WO2003010305A1 - Cellules souches immortalisées - Google Patents

Cellules souches immortalisées Download PDF

Info

Publication number
WO2003010305A1
WO2003010305A1 PCT/DK2002/000514 DK0200514W WO03010305A1 WO 2003010305 A1 WO2003010305 A1 WO 2003010305A1 DK 0200514 W DK0200514 W DK 0200514W WO 03010305 A1 WO03010305 A1 WO 03010305A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell line
stem cell
immortalized
bone marrow
Prior art date
Application number
PCT/DK2002/000514
Other languages
English (en)
Inventor
G. Jensen Thomas
Kassem Moustapha
I. S. Rattan Suresh
Original Assignee
Arhus Amt
Arhus Universitet
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 Arhus Amt, Arhus Universitet filed Critical Arhus Amt
Priority to EP02764564A priority Critical patent/EP1430115A1/fr
Publication of WO2003010305A1 publication Critical patent/WO2003010305A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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)
    • 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
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/04Immortalised 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13041Use of virus, viral particle or viral elements as a vector
    • C12N2740/13043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to the field of regeneration and functional restoratiori of damaged and diseased tissue and to the manufacture of immortalized stem cell lines.
  • Human bone marrow contains a population of non-hematopoietic cells. These cells known as marrow stromal cells (MSC) play a role in supporting the hematopoietic cells (hence the name stroma). However, it has been increasingly recognised that these cells include a population of stem cells that are able to differentiate into sev- eral mesenchymal cell types including: osteoblasts, chondrocytes, adipocytes, and fibroblasts in addition to the marrow-supporting stroma.
  • US 5,942,225 describes lineage-directed induction of human mesenchymal stem cell differentiation. The molecular mechanisms controlling the differentiation of MSC into lineage-specific phenotype is not completely known. Detailed molecular studies on the cellular dif- ferentiation of MSC have been obstructed by the inability to obtain normal cells in a number large enough to allow such studies.
  • MSC Mesenchymal stem cells
  • MSC Mesenchymal stem cells
  • limb-bud mesoderm tissue that generates bone, cartilage, fat, skeletal muscle and possibly endothelium.
  • Mesoderm also differentiates to visceral mesoderm, which can give rise to cardiac muscle, smooth muscle, or blood islands consisting of endothelium and hematopoietic progenitor cells.
  • Primitive mesodermal or mesenchymal stem cells therefore, could provide a source for a number of cell and tissue types.
  • a tissue specific cell that has been named a stem cell is the mesenchymal stem cell, initially described by Fridenshtein, 1992.
  • T-antigen (T) gene has been tradi- tionally employed as an immortalizing agent (Hicok et al, 1998). Immortalization using the large T-antigen gene is known to induced several changes in the normal cell cycle machinery that may affect cells differentiation capacity and thus findings from T + -cell lines will be of limited value to understand the normal human physiology.
  • Telomeres are repetitive DNA sequences at the end of chromosomes that protect the ends of the chromosomes against degradation and prevent ligation of the ends of DNA by DNA-repair enzymes.
  • the limited ability of normal cells for extensive proliferation has been suggested to be due the successive shortening of the telomeres during cell replication.
  • a strong evidence supporting this hypothesis is the recent demonstration of an extended life span of human fibroblasts by forced expression of the human telomeric subunit gene (TRT) (Bodnar et al., 1998). The extended life span was not associated with evidence of transformation or tumor-genesis. Also, these cells maintain a pattern of onco-gene and tumor suppressor gene expression similar to normal cells.
  • the inventors have employed a retroviral vector containing the human TRT gene to create an immortalized human marrow stromal cell line. It is demonstrated that MSCs overexpressing the human TRT gene exhibit an extended life span in culture and are able to differentiate into multiple lineages (e.g. osteo- blasts, adipocytes and chondrocytes).
  • multiple lineages e.g. osteo- blasts, adipocytes and chondrocytes.
  • the present invention relates to a method of immortalizing human stem cells, comprising the following steps,
  • telomeric repeat subunit hTRT
  • an immortalized cell line wherein said immortalized cell line has substantially identical characteristics and properties when compared to the bone marrow stromal cells of step a).
  • the invention also concerns the use of the method.
  • the invention discloses an immortalized stem cell line obtainable by the method of the invention and the use of said cell line.
  • Figure 1 shows the growth curve of immortalized cells compared to control cells. 980 controls and 980 TRT cells were grown in culture and their proliferative potential was measured as population doublings (PD).
  • Figure 2 shows a telomerase assay for TERT cells.
  • Figure 3 shows the DNA-sequence of the TERT coding fragment, which was incorporated into a retroviral vector. The fragment is from position 4215 to position 7668. Insert info: 4215 EcoR1 site, 4225 start of hTRT ORF (ATG codon), 7620 end of hTRT ORF, 7621 termination codon (TGA), 7626 end of hTRT cDNA sequence, 7668 EcoR1 site.
  • telomeric repeat subunit TRT
  • telomeric repeat subunit hTRT
  • Step a) obtaining an immortal stem cell line, wherein said immortalized cell line has substantially identical characteristics and properties when compared to the bone marrow stromal cells of step a).
  • the human bone marrow stromal cells are cultured in accordance with standard conventional cell culturing methods known to the skilled person. An example of the method is described in the section "Examples" of the present specification.
  • the hMSC cell cultures of the invention are transducted with a retroviral vector.
  • the transduction method is preferably performed by the following steps:
