CN114958921A - Method for improving stability and function of regulatory T cells - Google Patents

Method for improving stability and function of regulatory T cells Download PDF

Info

Publication number
CN114958921A
CN114958921A CN202210407362.7A CN202210407362A CN114958921A CN 114958921 A CN114958921 A CN 114958921A CN 202210407362 A CN202210407362 A CN 202210407362A CN 114958921 A CN114958921 A CN 114958921A
Authority
CN
China
Prior art keywords
vector
beta
tgf
nucleic acid
cells
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
CN202210407362.7A
Other languages
Chinese (zh)
Inventor
徐建青
张晓燕
李昂
高戎戎
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.)
Zhongshan Hospital Fudan University
Original Assignee
Zhongshan Hospital Fudan University
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 Zhongshan Hospital Fudan University filed Critical Zhongshan Hospital Fudan University
Priority to CN202210407362.7A priority Critical patent/CN114958921A/en
Publication of CN114958921A publication Critical patent/CN114958921A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5428IL-10
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0637Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Hematology (AREA)
  • Transplantation (AREA)
  • Developmental Biology & Embryology (AREA)
  • Epidemiology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention provides a method for improving the stability and the immune regulation function of regulatory T cells based on the combination of a transcription factor inhibiting T-beta technology and cytokine overexpression. Specifically, the transcription factor T-beta inhibiting technology comprises hairpin RNA interference technology, and the cell factors comprise TGF-beta and IL-10. The technology provided by the invention can effectively maintain the stability of the regulatory T cells in the inflammatory environment for a long time, has a remarkable maintaining effect especially on the genetics and functional stability of the inducible regulatory T cells, has an important revelation value for solving the scientific problem of instability of the inflammatory environment function of the regulatory T cells, and has a wide application prospect in the fields of treating autoimmune diseases, intervening organ transplant rejection and the like.

Description

Method for improving stability and function of regulatory T cells
Technical Field
The invention relates to the field of biotechnology and medicine, in particular to a method for improving stability and function of induced regulatory T cells.
Background
Regulatory T cells (tregs) are a group of lymphocytes that down-regulate the immune response of the body, maintain immune homeostasis by down-regulating the proliferation and function of immune effector cells, maintain the body's tolerance to self or foreign antigens, and prevent excessive immune responses. For a long time, systemic immunosuppressive drugs, hormones, and the like have been mainly used for clinical treatments of diseases caused by excessive immune response, such as allergic diseases, autoimmune diseases, and organ transplant rejection. Most of the drugs are taken for life, and are easy to induce liver injury, bone marrow suppression, metabolic disorder, increase the risk of bacterial and fungal infection and tumor. In recent years, clinical trials of Treg cell intervention are gradually developed at home and abroad aiming at the diseases, so as to replace or reduce the dosage of systemic immunosuppressive drugs or hormones and assist immune reconstitution.
The Treg Cell subpopulation selected by the current immunotherapy clinical test project is relatively single, and Natural Regulatory T cells (ntreglatory T cells, ntregs) with stable epigenetics from thymus are mostly adopted to ensure the lineage and functional stability of the Treg cells in the environment of rejection inflammation in vivo. Due to nTreg (CD 4) + CD25 + ) The proportion of mononuclear cells (PBMC) in the peripheral blood cells is very low (0.1-0.7%), the peripheral blood of a patient needs to be collected in a large amount, and the compliance and the safety of the patient are reduced. Furthermore, the technology of in vitro expansion of Treg cells by PBMCThe operation method is mainly used for promoting cell proliferation by adjusting the dosage of rapamycin and IL-2 cytokines in a culture system, and the maturity of an amplification technology still has a larger space for improvement.
Induced Regulatory T cells (iTreg), which have been discovered in recent years, have a strong ability to mediate recognition tolerance and mucosal and environmental tolerance of foreign antigens, and can exert a good immune negative Regulatory function in transplantation tolerance, allergic reactions and chronic inflammation. From Naive CD4 + The iTreg cells obtained by the T cell induced differentiation have sufficient initial cell number and short amplification period required by an in-vitro amplification culture system, and are expected to break through the restriction of in-vitro large-scale amplification of the nTreg cells. The TGF-beta signal pathway regulates and controls key molecules of iTreg cell differentiation, including expression and phosphorylation of transcription factor STAT3 and methylation of Foxp3 gene, so that an iTreg culture system is seriously dependent on exogenously added cell factor TGF-beta, a large amount of the latter increases the production cost of cells, and factors such as addition time phase, half-life period, functional stability and the like also need to be comprehensively considered so as to further optimize the controllability of an amplification system. The international clinical trial of first using ex vivo expanded iTreg cells to intervene in transplant rejection was conducted by the Margaret l. Mainly observe the safety and the effectiveness of the iTreg in negatively regulating the host rejection reaction resistance of the transplanted graft after the hematopoietic stem cell transplantation. Due to the large amount of blood collected from donors, only 14 out of 55 donors initially screened into the group eventually received such large amounts of blood collected into clinical trials. The iTreg cells account for about 44% at the end point of the in vitro amplification culture system, and the capability of in vitro negative regulation and control of the proliferation of effector cells is weaker, so that the iTreg cell amplification system is prompted to be further optimized. The validity of the clinical trial results has not been determined.
Due to the low epigenetic stability of iTreg, Foxp3 expression is easily lost in the context of high inflammation and is then polarized towards inflammatory effector T cells, leading to an increased risk of inducing inflammation. In addition, autoimmune diseases or organ transplant rejectants are in an inflammatory immune environment for a long time in vivo, the functions of self-separated Treg cells are low, and the clinical application has a larger challenge. Therefore, how to effectively maintain the phenotype and functional stability of the iTreg cells and further improve the immune negative regulation function of the iTreg cells is the key point for breaking through the technical bottleneck of the immune therapy of the Treg cells.
Disclosure of Invention
In order to solve the above scientific and technical problems, the present invention provides in a first aspect a method for increasing induction, phenotypic stability and negative immunoregulatory function of a regulatory T immune cell, said method comprising inhibiting expression of transcription factor T-beta or knocking out TBX21 gene.
In certain embodiments, the nucleic acid encoding the transcription factor T-beta comprises SEQ ID NO 1, a degenerate sequence thereof, or a spliceosome nucleotide sequence thereof.
In certain embodiments, the transcription factor T-beta protein comprises the amino acid sequence of SEQ ID NO 2, a spliceosome thereof, or a variant thereof.
In certain embodiments, the variant refers to a protein that is functionally identical or similar to SEQ ID No. 2 or a spliceosome thereof, resulting from substitution and/or deletion and/or addition of at least one amino acid in the amino acid sequence shown in SEQ ID No. 2 or a spliceosome thereof.
In certain embodiments, methods or products for the inhibition of the expression of transcription factor T-beta include, but are not limited to, small RNAs or small RNAs of multi-target multi-hairpin structures, ribozymes, gene targeting, protein inhibitors and compound inhibitors, and the like; preferably, siRNA interference through hairpin structure; more preferably, the siRNA has a sequence shown in SEQ ID NO 3-8.
In certain embodiments, the methods or products for knocking out the TBX21 gene are selected from CRISPR/Cas9 technology, zinc finger nucleases, TALENs, and the like.
In certain embodiments, the method of the first aspect of the invention further comprises the step of adding or overexpressing a combination of one or more cytokines.
In some embodiments, the combination of multiple cytokines may be achieved by adding multiple cytokines simultaneously, or by overexpressing multiple cytokine genes simultaneously, or by adding and overexpressing a combination of multiple cytokines or their fragments, or by adding fusion proteins of multiple cytokines or their fragments, or by overexpressing multiple cytokines or their fragments, to enhance induction, phenotypic stability, and immunomodulatory function of regulatory T-immune cells.
In certain embodiments, the cytokines include, but are not limited to, natural or recombinant interleukins, interferons, colony stimulating factors, erythropoietins, epidermal growth factors, nerve growth factors, basic fibroblast growth factors, tumor necrosis factors and antagonists thereof, thrombopoietin, platelet-derived growth factors, stem cell factors, vascular endothelial growth factors and antagonists thereof; preferably, the cytokine is TGF-beta, IL-10 or a combination thereof.
In certain embodiments, the TGF- β is selected from TGF- β 1, TGF- β 2, TGF- β 3, and their corresponding precursor molecules (per-pro-TGF- β), polypeptide chain precursors (pro-TGF- β), and mature forms (mTGF- β).
In certain embodiments, the TGF- β is TGF- β 1, the nucleic acid encoding TGF- β 1 comprises SEQ ID NO 9, a degenerate sequence thereof, or a spliceosome nucleotide sequence thereof, and the TGF- β 1 protein comprises the amino acid sequence of SEQ ID NO 10, a spliceosome or a variant thereof;
in certain embodiments, the variant refers to a protein that is functionally identical or similar to SEQ ID NO. 10 or a spliceosome thereof, which is obtained by substituting and/or deleting and/or adding at least one amino acid in the amino acid sequence shown in SEQ ID NO. 10 or the spliceosome amino acid sequence thereof.
In certain embodiments, the TGF- β is mTGF- β 1, the mTGF- β 1 encoding nucleic acid comprises SEQ ID NO 11, a degenerate sequence thereof, or a splice nucleotide sequence thereof, and the mTGF- β 1 protein comprises SEQ ID NO 12, a splice thereof, or a variant amino acid sequence thereof.
In certain embodiments, the variant refers to a protein that is functionally identical or similar to SEQ ID No.12 or a spliceosome thereof, which is obtained by substitution and/or deletion and/or addition of at least one amino acid in the amino acid sequence shown in SEQ ID No.12 or a spliceosome thereof.
In certain embodiments, the IL-10 encoding nucleic acid has the nucleotide sequence of SEQ ID NO 13, a degenerate sequence thereof, or a splice thereof, and the mTGF- β 1 protein comprises the amino acid sequence set forth in SEQ ID NO 14, a splice thereof, or a variant thereof.
In certain embodiments, the variant refers to a protein that is functionally identical or similar to SEQ ID NO. 14 or a spliceosome thereof, which is obtained by substitution and/or deletion and/or addition of at least one amino acid in the amino acid sequence shown in SEQ ID NO. 14 or a spliceosome amino acid sequence thereof.
In certain embodiments, the methods of the first aspect of the invention further comprise the combination of overexpression, interference or knock-out of 1 or more other genes, including BCL-6, including GATA-3, ROR γ T, ROR α, etc., to induce Follicular Regulatory T cells (Tfr) or to further enhance the phenotype and function of tregs.
In certain embodiments, the methods further express the homing molecules CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CX3CR1, or a combination of homing molecules to promote chemotaxis of tregs in vivo to the disease site, enhancing therapeutic efficacy; preferably, the homing molecule is expressed on a viral vector or an mRNA vector.
In certain embodiments, the method further expresses or associates a molecule capable of recognizing a disease site or transplanted organ specific target, preferably, the molecule is a chimeric antigen receptor molecule, a ligand for a tissue specific receptor.
In certain embodiments, the interference or knock-out sequence, overexpressed gene, cytokine combination, may be expressed separately in combination after different vectors, or multiple genes or interference sequences may be expressed simultaneously in the same vector.
In certain embodiments, the vector comprises an adenoviral vector, an adeno-associated viral vector, a poxvirus vector, a retroviral vector, a herpes viral vector, an RNA viral vector, an EB viral vector, a baculovirus vector, a phage vector, an animal viral vector, a plant viral vector, a DNA plasmid vector, an RNA vector, preferably the vector is a DNA plasmid vector, more preferably the vector is a lentiviral vector.
In a second aspect, the invention provides the use of the regulatory T immune cells obtained by culturing according to the method of the first aspect of the invention in the preparation of a medicament for the treatment of autoimmune diseases, for inducing immune tolerance after organ transplantation.
In certain embodiments, the autoimmune disease comprises systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, myasthenia gravis, inflammatory bowel disease, type I diabetes, autoimmune hepatitis, polyneuritis, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, or the like;
in certain embodiments, the organ transplantation comprises allogenic and xenogenic heart, liver, kidney transplantation, and the like; preferably, the xenotransplantation is organ transplantation from porcine origin to humans.
In certain embodiments, the regulatory T immune cells can be used alone or in combination with other drugs.
In a third aspect, the invention provides a vector or combination of vectors for use in improving the stability of a regulatory T immune cell.
In certain embodiments, the vector or combination of vectors is selected from the following: I) a vector comprising a T-beta shRNA; II) a vector comprising a TGF- β encoding nucleic acid; III) a vector comprising a nucleic acid encoding IL-10; IV) a vector comprising both T-beta shRNA and TGF-beta encoding nucleic acid; v) a vector comprising both T-beta shRNA and IL-10 encoding nucleic acid; VI) a vector comprising both a TGF-beta encoding nucleic acid and an IL-10 encoding nucleic acid; VII) vectors comprising both T-beta shRNA, TGF-beta encoding nucleic acid, and IL-10 encoding nucleic acid; VIII) a vector combination comprising a vector of T-beta shRNA and a TGF-beta encoding nucleic acid, a vector comprising an IL-10 encoding nucleic acid; IX) a vector combination comprising a vector of T-beta shRNA and IL-10 encoding nucleic acid, a vector comprising TGF-beta encoding nucleic acid; x) vector combinations comprising T-beta shRNA vectors, vectors comprising TGF-beta encoding nucleic acid and IL-10 encoding nucleic acid; XI) vector combinations comprising T-beta shRNA vectors, vectors comprising TGF-beta encoding nucleic acids, and vectors comprising IL-10 encoding nucleic acids.
In a fourth aspect, the invention provides the use of a vector or combination of vectors according to the third aspect of the invention for inducing iTreg cells.
The invention has the beneficial effects that:
1) the single vector or the combined vector containing shRNA, mTGF-beta 1 and IL-10 targeting transcription factor T-beta is used for the naive CD4 + T induced iTreg, fine maintenance induced iTreg leave the phenotype and the functional stability of culture system after, induced iTreg has stronger immunosuppressive function than traditional induced iTreg.
2) The induction method provided by the invention can improve the induction efficiency of iTregs to a greater extent.
3) By transfecting the cell factor coding gene by the virus and carrying out exogenous expression in a host, the use amount of expensive cell factors in an induction culture system is saved, and the culture cost is reduced to the maximum extent.
4) The method provided by the invention provides higher feasibility for industrial amplification production of iTregs as clinical therapeutic cell drugs, and lays a good foundation for better application of iTregs in clinical therapy.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.
FIG. 1 characteristic phenotypic analysis of in vitro expanded iTregs. FIG. 1A shows the detection of IFN-gamma secretion in iTreg culture supernatant by ELISA method, FIG. 1B shows the iTreg expression transcription factor Foxp3 and T-beta level at day 5 of culture, FIG. 1B shows the expression of iTreg cell Tbet and Foxp3 at the upper right and lower left, respectively, and the co-expression of Tbet and Foxp3 at the lower right.
FIG. 2, shT-beta regulation iTreg effect verification. FIG. 2A is a schematic diagram of the construction of an interfering RNA (shT-beta) plasmid carrying T-beta molecules, the constructed plasmid and a plasmid expressing T-beta molecules (carrying GFP reporter genes) are co-transfected into HEK293T cells (FIG. 2B) in 6 different proportions, the left image is transfected cells under a fluorescence microscope, and the right image is the expression inhibition rate of T-beta molecules.
Flow analysis of the inhibitory effect of shT-beta lentivirus infection on transcription factor T-beta in vitro expanded iTregs is shown in FIG. 2C, where cells in mCherry positive gate in the left panel of FIG. 2C represent iTreg cells with shT-beta lentivirus infection, and cells in mCherry negative gate represent uninfected control group; FIG. 2C right shows T-beta expression in the two groups of cells. IFN-. gamma.expression in the supernatants of both groups was detected by ELISA (FIG. 2D). shT-beta regulated flow detection results of iTreg inhibiting CD8 effector T cell proliferation are shown in figure 2E, and the abscissa is iTreg and CD8 + Mixing ratio of T cells, ordinate iTreg to CD8 + Inhibition of proliferation of T cells; the expression of IFN-gamma in the supernatants of the suppressor cells detected by ELISA is shown in FIG. 2F, with the abscissa of iTreg and CD8 + T cells were mixed at different ratios.
FIG. 3 shows the effect of combined regulation of iTreg by overexpression of mTGF-beta 1 and shT-beta. The mature TGF-. beta.sequence and the shT-beta sequence were ligated in tandem on the same vector (FIG. 3A), and the group of cells transduced with the constructed plasmid was designated SH-TGF. The expression levels of the transcription factors Foxp3 (FIG. 3B) and T-beta (FIG. 3C) in cells, as well as TGF- β in cell culture supernatant (FIG. 3D), were examined at various times during iTreg culture. The time of detection of Foxp3 was day 5, day 10 and day 30 of induction culture, both day 5. The inhibitory effect of induced iTreg cells on CD8 effector T cell proliferation is shown in fig. 3E, with non-induced iTreg cells as a control.
FIG. 4 validation of the effect of over-expressing IL-10 in modulating iTreg. The construction scheme of the IL-10 overexpression plasmid is shown in FIG. 4A; on the fifth day after IL-10-carrying lentiviruses infect iTregs, the flow analysis result of the intracellular IL-10 factor expression is shown in figure 4B, and the ELISA detection result of IL-10 secretion in cell supernatant is shown in figure 4C. The results of the RTCA experiment of killing a small cell lung cancer cell line overexpressing CD19 (NCI-H292-CD19) by iTreg suppressor T cells (CD19-CART) cultured on the tenth day are shown in FIG. 4D; the inhibition of CD8 effector T cell proliferation results are shown in fig. 4E, with the abscissa representing the effective target ratio and the abscissa representing the proliferation inhibition rate.
FIG. 5 shows that the effect of the TGF-beta and IL-10 dual factor combined regulation iTreg is verified. iTreg (TGF-beta) jointly regulated by TGF-beta and IL-10 double factors&IL-10-iTreg) on CD19-CART killing NCI-H292-CD19 cells as shown in FIG. 5A, the abscissa is time and the ordinate is NCI-292 cell proliferation parameters. FIG. 5B is TGF-. beta.&The result of the experiment that the IL-10-iTreg inhibits the proliferation of CD8 effector T cells, the abscissa is the effective target ratio, and the ordinate is the proliferation inhibition rate. FIG. 5C shows TGF-. beta.&Supernatant and CD8 secreted by IL-10-iTreg + Flow detection results of T cell co-incubation and further induction of apoptosis, abscissa co-incubation time, cell proportion of mid-stage apoptosis shown on the left of FIG. 5C, and cell proportion of late-stage apoptosis shown on the right of FIG. 5C.
FIG. 6 shows that the effect of shRNA, TGF-beta and IL-10 combined regulation iTreg on GVHD intervention is verified. The GVHD model construction and iTreg treatment are shown in fig. 6A, 2.0GY indicates irradiation of mice with gamma rays of 2.0 intensity, the experiment was divided into four groups, and the four groups of mice were irradiated with the same intensity for the same time, wherein: (1) irradiation control group: a control group which was irradiated with gamma rays only and did not receive cell return; (2) model group: human PBMC (3X 10) were returned alone 6 Control group of/only) (3) iTreg control group: human PBMC (3X 10) were reinfused 6 /+ iTreg cells (6X 10) carrying empty control lentivirus infection 6 Control group of/only) (4) SH-iTreg group: human PBMC (3X 10) were reinfused 6 (6X 10) + iTreg cells carrying sh-T-beta, TGF beta and IL-10 lentivirus infection 6 Experimental group/only). Fig. 6B reflects weight loss and fig. 6C reflects survival of mice in each group. FIG. 6D shows the pathology of the day 14 mice in each group, with tissues including skin, lung, liver and small intestine.
Detailed Description
The present invention is further described below, and the embodiments described in the present description are only exemplary and do not limit the scope of the present invention. It will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention, and it is intended that all such changes and modifications be considered as within the scope of the invention.
Definition of terms
As used herein, the term "Treg" is a regulatory T cell, a subset of T cells that control autoimmune reactivity in vivo, and can be divided into naturally occurring regulatory T cells (ntregs) and inducible regulatory T cells (itrregs), such as Th3, Tr1, and further, CD8 tregs, NKT cells, etc., that are closely related to the development of autoimmune diseases.
As used herein, the term "T-beta", also known as "TBX 21(T-box transcription factor 21)" is a novel transcription factor of the T-box gene family, whose T-box binding domain contains 189 amino acids. T-beta selection is specifically expressed in Th1 cell, regulates its development and differentiation by initiating Th1 genetic program, and inhibits synthesis of Th2 type cell factor.
