WO2007097494A1 - De-differentiation of astrocytes into neural stem cell using bmi-1 - Google Patents

De-differentiation of astrocytes into neural stem cell using bmi-1 Download PDF

Info

Publication number
WO2007097494A1
WO2007097494A1 PCT/KR2006/001350 KR2006001350W WO2007097494A1 WO 2007097494 A1 WO2007097494 A1 WO 2007097494A1 KR 2006001350 W KR2006001350 W KR 2006001350W WO 2007097494 A1 WO2007097494 A1 WO 2007097494A1
Authority
WO
WIPO (PCT)
Prior art keywords
bmi
neural stem
astrocytes
stem cells
cells
Prior art date
Application number
PCT/KR2006/001350
Other languages
French (fr)
Inventor
Seungkwon You
Jai Hee Moon
Byung Sun Yoon
Ki Dong Kim
Gyuman Park
Eun Kyung Jun
Bona Kim
Seung Jun Yoo
Sung Sik Kwak
Isaac Maeng
Original Assignee
Imgen Co., Ltd.
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 Imgen Co., Ltd. filed Critical Imgen Co., Ltd.
Priority to EP06732855A priority Critical patent/EP1987148A4/en
Priority to JP2008557196A priority patent/JP2009528050A/en
Publication of WO2007097494A1 publication Critical patent/WO2007097494A1/en

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
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • 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
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/08Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from cells of the nervous system

