CN114645021A - Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof - Google Patents

Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof Download PDF

Info

Publication number
CN114645021A
CN114645021A CN202011520308.0A CN202011520308A CN114645021A CN 114645021 A CN114645021 A CN 114645021A CN 202011520308 A CN202011520308 A CN 202011520308A CN 114645021 A CN114645021 A CN 114645021A
Authority
CN
China
Prior art keywords
shrna
hla
pluripotent stem
seq
mir
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
CN202011520308.0A
Other languages
Chinese (zh)
Inventor
王淋立
陈月花
莫健
杨建国
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.)
Future Intelligent Regenerative Medicine Research Institute Guangzhou Co ltd
Original Assignee
Future Intelligent Regenerative Medicine Research Institute Guangzhou 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 Future Intelligent Regenerative Medicine Research Institute Guangzhou Co ltd filed Critical Future Intelligent Regenerative Medicine Research Institute Guangzhou Co ltd
Priority to CN202011520308.0A priority Critical patent/CN114645021A/en
Publication of CN114645021A publication Critical patent/CN114645021A/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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N5/0608Germ cells
    • C12N5/0611Primordial germ cells, e.g. embryonic germ cells [EG]
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • 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
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • 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

Abstract

The invention discloses a pluripotent stem cell expressing a targeted CD47 inhibitory factor, a derivative thereof and application thereof. An expression sequence of a CD47 inhibitor is introduced into the genome of the pluripotent stem cell or the derivative thereof, wherein the CD47 inhibitor is at least one of shRNA and/or shRNA-miR of a targeted CD 47. After the CD47 inhibitor expressed by the CD47 pluripotent stem cells or the derivatives thereof is encapsulated by exosomes, the exosomes carry the CD47 inhibitor to be combined with target cells, the CD47 inhibitor contained in the exosomes is released, so that a CD47 channel is blocked, immunosuppression is relieved, the immune system is activated, the activity of NK cells is recovered, and the exosomes can effectively eliminate tumor cells.