  • step b5) transducing human bone marrow stromal cells with the packaging cells of step b4), said cells comprising said retroviral vector.
  • transduction is intended to cover the transfer of bacterial genes (DNA) from one bacterium to another by means of a bacterial virus (bacteriophage).
  • DNA bacterial genes
  • bacteriophage bacterial virus
  • transduction There exist two kinds of transduction, namely specialised and general.
  • specialised transduction a restricted group of host genes becomes integrated into the virus genome.
  • guest genes usually replace some of the virus genes and are subsequently transferred to a second bacterium.
  • host genes become a part of the mature virus particle in place of, or in addition to the virus DNA.
  • the genes can come from virtually any portion of the host genome and this material does not become directly integrated into the virus genome.
  • the packaging cells are cells in which the retroviral vector of the invention is produced.
  • it means a cell line capable of producing a vector which can transduce human cells.
  • the packaging cell line may be cells, such as PA317 or Phenix.
  • the packaging cell line may be PG13 cells.
  • This particular cell line is made in mouse embryo fibroblasts by transfection with the Gibbon Ape leukemia virus (GALV) env gene and the Moloney murine leukemia virus gag and pol genes.
  • the PG13 cells are commercially available and may be obtained from ATCC, Rockville, MD.
  • the PG13 cells may be cultured for 5-15 days, such as 7- 13 days, for example 9-11 days before they are used to produce the retroviral vector.
  • vector is meant as an agent used to carry new genes into cells.
  • Vectors may be plasmids, viruses or bacteria.
  • a "retrovirus” in the present context is used to carry new genes into cells. These molecules become part of that cells genome.
  • Retroviruses are divided into three subfamilies based primarily on their association with disease: oncovirinae, lentivirinae and spumavirinae.
  • the retroviruses most widely used for gene transfer is Moloney murine leukemia virus which belong to the oncovirinae group.
  • MFG Moloney murine leukemia virus
  • the retroviral vector of the present invention comprises at least a fragment of the catalytic subunit of the human telomeric repeat (hTRT) gene or a variant thereof.
  • a retrovirus (from the Latin word retrovir, which means "backward man") is an onco- genic, single-stranded, diploid RNA (ribonucleic acid) virus containing (+) RNA in its virion and propagate through a double-helical DNA intermediate. It is called a retrovirus because its genetic information flows from RNA to DNA (reverse of normal). That is the virus contains an enzyme that allows the production of DNA using RNA as a template. Retroviruses can only infect cells in which DNA is replicating.
  • the telomerase gene inserted into the retroviral vector is a hTERT cDNA fragment.
  • a 3452 base pair EcoR1 fragment from pGRN145 (Geron), or a variant thereof comprising all the coding sequences may be employed.
  • the important features of said vector are the efficiency of gene transfer, the ability to become integrated in the genome and the possibility for long term gene expression.
  • retroviral vectors are best suited for the purpose of TERT-immortalization of stromal cells due to the efficient integration.
  • other vectors based on the Moloney murine leukemia virus or for example lentiviral vectors is within the scope of the present invention.
  • the retroviral vector is driven by a Moloney murine leukemia virus long terminal repeat (GCSam).
  • GCSam Moloney murine leukemia virus long terminal repeat
  • telomere in the present context a variant means a functional equivalent of telomerase as identified by the sequence in Figure 3 (3452 amino acids corresponding to the TERT protein), and functional equivalents of a fragment of telomerase comprising a predetermined amino acid sequence are defined as:
  • fragments comprising an amino acid sequence capable of being recognised by an antibody also capable of recognising the predetermined amino acid sequence, and/or
  • fragments comprising an amino acid sequence capable of binding to a receptor moiety also capable of binding the predetermined amino acid se- quence, and/or iii) fragments having at least a substantially similar catalytic effect as the fragment of telomerase comprising said predetermined amino acid sequence.
  • the fragment of telomerase according to the present invention may in one embodiment comprise less than 5000 amino acid residues, such as less than 4500 amino acid residues, for example less than 4000 amino acid residues, such as less than 3500 amino acid residues, for example less than 3000 amino acid residues, such as less than 2500 amino acid residues, for example less than 2000 amino acid residues.
  • Any functional equivalent may be obtained by addition, substitution or deletion of at least one amino acid.
  • amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated by the inventors that various changes may be made in the peptide sequences of the disclosed compositions, or corresponding DNA sequences which encode said peptides without appreciable loss of their biological utility or activity. In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art.
  • transfection refers to a special case of transformation in which an appropriate recipient strain of bacteria is exposed to (free) DNA isolated from a transducing phage with the "take-up" of that DNA by some of the bacteria and consequent production and release of complete virus particles.
  • the process involves the direct transfer of genetic material from donor to recipient.
  • the retroviral vector of the invention may be transfected into GGP+E86 packaging cell lines. These cell lines are made in NIH3T3 mouse fibroblasts by transfection with the gag, pol and env genes (Markovitz et al., 1988). Cells are obtained from ATCC, Rockville, MD.
  • transfected cells GGP + E86 cells containing the retroviral vector are hereafter used to transduce the PG13 packaging cells.