Definitions

  • Neural stem cells are a subtype of progenitor cells in the nervous system that has the ability to differentiate into astrocytes, oligodendrocytes, and neurons. Originating from the Central Nervous System (CNS) and the Peripheral Nervous System (PNS) , neural stem cells form multicellular neurospheres, which differentiate into glial lineage and neural lineage cells under respective sets of conditions (Sally Temple et al. 2001). These neural stem cells are used in the treatment of incurable diseases, and are being studied as a potential method for cell treatment. Extensive research has been conducted on neural stem cells because they are adult stem cells that entail few ethical problems . Active research on de- differentiation, which has been conducted recently, is increasing the importance of adult stem cells.
  • NN Bmi-l one transcription factor which is known to regulate properties of NSCs.
  • the selected factor identified as NN Bmi-l
  • NN Bmi-l is one of the proteins accounting for the regulation of histone modification and cell cycle regulators (Jan W. et al., 1999).
  • a cdkn2a/INK4A locus is reported as a Bmi-1 target.
  • Brt ⁇ i-1 is known as being a transcriptional inhibitor of the target gene.
  • the Bmi-1 gene has been reported to be expressed in neural stem cells and play a role in the self-renewal of neural stem cells (Anna V. M. et al., 2003, 2005, In-Kyoung Park et al., 2004).
  • An object of the invention is to provide a composition for inducing the de-differentiation of astrocytes into neural stem cells, comprising a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
  • Another object of the invention is to provide a method of inducing the de-differentiation of astrocytes into neural stem cells, comprising the treatment of astrocytes with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
  • a further object of the invention is to provide neural stem cells produced using the method.
  • Still a further object of the invention is to provide a method of differentiating the de-differentiated neural stem cells into astrocytes, oligodendrocytes and neurons.
  • FIG. 1 shows the overexpression of the Bmi-1 targets pl6 Ink4a and pl9 Arf f analyzed using immunocytochemistry
  • FIG. 2 shows the induction of de-differentiation, in which sphere formation (A) and direct sphere formation (B) are indicated (direct spheres are formed when single cells are cultured under conditions suitable for neural stem cells and spheres are formed when single cells are cultured under conditions suitable for neural stem cells only after being attached under conditions suitable for the culture of astrocytes, and stabilized for 12 hours) ;
  • FIG. 3 shows the expression of (a) neural stem cell markers as analyzed through immunocytochemistry, in which neural stem cells (A to C) and neural stem cell-like cells (D to F) are differentiated;
  • FIG. 4 shows the expression of neural stem cell-specific markers as measured through RT-PCR (Ast: astrocytes, NSC: neural stem cell, Ast-Bmi-1: astrocyte+Bmi-1, NSCLC: neural stem cell-like cell) ; and
  • FIG. 5 shows in vitro differentiation of neural stem cells (A-C) and neural stem cell-like cells (D-G) into astrocytes, oligodendrocytes and neurons.
  • the present invention relates to a composition capable of inducing the de-differentiation of astrocytes into neural stem cells, which comprises a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
  • neural stem cell-like cells is intended to indicate multipotent stem cells, dedifferentiated from somatic cells capable of differentiating into astrocytes, oligodendrocytes, and neurons.
  • neural stem cell-like cells are also described as neural stem cells.
  • Brai-1 is provided in the form of a protein or a nucleic acid that codes for the Bmi- 1 protein. As long as it is originated from mammals such as humans, horses, sheep, pigs, goats, camels, antelopes, and dogs, any Bmi-1 may be used in the composition of the present invention.
  • the Bmi-1 protein of the present invention, used for de-differentiation into neural cells may be a wild-type or a variant thereof.
  • Bmi-1 protein variant is intended to refer to Bmi-1 proteins, occurring naturally or artificially, which are different in amino acid sequence from the wild- type due to the deletion, insertion, non-conservative substitution or conservative substitution of amino acids, or combinations thereof.
  • the variant is a functional equivalent which has the same biological activity as the native protein although its physical and/or chemical properties are modified.
  • the variant is increased in structural stability to physical and chemical environments or in physiological activity.
  • the Bmi-1 is in the form of a nucleic acid material comprising a nucleotide sequence coding for the Bmi-1 protein.
  • the nucleotide sequence that encodes a Bmi-1 protein is a wild-type or a variant thereof which, naturally occurring or artificially synthesized, is different in amino acid sequence from the wild-type due to the deletion, insertion, non-conservative substitution or conservative substitution of bases, or combinations thereof.
  • the nucleotide sequence that encodes a Bmi-1 protein can be either a single or a double strand consisting of a DNA molecule (genome, cDNA) or an RNA molecule.
  • the nucleic acid material containing a nucleotide sequence encoding a Bmi-1 protein is a vector that allows a Bmi-1 protein to be expressed.
  • vector is intended to refer to a DNA construct containing a DNA sequence which is operably linked to a control sequence capable of effecting the expression of the DNA in a suitable host cell.
  • operably linked is intended to refer to a functional linkage between a nucleic acid expression control sequence and a second nucleic acid sequence coding for a target protein in such a manner as to enable general functionality.
  • the operable linkage to a recombinant vector may be prepared using a genetic recombinant technique that is well known in the art, and site-specific DNA cleavage and ligation may be carried out using enzymes that are generally known in the art.
  • a vector suitable for use in the present invention includes a signal sequence or a leader sequence for membrane targeting or secretion as well as expression regulatory elements, such as a promoter, an operator, an initiation codon, a stop codon, a polyadenylation signal and an enhancer, and can be constructed in various forms depending on the purpose thereof.
  • the promoter of the vector may be constitutive or inducible.
  • expression vectors include a selectable marker that allows the selection of host cells containing the vector, and replicable expression vectors include a replication origin.
  • the vector may be self-replicable, or may be integrated into the DNA of a host cell.
  • the vector useful in the present invention may be a plasmid vector, a cosmid vector, or a viral vector, with preference for a viral vector.
  • the viral vector includes vectors originated from retroviruses such as HIV
  • Necrosis Virus RSV (Rous Sarcoma Virus), MMTV (Mouse Mammary Tumor Virus), etc.
  • Adeno-associated viruses adeno-associated viruses
  • Herpes Simplex virus Herpes Simplex virus, but are not limited thereto.
  • a pBabe puro vector derived from a Moloney leukemia virus-based virus vector, carrying a selection marker for puromycin, was used.
  • the nucleic acid material containing the nucleotide sequence coding for the Bmi-1 protein can be introduced into cells in the form of a vector as naked DNA (Wolff et al. Science, 247:1465-8, 1990: Wolff et al. J. Cell Sci. 103:1249-59, 1992), or with the aid of a liposome or a cationic polymer.
  • a liposome is a phospholipid membrane made of cationic phospholipids such as DOTMA and DOTAP.
  • a cationic liposome when mixed with a negatively charged nucleotide at a certain ratio, is formed into a nucleic acid-liposome complex.
  • the nucleic acid material containing a nucleotide sequence that encodes the Bmi-1 protein may be a virus which expresses the Bmi-1 protein therein.