Description

Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof
Technical Field
The invention belongs to the technical field of genetic engineering, and particularly relates to a pluripotent stem cell expressing a targeted CD47 inhibitory factor, a derivative thereof and application thereof.
Background
CD47 (i.e., an integrin-associated protein, IAP), is a transmembrane glycoprotein that is widely expressed on the cell surface and belongs to the immunoglobulin superfamily. CD47 has a molecular weight between 47-55kD and comprises 1 extracellular Ig-like variable domain, 5 highly hydrophobically extended transmembrane segments and 1 carboxy-terminal cytoplasmic tail spliced selectively. CD47 is a pluripotent molecule whose primary functions include:
(1) CD47 binds to CD47 ligand signal-regulating protein (SIRP. alpha.) and interacts with integrins to regulate cell-cell communication. CD47 binds to SIRP alpha on the surface of macrophages, activates tyrosine phosphorylase, and inhibits the aggregation of myosin at coordination sites under macrophage synaptic membrane. Thus, if CD47 is deleted, old or damaged cells will be engulfed.
(2) CD47 binds to thrombospondin-1 and is involved in the interaction of cells with extracellular machinery.
(3) CD47 is closely related to cellular processes such as apoptosis, proliferation, adhesion, migration, etc., plays a key role in immune and cardiovascular responses, and is an important target for cardiovascular disease treatment.
(4) CD47 is associated with tumor immune escape and has a major impact on the microenvironment of tumor cells.
CD47 has now been found to be aberrantly expressed in a variety of tumor types, including Acute Myeloid Leukemia (AML), Chronic Myeloid Leukemia (CML), Acute Lymphocytic Leukemia (ALL), non-hodgkin's lymphoma (NHL), Multiple Myeloma (MM), bladder cancer, and other solid tumors. Thus, CD47 also exerts an inhibitory effect during antigen presentation to adaptive immune T cells by innate immune cells such as dendritic cells. In recent years, the gradual rise of CD47 in tumor immunotherapy is a potential effective and widely applied target of tumor immunotherapy, so that a plurality of inhibitors have been developed, and can specifically block a CD 47-SIRPa signal pathway to enhance the immune response to tumors.
On the other hand, in the field of cell therapy, the problem of immunological compatibility of allogens remains a big problem. In recent years, a plurality of reports have been provided that the deletion expression of genes on the cell surfaces of HLA-I and HLA-II or the genes thereof is realized by knocking out genes such as B2M, CIITA and the like, so that the cells have immune tolerance or escape T/B cell specific immune response, and universal PSCs with immune compatibility are generated, thereby laying an important foundation for the application of wider universal PSCs source cells, tissues and organs. Also, cells have been reported to overexpress CTLA4-Ig, PD-L1 and thereby inhibit allogeneic immune rejection. However, these approaches are either not fully immune compatible, and still allow for immunological rejection of the allogens by other routes; or completely eliminate the allogeneic immune rejection response, but simultaneously make the cells of the donor-derived transplant lose the antigen presenting capability, which brings great risk of diseases such as tumorigenicity and virus infection to the recipient.
Therefore, it is also reported that, when the B2M is not directly knocked out, the HLA-A, HLA-B is knocked out or the CIITA is knocked out together, the HLA-C is kept, 12 HLA-C immune matching antigens covering more than 90% of people are constructed, so that the transplanted cells still have a certain degree of antigen presenting function, and the inherent immune response of NK cells can be inhibited through the HLA-C. However, in the cells, the antigen type presented by HLA-I antigen is reduced by more than two thirds, the integrity of the presented antigen is reduced irreversibly, the presenting of various tumor, virus and other disease antigens has great bias, the risk of diseases such as tumor and virus infection is still kept to a certain extent, and the pathogenic risk is higher under the condition that CIITA is knocked out simultaneously; secondly, 12 high-frequency immune match HLA-C antigen species are very different, and the part of the area can only account for 70 percent by verification and calculation, while the HLA data of large sample size which is not authoritative currently in China, Indian and other big countries is displayed, so that the prepared general PSCs are still subjected to huge match vacancy tests; thirdly, the method can go through repeated gene editing for a plurality of times, at least two rounds of single cell isolation culture meters are needed according to each gene editing, the whole process needs at least more than six rounds of single cell isolation culture, and the processes are inevitable and cause various unpredictable mutations of cells due to multiple times of gene editing off-target or unstable chromatin or due to passage proliferation of a large number of single cells, thereby further inducing various problems of carcinogenesis, metabolic diseases and the like. It follows that such immuno-compatible schemes are also a matter of convenience in the "transition period", and many problems remain that are not better solved.
In addition, inducing killing of the suicide gene after donor tissue and cell disease has been induced, which results in serious tissue necrosis, cytokine storm and other unpredictable disease risk problems, and it is a big problem that proper donor cells, tissues and organs do not exist after the cell death of the design.
Disclosure of Invention
The purpose of the present invention is to provide a pluripotent stem cell or a derivative thereof.
The invention also aims to provide application of the pluripotent stem cells or the derivatives thereof and exosomes secreted by the pluripotent stem cells in preparation of anti-tumor therapeutic preparations or drugs.
It is another object of the invention to provide an exosome.
The technical scheme adopted by the invention is as follows:
in a first aspect of the present invention, there is provided:
a pluripotent stem cell or a derivative thereof, wherein a genome of the pluripotent stem cell or the derivative thereof has an expression sequence of a CD47 inhibitory factor introduced therein; the CD47 inhibitor is at least one of shRNA and/or shRNA-miR of targeting CD 47.
The inventor finds that after an shRNA/shRNA-miR expression sequence of a CD47 inhibitor is introduced into a pluripotent stem cell or a derivative thereof, exosomes secreted in the pluripotent stem cell or the derivative thereof contain high-abundance shRNA/shmiRNA of the CD47 inhibitor targeting CD47 and mature siRNA, and the exosomes carry the inhibitors, are combined with a target cell and then released, so that a CD47 channel is blocked, immunosuppression is relieved, the immune system is activated, the activity of NK cells is restored, and the tumor cells can be effectively eliminated.
Further, the sequence of the shRNA and/or shRNA-miR of the targeting CD47 is shown in SEQ ID NO. 1-SEQ ID NO. 2.
In a second aspect of the present invention, there is provided:
a pluripotent stem cell or a derivative thereof, wherein a gene group of the pluripotent stem cell or the derivative thereof has an expression sequence of a CD47 inhibitory factor introduced therein; the CD47 inhibitor is at least one of shRNA and/or shRNA-miR of targeting CD 47.
The B2M gene and/or CIITA gene of the genome of the pluripotent stem cell or the derivative thereof is knocked out.
When B2M and CIITA genes are knocked out, the influence of HLA-I and HLA-II molecules is completely eliminated, and therefore, the tumor treatment effect is optimal.
In a third aspect of the present invention, there is provided:
a pluripotent stem cell or a derivative thereof, wherein a gene group of the pluripotent stem cell or the derivative thereof has an expression sequence of a CD47 inhibitory factor introduced therein; the CD47 inhibitor is at least one of shRNA and/or shRNA-miR of targeting CD 47.
A first nucleic acid molecule is introduced into the genome of the pluripotent stem cell or the derivative thereof; and the pluripotent stem cell or the derivative thereof has a second nucleic acid molecule introduced into the 3' UTR region of the immune response-related gene; the first nucleic acid molecule encodes a small nucleic acid molecule that mediates RNA interference, the small nucleic acid molecule specifically targets the transcript of the second nucleic acid molecule, and the small nucleic acid molecule does not target any other mRNA or incrna of the pluripotent stem cell or a derivative thereof.
In the technical scheme, the small nucleic acid molecule coded by the first nucleic acid molecule can be specifically combined with a transcription product of a second nucleic acid molecule introduced into a 3' UTR region of an immune response related gene, so that an RNA interference program is started, mRNA of the immune response related gene is degraded or silenced, and the expression of the immune response related gene is blocked, so that the cell has the immune compatibility characteristic, and the allogeneic immune rejection response reaction can be eliminated or reduced. Moreover, the RNA interference program only acts on such engineered pluripotent stem cells or derivatives thereof, and thus, when such cells or derivatives are transplanted into a recipient, RNA interference of the immune response-related gene mediated by the small nucleic acid molecule encoded by the first nucleic acid molecule and the second nucleic acid molecule introduced at the 3 'UTR of the immune response-related gene only acts on the donor cells, without interfering with the genome of the recipient's cells.
Further, the small nucleic acid molecule includes short interfering nucleic acid, short interfering RNA, and double-stranded RNA, and preferably at least one of miRNA, shRNA, and shRNA-miR.
Further, the pluripotent stem cell or the derivative thereof is derived from a human; the sequence of the small nucleic acid molecule is a random sequence of a non-human species that does not target any mRNA or incrna of a human.
The small nucleic acid molecule is preferably derived from caenorhabditis elegans. For example:
5’-TTGTACTACACAAAAGTACTG-3’(SEQ ID NO.102)
5’-TCACAACCTCCTAGAAAGAGTAGA-3’(SEQ ID NO.103)。
further, the second nucleic acid molecule comprises a reverse complement of at least 3 repeated small nucleic acid molecule sequences, preferably 6-10 repeated small nucleic acid molecule sequences.
Furthermore, an inducible gene expression system is introduced into the genome of the pluripotent stem cell or the derivative thereof for regulating the expression of the first nucleic acid molecule.
In the technical scheme, the inducible gene expression system is regulated and controlled by an exogenous inducer, and the opening and closing of the inducible gene expression system are controlled by adjusting the addition amount, the duration action time and the type of the exogenous inducer, so that the expression quantity of small nucleic acid molecules is controlled.
When the inducible gene expression system is started, the transcription product of the small nucleic acid molecule which is normally expressed and the second nucleic acid molecule which is introduced into the 3' UTR region of the immune response related gene is specifically combined, so that an RNA interference program is started, mRNA of the immune response related gene is degraded or silenced, and the expression of the immune response related gene is blocked. Thus, when such cells or derivatives are transplanted into a recipient, the allogeneic immune rejection response may be eliminated or reduced, and the immune compatibility between the transplant and the recipient may be improved.
When the graft suffers from pathological changes, the inducible gene expression system can be closed by adding an exogenous inducer, so that the expression of small nucleic acid molecules is closed, the interference effect of the small nucleic acid on mRNA of the immune response related gene is stopped, the normal expression of the immune response related gene is recovered, the antigen presenting capability of graft cells is recovered, and the receptor can remove the pathological graft, so that the clinical safety of the pluripotent stem cells or the derivatives thereof is improved, and the value of the pluripotent stem cells in clinical application is greatly expanded.
Moreover, the RNA interference program only acts on such engineered pluripotent stem cells or derivatives thereof, and thus, when such cells or derivatives are transplanted into a recipient, RNA interference of the immune response-related gene mediated by the small nucleic acid molecule encoded by the first nucleic acid molecule and the second nucleic acid molecule introduced at the 3 'UTR of the immune response-related gene only acts on the donor cells, without interfering with the genome of the recipient's cells.
In a fourth aspect of the present invention, there is provided:
a pluripotent stem cell or a derivative thereof, wherein a genome of the pluripotent stem cell or the derivative thereof has an expression sequence of a CD47 inhibitory factor introduced therein; the CD47 inhibitor is at least one of shRNA and/or shRNA-miR of targeting CD 47.
The genome of the pluripotent stem cell or the derivative thereof is also introduced with an expression sequence of at least one immune compatible molecule for regulating the expression of genes related to immune response in the pluripotent stem cell or the derivative thereof.
In the technical scheme, the immune compatible molecules can regulate and control the expression of genes related to immune response in the pluripotent stem cells or the derivatives thereof, so that the pluripotent stem cells or the derivatives thereof have low immunogenicity, and can eliminate or reduce allogeneic immune rejection response and improve the immune compatibility between a graft and a recipient when the pluripotent stem cells or the derivatives thereof are transplanted into the recipient. The transplant can continuously express shRNA/shRNA-miR of a target CD47 in a receptor endogenously, after the inhibition factors are wrapped by an exosome, the exosome carries the inhibition factors to be combined with a target cell and then releases the target cell, so that a CD47 pathway is blocked, immunosuppression is relieved, an immune system is activated, NK cell activity is recovered, and tumor cells can be effectively eliminated.
Furthermore, an inducible gene expression system is also introduced into the genome of the pluripotent stem cell or the derivative thereof and is used for regulating and controlling the expression of the immune compatible molecules.
In the technical scheme, the inducible gene expression system is regulated by an exogenous inducer, the on and off of the inducible gene expression system are controlled by adjusting the addition amount, the duration action time and the type of the exogenous inducer, and the expression quantity of an immune compatible molecular expression sequence is further controlled, so that the reversible regulation of the immune compatibility of the pluripotent stem cells or derivatives thereof is realized. When the immune compatible molecule is normally expressed, the expression of genes related to immune response in the pluripotent stem cell or the derivative thereof is inhibited or overexpressed, so that when allogeneic cell therapy is performed, allogeneic immune rejection response can be eliminated or reduced, and the immune compatibility between the donor cell and the recipient can be improved. When the donor cell is diseased, the expression of the immune compatible molecules can be closed by induction of an exogenous inducer, so that the HLA class I molecules can be reversibly re-expressed on the surface of the donor cell, the antigen presenting capability of the donor cell is recovered, and then the receptor immune system enables the receptor to eliminate the diseased cell by identifying unmatched HLA class I molecules or by cross HLA class I molecule antigen presenting (antigen presenting/identifying between classical incompatible HLA) mutated molecules, so that the clinical safety of the general pluripotent stem cell or the derivative thereof is improved, and the value of the general pluripotent stem cell in clinical application is greatly expanded.
With respect to the third and fourth aspects of the present invention, further, the genes associated with immune response include:
(1) major histocompatibility complex genes including at least one of HLA-A, HLA-B, HLA-C, HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1 and HLA-DPB 1;
(2) major histocompatibility complex-associated genes including at least one of B2M and CIITA.
Still further, the immune-compatible molecule comprises at least one of:
(1) immune tolerance-related genes, including HLA-G;
(2) HLA-C molecules, including HLA-C multiple alleles of which the proportion in the population is over 90 percent in total, or fusion protein genes consisting of the HLA-C multiple alleles of which the proportion is over 90 percent and B2M;
(3) shRNA and/or shRNA-miR targeting a major histocompatibility complex gene including at least one of HLA-A, HLA-B, HLA-C, HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1 and HLA-DPB 1;
(4) an shRNA and/or shRNA-miR targeting a major histocompatibility complex-associated gene comprising at least one of B2M and CIITA.
Furthermore, the target sequence of the shRNA and/or shRNA-miR targeting B2M is selected from one of SEQ ID NO. 10-SEQ ID NO. 12;
the target sequence of the shRNA and/or shRNA-miR of the targeting CIITA is selected from one of SEQ ID NO. 13-SEQ ID NO. 15;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-A is selected from one of SEQ ID NO. 16-SEQ ID NO. 18;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-B is selected from one of SEQ ID NO. 19-SEQ ID NO. 21;
the target sequence of the shRNA and/or shRNA-miR targeting the HLA-C is selected from one of SEQ ID NO. 22-SEQ ID NO. 24;
the target sequence of the shRNA and/or shRNA-miR targeting the HLA-DRA is selected from one of SEQ ID NO. 25-27;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB1 is selected from one of SEQ ID NO. 28-SEQ ID NO. 30;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB3 is selected from one of SEQ ID NO. 31-SEQ ID NO. 32;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB4 is selected from one of SEQ ID NO. 33-SEQ ID NO. 35;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB5 is selected from one of SEQ ID NO. 36-SEQ ID NO. 38;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DQA1 is selected from one of SEQ ID NO. 39-SEQ ID NO. 41;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DQB1 is selected from one of SEQ ID NO. 42-SEQ ID NO. 44;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DPA1 is selected from one of SEQ ID NO. 45-SEQ ID NO. 47;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DPB1 is selected from one of SEQ ID NO. 48-SEQ ID NO. 51.