  • the transduction of the human bone marrow stromal cells is at a time of division of said cells. It is preferred that the transduction is performed when the human bone marrow stromal cells have a density of 5-25%, preferably 10-20%. In the present context "density" is measured as the number of cells per predefined surface area of culture plates. The method of counting the cells in a counting chamber is known to the person skilled in the art.
  • the transduction of the human bone marrow stromal cells is performed by using centrifugation.
  • the centrifugation may be performed in the range between 200-2000 g, such as between 300-1800 g, for example between
  • the temperature during centrifugation may be between 25- 37 °C, such as between 27-35 °C, for example between 29-33 °C.
  • the centrifugation is performed for a period of between 5-55 minutes, such as between 10-50 minutes, for example between 15-45 minutes, such as between 20-40 minutes, for example between 25-35 minutes.
  • the centrifugation is performed at 1000 x g and 32 °C for 30 minutes. It is speculated that the centrifugation step is crucial to obtaining the immortal stem cell line.
  • the present inventors have found that the centrifugation step generally increases transduction ten-fold, such as nine-fold, for example eight-fold, such as seven-fold, for example six-fold, such as five-fold, for example four-fold, such as three-fold, for example two-fold.
  • the probability of obtaining immortalization is directly correlated to the transduction frequency.
  • an immortal stem cell line wherein the immortalized cell line has substantially identical characteristics and properties when compared to the bone marrow stromal cells of step a), i.e. bone marrow stromal cell which have not been immortalized.
  • variants include those sequences substantially identical (determined as described below) to a telomerase sequence encoding polypeptides that are either mutants of wild type telomerase polypeptides or retain the function of the telomerase polypeptide (e.g., resulting from conservative substitutions of amino acids in the telomerase polypeptide).
  • variants can be those that encode dominant negative mutants as described below.
  • nucleic acid sequences or polypeptides are said to be “identical” if the sequence of nucleotides or amino acid residues, respectively, in the two sequences is the same when aligned for maximum correspondence as described below.
  • the terms “identical” or percent “identity,” in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or sub-sequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence over a comparison window, as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
  • sequence identity When percentage of sequence identity is used in reference to proteins or peptides, it is recognised that residue positions that are not identical often differ by conservative amino acid substitutions, where amino acids residues are substituted for other amino acid residues with similar chemical properties (e.g., charge or hydrophobicity) and therefore do not change the functional properties of the molecule. Where sequences differ in conservative substitutions, the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution.
  • substantially identical in the context of two nucleic acids or polypeptides, refers to sequences or sub-sequences that have at least 60%, preferably 80%, most preferably 90-95% nucleotide or amino acid residue identity when aligned for maximum correspondence over a comparison window as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
  • This definition also refers to the complement of a test sequence, which has substantial sequence or sub-sequence complementarity when the test sequence has substantial identity to a reference sequence.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, sub-sequence co-ordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • Constantly modified variants applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine
  • each silent variation of a nucleic acid which encodes a polypeptide is implicit in each described sequence.
  • the identity may be assessed by characterising and comparing the cell expression of certain cell markers, such as collagen type I or Alkaline phosphatase or any other relevant cell marker.
  • the present invention overcomes the prior art problem of having immortalized cells differing molecularly and possibly also phenotypically from the initial cells used for immortalisation. This is an advantage when employing the immortalized stem cells of the present invention, for example for medical purposes, since it is important to apply immortalized cells identical to the originals to achieve successful applications.
  • the human bone marrow stromal cells are capable of differentiating into cell types selected from, but not limited to the groups consisting of osteoblastic, adipocytic or chondrocytic phenotype.
  • the invention concerns an immortalized stem cell line obtainable by the method as described herein above.
  • the immortalized stem cell line is characterised in that it comprises a hTRT (human telomeric repeat gene) vector.
  • each cell of the transducted stromal cell line comprises a vector of the invention.
  • the transduction is performed when the human bone marrow stromal cells have a density of 5-25%, preferably 10-20% in culture.
  • a cell is defined as immortal when it has exceeded the double life span of the normal cell (i.e. non-immortalized cell).
  • the maximal life span of a normal diploid cell in vitro varies dependent on the cell type (fetal versus adult cell) and culture conditions.
  • the maximum life span of cultured normal cells in vitro is approximately 60-80 population doublings.
  • keratinocytes may divide around 80 times, fibroblasts more than 50 times, and lymphocytes about 20 times.
  • Normal bone marrowstromal cells may exhibit a maximal life span of 30-40 population doublings.
  • a stromal cell line may continuously grow past 350 population doublings and may still maintain a normal growth rate characteristic of young cells.
  • a crucial aspect of the present invention is that the transduction of the human bone marrow stromal cells is performed using centrifugation as described above.
  • the present invention further relates to the use of the method for immortalizing stem cells as well as the use for an immortalized stem cell line according to the invention.
