed in the brain to treat a neurodegenerative disease. Relatedly, ES cells can be used to screen factors to determine which factors produce derivative (more differentiated cells. Many standard means to determine the presence of a more differentiated cell are well known in the art.
  • FGF, SF and LIF have been shown herein to be critical for making ES cells. However, as noted above for FGF, other members of the respective growth factor family could also be used to make ES cells. Thus, later discovered members of each family can merely be substituted to determined if the new factor enhances the growth and allows the continued proliferation of PGCs or embryonic ectoderm cells to form ES cells. For example, if a new member of the LIF family is discovered, the new LIF is merely combined with SF and FGF to determine if the new family member enhances the growth and allows the continued proliferation of PGCs or embryonic ectoderm cells. Thus, this invention provides the use of family members and a method of screening family members for activity.
  • Likewise, additional growth factors may be found useful in enhancing the growth and proliferation of PGCs or embryonic ectoderm cells from various animals. This invention provides combining FGF, SF and LIF with other growth factors to obtain or enhance the production of ES cells. Thus, a method of screening other growth factors for the ability to promote PGCs and embryonic ectoderm cells to form ES cells is also provided. In this regard, IL-11, IL-6, CNTF, NGF, IGFII flt3/flk-2 ligand and members of the Bone Morphogenetic Protein family are good screening candidates and can be used to promote ES cell formation.
  • EXAMPLES
  • All the cell types and other materials listed below can be obtained through available sources and/or through routine methods.
  • Materials and Methods
  • Feeder Cells
  • The SI/SI4 cell line, derived from a homozygous null SI/SI mouse embryo, and its derivative, SI4-m220, which stably expresses only membrane bound murine SF lacking exon 6 encoding the proteolytic cleavage site, were obtained from Dr. David Williams (Howard Hughes Medical Institute, Indiana University Medical School). Other cell lines which produce adequate SF can be substituted for SI/SI4, for example mouse or human embryo fibroblasts or cell lines or somatic cell lines from gonads or genital ridges. Combinations of feeder cells can also be utilized. They were maintained in DMEM with 10% calf serum and 50 ug/ml gentamicin. For making feeder layers they were irradiated (500 rads) and plated at a density of 2×105 per well of 24-well plates (Falcon) in the same medium, 24 hrs before use. Wells were pre-treated with 1% gelatin. STO cells stably transfected with human LIF and the bacterial neor gene (SLN) were obtained from Dr. Allan Bradley.
  • Primary Cultures of PGCs
  • Embryos were from ICR females mated with (C57BLxDBA)F1 males. Noon of the day of plug is 0.5 day post coitum (p.c.). The caudal region of 8.5 day p.c. embryos (between the last somite and the base of the allantois) was dissociated into single cells by incubation at 37° C. with 0.05% trypsin, 0.02% EDTA in CA++/MG++ free Dulbecco's phosphate-buffered saline (PBS) for about 10 mins with gentle pipetting. At this stage there are between about 149 and 379 PGCs in each embryo (Mintz and Russell, 1957). Cells from the equivalent of 0.5 embryo were seeded into a well containing feeder cells as above and 1 ml of DMEM, 2mM glutamine, 1 mM sodium pyruvate, 100 i.u./ml penicillin and 100 ug/ml streptomycin and 15% fetal bovine serum (PGC culture medium). Finely minced fragments of genital ridges from 1.5 and 12.5 day p.c. embryos were trypsinized as above and plated at a concentration of 0.1 embryo per well. Growth factors were added at the time of seeding, usually at the following concentrations, which were shown to be optimal for PGC proliferation; recombinant human LIF and bFGF (10-20 ng/ml) and soluble rat SF (60 ng/ml). The medium was changed every day.
  • Secondary Culture of PGC
  • Primary cultures were trypsinized and reseeded into wells containing SI4-m220 feeder layers in PGC culture medium. For further subculture, rounded colonies of densely packed ES-like cells were carefully picked up in a finely drawn pipette and trypsinized in a microdrop under mineral oil before seeding into wells containing feeder cells as above. After several subcultures in this way, cultures were passaged without picking individual colonies.
  • Alkaline Phosphatase (AP) Staining
  • This was carried out as described (Matsui et al. 1991). After staining, AP positive cells were counted using an inverted microscope.
  • SSEA-1 Staining
  • PGC cultures on SI4-m220 feeder cells on a chamber slide (Nunc) were washed twice with PBS containing 2% calf serum, 0.1% sodium azide and then incubated with mouse monoclonal antibody SSEA-1 (1:100 dilution) on ice for 30 min. After washing with PBS, cells were incubated for 30 mins with FITC-conjugated Fab′ fragment of goat anti mouse IgG (H+L) (Cappell, 1:5 dilution). After washing in PBS, cells were fixed in 4% paraformaldehyde before staining for AP.
  • Tumors in Nude Mice
  • Approximately 2×106 cells from three independent lines were injected subcutaneously into nude mice (three mice per line). After three weeks tumors were fixed in Bouin's fixative, processed for histology and sections stained with haematoxylin and cosin.
  • Chimera Formation
  • Ten to fifteen cells from two independent lines derived from 8.5 day p.c. embryos were injected into the blastocoel of 3.5 day p.c. blastocysts of either ICR or C57BL/6 mice. These were returned to the uteri of 2.5 day p.c. pseudopregnant foster mothers.
  • Culture of Murine PGCs in the Presence of Growth Factors
  • Initial experiments used SI/SI4 cells derived from a homozygous null SI/SI mutant mouse as a feeder layer for the culture of cells dissociated from the posterior of 8.5 days p.c. embryos, and AP staining as a marker for PGCs (FIG. 1A). As shown previously (Matsui et al., 1991), soluble SF and LIF act synergistically on PGCs. Addition of bFGF further enhances growth, and the cells continue to increase in number until day 5 culture, i.e. one day longer than usual. The effect of bFGF alone is small, and both SF and LIF are needed in addition to bFGF for maximal effect on PGC growth (FIG. 1A, B). A variety of other growth factors, including human activin, Bone Morphogenetic Protein-4, BNGF, and PDGF at 10 and 50 ng/ml had no effect in the presence of SF and LIF.
  • Membrane associated SF seems to play an important role in PGC proliferation since SId mouse mutants which make only soluble SF have a reduced number of PGCs in vivo, and membrane associated SF is more effective than soluble SF in supporting PGC growth and survival in culture (Dolci et al., 1991; Matsui et al., 1991). To test the effect of added factors in the presence of membrane associated SF, 8.5 day p.c. PGCs were cultured on SI4-m220 feeder cells which express only membrane associated SF (Matsui et al., 1991; Toksoz et al., 1991). Both LIF and bFGF separately enhance PGC growth on SI4 -m220 feeder cells, with added soluble rSF. However, when LIF and bFGF are added together, PGC growth is dramatically stimulated and the cells continue to proliferate through to day 6 in culture (FIG. 1C). The cells survive until day 8, at which time the feeder layer deteriorates, but they can be trypsinized and subcultured (see below).
  • Pregonadal PGCs are motile in vivo, and when cultured with LIF on a SI4 -m220 feeder layer they form burst colonies of cells with a flattened and polarized morphology, characteristic of motile cells (FIG. 2A). In contrast, PGCs cultured on a SI4-m220 feeder layer with soluble SF and bFGF or with bFGF and LIF (FIG. 2B, C), form discrete colonies of tightly packed cells. These colonies increase in size over day 6 in culture only when both bFGF and LIF are present (FIG. 2C).
  • To determine whether PGCs and their descendants continue to proliferate in culture, primary colonies of PGCs were typrsinized after 6 days in culture and replated on a fresh SI4 -m220 feeder layer with added growth factors. By day 6 in secondary culture, large colonies of densely packed AP positive cells resembling embryonic stem (ES) cells are present (FIG. 2D, E; FIG. 4, A), with an overall plating efficiency of about 5%. These colonies are also positive for the expression of the antigen SSEA-1, a characteristic of PGCs (Donovan et al., 1986) and undifferentiated embryonal carcinoma and ES cells (Solter and Knowles, 1978) (FIG. 2 G, H). Although the growth of primary cultures is strictly dependent on the presence of LIF and bFGF, secondary colonies can form in the absence of these factors (Table 1), indicating a reduced exogenous growth factor requirement for the descendants of PGCs after subculture. Most of the colonies show strong, uniform AP staining. However, some colonies contain only a small number of strongly stained cells, surrounded by cells which are weakly stained or negative (FIG. 2E). In many cases these negative cells are larger and have a more flattened morphology than the AP positive cells. For further subculture, individual colonies of cells with a distinctive, tightly packed, ES cell-like morphology were picked up in a micropipette, trypinized and replated on a fresh feeder layer with added factors. Such colonies can be subcultured at least ten times and continue to give rise to colonies of similar morphology. In later passages, these cultures were transferred to feeder layers of STO cells in medium without added factors normally used for blastocyst-derived ES cell culture (Robertson, 1987). Under these conditions they continue to proliferate in an undifferentiated state, for a total of at least 20 passages.
  • Two independent lines at passage 14 (1/14, 2/14) and one at passage 20 (3/20) were karyotyped. Most cells had a normal or near normal XY karyotype, but in two lines (2/14 and 3/20) there was a significant proportion of trisomic cells.
  • Long Term Culture of PGC-Derived Cells from Genital Ridges
  • Since transplantable teratocarcinomas can be induced experimentally by grafting genital ridges from 11.5 or 12.5 days p.c. male embryos of the 129 strain to an ectopic site, we tested the possibility that ES-like cells can be obtained from genital ridges in culture. Genital ridges were trysinized and the cells plated on an SI4 -m220 feeder layer with soluble SF, LIF and bFGF. The number of PGCs initially declines but increases after 3 days, and by 6 days colonies of densely packed, AP positive cells can be seen (FIG. 2F). If cells from male and female 12.5 days p.c. genital ridges are cultured separately, male PGCs increase in number and form colonies. In contrast, only a few female PGCs form colonies (FIG. 3). The differentiation capacity of genital ridge-derived colonies has not so far been tested.
  • Differentiation of PGC-Derived ES Cells in vitro and in Nude Mice
  • Four independent lines of undifferentiated cells derived from 8.5 day embryos and cultured onto STO feeder layers were trypsinized and pipetted gently to generate small clumps of cells which were then placed in bacteriological plastic dishes. After five to seven days most of the clumps differentiated into typical simple or cystic embryoid bodies (EBs), with a clear outer layer of extraembryonic endoderm cells (FIG. 4, B). When these EBs were returned to tissue culture plastic dishes they rapidly attached and over two weeks gave rise to a variety of cell types, including extraembryonic endoderm, spontaneously contracting muscle, nerve and endothelial and fibroblast-like cells.
  • Three of these four lines, at passages 9 and 15 on STO cells, were injected subcutaneously into nude mice. Each line gave rise to multiple, well-differentiated teratocarcinomas, containing a wide variety of tissues, including keratinized, secretory and ciliated epithelium, neuroepithelium and pigmented epithelium, cartilage, bone, and muscle, as well as nests of undifferentiated embryonic cells (FIG. 4, C-E).
  • PGC-Derived ES Cells can Contributed to Chimeras in vivo
  • To test whether the descendants of PGCs in culture are able to contributed to chimeras in vivo, 10-15 cells with an ES-like morphology from two independent early passage culture derived from 8.5 day embryos and cultured on either SI4m220 cells or STO cells were injected into host ICR or C57BL/6 blastocysts. From a total of 21 pups born, four were chimeric, as judged by coat color, but only two were extensive, with approximately 50 and 90% chimerism. The 50% coat color chimera, generated by injecting cells from the 4th passage on STO cells into an ICR blastocyst, died at 11 days after birth and showed stunted growth and skeletal abnormalities. The 90% coat color chimera, obtained by injecting cells from the 6th passage on STO cells into a C57BL/6 blastocyst, had no obvious abnormalities.
  • Germ Line Transmission
  • Materials and Methods
  • PGC Culture
  • Cultures are initiated as described above by dissecting C57BL/6 8.5 days p.c. embryos free of extraembryonic tissues. Fragments comprising the posterior third of the embryo (from the base of the allantois to the first somite) are then pooled, rinsed with Dulbecco's Ca++•Mg++ free phosphate buffered saline (PBS) and dissociated with 0.25% trypsin, 1 mM EDTA (GIBCO) and gentle pipetting. This single cell suspension is then plated in 0.1% gelatin coated 24 well dishes (Coming) with irradiated SI/SI4 m220 cells as feeder layers at a concentration of approximately 0.5 embryo equivalents per well. The cultures were grown in Dulbecco's modified Eagle's medium (DMEM) (Specialty Media, Lavallette, N.J.) supplemented with 0.01 mM non-essential amino acids (GIBCO), 2 mM glutamine (GIBCO), 50 ,ug/ml gentamycin (Sigma), 15% fetal bovine serum (selected batches, Hyclone) and 0.1 mM 2-mercaptoethanol (Sigma). For these primary culture, the medium is additionally supplemented with soluble recombinant rat SF at 60 ng/ml, bFGF at 20 ng/ml (GIBCO), and LIF at 20 ng/ml. After 6 days some of the cultures are stained for alkaline phosphatase (AP) as described above in order to assess the survival and proliferation of PGCs. After 10 days, parallel cultures are dissociated into single cells and plated onto mouse embryo fibroblast (mef) feeder layers with LIF (ESGRO, GIBCO 1000 U/ml). These cultures are monitored for the appearance of colonies of EG cells. Individual EG colonies are isolated with a micropipette and lines established. EG cultures are then maintained in the same manner as ES cell lines with irradiated mefs as feeder cells and LIF (Smith et al., 1988 and Williams et al., 1988).
  • Blastocyst Injection
  • Ten to twenty EG cells at passage numbers 6 to 10 were injected into 3.5 days p.c. blastocysts from BALB/c mice. Foster mothers were (C57BL/6×DBA)F1 females mated to vasectomized Swiss Webster males. Injected blastocysts were transferred to the uterus of 2.5 days p.c. foster mothers (Hogan et al., 1986) and chimeric pups were identified by their coat color. Chimeras were bred to either BALB/c or ICR mice and germ line transmission was judged on the day of birth by the presence of eye pigment.
  • Results
  • Two ES lines derived from 8.5 day C57BL/6 embryos produced chimeras which transmitted the C57BL/6 genome through mature sperm (germ line transmission). This was judged by mating male chimeras with albino females and observing the production of pigmented pups. With the ES cell line known as TGC8.510, nine chimeric males were obtained and four produced pigmented pups. With the ES cell line known as TGC8.519 six chimeric males were obtained and one of these produced pigmented pups.
  • Generation of ES Cells from other Mammals
  • ES cells from other mammals can be produced using the methods described above for murine. The mammalian cell of choice is simply substituted for murine and the murine methods are duplicated. The appropriate species specific growth factors (e.g. SF) can be substituted for murine growth factors as necessary. Any additional growth factors which can promote the formation of ES cells can be determined by adding the growth factors to FGF, LIF, and SF as described above and monitored for an effect on ES formation.
  • Method for the Isolation of Pluripotential Stem Cells from Human Primordial Germ Cells and Human Embryonic (Fetal) Gonads
  • The above methods for isolation of ES cells from murine embryos were repeated for isolation of ES cells from human embryos. Specifically, testes were dissected from a 10.5 week human embryo. Younger or older embryos represent alternative sources. The preferred age range is between 8.5 weeks and 22 weeks. Tissue was rinsed in buffered saline, and incubated in trypsin solution (0.25% trypsin, 1 mM EDTA in Ca++/MG++ fre HEPES buffered saline) for 10 minutes at 37° C. This tissue was dissociated by pipetting and the cells plated into wells of a 24 well tray containing irradiated feeder cells. In this experiment the feeder cells were SI/SI mouse fibroblasts transfected with human membrane associated Stem Cell Factor (SI4h220 cells from Dr. David Williams, HHMI, Indiana State University School of Medicine). An alternative feeder layer would consist of a mixture of mouse or human embryo fibroblasts and SI4h220 cells, to provide a more coherent layer for long term cell attachment. The culture medium consists of Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum supplemented with 10 ng/ml human bFGF, 60 ng/ml human Stem Cell Factor and 10 ng/ml human LIF. Alternatively, the amounts of bFGF can be increased (e.g. 20 ng/ml). Other alternative or additional supplements can be added at this time, for example IF-6, IL-ll, CNTF, NGF, IGFII, flt3/flk-2 ligand, and/or members of the Bone Morphogenetic Protein family. The cultures were maintained for 5 days, with daily addition of fresh growth factors. Longer culture could also be utilized, e.g. 5 to 20 days.
  • After 5 days, cultures were dissociated with trypsin solution as before and seeded into wells containing a feeder layer of irradiated mouse embryo fibroblasts. The medium was supplemented with growth factors daily as above. The addition of growth factors to the culture medium at this stage can be utilized, and a feeder layer of a mixture of mouse of human fibroblast and SI4h220 cells can be substituted.
  • After 10 days the cultures were fixed and stained for alkaline phosphatase activity. Colonies of cells expressing high levels of alkaline phosphatase and closely resembling primordial germ cells of the mouse embryo were detected in many wells (see FIG. 5). Closely packed clusters of cells were present in some colonies (arrow in FIG. 5). In cultures of mouse embryo germ cells these colonies give rise to lines of pluripotential embryonic stem cells. Therefore, the identified human cells can give rise to cell lines.
  • Method for the Isolation of Embryonic Stem Cell Lines Form Postnatal Mammalian Testis
  • Testes are dissected and the tunica removed. The testes are then incubated at 32° C. with mild shaking in buffered saline containing bovine serum albumin and collagenase (final concentration approximately 0.5 mg/ml). When the tissue has dissociated the tubules are allowed to settle out and then washed in saline several times. The collagenase treatment is repeated to remove all the cells surrounding the tubules (Leydig cells and connective tissue). The tubules are then washed and treated with hyaluronidase in buffered saline (final concentration approximately 0.5 mg/ml) at 32° C. until the tubules are free of adherent material. The tubules are washed and placed onto tissue culture dishes coated with Poly-L-lysine. The Sertoli cells attach strongly to the dish and spread out, while the germ cells remain in suspension. The germ cells are collected and plated onto a layer or irradiated feeder cells comprising membrane bound and soluble stem cell factor, LIF and basic FGF as described above.
  • Generation of Chimeras using Non-Murine ES Cells
  • Chimeras utilizing non-murine non-human ES cells can likewise be produced utilizing the methods for murine described above and simply substituting the appropriate non-murine blastocyst for the species of ES utilized.
  • Throughout this application various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.
  • The preceding examples are intended to illustrate but not limit the invention. While they are typical of those that might be used, other procedures known to those skilled in the art may be alternatively employed.
    TABLE 1
    Growth Factor Requirements for Secondary
    Cultures of PCG-Derived Cells
    SF + LIF +
    Days in SF→SF + LIF + bFGF→SF + SF + LIF +
    Culture LIF + bFGF LIF + bFGF bFGF→SF
    1 112 ± 16 cells 116 ± 20 cells 142 ± 18 cells
    3 0.9 ± 0.6 colonies 4.6 ± 1.1 colonies 5.6 ± 0.8 colonies
    5 0.5 ± 0.4 colonies 6.9 ± 1.2 colonies 6.6 ± 1.3 colonies

    PGCs from 8.5 dpc embryos were cultured for 6 days on SI4 -m220 cells
    # in the presence of either soluble rat SF alone or with soluble rat SF, LIF, and
    # bFGF. Cultures were trypsinized and seeded into wells
    # containing SI4 -m220 feeder cells with either soluble rat SF alone
    # or soluble rat SF, LIF, and bFGF. Cultures were fixed and AP-positive
    # cells (day 1) or colonies (days 2 and 5) counted. Numbers are
    # mean ± SEM from four experiments. Secondary cultures show a reduced
    # growth factor requirement compared with primary cultures.
  • REFERENCES
    • Bogler, O., Wren, D., Barnett, S. C., Land, H. and Noble, M. (1990). Cooperation between two growth factors promotes extended self-renewal and inhibits differentiations of oligodendrocyte-type-2 astrocyte (0-2A) progenitor cells. Proc. Natl. Acad. Sci. USA. 87, 6368-6372.
    • Dolci, S., Williams, D.E., Ernst, M. K., Resnick, J. L., Brannan, C. I., Lock, L. F., Lyman, S. D., Boswell, H. S. and Donovan, P. J. (1991). Requirement for mast cell growth factor for primordial germ cell survival in culture. Nature 352, 809-811.
    • Donovan, P. J., Stott, D., Cairns, L. A., Heasman, J. and Wylie, C. C. (1986). Migratory and postmigratory mouse primordial germ cells behave differently in culture. Cell 44, 831-838.
    • Ffrench-Constant, C., Hollingsworth, A., Heasman, J. and Wylie, C. C. (1991). Response to fibronectin of mouse primordial germ cells before, during and after migration. Development 113, 1365-1373.
    • Godin, I. and Wylie, C.C. (1991). TGFβ-1 inhibits proliferation and has a chemotropic effect on mouse primordial germ cells in culture. Development 113, 1451-1457.
    • Godin, I., Deed, R., Cooke, J., Zsebo, K., Dexter, M. and Wylie, C. C. (1991), Effects of the steel gene product on mouse primordial germ cells in culture. Nature 352, 807-809.
    • Ginsberg, M., Snow, M. H. L. and McLaren, A. (1990). Primordial germ cells in the mouse embryo during gastrulation. Development 110, 521-528.
    • Hogan, B. L. M., Costantini, F., and Lacy, E. (1986). Manipulating the Mouse Embryo: A Laboratory Manual. Cold Spring Harbor Publications, Cold Spring Harbor, N.Y.
    • Lawson, K. A. and Pederson, R. A. (1992). Clonal analysis of cell fate during gastrulation and early neurulation in the mouse in CIBA Foundation Symposium 165 Post Implantation Development in the Mouse John Wiley and Sons.
    • Mann, J. R., Gadi, I., Harbison, M. L., Abbondanzo, S. J. and Stewart, C. L. (1990). Androgenetic mouse embryonic stem cells are pluripotent and cause skeletal defects in chimeras: implications for genetic imprinting. Cell 62, 251-260.
    • Manova, K. and Bachvarova, R. F. (1991). Expression of c-kit encoded at the W locus of mice in developing embryonic germ cells and presumptive melanoblasts. Dev. Biol. 146, 312-324.
    • Matsui, Y., Toksok, D., Nishikawa, S., Nishikawa, S-I, Williams, D., Zsebo, K. and Hogan, B. L. M. (1991). Effect of steel factor and leukemia inhibitory factor on murine primordial germ cells in culture. Nature 353, 750-752.
    • Mintz, B. and Russell, E. S. (1957). Gene-induced embryological modifications of primordial germ cells in the mouse. J. Exp. Zoology 134, 207-230.
    • Noble, M., Murray, K., Stroobant, P., Waterfield, M. D., and Riddle, P. (1988). Platelet-derived growth factor promotes division and motilty and inhibits premature differentiation of the oligodendrocyte/type-2 astrocyte progenitor cell. Nature 333, 560-562.
    • Noguehi, T. and Stevens, L. C.(1992). Primordial germ cell proliferation in fetal testes in mouse strains with high and low incidences of congenital testicular teratomas J. Natl. Cancer Inst. 69, 907-913.
    • Raff, M. C., Lillien, L. E., Richardson, W. D., Burne, J. F. and Noble, M. D. (1988). Platelet-derived growth factor from astrocytes drives the clock that times oligodendrocyte development in culture. Nature 333, 562-565.
    • Robertson, E. J. (1987). Embryo-derived stem cell lines in teratocarcinomas and embryonic stem cells: a practical approach. Ed. E. J. Robertson, IRL Press, Oxford pp. 71-112.
    • Robertson, E. J. (1987). Teratocarcinomas and embryonic stem cells: a practical approach, Ed. E. J. Robertson, IRL Press, Oxford.
    • Sedivy, J. M., and A. Joyner (1992). Gene Targeting, W. H. Freeman and Co.
    • Solter, D., Adams, N., Damjanov, I. and Koprowski, H. (1975). Control of teratocarcinogenesis in teratomas and differentiation, Eds. M. I. Sherman and D. Solter, Academic Press.
    • Solter, D. and Knowles, B. B. (1978). Monoclonal antibody defining a stage-specific mouse embryonic antigen (SSEA-1) Proc. Natl. Acad. Sci. USA 75, 5565-5569.
    • Soriano, P. and Jaenisch, R. (1986). Retroviruses as probes for mammalian development: allocation of cells to the somatic and germ cell lineages. Cell 46, 19-29.
    • Stevens, L. C. (1983). The origin and development of testicular, ovarian, and embryo-derived teratomas. Cold Spring Harbor Conferences on Cell Proliferation Vol. 10 Teratocarcinoma Stem Cells. Eds. Silver, L. M., Martin, G. R. and S. Strickland. 10, 23-36.
    • Stevens, L. C. and Makensen, J. A. (1961). Genetic and environmental influences on teratogenesis in mice. J. Natl. Cancer Inst. 27, 443-453.
    • Surani, M. A., Kothary, R., Allen, N. D., Singh, P. B., Fundele, R., Ferguson-Smith, A. C. and Barton, S. C. (1990). Genome imprinting and development in the mouse development. Suppl., 89-98.
    • Tam, P. P. L. and Snow, M. H. L. (1981). Proliferation and migration of primordial germ cells during compensatory growth in mouse embryos. J. Embryol. Exp. Morph. 64, 133-147.
    • Toksoz, D., Zsebo, K. M., Smith, K. A., Hu, S., Brankow, D., Suggs, S. V., Martin, F. H. and Williams, D. A. (1992). Support of human hematopoiesis in long-term bone marrow cultures by murine stromal cells selectively expressing the membrane-bound and secreted forms of the human homolog of the steel product, stem cell factor. Proc. Natl. Acad. Sci. USA, in press.
    • Yamaguchi, T. P., Conlon, R. A., and J. Rossant (1992). Expression of the fibroblast growth factor receptor FGFR-l/flg during gastrulation and segmentation in the mouse embryo. Development 152:75-88.

Claims (36)

1. An isolated non-mouse mammalian pluripotential embryonic stem cell which can:
(a) be maintained on feeder layers for at least 20 passages; and
(b) give rise to embryoid bodies and multiple differentiated cell phenotypes in monolayer culture.
2. The embryonic stem cell of claim 1, having a mutation which renders a gene non-functional.
3. The embryonic stem cell of claim 1, having an insertion of a functional gene.
4. An isolated human pluripotential embryonic stem cell which can:
(a) be maintained on feeder layers for at least 20 passages; and
(b) give rise to embryoid bodies and multiple differentiated cell phenotypes in monolayer culture.
5. A composition comprising:
(a) mammalian pluripotential embryonic stem cells; and
(b) a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cell.
6. The method of claim 5, wherein the fibroblast growth factor is basic fibroblast growth factor.
7. A composition comprising:
(a) human pluripotential embryonic stem cells; and
(b) a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cell.
8. A composition comprising:
(a) mammalian primordial germ cells; and
(b) a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cells and the formation of pluripotent embryonic stem cells from the primordial germ cell.
9. The composition of claim 8, wherein the fibroblast growth factor is basic fibroblast growth factor.
10. A composition comprising:
(a) embryonic ectoderm cells; and
(b) fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cells and the formation of pluripotent embryonic stem cells from the embryonic ectoderm cells.
11. A composition comprising:
(a) germ cells; and
(b) fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of the cells and the formation of pluripotent embryonic stem cells from the germ cells.
12. The composition of claim 11, wherein the fibroblast growth factor is basic fibroblast growth factor.
13. A composition comprising a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor in amounts to enhance the growth and allow the continued proliferation of primordial germ cells and the formation of pluripotent embryonic stem cells from he primordial germ cells.
14. The composition of claim 13, wherein the fibroblast growth factor is basic fibroblast growth factor.
15. A method of making a mammalian pluripotential embryonic stem cell comprising culturing a primordial germ cell in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor, thereby making a pluripotential embryonic stem cell from a primordial germ cell.
16. The method of claim 15 wherein the primordial germ cell is derived from a human.
17. The method of claim 15, wherein the primordial germ cell is derived from a mouse.
18. A pluripotential embryonic stem cell produced by the method of claim 15.
19. A human pluripotential embryonic stem cell produced by the method of claim 16.
20. A method of making a mammalian pluripotential embryonic stem cell comprising culturing an embryonic ectoderm cell in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor, thereby making a pluripotential embryonic stem cell from an embryonic ectoderm cell.
21. The method of claim 20, wherein the embryonic ectoderm cell is derived from a human.
22. The method of claim 20, wherein the embryonic ectoderm cell is derived from a mouse.
23. A pluripotential embryonic stem cell produced by the method of claim 20.
24. A human pluripotential embryonic stem cell produced by the method of claim 21.
25. A method of making a mammalian pluripotential embryonic stem cell comprising culturing a germ cell in a composition comprising a growth enhancing amount of basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor, thereby making a pluripotential embryonic stem cell from a germ cell.
26. The method of claim 25, wherein the germ cell is derived from a human.
27. A pluripotential embryonic stem cell produced by the method of claim 25.
28. A pluripotential embryonic stem cell produced by the method of claim 26.
29. A method of screening for a cell which can be promoted to become a pluripotential embryonic stem cell comprising culturing the cell in a composition comprising basic fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, and soluble steel factor in amounts to enhance the growth and allow the formation of pluripotential embryonic stem cells, and determining which cells become embryonic stem cells.
30. A method of using a non-human pluripotential embryonic stem cell of claim 1 to contribute to chimeras in vivo comprising injecting the cell into a blastocyst and growing the blastocyst in a foster mother.
31. A method of using a non-human embryonic stem cell of claim 1 to contribute to chimeras in vivo, comprising aggregating the cell with a morula stage embryo and growing the embryo in a foster mother.
32. A method of screen factors for the ability to promote the formation of pluripotential embryonic stem cells, comprising culturing primordial germ cells or embryonic ectoderm cells in a feeder layer comprising a fibroblast growth factor, leukemia inhibitory factor, membrane associated steel factor, soluble steel factor and the growth factor to be screened, and determining the formation of pluripotential embryonic stem cells.
33. A method of obtaining a cell for therapy comprising deriving a cell from the pluripotential embryonic stem cell of claim 1 and determine whether the derivative cell can be utilized for therapy.
34. A method of obtaining a cell for therapy comprising deriving a cell from the pluripotential embryonic stem cell of claim 4 and determining whether the derivative cell can be utilized for therapy.
35. A method of screening a factor for the ability to derive a cell from the pluripotential embryonic stem cell comprising adding the factor to the pluripotential embryonic stem cell of claim 1 and determining whether a derivative cell is formed.
36. A method of screening a factor for the ability to derive a cell from the pluripotential embryonic stem cell comprising adding the factor to the pluripotential embryonic stem cell of claim 4 and determining whether a derivative cell is formed.
US11/550,713 1992-10-08 2006-10-18 Pluripotential embryonic stem cells and methods of making same Abandoned US20070178590A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/550,713 US20070178590A1 (en) 1992-10-08 2006-10-18 Pluripotential embryonic stem cells and methods of making same