In the first to fourth aspects of the present invention, further, the genome of the pluripotent stem cell or the derivative thereof is further introduced with shRNA and/or miRNA processing complex-associated gene and/or anti-interferon effector molecule, wherein: the shRNA and/or miRNA processing complex related gene comprises at least one of Drosha, Ago1, Ago2, Dicer1, Exportin-5, TRBP (TARBP2), PACT (PRKRA) and DGCR 8; the anti-interferon effector molecule is shRNA and/or shRNA-miR of at least one of target PKR, 2-5As, IRF-3 and IRF-7.
Furthermore, the target sequence of the shRNA and/or shRNA-miR targeting the PKR is selected from one of SEQ ID NO. 51-SEQ ID NO. 53;
the target sequence of the shRNA and/or shRNA-miR targeting 2-5As is selected from one of SEQ ID NO. 54-SEQ ID NO. 62;
the target sequence of the shRNA and/or shRNA-miR of the target IRF-3 is selected from one of SEQ ID NO. 63-SEQ ID NO. 65;
the target sequence of the shRNA and/or shRNA-miR of the target IRF-7 is selected from one of SEQ ID NO. 66-SEQ ID NO. 68.
With regard to the third and fourth aspects of the present invention, the inducible gene expression system includes at least one of a Tet-Off system, a dimer inducible expression system.
When the inducible gene expression system employed is the Tet-Off system, the expression of small nucleic acid molecules in the cell or derivative thereof can be controlled by the addition of the exogenous inducer tetracycline (Doxycyline, Dox). After the pluripotent stem cells or the derivatives thereof are transplanted into a donor, the expression level of the small nucleic acid molecules can be gradually reduced even by adjusting the addition amount of Dox, so that the cells can gradually express low-concentration immune-related genes to stimulate the donor, and the donor can gradually generate tolerance to the transplanted cells or the derivatives thereof, and finally stable tolerance is achieved. In general, Dox is added in an amount of 0 to 100 uM.
In the first to fourth aspects of the present invention, further, the genome of the pluripotent stem cell or the derivative thereof further introduces an exosome-processing synthetic gene comprising at least one of STEAP3, Syndevan-4, an L-aspartate oxidase fragment, CD63-L7Ae and Cx43S 368A.
The secretion efficiency of the exosome and the encapsulation efficiency of the exosome on shRNA, shRNA-miR and mature siRNA can be improved by introducing the exosome processing synthetic gene into the genome of the stem cell or the stem cell derivative.
The dimer inducible expression system specifically refers to: dimerized inducers or dimers are used to recombine active transcription factors on inactive fusion proteins. The most commonly used system is rapamycin (rapamydn), a natural product, or an analog that is biologically inactive, as the drug for dimerization. The rapamycin (or analog) sibling protein FKBP12 (the protein to which FKBP binds to FK 506) and a large serine-threonine protein kinase, known as FRAP (FRBP-rapamycin associated protein, mTOR (mammalian target of rapamycin)), have high affinity and function to bind to both proteins, thus bringing them together as a heterologous dimer. To regulate transcription of a target gene, a DNA binding domain is fused to one or more FKBP domains and a transcription repressing domain is fused to amino acid position 93 of FRAP, designated FRB, which is sufficient to bind the FKBP-rapamycin complex. Dimerization of these two fusion proteins can only occur in the presence of rapamycin. Thus inhibiting transcription of genes having sites that bind to the DNA binding region.
In the first to fourth aspects of the present invention, further, the expression frameworks of the shRNA and/or shRNA-miR targeting CD47, major histocompatibility complex gene, major histocompatibility complex-related gene, and anti-interferon effector molecule are as follows:
the shRNA expression framework is as follows: the gene sequence sequentially comprises an shRNA sequence, a stem-loop sequence, a reverse complementary sequence of the shRNA sequence and Poly T from 5 'to 3';
wherein the shRNA sequence, the stem-loop sequence and the reverse complementary sequence of the shRNA sequence form a hairpin structure; poly T is a transcription terminator of RNA polymerase III;
shRNA-miR expression framework: and replacing the shRNA target sequence in the shRNA expression frame by using a shRNA-miR sequence.
In the first to fourth aspects of the present invention, further, the first nucleic acid molecule, the expression sequence of CD47 repressor, the expression sequence of an immune-compatible molecule, the shRNA and/or miRNA processing complex-related gene, the anti-interferon effector molecule, the inducible gene expression system, the introduction of the exosome processing synthetic gene are introduced by a method of viral vector interference, non-viral vector transfection or gene editing including gene knock-in.
In the first to fourth aspects of the present invention, further, the introduction site of the expression sequence of CD47 suppressor, the exosome processing synthetic gene, the expression sequence of immune-compatible molecule, shRNA and/or miRNA processing complex-related gene, anti-interferon effector molecule, inducible gene expression system is a genome safety site. Preferably one or more of AAVS1 safety site, eGSH safety site and H11 safety site.
With respect to the first to fourth aspects of the present invention, further, the pluripotent stem cells include embryonic stem cells, embryonic germ cells, embryonic carcinoma cells, or induced pluripotent stem cells; the pluripotent stem cell derivative comprises an adult stem cell, each germ layer cell or a tissue or organ into which the pluripotent stem cell is differentiated; the adult stem cells include mesenchymal stem cells or neural stem cells.
In a fifth aspect of the present invention, there is provided:
the application of the pluripotent stem cells or the derivatives thereof and the exosomes secreted by the pluripotent stem cells in preparing anti-tumor therapeutic preparations or medicaments.
Further, the application of the pluripotent stem cells or the derivatives thereof and the secreted exosomes thereof in preparing CD47 high-expression tumor treatment preparations or medicines.
In a sixth aspect of the present invention, there is provided:
an exosome secreted by the pluripotent stem cell or a derivative thereof.
The invention has the beneficial effects that:
1. the pluripotent stem cells or the derivatives thereof provided by the first aspect of the invention can be applied to autologous cell-induced iPSCs or MSCs low-immunogenicity cells. After an expression sequence of shRNA/shRNA-miR of a CD47 inhibition factor targeting CD47 is introduced into an iPSCs genome induced by autologous cells, the iPSCs can express a large amount of shRNA/shRNA-miR of a targeting CD47 and are wrapped by exosomes secreted by the cells. Exosomes carry these inhibitory factors to bind to target cells and then release them, thereby blocking the CD47 pathway, relieving immunosuppression, activating the immune system, and restoring NK cell activity, enabling them to effectively eliminate tumor cells.
2. The pluripotent stem cells or derivatives thereof provided by the second aspect of the invention may also be used in allogeneic cell therapy. Because the B2M and CIITA genes in the pluripotent stem cells or the derivatives thereof are knocked out, the pluripotent stem cells or the derivatives thereof have low immunogenicity, and when the pluripotent stem cells or the derivatives thereof are transplanted into a recipient, RNA interference aiming at immune response related genes, mediated by transcription products of a small nucleic acid molecule coded by a first nucleic acid molecule and a second nucleic acid molecule, only acts on donor cells and does not interfere with the genome of cells of the recipient. Improving the immune compatibility between the graft and the recipient. The transplant can continuously express shRNA/shRNA-miR of a target CD47 in a receptor endogenous source, after the inhibition factors are wrapped by an exosome, the exosome carries the inhibition factors to be combined with a target cell, and then the inhibition factors are released, so that a CD47 pathway is blocked, immunosuppression is relieved, an immune system is activated, NK cell activity is recovered, and the tumor cell can be effectively eliminated.
3. The pluripotent stem cells or derivatives thereof provided by the third aspect of the invention have immune compatibility, and can eliminate or reduce allogeneic immune rejection response. Furthermore, the RNA interference program of the pluripotent stem cell or the derivative thereof acts only on such an engineered pluripotent stem cell or the derivative thereof. Thus, when such cells or derivatives are transplanted into a recipient, RNA interference directed to a gene associated with an immune response mediated by the transcript of the small nucleic acid molecule encoded by the first nucleic acid molecule and the second nucleic acid molecule acts only on the donor cell and does not interfere with the genome of the recipient cell.
Furthermore, an inducible gene expression system is introduced into the genome of the pluripotent stem cell or the derivative thereof, and is used for regulating the expression of the first nucleic acid molecule, so that the immune compatibility and reversibility of the pluripotent stem cell or the derivative thereof are realized.
4. The pluripotent stem cell or the derivative thereof provided by the fourth aspect of the invention has an immune-compatible molecule expression sequence introduced into its genome, so that the pluripotent stem cell or the derivative thereof has low immunogenicity, and when the pluripotent stem cell or the derivative thereof is transplanted into a recipient, RNA interference directed to an immune response-related gene mediated by a transcription product of a small nucleic acid molecule encoded by a first nucleic acid molecule and a second nucleic acid molecule only acts on a donor cell, and does not interfere with the genome of the recipient cell. Improving the immune compatibility between the graft and the recipient. The transplant can continuously express shRNA/shRNA-miR of a target CD47 in a receptor endogenous source, after the inhibition factors are wrapped by an exosome, the exosome carries the inhibition factors to be combined with a target cell, and then the inhibition factors are released, so that a CD47 pathway is blocked, immunosuppression is relieved, an immune system is activated, NK cell activity is recovered, and the tumor cell can be effectively eliminated.
Furthermore, the genome of the pluripotent stem cell or the derivative thereof provided by the fourth aspect of the invention is also introduced with an inducible gene expression system, the inducible gene expression system can regulate and control the expression of the immune compatible molecules, and the inducible gene expression system is regulated and controlled by an exogenous inducer, so that the on and off of the inducible gene expression system are controlled by adjusting the addition amount, the sustained action time and the type of the exogenous inducer, and the expression quantity of the expression sequence of the immune compatible molecules is further controlled, thereby realizing the reversible regulation and control of the immune compatibility of the pluripotent stem cell or the derivative thereof. When the inducible gene expression system is started, the transcription product of the small molecule nucleic acid which is normally expressed and a second nucleic acid molecule which is introduced into the 3' UTR region of the immune response related gene is specifically combined, so that an RNA interference program is started, mRNA of the immune response related gene is degraded or silenced, and the expression of the immune response related gene is blocked. Thus, when such cells or derivatives are transplanted into a recipient, the allogeneic immune rejection response may be eliminated or reduced, and the immune compatibility between the transplant and the recipient may be improved. When the graft suffers from pathological changes, the inducible gene expression system can be closed by adding an exogenous inducer, so that the expression of small nucleic acid molecules and the interference effect of the small nucleic acid on mRNA of the immune response related gene are stopped, the normal expression of the immune related gene is recovered, the antigen presenting capability of graft cells is further recovered, and the receptor can remove the pathological graft, thereby improving the clinical safety of the pluripotent stem cells or the derivatives thereof, and greatly expanding the value of the pluripotent stem cells or the derivatives thereof in clinical application.
5. The pluripotent stem cells or the derivatives thereof can gradually reduce the expression amount of small nucleic acid molecules in the pluripotent stem cells or the derivatives thereof by adjusting the addition amount and the sustained action time of the exogenous inducer, so that the donor cells can gradually express low-concentration immune-related genes to stimulate the donor, and the donor can gradually generate tolerance to transplanted cells or the derivatives thereof, and finally stable tolerance is achieved. In this case, even though mismatched HLA class I molecules are expressed on the surface of the graft cells, they are compatible with the recipient immune system.
Drawings
Figure 1 is a plasmid map of Cas9 (D10A).
FIG. 2 is a plasmid map of sgRNA Clone AAVS 1-1.
FIG. 3 is a plasmid map of sgRNA Clone AAVS 1-2.
FIG. 4 is a plasmid map of sgRNA clone B2M-1.
FIG. 5 is a plasmid map of sgRNA clone B2M-2.
FIG. 6 is a plasmid map of sgRNA clone B2M-3.
FIG. 7 is a plasmid map of sgRNA clone B2M-4.
FIG. 8 is a plasmid map of sgRNA clone CIITA-1.
FIG. 9 is a plasmid map of sgRNA clone CIITA-2.
FIG. 10 is a plasmid map of sgRNA clone CIITA-3.
FIG. 11 is a plasmid map of sgRNA clone CIITA-4.
FIG. 12 is a plasmid map of AAVS1KI Vector (shRNA, constitutive).
FIG. 13 is a plasmid map of AAVS1KI Vector (shRNA, inducible).
FIG. 14 is a plasmid map of AAVS1KI Vector (shRNA-miR, constitutive).
FIG. 15 is a plasmid map of AAVS1KI Vector (shRNA-miR, inducible).
FIG. 16 is a plasmid map of B2M KI Vector.
FIG. 17 is a plasmid map of CIITA KI Vector.
Detailed Description
In order to make the objects, technical solutions and technical effects of the present invention more clear, the present invention will be described in further detail with reference to specific embodiments. It should be understood that the detailed description and specific examples, while indicating the preferred embodiment of the invention, are intended for purposes of illustration only and are not intended to limit the scope of the invention.
The experimental materials and reagents used are, unless otherwise specified, all consumables and reagents which are conventionally available from commercial sources.
Experimental materials:
selection of CD47 inhibitors
CD47 inhibitor: shRNA or shRNA-miR targeting CD 47.
The sequence of the shRNA or shRNA-miR sequence targeting CD47 used in the embodiment of the invention is shown in Table 1.
TABLE 1 sequences of shRNA or shRNA-miR sequences targeting CD47
Figure BDA0002848707890000081
2. Construction of Small nucleic acid molecules
The sequence of the small nucleic acid molecule is a random sequence of a non-human species that does not target any mRNA or incrna of a human, preferably derived from caenorhabditis elegans.
The sequence of the small nucleic acid molecules used in this example was:
5’-TTGTACTACACAAAAGTACTG-3’(SEQ ID NO.102);
designing a first nucleic acid molecule and a second nucleic acid molecule according to the small nucleic acid molecules, wherein the first nucleic acid molecule and the second nucleic acid molecule are respectively as follows:
a first nucleic acid molecule (i.e., the shRNA expression framework or shRNA-miR expression framework of a small nucleic acid molecule):
(1) the sequence composition of the shRNA expression framework of the small nucleic acid molecule is as follows:
5’-CCACTCCCTATCAGTGATAGAGAAAAGTGAAAGTCGAGTTTACCACTCCCTATCAGTGATAGAGAAAAGTGAAAGTCGAGTTTACCACTCCCTATCAGTGATAGAGAAAAGTGAAAGTCGAGTTTACCACTCCCTATCAGTGATAGAGAAAAGTGAAAGTCGAGCTCGGTACCCGGGTCGAGGTAGGCGTGTACGGTGGGAGGCCTATATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCGCCTGGAGACGCCATCCACGCTGTTTTGACCTCCATAGAAGACACCGGGACCGATCCAGCCTGCTAGCGCCACC(SEQ ID NO.4)N1...N21TTCAAGAGA(SEQ ID NO.5)N22...N42TTTTTT-3’。
wherein the content of the first and second substances,
a)N1...N21is the sequence of the small nucleic acid molecule, N22...N42The reverse complementary sequence of the small nucleic acid molecule sequence;
b) if the plasmid needs to express shRNA of a plurality of genes, each gene corresponds to a shRNA expression frame and then is connected seamlessly;
c) constitutive shRNA plasmids with different resistance genes only have different resistance genes and have the same other sequences;
d) n represents A or T or G or C base.
(2) shRNA-miR expression framework of small nucleic acid molecules: the small nucleic acid molecule sequence is used for replacing a target sequence in microRNA-30 or microRNA-155 to obtain the target sequence. The specific sequence is as follows:
5’-GAGGCTTCAGTACTTTACAGAATCGTTGCCTGCACATCTTGGAAACACTTGCTGGGATTACTTCTTCAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTGACAGTGAGCG(SEQ ID NO.6)M1N1...N21TAGTGAAGCCACAGATGTA(SEQ ID NO.7)N22...N42M2TGCCTACTGCCTCGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTACTAAAACTGAATACCTTGCTATCTCTTTGATACATTTTTACAAAGCTGAATTAAAATGGTATAAAT-3’(SEQ ID NO.8)。
wherein the content of the first and second substances,
a)N1...N21is a small nucleic acid molecule sequence, N22...N42Is a reverse complementary sequence of a small nucleic acid molecule sequence;
b) if the plasmid needs to express shRNA-miR of a plurality of genes, each gene corresponds to a shRNA-miR expression frame and is then connected seamlessly;
c) constitutive shRNA-miR plasmids with different resistance genes only have different resistance genes and have the same other sequences;
d) n represents A or T or G or C base, M base represents A or C base;
e) if N is present1Is a G base, then M1Is A base; otherwise M1Is a C base;
f)M1base and M2And (3) base complementation.
A second nucleic acid molecule: comprises a reverse complementary sequence of at least 3 repeated small nucleic acid molecule sequences, preferably a reverse complementary sequence of 6-10 repeated small nucleic acid molecule sequences. The reverse complement of the small nucleic acid molecule sequence can be linked by a random Linker sequence.
As an embodiment of the invention, the second nucleic acid molecule is formed by connecting the random sequence of the first 10nt and the reverse complement of the sequence of 8 repeated small nucleic acid molecules through a random linker sequence (CGTA):
5’-atTCTAGATACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTACAGTACTTTTGTGTAGTACAACGTA-3’(SEQ ID NO.9)。
3. selection of immune compatible molecules
The types and sequences of the immune-compatible molecules used in the examples of the present invention are shown in tables 2 and 3.
TABLE 2 types and roles of immune-compatible molecules
Figure BDA0002848707890000091
Figure BDA0002848707890000101
TABLE 3 target sequences for immune-compatible molecules
Figure BDA0002848707890000102
Figure BDA0002848707890000111
Figure BDA0002848707890000121
Figure BDA0002848707890000131
shRNA or shRNA-miR immune compatible molecule sequences of the experimental groups in the following tables 8-9 are shRNA or shRNA-miR immune compatible molecules constructed by adopting target sequences in the table 3. Those skilled in the art will understand that: the shRNA or shRNA-miR immune compatible molecule constructed by other target sequences can also realize the technical effect of the invention and all fall into the protection scope of the claims of the invention.
Selection of shRNA/miRNA processing Complex genes and anti-Interferon Effector molecules
(1) selection of shRNA/miRNA processing Complex genes
shRNA/miRNA processing complex genes used in the present invention include shRNA and/or miRNA processing machinery that can induce off-expression. The shRNA and/or miRNA processor capable of inducing closed expression specifically comprises: drosha (Access number: NM-001100412), Ago1(Access number: NM-012199), Ago2(Access number: NM-001164623), Dicer1(Access number: NM-001195573), export-5 (Access number: NM-020750), TRBP (Access number: NM-134323), PACT (Access number: NM-003690) and DGCR8(Access number: NM-022720).
(2) Selection of anti-Interferon Effector molecules
The anti-interferon effector molecules used in the invention comprise shRNA and/or shRNA-miR expression sequences which can induce closed expression and aim at inhibiting PKR, 2-5As, IRF-3 and IRF-7 genes so As to reduce the interferon reaction induced by dsRNA and avoid generating cytotoxicity.
Wherein the target sequences of the anti-interferon effector molecules are shown in Table 4.
TABLE 4 target sequences for anti-interferon effector molecules
Figure BDA0002848707890000132
Figure BDA0002848707890000141
The anti-interferon effector molecule sequences of the experimental groups in the following tables 8-9 are all shRNA or shRNA-miR constructed by adopting the target sequences in the table 4. Those skilled in the art will understand that: the technical effect of the invention can be realized by shRNA or shRNA-miR anti-interferon effector molecules constructed by other target sequences, and the shRNA or shRNA-miR anti-interferon effector molecules fall into the protection scope of the claims of the invention.
5. Selection of exosome processing synthetic genes
The exosome processing synthetic gene is at least one selected from STEAP3(NM _182915), Syndecano-4 (NM _002999), L-aspartate oxidase fragment (SEQ ID NO.69), CD63-L7Ae (SEQ ID NO.70) and Cx43S 368A. Wherein Cx43S368A is obtained by mutating S (serine) to A (alanine) at position 368 of Cx43 (NM-000165).
6. Selection of Stem cell vectors
The stem cell carrier in the embodiment of the invention is a pluripotent stem cell, and the pluripotent stem cell can be selected from Embryonic Stem Cells (ESCs), Induced Pluripotent Stem Cells (iPSCs) and other forms of pluripotent stem cells, such as hPSCs-MSCs, NSCs and EBs cells.
Wherein the preparation method of the pluripotent stem cells comprises the following steps:
ESCs: HN4 cells were selected and purchased from Shanghai department of sciences.
And (3) iPSCs: using an episomal-iPSCs induction system (6F/BM1-4C), pE3.1-OG- -KS and pE3.1-L-Myc- -hmiR302cluster were electrotransferred into somatic cells, RM1 medium was cultured for 2 days, 2 days with 2uM Parnate-containing BioCISO-BM1 medium, 2 days with 2uM Parnate, 0.25mM sodium butyrate, 3uM CHIR99021 and 0.5uM PD 03254901-containing BioCISO-BM1 medium, and then cultured continuously with BioCISO medium without other substances for about 17 days, iPSCs clonal cells were picked, and the picked iPSCs clonal cells were purified, digested, and passaged to obtain stable iPSCs. The specific construction method is as follows: stem Cell Res ther.2017nov 2; 8(1):245.
hPSCs-MSCs: iPSCs were cultured for 25 days in BioCISO medium containing 10uM TGF β inhibitor SB431542 until 80-90 cell confluence (2mg/mL Dispase), 1:3 passaging was performed on Matrigel-coated plates, followed by ESC-MSC medium (knockout DMEM medium containing 10% KSR, NEAA, diabody, glutamine, β -mercaptoethanol, 10ng/mL bFGF and SB-431542), fluid was changed every day, and culture was continued for 20 days until 80-90 cell confluence (1:3 passaging), and specific construction methods were as follows: proc Natl Acad Sci U S A.2015; 112(2):530-535.
NSCs: iPSCs are cultured for 14 days in an induction medium (knockout DMEM medium containing 10% KSR and TGF-beta inhibitor and BMP4 inhibitor), and rose annular nerve cells are picked and cultured in a low-adhesion culture plate. The medium in the low adhesion plates included DMEM/F12 (with 1% N2, Invitrogen) and Neurobasal medium (with 2% B27, Invitrogen) in a ratio of 1:1, along with 20ng/ml bFGF and 20ng/ml EGF. Digestion passages were performed using Accutase. The specific construction method is as follows: FASEB J.2014; 28(11):4642-4656.
EBs cells: the iPSCs with cell confluency of 95% were digested with BioC-PDE1 for 6min, and the cells were scraped into clumps by mechanical scraping, and the clumps were settled. The settled cell pellet was transferred to a low adhesion culture plate and cultured for 7 days using BioCISO-EB1, with fluid changes every other day. After 7 days, the cells were transferred to a Matrigel-coated plate and subjected to adherent culture using a BioCISO medium for 7 days, thereby obtaining Embryoid Bodies (EBs) having an inner, middle and outer mesoderm structure. The specific construction method is as follows: stem Cell Res ther.2017nov 2; 8(1):245.
The pluripotent stem cell derivative also includes adult stem cells, each germ layer cell or tissue, organ into which the pluripotent stem cells are differentiated; the adult stem cells include mesenchymal stem cells or neural stem cells.
Constructing a plasmid vector:
plasmid vectors used in the examples of the present invention include: cas9(D10A) plasmid, sgRNA plasmid, Donor fragment.
The genes in the plasmids are edited by a CRISPR-Cas9 gene editing system, the used Cas9 protein is Cas9(D10A), Cas9(D10A) is combined with sgRNA, the sgRNA is responsible for specifically recognizing a target sequence (genome DNA of a cell vector), and then the Cas9(D10A) performs single-strand cleavage on the target sequence. Double Strand breaks in genomic DNA (DSB) must occur, and two Cas9(D10A)/sgRNA groups must cleave both strands of genomic DNA of the cell vector separately, and not too far apart. The Cas9(D10A)/sgRNA scheme has the advantage of higher specificity and lower probability of off-target compared to the Cas 9/sgRNA scheme.
Wherein, the specific sequence of the sgRNA is as follows:
sgRNA-AAVS1-1:5’-TATAAGGTGGTCCCAGCTCG-3’(SEQ ID NO.71);
sgRNA-AAVS1-2:5’-AGGGCCGGTTAATGTGGCTC-3’(SEQ ID NO.72);
sgRNA-B2M-1:5’-CTCCTGTTATATTCTAGAAC-3’(SEQ ID NO.73);
sgRNA-B2M-2:5’-TTTCAGCATCAATGTACCCT-3’(SEQ ID NO.74);
sgRNA-B2M-3:5’-CGCGAGCACAGCTAAGGCCA-3’(SEQ ID NO.75);
sgRNA-B2M-4:5’–ACTCTCTCTTTCTGGCCTGG-3’(SEQ ID NO.76);
sgRNA-CIITA-1:5’-GGCACTCAGAAGACACTGAT-3’(SEQ ID NO.77);
sgRNA-CIITA-2:5’–AAGGTGTCTGGTCGGAGAGC-3’(SEQ ID NO.78);
sgRNA-CIITA-3:5’–ACCCAGCAGGGCGTGGAGCC-3’(SEQ ID NO.79);
sgRNA-CIITA-4:5’–GTCAGAGCCCCAAGGTAAAA-3’(SEQ ID NO.80)。
the specific method for constructing the plasmid vector comprises the following steps:
(1) cas9(D10A) plasmid: obtained directly from an Addgene (plasma 41816, Addgene) subscription.
(2) sgRNA plasmid: the Plasmid was constructed by putting different target sequences into original blank plasmids (Plasmid 41824, Addgene).
Wherein the target sequence put into the sgRNA plasmid comprises the sequence of the immune compatible molecule, the sequence of shRNA/miRNA processing complex gene, the sequence of anti-interferon effector molecule and the sequence of exosome processing synthetic gene.
(3) Donor fragment (KI plasmid):
a) designing a PCR primer, and amplifying by using a pUC18 plasmid (Takara, Code No.3218) as a template and a high fidelity enzyme (Nanjing Nozan organism, P505-d1) through a PCR method to obtain an Amp (R) -pUC origin fragment;
b) extracting genome DNA of human cells and designing corresponding primers, and then amplifying by using high-fidelity enzyme through a PCR method by taking the genome DNA of human as a template to obtain a recombinant arm;
c) designing PCR amplification primers of KI (Knock-in) plasmid elements, and then carrying out high-fidelity enzyme amplification by using plasmids containing the KI plasmid elements as templates to obtain KI plasmid elements (subclones);
d) the amplified Amp (R) -pUC origin fragment, recombination arm and KI plasmid components are connected by using multi-fragment recombinase (Nanjing Novozam organism, C113-02) or overlap PCR to form a complete circular plasmid.
Wherein the recombination arms comprise a B2M recombination arm, a CIITA recombination arm and an AAVS1 recombination arm.
The sequence of the B2M recombinant arm is shown as B2M-HR-L (SEQ ID NO.81) and B2M-HR-R (SEQ ID NO. 82).
The sequence of the CIITA recombination arm is shown as CIITA-HR-L (SEQ ID NO.83) and CIITA-HR-R (SEQ ID NO. 84).
The sequence of the AAVS1 recombination arm is shown as AAVS1-HR-L (SEQ ID NO.85) and AAVS1-HR-R (SEQ ID NO. 86).
The plasmids prepared by the embodiment of the invention can be divided into constitutive plasmids and inducible plasmids according to the expression frame type in the plasmids.
The expression frame in the constitutive plasmid comprises shRNA constitutive expression frame and shRNAMIR constitutive expression frame. The expression function of the Donor fragment obtained by enzyme digestion of the constitutive plasmid cannot be regulated after knocking in the stem cell vector genome DNA.
The expression frames in the inducible plasmid comprise shRNA inducible expression frames and shRNAMIR inducible expression frames. After the Donor fragment obtained from the inducible plasmid is digested by enzyme and the stem cell vector genome DNA is knocked in, the expression function of the fragment can be regulated and controlled by adding an inducer, which is equivalent to adding a switch for turning on or off the expression function.
The specific sequence requirements and structural composition of the expression frameworks described above are as follows.
(1) The sequence composition of the shRNA constitutive expression framework is:
5’-GAGGGCCTATTTCCCATGATTCCTTCATATTTGCATATACGATACAAGGCTGTTAGAGAGATAATTGGAATTAATTTGACTGTAAACACAAAGATATTAGTACAAAATACGTGACGTAGAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGTAACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACgctagcgccacc(SEQ ID NO.87)N1...N21TTCAAGAGAN22...N42TTTTTT-3’;
wherein the content of the first and second substances,
a)N1...N21shRNA target sequence which is the above-mentioned target sequence, N22...N42The reverse complementary sequence of the shRNA target sequence of the target sequence;
b) when plasmids constructed by using the shRNA constitutive expression frame need to express shRNAs of a plurality of genes, each gene corresponds to a shRNA expression frame respectively and then is connected seamlessly;
c) when the shRNA constitutive expression frame needs to carry different resistance genes, only the resistance gene sequences in the shRNA constitutive expression frame are different, and other sequences are the same;
d) n represents A or T or G or C base.
(2) The sequence composition of the shrmamir constitutive expression framework is:
5’-GAGGCTTCAGTACTTTACAGAATCGTTGCCTGCACATCTTGGAAACACTTGCTGGGATTACTTCTTCAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTGACAGTGAGCGM1N1...N21TAGTGAAGCCACAGATGTAN22...N42M2TGCCTACTGCCTCGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTACTAAAACTGAATACCTTGCTATCTCTTTGATACATTTTTACAAAGCTGAATTAAAATGGTATAAAT-3’;
wherein the content of the first and second substances,
g)N1...N21shRNAMIR target sequence, N, being a target sequence as described above22...N42The reverse complement of the shrnmanir target sequence of the above target sequence;
h) when a plasmid constructed by using the shRNAMIR constitutive expression framework needs to express shRNAMIR of a plurality of genes, each gene corresponds to one shRNAMIR expression framework respectively and then is connected seamlessly;
i) when the shRNAMIR constitutive expression framework needs to carry different resistance genes, only the resistance gene sequences in the shRNAMIR constitutive expression framework are different, and other sequences are the same;
j) n represents A or T or G or C base, M base represents A or C base;
k) if N is present1Is a G base, then M1Is A base; otherwise M1Is a C base;
l)M1base and M2And (3) base complementation.
(3) The sequence composition of the shRNA inducible expression framework is:
5’-GAGGGCCTATTTCCCATGATTCCTTCATATTTGCATATACGATACAAGGCTGTTAGAGAGATAATTGGAATTAATTTGACTGTAAACACAAAGATATTAGTACAAAATACGTGACGTAGAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGTAACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACtttaccactccctatcagtgatagagaaaagtgaaagtcgagtttaccactccctatcagtgatagagaaaagtgaaagtcgagtttaccactccctatcagtgatagagaaaagtgaaagtcgagtttaccactccctatcagtgatagagaaaagtgaaagtcgagtttaccactccctatcagtgatagagaaaagtgaaagtcgagtttaccactccctatcagtgatagagaaaagtgaaagtcgagtttaccactccctatcagtgatagagaaaagtgaaagtcgagctcggtacccgggtcgaggtaggcgtgtacggtgggaggcctatataagcagagctcgtttagtgaaccgtcagatcgcctggagacgccatccacgctgttttgacctccatagaagacaccgggaccgatccagcctgctagcgccacc(SEQ ID NO.88)N1...N21TTCAAGAGAN22...N42TTTTTT-3’;
wherein the content of the first and second substances,
e)N1...N21the shRNA target sequence corresponding to the target sequence, N22...N42The shRNA target sequence and/or small nucleic acid corresponding to the target sequenceThe reverse complement of the molecule;
f) when plasmids constructed by using the shRNA constitutive expression frame need to express shRNAs of a plurality of genes, each gene corresponds to a shRNA expression frame respectively and then is connected seamlessly;
g) when the constitutive expression framework of the shRNA needs to be provided with different resistance genes, only the resistance gene sequences in the constitutive expression framework of the shRNA are different, and other sequences are the same;
h) n represents A or T or G or C base.
(4) The sequence composition of the shrnmamir inducible expression framework is shown as that of the shrnmamir constitutive expression framework.
The plasmid map of Cas9(D10A) constructed by the method is shown in figure 1, the plasmid map of sgRNA of AAVS1 safety site is shown in figure 2-3, the plasmid map of sgRNA of B2M gene is shown in figure 4-7, the plasmid map of sgRNA of CIITA gene is shown in figure 8-11, the plasmid map (constitutive and inducible) of constructed AAVS1KI is shown in figure 12-15, the plasmid map of B2M KI is shown in figure 16, and the plasmid map of CIITA KI is shown in figure 17.
Construction of Stem cell vectors
In the technical scheme of the invention, the safety sites knocked in the genome of the constructed stem cell vector comprise an AAVS1 safety site, an eGSH safety site and an H11 safety site.
The single-cell cloning operation of the knock-in of the AAVS1 gene comprises the following steps:
a) electric transfer program:
donor cell preparation: human pluripotent stem cells
The kit comprises: human Stem Cell
Figure BDA0002848707890000181
Kit
1
The instrument comprises the following steps: electric rotating instrument
Culture medium: BioCISO
Induction of plasmid: the corresponding plasmids constructed in the above examples.
b) The transformed human pluripotent stem cells are screened in a double antibiotic medium containing G418 and puro.
c) And (4) carrying out single cell clone screening and culture to obtain a single cell clone strain.
d) The obtained single-cell clone was cultured.
Wherein, the culture reagent of the single cell clone strain comprises:
the culture medium is as follows: BioCISO medium, 300. mu.g/ml G418 and 0.5. mu.g/ml puro. The culture medium needs to be placed at room temperature in advance, placed for 30-60 minutes in a dark condition until the room temperature is recovered, but not placed at 37 ℃ for preheating, so that the activity of the biological molecules is prevented from being reduced.
Matrix glue: hESC grade Matrigel. Before passage or cell recovery, adding the Matrigel working solution into a cell culture bottle dish and shaking up to ensure that the Matrigel completely submerges the bottom of the culture bottle dish and any Matrigel cannot be dried off before use. To ensure that cells adhere better and survive, Matrigel was placed in a 37 ℃ incubator for a period of time: 1:100 Xmatrigel can not be less than 0.5 hour; the 1:200 Xmatrigel cannot be less than 2 hours.
Digestion solution: EDTA was dissolved using DPBS to a final concentration of 0.5mM, pH 7.4. The EDTA cannot be diluted with water, otherwise the cells die due to reduced osmotic pressure.
Freezing and storing liquid: 60% BioCISO, 30% ESCS grade FBS, and 10% DMSO.
The culture step of the single-cell clone adopts the conventional subculture maintaining process in the field.
Wherein, the optimal passage time is that the overall confluency of the cells reaches 80 to 90 percent.
The optimal ratio of passage is 1: 4-1: 7, and the optimal confluency of the next day after passage is maintained at 20-30%.
The specific passage operation steps in the invention are as follows:
a) discarding the Matrigel in the coated cell culture flask, adding appropriate amount of the culture medium, and placing at 37 deg.C and 5% CO2Incubation in an incubator;
b) when the cells meet the passage requirement, removing culture medium supernatant, and adding a proper amount of 0.5mM EDTA digestive juice into a cell bottle dish;
c) the cells were incubated at 37 ℃ with 5% CO2Incubating in an incubator for 5-10 minutes (digesting until most cells are observed to shrink and become round under a microscope but not float), blowing the cells to separate the cells from the wall, sucking the cell suspension into a centrifugal tube, and centrifuging for 5 minutes at 200 g;
d) centrifuging, discarding supernatant, suspending the cells with culture medium, repeatedly blowing the cells until uniformly mixing, transferring the cells to a petrigel-coated bottle dish, shaking, observing under a mirror without abnormality, shaking, and standing at 37 deg.C and 5% CO2Culturing in an incubator;
e) observing the adherent survival state of the cells the next day, sucking off the culture medium, and changing the culture medium on time every day.
The gene knock-in detection method comprises the following steps:
1. single cell clone AAVS1 gene knock-in assay.
(1) AAVS1 gene knock-in assays.
The detection principle is as follows:
the cells treated by knock-in were tested for homozygote by PCR. Since the two Donor fragments have only difference in the sequences of the resistance genes, it is necessary to determine whether the cell is homozygous (the two chromosomes knock in the Donor fragments of different resistance genes), and it is only possible that the double-knocked-in cell is the correct homozygous by determining whether the genome of the cell contains the Donor fragments of the two resistance genes.
The detection method comprises the following steps:
one primer was designed inside (non-recombinant arm portion) the Donor plasmid (KI plasmid), and then the other primer was designed in the genome of the cell (non-recombinant arm portion). If the Donor fragment is inserted correctly in the genome, the target band will appear, otherwise no target band will appear.
The specific sequences of the primers and amplification conditions are shown in the following table.
TABLE 5 detailed sequences of primers for detection of knock-in of AAVS1 Gene and amplification conditions
Figure BDA0002848707890000191
(2) B2M and CIITA gene knock-in test.
The detection method of the knock-in of the second nucleic acid molecule at the 3' UTR of B2M and CIITA gene was the same as the detection principle of AAVS1, and the PCR detection conditions used were as shown in tables 6 and 7:
TABLE 6 detailed sequences of knock-in detection primers for the B2M Gene and amplification conditions
Figure BDA0002848707890000192
TABLE 7 specific sequences of CIITA Gene knock-in detection primers and amplification conditions
Figure BDA0002848707890000193
Figure BDA0002848707890000201
Selected plasmids were knocked into the stem cell vector genome using techniques conventional in the art:
according to the grouping in the following tables 8 and 9, the sequence of the CD47 inhibitory factor, the sequence of the immune compatible molecule, the sequence of the shRNA/miRNA processing complex gene, the sequence of the anti-interferon effector molecule and the exosome processing synthetic gene are knocked into the safe locus of the stem cell vector by adopting the conventional technology in the field, so as to obtain different types of constitutive stem cell expression vectors and inducible stem cell expression vectors, and detect the expression feasibility of the constitutive stem cell expression vectors and the inducible stem cell expression vectors.
TABLE 8 constitutive knock-in expression assay grouping
Figure BDA0002848707890000202
Wherein a "+" symbol indicates a knock-in of a gene or nucleic acid sequence and a "-" symbol indicates a knock-out of a gene.
General principles are as follows: shRNA (or shRNA-miR) of the targeted inhibition factor is put into a shRNA (or shRNA-miR) expression frame 2 of the corresponding plasmid, shRNA (or shRNA-miR) of the other inhibition factors are put into a shRNA (or shRNA-miR) expression frame 1 of the corresponding plasmid, and the gene sequence is put into an MCS.
B2M-3 ' UTR-miRNA-focus or CIITA-3 ' UTR-miRNA-focus, i.e. the second nucleic acid molecule (SEQ ID No.9), knocks into the 3 ' UTR region of the B2M and CIITA genes, respectively.
The B2M/CIITA-3 'UTR-shRNA is an shRNA expression framework of a small nucleic acid molecule, namely a first nucleic acid molecule, specifically targets a transcription product of a second nucleic acid molecule in a B2M gene and a CIITA gene 3' UTR region, and a knock-in site is a genome safety site AAVS 1.
B2M/CIITA-3 'UTR-shRNA-miR is an shRNA-miR expression framework of a small nucleic acid molecule, namely a first nucleic acid molecule, a transcription product of a second nucleic acid molecule in a targeting B2M gene and CIITA gene 3' UTR region, and a knock-in site is a genome safety site AAVS 1.
If multiple fragments need to be inserted into the expression cassette or MCS, they can be ligated together using EMCV IRESWT (SEQ ID NO.101) and then inserted.
The gene knock-in sgRNA plasmid was: sgRNA clone B2M-1, sgRNA clone B2M-2, sgRNA clone CIITA-1 and sgRNA clone CIITA-2.
sgRNA plasmids used for gene knockout were: sgRNA clone B2M-3, sgRNA clone B2M-4, sgRNA clone CIITA-3 and sgRNA clone CIITA-4.
(1) Aa1 grouping:
shRNA expression frame 2 of AAVS1KI Vector (shRNA, constitutive) plasmid placed into the shRNA sequence targeting CD 47.
(2) Aa2 grouping:
shRNA expression frame 2 of AAVS1KI Vector (shRNA, constitutive) plasmid is placed into shRNA sequence of targeting CD47, shRNA expression frame 1 is placed into the rest shRNA sequence (including target sequence of B2M/CIITA-3' UTR-shRNA), and MCS is placed into gene sequence.
B2M KI Vector was placed in B2M-3' UTR-miRNA-focus.
CIITA KI Vector was placed into CIITA-3' UTR-miRNA-focus.
(3) Aa3 grouping:
and (3) putting an shRNA sequence targeting CD47 into an shRNA expression frame 2 of an AAVS1KI Vector (shRNA, constitutive) plasmid, putting the rest shRNA target sequences into an shRNA expression frame 1, knocking out B2M and CIITA, and putting MCS into a gene sequence.
(4) Aa4 grouping:
shRNA expression frame 2 of AAVS1KI Vector (shRNA, constitutive) plasmid is placed into shRNA sequence of target CD47, shRNA expression frame 1 is placed into other shRNA target sequence, and MCS is placed into gene sequence.
(5) Ab1 groups:
shRNA-miR expression frame 2 of AAVS1KI Vector (shRNA-miR, constitutive) plasmid is placed into a shRNA-miR sequence targeting CD 47.
(6) Ab2 groups:
shRNA-miR expression frame 2 of AAVS1KI Vector (shRNA-miR, constitutive) plasmid is placed into a shRNA-miR sequence targeting CD47, shRNA-miR expression frame 1 is placed into the rest shRNA-miR target sequence (including target sequences of B2M/CIITA-3' UTR-shRNA-miR), and MCS is placed into a gene sequence.
B2M KI Vector was placed in B2M-3' UTR-miRNA-focus.
CIITA KI Vector was placed into CIITA-3' UTR-miRNA-focus.
(7) Ab3 groups:
shRNA-miR sequence of targeting CD47 is placed in shRNA-miR expression frame 2 of AAVS1KI Vector (shRNA-miR, constitutive type) plasmid, the rest shRNA-miR target sequence is placed in shRNA-miR expression frame 1, B2M and CIITA are knocked out, and MCS is placed in gene sequence.
(8) Ab4 groups:
shRNA-miR expression frame 2 of AAVS1KI Vector (shRNA-miR, constitutive) plasmid is placed into shRNA-miR sequence of targeting CD47, shRNA-miR expression frame 1 is placed into other shRNA-miR target sequence, and MCS is placed into gene sequence.
TABLE 9 inducible knock-in expression experiment grouping
Figure BDA0002848707890000221
Wherein a "+" symbol indicates a knock-in of a gene or nucleic acid sequence and a "-" symbol indicates a knock-out of a gene.
The general principle is shown above in the constitutive knock-in expression experimental group.
(1) B1 grouping:
shRNA expression frame 2 of AAVS1KI Vector (shRNA, inducible) plasmid is placed into shRNA sequence of targeting CD47, shRNA expression frame 1 is placed into other shRNA target sequence (including B2M/CIITA-3' UTR-shRNA target sequence), and MCS is placed into gene sequence. Adding a Tet-Off system induction system.
B2M KI Vector was placed in B2M-3' UTR-miRNA-focus.
CIITA KI Vector was placed in CIITA-3' UTR-miRNA-locus.
(2) B2 grouping:
shRNA-miR expression frame 2 of AAVS1KI Vector (shRNA-miR, inducible) plasmid is placed into shRNA-miR sequence of targeting CD47, shRNA-miR expression frame 1 is placed into other shRNA-miR target sequence (including target sequence of B2M/CIITA-3' UTR-shRNA-miR), and MCS is placed into gene sequence. Adding a Tet-Off system induction system.
B2M KI Vector was placed in B2M-3' UTR-miRNA-focus.
CIITA KI Vector was placed in CIITA-3' UTR-miRNA-locus.
(3) B3 grouping:
shRNA expression frame 2 of AAVS1KI Vector (shRNA, inducible) plasmid is placed into shRNA sequence of targeting CD47, shRNA expression frame 1 is placed into the rest shRNA target sequence (including B2M/CIITA-3' UTR-shRNA target sequence), and MCS is placed into gene sequence. Adding a Tet-Off system induction system.
(4) B4 grouping:
shRNA-miR expression frame 2 of AAVS1KI Vector (shRNA-miR, inducible) plasmid is placed into shRNA-miR sequence of targeting CD47, shRNA-miR expression frame 1 is placed into other shRNA-miR target sequence (including target sequence of B2M/CIITA-3' UTR-shRNA-miR), and MCS is placed into gene sequence. Adding a Tet-Off system induction system.
Wherein, when the constitutive knock-in and the inducible knock-in are used for immune compatible reconstruction, the constitutive knock-in and the inducible knock-in can be firstly reconstructed on hPSCs and then differentiated into derivatives of pluripotent stem cells for application after the reconstruction is finished; the immune compatibility can also be modified after differentiation of hPSCs into derivatives of pluripotent stem cells.
The Tet-Off system in the invention specifically comprises the following steps:
in the absence of tetracycline, the tTA protein continues to act on the tet promoter, resulting in sustained gene expression. This system is very useful in situations where it is desirable to maintain the transgene in a sustained expression state. When tetracycline is added, the tetracycline can change the structure of the tTA protein, so that the tTA protein cannot be combined with a promoter, and the expression level of a gene driven by the tTA protein is reduced. To keep the system in an "off" state, the tetracycline must be added continuously.
The sequence of the Tet-Off system and one or more immune compatible molecules is knocked into a genome safety site of the pluripotent stem cell, and the expression of the immune compatible molecules is accurately turned on or Off through the addition of tetracycline, so that the expression of major histocompatibility complex related genes in the pluripotent stem cell or the derivative thereof can be reversibly regulated and controlled.
The inhibition effect of the pluripotent stem cells or the derivatives thereof expressing the targeted CD47 inhibitor on CD47 is detected:
the experimental group protocols in tables 8 and 9 were knocked into the genome safety sites of iPSCs, MSCs, NSCs, and EBs cells at 37 ℃ with 0.5% CO2Culturing in an incubator, collecting culture medium supernatant, and extracting exosome in the culture supernatant by using an exosome extraction kit (BestBio, lot # BB-3901) for cells containing a knock-in exosome processing synthetic gene sequence. Centrifuging culture supernatant at 37 deg.C for 15min at 3000g, collecting supernatant, centrifuging at 37 deg.C for 20min at 10000g, collecting supernatant, adding extractive solution A at a ratio of 4:1, reversing the upper and lower parts for 1min, and standing at 37 deg.C overnight. Centrifuging at 4 deg.C for 60min at 10000g, and collecting precipitate.
And testing the blocking effect of the CD47 inhibitor expressed by the pluripotent stem cells by using a flow cytometry method. Exosomes containing CD47 inhibitor were added to the CHO cell culture medium, and CD 47-expressing CHO cells were cultured for 72 h. After digesting into single cells and washing the cells 2 times with PBS, FITC-labeled CD47 antibody fusion protein was added to the tube and incubated at 37 ℃ for 30 minutes. Flow cytometric analysis was performed using a flow cytometer. According to the Mean Fluorescence Intensity (MFI) of the staining, the effect of the CD47 inhibitor expressed by the pluripotent stem cells on inhibiting the expression of CD47 by CHO cells can be measured.
The N (control) group refers to CD 47-expressing CHO cells cultured without addition of CD47 suppressor or CD47 suppressor-containing exosomes.
The results of the tests of the respective experimental groups are shown in Table 10.
TABLE 10 flow cytometric assay of CD47 expression results for CD47 expressing cells
Figure BDA0002848707890000231
Figure BDA0002848707890000241
Independent samples were tested for T (/ p < 0.01).
As can be seen from the above table, the exosome-encapsulated CD47 inhibitor expressed by the pluripotent stem cells or derivatives thereof of the present invention can effectively inhibit the expression of CD47 in target cells. And the inhibition degree is relatively constant in each group, so that the CD47 inhibitor expressed by the pluripotent stem cell derivative is not influenced by cell differentiation morphology and other exogenous genes (immune compatibility modification).
Detection of antitumor Effect of pluripotent Stem cells expressing a targeting CD47 inhibitor or derivatives thereof:
the experimental group schemes in tables 8 and 9 are knocked into genome safety loci of iPSCs, MSCs, NSCs and EBs cells to obtain the cell expressing the CD47 inhibitory factor. The anti-tumor effect was examined by using LDH (lactate dehydrogenase) release method.
The detection principle is as follows:
NK cells attack tumor cells to cause tumor cell death, and the tumor cells cultured by culture supernatant of pluripotent stem cells which cannot express CD47 inhibitory factor are not recognized by the NK cells and can generate immune escape. Because NK cells kill target cellsHour, CD107aThe molecule is transported to the cell membrane surface, NK cell membrane surface CD107aThe natural expression frequency is very low, only 1.2% -5.8%, therefore, only CD107aThe NK cells with the killing activity are the NK cells with the positive expression of the molecules. Therefore, by detecting NK cell surface CD107aThe ability of NK cells to kill tumor cells can be obtained.
The specific detection steps are as follows:
(a) preparation of effector cells:
using MagniSortTM Human NK cell Enrichment Kit(InvitrogenTMAnd the cargo number: 8804 and 6819-74) kit for sorting and separating NK cells. The isolated NK cells were resuspended in RPMI1640 medium containing 10% FBS, the cells were counted by trypan blue staining, and concentrated to 1X 107cells/mL.
(b) Preparation of target cells:
the target cells are tumor cells (NIC lung cancer cells), NIC lung cancer cells are digested and resuspended, and cells are counted by trypan blue staining to prepare 1 × 107cells/mL of cell suspension.
(c) Mixing NK cells (effector cells) and target cells (E) at a certain ratio (E/NK: 10:1, 25:1, 50:1, E/NK is the ratio of target cells to effector cells NK), placing in 96-well plate, and adding 5% CO at 37 deg.C2The culture was incubated for 4h, and 100. mu.L of 1% NP-40 was added to the maximum release group (the maximum release group replaced 0.1% NP-40 for effector cells), without target cells. In the natural release group, 100. mu.L of 0.9% physiological saline was added, and no target cells were added. Centrifuging at 500rpm for 5min, collecting supernatant 100 μ L, adding substrate solution 100 μ L, reacting at room temperature for 10min, adding 0.1mol/L citric acid 25 μ L/well to terminate enzymatic reaction, and measuring optical density of each well with enzyme-linked detector at 570nm wavelength.
(d) And (4) calculating a result:
according to the formula:
Figure BDA0002848707890000251
the effect of the pluripotent stem cells or the derivatives thereof expressing the targeted CD47 inhibitory factor on the killing of tumor cells by NK cells is obtained.
The results of the tests of the respective experimental groups are shown in Table 11.
TABLE 11 Effect of CD47 inhibitory factor expressed in each experimental group on NK cell killing of tumor cells
Figure BDA0002848707890000252
Figure BDA0002848707890000261
The N (control) group refers to NK cells that were not treated with the culture supernatant of pluripotent stem cells expressing CD47 inhibitor. Independent samples were tested for T (./p < 0.01).
Through the experiments, the CD47 inhibiting factor which is expressed by the pluripotent stem cells or the derivatives thereof and is wrapped by exosomes can be proved to have the anti-tumor effect.
Pluripotent stem cells expressing targeted CD47 inhibitors find use in a variety of tumor therapies:
immune compatible cells (MSCs) from both B2M and CIITA gene knock-out protocol groups (Aa3, Ab3) were selected for testing.
In the humanized NSG mouse tumor model, each group of experimental cells was injected and the effect on the treatment of NIC lung cancer, MM myeloma, SCC gastric cancer was observed. To avoid the problem of immune compatibility, the immune cells used are derived from the same person as the derivative of hPSCs.
The method comprises the following specific steps:
in humanized NSG mice (The Jackson Laboratory (JAX)), The right axilla was injected subcutaneously with 5X 106Tumor (NIC lung carcinoma, MM myeloma, SCC gastric carcinoma) cells until the tumor grows to 60MM3At size, tail vein injection of 200uL PBS (containing human immune cells and 10 mg)6The pluripotent stem cell derivative expressing the exosome-encapsulated CD47 suppressor) for tumor therapy, wherein only the human immune cell-containing cells are injectedThe group served as a control group. Mice were sacrificed after 20 days and tumor sizes were compared between groups and statistical analysis of differences was performed.
The results are shown in Table 12.
TABLE 12 antitumor Effect of pluripotent Stem cells or derivatives thereof on various tumors in Each test group
Figure BDA0002848707890000262
Figure BDA0002848707890000271
Independent samples were tested for T (./p < 0.01).
Through the experiments, the stem cells expressing the CD47 inhibitory factor or the derivatives thereof prepared by the invention can be proved to be capable of effectively blocking CD47 to play an anti-tumor role.
Immune compatibility testing of pluripotent stem cells expressing targeted CD47 inhibitor:
by utilizing the characteristic of low immunogenicity of the MSCs, in a humanized NSG mouse tumor model, the MSCs are injected with hPSCs source immune compatibility capable of expressing CD47 inhibitory factor (shRNA of targeting CD 47), and the effect of tumor (MM melanoma) treatment is observed. Wherein the immunocyte and the MSCs derived from hPSCs are derived from non-same human.
The control group is an NSG mouse tumor model without MSCs cell injection;
the Dox addition group is: the addition of 0.5mg/mL Dox to the mouse diet was continued from the injection of inhibitor-expressing cells until the end of the experiment.
The results are shown in Table 13.
TABLE 13 reversible expression test results for immune-compatible molecule-inducible expression sets
Figure BDA0002848707890000272
Independent samples were tested for T (./p < 0.01).
The above experiments show that: MSCs that express only suppressive factors (group 2), which have low immunogenicity and can exist in foreign body for a certain time, can exert a certain tumor therapeutic effect, while those that are immunologically compatibly engineered (groups 3-7, including constitutive and reversible inducible immunocompatibilities), which have better immuno-compatibility effects, are present in vivo for a longer time (or can achieve long-term co-existence) than MSCs that are not immunologically compatibly engineered, which exert better tumor therapeutic effects, whereas group 4, which is a B2M and CIITA gene knock-out group, completely eliminates the influence of HLA-I and HLA-II molecules, and thus, has the best tumor therapeutic effects. However, there are groups 6-9 protocol settings because of their constitutive immune compatible modifications (knock-in/knock-out) and their inability to clear when a graft becomes mutated or otherwise undesirable. In groups 8-9, the mice injected with the suppressor cells were shown to have abolished the immune compatibility effect by administering Dox inducer (always administered) to the mice at the same time as the injection of the suppressor cells into the mice, which was found in vivo for a time period comparable to that of the MSCs without immune compatibility modification, and the tumor therapy effect was comparable to that of the MSCs without immune compatibility modification.
The embodiments of the present invention are not limited to the embodiments, and any other changes, modifications, substitutions, combinations, and simplifications which do not depart from the spirit and principle of the present invention should be construed as equivalents and are included in the scope of the present invention.
SEQUENCE LISTING
<110> future Chile regenerative medicine institute (Guangzhou) Co., Ltd
Wang Linli
<120> pluripotent stem cell expressing targeted CD47 inhibitory factor, and derivative and application thereof
<130>
<160> 103
<170> PatentIn version 3.5
<210> 1
<211> 21
<212> DNA
<213> human
<400> 1
ggtgattacc cagagatatc t 21
<210> 2
<211> 21
<212> DNA
<213> human
<400> 2
gcacatgcat cttctgtatg g 21
<210> 3
<211> 21
<212> DNA
<213> human
<400> 3
gcatcttctg tatggacaaa g 21
<210> 4
<211> 315
<212> DNA
<213> Artificial sequence
<400> 4
ccactcccta tcagtgatag agaaaagtga aagtcgagtt taccactccc tatcagtgat 60
agagaaaagt gaaagtcgag tttaccactc cctatcagtg atagagaaaa gtgaaagtcg 120
agtttaccac tccctatcag tgatagagaa aagtgaaagt cgagctcggt acccgggtcg 180
aggtaggcgt gtacggtggg aggcctatat aagcagagct cgtttagtga accgtcagat 240
cgcctggaga cgccatccac gctgttttga cctccataga agacaccggg accgatccag 300
cctgctagcg ccacc 315
<210> 5
<211> 9
<212> DNA
<213> Artificial sequence
<400> 5
ttcaagaga 9
<210> 6
<211> 119
<212> DNA
<213> Artificial sequence
<400> 6
gaggcttcag tactttacag aatcgttgcc tgcacatctt ggaaacactt gctgggatta 60
cttcttcagg ttaacccaac agaaggctaa agaaggtata ttgctgttga cagtgagcg 119
<210> 7
<211> 19
<212> DNA
<213> Artificial sequence
<400> 7
tagtgaagcc acagatgta 19
<210> 8
<211> 119
<212> DNA
<213> Artificial sequence
<400> 8
tgcctactgc ctcggacttc aaggggctac tttaggagca attatcttgt ttactaaaac 60
tgaatacctt gctatctctt tgatacattt ttacaaagct gaattaaaat ggtataaat 119
<210> 9
<211> 210
<212> DNA
<213> Artificial sequence
<400> 9
attctagata cagtactttt gtgtagtaca acgtacagta cttttgtgta gtacaacgta 60
cagtactttt gtgtagtaca acgtacagta cttttgtgta gtacaacgta cagtactttt 120
gtgtagtaca acgtacagta cttttgtgta gtacaacgta cagtactttt gtgtagtaca 180
acgtacagta cttttgtgta gtacaacgta 210
<210> 10
<211> 21
<212> DNA
<213> human
<400> 10
gggagcagag aattctctta t 21
<210> 11
<211> 21
<212> DNA
<213> human
<400> 11
ggagcagaga attctcttat c 21
<210> 12
<211> 21
<212> DNA
<213> human
<400> 12
gagcagagaa ttctcttatc c 21
<210> 13
<211> 21
<212> DNA
<213> human
<400> 13
gctacctgga gcttcttaac a 21
<210> 14
<211> 21
<212> DNA
<213> human
<400> 14
ggagcttctt aacagcgatg c 21
<210> 15
<211> 21
<212> DNA
<213> human
<400> 15
gggtctccag tatattcatc t 21
<210> 16
<211> 21
<212> DNA
<213> human
<400> 16
gctcccactc catgaggtat t 21
<210> 17
<211> 21
<212> DNA
<213> human
<400> 17
ggtatttctt cacatccgtg t 21
<210> 18
<211> 21
<212> DNA
<213> human
<400> 18
aggagacacg gaatgtgaag g 21
<210> 19
<211> 21
<212> DNA
<213> human
<400> 19
gctcccactc catgaggtat t 21
<210> 20
<211> 21
<212> DNA
<213> human
<400> 20
ggtatttcta cacctccgtg t 21
<210> 21
<211> 21
<212> DNA
<213> human
<400> 21
ggaccggaac acacagatct a 21
<210> 22
<211> 21
<212> DNA
<213> human
<400> 22
ttcttacttc cctaatgaag t 21
<210> 23
<211> 21
<212> DNA
<213> human
<400> 23
aagttaagaa cctgaatata a 21
<210> 24
<211> 21
<212> DNA
<213> human
<400> 24
aacctgaata taaatttgtg t 21
<210> 25
<211> 21
<212> DNA
<213> human
<400> 25
gggtctggtg ggcatcatta t 21
<210> 26
<211> 21
<212> DNA
<213> human
<400> 26
ggtctggtgg gcatcattat t 21
<210> 27
<211> 21
<212> DNA
<213> human
<400> 27
gcatcattat tgggaccatc t 21
<210> 28
<211> 21
<212> DNA
<213> human
<400> 28
gatgaccaca ttcaaggaag a 21
<210> 29
<211> 21
<212> DNA
<213> human
<400> 29
gaccacattc aaggaagaac t 21
<210> 30
<211> 21
<212> DNA
<213> human
<400> 30
gctttcctgc ttggcagtta t 21
<210> 31
<211> 21
<212> DNA
<213> human
<400> 31
gcgtaagtct gagtgtcatt t 21
<210> 32
<211> 21
<212> DNA
<213> human
<400> 32
gacaatttaa ggaagaatct t 21
<210> 33
<211> 21
<212> DNA
<213> human
<400> 33
ggccatagtt ctccctgatt g 21
<210> 34
<211> 21
<212> DNA
<213> human
<400> 34
gccatagttc tccctgattg a 21
<210> 35
<211> 21
<212> DNA
<213> human
<400> 35
gcagatgacc acattcaagg a 21
<210> 36
<211> 21
<212> DNA
<213> human
<400> 36
gcagcaggat aagtatgagt g 21
<210> 37
<211> 21
<212> DNA
<213> human
<400> 37
gcaggataag tatgagtgtc a 21
<210> 38
<211> 21
<212> DNA
<213> human
<400> 38
ggttcctgca cagagacatc t 21
<210> 39
<211> 21
<212> DNA
<213> human
<400> 39
ggatgtggaa cccacagata c 21
<210> 40
<211> 21
<212> DNA
<213> human
<400> 40
gatgtggaac ccacagatac a 21
<210> 41
<211> 21
<212> DNA
<213> human
<400> 41
gtggaaccca cagatacaga g 21
<210> 42
<211> 21
<212> DNA
<213> human
<400> 42
gggtagcaac tgtcaccttg a 21
<210> 43
<211> 21
<212> DNA
<213> human
<400> 43
ggatttcgtg ttccagttta a 21
<210> 44
<211> 21
<212> DNA
<213> human
<400> 44
gcatgtgcta cttcaccaac g 21
<210> 45
<211> 21
<212> DNA
<213> human
<400> 45
gctcacagtc atcaattata g 21
<210> 46
<211> 21
<212> DNA
<213> human
<400> 46
gccctgaaga cagaatgttc c 21
<210> 47
<211> 21
<212> DNA
<213> human
<400> 47
gcggaccatg tgtcaactta t 21
<210> 48
<211> 21
<212> DNA
<213> human
<400> 48
gcctgatagg acccatattc c 21
<210> 49
<211> 21
<212> DNA
<213> human
<400> 49
gcatccaata gacgtcattt g 21
<210> 50
<211> 21
<212> DNA
<213> human
<400> 50
gcgtcactgg cacagatata a 21
<210> 51
<211> 21
<212> DNA
<213> human
<400> 51
ggatggattt gattatgatc c 21
<210> 52
<211> 21
<212> DNA
<213> human
<400> 52
ggaccttgga acaatggatt g 21
<210> 53
<211> 21
<212> DNA
<213> human
<400> 53
gctaattctt gctgaacttc t 21
<210> 54
<211> 21
<212> DNA
<213> human
<400> 54
gcagttctgt tgccactctc t 21
<210> 55
<211> 21
<212> DNA
<213> human
<400> 55
gggagagttc atccaggaaa t 21
<210> 56
<211> 21
<212> DNA
<213> human
<400> 56
ggagagttca tccaggaaat t 21
<210> 57
<211> 21
<212> DNA
<213> human
<400> 57
gggttggttt atccaggaat a 21
<210> 58
<211> 21
<212> DNA
<213> human
<400> 58
ggatcagaag agaagccaac g 21
<210> 59
<211> 21
<212> DNA
<213> human
<400> 59
ggttcaccat ccaggtgttc a 21
<210> 60
<211> 21
<212> DNA
<213> human
<400> 60
ggaggaactt tgtgaacatt c 21
<210> 61
<211> 21
<212> DNA
<213> human
<400> 61
gctgtaagaa ggatgctttc a 21
<210> 62
<211> 21
<212> DNA
<213> human
<400> 62
gctgcaggca ggattgtttc a 21
<210> 63
<211> 19
<212> DNA
<213> human
<400> 63
gcctcgagtt tgagagcta 19
<210> 64
<211> 19
<212> DNA
<213> human
<400> 64
agacattctg gatgagtta 19
<210> 65
<211> 19
<212> DNA
<213> human
<400> 65
gggtctgtta cccaaagaa 19
<210> 66
<211> 21
<212> DNA
<213> human
<400> 66
ggacactggt tcaacacctg t 21
<210> 67
<211> 21
<212> DNA
<213> human
<400> 67
ggttcaacac ctgtgacttc a 21
<210> 68
<211> 21
<212> DNA
<213> human
<400> 68
acctgtgact tcatgtgtgc g 21
<210> 69
<211> 1607
<212> DNA
<213> human
<400> 69
atgaatactc tccctgaaca ttcatgtgac gtgttgatta tcggtagcgg cgcagccgga 60
ctttcactgg cgctacgcct ggctgaccag catcaggtca tcgttctaag taaggccggt 120
aacgaggttc aacattttat gcccagggcg gtattgccgc cgtgtttgat aaactgacag 180
cattgactcg catgtggaag acacattgat tgccggggct ggtatttgcg atcgccatgc 240
agttgaattt gtcgccagca atgcacgatc ctgtgtgcaa tggctaatcg accagggggt 300
gttgtttgat acccacattc aaccgaatgg cgaagaaagt taccatctga cccgtgaagg 360
tggacatagt caccgtcgta ttcttcatgc cgccgacgcc accggtagag aagtagaaac 420
cacgctggtg agcaaggcgc tgaaccatcc gaatattcgc gtgctggagc gcagcaacgc 480
ggttgatctg attgtttctg acaaaattgg cctgccgggc acgcgacggg ttgttggcgc 540
gtgggtatgg aaccgtaata aagaaacggt ggaaacctgc cacgcaaaag cggtggtgct 600
ggcaaccggc ggtgcgtcga aggtttatca gtacaccacc aatccggata tttcttctgg 660
cgatggcatt gctatggcgt ggcgcgcagg ctgccggttg ccaatctcga tttaatcagt 720
tccaccctac cgcgctatat cacccacagg cacgcaattt cctgttaaca gaagcactgc 780
gcggcgaggc gcttatctca agcgcccgga tggtacgcgt ttatccgatt ttgatgagcg 840
cggcgaactg ccccgcgcga tattgtcgcc cgcgccattg accatgaaat gaaacgcctc 900
ggcgcagatt gtatgttcct tgatatcagc cataagcccg ccgattttat tcgccagcat 960
ttcccgatga tttatgaaaa gctgctcggg ctgggattga tctcacacaa gaaccggtac 1020
cgattgtgcc tgctgcacat tatacctgcg gtggtgtaat ggttgatgat catgggcgta 1080
cggacgtcga gggcttgtat gccattggcg aggtgagtta taccggctta cacggcgcta 1140
accgcatggc ctcgaattca ttgctggagt gtctggtcta tggctggtcg gcggcggaag 1200
atatcaccag acgtatgcct tatgcccacg acatcagtac gttaccgccg tgggatgaaa 1260
gccgcgttga gaaccctgac gaacggtagt aattcagcat aactggcacg agctacgtct 1320
gtttatgtgg gattacgttg gcattgtgcg cacaacgaag cgcctggaac gcgccctgcg 1380
gcggataacc atgctccaac aagaaataga cgaatattac gcccatttcc gcgtctcaaa 1440
taatttgctg gagctgcgta atctggtaca ggttgccgag ttgattgttc gctgtgcaat 1500
gatgcgtaaa gagagtcggg gttgcatttc acgctggatt atccggaact gctcacccat 1560
tccggtccgt cgatccttcc cccggcaatc attacataaa cagataa 1607
<210> 70
<211> 411
<212> DNA
<213> human
<400> 70
ggaagtggtg ccggcaccgg cggcatgtac gtgcgcttcg aggtgcccga ggacatgcag 60
aacgaggccc tgagcctgct ggaaaaagtg cgcgagagcg gcaaagtgaa gaagggcacc 120
aacgaaacca ccaaggccgt ggaacggggc ctggccaagc tggtgtatat cgccgaggac 180
gtggaccccc ccgagattgt ggcccatctg cccctgctgt gcgaagagaa gaacgtgccc 240
tacatctacg tgaagtccaa gaacgacctg ggcagagccg tgggcatcga ggtgccatgt 300
gcctctgccg ccatcatcaa cgagggcgag ctgcggaaag aactgggcag cctggtggaa 360
aagatcaagg gcctgcagaa gggttccggt ggatccggtt ccggacgggc t 411
<210> 71
<211> 20
<212> DNA
<213> Artificial sequence
<400> 71
tataaggtgg tcccagctcg 20
<210> 72
<211> 20
<212> DNA
<213> Artificial sequence
<400> 72
agggccggtt aatgtggctc 20
<210> 73
<211> 20
<212> DNA
<213> Artificial sequence
<400> 73
ctcctgttat attctagaac 20
<210> 74
<211> 20
<212> DNA
<213> Artificial sequence
<400> 74
tttcagcatc aatgtaccct 20
<210> 75
<211> 20
<212> DNA
<213> Artificial sequence
<400> 75
cgcgagcaca gctaaggcca 20
<210> 76
<211> 20
<212> DNA
<213> Artificial sequence
<400> 76
actctctctt tctggcctgg 20
<210> 77
<211> 20
<212> DNA
<213> Artificial sequence
<400> 77
ggcactcaga agacactgat 20
<210> 78
<211> 20
<212> DNA
<213> Artificial sequence
<400> 78
aaggtgtctg gtcggagagc 20
<210> 79
<211> 20
<212> DNA
<213> Artificial sequence
<400> 79
acccagcagg gcgtggagcc 20
<210> 80
<211> 20
<212> DNA
<213> Artificial sequence
<400> 80
gtcagagccc caaggtaaaa 20
<210> 81
<211> 900
<212> DNA
<213> Artificial sequence
<400> 81
cctggacttc tccagtactt tctggctgga ttggtatctg aggctagtag gaagggcttg 60
ttcctgctgg gtagctctaa acaatgtatt catgggtagg aacagcagcc tattctgcca 120
gccttatttc taaccatttt agacatttgt tagtacatgg tattttaaaa gtaaaactta 180
atgtcttcct tttttttctc cactgtcttt ttcatagatc gagacatgta agcagcatca 240
tggaggtaag tttttgacct tgagaaaatg tttttgtttc actgtcctga ggactattta 300
tagacagctc taacatgata accctcacta tgtggagaac attgacagag taacatttta 360
gcagggaaag aagaatccta cagggtcatg ttcccttctc ctgtggagtg gcatgaagaa 420
ggtgtatggc cccaggtatg gccatattac tgaccctcta cagagagggc aaaggaactg 480
ccagtatggt attgcaggat aaaggcaggt ggttacccac attacctgca aggctttgat 540
ctttcttctg ccatttccac attggacatc tctgctgagg agagaaaatg aaccactctt 600
ttcctttgta taatgttgtt ttattcttca gacagaagag aggagttata cagctctgca 660
gacatcccat tcctgtatgg ggactgtgtt tgcctcttag aggttcccag gccactagag 720
gagataaagg gaaacagatt gttataactt gatataatga tactataata gatgtaacta 780
caaggagctc cagaagcaag agagagggag gaacttggac ttctctgcat ctttagttgg 840
agtccaaagg cttttcaatg aaattctact gcccagggta cattgatgct gaaaccccat 900
<210> 82
<211> 900
<212> DNA
<213> Artificial sequence
<400> 82
tcaaatctcc tgttatattc tagaacaggg aattgatttg ggagagcatc aggaaggtgg 60
atgatctgcc cagtcacact gttagtaaat tgtagagcca ggacctgaac tctaatatag 120
tcatgtgtta cttaatgacg gggacatgtt ctgagaaatg cttacacaaa cctaggtgtt 180
gtagcctact acacgcatag gctacatggt atagcctatt gctcctagac tacaaacctg 240
tacagcctgt tactgtactg aatactgtgg gcagttgtaa cacaatggta agtatttgtg 300
tatctaaaca tagaagttgc agtaaaaata tgctatttta atcttatgag accactgtca 360
tatatacagt ccatcattga ccaaaacatc atatcagcat tttttcttct aagattttgg 420
gagcaccaaa gggatacact aacaggatat actctttata atgggtttgg agaactgtct 480
gcagctactt cttttaaaaa ggtgatctac acagtagaaa ttagacaagt ttggtaatga 540
gatctgcaat ccaaataaaa taaattcatt gctaaccttt ttcttttctt ttcaggtttg 600
aagatgccgc atttggattg gatgaattcc aaattctgct tgcttgcttt ttaatattga 660
tatgcttata cacttacact ttatgcacaa aatgtagggt tataataatg ttaacatgga 720
catgatcttc tttataattc tactttgagt gctgtctcca tgtttgatgt atctgagcag 780
gttgctccac aggtagctct aggagggctg gcaacttaga ggtggggagc agagaattct 840
cttatccaac atcaacatct tggtcagatt tgaactcttc aatctcttgc actcaaagct 900
<210> 83
<211> 900
<212> DNA
<213> Artificial sequence
<400> 83
cttgacaagt ctcctgctcc tcactatgaa gatcactgtc ccccagccct gtgctccccg 60
cactgtgctg cacgtccacc tccattccac tgcccctccc atccccccat cttgatagca 120
cccttcccag gtgtcaagct gcccctccta gagtgtcctg cctaaacccc ctctcctggc 180
tcctcccgct acagcatgtt ctctgaggac actaaccacg ctggaccttg aactgggtac 240
ttgtggacac agctcttctc caggctgtat cccatgagcc tcagcatcct ggcacccggc 300
ccctgctggt tcagggttgg cccctgcccg gctgcggaat gaaccacatc ttgctctgct 360
gacagacaca ggcccggctc caggctcctt tagcgcccag ttgggtggat gcctggtggc 420
agctgcggtc cacccaggag ccccgaggcc ttctctgaag gacattgcgg acagccacgg 480
ccaggccaga gggagtgaca gaggcagccc cattctgcct gcccaggccc ctgccaccct 540
ggggagaaag tacttctttt tttttatttt tagacagagt ctcactgttg cccaggctgg 600
cgtgcagtgg tgcgatctgg gttcactgca acctccgcct cttgggttca agcgattctt 660
ctgcttcagc ctcccgagta gctgggacta caggcaccca ccatcatgtc tggctaattt 720
ttcattttta gtagagacag ggttttgcca tgttggccag gctggtctca aactcttgac 780
ctcaggtgat ccacccacct cagcctccca aagtgctggg attacaagcg tgagccactg 840
caccgggcca cagagaaagt acttctccac cctgctctcc gaccagacac cttgacaggg 900
<210> 84
<211> 900
<212> DNA
<213> Artificial sequence
<400> 84
cacaccgggc actcagaaga cactgatggg caacccccag cctgctaatt ccccagattg 60
caacaggctg ggcttcagtg gcagctgctt ttgtctatgg gactcaatgc actgacattg 120
ttggccaaag ccaaagctag gcctggccag atgcaccagc ccttagcagg gaaacagcta 180
atgggacact aatggggcgg tgagagggga acagactgga agcacagctt catttcctgt 240
gtcttttttc actacattat aaatgtctct ttaatgtcac aggcaggtcc agggtttgag 300
ttcataccct gttaccattt tggggtaccc actgctctgg ttatctaata tgtaacaagc 360
caccccaaat catagtggct taaaacaaca ctcacattta ttctgctcac atatctgtca 420
tttgagcagg gctcagcggg gacagctcct tctgtcctac tctgtgtcag gtggggcagc 480
ttgagggttg ggctggtgtc acctgaagac tcattcttct gtacgtctga caggcaatgc 540
tggctgttgg ctgggggcct cagtgccact acggaatagt tggctaggac ccctccatgt 600
gggctagttg ggcttcctca tagtatggtg gctgggttgg agggtgtccc aaaaagaaag 660
gaggggatag agagagacca cttttcataa cctagcctta gaagtcacac agtattactt 720
ctgctacata tatatgtttt aagaggcagg gtctcactct gtcgcccagt ctggaatgca 780
gtggtatgat cacggctcac tgcagcctca acctcctggg ctaagtgatc ctcccacctc 840
agcctcccga atagctggga ctacaggtgt gagtcaccaa gcccagttaa tctttagttt 900
<210> 85
<211> 804
<212> DNA
<213> Artificial sequence
<400> 85
tgctttctct gacctgcatt ctctcccctg ggcctgtgcc gctttctgtc tgcagcttgt 60
ggcctgggtc acctctacgg ctggcccaga tccttccctg ccgcctcctt caggttccgt 120
cttcctccac tccctcttcc ccttgctctc tgctgtgttg ctgcccaagg atgctctttc 180
cggagcactt ccttctcggc gctgcaccac gtgatgtcct ctgagcggat cctccccgtg 240
tctgggtcct ctccgggcat ctctcctccc tcacccaacc ccatgccgtc ttcactcgct 300
gggttccctt ttccttctcc ttctggggcc tgtgccatct ctcgtttctt aggatggcct 360
tctccgacgg atgtctccct tgcgtcccgc ctccccttct tgtaggcctg catcatcacc 420
gtttttctgg acaaccccaa agtaccccgt ctccctggct ttagccacct ctccatcctc 480
ttgctttctt tgcctggaca ccccgttctc ctgtggattc gggtcacctc tcactccttt 540
catttgggca gctcccctac cccccttacc tctctagtct gtgctagctc ttccagcccc 600
ctgtcatggc atcttccagg ggtccgagag ctcagctagt cttcttcctc caacccgggc 660
ccctatgtcc acttcaggac agcatgtttg ctgcctccag ggatcctgtg tccccgagct 720
gggaccacct tatattccca gggccggtta atgtggctct ggttctgggt acttttatct 780
gtcccctcca ccccacagtg gggc 804
<210> 86
<211> 837
<212> DNA
<213> Artificial sequence
<400> 86
actagggaca ggattggtga cagaaaagcc ccatccttag gcctcctcct tcctagtctc 60
ctgatattgg gtctaacccc cacctcctgt taggcagatt ccttatctgg tgacacaccc 120
ccatttcctg gagccatctc tctccttgcc agaacctcta aggtttgctt acgatggagc 180
cagagaggat cctgggaggg agagcttggc agggggtggg agggaagggg gggatgcgtg 240
acctgcccgg ttctcagtgg ccaccctgcg ctaccctctc ccagaacctg agctgctctg 300
acgcggccgt ctggtgcgtt tcactgatcc tggtgctgca gcttccttac acttcccaag 360
aggagaagca gtttggaaaa acaaaatcag aataagttgg tcctgagttc taactttggc 420
tcttcacctt tctagtcccc aatttatatt gttcctccgt gcgtcagttt tacctgtgag 480
ataaggccag tagccagccc cgtcctggca gggctgtggt gaggaggggg gtgtccgtgt 540
ggaaaactcc ctttgtgaga atggtgcgtc ctaggtgttc accaggtcgt ggccgcctct 600
actccctttc tctttctcca tccttctttc cttaaagagt ccccagtgct atctgggaca 660
tattcctccg cccagagcag ggtcccgctt ccctaaggcc ctgctctggg cttctgggtt 720
tgagtccttg gcaagcccag gagaggcgct caggcttccc tgtccccctt cctcgtccac 780
catctcatgc ccctggctct cctgcccctt ccctacaggg gttcctggct ctgctct 837
<210> 87
<211> 253
<212> DNA
<213> Artificial sequence
<400> 87
gagggcctat ttcccatgat tccttcatat ttgcatatac gatacaaggc tgttagagag 60
ataattggaa ttaatttgac tgtaaacaca aagatattag tacaaaatac gtgacgtaga 120
aagtaataat ttcttgggta gtttgcagtt ttaaaattat gttttaaaat ggactatcat 180
atgcttaccg taacttgaaa gtatttcgat ttcttggctt tatatatctt gtggaaagga 240
cgctagcgcc acc 253
<210> 88
<211> 686
<212> DNA
<213> Artificial sequence
<400> 88
gagggcctat ttcccatgat tccttcatat ttgcatatac gatacaaggc tgttagagag 60
ataattggaa ttaatttgac tgtaaacaca aagatattag tacaaaatac gtgacgtaga 120
aagtaataat ttcttgggta gtttgcagtt ttaaaattat gttttaaaat ggactatcat 180
atgcttaccg taacttgaaa gtatttcgat ttcttggctt tatatatctt gtggaaagga 240
ctttaccact ccctatcagt gatagagaaa agtgaaagtc gagtttacca ctccctatca 300
gtgatagaga aaagtgaaag tcgagtttac cactccctat cagtgataga gaaaagtgaa 360
agtcgagttt accactccct atcagtgata gagaaaagtg aaagtcgagt ttaccactcc 420
ctatcagtga tagagaaaag tgaaagtcga gtttaccact ccctatcagt gatagagaaa 480
agtgaaagtc gagtttacca ctccctatca gtgatagaga aaagtgaaag tcgagctcgg 540
tacccgggtc gaggtaggcg tgtacggtgg gaggcctata taagcagagc tcgtttagtg 600
aaccgtcaga tcgcctggag acgccatcca cgctgttttg acctccatag aagacaccgg 660
gaccgatcca gcctgctagc gccacc 686
<210> 89
<211> 22
<212> DNA
<213> Artificial sequence
<400> 89
ccatagctca gtctggtcta tc 22
<210> 90
<211> 22
<212> DNA
<213> Artificial sequence
<400> 90
ctcttcgtcc agatcatcct ga 22
<210> 91
<211> 22
<212> DNA
<213> Artificial sequence
<400> 91
ccatagctca gtctggtcta tc 22
<210> 92
<211> 20
<212> DNA
<213> Artificial sequence
<400> 92
cacaccttgc cgatgtcgag 20
<210> 93
<211> 24
<212> DNA
<213> Artificial sequence
<400> 93
gcactgaacg aacatctcaa gaag 24
<210> 94
<211> 22
<212> DNA
<213> Artificial sequence
<400> 94
ctcttcgtcc agatcatcct ga 22
<210> 95
<211> 24
<212> DNA
<213> Artificial sequence
<400> 95
gcactgaacg aacatctcaa gaag 24
<210> 96
<211> 20
<212> DNA
<213> Artificial sequence
<400> 96
cacaccttgc cgatgtcgag 20
<210> 97
<211> 22
<212> DNA
<213> Artificial sequence
<400> 97
tgctccgggt ttgtctcaga tg 22
<210> 98
<211> 22
<212> DNA
<213> Artificial sequence
<400> 98
ctcttcgtcc agatcatcct ga 22
<210> 99
<211> 22
<212> DNA
<213> Artificial sequence
<400> 99
tgctccgggt ttgtctcaga tg 22
<210> 100
<211> 20
<212> DNA
<213> Artificial sequence
<400> 100
cacaccttgc cgatgtcgag 20
<210> 101
<211> 590
<212> DNA
<213> Artificial sequence
<400> 101
cccctctccc tccccccccc ctaacgttac tggccgaagc cgcttggaat aaggccggtg 60
tgcgtttgtc tatatgttat tttccaccat attgccgtct tttggcaatg tgagggcccg 120
gaaacctggc cctgtcttct tgacgagcat tcctaggggt ctttcccctc tcgccaaagg 180
aatgcaaggt ctgttgaatg tcgtgaagga agcagttcct ctggaagctt cttgaagaca 240
aacaacgtct gtagcgaccc tttgcaggca gcggaacccc ccacctggcg acaggtgcct 300
ctgcggccaa aagccacgtg tataagatac acctgcaaag gcggcacaac cccagtgcca 360
cgttgtgagt tggatagttg tggaaagagt caaatggctc tcctcaagcg tattcaacaa 420
ggggctgaag gatgcccaga aggtacccca ttgtatggga tctgatctgg ggcctcggtg 480
cacatgcttt acatgtgttt agtcgaggtt aaaaaaacgt ctaggccccc cgaaccacgg 540
ggacgtggtt ttcctttgaa aaacacgatg ataatatggc cacaaccatg 590
<210> 102
<211> 21
<212> DNA
<213> Caenorhabditis elegans
<400> 102
ttgtactaca caaaagtact g 21
<210> 103
<211> 24
<212> DNA
<213> Caenorhabditis elegans
<400> 103
tcacaacctc ctagaaagag taga 24

Claims (22)

1. A pluripotent stem cell or a derivative thereof, wherein a CD47 inhibitor expression sequence is introduced into the genome of the pluripotent stem cell or the derivative thereof; the CD47 inhibitor is at least one of shRNA and/or shRNA-miR of targeting CD 47.
2. The pluripotent stem cell or the derivative thereof according to claim 1, wherein the sequence of the shRNA and/or shRNA-miR targeting CD47 is shown as SEQ ID No.1 to SEQ ID No. 2.
3. The pluripotent stem cell or the derivative thereof according to claim 1, wherein the B2M gene and/or the CIITA gene of the genome of the pluripotent stem cell or the derivative thereof is knocked out.
4. The pluripotent stem cell or the derivative thereof according to claim 1, wherein: the genome of the pluripotent stem cell or the derivative thereof is also introduced with a first nucleic acid molecule;
and, a second nucleic acid molecule is further introduced into the 3' UTR region of the immune response-associated gene in the pluripotent stem cell or the derivative thereof;
the first nucleic acid molecule encodes a small nucleic acid molecule that mediates RNA interference, which small nucleic acid molecule specifically targets the transcript of the second nucleic acid molecule, and which small nucleic acid molecule does not target any other mRNA or incrna of the pluripotent stem cell or a derivative thereof.
5. The pluripotent stem cell or the derivative thereof according to claim 4, wherein the small nucleic acid molecule comprises at least one of a short interfering nucleic acid, a short interfering RNA, a double-stranded RNA, preferably miRNA, shRNA-miR.
6. The pluripotent stem cells or the derivatives thereof according to claim 5, wherein the pluripotent stem cells or the derivatives thereof are derived from a human; the sequence of the small nucleic acid molecule is a random sequence of a non-human species that does not target any mRNA or incrna of humans.
7. The pluripotent stem cell or the derivative thereof according to claim 4, wherein the second nucleic acid molecule comprises the reverse complement of at least 3 repeats of the sequence of the small nucleic acid molecule, preferably 6 to 10 repeats of the sequence of the small nucleic acid molecule.
8. The pluripotent stem cells or the derivatives thereof according to claim 1, wherein the genome of the pluripotent stem cells or the derivatives thereof further comprises an expression sequence of at least one immune compatible molecule for regulating the expression of genes associated with an immune response in the pluripotent stem cells or the derivatives thereof.
9. The pluripotent stem cell or the derivative thereof according to claim 4 or 8, wherein the genes associated with the immune response comprise:
(1) major histocompatibility complex genes including at least one of HLA-A, HLA-B, HLA-C, HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1 and HLA-DPB 1;
(2) major histocompatibility complex-associated genes including at least one of B2M and CIITA.
10. The pluripotent stem cell or derivative thereof of claim 8, wherein the immune-compatible molecule comprises at least one of:
(1) immune tolerance-related genes, including HLA-G;
(2) HLA-C molecules, including HLA-C multiple alleles of which the proportion in the population is over 90 percent in total, or fusion protein genes consisting of the HLA-C multiple alleles of which the proportion is over 90 percent and B2M;
(3) shRNA and/or shRNA-miR targeting major histocompatibility complex genes including at least one of HLA-A, HLA-B, HLA-C, HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1 and HLA-DPB 1;
(4) shRNA and/or shRNA-miR targeting a major histocompatibility complex-associated gene that includes at least one of B2M and CIITA.
11. The pluripotent stem cell or derivative thereof of claim 10, wherein:
the target sequence of the shRNA and/or shRNA-miR targeting B2M is selected from one of SEQ ID NO. 10-SEQ ID NO. 12;
the target sequence of the shRNA and/or shRNA-miR of the targeting CIITA is selected from one of SEQ ID NO. 13-SEQ ID NO. 15;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-A is selected from one of SEQ ID NO. 16-SEQ ID NO. 18;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-B is selected from one of SEQ ID NO. 19-SEQ ID NO. 21;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-C is selected from one of SEQ ID NO. 22-SEQ ID NO. 24;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRA is selected from one of SEQ ID NO. 25-SEQ ID NO. 27;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB1 is selected from one of SEQ ID NO. 28-SEQ ID NO. 30;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB3 is selected from one of SEQ ID NO. 31-SEQ ID NO. 32;
the target sequence of the shRNA and/or shRNA-miR targeting HLA-DRB4 is selected from one of SEQ ID NO. 33-SEQ ID NO. 35;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DRB5 is selected from one of SEQ ID NO. 36-SEQ ID NO. 38;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DQA1 is selected from one of SEQ ID NO. 39-SEQ ID NO. 41;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DQB1 is selected from one of SEQ ID NO. 42-SEQ ID NO. 44;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DPA1 is selected from one of SEQ ID NO. 45-SEQ ID NO. 47;
the target sequence of the shRNA and/or shRNA-miR of the target HLA-DPB1 is selected from one of SEQ ID NO. 48-SEQ ID NO. 51.
12. The pluripotent stem cell or the derivative thereof according to any one of claims 1 to 11, wherein a shRNA and/or miRNA processing complex-associated gene and/or an anti-interferon effector molecule is further introduced into the genome of the pluripotent stem cell or the derivative thereof, wherein: the shRNA and/or miRNA processing complex related gene comprises at least one of Drosha, Ago1, Ago2, Dicer1, Exportin-5, TRBP (TARBP2), PACT (PRKRA) and DGCR 8; the anti-interferon effector molecule is shRNA and/or shRNA-miR of at least one of target PKR, 2-5As, IRF-3 and IRF-7.
13. The pluripotent stem cell or the derivative thereof according to claim 12, wherein the target sequence of the PKR-targeting shRNA and/or shRNA-miR is selected from one of SEQ ID No.51 to SEQ ID No. 53;
the target sequence of the shRNA and/or shRNA-miR targeting 2-5As is selected from one of SEQ ID NO. 54-SEQ ID NO. 62;
the target sequence of the shRNA and/or shRNA-miR of the target IRF-3 is selected from one of SEQ ID NO. 63-SEQ ID NO. 65;
the target sequence of the shRNA and/or shRNA-miR of the target IRF-7 is selected from one of SEQ ID NO. 66-SEQ ID NO. 68.
14. The pluripotent stem cell or the derivative thereof according to claim 2, 11 or 13, wherein the expression frameworks of the shRNA and/or shRNA-miR targeting CD47, major histocompatibility complex gene, major histocompatibility complex-related gene, anti-interferon effector molecule are as follows:
the shRNA expression framework is as follows: the gene sequence sequentially comprises an shRNA target sequence, a stem-loop sequence, a reverse complementary sequence of the shRNA target sequence and Poly T from 5 'to 3';
wherein the shRNA target sequence, the stem-loop sequence and the reverse complementary sequence of the shRNA target sequence form a hairpin structure; poly T is a transcription terminator of RNA polymerase III;
shRNA-miR expression framework: and replacing the shRNA target sequence in the shRNA expression frame by using a shRNA-miR target sequence.
15. The pluripotent stem cell or the derivative thereof according to claim 4, 8 or 12, wherein an inducible gene expression system is further introduced into the genome of the pluripotent stem cell or the derivative thereof for regulating the expression of the first nucleic acid molecule and/or the immune-compatible molecule and/or the shRNA and/or the miRNA processing complex-associated gene and/or the anti-interferon effector molecule.
16. The pluripotent stem cell or the derivative thereof of claim 15, wherein the inducible gene expression system comprises at least one of a Tet-Off system, a dimer inducible expression system.
17. The pluripotent stem cell or derivative thereof according to any one of claims 1 to 16, wherein the genome of the pluripotent stem cell or derivative thereof further comprises an exosome-processing synthetic gene comprising at least one of STEAP3, Syndevan-4, an L-aspartate oxidase fragment, CD63-L7Ae and Cx43S 368A.
18. The pluripotent stem cell or the derivative thereof according to any one of claims 1 to 17, wherein the expression sequence of the CD47 suppressor, the first nucleic acid molecule, the expression sequence of an immune-compatible molecule, the shRNA and/or miRNA processing complex-associated gene, the anti-interferon effector molecule, the inducible gene expression system, the exosome processing synthetic gene is introduced by viral vector interference, non-viral vector transfection or gene editing, preferably by knock-in.
19. The pluripotent stem cell or the derivative thereof according to any one of claims 1 to 17, wherein the expression sequence of the CD47 suppressor, the expression sequence of an immune-compatible molecule, the shRNA and/or miRNA processing complex-associated gene, the anti-interferon effector molecule, the inducible gene expression system, the introduction site of the exosome processing synthetic gene is a genome-safe site, preferably one or more of an AAVS 1-safe site, an eGSH-safe site, and an H11-safe site.
20. The pluripotent stem cell or derivative thereof of any one of claims 1 to 19, wherein:
the pluripotent stem cells comprise embryonic stem cells, embryonic germ cells, embryonic cancer cells, or induced pluripotent stem cells;
the pluripotent stem cell derivative comprises an adult stem cell, each germ layer cell or a tissue or organ into which the pluripotent stem cell is differentiated;
the adult stem cells include mesenchymal stem cells or neural stem cells.
21. Use of the pluripotent stem cells or derivatives thereof and exosomes secreted therefrom according to claim 20 for the preparation of an anti-tumor therapeutic agent or drug.
22. An exosome secreted from the pluripotent stem cell or derivative thereof of any one of claims 1 to 20.
CN202011520308.0A 2020-12-21 2020-12-21 Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof Pending CN114645021A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011520308.0A CN114645021A (en) 2020-12-21 2020-12-21 Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011520308.0A CN114645021A (en) 2020-12-21 2020-12-21 Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof

Publications (1)

Publication Number Publication Date
CN114645021A true CN114645021A (en) 2022-06-21

Family

ID=81990851

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011520308.0A Pending CN114645021A (en) 2020-12-21 2020-12-21 Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof

Country Status (1)

Country Link
CN (1) CN114645021A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110863015A (en) * 2019-11-25 2020-03-06 重庆精准生物产业技术研究院有限公司 Target cell with low sensitivity to T cell and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108368520A (en) * 2015-11-04 2018-08-03 菲特治疗公司 The genome project of pluripotent cell is transformed

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108368520A (en) * 2015-11-04 2018-08-03 菲特治疗公司 The genome project of pluripotent cell is transformed

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
FRANK R T等: "Concise review: stem cells as an emerging platform for antibody therapy of cancer", STEM CELLS, vol. 28, no. 11, 31 December 2010 (2010-12-31) *
汲晓沛等: "负性协同刺激分子B7-H4在C3H10T1/2移植治疗EAE中作用机制的研究", 万方数据, 14 November 2015 (2015-11-14) *
负性协同刺激分子B7-H4在C3H10T1/2移植治疗EAE中作用机制的研究:百度学术,B7-H4 AND 干细胞: "CD47与肿瘤免疫相关性研究进展", 广东医学, vol. 32, no. 18, 30 September 2011 (2011-09-30), pages 4 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110863015A (en) * 2019-11-25 2020-03-06 重庆精准生物产业技术研究院有限公司 Target cell with low sensitivity to T cell and preparation method and application thereof
CN110863015B (en) * 2019-11-25 2023-06-16 重庆精准生物产业技术研究院有限公司 Target cell with low sensitivity to T cells, preparation method and application thereof

Similar Documents

Publication Publication Date Title
Ding et al. PARP inhibition elicits STING-dependent antitumor immunity in Brca1-deficient ovarian cancer
CN112342196A (en) Immune-compatible reversible universal pluripotent stem cell and application thereof
KR20210010555A (en) Drug resistant immune cells and methods of use thereof
CN107523569A (en) The purposes and its related drugs of PDCD1 genes
CN114645021A (en) Pluripotent stem cell expressing targeted CD47 inhibitory factor, derivative and application thereof
CN114657133A (en) Pluripotent stem cell for expressing shRNA and/or shRNA-miR of targeted IL-4R alpha
CN114657136A (en) Pluripotent stem cell expressing shRNA and/or shRNA-miR of target PCSK9 or derivative thereof
CN114426953A (en) Pluripotent stem cell derivative for expressing IL-12 and application thereof
CN114645020A (en) Pluripotent stem cell expressing targeted NR4A1 inhibitory factor, and derivative and application thereof
CN114657132A (en) Pluripotent stem cell expressing Siglec-15 targeted inhibitory factor, derivative and application thereof
CN114645018A (en) Pluripotent stem cell expressing CD38 targeted inhibitory factor, derivative and application thereof
CN114657135A (en) Pluripotent stem cell expressing Tim-3 targeted inhibitory factor, derivative and application thereof
CN114657139A (en) Pluripotent stem cell expressing LAG-3 targeted inhibitory factor, derivative and application thereof
CN114657130A (en) Pluripotent stem cell expressing VEGF-A targeted inhibitory factor, derivative and application thereof
CN114107211A (en) Pluripotent stem cell and derivative thereof
CN114717193A (en) Pluripotent stem cell expressing shRNA and/or shRNA-miR targeting B7-H5 or derivative thereof
CN114657138A (en) Pluripotent stem cell expressing shRNA and/or shRNA-miR targeting B7-H4 or derivative thereof
CN114657137A (en) Pluripotent stem cell or derivative thereof for expressing BTLA-targeted shRNA and/or shRNA-miR
CN114717232A (en) Pluripotent stem cell expressing IRF-1 targeted inhibitory factor, derivative and application thereof
CN114657134A (en) Pluripotent stem cell expressing IgE-targeted shRNA and/or shRNA-miR or derivative thereof
CN114717192A (en) Pluripotent stem cell expressing Amyloid beta targeted inhibitory factor, derivative and application thereof
CN114457022A (en) Pluripotent stem cell expressing OX40 activated antibody and derivative and application thereof
CN114457026A (en) Pluripotent stem cell expressing 4-1BB activated antibody, derivative and application thereof
CN114457035A (en) Pluripotent stem cell expressing LAG-3 blocking substance or derivative thereof and application
CN114457023A (en) Pluripotent stem cell expressing CD28 activated antibody, derivative 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