  • virus is intended to refer to a Bmi-1-expressing virus which is prepared by transforming or transfecting a packaging cell with a viral vector carrying a nucleotide sequence coding for the Bmi-1 protein.
  • viruses useful in the preparation of the Bmi-1-expressing viruses according to the present invention include retroviruses, adenoviruses, adeno-associated viruses, and the Herpes Simplex virus, but are not limited thereto. Preferable are retroviruses.
  • a Bmi-1-expressing virus was prepared by transforming a recombinant pBabe puro vector carrying a nucleotide sequence coding for the Bmi-1 protein (pBabe puro Bmi-1) into PT67 packaging cells.
  • the present invention pertains to a method of dedifferentiating astrocytes into neural stem cells, comprising treatment with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence encoding the Bmi- 1 protein.
  • the method comprises the steps of (i) culturing astrocytes in a medium; (ii) treating the culture with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for the Bmi-1 protein; and (iii) inducing the de-differentiation of astrocytes into neural stem cells.
  • Any conventional culture medium for neural stem cells may be used as a culture medium for astrocytes in step (i) .
  • a culture medium contains a carbon source, a nitrogen source, and trace element ingredients.
  • the culture medium may include antibiotics, such as penicillin, streptomycin, and gentamicin.
  • antibiotics such as penicillin, streptomycin, and gentamicin.
  • Preferred is a culture medium containing bFGF.
  • the Bmi-1 protein or the nucleic acid material containing the nucleotide sequence coding for the Bmi-1 protein with which the cells are treated in step (ii) is as mentioned above.
  • the present invention pertains to neural stem cells prepared in accordance with the aforementioned method.
  • the neural stem cells prepared through the de-differentiation according to the present invention, express the neural stem cell-specific markers, Nestin, CD133, and Sox2 at the same level, and have the same ability to differentiate as general neural stem cells.
  • the neural stem cells prepared through de- differentiation according to the present invention feature self-renewal, as well.
  • the present invention pertains to a method of differentiating the neural stem cells, de-differentiated according to the aforementioned method, into astrocytes, oligodendrocytes, and neurons.
  • neural stem cells de-differentiated using the composition and method of the present invention
  • differentiation into astrocytes, neurons, and oligodendrocytes can be monitored by detecting the expression of markers specific for respective cells.
  • Mouse astrocytes were isolated from the CNS at E13.5 and cultured under suitable conditions. They were treated with trypsin (Gibco 0.05%) before incubation in modified Eagle's medium (DMEM (high glucose, HyClone) ) supplemented with 10% FBS (HyClone) , 1% penicillin/streptomycin, and 1% L-glutamine (Cambrex) . From the next day, the medium was replaced with a fresh one. Cells between passage one and three were used.
  • DMEM modified Eagle's medium
  • FBS HexClone
  • penicillin/streptomycin 1% penicillin/streptomycin
  • L-glutamine Limbrex
  • Neural stem cells were separated from mice (E13.5) and cultured as a control in Dulbecco's modified Eagle's medium/F12 (DMEM/F12, Gibco), supplemented with B27 serum-replacement (Gibco) , human recombinant basic FGF and human recombinant EGF (R&D) , containing insulin (Sigma), apo-transferrin (Sigma) , selenium (Sigma) , progesterone (Sigma) and penicillin/streptomycin (Cambrex) .
  • pBabe puro Bmi-1 prepared by inserting human Bmi-1 (NCBI accession No.: L13689 NM 024865) into a pBabe puro vector
  • pBabe puro were transfected into a PT67 packaging cell line (Clontech) using Lipofectamine
  • the primers used in the RT-PCR were adapted to amplify the mouse markers GAPDH, Nestin, and Sox2, and their base sequences are shown as follows.
  • De-differentiation was induced under the same culture condition used for neural stem cells.
  • Cell culture was conducted using two different methods. Cells were plated at a density of IxIO 5 cells/well in ⁇ -well plates and incubated for 12 hrs, after which the medium was replaced with a fresh Dulbecco' s modified Eagle's medium/Fl2 (N2) (DMEM/F12, Gibco) , supplemented with B27 serum-replacement
  • neurospheres were incubated overnight at 4 0 C in 20% sucrose with shaking. Then, the neurospheres were subjected to cryopreservation using an OCT compound in an
  • anti-GFAP Dako
  • anti-SlOO ⁇ Zymed
  • anti- ⁇ -tubulin III Covance
  • anti-Map2a Sigma
  • anti-TH Sigma
  • anti-04 R&D
  • anti-CNPase Cemicon
  • DMEM HyClone, high glucose
  • FBS HyClone
  • CNTF recombinant rat ciliary neurotrophic factor
  • Differentiation into neurons was induced by culturing the cells in a medium containing N2 and B27 serum- replacement, supplemented with human recombinant FGF, for 4 days and then in an FGF-free medium for 8 days.
  • cells were cultured for 7 ⁇ 14 days in the presence of 1 ⁇ 10 ⁇ M of RA (retinoic acid, Sigma) as a differentiation inducer.
  • RA retinoic acid, Sigma
  • cells were cultured for 7 ⁇ 14 days in the co-presence of 1 ⁇ 10 Mm of VPA (valproic acid, Sigma) and 1 ⁇ 10 ⁇ M of RA (retinoic acid, Sigma) to induce differentiation into neurons .
  • oligodendrocytes Differentiation into oligodendrocytes was induced by incubating in an N2 medium supplemented with B27 serum replacement in the presence of PDGF-AA (platelet derived growth factor-AA, R&D), T3 (3, 3, 5-triiodo-L-thyronine, Sigma) , human recombinant basic FGF and EGF (R&D) .
  • PDGF-AA platelet derived growth factor-AA, R&D
  • T3 3, 5-triiodo-L-thyronine, Sigma
  • R&D human recombinant basic FGF and EGF
  • the cells While being cultured under the aforementioned differentiation conditions, the cells were monitored for morphology, and immunocytochemistry using antibodies specific to respective differentiation markers was conducted to examine whether the differentiation proceeded accurately.
  • EXAMPLE 2 Results The Brai-1 gene was overexpressed using a retroviral vector transduction system in mouse astrocytes, which had been differentiated. The overexpression of the Bmi-1 gene was verified through Western blotting analysis while the Bmi-1 target genes pl ⁇ Ink4a and pl9 Art were not expressed
  • FIG. 1 (FIG. 1) .
  • de-differentiation was induced using conditions for culturing neural stem cells.
  • the astrocytes were seeded at a density of IxIO 5 cells/well in ⁇ -well plates, and 12 hrs later, the cells were incubated under conditions suitable for neural stem cells. Within 3 - 4 days of incubation, the cells were observed to have neural stem cell-like morphology. When the cells were plated at a density of 3 ⁇ lO 5 cells/plate in 60-mrn bacterial plates and cultured under the same conditions as neural stem cells, they were also observed to have the morphology of neural stem cells. On the contrary, when the cells transfected with the control vector pBabe puro EGFP were cultured, no neurospheres were formed (FIG.
  • neural stem cell-like cells In order to examine whether the neural stem cell-like cells could differentiate like neural stem cells, differentiation into astrocytes, oligodendrocytes and neurons was induced in the same manner as described above.
  • the neural stem cell-like cells were found to differentiate into the three types of cells like neural stem cells as analyzed with antibodies against the respective markers specific therefor. Identification was conducted with the expression of GFAP and SlOO for differentiation into astrocytes, with the expression of ⁇ -tubulin III (Tujl) and Map2a for differentiation into neurons and with the expression of 04 and CNPOase for differentiation into oligodendrocytes (FIG. 5) .
  • Bmi-1 is useful in inducing the de-differentiation of astrocytes into neural stem cells, and the de-differentiated neural stem cells can be used for the treatment of various diseases.
  • Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF Genes & Development 1999; 13:2678-2690 6.
  • Kishi Y Takahashi J, Koyanagi M, Morizane A, Okamoto Y, Horiguchi S, Tashiro K, Honjo T, Fujii S,

