CN111876443A - Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology - Google Patents

Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology Download PDF

Info

Publication number
CN111876443A
CN111876443A CN202010795950.3A CN202010795950A CN111876443A CN 111876443 A CN111876443 A CN 111876443A CN 202010795950 A CN202010795950 A CN 202010795950A CN 111876443 A CN111876443 A CN 111876443A
Authority
CN
China
Prior art keywords
cells
cell
nerve
neural cell
culture solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010795950.3A
Other languages
Chinese (zh)
Inventor
樊晋宇
池金鹏
赵亚琪
程雪
张诗薇
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shandong Tianchuan Precision Medical Technology Co Ltd
Original Assignee
Shandong Tianchuan Precision Medical Technology 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 Shandong Tianchuan Precision Medical Technology Co Ltd filed Critical Shandong Tianchuan Precision Medical Technology Co Ltd
Priority to CN202010795950.3A priority Critical patent/CN111876443A/en
Publication of CN111876443A publication Critical patent/CN111876443A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • 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
    • C12N15/66General methods for inserting a gene into a vector to form a recombinant vector using cleavage and ligation; Use of non-functional linkers or adaptors, e.g. linkers containing the sequence for a restriction endonuclease
    • 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
    • C12N5/0622Glial cells, e.g. astrocytes, oligodendrocytes; Schwann cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0656Adult fibroblasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1307Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from adult fibroblasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The invention provides a method for inducing somatic cell reprogramming to a nerve cell model by utilizing a transdifferentiation technology, and particularly relates to a method for directly transdifferentiating skin fibroblasts of a schizophrenia patient into nerve cells by utilizing a nerve cell development key transcription factor combination. The invention has the advantages that somatic cells can be successfully transdifferentiated into nerve cells, the transdifferentiation efficiency is high, a relatively accurate cell model can be established, the neural development process of schizophrenia can be simulated, and a new possibility is provided for further researching the pathogenesis of schizophrenia.

Description

Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology
Technical Field
The invention relates to the field of biotechnology, in particular to a method for inducing reprogramming of somatic cells to a neural cell model by utilizing a transdifferentiation technology.
Background
The central nervous system injury includes brain injury, spinal cord injury or neurodegenerative disease, etc., the treatment of central nervous system injury or degenerative disease is still a worldwide medical problem at present, the clinical main treatment methods for nerve injury include surgical removal of etiology, avoidance of secondary injury, protection of residual neuron function, and late rehabilitation treatment, etc., however, these methods have little effect on nerve regeneration, and the prognosis effect is still poor, which is related to adverse factors such as low neuronal regeneration function, existence of a barrier of scar tissue hyperplasia between the injured area and normal tissue, etc. Therefore, how to promote the regeneration of nerve cells and break through the regeneration barrier is an important focus of present neuroscientists and clinical researchers.
The current methods for promoting nerve cell regeneration mainly comprise:
1. by inducing traditional induced pluripotent stem cells (iPS), namely inducing somatic cells into stem cells and then transforming the stem cells into neurons or precursor cells, the central nervous system cells induced and differentiated by the iPS have autologous cell replacement therapy potential for various diseases. However, the use of iPS has the following problems: (1) a certain amount of iPS needs to be prepared in time, and a patient prepared for nerve transplantation can not wait for the process of long preparation time of the stem cells such as induction, differentiation, screening and the like in vitro; (2) the induction conversion rate of iPS cells is low, and the iPS cells need to be proliferated again, and the differentiation is uncertain in the process; (3) undifferentiated stem cells used for transplantation, if not differentiated in time, increase the risk of tumor formation in vivo.
2. Differentiation was directly induced by somatic cells in vitro. In recent years, a large number of scientific experiments show that part of cells can be directly induced into somatic cells through reprogramming of the cells by transcription factors under the mediation of vectors such as lentiviruses and retroviruses, and the time and uncertainty required by secondary induced differentiation are greatly reduced after the stem cell stage is passed. At present, researchers transfer two factors, namely Neurog2 and Dlx2, into astrocytes respectively, and prove that the astrocytes can be directly induced into mature neurons under the action of the two factors respectively and can form synaptic structures among the differentiated neurons. In addition, it has been found that fibroblasts have the ability to be directly transformed into cholinergic neurons under the combined influence of internal and external factors. After intensive research, it was found that adult skin fibroblasts can be directly and efficiently converted into high-purity motor neurons without passing through the induced pluripotent stem cell stage. However, researchers found that stem cell therapy was derived from cells of patients themselves during clinical transformation, but fibroblasts from patients with mental diseases (such as amyotrophic lateral sclerosis, schizophrenia, etc.) have poor viability after transformation compared with fibroblasts of normal people, and partial cell differentiation uncertainty always exists during the differentiation induction process of the fibroblasts, which all cause difficulty in practical application of stem cell therapy for neurological diseases.
Disclosure of Invention
In order to solve the problems of low transdifferentiation efficiency, high differentiation uncertainty, lack of mental disease models and the like in the process of transforming fibroblasts from patients with nervous system diseases into nerve cells in the prior art, the invention provides a method for inducing somatic cell reprogramming to a nerve cell model by using a transdifferentiation technology, and particularly relates to a method for directly transdifferentiating skin fibroblasts of patients with schizophrenia into nerve cells by using a nerve cell development key transcription factor combination.
More specifically, the method constructs a nerve induction related factor lentiviral plasmid, and amplifies ASCL1 and MYT1L fragments by a PCR method to connect the lentiviral vector with the plasmid. Recombinant plasmids pLVX-ASCL1 and pLVX-MYT1L are obtained by enzyme digestion, linkage, escherichia coli transformation and the like, and virus particles with good infection activity are combined to infect somatic cells to induce and form nerve-like cells.
In one embodiment, the method of the present invention comprises the steps of:
firstly, a nerve induction related factor lentivirus plasmid is constructed, a PCR method is used for amplification to obtain ASCL1 and MYT1L fragments, and the fragments are connected into a lentivirus vector pLVX-IRES-mCherry. And obtaining recombinant plasmids pLVX-ASCL1 and pLVX-MYT1L through enzyme digestion, chaining, escherichia coli transformation and the like, respectively combining the two recombinant plasmids serving as target plasmids with packaging plasmids pMD2.G and pSPAX2 to carry out lentivirus packaging to obtain viruses with infection activity, infecting skin fibroblasts of a patient with schizophrenia by using virus particle combinations with good infection activity, and further successfully inducing and cloning to obtain a nerve cell model.
The invention has the advantages that somatic cells can be successfully transdifferentiated into nerve cells, the transdifferentiation efficiency is high, a more accurate nerve disease cell model can be established, the nerve development process of schizophrenia can be simulated, and a new possibility is provided for further researching the pathogenesis of schizophrenia and the stem cell therapy thereof.
Drawings
The accompanying drawings, which are included to provide a further understanding of the invention and are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and together with the description serve to explain the principles of the invention and not to limit the invention. In the drawings:
FIG. 1 shows a comparison of 15-day survival and transformation rates of cells induced by various groups of genes;
FIG. 2 nerve cell induction formation process;
FIG. 3 induction of nerve cell protein level detection marker gene expression.
Detailed Description
The preferred embodiments of the present invention will be described in conjunction with the accompanying drawings, and it will be understood that they are described herein for the purpose of illustration and explanation and not limitation.
EXAMPLE 1 transdifferentiation of fibroblasts
Step 1, using cDNA reverse transcription of mRNA of healthy human whole blood as a template, designing primers according to sequence information in NCBI websites to amplify full-length genes of ASCL1, MYT1L, Scl1, Brn2 and Isx9, carrying out electrophoresis on PCR products by 8% agarose gel, cutting off bands with proper size for gel recovery, and carrying out double enzyme digestion on target bands recovered by the gel and a vector plasmid pLVX (purchased from Addgene). The double digestion system was allowed to react at 37 ℃ for 2-4 hours, and the plasmid was purified using a plasmid purification kit (purchased from OMEGA). The concentration of the purified plasmid was measured on a Nanodrop2000, the target fragment and the vector fragment were mixed at a ratio of 3:1, and ligation was performed overnight at 4 ℃. Transforming the connected plasmid through DH5 alpha competent cell, smearing on LB solid plate with ampicillin resistance, screening;
step 2, packaging lentivirus: transfection of 293T cells: 12-24 hours before transfection, 1 × 106293T cells in a logarithmic growth phase are inoculated into a cell culture bottle of 75CM2 and cultured in a 5% CO2 incubator at 37 ℃, and the cells can be used for transfection when the confluence degree is 40% -70%. One hour prior to transfection, cells were replaced with fresh medium and plasmid transfection was performed using the calcium phosphate method. Mu.g of recombinant plasmid, 10. mu.g of each of the viral packaging plasmids pMD2.G and pSPAX2 (both from Clontech), was added to ultrapure water in a total volume of 450. mu.l, 50. mu.l of 2.5mol/L CaCl2 was added and mixed well, and then mixed dropwise with 500. mu.l of 2X BES, left at room temperature for 15-30min, added well to HEK293T cells, and then cultured in a 37 5% CO2 incubator. After 8-16 hours, wash twice with PBS and replace with fresh medium. Collecting lentiviruses: collecting virus 48 hours after liquid change, adding culture solution containing slow virus into a 50ml centrifuge tube, centrifuging at 4 ℃ and 10000g for 15min to remove cell debris, adding supernatant into an ultrafiltration tube at 4 ℃ and 2700g, centrifuging for 30min, and concentrating virus solution to 600 mu l.
And 3, transferring the skin fibroblasts (donated by the psychiatric hospital in Henan province) of the schizophrenia patient to a cell culture bottle 24 hours before collecting the viruses, culturing in a 5% CO2 incubator at 37 ℃, and infecting when the cell confluency is 40% -70%. Before cell infection, fresh culture solution is replaced, and concentrated lentiviruses are respectively added into cells to be infected singly or in a 1:1 mixed mode (the total addition amount is 1.2 ml). After 36 hours, the medium was changed to a neural cell induction medium (DMEM/F12 medium supplemented with 1X N2 and B27, 20ng/ml β FGF, 20ng/ml EGF), and then observed every 2 days.
The experimental results are as follows:
the recombinant virus solutions are respectively added into the patient fibroblasts with the confluency of about 70-90% singly or in combination, and the cell survival rate and the transformation rate are shown in figure 1 after 15d observation. As shown in fig. 1, co-transfection of ASCL1 and MYT1L genes significantly improved the cell survival rate and transformation rate of fibroblasts reprogrammed to neural cells in schizophrenic patients. Based on this screening result, a group of cells co-transfected with ASCL1 and MYT1L genes was selected for the following observation and further experiments.
Morphology observation of co-transfected ASCL1 and MYT1L groups of cells: red fluorescence expression was observed after 24 hours. As shown in FIG. 2, 0Day is the cell state before infection, 5Day, the cell morphology begins to change, most cells gradually become slender, 10Day, the cells begin to have synapses and become nerve-like cells, 15Day, the cells can obviously be observed to grow slender synapses, most cells are transdifferentiated into nerve cells, and the differentiation efficiency reaches more than 80%.
Example 2 nerve cell identification
To identify whether the induced cells are nerve cells, MAP2 and GFAP two nerve marker genes are selected to carry out protein level identification on ASCL1 and MYT1L induced nerve cells by an immunofluorescence method. As shown in fig. 3: the mCherry is an exogenous gene inserted by cell expression and has red fluorescence; immunofluoresence is the detection result of immunofluorescence, and a secondary antibody is labeled by an FITC group and carries green fluorescence; DAPI is the result of staining the nuclei; merge is a composite image of three fluorescence pictures; the results showed that almost all induced cellular MAP2 and GFAP protein expression were positive.
It will be apparent to those skilled in the art that various changes and modifications may be made in the present invention without departing from the spirit and scope of the invention. Thus, if such modifications and variations of the present invention fall within the scope of the claims of the present invention and their equivalents, the present invention is also intended to include such modifications and variations.

Claims (3)

1. A method of inducing reprogramming of somatic cells to a neural cell model, the method comprising the steps of:
step 1, amplifying full-length genes of ASCL1 and MYT1L by using cDNA reverse transcribed by mRNA of healthy human whole blood as a template, performing electrophoresis on a PCR product by using 8% agarose gel, cutting off a band with a proper size for gel recovery, performing double enzyme digestion on a target band recovered by the gel and a vector plasmid pLVX, purifying, performing mixing and connection reaction according to the concentration ratio of a target fragment to a vector fragment of 3:1, transforming the connected plasmid by using DH5 alpha competent cells, coating the transformed plasmid on an LB solid plate with ampicillin resistance, and screening;
step 2, packaging lentivirus: transfection of 293T cells: taking 20 mu g of the recombinant plasmid obtained in the step 1, respectively taking 10 mu g of the virus packaging plasmids pMD2.G and pSPAX2, adding into ultrapure water to make the total volume be 450 mu L, and then adding 50 mu L of 2.5mol/L CaCl2Mixing, mixing with 500 μ l 2X BES drop by drop, standing at room temperature for 15-30min, adding into HEK293T cell, culturing at 37 deg.C in 5% CO2 incubator for 8-16 hr, washing with PBS twice, and replacing with fresh culture solution; collecting lentiviruses: collecting virus 48 hours after liquid change, centrifuging culture solution containing slow virus, removing cell debris, centrifuging supernatant, and concentrating virus solution to 600 μ l;
and 3, transferring the skin fibroblasts of the schizophrenia patient to a cell culture bottle 24 hours before collecting the viruses, infecting the skin fibroblasts when the confluence degree of the cells is 40-70%, changing fresh culture solution before infecting the cells, mixing and adding 600 mu L of two kinds of concentrated slow viruses of ASCL1 and MYT1L into the cells to be infected, changing a nerve cell induction culture solution after 30-36 hours, and changing the solution every 2 days until the fibroblasts are differentiated into the nerve cells.
2. The method of claim 1, wherein the neural cell induction culture solution of step 3 is prepared by the following method: DMEM/F12 medium was supplemented with 1X N2 and B27, 20ng/ml β FGF, 20ng/ml EGF.
3. A method of modeling a disease in a neural cell, wherein the neural cell is produced by the method of claim 1.
CN202010795950.3A 2020-08-10 2020-08-10 Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology Pending CN111876443A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010795950.3A CN111876443A (en) 2020-08-10 2020-08-10 Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010795950.3A CN111876443A (en) 2020-08-10 2020-08-10 Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology

Publications (1)

Publication Number Publication Date
CN111876443A true CN111876443A (en) 2020-11-03

Family

ID=73210369

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010795950.3A Pending CN111876443A (en) 2020-08-10 2020-08-10 Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology

Country Status (1)

Country Link
CN (1) CN111876443A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102286535A (en) * 2011-06-17 2011-12-21 中国人民解放军第二军医大学 Method for transdifferentiation of fibroblasts into hepatic stem cells
CN102796696A (en) * 2011-05-27 2012-11-28 复旦大学附属华山医院 Neurons directly induced from human skin cells and preparation method for neurons
CN105950557A (en) * 2016-04-22 2016-09-21 中国科学院生物物理研究所 Signal channel for controlling human neuron reprogramming and application of signal channel

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102796696A (en) * 2011-05-27 2012-11-28 复旦大学附属华山医院 Neurons directly induced from human skin cells and preparation method for neurons
CN102286535A (en) * 2011-06-17 2011-12-21 中国人民解放军第二军医大学 Method for transdifferentiation of fibroblasts into hepatic stem cells
CN105950557A (en) * 2016-04-22 2016-09-21 中国科学院生物物理研究所 Signal channel for controlling human neuron reprogramming and application of signal channel

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ENHANCED CONVERSION OF INDUCED NEURONAL CELLS (IN CELLS) FROM HU: "Enhanced conversion of induced neuronal cells (iN cells) from human fibroblasts: Utility in uncovering cellular deficits in mental illness-associated chromosomal abnormalities", 《NEUROSCIENCE RESEARCH》 *
SOHAM CHANDA等: "Generation of Induced Neuronal Cells by the Single Reprogramming Factor ASCL1", 《STEM CELL REPORTS》 *

Similar Documents

Publication Publication Date Title
EP2832851B1 (en) Immortalized stem cells and medicinal composition and medicinal preparation comprising product thereof as active ingredient
CN112194717B (en) Peptide segment Tr for promoting regeneration of cochlear hair cells and application thereof
WO2022166026A1 (en) Cre recombinase induction-based large-scale lentiviral gene drug preparation system, and method
Wang et al. Reprogramming glial cells into functional neurons for neuro-regeneration: challenges and promise
CN108588026A (en) The preparation method and its usage of the clinical grade mescenchymal stem cell of height expression IL10
CN103773771A (en) Transcription factor system as well as preparation method and application thereof
US7592170B2 (en) Selective precipitation of viruses using polyectrolytes
CN111876443A (en) Method for inducing reprogramming of somatic cells to neural cell model by using transdifferentiation technology
Peng et al. Astrocyte reprogramming in stroke: opportunities and challenges
CN113046330B (en) Lentivirus and medicine carrying erythroid gene editing system
CN110656087B (en) MANF gene modified umbilical cord mesenchymal stem cell and preparation method and application thereof
WO2021228050A1 (en) Method for inducing glial cells transdifferentiation into functional neurons, and application thereof
CN108456697A (en) Using slow virus carrier EF1 α promoter Optimal Expression ABCD1 gene therapy adrenoleukodystrophy diseases
CA3197178A1 (en) Neurod1 combination vector
CN113171369A (en) Application of polypyrimidine sequence binding protein in preparation of spinal cord injury repair drug
CN116437943A (en) Functional fragments, combinations and uses thereof for reprogramming
CN111073906A (en) Method for combining optogenetic and enkephalinase genes
CN115845030A (en) Application of nerve growth factor inducing protein or its gene
WO2024036839A1 (en) Gene-modified cell drug expressing pah, method for preparing same, and use thereof
Zhu et al. Establishment of a lentiviral vector encoding human HGF and the infection of human ADSCs
CN108795987A (en) A kind of neural stem cell preparation method and application of FerritinH, Bcl2 and EGFP gene combined modification
CN114941013B (en) Recombinant mesenchymal stem cells for treating diabetic pneumonia
CN111662876B (en) Method for inducing human fibroblast to reprogram to Leydig-like cell
CN116200343A (en) Bone marrow mesenchymal stem cell for efficiently synthesizing dopamine and preparation method and application thereof
CN101787359A (en) Recombinant lentivirus for carrying out targeted induction on osteogenic differentiated cell apoptosis as well as preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
RJ01 Rejection of invention patent application after publication

Application publication date: 20201103

RJ01 Rejection of invention patent application after publication