  • the present invention provides a method for using specifically differentiated cells for therapy, comprising administering the specifically differentiated cells to a patient in need thereof. It further provides for the use of genetically engineered pluripotent stem cells to selectively express an endogenous gene or a transgene, and for the use of MSCs grown in vivo for transplantation/administration into an animal to treat a disease.
  • genetically engineered pluripotent stem cells to selectively express an endogenous gene or a transgene
  • MSCs grown in vivo for transplantation/administration into an animal to treat a disease For example, neuroretinal cells derived from pluripotent stem or MSCs can be used to treat blindness, caused by among other things but not limited to neuroretinal disease, caused by among other things macular degeneration, diabetic reti- nopathy, glaucoma, retinitis pigmentosa.
  • the cells of the invention can be used to engraft a cell into a mammal, comprising administering autologous, allogenic or xenogenic cells, to restore or correct tissue specific metabolic, enzymatic, coagulation, structural or other functions to the mammal.
  • the present cells can be used to engraft a cell into a mammal, causing the differentiation in vivo of cell types, and for administering the differentiated stem cells into the mammal.
  • the present cells may be used to correct diseases, such as a genetic disease, degenerative disease, cardiovascular disease, metabolic storage disease, neural disease and/or a cancer disease.
  • the cells may be used to produce gingiva- like material for treatment of periodontal disease and in a further embodiment the present cells may be to be used to develop skin epithelial tissue derived from pluripotent stem cells utilized for skin grafting and plastic surgery. They may be used to produce blood ex-vivo for therapeutic use, or to produce human hematopoietic cells and/or blood in prenatal or postnatal animals for human use.
  • the cells may be used as a therapeutic to aid for example in the recovery of a patient from chemotherapy or radiation therapy due to the treatment of cancer, or be used in the treatment of auto-immune disease, or the cells may be used to induce tolerance in the recipient, such as in the treatment of AIDS or other infectious diseases.
  • the MSC differentiated into cardiomyocytes may be used to treat cardiac diseases including among others but not limited to myocarditis, car- diomyopathy, myocardial infarction, hypertension, atherosclorosis, heart valve dys- function.
  • a genetically engineered pluriipotent mammalian derived stem cell, or its differentiated progeny may be used to treat a disease with Central Nervous System (CNS) deficits or damage.
  • CNS Central Nervous System
  • the present pluripotent mammalian derived stem cell, or its neuronally related differentiated cell may be used to treat a disease with neural deficits or degeneration including among but not limited to stroke, Alz- heimer's, Parkinson's disease, Huntington's disease, AIDS associated dementia, spinal cord injury, metabolic diseases effecting the brain or other nerves.
  • a disease with neural deficits or degeneration including among but not limited to stroke, Alz- heimer's, Parkinson's disease, Huntington's disease, AIDS associated dementia, spinal cord injury, metabolic diseases effecting the brain or other nerves.
  • It is an object of the invention to provide a pluripotent mammalian derived stem cell, or their differentiated progeny such as stromal cells may be used to support the growth and differentiation of other cell types in vivo or in vitro, including but not limited to hematopoietic cells, pancreatic islet insulin-producing ⁇ cells, and hepato- cytes.
  • the present stem cell, or cartilage differentiated progeny may be used to treat a disease of the joints or cartilage including but not limited to cartilage tears, cartilage thinning, osteoarthritis.
  • the stem cells of the invention, or their osteoblast differentiated progeny may be used to ameliorate a process having deleterious effects on bone including among but not limited to bone fractures and non-healing fractures, "defects" in bones caused by the removal of primary bone tumors, or bone metastases spreading to said bones, such as in breast cancer or lung cancer. Bone loss
  • One such purpose may be for the treatment of bone loss associated with ageing and/or osteoporosis.
  • the immortalized cells will be able to replace cells lost during such diseases.
  • Another application of the invention is for use in the treatment of bone-fractures.
  • Prosthesis coating In the field of orthopaedic surgery, prosthesis loosing with age is a major problem for joint replacement technology. It is envisaged that coating the prosthesis surfaces with the immortalized cell line of the invention loaded on a carrier can increase the adhesion of the prosthesis to the bone and prevent loosening.
  • the cells are ideal candidates for deriving endocrine-hormone secreting cells (e.g. insulin-producing, growth hormone-producing) from the present cell line. This may be done either through induction of differentiation of stem cells into these cell types by chaning culture conditions or through transfection of the cells with genes responsible for hormone synthesis and secretion. Such modified cells may be used for treatment of hormone deficiency, for example diabetes mellitus or growth hormonedeficiency.
  • endocrine-hormone secreting cells e.g. insulin-producing, growth hormone-producing
  • the immortalized stem cell line may be used for tissue engineering, such as for creating bone matrix for the treatment of bone defects and non- healed fractures and for creating cartilage matrix for the treatment of cartilage defects and osteoarthritis.
  • the stem cells may be used for creating general artificial tissues in vitro, such as skin, cartilage, heart valves and vascular tissues, all which can be used in replacement therapy. It is also within the scope of the invention to use the immortalized cells to create a skin matrix for the treatment of skin defects (including cosmetic purposes) after skin burns.