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Materials For Medical Uses (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Disclosed are a composition and a method for inducing the de-differentiation of astrocytes into neural stem cells using Bmi-1. The de-differentiated neural stem cells have the ability to differentiate into astrocytes, neurons, and oligodendrocytes.

Description

DE-DIFFERENTIATION OF ASTROCYTES INTO NEURAL STEM CELL
USING BMI-I
Technical Field
Neural stem cells (NSCs) are a subtype of progenitor cells in the nervous system that has the ability to differentiate into astrocytes, oligodendrocytes, and neurons. Originating from the Central Nervous System (CNS) and the Peripheral Nervous System (PNS) , neural stem cells form multicellular neurospheres, which differentiate into glial lineage and neural lineage cells under respective sets of conditions (Sally Temple et al. 2001). These neural stem cells are used in the treatment of incurable diseases, and are being studied as a potential method for cell treatment. Extensive research has been conducted on neural stem cells because they are adult stem cells that entail few ethical problems . Active research on de- differentiation, which has been conducted recently, is increasing the importance of adult stem cells. Adult stem cells are easier to obtain than embryonic stem cells, but there are still many difficulties in the practical application thereof. In addition, immune rejection response can be a problem when using adult stem cells from another person. Therefore, the induction of de-differentiation using a patient's own cells could solve current problems. For this purpose, it is necessary to induce de- differentiation of cells that are already differentiated. Currently, extensive research is underway to induce de- differentiation using methods such as cell fusion and nuclear transfer. In another group using different methods, Alexis J. reported that cells isolated from the skin have the characteristics of neural stem cells when cultured under neural stem cell cultivation conditions (Lancet 2004) . In addition, Toru K. succeeded in de-differentiating oligodendrocyte precursors into neural stem cells (Genes & Development 2004) . This group has been publishing papers on this issue since 2000, and reported in a 2004 paper that gene expression at each stage is relevant to chromatin remodeling and histone modification.
Background Art
In the present invention, a solution to the problems of formerly introduced methods is sought and a novel proper usage method is established. In this invention, one transcription factor which is known to regulate properties of NSCs, is selected and overexpressed to study differentiation into neural stem cells. The selected factor, identified as NNBmi-l", is one of the proteins accounting for the regulation of histone modification and cell cycle regulators (Jan W. et al., 1999). A cdkn2a/INK4A locus is reported as a Bmi-1 target. Brtιi-1 is known as being a transcriptional inhibitor of the target gene. Recently, the Bmi-1 gene has been reported to be expressed in neural stem cells and play a role in the self-renewal of neural stem cells (Anna V. M. et al., 2003, 2005, In-Kyoung Park et al., 2004).
Leading to the present invention, intensive and thorough research, conducted by the present inventors with this background, resulted in the finding that the overexpression of Bmi-1 therein induces already differentiated astrocytes to de-differentiate into neural stem cell-like cells capable of self-renewal and differentiation into astrocytes, neurons, and oligodendrocytes .
Disclosure of the Invention
An object of the invention is to provide a composition for inducing the de-differentiation of astrocytes into neural stem cells, comprising a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
Another object of the invention is to provide a method of inducing the de-differentiation of astrocytes into neural stem cells, comprising the treatment of astrocytes with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
A further object of the invention is to provide neural stem cells produced using the method.
Still a further object of the invention is to provide a method of differentiating the de-differentiated neural stem cells into astrocytes, oligodendrocytes and neurons.
Brief Description of the Drawings
The above and other objects, features and other advantages of the present invention will be more clearly understood from the following detailed description taken in conjunction with the accompanying drawings, in which:
FIG. 1 shows the overexpression of the Bmi-1 targets pl6Ink4a and pl9Arf f analyzed using immunocytochemistry;
FIG. 2 shows the induction of de-differentiation, in which sphere formation (A) and direct sphere formation (B) are indicated (direct spheres are formed when single cells are cultured under conditions suitable for neural stem cells and spheres are formed when single cells are cultured under conditions suitable for neural stem cells only after being attached under conditions suitable for the culture of astrocytes, and stabilized for 12 hours) ;
FIG. 3 shows the expression of (a) neural stem cell markers as analyzed through immunocytochemistry, in which neural stem cells (A to C) and neural stem cell-like cells (D to F) are differentiated;
FIG. 4 shows the expression of neural stem cell- specific markers as measured through RT-PCR (Ast: astrocytes, NSC: neural stem cell, Ast-Bmi-1: astrocyte+Bmi-1, NSCLC: neural stem cell-like cell) ; and
FIG. 5 shows in vitro differentiation of neural stem cells (A-C) and neural stem cell-like cells (D-G) into astrocytes, oligodendrocytes and neurons.
Best Mode for Carrying Out the Invention
In accordance with an aspect thereof, the present invention relates to a composition capable of inducing the de-differentiation of astrocytes into neural stem cells, which comprises a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
As used herein, the term "neural stem cell-like cells" is intended to indicate multipotent stem cells, dedifferentiated from somatic cells capable of differentiating into astrocytes, oligodendrocytes, and neurons. In the present invention, neural stem cell-like cells are also described as neural stem cells.
It is newly disclosed in the present invention that when Bmi-1 is overexpressed, astrocytes, although already differentiated, can de-differentiate into multipotent neural stem cell-like cells.
In the present invention, Brai-1 is provided in the form of a protein or a nucleic acid that codes for the Bmi- 1 protein. As long as it is originated from mammals such as humans, horses, sheep, pigs, goats, camels, antelopes, and dogs, any Bmi-1 may be used in the composition of the present invention. In addition, the Bmi-1 protein of the present invention, used for de-differentiation into neural cells, may be a wild-type or a variant thereof.
The term "Bmi-1 protein variant" is intended to refer to Bmi-1 proteins, occurring naturally or artificially, which are different in amino acid sequence from the wild- type due to the deletion, insertion, non-conservative substitution or conservative substitution of amino acids, or combinations thereof. The variant is a functional equivalent which has the same biological activity as the native protein although its physical and/or chemical properties are modified. Preferably, the variant is increased in structural stability to physical and chemical environments or in physiological activity.
In a preferable embodiment of the present invention, the Bmi-1 is in the form of a nucleic acid material comprising a nucleotide sequence coding for the Bmi-1 protein.
The nucleotide sequence that encodes a Bmi-1 protein is a wild-type or a variant thereof which, naturally occurring or artificially synthesized, is different in amino acid sequence from the wild-type due to the deletion, insertion, non-conservative substitution or conservative substitution of bases, or combinations thereof.
The nucleotide sequence that encodes a Bmi-1 protein can be either a single or a double strand consisting of a DNA molecule (genome, cDNA) or an RNA molecule.
In a preferred embodiment of the present invention, the nucleic acid material containing a nucleotide sequence encoding a Bmi-1 protein is a vector that allows a Bmi-1 protein to be expressed.
The term "vector", as used herein, is intended to refer to a DNA construct containing a DNA sequence which is operably linked to a control sequence capable of effecting the expression of the DNA in a suitable host cell.
The term "operably linked", as used herein, is intended to refer to a functional linkage between a nucleic acid expression control sequence and a second nucleic acid sequence coding for a target protein in such a manner as to enable general functionality. The operable linkage to a recombinant vector may be prepared using a genetic recombinant technique that is well known in the art, and site-specific DNA cleavage and ligation may be carried out using enzymes that are generally known in the art.
A vector suitable for use in the present invention includes a signal sequence or a leader sequence for membrane targeting or secretion as well as expression regulatory elements, such as a promoter, an operator, an initiation codon, a stop codon, a polyadenylation signal and an enhancer, and can be constructed in various forms depending on the purpose thereof. The promoter of the vector may be constitutive or inducible. In addition, expression vectors include a selectable marker that allows the selection of host cells containing the vector, and replicable expression vectors include a replication origin. The vector may be self-replicable, or may be integrated into the DNA of a host cell.
The vector useful in the present invention may be a plasmid vector, a cosmid vector, or a viral vector, with preference for a viral vector. Examples of the viral vector includes vectors originated from retroviruses such as HIV
(Human Immunodeficiency Virus) , MLV (Murine Leukemia
Virus) , ASLV (Avian Sarcoma/Leukosis Virus) , SNV (Spleen
Necrosis Virus), RSV (Rous Sarcoma Virus), MMTV (Mouse Mammary Tumor Virus), etc., Adeno-associated viruses, and Herpes Simplex virus, but are not limited thereto. In one practical example of the present invention, a pBabe puro vector, derived from a Moloney leukemia virus-based virus vector, carrying a selection marker for puromycin, was used.
The nucleic acid material containing the nucleotide sequence coding for the Bmi-1 protein can be introduced into cells in the form of a vector as naked DNA (Wolff et al. Science, 247:1465-8, 1990: Wolff et al. J. Cell Sci. 103:1249-59, 1992), or with the aid of a liposome or a cationic polymer. For use in gene transfection, a liposome is a phospholipid membrane made of cationic phospholipids such as DOTMA and DOTAP. A cationic liposome, when mixed with a negatively charged nucleotide at a certain ratio, is formed into a nucleic acid-liposome complex. In another preferred embodiment of the present invention, the nucleic acid material containing a nucleotide sequence that encodes the Bmi-1 protein may be a virus which expresses the Bmi-1 protein therein.
The term "virus", as used herein, is intended to refer to a Bmi-1-expressing virus which is prepared by transforming or transfecting a packaging cell with a viral vector carrying a nucleotide sequence coding for the Bmi-1 protein.
Examples of viruses ,useful in the preparation of the Bmi-1-expressing viruses according to the present invention include retroviruses, adenoviruses, adeno-associated viruses, and the Herpes Simplex virus, but are not limited thereto. Preferable are retroviruses. In the following examples, a Bmi-1-expressing virus was prepared by transforming a recombinant pBabe puro vector carrying a nucleotide sequence coding for the Bmi-1 protein (pBabe puro Bmi-1) into PT67 packaging cells.
In accordance with another aspect thereof, the present invention pertains to a method of dedifferentiating astrocytes into neural stem cells, comprising treatment with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence encoding the Bmi- 1 protein.
In greater detail, the method comprises the steps of (i) culturing astrocytes in a medium; (ii) treating the culture with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for the Bmi-1 protein; and (iii) inducing the de-differentiation of astrocytes into neural stem cells.
Any conventional culture medium for neural stem cells may be used as a culture medium for astrocytes in step (i) .
Generally, a culture medium contains a carbon source, a nitrogen source, and trace element ingredients. In addition, the culture medium may include antibiotics, such as penicillin, streptomycin, and gentamicin. Preferred is a culture medium containing bFGF.
The Bmi-1 protein or the nucleic acid material containing the nucleotide sequence coding for the Bmi-1 protein with which the cells are treated in step (ii) is as mentioned above.
In accordance with a further aspect thereof, the present invention pertains to neural stem cells prepared in accordance with the aforementioned method.
It has been confirmed that the neural stem cells, prepared through the de-differentiation according to the present invention, express the neural stem cell-specific markers, Nestin, CD133, and Sox2 at the same level, and have the same ability to differentiate as general neural stem cells. The neural stem cells prepared through de- differentiation according to the present invention feature self-renewal, as well.
In accordance with still a further aspect thereof, the present invention pertains to a method of differentiating the neural stem cells, de-differentiated according to the aforementioned method, into astrocytes, oligodendrocytes, and neurons.
When the neural stem cells, de-differentiated using the composition and method of the present invention, are placed under respective differentiation conditions, differentiation into astrocytes, neurons, and oligodendrocytes can be monitored by detecting the expression of markers specific for respective cells.
A better understanding of the present invention may be obtained through the following examples, which are set forth to illustrate, but are not to be construed as limiting the present invention.
EXAMPLE 1: Experiment Method
1. Cultivation of mouse astrocytes and mouse neural stem cell
Mouse astrocytes were isolated from the CNS at E13.5 and cultured under suitable conditions. They were treated with trypsin (Gibco 0.05%) before incubation in modified Eagle's medium (DMEM (high glucose, HyClone) ) supplemented with 10% FBS (HyClone) , 1% penicillin/streptomycin, and 1% L-glutamine (Cambrex) . From the next day, the medium was replaced with a fresh one. Cells between passage one and three were used. Neural stem cells were separated from mice (E13.5) and cultured as a control in Dulbecco's modified Eagle's medium/F12 (DMEM/F12, Gibco), supplemented with B27 serum-replacement (Gibco) , human recombinant basic FGF and human recombinant EGF (R&D) , containing insulin (Sigma), apo-transferrin (Sigma) , selenium (Sigma) , progesterone (Sigma) and penicillin/streptomycin (Cambrex) .
2. Retroviral-mediated infection
pBabe puro Bmi-1 (prepared by inserting human Bmi-1 (NCBI accession No.: L13689 NM 024865) into a pBabe puro vector) and pBabe puro were transfected into a PT67 packaging cell line (Clontech) using Lipofectamine
(Invitrogen) and selected in the presence of puromycin (3 μg/ml) (BD science) . After the transformed cell line had grown to 90% or higher confluency, the supernatant was passed through a filter (0.45 μm) (Millipore) to remove cell debris, after which the supernatant was twice allowed to infect the astrocytes, which were separated at intervals of 10 hrs using polybrene (Sigma) . Selection was subsequently conducted in the presence of puromycin (0.5 μg/ml) .
3. Western blot analysis and semi-quantitative PCR
Total cell extracts were isolated from the cells using RIPA buffer, followed by Bradford assay (Bio-rad) to determine the protein concentrations thereof. 50 μg of each cell extract was subjected to 10% or 4~12% precasted SDS- PAGE (Invitrogen) . The proteins thus separated were transferred onto a PVDF membrane (Millipore) and blocked with 3~5% skimmed milk TBST. Incubation with anti-Bmi-1 (Upstate) anti-plβINK4A (Santacruz), anti-pl^ (Novous), and α-tubulin (sigma) was done at 4°C overnight with shaking, followed by reaction with HRP conjugated anti-mouse IgG, anti-rabbit IgG (Zymed) at RT. A Supersignal West Pico kit
(Pierce) was used for detection. Total RNA was isolated from each cell line using Trizol (Invitrogen) . cDNA was synthesized from 500 ng of the total RNA in the presence of oligo d(T) 12-18-mer (Invitrogen) using Superscriptase II reverse transcriptase (Invitrogen) . RT-PCR was performed with 1 μl of the cDNA in the presence of 10 pmol of each of suitable primers using a PCR premix (IU Tag DNA polymerase, 250 μM dNTPs, 10 mM Tris-HCl, 40 mM KCl and 1.5 mM MgCl2 Bioneer, Korea). The primers used in the RT-PCR were adapted to amplify the mouse markers GAPDH, Nestin, and Sox2, and their base sequences are shown as follows.
Primers for mouse GAPDH
Forward: 5'-GATGACATCAAGAAGGTGGTGAAG-SMSEQ ID NO.:1) Reverse: 5'-GTTGCTGTAGCCGTATTCATTGTC-SMSEQ ID NO.^) Primers for mouse nestin
Forward: 5'-GGCATCCCTGAATTACCCAA-S' (SEQ ID NO.: 3) Reverse: 5'-AGCTCATGGGCATCTGTCAA-S' (SEQ ID NO.: 4) Primers for mouse sox2 Forward: 5'-AGTGGTACGTTAGGCGCTTC-S' (SEQ ID NO.: 5)
Reverse: 5'-TGCCTTAAACAAGACCACGA-S' (SEQ ID NO. 6)
4. Induction of de-differentiation
De-differentiation was induced under the same culture condition used for neural stem cells. Cell culture was conducted using two different methods. Cells were plated at a density of IxIO5 cells/well in β-well plates and incubated for 12 hrs, after which the medium was replaced with a fresh Dulbecco' s modified Eagle's medium/Fl2 (N2) (DMEM/F12, Gibco) , supplemented with B27 serum-replacement
(Gibco) , human recombinant basic FGF and human recombinant EGF (R&D) , containing insulin (Sigma) , apo-transferrin
(Sigma) , selenium (Sigma) , progesterone (Sigma) and penicillin/streptomycin (Cambrex) . Cells were treated with bFGF every day, and the medium was replaced with a fresh onemedium every other day. Alternatively, cells which had been cultured under proper conditions were trypsinized and seeded at a density of 3χlO5 cells in a 60-mm bacterial culture plate. Dulbecco' s modified Eagle's medium/F12 (N2) (DMEM/F12, Gibco) , supplemented with B27 serum-replacement
(Gibco) , human recombinant basic FGF and human recombinant EGF (R&D) , containing insulin (Sigma) , apo-transferrin
(Sigma) , selenium (Sigma) , progesterone (Sigma) and penicillin/streptomycin (Cambrex) , was used as a culture medium.
5. Determination of overexpression and respective markers through immunocytochemistry
After fixation with 4% paraformaldehyde (EMS) at 40C for one hour, neurospheres were incubated overnight at 40C in 20% sucrose with shaking. Then, the neurospheres were subjected to cryopreservation using an OCT compound in an
8-well chamber slide (Nunc) and sectioned 8~10 μm thick before staining. After blocking with PBS containing 10% normal goat serum (Jackson ImmunoResearch) +0.1% BSA
(Sigma) +0.3% Triton X-100 (Sigma), the sections were incubated with anti-nestin (Chemicon) , anti-CD133 (MACS) and anti-Sox2 (Sigma) at 4°C overnight and then with anti- mouse-cy3 (Jackson ImmunoResearch) , anti-rabbit-FITC (Molecular probe) , and anti-goat-cy3 (Zymed) at RT, finally followed by nuclear staining with DAPI (Sigma) . A Zeiss confocal lens (Carl Zeiss) was used for examination after staining. Differentiated cells were stained in the same manner as described above. In this regard, anti-GFAP (Dako), anti-SlOOβ (Zymed), anti-β-tubulin III (Covance) , anti-Map2a (Sigma), anti-TH (Sigma), anti-04 (R&D) , and anti-CNPase (Chemicon) were used as antibodies.
6. In vitro differentiation
After being coated with PLO (poly-L-ornithine) (Sigma) and laminin or fibronectin (Sigma) , cells were cultured under the differentiation conditions given below.
