US20010024825A1 - Primate embryonic stem cells - Google Patents

Primate embryonic stem cells Download PDF

Info

Publication number
US20010024825A1
US20010024825A1 US09/761,289 US76128901A US2001024825A1 US 20010024825 A1 US20010024825 A1 US 20010024825A1 US 76128901 A US76128901 A US 76128901A US 2001024825 A1 US2001024825 A1 US 2001024825A1
Authority
US
United States
Prior art keywords
cells
human
primate
cell
ssea
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
US09/761,289
Inventor
James Thomson
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.)
Wisconsin Alumni Research Foundation
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23484566&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20010024825(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US09/761,289 priority Critical patent/US20010024825A1/en
Application filed by Individual filed Critical Individual
Publication of US20010024825A1 publication Critical patent/US20010024825A1/en
Priority to US09/982,637 priority patent/US7029913B2/en
Priority to US10/430,496 priority patent/US20060040383A1/en
Priority to US11/033,335 priority patent/US20050158854A1/en
Priority to US11/036,245 priority patent/US7582479B2/en
Priority to US12/047,135 priority patent/US7781216B2/en
Priority to US12/822,004 priority patent/US8273569B2/en
Priority to US13/595,587 priority patent/US20120328582A1/en
Assigned to WISCONSIN ALUMNI RESEARCH FOUNDATION reassignment WISCONSIN ALUMNI RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THOMSON, JAMES
Priority to US14/281,341 priority patent/US20150056698A1/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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • 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
    • C12N5/0605Cells from extra-embryonic tissues, e.g. placenta, amnion, yolk sac, Wharton's jelly
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • 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
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells

Definitions

  • the field of the present invention is stem cell cultures.
  • the field of the present invention is primate embryonic stem cell cultures.
  • stem cells are undifferentiated cells which can give rise to a succession of mature functional cells.
  • a hematopoietic stem cell may give rise to any of the different types of terminally differentiated blood cells.
  • Embryonic stem (ES) cells are derived from the embryo and are pluripotent, thus possessing the capability of developing into any organ or tissue type or, at least potentially, into a complete embryo.
  • Mouse ES cells are undifferentiated, pluripotent cells derived in vitro from preimplantation embryos (Evans, et al. Nature 292:154-159, 1981; Martin, Proc. Natl. Acad. Sci. USA 78:7634-7638, 1981) or from fetal germ cells (Matsui, et al., Cell 70:841-847, 1992).
  • Mouse ES cells maintain an undifferentiated state through serial passages when cultured in the presence of fibroblast feeder layers in the presence of Leukemia Inhibitory Factor (LIF) (Williams, et al., Nature 336:684-687, 1988). If LIF is removed, mouse ES cells differentiate.
  • LIF Leukemia Inhibitory Factor
  • Mouse ES cells cultured in non-attaching conditions aggregate and differentiate into simple embryoid bodies, with an outer layer of endoderm and an inner core of primitive ectoderm. If these embryoid bodies are then allowed to attach onto a tissue culture surface, disorganized differentiation occurs of various cell types, including nerves, blood cells, muscle, and cartilage (Martin, 1981, supra; Doetschman, et al., J. Embryol. Exp. Morph. 87:27-45, 1985).
  • Mouse ES cells injected into syngeneic mice form teratocarcinomas that exhibit disorganized differentiation, often with representatives of all three embryonic germ layers. Mouse ES cells combined into chimeras with normal preimplantation embryos and returned to the uterus participate in normal development (Richard, et al., Cytogenet. Cell Genet. 65:169-171, 1994).
  • mouse ES cells to contribute to functional germ cells in chimeras provides a method for introducing site-specific mutations into mouse lines.
  • homologous recombination can be used to derive ES cell lines with planned alterations of specific genes. These genetically altered cells can be used to form chimeras with normal embryos and chimeric animals are recovered. If the ES cells contribute to the germ line in the chimeric animal, then in the next generation a mouse line for the planned mutation is established.
  • mouse ES cells have the potential to differentiate into any cell type in the body
  • mouse ES cells allow the in vitro study of the mechanisms controlling the differentiation of specific cells or tissues.
  • the study of mouse ES cells provides clues to understanding the differentiation of general mammalian tissues, dramatic differences in primate and mouse development of specific lineages limits the usefulness of mouse ES cells as a model of human development.
  • Mouse and primate embryos differ meaningfully in the timing of expression of the embryonic genome, in the formation of an egg cylinder versus an embryonic disc (Kaufman, The Atlas of Mouse Development , London: Academic Press, 1992), in the proposed derivation of some early lineages (O'Rahilly & Muller, Developmental Stages in Human Embryos , Washington: Carnegie Institution of Washington, 1987), and in the structure and function in the extraembryonic membranes and placenta (Mossman, Vertebrate Fetal Membranes , New Brunswick: Rutgers, 1987). Other tissues differ in growth factor requirements for development (e.g.
  • primate ES cells lines will provide a faithful model for understanding the differentiation of primate tissues in general and human tissues in particular.
  • the placenta provides just one example of how primate ES cells will provide an accurate model of human development that cannot be provided by ES cells from other species.
  • the placenta and extraembryonic membranes differ dramatically between mice and humans. Structurally, the mouse placenta is classified as labyrinthine, whereas the human and the rhesus monkey placenta are classified as villous.
  • Chorionic gonadotropin, expressed by the trophoblast is an essential molecule involved in maternal recognition of pregnancy in all primates, including humans (Hearn, J Reprod Fertil 76:809-819, 1986; Hearn et al., J Reprod Fert 92:497-509, 1991).
  • Trophoblast secretion of chorionic gonadotropin in primates maintains the corpus luteum of pregnancy and, thus, progesterone secretion. Without progesterone, pregnancy fails. Yet mouse trophoblast produces no chorionic gonadotropin, and mice use entirely different mechanisms for pregnancy maintenance (Hearn et al., “Normal and abnormal embryo-fetal development in mammals,” In: Lamming E, ed. Marshall's Physiology of Reproduction. 4th ed. Edinburgh, N.Y.: Churchill Livingstone, 535-676, 1994).
  • An immortal, euploid, primate ES cell line with the developmental potential to form trophoblast in vitro, will allow the study of the ontogeny and function of genes such as chorionic gonadotropin which are critically important in human pregnancy. Indeed, the differentiation of any tissue for which there are significant differences between mice and primates will be more accurately reflected in vitro by primate ES cells than by mouse ES cells.
  • the major in vitro models for studying trophoblast function include human choriocarcinoma cells, which are malignant cells that may not faithfully reflect normal trophectoderm; short-term primary cultures of human and non-human primate cytotrophoblast, which in present culture conditions quickly form non-dividing syncytial trophoblast; and in vitro culture of preimplantation non-human primate embryos (Hearn, et al., J. Endocrinol. 119:249-255, 1988; Coutifaris, et al., Ann. NY Acad. Sci. 191-201, 1994).
  • An immortal, euploid, non-human primate embryonic stem (ES) cell line with the developmental potential to form trophectoderm offers significant advantages over present in vitro models of human trophectoderm development and function, as trophoblast-specific genes such as chorionic gonadotropin could be stably altered in the ES cells and then studied during differentiation to trophectoderm.
  • ES embryonic stem
  • EC human embryonic carcinoma
  • EC cells can be induced to differentiate in culture, and the differentiation is characterized by the loss of specific cell surface markers (SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81) and the appearance of new markers (Andrews, et al., 1987, supra).
  • SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 specific cell surface markers
  • Human EC cells will form teratocarcinomas with derivatives of multiple embryonic lineages in tumors in nude mice.
  • the range of differentiation of these human EC cells is limited compared to the range of differentiation obtained with mouse ES cells, and all EC cell lines derived to date are aneuploid (Andrews, et al., 1987, supra).
  • teratocarcinomas are tumors derived from germ cells, and although germ cells (like ES cells) are theoretically totipotent (i.e. capable of forming all cell types in the body), the more limited developmental potential and the abnormal karyotypes of EC cells are thought to result from selective pressures in the teratocarcinoma tumor environment (Roimpuls & Papaioannou, Cell Differ 15:155-161, 1984).
  • ES cells are thought to retain greater developmental potential because they are derived from normal embryonic cells in vitro, without the selective pressures of the teratocarcinoma environment. Nonetheless, mouse EC cells and mouse ES cells share the same unique combination of cell surface markers (SSEA-1 (+), SSEA-3 ( ⁇ ), SSEA-4 ( ⁇ ), and alkaline phosphatase (+)).
  • Pluripotent cell lines have also been derived from preimplantation embryos of several domestic and laboratory animals species (Evans, et al., Theriogenology 33(1):125-128, 1990; Evans, et al., Theriogenology 33(1):125-128, 1990; Notarianni, et al., J. Reprod. Fertil. 41(Suppl.):51-56, 1990; Giles, et al., Mol. Reprod. Dev. 36:130-138, 1993; Graves, et al., Mol. Reprod. Dev. 36:424-433, 1993; Sukoyan, et al., Mol. Reprod. Dev.
  • ES cells lines are true ES cells lines.
  • True ES cells should: (i) be capable of indefinite proliferation in vitro in an undifferentiated state; (ii) maintain a normal karyotype through prolonged culture; and (iii) maintain the potential to differentiate to derivatives of all three embryonic germ layers (endoderm, mesoderm, and ectoderm) even after prolonged culture. Strong evidence of these required properties have been published only for rodents ES cells including mouse (Evans & Kaufman, Nature 292:154-156, 1981; Martin, Proc Natl Acad Sci USA 78:7634-7638, 1981) hamster (Doetschmanet al.
  • the present invention is a purified preparation of primate embryonic stem cells.
  • the primate ES cell lines are true ES cell lines in that they: (i) are capable of indefinite proliferation in vitro in an undifferentiated state; (ii) are capable of differentiation to derivatives of all three embryonic germ layers (endoderm, mesoderm, and ectoderm) even after prolonged culture; and (iii) maintain a normal karyotype throughout prolonged culture.
  • the true primate ES cells lines are therefore pluripotent.
  • primate ES cell lines are preferably negative for the SSEA-1 marker, preferably positive for the SSEA-3 marker, and positive for the SSEA-4 marker.
  • the primate ES cell lines are also positive for the TRA-1-60, and TRA-1-81 markers, as well as positive for the alkaline phosphatase marker.
  • the primate ES cell lines continue to proliferate in an undifferentiated state after continuous culture for at least one year.
  • the cells remain euploid after proliferation in an undifferentiated state.
  • the cells can differentiate to trophoblast in vitro and express chorionic gonadotropin.
  • the present invention is also a purified preparation of primate embryonic stem cells that has the ability to differentiate into cells derived from mesoderm, endoderm, and ectoderm germ layers after the cells have been injected into an immunocompromised mouse, such as a SCID mouse.
  • the present invention is also a method of isolating a primate embryonic stem cell line.
  • the method comprises the steps of isolating a primate blastocyst, isolating cells from the inner cellular mass (ICM) of the blastocyst, plating the ICM cells on a fibroblast layer (wherein ICM-derived cell masses are formed) removing an ICM-derived cell mass and dissociating the mass into dissociated cells, replating the dissociated cells on embryonic feeder cells and selecting colonies with compact morphology containing cells with a high nucleus/cytoplasm ratio, and prominent nucleoli. The cells of the selected colonies are then cultured.
  • ICM inner cellular mass
  • FIG. 1 is a photomicrograph illustrating normal XY karyotype of rhesus ES cell line R278.5 after 11 months of continuous culture.
  • Photograph 2 A demonstrates the distinct cell borders, high nucleus to cytoplasm ratio, and prominent nucleoli of undifferentiated rhesus ES cells.
  • Photographs 2 B- 2 D shows differentiated cells eight days after plating R278.5 cells on gel treated tissue culture plastic (with 10 3 units/ml added human LIF). Cells of these three distinct morphologies are consistently present when R278.5 cells are allowed to differentiate at low density without fibroblasts either in the presence or absence of soluble human LIF.
  • Photograph 3 A shows Alkaline Phosphatase (+);
  • Photograph 3 B shows SSEA-1 ( ⁇ );
  • Photograph 3 C shows SSEA-3 (+);
  • Photograph 3 D shows SSEA-4 (+);
  • Photograph 3 E shows TRA-1-60 (+); and
  • Photograph 3 F shows TRA-1-81 (+).
  • FIGS. 4 A- 4 B are photographs illustrating expression of ⁇ -fetoprotein mRNA and ⁇ - and ⁇ -chorionic gonadotrophin mRNA expression in rhesus ES cells (R278.5) allowed to differentiate in culture.
  • FIGS. 5 A- 5 F include six photomicrographs of sections of tumors formed by injection of 0.5 ⁇ 10 6 rhesus ES (R278.5) cells into the hindleg muscles of SCID mice and analyzed 15 weeks later.
  • Photograph 5 A shows a low power field demonstrating disorganized differentiation of multiple cell types.
  • Photograph 5 C shows stratified squamous epithelium with several hair follicles.
  • FIGS. 6 A- 6 B include photographs of an embryoid Body. This embryoid body was formed from a marmoset ES cell line (Cj62) that had been continuously passaged in vitro for over 6 months.
  • Photograph 6 A (above) shows a section of the anterior 1 ⁇ 3 of the embryonic disc. Note the primitive ectoderm (E) forms a distinct cell layer from the underlying primitive endoderm (e), with no mixing of the cell layers. Note also that amnion (a) is composed of two distinct layers; the inner layer is continuous with the primitive ectoderm at the margins.
  • Photograph 6 B (below) shows a section in the caudal 1 ⁇ 3 of embryonic disc. Note central groove (arrow) and mixing of primitive ectoderm and endoderm representing early primitive streak formation, indicating the beginning of gastrulation. 400X, toluidine blue stain.
  • the present invention is a pluripotent, immortal euploid primate ES cell line, as exemplified by the isolation of ES cell lines from two primate species, the common marmoset ( Callithrix jacchus ) and the rhesus monkey ( Macaca mulatta ). Primate embryonic stem cells are useful for:
  • primate ES cells By manipulating culture conditions, primate ES cells, human and nonhuman, can be induced to differentiate to specific cell types, such as blood cells, neuron cells, or muscle cells.
  • primate ES cells can be allowed to differentiate in tumors in SCID mice, the tumors can be disassociated, and the specific differentiated cell types of interest can be selected by the usage of lineage specific markers through the use of fluorescent activated cell sorting (FACS) or other sorting method or by direct microdissection of tissues of interest.
  • FACS fluorescent activated cell sorting
  • These differentiated cells could then be transplanted back to the adult animal to treat specific diseases, such as hematopoietic disorders, endocrine deficiencies, degenerative neurological disorders or hair loss.
  • Macaques and marmosets were used as exemplary species for isolation of a primate ES cell line.
  • Macaques such as the rhesus monkey, are Old World species that are the major primates used in biomedical research. They are relatively large (about 7-10 kg). Males take 4-5 years to mature, and females have single young.
  • rhesus monkey true ES cell lines provide a very accurate in vitro model for human differentiation.
  • Rhesus monkey ES cell lines and rhesus monkeys will be particularly useful in the testing of the safety and efficacy of the transplantation of differentiated cell types into whole animals for the treatment of specific diseases or conditions.
  • the techniques developed for the rhesus ES cell lines model the generation, characterization and manipulation of human ES cell lines.
  • the common marmoset ( Callithrix jacchus ) is a New World primate species with reproductive characteristics that make it an excellent choice for ES cell derivation.
  • Marmosets are small (about 350-400 g), have a short gestation period (144 days), reach sexual maturity in about 18 months, and routinely have twins or triplets. Unlike in macaques, it is possible to routinely synchronize ovarian cycles in the marmoset with prostaglandin analogs, making collection of age-matched embryos from multiple females possible, and allowing efficient embryo transfer to synchronized recipients with 70%-80% of embryos transferred resulting in pregnancies. Because of these reproductive characteristics that allow for the routine efficient transfer of multiple embryos, marmosets provide an excellent primate species in which to generate transgenic models for human diseases.
  • human ES cell lines will be permanent cell lines that will also be distinguished from all other permanent human cell lines by their normal karyotype and the expression of the same combination of cell surface markers (alkaline phosphotase, preferably SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81) that characterize other primate ES cell lines.
  • a normal karyotype and the expression of this combination of cell surface markers will be defining properties of true human ES cell lines, regardless of the method used for their isolation and regardless of their tissue of origin.
  • ES medium consists of 80% Dulbecco's modified Eagle's medium (DMEM; no pyruvate, high glucose formulation, Gibco BRL), with 20% fetal bovine serum (FBS; Hyclone), 0.1 mM ⁇ -mercaptoethanol (Sigma), 1% non-essential amino acid stock (Gibco BRL).
  • FBS batches are compared by testing clonal plating efficiency of a low passage mouse ES cell line (ES jt3 ), a cell line developed just for the purpose of this test.
  • FBS batches must be compared because it has been found that batches vary dramatically in their ability to support embryonic cell growth, but any other method of assaying the competence of FBS batches for support of embryonic cells will work as an alternative.
  • ES cells are isolated on a confluent layer of murine embryonic fibroblast in the presence of ES cell medium.
  • Embryonic fibroblasts are preferably obtained from 12 day old fetuses from outbred CF1 mice (SASCO), but other strains may be used as an alternative.
  • Tissue culture dishes are preferably treated with 0.1% gelatin (type I; Sigma).
  • Expanded blastocysts are collected by non-surgical uterine flushing at six days after ovulation. This procedure routinely results in the recovery of an average 0.4 to 0.6 viable embryos per rhesus monkey per month, Seshagiri et al. Am J Primatol 29:81-91, 1993.
  • the day of ovulation is taken as the day preceding a plasma progesterone concentration of 10 ng/ml or more.
  • expanded blastocysts are recovered by a non-surgical uterine flush procedure, Thomson et al. “Non-surgical uterine stage preimplantation embryo collection from the common marmoset,” J Med Primatol, 23:333-336 (1994). This procedure results in the average production of 1.0 viable embryos per marmoset per month.
  • the zona pellucida is removed from blastocysts by brief exposure to pronase (Sigma).
  • pronase for immunosurgery, blastocysts are exposed to a 1:50 dilution of rabbit anti-marmoset spleen cell antiserum (for marmoset blastocysts) or a 1:50 dilution of rabbit anti-rhesus monkey (for rhesus monkey blastocysts) in DMEM for 30 minutes, then washed for 5 minutes three times in DMEM, then exposed to a 1:5 dilution of Guinea pig complement (Gibco) for 3 minutes.
  • lysed trophectoderm cells are removed from the intact inner cell mass (ICM) by gentle pipetting, and the ICM plated on mouse inactivated (3000 rads gamma irradiation) embryonic fibroblasts.
  • ICM-derived masses are removed from endoderm outgrowths with a micropipette with direct observation under a stereo microscope, exposed to 0.05% Trypsin-EDTA (Gibco) supplemented with 1% chicken serum for 3-5 minutes and gently dissociated by gentle pipetting through a flame polished micropipette.
  • Trypsin-EDTA Gibco
  • Dissociated cells are replated on embryonic feeder layers in fresh ES medium, and observed for colony formation. Colonies demonstrating ES-like morphology are individually selected, and split again as described above. The ES-like morphology is defined as compact colonies having a high nucleus to cytoplasm ratio and prominent nucleoli. Resulting ES cells are then routinely split by brief trypsinization or exposure to Dulbecco's Phosphate Buffered Saline (without calcium or magnesium and with 2 mM EDTA) every 1-2 weeks as the cultures become dense. Early passage cells are also frozen and stored in liquid nitrogen.
  • Cell lines may be karyotyped with a standard G-banding technique (such as by the Cytogenetics Laboratory of the University of Wisconsin State Hygiene Laboratory, which provides routine karyotyping services) and compared to published karyotypes for the primate species.
  • G-banding technique such as by the Cytogenetics Laboratory of the University of Wisconsin State Hygiene Laboratory, which provides routine karyotyping services
  • Isolation of ES cell lines from other primate species would follow a similar procedure, except that the rate of development to blastocyst can vary by a few days between species, and the rate of development of the cultured ICMs will vary between species. For example, six days after ovulation, rhesus monkey embryos are at the expanded blastocyst stage, whereas marmoset embryos don't reach the same stage until 7-8 days after ovulation. The Rhesus ES cell lines were obtained by splitting the ICM-derived cells for the first time at 7-16 days after immunosurgery; whereas the marmoset ES cells were derived with the initial split at 7-10 days after immunosurgery. Because other primates also vary in their developmental rate, the timing of embryo collection, and the timing of the initial ICM split will vary between primate species, but the same techniques and culture conditions will allow ES cell isolation.
  • human ES cells that are derived from preimplantation embryos will be derived from in vitro fertilized (IVF) embryos.
  • IVF in vitro fertilized
  • Experiments on unused (spare) human IVF-produced embryos are allowed in many countries, such as Singapore and the United Kingdom, if the embryos are less than 14 days old. Only high quality embryos are suitable for ES isolation.
  • Present defined culture conditions for culturing the one cell human embryo to the expanded blastocyst are suboptimal but practicable, Bongso et al., Hum Reprod 4:706-713, 1989.
  • Primate embryonic stem cells share features with the primate ICM and with pluripotent human embryonal carcinoma cells.
  • Putative primate ES cells may therefore be characterized by morphology and by the expression of cell surface markers characteristic of human EC cells. Additionally, putative primate ES cells may be characterized by developmental potential, karyotype and immortality.
  • the colony morphology of primate embryonic stem cell lines is similar to, but distinct from, mouse embryonic stem cells. Both mouse and primate ES cells have the characteristic features of undifferentiated stem cells, with high nuclear/cytoplasmic ratios, prominent nucleoli, and compact colony formation. The colonies of primate ES cells are flatter than mouse ES cell colonies and individual primate ES cells can be easily distinguished.
  • reference character A indicates a phase contrast photomicrograph of cell line R278.5 demonstrating the characteristic primate ES cell morphology.
  • a primate ES cell line of the present invention is distinct from mouse ES cell lines by the presence or absence of the cell surface markers described below.
  • One set of glycolipid cell surface markers is known as the Stage-specific embryonic antigens 1 through 4. These antigens can be identified using antibodies for SSEA 1, preferably SSEA-3 and SSEA-4 which are available from the Developmental Studies Hybridoma Bank of the National Institute of Child Health and Human Development.
  • SSEA 1-60 and TRA-1-81 designate antibodies from hybridomas developed by Peter Andrews of the University of Sheffield and are described in Andrews et al., “Cell lines from human germ cell tumors,” In: Robertson E, ed. Teratocarcinomas and Embryonic Stem Cells: A Practical Approach . Oxford: IRL Press, 207-246, 1987.
  • NTERA-2 cl. D1 a pluripotent human EC cell line (gift of Peter Andrews), may be used as a negative control for SSEA-1, and as a positive control for SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81. This cell line was chosen for positive control only because it has been extensively studied and reported in the literature, but other human EC cell lines may be used as well.
  • Mouse ES cells (ES jt3 ) are used as a positive control for SSEA-1, and for a negative control for SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81.
  • Other routine negative controls include omission of the primary or secondary antibody and substitution of a primary antibody with an unrelated specificity.
  • Alkaline phosphatase may be detected following fixation of cells with 4% para-formaldehyde using “Vector Red” (Vector Laboratories) as a substrate, as described by the manufacturer (Vector Laboratories).
  • Vector Red Vector Laboratories
  • the precipitate formed by this substrate is red when viewed with a rhodamine filter system, providing substantial amplification over light microscopy.
  • Table 1 diagrams a comparison of mouse ES cells, primate ES cells, and human EC cells.
  • the only cells reported to express the combination of markers SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 other than primate ES cells are human EC cells.
  • the globo-series glycolipids SSEA-3 and SSEA-4 are consistently present on human EC cells, and are of diagnostic value in distinguishing human EC cell tumors from human yolk sac carcinomas, choriocarcinomas, and other lineages which lack these markers, Wenk et al., Int J Cancer 58:108-115, 1994.
  • TRA-1-60 and TRA-1-81 antigens have been studied extensively on a particular pluripotent human EC cell line, NTERA-2 CL. D1, Andrews et al, supra. Differentiation of NTERA-2 CL. D1 cells in vitro results in the loss of SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 expression and the increased expression of the lacto-series glycolipid SSEA-1, Andrews et al, supra. This contrasts with undifferentiated mouse ES cells, which express SSEA-1, and neither SSEA-3 nor SSEA-4. Although the function of these antigens are unknown, their shared expression by R278.5 cells and human EC cells suggests a close embryological similarity. Alkaline phosphatase will also be present on all primate ES cells. A successful primate ES cell culture of the present invention will correlate with the cell surface markers found in the rhesus macaque and marmoset cell lines described in Table 1.
  • the rhesus macaque and marmoset cell lines are identical to human EC cell lines for the 5 described markers. Therefore, a successful primate ES cell culture will also mimic human EC cells. However, there are other ways to discriminate ES cells from EC cells. For example, the primate ES cell line has a normal karyotype and the human EC cell line is aneuploid.
  • FIG. 3 the photographs labelled A through F demonstrate the characteristic staining of these markers on a rhesus monkey ES cell line designated R278.5.
  • TABLE 1 Mouse C. jacchus M. mulatta Human EC ES ES ES (NTERA-2 cl.D1) SSEA-1 + ⁇ ⁇ ⁇ SSEA-3 ⁇ + + + SSEA-4 ⁇ + + + Tra-1-60 ⁇ + + + Tra-1-81 ⁇ + + + + +
  • Primate ES cells of the present invention are pluripotent.
  • pluripotent we mean that the cell has the ability to develop into any cell derived from the three main germ cell layers or an embryo itself.
  • a successful primate ES cell line will differentiate into cells derived from all three embryonic germ layers including: bone, cartilage, smooth muscle, striated muscle, and hematopoietic cells (mesoderm); liver, primitive gut and respiratory epithelium (endoderm); neurons, glial cells, hair follicles, and tooth buds (ectoderm).
  • This experiment can be accomplished by injecting approximately 0.5-1.0 ⁇ 10 6 primate ES cells into the rear leg muscles of 8-12 week old male SCID mice.
  • the resulting tumors can be fixed in 4% paraformaldehyde and examined histologically after paraffin embedding at 8-16 weeks of development.
  • photomicrographs designated A-F are of sections of tumors formed by injection of rhesus ES cells into the hind leg muscles of SCID mice and analyzed 15 weeks later demonstrating cartilage, smooth muscle, and striated muscle (mesoderm); stratified squamous epithelium with hair follicles, neural tube with ventricular, intermediate, and mantle layers (ectoderm); ciliated columnar epithelium and villi lined by absorptive enterocytes and mucus-secreting goblet cells (endoderm).
  • a successful nonhuman primate ES cell line will have the ability to participate in normal development when combined in chimeras with normal preimplantation embryos. Chimeras between preimplantation nonhuman primate embryos and nonhuman primate ES cells can be formed by routine methods in several ways.
  • injection chimeras 10-15 nonhuman primate ES cells can be microinjected into the cavity of an expanded nonhuman primate blastocyst;
  • aggregation chimeras nonhuman primate morulae can be co-cultured on a lawn of nonhuman primate ES cells and allowed to aggregate; and
  • tetraploid chimeras 10-15 nonhuman primate ES cells can be aggregated with tetraploid nonhuman primate morulae obtained by electrofusion of 2-cell embryos, or incubation of morulae in the cytoskeletal inhibitor cholchicine.
  • the chimeras can be returned to the uterus of a female nonhuman primate and allowed to develop to term, and the ES cells will contribute to normal differentiated tissues derived from all three embryonic germ layers and to germ cells. Because nonhuman primate ES can be genetically manipulated prior to chimera formation by standard techniques, chimera formation followed by embryo transfer can lead to the production of transgenic nonhuman primates.
  • Successful primate ES cell lines have normal karyotypes. Both XX and XY cells lines will be derived. The normal karyotypes in primate ES cell lines will be in contrast to the abnormal karyotype found in human embryonal carcinoma (EC), which are derived from spontaneously arising human germ cell tumors (teratocarcinomas). Human embryonal carcinoma cells have a limited ability to differentiate into multiple cell types and represent the closest existing cell lines to primate ES cells. Although tumor-derived human embryonal carcinoma cell lines have some properties in common with embryonic stem cell lines, all human embryonal carcinoma cell lines derived to date are aneuploid.
  • EC human embryonal carcinoma
  • primate ES cell lines and human EC cell lines can be distinguished by the normal karyotypes found in primate ES cell lines and the abnormal karyotypes found in human EC lines.
  • normal karyotype it is meant that all chromosomes normally characteristic of the species are present and have not been noticeably altered.
  • Immortal cells are capable of continuous indefinite replication in vitro. Continued proliferation for longer than one year of culture is a sufficient evidence for immortality, as primary cell cultures without this property fail to continuously divide for this length of time (Freshney, Culture of animal cells . New York: Wiley-Liss, 1994). Primate ES cells will continue to proliferate in vitro with the culture conditions described above for longer than one year, and will maintain the developmental potential to contribute all three embryonic germ layers. This developmental potential can be demonstrated by the injection of ES cells that have been cultured for a prolonged period (over a year) into SCID mice and then histologically examining the resulting tumors. Although karyotypic changes can occur randomly with prolonged culture, some primate ES cells will maintain a normal karyotype for longer than a year of continuous culture.
  • LIF Leukemia inhibitory factor
  • primate ES cells of the present invention spontaneously differentiate and will produce chorionic gonadotropin, indicating trophoblast differentiation (a component of the placenta) and produce ⁇ -fetoprotein, indicating endoderm differentiation.
  • Chorionic gonadotropin activity can be assayed in the medium conditioned by differentiated cells by Leydig cell bioassay, Seshagiri & Hearn, Hum Reprod 8:279-287, 1992.
  • RNA can be prepared by guanidine isothiocyanate-phenol/chloroform extraction (1) from approximately 0.2 ⁇ 10 6 differentiated cells and from 0.2 ⁇ 10 6 undifferentiated cells.
  • the relative levels of the mRNA for ⁇ -fetoprotein and the ⁇ - and ⁇ -subunit of chorionic gonadotropin relative to glyceraldehyde-3-phosphate dehydrogenase can be determined by semi-quantitative Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR).
  • the PCR primers for glyceraldehyde 3-phosphate dehydrogenase are based on the human cDNA sequence, and do not amplify mouse G3PDH mRNA under our conditions.
  • the identity of the ⁇ -fetoprotein, CG ⁇ and CG ⁇ cDNAs can be verified by subcloning and sequencing.
  • the number of amplification rounds which produced linear increases in target cDNAs and the relation between input RNA and amount of PCR product is empirically determined as by Golos et al. Samples were fractionated in 3% Nusieve (FMC, Rockland, Me.) agarose gels (1X TBE running buffer) and DNA bands of interest were cut out, melted at 65° C. in 0.5 ml TE, and radioactivity determined by liquid scintillation counting. The ratio of counts per minute in a specific PCR product relative to cpm of G3PDH PCR product is used to estimate the relative levels of a mRNAs among differentiated and undifferentiated cells.
  • Nusieve FMC, Rockland, Me.
  • WRPRC Wisconsin Regional Primate Research Center
  • R278.5 three independent embryonic stem cell lines from two rhesus monkey blastocysts (R278.5, R366, and R367).
  • R278.5 remains undifferentiated and continues to proliferate after continuous culture for over one year.
  • R278.5 cells have also been frozen and successfully thawed with the recovery of viable cells.
  • R278.5 cells The morphology and cell surface markers of R278.5 cells are indistinguishable from human EC cells, and differ significantly from mouse ES cells.
  • R278.5 cells have a high nucleus/cytoplasm ratio and prominent nucleoli, but rather than forming compact, piled-up colonies with indistinct cell borders similar to mouse ES cells, R278.5 cells form flatter colonies with individual, distinct cells (FIG. 2 A).
  • R278.5 cells express the SSEA-3, SSEA-4, TRA-1-60, and TRA-81 antigens (FIG. 3 and Table 1), none of which are expressed by mouse ES cells.
  • the only cells known to express the combination of markers SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 other than primate ES cells are human EC cells.
  • the globo-series glycolipids SSEA-3 and SSEA-4 are consistently present on human EC cells, and are of diagnostic value in distinguishing human EC cell tumors from yolk sac carcinomas, choriocarcinomas and other stem cells derived from human germ cell tumors which lack these markers, Wenk et al, Int J Cancer 58:108-115, 1994.
  • TRA-1-60 and TRA-1-81 antigens have been studied extensively on a particular pluripotent human EC cell line, NTERA-2 CL. D1 (Andrews et al.). Differentiation of NTERA-2 CL. D1 cells in vitro results in the loss of SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 expression and the increased expression of the lacto-series glycolipid SSEA-1. Undifferentiated mouse ES cells, on the other hand, express SSEA-1, and not SSEA-3, SSEA-4, TRA-1-60 or TRA-1-81 (Wenk et al.). Although the function of these antigens is unknown, their expression by R278.5 cells suggests a close embryological similarity between primate ES cells and human EC cells, and fundamental differences between primate ES cells and mouse ES cells.
  • R278.5 cells also express alkaline phosphatase.
  • alkaline phosphatase The expression of alkaline phosphatase is shared by both primate and mouse ES cells, and relatively few embryonic cells express this enzyme.
  • Positive cells include the ICM and primitive ectoderm (which are the most similar embryonic cells in the intact embryo to ES cells), germ cells, (which are totipotent), and a very limited number of neural precursors, Kaufman M H. The atlas of mouse development . London: Academic Press, 1992. Cells not expressing this enzyme will not be primate ES cells.
  • R366 and R367 Since the derivation of the R278.5 cell line, we have derived two additional rhesus ES cell lines (R366 and R367) on embryonic fibroblasts without any exogenously added LIF at initial derivation. R366 and R367 cells, like R278.5 cells, continue to proliferate on embryonic fibroblasts without exogenously added LIF and differentiate in the absence of fibroblasts, regardless of the presence of added LIF. RT-PCR performed on mRNA from spontaneously differentiated R278.5 cells revealed ⁇ -fetoprotein mRNA (FIG. 4).
  • ⁇ -fetoprotein is a specific marker for endoderm, and is expressed by both extraembryonic (yolk sac) and embryonic (fetal liver and intestines) endoderm-derived tissues.
  • Epithelial cells resembling extraembryonic endoderm are present in cells differentiated in vitro from R278.5 cells (FIG. 2).
  • Bioactive CG (3.89 mI units/ml) was present in culture medium collected from differentiated cells, but not in medium collected from undifferentiated cells (less than 0.03 mI units/ml), indicating the differentiation of trophoblast, a trophectoderm derivative.
  • the relative level of the CG ⁇ mRNA increased 23.9-fold after differentiation (FIG. 4).
  • Such structures included gut lined by villi with both absorptive enterocytes and mucus-secreting goblet cells, and sometimes encircled by layers of smooth muscle in the same orientation as muscularis mucosae (circular) and muscularis (outer longitudinal layer and inner circular layer); neural tubes with ventricular, intermediate, and mantle layers; and hair follicles with hair shafts (FIG. 5).
  • R278.5 cells The essential characteristics that define R278.5 cells as ES cells include: indefinite (greater than one year) undifferentiated proliferation in vitro, normal karyotype, and potential to differentiate to derivatives of trophectoderm and all three embryonic germ layers.
  • indefinite greater than one year
  • the last cells capable of contributing to derivatives of both trophectoderm and ICM are early ICM cells.
  • the timing of commitment to ICM or trophectoderm has not been established for any primate species, but the potential of rhesus ES cells to contribute to derivatives of both suggests that they most closely resemble early totipotent embryonic cells.
  • the ability of rhesus ES cells to form trophoblast in vitro distinguishes primate ES cell lines from mouse ES cells.
  • Mouse ES cell have not been demonstrated to form trophoblast in vitro, and mouse trophoblast does not produce gonadotropin.
  • Rhesus ES cells and mouse ES cells do demonstrate the similar wide range of differentiation in tumors that distinguishes ES cells from EC cells.
  • the development of structures composed of multiple cell types such as hair follicles, which require inductive interactions between the embryonic epidermis and underlying mesenchyme, demonstrates the ability of rhesus ES cells to participate in complex developmental processes.
  • the rhesus ES lines R366 and R367 have also been further cultured and analyzed. Both lines have a normal XY karyotype and were proliferated in an undifferentiated state for about three months prior to freezing for later analysis. Samples of each of the cell lines R366 and R367 were injected into SCID mice which then formed teratomas identical to those formed by R278.5 cells. An additional rhesus cell line R394 having a normal XX karyotype was also recovered. All three of these cell lines, R366, R367 and R394 are identical in morphology, growth characteristics, culture requirements and in vitro differentiation characteristics, i.e. the trait of differentiation to multiple cell types in the absence of fibroblasts, to cell line 278.5.
  • fibroblasts for co-culture are not critical.
  • Several fibroblast cell lines have been tested both with rhesus line R278.5 and with the marmoset cell lines described below.
  • the fibroblasts tested include mouse STO cells (ATCC 56-X), mouse 3T3 cells (ATCC 48-X), primary rhesus monkey embryonic fibroblasts derived from 36 day rhesus fetuses, and mouse Sl/Sl 4 cells, which are deficient in the steel factor. All these fibroblast cell lines were capable of maintaining the stem cell lines in an undifferentiated state. Most rapid proliferation of the stem cells was observed using primary mouse embryonic fibroblasts.
  • Rhesus ES cells will be important for elucidating the mechanisms that control the differentiation of specific primate cell types. Given the close evolutionary distance and the developmental and physiological similarities between humans and rhesus monkeys, the mechanisms controlling the differentiation of rhesus cells will be very similar to the mechanisms controlling the differentiation of human cells. The importance of elucidating these mechanisms is that once they are understood, it will be possible to direct primate ES cells to differentiate to specific cell types in vitro, and these specific cell types can be used for transplantation to treat specific diseases.
  • ES cells have the developmental potential to give rise to any differentiated cell type, any disease that results in part or in whole from the failure (either genetic or acquired) of specific cell types will be potentially treatable through the transplantation of cells derived from ES cells.
  • Rhesus ES cells and rhesus monkeys will be invaluable for testing the efficacy and safety of the transplantation of specific cell types derived from ES cells.
  • a few examples of human diseases potentially treatable by this approach with human ES cells include degenerative neurological disorders such as Parkinson's disease (dopanergic neurons), juvenile onset diabetes (pancreatic ⁇ -islet cells) or Acquired Immunodeficiency Disease (lymphocytes).
  • undifferentiated ES cells can proliferate indefinitely in vitro, they can be genetically manipulated with standard techniques either to prevent immune rejection after transplantation, or to give them new genetic properties to combat specific diseases.
  • cells derived from rhesus monkey ES cells or other non-human primate ES cells could be used for transplantation to humans to treat specific diseases.
  • Cj11 was cultured continuously for over 14 months, and then frozen for later analysis.
  • the Cj11 cell line and other marmoset ES cell lines have been successfully frozen and then thawed with the recovery of viable cells.
  • These cells have a high nuclear/cytoplasmic ratio, prominent nucleoli, and a compact colony morphology similar to the pluripotent human embryonal carcinoma (EC) cell line NT2/D2.
  • the putative marmoset ES cells When the putative marmoset ES cells were removed from fibroblast feeders, they differentiated into cells of several distinct morphologies. Among the differentiated cells, trophectoderm is indicated by the secretion of chorionic gonadotropin and the presence of the chorionic gonadotropin ⁇ -subunit mRNA. 12.7 mIU/ml luteinizing hormone (LH) activity was measured in the WRPRC core assay lab using a mouse Leydig cell bioassay in medium conditioned 24 hours by putative ES cells allowed to differentiate for one week. Note that chorionic gonadotrophin has both LH and FSH activity, and is routinely measured by LH assays. Control medium from undifferentiated ES cells had less than 1 mIU/ml LH activity.
  • LH luteinizing hormone
  • RT-PCR reverse transcriptase-polymerase chain reaction
  • Endoderm differentiation (probably extraembryonic endoderm) was indicated by the presence of ⁇ -fetoprotein mRNA, detected by RT-PCR.
  • the embryonic disc was composed of a polarized, columnar epithelial epiblast (primitive ectoderm) layer separated from a visceral endoderm (primitive endoderm) layer. Electron microscopy of the epiblast revealed apical junctional complexes, apical microvilli, subapical intermediate filaments, and a basement membrane separating the epiblast from underlying visceral endoderm. All of these elements are features of the normal embryonic disc.
  • the amnion was composed of an inner squamous (ectoderm) layer continuous with the epiblast and an outer mesoderm layer.
  • the bilayered yolk sac had occasional endothelial-lined spaces containing possible hematopoietic precursors.
  • marmoset cells identify these marmoset cells as primate ES cells similar to the rhesus ES cells. Since the last cells in the mammalian embryo capable of contributing to both trophectoderm derivatives and endoderm derivatives are the totipotent cells of the early ICM, the ability of marmoset ES cells to contribute to both trophoblast and endoderm demonstrates their similarities to early totipotent embryonic cells of the intact embryo. The formation of embryoid bodies by marmoset ES cells, with remarkable structural similarities to the early post-implantation primate embryo, demonstrates the potential of marmoset ES cells to participate in complex developmental processes requiring the interaction of multiple cell types.
  • marmoset ES cells Given the reproductive characteristics of the common marmoset described above (efficient embryo transfer, multiple young, short generation time), marmoset ES cells will be particularly useful for the generation of transgenic primates. Although mice have provided invaluable insights into gene function and regulation, the anatomical and physiological differences between humans and mice limit the usefulness of transgenic mouse models of human diseases. Transgenic primates, in addition to providing insights into the pathogenesis of specific diseases, will provide accurate animal models to test the efficacy and safety of specific treatments.