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US07/958,562 US5453357A (en) 1992-10-08 1992-10-08 Pluripotential embryonic stem cells and methods of making same
US08/217,921 US5690926A (en) 1992-10-08 1994-03-25 Pluripotential embryonic cells and methods of making same
US08/813,829 US7153684B1 (en) 1992-10-08 1997-03-06 Pluripotential embryonic stem cells and methods of making same
US10/327,400 US20040071672A1 (en) 1992-10-08 2002-12-20 Pluripotential embryonic stem cells and methods of making same
US11/550,713 US20070178590A1 (en) 1992-10-08 2006-10-18 Pluripotential embryonic stem cells and methods of making same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/327,400 Continuation US20040071672A1 (en) 1992-10-08 2002-12-20 Pluripotential embryonic stem cells and methods of making same

Publications (1)

Publication Number Publication Date
US20070178590A1 true US20070178590A1 (en) 2007-08-02

Family

ID=32074262

Family Applications (3)

Application Number Title Priority Date Filing Date
US08/813,829 Expired - Fee Related US7153684B1 (en) 1992-10-08 1997-03-06 Pluripotential embryonic stem cells and methods of making same
US10/327,400 Abandoned US20040071672A1 (en) 1992-10-08 2002-12-20 Pluripotential embryonic stem cells and methods of making same
US11/550,713 Abandoned US20070178590A1 (en) 1992-10-08 2006-10-18 Pluripotential embryonic stem cells and methods of making same

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/813,829 Expired - Fee Related US7153684B1 (en) 1992-10-08 1997-03-06 Pluripotential embryonic stem cells and methods of making same
US10/327,400 Abandoned US20040071672A1 (en) 1992-10-08 2002-12-20 Pluripotential embryonic stem cells and methods of making same

Country Status (1)

Country Link
US (3) US7153684B1 (en)

Families Citing this family (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9807935D0 (en) * 1998-04-14 1998-06-10 Univ Edinburgh Lineage specific cells and progenitor cells
US20100173410A1 (en) * 2000-03-09 2010-07-08 Wicell Research Institute, Inc. Cultivation of Primate Embryonic Stem Cells
DK2722387T3 (en) * 2003-12-23 2020-01-20 Viacyte Inc Definitely endoderm
US7985585B2 (en) 2004-07-09 2011-07-26 Viacyte, Inc. Preprimitive streak and mesendoderm cells
US7541185B2 (en) * 2003-12-23 2009-06-02 Cythera, Inc. Methods for identifying factors for differentiating definitive endoderm
US20050266554A1 (en) * 2004-04-27 2005-12-01 D Amour Kevin A PDX1 expressing endoderm
WO2007059007A2 (en) * 2005-11-14 2007-05-24 Cythera, Inc. Markers of definitive endoderm
US8647873B2 (en) 2004-04-27 2014-02-11 Viacyte, Inc. PDX1 expressing endoderm
US7625753B2 (en) * 2003-12-23 2009-12-01 Cythera, Inc. Expansion of definitive endoderm cells
CN103146649B (en) * 2004-06-09 2018-05-22 爱丁堡大学管理处 Neural stem cell
AU2005272681B2 (en) * 2004-08-13 2009-11-19 University Of Georgia Research Foundation, Inc. Compositions and methods for self-renewal and differentiation in human embryonic stem cells
GB2441488C (en) * 2005-06-22 2011-10-05 Geron Corp Suspension culture of human embryonic stem cells
ES2687233T3 (en) 2005-10-27 2018-10-24 Viacyte, Inc. Endoderm of the dorsal and ventral proximal intestine expressing PDX-1
SG170068A1 (en) 2006-03-02 2011-04-29 Cythera Inc Endocrine precursor cells, pancreatic hormone-expressing cells and methods of production
US11254916B2 (en) 2006-03-02 2022-02-22 Viacyte, Inc. Methods of making and using PDX1-positive pancreatic endoderm cells
US7695965B2 (en) 2006-03-02 2010-04-13 Cythera, Inc. Methods of producing pancreatic hormones
CN102660495A (en) 2006-04-14 2012-09-12 先进细胞技术公司 Hemangio-colony forming cells
WO2007127454A2 (en) * 2006-04-28 2007-11-08 Cythera, Inc. Hepatocyte lineage cells
WO2007146105A2 (en) * 2006-06-05 2007-12-21 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
US20100323442A1 (en) * 2006-10-17 2010-12-23 Emmanuel Edward Baetge Modulation of the phosphatidylinositol-3-kinase pathway in the differentiation of human embryonic stem cells
US7695963B2 (en) 2007-09-24 2010-04-13 Cythera, Inc. Methods for increasing definitive endoderm production
US20110111492A1 (en) 2008-01-18 2011-05-12 Regents Of The University Of Minnesota Stem Cell Aggregates and Methods for Making and Using
EP2356227B1 (en) 2008-11-14 2018-03-28 Viacyte, Inc. Encapsulation of pancreatic cells derived from human pluripotent stem cells
WO2010075500A1 (en) 2008-12-23 2010-07-01 Stemcells California, Inc Target populations of oligodendrocyte precursor cells and methods of making and using same
CN102625829B (en) 2009-07-21 2015-05-06 Abt控股公司 Use of stem cells to reduce leukocyte extravasation
ES2690199T3 (en) 2009-07-21 2018-11-19 Abt Holding Company Use of stem cells to reduce leukocyte extravasation
KR101900507B1 (en) 2010-02-25 2018-09-19 에이비티 홀딩 컴퍼니 Modulation of angiogenesis
WO2011106521A1 (en) 2010-02-25 2011-09-01 Abt Holding Company Modulation of macrophage activation
CA2798895A1 (en) 2010-05-12 2011-11-17 Abt Holding Company Modulation of splenocytes in cell therapy
US9090878B2 (en) 2010-06-17 2015-07-28 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
US8609406B2 (en) 2010-08-24 2013-12-17 Regents Of The University Of Minnesota Non-static suspension culture of cell aggregates
US9677042B2 (en) 2010-10-08 2017-06-13 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
WO2012103012A1 (en) 2011-01-24 2012-08-02 The Board Of Trustees Of The Leland Stanford Junior University Methods for generating inner ear cells in vitro
SG10201914092XA (en) 2011-06-06 2020-03-30 ReGenesys BVBA Expansion of stem cells in hollow fiber bioreactors
GB201119335D0 (en) 2011-11-09 2011-12-21 Univ Leuven Kath Hepatitis virus infectable stem cells
HUE043861T2 (en) 2012-05-25 2019-09-30 Univ California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
CA2899507A1 (en) 2013-03-14 2014-09-25 The Regents Of The University Of California In vitro production of medial ganglionic eminence precursor cells
HUE052516T2 (en) 2013-04-12 2021-05-28 Houston Methodist Hospital Improving organs for transplantation
ES2753323T3 (en) 2013-04-30 2020-04-08 Univ Leuven Kath Cell therapy for myelodysplastic syndromes
US9932607B2 (en) 2013-11-15 2018-04-03 The Board Of Trustees Of The Leland Stanford Junior University Site-specific integration of transgenes into human cells
WO2015073918A1 (en) 2013-11-16 2015-05-21 Terumo Bct, Inc. Expanding cells in a bioreactor
WO2015071474A2 (en) 2013-11-18 2015-05-21 Crispr Therapeutics Ag Crispr-cas system materials and methods
WO2015089277A1 (en) 2013-12-12 2015-06-18 The Regents Of The University Of California Methods and compositions for modifying a single stranded target nucleic acid
US9733236B2 (en) 2014-04-10 2017-08-15 James L. Sherley Methods for determining the effect of an agent on tissue stem cells
EP3188763B1 (en) 2014-09-02 2020-05-13 The Regents of The University of California Methods and compositions for rna-directed target dna modification
WO2017053312A1 (en) 2015-09-21 2017-03-30 The Regents Of The University Of California Compositions and methods for target nucleic acid modification
WO2017064546A1 (en) 2015-09-24 2017-04-20 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
US20200325449A1 (en) 2016-06-02 2020-10-15 The Cleveland Clinic Foundation Complement inhibition for improving cell viability
US10828330B2 (en) 2017-02-22 2020-11-10 IO Bioscience, Inc. Nucleic acid constructs comprising gene editing multi-sites and uses thereof
MA50578A (en) 2017-11-09 2021-09-15 Vertex Pharma CRISPR / CAS SYSTEMS FOR THE TREATMENT OF DMD
CA3084825A1 (en) 2017-12-14 2019-06-20 Crispr Therapeutics Ag Novel rna-programmable endonuclease systems and their use in genome editing and other applications
EP3768834A1 (en) 2018-03-19 2021-01-27 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and uses thereof
EP3572512A1 (en) 2018-05-24 2019-11-27 B.R.A.I.N. Ag A method for engineering a protein
CA3108275A1 (en) 2018-08-10 2020-02-13 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
GB2595606B (en) 2019-03-07 2022-09-21 Univ California CRISPR-Cas effector polypeptides and methods of use thereof
WO2020209959A1 (en) 2019-03-08 2020-10-15 Crispr Therapeutics Ag Nucleobase-editing fusion protein systems, compositions, and uses thereof
AU2020239225A1 (en) 2019-03-12 2021-09-30 Bayer Healthcare Llc Novel high fidelity RNA-programmable endonuclease systems and uses thereof
US20210047649A1 (en) 2019-05-08 2021-02-18 Vertex Pharmaceuticals Incorporated Crispr/cas all-in-two vector systems for treatment of dmd
EP3990624A1 (en) 2019-06-25 2022-05-04 Vertex Pharmaceuticals Incorporated Enhanced differentiation of beta cells
WO2021030424A1 (en) 2019-08-13 2021-02-18 Semma Therapeutics, Inc. Pancreatic differentiation
WO2021250058A2 (en) 2020-06-12 2021-12-16 Bayer Aktiengesellschaft CRISPR-Cas12a DIRECTED RANDOM MUTAGENESIS AGENTS AND METHODS
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
CA3222950A1 (en) 2021-06-11 2022-12-15 Bayer Aktiengesellschaft Type v rna programmable endonuclease systems
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023070019A1 (en) 2021-10-21 2023-04-27 Vertex Pharmaceuticals Incorporated Hypoimmune cells
WO2023077140A2 (en) 2021-11-01 2023-05-04 Vertex Pharmaceuticals Incorporated Stem cell derived pancreatic islet differentiation
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023167986A1 (en) 2022-03-02 2023-09-07 Lineage Cell Therapeutics, Inc. Methods and compositions for treating hearing loss
WO2023211857A1 (en) 2022-04-25 2023-11-02 Lineage Cell Therapeutics, Inc. Methods and compositions for treating vision loss
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5166065A (en) * 1988-08-04 1992-11-24 Amrad Corporation Limited In vitro propagation of embryonic stem cells
US5334702A (en) * 1991-03-25 1994-08-02 University Of Illinois Compositions which are immunologically crossreactive with antibodies and preparative methods therefor
US5340740A (en) * 1992-05-15 1994-08-23 North Carolina State University Method of producing an avian embryonic stem cell culture and the avian embryonic stem cell culture produced by the process
US5453357A (en) * 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5523226A (en) * 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US5605803A (en) * 1989-03-03 1997-02-25 University Of Virginia Alumni Patents Foundation Human sperm diagnostic
US5639618A (en) * 1994-05-13 1997-06-17 Plurion, Inc. Method of isolating a lineage specific stem cell in vitro
US5690926A (en) * 1992-10-08 1997-11-25 Vanderbilt University Pluripotential embryonic cells and methods of making same
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6090622A (en) * 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
US6245566B1 (en) * 1997-03-31 2001-06-12 The Johns Hopkins University School Of Medicine Human embryonic germ cell line and methods of use
US6521671B1 (en) * 1999-03-05 2003-02-18 Idemitsu Kosan Co., Ltd. Micelle dispersion and method for preparation thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU650616B2 (en) 1988-09-21 1994-06-30 Animal Biotechnology Cambridge Limited Derivation of pluripotential embryonic cell lines from domestic animals
WO1993017032A1 (en) 1992-02-24 1993-09-02 The Trustees Of The University Of Pennsylvania Techniques and intermediates for preparing non-peptide peptidomimetics
JP2000502910A (en) 1996-01-09 2000-03-14 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Bovine embryonic stem-like cells for producing transgenic ungulates, and methods of making and using the cells

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5166065A (en) * 1988-08-04 1992-11-24 Amrad Corporation Limited In vitro propagation of embryonic stem cells
US5605803A (en) * 1989-03-03 1997-02-25 University Of Virginia Alumni Patents Foundation Human sperm diagnostic
US5334702A (en) * 1991-03-25 1994-08-02 University Of Illinois Compositions which are immunologically crossreactive with antibodies and preparative methods therefor
US5340740A (en) * 1992-05-15 1994-08-23 North Carolina State University Method of producing an avian embryonic stem cell culture and the avian embryonic stem cell culture produced by the process
US5690926A (en) * 1992-10-08 1997-11-25 Vanderbilt University Pluripotential embryonic cells and methods of making same
US5453357A (en) * 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5670372A (en) * 1992-10-08 1997-09-23 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5523226A (en) * 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US5639618A (en) * 1994-05-13 1997-06-17 Plurion, Inc. Method of isolating a lineage specific stem cell in vitro
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6200806B1 (en) * 1995-01-20 2001-03-13 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6090622A (en) * 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
US6245566B1 (en) * 1997-03-31 2001-06-12 The Johns Hopkins University School Of Medicine Human embryonic germ cell line and methods of use
US6331406B1 (en) * 1997-03-31 2001-12-18 The John Hopkins University School Of Medicine Human enbryonic germ cell and methods of use
US6521671B1 (en) * 1999-03-05 2003-02-18 Idemitsu Kosan Co., Ltd. Micelle dispersion and method for preparation thereof

Also Published As

Publication number Publication date
US7153684B1 (en) 2006-12-26
US20040071672A1 (en) 2004-04-15

Similar Documents

Publication Publication Date Title
US5690926A (en) Pluripotential embryonic cells and methods of making same
US7153684B1 (en) Pluripotential embryonic stem cells and methods of making same
US5453357A (en) Pluripotential embryonic stem cells and methods of making same
Matsui et al. Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture
Mitalipova et al. Pluripotency of bovine embryonic cell line derived from precompacting embryos
Prelle et al. Pluripotent stem cells–model of embryonic development, tool for gene targeting, and basis of cell therapy
JP5456467B2 (en) Pluripotent cells from rats and other species
Labosky et al. Embryonic germ cell lines and their derivation from mouse primordial germ cells
Donovan The germ cell-the mother of all stem cells
US20030113910A1 (en) Pluripotent stem cells derived without the use of embryos or fetal tissue
US20070192881A1 (en) Culture conditions and growth factors affecting fate determination, self-renewal and expansion of rat spermatogonial stem cells
JP4314372B2 (en) Method for producing testicular cell-derived pluripotent stem cells
RU2215029C2 (en) Method for obtaining established embryonic germ cell line in poultry, line of chicken embryonic germ cells, method for obtaining somatic or sexual chimeras, method for transfection of foreign gene in embryonic germ cells
Dattena et al. Isolation, culture, and characterization of embryonic cell lines from vitrified sheep blastocysts
Petitte et al. The origin of the avian germ line and transgenesis in birds
US20110044954A1 (en) Methods of producing germ-like cells and related therapies
JP2007501625A (en) Avian spermatogonial stem cell culture method and avian spermatogonial stem cell obtained thereby
Donovan et al. Towards the use of primordial germ cells for germline modification
Stevens Teratocarcinogenesis and spontaneous parthenogenesis in mice
US20070053890A1 (en) Canine embryonic stem cells
JP2002176973A (en) Mammalian embryonic stem cell and method for establishing the same and subculture method for the same
Kyuwa et al. Characterization of embryonic stem-like cell lines derived from embryoid bodies
Shamblott et al. Pluripotent stem cells
Kawase et al. Effects of feeder cells on the establishment of ES cells from mice and rats: The difference between allogenetic and xenogenetic feeders
Odorico et al. Human and murine embryonic stem cell lines: windows to early mammalian development

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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