  • the immortalized stem cells of the invention may be used in the production and secretion of growth factors, such as VEGF (Vascular Endothelial Growth Factor),
  • PDGF Plater Derived Growth Factor
  • IGFs Insulin-like growth factors
  • the immortalized stem cells are used as "metabolic sink".
  • the immortalized stem cells of the present invention are capable of metabolising substances accumulating during the course of various diseases.
  • metabolites could be Phenylalanine in phenylke- tonuri (PKU); Ornithine in Gyrate Atrophy or Adenosine in Adenosine deaminase (ADA) deficiency, cholesterol in hypercholestremic states.
  • the immortalized cells may produce metabolic enzymes, such as Adenosine Deaminase (ADA), Ornithin Aminotransferase (OAT) or Phenylalanine Hydroxylase (PAH).
  • ADA Adenosine Deaminase
  • OAT Ornithin Aminotransferase
  • PAH Phenylalanine Hydroxylase
  • the use of the immortalized stem cells may be for drug delivery of compounds, such as Coagulation factor VIII (Hemophilia A), Coagulation factor IX (Hemophilia B), Erythropoietin (EPO), insulin, leptin, angiostat- ins/endostatins, human growth hormone and/or interleukins (II).
  • compounds such as Coagulation factor VIII (Hemophilia A), Coagulation factor IX (Hemophilia B), Erythropoietin (EPO), insulin, leptin, angiostat- ins/endostatins, human growth hormone and/or interleukins (II).
  • the main routes of drug delivery, in the treatment methods are intravenous, oral, and topical.
  • Other drug-administration methods such as subcutaneous injection or via inhalation, which are effective to deliver the drug to a target site or to introduce the drug into the blood stream, are also contemplated.
  • the administration is systemic.
  • the cells are intravenously injected, whereby the cells are positioned with for example bone marrow or bone structures.
  • Immortalized stem cells may be implanted subcutanously, such as in immunoprotective capsules or in bone structures.
  • the immortalized stem cells are used for drug testing, such as for identifying drugs acting on mesenchymal stem cells and/or drugs enhancing osteoblast differentiation, chondrocyte differentiation, neuronal cell differentiation and/or muscle cell differentiation,.
  • drug testing such as for identifying drugs acting on mesenchymal stem cells and/or drugs enhancing osteoblast differentiation, chondrocyte differentiation, neuronal cell differentiation and/or muscle cell differentiation.
  • This provides for a unique opportunity of testing drugs on normal human cells capable of differentiation in a multitude of cell types.
  • the present invention represents a good alternative for drug testing on small animals, and is beneficial in drug discovery studies.
  • the immortalized stem cells may be employed in gene therapy.
  • Genetic material such as genes or functional parts thereof may be introduced in constructs as a single DNA molecule encoding all of the genes, or different DNA molecules having one or more genes.
  • the constructs may be introduced simultane- ously or consecutively, each with the same or different markers.
  • the gene may be linked to the complex as such or protected by any suitable system normally used for transfection, such as viral vectors of the invention or artificial viral envelope, liposomes or micellas, wherein the system is linked to the complex.
  • virus vectors which allow transfection and random integration of the virus into the host.
  • Routes and modes of administering the vector include injection, e.g intravascularly or intramuscularly, inhalation, or other paren- teral administration.
  • the immortalized cells are used for large scale production of pure proteins which may then be used in crystallographic characterization.
  • hMSc human bone marrow stromal cells
  • Modified essential medium (MEM) (Life Technology, Denmark) containing 10% Foetal Calf Serum (FCS) (Bio-Whittaker, Belgium) was used for all cell types.
  • MEM containing 15% Normal Horse Serum (Sigma, USA) and 10 "7 M Dexamethasone (Sigma, USA), and MEM containing 290 nM Ascorbic Acid, were used respectively to induce adipocytic differentiation and mineralisation.
  • 1.25-vitamin D 3 (vit. D) (Hoffman La Roche, Denmark) treatment was carried out for 48 hrs with a concentration of 10 "8 M in standard medium.
  • telomerase gene and the green fluorescence protein (GFP) gene (as control) inserted in the GSsam retroviral vector (from T. G. Jensen, Aarhus, Denmark) were used to immortalise hMSc.
  • These retroviral vectors are driven by a Moloney murine leukemia virus (MoMLV) long terminal repeat (LTR), and they were packaged in the gibbon ape leukemia virus (GALV) PG13 packaging cell line.
  • MoMLV Moloney murine leukemia virus
  • LTR long terminal repeat
  • GLV gibbon ape leukemia virus
  • Supematants from the PG13 virus producing cells were collected and used to transduced hMSc. 9 days old in culture hMSc plated on a 6 well/plate were exposed to the virus supematants containing 8 ⁇ g/ml polybrene.
  • the plates were centrifuged for 30 min. at 1000xg at 32°C, and then incubated for 2 hrs. at 37°C. Medium was then changed and cells were monitored for GFP expression during the next two days, in order to asses the quality of transduction. Cell medium was changed once a week and the cells were passaged at confluence.
  • DMEM Dulbecco's modified Eagle's medium
  • DMEM Dulbecco's modified Eagle's medium
  • penicillin and streptomycin Gibco, Life Tech- nology
  • 2 mM of L-glutamine Gibco, Life Technology
  • Retroviral vectors and packaging cell lines The vector pGRN1212 containing the human TERT gene (hTERT) was kindly provided by Geron Corp., Ca, USA (see figure 3).
  • the retroviral vector GCsamTERT which does not contain a neo gene was constructed by inserting the hTERT gene, isolated from pGRN1212, into the retrovi- ral vector GCsam (Chuah et al, 1995) . In this vector the expression of the transgene is driven from a Moloney Murine Leukemia Virus LTR.
  • GCsamTERT was packaged in PG13 as described (Onodera et al, 1997).