Abstract

A purified preparation of primate embryonic stem cells is disclosed. This preparation is characterized by the following cell surface markers: SSEA-1 (−); SSEA-4 (+); TRA-1-60 (+); TRA-1-81 (+); and alkaline phosphatase (+). In a particularly advantageous embodiment, the cells of the preparation are human embryonic stem cells, have normal karyotypes, and continue to proliferate in an undifferentiated state after continuous culture for eleven months. The embryonic stem cell lines also retain the ability, throughout the culture, to form trophoblast and to differentiate into all tissues derived from all three embryonic germ layers (endoderm, mesoderm and ectoderm). A method for isolating a primate embryonic stem cell line is also disclosed.

Description

    CROSS REFERENCES TO RELATED APPLICATIONS
  • This application is a divisional of U.S. Ser. No. 08/591,246 which was filed on Jan. 18, 1996, and is a continuation-in-part of U.S. Ser. No. 08/376,327 which was filed on Jan. 20, 1995.[0001]
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
  • [0002] This invention was made with United States government support awarded by NIH NCRR Grant No. RR00167. The United States government has certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • In general, the field of the present invention is stem cell cultures. Specifically, the field of the present invention is primate embryonic stem cell cultures. [0003]
  • In general, stem cells are undifferentiated cells which can give rise to a succession of mature functional cells. For example, a hematopoietic stem cell may give rise to any of the different types of terminally differentiated blood cells. Embryonic stem (ES) cells are derived from the embryo and are pluripotent, thus possessing the capability of developing into any organ or tissue type or, at least potentially, into a complete embryo. [0004]
  • One of the seminal achievements of mammalian embryology of the last decade is the routine insertion of specific genes into the mouse genome through the use of mouse ES cells. This alteration has created a bridge between the in vitro manipulations of molecular biology and an understanding of gene function in the intact animal. Mouse ES cells are undifferentiated, pluripotent cells derived in vitro from preimplantation embryos (Evans, et al. [0005] Nature 292:154-159, 1981; Martin, Proc. Natl. Acad. Sci. USA 78:7634-7638, 1981) or from fetal germ cells (Matsui, et al., Cell 70:841-847, 1992). Mouse ES cells maintain an undifferentiated state through serial passages when cultured in the presence of fibroblast feeder layers in the presence of Leukemia Inhibitory Factor (LIF) (Williams, et al., Nature 336:684-687, 1988). If LIF is removed, mouse ES cells differentiate.
  • Mouse ES cells cultured in non-attaching conditions aggregate and differentiate into simple embryoid bodies, with an outer layer of endoderm and an inner core of primitive ectoderm. If these embryoid bodies are then allowed to attach onto a tissue culture surface, disorganized differentiation occurs of various cell types, including nerves, blood cells, muscle, and cartilage (Martin, 1981, supra; Doetschman, et al., [0006] J. Embryol. Exp. Morph. 87:27-45, 1985). Mouse ES cells injected into syngeneic mice form teratocarcinomas that exhibit disorganized differentiation, often with representatives of all three embryonic germ layers. Mouse ES cells combined into chimeras with normal preimplantation embryos and returned to the uterus participate in normal development (Richard, et al., Cytogenet. Cell Genet. 65:169-171, 1994).
  • The ability of mouse ES cells to contribute to functional germ cells in chimeras provides a method for introducing site-specific mutations into mouse lines. With appropriate transfection and selection strategies, homologous recombination can be used to derive ES cell lines with planned alterations of specific genes. These genetically altered cells can be used to form chimeras with normal embryos and chimeric animals are recovered. If the ES cells contribute to the germ line in the chimeric animal, then in the next generation a mouse line for the planned mutation is established. [0007]
  • Because mouse ES cells have the potential to differentiate into any cell type in the body, mouse ES cells allow the in vitro study of the mechanisms controlling the differentiation of specific cells or tissues. Although the study of mouse ES cells provides clues to understanding the differentiation of general mammalian tissues, dramatic differences in primate and mouse development of specific lineages limits the usefulness of mouse ES cells as a model of human development. Mouse and primate embryos differ meaningfully in the timing of expression of the embryonic genome, in the formation of an egg cylinder versus an embryonic disc (Kaufman, [0008] The Atlas of Mouse Development, London: Academic Press, 1992), in the proposed derivation of some early lineages (O'Rahilly & Muller, Developmental Stages in Human Embryos, Washington: Carnegie Institution of Washington, 1987), and in the structure and function in the extraembryonic membranes and placenta (Mossman, Vertebrate Fetal Membranes, New Brunswick: Rutgers, 1987). Other tissues differ in growth factor requirements for development (e.g. the hematopoietic system (Lapidot et al., Lab An Sci 43:147-149, 1994)), and in adult structure and function (e.g. the central nervous system). Because humans are primates, and development is remarkably similar among primates, primate ES cells lines will provide a faithful model for understanding the differentiation of primate tissues in general and human tissues in particular.
  • The placenta provides just one example of how primate ES cells will provide an accurate model of human development that cannot be provided by ES cells from other species. The placenta and extraembryonic membranes differ dramatically between mice and humans. Structurally, the mouse placenta is classified as labyrinthine, whereas the human and the rhesus monkey placenta are classified as villous. Chorionic gonadotropin, expressed by the trophoblast, is an essential molecule involved in maternal recognition of pregnancy in all primates, including humans (Hearn, [0009] J Reprod Fertil 76:809-819, 1986; Hearn et al., J Reprod Fert 92:497-509, 1991). Trophoblast secretion of chorionic gonadotropin in primates maintains the corpus luteum of pregnancy and, thus, progesterone secretion. Without progesterone, pregnancy fails. Yet mouse trophoblast produces no chorionic gonadotropin, and mice use entirely different mechanisms for pregnancy maintenance (Hearn et al., “Normal and abnormal embryo-fetal development in mammals,” In: Lamming E, ed. Marshall's Physiology of Reproduction. 4th ed. Edinburgh, N.Y.: Churchill Livingstone, 535-676, 1994). An immortal, euploid, primate ES cell line with the developmental potential to form trophoblast in vitro, will allow the study of the ontogeny and function of genes such as chorionic gonadotropin which are critically important in human pregnancy. Indeed, the differentiation of any tissue for which there are significant differences between mice and primates will be more accurately reflected in vitro by primate ES cells than by mouse ES cells.
  • The major in vitro models for studying trophoblast function include human choriocarcinoma cells, which are malignant cells that may not faithfully reflect normal trophectoderm; short-term primary cultures of human and non-human primate cytotrophoblast, which in present culture conditions quickly form non-dividing syncytial trophoblast; and in vitro culture of preimplantation non-human primate embryos (Hearn, et al., [0010] J. Endocrinol. 119:249-255, 1988; Coutifaris, et al., Ann. NY Acad. Sci. 191-201, 1994). An immortal, euploid, non-human primate embryonic stem (ES) cell line with the developmental potential to form trophectoderm offers significant advantages over present in vitro models of human trophectoderm development and function, as trophoblast-specific genes such as chorionic gonadotropin could be stably altered in the ES cells and then studied during differentiation to trophectoderm.
  • The cell lines currently available that resembles primate ES cells most closely are human embryonic carcinoma (EC) cells, which are pluripotent, immortal cells derived from teratocarcinomas (Andrews, et al., [0011] Lab. Invest. 50(2):147-162, 1984; Andrews, et al., in: Robertson E., ed. Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. Oxford: IRL press, pp. 207-246, 1987). EC cells can be induced to differentiate in culture, and the differentiation is characterized by the loss of specific cell surface markers (SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81) and the appearance of new markers (Andrews, et al., 1987, supra). Human EC cells will form teratocarcinomas with derivatives of multiple embryonic lineages in tumors in nude mice. However, the range of differentiation of these human EC cells is limited compared to the range of differentiation obtained with mouse ES cells, and all EC cell lines derived to date are aneuploid (Andrews, et al., 1987, supra). Similar mouse EC cell lines have been derived from teratocarcinomas, and, in general their developmental potential is much more limited than mouse ES cells (Rossant, et al., Cell Differ. 15:155-161, 1984). Teratocarcinomas are tumors derived from germ cells, and although germ cells (like ES cells) are theoretically totipotent (i.e. capable of forming all cell types in the body), the more limited developmental potential and the abnormal karyotypes of EC cells are thought to result from selective pressures in the teratocarcinoma tumor environment (Rossant & Papaioannou, Cell Differ 15:155-161, 1984). ES cells, on the other hand, are thought to retain greater developmental potential because they are derived from normal embryonic cells in vitro, without the selective pressures of the teratocarcinoma environment. Nonetheless, mouse EC cells and mouse ES cells share the same unique combination of cell surface markers (SSEA-1 (+), SSEA-3 (−), SSEA-4 (−), and alkaline phosphatase (+)).
  • Pluripotent cell lines have also been derived from preimplantation embryos of several domestic and laboratory animals species (Evans, et al., [0012] Theriogenology 33(1):125-128, 1990; Evans, et al., Theriogenology 33(1):125-128, 1990; Notarianni, et al., J. Reprod. Fertil. 41(Suppl.):51-56, 1990; Giles, et al., Mol. Reprod. Dev. 36:130-138, 1993; Graves, et al., Mol. Reprod. Dev. 36:424-433, 1993; Sukoyan, et al., Mol. Reprod. Dev. 33:418-431, 1992; Sukoyan, et al., Mol. Reprod. Dev. 36:148-158, 1993; Iannaccone, et al., Dev. Biol. 163:288-292, 1994).
  • Whether or not these cell lines are true ES cells lines is a subject about which there may be some difference of opinion. True ES cells should: (i) be capable of indefinite proliferation in vitro in an undifferentiated state; (ii) maintain a normal karyotype through prolonged culture; and (iii) maintain the potential to differentiate to derivatives of all three embryonic germ layers (endoderm, mesoderm, and ectoderm) even after prolonged culture. Strong evidence of these required properties have been published only for rodents ES cells including mouse (Evans & Kaufman, [0013] Nature 292:154-156, 1981; Martin, Proc Natl Acad Sci USA 78:7634-7638, 1981) hamster (Doetschmanet al. Dev Biol 127:224-227, 1988), and rat (Iannaccone et al. Dev Biol 163:288-292, 1994), and less conclusively for rabbit ES cells (Gileset al. Mol Reprod Dev 36:130-138, 1993; Graves & Moreadith, Mol Reprod Dev 36:424-433, 1993). However, only established ES cell lines from the rat (Iannaccone, et al., 1994, supra) and the mouse (Bradley, et al., Nature 309:255-256, 1984) have been reported to participate in normal development in chimeras. There are no reports of the derivation of any primate ES cell line.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention is a purified preparation of primate embryonic stem cells. The primate ES cell lines are true ES cell lines in that they: (i) are capable of indefinite proliferation in vitro in an undifferentiated state; (ii) are capable of differentiation to derivatives of all three embryonic germ layers (endoderm, mesoderm, and ectoderm) even after prolonged culture; and (iii) maintain a normal karyotype throughout prolonged culture. The true primate ES cells lines are therefore pluripotent. [0014]
  • The present invention is also summarized in that primate ES cell lines are preferably negative for the SSEA-1 marker, preferably positive for the SSEA-3 marker, and positive for the SSEA-4 marker. The primate ES cell lines are also positive for the TRA-1-60, and TRA-1-81 markers, as well as positive for the alkaline phosphatase marker. [0015]
  • It is an advantageous feature of the present invention that the primate ES cell lines continue to proliferate in an undifferentiated state after continuous culture for at least one year. In a particularly advantageous embodiment, the cells remain euploid after proliferation in an undifferentiated state. [0016]
  • It is a feature of the primate ES cell lines in accordance with the present invention that the cells can differentiate to trophoblast in vitro and express chorionic gonadotropin. [0017]
  • The present invention is also a purified preparation of primate embryonic stem cells that has the ability to differentiate into cells derived from mesoderm, endoderm, and ectoderm germ layers after the cells have been injected into an immunocompromised mouse, such as a SCID mouse. [0018]
  • The present invention is also a method of isolating a primate embryonic stem cell line. The method comprises the steps of isolating a primate blastocyst, isolating cells from the inner cellular mass (ICM) of the blastocyst, plating the ICM cells on a fibroblast layer (wherein ICM-derived cell masses are formed) removing an ICM-derived cell mass and dissociating the mass into dissociated cells, replating the dissociated cells on embryonic feeder cells and selecting colonies with compact morphology containing cells with a high nucleus/cytoplasm ratio, and prominent nucleoli. The cells of the selected colonies are then cultured. [0019]
  • It is an object of the present invention to provide a primate embryonic stem cell line. [0020]
  • It is an object of the present invention to provide a primate embryonic stem cell line characterized by the following markers: alkaline phosphatase(+); SSEA-1(−); preferably SSEA-3(+); SSEA-4(+); TRA-1-60(+); and TRA-1-81(+). [0021]
  • It is an object of the present invention to provide a primate embryonic stem cell line capable of proliferation in an undifferentiated state after continuous culture for at least one year. Preferably, these cells remain euploid. [0022]
  • It is another object of the present invention to provide a primate embryonic stem cell line wherein the cells differentiate into cells derived from mesoderm, endoderm, and ectoderm germ layers when the cells are injected into an immunocompromised mouse. [0023]
  • Other objects, features, and advantages of the present invention will become obvious after study of the specification, drawings, and claims.[0024]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a photomicrograph illustrating normal XY karyotype of rhesus ES cell line R278.5 after 11 months of continuous culture. [0025]
  • FIGS. [0026] 2A-2D are a set of phase-contrast photomicrographs demonstrating the morphology of undifferentiated rhesus ES (R278.5) cells and of cells differentiated from R278.5 in vitro (bar=100μ). Photograph 2A demonstrates the distinct cell borders, high nucleus to cytoplasm ratio, and prominent nucleoli of undifferentiated rhesus ES cells. Photographs 2B-2D shows differentiated cells eight days after plating R278.5 cells on gel treated tissue culture plastic (with 103 units/ml added human LIF). Cells of these three distinct morphologies are consistently present when R278.5 cells are allowed to differentiate at low density without fibroblasts either in the presence or absence of soluble human LIF.
  • FIGS. [0027] 3A-F are photomicrographs demonstrating the expression of cell surface markers on undifferentiated rhesus ES (R278.5) cells (bar=100μ). Photograph 3A shows Alkaline Phosphatase (+); Photograph 3B shows SSEA-1 (−); Photograph 3C shows SSEA-3 (+); Photograph 3D shows SSEA-4 (+); Photograph 3E shows TRA-1-60 (+); and Photograph 3F shows TRA-1-81 (+).
  • FIGS. [0028] 4A-4B are photographs illustrating expression of α-fetoprotein mRNA and α- and β-chorionic gonadotrophin mRNA expression in rhesus ES cells (R278.5) allowed to differentiate in culture.
  • FIGS. [0029] 5A-5F include six photomicrographs of sections of tumors formed by injection of 0.5×106 rhesus ES (R278.5) cells into the hindleg muscles of SCID mice and analyzed 15 weeks later. Photograph 5A shows a low power field demonstrating disorganized differentiation of multiple cell types. A gut-like structure is encircled by smooth muscle(s), and elsewhere foci of cartilage (c) are present (bar=400μ); Photograph 5B shows striated muscle (bar=40μ); Photograph 5C shows stratified squamous epithelium with several hair follicles. The labeled hair follicle (f) has a visible hair shaft (bar=200μ); Photograph 5D shows stratified layers of neural cells in the pattern of a developing neural tube. An upper “ventricular” layer, containing numerous mitotic figures (arrows), overlies a lower “mantle” layer. (bar=100μ); Photograph 5E shows ciliated columnar epithelium (bar=40μ); Photograph 5F shows villi covered with columnar epithelium with interspersed mucus-secreting goblet cells (bar=200μ).
  • FIGS. [0030] 6A-6B include photographs of an embryoid Body. This embryoid body was formed from a marmoset ES cell line (Cj62) that had been continuously passaged in vitro for over 6 months. Photograph 6A (above) shows a section of the anterior ⅓ of the embryonic disc. Note the primitive ectoderm (E) forms a distinct cell layer from the underlying primitive endoderm (e), with no mixing of the cell layers. Note also that amnion (a) is composed of two distinct layers; the inner layer is continuous with the primitive ectoderm at the margins. Photograph 6B (below) shows a section in the caudal ⅓ of embryonic disc. Note central groove (arrow) and mixing of primitive ectoderm and endoderm representing early primitive streak formation, indicating the beginning of gastrulation. 400X, toluidine blue stain.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • (1) In General [0031]
  • (a) Uses of Primate ES Cells [0032]
  • The present invention is a pluripotent, immortal euploid primate ES cell line, as exemplified by the isolation of ES cell lines from two primate species, the common marmoset ([0033] Callithrix jacchus) and the rhesus monkey (Macaca mulatta). Primate embryonic stem cells are useful for:
  • (i) Generating transgenic non-human primates for models of specific human genetic diseases. Primate embryonic stem cells will allow the generation of primate tissue or animal models for any human genetic disease for which the responsible gene has been cloned. The human genome project will identify an increasing number of genes related to human disease, but will not always provide insights into gene function. Transgenic nonhuman primates will be essential for elucidating mechanisms of disease and for testing new therapies. [0034]
  • (ii) Tissue transplantation. By manipulating culture conditions, primate ES cells, human and nonhuman, can be induced to differentiate to specific cell types, such as blood cells, neuron cells, or muscle cells. Alternatively, primate ES cells can be allowed to differentiate in tumors in SCID mice, the tumors can be disassociated, and the specific differentiated cell types of interest can be selected by the usage of lineage specific markers through the use of fluorescent activated cell sorting (FACS) or other sorting method or by direct microdissection of tissues of interest. These differentiated cells could then be transplanted back to the adult animal to treat specific diseases, such as hematopoietic disorders, endocrine deficiencies, degenerative neurological disorders or hair loss. [0035]
  • (b) Selection of Model Species [0036]
  • Macaques and marmosets were used as exemplary species for isolation of a primate ES cell line. Macaques, such as the rhesus monkey, are Old World species that are the major primates used in biomedical research. They are relatively large (about 7-10 kg). Males take 4-5 years to mature, and females have single young. Because of the extremely close anatomical and physiological similarities between humans and rhesus monkeys, rhesus monkey true ES cell lines provide a very accurate in vitro model for human differentiation. Rhesus monkey ES cell lines and rhesus monkeys will be particularly useful in the testing of the safety and efficacy of the transplantation of differentiated cell types into whole animals for the treatment of specific diseases or conditions. In addition, the techniques developed for the rhesus ES cell lines model the generation, characterization and manipulation of human ES cell lines. [0037]
  • The common marmoset ([0038] Callithrix jacchus) is a New World primate species with reproductive characteristics that make it an excellent choice for ES cell derivation. Marmosets are small (about 350-400 g), have a short gestation period (144 days), reach sexual maturity in about 18 months, and routinely have twins or triplets. Unlike in macaques, it is possible to routinely synchronize ovarian cycles in the marmoset with prostaglandin analogs, making collection of age-matched embryos from multiple females possible, and allowing efficient embryo transfer to synchronized recipients with 70%-80% of embryos transferred resulting in pregnancies. Because of these reproductive characteristics that allow for the routine efficient transfer of multiple embryos, marmosets provide an excellent primate species in which to generate transgenic models for human diseases.
  • There are approximately 200 primate species in the world. The most fundamental division that divides higher primates is between Old World and New world species. The evolutionary distance between the rhesus monkey and the common marmoset is far greater than the evolutionary distance between humans and rhesus monkeys. Because it is here demonstrated that it is possible to isolate ES cell lines from a representative species of both the Old World and New World group using similar conditions, the techniques described below may be used successfully in deriving ES cell lines in other higher primates as well. Given the close evolutionary distance between rhesus macaques and humans, and the fact that feeder-dependent human EC cell lines can be grown in conditions similar to those that support primate ES cell lines, the same growth conditions will allow the isolation and growth of human ES cells. In addition, human ES cell lines will be permanent cell lines that will also be distinguished from all other permanent human cell lines by their normal karyotype and the expression of the same combination of cell surface markers (alkaline phosphotase, preferably SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81) that characterize other primate ES cell lines. A normal karyotype and the expression of this combination of cell surface markers will be defining properties of true human ES cell lines, regardless of the method used for their isolation and regardless of their tissue of origin. [0039]
  • No other primate (human or non-human) ES cell line is known to exist. The only published permanent, euploid, embryo-derived cell lines that have been convincingly demonstrated to differentiate into derivatives of all three germ layers have been derived from rodents (the mouse, rat, and hamster), and possibly from rabbit. The published reports of embryo-derived cell lines from domestic species have failed to convincingly demonstrate differentiation of derivatives of all three embryonic germ layers or have not been permanent cell lines. Research groups in Britain and Singapore are informally reported, later than the work described here, to have attempted to derive human ES cell lines from surplus in vitro fertilization-produced human embryos, although they have not yet reported success in demonstrating pluripotency of their cells and have failed to isolate permanent cell lines. In the only published report on attempts to isolate human ES cells, conditions were used (LIF in the absence of fibroblast feeder layers) that the results below will indicate will not result in primate ES cells which can remain in an undifferentiated state. It is not surprising, then that the cells grown out of human ICMs failed to continue to proliferate after 1 or 2 subcultures, Bongso et al. [0040] Hum. Reprod. 9:2100-2117 (1994).
  • (2) Embryonic Stem Cell Isolation [0041]
  • A preferable medium for isolation of embryonic stem cells is “ES medium.” ES medium consists of 80% Dulbecco's modified Eagle's medium (DMEM; no pyruvate, high glucose formulation, Gibco BRL), with 20% fetal bovine serum (FBS; Hyclone), 0.1 mM β-mercaptoethanol (Sigma), 1% non-essential amino acid stock (Gibco BRL). Preferably, fetal bovine serum batches are compared by testing clonal plating efficiency of a low passage mouse ES cell line (ES[0042] jt3), a cell line developed just for the purpose of this test. FBS batches must be compared because it has been found that batches vary dramatically in their ability to support embryonic cell growth, but any other method of assaying the competence of FBS batches for support of embryonic cells will work as an alternative.
  • Primate ES cells are isolated on a confluent layer of murine embryonic fibroblast in the presence of ES cell medium. Embryonic fibroblasts are preferably obtained from 12 day old fetuses from outbred CF1 mice (SASCO), but other strains may be used as an alternative. Tissue culture dishes are preferably treated with 0.1% gelatin (type I; Sigma). [0043]
  • For rhesus monkey embryos, adult female rhesus monkeys (greater than four years old) demonstrating normal ovarian cycles are observed daily for evidence of menstrual bleeding ([0044] day 1 of cycle=the day of onset of menses). Blood samples are drawn daily during the follicular phase starting from day 8 of the menstrual cycle, and serum concentrations of luteinizing hormone are determined by radioimmunoassay. The female is paired with a male rhesus monkey of proven fertility from day 9 of the menstrual cycle until 48 hours after the luteinizing hormone surge; ovulation is taken as the day following the luteinizing hormone surge. Expanded blastocysts are collected by non-surgical uterine flushing at six days after ovulation. This procedure routinely results in the recovery of an average 0.4 to 0.6 viable embryos per rhesus monkey per month, Seshagiri et al. Am J Primatol 29:81-91, 1993.
  • For marmoset embryos, adult female marmosets (greater than two years of age) demonstrating regular ovarian cycles are maintained in family groups, with a fertile male and up to five progeny. Ovarian cycles are controlled by intramuscular injection of 0.75 g of the prostaglandin PGF2a analog cloprostenol (Estrumate, Mobay Corp, Shawnee, Kans.) during the middle to late luteal phase. Blood samples are drawn on day 0 (immediately before cloprostenol injection), and on [0045] days 3, 7, 9, 11, and 13. Plasma progesterone concentrations are determined by ELISA. The day of ovulation is taken as the day preceding a plasma progesterone concentration of 10 ng/ml or more. At eight days after ovulation, expanded blastocysts are recovered by a non-surgical uterine flush procedure, Thomson et al. “Non-surgical uterine stage preimplantation embryo collection from the common marmoset,” J Med Primatol, 23:333-336 (1994). This procedure results in the average production of 1.0 viable embryos per marmoset per month.
  • The zona pellucida is removed from blastocysts by brief exposure to pronase (Sigma). For immunosurgery, blastocysts are exposed to a 1:50 dilution of rabbit anti-marmoset spleen cell antiserum (for marmoset blastocysts) or a 1:50 dilution of rabbit anti-rhesus monkey (for rhesus monkey blastocysts) in DMEM for 30 minutes, then washed for 5 minutes three times in DMEM, then exposed to a 1:5 dilution of Guinea pig complement (Gibco) for 3 minutes. [0046]
  • After two further washes in DMEM, lysed trophectoderm cells are removed from the intact inner cell mass (ICM) by gentle pipetting, and the ICM plated on mouse inactivated (3000 rads gamma irradiation) embryonic fibroblasts. [0047]
  • After 7-21 days, ICM-derived masses are removed from endoderm outgrowths with a micropipette with direct observation under a stereo microscope, exposed to 0.05% Trypsin-EDTA (Gibco) supplemented with 1% chicken serum for 3-5 minutes and gently dissociated by gentle pipetting through a flame polished micropipette. [0048]
  • Dissociated cells are replated on embryonic feeder layers in fresh ES medium, and observed for colony formation. Colonies demonstrating ES-like morphology are individually selected, and split again as described above. The ES-like morphology is defined as compact colonies having a high nucleus to cytoplasm ratio and prominent nucleoli. Resulting ES cells are then routinely split by brief trypsinization or exposure to Dulbecco's Phosphate Buffered Saline (without calcium or magnesium and with 2 mM EDTA) every 1-2 weeks as the cultures become dense. Early passage cells are also frozen and stored in liquid nitrogen. [0049]
  • Cell lines may be karyotyped with a standard G-banding technique (such as by the Cytogenetics Laboratory of the University of Wisconsin State Hygiene Laboratory, which provides routine karyotyping services) and compared to published karyotypes for the primate species. [0050]
  • Isolation of ES cell lines from other primate species would follow a similar procedure, except that the rate of development to blastocyst can vary by a few days between species, and the rate of development of the cultured ICMs will vary between species. For example, six days after ovulation, rhesus monkey embryos are at the expanded blastocyst stage, whereas marmoset embryos don't reach the same stage until 7-8 days after ovulation. The Rhesus ES cell lines were obtained by splitting the ICM-derived cells for the first time at 7-16 days after immunosurgery; whereas the marmoset ES cells were derived with the initial split at 7-10 days after immunosurgery. Because other primates also vary in their developmental rate, the timing of embryo collection, and the timing of the initial ICM split will vary between primate species, but the same techniques and culture conditions will allow ES cell isolation. [0051]
  • Because ethical considerations in the U.S. do not allow the recovery of human in vivo fertilized preimplantation embryos from the uterus, human ES cells that are derived from preimplantation embryos will be derived from in vitro fertilized (IVF) embryos. Experiments on unused (spare) human IVF-produced embryos are allowed in many countries, such as Singapore and the United Kingdom, if the embryos are less than 14 days old. Only high quality embryos are suitable for ES isolation. Present defined culture conditions for culturing the one cell human embryo to the expanded blastocyst are suboptimal but practicable, Bongso et al., [0052] Hum Reprod 4:706-713, 1989. Co-culturing of human embryos with human oviductal cells results in the production of high blastocyst quality. IVF-derived expanded human blastocysts grown in cellular co-culture, or in improved defined medium, will allow the isolation of human ES cells with the same procedures described above for nonhuman primates.
  • (3) Defining Characteristics of Primate ES Cells [0053]
  • Primate embryonic stem cells share features with the primate ICM and with pluripotent human embryonal carcinoma cells. Putative primate ES cells may therefore be characterized by morphology and by the expression of cell surface markers characteristic of human EC cells. Additionally, putative primate ES cells may be characterized by developmental potential, karyotype and immortality. [0054]
  • (a) Morphology [0055]
  • The colony morphology of primate embryonic stem cell lines is similar to, but distinct from, mouse embryonic stem cells. Both mouse and primate ES cells have the characteristic features of undifferentiated stem cells, with high nuclear/cytoplasmic ratios, prominent nucleoli, and compact colony formation. The colonies of primate ES cells are flatter than mouse ES cell colonies and individual primate ES cells can be easily distinguished. In FIG. 2, reference character A indicates a phase contrast photomicrograph of cell line R278.5 demonstrating the characteristic primate ES cell morphology. [0056]
  • (b) Cell Surface Markers [0057]
  • A primate ES cell line of the present invention is distinct from mouse ES cell lines by the presence or absence of the cell surface markers described below. [0058]
  • One set of glycolipid cell surface markers is known as the Stage-specific [0059] embryonic antigens 1 through 4. These antigens can be identified using antibodies for SSEA 1, preferably SSEA-3 and SSEA-4 which are available from the Developmental Studies Hybridoma Bank of the National Institute of Child Health and Human Development. The cell surface markers referred to as TRA-1-60 and TRA-1-81 designate antibodies from hybridomas developed by Peter Andrews of the University of Sheffield and are described in Andrews et al., “Cell lines from human germ cell tumors,” In: Robertson E, ed. Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. Oxford: IRL Press, 207-246, 1987. The antibodies were localized with a biotinylated secondary antibody and then an avidin/biotinylated horseradish peroxidase complex (Vectastain ABC System, Vector Laboratories). Alternatively, it should also be understood that other antibodies for these same cell surface markers can be generated. NTERA-2 cl. D1, a pluripotent human EC cell line (gift of Peter Andrews), may be used as a negative control for SSEA-1, and as a positive control for SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81. This cell line was chosen for positive control only because it has been extensively studied and reported in the literature, but other human EC cell lines may be used as well.
  • Mouse ES cells (ES[0060] jt3) are used as a positive control for SSEA-1, and for a negative control for SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81. Other routine negative controls include omission of the primary or secondary antibody and substitution of a primary antibody with an unrelated specificity.
  • Alkaline phosphatase may be detected following fixation of cells with 4% para-formaldehyde using “Vector Red” (Vector Laboratories) as a substrate, as described by the manufacturer (Vector Laboratories). The precipitate formed by this substrate is red when viewed with a rhodamine filter system, providing substantial amplification over light microscopy. [0061]
  • Table 1 diagrams a comparison of mouse ES cells, primate ES cells, and human EC cells. The only cells reported to express the combination of markers SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 other than primate ES cells are human EC cells. The globo-series glycolipids SSEA-3 and SSEA-4 are consistently present on human EC cells, and are of diagnostic value in distinguishing human EC cell tumors from human yolk sac carcinomas, choriocarcinomas, and other lineages which lack these markers, Wenk et al., [0062] Int J Cancer 58:108-115, 1994. A recent survey found SSEA-3 and SSEA-4 to be present on all of over 40 human EC cell lines examined, Wenk et al. TRA-1-60 and TRA-1-81 antigens have been studied extensively on a particular pluripotent human EC cell line, NTERA-2 CL. D1, Andrews et al, supra. Differentiation of NTERA-2 CL. D1 cells in vitro results in the loss of SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 expression and the increased expression of the lacto-series glycolipid SSEA-1, Andrews et al, supra. This contrasts with undifferentiated mouse ES cells, which express SSEA-1, and neither SSEA-3 nor SSEA-4. Although the function of these antigens are unknown, their shared expression by R278.5 cells and human EC cells suggests a close embryological similarity. Alkaline phosphatase will also be present on all primate ES cells. A successful primate ES cell culture of the present invention will correlate with the cell surface markers found in the rhesus macaque and marmoset cell lines described in Table 1.
  • As disclosed below in Table 1, the rhesus macaque and marmoset cell lines are identical to human EC cell lines for the 5 described markers. Therefore, a successful primate ES cell culture will also mimic human EC cells. However, there are other ways to discriminate ES cells from EC cells. For example, the primate ES cell line has a normal karyotype and the human EC cell line is aneuploid. [0063]
  • In FIG. 3, the photographs labelled A through F demonstrate the characteristic staining of these markers on a rhesus monkey ES cell line designated R278.5. [0064]
    TABLE 1
    Mouse C. jacchus M. mulatta Human EC
    ES ES ES (NTERA-2 cl.D1)
    SSEA-1 +
    SSEA-3 + + +
    SSEA-4 + + +
    Tra-1-60 + + +
    Tra-1-81 + + +
  • (c) Developmental Potential [0065]
  • Primate ES cells of the present invention are pluripotent. By “pluripotent” we mean that the cell has the ability to develop into any cell derived from the three main germ cell layers or an embryo itself. When injected into SCID mice, a successful primate ES cell line will differentiate into cells derived from all three embryonic germ layers including: bone, cartilage, smooth muscle, striated muscle, and hematopoietic cells (mesoderm); liver, primitive gut and respiratory epithelium (endoderm); neurons, glial cells, hair follicles, and tooth buds (ectoderm). [0066]
  • This experiment can be accomplished by injecting approximately 0.5-1.0×10[0067] 6 primate ES cells into the rear leg muscles of 8-12 week old male SCID mice. The resulting tumors can be fixed in 4% paraformaldehyde and examined histologically after paraffin embedding at 8-16 weeks of development. In FIG. 4, photomicrographs designated A-F are of sections of tumors formed by injection of rhesus ES cells into the hind leg muscles of SCID mice and analyzed 15 weeks later demonstrating cartilage, smooth muscle, and striated muscle (mesoderm); stratified squamous epithelium with hair follicles, neural tube with ventricular, intermediate, and mantle layers (ectoderm); ciliated columnar epithelium and villi lined by absorptive enterocytes and mucus-secreting goblet cells (endoderm).
  • A successful nonhuman primate ES cell line will have the ability to participate in normal development when combined in chimeras with normal preimplantation embryos. Chimeras between preimplantation nonhuman primate embryos and nonhuman primate ES cells can be formed by routine methods in several ways. (i) injection chimeras: 10-15 nonhuman primate ES cells can be microinjected into the cavity of an expanded nonhuman primate blastocyst; (ii) aggregation chimeras: nonhuman primate morulae can be co-cultured on a lawn of nonhuman primate ES cells and allowed to aggregate; and (iii) tetraploid chimeras: 10-15 nonhuman primate ES cells can be aggregated with tetraploid nonhuman primate morulae obtained by electrofusion of 2-cell embryos, or incubation of morulae in the cytoskeletal inhibitor cholchicine. The chimeras can be returned to the uterus of a female nonhuman primate and allowed to develop to term, and the ES cells will contribute to normal differentiated tissues derived from all three embryonic germ layers and to germ cells. Because nonhuman primate ES can be genetically manipulated prior to chimera formation by standard techniques, chimera formation followed by embryo transfer can lead to the production of transgenic nonhuman primates. [0068]
  • (d) Karyotype [0069]
  • Successful primate ES cell lines have normal karyotypes. Both XX and XY cells lines will be derived. The normal karyotypes in primate ES cell lines will be in contrast to the abnormal karyotype found in human embryonal carcinoma (EC), which are derived from spontaneously arising human germ cell tumors (teratocarcinomas). Human embryonal carcinoma cells have a limited ability to differentiate into multiple cell types and represent the closest existing cell lines to primate ES cells. Although tumor-derived human embryonal carcinoma cell lines have some properties in common with embryonic stem cell lines, all human embryonal carcinoma cell lines derived to date are aneuploid. Thus, primate ES cell lines and human EC cell lines can be distinguished by the normal karyotypes found in primate ES cell lines and the abnormal karyotypes found in human EC lines. By “normal karyotype” it is meant that all chromosomes normally characteristic of the species are present and have not been noticeably altered. [0070]
  • Because of the abnormal karyotypes of human embryonal carcinoma cells, it is not clear how accurately their differentiation reflects normal differentiation. The range of embryonic and extraembryonic differentiation observed with primate ES cells will typically exceed that observed in any human embryonal carcinoma cell line, and the normal karyotypes of the primate ES cells suggests that this differentiation accurately recapitulates normal differentiation. [0071]
  • (e) Immortality [0072]
  • Immortal cells are capable of continuous indefinite replication in vitro. Continued proliferation for longer than one year of culture is a sufficient evidence for immortality, as primary cell cultures without this property fail to continuously divide for this length of time (Freshney, [0073] Culture of animal cells. New York: Wiley-Liss, 1994). Primate ES cells will continue to proliferate in vitro with the culture conditions described above for longer than one year, and will maintain the developmental potential to contribute all three embryonic germ layers. This developmental potential can be demonstrated by the injection of ES cells that have been cultured for a prolonged period (over a year) into SCID mice and then histologically examining the resulting tumors. Although karyotypic changes can occur randomly with prolonged culture, some primate ES cells will maintain a normal karyotype for longer than a year of continuous culture.
  • (f) Culture Conditions [0074]
  • Growth factor requirements to prevent differentiation are different for the primate ES cell line of the present invention than the requirements for mouse ES cell lines. In the absence of fibroblast feeder layers, Leukemia inhibitory factor (LIF) is necessary and sufficient to prevent differentiation of mouse ES cells and to allow their continuous passage. Large concentrations of cloned LIF fail to prevent differentiation of primate ES cell lines in the absence of fibroblast feeder layers. In this regard, primate ES stem cells are again more similar to human EC cells than to mouse ES cells, as the growth of feeder-dependent human EC cells lines is not supported by LIF in the absence of fibroblasts. [0075]
  • (g) Differentiation to Extra Embryonic Tissues [0076]
  • When grown on embryonic fibroblasts and allowed to grow for two weeks after achieving confluence (i.e., continuously covering the culture surface), primate ES cells of the present invention spontaneously differentiate and will produce chorionic gonadotropin, indicating trophoblast differentiation (a component of the placenta) and produce α-fetoprotein, indicating endoderm differentiation. Chorionic gonadotropin activity can be assayed in the medium conditioned by differentiated cells by Leydig cell bioassay, Seshagiri & Hearn, [0077] Hum Reprod 8:279-287, 1992. For mRNA analysis, RNA can be prepared by guanidine isothiocyanate-phenol/chloroform extraction (1) from approximately 0.2×106 differentiated cells and from 0.2×106 undifferentiated cells. The relative levels of the mRNA for α-fetoprotein and the α- and β-subunit of chorionic gonadotropin relative to glyceraldehyde-3-phosphate dehydrogenase can be determined by semi-quantitative Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR). The PCR primers for glyceraldehyde 3-phosphate dehydrogenase (G3PDH), obtained from Clontech (Palo Alto, Calif.), are based on the human cDNA sequence, and do not amplify mouse G3PDH mRNA under our conditions. Primers for the α-fetoprotein mRNA are based on the human sequence and flank the 7th intron (5′ primer=(5′) GCTGGATTGTCTGCAGGATGGGGAA (SEQ ID NO: 1); 3′ primer=(5′) TCCCCTGAAGAAAATTGGTTAAAAT (SEQ ID NO: 2)). They amplify a cDNA of 216 nucleotides. Primers for the β-subunit of chorionic gonadotropin flank the second intron (5′ primer=(5′) ggatc CACCGTCAACACCACCATCTGTGC (SEQ ID NO: 3); 3′ primer=(5′) ggatc CACAGGTCAAAGGGTGGTCCTTGGG (SEQ ID NO: 4)) (nucleotides added to the hCGb sequence to facilitate sub-cloning are shown in lower case italics). They amplify a cDNA of 262 base pairs. The primers for the CGα subunit can be based on sequences of the first and fourth exon of the rhesus gene (5′ primer=(5′) gggaattc GCAGTTACTGAGAACTCACAAG (SEQ ID NO: 5); 3′ primer=(5′) gggaattc GAAGCATGTCAAAGTGGTATGG (SEQ ID NO: 6)) and amplify a cDNA of 556 base pairs. The identity of the α-fetoprotein, CGα and CGβ cDNAs can be verified by subcloning and sequencing.
  • For Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR), 1 to 5 μl of total R278.5 RNA can be reverse transcribed as described Golos et al. [0078] Endocrinology 133(4):1744-1752, 1993, and one to 20 μl of reverse transcription reaction was then subjected to the polymerase chain reaction in a mixture containing 1-12.5 pmol of each G3PDH primer, 10-25 pmol of each mRNA specific primer, 0.25 mM dNTPs (Pharmacia, Piscataway, N.J.), 1X AmpliTaq buffer (final reaction concentrations=10 mM Tris, pH 8.3, 50 mM KCl, 1.5 mM MgCl2, 0.001% (w/v) gelatin) 2.5 μCi of deoxycytidine 5′a[32P]triphosphate (DuPont, Boston, Mass.), 10% glycerol and 1.25 U of AmpliTaq (Perkin-Elmer, Oak Brook, Ill.) in a total volume of 50 μl. The number of amplification rounds which produced linear increases in target cDNAs and the relation between input RNA and amount of PCR product is empirically determined as by Golos et al. Samples were fractionated in 3% Nusieve (FMC, Rockland, Me.) agarose gels (1X TBE running buffer) and DNA bands of interest were cut out, melted at 65° C. in 0.5 ml TE, and radioactivity determined by liquid scintillation counting. The ratio of counts per minute in a specific PCR product relative to cpm of G3PDH PCR product is used to estimate the relative levels of a mRNAs among differentiated and undifferentiated cells.
  • The ability to differentiate into trophectoderm in vitro and the ability of these differentiated cells to produce chorionic gonadotropin distinguishes the primate ES cell line of the present invention from all other published ES cell lines. [0079]
  • EXAMPLES
  • (1) Animals and Embryos [0080]
  • As described above, we have developed a technique for non-surgical, uterine-stage embryo recovery from the rhesus macaque and the common marmoset. [0081]
  • To supply rhesus embryos to interested investigators, The Wisconsin Regional Primate Research Center (WRPRC) provides a preimplantation embryo recovery service for the rhesus monkey, using the non-surgical flush procedure described above. During 1994, 151 uterine flushes were attempted from rhesus monkeys, yielding 80 viable embryos (0.53 embryos per flush attempt). [0082]
  • By synchronizing the reproductive cycles of several marmosets, significant numbers of in vivo produced, age-matched, preimplantation primate embryos were studied in controlled experiments for the first time. Using marmosets from the self-sustaining colony (250 animals) of the Wisconsin Regional Primate Research Center (WRPRC), we recovered 54 viable morulae or blastocysts, 7 unfertilized oocytes or degenerate embryos, and 5 empty zonae pellucidae in a total of 54 flush attempts (1.0 viable embryo-flush attempt). Marmosets have a 28 day ovarian cycle, and because this is a non-surgical procedure, females can be flushed on consecutive months, dramatically increasing the embryo yield compared to surgical techniques which require months of rest between collections. [0083]
  • (2) Rhesus Macaque Embryonic Stem Cells [0084]
  • Using the techniques described above, we have derived three independent embryonic stem cell lines from two rhesus monkey blastocysts (R278.5, R366, and R367). One of these, R278.5, remains undifferentiated and continues to proliferate after continuous culture for over one year. R278.5 cells have also been frozen and successfully thawed with the recovery of viable cells. [0085]
  • The morphology and cell surface markers of R278.5 cells are indistinguishable from human EC cells, and differ significantly from mouse ES cells. R278.5 cells have a high nucleus/cytoplasm ratio and prominent nucleoli, but rather than forming compact, piled-up colonies with indistinct cell borders similar to mouse ES cells, R278.5 cells form flatter colonies with individual, distinct cells (FIG. 2 A). R278.5 cells express the SSEA-3, SSEA-4, TRA-1-60, and TRA-81 antigens (FIG. 3 and Table 1), none of which are expressed by mouse ES cells. The only cells known to express the combination of markers SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 other than primate ES cells are human EC cells. The globo-series glycolipids SSEA-3 and SSEA-4 are consistently present on human EC cells, and are of diagnostic value in distinguishing human EC cell tumors from yolk sac carcinomas, choriocarcinomas and other stem cells derived from human germ cell tumors which lack these markers, Wenk et al, [0086] Int J Cancer 58:108-115, 1994. A recent survey found SSEA-3 and SSEA-4 to be present on all of over 40 human EC cell lines examined (Wenk et al.).
  • TRA-1-60 and TRA-1-81 antigens have been studied extensively on a particular pluripotent human EC cell line, NTERA-2 CL. D1 (Andrews et al.). Differentiation of NTERA-2 CL. D1 cells in vitro results in the loss of SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81 expression and the increased expression of the lacto-series glycolipid SSEA-1. Undifferentiated mouse ES cells, on the other hand, express SSEA-1, and not SSEA-3, SSEA-4, TRA-1-60 or TRA-1-81 (Wenk et al.). Although the function of these antigens is unknown, their expression by R278.5 cells suggests a close embryological similarity between primate ES cells and human EC cells, and fundamental differences between primate ES cells and mouse ES cells. [0087]
  • R278.5 cells also express alkaline phosphatase. The expression of alkaline phosphatase is shared by both primate and mouse ES cells, and relatively few embryonic cells express this enzyme. Positive cells include the ICM and primitive ectoderm (which are the most similar embryonic cells in the intact embryo to ES cells), germ cells, (which are totipotent), and a very limited number of neural precursors, Kaufman M H. [0088] The atlas of mouse development. London: Academic Press, 1992. Cells not expressing this enzyme will not be primate ES cells.
  • Although cloned human LIF was present in the medium at cell line derivation and for initial passages, R278.5 cells grown on mouse embryonic fibroblasts without exogenous LIF remain undifferentiated and continued to proliferate. R278.5 cells plated on gelatin-treated tissue culture plates without fibroblasts differentiated to multiple cell types or failed to attach and died, regardless of the presence or absence of exogenously added human LIF (FIG. 2). Up to 10[0089] 4 units/ml human LIF fails to prevent differentiation. In addition, added LIF fails to increase the cloning efficiency or proliferation rate of R278.5 cells on fibroblasts. Since the derivation of the R278.5 cell line, we have derived two additional rhesus ES cell lines (R366 and R367) on embryonic fibroblasts without any exogenously added LIF at initial derivation. R366 and R367 cells, like R278.5 cells, continue to proliferate on embryonic fibroblasts without exogenously added LIF and differentiate in the absence of fibroblasts, regardless of the presence of added LIF. RT-PCR performed on mRNA from spontaneously differentiated R278.5 cells revealed α-fetoprotein mRNA (FIG. 4). α-fetoprotein is a specific marker for endoderm, and is expressed by both extraembryonic (yolk sac) and embryonic (fetal liver and intestines) endoderm-derived tissues. Epithelial cells resembling extraembryonic endoderm are present in cells differentiated in vitro from R278.5 cells (FIG. 2). Bioactive CG (3.89 mI units/ml) was present in culture medium collected from differentiated cells, but not in medium collected from undifferentiated cells (less than 0.03 mI units/ml), indicating the differentiation of trophoblast, a trophectoderm derivative. The relative level of the CGα mRNA increased 23.9-fold after differentiation (FIG. 4).
  • All SCID mice injected with R278.5 cells in either intra-muscular or intra-testicular sites formed tumors, and tumors in both sites demonstrated a similar range of differentiation. The oldest tumors examined (15 weeks) had the most advanced differentiation, and all had abundant, unambiguous derivatives of all three embryonic germ layers, including gut and respiratory epithelium (endoderm); bone, cartilage, smooth muscle, striated muscle (mesoderm); ganglia, glia, neural precursors, and stratified squamous epithelium (ectoderm), and other unidentified cell types (FIG. 5). In addition to individual cell types, there was organized development of some structures which require complex interactions between different cell types. Such structures included gut lined by villi with both absorptive enterocytes and mucus-secreting goblet cells, and sometimes encircled by layers of smooth muscle in the same orientation as muscularis mucosae (circular) and muscularis (outer longitudinal layer and inner circular layer); neural tubes with ventricular, intermediate, and mantle layers; and hair follicles with hair shafts (FIG. 5). [0090]
  • The essential characteristics that define R278.5 cells as ES cells include: indefinite (greater than one year) undifferentiated proliferation in vitro, normal karyotype, and potential to differentiate to derivatives of trophectoderm and all three embryonic germ layers. In the mouse embryo, the last cells capable of contributing to derivatives of both trophectoderm and ICM are early ICM cells. The timing of commitment to ICM or trophectoderm has not been established for any primate species, but the potential of rhesus ES cells to contribute to derivatives of both suggests that they most closely resemble early totipotent embryonic cells. The ability of rhesus ES cells to form trophoblast in vitro distinguishes primate ES cell lines from mouse ES cells. Mouse ES cell have not been demonstrated to form trophoblast in vitro, and mouse trophoblast does not produce gonadotropin. Rhesus ES cells and mouse ES cells do demonstrate the similar wide range of differentiation in tumors that distinguishes ES cells from EC cells. The development of structures composed of multiple cell types such as hair follicles, which require inductive interactions between the embryonic epidermis and underlying mesenchyme, demonstrates the ability of rhesus ES cells to participate in complex developmental processes. [0091]
  • The rhesus ES lines R366 and R367 have also been further cultured and analyzed. Both lines have a normal XY karyotype and were proliferated in an undifferentiated state for about three months prior to freezing for later analysis. Samples of each of the cell lines R366 and R367 were injected into SCID mice which then formed teratomas identical to those formed by R278.5 cells. An additional rhesus cell line R394 having a normal XX karyotype was also recovered. All three of these cell lines, R366, R367 and R394 are identical in morphology, growth characteristics, culture requirements and in vitro differentiation characteristics, i.e. the trait of differentiation to multiple cell types in the absence of fibroblasts, to cell line 278.5. [0092]
  • It has been determined that LIF is not required either to derive or proliferate these ES cultures. Each of the cell lines R366, R367 and R394 were derived and cultured without exogenous LIF. [0093]
  • It has also been demonstrated that the particular source of fibroblasts for co-culture is not critical. Several fibroblast cell lines have been tested both with rhesus line R278.5 and with the marmoset cell lines described below. The fibroblasts tested include mouse STO cells (ATCC 56-X), mouse 3T3 cells (ATCC 48-X), primary rhesus monkey embryonic fibroblasts derived from 36 day rhesus fetuses, and mouse Sl/Sl[0094] 4 cells, which are deficient in the steel factor. All these fibroblast cell lines were capable of maintaining the stem cell lines in an undifferentiated state. Most rapid proliferation of the stem cells was observed using primary mouse embryonic fibroblasts.
  • Unlike mouse ES cells, neither rhesus ES cells nor feeder-dependent human EC cells remain undifferentiated and proliferate in the presence of soluble human LIF without fibroblasts. The factors that fibroblasts produce that prevent the differentiation of rhesus ES cells or feeder-dependent human EC cells are unknown, but the lack of a dependence on LIF is another characteristic that distinguishes primate ES cells from mouse ES cells. The growth of rhesus monkey ES cells in culture conditions similar to those required by feeder-dependent human EC cells, and the identical morphology and cell surface markers of rhesus ES cells and human EC cells, suggests that similar culture conditions will support human ES cells. [0095]
  • Rhesus ES cells will be important for elucidating the mechanisms that control the differentiation of specific primate cell types. Given the close evolutionary distance and the developmental and physiological similarities between humans and rhesus monkeys, the mechanisms controlling the differentiation of rhesus cells will be very similar to the mechanisms controlling the differentiation of human cells. The importance of elucidating these mechanisms is that once they are understood, it will be possible to direct primate ES cells to differentiate to specific cell types in vitro, and these specific cell types can be used for transplantation to treat specific diseases. [0096]
  • Because ES cells have the developmental potential to give rise to any differentiated cell type, any disease that results in part or in whole from the failure (either genetic or acquired) of specific cell types will be potentially treatable through the transplantation of cells derived from ES cells. Rhesus ES cells and rhesus monkeys will be invaluable for testing the efficacy and safety of the transplantation of specific cell types derived from ES cells. A few examples of human diseases potentially treatable by this approach with human ES cells include degenerative neurological disorders such as Parkinson's disease (dopanergic neurons), juvenile onset diabetes (pancreatic β-islet cells) or Acquired Immunodeficiency Disease (lymphocytes). Because undifferentiated ES cells can proliferate indefinitely in vitro, they can be genetically manipulated with standard techniques either to prevent immune rejection after transplantation, or to give them new genetic properties to combat specific diseases. For specific cell types where immune rejection can be prevented, cells derived from rhesus monkey ES cells or other non-human primate ES cells could be used for transplantation to humans to treat specific diseases. [0097]
  • (3) Marmoset Embryonic Stem Cells [0098]
  • Our method for creating an embryonic stem cell line is described above. Using isolated ICM's derived by immunosurgery from marmoset blastocysts, we have isolated 7 putative ES cell lines, each of which have been cultured for over 6 months. [0099]
  • One of these, Cj11, was cultured continuously for over 14 months, and then frozen for later analysis. The Cj11 cell line and other marmoset ES cell lines have been successfully frozen and then thawed with the recovery of viable cells. These cells have a high nuclear/cytoplasmic ratio, prominent nucleoli, and a compact colony morphology similar to the pluripotent human embryonal carcinoma (EC) cell line NT2/D2. [0100]
  • Four of the cell lines we have isolated have normal XX karyotypes, and one has a normal XY karyotype (Karyotypes were performed by Dr. Charles Harris, University of Wisconsin). These cells were positive for a series of cell surface markers (alkaline phosphatase, SSEA-3, SSEA-4, TRA-1-60, and TRA-1-81) that in combination are definitive markers for undifferentiated human embryonal carcinoma cells (EC) cells and primate ES cells. In particular, these markers distinguish EC cells from the earliest lineages to differentiate in the human preimplantation embryo, trophectoderm (represented by BeWO choriocarcinoma cells) and extraembryonic endoderm (represented by 1411H yolk sac carcinoma cells). [0101]
  • When the putative marmoset ES cells were removed from fibroblast feeders, they differentiated into cells of several distinct morphologies. Among the differentiated cells, trophectoderm is indicated by the secretion of chorionic gonadotropin and the presence of the chorionic gonadotropin β-subunit mRNA. 12.7 mIU/ml luteinizing hormone (LH) activity was measured in the WRPRC core assay lab using a mouse Leydig cell bioassay in medium conditioned 24 hours by putative ES cells allowed to differentiate for one week. Note that chorionic gonadotrophin has both LH and FSH activity, and is routinely measured by LH assays. Control medium from undifferentiated ES cells had less than 1 mIU/ml LH activity. [0102]
  • Chorionic gonadotropin β-subunit mRNA was detected by reverse transcriptase-polymerase chain reaction (RT-PCR). DNA sequencing confirmed the identity of the chorionic gonadotrophin β-subunit. [0103]
  • Endoderm differentiation (probably extraembryonic endoderm) was indicated by the presence of α-fetoprotein mRNA, detected by RT-PCR. [0104]
  • When the marmoset ES cells were grown in high densities, over a period of weeks epithelial cells differentiated and covered the culture dish. The remaining groups of undifferentiated cells rounded up into compact balls and then formed embryoid bodies (as shown in FIG. 6) that recapitulated early development with remarkable fidelity. Over 3-4 weeks, some of the embryoid bodies formed a bilaterally symmetric pyriform embryonic disc, an amnion, a yolk sac, and a mesoblast outgrowth attaching the caudal pole of the amnion to the culture dish. [0105]
  • Histological and ultrastructural examination of one of these embryoid bodies (formed from a cell line that had been passaged continuously for 6 months) revealed a remarkable resemblance to a stage 6-7 post-implantation embryo. The embryonic disc was composed of a polarized, columnar epithelial epiblast (primitive ectoderm) layer separated from a visceral endoderm (primitive endoderm) layer. Electron microscopy of the epiblast revealed apical junctional complexes, apical microvilli, subapical intermediate filaments, and a basement membrane separating the epiblast from underlying visceral endoderm. All of these elements are features of the normal embryonic disc. In the caudal third of the embryonic disc, there was a midline groove, disruption of the basement membrane, and mixing of epiblast cells with underlying endodermal cells (early primitive streak). The amnion was composed of an inner squamous (ectoderm) layer continuous with the epiblast and an outer mesoderm layer. The bilayered yolk sac had occasional endothelial-lined spaces containing possible hematopoietic precursors. [0106]
  • The morphology, immortality, karyotype, and cell surface markers of these marmoset cells identify these marmoset cells as primate ES cells similar to the rhesus ES cells. Since the last cells in the mammalian embryo capable of contributing to both trophectoderm derivatives and endoderm derivatives are the totipotent cells of the early ICM, the ability of marmoset ES cells to contribute to both trophoblast and endoderm demonstrates their similarities to early totipotent embryonic cells of the intact embryo. The formation of embryoid bodies by marmoset ES cells, with remarkable structural similarities to the early post-implantation primate embryo, demonstrates the potential of marmoset ES cells to participate in complex developmental processes requiring the interaction of multiple cell types. [0107]
  • Given the reproductive characteristics of the common marmoset described above (efficient embryo transfer, multiple young, short generation time), marmoset ES cells will be particularly useful for the generation of transgenic primates. Although mice have provided invaluable insights into gene function and regulation, the anatomical and physiological differences between humans and mice limit the usefulness of transgenic mouse models of human diseases. Transgenic primates, in addition to providing insights into the pathogenesis of specific diseases, will provide accurate animal models to test the efficacy and safety of specific treatments. [0108]
  • 1 6 25 base pairs nucleic acid double linear DNA (genomic) not provided 1 GCTGGATTGT CTGCAGGATG GGGAA 25 25 base pairs nucleic acid double linear DNA (genomic) not provided 2 TCCCCTGAAG AAAATTGGTT AAAAT 25 29 base pairs nucleic acid double linear DNA (genomic) not provided 3 GGATCCACCG TCAACACCAC CATCTGTGC 29 30 base pairs nucleic acid double linear DNA (genomic) not provided 4 GGATCCACAG GTCAAAGGGT GGTCCTTGGG 30 30 base pairs nucleic acid double linear DNA (genomic) not provided 5 GGGAATTCGC AGTTACTGAG AACTCACAAG 30 30 base pairs nucleic acid double linear DNA (genomic) not provided 6 GGGAATTCGA AGCATGTCAA AGTGGTATGG 30