For differentiation into astrocytes, cell culture was conducted for 5-7 days in DMEM (HyClone, high glucose) supplemented with 10% FBS (HyClone) in the presence of human recombinant bFGF and EGF (R&D) or of CNTF (recombinant rat ciliary neurotrophic factor) (Upstate) .
Differentiation into neurons was induced by culturing the cells in a medium containing N2 and B27 serum- replacement, supplemented with human recombinant FGF, for 4 days and then in an FGF-free medium for 8 days. Alternatively, cells were cultured for 7 ~ 14 days in the presence of 1 ~ 10 μM of RA (retinoic acid, Sigma) as a differentiation inducer. As an additional alternative, cells were cultured for 7 ~ 14 days in the co-presence of 1 ~ 10 Mm of VPA (valproic acid, Sigma) and 1 ~ 10 μM of RA (retinoic acid, Sigma) to induce differentiation into neurons .
Differentiation into oligodendrocytes was induced by incubating in an N2 medium supplemented with B27 serum replacement in the presence of PDGF-AA (platelet derived growth factor-AA, R&D), T3 (3, 3, 5-triiodo-L-thyronine, Sigma) , human recombinant basic FGF and EGF (R&D) .
While being cultured under the aforementioned differentiation conditions, the cells were monitored for morphology, and immunocytochemistry using antibodies specific to respective differentiation markers was conducted to examine whether the differentiation proceeded accurately.
EXAMPLE 2: Results The Brai-1 gene was overexpressed using a retroviral vector transduction system in mouse astrocytes, which had been differentiated. The overexpression of the Bmi-1 gene was verified through Western blotting analysis while the Bmi-1 target genes plβInk4a and pl9Art were not expressed
(FIG. 1) .
Then, de-differentiation was induced using conditions for culturing neural stem cells. The astrocytes were seeded at a density of IxIO5 cells/well in β-well plates, and 12 hrs later, the cells were incubated under conditions suitable for neural stem cells. Within 3 - 4 days of incubation, the cells were observed to have neural stem cell-like morphology. When the cells were plated at a density of 3χlO5 cells/plate in 60-mrn bacterial plates and cultured under the same conditions as neural stem cells, they were also observed to have the morphology of neural stem cells. On the contrary, when the cells transfected with the control vector pBabe puro EGFP were cultured, no neurospheres were formed (FIG. 2) . On day 6, the neurospheres were observed to have the same morphology as neural stem cells when they were transferred to and cultured in new plates . In order to examine whether the cells de-differentiated with the Bmi-1 gene ensure self- renewal, a subsphere formation assay was conducted. As a result, spheres were formed in single cells one week later (data not shown) .
In order to find out whether these cells had the same characteristics as neural stem cells, immunocytochemistry was performed using respective markers. In this regard, the markers nestin, CD133 and Sox2, which are most abundantly expressed in neural stem cells, were also observed in the neural stem cell-like cells after staining. Also, the cryo- section of the neurospheres thus formed ensured the expression of nestin, CD133 and Sox2 (FIG. 3) . Also, nestin and sox2, both expressed in neural stem cells, were found to be expressed in the cells dedifferentiated according to the present invention, as analyzed through RT-PCR. Therefore, the cells dedifferentiated according to the present invention were judged to be neural stem cell-like cells (FIG. 4) .
In order to examine whether the neural stem cell-like cells could differentiate like neural stem cells, differentiation into astrocytes, oligodendrocytes and neurons was induced in the same manner as described above. The neural stem cell-like cells were found to differentiate into the three types of cells like neural stem cells as analyzed with antibodies against the respective markers specific therefor. Identification was conducted with the expression of GFAP and SlOO for differentiation into astrocytes, with the expression of β-tubulin III (Tujl) and Map2a for differentiation into neurons and with the expression of 04 and CNPOase for differentiation into oligodendrocytes (FIG. 5) .
Taken together, the data obtained through this study demonstrate that the Bmi-1 gene plays a critical role in the de-differentiation of astrocytes into neural stem cell- like cells, and that these de-differentiated neural stem cell-like cells can differentiate back into astrocytes, oligodendrocytes, and neurons.
Industrial Applicability
As described hitherto, Bmi-1 is useful in inducing the de-differentiation of astrocytes into neural stem cells, and the de-differentiated neural stem cells can be used for the treatment of various diseases.
Bibliography of Prior Art
1. Toru Kondo, martin Raff. Chromatin remodeling and histone modification in the conversion of oligodendrocyte precursors to neural stem cells Genes & Development 2004;18: 2963-2972 2. Alexis Joannides, Phil Gaughwin, Chistof Shwiening, Henry Majed, Jane Sterling, Alastair Compston, siddharthan Chandran. Effiencient generation of neural precursors from adult human skin: astrocytes promote neurogenesis from skin-derived stem cells Lancet 2004;363:172-178
3. Sally Temple. The development of neural stem cells Nature 2001; 414 : 112-117 4. Anna V. Molofsky, Ricardo Pardal, Toshihide Iwashita, In-Kyung Park, Michael F. Clarke & Sean J. Morrison. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation Nature 2003/425:962-967 5. Jacqueline J. L. Jacobs, Blanca Scheijen, Jan- Wi11em Voncken, Karin Kieboom, Anton Berns, and Maarten van Lohuizen. Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF Genes & Development 1999; 13:2678-2690 6. Si'lvia Bea, Frederic Tort, Magda Pinyol, Xavier Puig, Luis Herna'ndez, Silvia Herna'ndez, Pedro L. Ferna'ndez, Maarten van Lohuizen, Dolors Colomer, and Elias Campo. BMI-1 Gene Amplification and Overexpression in Hematological Malignancies Occur Mainly in Mantle Cell Lymphomas. 2001; 61:2409-412
7. Dusan Zencak, Merel Lingbeek, Corinne Kostic, Meriem Tekaya, Ellen Tanger, Dana Hornfeld, Muriel Jaquet, Francis L. Munier, Daniel F.Schorderet, Maarten van Lohuizen, and Yvan Arsenijevic. Bmil Loss Produces an Increase in Astroglial Cells and a Decrease in Neural Stem Cell Population and Proliferation. The Journal of Neuroscience 2005; 25: 5774-783
8. Carly Leung, Merel Lingbeek, Olga Shakhova, James Liu, Ellen Tanger, Parvin Saremaslani, Maarten van Lohuizen & Silvia Marino. Bmil is essential for cerebellar development and is overexpressed in human medulloblastomas Nature 2004; 428 : 337-341
9. Atsushi Iwama, Hideyuki Oguro, Masamitsu Negishi, Yuko Kato, Youhei Morita,Hiroko Tsukui, Hideo Ema, Takehiko Kamijo, Yuko Katoh-Fukui, Haruhiko Koseki, Maarten van Lohuizen, and Hiromitsu Nakauchi. Enhanced Self-Renewal of Hematopoietic Stem Cells Mediated by the Polycomb Gene Product Bmi-1 of transcriptional activators 2004 ; 21 : 843-851
10. Katalin Schlett and Emilia Madara'sz. Retinoic Acid Induced Neural Differentiation in a Neuroectodermal
Cell Line Immortalized by p53 Deficiency. Journal of Neuroscience Research 1997; 47 : 405-15
11. Barbara Ahlemeyer, Eveline Baumgart-Vogt . Optimized protocols for the simultaneous preparation of primary neuronal cultures of the neocortex, hippocampus and cerebellum from individual newborn (PO.5) C57B1/6J mice. Journal of Neuroscience Methods 2005
12. Tomoya Kitayama, Masanori Yoneyama, Keisuke Tamaki, and Yukio Yoneda. Regulation of Neuronal Differentiation by N-Methyl-D-Aspartate Receptors Expressed in Neural Progenitor Cells Isolated From Adult Mouse Hippocampus Journal of Neuroscience Research 2004:76:599-12
13. Yingwei Mao and Angel W. -M. Lee. A novel role for Gab2 in bFGF-mediated cell survival during retinoic acid- induced neuronal differentiation. The Journal of Cell Biology 2005; 170: 305-16
14. Mireya Marin-Husstege, Michela Muggironi, Aixiao Liu, Patrizia Casaccia-Bonnefi. Histone Deacetylase Activity Is Necessary for Oligodendrocyte Lineage Progression. The Journal of Neuroscience, 2002 ; 22 : 10333- 0345
15. Anna V. Molofsky, Shenghui He, Mohammad Bydon, Sean J. Morrison, Ricardo Pardal. Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the pl6Ink4a and pl9Arf senescence pathways. Genes & Development 2005; 19: 1432-1437
16. Sopia W. M. bruggeman, Merel E. Valk-Lingbeek, Petra P.M. van der Stoop, Jacqueline J. L., Jacobs, Karin kieboom, Ellen Tanger, Danielle Hulsman, Carly Leung, Yvan Arsenijevic, Silvia Marino and Maarten van Lohuizen. Ink4a and Arf differentiallu affect cell proliferation and neural stem cell self-renewal in Bmi-1-deficient mice. Genes & Development 2005; 19: 1438-1443
17. Kishi Y, Takahashi J, Koyanagi M, Morizane A, Okamoto Y, Horiguchi S, Tashiro K, Honjo T, Fujii S,
Hashimoto N.. Estrogen promotes differentiation and survival of dopaminergic neurons derived from neural stem cells Journal of neuroscience research 2005/79:279-286
18. Hsieh J, Nakashima K, Kuwabara T, Mejia E, Gage FH. Histone deacetylase inhibitor-mediated neuronal differentiation of multipotent adult neural progenitor cells PNAS 2004; 101 : 16659-16664
19. Tarasenko YI, Yu Y, Jordan PM, Bottenstein J, Wu P. Effect of growth factors on proliferation and phenotypic differentiation of human fetal neural stem cells Journal of neuroscience research 2004; 78 : 625-636
20. Ping Wu, Yevgeniy. Tarasenko, Yanping Gu, Li-Yen M. Huang, Richard E. Coggeshall and Yongjia Yu Region- specific generation of cholinergic neurons from fetal human neural stem cells grafted in adult rat. Nature neuroscience 2005;5:1271-1278
21. Hu X, Jin L, Feng L. Erkl/2 but not PI3K pathway is required for neurotrophin 3-induced oligodendrocyte differentiation of post-natal neural stem cells. Journal of Neurochemistry 2004; 90 : 1339-1347 22. In-Kyung Park, Sean J. Morrison, and Michael F. Clarke. Bmil, stem cells, and senescence regulation. J.Clin. Invest. 2004; 113: 175-179