As used herein, the term "shRNA" is a "short hairpin RNA" comprising two short inverted repeats. The shRNA carried in the shRNA expression vector comprises two short inverted repeat sequences, the middle of the two short inverted repeat sequences is separated by a stem-loop sequence to form a hairpin structure, and the hairpin structure is controlled by a pol III promoter. Followed by 5-6 Ts as transcription terminators for RNA polymerase III. As used herein, "shT-beta" refers to a T-beta targeted shRNA.
As used herein, the term "TGF- β 1" cytokine is a member of the TGF- β family, and forms of TGF- β 1 existence include precursor molecules (per-pro-TGF- β 1), polypeptide chain precursors (pro-TGF- β 1, sometimes also referred to as LAP-TGF- β 1), and mature forms (mTGF- β 1), with the non-exclusive indication in the examples of this patent that TGF- β 1 is its mature form mTGF- β 1.
As used herein, the term "IL-10" is interleukin 10, a multi-cellular source, multifunctional cytokine that regulates the growth and differentiation of cells, participates in inflammatory and immune responses, and is a recognized inflammatory and immunosuppressive factor.
As used herein, the term "lentiviral vector" is a gene delivery vehicle based on human immunodeficiency virus type I (HIV-1), which is effective in infecting a variety of cell types, including neuronal, hepatic, cardiac, tumor, endothelial, stem cells, and the like, and has the ability to infect both dividing and non-dividing cells. The vector can effectively integrate the exogenous gene into the host chromosome, thereby achieving persistent expression.
As used herein, the term "CD 4 + T cells "are a type of T lymphocytes that can be divided into different classes or subpopulations based on the presence of a specific protein molecule on their surface. CD4+ T cells are surface CD4 + T lymphocytes of T molecules. As used herein, "naive CD4+ T cells" refers to naive CD4 + T cells, also called naive CD4+ T cells, mature in the thymus and migrate to peripheral lymphoid tissues such as spleen, lymph nodes, and the like.
As used herein, the term "ELISA" is an enzyme linked immunosorbent assay (enzyme linked immunosorbent assay) which refers to a qualitative and quantitative detection method in which soluble antigen or antibody is bound to a solid support such as polystyrene, and an immune reaction is carried out using the specific binding of antigen to antibody.
As used herein, the term "RTCA" refers to a real-time label-free dynamic cell analysis technique by integrating a microelectronic cell sensor chip into the bottom of a cell detection plate to construct a cell impedance detection sensor system that tracks changes in cell morphology and proliferation and differentiation, dynamically and quantitatively in real time. When the adherent cells growing on the surface of the microelectrode cause the change of the interfacial impedance of the adherent electrodes, the change is related to the real-time functional state change of the cells, and biological information related to the physiological functions of the cells, including cell growth, extension, morphological change, death, adherence and the like, can be obtained through real-time dynamic electrode impedance detection.
The patent is described in further detail below with reference to the accompanying drawings and specific experiments. Unless otherwise indicated, all reagents, apparatus, devices and methods used in this patent are conventional and commercially available reagents, apparatus, devices and methods in the art.
The iTreg induction and the amplification culture in the embodiment and all the embodiments of the invention are carried out according to the following methods: naive CD4 was sorted from PBMC by magnetic bead negative selection kit (Stemcell, 17555) + T cells. Obtain naive CD4 + After T cells, induction culture of iTreg was performed. The induction culture medium comprises IL-2500 IU/ml (Peprotech, AF-200-02-1MG), TGF-beta 5ng/ml (Peprotech, 100-21-10UG), rapamycin 100nM (Selleck, S1039), a basic culture medium X-VIVO-15(Lonza, 04-418Q) is added, and the induction culture medium is used for culture five days before induction; amplification medium: IL-2500 IU/ml and rapamycin 100nM are added into a basic culture medium X-VIVO-15, and after lentivirus transduction, an amplification culture medium is supplemented in half every other day; naive CD4 separated from magnetic beads + T cells were counted and induction medium was used to adjust cell density to the desired density. The cell suspension was added to a prepared 96-well plate, 50. mu.l of cell suspension per well. Approximately 110,000 and 130,000 cells per well, in a total volume of 200. mu.l. The beads were added with CD3/28 beads at a 1:1 ratio to the cells, which were then replenished once a week. After 2-3 days of culture, the growth of the cells was observed under a microscope, at which time small cell masses were observed. When cultured to a suitable time point, samples can be taken for RNA extraction or flow cytometric assay, followed by lentiviral transduction according to different experimental objectives.
Lentiviral packaging and transduction in the embodiments of the invention, all examples, was performed as follows.
And (3) slow virus packaging: freshly resuscitated HEK293T (ATCC) cells in a 6cm cell culture dish, and the cells grown in the dish at a ratio of 1:4 (about 1X 10) 24h before transfection 7 ) Transfection was initiated at a growth density of about 60% for 293T cells passaged on 10cm cell culture dishes. Half-replaced 5ml of fresh cell culture medium 2h before transfection. A clean 1.5ml EP tube (labeled tube A) was taken and 30ul of MIRUS solution was added. Another clean EP tube (labeled as tube B) was added with 30ug of a DNA mixture (containing the desired plasmid, and the structure is shown in FIG. 2A, FIG. 3A, and FIG. 4B). Mixing the liquid of tube B and tube A, standing at room temperature for 15min, adding dropwise the mixture into 10cm cell culture dish, slightly mixing, standing at 37 deg.C and 5% CO 2 Culturing in an incubator. After 6h of transfection, the medium was changed to 15ml fresh medium in a biosafety cabinet. Collecting cell culture medium containing virus 48h after transfection in 50ml centrifuge tube, centrifuging at 4 deg.C and 2500rpm for 5min, collecting supernatant, and collectingThe mixture was filtered through a 0.45 μm filter. The supernatant was centrifuged at 27000rpm for 2h at 4 ℃. After the end of the process, the supernatant was discarded, the centrifuge tube was placed upside down on a paper towel, dried in a biosafety cabinet for 5min, then the virus precipitate was dissolved in 50. mu.l of serum-free DMEM, and placed in a refrigerator at 4 ℃ overnight. HEK293T cells (12-well plate, 10) were used 5 Cells/well) for virus titer detection.
Lentivirus transduction: mixing 5E5 (5X 10) 5 ) Personal/well stimulation of day one naive CD4 + T is placed in a 96U bottom plate, and 50 ul/well of the concentrated virus solution is taken. Centrifuging for 2h at 1000g, removing supernatant in the holes, replacing new induction culture medium, and performing induction culture. The first induced iTreg, the control group infected with the unloaded plasmid lentivirus, and the experimental group infected with the shT-beta fragment plasmid (as shown in FIG. 2A, see SEQ ID NO:15) lentivirus. The second induction culture of iTreg, control infected with plasmid-free lentivirus, experimental infected with a lentivirus containing both shT-beta fragment and a plasmid containing mTGF-beta 1(SEQ ID NO:11) nucleic acid sequence (as shown in FIG. 3A). Wherein the sequence of shRNA obtained by transcription of the shT-beta fragment plasmid is shown in SEQ ID NO. 3-8. In the third batch of induced cultured iTregs, the control group was infected with lentivirus carrying plasmid, and the control group was simultaneously infected with two lentiviruses respectively containing shTbeta fragment and mTGF-beta 1 nucleic acid sequence (as shown in FIG. 3A) and plasmid carrying IL-10 nucleic acid sequence alone (as shown in FIG. 4A).
The experiments for inhibiting proliferation and killing of iTreg and the experiments for inhibiting killing are carried out according to the following methods.
Proliferation inhibition assay: separation of CD8 from human peripheral blood PBMC by magnetic bead sorting + A T cell; CD8 + T cells were stained with efluor670 (Thermofeisher, 65-0840-90) proliferation fuel; CD3/28 stimulation magnetic beads and CD8 + T cells are stimulated by mixing 1:10 in a system of 100 mu L in a 5E 4/hole and 96U bottom hole plate; 50 mu L of diluted iTreg with the same ratio as the iTreg and CD8 are added into each hole + T is 1:1, 1:2,1:4, 1:8, 1:16, 1:32, 1:64, 1: 128; in addition, a hole without magnetic bead stimulation CD8 is separated + T cells, and magnetic bead-alone stimulated CD8 + T cells, 150 ul/well; after 5 days of incubation, CD4 flow staining was performed, and flow staining was performedAnd (4) detecting.
Inhibition and killing experiment: the small cell lung cancer cell line NCI-H292 overexpressing CD19 was plated in RTCA well plates at 1E 4/well; effector cells chimeric to the CD19 antigen receptor (CD19-CART) were incubated with iTreg cells for 24 hours at a cell-to-cell ratio of: CD 19-CART: NCI-H292-CD19 cells were 2:1, the ratio of iTreg cells to CD19-CART cells was 1:2,1:4, 1: 8; the cells that completed the co-incubation were added together to the RTCA well plates plated with NCI-H292-CD19 cells and the data results were analyzed after 24 hours.
Example 1 characteristic phenotypic analysis of in vitro expanded iTregs
According to the characteristic that iTregs are unstable under the stimulation of inflammatory environment, the inflammatory environment is simulated by adding 50ng/ml IL-6 proinflammatory cytokines into an iTreg in-vitro amplification system, and IFN-gamma secreted by the iTregs is detected by an ELISA method, and the result is shown in figure 1A. One week after induction, the expression of transcription factors such as Foxp3, Tbet, etc. of iTreg was detected by flow assay, as shown in FIG. 1B. From the results in fig. 1, it can be seen that the conventionally induced iTreg can secrete a certain amount of IFN- γ at the initial stage of induction, and there are a large population of cells in which Foxp3 and Tbet are co-expressed, which further verifies the instability deep molecular mechanism of the iTreg that it is prone to polarization to Th1 pro-inflammatory cells in an inflammatory environment and further loses Foxp3 expression.
Example 2 shT-beta Regulation iTreg Effect verification
Aiming at the unstable factor that iTreg is easy to polarize towards Th1 or secrete Th 1-class related proinflammatory cytokines in an inflammatory environment, a hairpin-structure interference RNA (shT-beta) aiming at a Th1 characteristic transcription factor Tbeta gene is constructed, and the interference effect of shT-beta is indicated by using a reporter gene mCherry shown in figure 2A. shT-beta plasmid and T-beta plasmid with EGFP reporter gene are jointly transfected into HEK293T cells, if shT-beta has interference effect on T-beta target sequence, the EGFP protein expression strength of the T-beta expression level can be directly reported to indicate, so that the interference efficiency of shT-beta on T-beta is verified, and the constructed shT-beta has strong inhibition effect on exogenous over-expressed T-beta as shown in figure 2B.
For further verification of shT-beta internal sourceT-beta inhibitory Effect of sex, we will sort the negative CD4 + The following day of induction of T cells into iTreg and stimulation with magnetic beads, infecting the lentivirus with shT-beta vector by the lentivirus system, removing the magnetic beads on the third day of stimulation, and detecting the expression level of T-beta by flow-type after culturing for 5 days, the result is shown in FIG. 2C, wherein GFP is used as the reference + The subpopulation indicates cells successfully expressing shT-beta interfering RNA, GFP - The subgroup indicates cells which do not successfully express shT-beta interfering RNA, the result in the figure shows that shTbeta also has a strong inhibitory effect on endogenous T-beta, and the situation that IFN-gamma is secreted in iTreg supernatant is further detected by ELISA, as shown in figure 2D, the shT-beta interfering group hardly secretes IFN-gamma.
To further verify the functional superiority of T-beta interference, shT-beta-iTreg and WT-iTreg are subjected to inhibition tests, and the results show that the shT-beta-iTreg inhibition effect is superior (FIG. 2E), and the amount of IFN-gamma secreted by iTreg under the inflammatory environment is obviously reduced (FIG. 2F).
Example 3 verification of the Effect of over-expressing mTGF-beta 1 and shT-beta combined regulation iTreg
We designed a nucleic acid sequence expressing TGF-beta 1 mature peptide, which is composed of signal peptide and TGF-beta 1 mature peptide and named as mTGF-beta, and constructed the nucleic acid sequence on shT-beta expression vector (figure 3A), and tested the stable expression maintaining effect of mTGF-beta on Foxp3, the long-term stable expression of mTGF-beta on Foxp3, and after 5 days of infecting over-expression virus vector, remove TGF-beta 1 cell factor in culture medium system, and cultured for one month, compared with WT group, the mTGF-beta group has obvious maintaining advantage on Foxp3 stable expression, and the result is shown in figure 3B. Furthermore, the shTbet inhibition effect of the flow detection is more remarkable (fig. 4C). ELISA detected significant increase in mTGF- β overexpression levels (fig. 3D). And the mTGF-beta overexpression is improved on the iTreg inhibition function, and the result is shown in figure 3E.
Example 4 validation of the Effect of over-expressing IL-10 regulatory iTregs
To further improve the inhibitory ability of iTreg, we cloned the CDS region of IL-10 gene into a lentiviral vector and constructed as shown in fig. 4A. And after successful transduction and induction of iTreg cells for one week, IL-10 expression was detected by ELISA (fig. 4B) and flow assay (fig. 4C). The result shows that the IL-10 expression vector has a good overexpression effect, the functional effect of over-expression IL-10 generation is further verified, an RTCA inhibition and killing test is carried out, the result is shown in 4D, the traditional induced iTreg has almost no effect of inhibiting the specificity killing of allogenic CAR-T, and the iTreg group of over-expression IL-10 has a good inhibition and killing effect. Moreover, the ability of the iTreg cells overexpressing IL-10 to suppress the proliferation of effector cells was also significantly improved (FIG. 4E).
Example 5 Effect verification of TGF-beta and IL-10 double-factor combined regulation iTreg
In order to further improve the stability and the inhibition effect of iTreg, the CDS region of the IL-10 gene and the mTGF-beta gene are jointly cloned on a lentiviral vector, and lentiviral transduction is carried out to obtain TGF-beta of a co-expression double cell factor&IL-10-iTreg, RTCA inhibitory killing assay showed (FIG. 5A), TGF-beta specific killing effect of inhibiting allogeneic CAR-T compared to traditionally induced iTreg cells&The IL-10-iTreg group had significantly greater inhibitory potency. We performed proliferation inhibition experiments with the effective target ratios 1:64 and 1:128, as shown in FIG. 5B, from CD8 + According to the experimental result of T cell proliferation inhibition, the two-factor group has obvious inhibition function advantage, and meanwhile, in order to detect the iTreg paracrine function of over-expressing IL-10 and TGF-beta, the iTreg cells are cultured for 3 days, and supernatant and CD8 are obtained + T cells were co-incubated and tested for apoptosis, as shown in FIG. 5C, and we found TGF-. beta.&The IL-10-iTreg group entered metaphase apoptosis at the earliest day 3, TGF-. beta.at day 5&The late apoptosis of the IL-10-iTreg group is obviously higher than that of the control group, which shows that the combination of two factors greatly improves the lateral secretion inhibition function of the iTreg.
Example 6 validation of Effect of shRNA, TGF-beta and IL-10 combined regulation iTreg on intervention GVHD
The SH-iTreg induced by T-beta interference, TGF beta and IL-10 overexpression method and the traditional control iTreg in the patent are used for evaluating the treatment effect of the mouse graft-versus-host (GVHD), and fig. 6A shows the construction of a GVHD model and the iTreg treatment method.
The specific implementation steps are as follows: in the orbital veinIrradiating NSG mice with 2.0G gamma rays 1 day before injection, and weighing the first weight; the normal group is a group which only receives irradiation and does not return cells; PBMC (3X 10) from human peripheral blood mononuclear cells 6 Mice) and iTreg (6X 10) 6 Mice) mixed injection; weigh and record GVHD symptoms every other day until day 20; peripheral blood was drawn on days 7, 14 and one month for flow staining; 3 mice per group were sacrificed on day 14 and skin, lung, liver and small intestine were taken for immunohistochemical staining. The weight and survival rate of the model group only transfused with PBMC and the iTreg control group are in obvious descending trend, the GVHD mice treated by the SH-iTreg group induced by the method provided by the invention have lower weight descending rate (figure 6B), and the survival condition of the mice is obviously improved (figure 6C).
Model group mice infused with PBMC showed significant GVHD, with lymphocyte infiltration in the liver and lung, severe atrophy of small intestine villi, severe discontinuation of skin basal layer cells, lymphocyte infiltration in the liver and lung of iTreg control group, even more severe small intestine villi, severe separation of skin layers, severe discontinuation of basal layer cells, depletion of adipose layer, SH-iTreg showed the lightest pathological symptoms of GVHD, as shown in fig. 6D.
The above description is only a preferred embodiment of the present invention, and is not intended to limit the present invention, and any simple modifications, changes and equivalent structural changes made to the above embodiment according to the technical essence of the present invention still fall within the protection scope of the technical solution of the present invention.
SEQUENCE LISTING
<110> Zhongshan Hospital affiliated to Fudan university
<120> a method for improving the stability and function of regulatory T cells
<130> 202203
<160> 15
<170> PatentIn version 3.3
<210> 1
<211> 2583
<212> DNA
<213> Homo sapiens
<400> 1
agtgacagcg gcccgctgga gaggaagccc gagagctgcc gcgcgcctgc cggacgaggg 60
cgtagaagcc aggcgtcaga gcccgggctc cggtggggtc ccccacccgg ccctcgggtc 120
ccccgccccc tgctccctgc ccatcccagc ccacgcgacc ctctcgcgcg cggaggggcg 180
ggtcctcgac ggctacggga aggtgccagc ccgccccgga tgggcatcgt ggagccgggt 240
tgcggagaca tgctgacggg caccgagccg atgccgggga gcgacgaggg ccgggcgcct 300
ggcgccgacc cgcagcaccg ctacttctac ccggagccgg gcgcgcagga cgcggacgag 360
cgtcgcgggg gcggcagcct ggggtctccc tacccggggg gcgccttggt gcccgccccg 420
ccgagccgct tccttggagc ctacgcctac ccgccgcgac cccaggcggc cggcttcccc 480
ggcgcgggcg agtccttccc gccgcccgcg gacgccgagg gctaccagcc gggcgagggc 540
tacgccgccc cggacccgcg cgccgggctc tacccggggc cgcgtgagga ctacgcgcta 600
cccgcgggac tggaggtgtc ggggaaactg agggtcgcgc tcaacaacca cctgttgtgg 660
tccaagttta atcagcacca gacagagatg atcatcacca agcagggacg gcggatgttc 720
ccattcctgt catttactgt ggccgggctg gagcccacca gccactacag gatgtttgtg 780
gacgtggtct tggtggacca gcaccactgg cggtaccaga gcggcaagtg ggtgcagtgt 840
ggaaaggccg agggcagcat gccaggaaac cgcctgtacg tccacccgga ctcccccaac 900
acaggagcgc actggatgcg ccaggaagtt tcatttggga aactaaagct cacaaacaac 960
aagggggcgt ccaacaatgt gacccagatg attgtgctcc agtccctcca taagtaccag 1020
ccccggctgc atatcgttga ggtgaacgac ggagagccag aggcagcctg caacgcttcc 1080
aacacgcata tctttacttt ccaagaaacc cagttcattg ccgtgactgc ctaccagaat 1140
gccgagatta ctcagctgaa aattgataat aacccctttg ccaaaggatt ccgggagaac 1200
tttgagtcca tgtacacatc tgttgacacc agcatcccct ccccgcctgg acccaactgt 1260
caattccttg ggggagatca ctactctcct ctcctaccca accagtatcc tgttcccagc 1320
cgcttctacc ccgaccttcc tggccaggcg aaggatgtgg ttccccaggc ttactggctg 1380
ggggcccccc gggaccacag ctatgaggct gagtttcgag cagtcagcat gaagcctgca 1440
ttcttgccct ctgcccctgg gcccaccatg tcctactacc gaggccagga ggtcctggca 1500
cctggagctg gctggcctgt ggcaccccag taccctccca agatgggccc ggccagctgg 1560
ttccgcccta tgcggactct gcccatggaa cccggccctg gaggctcaga gggacgggga 1620
ccagaggacc agggtccccc cttggtgtgg actgagattg cccccatccg gccggaatcc 1680
agtgattcag gactgggcga aggagactct aagaggaggc gcgtgtcccc ctatccttcc 1740
agtggtgaca gctcctcccc tgctggggcc ccttctcctt ttgataagga agctgaagga 1800
cagttttata actattttcc caactgagca gatgacatga tgaaaggaac agaaacagtg 1860
ttattaggtt ggaggacacc gactaatttg ggaaacggat gaaggactga gaaggccccc 1920
gctccctctg gcccttctct gtttagtagt tggttgggga agtggggctc aagaaggatt 1980
ttggggttca ccagatgctt cctggcccac gatgaaacct gagaggggtg tccccttgcc 2040
ccatcctctg ccctaactac agtcgtttac ctggtgctgc gtcttgcttt tggtttccag 2100
ctggagaaaa gaagacaaga aagtcttggg catgaaggag ctttttgcat ctagtgggtg 2160
ggaggggtca ggtgtgggac atgggagcag gagactccac tttcttcctt tgtacagtaa 2220
ctttcaacct tttcgttggc atgtgtgtta atccctgatc caaaaagaac aaatacacgt 2280
atgttataac catcagcccg ccagggtcag ggaaaggact cacctgactt tggacagctg 2340
gcctgggctc cccctgctca aacacagtgg ggatcagaga aaaggggctg gaaagggggg 2400
aatggcccac atctcaagaa gcaagatatt gtttgtggtg gttgtgtgtg ggtgtgtgtt 2460
ttttcttttt ctttcttttt attttttttg aatgggggag gctatttatt gtactgagag 2520
tggtgtctgg atatattcct tttgtcttca tcactttctg aaaataaaca taaaactgtt 2580
gaa 2583
<210> 2
<211> 535
<212> PRT
<213> Homo sapiens
<400> 2
Met Gly Ile Val Glu Pro Gly Cys Gly Asp Met Leu Thr Gly Thr Glu
1 5 10 15
Pro Met Pro Gly Ser Asp Glu Gly Arg Ala Pro Gly Ala Asp Pro Gln
20 25 30
His Arg Tyr Phe Tyr Pro Glu Pro Gly Ala Gln Asp Ala Asp Glu Arg
35 40 45
Arg Gly Gly Gly Ser Leu Gly Ser Pro Tyr Pro Gly Gly Ala Leu Val
50 55 60
Pro Ala Pro Pro Ser Arg Phe Leu Gly Ala Tyr Ala Tyr Pro Pro Arg
65 70 75 80
Pro Gln Ala Ala Gly Phe Pro Gly Ala Gly Glu Ser Phe Pro Pro Pro
85 90 95
Ala Asp Ala Glu Gly Tyr Gln Pro Gly Glu Gly Tyr Ala Ala Pro Asp
100 105 110
Pro Arg Ala Gly Leu Tyr Pro Gly Pro Arg Glu Asp Tyr Ala Leu Pro
115 120 125
Ala Gly Leu Glu Val Ser Gly Lys Leu Arg Val Ala Leu Asn Asn His
130 135 140
Leu Leu Trp Ser Lys Phe Asn Gln His Gln Thr Glu Met Ile Ile Thr
145 150 155 160
Lys Gln Gly Arg Arg Met Phe Pro Phe Leu Ser Phe Thr Val Ala Gly
165 170 175
Leu Glu Pro Thr Ser His Tyr Arg Met Phe Val Asp Val Val Leu Val
180 185 190
Asp Gln His His Trp Arg Tyr Gln Ser Gly Lys Trp Val Gln Cys Gly
195 200 205
Lys Ala Glu Gly Ser Met Pro Gly Asn Arg Leu Tyr Val His Pro Asp
210 215 220
Ser Pro Asn Thr Gly Ala His Trp Met Arg Gln Glu Val Ser Phe Gly
225 230 235 240
Lys Leu Lys Leu Thr Asn Asn Lys Gly Ala Ser Asn Asn Val Thr Gln
245 250 255
Met Ile Val Leu Gln Ser Leu His Lys Tyr Gln Pro Arg Leu His Ile
260 265 270
Val Glu Val Asn Asp Gly Glu Pro Glu Ala Ala Cys Asn Ala Ser Asn
275 280 285
Thr His Ile Phe Thr Phe Gln Glu Thr Gln Phe Ile Ala Val Thr Ala
290 295 300
Tyr Gln Asn Ala Glu Ile Thr Gln Leu Lys Ile Asp Asn Asn Pro Phe
305 310 315 320
Ala Lys Gly Phe Arg Glu Asn Phe Glu Ser Met Tyr Thr Ser Val Asp
325 330 335
Thr Ser Ile Pro Ser Pro Pro Gly Pro Asn Cys Gln Phe Leu Gly Gly
340 345 350
Asp His Tyr Ser Pro Leu Leu Pro Asn Gln Tyr Pro Val Pro Ser Arg
355 360 365
Phe Tyr Pro Asp Leu Pro Gly Gln Ala Lys Asp Val Val Pro Gln Ala
370 375 380
Tyr Trp Leu Gly Ala Pro Arg Asp His Ser Tyr Glu Ala Glu Phe Arg
385 390 395 400
Ala Val Ser Met Lys Pro Ala Phe Leu Pro Ser Ala Pro Gly Pro Thr
405 410 415
Met Ser Tyr Tyr Arg Gly Gln Glu Val Leu Ala Pro Gly Ala Gly Trp
420 425 430
Pro Val Ala Pro Gln Tyr Pro Pro Lys Met Gly Pro Ala Ser Trp Phe
435 440 445
Arg Pro Met Arg Thr Leu Pro Met Glu Pro Gly Pro Gly Gly Ser Glu
450 455 460
Gly Arg Gly Pro Glu Asp Gln Gly Pro Pro Leu Val Trp Thr Glu Ile
465 470 475 480
Ala Pro Ile Arg Pro Glu Ser Ser Asp Ser Gly Leu Gly Glu Gly Asp
485 490 495
Ser Lys Arg Arg Arg Val Ser Pro Tyr Pro Ser Ser Gly Asp Ser Ser
500 505 510
Ser Pro Ala Gly Ala Pro Ser Pro Phe Asp Lys Glu Ala Glu Gly Gln
515 520 525
Phe Tyr Asn Tyr Phe Pro Asn
530 535
<210> 3
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 3
gtggaggtgt gtgaagtta 19
<210> 4
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 4
taacttcaca cacctccac 19
<210> 5
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 5
tgctcgaatt gacagaaaa 19
<210> 6
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 6
ttttctgtca attcgagca 19
<210> 7
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 7
catccttcga gttgaatat 19
<210> 8
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 8
atattcaact cgaaggatg 19
<210> 9
<211> 1173
<212> DNA
<213> Homo sapiens
<400> 9
atgccgccct ccgggctgcg gctgctgccg ctgctgctac cgctgctgtg gctactggtg 60
ctgacgcctg gccggccggc cgcgggacta tccacctgca agactatcga catggagctg 120
gtgaagcgga agcgcatcga ggccatccgc ggccagatcc tgtccaagct gcggctcgcc 180
agccccccga gccaggggga ggtgccgccc ggcccgctgc ccgaggccgt gctcgccctg 240
tacaacagca cccgcgaccg ggtggccggg gagagtgcag aaccggagcc cgagcctgag 300
gccgactact acgccaagga ggtcacccgc gtgctaatgg tggaaaccca caacgaaatc 360
tatgacaagt tcaagcagag tacacacagc atatatatgt tcttcaacac atcagagctc 420
cgagaagcgg tacctgaacc cgtgttgctc tcccgggcag agctgcgtct gctgaggctc 480
aagttaaaag tggagcagca cgtggagctg taccagaaat acagcaacaa ttcctggcga 540
tacctcagca accggctgct ggcacccagc gactcgccag agtggttatc ttttgatgtc 600
accggagttg tgcggcagtg gttgagccgt ggaggggaaa ttgagggctt tcgccttagc 660
gcccactgct cctgtgacag cagggataac acactgcaag tggacatcaa cgggttcact 720
accggccgcc gaggtgacct ggccaccatt catggcatga accggccttt cctgcttctc 780
atggccaccc cgctggagag ggcccagcat ctgcaaagct cccggcaccg ccgagccctg 840
gacaccaact attgcttcag ctccacggag aagaactgct gcgtgcggca gctgtacatt 900
gacttccgca aggacctcgg ctggaagtgg atccacgagc ccaagggcta ccatgccaac 960
ttctgcctcg ggccctgccc ctacatttgg agcctggaca cgcagtacag caaggtcctg 1020
gccctgtaca accagcataa cccgggcgcc tcggcggcgc cgtgctgcgt gccgcaggcg 1080
ctggagccgc tgcccatcgt gtactacgtg ggccgcaagc ccaaggtgga gcagctgtcc 1140
aacatgatcg tgcgctcctg caagtgcagc tga 1173
<210> 10
<211> 390
<212> PRT
<213> Homo sapiens
<400> 10
Met Pro Pro Ser Gly Leu Arg Leu Leu Pro Leu Leu Leu Pro Leu Leu
1 5 10 15
Trp Leu Leu Val Leu Thr Pro Gly Arg Pro Ala Ala Gly Leu Ser Thr
20 25 30
Cys Lys Thr Ile Asp Met Glu Leu Val Lys Arg Lys Arg Ile Glu Ala
35 40 45
Ile Arg Gly Gln Ile Leu Ser Lys Leu Arg Leu Ala Ser Pro Pro Ser
50 55 60
Gln Gly Glu Val Pro Pro Gly Pro Leu Pro Glu Ala Val Leu Ala Leu
65 70 75 80
Tyr Asn Ser Thr Arg Asp Arg Val Ala Gly Glu Ser Ala Glu Pro Glu
85 90 95
Pro Glu Pro Glu Ala Asp Tyr Tyr Ala Lys Glu Val Thr Arg Val Leu
100 105 110
Met Val Glu Thr His Asn Glu Ile Tyr Asp Lys Phe Lys Gln Ser Thr
115 120 125
His Ser Ile Tyr Met Phe Phe Asn Thr Ser Glu Leu Arg Glu Ala Val
130 135 140
Pro Glu Pro Val Leu Leu Ser Arg Ala Glu Leu Arg Leu Leu Arg Leu
145 150 155 160
Lys Leu Lys Val Glu Gln His Val Glu Leu Tyr Gln Lys Tyr Ser Asn
165 170 175
Asn Ser Trp Arg Tyr Leu Ser Asn Arg Leu Leu Ala Pro Ser Asp Ser
180 185 190
Pro Glu Trp Leu Ser Phe Asp Val Thr Gly Val Val Arg Gln Trp Leu
195 200 205
Ser Arg Gly Gly Glu Ile Glu Gly Phe Arg Leu Ser Ala His Cys Ser
210 215 220
Cys Asp Ser Arg Asp Asn Thr Leu Gln Val Asp Ile Asn Gly Phe Thr
225 230 235 240
Thr Gly Arg Arg Gly Asp Leu Ala Thr Ile His Gly Met Asn Arg Pro
245 250 255
Phe Leu Leu Leu Met Ala Thr Pro Leu Glu Arg Ala Gln His Leu Gln
260 265 270
Ser Ser Arg His Arg Arg Ala Leu Asp Thr Asn Tyr Cys Phe Ser Ser
275 280 285
Thr Glu Lys Asn Cys Cys Val Arg Gln Leu Tyr Ile Asp Phe Arg Lys
290 295 300
Asp Leu Gly Trp Lys Trp Ile His Glu Pro Lys Gly Tyr His Ala Asn
305 310 315 320
Phe Cys Leu Gly Pro Cys Pro Tyr Ile Trp Ser Leu Asp Thr Gln Tyr
325 330 335
Ser Lys Val Leu Ala Leu Tyr Asn Gln His Asn Pro Gly Ala Ser Ala
340 345 350
Ala Pro Cys Cys Val Pro Gln Ala Leu Glu Pro Leu Pro Ile Val Tyr
355 360 365
Tyr Val Gly Arg Lys Pro Lys Val Glu Gln Leu Ser Asn Met Ile Val
370 375 380
Arg Ser Cys Lys Cys Ser
385 390
<210> 11
<211> 423
<212> DNA
<213> Homo sapiens
<400> 11
atgccaccta gcggcctgag actgctccca ctgctcctcc ctctcctctg gctcctggtg 60
ctcacacccg gcagaccagc cgccggcctc gacacaaact actgcttctc tagcaccgag 120
aagaactgct gcgtgagaca gctgtacatt gacttccgca aggacctggg ctggaagtgg 180
attcacgagc ctaagggcta ccacgccaac ttctgcctgg gcccttgccc atacatttgg 240
agcctcgaca cacagtactc taaggtgctc gccctgtaca accagcacaa ccccggcgcc 300
agcgccgccc cttgctgcgt gcctcaggcc ctcgaaccac tgcctatcgt gtactacgtg 360
ggcaggaagc ctaaggtgga gcagctgtct aacatgatcg tgcggtcttg caagtgctct 420
tga 423
<210> 12
<211> 141
<212> PRT
<213> Homo sapiens
<400> 12
Met Pro Pro Ser Gly Leu Arg Leu Leu Pro Leu Leu Leu Pro Leu Leu
1 5 10 15
Trp Leu Leu Val Leu Thr Pro Gly Arg Pro Ala Ala Gly Ala Leu Asp
20 25 30
Thr Asn Tyr Cys Phe Ser Ser Thr Glu Lys Asn Cys Cys Val Arg Gln
35 40 45
Leu Tyr Ile Asp Phe Arg Lys Asp Leu Gly Trp Lys Trp Ile His Glu
50 55 60
Pro Lys Gly Tyr His Ala Asn Phe Cys Leu Gly Pro Cys Pro Tyr Ile
65 70 75 80
Trp Ser Leu Asp Thr Gln Tyr Ser Lys Val Leu Ala Leu Tyr Asn Gln
85 90 95
His Asn Pro Gly Ala Ser Ala Ala Pro Cys Cys Val Pro Gln Ala Leu
100 105 110
Glu Pro Leu Pro Ile Val Tyr Tyr Val Gly Arg Lys Pro Lys Val Glu
115 120 125
Gln Leu Ser Asn Met Ile Val Arg Ser Cys Lys Cys Ser
130 135 140
<210> 13
<211> 537
<212> DNA
<213> Homo sapiens
<400> 13
atgcacagct cagcactgct ctgttgcctg gtcctcctga ctggggtgag ggccagccca 60
ggccagggca cccagtctga gaacagctgc acccacttcc caggcaacct gcctaacatg 120
cttcgagatc tccgagatgc cttcagcaga gtgaagactt tctttcaaat gaaggatcag 180
ctggacaact tgttgttaaa ggagtccttg ctggaggact ttaagggtta cctgggttgc 240
caagccttgt ctgagatgat ccagttttac ctggaggagg tgatgcccca agctgagaac 300
caagacccag acatcaaggc gcatgtgaac tccctggggg agaacctgaa gaccctcagg 360
ctgaggctac ggcgctgtca tcgatttctt ccctgtgaaa acaagagcaa ggccgtggag 420
caggtgaaga atgcctttaa taagctccaa gagaaaggca tctacaaagc catgagtgag 480
tttgacatct tcatcaacta catagaagcc tacatgacaa tgaagatacg aaactga 537
<210> 14
<211> 178
<212> PRT
<213> Homo sapiens
<400> 14
Met His Ser Ser Ala Leu Leu Cys Cys Leu Val Leu Leu Thr Gly Val
1 5 10 15
Arg Ala Ser Pro Gly Gln Gly Thr Gln Ser Glu Asn Ser Cys Thr His
20 25 30
Phe Pro Gly Asn Leu Pro Asn Met Leu Arg Asp Leu Arg Asp Ala Phe
35 40 45
Ser Arg Val Lys Thr Phe Phe Gln Met Lys Asp Gln Leu Asp Asn Leu
50 55 60
Leu Leu Lys Glu Ser Leu Leu Glu Asp Phe Lys Gly Tyr Leu Gly Cys
65 70 75 80
Gln Ala Leu Ser Glu Met Ile Gln Phe Tyr Leu Glu Glu Val Met Pro
85 90 95
Gln Ala Glu Asn Gln Asp Pro Asp Ile Lys Ala His Val Asn Ser Leu
100 105 110
Gly Glu Asn Leu Lys Thr Leu Arg Leu Arg Leu Arg Arg Cys His Arg
115 120 125
Phe Leu Pro Cys Glu Asn Lys Ser Lys Ala Val Glu Gln Val Lys Asn
130 135 140
Ala Phe Asn Lys Leu Gln Glu Lys Gly Ile Tyr Lys Ala Met Ser Glu
145 150 155 160
Phe Asp Ile Phe Ile Asn Tyr Ile Glu Ala Tyr Met Thr Met Lys Ile
165 170 175
Arg Asn
<210> 15
<211> 911
<212> DNA
<213> Homo sapiens
<400> 15
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
ccgcgcgatc cgtggaggtg tgtgaagtta ttcaagagat aacttcacac acctccactt 300
ttttgggagg gcctatttcc catgattcct tcatatttgc atatacgata caaggctgtt 360
agagagataa ttggaattaa tttgactgta aacacaaaga tattagtaca aaatacgtga 420
cgtagaaagt aataatttct tgggtagttt gcagttttaa aattatgttt taaaatggac 480
tatcatatgc ttaccgtaac ttgaaagtat ttcgatttct tggctttata tatcttgtgg 540
aaaggaccca gcgatcctgc tcgaattgac agaaaattca agagattttc tgtcaattcg 600
agcatttttt ggagggccta tttcccatga ttccttcata tttgcatata cgatacaagg 660
ctgttagaga gataattgga attaatttga ctgtaaacac aaagatatta gtacaaaata 720
cgtgacgtag aaagtaataa tttcttgggt agtttgcagt tttaaaatta tgttttaaaa 780
tggactatca tatgcttacc gtaacttgaa agtatttcga tttcttggct ttatatatct 840
tgtggaaagg acgatcccat ccttcgagtt gaatatttca agagaatatt caactcgaag 900
gatgtttttt a 911

Claims (16)

1. A method for increasing induction, phenotypic stability and immunomodulatory function of a regulatory T immune cell comprising inhibiting expression of transcription factor T-beta or knocking out TBX21 gene.
2. The method of claim 1, wherein the nucleic acid encoding the transcription factor T-beta comprises SEQ ID NO 1, a degenerate sequence thereof, or a spliceosome nucleotide sequence thereof.
3. The method of claims 1-2, wherein the transcription factor T-beta protein comprises the amino acid sequence of SEQ ID No. 2, a spliceosome thereof, or a variant thereof.
4. The method of claim 1, wherein the method or product for inhibiting the expression of the transcription factor T-beta is selected from the group consisting of small RNAs or small RNAs of multi-target multi-hairpin structures, ribozymes, gene targeting, protein inhibitors and compound inhibitors; preferably, siRNA interference through hairpin structure; more preferably, the siRNA has a sequence shown in SEQ ID NO 3-8.
5. The method according to claim 1, wherein the method or product for knocking out the TBX21 gene is selected from CRISPR/Cas9 technology, zinc finger nucleases, TALENs.
6. The method of claims 1-5, further comprising the step of adding or overexpressing a combination of one or more cytokines.
7. The method of claim 6, wherein the cytokine is selected from the group consisting of natural or recombinant interleukins, interferons, colony stimulating factors, erythropoietins, epidermal growth factors, nerve growth factors, basic fibroblast growth factors, tumor necrosis factors, thrombopoietin, platelet-derived growth factors, stem cell factors, vascular endothelial growth factors; preferably, the additional or overexpressed cytokine is TGF- β, IL-10, or a combination thereof.
8. The method according to claim 7, wherein the TGF- β is selected from TGF- β 1, TGF- β 2, TGF- β 3 and their corresponding precursor molecules (per-pro-TGF- β), polypeptide chain precursors (pro-TGF- β) and mature forms (mTGF- β); preferably, the TGF-beta is TGF-beta 1, the encoding nucleic acid of the TGF-beta 1 comprises SEQ ID NO 9, a degenerate sequence thereof or a spliceosome nucleotide sequence thereof, and the TGF-beta 1 protein comprises an amino acid sequence of SEQ ID NO 10, a spliceosome or a variant thereof; more preferably, the TGF- β is mTGF- β 1, the mTGF- β 1 encoding nucleic acid comprises SEQ ID NO 11, a degenerate sequence thereof, or a splice nucleotide sequence thereof, and the mTGF- β 1 protein comprises SEQ ID NO 12, a splice thereof, or a variant amino acid sequence thereof.
9. The method according to claim 7, characterized in that said IL-10 encoding nucleic acid has the nucleotide sequence of SEQ ID NO 13, a degenerate sequence thereof or a spliceosome thereof, and said IL-10 protein comprises the amino acid sequence shown in SEQ ID NO 14, a spliceosome thereof or a variant thereof.
10. The method of any one of claims 1-8, further comprising overexpression, interference, or knock-out of 1 or more additional genes in combination, wherein the overexpressed genes comprise BCL-6 and the interfered or knocked-out genes comprise GATA-3, ROR γ t, ROR α.
11. The method of any one of claims 1 to 10, wherein the method further expresses the homing molecule CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CX3CR1 or a combination of homing molecules to promote Treg chemotaxis in vivo to the disease site and to increase the therapeutic effect; preferably, the homing molecule is expressed on a viral vector or an mRNA vector.
12. The method according to any one of claims 1 to 11, wherein the method further expresses or links a molecule capable of recognizing a disease site or transplanted organ specific target, preferably wherein the molecule is a chimeric antigen receptor molecule, a ligand for a tissue specific receptor.
13. The method of any one of claims 1-12, wherein the interfering or knockout sequences, overexpressed genes, cytokine combinations, can be expressed separately in combination after different vectors, or multiple genes or interfering sequences can be expressed simultaneously in the same vector; the vector comprises an adenovirus vector, an adeno-associated virus vector, a pox virus vector, a retrovirus vector, a herpes virus vector, an RNA virus vector, an EB virus vector, a baculovirus vector, a phage vector, an animal virus vector, a plant virus vector, a DNA plasmid vector and an RNA vector, preferably the vector is a DNA plasmid vector, and more preferably the vector is a lentivirus vector.
14. Use of the regulatory T immune cells cultured according to the method of claims 1-13 for the preparation of a medicament for the treatment of autoimmune diseases, inducing immune tolerance after organ transplantation, wherein the autoimmune diseases include systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, myasthenia gravis, inflammatory bowel disease, type I diabetes, autoimmune hepatitis, polyneuritis, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia; the organ transplantation comprises allogenic and xenogenic heart, liver and kidney transplantation; the regulatory T immune cells can be used alone or in combination with other drugs.
15. A vector or combination of vectors for use in increasing the stability of a regulatory T immune cell, wherein said vector or combination of vectors is selected from the group consisting of: I) a vector comprising a T-beta shRNA; II) a vector comprising a TGF- β encoding nucleic acid; III) a vector comprising a nucleic acid encoding IL-10; IV) a vector comprising both T-beta shRNA and TGF-beta encoding nucleic acid; v) a vector comprising both T-beta shRNA and IL-10 encoding nucleic acid; VI) a vector comprising both a TGF-beta encoding nucleic acid and an IL-10 encoding nucleic acid; VII) vectors comprising both T-beta shRNA, TGF-beta encoding nucleic acid, and IL-10 encoding nucleic acid; VIII) a vector combination comprising a vector of T-beta shRNA and a TGF-beta encoding nucleic acid, a vector comprising an IL-10 encoding nucleic acid; IX) a vector combination comprising a vector of T-beta shRNA and IL-10 encoding nucleic acid, a vector comprising TGF-beta encoding nucleic acid; x) vector combinations comprising first a T-beta shRNA vector, a vector comprising a TGF-beta encoding nucleic acid and an IL-10 encoding nucleic acid; XI) vector combinations comprising T-beta shRNA vectors, vectors comprising TGF-beta encoding nucleic acids, and vectors comprising IL-10 encoding nucleic acids.
16. Use of the vector or vector combination of claim 15 for inducing T immune cells of the regulatory type.
CN202210407362.7A 2022-04-18 2022-04-18 Method for improving stability and function of regulatory T cells Pending CN114958921A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210407362.7A CN114958921A (en) 2022-04-18 2022-04-18 Method for improving stability and function of regulatory T cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210407362.7A CN114958921A (en) 2022-04-18 2022-04-18 Method for improving stability and function of regulatory T cells

Publications (1)

Publication Number Publication Date
CN114958921A true CN114958921A (en) 2022-08-30

Family

ID=82977692

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210407362.7A Pending CN114958921A (en) 2022-04-18 2022-04-18 Method for improving stability and function of regulatory T cells

Country Status (1)

Country Link
CN (1) CN114958921A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101585823A (en) * 2009-06-09 2009-11-25 华中科技大学 New immunosuppressive agent extracted from Honghuoma and extraction method and use thereof
CN103314012A (en) * 2010-08-20 2013-09-18 李尚揆 Fusion protein having transcription factor transactivation-regulating domain and protein transduction domain, and transcription factor function inhibitor comprising the same
US20220025001A1 (en) * 2016-04-28 2022-01-27 The Trustees Of Dartmouth College Nucleic acid constructs for co-expression of chimeric antigen receptor and transcription factor, cells containing and therapeutic use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101585823A (en) * 2009-06-09 2009-11-25 华中科技大学 New immunosuppressive agent extracted from Honghuoma and extraction method and use thereof
CN103314012A (en) * 2010-08-20 2013-09-18 李尚揆 Fusion protein having transcription factor transactivation-regulating domain and protein transduction domain, and transcription factor function inhibitor comprising the same
US20220025001A1 (en) * 2016-04-28 2022-01-27 The Trustees Of Dartmouth College Nucleic acid constructs for co-expression of chimeric antigen receptor and transcription factor, cells containing and therapeutic use thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
PRESSER等: "Coexpression of TGF- 1 and IL-10 Enables Regulatory T Cells to Completely Suppress Airway Hyperreactivity", THE JOURNAL OF IMMUNOLOGY, vol. 181, 31 December 2008 (2008-12-31), pages 7751, XP055852258, DOI: 10.4049/jimmunol.181.11.7751 *
WANG等: "siRNA‑mediated knockdown of T‑bet and RORγt contributes to decreased inflammation in pre‑eclampsia", 《MOLECULAR MEDICINE REPORTS》, vol. 16, pages 6368 - 6375 *
WANG等: "siRNA‑mediated knockdown of T‑bet and RORγt contributes to decreased inflammation in pre‑eclampsia", MOLECULAR MEDICINE REPORTS, vol. 16, 31 December 2017 (2017-12-31), pages 6368 - 6375 *
XIONG等: "T-bet Regulates Natural Regulatory T Cell Axerent Lymphatic Migration and Suppressive Function", 《THE JOURNAL OF IMMUNOLOGY》, vol. 196, no. 6, pages 2526 *
尹创新等: "调节性 T 细胞及其细胞因子在自身免疫性疾病中的研究进展", 医学综述, vol. 27, no. 7, 30 April 2021 (2021-04-30), pages 1285 *
尹创新等: "调节性T 细胞及其细胞因子在自身免疫性疾病中的研究进展", 《医学综述》, vol. 27, no. 7, pages 1285 *

Similar Documents

Publication Publication Date Title
Li et al. Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
KR20080056266A (en) Method for production of t cell population
CN111690050B (en) TCR recognizing EBV-LMP2 antigen and corresponding nucleic acid molecule, vector, cell and drug
KR20210111747A (en) Methods for isolating and propagating cells
US11723922B2 (en) CXCR6-transduced T cells for targeted tumor therapy
KR20210111746A (en) Methods for isolating and propagating cells
Kobayashi et al. How hematopoietic stem/progenitors and their niche sense and respond to infectious stress
WO2021259334A1 (en) Self-regulating chimeric antigen receptor and application thereof in tumor immunity
CN114729320B (en) Compositions, methods and uses for reprogramming cells to dendritic cell type 2 capable of presenting antigen
Pandit et al. Step-dose IL-7 treatment promotes systemic expansion of T cells and alters immune cell landscape in blood and lymph nodes
US20240052312A1 (en) Composition for reprogramming cells into plasmacytoid dendritic cells or interferon type i-producing cells, methods and uses thereof
CN107217041B (en) DC cell with high antigen presentation and antigen-specific T cell, and preparation method and application thereof
CN111588840A (en) Application of histone deubiquitinating enzyme in preparation of medicine for treating systemic lupus erythematosus
CN117106061B (en) TCR (T cell receptor) targeting cytomegalovirus antigen, T cell expressing TCR and application of TCR
CN114958921A (en) Method for improving stability and function of regulatory T cells
CN116376943A (en) NK cell for resisting HLA-G
CN114853902B (en) Chimeric antigen receptor, expression gene thereof, CAR modified NK cells and application thereof
CN115028737A (en) NK cell culture feeder cell
Hnátková et al. The biological environment of Hodgkin’s lymphoma and the role of the chemokine CCL17/TARC
CN113789333A (en) Application of Chi3l1 in regulating and controlling hUC-MSCs to inhibit Th17 differentiation-mediated immunoregulation effect
EP2267118A1 (en) Method for production of transfected cell
CN113244269A (en) Application of macrophage in treating obesity and regulating and controlling blood sugar
CN115595310A (en) NK cell trophoblast, preparation method and application thereof
CN110760542B (en) Plasmid for coexpression of ZNF580 and VEGF165 double genes and application thereof
CN110129372B (en) Construction method of RFFT1 cells

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