Claims (11)

I claim:
1. A purified preparation of human embryonic stem cells which (i) is capable of proliferation in an in vitro culture for over one year, (ii) maintains a karyotype in which all the chromosomes characteristic of the human species are present and not noticeably altered through prolonged culture, (iii) maintains the potential to differentiate to derivatives of endoderm, mesoderm, and ectoderm tissues throughout the culture, and (iv) are inhibited from differentiation when cultured on a fibroblast feeder layer.
2. The preparation of
claim 1
, wherein the stem cells will spontaneously differentiate to trophoblast and produce chorionic gonadotropin when cultured to high density.
3. A purified preparation of human embryonic stem cells wherein the cells are essentially negative for the SSEA-1 marker, positive for the SSEA-4 marker, express alkaline phosphatase activity, are pluripotent, and have karyotypes which includes the presence of all of the chromosomes characteristic of the human species and in which none of the chromosomes are noticeably altered.
4. The preparation of
claim 3
, wherein the cells are positive for the TRA-1-60, and TRA-1-81 markers.
5. The preparation of
claim 3
, wherein the cells continue to proliferate in an undifferentiated state after continuous culture for at least one year.
6. The preparation of
claim 3
, wherein the cells will differentiate to trophoblast when cultured beyond confluence and will produce chorionic gonadotropin.
7. The preparation of
claim 3
, wherein the cells remain euploid for more than one year of continuous culture.
8. The preparation of
claim 3
, wherein the cells differentiate into cells derived from mesoderm, endoderm and ectoderm germ layers when the cells are injected into a SCID mouse.
9. A method of isolating a human embryonic stem cell line, comprising the steps of:
(a) isolating a human blastocyst;
(b) isolating cells from the inner cell mass of the blastocyte of (a);
(c) plating the inner cell mass cells on embryonic fibroblasts, wherein inner cell mass-derived cell masses are formed;
(d) dissociating the mass into dissociated cells;
(e) replating the dissociated cells on embryonic feeder cells;
(f) selecting colonies with compact morphologies and cells with high nucleus to cytoplasm ratios and prominent nucleoli; and
(g) culturing the cells of the selected colonies.
10. A method as claimed in
claim 9
, further comprising maintaining the isolated cells on a fibroblast feeder layer to prevent differentiation.
11. A cell line developed by the method of step 9.
US09/761,289 1995-01-20 2001-01-16 Primate embryonic stem cells Abandoned US20010024825A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US09/761,289 US20010024825A1 (en) 1995-01-20 2001-01-16 Primate embryonic stem cells
US09/982,637 US7029913B2 (en) 1995-01-20 2001-10-18 Primate embryonic stem cells
US10/430,496 US20060040383A1 (en) 1995-01-20 2003-05-06 Primate embryonic stem cells
US11/033,335 US20050158854A1 (en) 1995-01-20 2005-01-11 Primate embryonic stem cells
US11/036,245 US7582479B2 (en) 1995-01-20 2005-01-14 Primate embryonic stem cell line
US12/047,135 US7781216B2 (en) 1995-01-20 2008-03-12 Spontaneous differentiation of human embryonic stem cells in culture
US12/822,004 US8273569B2 (en) 1995-01-20 2010-06-23 Preparation of human embryonic stem cells
US13/595,587 US20120328582A1 (en) 1995-01-20 2012-08-27 Primate Embryonic Stem Cells
US14/281,341 US20150056698A1 (en) 1995-01-20 2014-05-19 Primate embryonic stem cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US37632795A 1995-01-20 1995-01-20
US08/591,246 US5843780A (en) 1995-01-20 1996-01-18 Primate embryonic stem cells
US09/106,390 US6200806B1 (en) 1995-01-20 1998-06-26 Primate embryonic stem cells
US09/761,289 US20010024825A1 (en) 1995-01-20 2001-01-16 Primate embryonic stem cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/106,390 Continuation US6200806B1 (en) 1995-01-20 1998-06-26 Primate embryonic stem cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US09/982,637 Continuation US7029913B2 (en) 1995-01-20 2001-10-18 Primate embryonic stem cells

Publications (1)

Publication Number Publication Date
US20010024825A1 true US20010024825A1 (en) 2001-09-27

Family

ID=23484566

Family Applications (11)

Application Number Title Priority Date Filing Date
US08/591,246 Expired - Lifetime US5843780A (en) 1995-01-20 1996-01-18 Primate embryonic stem cells
US09/106,390 Expired - Lifetime US6200806B1 (en) 1995-01-20 1998-06-26 Primate embryonic stem cells
US09/761,289 Abandoned US20010024825A1 (en) 1995-01-20 2001-01-16 Primate embryonic stem cells
US09/982,637 Expired - Fee Related US7029913B2 (en) 1995-01-20 2001-10-18 Primate embryonic stem cells
US10/430,496 Abandoned US20060040383A1 (en) 1995-01-20 2003-05-06 Primate embryonic stem cells
US11/033,335 Abandoned US20050158854A1 (en) 1995-01-20 2005-01-11 Primate embryonic stem cells
US11/036,245 Expired - Fee Related US7582479B2 (en) 1995-01-20 2005-01-14 Primate embryonic stem cell line
US12/047,135 Expired - Fee Related US7781216B2 (en) 1995-01-20 2008-03-12 Spontaneous differentiation of human embryonic stem cells in culture
US12/822,004 Expired - Fee Related US8273569B2 (en) 1995-01-20 2010-06-23 Preparation of human embryonic stem cells
US13/595,587 Abandoned US20120328582A1 (en) 1995-01-20 2012-08-27 Primate Embryonic Stem Cells
US14/281,341 Abandoned US20150056698A1 (en) 1995-01-20 2014-05-19 Primate embryonic stem cells

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/591,246 Expired - Lifetime US5843780A (en) 1995-01-20 1996-01-18 Primate embryonic stem cells
US09/106,390 Expired - Lifetime US6200806B1 (en) 1995-01-20 1998-06-26 Primate embryonic stem cells

Family Applications After (8)

Application Number Title Priority Date Filing Date
US09/982,637 Expired - Fee Related US7029913B2 (en) 1995-01-20 2001-10-18 Primate embryonic stem cells
US10/430,496 Abandoned US20060040383A1 (en) 1995-01-20 2003-05-06 Primate embryonic stem cells
US11/033,335 Abandoned US20050158854A1 (en) 1995-01-20 2005-01-11 Primate embryonic stem cells
US11/036,245 Expired - Fee Related US7582479B2 (en) 1995-01-20 2005-01-14 Primate embryonic stem cell line
US12/047,135 Expired - Fee Related US7781216B2 (en) 1995-01-20 2008-03-12 Spontaneous differentiation of human embryonic stem cells in culture
US12/822,004 Expired - Fee Related US8273569B2 (en) 1995-01-20 2010-06-23 Preparation of human embryonic stem cells
US13/595,587 Abandoned US20120328582A1 (en) 1995-01-20 2012-08-27 Primate Embryonic Stem Cells
US14/281,341 Abandoned US20150056698A1 (en) 1995-01-20 2014-05-19 Primate embryonic stem cells

Country Status (5)

Country Link
US (11) US5843780A (en)
EP (2) EP1640448A3 (en)
AU (1) AU4758496A (en)
CA (1) CA2190528C (en)
WO (1) WO1996022362A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030079536A1 (en) * 2001-09-10 2003-05-01 Frank Fischer Method and system for monitoring a tire air pressure
US20030207448A1 (en) * 2002-05-06 2003-11-06 Revera Gregory Henry Methodologies for the creation of pluripotent or multipotent human stem cells without creating or destroying a human embryo
US20040014206A1 (en) * 1999-10-28 2004-01-22 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20040018617A1 (en) * 2002-07-26 2004-01-29 Shiaw-Min Hwang Somatic pluripotent cells
WO2004038012A1 (en) * 2002-10-25 2004-05-06 Hunan Hui-Lin Life Technology Co. Ltd The feeder cell layer for in vitro culturing human embryonic stem cells and the method for culturing embryonic stem cells
US20040107453A1 (en) * 2001-02-14 2004-06-03 Furcht Leo T Multipotent adult stem cells, sources thereof, methods of obtaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US20050059145A1 (en) * 2001-12-21 2005-03-17 Luc Schoonjans Compositions for the in vitro derivation and culture of embryonic stem (es) cell lines with germline transmission capability and for the culture of adult stem cells
US20050100924A1 (en) * 2003-11-06 2005-05-12 National University Of Singapore C/EBPalpha gene targeting constructs and uses thereof
US20050118561A1 (en) * 2003-12-02 2005-06-02 Catholic Healthcare West Compositions and methods for propagation of neural progenitor cells
US20050181502A1 (en) * 1999-08-05 2005-08-18 Athersys, Inc. Multipotent adult stem cells and methods for isolation
US20060263879A1 (en) * 2004-12-30 2006-11-23 Stemlifeline, Inc. Methods and systems relating to embryonic stem cell lines
US20060275899A1 (en) * 2004-12-30 2006-12-07 Stemlifeline, Inc. Methods and compositions relating to embryonic stem cell lines
US20070269412A1 (en) * 2003-12-02 2007-11-22 Celavie Biosciences, Llc Pluripotent cells
US20090098650A1 (en) * 2001-12-21 2009-04-16 Thromb-X N.V. Compositions for the in vitro derivation and culture of embryonic stem (es) cell lines with germline transmission capability and for the culture of adult stem cells
US20100150876A1 (en) * 2006-11-24 2010-06-17 Regents Of The Univeristy Of Minnesota Endodermal progenitor cells
US8252280B1 (en) 1999-08-05 2012-08-28 Regents Of The University Of Minnesota MAPC generation of muscle
US20130130387A1 (en) * 2010-07-27 2013-05-23 Technion Research & Development Foundation Limited Method for generating induced pluripotent stem cells from keratinocytes derived from plucked hair follicles
US9144585B2 (en) 2010-07-27 2015-09-29 Technion Research & Development Foundation Limited Isolated mesenchymal progenitor cells and extracellular matrix produced thereby
US10638734B2 (en) 2004-01-05 2020-05-05 Abt Holding Company Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof

Families Citing this family (789)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030032178A1 (en) * 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
US7153684B1 (en) * 1992-10-08 2006-12-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
EP0688358A4 (en) * 1993-03-12 1997-10-01 Univ Creighton Improved vectors for gene therapy
US20040071637A1 (en) * 1993-04-27 2004-04-15 Elia James P. Method for repairing a damaged portion of a human organ
US5874301A (en) * 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US9068164B1 (en) 1995-02-02 2015-06-30 Tristem Trading (Cyprus) Limited Method of preparing an undifferentiated cell
GB9502022D0 (en) * 1995-02-02 1995-03-22 Abuljadayel Ilham M S A method for preparing lymphohaematopoietic progenitor cells
US7410773B2 (en) * 1995-02-02 2008-08-12 Ghazi Jaswinder Dhoot Method of preparing an undifferentiated cell
US7112440B2 (en) 1995-02-02 2006-09-26 Ghazi Jaswinder Dhoot Method of increasing the relative number of CD45 low cells in a cell population
GB9518606D0 (en) * 1995-09-12 1995-11-15 Inst Of Psychiatry Neural transplantation
US20030190753A1 (en) * 1995-11-09 2003-10-09 Nature Technology Corporation Vectors for gene transfer
US7544511B2 (en) * 1996-09-25 2009-06-09 Neuralstem Biopharmaceuticals Ltd. Stable neural stem cell line methods
AU3392697A (en) * 1996-06-14 1998-01-07 Regents Of The University Of California, The (in vitro) derivation and culture of primate pluripotent stem cells and therapeutic uses thereof
US7696404B2 (en) * 1996-08-19 2010-04-13 Advanced Cell Technology, Inc. Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
US20020194637A1 (en) * 2001-06-06 2002-12-19 University Of Massachussetts Embryonic or stem-like cell lines produced by cross species nuclear transplantation
US6331406B1 (en) * 1997-03-31 2001-12-18 The John Hopkins University School Of Medicine Human enbryonic germ cell and methods of use
US6090622A (en) * 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
DE69840439D1 (en) 1997-04-24 2009-02-26 Univ Washington TARGETED GENERIC CHANGE WITH PARVOVIRAL VECTORS
WO1999001763A2 (en) * 1997-07-02 1999-01-14 Board Of Regents, The University Of Texas System p53 AS A REGULATOR OF CELL DIFFERENTIATION
US6800480B1 (en) 1997-10-23 2004-10-05 Geron Corporation Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
WO1999027076A1 (en) * 1997-11-25 1999-06-03 Arc Genomic Research Pluripotent embryonic stem cells and methods of obtaining them
US20050096274A1 (en) * 1998-04-07 2005-05-05 Lough John W. Bone morphogenetic protein and fibroblast growth factor compositions and methods for the induction of cardiogenesis
US20080206206A1 (en) 1998-05-07 2008-08-28 University Of South Florida Bone marrow-derived neuronal cells
AU3888699A (en) 1998-05-07 1999-11-23 University Of South Florida Bone marrow cells as a source of neurons for brain and spinal cord repair
US6767737B1 (en) * 1998-08-31 2004-07-27 New York University Stem cells bearing an FGF receptor on the cell surface
AU3881499A (en) * 1998-09-01 2000-03-21 Wisconsin Alumni Research Foundation Primate embryonic stem cells with compatible histocompatibility genes
WO2000017325A1 (en) * 1998-09-23 2000-03-30 Mount Sinai Hospital Trophoblast cell preparations
US7410798B2 (en) 2001-01-10 2008-08-12 Geron Corporation Culture system for rapid expansion of human embryonic stem cells
US7413904B2 (en) * 1998-10-23 2008-08-19 Geron Corporation Human embryonic stem cells having genetic modifications
US20020168766A1 (en) * 2000-01-11 2002-11-14 Gold Joseph D. Genetically altered human pluripotent stem cells
US6667176B1 (en) * 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
JP2002529070A (en) * 1998-11-09 2002-09-10 モナシュ・ユニヴァーシティ Embryonic stem cells
GB9907243D0 (en) 1999-03-29 1999-05-26 Reneuron Ltd Therapy
US7371400B2 (en) 2001-01-02 2008-05-13 The General Hospital Corporation Multilayer device for tissue engineering
EP1187909B1 (en) * 1999-04-30 2005-02-02 Massachusetts General Hospital Fabrication of threedimensional vascularized tissue using microfabricated two-dimensional molds
US7759113B2 (en) * 1999-04-30 2010-07-20 The General Hospital Corporation Fabrication of tissue lamina using microfabricated two-dimensional molds
IL129966A (en) * 1999-05-14 2009-12-24 Technion Res & Dev Foundation ISOLATED HUMAN EMBRYOID BODIES (hEB) DERIVED FROM HUMAN EMBRYONIC STEM CELLS
IL148547A0 (en) * 1999-09-14 2002-09-12 Univ Massachusetts Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
WO2001021767A2 (en) * 1999-09-24 2001-03-29 Morphogen Pharmaceuticals, Inc. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
AUPQ307399A0 (en) * 1999-09-24 1999-10-21 Luminis Pty Limited Cell cycle control
AU782385B2 (en) * 1999-10-15 2005-07-21 Advanced Cell Technology, Inc. Methods of producing differentiated progenitor cells and lineage-defective embryonic stem cells
BR0015264A (en) * 1999-11-02 2002-10-15 Univ Massachusetts Public Inst Use of haploid genomes for diagnosis, genetic modification and multiplication
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
AU2103601A (en) * 1999-12-17 2001-06-25 Oregon Health And Science University Methods for producing transgenic animals
US20020035735A1 (en) * 2000-01-07 2002-03-21 Gerald Schatten Clonal propagation of primate offspring by embryo splitting
US7455983B2 (en) * 2000-01-11 2008-11-25 Geron Corporation Medium for growing human embryonic stem cells
US20050042749A1 (en) * 2001-05-16 2005-02-24 Carpenter Melissa K. Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
US20030134413A1 (en) * 2000-01-14 2003-07-17 Rathjen Peter David Cell production
US6602711B1 (en) * 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells
US7541184B2 (en) * 2000-02-24 2009-06-02 Invitrogen Corporation Activation and expansion of cells
US7005252B1 (en) 2000-03-09 2006-02-28 Wisconsin Alumni Research Foundation Serum free cultivation of primate embryonic stem cells
US7439064B2 (en) * 2000-03-09 2008-10-21 Wicell Research Institute, Inc. Cultivation of human embryonic stem cells in the absence of feeder cells or without conditioned medium
US6619693B1 (en) * 2000-03-10 2003-09-16 Days Corporation Apparatus and method for automatically leveling an object
US7226057B2 (en) * 2000-03-10 2007-06-05 Days Corporation Apparatus and method for automatically leveling an object
JP4889902B2 (en) 2000-03-14 2012-03-07 イーエス・セル・インターナショナル・プライヴェート・リミテッド Method for producing human neural progenitor cells from human embryonic stem (hES) cells, method for producing neurons using the method, method for producing oligodendrocytes or astrocytes
US6458589B1 (en) 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
US7776021B2 (en) * 2000-04-28 2010-08-17 The Charles Stark Draper Laboratory Micromachined bilayer unit for filtration of small molecules
US6828145B2 (en) 2000-05-10 2004-12-07 Cedars-Sinai Medical Center Method for the isolation of stem cells by immuno-labeling with HLA/MHC gene product marker
NL1017973C2 (en) 2000-05-10 2002-11-08 Tristem Trading Cyprus Ltd Design.
US8273570B2 (en) * 2000-05-16 2012-09-25 Riken Process of inducing differentiation of embryonic cell to cell expressing neural surface marker using OP9 or PA6 cells
US7250294B2 (en) 2000-05-17 2007-07-31 Geron Corporation Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
KR100903755B1 (en) * 2000-05-17 2009-06-18 제론 코포레이션 Neural progenitor cell populations
CA2381065C (en) * 2000-06-01 2007-06-05 Japan Science And Technology Corporation Method for enrichment and/or isolation of dopaminergic neurons
CA2411849A1 (en) 2000-06-05 2001-12-13 The Burnham Institute Methods of differentiating and protecting cells by modulating the p38/mef2 pathway
DK1290444T3 (en) 2000-06-14 2010-01-18 Vistagen Inc Toxicity typing using liver stem cells
JP5014535B2 (en) * 2000-06-15 2012-08-29 田辺三菱製薬株式会社 Cynomolgus monkey-derived embryonic stem cells
AU6842001A (en) * 2000-06-15 2001-12-24 Tanja Dominko Pluripotent mammalian cells
WO2001096533A1 (en) * 2000-06-15 2001-12-20 Tanabe Seiyaku Co., Ltd. Monkey-origin embryonic stem cells
CA2411914C (en) * 2000-06-20 2012-08-21 Es Cell International Pte Ltd Method of controlling differentiation of embryonic stem (es) cells by culturing es cells in the presence of bmp-2 pathway antagonists
CA2417356A1 (en) * 2000-08-01 2002-02-07 Yissum Research Development Company Directed differentiation of embryonic cells
AU2001284923A1 (en) * 2000-08-15 2002-02-25 Geron Corporation Reprogramming cells for enhanced differentiation capacity using pluripotent stemcells
US20040053869A1 (en) * 2000-08-19 2004-03-18 Peter Andrews Stem cell differentiation
AU785428B2 (en) 2000-08-30 2007-05-17 Maria Biotech Co., Ltd Human embryonic stem cells derived from frozen-thawed embryo
US6534052B1 (en) 2000-09-05 2003-03-18 Yong-Fu Xiao Cardiac function comprising implantation of embryonic stem cell in which differentiation has been initiated
US6607720B1 (en) 2000-09-05 2003-08-19 Yong-Fu Xiao Genetically altered mammalian embryonic stem cells, their living progeny, and their therapeutic application for improving cardiac function after myocardial infarction
JPWO2002022789A1 (en) * 2000-09-14 2004-01-22 結城 惇 Method for producing normal parenchymal cells, tissues or organs using a biological incubator
AUPR095200A0 (en) * 2000-10-24 2000-11-16 Bresagen Limited Cell production
US20040072343A1 (en) * 2000-11-09 2004-04-15 Verma Paul John Cell reprogramming
IL155728A0 (en) * 2000-11-22 2003-11-23 Geron Corp Tolerizing allografts of pluripotent stem cells
US6576464B2 (en) * 2000-11-27 2003-06-10 Geron Corporation Methods for providing differentiated stem cells
US6921665B2 (en) * 2000-11-27 2005-07-26 Roslin Institute (Edinburgh) Selective antibody targeting of undifferentiated stem cells
IL156234A0 (en) * 2000-11-30 2004-01-04 Stemron Inc Isolated homozygous stem cells, differentiated cells derived therefrom, and materials and methods for making and using same
US20030027331A1 (en) * 2000-11-30 2003-02-06 Yan Wen Liang Isolated homozygous stem cells, differentiated cells derived therefrom, and materials and methods for making and using same
US20020068046A1 (en) * 2000-12-04 2002-06-06 Jianwu Dai Use of stem cells derived from dermal skin
AU2002251681A1 (en) * 2000-12-05 2002-08-28 Layton Bioscience Inc. Production and use of dopaminergic cells to treat dopaminergic deficiencies
KR20030088023A (en) 2001-01-02 2003-11-15 스템론 인크. A Method for Producing a Population of Homozygous Stem Cells Having a Pre-selected Immunotype and/or Genotype, Cells Suitable for Transplant Derived therefrom, and Materials and Methods Using Same
EP1366148A2 (en) * 2001-01-24 2003-12-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE DEPARTMENT OF HEALTH & HUMAN SERVICES Differentiation of stem cells to pancreatic endocrine cells
AUPR349501A0 (en) * 2001-03-02 2001-03-29 Bresagen Limited Cellular production control
US7126039B2 (en) * 2001-03-21 2006-10-24 Geron Corporation Animal tissue with carbohydrate antigens compatible for human transplantation
US7838292B1 (en) * 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
CA2442177A1 (en) * 2001-03-29 2002-10-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
EP1390076A4 (en) * 2001-04-27 2004-12-15 Xcyte Therapies Inc Maturation of antigen-presenting cells using activated t cells
US20030211605A1 (en) * 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
US20050176665A1 (en) * 2001-05-18 2005-08-11 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hairless (HR) gene expression using short interfering nucleic acid (siNA)
CA2453068C (en) * 2001-07-06 2018-02-20 Geron Corporation Mesenchymal cells and osteoblasts from human embryonic stem cell
US7732199B2 (en) 2001-07-12 2010-06-08 Geron Corporation Process for making transplantable cardiomyocytes from human embryonic stem cells
US7425448B2 (en) * 2001-07-12 2008-09-16 Geron Corporation Cardiomyocyte precursors from human embryonic stem cells
US20030017587A1 (en) * 2001-07-18 2003-01-23 Rader William C. Embryonic stem cells, clinical applications and methods for expanding in vitro
DE10136719A1 (en) * 2001-07-27 2003-02-20 Georg S Wengler Recovering omnipotent human embryonic stem cells, comprises separating them from a blastocyte which remains viable, useful for gene therapy especially autotransplants
CA2456981C (en) 2001-08-06 2012-02-28 Bresagen, Inc. Alternative compositions and methods for the culture of stem cells
US20040092013A1 (en) * 2001-08-14 2004-05-13 Snyder Evan Y. Method of treating alzheimer's disease with cell therapy
US20030211603A1 (en) * 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
AU2002334378B2 (en) * 2001-08-23 2008-02-28 Reliance Life Sciences Pvt.,Ltd Isolation of inner cell mass for the establishment of human embryonic stem cell (hESC) lines
US7588937B2 (en) 2001-10-03 2009-09-15 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US8153424B2 (en) * 2001-10-03 2012-04-10 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US6887706B2 (en) * 2001-10-03 2005-05-03 Wisconsin Alumni Research Foundation Method of in vitro differentiation of transplantable neural precursor cells from primate embryonic stem cells
CA2465173C (en) * 2001-11-02 2010-04-20 Wisconsin Alumni Research Foundation Endothelial cells derived from primate embryonic stem cells
US6759244B2 (en) * 2001-11-08 2004-07-06 Art Institute Of New York And New Jersey, Inc. Composite blastocysts (CBs) from aggregates of dissociated cells of non-viable pre-embryos
US7491690B2 (en) * 2001-11-14 2009-02-17 Northwestern University Self-assembly and mineralization of peptide-amphiphile nanofibers
AU2002363659B2 (en) * 2001-11-15 2008-09-25 Children's Medical Center Corporation Methods of isolation, expansion and differentiation of fetal stem cells from chorionic villus, amniotic fluid, and placenta and therapeutic uses thereof
KR101008868B1 (en) * 2001-12-07 2011-01-17 제론 코포레이션 Islet cells from human embryonic stem cells
US20040224403A1 (en) * 2001-12-07 2004-11-11 Robarts Research Institute Reconstituting hematopoietic cell function using human embryonic stem cells
US7799324B2 (en) * 2001-12-07 2010-09-21 Geron Corporation Using undifferentiated embryonic stem cells to control the immune system
IL162132A0 (en) * 2001-12-07 2005-11-20 Geron Corp Chondrocyte precursors derived from human embryonic stem cells
KR101457742B1 (en) 2001-12-07 2014-11-03 아스테리아스 바이오세라퓨틱스, 인크. Hematopoietic cells from human embryonic stem cells
US20030113910A1 (en) * 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
CA2471540A1 (en) * 2001-12-28 2003-07-10 Cellartis Ab A method for the establishment of a pluripotent human blastocyst-derived stem cell line
US7190781B2 (en) * 2002-01-04 2007-03-13 Telefonaktiebolaget Lm Ericsson (Publ) Message transfer part point code mapping method and node
US20040043482A1 (en) * 2002-01-04 2004-03-04 Kye-Hyung Paik Method of producing stem cell lines
US20050090004A1 (en) * 2003-01-16 2005-04-28 Sayre Chauncey B. Stem cell maturation for all tissue lines
US20050170506A1 (en) * 2002-01-16 2005-08-04 Primegen Biotech Llc Therapeutic reprogramming, hybrid stem cells and maturation
US20030134422A1 (en) * 2002-01-16 2003-07-17 Sayre Chauncey Bigelow Stem cell maturation for all tissue lines
US20030162290A1 (en) * 2002-01-25 2003-08-28 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
AU2003238620B2 (en) * 2002-01-31 2008-11-06 Asahi Techno Glass Corporation Liquid for frozen storage of primate embryo stem cells and frozen storage method
WO2003066839A1 (en) * 2002-02-05 2003-08-14 Rappaport Family Institute For Research In The Medical Sciences Lineage committed stem cells selected for telomerase promoter activity
ATE397654T1 (en) * 2002-02-06 2008-06-15 Stiftung Caesar PLURIPOTENT EMBRYON-LIKE STEM CELLS DERIVED FROM TEETH AND USES THEREOF
US7371719B2 (en) * 2002-02-15 2008-05-13 Northwestern University Self-assembly of peptide-amphiphile nanofibers under physiological conditions
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20030161818A1 (en) * 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
WO2003078967A2 (en) * 2002-03-12 2003-09-25 Oregon Health & Science University Stem cell selection and differentiation
WO2003075648A1 (en) * 2002-03-13 2003-09-18 Fundacion Ivi Para El Estudio De La Reproduccion Humana (Fivier) Method of producing cell lines
US7148062B2 (en) * 2002-03-15 2006-12-12 Wicell Research Institute, Inc. Method for generating primate trophoblasts
IL163949A0 (en) * 2002-03-15 2005-12-18 Wisconsin Alumni Res Found Method of identifying genes controlling differentiation
GB0207440D0 (en) * 2002-03-28 2002-05-08 Ppl Therapeutics Scotland Ltd Tolerogenic antigen-presenting cells
US20040111285A1 (en) * 2002-04-09 2004-06-10 Mark Germain Method for human pluripotent stem cells
AU2003239176A1 (en) * 2002-04-25 2003-11-10 Wisconsin Alumni Research Foundation Neurodegenerative disorder treatment using gdnf secreting neural cells
US6816665B2 (en) * 2002-05-09 2004-11-09 Lynx Photonic Networks Inc. Constant power operation thermo-optic switch
AU2003304106C1 (en) * 2002-05-17 2010-10-28 Mount Sinai School Of Medicine Of New York University Mesoderm and definitive endoderm cell populations
US20060003446A1 (en) 2002-05-17 2006-01-05 Gordon Keller Mesoderm and definitive endoderm cell populations
EP1513928B1 (en) 2002-05-24 2019-08-21 Advanced Cell Technology, Inc. A bank of stem cells for producing cells for transplantation having hla antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
WO2003104423A2 (en) * 2002-06-11 2003-12-18 Roy Ogle Meningeal-derived stem cells
WO2003106640A2 (en) * 2002-06-14 2003-12-24 Case Western Reserve University Cell targeting methods and compositions
US7285415B2 (en) 2002-07-11 2007-10-23 The Regents Of The University Of California Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US20050101014A1 (en) * 2002-07-11 2005-05-12 Keirstead Hans S. Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
WO2004009758A2 (en) * 2002-07-23 2004-01-29 Nanodiagnostics, Inc. Embryonic stem cell markers and uses thereof
WO2007116408A2 (en) * 2006-04-11 2007-10-18 Nanodiagnostics Israel Ltd. Pluripotent stem cells characterized by expression of germline specific genes
US20110158966A1 (en) * 2002-07-23 2011-06-30 Judith Seligman Stem cells characterized by expression of germline specific genes
US8876532B2 (en) 2002-07-31 2014-11-04 Dentsply International Inc. Bone repair putty
US7462448B2 (en) * 2002-08-02 2008-12-09 Stratatech Corporation Species specific DNA detection
WO2004015077A2 (en) * 2002-08-08 2004-02-19 University Of Georgia Research Foundation, Inc. Compositions and methods for neural differentiation of embryonic stem cells
AU2003249749A1 (en) * 2002-08-09 2004-02-25 Dr. H. Zech Gmbh Method for producing cell lines and organs by means of differentiable cells
US7534761B1 (en) 2002-08-21 2009-05-19 North Western University Charged peptide-amphiphile solutions and self-assembled peptide nanofiber networks formed therefrom
CA2404540A1 (en) * 2002-09-20 2004-03-20 Institut Pasteur A method for measuring a marker indicative of the exposure of a patient to nicotine; a kit for measuring such a marker
AU2003276924A1 (en) * 2002-09-25 2004-04-19 Bresagen, Inc. Compositions and methods for enrichment of neural stem cells using ceramide analogs
US7267981B2 (en) * 2002-10-07 2007-09-11 Technion Research & Development Foundation Ltd. Human foreskin fibroblasts for culturing ES cells
US7554021B2 (en) * 2002-11-12 2009-06-30 Northwestern University Composition and method for self-assembly and mineralization of peptide amphiphiles
WO2004046167A2 (en) 2002-11-14 2004-06-03 Northwestern University Synthesis and self-assembly of abc triblock bola peptide
AU2003302701A1 (en) * 2002-11-29 2004-06-23 Technion Research And Development Foundation Ltd. Method of dynamically culturing embryonic stem cells
US20040110286A1 (en) * 2002-12-06 2004-06-10 The John P. Robarts Research Institute Method for making hematopoietic cells
ES2571355T3 (en) 2002-12-16 2016-05-24 Technion Res & Dev Foundation Culture system without feeder cells or xenocontaminants for human embryonic stem cells
AU2003303741A1 (en) * 2002-12-18 2004-09-17 Bresagen, Inc. Compositions and methods for neural cell production and stabilization
EP1589814B1 (en) 2003-01-16 2009-08-12 The General Hospital Corporation Use of three-dimensional microfabricated tissue engineered systems for pharmacologic applications
WO2004072104A2 (en) * 2003-02-11 2004-08-26 Northwestern University Methods and materials for nanocrystalline surface coatings and attachment of peptide amphiphile nanofibers thereon
KR100763477B1 (en) * 2003-03-12 2007-10-04 리라이언스 라이프 사이언시스 프라이빗. 리미티드 Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells
US20030224411A1 (en) * 2003-03-13 2003-12-04 Stanton Lawrence W. Genes that are up- or down-regulated during differentiation of human embryonic stem cells
US7153650B2 (en) * 2003-03-13 2006-12-26 Geron Corporation Marker system for preparing and characterizing high-quality human embryonic stem cells
CA2520861A1 (en) * 2003-03-27 2004-10-14 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic pathway
JP4502317B2 (en) * 2003-03-28 2010-07-14 独立行政法人農業生物資源研究所 Method for selecting undifferentiated cells and use thereof
US20040191839A1 (en) * 2003-03-28 2004-09-30 National Institute Of Agrobiological Sciences Japan Methods for sorting undifferentiated cells and uses thereof
JP2004298108A (en) * 2003-03-31 2004-10-28 Japan Science & Technology Agency Method for producing lens cell, and lens cell obtained by the method
CA2520023C (en) 2003-04-08 2013-02-05 Yeda Research And Development Co. Ltd Stem cells having increased sensitivity to sdf-1 and methods of generating and using same
FR2853551B1 (en) 2003-04-09 2006-08-04 Lab Francais Du Fractionnement STABILIZING FORMULATION FOR IMMUNOGLOBULIN G COMPOSITIONS IN LIQUID FORM AND LYOPHILIZED FORM
US20060037086A1 (en) * 2003-04-09 2006-02-16 Schatten Gerald P Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals
JP2006522609A (en) * 2003-04-09 2006-10-05 マギー−ウイメンズ・ヘルス・コーポレイション A method for correcting spindle defects associated with somatic cell nuclear transfer in animals
IL155783A (en) 2003-05-05 2010-11-30 Technion Res & Dev Foundation Multicellular systems of pluripotent human embryonic stem cells and cancer cells and uses thereof
US20070020608A1 (en) * 2003-05-08 2007-01-25 Peter Eriksson Method for the generation of neural progenitor cells
EP1625213A2 (en) * 2003-05-08 2006-02-15 Cellartis AB A method for efficient transfer of human blastocyst-derived stem cells (hbs cells) from a feeder-supported to a feeder-free culture system
US9567591B2 (en) 2003-05-15 2017-02-14 Mello Biotechnology, Inc. Generation of human embryonic stem-like cells using intronic RNA
US20090203141A1 (en) * 2003-05-15 2009-08-13 Shi-Lung Lin Generation of tumor-free embryonic stem-like pluripotent cells using inducible recombinant RNA agents
US7960166B2 (en) 2003-05-21 2011-06-14 The General Hospital Corporation Microfabricated compositions and processes for engineering tissues containing multiple cell types
WO2004108882A2 (en) * 2003-06-11 2004-12-16 Yeda Research And Development Co. Ltd. Neural stem cells and methods of generating and utilizing same
EP1702062A2 (en) * 2003-06-11 2006-09-20 Jan Remmereit Differentiation of stem cells for therapeutic use
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
EP1641913B1 (en) 2003-06-27 2016-01-06 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US20050019907A1 (en) * 2003-07-22 2005-01-27 Santiago Munne Obtaining normal disomic stem cells from chromosomally abnormal embryos
EP3100611B1 (en) 2003-08-01 2018-09-12 Stratatech Corporation Human skin equivalents expressing exogenous polypeptides
US20050042595A1 (en) * 2003-08-14 2005-02-24 Martin Haas Banking of multipotent amniotic fetal stem cells
US7820439B2 (en) * 2003-09-03 2010-10-26 Reliance Life Sciences Pvt Ltd. In vitro generation of GABAergic neurons from pluripotent stem cells
US20050054100A1 (en) * 2003-09-08 2005-03-10 Rennard Stephen I. Methods for fibroblast differentiation
WO2005033297A1 (en) * 2003-09-19 2005-04-14 The Rockefeller University Compositions, methods and kits relating to reprogramming adult differentiated cells and production of embryonic stem cell-like cells
WO2005032251A1 (en) * 2003-10-09 2005-04-14 I.M.T. Interface Multigrad Technology Ltd. Method for freezing, thawing and transplantation of viable cartilage
IL158868A0 (en) * 2003-11-13 2004-05-12 Yeda Res & Dev Methods of generating and using stem cells enriched with immature primitive progenitor
US20050123525A1 (en) * 2003-11-13 2005-06-09 Ulrich Martin Composition and method for inducing immune tolerance towards cell, tissue and/or organ transplants
US20050106725A1 (en) * 2003-11-19 2005-05-19 Palecek Sean P. Method of reducing cell differentiation
US20080026460A1 (en) * 2006-06-20 2008-01-31 Palecek Sean P Method for culturing stem cells
US20080220520A1 (en) * 2003-11-19 2008-09-11 Palecek Sean P Cryopreservation of human embryonic stem cells in microwells
US20050106554A1 (en) * 2003-11-19 2005-05-19 Palecek Sean P. Cryopreservation of pluripotent stem cells
US7790039B2 (en) * 2003-11-24 2010-09-07 Northwest Biotherapeutics, Inc. Tangential flow filtration devices and methods for stem cell enrichment
US7682828B2 (en) * 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
KR20070004560A (en) * 2003-12-05 2007-01-09 노오쓰웨스턴 유니버시티 Self-assembling peptide amphiphiles and related methods for growth factor delivery
JP4741510B2 (en) * 2003-12-05 2011-08-03 ノースウエスタン ユニバーシティ Branched peptide amphiphiles, related epitope compounds and their self-assembled structures
TWI280280B (en) * 2003-12-09 2007-05-01 Ind Tech Res Inst Culture system and method for expansion and undifferentiated growth of human embryonic stem cells
US20060030042A1 (en) * 2003-12-19 2006-02-09 Ali Brivanlou Maintenance of embryonic stem cells by the GSK-3 inhibitor 6-bromoindirubin-3'-oxime
EP1709159B1 (en) 2003-12-23 2019-05-15 Viacyte, Inc. Definitive endoderm
US8586357B2 (en) * 2003-12-23 2013-11-19 Viacyte, Inc. Markers of definitive 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
US20050214257A1 (en) 2003-12-23 2005-09-29 Northwestern University Compositions and methods for controlling stem cell and tumor cell differentiation, growth, and formation
US7625753B2 (en) * 2003-12-23 2009-12-01 Cythera, Inc. Expansion of definitive endoderm cells
US8647873B2 (en) 2004-04-27 2014-02-11 Viacyte, Inc. PDX1 expressing endoderm
US20050233446A1 (en) * 2003-12-31 2005-10-20 Parsons Xuejun H Defined media for stem cell culture
NZ548623A (en) 2004-01-02 2010-04-30 Advanced Cell Tech Inc Novel culture systems for ex vivo development of stem cells in telolecithal or eutelolecithal eggs
AU2005209169B2 (en) 2004-01-16 2010-12-16 Carnegie Mellon University Cellular labeling for nuclear magnetic resonance techniques
WO2005071066A1 (en) * 2004-01-23 2005-08-04 Board Of Regents, The University Of Texas System Methods and compositions for preparing pancreatic insulin secreting cells
US20050186672A1 (en) * 2004-01-27 2005-08-25 Reliance Life Sciences Pvt. Ltd. Tissue system with undifferentiated stem cells derived from corneal limbus
JP4777908B2 (en) * 2004-02-02 2011-09-21 コア・ダイナミクス・リミテッド Biological materials and methods and solutions for storage of biological materials
WO2005073652A2 (en) * 2004-02-02 2005-08-11 I.M.T. Interface Multigrad Technology Ltd. Apparatus, system and method for lyophilization
WO2005080551A2 (en) * 2004-02-12 2005-09-01 University Of Newcastle Upon Tyne Stem cells
US8187875B2 (en) 2004-02-26 2012-05-29 Reliance Life Sciences Pvt. Ltd. Dopaminergic neurons derived from corneal limbus, methods of isolation and uses thereof
US20060216821A1 (en) * 2004-02-26 2006-09-28 Reliance Life Sciences Pvt. Ltd. Pluripotent embryonic-like stem cells derived from corneal limbus, methods of isolation and uses thereof
KR20080036636A (en) * 2004-02-26 2008-04-28 리라이언스 라이프 사이언시스 프라이빗. 리미티드 Pluripotent embryonic-like stem cells derived from corneal limbus, methods of isolation and uses thereof
US20050214938A1 (en) * 2004-03-26 2005-09-29 Gold Joseph D Cardiac bodies: clusters of spontaneously contracting cells for regenerating cardiac function
US7452718B2 (en) * 2004-03-26 2008-11-18 Geron Corporation Direct differentiation method for making cardiomyocytes from human embryonic stem cells
US7622108B2 (en) 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
CA2563518C (en) 2004-04-23 2014-09-02 Bioe, Inc. Multi-lineage progenitor cells
EP2377922B1 (en) 2004-04-27 2020-04-08 Viacyte, Inc. PDX1 expressing endoderm
AU2005243158B2 (en) * 2004-05-07 2010-03-25 Wisconsin Alumni Research Foundation Method of forming mesenchymal stem cells from embryonic stem cells
WO2005120591A1 (en) * 2004-06-07 2005-12-22 I.M.T. Interface Multigrad Technology Ltd. Method for sterilization of biological preparations
GB2414995B (en) 2004-06-09 2006-11-15 Univ Edinburgh Neural stem cells
US20060008451A1 (en) * 2004-07-06 2006-01-12 Michigan State University In vivo methods for effecting tissue specific differentiation of embryonic stem cells
CA2573354C (en) 2004-07-09 2015-02-24 Cythera, Inc. Preprimitive streak and mesendoderm cells
JP5687816B2 (en) 2004-07-09 2015-03-25 ヴィアサイト,インコーポレイテッド Methods for identifying factors for differentiating definitive endoderm
AU2005271723B2 (en) 2004-07-13 2010-12-16 Asterias Biotherapeutics, Inc. Medium for growing human embryonic stem cells
EP1781776A2 (en) * 2004-07-29 2007-05-09 Stem Cell Innovations, Inc. Differentiation of stem cells
WO2006016372A1 (en) * 2004-08-12 2006-02-16 I.M.T. Interface Multigrad Technology Ltd. Method and apparatus for freezing or thawing of a biological material
MX2007001772A (en) * 2004-08-13 2007-07-11 Univ Georgia Res Found Compositions and methods for self-renewal and differentiation in human embryonic stem cells.
EP1802744A4 (en) * 2004-09-03 2008-06-18 Agency Science Tech & Res Method for maintaining pluripotency of stem/progenitor cells
WO2006029093A1 (en) * 2004-09-07 2006-03-16 Central Institute For Experimental Animals Common marmoset embryonic stem cell lines
AU2005286097A1 (en) * 2004-09-21 2006-03-30 Nvr Labs. Ltd. Compositions and methods for stem cell expansion and differentiation
DK1645626T3 (en) * 2004-09-30 2008-01-21 Reneuron Ltd cell Line
US20070077654A1 (en) * 2004-11-01 2007-04-05 Thomson James A Platelets from stem cells
EP2960328A1 (en) 2004-11-04 2015-12-30 Ocata Therapeutics, Inc. Derivation of embryonic stem cells
US7893315B2 (en) * 2004-11-04 2011-02-22 Advanced Cell Technology, Inc. Derivation of embryonic stem cells and embryo-derived cells
WO2006053378A1 (en) * 2004-11-16 2006-05-26 Sydney Ifv Limited Derivation and culture of human embryo-derived cells
KR101429284B1 (en) 2004-11-17 2014-08-11 뉴럴스템, 인크. Transplantation of Human Neural Cells for Treatment of Neurodegenerative Conditions
US8017395B2 (en) 2004-12-17 2011-09-13 Lifescan, Inc. Seeding cells on porous supports
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
WO2006101548A2 (en) * 2004-12-21 2006-09-28 Ethicon, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
WO2006083394A2 (en) * 2004-12-21 2006-08-10 Ethicon, Inc. Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
CA2592435C (en) 2004-12-23 2017-03-28 Ethicon, Incorporated Treatment of stroke and other acute neural degenerative disorders using postpartum derived cells
DE102004062184B4 (en) * 2004-12-23 2013-08-01 Wolfgang Würfel Embryo-preserving production of pluripotent embryonic stem cells, stem cells thus obtained and use thereof
EP1838843B1 (en) 2004-12-23 2019-07-10 Viacyte, Inc. Expansion of definitive endoderm cells
PL1831356T3 (en) 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8597947B2 (en) * 2004-12-29 2013-12-03 Hadasit Medical Research Services & Development Limited Undifferentiated stem cell culture systems
WO2006070370A2 (en) * 2004-12-29 2006-07-06 Hadasit Medical Research Services & Development Limited Stem cells culture systems
WO2006079036A2 (en) * 2005-01-21 2006-07-27 Northwestern University Methods and compositions for encapsulation of cells
US7022518B1 (en) 2005-01-31 2006-04-04 Glen Feye Apparatus and method for co-culturing of cells
WO2006084314A1 (en) * 2005-02-09 2006-08-17 Australian Stem Cell Centre Limited Stem cell populations and classification system
WO2006090372A2 (en) * 2005-02-22 2006-08-31 I.M.T. Interface Multigrad Technology Ltd. Preserved viable cartilage, method for its preservation, and system and devices used therefor
US7851445B2 (en) * 2005-03-04 2010-12-14 Northwestern University Angiogenic heparin-binding epitopes, peptide amphiphiles, self-assembled compositions and related methods of use
US20070048865A1 (en) * 2005-03-15 2007-03-01 Kenzaburo Tani Method for acceleration of stem cell differentiation
JP2008532550A (en) 2005-03-17 2008-08-21 ストラタテック コーポレーション Skin substitute with improved purity
US8012751B2 (en) * 2005-03-31 2011-09-06 Wisconsin Alumni Research Foundation Differentiation of pluripotent embryonic stem cells
WO2007021321A2 (en) 2005-04-01 2007-02-22 Wake Forest University Health Sciences Transcriptional profiling of stem cells and their multilineage differentiation
RU2007146777A (en) * 2005-05-17 2009-06-27 Релайанс Лайф Сайенсиз Пвт Лтд (In) OBTAINING A LINE OF HUMAN EMBRYONIC STEM CELLS USING MAMMAL CELLS
AU2006202209B2 (en) * 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
WO2006133052A2 (en) * 2005-06-08 2006-12-14 Centocor, Inc. A cellular therapy for ocular degeneration
WO2007002086A2 (en) * 2005-06-22 2007-01-04 Geron Corporation Suspension culture of human embryonic stem cells
US20060292694A1 (en) * 2005-06-22 2006-12-28 Roslin Institute Reporter hepatocytes and other cells for drug screening and toxicity testing
WO2007002136A2 (en) 2005-06-22 2007-01-04 Geron Corporation Differentiation of primate pluripotent stem cells to cardiomyocyte-lineage cells
US20060292695A1 (en) * 2005-06-22 2006-12-28 Roslin Institute Methods and kits for drug screening and toxicity testing using promoter-reporter cells derived from embryonic stem cells
WO2007002568A1 (en) * 2005-06-22 2007-01-04 Geron Corporation Reporter hepatocytes and other cells for drug screening and toxicity testing
US20070026520A1 (en) * 2005-07-29 2007-02-01 Kelly James H Novel cells, compositions, and methods
WO2007015252A2 (en) * 2005-08-03 2007-02-08 I.M.T. Interface Multigrad Technology Ltd. Somatic cells for use in cell therapy
EP1924606A4 (en) 2005-08-25 2010-01-13 Repair Technologies Inc Devices, compositions and methods for the protection and repair of cells and tissues
EP1962719A4 (en) 2005-08-29 2011-05-04 Technion Res And Dev Of Foundation Ltd Media for culturing stem cells
WO2008020815A1 (en) 2006-08-15 2008-02-21 Agency For Science, Technology And Research Mesenchymal stem cell conditioned medium
EP2796544B1 (en) 2005-09-09 2019-04-03 Duke University Tissue engineering methods and compositions
ES2687233T3 (en) 2005-10-27 2018-10-24 Viacyte, Inc. Endoderm of the dorsal and ventral proximal intestine expressing PDX-1
US7413900B2 (en) * 2005-10-31 2008-08-19 President And Fellows Of Harvard College Immortalized fibroblasts
AU2006315668B2 (en) 2005-11-10 2013-06-20 Genervon Biopharmaceuticals Llc MNTF differentiation and growth of stem cells
US7521221B2 (en) * 2005-11-21 2009-04-21 Board Of Trustees Of The University Of Arknasas Staphylococcus aureus strain CYL1892
PL2535403T3 (en) 2005-12-08 2019-10-31 Univ Louisville Res Found Inc Very small embryonic-like (VSEL) stem cells and methods of isolating and using the same
US9155762B2 (en) * 2005-12-08 2015-10-13 University Of Louisville Research Foundation, Inc. Uses and isolation of stem cells from bone marrow
US8129187B2 (en) * 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
CN103113463B (en) * 2005-12-13 2015-02-18 国立大学法人京都大学 Nuclear reprogramming factor
US20090227032A1 (en) * 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US8278104B2 (en) * 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
PL1971681T3 (en) 2005-12-16 2018-01-31 Depuy Synthes Products Inc Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
WO2007073552A1 (en) 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
WO2007076522A2 (en) * 2005-12-28 2007-07-05 Ethicon, Incorporated Treatment of peripheral vascular disease using postpartum-derived cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
WO2007101130A2 (en) 2006-02-23 2007-09-07 Novocell, Inc. Compositions and methods useful for culturing differentiable cells
SG10201405380QA (en) 2006-03-02 2014-10-30 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
JP5468782B2 (en) 2006-03-02 2014-04-09 エージェンシー フォー サイエンス,テクノロジー アンド リサーチ Methods for cancer treatment and stem cell regulation
EP2422800A3 (en) 2006-03-07 2013-03-06 Geeta Shroff Compositions comprising human embryonic stem cells and their derivatives, methods of use, and methods of preparation
US20070269411A1 (en) * 2006-03-10 2007-11-22 Wei Sun Porous silicon materials and devices
HUE036378T2 (en) * 2006-04-10 2018-07-30 Wisconsin Alumni Res Found Methods for using human embryonic stem cells to evaluate toxicity of pharmaceutical compounds & other chemicals
WO2007120911A2 (en) * 2006-04-14 2007-10-25 Carnegie Mellon University Cellular labeling and quantification for nuclear magnetic resonance techniques
WO2008054509A2 (en) * 2006-04-14 2008-05-08 Celsense, Inc. Methods for assessing cell labeling
WO2007121443A2 (en) 2006-04-17 2007-10-25 Bioe, Inc. Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
JP5149791B2 (en) 2006-04-28 2013-02-20 第一三共株式会社 Method for inducing differentiation of cardiomyocytes from pluripotent stem cells
US7989204B2 (en) 2006-04-28 2011-08-02 Viacyte, Inc. Hepatocyte lineage cells
EP2021462B1 (en) 2006-04-28 2019-01-09 Lifescan, Inc. Differentiation of human embryonic stem cells
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
DK2656832T3 (en) 2006-05-11 2019-04-08 Regenics As Compositions for use in wound healing
US7964402B2 (en) 2006-05-25 2011-06-21 Sanford-Burnham Medical Research Institute Methods for culture and production of single cell populations of human embryonic stem cells
US20090298169A1 (en) * 2006-06-02 2009-12-03 The University Of Georgia Research Foundation Pancreatic and Liver Endoderm Cells and Tissue by Differentiation of Definitive Endoderm Cells Obtained from Human Embryonic Stems
US8415153B2 (en) 2006-06-19 2013-04-09 Geron Corporation Differentiation and enrichment of islet-like cells from human pluripotent stem cells
US20080003676A1 (en) * 2006-06-26 2008-01-03 Millipore Corporation Growth of embryonic stem cells
US20080124276A1 (en) * 2006-07-24 2008-05-29 Lifeline Cell Technology Synthetic cornea from retinal stem cells
CA2661232A1 (en) * 2006-08-31 2008-03-06 The University Of Louisville Research Foundation, Inc. Transcription factors for differentiation of adult human olfactory progenitor 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
US8685720B2 (en) * 2006-11-03 2014-04-01 The Trustees Of Princeton University Engineered cellular pathways for programmed autoregulation of differentiation
US20080108044A1 (en) * 2006-11-08 2008-05-08 Deepika Rajesh In vitro differentiation of hematopoietic cells from primate embryonic stem cells
US7883698B2 (en) * 2007-01-17 2011-02-08 Maria Michejda Isolation and preservation of fetal hematopoietic and mesencymal system cells from non-controversial materials and/or tissues resulting from miscarriages and methods of therapeutic use
US20100068806A1 (en) 2007-01-18 2010-03-18 Suomen Punainen Risti, Veripalvelu Novel methods and reagents directed to production of cells
JP2010516240A (en) 2007-01-18 2010-05-20 スオメン プナイネン リスティ,ヴェリパルベル Novel carbohydrates from human cells and methods for their analysis and modification
US8084023B2 (en) * 2007-01-22 2011-12-27 The Board Of Trustees Of The University Of Arkansas Maintenance and propagation of mesenchymal stem cells
KR20090115142A (en) * 2007-01-30 2009-11-04 유니버시티 오브 조지아 리서치 파운데이션, 인코포레이티드 Early mesoderm cells, a stable population of mesendoderm cells that has utility for generation of endoderm and mesoderm lineages and multipotent migratory cells(mmc)
WO2008121437A2 (en) * 2007-02-08 2008-10-09 The Burnham Institute For Medical Research Trophinin-binding peptides and uses thereof
EP3190178A1 (en) 2007-02-23 2017-07-12 Astellas Institute for Regenerative Medicine Highly efficient methods for reprogramming differentiated cells and for generating animals and embryonic stem cells from reprogrammed cells
AU2008231020B2 (en) 2007-03-23 2013-09-05 Wisconsin Alumni Research Foundation Somatic cell reprogramming
CA2678901C (en) 2007-03-26 2017-11-14 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Methods for modulating embryonic stem cell differentiation
US20080267874A1 (en) * 2007-03-28 2008-10-30 The Buck Institute For Age Research Targeted Neuronal And Glial Human Embryonic Stem Cell Line
CA3071055A1 (en) 2007-04-07 2008-10-16 Whitehead Institute For Biomedical Research Reprogramming of somatic cells
US8076295B2 (en) * 2007-04-17 2011-12-13 Nanotope, Inc. Peptide amphiphiles having improved solubility and methods of using same
JP5395058B2 (en) * 2007-04-18 2014-01-22 ハダシット メディカル リサーチ サーヴィシーズ アンド ディヴェロップメント リミテッド Retinal pigment epithelial cells derived from stem cells
WO2008150030A1 (en) * 2007-06-07 2008-12-11 Keiichi Fukuda Method of inducing differentiation into myocardial cells using g-csf
JP2008307007A (en) * 2007-06-15 2008-12-25 Bayer Schering Pharma Ag Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
US9213999B2 (en) * 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
DK2173863T3 (en) 2007-06-29 2019-01-21 Fujifilm Cellular Dynamics Inc Automated method and apparatus for embryonic stem cell culture
KR101732952B1 (en) * 2007-07-01 2017-05-08 라이프스캔, 인코포레이티드 Single pluripotent stem cell culture
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
AU2008275578B2 (en) * 2007-07-10 2014-04-10 Carnegie Mellon University Compositions and methods for producing cellular labels for nuclear magnetic resonance techniques
CN101861386A (en) 2007-07-18 2010-10-13 生命扫描有限公司 The differentiation of human embryo stem cell
CN101835479A (en) * 2007-07-25 2010-09-15 佰欧益有限公司 Differentiation of multi-lineage progenitor cells to chondrocytes
DK2185693T3 (en) 2007-07-31 2019-09-23 Lifescan Inc DIFFERENTIZING HUMAN EMBRYONIC STEM CELLS
ES2665434T3 (en) 2007-07-31 2018-04-25 Lifescan, Inc. Differentiation of pluripotent stem cells using human feeder cells
WO2009026106A1 (en) 2007-08-16 2009-02-26 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Methods for promoting stem cell proliferation and survival
CA2698091C (en) 2007-08-31 2018-07-03 Brett Chevalier Wnt pathway stimulation in reprogramming somatic cells
US10925903B2 (en) 2007-09-13 2021-02-23 Reprobiogen Inc. Use of cells derived from first trimester umbilical cord tissue
US7695963B2 (en) 2007-09-24 2010-04-13 Cythera, Inc. Methods for increasing definitive endoderm production
US20110236971A2 (en) * 2007-09-25 2011-09-29 Maksym Vodyanyk Generation of Clonal Mesenchymal Progenitors and Mesenchymal Stem Cell Lines Under Serum-Free Conditions
US8034329B2 (en) 2007-10-05 2011-10-11 Advanced Technologies And Regenerative Medicine, Llc Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8623650B2 (en) 2007-10-19 2014-01-07 Viacyte, Inc. Methods and compositions for feeder-free pluripotent stem cell media containing human serum
CN101978046A (en) * 2007-10-30 2011-02-16 路易斯维尔大学研究基金会有限公司 Uses and isolation of very small embryonic-like (vsel) stem cells
CN107574142B (en) * 2007-11-27 2021-07-06 生命扫描有限公司 Differentiation of human embryonic stem cells
WO2009076529A1 (en) * 2007-12-11 2009-06-18 Research Development Foundation Small molecules for neuronal differentiation of embryonic stem cells
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
SG187493A1 (en) 2008-01-18 2013-02-28 Univ Minnesota Stem cell aggregates and methods for making and using
CN101939362A (en) 2008-01-30 2011-01-05 杰龙公司 Synthetic surfaces for culturing stem cell derived oligodendrocyte progenitor cells
KR101809863B1 (en) 2008-01-30 2017-12-15 아스테리아스 바이오세라퓨틱스, 인크. Synthetic surfaces for culturing stem cell derived cardiomyocytes
WO2009098698A2 (en) 2008-02-07 2009-08-13 Shahar Cohen Compartmental extract compositions for tissue engineering
US20090202978A1 (en) * 2008-02-13 2009-08-13 Ginadi Shaham Method and apparatus for freezing of a biological material
US20100087002A1 (en) * 2008-02-21 2010-04-08 Benjamin Fryer Methods, Surface Modified Plates and Compositions for Cell Attachment, Cultivation and Detachment
CA2715878C (en) 2008-02-21 2017-06-13 Centocor Ortho Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
EP2100954A1 (en) 2008-03-10 2009-09-16 Assistance Publique - Hopitaux de Paris Method for generating primate cardiac progenitor cells for clinical use from primate embryonic stem cells, and their applications
DE102008013854A1 (en) * 2008-03-12 2009-09-24 Siemens Aktiengesellschaft Catheter and associated medical examination and treatment facility
WO2009116951A2 (en) 2008-03-17 2009-09-24 Agency For Science, Technology And Research Microcarriers for stem cell culture
CA2718904C (en) 2008-03-17 2017-01-03 The Scripps Research Institute Combined chemical and genetic approaches for generation of induced pluripotent stem cells
EP2268794B1 (en) 2008-03-27 2017-09-13 Asterias Biotherapeutics, Inc. Differentiation of primate pluripotent stem cells to hematopoietic lineage cells
US8338170B2 (en) 2008-04-21 2012-12-25 Viacyte, Inc. Methods for purifying endoderm and pancreatic endoderm cells derived from human embryonic stem cells
WO2009131568A1 (en) 2008-04-21 2009-10-29 Cythera, Inc. Methods for purifying endoderm and pancreatic endoderm cells derived from human embryonic stem cells
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US7939322B2 (en) 2008-04-24 2011-05-10 Centocor Ortho Biotech Inc. Cells expressing pluripotency markers and expressing markers characteristic of the definitive endoderm
EP3851520A1 (en) 2008-04-30 2021-07-21 SanBio, Inc. Neural regenerating cells with alterations in dna methylation
JP5346925B2 (en) * 2008-05-02 2013-11-20 国立大学法人京都大学 Nuclear initialization method
AU2009241762B2 (en) * 2008-05-02 2015-07-16 Celsense Inc. Compositions and methods for producing emulsions for nuclear magnetic resonance techniques and other applications
WO2009136867A1 (en) * 2008-05-06 2009-11-12 Agency For Science, Technology And Research Method of effecting de-differentiation of a cell
US9394538B2 (en) 2008-05-07 2016-07-19 Shi-Lung Lin Development of universal cancer drugs and vaccines
ES2613965T3 (en) 2008-05-09 2017-05-29 Regenics As Cell extracts
EP2297298A4 (en) * 2008-05-09 2011-10-05 Vistagen Therapeutics Inc Pancreatic endocrine progenitor cells derived from pluripotent stem cells
US20120052003A9 (en) * 2008-05-16 2012-03-01 Szalay Aladar A Microorganisms for preventing and treating neoplasms accompanying cellular therapy
EP2294187A2 (en) * 2008-05-21 2011-03-16 BioE LLC Differentiation of multi-lineage progenitor cells to pancreatic cells
DK2297319T3 (en) 2008-06-03 2015-10-19 Viacyte Inc GROWTH FACTORS FOR PREPARING DEFINITIVE ENDODERM
US20090298178A1 (en) * 2008-06-03 2009-12-03 D Amour Kevin Allen Growth factors for production of definitive endoderm
EP3279314A1 (en) 2008-06-04 2018-02-07 Cellular Dynamics International, Inc. Methods for the production of ips cells using non-viral approach
EP2300611B1 (en) 2008-06-13 2017-08-09 Whitehead Institute for Biomedical Research Programming and reprogramming of cells
US8871900B2 (en) 2008-06-16 2014-10-28 University Of Rochester Fibroblast growth factor (FGF) analogs and uses thereof
US8669048B2 (en) 2008-06-24 2014-03-11 Parkinson's Institute Pluripotent cell lines and methods of use thereof
KR20180018839A (en) 2008-06-30 2018-02-21 얀센 바이오테크 인코포레이티드 Differentiation of pluripotent stem cells
US20110305672A1 (en) 2008-07-25 2011-12-15 University Of Georgia Research Foundation, Inc. COMPOSITIONS FOR MESODERM DERIVED ISL1+ MULTIPOTENT CELLS (IMPs), EPICARDIAL PROGENITOR CELLS (EPCs) AND MULTIPOTENT CD56C CELLS (C56Cs) AND METHODS OF PRODUCING AND USING SAME
US20100028307A1 (en) * 2008-07-31 2010-02-04 O'neil John J Pluripotent stem cell differentiation
JP2012507289A (en) 2008-10-31 2012-03-29 ヤンセン バイオテツク,インコーポレーテツド Differentiation of human embryonic stem cells into the pancreatic endocrine system
US9234178B2 (en) 2008-10-31 2016-01-12 Janssen Biotech, Inc. Differentiation of human pluripotent stem cells
CA2907326A1 (en) 2008-11-04 2010-05-14 Chad Green Stem cell aggregate suspension compositions and methods for differentiation thereof
US8956867B2 (en) * 2008-11-07 2015-02-17 Wisconsin Alumni Research Foundation Method for culturing stem cells
EP2356227B1 (en) 2008-11-14 2018-03-28 Viacyte, Inc. Encapsulation of pancreatic cells derived from human pluripotent stem cells
MX356756B (en) 2008-11-20 2018-06-11 Centocor Ortho Biotech Inc Pluripotent stem cell culture on micro-carriers.
EP2366022B1 (en) * 2008-11-20 2016-04-27 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
BRPI0922572A2 (en) 2008-12-17 2019-09-24 Scripps Research Inst method for culturing pluripotent cells, pluripotent mammalian cell culture, cell culture medium, isolated pluripotent animal cell, and method for increasing pluripotence of a mammalian cell.
EP2379087B1 (en) * 2008-12-19 2014-08-20 DePuy Synthes Products, LLC Umbilical cord tissue derived cells for treating neuropathic pain and spasticity
US10557116B2 (en) 2008-12-19 2020-02-11 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
EP2379089B1 (en) * 2008-12-19 2019-04-17 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue following injury
CN102325878A (en) 2008-12-23 2012-01-18 斯特姆塞尔思加利福尼亚有限公司 Target populations of oligodendrocyte precursor cells and methods of making and using same
US20120202287A1 (en) 2009-01-22 2012-08-09 David Roger Adams Stem Cell Culture Methods
US20100209399A1 (en) * 2009-02-13 2010-08-19 Celavie Biosciences, Llc Brain-derived stem cells for repair of musculoskeletal system in vertebrate subjects
EP2233566A1 (en) 2009-03-17 2010-09-29 Vrije Universiteit Brussel Generation of pancreatic progenitor cells
AU2010229651B2 (en) 2009-03-26 2014-05-08 Advanced Technologies And Regenerative Medicine, Llc Human umbilical cord tissue cells as therapy for Alzheimer' s disease
GB0906424D0 (en) 2009-04-09 2009-05-20 Antoxis Ltd Use of compounds for differentiation of cells
WO2010120830A1 (en) * 2009-04-13 2010-10-21 Northwestern University Novel peptide-based scaffolds for cartilage regeneration and methods for their use
US9109245B2 (en) 2009-04-22 2015-08-18 Viacyte, Inc. Cell compositions derived from dedifferentiated reprogrammed cells
DK2421957T3 (en) * 2009-04-22 2021-01-25 Viacyte Inc CELL COMPOSITIONS DERIVED FROM DEDIFFERENTIATED PROGRAMMED CELLS
WO2010144696A1 (en) 2009-06-11 2010-12-16 Burnham Institute For Medical Research Directed differentiation of stem cells
US8703822B2 (en) 2009-06-12 2014-04-22 Nanyang Polytechnic Method for removing undifferentiated and dedifferentiated stem cell
WO2010144780A1 (en) * 2009-06-12 2010-12-16 University Of Kansas Compositions and methods for establishing and maintaining stem cells in an undifferentiated state
GB0911060D0 (en) 2009-06-26 2009-08-12 Ge Healthcare Uk Ltd Methods for predicting the toxicity of a chemical
WO2011005326A1 (en) 2009-07-09 2011-01-13 Massachusetts Institute Of Technology Methods and compositions for increased safety of stem cell-derived populations
JP5819825B2 (en) * 2009-07-20 2015-11-24 ヤンセン バイオテツク,インコーポレーテツド Differentiation of human embryonic stem cells
US8785185B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
JP6219568B2 (en) 2009-07-20 2017-10-25 ヤンセン バイオテツク,インコーポレーテツド Differentiation of human embryonic stem cells
AU2010276201B2 (en) 2009-07-21 2013-10-17 Abt Holding Company Use of stem cells to reduce leukocyte extravasation
EP2456860B1 (en) 2009-07-21 2018-09-05 ABT Holding Company Use of stem cells to reduce leukocyte extravasation
WO2011017315A2 (en) 2009-08-03 2011-02-10 Recombinetics, Inc. Methods and compositions for targeted gene modification
JP5761816B2 (en) 2009-08-12 2015-08-12 国立大学法人京都大学 Differentiation induction method from pluripotent stem cells to neural progenitor cells
EP2467470A2 (en) 2009-08-17 2012-06-27 Technion Research & Development Foundation Ltd. Pericyte progenitor cells and methods of generating and using same
ES2399711T3 (en) 2009-08-22 2013-04-02 The Board Of Trustees Of The University Of The Leland Stanford Junior University Obtaining images and evaluation of embryos, oocytes and stem cells
AU2010289423B2 (en) 2009-09-04 2014-03-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods for enhancing genome stability and telomere elongation in embryonic stem cells
MX337982B (en) 2009-10-16 2016-03-30 Scripps Research Inst Induction of pluripotent cells.
AU2010315712B2 (en) 2009-10-19 2014-04-17 FUJIFILM Cellular Dynamics, Inc. Cardiomyocyte production
EP2494035B1 (en) * 2009-10-29 2018-02-28 Janssen Biotech, Inc. Pluripotent stem cells
WO2011057249A2 (en) 2009-11-09 2011-05-12 The Brigham And Women's Hospital, Inc. Treatment of heart disease
CA2783437C (en) 2009-11-12 2020-08-04 Technion Research & Development Foundation Ltd. Culture media, cell cultures and methods of culturing pluripotent stem cells in an undifferentiated state
WO2011068879A2 (en) * 2009-12-02 2011-06-09 Research Development Foundation Selection of stem cell clones with defined differentiation capabilities
FI20096288A0 (en) 2009-12-04 2009-12-04 Kristiina Rajala Formulations and Methods for Culturing Stem Cells
CN102725399B (en) 2009-12-18 2015-03-11 上海赛傲生物技术有限公司 Materials and methods for generating pluripotent stem cells
RU2664864C1 (en) 2009-12-23 2018-08-23 Янссен Байотек, Инк. Ways to increase expression of ngn3 and nkx6.1 in pancreatic endocrine cells
MX343786B (en) 2009-12-23 2016-11-22 Janssen Biotech Inc Differentiation of human embryonic stem cells.
WO2011091944A1 (en) 2010-01-26 2011-08-04 Université Libre de Bruxelles Tools for isolating and following cardiovascular progenitor cells
EP2533859B1 (en) 2010-02-10 2016-04-06 Nayacure Therapeutics Ltd Pharmaceutical compositions for the treatment and prevention of cancer
BR112012021451B8 (en) 2010-02-25 2021-05-25 Abt Holding Co use of cells that have a desired efficiency for expression and/or secretion of the pro-angiogenic factors vegf, cxcl5 and il8, methods to build a cell bank, to develop drug, and to increase the expression of one or more pro-angiogenic factors angiogenic in a cell performed in vitro, and composition
SG10201501402UA (en) 2010-02-25 2015-04-29 Abt Holding Co Modulation of macrophage activation
JP6013196B2 (en) 2010-03-01 2016-10-25 ヤンセン バイオテツク,インコーポレーテツド Method for purifying cells derived from pluripotent stem cells
US8662085B2 (en) * 2010-03-02 2014-03-04 Siemens Aktiengesellschaft Magnetic nanoparticle and group of nanoparticles
WO2011119637A1 (en) 2010-03-22 2011-09-29 Stemina Biomarker Discovery, Inc. Predicting human developmental toxicity of pharmaceuticals using human stem-like cells and metabolomics
WO2011119507A2 (en) 2010-03-25 2011-09-29 International Stem Cell Corporation Method of altering the differentiative state of a cell and compositions thereof
EP3199623B1 (en) 2010-03-31 2021-07-28 The Scripps Research Institute Reprogramming cells
US9029145B2 (en) 2010-04-08 2015-05-12 The University Court Of The University Of Edinburgh Chondrogenic progenitor cells, protocol for derivation of cells and uses thereof
US8927274B2 (en) 2010-04-12 2015-01-06 Technion Research & Development Foundation Limited Populations of pancreatic progenitor cells and methods of isolating and using same
US8815592B2 (en) 2010-04-21 2014-08-26 Research Development Foundation Methods and compositions related to dopaminergic neuronal cells
JP5841322B2 (en) 2010-04-22 2016-01-13 オレゴン ヘルス アンド サイエンス ユニバーシティ Fumaryl acetoacetate hydrolase (FAH) deficient pig and use thereof
PT2561078T (en) 2010-04-23 2018-12-03 Cold Spring Harbor Laboratory Novel structurally designed shrnas
AU2011249478B2 (en) 2010-05-06 2014-12-04 Regenics As Use of cellular extracts for skin rejuvenation
CN102242146B (en) * 2010-05-10 2015-11-25 高丽大学校产学协力团 Composition and the method with its generation generate induced pluripotent stem cells
SG185526A1 (en) 2010-05-12 2012-12-28 Abt Holding Co Modulation of splenocytes in cell therapy
RU2587634C2 (en) 2010-05-12 2016-06-20 Янссен Байотек, Инк. Differentiation of human embryo stem cells
EP2580320B1 (en) 2010-06-14 2018-08-01 The Scripps Research Institute Reprogramming of cells to a new fate
WO2011158125A2 (en) 2010-06-17 2011-12-22 Katholieke Universiteit Leuven Methods for differentiating cells into hepatic stellate cells and hepatic sinusoidal endothelial cells, cells produced by the methods, and methods for using the cells
US9365826B2 (en) 2010-06-18 2016-06-14 Cellular Dynamics International, Inc. Cardiomyocyte medium with dialyzed serum
CA2802896C (en) 2010-07-01 2021-05-11 Regenerative Research Foundation Methods for culturing undifferentiated cells using sustained release compositions
CA2804595C (en) 2010-07-07 2018-11-13 Cellular Dynamics International, Inc. Endothelial cell production by programming
WO2012009377A2 (en) 2010-07-12 2012-01-19 University Of Southern California Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
JP5896360B2 (en) 2010-07-21 2016-04-13 国立大学法人京都大学 Method for inducing differentiation from human pluripotent stem cells to intermediate mesoderm cells
US20130122589A1 (en) 2010-07-26 2013-05-16 The University Of Manchester Targeted differentiation of stem cells
AU2011282642B2 (en) 2010-07-28 2015-06-11 Shinn-Zong Lin Methods for treating and/or reversing neurodegenerative diseases and/or disorders
SG10201506264QA (en) 2010-08-12 2015-09-29 Janssen Biotech Inc Treatment of diabetes with pancreatic endocrine precursor cells
CN103168236B (en) 2010-08-23 2016-01-20 哈佛大学管理委员会 For the light genetics probe that membrane potential measures
AU2011293440B2 (en) 2010-08-24 2016-05-05 Katholieke Universiteit Leuven Non-static suspension culture of cell aggregates
KR101851956B1 (en) 2010-08-31 2018-04-25 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
ES2585028T3 (en) 2010-08-31 2016-10-03 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
ES2659393T3 (en) 2010-08-31 2018-03-15 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
CA2809374C (en) 2010-09-01 2021-02-23 Thomas Jefferson University A composition comprising a retinoic acid receptor gamma agonist and its use for muscle repair and regeneration
US10214722B2 (en) 2010-09-07 2019-02-26 Technion Research & Development Foundation Limited Methods for expanding and maintaining human pluripotent stem cells (PSCs) in an undifferentiated state in a single cell suspension culture
CA2810444A1 (en) 2010-09-07 2012-03-15 The Board Of Regents Of The University Of Texas System Tissue-specific differentiation matrices and uses thereof
WO2012037456A1 (en) 2010-09-17 2012-03-22 President And Fellows Of Harvard College Functional genomics assay for characterizing pluripotent stem cell utility and safety
AU2011308567B2 (en) 2010-10-01 2015-09-03 Fundacion Centro Nacional De Investigaciones Oncologicas, Carlos Iii Manipulation of stem cell function by p53 isoforms
US9725689B2 (en) 2010-10-08 2017-08-08 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
CA2814860C (en) 2010-10-22 2020-08-25 Biotime Inc. Methods of modifying transcriptional regulatory networks in stem cells
EP2655601A4 (en) 2010-12-22 2014-09-10 Fate Therapeutics Inc Cell culture platform for single cell sorting and enhanced reprogramming of ipscs
WO2012091978A2 (en) 2010-12-31 2012-07-05 University Of Georgia Research Foundation, Inc. Differentiation of human pluripotent stem cells to multipotent neural crest cells
JP6012629B2 (en) 2011-01-25 2016-10-25 ユニヴェルシテ カソリック ド ルーヴァンUniversite Catholique De Louvain Compositions and methods for cell transplantation
WO2012105979A1 (en) 2011-02-03 2012-08-09 Empire Technology Development Llc 3d trophoblast matrix for preparing organ-specific stem cells
US9574179B2 (en) 2011-02-08 2017-02-21 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
JP5995247B2 (en) 2011-02-23 2016-09-21 国立大学法人京都大学 Method for producing dendritic cells from pluripotent stem cells
WO2012116185A1 (en) 2011-02-23 2012-08-30 The Board Of Trustees Of The Leland Stanford Junior University Methods of detecting aneuploidy in human embryos
EP2681555B1 (en) 2011-02-25 2020-04-08 Benaroya Research Institute Detection of an allergic disorder
DK2681306T3 (en) 2011-02-28 2019-04-23 Harvard College CELL CULTURE SYSTEM
EP2801377B1 (en) 2011-03-04 2019-08-07 The Regents of The University of California Hydrogel comprising cells for local release of growth factors to mediate motor recovery after stroke
US20140329314A1 (en) 2011-03-29 2014-11-06 Christopher O'Sullivan Enriched populations of cardiomyocyte lineage cells from pluripotent stem cells
JP5840855B2 (en) 2011-03-30 2016-01-06 学校法人東京女子医科大学 Method for producing myocardial sheet from embryonic stem cells
DK2694644T3 (en) 2011-03-30 2018-04-16 Cellular Dynamics Int Inc Priming of pluripotent stem cells for neural differentiation
US8785190B2 (en) 2011-04-06 2014-07-22 Sanbio, Inc. Methods and compositions for modulating peripheral immune function
JP5745919B2 (en) * 2011-04-28 2015-07-08 浜松ホトニクス株式会社 Cell analysis method, cell analysis apparatus, and cell analysis program
US10478206B2 (en) 2011-04-29 2019-11-19 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
PL2718416T3 (en) 2011-06-06 2020-05-18 ReGenesys BVBA Expansion of stem cells in hollow fiber bioreactors
GB201110331D0 (en) 2011-06-16 2011-08-03 Isis Innovation Method of cryopreserving pluripotent stem cells
WO2013010045A1 (en) 2011-07-12 2013-01-17 Biotime Inc. Novel methods and formulations for orthopedic cell therapy
WO2013010965A1 (en) 2011-07-15 2013-01-24 Universite Libre De Bruxelles Generation of mesodermal cells from pluripotent stem cells
CA2841985C (en) 2011-07-29 2019-09-17 Cellular Dynamics International, Inc. Metabolic maturation in stem cell-derived tissue cells
WO2013021389A2 (en) 2011-08-09 2013-02-14 Yeda Research And Development Co.Ltd. Downregulation of mir-7 for promotion of beta cell differentiation and insulin production
EP3578042A1 (en) 2011-08-26 2019-12-11 Yecuris Corporation Fumarylacetoacetate hydrolase (fah)-deficient and immunodeficient rats and uses thereof
BR112014007676A2 (en) 2011-09-30 2017-04-18 The Univ Of Miami isolated renal stem cells from kidney
GB201117469D0 (en) 2011-10-10 2011-11-23 Cell Guidance Systems Ltd Culture media for pluripotent stem cells
WO2013058403A1 (en) 2011-10-21 2013-04-25 国立大学法人京都大学 Method for culturing pluripotency-maintained singly dispersed cells by means of laminar flow
GB201119335D0 (en) 2011-11-09 2011-12-21 Univ Leuven Kath Hepatitis virus infectable stem cells
EP2594635A1 (en) 2011-11-18 2013-05-22 Univercell Biosolutions Method for generating primate cardiovascular progenitor cells for clinical and drug cells testing use from primate embryonic stem cells or embryonic-like state cells, and their applications
WO2013077423A1 (en) 2011-11-25 2013-05-30 国立大学法人京都大学 Method for culturing pluripotent stem cell
US10238755B2 (en) 2011-11-30 2019-03-26 The Wistar Institute Of Anatomy And Biology Methods and compositions for regulation of cell aging, carcinogenesis and reprogramming
WO2013082509A1 (en) 2011-12-01 2013-06-06 The New York Stem Cell Foundation Automated system for producing induced pluripotent stem cells or differentiated cells
WO2013082106A1 (en) 2011-12-02 2013-06-06 The General Hospital Corporation Differentiation into brown adipocytes
KR20210134808A (en) 2011-12-05 2021-11-10 팩터 바이오사이언스 인크. Methods and products for transfecting cells
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
US8497124B2 (en) * 2011-12-05 2013-07-30 Factor Bioscience Inc. Methods and products for reprogramming cells to a less differentiated state
WO2013094771A1 (en) 2011-12-19 2013-06-27 Kyoto University Method for inducing differentiation of human pluripotent stem cells into intermediate mesoderm cells
RU2705001C2 (en) 2011-12-22 2019-11-01 Янссен Байотек, Инк. Differentiation of human embryonic stem cells into single-hormonal insulin-positive cells
SG11201403465PA (en) 2011-12-23 2014-10-30 Atrm Llc Detection of human umbilical cord tissue-derived cells
EP2615176A1 (en) 2012-01-11 2013-07-17 Paul-Ehrlich-Institut Bundesamt für Sera und Impfstoffe Novel pseudotyped lentiviral particles and their use in the in vitro targeted transduction of undifferentiated pluripotent human embryonic stem cells and induced pluripotent stem cells
ES2833280T3 (en) 2012-01-13 2021-06-14 Massachusetts Gen Hospital Isolated human lung progenitor cells and their uses
US9775890B2 (en) 2012-01-25 2017-10-03 Université Catholique de Louvain Factor Xa inhibitor used with liver-derived progenitor cells
US9295756B2 (en) 2012-02-01 2016-03-29 Nayacure Therapeutics Ltd. Methods for inducing immune tolerance to organ transplants
WO2013124816A2 (en) 2012-02-22 2013-08-29 Brainstem Biotec Ltd. Generation of neural stem cells and motor neurons
CA2866590A1 (en) 2012-03-07 2013-09-12 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
WO2013151186A1 (en) 2012-04-06 2013-10-10 国立大学法人京都大学 Method for inducing erythropoietin-producing cell
RU2014147015A (en) 2012-04-24 2016-06-10 Интернэшнл Стем Селл Корпорейшн OBTAINING NERVOUS STEM CELLS AND DOPAMINERGIC NEURONS FROM HUMAN PLURIPOTENT STEM CELLS
WO2013163296A1 (en) 2012-04-24 2013-10-31 The Brigham And Women's Hospital, Inc. Generating pluripotent cells de novo
LT2844739T (en) 2012-04-30 2019-10-10 University Health Network METHODS AND COMPOSITIONS FOR GENERATING PANCREATIC PROGENITORS AND FUNCTIONAL BETA CELLS FROM hPSCs
JP5432322B2 (en) 2012-05-08 2014-03-05 株式会社大塚製薬工場 Mammalian cell suspension for prevention of pulmonary embolism containing trehalose
TR201806812T4 (en) 2012-05-25 2018-06-21 Charpentier Emmanuelle Methods and compositions for RNA-directed target DNA modification and for RNA-directed transcription modification.
CA2875038A1 (en) 2012-05-31 2013-12-05 Auxogyn, Inc. In vitro embryo blastocyst prediction methods
JP6469003B2 (en) 2012-06-08 2019-02-13 ヤンセン バイオテツク,インコーポレーテツド Differentiation of human embryonic stem cells into pancreatic endocrine cells
KR102055438B1 (en) 2012-07-31 2019-12-19 에이지엑스 쎄라퓨틱스, 인크. Hla g-modified cells and methods
EP2925332A4 (en) 2012-08-20 2016-12-21 Boris Markosian Placental vaccination therapy for cancer
WO2014052912A1 (en) 2012-09-28 2014-04-03 Scripps Health Methods of differentiating stem cells into chondrocytes
AU2013237760A1 (en) 2012-10-08 2014-04-24 Biotime, Inc. Differentiated progeny of clonal progenitor cell lines
EP2912166B1 (en) 2012-10-29 2019-05-01 Scripps Health Methods of producing pluripotent stem cells from chondrocytes
JP6324395B2 (en) 2012-10-29 2018-05-16 スクリップス ヘルス Method of transplanting chondrocytes
CN104769112A (en) 2012-11-01 2015-07-08 菲克特生物科学股份有限公司 Methods and products for expressing proteins in cells
CA2888064C (en) 2012-11-02 2023-05-30 Stemina Biomarker Discovery, Inc. Predicting human developmental toxicity of pharmaceuticals using human stem-like cells and metabolomic ratios
AU2013248265B2 (en) 2012-11-08 2018-11-01 Viacyte, Inc. Scalable primate pluripotent stem cell aggregate suspension culture and differentiation thereof
EP3459522B1 (en) 2012-12-10 2021-02-17 Regenics AS Use of egg cellular extracts for wound treatment
SG11201505152PA (en) 2012-12-28 2015-08-28 Univ Kyoto Method for inducing astrocytes
JP6557146B2 (en) 2012-12-31 2019-08-07 ヤンセン バイオテツク,インコーポレーテツド Culture of human embryonic stem cells at the air-liquid interface for differentiation from pluripotent stem cells to pancreatic endocrine cells
EP4039798A1 (en) * 2012-12-31 2022-08-10 Janssen Biotech, Inc. Suspension and clustering of human pluripotent cells
EP2938723B1 (en) 2012-12-31 2023-02-01 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
ES2755878T3 (en) 2013-02-01 2020-04-24 Ares Trading Sa Abnormal phenotypes of singamia observed with imaging at pre-established time intervals for early identification of embryos with inferior development potential
US20160002599A1 (en) 2013-02-08 2016-01-07 Kyoto University Production methods for megakaryocytes and platelets
JP2016508520A (en) 2013-02-15 2016-03-22 インターナショナル ステム セル コーポレイション Use of neurons derived from human pluripotent stem cells for the treatment of neurodegenerative diseases
US20140242595A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
EP2966163B1 (en) 2013-03-06 2018-01-17 Kyoto University Culture system for pluripotent stem cells and method for subculturing pluripotent stem cells
WO2014143383A1 (en) 2013-03-13 2014-09-18 Agilent Technologies, Inc. Transposome tethered to a gene delivery vehicle
US8859286B2 (en) 2013-03-14 2014-10-14 Viacyte, Inc. In vitro differentiation of pluripotent stem cells to pancreatic endoderm cells (PEC) and endocrine cells
CA2899507A1 (en) 2013-03-14 2014-09-25 The Regents Of The University Of California In vitro production of medial ganglionic eminence precursor cells
JP7182852B2 (en) 2013-03-15 2022-12-05 ホワイトヘッド・インスティテュート・フォー・バイオメディカル・リサーチ A Cellular Discovery Platform for Neurodegenerative Diseases
EP2977449B1 (en) 2013-03-21 2020-02-26 Kyoto University Pluripotent stem cell for neuronal differentiation induction
EP2980207B1 (en) 2013-03-25 2018-12-05 Foundation for Biomedical Research and Innovation at Kobe Cell sorting method
JP6602288B2 (en) 2013-04-03 2019-11-06 フジフィルム セルラー ダイナミクス,インコーポレイテッド Methods and compositions for culturing endoderm progenitor cells in suspension
SG10201708195RA (en) 2013-04-05 2017-11-29 Univ Health Network Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
EP2983728B1 (en) 2013-04-10 2018-06-06 Agency For Science, Technology And Research Polycaprolactone microcarriers for stem cell culture and fabrication thereof
WO2014168264A1 (en) 2013-04-12 2014-10-16 国立大学法人京都大学 Method for inducing alveolar epithelium progenitor cells
ES2827250T3 (en) 2013-04-12 2021-05-20 Houston Methodist Hospital Improvement of organs for transplantation
CN105531365A (en) 2013-04-23 2016-04-27 耶达研究及发展有限公司 Isolated naive pluripotent stem cells and methods of generating same
DK2992088T3 (en) 2013-04-30 2019-11-11 Univ Leuven Kath CELL THERAPY FOR MYELODYSPLASTIC SYNDROMES
WO2014185358A1 (en) 2013-05-14 2014-11-20 国立大学法人京都大学 Efficient myocardial cell induction method
US9758763B2 (en) 2013-05-29 2017-09-12 The Regents Of The University Of California Methods and compositions for somatic cell proliferation and viability
WO2014197421A1 (en) 2013-06-05 2014-12-11 Biotime, Inc. Compositions and methods for induced tissue regeneration in mammalian species
US10119122B2 (en) 2013-06-10 2018-11-06 Academisch Ziekenhuis Leiden Differentiation and expansion of endothelial cells from pluripotent stem cells and the in vitro formation of vasculature like structures
EP3008229B1 (en) 2013-06-10 2020-05-27 President and Fellows of Harvard College Early developmental genomic assay for characterizing pluripotent stem cell utility and safety
CA2915085C (en) 2013-06-11 2021-04-27 Kyoto University Method for producing renal progenitor cells and drug comprising the same
US20170029778A1 (en) 2013-06-11 2017-02-02 President And Fellows Of Harvard College Sc-beta cells and compositions and methods for generating the same
WO2014201254A1 (en) 2013-06-12 2014-12-18 University Of Washington Through Its Center For Commercialization Methods for maturing cardiomyocytes and uses thereof
US10240126B2 (en) 2013-06-12 2019-03-26 Kyoto University Induced pluripotent stem cell selection method and method for inducing differentiation to blood cells
WO2014208053A1 (en) 2013-06-28 2014-12-31 株式会社大塚製薬工場 Trehalose and dextran-containing solution for transplanting mammalian cells
JP6378183B2 (en) 2013-08-07 2018-08-22 国立大学法人京都大学 Method for producing pancreatic hormone-producing cells
WO2015023720A1 (en) 2013-08-16 2015-02-19 Yale University Epithelial cell differentiation of human mesenchymal stromal cells
EP3569699A1 (en) 2013-09-04 2019-11-20 Otsuka Pharmaceutical Factory, Inc. Method for preparing pluripotent stem cells
MY184219A (en) 2013-09-05 2021-03-26 Univ Kyoto New method for inducing dopamine-producing neural precursor cells
AU2014321101B2 (en) 2013-09-13 2020-12-03 University Health Network Methods and compositions for generating epicardium cells
CN105960453A (en) 2013-10-01 2016-09-21 卡迪马干细胞有限公司 Directed differentiation of astrocytes from human pluripotent stem cells for use in drug screening and the treatment of amyotrophic lateral sclerosis (ALS)
WO2015064754A1 (en) 2013-11-01 2015-05-07 国立大学法人京都大学 Novel chondrocyte induction method
US11767507B2 (en) 2013-11-08 2023-09-26 The Mclean Hospital Corporation Methods for efficient generation of GABAergic interneurons from pluripotent stem cells
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
WO2015073913A1 (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
WO2015087231A1 (en) 2013-12-11 2015-06-18 Pfizer Limited Method for producing retinal pigment epithelial cells
EP3835419A1 (en) 2013-12-12 2021-06-16 The Regents of The University of California Methods and compositions for modifying a single stranded target nucleic acid
EP2896688A1 (en) 2014-01-20 2015-07-22 Centre National de la Recherche Scientifique (CNRS) A method of producing beta pancreatic cells from progenitor cells through the use of hydrogen peroxide
EP3096713B1 (en) 2014-01-23 2022-09-21 President and Fellows of Harvard College Engineered polymeric valves, tubular structures and sheets
FI3690056T3 (en) 2014-01-31 2023-03-19 Factor Bioscience Inc Methods and products for nucleic acid production and delivery
WO2015119642A1 (en) 2014-02-10 2015-08-13 The Johns Hopkins University Low oxygen tension enhances endothelial fate of human pluripotent stem cells
US11078462B2 (en) 2014-02-18 2021-08-03 ReCyte Therapeutics, Inc. Perivascular stromal cells from primate pluripotent stem cells
KR102340553B1 (en) 2014-03-04 2021-12-21 페이트 세러퓨틱스, 인코포레이티드 Improved reprogramming methods and cell culture platforms
WO2015140005A1 (en) 2014-03-19 2015-09-24 Ifom Fondazione Istituto Firc Di Oncologia Molecolare Method of generation of pluripotent cells
WO2015140257A1 (en) 2014-03-19 2015-09-24 INSERM (Institut National de la Santé et de la Recherche Médicale) A method for inducing human cholangiocyte differentiation
CN112111446A (en) 2014-03-19 2020-12-22 V 细胞治疗公司 Methods relating to pluripotent cells
ES2837840T3 (en) 2014-03-20 2021-07-01 Ares Trading Sa Quantitative measurement of developmental kinetics of human morula and blastocyst morphology
ES2939807T3 (en) 2014-03-21 2023-04-27 Fujifilm Cellular Dynamics Inc Production of midbrain dopaminergic neurons and methods for their utilization
US20170107486A1 (en) 2014-04-21 2017-04-20 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
CA2949056A1 (en) 2014-05-16 2015-11-19 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
EP3149155B1 (en) 2014-06-02 2020-09-23 Kadimastem Ltd. Methods of inducing myelination and maturation of oligodendrocytes
US10240127B2 (en) 2014-07-03 2019-03-26 ReCyte Therapeutics, Inc. Exosomes from clonal progenitor cells
EP3170901B1 (en) 2014-07-14 2021-06-02 Chugai Seiyaku Kabushiki Kaisha Method for producing dendritic cells from stem cells
CN108064274A (en) 2014-07-30 2018-05-22 耶达研究及发展有限公司 For cultivating the culture medium of multipotential stem cell
WO2016025510A1 (en) 2014-08-12 2016-02-18 Rappolee Daniel A Systems and methods to detect stem cell stress and uses thereof
US10596200B2 (en) 2014-08-22 2020-03-24 Procella Therapeutics Ab Use of LIFR or FGFR3 as a cell surface marker for isolating human cardiac ventricular progenitor cells
US10597637B2 (en) 2014-08-22 2020-03-24 Procella Therapeutics Ab Use of jagged 1/frizzled 4 as a cell surface marker for isolating human cardiac ventricular progenitor 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
WO2016040794A1 (en) 2014-09-12 2016-03-17 Whitehead Institute For Biomedical Research Cells expressing apolipoprotein e and uses thereof
KR102473092B1 (en) 2014-09-15 2022-12-01 칠드런'즈 메디컬 센터 코포레이션 Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
US9371516B2 (en) 2014-09-19 2016-06-21 Regenerative Medical Solutions, Inc. Compositions and methods for differentiating stem cells into cell populations comprising beta-like cells
US10047341B2 (en) 2014-10-14 2018-08-14 FUJIFILM Cellular Dynamics, Inc. Generation of keratinocytes from pluripotent stem cells and maintenance of keratinocyte cultures
KR20190060016A (en) 2014-10-20 2019-05-31 뉴럴스템, 인크. Stable neural stem cells comprising an exogenous polynucleotide coding for a growth factor and methods of use thereof
EP3868873A1 (en) 2014-10-24 2021-08-25 Sumitomo Dainippon Pharma Co., Ltd. Production method for retinal tissue
EP3220945A2 (en) 2014-11-17 2017-09-27 Yeda Research and Development Co., Ltd. Methods of treating diseases related to mitochondrial function
EP3223831B1 (en) 2014-11-25 2020-06-24 President and Fellows of Harvard College Methods for generation of podocytes from pluripotent stem cells and cells produced by the same
JP6800854B2 (en) 2014-12-19 2020-12-16 ヤンセン バイオテツク,インコーポレーテツド Suspension culture of pluripotent stem cells
WO2016103269A1 (en) 2014-12-23 2016-06-30 Ramot At Tel-Aviv University Ltd. Populations of neural progenitor cells and methods of producing and using same
JP6882981B2 (en) 2014-12-30 2021-06-02 セル キュア ニューロサイエンシズ リミテッド RPE cell population and how to make it
ES2880346T3 (en) 2014-12-30 2021-11-24 Cell Cure Neurosciences Ltd Evaluation of retinal pigment epithelial cell populations
WO2016118824A1 (en) 2015-01-22 2016-07-28 Regenerative Medical Solutions, Inc. Markers for differentiation of stem cells into differentiated cell populations
AU2016218977C1 (en) 2015-02-13 2023-03-23 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10561687B2 (en) 2015-02-17 2020-02-18 University Health Network Methods for making and using sinoatrial node-like pacemaker cardiomyocytes and ventricular-like cardiomyocytes
EP3059307B2 (en) 2015-02-20 2022-05-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of a laminin for differentiating pluripotent cells into hepatocyte lineage cells
WO2016143803A1 (en) 2015-03-06 2016-09-15 国立大学法人京都大学 Method for inducing differentiation of alveolar epithelial cells
CA2979293C (en) 2015-03-11 2022-01-04 Timothy J. Kieffer Pancreatic endocrine progenitor cell therapies for the treatment of obesity and type 2 diabetes (t2d)
WO2016185457A1 (en) 2015-05-19 2016-11-24 Yeda Research And Development Co. Ltd. Methods of promoting lymphangiogenesis
CN108138129B (en) 2015-07-17 2021-11-16 国立大学法人京都大学 Method for inducing vascular endothelial cells
US11230696B2 (en) 2015-07-29 2022-01-25 Hadasit Medical Research Services And Development Ltd. Large scale production of retinal pigment epithelial cells
CN108138144A (en) 2015-08-05 2018-06-08 细胞治疗神经科学有限公司 The preparation of retinal pigment epithelium
EA201890442A1 (en) 2015-08-05 2018-07-31 Селл Кьюр Нейросайансес Лтд. OBTAINING PHOTORECEPTORS FOR THE TREATMENT OF RETITAL DISEASES
US11312940B2 (en) 2015-08-31 2022-04-26 University Of Louisville Research Foundation, Inc. Progenitor cells and methods for preparing and using the same
US11162070B2 (en) 2015-09-08 2021-11-02 FUJIFILM Cellular Dynamics, Inc. MACS-based purification of stem cell-derived retinal pigment epithelium
US11649432B2 (en) 2015-09-08 2023-05-16 Sumitomo Pharma Co., Ltd. Method for producing retinal pigment epithelial cells
CN114807035B (en) 2015-09-08 2024-02-02 (由卫生与公众服务部部长代表的)美利坚合众国 Reproducible differentiation method of clinical grade retinal pigment epithelial cells
MX2018003036A (en) 2015-09-11 2018-05-02 Astellas Pharma Inc Method for producing kidney progenitor cells.
US20190048340A1 (en) 2015-09-24 2019-02-14 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
EP3362570A4 (en) 2015-10-16 2019-03-20 Fate Therapeutics, Inc. Platform for the induction & maintenance of ground state pluripotency
US10093903B2 (en) 2015-10-19 2018-10-09 FUJIFILM Cellular Dynamics, Inc. Production of virus-receptive pluripotent stem cell (PSC)-derived hepatocytes
US10865381B2 (en) 2015-10-20 2020-12-15 FUJIFILM Cellular Dynamics, Inc. Multi-lineage hematopoietic precursor cell production by genetic programming
WO2017072763A1 (en) 2015-10-26 2017-05-04 Cell Cure Neurosciences Ltd. Preparation of retinal pigment epithelium cells
CA3003145A1 (en) 2015-10-30 2017-05-04 Gay M. Crooks Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells
BR112018009891A2 (en) 2015-11-18 2018-12-26 University Of Georgia Research Foundation, Inc. extracellular neural cell vesicles
EP3387112A4 (en) 2015-12-07 2019-08-21 BioTime, Inc. Methods for the re-derivation of diverse pluripotent stem cell-derived brown fat cells
WO2017117333A1 (en) 2015-12-30 2017-07-06 Cellular Dynamics International, Inc. Microtissue formation using stem cell-derived human hepatocytes
US10612094B2 (en) 2016-02-19 2020-04-07 Procella Therapeutics Ab Genetic markers for engraftment of human cardiac ventricular progenitor cells
EP3423158B1 (en) 2016-02-24 2023-11-15 The Rockefeller University Embryonic cell-based therapeutic candidate screening systems, models for huntington's disease and uses thereof
US11072777B2 (en) 2016-03-04 2021-07-27 University Of Louisville Research Foundation, Inc. Methods and compositions for ex vivo expansion of very small embryonic-like stem cells (VSELs)
US20190216891A1 (en) 2016-03-06 2019-07-18 Yeda Research And Development Co., Ltd. Method for modulating myelination
US11534466B2 (en) 2016-03-09 2022-12-27 Aal Scientifics, Inc. Pancreatic stem cells and uses thereof
WO2017176810A1 (en) 2016-04-04 2017-10-12 Biotime, Inc. Pluripotent stem cell-derived 3d retinal tissue and uses thereof
MA45479A (en) 2016-04-14 2019-02-20 Janssen Biotech Inc DIFFERENTIATION OF PLURIPOTENT STEM CELLS IN ENDODERMAL CELLS OF MIDDLE INTESTINE
AU2017254268B2 (en) 2016-04-22 2023-03-16 Kyoto University Method for producing dopamine-producing neural precursor cells
WO2017202814A1 (en) 2016-05-24 2017-11-30 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of neuropathological disorders characterized by a loss of cortical neurons
US11458194B2 (en) 2016-05-25 2022-10-04 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and compositions for treating cancers
WO2017210537A1 (en) 2016-06-02 2017-12-07 The Cleveland Clinic Foundation Complement inhibition for improving cell viability
EP3472321A2 (en) 2016-06-17 2019-04-24 Genesis Technologies Limited Crispr-cas system, materials and methods
MA45502A (en) 2016-06-21 2019-04-24 Janssen Biotech Inc GENERATION OF FUNCTIONAL BETA CELLS DERIVED FROM HUMAN PLURIPOTENT STEM CELLS WITH GLUCOSE-DEPENDENT MITOCHONDRIAL RESPIRATION AND TWO-PHASE INSULIN SECRETION RESPONSE
WO2018002290A1 (en) 2016-07-01 2018-01-04 Centre National De La Recherche Scientifique (Cnrs) Amplifying beta cell differentiation with small molecules bet (bromodomain and extraterminal family of bromodomain-containing proteins) inhibitors
JP7099967B2 (en) 2016-07-01 2022-07-12 リサーチ ディベロップメント ファウンデーション Elimination of Proliferative Cells from Stem Cell-Derived Grafts
EP3500664B1 (en) 2016-08-16 2021-09-22 FUJIFILM Cellular Dynamics, Inc. Methods for differentiating pluripotent cells
CN116115629A (en) 2016-08-17 2023-05-16 菲克特生物科学股份有限公司 Nucleic acid products and methods of administration thereof
CA3038690A1 (en) 2016-10-05 2018-04-12 FUJIFILM Cellular Dynamics, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
JP2019533703A (en) 2016-11-02 2019-11-21 エーエーエル サイエンティフィックス,インコーポレイテッド Non-mesenchymal human lung stem cells and methods for their use to treat respiratory diseases
US11540507B2 (en) 2016-11-04 2023-01-03 The University Of Tokyo Solution for cryopreservation of animal cells or animal tissues, cryopreserved product, and cryopreservation method
JP7088564B2 (en) 2016-11-10 2022-06-21 ヴィアサイト,インコーポレイテッド PDX1 pancreatic endoderm cells and methods thereof in a cell delivery device
AU2017363689B2 (en) 2016-11-25 2023-11-23 Riken Cell population for transplantation and method for producing same
US10508263B2 (en) 2016-11-29 2019-12-17 Procella Therapeutics Ab Methods for isolating human cardiac ventricular progenitor cells
AU2017377309B9 (en) 2016-12-14 2021-06-03 Otsuka Pharmaceutical Factory, Inc. Mammalian cell cryopreservation liquid
US10828330B2 (en) 2017-02-22 2020-11-10 IO Bioscience, Inc. Nucleic acid constructs comprising gene editing multi-sites and uses thereof
US11618883B2 (en) 2017-03-08 2023-04-04 Sumitomo Pharma Co., Ltd. Method for producing retinal pigment epithelial cells
JP2020511539A (en) 2017-03-16 2020-04-16 リネージ セル セラピューティクス インコーポレイテッド Method to measure the therapeutic effect of retinal disease treatment
US20200131473A1 (en) 2017-03-20 2020-04-30 Ifom Fondazione Istituto Firc Di Oncologia Molecolare Method of generating 2 cell-like stem cells
DK3612557T3 (en) 2017-04-18 2022-04-19 Fujifilm Cellular Dynamics Inc ANTIGEN-SPECIFIC IMMUNE EFFECTOR CELLS
US20210130849A1 (en) 2017-04-20 2021-05-06 Oregon Health & Science University Human gene correction
US20200172969A1 (en) 2017-06-19 2020-06-04 Foundation For Biomedical Research And Innovation At Kobe Method for predicting differentiation ability of pluripotent stem cell, and reagent for same
EP3656852A4 (en) 2017-07-20 2021-04-21 Riken Method for maturation of retinal tissue containing continuous epithelium
KR20200029479A (en) 2017-07-20 2020-03-18 고쿠리쓰 겐큐 가이하쓰 호징 리가가쿠 겐큐소 Methods of preservation of nerve tissue
AU2018308976A1 (en) 2017-07-31 2020-02-20 Biotime, Inc. Compositions and methods for restoring or preventing loss of vision caused by disease or traumatic injury
JP7275109B2 (en) 2017-08-23 2023-05-17 プロセラ セラピューティクス アーベー Use of Neuropilin-1 (NRP1) as a Cell Surface Marker to Isolate Human Ventricular Progenitor Cells
AU2018330694A1 (en) 2017-09-08 2020-03-19 Riken Cell aggregate including retinal tissue and production method therefor
CA3075883A1 (en) 2017-09-14 2019-03-21 Riken Method for amplifying cone photoreceptors or rod photoreceptors using dorsalization signal transmitter or ventralization signal transmitter
WO2019054514A1 (en) 2017-09-14 2019-03-21 国立研究開発法人理化学研究所 Method for producing retinal tissues
WO2019092505A1 (en) 2017-11-09 2019-05-16 Casebia Therapeutics Llp Self-inactivating (sin) crispr/cas or crispr/cpf1 systems and uses thereof
WO2019096787A1 (en) 2017-11-14 2019-05-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Regulatory t cells genetically modified for the lymphotoxin alpha gene and uses thereof
CN111630155A (en) 2017-11-15 2020-09-04 森玛治疗公司 Islet cell preparative compositions and methods of use
JP7297674B2 (en) 2017-11-24 2023-06-26 住友化学株式会社 Method for producing cell mass containing pituitary tissue and cell mass thereof
AU2018371437A1 (en) 2017-11-24 2020-07-02 Sumitomo Chemical Company, Limited Production method for cell mass including neural cells/tissue and non-neural epithelial tissue, and cell mass from same
CA3083158A1 (en) 2017-11-24 2019-05-31 Institut National De La Sante Et De La Recherche Medicale (Inserm) Methods and compositions for treating cancers
EP3724332A1 (en) 2017-12-14 2020-10-21 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and their use in genome editing and other applications
SG11202005795TA (en) 2017-12-29 2020-07-29 Cell Cure Neurosciences Ltd Retinal pigment epithelium cell compositions
CN111788303A (en) 2018-02-19 2020-10-16 大日本住友制药株式会社 Cell aggregate, mixture of cell aggregates, and method for producing same
WO2019183150A1 (en) 2018-03-19 2019-09-26 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and uses thereof
KR20200139210A (en) 2018-03-30 2020-12-11 고쿠리츠 다이가쿠 호진 교토 다이가쿠 Heterocyclic compound
CN111918961B (en) 2018-03-30 2023-10-24 国立大学法人京都大学 Myocardial cell maturation promoter
JP2021521792A (en) 2018-04-20 2021-08-30 フジフィルム セルラー ダイナミクス,インコーポレイテッド Differentiation method of eyeball cells and their use
US20210123016A1 (en) 2018-05-02 2021-04-29 Novartis Ag Regulators of human pluripotent stem cells and uses thereof
EP3572512A1 (en) 2018-05-24 2019-11-27 B.R.A.I.N. Ag A method for engineering a protein
TW202019458A (en) 2018-06-27 2020-06-01 美商裘美娜治療公司 Heparin-associated polypeptides and uses thereof
JP7285015B2 (en) 2018-07-19 2023-06-01 国立大学法人京都大学 Lamellar cartilage derived from pluripotent stem cells and method for producing the same
JP7357369B2 (en) 2018-07-23 2023-10-06 国立大学法人京都大学 Novel renal progenitor cell marker and method for enriching renal progenitor cells using it
CN112703011A (en) 2018-08-06 2021-04-23 国家医疗保健研究所 Methods and compositions for treating cancer
EP3833365A4 (en) 2018-08-10 2022-05-11 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
WO2020039732A1 (en) 2018-08-24 2020-02-27 住友化学株式会社 Cell aggregation including olfactory neuron or precursor cell thereof, and method for producing same
CA3112902A1 (en) 2018-09-28 2020-04-02 Otsuka Pharmaceutical Factory, Inc. Mammal cell preserving solution containing acarbose or stachyose
JP7437766B2 (en) 2018-10-31 2024-02-26 国立大学法人京都大学 Method for producing pluripotent stem cells that are free from mesendoderm differentiation resistance
US20220010271A1 (en) 2018-11-15 2022-01-13 Jsr Corporation Method for producing brain organoids
JP2022513073A (en) 2018-11-19 2022-02-07 ザ ユナイテッド ステイツ オブ アメリカ アズ リプリゼンテッド バイ ザ セクレタリー、デパートメント オブ ヘルス アンド ヒューマン サービシーズ Biodegradable tissue replacement implants and their use
US20210380950A1 (en) 2018-11-28 2021-12-09 Milica Radisic Methods for tissue generation
EP3901253A4 (en) 2018-12-20 2022-09-07 Sumitomo Chemical Company Limited Embryonic erythroblast-containing cell population and method for producing same, cell culture composition, and compound test method
WO2020130147A1 (en) 2018-12-21 2020-06-25 国立大学法人京都大学 Lubricin-localized cartilage-like tissue, method for producing same and composition comprising same for treating articular cartilage damage
EP3903789A4 (en) 2018-12-28 2022-10-05 Riken Therapeutic drug for disease accompanied by disorders in retinal system cells or retinal tissue
WO2020152686A1 (en) 2019-01-23 2020-07-30 Yeda Research And Development Co. Ltd. Culture media for pluripotent stem cells
US20220104481A1 (en) 2019-02-13 2022-04-07 Tigenix, S.A.U. Cryopreservation of stem cells
JP2022522187A (en) 2019-02-27 2022-04-14 タイジェニックス、ソシエダッド、アノニマ、ウニペルソナル Improved stem cell population for allogeneic therapy
MX2021010559A (en) 2019-03-07 2021-12-15 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
MA55297A (en) 2019-03-12 2022-01-19 Bayer Healthcare Llc NOVEL HIGH-FIDELITY PROGRAMMABLE RNA ENDONUCLEASE SYSTEMS AND THEIR USES
CA3132804A1 (en) 2019-03-13 2020-09-17 Sumitomo Dainippon Pharma Co., Ltd. Method for evaluating quality of transplant neural retina, and transplant neural retina sheet
WO2020204149A1 (en) 2019-03-29 2020-10-08 公立大学法人横浜市立大学 Screening method and toxicity evaluation method
AU2020260894A1 (en) 2019-04-26 2021-11-18 Riken Composite including neural retina, retinal pigment epithelial cells, and hydrogel, and method for producing same
AU2020263769B2 (en) 2019-04-26 2023-07-06 Otsuka Pharmaceutical Factory, Inc. Trehalose-containing liquid for mammalian cell preservation
US20210047649A1 (en) 2019-05-08 2021-02-18 Vertex Pharmaceuticals Incorporated Crispr/cas all-in-two vector systems for treatment of dmd
CA3138348A1 (en) 2019-05-09 2020-11-12 FUJIFILM Cellular Dynamics, Inc. Methods for the production of hepatocytes
EP3969569A1 (en) 2019-05-14 2022-03-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Regulatory t cells targeted by lymphotoxin alpha blocking agent and uses thereof
EP3973049A1 (en) 2019-05-22 2022-03-30 Hadasit Medical Research Services and Development Ltd. Methods of culturing human pluripotent cells
EP3754014A1 (en) 2019-06-21 2020-12-23 Centre d'Etude des Cellules Souches (CECS) Automated method for preparing retinal pigment epithelium cells
WO2020264072A1 (en) 2019-06-25 2020-12-30 Semma Therapeutics, Inc. Enhanced differentiation of beta cells
JP7385244B2 (en) 2019-06-27 2023-11-22 国立大学法人 東京大学 Method for isolating pancreatic progenitor cells
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
WO2021030424A1 (en) 2019-08-13 2021-02-18 Semma Therapeutics, Inc. Pancreatic differentiation
KR20220052946A (en) 2019-09-06 2022-04-28 각고호우징 게이오기주크 Method for producing cell aggregates containing glial progenitor cells
WO2021069593A1 (en) 2019-10-09 2021-04-15 INSERM (Institut National de la Santé et de la Recherche Médicale) T cells modified to express mutated cxcr4 or partially deleted and uses thereof
EP4063495A4 (en) 2019-11-20 2023-12-27 Sumitomo Pharma Co., Ltd. Method for freezing cell aggregates
EP4063496A1 (en) 2019-11-20 2022-09-28 Sumitomo Pharma Co., Ltd. Method for freezing neural cells
CA3162755A1 (en) 2019-11-25 2021-06-03 Kyoto University T-cell master cell bank
US20220409652A1 (en) 2019-11-29 2022-12-29 Novadip Biosciences miRNA-BASED PHARMACEUTICAL COMPOSITIONS AND USES THEREOF FOR THE PREVENTION AND THE TREATMENT OF TISSUE DISORDERS
MX2022006467A (en) 2019-11-29 2022-08-08 Novadip Biosciences Biomaterials for the prevention and the treatment of tissue disorders.
EP4074321A4 (en) 2019-12-12 2024-01-03 Univ Chiba Nat Univ Corp Freeze-dried preparation containing megakaryocytes and platelets
EP3875581A1 (en) 2020-03-02 2021-09-08 Centre d'Etude des Cellules Souches (CECS) Automated method for preparing keratinocytes
EP3875580A1 (en) 2020-03-02 2021-09-08 Centre d'Etude des Cellules Souches (CECS) Methods for preparing keratinocytes
US20230407264A1 (en) 2020-03-13 2023-12-21 Goliver Therapeutics Hepatic Stem-Like Cells for the Treatment and/or the Prevention of Liver Disorders
CN115885035A (en) 2020-03-19 2023-03-31 千纸鹤治疗公司 Method for purifying cardiomyocytes
JPWO2021187602A1 (en) 2020-03-19 2021-09-23
EP4132479A1 (en) 2020-04-07 2023-02-15 Ramot at Tel-Aviv University Ltd. Cannabidiol-containing compositions and uses thereof
IL298254A (en) 2020-05-29 2023-01-01 Fujifilm Cellular Dynamics Inc Bilayer of retinal pigmented epithelium and photoreceptors and use thereof
CN116033912A (en) 2020-05-29 2023-04-28 富士胶片细胞动力公司 Retinal pigment epithelium and photoreceptor double cell aggregates and methods of use thereof
US20230235319A1 (en) 2020-06-12 2023-07-27 Bayer Aktiengesellschaft Crispr-cas12a directed random mutagenesis agents and methods
EP4180516A4 (en) 2020-07-13 2024-01-17 Univ Kyoto Skeletal muscle precursor cells and method for purifying same, composition for treating myogenic diseases, and method for producing cell group containing skeletal muscle precursor cells
CA3194364A1 (en) 2020-09-11 2022-03-17 Sumitomo Pharma Co., Ltd. Medium for tissue for transplantation
CN116157512A (en) 2020-09-11 2023-05-23 国立研究开发法人理化学研究所 Composite comprising cell aggregate containing neural retina and matrix, and method for producing same
AU2021350981A1 (en) 2020-09-24 2023-06-08 Beijing Institute For Stem Cell And Regenerative Medicine Activated pluripotent stem cell, and preparation method therefor and use thereof
US20230399622A1 (en) 2020-10-16 2023-12-14 Fundació Centre De Regulació Genòmica Therapy for degenerative disease and tissue damage
CN112538458A (en) 2020-11-26 2021-03-23 北京赛尔湃腾科技咨询合伙企业(有限合伙) Method for reprogramming cells
EP4005577A1 (en) 2020-11-26 2022-06-01 Novadip Biosciences Cellular and/or extracellular extracts for preventing and/or treating cancer and/or inflammation
CN116583595A (en) 2020-12-23 2023-08-11 三井化学株式会社 Culture member and use thereof
JPWO2022191216A1 (en) 2021-03-09 2022-09-15
WO2022207889A1 (en) 2021-04-01 2022-10-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Liver organoid manufacturing methods, liver organoids obtained with the same, and uses thereof
KR20230165846A (en) 2021-04-07 2023-12-05 후지필름 셀룰러 다이내믹스, 인코포레이티드 Dopaminergic progenitor cells and methods of use
JP2024513520A (en) 2021-04-11 2024-03-25 プレジデント アンド フェローズ オブ ハーバード カレッジ Cardiomyocytes and compositions and methods for producing them
CN117242173A (en) 2021-05-03 2023-12-15 安斯泰来再生医药协会 Method for producing mature corneal endothelial cells
TW202309268A (en) 2021-05-07 2023-03-01 安斯泰來再生醫藥協會 Methods of generating mature hepatocytes
CA3220602A1 (en) 2021-05-28 2022-12-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods to generate macular, central and peripheral retinal pigment epithelial cells
AU2022282379A1 (en) 2021-05-28 2023-11-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Biodegradable tissue scaffold with secondary matrix to host weakly adherent cells
WO2022258511A1 (en) 2021-06-07 2022-12-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for generating highly functional hepatocytes by differentiating hepatoblasts
TW202303145A (en) 2021-06-09 2023-01-16 美商譜系細胞治療公司 Methods and compositions for treating retinal diseases and conditions
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
WO2022265086A1 (en) 2021-06-17 2022-12-22 国立大学法人京都大学 Method for producing cerebral cortical cell preparation derived from human pluripotent stem cells
KR20240038013A (en) 2021-07-28 2024-03-22 리니지 셀 테라퓨틱스, 인크. Expansion of retinal pigment epithelial cells
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023039588A1 (en) 2021-09-13 2023-03-16 FUJIFILM Cellular Dynamics, Inc. Methods for the production of committed cardiac progenitor cells
WO2023049826A1 (en) 2021-09-23 2023-03-30 President And Fellows Of Harvard College Genetically encoded voltage indicators and uses thereof
US20230226116A1 (en) 2021-10-20 2023-07-20 University Of Rochester Method for rejuvenating glial progenitor cells and rejuvenated glial progenitor cells per se
US20230277600A1 (en) 2021-10-20 2023-09-07 University Of Rochester Treatment Of Age-Related White Matter Loss By Competitive Replacement Of Glial Cells
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
WO2023095149A1 (en) 2021-11-29 2023-06-01 Ramot At Tel-Aviv University Ltd. Methods and compositions for treating spinal cord injury
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
WO2023178239A1 (en) 2022-03-16 2023-09-21 The Children's Medical Center Corporation Hpsc-derived articular chondrocyte compositions, systems and methods of use thereof
US20230330264A1 (en) 2022-04-15 2023-10-19 Smartcella Solutions Ab COMPOSITIONS AND METHODS FOR EXOSOME-MEDIATED DELIVERY OF mRNA AGENTS
WO2023211857A1 (en) 2022-04-25 2023-11-02 Lineage Cell Therapeutics, Inc. Methods and compositions for treating vision loss
WO2023215455A1 (en) 2022-05-05 2023-11-09 University Of Rochester Dual macroglial-microglial approach towards therapeutic cell replacement in neurodegenerative and neuropsychiatric disease
WO2023230391A1 (en) 2022-05-26 2023-11-30 Tender Food, Inc. Plant-based shredded meat products, and methods of producing the same
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2024059358A1 (en) 2022-09-16 2024-03-21 Tender Food, Inc. Plant and animal cell blended meat products and methods of producing the same
WO2024073776A1 (en) 2022-09-30 2024-04-04 FUJIFILM Cellular Dynamics, Inc. Methods for the production of cardiac fibroblasts

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3060078A (en) * 1960-12-02 1962-10-23 Burlington Industries Inc Bonding of polyethylene terephthalate fibers to certain rubbers
GB1051340A (en) * 1962-09-21 1900-01-01
US20030032178A1 (en) * 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
EP0380646B1 (en) 1988-08-04 1999-01-20 Amrad Corporation Limited Use of leukaemia inhibitory factor (lif) for the (in vitro) propagation of embryonic stem cells
JP2750464B2 (en) * 1990-01-30 1998-05-13 日本ゼオン株式会社 Method for producing fiber-rubber composite
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
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
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
WO1994003585A1 (en) * 1992-08-04 1994-02-17 Commonwealth Scientific And Industrial Research Organisation A method for maintaining embryonic stem cells and avian factor useful for same
US5453357A (en) * 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5690926A (en) * 1992-10-08 1997-11-25 Vanderbilt University Pluripotential embryonic cells and methods of making same
US5523226A (en) * 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US5591625A (en) * 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US5449620A (en) * 1994-01-25 1995-09-12 Thomas Jefferson University Apparatus and method for culturing embryonic stem cells
US5541081A (en) * 1994-03-22 1996-07-30 President And Fellows Of Harvard College Process for assessing oocyte and embryo quality
JP3496292B2 (en) * 1994-09-30 2004-02-09 日本ゼオン株式会社 Composites of nitrile group-containing highly saturated copolymer rubber and fiber
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5654099A (en) * 1996-02-21 1997-08-05 Dayco Products, Inc. Method of improving adhesion between alkylated chlorosulfonated polyethylene (ACSM) or chlorosulfonated polyethylene (CSM), and resorcinol formaldehyde latex (RFL) treated polyester cord
CA2417356A1 (en) * 2000-08-01 2002-02-07 Yissum Research Development Company Directed differentiation of embryonic cells
US20070077654A1 (en) * 2004-11-01 2007-04-05 Thomson James A Platelets from stem cells

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8252280B1 (en) 1999-08-05 2012-08-28 Regents Of The University Of Minnesota MAPC generation of muscle
US10226485B2 (en) 1999-08-05 2019-03-12 Abt Holding Company Multipotent adult stem cells and methods for isolation
US7659118B2 (en) 1999-08-05 2010-02-09 Abt Holding Company Multipotent adult stem cells
US7015037B1 (en) 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US20050181502A1 (en) * 1999-08-05 2005-08-18 Athersys, Inc. Multipotent adult stem cells and methods for isolation
US9249388B2 (en) 1999-10-28 2016-02-02 University Of Massachusetts Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US8273571B2 (en) 1999-10-28 2012-09-25 Advanced Cell Technology, Inc. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US7951591B2 (en) 1999-10-28 2011-05-31 Advanced Cell Technology, Inc. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20080075702A1 (en) * 1999-10-28 2008-03-27 University Of Massachusetts, As Represented By Its Amherst Campus Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20040014206A1 (en) * 1999-10-28 2004-01-22 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20040107453A1 (en) * 2001-02-14 2004-06-03 Furcht Leo T Multipotent adult stem cells, sources thereof, methods of obtaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US7838289B2 (en) 2001-02-14 2010-11-23 Abt Holding Company Assay utilizing multipotent adult stem cells
US20030079536A1 (en) * 2001-09-10 2003-05-01 Frank Fischer Method and system for monitoring a tire air pressure
US8697444B2 (en) 2001-12-21 2014-04-15 Thrombogenics N.V. Compositions for the in vitro derivation and culture of embryonic stem (ES) cell lines with germline transmission capability and for the culture of adult stem cells
US20090098650A1 (en) * 2001-12-21 2009-04-16 Thromb-X N.V. Compositions for the in vitro derivation and culture of embryonic stem (es) cell lines with germline transmission capability and for the culture of adult stem cells
US8993323B2 (en) 2001-12-21 2015-03-31 Thrombogenics N.V. Compositions for the in vitro derivation and culture of embryonic stem (ES) cell lines with germline transmission capability and for the culture of adult stem cells
US20050059145A1 (en) * 2001-12-21 2005-03-17 Luc Schoonjans Compositions for the in vitro derivation and culture of embryonic stem (es) cell lines with germline transmission capability and for the culture of adult stem cells
US20030207448A1 (en) * 2002-05-06 2003-11-06 Revera Gregory Henry Methodologies for the creation of pluripotent or multipotent human stem cells without creating or destroying a human embryo
US20040018617A1 (en) * 2002-07-26 2004-01-29 Shiaw-Min Hwang Somatic pluripotent cells
US7422736B2 (en) * 2002-07-26 2008-09-09 Food Industry Research And Development Institute Somatic pluripotent cells
WO2004038012A1 (en) * 2002-10-25 2004-05-06 Hunan Hui-Lin Life Technology Co. Ltd The feeder cell layer for in vitro culturing human embryonic stem cells and the method for culturing embryonic stem cells
US20050100924A1 (en) * 2003-11-06 2005-05-12 National University Of Singapore C/EBPalpha gene targeting constructs and uses thereof
US20050118561A1 (en) * 2003-12-02 2005-06-02 Catholic Healthcare West Compositions and methods for propagation of neural progenitor cells
US20070269412A1 (en) * 2003-12-02 2007-11-22 Celavie Biosciences, Llc Pluripotent cells
US8367406B2 (en) 2003-12-02 2013-02-05 Celavie Biosciences, Llc Pluripotent cells
US9834751B2 (en) 2003-12-02 2017-12-05 Celavie Biosciences, Llc Pluripotent cells
US7632681B2 (en) 2003-12-02 2009-12-15 Celavie Biosciences, Llc Compositions and methods for propagation of neural progenitor cells
US20090280097A1 (en) * 2003-12-02 2009-11-12 Celavie Biosciences, Llc Pluripotent cells
US9439932B2 (en) 2003-12-02 2016-09-13 Celavie Biosciences, Llc Pluripotent cells
US10638734B2 (en) 2004-01-05 2020-05-05 Abt Holding Company Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US20060275899A1 (en) * 2004-12-30 2006-12-07 Stemlifeline, Inc. Methods and compositions relating to embryonic stem cell lines
US20060263879A1 (en) * 2004-12-30 2006-11-23 Stemlifeline, Inc. Methods and systems relating to embryonic stem cell lines
US9005964B2 (en) 2006-11-24 2015-04-14 Regents Of The University Of Minnesota Endodermal progenitor cells
US20100150876A1 (en) * 2006-11-24 2010-06-17 Regents Of The Univeristy Of Minnesota Endodermal progenitor cells
US9144585B2 (en) 2010-07-27 2015-09-29 Technion Research & Development Foundation Limited Isolated mesenchymal progenitor cells and extracellular matrix produced thereby
US20130130387A1 (en) * 2010-07-27 2013-05-23 Technion Research & Development Foundation Limited Method for generating induced pluripotent stem cells from keratinocytes derived from plucked hair follicles

Also Published As

Publication number Publication date
US20050158854A1 (en) 2005-07-21
US7781216B2 (en) 2010-08-24
US7582479B2 (en) 2009-09-01
US20110256623A1 (en) 2011-10-20
EP1640448A2 (en) 2006-03-29
US20030008392A1 (en) 2003-01-09
CA2190528C (en) 2010-04-27
US20060040383A1 (en) 2006-02-23
EP0770125A1 (en) 1997-05-02
US20120328582A1 (en) 2012-12-27
US7029913B2 (en) 2006-04-18
US5843780A (en) 1998-12-01
AU4758496A (en) 1996-08-07
CA2190528A1 (en) 1996-07-25
US20150056698A1 (en) 2015-02-26
US20050164381A1 (en) 2005-07-28
US6200806B1 (en) 2001-03-13
EP0770125A4 (en) 1998-01-14
EP1640448A3 (en) 2006-04-26
US8273569B2 (en) 2012-09-25
WO1996022362A1 (en) 1996-07-25
US20090011503A1 (en) 2009-01-08

Similar Documents

Publication Publication Date Title
US7582479B2 (en) Primate embryonic stem cell line
Thomson et al. Isolation of a primate embryonic stem cell line.
Martin Teratocarcinomas as a model system for the study of embryogenesis and neoplasia
CA2456981C (en) Alternative compositions and methods for the culture of stem cells
AU2016324669B2 (en) Culture method for differentiating primordial germ cells into functionally mature oocytes
US9255247B2 (en) Methods of deriving mammalian pluripotent stem cell lines
Strelchenko Bovine pluripotent stem cells
US20020160509A1 (en) Embryonic stem cells
Suemori et al. Establishment of the Embryo‐derived Stem (ES) Cell Lines from Mouse Blastocysts: Effects of the Feeder Cell Layer.
EP1263932A1 (en) Embryonic stem cells and neural progenitor cells derived therefrom
Andrews et al. Embryonal carcinoma cells as embryonic stem cells
US20030073234A1 (en) Clonal human embryonic stem cell lines and methods of generating same
JP2007516720A (en) Embryonic stem cell line and method for producing the same
Fan et al. A modified culture medium increases blastocyst formation and the efficiency of human embryonic stem cell derivation from poor-quality embryos
Pall et al. Establishment of an embryonic stem cell line from blastocyst stage mouse embryos
JP2002536976A (en) Pluripotent cells-1
Amit et al. Isolation and maintenance of primate ES cells
Vrana et al. Colloquium Nonhuman primate parthenogenetic stem cells
Draper et al. Human embryonal
JPH08116966A (en) Bovine embryonic stem cell and its preparation

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: WISCONSIN ALUMNI RESEARCH FOUNDATION, WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THOMSON, JAMES;REEL/FRAME:031347/0603

Effective date: 19960702