  • GCsamTERT DNA was transfected into GP+E86 (Markovitz et al., 1988) by calcium phosphate co-precipitation (Mam- malian transfection kit, Stratagene, La Jolla, CA). Supernatant from transfected cells was harvested 16 hours later, supplemented with polybrene to a concentration of 8 ⁇ g/ml, passed through a 0.45 ⁇ m filter, and used to transduce PG13 cells by using a centrifugation technique (Bunnell et al, 1995). A population of the packaging cell line was used for production of the retroviral vector.
  • the retroviral vector GCsamGFP containing the eGFP gene, was constructed and packaged as described previously (Jensen et al, 2000).
  • Transduction of stroma cells was performed using a centrifugation technique (Bunnell et al, 1995). Polybrene (8 /g/ml) was added to fresh supernatant from packaging cell lines cultivated at 37 °C, and the supematants were passed through a 0.45 ⁇ m filter. Stroma cells, seeded the day before at a con- fluency of 10-20%, in a well of a 6 well plate (NUNC) were treated with 2 ml of retroviral supernatant. Plates were centrifuged 30 minutes at 32 °C at 1000 x g and then incubated at 37 °C 16 hours. Thereafter, the supematants were removed, and cells were refed with fresh complete media.
  • NUNC 6 well plate
  • telomerase activity was assessed by TRAP using the TRAPeze Telomerase Detection Kit, Oncor, USA, based on the protocol first described by Kim et al. Briefly, 100000 cells were pelleted and lysated in 30 ⁇ l of a CHAPS containing lysis buffer. 2 ⁇ l of the lysate were incubated for 30 min. at 30 °C to promote amplification. The amplification products were then subjected to 2 step PCR and the PCR products were visualised with an Egle Eye System after PAGE and staining in Ethidium Bromide solution.
  • hMSc were stained for ⁇ -galactosidase.
  • Cells were washed in PBS, fixed in 2% formaldehyde/0.2% gluta- raldehyde in PBS for 3-5 min at room temperature. The cells were then incubated at 37 °C with freshly made D-galactosidase stain solution (1 mg 5-bromo-4-chloro-3- indolyl ⁇ -D-galactosidase [Sigma, USA] per ml, 40 mM Citric acid sodium phosphate, pH. 6.0, 5 mM Potassium ferrocyanide, 150 mM NaCI, and 2 mM MgCI 2 ). Staining reaction was stopped after 16 hrs. incubation.
  • RNA extraction cDNA synthesis, and RT-PCR.
  • PCR was run using Amplitaq gold, buffer, and reagents from PE Biosystem, Denmark, in a final volume of 20 ⁇ l per sample [10x PCR buffer, MgCI 2 1.5 mM, dNTP 0.2 mM each dATP, dGTP, dCTP, and dTTP, and Amlitaq Gold polymerase
  • Adipocvtic differentiation Cells grown in adipocvtic medium for 14 days were fixed in 4% neutral buffer formalin for 10 min. followed by a wash with 3% isopropanol. Cells were then stained with an Oil red O solution (0.5 g Oil red O, 100 ml isopropanol, and 67 ml water) for 1 hr at room temperature.
  • Oil red O solution 0.5 g Oil red O, 100 ml isopropanol, and 67 ml water
  • HA/TCP Hydroxyapatite/tricalcium phosphate
  • Transplantation was performed under general anathesia achieved by intraperitoneal injection of ketamin (2.5 mg/ml) and xylazin (4 mg/ml) in phyisiological saline at 0.1 ml/g body weight. Mice were shaved and the skin was disinfected with 70% ethanol prior to operation. Two midlongitudinal skin incisions of about 1 cm were made on the dorsal surface of each mouse and subcutaneous pockets were formed by blunt dissection. A singel transplant was placed into each pocket. The incision were closed. After 8 weeks the transplants were recovered, fixed in ethanol (70%) and embedded unde- calcified in methyl-methacrylate. Tissue sections were cut 7.5 urn thick and were stained for Goldner-trichrome.
  • telomere activity was not detectable in different strains of normal human MSC including the parental strain of the TRT 1" - MSC.
  • the integration site and copy number of the ectopic hTERT gene were determined using fluorescent in situ hybridization (FISH). Twenty metaphases and 100 nuclei were evaluated. The ectopic hTERT was found to be integrated at chromosome 5q23-31. All nuclei showed only one copy of ectopic hTERT and two copies of endogenous hTERT.
  • ⁇ -galactosidase ( ⁇ -gal) activity at neutral pH was employed as a marker of senescent phenotype.
  • ⁇ -galactosidase ( ⁇ -gal) activity at neutral pH was employed.
  • the senescent phenotype of normal MSC was associated with the positive staining for ⁇ -gal in more than 90% of the cells.
  • Staining for the ⁇ -gal was performed regularly at different population doubling levels of the TRT + -MSC. Only a small and a stable proportion (less than 10%) of the cells stained positive for ⁇ -gal suggesting the maintenance of a "young" phe- notype in spite of the extensive proliferation.
  • TRT + -MSC In order to investigate the cellular phenotype characteristics of TRT + -MSC, the ability of the cells to express known osteoblastic phenotype markers was examined. TRT+-MSC were found to express constitutively known osteoblast-specific genes: alkaline phosphatease (AP), osteopontin, core binding factor 1 (CBFA-1) and osteo- calcin (OC) in presence of calcitriol (10 "7 M). The expression levels of these osteo- blast-specific genes were found to be stable during the extended period of growth in culture. In addition to mRNA expression, MSCs stained positive for AP and type I collagen (Coll) confirming the observed findings on the mRNA level. Employing an assay for in vitro mineralization, the cells were able to form a mineralized matrix in vitro at confluency as demonstrated by staining by alizarin red.
  • AP alkaline phosphatease
  • CBFA-1 core binding factor 1
  • OC osteo- calcin
  • TRT + - MSC maintained the adipocytic-forming capacity.
  • TRT + -MSC were incubated with adipogenic medium containing normal horse serum and dexamethasone (10 "7 M). After incubation for 2 weeks, TRT + -MSC were able to form morphologically recognisable adipoctyes with oil-filled cytoplasm and showed positive histochemical staining for oil red O.
  • the cells expressed constitutively lipoprotein lipase and proxisome proliferator activator receptor-gamma (PPAR- ⁇ ) known adipocyte-specific genes.
  • PPAR- ⁇ proxisome proliferator activator receptor-gamma
  • the differentiation potential of hTERT-MSC were tested by culturing the cells in different combinations of growth factors and cyokines to induce a lineage specific phenotype.
  • the cells were able to express markers for neuronal cells, myocytic cells, endothelial cells and B-lymphocytes.