Claims

Claims
1. A composition for inducing de-differentiation of astrocytes into neural stem cells, comprising a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
2. The composition as set forth in claim 1, wherein the nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein is a vector that allows the Bmi- 1 protein to be expressed.
3. The composition as set forth in claim 1, wherein the nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein is a virus that expresses the Bmi-1 protein.
4. The composition as set forth in claim 1, wherein the de-differentiated neural stem cells have ability to differentiate into astrocytes, neurons and oligodendrocytes .
5. A method for inducing the de-differentiation of astrocytes into neural stem cells by treating the astrocytes with a Bmi-1 protein or a nucleic acid material containing a nucleotide sequence coding for a Bmi-1 protein.
6. The method as set forth in claim 5, wherein the de-differentiated neural stem cells have ability to differentiate into astrocytes, neurons and oligodendrocytes .
7. The method as set forth in claim 5, comprising steps of:
(i) culturing astrocytes in a medium; (ii) treating the astrocytes with the Bmi-1 protein or the nucleic acid material; and
(iii) inducing the astrocytes to differentiate into neural stem cells.
8. The medium as set forth in claim 7, wherein the medium of step (i) contains bFGF.
9. A neural stem cell, produced using the method of claim 5.
10. A method of differentiating the neural stem cells produced using the method of claim 5 into astrocytes, neurons, and oligodendrocytes.
PCT/KR2006/001350 2006-02-27 2006-04-12 De-differentiation of astrocytes into neural stem cell using bmi-1 WO2007097494A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP06732855A EP1987148A4 (en) 2006-02-27 2006-04-12 De-differentiation of astrocytes into neural stem cell using bmi-1
JP2008557196A JP2009528050A (en) 2006-02-27 2006-04-12 Dedifferentiation of astrocytes into neural stem cells using Bmi-1

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2006-0019018 2006-02-27
KR20060019018 2006-02-27

Publications (1)

Publication Number Publication Date
WO2007097494A1 true WO2007097494A1 (en) 2007-08-30

Family

ID=38437522

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2006/001350 WO2007097494A1 (en) 2006-02-27 2006-04-12 De-differentiation of astrocytes into neural stem cell using bmi-1

Country Status (5)

Country Link
EP (1) EP1987148A4 (en)
JP (1) JP2009528050A (en)
KR (1) KR100844971B1 (en)
CN (1) CN101389761A (en)
WO (1) WO2007097494A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009079007A1 (en) * 2007-12-17 2009-06-25 Gliamed, Inc. Stem-like cells and method for reprogramming adult mammalian somatic cells
US8048999B2 (en) 2005-12-13 2011-11-01 Kyoto University Nuclear reprogramming factor
US8058065B2 (en) 2005-12-13 2011-11-15 Kyoto University Oct3/4, Klf4, c-Myc and Sox2 produce induced pluripotent stem cells
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8211697B2 (en) 2007-06-15 2012-07-03 Kyoto University Induced pluripotent stem cells produced using reprogramming factors and a rho kinase inhibitor or a histone deacetylase inhibitor
US8791248B2 (en) 2007-12-10 2014-07-29 Kyoto University Nuclear reprogramming factor comprising miRNA and a protein factor
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
US9453205B2 (en) 2009-10-31 2016-09-27 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US9499797B2 (en) 2008-05-02 2016-11-22 Kyoto University Method of making induced pluripotent stem cells
US9528087B2 (en) 2009-10-31 2016-12-27 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
US9885014B2 (en) 2010-05-13 2018-02-06 The Regents Of The University Of California Re-conditioned serum-free culture medium composition obtained by culturing mammalian cells in Fibromodulin

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011135864A (en) 2009-12-30 2011-07-14 Korea Univ Research & Business Foundation Composition for retrodifferentiating somatic cell to embryonic stem cell-like cell, by using oct4 in combination with bmi1 or upstream regulator thereof, and method for generating embryonic stem cell-like cell using the same
KR101158402B1 (en) * 2009-12-30 2012-06-22 주식회사 일진케이제이 Generation composition for induced pluripotent stem cells with Bmi1 and Oct4, and method of manufacturing induced pluripotent stem cells using the same
CN102242146B (en) * 2010-05-10 2015-11-25 高丽大学校产学协力团 Composition and the method with its generation generate induced pluripotent stem cells
CN106754728B (en) * 2016-12-30 2020-05-15 中国人民解放军总医院 Metanephric mesenchymal cell line and preparation method and application thereof
CN109674809B (en) * 2018-12-27 2022-08-09 吉林大学 Composition containing miR-124-3P and application thereof in medicine for inducing neuron formation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6689583B1 (en) * 1995-05-10 2004-02-10 Boehringer Ingelheim International Gmbh Chromatin regulator genes
US6897061B1 (en) * 2000-06-16 2005-05-24 Spinal Cord Society Transdifferentiation of glial cells
US6919204B2 (en) * 2000-09-29 2005-07-19 Sangamo Biosciences, Inc. Modulation of gene expression using localization domains

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003033179A (en) * 2001-07-05 2003-02-04 Asahi Kasei Corp Vector for reversible transduction of gene
KR20010099203A (en) * 2001-09-11 2001-11-09 이상훈 Method for differentiation of neural stem cell to dopaminergic neuron

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6689583B1 (en) * 1995-05-10 2004-02-10 Boehringer Ingelheim International Gmbh Chromatin regulator genes
US6897061B1 (en) * 2000-06-16 2005-05-24 Spinal Cord Society Transdifferentiation of glial cells
US6919204B2 (en) * 2000-09-29 2005-07-19 Sangamo Biosciences, Inc. Modulation of gene expression using localization domains

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
LANG B. ET AL., NEUROSCIENCE, vol. 128, no. 4, 2004, pages 775 - 783, XP004586778 *
MOLOFSKY A.V. ET AL., GENES DEV., vol. 19, no. 12, 15 June 2005 (2005-06-15), pages 1432 - 1437, XP002511653 *
MOLOFSKY A.V. ET AL., NATURE, vol. 425, no. 6961, 30 October 2003 (2003-10-30), pages 962 - 967, XP002324138 *
See also references of EP1987148A4 *
ZENCAK D. ET AL., J. NEUROSCI., vol. 25, no. 24, 15 June 2005 (2005-06-15), pages 5774 - 5783, XP002533121 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US8048999B2 (en) 2005-12-13 2011-11-01 Kyoto University Nuclear reprogramming factor
US8058065B2 (en) 2005-12-13 2011-11-15 Kyoto University Oct3/4, Klf4, c-Myc and Sox2 produce induced pluripotent stem cells
US8257941B2 (en) 2007-06-15 2012-09-04 Kyoto University Methods and platforms for drug discovery using induced pluripotent stem cells
US8211697B2 (en) 2007-06-15 2012-07-03 Kyoto University Induced pluripotent stem cells produced using reprogramming factors and a rho kinase inhibitor or a histone deacetylase inhibitor
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
US9714433B2 (en) 2007-06-15 2017-07-25 Kyoto University Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
US8791248B2 (en) 2007-12-10 2014-07-29 Kyoto University Nuclear reprogramming factor comprising miRNA and a protein factor
US20100330063A1 (en) * 2007-12-17 2010-12-30 Gliamed, Inc. Stem-like cells, method for de-differentiating mammalian somatic cells into stem-like cells, and method for differentiating stem-like cells
WO2009079007A1 (en) * 2007-12-17 2009-06-25 Gliamed, Inc. Stem-like cells and method for reprogramming adult mammalian somatic cells
US9499797B2 (en) 2008-05-02 2016-11-22 Kyoto University Method of making induced pluripotent stem cells
US9453205B2 (en) 2009-10-31 2016-09-27 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US9528087B2 (en) 2009-10-31 2016-12-27 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US10017737B2 (en) 2009-10-31 2018-07-10 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US10131879B2 (en) 2009-10-31 2018-11-20 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US10260046B2 (en) 2009-10-31 2019-04-16 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US10557123B2 (en) 2009-10-31 2020-02-11 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US10563176B2 (en) 2009-10-31 2020-02-18 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US11795439B2 (en) 2009-10-31 2023-10-24 Genesis Technologies Limited Methods for reprogramming cells and uses thereof
US9885014B2 (en) 2010-05-13 2018-02-06 The Regents Of The University Of California Re-conditioned serum-free culture medium composition obtained by culturing mammalian cells in Fibromodulin

Also Published As

Publication number Publication date
KR20070089089A (en) 2007-08-30
KR100844971B1 (en) 2008-07-09
EP1987148A1 (en) 2008-11-05
EP1987148A4 (en) 2009-08-05
CN101389761A (en) 2009-03-18
JP2009528050A (en) 2009-08-06

Similar Documents

Publication Publication Date Title
WO2007097494A1 (en) De-differentiation of astrocytes into neural stem cell using bmi-1
US9249391B2 (en) Methods of generating neural stem cells
US11512285B2 (en) Method of preparing induced neural stem cells reprogrammed from non-neuronal cells using HMGA2
US20130280808A1 (en) COMPOSITION FOR REPROGRAMMING SOMATIC CELLS TO GENERATE INDUCED PLURIPOTENT STEM CELLS, COMPRISING Oct4 IN COMBINATION WITH Bmi1 OR ITS UPSTREAM REGULATOR, AND METHOD FOR GENERATING INDUCED PLURIPOTENT STEM CELLS USING THE SAME
KR100794362B1 (en) De-differentiation of astrocytes into neural stem cell using Nanog
KR100794359B1 (en) De-differentiation of astrocytes into neural stem cell using Shh
Lubetzki et al. Clonal segregation of oligodendrocytes and astrocytes during in vitro differentiation of glial progenitor cells
KR101211624B1 (en) Generation composition for induced pluripotent stem cells with Shh, FGFR tyrosine kinase inhibitor, MEK inhibitor and GSK inhibitor treatment and method of manufacturing induced pluripotent stem cells using the same
KR101211610B1 (en) Generation composition for induced pluripotent stem cells with Bmi1, MEK inhibitor and GSK inhibitor treatment and method of manufacturing induced pluripotent stem cells using the same
KR101188878B1 (en) Generation composition for induced pluripotent stem cells with purmorphamine treatment and Oct4, and method of manufacturing induced pluripotent stem cells using the same
KR101211962B1 (en) Generation composition for induced pluripotent stem cells with Bmi1, G9aHMTase inhibitor and DMNT inhibitor treatment and method of manufacturing induced pluripotent stem cells using the same
KR101158402B1 (en) Generation composition for induced pluripotent stem cells with Bmi1 and Oct4, and method of manufacturing induced pluripotent stem cells using the same
KR101188876B1 (en) Generation composition for induced pluripotent stem cells with Shh and Oct4, and method of manufacturing induced pluripotent stem cells using the same
KR101188877B1 (en) Generation composition for induced pluripotent stem cells with oxysterols treatment and Oct4, and method of manufacturing induced pluripotent stem cells using the same
KR20110124105A (en) Method of manufacturing induced epiblast stem cells with bmi1 and induced pluripotent stem cells using the same
KR20110124103A (en) Generation composition for induced pluripotent stem cells with bmi1 and histone deacetylase inhibitor treatment and method of manufacturing induced pluripotent stem cells using the same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006732855

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200680053500.5

Country of ref document: CN

Ref document number: 2008557196

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE