WO2000029002A2 - Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines - Google Patents

Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines Download PDF

Info

Publication number
WO2000029002A2
WO2000029002A2 PCT/US1999/026927 US9926927W WO0029002A2 WO 2000029002 A2 WO2000029002 A2 WO 2000029002A2 US 9926927 W US9926927 W US 9926927W WO 0029002 A2 WO0029002 A2 WO 0029002A2
Authority
WO
WIPO (PCT)
Prior art keywords
human
cells
mesenchymal stem
stem cells
organ
Prior art date
Application number
PCT/US1999/026927
Other languages
English (en)
Other versions
WO2000029002A3 (fr
Inventor
Mark Thiede
Alan Flake
Original Assignee
Osiris Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Osiris Therapeutics, Inc. filed Critical Osiris Therapeutics, Inc.
Priority to AU14780/00A priority Critical patent/AU1478000A/en
Priority to EP99972103A priority patent/EP1128836A2/fr
Priority to CA002348514A priority patent/CA2348514A1/fr
Priority to JP2000582048A priority patent/JP2002529509A/ja
Publication of WO2000029002A2 publication Critical patent/WO2000029002A2/fr
Publication of WO2000029002A3 publication Critical patent/WO2000029002A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)

Definitions

  • the present invention relates to the field of cell therapy, and more particularly to the field of in vivo gene therapy by administering mesenchymal stem cells.
  • Certain diseases including inherited metabolic diseases, may produce irreversible damage to the fetus before birth.
  • In utero hematopoietic stem cell transplantation is a potential therapy for a large number of immunodeficiency diseases, hemoglobinopathies and others.
  • Advantages for the fetal recipient include potential immunologic tolerance for transplanted cells, and rapid expansion of the hematopoeitic compartment with formation of new "niches" for the competitive engraftment of donor cells.
  • the existence of natural models of hematopoietic chimerism in dizygotic twins which share cross placental circulation during development supports the potential to achieve high levels of donor cell chimerism with prenatal transplantation in recipients with normal hematopoiesis. Experimental work in sheep and other animal models has.
  • Mesenchymal stem cells are the formative pluripotential cells found inter alia in bone marrow, blood, dermis and periosteum that are capable of differentiation into any of the mesenchymal or connective tissues, for example, bone, cartilage, muscle, stroma, tendon, and fat.
  • This homogeneous population of cells can be passaged in culture and may be characterized by the lack of hematopoietic cell markers and by the presence of a unique set of surface antigens. Under specific conditions they have been induced to form bone, cartilage, adipose tissue, tendon, and muscle, and in their undifferentiated state, resemble roughly stromal fibroblasts and can support hematopoiesis as evidenced by the support of LT-CIC in long term bone marrow culture. Preliminary in vivo studies suggest that these cells home to bone marrow of post-natal recipients after intravenous administration and can accelerate constitution after myeloablative conditioning regimes.
  • Another object of the invention is to increase donor cell engraftment in fetal recipients. Another object is to regenerate damaged or diseased tissue by providing to a fetal recipient donor cells which can differentiate in situ. Yet another object is to prepare chimeric organs and tissues. Still another object is to treat a recipient by administering mesenchymal stem cells ii which , has. been ' modified "to express a therapeutic gene product.
  • MSCs mesenchymal stem cells transplanted into a fetus in utero then were distributed throughout the fetus.
  • the MSCs remained viable and differentiated into cellular types appropriate for the tissue or organ in which they engrafted. Surprisingly, the MSCs and their differentiated progeny were not rejected by immunocompetent hosts.
  • the MSCs can be used for cellular therapy and tissue engineering.
  • Normal MSCs express and secrete a number of cytokises, including G-CSF, SCF, LIF, M-CSF, IL-6, and IL-11. (Haynesworth, et al, J. Cell Physiol., Vol. 166, No. 3, pgs. 585-592 (March 1996)).
  • cytokises including G-CSF, SCF, LIF, M-CSF, IL-6, and IL-11.
  • transduced mesenchymal stem cells which have been modified to carry exogenous genetic material of interest, are induced to differentiate, the progeny cells also carry the new genetic material.
  • These transduced cells are able to express the exogenous gene product.
  • transduced mesenchymal stem cells and the cells differentiated therefrom can be used for applications where treatment using such modified cells is beneficial.
  • these modified cells can be used as a delivery system for therapeutic proteins encoded by the exogenous gene for treatment of inherited and/or acquired disorders of blood coagulation and wound healing.
  • the present invention provides a method of obtaining genetically modified mesenchymal stem cells, comprising transducing mesenchymal stem cells with exogenous genetic material and placing the transduced cells under conditions suitable for differentiation of the mesenchymal stem cells into lineages which then contain the exogenous genetic material.
  • the present invention is directed to a method for effecting gene therapy in vivo by administrating human mesenchymal stem cells in utero.
  • the mesenchymal stem cells can differentiate into specific ' cell liheagesiidepen ⁇ -ing ⁇ i-uthe. environment and integrate with like tissue to effect repair of tissue defects.
  • the mesenchymal stem cells can be transduced with exogenous genetic material such that a gene product will be expressed by the mesenchymal stem cell or its differentiated progeny in vivo to provide a desired therapeutic effect.
  • the present invention involves a method for treating a subject in need thereof by administering human mesenchymal stem cells in an amount effective to enhance the in vivo distribution and engraftment of mesenchymal stem cells.
  • the mesenchymal stem cells can be transduced with exogenous genetic material such that a gene product will be expressed by the mesenchymal stem cell or its differentiated progeny in vivo to provide a desired therapeutic effect.
  • Figure 1 is a photograph of liver tissue at 9 weeks after mesenchymal stem cells were injected intraperitoneally at 65 days gestation.
  • Figure 2 is a photograph of bone marrow at 9 weeks after mesenchymal stem cells were injected intraperitoneally at 85 days gestation.
  • Figure 3 is a photograph of heart tissue at 9 weeks after mesenchymal stem cells were injected intraperitoneally at 65 days gestation.
  • Figure 4 is a photograph of thymus tissue at 9 weeks after mesenchymal stem cells were injected intraperitoneally at 85 days gestation.
  • Figure 5 shows results of PCR analysis for human-specific ' ⁇ -2 sculpturecr ⁇ glob ⁇ iri performed on tissue samples of liver (Lv), spleen (Sp), bone marrow (BM), thymus (Th), lung (Lg), brain (Br), and blood (Bd).
  • Figure 6A is a photograph of a gel showing the presence of human ⁇ -2 microglobulin DNA in liver, spleen, lung, bone marrow, thymus, brain, and blood isolated from sheep fetuses at 65 or 85 days gestation, wherein the sheep fetuses were given human mesenchymal stem cells in utero.
  • Figures 6B, 6C, 6D, 6E, and 6F are photographs of slides showing the presence of human cells in sheep fetal liver, spleen, bone marrow, thymus, and lung, respectively.
  • Figure 7A is a photograph of a slide showing human mesenchymal stem cells, in the cardiac muscle of sheep fetuses stained with anti-human ⁇ -2 microglobulin.
  • Figures 7B and 7C are photographs of slides showing human cells in the cardiac muscle of sheep fetuses stained with anti-human ⁇ -2 microglobulin and
  • Figures 8A and 8B are photographs of slides showing the presence of human ⁇ -2 microglobulin through nickel chloride staining in cartilage lacunae of lambs given human mesenchymal stem cells in utero at 65 days gestation, and wherein the cartilage was harvested at 2 months and 5 months after transplantation, respectively.
  • Figure 9 shows photographs of slides of human cells, contacted with human- specific anti-CD23 antibody, found in the bone marrow of sheep at 5 months after in utero transplantation of human mesenchymal stem cells.
  • Figure 9A is a control showing normal ovine tissue contacted with human-specific anti-CD23 antibody.
  • Figures 9B through 9D show CD23+ human cells in ovine bone marrow at increasing magnification.
  • Figure 10 shows photographs of slides of human cells, contacted with human-specific anti-CD74 antibody, found in the bone marrow of sheep at 5 months after in utero transplantation of human mesenchymal stem cells.
  • Figure 10A is a control showing normal ovine tissue contacted with human-specific anti-CD74 antibody.
  • Figures 10B through 10D show CD74+ human " : cells . ⁇ h.Ovihe%ymus;.a ⁇ increasing magnification.
  • Figures 11A and 11B are photographs of slides showing human ⁇ -2 microglobulin positive cells in the central nervous system of sheep at 5 months after the sheep were given mesenchymal stem cells in utero.
  • the present invention relates generally to the use of human mesenchymal stem cells and to compositions comprising human mesenchymal stem cells for in utero administration.
  • mesenchymal stem cells can be recovered from other cells in the bone marrow or other mesenchymal stem cell source.
  • Bone marrow cells may be obtained from iliac crest, femora, tibiae, spine, rib or other medullary spaces.
  • Other sources of human mesenchymal stem cells include embryonic yolk sac, placenta, umbilical cord, fetal and adolescent skin, and blood.
  • the presence of mesenchymal stem cells in the culture colonies may be verified by specific cell surface markers which are identified with unique monoclonal antibodies, see, e.g., U.S. Patent No.
  • the mesenchymal stem cell populations can be autologous, allogeneic, or xenogeneic to the recipient.
  • mesenchymal stem cells are from the same species as the recipient.
  • the MSCs are human in origin.
  • a method of isolating mesenchymal stem cells comprises the steps of providing a tissue specimen containing mesenchymal stem cells, preferably bone marrow; isolating the mesenchymal stem cells from the specimen, for example by density gradient centrifugation: adding the isolated cells to a medium which contains factors that stimulate mesenchymal stem cell growth without differentiation and allows, when cultured, for the selective adherence of only the mesenchymal stem cells to a substrate surface; culturing the specimen-medium mixture; and removing the non-adherent matter from the substrate surface.
  • the isolated mesenchymal stem cells are culture expanded in appropriate media, i.e. cultured by methods which favor cell growth of the enriched cell populations.
  • the cells are plated at a density of 0.05-2xl0 5 cells/cm 2 , preferably at a density of 0.5 -10 x 10 4 cells/cm 2 .
  • the cells may be characterized prior to, during, and after culture to determine the composition of the cell population, for example by flow cytometric analysis (FACS).
  • FACS flow cytometric analysis
  • the human mesenchymal stem cells can be stained with human mesenchymal stem cell-specific monoclonal antibodies.
  • the culture conditions such as temperature, pH, and the like, are those previously used with the cells utilized in this invention and will be apparent to one of skill in the art.
  • the mesenchymal stem cells produced according to the methods described herein can be used to provide a reliable and constant source of mesenchymal stem cells for individuals in need thereof, e.g. those in need of cellular therapy, tissue engineering or regeneration and gene therapy.
  • Another aspect of the present invention relates to the introduction of genes into the mesenchymal stem cells such that the mesenchymal stem cells and progeny of the cells carry the new genetic material.
  • the mesenchymal stem cells can be modified with genetic material of interest.
  • the mesenchymal stem cells are able to express the product of the gene expression and, with a signal sequence, secrete the expression product. These modified cells can then be administered to a target, i.e., in need of mesenchymal stem cells or the gene expression product, where the expressed product will have a beneficial effect.
  • mesenchymal stem cells may be genetically engineered to express therapeutic proteins. Those that may be mentioned include providing continuous delivery of insulin, which at present must be isolated from the pancreas and extensively purified or manufactured in vitro recombinantly and then injected into the body by those whose insulin production or utilization is impaired. Genetically engineered human mesenchymal stem cells can also be used for the production of clotting factors. Persons suffering from hemophilia A lack a protein called Factor VIII, which is involved in clotting.
  • a recombinant Factor VIII product is now available and is administered by injection (Kogenate®, Bayer, Berkeley, CA)Jncorporation of genetic material of interest into human stem cells and other types of cells is particularly valuable in the treatment of inherited and acquired disease.
  • Inherited disorders that could be treated in this way include disorders of amino acid metabolism, such as Fabry's Disease, Gaucher's Disease, histidinurea or familial hypocholesterolemia; and disorders of nucleic acid metabolism, such as hereditary orotic aciduris.
  • MSCs express exogenous genes at high levels for long periods. This expression can continue through and after terminal differentiation.
  • the mesenchymal stem cells may be genetically modified by incorporation of genetic material into the cells, for example using recombinant expression vectors.
  • recombinant expression vector refers to a transcriptional unit comprising an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription initiation and termination sequences.
  • Structural units intended for use in eukaryotic expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host cell.
  • recombinant protein may include an N-terminal methionine residue. This residue may or may not be subsequently cleaved from the expressed recombinant protein to provide a final product.
  • the mesenchymal stem cells thus may have stably integrated a recombinant transcriptional unit into chromosomal DNA or carry the recombinant transcriptional unit as a component of a resident plasmid.
  • Cells may be engineered with a polynucleotide (DNA or RNA) encoding a polypeptide ex vivo, for example.
  • Cells may be engineered by procedures known in the art :;by ' ,use .of a retroviral particle ' , containing RNA encoding a polypeptide.
  • Retroviruses from which the retroviral plasmid vectors hereinabove mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
  • the retroviral plasmid vector is MGIN, derived from murine embryonic stem cells. Generally regarding retroviral mediated gene transfer, see McLachlin et al.(1990).
  • a preferred retroviral packaging cell line is described in U.S. Patent No. 5,910,434, the contents of which are incorporated herein by reference. Such cell line permits very high levels of transfection, i.e., greater than 80%.
  • the nucleic acid sequence encoding the polypeptide is under the control of a suitable promoter.
  • suitable promoters which may be employed include, but are not limited to, TRAP promoter, adeno viral promoters, such as the adeno viral major late promoter; the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral long terminal repeats (LTRs); ITRs; the ⁇ -actin promoter; and human growth hormone promoters; the GPIIb promoter.
  • TRAP promoter adeno viral promoters, such as the adeno viral major late
  • the promoter also may be the native promoter that controls the gene encoding the polypeptide. These vectors also make it possible to regulate the production of the polypeptide by the engineered progenitor cells. The selection of a suitable promoter will be apparent to those skilled in the art.
  • the retroviral LTR is preferred.
  • vehicles other than retroviruses to genetically engineer or modify the mesenchymal stem cells.
  • Genetic information of interest can be introduced by means of any virus which can express the new genetic mater ⁇ afin such cells.
  • SV40 herpes virus, adenovirus, adeno-associated virus, and human papillomavirus can be used for this purpose.
  • the expression vectors may contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed cells such as dihydrofolate reductase, neomycin resistance or green fluorescent protein (GFP).
  • selectable marker genes such as dihydrofolate reductase, neomycin resistance or green fluorescent protein (GFP).
  • the mesenchymal stem cells may be transfected through other means known in the art.
  • Such means include, but are not limited to transfection mediated by calcium phosphate or DEAE-dextran; transfection mediated by the polycation Polybrene (Kawai and Nishizawa 1984; Chaney et al. 1986); protoplast fusion (Robert de Saint Vincent et al. 1981; Schaffher 1980; Rassoulzadegan et al. 1982); electroporation (Neumann et al. 1982; Zimmermann 1982; Boggs et al. 1986); liposomes (see, e.g.
  • the present invention further makes it possible to genetically engineer mesenchymal stem cells in such a manner that they produce, in vitro or in vivo polypeptides, hormones and proteins not normally produced in human mesenchymal stem cells in biologically significant amounts or produced in small amounts but in situations in which increased expression would lead to a therapeutic benefit. These products then would be secreted into the surrounding media or purified from the cells.
  • the human mesenchymal stem cells formed in this way can serve as continuous short-term or long-term production systems of the expressed substance.
  • the cells could be modified such that a protein normally expressed will be expressed at much lower levels. This can be accomplished with antisense nucleic acid technology, catalytic enzyme expression, single chain antibody expression, and the like.
  • This technology may be used to produce additional copies of essential geheS to allow augmented expression by the mesenchymal stem cells of certain gene products in vivo.
  • genes can be, for example, hormones, matrix proteins, cell membrane proteins, cytokines, adhesion molecules, "rebuilding" proteins important in tissue repair.
  • the expression of the exogenous genetic material in vivo is often referred to as "gene therapy”.
  • Disease states and procedures for which such treatments have application include genetic disorders and diseases of blood and the immune system.
  • Cell delivery of the transformed cells may be effected using various methods and includes intravenous or intraperitoneal infusion and direct depot injection into periosteal, bone marrow and subcutaneous sites.
  • the mesenchymal stem cells of the present invention may be administered to the fetus using methods generally known in the art.
  • the present invention provides a method of modifying fetal organs of a first species by administering mesenchymal stem cells of a second species to a fetus of the first species in utero.
  • human mesenchymal stem cells are administered to a non-human fetus in utero.
  • the scope of this aspect of the present invention is not to be limited to any theoretical reasoning, it is believed that the non-human fetal organs may be less immunogenic, in the context of subsequent transplants into humans than unmodified organs. (See, for example, published PCT Application No. WO99/47163.) Thus, the modified non-human organs may be more suitable for transplantation.
  • This aspect of the present invention is applicable particularly to the transplantation of animal organs into humans.
  • organs include, but are not limited to, the heart, pancreas, kidney, skin, liver, thymus, spleen, bone marrow, cartilage, and bone.
  • human mesenchymal stem cells are administered to a non-human animal fetus.
  • the MSCs preferably are autologous to the human patient. After birth, the non-human animal is raised to an appropriate age and/or size such that the size of the organ or organs to be transplanted approximates the size of the organ or organs required by the human.
  • the modified organ(s) then is (are) harvested and transplanted into the human patient.
  • mesenchymal stem cells from a pediatric patient with a severe congenital heart defect are administered to a pig fetus. After birth, the pig would be raised to an appropriate age and/or size such that its heart approximates the size of the patient's heart. The modified heart then would be removed and transplanted into the pediatric patient.
  • MSCs mesenchymal stem cells
  • the MSCs were cultured in Control Media at 37°C in a humidified atmosphere containing 5% CO 2 . Upon reaching near- confluence, the cells were detached with 0.25% trypsin containing ImM EDTA (Gibco/BRL) for 5 minutes at 37°C. The cells were washed with Control Media and resuspended at 5 x 10 6 MSCs/mL in Control Media containing 5% DMSO (Sigma Chemical Co., St. Louis, MO). The cells were then stored in liquid nitrogen for use in the subsequent experiments.
  • MSC's human mesenchymal stem cells
  • the fetal liver remains the primary hematopoietic organ but stromal elements can be seen in the marrow with a few hematopoietic cells.
  • both fetal liver and bone marrow hematopoiesis are both active.
  • the day 65 fetus does not mount an immune response to non-self hematopoietic cells, whereas the day 85 fetus is immune competent and rejects hematopoietic cell transplants routinely.
  • Recipients were sacrificed at 7 or 14 days after transplantation and the liver, spleen, bone marrow, thymus, lung, brain and blood were analyzed by PCR for human specific ⁇ -2 microglobulin. Tissues positive for human sequence were confirmed by immunohistochemistry for morphologic assessment by staining for human ⁇ -2 microglobulin with secondary staining with horseradish peroxidase for visualization.
  • Fetal lambs at 65 or 85 days gestational age underwent transuterine-intraperitoneal or intravascular injection of 5 x 10 6 , 50 x 10 6 MSCs/fetus, respectively.
  • the ewes underwent horizontal hysterotomy with the use of electrocautery and Babcock clamps to control bleeding at the cut uterus margin.
  • the hind limbs and base of the umbilical cord were exteriorized and intravascular injection performed.
  • the fetal hind limbs and umbilical cord were returned to the amniotic cavity. Amniotic fluid volume was restored with warm, sterile lactated Ringer's solution and the hysterotomy closed with a TA-90 stapler.
  • the maternal abdomen was closed in layers and dressed with colloidin.
  • the ewe was placed in a movable cage where she was monitored until completely recovered from anesthesia. The ewes were continuously monitored until completely alert, and were able to stand, eat and drink. At this time, the animal was transported back to her holding room.
  • Buprenorphine was administered to alleviate postoperative pain. The animals were checked by the investigator's team 4-8 hours later, and then once or twice daily. Further doses of buprenorphine were administered every 8 hours as needed for pain. Also, antibiotic (liquamycin) was given daily for 5 days.
  • Fetal tissues were fixed overnight in 10% neutral buffered formalin at 4°C and paraffin embedded. For isolation of total cellular DNA, samples from each tissue were snap frozen in liquid nitrogen, and stored at -80°C for later DNA extraction.
  • Immunohistochemistry Serial 5um sections were obtained from each of the paraffin embedded tissues using a 30/50 microtome. Sections were deparaffinated, dehydrated, and rehydrated and then subjected to microwave antigen retrieval. Sections were then stained immunohistochemically for human class I antigen and SH2/SH3 antigen. The latter two antigens are found on MSCs. (See U.S. Patent No. 5,837,539.)
  • Total cellular DNA from the organs mentioned above were isolated using DNAzol. Specific primers for human class I antigen were selected based on the published human class I sequence.
  • lug DNA were added to each 0.65 mL microcentrifuge tube and placed on ice. A master mix was prepared and added on ice such that the final concentration of reagents for each sample was 2.5U Amplitaq Gold DNA polymerase (Perkin Elmer, Norwalk, CT). 200 uM deoxytriphosphates (dNTP's, Pharmacia, Piscataway, NJ), 50mM KC1, lOmM Tris-Cl (pH 8.3 at 22°C), 1.5mM MgCl 2 , 0.01% gelatin, and luM upstream and downstream primers.
  • thermocycler block reached 94°C, when the samples were immediately placed into the block for 9 minutes.
  • Samples were amplified for 40 cycles of 30 seconds at 94°C followed by 30 seconds of primer annealing followed by 1 minute of extension at 72°C.
  • samples were incubated for 5 minutes at 72°C.
  • PCR products were subjected to electrophoresis through a 2.5% NuSieve/1% Seakem agarose gel containing 0.5ug ethidium bromide/mL in IX Tris acetate running buffer. The gels were illuminated with UV 280-nm light and photographed with type 55 positive/negative Polaroid film. The negative was scanned transmissively and the intensities of the bands determined using the Intelligent Quantifier densitometer. Band intensities was compared to standard curves generated with known concentrations of human DNA.
  • the fetal sheep were sacrificed at 1 or 2 weeks or 2 or 5 months after injection and the liver, spleen, lung, bone marrow, thymus, brain, heart, skeletal muscle, cartilage, and blood were harvested and analyzed for the presence of human cells.
  • the fetal tails were docked at the time of MSC injection and the tail wounds harvested at 1 week or 2 months after wounding for DNA isolation and immunohistochemistry.
  • DNA Isolation Total cellular DNA from the organs mentioned above was isolated using DNAzol (Molecular Resource Center, Inc., Cincinnati, OH). In brief, approximately 100 mg of tissue was homogenized in lmL of DNA zol. The DNA was precipitated with 0.5mL of 100% ethanol. The DNA precipitate was pelleted by centrifugation and then washed twice with 95% ethanol. The DNA pellet was then dissolved in sterile water.
  • PCR Analysis To screen the ovine tissues for the presence of human cells, total cellular DNA was subjected to PCR analysis for human specific ⁇ -2 microglobulin using a modification of previously described methods (Gilliland, et al., Proc. Nat. Acad. Sci., Vol. 87, pgs.
  • PCR products were subjected to electrophoresis through a 2.5% NuSieve/1% Seakern agarose gel containing 0.5ug ethidium bromide/mL in IX Tris acetate running buffer. The gels were illuminated with UV 280-nm light and photographed with type 55 positive/negative Polaroid film.
  • Immunohistochemistry To verify the PCR results, inrmunohistochemistry for human ⁇ -2 microglobulin was performed as previously described.
  • Tissue Unmasking Fluid Ted Pella, Redding, CA
  • Blocking for 30 minutes at room temperature (RT) was performed using non- immune serum from the species in which the primary antibody was raised (1 :20 dilution), followed by a 12 hour incubation with the specific primary antibody.
  • the primary antibody dilutions used were as follows: human ⁇ -2 microglobulin (Pharmingen International, San Diego, CA, 1 :200); human CD74 (Pharmingen, 1 : 10); or human CD23 (Vector Laboratory, Burlingame, CA, 1:10).
  • the slides were then washed with PBS followed by a second blocking step with methanol containing 0.3% hydrogen peroxide for 30 minutes at room temperature. Slides were then rinsed with deionized water, then PBS, followed by incubation with biotinylated secondary antibody (1 :200 dilution) for 30 min. at RT. The slides were washed with PBS and avidin-biotin complex added for 45 min. at RT. The slides were then rinsed well in PBS, developed with the chromagen 3,3'-diaminobenzidine. For sections stained for human ⁇ -2 microglobulin, CD74, and CD23 the slides then were lightly counterstained with hematoxylin.
  • the human ⁇ -2 microglobulin was developed first using nickel chloride as the chromagen, and then subjected to a secondary immunohistochemical staining for SERCA-2 (Vector Laboratory, 1 :50 dilution) or GFAP (Vector Laboratory, 1 :100 dilution), respectively, as previously described (Van Der Loos, et al., Histochem. J., Vol. 25, pgs. 1-1 1 (1993); VanDer Loos, et al., J. Hist. Cyto., Vol. 42, pgs. 289-294 (1994)). Secondary staining was developed using Vector VIP substrate kit (Vector Laboratory). No counterstaining was performed on these double-stained slides.
  • PCR for human specific ⁇ -2 microglobulin DNA sequences was performed on DNA isolated from liver, spleen, lung, bone marrow, thymus, brain, heart, skeletal muscle, and blood from fetuses transplanted at either 65 or 85 days gestation. Tissue was harvested at 2 weeks, 2 months, or 5 months after in utero transplantation. Two weeks after transplantation human ⁇ -2 microglobulin DNA was detected in all tissues examined in fetal sheep transplanted at 65 and 85 days gestation ( Figure 6A and Table 1), with the exception of skeletal muscle. Cartilage was not examined.
  • Human MSCs could often be appreciated in a single high-power field ( Figures 6B and 6C) in these tissues.
  • Human cells were also identified in non-lymphohematopoietic sites including the heart, skeletal muscle, cartilage, perivascular areas of the CNS, and lung ( Figure 6F). Five months after transplantation, human cells continued to be present in multiple tissues including the bone marrow, thymus, cartilage, heart, skeletal muscle, and brain.
  • Differentiation of human MSCs in various tissues following transplantation was assessed by one of three techniques: 1) characteristic morphology on anti- human ⁇ -2 microglobulin staining; 2) immunohistochemical double-staining for anti-human ⁇ -2 microglobulin and a second non-human specific differentiation marker; or 3) when available, positive staining with human specific differentiation markers proven to not cross-react with sheep cells.
  • site- specific differentiation was confirmed for human cardiomyocytes, chondrocytes, bone marrow stromal cells, thymic stromal cells, and skeletal myocytes. Human cells were identified in the CNS.
  • Cardiomyocyte Differentiation To assess differentiation of human MSCs found in cardiac muscle, double- staining immunohistochemistry was performed using anti-human ⁇ -2 microglobulin and anti-SERCA-2 (Smooth Endoplasmic Reticulum ATPase). At 2 and 5 months after in utero transplantation, human cells were detected in the cardiac muscle of fetuses transplanted at 65 and 85 days gestation. These cells had similar morphology to the surrounding ovine cardiomyocytes and also double-stained with human ⁇ -2 microglobulin and SERCA-2, consistent with human cardiomyocyte differentiation ( Figures 7B and 7C).
  • Chondrocyte differentiation was identified by the finding of human ⁇ -2 microglobulin positive cells in cartilage lacunae of lambs transplanted at 65 days and harvested at 2 months or 5 months after transplantation. Immunohistochemistry was performed using a nickel chloride-based developing technique giving the particulate appearance observed ( Figures 8A and 8B). The immunohistochemical identification of human cells within the lacunae of cartilage specimens that were DNA PCR positive for human ⁇ -2 microglobulin sequences represents clear evidence of human chondrocyte differentiation.
  • Bone Marrow Stromal Differentiation To assess differentiation of human MSCs found in the bone marrow immunohistochemistry was performed using a human specific anti-CD23 antibody (Pharmingen, San Diego, CA, mouse IgGi, Clone M-L233).
  • CD23 is the low affinity IgE receptor and has been shown to be expressed on a variety of cell types including bone marrow stromal cells (Huang, et al, Blood, Vol. 85, pgs. 3704-3712 (1995); Fourcade, et al., European Cytokine Network, Vol. 3, pgs. 539-543 (1992)).
  • tail wounds were created in five 65 day gestation fetal sheep at the time of MSC injection. One animal was sacrificed at one week and four animals at 2 months. Human ⁇ -2 microglobulin DNA was detected by PCR in the one tail wound at 1 week and in one of four tail wounds at 2 months. The PCR results were verified by human ⁇ -2 microglobulin immunihistochemistry (data not shown). The cells expressing human ⁇ -2 microglobulin in the tail wound appeared in the dermis and dermal appendages and had the morphologic appearance of fibroblasts consistent with participation in the wound healing response.
  • Mesenchymal stem cells are of increasing interest to the emerging fields of tissue engineering, cellular transplantation, and gene therapy because of their availability in bone marrow, their relative ease of expansion in culture, their amenability to genetic manipulation, and most importantly, their capacity for differentiation into multiple mesenchymal tissues. These properties support potential clinical applications of: 1) large scale tissue engineering particularly for repair of musculoskeletal injury; 2) cellular therapy for diseases of mesenchymal origin such as muscular dystrophy, osteoporosis, osteogenesis imperfecta, and collagen disorders; 3) bone marrow conditioning to facilitate engraftment of autologous or allogeneic hematopoietic stem cells; and 4) gene therapy. Prenatal MSC transplantation may provide a "reservoir" of normal stem cells to replace defective cells as they become damaged in degenerative diseases with progressive cellular and organ damage.
  • mice There have been two studies in mice in which cultured mouse adherent cell populations have been transplanted and documented to persist following transplantation.
  • cells from transgenic mice expressing a human mini-gene for collagen I were used as mesenchymal progenitor donors and the fate of the cells followed after transplantation into irradiated mice (Pereira, et al., Proc. Nat. Acad. Sci., Vol. 92, pgs. 4857-4861 (1995)).
  • Donor cells were detected in bone marrow, spleen, bone cartilage, and lung up to 5 months later by PCR for the human mini-gene, and a PCR in situ assay on lung indicated that the donor cells diffusely populated the parenchyma.
  • Reverse transcription-PCR assays indicated that the marker collagen I gene was expressed in a tissue-specific manner.
  • a second study transplanted either cultured adherent cells or whole bone marrow into irradiated mice with a phenotype of fragile bones resembling osteogenesis imperfecta caused by expression of the human minigene for type I collagen (Pereira, et al., Proc. Nat. Acad. Sci., Vol. 95, pgs. 1142-1147 (1998)).
  • fluorescense in situ hybridization assays for the Y chromosome indicated that, after 2.5 months, donor male cells accounted for 4-19% of the fibroblasts or fibroblast- like cells obtained in primary cultures of the lung, calvaria, cartilage, long bone, tail, and skin.
  • MSCs although very large cells, can be transplanted, and are capable of homing to and engrafting in multiple tissues, even when transplanted into the fetal peritoneal cavity. This requires the transplanted MSC to cross endothelial barriers, integrate into host tissue microenvironments, and survive with available growth factors and regulatory signals.
  • This may be a function of the ability of specific microenvironments to support the engraftment and differentiation of MSCs, or alternatively, the loss of engraftment from some tissues may be due to heterogeneity, of me transplanted population with respect to differentiation potential or replicative capacity. Immune mediated rejection is less likely since the pattern of engraftment was not limited to immune privileged sites, and an immune mechanism should result in eradication of donor cells.
  • MSCs are capable of site specific, multipotential differentiation and tissue integration following transplantation.
  • Human MSCs have been shown in vitro to differentiate into adipocytic, chondrocytic, or osteocytic lineages (Pittenger, supra). Less well characterized MSC populations from other species have been induced in vitro toward myocytic differentiation.
  • This study confirms in vivo chondrocytic differentiation and for the first time clearly demonstrates in vivo cardiomyocytic and myocytic differentiation of a defined human MSC population.
  • MSCs derived from bone marrow from multiple species have been demonstrated to support hematopoiesis with equal or greater efficacy than stromal layers formed in long term Dexter cultures.
  • CD23 has been identified as a low affinity IgE receptor as well as a functional CD21 ligand (Huang, et al., 1995; Aubry, et al., Cell, Vol.
  • Human MSCs are known to express Class I HLA antigen but do not express Class II, which may limit immune recognition.
  • thymic stromal cells are known to participate in thymocyte positive and negative selection and host thymic antigen presenting cells are capable of facilitating clonal deletion of donor reactive lymphocytes after in utero HSC transplantation (Kim, et al., J. Pediatr. Surg., Vol. 34, pgs.

Landscapes

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

Abstract

La présente invention se rapporte à des cellules embryonnaires mésenchymateuses destinées à une transplantation intra-utérine. Ces cellules embryonnaires mésenchymateuses peuvent être utilisées dans une méthode de traitement d'un foetus, ou dans une méthode de greffe de cellules embryonnaires mésenchymateuses, ou encore dans une méthode de préparation d'un organe en vue de sa transplantation.
PCT/US1999/026927 1998-11-13 1999-11-12 Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines WO2000029002A2 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU14780/00A AU1478000A (en) 1998-11-13 1999-11-12 (in utero) transplantation of human mesenchymal stem cells
EP99972103A EP1128836A2 (fr) 1998-11-13 1999-11-12 Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines
CA002348514A CA2348514A1 (fr) 1998-11-13 1999-11-12 Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines
JP2000582048A JP2002529509A (ja) 1998-11-13 1999-11-12 ヒト間葉幹細胞の子宮内移植で胎児を処置し間葉幹細胞を移植し移植器官を調製する方法と調製された雑種器官

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10835798P 1998-11-13 1998-11-13
US60/108,357 1998-11-13

Publications (2)

Publication Number Publication Date
WO2000029002A2 true WO2000029002A2 (fr) 2000-05-25
WO2000029002A3 WO2000029002A3 (fr) 2000-10-05

Family

ID=22321740

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/026927 WO2000029002A2 (fr) 1998-11-13 1999-11-12 Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines

Country Status (5)

Country Link
EP (1) EP1128836A2 (fr)
JP (1) JP2002529509A (fr)
AU (1) AU1478000A (fr)
CA (1) CA2348514A1 (fr)
WO (1) WO2000029002A2 (fr)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001096865A1 (fr) * 2000-06-14 2001-12-20 Vistagen, Inc. Typage de toxicite au moyen de cellules souches mesenchymateuses
WO2002040061A2 (fr) * 2000-11-15 2002-05-23 Osiris Therapeutics, Inc. Animaux immunocompetents, y compris sous forme d'implants xenogeniques de cellules embryonnaires mesenchymateuses
WO2003047607A1 (fr) * 2001-12-06 2003-06-12 Sankyo Company, Limited Compositions medicales contenant des cellules amniotiques humaines
US8143009B2 (en) 2000-06-14 2012-03-27 Vistagen, Inc. Toxicity typing using liver stem cells
CN106806947A (zh) * 2017-01-13 2017-06-09 宁夏医科大学总医院 一种低免疫原性组织工程皮肤构建方法
WO2019040747A1 (fr) * 2017-08-23 2019-02-28 Wake Forest University Health Sciences Transplantation in utero de cellules exprimant le facteur viii pour le traitement de l'hémophilie
WO2020205969A1 (fr) * 2019-04-02 2020-10-08 The General Hospital Corporation Procédés pour améliorer la régénération de lymphocytes t
CN113215084A (zh) * 2021-06-11 2021-08-06 中国农业科学院兰州兽医研究所 一种羊胎儿皮肤成纤维细胞、其分离方法与应用
CN113941032A (zh) * 2020-07-16 2022-01-18 北京中卫医正科技有限公司 一种间充质干细胞复合补片及其制备方法与应用
US11608486B2 (en) 2015-07-02 2023-03-21 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11634677B2 (en) 2016-06-07 2023-04-25 Terumo Bct, Inc. Coating a bioreactor in a cell expansion system
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11667881B2 (en) 2014-09-26 2023-06-06 Terumo Bct, Inc. Scheduled feed
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11795432B2 (en) 2014-03-25 2023-10-24 Terumo Bct, Inc. Passive replacement of media
US11965175B2 (en) 2016-05-25 2024-04-23 Terumo Bct, Inc. Cell expansion
US11999929B2 (en) 2020-04-10 2024-06-04 Terumo Bct, Inc. Methods and systems for coating a cell growth surface

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2676546A1 (fr) * 2006-04-24 2007-11-08 Stemcell Institute Inc. Procede de preparation d'un organe en vue d'une transplantation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026884A1 (fr) * 1993-05-14 1994-11-24 Biotechnology Research And Development Corporation Cellules souches embryonnaires destinees a la reproduction d'ongules chimeres et transgeniques
WO1997010348A1 (fr) * 1995-09-14 1997-03-20 The Regents Of The University Of California Procedes d'induction de types cellulaires selectionnes
WO1999046366A1 (fr) * 1998-03-13 1999-09-16 Osiris Therapeutics, Inc. Utilisations de cellules souches humaines mesenchymateuses non autologues

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996028967A1 (fr) * 1995-03-17 1996-09-26 Chihiro Koike Mammiferes transgeniques autres que des primates, ou des serotypes de primates superieurs ont ete exprimes par un transfert de genes etrangers et procede pour creer ceux-ci

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026884A1 (fr) * 1993-05-14 1994-11-24 Biotechnology Research And Development Corporation Cellules souches embryonnaires destinees a la reproduction d'ongules chimeres et transgeniques
WO1997010348A1 (fr) * 1995-09-14 1997-03-20 The Regents Of The University Of California Procedes d'induction de types cellulaires selectionnes
WO1999046366A1 (fr) * 1998-03-13 1999-09-16 Osiris Therapeutics, Inc. Utilisations de cellules souches humaines mesenchymateuses non autologues

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
ALMEIDA-PORADA G ET AL: "Human bone marrow stromal cell progenitors are able to induce donor specific tolerance after in utero transplantation." BLOOD, vol. 94, no. 10 SUPPL. 1 PART 1, 15 November 1999 (1999-11-15), page 40a XP002139877 41th annual meeting of the American Society of Hematology, 3-7/12-1999 *
DEVINE S ET AL.: "Studies of mesenchymal stem cells in non-human primates: Evaluation of toxicity and engraftment" BLOOD, vol. 94, no. 10 Suppl. 1, 15 November 1999 (1999-11-15), page 391a XP002139790 41th annual meeting of the American Society of Hematology, 3-7/12/1999 *
LIECHTY K W ET AL.: "Distribution and engraftment of human mesenchymal stem cells (MSC) after in utero transplantation in fetal sheep." BLOOD, vol. 92, no. 10 Suppl. 1, 15 November 1998 (1998-11-15), page 117A XP000914604 40th annual meeting of the American Society of Hematology, 4-8/12/1998 *
PEREIRA R F ET AL.: "Cultured adherent cells from marrow can serve as long-lasting precursor cells for bone, cartilage, and lung in irradiated mice" PROC. NAT'L. ACAD. SCI. USA, vol. 92, May 1995 (1995-05), pages 4857-4861, XP002139789 cited in the application *
PROCKOP D J: "Marrow stromal cells as stem cells for nonhematopoietic tissues" SCIENCE, vol. 276, no. 5309, 4 April 1997 (1997-04-04), pages 71-74, XP002139788 *
SROUR E F ET AL.: "Sustained human hematopoiesis in sheep transplanted in utero during early gestation with fractionated adult human bone marrow cells" BLOOD, vol. 79, no. 6, 15 March 1992 (1992-03-15), pages 1404-1412, XP000569644 *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8143009B2 (en) 2000-06-14 2012-03-27 Vistagen, Inc. Toxicity typing using liver stem cells
US8512957B2 (en) 2000-06-14 2013-08-20 Vistagen Therapeutics, Inc. Toxicity typing using liver stem cells
WO2001096865A1 (fr) * 2000-06-14 2001-12-20 Vistagen, Inc. Typage de toxicite au moyen de cellules souches mesenchymateuses
WO2002040061A2 (fr) * 2000-11-15 2002-05-23 Osiris Therapeutics, Inc. Animaux immunocompetents, y compris sous forme d'implants xenogeniques de cellules embryonnaires mesenchymateuses
WO2002040061A3 (fr) * 2000-11-15 2002-08-22 Osiris Therapeutics Inc Animaux immunocompetents, y compris sous forme d'implants xenogeniques de cellules embryonnaires mesenchymateuses
WO2003047607A1 (fr) * 2001-12-06 2003-06-12 Sankyo Company, Limited Compositions medicales contenant des cellules amniotiques humaines
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11773363B2 (en) 2010-10-08 2023-10-03 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11746319B2 (en) 2010-10-08 2023-09-05 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11708554B2 (en) 2013-11-16 2023-07-25 Terumo Bct, Inc. Expanding cells in a bioreactor
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11795432B2 (en) 2014-03-25 2023-10-24 Terumo Bct, Inc. Passive replacement of media
US11667881B2 (en) 2014-09-26 2023-06-06 Terumo Bct, Inc. Scheduled feed
US11608486B2 (en) 2015-07-02 2023-03-21 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11965175B2 (en) 2016-05-25 2024-04-23 Terumo Bct, Inc. Cell expansion
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11634677B2 (en) 2016-06-07 2023-04-25 Terumo Bct, Inc. Coating a bioreactor in a cell expansion system
CN106806947A (zh) * 2017-01-13 2017-06-09 宁夏医科大学总医院 一种低免疫原性组织工程皮肤构建方法
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
WO2019040747A1 (fr) * 2017-08-23 2019-02-28 Wake Forest University Health Sciences Transplantation in utero de cellules exprimant le facteur viii pour le traitement de l'hémophilie
WO2020205969A1 (fr) * 2019-04-02 2020-10-08 The General Hospital Corporation Procédés pour améliorer la régénération de lymphocytes t
US11999929B2 (en) 2020-04-10 2024-06-04 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
CN113941032A (zh) * 2020-07-16 2022-01-18 北京中卫医正科技有限公司 一种间充质干细胞复合补片及其制备方法与应用
CN113941032B (zh) * 2020-07-16 2023-04-07 北京中卫医正科技有限公司 一种间充质干细胞复合补片及其制备方法与应用
CN113215084A (zh) * 2021-06-11 2021-08-06 中国农业科学院兰州兽医研究所 一种羊胎儿皮肤成纤维细胞、其分离方法与应用

Also Published As

Publication number Publication date
EP1128836A2 (fr) 2001-09-05
CA2348514A1 (fr) 2000-05-25
JP2002529509A (ja) 2002-09-10
AU1478000A (en) 2000-06-05
WO2000029002A3 (fr) 2000-10-05

Similar Documents

Publication Publication Date Title
EP1128836A2 (fr) Transplantation intra-uterine de cellules embryonnaires mesenchymateuses humaines
Mackenzie et al. Human mesenchymal stem cells persist, demonstrate site-specific multipotential differentiation, and are present in sites of wound healing and tissue regeneration after transplantation into fetal sheep
JP2021126129A (ja) 疾患を処置するための遺伝子改変された細胞、組織および臓器
EP2366775B1 (fr) Procédés de développement cellulaire et utilisations thérapeutiques des cellules et des milieux conditionnés produits de cette maniere
JP4950661B2 (ja) 分娩後由来細胞類を使用する、神経組織の再生および修復
US7135171B2 (en) Endothelial precursor cells for enhancing and restoring vascular function
US20120014929A1 (en) Stem cells for treating lung diseases
AU701752B2 (en) Yolk sac stem cells and their uses
US20010049139A1 (en) Hepatic regeneration from hematopoietic stem cells
JP2013172725A (ja) 治療用細胞または細胞培養物を調製する方法
KR20080051191A (ko) 단리된 동종접합 간세포, 그로부터 유래된 분화 세포 및이들을 제조 및 사용하기 위한 물질 및 방법
CN104736160A (zh) 肺及肺部疾病和病症的促炎介质的hUTC调节
WO2006078782A9 (fr) Compositions contenant des cellules d'agm et procedes pour les utiliser
JP2005151907A (ja) 胎盤又は羊膜由来ヒト幹細胞及びその樹立方法並びに臓器への分化誘導方法
CN101558151B (zh) 细胞扩增方法和藉此产生的细胞和条件培养基用于治疗的用途
US20160158292A1 (en) Method and apparatus for recovery of umbilical cord tissue derived regenerative cells and uses thereof
JP2009017891A (ja) 多能性成体幹細胞、その起源、それを得る方法および維持する方法、それを分化させる方法、その使用法、ならびにそれ由来の細胞
WO2001071016A1 (fr) Cellules souches pluripotentielles
WO2007005595A1 (fr) Cellules de progéniteur associées à la grossesse
WO1998012304A1 (fr) Systeme de mise en culture de cellules souches hematopoietiques
JPH06508762A (ja) 卵黄嚢幹細胞
US20040053272A1 (en) Methods of constructing a model of cellular development and differentiation using homozygous stem cell systems, methods of assessing and cataloging proteins expressed therein, cDNA libraries generated therefrom, and materials and methods using same
Péault et al. Haematopoietic stem cell emergence and development in the human embryo and fetus; perspectives for blood cell therapies in utero
JP2003530899A (ja) 造血を増強する方法
Grinnemo Cell transplantation with human mesenchymal or embryonic stem cells to the heart: Experimental, molecular, immunological and echocardiographic studies

Legal Events

Date Code Title Description
ENP Entry into the national phase in:

Ref country code: AU

Ref document number: 2000 14780

Kind code of ref document: A

Format of ref document f/p: F

AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 1999972103

Country of ref document: EP

ENP Entry into the national phase in:

Ref country code: CA

Ref document number: 2348514

Kind code of ref document: A

Format of ref document f/p: F

Ref document number: 2348514

Country of ref document: CA

ENP Entry into the national phase in:

Ref country code: JP

Ref document number: 2000 582048

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1999972103

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 09830139

Country of ref document: US

WWW Wipo information: withdrawn in national office

Ref document number: 1999972103

Country of ref document: EP