Abstract

L'invention concerne un procédé pour immortaliser des cellules souches humaines par la culture de cellules du stroma de la moëlle osseuse humaine (hMSC) et par la transduction des cultures cellulaires au moyen d'un vecteur rétroviral, comprenant le gène de sous-unité de répétition télomérique humaine (hTRT). La présente invention porte également sur une ligne de cellule souche immortalisée et sur son utilisation. Les cellules souches ainsi immortalisées peuvent, par exemple, être utilisées dans le traitement de fractures osseuses, de perte osseuse associée au vieillissement et/ou à l'ostéoporose, et dans l'ingéniérie tissulaire.
PCT/DK2002/000514 2001-07-27 2002-07-26 Cellules souches immortalisées WO2003010305A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP02764564A EP1430115A1 (fr) 2001-07-27 2002-07-26 Cellules souches immortalis es

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200101148 2001-07-27
DKPA200101148 2001-07-27

Publications (1)

Publication Number Publication Date
WO2003010305A1 true WO2003010305A1 (fr) 2003-02-06

Family

ID=8160642

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2002/000514 WO2003010305A1 (fr) 2001-07-27 2002-07-26 Cellules souches immortalisées

Country Status (2)

Country Link
EP (1) EP1430115A1 (fr)
WO (1) WO2003010305A1 (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1447443A1 (fr) * 2001-10-31 2004-08-18 Renomedix Institute Inc. Cellules mesenchymateuses immortalisees et leur utilisation
EP1727892A1 (fr) * 2004-03-22 2006-12-06 Osiris Therapeutics, Inc. Cellules souches mesenchymateuses et leurs utilisations
CN100408677C (zh) * 2003-04-09 2008-08-06 中国人民解放军军事医学科学院野战输血研究所 一种bdnf基因修饰的间充质干细胞的构建方法
EP2163243A1 (fr) 2008-09-12 2010-03-17 Biocompatibles UK Limited Traitement de l'infarctus du myocarde (AMI) utilisant des cellules encapsulées codant et sécrétant des peptides GLP-1 ou analogues associés
EP2174655A1 (fr) 2006-05-10 2010-04-14 Biocompatibles UK Limited Microcapsules sphériques contenant des peptides du type GLP-1, leur production et leur utilisation
EP2223696A1 (fr) * 2007-05-24 2010-09-01 Apceth GmbH & Co. KG Cellules souches mésenchymales CD34- pour l'utilisation dans la thérapie génique visant la guérison de blessures
EP2251028A1 (fr) 2009-05-12 2010-11-17 Biocompatibles Uk Ltd. Traitement des maladies des yeux utilisant des cellules encapsulées codant et sécrétant un facteur anti-angiogénique et/ou un facteur neuroprotecteur
WO2012034704A1 (fr) 2010-09-15 2012-03-22 Biocompatibles Uk Ltd. Traitement de maladies vasculaires à l'aide de cellules encapsulées codant pour le glp-1 et le sécrétant, ou un fragment ou un variant de celui-ci
US8431533B2 (en) 2005-09-22 2013-04-30 Biocompatibles Uk Ltd. GLP-1 fusion peptides, their production and use
CN109022355A (zh) * 2018-08-30 2018-12-18 丰泽康生物医药(深圳)有限公司 一种提高间充质干细胞向软骨细胞分化的培养方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001011011A2 (fr) * 1999-08-05 2001-02-15 Mcl Llc Cellules souches adultes toutes-puissantes et procede d'isolement

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001011011A2 (fr) * 1999-08-05 2001-02-15 Mcl Llc Cellules souches adultes toutes-puissantes et procede d'isolement

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ANDREA G.BODNAR ET AL: "Extension of life-span by introduction of telomerase into normal human cells", SCIENCE, vol. 279, January 1998 (1998-01-01), pages 349 - 352, XP002902750 *
BUNNELL ET AL: "HIGH-EFFICIENCY RETROVIRAL MEDIATED GENE TRANSFER INTO HUMAN AND NONHUMAN PRIMATE PERIPHERAL BLOOD LYMPHOCYTES", NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 92, 1 August 1995 (1995-08-01), pages 7739 - 7743, XP002081773, ISSN: 0027-8424 *
JANNE L.SIMONSEN ET AL: "Telomerase expression extends the proliferactive life-span and maintains the osteogenic potential of human bone marrow stromal cells", NATURE BIOTECHNOLOGY, vol. 20, June 2002 (2002-06-01), pages 592 - 596, XP002902749 *
ROSADA ET AL: "Establishment and Characterization of an Immortalized Pluripotent Telomerase-posotive Human Bone Marrow Stromal Cell Line", JOURNAL OF BONE AND MINERAL RESEARCH, SUPPL.1, vol. 15, September 2000 (2000-09-01), XP002902748 *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1447443A4 (fr) * 2001-10-31 2006-06-07 Renomedix Inst Inc Cellules mesenchymateuses immortalisees et leur utilisation
EP1447443A1 (fr) * 2001-10-31 2004-08-18 Renomedix Institute Inc. Cellules mesenchymateuses immortalisees et leur utilisation
CN100408677C (zh) * 2003-04-09 2008-08-06 中国人民解放军军事医学科学院野战输血研究所 一种bdnf基因修饰的间充质干细胞的构建方法
EP1727892A4 (fr) * 2004-03-22 2007-08-22 Osiris Therapeutics Inc Cellules souches mesenchymateuses et leurs utilisations
US9694035B2 (en) 2004-03-22 2017-07-04 Mesoblast International Sarl Mesenchymal stem cells and uses therefor
US11389484B2 (en) 2004-03-22 2022-07-19 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
EP1727892A1 (fr) * 2004-03-22 2006-12-06 Osiris Therapeutics, Inc. Cellules souches mesenchymateuses et leurs utilisations
US10960025B2 (en) 2004-03-22 2021-03-30 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10828334B1 (en) 2004-03-22 2020-11-10 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10729727B2 (en) 2004-03-22 2020-08-04 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10716814B2 (en) 2004-03-22 2020-07-21 Mesoblast International Sàrl Mesenchymal stem cells and uses therefor
US10668101B2 (en) 2004-03-22 2020-06-02 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US9943547B2 (en) 2004-03-22 2018-04-17 Mesoblast International Sàrl Mesenchymal stem cells and uses therefor
US8431533B2 (en) 2005-09-22 2013-04-30 Biocompatibles Uk Ltd. GLP-1 fusion peptides, their production and use
US8853159B2 (en) 2005-09-22 2014-10-07 Biocompatibles Uk Ltd GLP-1 Fusion Peptides
EP2174655A1 (fr) 2006-05-10 2010-04-14 Biocompatibles UK Limited Microcapsules sphériques contenant des peptides du type GLP-1, leur production et leur utilisation
US8790705B2 (en) 2006-05-10 2014-07-29 Biocompatibles Uk Ltd. Spherical microcapsules comprising GLP-1 peptides, their production and use
EP2851422A3 (fr) * 2007-05-24 2015-06-24 Apceth GmbH & Co. KG Cellules souches CD34neg pour le traitement des désordres gastrointestinaux
CN103263439B (zh) * 2007-05-24 2015-04-22 埃普塞斯有限责任两合公司 Cd34干细胞相关方法和组合物
CN103263439A (zh) * 2007-05-24 2013-08-28 埃普塞斯有限责任两合公司 Cd34干细胞相关方法和组合物
US7998472B2 (en) 2007-05-24 2011-08-16 Apceth Gmbh & Co. Kg CD34 stem cell-related methods and compositions
EP2223696A1 (fr) * 2007-05-24 2010-09-01 Apceth GmbH & Co. KG Cellules souches mésenchymales CD34- pour l'utilisation dans la thérapie génique visant la guérison de blessures
EP2163243A1 (fr) 2008-09-12 2010-03-17 Biocompatibles UK Limited Traitement de l'infarctus du myocarde (AMI) utilisant des cellules encapsulées codant et sécrétant des peptides GLP-1 ou analogues associés
EP2251028A1 (fr) 2009-05-12 2010-11-17 Biocompatibles Uk Ltd. Traitement des maladies des yeux utilisant des cellules encapsulées codant et sécrétant un facteur anti-angiogénique et/ou un facteur neuroprotecteur
WO2012034704A1 (fr) 2010-09-15 2012-03-22 Biocompatibles Uk Ltd. Traitement de maladies vasculaires à l'aide de cellules encapsulées codant pour le glp-1 et le sécrétant, ou un fragment ou un variant de celui-ci
CN109022355A (zh) * 2018-08-30 2018-12-18 丰泽康生物医药(深圳)有限公司 一种提高间充质干细胞向软骨细胞分化的培养方法
CN109022355B (zh) * 2018-08-30 2020-12-08 丰泽康生物医药(深圳)有限公司 一种提高间充质干细胞向软骨细胞分化的培养方法

Also Published As

Publication number Publication date
EP1430115A1 (fr) 2004-06-23

Similar Documents

Publication Publication Date Title
US20030049236A1 (en) Immortalized stem cells
Mosca et al. Mesenchymal stem cells as vehicles for gene delivery.
US5460959A (en) Transduced fibroblasts
US5591625A (en) Transduced mesenchymal stem cells
US5529774A (en) In vivo transfer of the HSV-TK gene implanted retroviral producer cells
Naffakh et al. Sustained delivery of erythropoietin in mice by genetically modified skin fibroblasts.
JP3953399B2 (ja) 不死化骨髄間葉系幹細胞
JPH07503121A (ja) 内皮細胞の遺伝的変性
Levy et al. Optimised retroviral infection of human epidermal keratinocytes: long-term expression of transduced integrin gene following grafting on to SCID mice
CA2383253A1 (fr) Immortalisation conditionnelle de cellules
WO2003010305A1 (fr) Cellules souches immortalisées
EP2213730B1 (fr) Adipocytes primaires cultivés pour thérapie génique
CN110760480A (zh) 一种抗肿瘤nk细胞及其制备方法
JP4860612B2 (ja) 脂肪細胞あるいは前駆脂肪細胞への遺伝子導入方法
CN109072220A (zh) 由成纤维细胞直接制造心脏祖细胞和心肌细胞的方法
JP2009201518A (ja) Lcat欠損症の遺伝子治療用細胞並びにこれを産生するのに用いられる複製欠損性レトロウイルスベクター及びプラスミド
CN109402061B (zh) 一种可以延缓细胞衰老及抵抗恶性转化的人间充质干细胞及其制备方法和应用
Miller et al. Transfer of genes into human somatic cells using retrovirus vectors
DE10019075B4 (de) Verwendung von CD34 oder einem davon abgeleiteten Polypeptid als Zell-Oberflächen- bzw. Gentransfer-Marker
WO2001048150A1 (fr) Cellules pouvant se differencier en cellules du muscle cardiaque
WO2020191115A1 (fr) Procédé d'amélioration de la survie in vivo de cellules souches mésenchymateuses
JP2018201499A (ja) 安全性と抗炎症作用を高めた間葉系幹細胞
AU2002258248B8 (en) Gene transfer into primate embryonic stem cells using VSV-G pseudotyped simian immunodeficiency virus vector
Mosca Joseph D. Mosca, Ph. D.", J Kyle Hendricks, MS", Diana Buyaner, MS", Janice Davis-Sproul, BS", Lei-Chieh Chuang, MS", Manas K. Majumdar, Ph. D.", Rajesh Chopra, Ph. D.", Mark A. Thiede, Ph. D.", Uwe Junker, MD", Richard J. Rigg, Ph. D.", Sean P.
Zhang et al. Mesenchymal stem cells

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ 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)
WWE Wipo information: entry into national phase

Ref document number: 2002764564

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 2002764564

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2002764564

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP