WO2015172659A1 - Uses of il-17 in enhancing immune-suppression function of mesenchymal stem cells - Google Patents

Uses of il-17 in enhancing immune-suppression function of mesenchymal stem cells Download PDF

Info

Publication number
WO2015172659A1
WO2015172659A1 PCT/CN2015/077862 CN2015077862W WO2015172659A1 WO 2015172659 A1 WO2015172659 A1 WO 2015172659A1 CN 2015077862 W CN2015077862 W CN 2015077862W WO 2015172659 A1 WO2015172659 A1 WO 2015172659A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
msc
ifnγ
tnfα
protein
Prior art date
Application number
PCT/CN2015/077862
Other languages
French (fr)
Chinese (zh)
Inventor
时玉舫
韩晓燕
王莹
Original Assignee
中国科学院上海生命科学研究院
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 中国科学院上海生命科学研究院 filed Critical 中国科学院上海生命科学研究院
Publication of WO2015172659A1 publication Critical patent/WO2015172659A1/en

Links

Images

Classifications

    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • the present invention belongs to the field of biomedicine, and in particular, the present invention relates to the use of IL-17 for enhancing the immunosuppressive function of mesenchymal stem cells.
  • MSC Mesenchymal stem cells
  • mesenchymal stem cells are important members of the stem cell family. They are derived from the mesoderm and ectoderm in early development and belong to pluripotent stem cells. MSCs are originally found in the bone marrow because of their multi-directional differentiation potential and hematopoiesis. Supporting and promoting stem cell implantation, immune regulation and self-replication are increasingly attracting attention. For example, mesenchymal stem cells can differentiate into various tissue cells such as fat, bone, cartilage, muscle, tendon, ligament, nerve, liver, heart muscle, endothelium, etc. after continuous induction and cryopreservation. It still has multi-directional differentiation potential and can be used as an ideal seed cell for the repair of tissue and organ damage caused by aging and lesions.
  • MSCs The immunosuppressive mechanism of MSCs for T cells has been extensively studied both in vivo and in vitro. It is generally believed that MSCs inhibit T cells by not inducing T cell apoptosis, but in the cell cycle of T cells. Stay in G0/G1 period. Studies have also shown that MSC can promote the apoptosis of activated T cells, but can promote the survival of T cells in resting state, which also indicates that MSC exerts immunosuppression depending on the stimulation of inflammatory cytokines released by T cell activation.
  • Interleukin-17A is one of more than 30 kinds of interleukins that have been discovered so far, ranking 17th according to the serial number.
  • IL-17 is secreted by CD4+ T cells and can induce the synthesis and secretion of IL-6, IL-8, G-CSF and PGE2 by epithelial cells, endothelial cells and fibroblasts, and promote the expression of ICAM-1.
  • IL-17 has been found to be a proinflammatory cytokine mainly produced by activated T cells, which can promote the activation of T cells and stimulate the production of various cytokines such as IL-6 and IL in epithelial cells, endothelial cells and fibroblasts. -8, granulocyte-macrophage stimulating factor (GM-CSF) and chemical stimulating hormone 1 and cellular adhesion molecule 1 (CAM-1), resulting in inflammation.
  • IL-17 is an early promoter of T cell-induced inflammatory responses that amplify the inflammatory response by promoting the release of pro-inflammatory cytokines.
  • IL-17 After binding to the receptor, IL-17 exerts its biological effects through the MAP kinase pathway and the nuclear factor kB (NF-kB) pathway. Th17 cells secrete IL-17A, IL-17F, IL-6, and tumor necrosis factora (TNF- ⁇ ). These cytokines can collectively mobilize, recruit, and activate neutrophils. IL-17 produced by Th17 cells can effectively mediate the excitatory process of neutrophil mobilization, thereby effectively mediating the inflammatory response of tissues.
  • NF-kB nuclear factor kB
  • IL-17 can act on a variety of cell types, thereby promoting the expression of other cytokines, chemokines and metalloproteinases, including TNF ⁇ , IL-1 ⁇ , IL-6, GM-CSF, G-CSF, CXCL1, MMP-3, and the like. At the same time, IL-17 can also promote the expression of multiple target genes in combination with other cytokines such as TNF ⁇ . In vivo experimental studies have revealed the important role of IL-17 family cytokines in the body's antimicrobial infection. Although IL-17 protects against bacterial and fungal infections in the body, over-activation of the IL-17 signaling pathway leads to autoimmune diseases.
  • IL-17 is significantly elevated, and many studies have shown that IL-17 is involved in the pathogenesis of many autoimmune diseases, including MS (multiple sclerosis), RA (rheumatoid arthritis), and IBD ( Inflammatory bowel disease).
  • Another object of the invention is to provide the use of an interleukin-17 antagonist.
  • Another object of the present invention is to provide an IL-17-treated mesenchymal stem which has improved immunosuppressive ability. cell.
  • an interleukin-17, a derivative of interleukin-17 or an agonist thereof for the preparation of a formulation or kit for:
  • the interleukin-17 is a mammalian interleukin-17.
  • the interleukin-17 is human interleukin-17.
  • amino acid sequence of interleukin-17 is as shown in SEQ ID NO.: 1.
  • the interleukin-17 derivative comprises a modified interleukin-17 molecule, a protein molecule having an amino acid sequence homologous to native interleukin-17 and having natural interleukin-17 activity, and dimerization of interleukin-17.
  • the modified interleukin-17 molecule is PEGylated interleukin-17.
  • the "protein molecule having an amino acid sequence homologous to natural interleukin-17 and having natural interleukin-17 activity means that the amino acid sequence thereof has ⁇ 85% as compared with SEQ ID NO.: Derived, preferably > 90% homology, more preferably > 95% homology, optimally > 98% homology; and a protein molecule having native interleukin-17 activity.
  • the "immunosuppressive function of mesenchymal stem cells” refers to an immunosuppressive function of mesenchymal stem cells to T cells.
  • the formulation or kit further comprises IFNy and/or TNF[alpha].
  • the agonist means a substance capable of increasing the activity and/or content of interleukin-17 or a derivative thereof in vivo or in vitro.
  • the substance may be a synthetic or natural compound, protein, nucleotide or the like.
  • the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
  • the hepatitis comprises autoimmune hepatitis, viral hepatitis;
  • the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
  • the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
  • an interleukin-17 antagonist for the preparation of a formulation or kit for:
  • the antagonist of interleukin-17 refers to a substance capable of reducing the activity of interleukin-17 in vivo or in vitro.
  • the antagonist of interleukin-17 may be a small miRNA, an anti-interleukin-17 antibody or an iNOS inhibitor, an Act1 protein inhibitor, an AUF1 protein agonist, an interleukin-17 receptor inhibitor, NF ⁇ B. Inhibitor, TRAF6 inhibitor.
  • the formulation comprises a pharmaceutical composition, a nutraceutical composition, a food composition, or an experimental agent.
  • the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
  • the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
  • composition comprising interleukin-17 or a derivative thereof, IFNy and TNFa.
  • the molar ratio of the interleukin-17 or its derivative, IFN ⁇ and TNF ⁇ is from 1-10:10:10.
  • the composition comprises a solid formulation, a liquid formulation, preferably in the form of a dry powder or solution.
  • the composition is a pharmaceutical composition.
  • an isolated population of MSC cells consisting of or consisting essentially of MSC cells, and said MSC cells having enhanced ability to inhibit T cell proliferation,
  • the MSC cells are selected from the group consisting of:
  • an in vitro pretreated MSC cell population wherein the pretreatment refers to simultaneous, sequential or sequential treatment with (i) interleukin-17 or a derivative thereof, (ii) IFN ⁇ and (iii) TNF ⁇ ;
  • the conditions are as follows: 0.5-1000 ng/ml (preferably 1-200 ng/ml, more preferably 1-20 ng/ml, optimally 1-10 ng/ml) IFN ⁇ , 0.5-1000 ng/ml (preferably 1-200 ng/ml, more preferably 1-20 ng/ml, optimally 1-10 ng/ml) of TNF ⁇ , and 0.2-1000 ng/ml (preferably 0.5-) 200 ng/ml, more preferably 1-20 ng/ml, optimally 1-10 ng/ml) interleukin-17.
  • 1-2 ng/ml IFN ⁇ , 1-2 ng/ml TNF ⁇ , and 0.5-2 ng/ml interleukin-17 are used in the pretreatment; or 10 ng/ml IFN ⁇ , 10 ng/ml TNF ⁇ , and 10 ng are used. /ml IL-17.
  • the enhanced ability to inhibit T cell proliferation refers to I1/I0 ⁇ 1.5 (preferably ⁇ 2, more preferably ⁇ 4), wherein I1 is the proliferation of T cells by the MSC cells. Percent inhibition rate; while I0 is the percent inhibition of T cell proliferation by wild-type MSC cells of the same species in the control group.
  • said "consisting essentially of” means that the MSC cells comprise at least 90%, preferably at least 95%, more preferably at least 99% of the total number of cells in said population of cells.
  • the MSC cell population has the characteristic that, after administration of the MSC cell population to an animal, a change in the animal from the group of the following occurs:
  • the preprocessing process includes the steps of:
  • the cells After trypsinizing the MSC cells, the cells are cultured until the cells are attached, and then the cytokines at the above concentrations are added, and after further culture for 6-24 hours, the cell population is further digested and collected.
  • the MSC cells are bone marrow-derived MSC cells, umbilical cord-derived MSC cells, adipose-derived MSC cells, placental-derived MSC cells, and/or pulp-derived MSC cells.
  • a genetically engineered cell strain which is genetically engineered to result in overexpression of an endogenous Actl protein and/or a decrease in AUF1 protein or activity.
  • the cell strain is a mammalian MSC cell line.
  • the cell strain is for enhancing the immunosuppressive function of mesenchymal stem cells.
  • an isolated protein complex is provided, the protein complex being a protein complex bound by an Act1 protein and an AUF1 protein.
  • the protein complex has a molecular weight of from 80 KD to 130 KD.
  • a seventh aspect of the invention provides the use of the protein complex of the sixth aspect of the invention for screening a drug or a compound which promotes or inhibits the formation of the Act1 protein and the AUF1 protein Complex.
  • the medicament is for:
  • the application comprises the steps of:
  • the screening further comprises a positive control group, preferably IL17 is added to the positive control group.
  • IL17 enhances the ability of two proteins to form a complex.
  • the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
  • the hepatitis comprises autoimmune hepatitis, viral hepatitis;
  • the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
  • the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
  • kits comprising the following components:
  • components (a), (b) and (c) are respectively located in one or more different containers or in the same container.
  • the description describes: the kit is for:
  • the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
  • the hepatitis comprises autoimmune hepatitis, viral hepatitis;
  • the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
  • the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
  • kits comprising the pharmaceutical composition according to claim 3 and instructions, wherein the pharmaceutical composition is described for:
  • the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
  • the hepatitis comprises autoimmune hepatitis, viral hepatitis;
  • the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
  • the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
  • a method of treating hepatitis or liver damage comprising the steps of:
  • a therapeutically effective amount of MSC cells is administered to the subject in need thereof.
  • the liver damage is cirrhosis.
  • the subject is a mammal (e.g., a human).
  • the application is an intravenous infusion.
  • the MSC cell is the MSC cell population of the fourth aspect of the invention.
  • the MSC cell is a pre-treated MSC cell population, wherein the pretreatment refers to (i) interleukin-17 or a derivative thereof, (ii) IFN ⁇ , and (iii) TNF ⁇ simultaneously. Process in sequence or sequentially.
  • the MSC cells are in vitro pretreated MSC cell populations, wherein the pretreatment refers to simultaneous, sequential or sequential treatment with IFN ⁇ and TNF ⁇ .
  • the density of the MSC cell population during the pretreatment is 1 ⁇ 10 4 cells/ml-5 ⁇ 10 6 cells/ml, preferably 5 ⁇ 10 4 cells/ml-5 ⁇ 10 5 cells. /ml.
  • the concentration of interleukin-17 during the pretreatment is from 0.5 to 1000 ng/ml, preferably from 1 to 100 ng/ml.
  • the concentration of IFN ⁇ during the pretreatment is from 0.5 to 1000 ng/ml, preferably from 1 to 100 ng/ml.
  • the concentration of TNF ⁇ in the pretreatment is from 0.5 to 1000 ng/ml, preferably from 1 to 100 ng/ml.
  • FIGS. 1A, 1B, 1C, 1D, 1E, 1F, 1G, and 1H showing that IL-17 enhances the immunosuppressive function of MSCs.
  • Figure 2 comprising Figures 2A and 2B, shows that IL-17 enhances the immunosuppressive properties of MSCs depending on the activity of iNOS.
  • Figure 3 shows that IL-17 enhances the immunosuppressive function of MSCs independent of the effects on T cell apoptosis.
  • Figure 4 comprising Figures 4A, 4B, 4C, 4D, 4E, 4F, and 4G, shows that IL-17 synergistic inflammatory cytokines up-regulate the expression of iNOS in MSCs.
  • Figure 5 comprising Figures 5A, 5B, 5C, and 5D, shows that IL-17 plays an important role in enhancing immunosuppression and gene expression in MSCs requiring Act1 involvement.
  • Figure 6 includes Figures 6A, 6B, and 6C showing that IL-17, IFNy, and TNF[alpha] pretreated MSCs are effective in treating ConA-induced liver damage.
  • Figure 7 includes Figures 7A, 7B, 7C, and 7D showing that the therapeutic effects of IL-17, IFN ⁇ , and TNF ⁇ pretreated MSCs on CIH depend on their immunosuppressive function on T cells without affecting T cell subsets. proportion.
  • Panels A-E in Figure 8 show that the therapeutic effect of IL-17, IFNy and TNF[alpha] pretreated MSCs on CIH is dependent on the expression of iNOS.
  • Figure 9 includes Figures 9A, 9B, 9C, 9D, 9E, and 9F showing that IL-17 can reverse the inhibition of gene expression by the RNA binding protein AUF1.
  • Panels A-D of Figure 10 show that IL-17 enhances the stability of iNOS mRNA by modulating the level of AUF1.
  • Panels A-B of Figure 11 show that AUF1 is a key molecule that mediates IL-17 signaling.
  • Panels A-D of Figure 12 show that AUF1 is a key molecule that mediates IL-17 enhancing MSC treatment of CIH.
  • IL-17 can significantly enhance the immunosuppressive function of mesenchymal stem cells and inhibit the proliferation of T cells; this function enhances the mRNA of iNOS by decreasing the expression level of the RNA binding protein AUF1.
  • the stability is achieved by up-regulating the expression of iNOS in MSC.
  • IL-17 pretreated MSC can effectively treat CONA-induced liver injury. The present invention has been completed on this basis.
  • treating refers to the administration of interleukin-17 of the present invention to a subject in need of treatment for the purpose of curing, alleviating, ameliorating, alleviating, affecting the disease, symptoms, and disease predisposition of the subject.
  • terapéutica subject refers to rats, humans, and other mammals.
  • terapéuticaally effective amount refers to an amount of interleukin-17 that is capable of achieving a therapeutic purpose in a subject. It will be understood by one of ordinary skill in the art that the “therapeutically effective amount” may vary depending on the route of administration of interleukin-17, the pharmaceutical excipients used, and the combination with other drugs.
  • MSC Mesenchymal stem cells
  • the inventors' research results show that MSC exerts an immunosuppressive function on T cells through the combined action of nitric oxide (NO) and chemokines.
  • T cells secrete a large number of inflammatory cytokines, including IFN ⁇ , TNF ⁇ , IL-1 ⁇ and IL-1 ⁇ , after activation of their receptor (TCR). These inflammatory cytokines stimulate MSCs to secrete large amounts of iNOS and chemokines.
  • Chemokines are capable of recruiting T cells to the periphery of MSCs and inhibiting T cell proliferation through the metabolite NO of iNOS.
  • the transcription factors C/EBP ⁇ and STAT1 are critical in the induction of iNOS production.
  • the present inventors also studied different mechanisms by which MSCs of different species are mediated in immunosuppression of T cells.
  • iNOS is a key molecule for MSCs to suppress T cells
  • IDO is a key molecule for MSCs to inhibit T cells.
  • HLA-G5, TGF- ⁇ and IL-10 are key molecules that mediate the immunosuppression of human MSCs.
  • immunosuppressive factor refers to a factor or molecule that can downregulate the body's excessive immune response. Excessive immune response, including B cells, T cells, natural killer cells, macrophages, dendritic cells, neutrophils and other cell-mediated immune responses beyond the normal range of the body, may lead to inflammatory diseases, such as allergic reactions , autoimmune diseases, etc.
  • Preferred immunosuppressive factors in the present invention include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin and/or B7- H4.
  • Interleukin-17 or its derivative and preparation method thereof are examples of Interleukin-17 or its derivative and preparation method thereof.
  • Interleukin-17 or "IL-17” refers to a protein which has (a) and Zhengbin Yao et al. Human IL-17: a novel cytokine derived from T cells. J. Immunol. 155(12), 5483-5486 (1995); and the basic amino acid sequence of human/mouse interleukin-17 described in Submitted (11-DEC-1995) Jacqueline Kennedy, Immunology, DNAX Research Institute and (b) with natural Interleukin-17 has the same biological activity.
  • the interleukin-17 of the present invention includes, but is not limited to, human interleukin-17, recombinant human interleukin, murine interleukin-17, and/or recombinant murine interleukin-17.
  • the amino acid sequence of interleukin-17 is:
  • VLRREPPHCP NSFRLEKILV SVGCTCVTPI VHHVA SEQ ID NO.: 1
  • IL-17 signaling pathway activates the expression of many pro-inflammatory genes, similar to the activation of some classical innate immune receptors, including IL-1R (IL-1 receptor) and TLR (Toll-like receptor). Wait. Similar to IL-1R and TLR, IL-17RA binds to IL-17 and interacts with the adaptor proteins Act1 and TRAF6 to activate NF ⁇ B signaling pathway and promote target gene expression. IL-17 activates the NF ⁇ B pathway and activates the MAPK signaling pathway. Activation of MAPK leads to activation of AP1 and promotes transcription of target genes. In addition, MAPK activation can also promote gene expression by enhancing mRNA stability. For IL-17, enhancing mRNA stability is an important part of its promotion of gene expression. Among them, Act1 is an important molecule that mediates IL-17 to enhance mRNA stability.
  • IL-17 is mainly secreted by Th17 cells, and can also be produced by other cells, including ⁇ T cells, NKT cells, NK cells, neutrophils and eosinophils.
  • IL-17 is involved in the pathogenesis of many autoimmune diseases.
  • the inventors have found that IL-17 can enhance the immunosuppressive function of MSC in the presence of MSC, thereby effectively inhibiting T cell proliferation, indicating that IL-17 can inhibit the immune response in the presence of MSC.
  • substantially identical amino acid sequence refers to a difference in sequence or caused by one or more amino acid changes (deletion, addition, substitution), but such alteration does not substantially reduce its biological activity, ie, by binding to IL- 17 target cell receptors and biological functions.
  • Any interleukin-17 that meets the "substantially identical" requirement is included in the invention, whether it is glycosylated (ie derived from natural or derived from eukaryotic expression systems) or non-glycosylated (ie source) In prokaryotic expression systems or chemically synthesized).
  • Interleukin-17 also includes PEGylated IL-17 and covalently modified IL-17 protein.
  • PEG polyethylene glycols
  • various activated polyethylene glycols (PEG) having a molecular weight of 5,000 to 100,000 can be used to polymerize IL-17 to prolong its half-life.
  • PEG polyethylene glycols
  • the interleukin-17 of the present invention can be cloned and expressed by genetic recombination techniques.
  • Host cells for expression include prokaryotic cells, yeast cells, or higher eukaryotic cells.
  • eukaryotic cells such as filamentous fungi or yeast are equally suitable for expressing or cloning the interleukin-17 of the present invention.
  • the host cell of the interleukin-17 of the present invention for expressing glycosylation is derived from a multicellular organism. Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, plant cells. Examples of suitable mammalian host cells include Chinese hamster ovary cells (CHO), COS cells. One of ordinary skill in the art will know how to select a suitable host cell.
  • the above host cells are transfected or transformed with an interleukin-17 expression vector or a cloning vector, and can be cultured in a conventional nutrient medium, which is modified to be suitable for inducing a promoter and a selective transformant. (selecting transformant) or amplifying the interleukin-17 encoding gene sequence. Selection of culture conditions such as medium, temperature, pH, etc. will be known to those of ordinary skill in the art. General principles, protocols, and techniques for how to maximize cell culture fertility can be found in Mammalian Cell Biotechnology: a Practical Approach, M. Butler, ed. (IRL Press, 1991) and Sambrook et al., supra.
  • the interleukin-17 of the present invention can be directly expressed not only by genetic recombination, but also by forming a fusion polypeptide with a heterologous polypeptide, which may be a signal sequence located at the N-terminus of the mature protein or polypeptide, or may be located in a mature protein or polypeptide. N-terminal other polypeptide fragments with specific cleavage sites.
  • the DNA sequence encoding the IL-17 dimer or fusion protein of the present invention can be all synthetically synthesized.
  • the coding DNA sequences of the IL-17 first monomer and/or the IL-17 second monomer can also be obtained by PCR amplification or synthesis and then spliced together to form a DNA sequence encoding the fusion protein of the present invention.
  • the IL-17 dimer coding sequence can be engineered, for example, using host cell-preferred codons to eliminate sequences that are detrimental to gene transcription and translation.
  • the yeast cell or mammalian cell-preferred codon can be used, and the IL-17 dimer gene can be detected by computer DNA software, and the sequence which is not conducive to gene transcription and translation in the gene, including the inclusion, is excluded. Sub-shearing site, transcription termination sequence, and the like.
  • the DNA sequence encoding the novel fusion protein of the present invention After obtaining the DNA sequence encoding the novel fusion protein of the present invention, it is ligated into a suitable expression vector and transferred to a suitable host cell. Finally, the transformed host cells are cultured, and the novel fusion protein of the present invention is obtained by isolation and purification.
  • vector includes plasmids, cosmids, expression vectors, cloning vectors, viral vectors, and the like.
  • various carriers known in the art such as commercially available carriers can be used.
  • a commercially available vector is selected, and then a nucleotide sequence encoding a novel fusion protein of the present invention is operably linked to an expression control sequence to form a protein expression vector.
  • operably linked refers to a condition in which portions of a linear DNA sequence are capable of affecting the activity of other portions of the same linear DNA sequence. For example, if a signal peptide DNA is expressed as a precursor and is involved in the secretion of a polypeptide, then the signal peptide (secretion leader sequence) DNA is operably linked to the polypeptide DNA; if the promoter controls the transcription of the sequence, then it is operably linked to A coding sequence; if the ribosome binding site is placed at a position that enables translation, then it is operably linked to the coding sequence.
  • “operably linked to” means adjacent, and for secretory leader sequences means adjacent in the reading frame.
  • the term "host cell” includes prokaryotic cells and eukaryotic cells.
  • prokaryotic host cells include Escherichia coli, Bacillus subtilis and the like.
  • eukaryotic host cells include yeast cells, insect cells, and mammalian cells.
  • the host cell is a eukaryotic cell, more preferably a mammalian cell.
  • the cell After obtaining the transformed host cell, the cell can be cultured under conditions suitable for expression of the fusion protein of the present invention to express the fusion protein.
  • the expressed fusion protein is then isolated.
  • compositions of the present invention comprise a safe or effective amount of the IL-17 or derivative thereof of the present invention and a pharmaceutically acceptable excipient or carrier.
  • safe, effective amount it is meant that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 0.001 to 1000 mg of IL-17 or a derivative/agent thereof, preferably 0.05 to 300 mg of IL-17 or a derivative/agent thereof, more preferably 0.5 to 200 mg of IL-17 or Its derivatives/agents.
  • the IL-17 or a derivative thereof of the present invention and a pharmacologically acceptable salt thereof can be formulated into various preparations comprising the IL-17 or derivative thereof of the present invention or a pharmacologically acceptable amount thereof in a safe and effective amount Salts and pharmaceutically acceptable excipients or carriers.
  • safe, effective amount it is meant that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the safe and effective amount of the compound is determined according to the specific conditions such as the age, condition, and course of treatment of the subject.
  • “Pharmacologically acceptable excipient or carrier” means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity .
  • “compatibility” it is meant herein that the components of the composition are capable of intermixing with the compounds of the invention and with each other without significantly reducing the potency of the compound.
  • Examples of pharmaceutically acceptable excipients or carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyol (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifier (such as ), a wetting agent (such as sodium lauryl sulfate), a coloring agent, a flavoring agent, a stabilizer, an antioxidant, a preservative, a pyrogen-free water, and the like.
  • cellulose and its derivatives such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose
  • IL-17 or a derivative thereof of the present invention when administered, it can be administered orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously) or topically.
  • the IL-17 or derivative thereof of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • the microcapsule containing the interleukin-17 of the present invention or a derivative thereof can be used for the sustained release administration of the interleukin-17 of the present invention.
  • the microcapsule sustained release drug delivery technology of recombinant protein has been successfully applied to recombinant human growth hormone (rhGH), recombinant human interferon (rhIFN), interleukin-2 and MNrgp120 (Johnson et al., Nat. Med., 2:795- 799 (1996); Yasuda, Biomed. Ther 27: 1221-1223 (1993); WO 97/03692, WO 96/40072, WO 96/07399; US Pat. No. 5,654,010.
  • the sustained release preparation of the interleukin-17 of the present invention or a derivative thereof can be produced by a lactic acid glycolic acid high polymer (PLGA) having good biocompatibility and broad biodegradability.
  • PLGA lactic acid glycolic acid high polymer
  • the degradation products of PLGA, lactic acid and glycolic acid can be quickly eliminated by the human body.
  • the degradation ability of the polymer can be extended from several months to several years depending on its molecular weight and composition (Lewis, "Controlled release of bioactive agents form lactide/glycolide polymer," in: M. Chasin and R. Langer (Eds.), Biodegradable Polymers as Drug Delivery Systems (Marcel Dekker: New York, 1990), pp. 1-41)).
  • a safe and effective amount of IL-17 or a dimer thereof of the present invention is applied to a mammal (e.g., a human) in need of treatment, wherein the dose at the time of administration is a pharmaceutically effective effective dose for 60 kg.
  • the dose is usually from 0.01 to 300 mg, preferably from 0.5 to 100 mg.
  • specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • IL-17 can significantly enhance the immunosuppressive function of mesenchymal stem cells.
  • IL-17 has the ability to inhibit the proliferation of T cells.
  • the reagents and experimental materials used in the present invention are commercially available, and the sources of the biological materials are as follows.
  • Recombinant mouse IFN ⁇ , TNF ⁇ , IL-17A, anti-IL-17A antibody was purchased from eBiosciences (La Jolla, CA); recombinant mouse IL-2 was purchased from R&D Systems (Minneapolis, MN); anti- ⁇ -actin, GAPDH, iNOS , p65, p-I ⁇ B ⁇ , p-p65, p-JNK, p-ERK1/2 antibody were purchased from Cell Signaling Technology (Danvers, MA); anti-Act1 antibody was purchased from Santa Cruz Biotechnology (Dallas, TX); L-NMMA, Propidium iodide, actinomycin D was purchased from Sigma-Aldrich (St. Louis, MO); Concanavaline A (ConA) was purchased from Vector Labs (Burlingame, CA).
  • C57BL/6 mice were purchased from Shanghai Slack Laboratory Animal Co., Ltd. The animals were raised in a clean environment at the Experimental Animal Science Department of the School of Medicine, Shanghai Jiaotong University.
  • mice were sacrificed by cervical dislocation and then immersed in 75% alcohol for 3-5 minutes.
  • the two hind legs of the mice were cut with surgical scissors, and two of the tibia and femur were taken and immersed in DMEM complete medium (containing twice as much penicillin and streptomycin), and the muscle tissue around the bone was removed with surgical scissors. Cut the ends of the removed bones to leave the cavity, see the bone marrow; use a 10 ml syringe to draw 5-10 ml of DMEM medium (without FBS) to the bone cavity and rinse the bone marrow into a 50 ml centrifuge tube.
  • the selected clones were characterized for in vitro differentiation (osteogenesis and adipogenesis) and surface markers, and the mesenchymal stem cells used in all experiments were used before 20 generations. All mesenchymal stem cells were cultured using DMEM complete medium.
  • Mouse mononuclear cell suspension was counted and cultured in a 10 cm culture dish at a density of 1 ⁇ 10 6 cells/ml.
  • Anti-mouse CD3 antibody (1 ⁇ g/ml) and anti-mouse CD28 antibody were added to the 1640 complete medium. (1 ⁇ g/ml).
  • the cells were collected and centrifuged for 500 g for 5 minutes, and the supernatant was collected as Sup-CD3 (small mouse spleen cell culture supernatant stimulated with anti-CD3 and anti-CD28 antibody for 48 hours) while collecting the cell pellet.
  • the cell pellet was resuspended in 1640 complete medium, counted and cultured in a 10 cm culture dish at a density of 1 ⁇ 10 6 cells/ml (without anti-CD3 and anti-CD28 antibodies), and IL-2 (200 U/ml was added to the culture solution). After 48 hours of incubation, the cells grow into a mass, which is T cell blasts.
  • 3H-Tdr (0.5 ⁇ Ci per well) was added to a 96-well plate for detecting T cell proliferation, and cultivation was continued in an incubator, and after 6 hours, the 96-well plate was placed in a -80 °C refrigerator. Until the day of the test, the plate was taken out and placed in an oven at 37 ° C to transfer all the isotope taken up by the cells to the glass cellulose membrane. The membrane was dried using a microwave oven and then added to the scintillation fluid. The cpm value was read on a Wallac MicroBeta liquid scintillation counter to measure the amount of 3H incorporated.
  • the primer sequences used are as follows:
  • Reverse primer 5'-CATTGGAAGTGAAGCGTTTCG-3' (SEQ ID NO.: 22).
  • mesenchymal stem cells were passaged to 5 6-well plates: 1.5 ⁇ 10 5 cells/well, 1.6 ml of complete medium was added to each well, and the cells were placed overnight to allow the cells to adhere.
  • IFN ⁇ +TNF ⁇ was added to the 6-well plate, and IFN ⁇ +TNF ⁇ +IL-17 cytokine was stimulated to a final concentration of 10 ng/ml.
  • Act.D (5 ⁇ g/ml) was added to the 6-well plate at 18 o'clock in the evening, and RNA was collected at 0, 0.5 hr, 1 hr, 2 hr, 3 hr, and 4 hr for 5 time points, each time point corresponding to each treatment. There are three duplicate wells, and the lysate is added and mixed, and then placed at -80 °C.
  • RNA per repeat at each time point was expressed as ratio to ⁇ -actin/GAPDH, with average RNA at time 0 Using the amount as a reference, the percentage of the amount of repeating-well RNA in each of the remaining time points relative to the zero-point reference value was calculated.
  • Plasmid transfection was performed by electroporation using the kit Amaxa Cell Line Nucleofector Kit V, with reference to its instructions, as follows.
  • Plasmid shCTRL, shAUF1, shAct1, GFP plasmid (included in the kit).
  • the cells were resuspended in 100 ⁇ l of Solution, and 2 ⁇ g of the corresponding plasmid was added to each centrifuge tube, which was gently blown.
  • the well plate was taken out, and the cells transfected with the GFP plasmid were observed by fluorescence microscopy. It can be seen that more than 80% of the cells emitted green fluorescence, that is, the transfection efficiency was over 80%. All 6-well plates were exchanged.
  • the cells can be collected by cell scraping or trypsinization. After washing with PBS, the collected cells are added with a certain amount of RIPA lysate, mixed by blowing, and placed on ice for 30 minutes, shaking for 3-4 times; Transfer at high speed 13,000 rpm for 30 min at low temperature; transfer the supernatant to a new centrifuge tube and place on ice for later use. Protein concentration was determined using Bio-Rad's Protein Assay Dye Reagent Concentrate (Bradford method). SDS-polyacrylamide gel was prepared by using 8% separating glue and 5% laminated glue. The sample was added to 5 ⁇ loading buffer by volume ratio, and boiled at 100 ° C for 10 minutes, then cooled at room temperature and loaded.
  • Each sample contains about 50-100 ⁇ g of protein; each gel contains a protein marker; the electrode is inserted and electrophoresis is started, first 80V electrophoresis for 30min, then 120V electrophoresis for 60min.
  • the laminated gel is removed and placed in an electrotransfer buffer; the NC membrane (nitrocellulose membrane) is placed in a transfer buffer for about 10 minutes; from bottom to top, the sponge, filter paper, and membrane are placed.
  • each layer should pay attention to discharge air bubbles, connect the transfer device, and then turn on the power, constant voltage 100V, 2 hours; after the transfer is finished, turn off the power, remove the membrane, according to the protein Marker and molecular weight of the target protein cut out the strip at the corresponding position and mark it.
  • the first antibody was shaken overnight at 4 °C.
  • the film was taken out the next day and then washed 3 times with 1 x TBST for 10 minutes each time.
  • the cells were collected by trypsinization. After washing with PBS, the collected cells were added with a certain amount of RIPA lysate, mixed by blowing, and placed on ice for 30 minutes, shaking for 3-4 times; then high speed 13,000 rpm Centrifuge at low temperature for 30 min; transfer the supernatant to a new centrifuge tube and let it stand on ice. Protein concentration was determined using Bio-Rad's Protein Assay Dye Reagent Concentrate (Bradford method). The above protein cleavage supernatant was incubated with protein G sepharose beads to remove non-specific binding, and then the supernatant was taken after high-speed centrifugation.
  • Mesenchymal stem cells were pretreated with cytokines for 12-16 hours in advance: WT MSC, iNOS-/-MSC or auf1-/-MSC administered IFN ⁇ +TNF ⁇ /IFN ⁇ +TNF ⁇ +IL-17 (10 ng/ml) cells Factor stimulation.
  • mice were intravenously administered with 15 mg/kg of ConA.
  • mice Eight hours after ConA injection, the mice were sacrificed, serum was collected, serum ALT levels were measured, and one leaf liver was immersed in 10% formalin for pathological section H&E staining; the remaining livers were all used to isolate mononuclear cells.
  • Detection of ALT in mouse serum Refer to the instructions of the ALT test kit produced by Shanghai Yihua Company for operation.
  • GFP-MSC was pretreated with cytokine IFN ⁇ +TNF ⁇ /IFN ⁇ +TNF ⁇ +IL-17 (10ng/ml) for 12-16 hours in advance; 30 minutes after ConA injection, each group received GFP-MSC intravenous injection, each treatment. Mice were treated with 5 x 10 5 cells. Mice were sacrificed 8 hours after ConA injection.
  • the cells were resuspended in 100 ⁇ l of FACS buffer for staining at 1 ⁇ 10 6 .
  • fluorescently labeled antibodies eg PE-labeled anti-mouse IL-17RA antibody, PE-labeled anti-mouse CD3 antibody, PerCP/Cy5.5-labeled anti-mouse CD4 antibody, APC-labeled anti-mouse CD8 antibody, APC-labeled antibody Mouse CD45 antibody, APC-labeled anti-mouse CD25 antibody, FITC-labeled anti-mouse CD3 antibody
  • the cells were resuspended in 400 ⁇ l of FACS buffer and analyzed by FACS Calibur flow cytometry.
  • Example 1 IL-17 can enhance the immunosuppressive function of MSC
  • mesenchymal stem cells are not intrinsic, but is obtained under the induction of inflammatory cytokines including IFN ⁇ , TNF ⁇ , IL-1 ⁇ and IL-1 ⁇ .
  • Mesenchymal stem cells secrete a large amount of iNOS and chemokines under the stimulation of inflammatory cytokines IFN ⁇ and TNF ⁇ .
  • Chemokines recruit T cells to the periphery of mesenchymal stem cells, and the NO, a metabolite of iNOS, inhibits T cell proliferation. effect.
  • IL-17 is a pleiotropic pro-inflammatory cytokine that plays an important role in the pathogenesis of many infectious diseases, inflammatory diseases and autoimmune diseases.
  • the present inventors added inflammatory cytokines IFN ⁇ and TNF ⁇ to mesenchymal stem cells cultured in vitro, or added IL-17 thereto, and observed the effects of these cytokines on the immunosuppressive ability of mesenchymal stem cells.
  • the present inventors have found that mesenchymal stem cells can only inhibit the proliferation of T cells in the presence of IFN ⁇ and TNF ⁇ , while IL-17 can significantly enhance the immunosuppression of mesenchymal stem cells induced by IFN ⁇ and TNF ⁇ . Capacity, this effect is manifested by a significant decrease in T cell proliferation (Fig. 1A).
  • IL-17 activates downstream signaling pathways by binding to heterodimers composed of IL-17RA and IL-17RC on the cell surface. Since IL-17 can act on mesenchymal stem cells and significantly enhance its immunosuppressive ability, the present inventors examined the expression of IL-17RA and IL-17RC in mesenchymal stem cells. The results showed that mesenchymal stem cells can constitutively express IL-17RA and IL-17RC (Fig. 1B, C).
  • IL-17 can significantly enhance the immunosuppressive function of mesenchymal stem cells when the levels of IFN ⁇ and TNF ⁇ are only 1-2 ng/ml (Fig. 1D, 4E). Even when the levels of IFN ⁇ and TNF ⁇ are high (10-20 ng/ml), IL-17 can improve the immunosuppressive function of mesenchymal stem cells.
  • the present inventors also observed the effect of different doses of IL-17 on the property of enhancing the immunosuppressive ability of mesenchymal stem cells. The results showed that approximately 0.5 ng/ml of IL-17 significantly enhanced the immunosuppressive function of mesenchymal stem cells (Fig. 1F).
  • IL-17 is involved in the inhibition of T cell proliferation by MSCs in a T cell-mediated immune response
  • the inventors co-cultured MSCs with anti-CD3 activated splenocytes and added anti to the co-culture system.
  • -IL-17A antibody The results showed that MSC can significantly inhibit the proliferation of activated spleen cells, while anti-IL-17A The antibody can largely restore the proliferation of activated splenocytes, reversing the inhibition of MSC (Fig. 1G).
  • IL-17 can significantly enhance MSC immunity against T cells in T cell-mediated immune responses, particularly when the levels of inflammatory cytokines IFN ⁇ and TNF ⁇ present in the immune microenvironment are low. Suppress function.
  • the present inventors also examined the effect of IL-17 on its immunosuppressive function on adipose-derived MSCs, and obtained similar results to bone marrow-derived MSCs (Fig. 1H), namely IL- 17 also enhances the immunosuppressive function of adipose-derived MSCs, suggesting that this function of IL-17 may not depend on the source of MSCs.
  • Figure 1 shows that IL-17 enhances the immunosuppressive function of MSCs.
  • A MSCs were first stimulated with different combinations of cytokines IFN ⁇ , TNF ⁇ and IL-17 (each concentration of 2 ng/ml) for 12 hours, and then co-cultured with T cell blasts (MSC and T cell). The ratio of blasts was 1:20), and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation.
  • B The expression of IL-17RA and IL-17RC on MSC and Raw264.7 cells (positive control) was detected by real-time fluorescent quantitative PCR.
  • C Flow cytometry was used to detect the expression of IL-17RA on MSC surface.
  • the MSCs were first stimulated with the cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 for 12 hours (the concentration of IFN ⁇ and TNF ⁇ was diluted in a gradient, the IL-17 concentration was fixed at 10 ng/ml), and then And T cell blasts (requires the addition of IL-2 to proliferate, and does not produce inflammatory cytokines during proliferation).
  • D or A1.1 cells (T cell hybridoma cells, can proliferate independently, and do not produce inflammatory cytokines during proliferation, the preparation method of the cells can be referred to the literature (Nature.
  • MSCs were co-cultured with anti-CD3 and anti-CD28-activated splenocytes at a ratio of 1:40 or 1:20, or Anti-IL-17A was added to the culture medium, and 48 hours later, spleen cell proliferation was measured by 3 H-Tdr incorporation method.
  • H Next, BMMSC or ADSC was treated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-. 17 (10 ng/ml for each factor) was stimulated for 12 hours, then co-cultured with A1.1 cells (1:10 ratio of MSC to A1.1), and after 12 hours, 3 H-Tdr T cell proliferation was determined by incorporation assay. Results are expressed as mean Number ⁇ standard deviation, BMMSC: bone marrow-derived MSC; ADSC: fat-derived MSC. The data shown represents the results of three experiments.
  • Example 2 IL-17 enhances the immunosuppressive properties of MSCs depending on the activity of iNOS
  • IL-17 can significantly enhance the immunosuppressive function of MSCs against T cell proliferation, and the inventors further studied the related mechanisms.
  • Previous studies have suggested that IL-17 can promote the proliferation of human MSCs. Therefore, the inventors first studied whether IL-17 affects the immunosuppressive function by affecting the proliferation of MSC.
  • the present inventors stimulated MSCs with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 for 24 hours, and then detected the proliferation of MSCs.
  • the present inventors have found that the addition of IL-17 inhibits the proliferation of MSCs while being stimulated by IFN ⁇ and TNF ⁇ (Fig. 2A). Therefore, IL-17 does not enhance its immunosuppressive ability by affecting the proliferation of MSCs.
  • iNOS is an important molecule mediating the immunosuppression of mouse MSCs.
  • the present inventors examined whether this effect of IL-17 enhancing MSC immunosuppression is dependent on the activity of iNOS. The results showed that the effect of IL-17 was completely reversed by the iNOS inhibitor L-NMMA (Fig. 2B), indicating that iNOS plays a crucial role in it.
  • Figure 2 shows that IL-17 enhances the immunosuppressive properties of MSCs depending on the activity of iNOS.
  • A MSC was treated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (concentration of IFN ⁇ +TNF ⁇ was diluted, IL-17 concentration was fixed at 10 ng/ml) for 24 hours, then 3H-Tdr was used. T cell proliferation was determined by incorporation.
  • B MSC was stimulated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each concentration was 2ng/ml) for 12 hours, and then co-cultured with A1.1 cells (MSC and The ratio of A1.1 was 1:10), or the iNOS inhibitor L-NMMA was added to the medium, and 12 hours later, T cell proliferation was measured by 3 H-Tdr incorporation. All data represent the results of three independent experiments.
  • Example 3 IL-17 enhances the immunosuppressive function of MSC independent of the effect on T cell apoptosis
  • IL-17 can synergize with the inflammatory cytokines IFN ⁇ and TNF ⁇ to enhance the immunosuppressive function of MSC, and this function is mainly reflected in the inhibition of T cell proliferation and depends on the activity of iNOS (Fig. 1, Fig. 2). .
  • MSCs can induce lymphocyte apoptosis in the presence of inflammatory cytokines IFN ⁇ and TNF ⁇ , a process that is also dependent on NO.
  • the inventors pre-stimulated MSC with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 for 12 hours, and then Co-culture was performed with A1.1 cells at a ratio of 1:10, and A1.1 cells were collected 12 hours later to detect apoptosis.
  • the present inventors have found that the ratio of apoptotic cells in A1.1 cells is less than 0.5% in the case of single culture; and when co-cultured with untreated MSCs, the proportion of apoptotic cells is significantly increased, the ratio is 3-5%.
  • Figure 3 shows that IL-17 enhances the immunosuppressive function of MSCs independent of the effects on T cell apoptosis.
  • the MSC was stimulated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 for 12 hours (the concentration of IFN ⁇ and TNF ⁇ was 5 ng/ml or 10 ng/ml, the concentration of IL-17 was always 10 ng/ml), and then The ratio of 1:10 was co-cultured with A1.1 cells. After 12 hours, A1.1 cells were collected, and after PI staining, the DNA content was analyzed by flow cytometry.
  • Example 4 IL-17 synergistic inflammatory cytokines up-regulate the expression of iNOS in MSC
  • iNOS and chemokines are the core molecules that mediate MSC immunosuppressive function, and that MSCs that initiate expression of iNOS and some chemokines require induction of the inflammatory cytokines IFN ⁇ and TNF ⁇ .
  • the above results of the present inventors show that the property of IL-17 to enhance MSC immunosuppression depends on the activity of iNOS.
  • the present inventors treated MSCs with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17, and then used real-time PCR and Western Blot to determine the expression of iNOS and chemokines.
  • IL-17 was able to significantly promote MSC expression of iNOS in combination with IFN ⁇ and TNF ⁇ at both mRNA and protein levels (Fig. 4A, B, C, D).
  • the present inventors also detected mRNA expression of chemokines involved in immunosuppression of MSC, including CCL2, CCL5, CXCL9, and CXCL10, and found that IL-17 does not cooperate with IFN ⁇ and TNF ⁇ to alter the expression of these chemokines. (Fig. 4E).
  • chemokines involved in immunosuppression of MSC including CCL2, CCL5, CXCL9, and CXCL10
  • the inventors incubated the activated spleen cell supernatant with anti-IL-17A for a period of time to neutralize the supernatant.
  • IL-17 is then used to stimulate MSCs to detect the expression of some immunoregulatory genes in MSCs.
  • the present inventors have found that activated spleen cell supernatant can significantly induce MSC expression of iNOS, CCL2, CCL5, CXCL9 and CXCL10; neutralizing the supernatant of IL-17 does not induce MSC to express iNOS well (Fig.
  • Figure 4 shows that IL-17 synergistic inflammatory cytokines up-regulate the expression of iNOS in MSCs.
  • MSCs were stimulated with different combinations of several cytokines of IFN ⁇ , TNF ⁇ and IL-17 (each cytokine concentration was 10 ng/ml), and RNA was extracted from cells after 12 hours, using Real-Time PCR. The method was used to detect the expression of several mRNAs of iNOS, CCL2, CCL5, CXCL9 and CXCL10.
  • B MSC with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each cytokine concentration was 10 ng/ml) was treated at different times, and the protein was extracted from the cells, and the protein expression of iNOS was detected by Western Blot. (C).
  • MSCs were stimulated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 for 24 hours (the concentration of IFN ⁇ and TNF ⁇ was diluted in a gradient, IL-17 concentration was fixed at 10 ng/ml), and the cell supernatant was collected. The amount of NO was measured using a Greiss reagent. (D). MSCs were stimulated with different combinations of cytokines of IFN ⁇ , TNF ⁇ and IL-17 (each cytokine concentration was 10 ng/ml). After 24 hours, the cells were extracted and the protein expression of iNOS was detected by Western Blot. (F).
  • MSCs were stimulated with untreated or anti-IL-17A-treated activated splenocyte supernatant for 24 hours, cell extract proteins were collected, and protein expression of iNOS was detected by Western Blot. (G). MSCs were stimulated with untreated or anti-IL-17A-treated activated splenocyte supernatant for 12 hours, and the cells were harvested for RNA extraction. Real-Time PCR was used to detect iNOS, CCL2, CCL5, CXCL9, and CXCL10. Expression of mRNA.
  • Example 5 Act1 is a key molecule mediating IL-17 to enhance MSC immunosuppression and related gene expression
  • IL-17 binds to heterodimers composed of IL-17RA and IL-17RC on the cell surface to activate downstream signaling pathways, while IL-17 binds to IL-17RA and IL-17RC.
  • binding of the adaptor protein Act1 to IL-17R is a central step in activating the downstream signaling pathway of IL-17.
  • IL-17 regulates target genes primarily by activating the MAPK and NF ⁇ B pathways. It has been reported that IL-17 function and signaling have obvious defects in MEF (mouse embryonic fibroblasts) in which Act1 gene is deleted.
  • the present inventors established Act1knockdown stable cells and corresponding control cells, and compared the phosphorylation of MAPK and NF ⁇ B signaling pathway proteins in the two cells after IL-17 stimulation. Similar to Act1 -/- MEF, the phosphorylation levels of I ⁇ B ⁇ , ERK, JNK, and p65 were significantly reduced in Act1knockdown MSCs after stimulation with IL-17, and IL-17 signaling was significantly inhibited (Fig. 5A).
  • the present inventors also studied the changes in immunosuppressive function and related immunoregulatory gene expression of MSCs stimulated by IFN ⁇ , TNF ⁇ and IL-17 after Act1knockdown.
  • the results showed that IL-17 could not effectively increase the level of iNOS expression induced by IFN ⁇ and TNF ⁇ after Act1knockdown (Fig. 5B, C), and IL-17 could not enhance the immunosuppressive function of MSC (Fig. 5D).
  • Figure 5 shows that the role of IL-17 in enhancing enhanced immunosuppression and gene expression in MSC requires the involvement of Act1.
  • (B). shCTRL MSC and shAct1 MSC were treated with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each cytokine concentration was 10 ng/ml) for 24 hours, and the protein was extracted and the protein of iNOS was detected by Western Blot. expression. (C).
  • shCTRL MSC and shAct1 MSC were stimulated with different combinations of cytokines of IFN ⁇ , TNF ⁇ and IL-17 (each cytokine concentration was 10 ng/ml). After 12 hours, the cells were extracted and RNA was used, and Real-Time PCR was used. The method detects the expression of iNOS mRNA.
  • D shCTRL MSC or shAct1MSC was stimulated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each concentration was 2 ng/ml) for 12 hours, and then co-cultured with A1.1 cells. (The ratio of MSC to A1.1 was 1:10), and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation. All data represent the results of three independent experiments.
  • Example 6 IL-17, IFN ⁇ and TNF ⁇ pretreated MSCs can effectively treat ConA-induced liver injury
  • CIH ConA-induced hepatitis
  • the present inventors further adopted the ConA-induced hepatitis (CIH) model to explore the role of IL-17 in the regulation of MSC immunosuppressive function in the treatment of diseases.
  • CIH is mainly a T cell-mediated acute hepatitis model, which is a good model for human acute fulminant hepatitis and viral hepatitis.
  • the activation and proliferation of T cells and the destruction of hepatocytes are important pathogenic mechanisms.
  • Many reports suggest that ConA-induced liver damage can be significantly alleviated by drugs that suppress the immune response or remove specific T cell subsets.
  • IL-17 Surrounding IL-17 can significantly enhance the ability of MSCs to inhibit T cell proliferation.
  • the inventors administered CIH mice to different pretreated GFP-MSCs (MSCs isolated from the bone marrow of GFP transgenic mice) and detected GFP-MSC aggregation in liver tissue approximately 7.5 hours after infusion. Studies have shown that MSC can locate damaged liver sites ( Figure 6A).
  • the inventors stimulated MSCs with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 for 12 hours in advance, and infused these MSCs into mice after 30 minutes of ConA injection. The degree of liver damage in the mice was observed and examined.
  • Figure 6 shows that IL-17, IFN ⁇ and TNF ⁇ pretreated MSCs are effective in treating ConA-induced liver damage.
  • Example 7 The therapeutic effect of IL-17, IFN ⁇ and TNF ⁇ pretreated MSCs on CIH depends on their immunosuppressive function on T cells without affecting the proportion of T cell subsets.
  • MSCs pretreated based on IL-17, IFN ⁇ and TNF ⁇ have a good therapeutic effect on ConA-induced liver injury, and the inventors further analyzed the specific mechanism of such pretreatment of MSC to regulate CIH.
  • the present inventors isolated mononuclear cells from the liver tissue of mice, and found that the number of liver mononuclear cells in mice treated with L-17, IFN ⁇ and TNF ⁇ pretreated MSCs was significantly reduced (Fig. 7A); Cytological analysis revealed a significant decrease in the number of T cells in CD4 + and CD8 + (Fig. 7A).
  • IL-17, IFN ⁇ and TNF ⁇ pretreated MSCs have a strong immunosuppressive function in inhibiting T cell proliferation in liver tissue after liver injury, and thus T cell-mediated immune response and damage to liver cells. Both are significantly weakened.
  • the immunomodulatory function of MSC is not only reflected in its immunosuppression of T cells, but also in the regulation of T cell subset differentiation.
  • MSC can induce Th17 cells to differentiate into Treg cells, thereby reducing the immune response.
  • the inventors further investigated whether the therapeutic effects of IL-17, IFN ⁇ , and TNF ⁇ -pretreated MSCs observed in the CIH model are also related to the effect of MSCs on T cell subset differentiation.
  • the present inventors isolated mononuclear cells from the livers of mice of different treatment groups, and detected changes in T cell subsets after stimulation with PMA and inomycin in vitro.
  • Figure 7 shows that the therapeutic effect of IL-17, IFNy and TNF[alpha] pretreated MSCs on CIH is dependent on their immunosuppressive function on T cells without affecting the proportion of T cell subsets.
  • mice were intravenously injected with MSCs pretreated with different cytokines (5 mice per group, 5 ⁇ 10 5 cells per mouse). Eight hours later, the mice were sacrificed and the liver was taken for follow-up testing.
  • A Mononuclear cells were isolated and counted after grinding the liver. The ratio of CD4 + and CD8 + T cells was analyzed by flow cytometry and converted to an absolute number.
  • B Flow cytometry analysis of the proportion of CD4 + and CD8 + T cells in the liver. (C and D).
  • Example 8 The therapeutic effect of IL-17, IFN ⁇ and TNF ⁇ pretreated MSCs on CIH depends on the expression of iNOS
  • IL-17 enhances the immunosuppressive effect of MSCs on iNOS activity in vitro (Fig. 2B), the inventors further applied in vivo experiments to verify the central role of iNOS in this process.
  • the inventors used iNOS -/- MSCs instead of WT MSCs and stimulated cells with a combination of different factors, IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17, and then used the different pretreatments of iNOS -/- MSCs. To treat CIH mice, observe the efficacy.
  • Figure 8 shows that the therapeutic effect of IL-17, IFN ⁇ and TNF ⁇ pretreated MSCs on CIH is dependent on the expression of iNOS.
  • Example 9 IL-17 can reverse the inhibition of gene expression by the RNA binding protein AUF1
  • IL-17 synergistic inflammatory cytokines IFN ⁇ and TNF ⁇ promote MSC expression of iNOS The control of the amount of mRNA of many immune molecules produced in an immune response is critical, and once the mRNA is excessively accumulated, it causes excessive activation of immune cells, which in turn triggers a hypersensitivity reaction.
  • mRNA accumulation can be regulated by a variety of mechanisms, one of the most important mechanisms is the interaction of mRNA binding protein and mRNA to accelerate the degradation of mRNA.
  • activation of signaling pathways often promotes the expression of some genes, and this promotion needs to be achieved by enhancing the stability of the mRNA.
  • IL-17 is no exception. IL-17 promotes the expression of some inflammatory molecules by increasing the stability of mRNA. Nevertheless, the mechanism of action of IL-17 to improve mRNA stability has not been fully elucidated.
  • the mRNA binding protein plays a crucial role in regulating the stability of mRNA, and it has been reported that the RNA binding protein AUF1 can negatively regulate the stability of iNOS mRNA, and knocking down AUF1 can significantly increase the expression of iNOS mRNA, so the inventors It was investigated whether AUF1, an RNA-binding protein, is involved in IL-17 regulation of iNOS gene expression in MSCs.
  • the present inventors isolated MSCs from the bone marrow of auf1 -/- mice and compared the expression of iNOS in WT MSCs and auf1 -/- MSCs after stimulation with IFN ⁇ + TNF ⁇ or IFN ⁇ + TNF ⁇ + IL-17.
  • the results showed that although IL-17 can up-regulate the expression of iNOS mRNA and protein in WTMSC in combination with IFN ⁇ and TNF ⁇ , only IFN ⁇ and TNF ⁇ can induce MSC to produce large amounts of iNOS in auf1 -/- MSC (Fig. 9A, B). IL-17 does not reflect obvious synergy. This phenomenon was also verified in the AUF1 knockdown MSC.
  • IL-17 was also significantly attenuated in the AUF1 knockdown MSC (Fig. 9C, D, E). Not only at the level of gene expression, the inventors found a similar phenomenon in the enhancement of immunosuppression by IL-17: in WTMSC, IL-17 can enhance the immunosuppression induced by IFN ⁇ and TNF ⁇ ; however, in auf1 - /- In MSC, only IFN ⁇ and TNF ⁇ can induce the strongest immunosuppression without the addition of IL-17 (Fig. 9F).
  • IL-17 can reverse the inhibition of iNOS mRNA expression by AUF1 in WT MSCs; when AUF1 is deleted, iNOS mRNA induced by IFN ⁇ and TNF ⁇ loses the inhibitory effect of AUF1 on its expression.
  • the promotion of IL-17 is significantly attenuated; AUF1 is likely to be a target for IL-17 to play a role in enhancing gene expression.
  • Figure 9 shows that IL-17 can reverse the inhibition of gene expression by the RNA binding protein AUF1.
  • WT MSC or auf1-/-MSCs were stimulated with different combinations of cytokines of IFN ⁇ , TNF ⁇ and IL-17 (each cytokine concentration was 10 ng/ml), and RNA was extracted after 12 hours. The expression of iNOS mRNA was detected by Real-Time PCR.
  • B WT MSC or auf1-/-MSC was stimulated with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each cytokine concentration was 10 ng/ml) for 24 hours, and the cells were extracted and the protein was detected by Western Blot. Protein expression.
  • C Protein expression.
  • shCTRL MSC or shAUF1 MSC was stimulated with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each cytokine concentration was 10 ng/ml) for 12 or 24 hours, and the cells were extracted and Western blot was used to detect iNOS. Protein. (F).
  • WTMSC or auf1-/-MSC was stimulated with cytokine IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each concentration was 2ng/ml) for 12 hours, and then it was combined with A1.1.
  • the cells were co-cultured (the ratio of MSC to A1.1 was 1:10), and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation. Results are expressed as mean ⁇ standard deviation. The data shown represents the results of four independent experiments.
  • Example 10 IL-17 enhances the stability of iNOS mRNA by modulating the level of AUF1
  • IL-17 can synergize with the inflammatory cytokines IFN ⁇ and TNF ⁇ to increase the expression of iNOS in the immunosuppressive function of MSC, but in the absence of AUF1, IL-17 does not reflect this synergistic effect, mainly due to IFN ⁇ . And TNF ⁇ can induce the strongest immunosuppression of auf1 -/- MSC.
  • IL-17 can reverse the degradation of iNOS mRNA caused by AUF1, AUF1 is likely to be a key target for IL-17 to play a role.
  • the inventors then studied the molecular mechanism.
  • the present inventors examined changes in AUF1 expression levels of MSCs after stimulation with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17. The results showed that the addition of IL-17 significantly reduced the protein level of AUF1 after stimulation with IFN ⁇ and TNF ⁇ (Fig. 10D).
  • IL-17 enhances the stability of iNOS mRNA by decreasing the level of AUF1, which is a key molecule that enhances immunosuppression and gene expression in IL-17.
  • Figure 10 shows that IL-17 enhances the stability of iNOS mRNA by modulating the level of AUF1.
  • MSCs were treated with IFN ⁇ +TNF ⁇ or IFN ⁇ +TNF ⁇ +IL-17 (each cytokine concentration was 10 ng/ml), and the cell extract protein was collected at the specified time point after the addition of cytokines, using Western Blot. Protein expression of AUF1 at each time point was examined.
  • Example 11 AUF1 is a key molecule mediating IL-17 signaling
  • IL-17 was unable to up - regulate the expression of the auf1 -/- MSC immunoregulatory gene in combination with the inflammatory cytokines IFN ⁇ and TNF ⁇ ; the same phenomenon was observed in Act1 -/- MSC (Fig. 5).
  • Act1 interacts with the IL-17 receptor, which mediates the downstream signaling, so Act1 is one of the key molecules in the IL-17 signaling pathway.
  • the inventors first compared the difference in phosphorylation of important signaling molecules in the IL-17 signaling pathway by AUF1 knockdown MSC (shAUF1 MSC) and its normal control cells (shCTRL MSC) after IL-17 stimulation.
  • the present inventors have found that in the case of AUF1 knockdown, phosphorylation of the key molecules p65 (NF ⁇ B pathway) and ERK (MAPK pathway) of the IL-17 signaling pathway is significantly attenuated after siRNA is stimulated by IL-17 (Fig. 11A). It is suggested that IL-17 signal transduction is inhibited, and AUF1 may be involved in IL-17 signaling.
  • AUF1 interacts with Act1 during the initiation of IL-17 signal transduction, thereby initiating activation of the downstream NF ⁇ B pathway and the MAPK pathway, thereby inducing expression of the target gene.
  • AUF1 becomes a target of IL-17 regulation, and IL-17 enhances the stability of mRNA of the target gene by decreasing the level of AUF1. Thereby promoting gene expression.
  • Figure 11 shows that AUF1 is a key molecule that mediates IL-17 signaling.
  • AUF1 knockdown cells (shAUF1 MSC) and corresponding control cells (shCTRL MSC) were treated with IL-17 (10 ng/ml) for a specified time, cells at different time points were collected, and proteins were extracted, Western Blot method. The expression of p-p65 and p-ERK protein was detected. P- stands for phosphorylation.
  • B MSCs were treated with IL-17 (10 ng/ml), and cell extract proteins were collected at the indicated time points after addition of cytokines, and immunologically co-immunized with IgG (control) or anti-mouse Act1 antibody and protein cleavage products. Precipitation, Western Blot detection of Act1 and AUF1.
  • Example 12 AUF1 is a key molecule mediating the effect of IL-17 on MSC treatment of CIH
  • IL-17 plays a key role in enhancing MSC immunosuppressive function (Fig. 6).
  • the present inventors pretreated WT MSC and auf1 -/- MSC with IFN ⁇ +TNF ⁇ , IFN ⁇ +TNF ⁇ +IL-17 for 12 hours, respectively, and then intravenously injected these differently treated cells into CIH mice to observe the therapeutic effect.
  • the present inventors found that IL-17 can significantly enhance the efficacy of WT MSCs for CIH, serum ALT levels, mononuclear cells in the liver, number of CD4 + T cells and CD8 + T cells, and degree of liver necrosis, consistent with previous results. Significant decline.
  • IFN ⁇ +TNF ⁇ +IL-17 pretreated MSC significantly enhanced the efficacy of WT MSCs on CIH: the mean level of serum ALT decreased from 8000 U/L to 2000 U/L in the untreated group; mononuclear cells in the liver were from the untreated group. 15 ⁇ 10 5 /g liver was reduced to 5 ⁇ 10 5 /g liver; CD4 + T cells were reduced from 3 ⁇ 10 5 /g liver in the untreated group to 1 ⁇ 10 5 /g liver; CD8 + T cells were untreated The group's 3.7 ⁇ 10 5 /gliver was reduced to 1 ⁇ 10 5 /g liver.
  • Figure 12 shows that AUF1 is a key molecule that mediates the effect of IL-17 on MSC treatment of CIH.
  • Example 13 inflammatory factor pretreatment of MSCs can effectively treat cirrhosis
  • C57BL/6 mice were given peritoneal carbon tetrachloride (CCL 4 ) weekly for 8 weeks to establish a model of cirrhosis.
  • mice treated with different inflammatory factors (10 ng/ml IFN ⁇ +10 ng/ml TNF ⁇ or 10 ng/ml IFN ⁇ +10 ng/ml TNF ⁇ +10 ng/ml IL-17) were administered to mice at the 8th week of the cirrhosis model.
  • Stem cells mesensymal stem cells, MSCs
  • untreated MSCs were used to observe the therapeutic effects of differently treated MSCs on cirrhosis.
  • TB total bilirubin
  • ALT alanine aminotrans-ferase
  • AST aspartate amino-transferase
  • Infusion of untreated MSCs (1 ⁇ 10 6 ) into cirrhotic model mice can effectively inhibit the levels of TB, ALT and AST in the serum of cirrhotic mice and significantly increase the level of albumin.
  • serum TB levels in cirrhotic mice were reduced by 50-60%; ALT levels were reduced by approximately 50%; AST levels were reduced by 60%; and albumin levels were increased by 5%.
  • MSCs (1 ⁇ 10 6 ) pretreated with inflammatory factor IFN ⁇ +TNF ⁇ for 12 hours, 7 days after infusion into cirrhotic mice, serum TB levels in cirrhotic mice decreased by about 70%; ALT levels decreased. 70-80%; AST levels are reduced by about 70%; albumin levels are increased by 20%. Shows better treatment of cirrhosis. Therefore, inflammatory factors pretreatment of MSCs can effectively improve the therapeutic effect of MSCs on cirrhosis.
  • IFN ⁇ +TNF ⁇ +IL-17 pretreated MSCs (1 ⁇ 10 6 ), and after 7 days of infusion into cirrhotic mice, serum TB levels in cirrhotic mice decreased by 85%; ALT levels decreased by about 80-85%. The AST level was reduced by about 80-90%; the albumin level was increased by 30%.
  • the addition of IL-17 showed a significant synergistic effect and could significantly improve the therapeutic effect of MSCs on cirrhosis.
  • IL-17 enhances the immunosuppression of MSCs.
  • IL-17 is recognized as an inflammatory factor that promotes immune responses.
  • Many studies have shown that IL-17 is a causative agent of various inflammatory diseases and autoimmune diseases, including rheumatoid arthritis, multiple sclerosis and inflammation. Sexual bowel disease, etc.
  • IL-17 is highly expressed in the serum and tissues of many autoimmune patients, and the symptoms of these autoimmune diseases can be significantly alleviated by using IL-17 neutralizing antibodies or knocking out the IL-17 gene.
  • IL-17 does not promote an immune response in all pathological conditions.
  • DSS distal endothelial suppression serum
  • IL-17 neutralizing antibodies or knocking out the IL-17 gene may accelerate disease progression.
  • MSC extran sulfate sodium
  • IL-17 can exert an immunosuppressive effect in the presence of MSC, and immunosuppression of MSCs when the present inventors neutralize IL-17 in a MSC-T cell co-culture system with a neutralizing antibody. The function will be damaged.
  • ConA-induced liver injury in mice is a classic animal model that mimics human viral or autoimmune acute hepatitis.
  • the acute immune response plays a leading role in mediating liver damage.
  • a variety of immune cells T lymphocytes, macrophages, NK cells
  • immune molecules cytokines IFN ⁇ , TNF ⁇ , IL-10, IL-22, IL-25, etc.
  • Inhibiting an immune response can be a very effective treatment for CIH.
  • the present inventors employed mouse bone marrow-derived MSCs and attempted to apply the IL-17-enhanced MSC immunosuppression property to its treatment of CIH.
  • the present inventors have shown that knockdown of AUF1 can promote the expression of iNOS induced by IFN ⁇ and TNF ⁇ in MSC, suggesting the importance of AUF1 for regulation of gene expression in MSC.
  • the inventors examined the effect of IL-17 on the stability of iNOS mRNA, and showed that IL-17 significantly enhanced the stability of iNOS mRNA induced by IFN ⁇ and TNF ⁇ .
  • IL-17 does not synergize IFN ⁇ and TNF ⁇ to enhance iNOS gene expression, and does not further enhance iNOS mRNA stability.
  • the results of co-immunoprecipitation also showed that the interaction between AUF1 and Act1 was significantly enhanced 15 min after IL-17 stimulation.
  • the present inventors also studied the changes in IL-17 signaling pathway protein activation in AUF1 knockdown MSCs, and the results showed that phosphorylation of p65, ERK and the like in the IL-17 signaling pathway was significantly decreased after AUF1 was knocked down, suggesting that AUF1 Knockdown caused impaired IL-17 signal transduction.
  • AUF1 plays a dual role in IL-17-enhanced MSC gene expression: AUF1 is required for IL-17 initiation signaling; whereas when IL-17 synergizes with other cytokines, AUF1 is also IL- The target of 17, IL-17, promotes the stability of related mRNA by decreasing the level of AUF1. Therefore, the present inventors have found for the first time that AUF1 plays an important role in mediating IL-17 enhancing the expression of immunosuppressive genes, and lays a foundation for further elucidating the molecular mechanism of IL-17 action.
  • IL-17 can enhance the immunosuppressive function of MSC.
  • This effect of IL-17 is mainly achieved by reversing the degradation of mRNA by the RNA binding protein AUF1.
  • the present inventors believe that an in-depth study of IL-17 regulation of MSC immunosuppression will facilitate the better application of MSCs in clinical practice.

Abstract

Provided are uses of IL-17 in enhancing an immune-suppression function of mesenchymal stem cells. In particular, the present invention provides uses of an interleukin-17, a derivative of interleukin-17 or an agonist thereof for preparing a preparation or kit for enhancing an immune-suppression function of the mesenchymal stem cells; up-regulating the expression of imunosuppressive factors in the mesenchymal stem cells; enhancing the stability of mRNAs of the immunosuppressive factors; reducing the expression level of RNA-binding protein AUF1; inhibiting the proliferation of T cells; and treating hepatitis or liver damage.

Description

IL-17在提高间充质干细胞免疫抑制功能中的应用Application of IL-17 in improving the immunosuppressive function of mesenchymal stem cells 技术领域Technical field
本发明属于生物医药领域,具体地说,本发明涉及IL-17在提高间充质干细胞免疫抑制功能中的应用。The present invention belongs to the field of biomedicine, and in particular, the present invention relates to the use of IL-17 for enhancing the immunosuppressive function of mesenchymal stem cells.
背景技术Background technique
间充质干细胞(mesenchymal stem cells,MSC)是干细胞家族的重要成员,来源于发育早期的中胚层和外胚层,属于多能干细胞,MSC最初在骨髓中发现,因其具有多向分化潜能、造血支持和促进干细胞植入、免疫调控和自我复制等特点而日益受到人们的关注。如间充质干细胞在体内或体外特定的诱导条件下,可分化为脂肪、骨、软骨、肌肉、肌腱、韧带、神经、肝、心肌、内皮等多种组织细胞,连续传代培养和冷冻保存后仍具有多向分化潜能,可作为理想的种子细胞用于衰老和病变引起的组织器官损伤修复。Mesenchymal stem cells (MSC) are important members of the stem cell family. They are derived from the mesoderm and ectoderm in early development and belong to pluripotent stem cells. MSCs are originally found in the bone marrow because of their multi-directional differentiation potential and hematopoiesis. Supporting and promoting stem cell implantation, immune regulation and self-replication are increasingly attracting attention. For example, mesenchymal stem cells can differentiate into various tissue cells such as fat, bone, cartilage, muscle, tendon, ligament, nerve, liver, heart muscle, endothelium, etc. after continuous induction and cryopreservation. It still has multi-directional differentiation potential and can be used as an ideal seed cell for the repair of tissue and organ damage caused by aging and lesions.
无论在体内实验还是在体外实验中,MSC对于T细胞的免疫抑制机制已经得到了广泛的研究,目前普遍认为MSC对于T细胞的抑制不是通过诱导T细胞凋亡,而是使得T细胞的细胞周期停留在G0/G1期。也有研究表明MSC可以促进激活的T细胞的凋亡,但可以促进静息状态下的T细胞的存活,这也说明MSC发挥免疫抑制依赖于T细胞活化时所释放的炎症细胞因子的刺激。The immunosuppressive mechanism of MSCs for T cells has been extensively studied both in vivo and in vitro. It is generally believed that MSCs inhibit T cells by not inducing T cell apoptosis, but in the cell cycle of T cells. Stay in G0/G1 period. Studies have also shown that MSC can promote the apoptosis of activated T cells, but can promote the survival of T cells in resting state, which also indicates that MSC exerts immunosuppression depending on the stimulation of inflammatory cytokines released by T cell activation.
白介素-17A(interleukin 17A,IL-17A,白细胞介素17,简称为IL-17)是目前已发现的30余种白细胞介素之一,按序号排在第17位。IL-17由CD4+T细胞分泌,能够诱导上皮细胞、内皮细胞、成纤维细胞合成分泌IL-6、IL-8、G-CSF、PGE2,促进ICAM-1的表达。近年来发现IL-17是一种主要由活化的T细胞产生的致炎细胞因子,可以促进T细胞的激活和刺激上皮细胞、内皮细胞、成纤维细胞产生多种细胞因子如IL-6、IL-8、粒细胞-巨噬细胞刺激因子(GM-CSF)和化学增活素及细胞黏附分子1(cellular adhesion molecule 1,CAM-1),从而导致炎症的产生。IL-17是T细胞诱导的炎症反应的早期启动因子,可以通过促进释放前炎性细胞因子来放大炎症反应。IL-17与受体结合后,可通过MAP激酶途径和核转录因子kB(nuclearfactor kB,NF-kB)途径发挥其生物学作用。Th17细胞能够分泌产生IL-17A、IL-17F、IL-6以及肿瘤坏死因子α(tumor necrosis factora,TNF-α)等,这些细胞因子可以集体动员、募集及活化中性粒细胞。Th17细胞产生的IL-17能有效地介导中性粒细胞动员的兴奋过程,从而有效地介导了组织的炎症反应。Interleukin-17A (interleukin 17A, IL-17A, interleukin-17, abbreviated as IL-17) is one of more than 30 kinds of interleukins that have been discovered so far, ranking 17th according to the serial number. IL-17 is secreted by CD4+ T cells and can induce the synthesis and secretion of IL-6, IL-8, G-CSF and PGE2 by epithelial cells, endothelial cells and fibroblasts, and promote the expression of ICAM-1. In recent years, IL-17 has been found to be a proinflammatory cytokine mainly produced by activated T cells, which can promote the activation of T cells and stimulate the production of various cytokines such as IL-6 and IL in epithelial cells, endothelial cells and fibroblasts. -8, granulocyte-macrophage stimulating factor (GM-CSF) and chemical stimulating hormone 1 and cellular adhesion molecule 1 (CAM-1), resulting in inflammation. IL-17 is an early promoter of T cell-induced inflammatory responses that amplify the inflammatory response by promoting the release of pro-inflammatory cytokines. After binding to the receptor, IL-17 exerts its biological effects through the MAP kinase pathway and the nuclear factor kB (NF-kB) pathway. Th17 cells secrete IL-17A, IL-17F, IL-6, and tumor necrosis factora (TNF-α). These cytokines can collectively mobilize, recruit, and activate neutrophils. IL-17 produced by Th17 cells can effectively mediate the excitatory process of neutrophil mobilization, thereby effectively mediating the inflammatory response of tissues.
IL-17作为一种促炎症细胞因子,能够作用于多种细胞类型,进而促进其他一些细胞因子、趋化因子和金属蛋白酶的表达,包括TNFα、IL-1β、IL-6、GM-CSF、G-CSF、CXCL1、MMP-3等。同时,IL-17还可以和其他一些细胞因子如TNFα协同促进多种靶基因的表达。体内的实验研究揭示了IL-17家族细胞因子在机体抗微生物感染中所发挥的重要作用。尽管IL-17在机体细菌和真菌感染中起到保护作用,但是IL-17信号通路的过度激活则会导致自身免疫病的发生。在人自身免疫病中,IL-17是明显上升的,许多研究表明IL-17参与了诸多自身免疫病的发病进程,包括MS(多发性硬化)、RA(类风湿性关节炎)和IBD(炎症性肠病)。As a pro-inflammatory cytokine, IL-17 can act on a variety of cell types, thereby promoting the expression of other cytokines, chemokines and metalloproteinases, including TNFα, IL-1β, IL-6, GM-CSF, G-CSF, CXCL1, MMP-3, and the like. At the same time, IL-17 can also promote the expression of multiple target genes in combination with other cytokines such as TNFα. In vivo experimental studies have revealed the important role of IL-17 family cytokines in the body's antimicrobial infection. Although IL-17 protects against bacterial and fungal infections in the body, over-activation of the IL-17 signaling pathway leads to autoimmune diseases. In human autoimmune diseases, IL-17 is significantly elevated, and many studies have shown that IL-17 is involved in the pathogenesis of many autoimmune diseases, including MS (multiple sclerosis), RA (rheumatoid arthritis), and IBD ( Inflammatory bowel disease).
发明内容Summary of the invention
本发明的目的在于提供白介素-17、白介素-17的衍生物或其激动剂的用途。It is an object of the present invention to provide use of interleukin-17, a derivative of interleukin-17 or an agonist thereof.
本发明的另一目的是提供白介素-17拮抗剂的用途。Another object of the invention is to provide the use of an interleukin-17 antagonist.
本发明的另一目的是提供一种经IL-17处理的提高了免疫抑制能力的间充质干 细胞。Another object of the present invention is to provide an IL-17-treated mesenchymal stem which has improved immunosuppressive ability. cell.
本发明的第一方面,提供了一种白介素-17、白介素-17的衍生物或其激动剂的用途,用于制备制剂或试剂盒,所述制剂或试剂盒用于:In a first aspect of the invention, there is provided a use of an interleukin-17, a derivative of interleukin-17 or an agonist thereof for the preparation of a formulation or kit for:
(1)提高间充质干细胞的免疫抑制功能;和/或(1) improving the immunosuppressive function of mesenchymal stem cells; and/or
(2)上调MSC中免疫抑制因子的表达;和/或(2) up-regulating the expression of immunosuppressive factors in MSC; and/or
(3)增强MSC中免疫抑制因子的mRNA的稳定性;和/或(3) enhancing the stability of mRNA of immunosuppressive factors in MSC; and/or
(4)降低RNA结合蛋白AUF1的表达水平;和/或(4) reducing the expression level of the RNA binding protein AUF1; and/or
(5)抑制T细胞的增殖;和/或(5) inhibiting the proliferation of T cells; and/or
(6)治疗肝炎或肝损伤。(6) Treatment of hepatitis or liver damage.
在另一优选例中,所述白介素-17是哺乳动物的白介素-17。In another preferred embodiment, the interleukin-17 is a mammalian interleukin-17.
在另一优选例中,所述白介素-17是人白介素-17。In another preferred embodiment, the interleukin-17 is human interleukin-17.
在另一优选例中,所述白介素-17的氨基酸序列如SEQ ID NO.:1所示。In another preferred embodiment, the amino acid sequence of interleukin-17 is as shown in SEQ ID NO.: 1.
在另一优选例中,所述白介素-17的衍生物包括经修饰的白介素-17分子、氨基酸序列与天然白介素-17同源且具有天然白介素-17活性的蛋白分子、白介素-17的二聚体或多聚体、含有白介素-17氨基酸序列的融合蛋白。In another preferred embodiment, the interleukin-17 derivative comprises a modified interleukin-17 molecule, a protein molecule having an amino acid sequence homologous to native interleukin-17 and having natural interleukin-17 activity, and dimerization of interleukin-17. A body or multimer, a fusion protein containing an interleukin-17 amino acid sequence.
在另一优选例中,所述经修饰的白介素-17分子是PEG化的白介素-17。In another preferred embodiment, the modified interleukin-17 molecule is PEGylated interleukin-17.
在另一优选例中,所述“氨基酸序列与天然白介素-17同源且具有天然白介素-17活性的蛋白分子”是指其氨基酸序列与SEQ ID NO.:1相比具有≥85%的同源性,较佳地≥90%的同源性,更佳地≥95%的同源性,最佳地≥98%同源性;并且具有天然白介素-17活性的蛋白分子。In another preferred embodiment, the "protein molecule having an amino acid sequence homologous to natural interleukin-17 and having natural interleukin-17 activity" means that the amino acid sequence thereof has ≥85% as compared with SEQ ID NO.: Derived, preferably > 90% homology, more preferably > 95% homology, optimally > 98% homology; and a protein molecule having native interleukin-17 activity.
在另一优选例中,所述“间充质干细胞的免疫抑制功能”是指间充质干细胞对T细胞的免疫抑制功能。In another preferred embodiment, the "immunosuppressive function of mesenchymal stem cells" refers to an immunosuppressive function of mesenchymal stem cells to T cells.
在另一优选例中,所述的制剂或试剂盒中还包含IFNγ和/或TNFα。In another preferred embodiment, the formulation or kit further comprises IFNy and/or TNF[alpha].
在另一优选例中,所述激动剂是指能够在体内或体外提高白介素-17或其衍生物的活性和/或含量的物质。所述物质可以为人工合成的或天然的化合物、蛋白、核苷酸等。In another preferred embodiment, the agonist means a substance capable of increasing the activity and/or content of interleukin-17 or a derivative thereof in vivo or in vitro. The substance may be a synthetic or natural compound, protein, nucleotide or the like.
在另一优选例中,所述MSC细胞为骨髓来源的MSC细胞、脐带来源MSC细胞、脂肪来源MSC细胞、胎盘来源MSC细胞、和/或牙髓来源MSC细胞。In another preferred embodiment, the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
在另一优选例中,所述肝炎包括自身免疫性肝炎、病毒性肝炎;所述肝损伤包括酒精性肝损伤、药物诱导性肝损伤、肝纤维化、肝硬化。In another preferred embodiment, the hepatitis comprises autoimmune hepatitis, viral hepatitis; the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
在另一优选例中,所述免疫抑制因子包括iNOS、吲哚胺-2,3-双加氧酶、PGE2、TSG6、IL-6、HO-1、IL-10、PD-L1、Galetin和/或B7-H4。In another preferred embodiment, the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
本发明的第二方面,提供了一种白介素-17拮抗剂的的用途,用于制备制剂或试剂盒,所述制剂或试剂盒用于:In a second aspect of the invention, there is provided a use of an interleukin-17 antagonist for the preparation of a formulation or kit for:
(1)降低间充质干细胞的免疫抑制功能;和/或(1) reducing the immunosuppressive function of mesenchymal stem cells; and/or
(2)下调MSC中免疫抑制因子的表达;和/或(2) down-regulating the expression of immunosuppressive factors in MSC; and/or
(3)降低MSC中免疫抑制因子的mRNA的稳定性;和/或(3) reducing the stability of mRNA of immunosuppressive factors in MSC; and/or
(4)增强RNA结合蛋白AUF1的表达水平;和/或(4) enhancing the expression level of the RNA binding protein AUF1; and/or
(5)促进T细胞的增殖。(5) Promote the proliferation of T cells.
在另一优选例中,所述白介素-17的拮抗剂是指能够降低体内或体外白介素-17活性的物质。In another preferred embodiment, the antagonist of interleukin-17 refers to a substance capable of reducing the activity of interleukin-17 in vivo or in vitro.
在另一优选例中,所述白介素-17的拮抗剂可以为小miRNA、抗白介素-17抗体或iNOS抑制剂、Act1蛋白抑制剂、AUF1蛋白激动剂、白介素-17受体的抑制剂、NFκB抑制剂、TRAF6抑制剂。In another preferred embodiment, the antagonist of interleukin-17 may be a small miRNA, an anti-interleukin-17 antibody or an iNOS inhibitor, an Act1 protein inhibitor, an AUF1 protein agonist, an interleukin-17 receptor inhibitor, NFκB. Inhibitor, TRAF6 inhibitor.
在另一优选例中,所述的制剂包括药物组合物、保健品组合物、食品组合物、或实验试剂。 In another preferred embodiment, the formulation comprises a pharmaceutical composition, a nutraceutical composition, a food composition, or an experimental agent.
在另一优选例中,所述MSC细胞为骨髓来源的MSC细胞、脐带来源MSC细胞、脂肪来源MSC细胞、胎盘来源MSC细胞、和/或牙髓来源MSC细胞。In another preferred embodiment, the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
在另一优选例中,所述免疫抑制因子包括iNOS、吲哚胺-2,3-双加氧酶、PGE2、TSG6、IL-6、HO-1、IL-10、PD-L1、Galetin和/或B7-H4。In another preferred embodiment, the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
本发明的第三方面,提供了一种组合物,所述组合物包括白介素-17或其衍生物、IFNγ和TNFα。In a third aspect of the invention, there is provided a composition comprising interleukin-17 or a derivative thereof, IFNy and TNFa.
在另一优选例中,所述白介素-17或其衍生物、IFNγ和TNFα的摩尔比为:1-10:10:10。In another preferred embodiment, the molar ratio of the interleukin-17 or its derivative, IFNγ and TNFα is from 1-10:10:10.
在另一优选例中,所述组合物包括固态制剂、液态制剂,较佳地为干粉或溶液形式。In another preferred embodiment, the composition comprises a solid formulation, a liquid formulation, preferably in the form of a dry powder or solution.
在另一优选例中,所述的组合物为药物组合物。In another preferred embodiment, the composition is a pharmaceutical composition.
本发明的第四方面,提供了一种分离的MSC细胞群,所述MSC细胞群由MSC细胞构成或基本上由MSC细胞构成,并且所述的MSC细胞具有增强的抑制T细胞增殖的能力,In a fourth aspect of the invention, there is provided an isolated population of MSC cells consisting of or consisting essentially of MSC cells, and said MSC cells having enhanced ability to inhibit T cell proliferation,
并且,所述的MSC细胞选自下组:And, the MSC cells are selected from the group consisting of:
(1)体外经预处理的MSC细胞群,其中所述预处理指用(i)白介素-17或其衍生物、(ii)IFNγ和(iii)TNFα同时、依次或先后进行处理;(1) an in vitro pretreated MSC cell population, wherein the pretreatment refers to simultaneous, sequential or sequential treatment with (i) interleukin-17 or a derivative thereof, (ii) IFNγ and (iii) TNFα;
(2)Act1蛋白过表达和/或AUF1蛋白缺失或活性降低的细胞群;和(2) a cell population in which Act1 protein is overexpressed and/or AUF1 protein is deleted or reduced in activity;
(3)体外经预处理的MSC细胞群,其中所述预处理指用IFNγ和TNFα进行处理;(3) an in vitro pretreated MSC cell population, wherein the pretreatment refers to treatment with IFNγ and TNFα;
(4)组(1)、组(2)和组(3)的组合。(4) A combination of group (1), group (2), and group (3).
在另一优选例中,所述预处理中,条件如下:0.5-1000ng/ml(较佳地1-200ng/ml,更佳地1-20ng/ml,最佳地1-10ng/ml)的IFNγ、0.5-1000ng/ml(较佳地1-200ng/ml,更佳地1-20ng/ml,最佳地1-10ng/ml)的TNFα,和0.2-1000ng/ml(较佳地0.5-200ng/ml,更佳地1-20ng/ml,最佳地1-10ng/ml)白介素-17。In another preferred embodiment, in the pretreatment, the conditions are as follows: 0.5-1000 ng/ml (preferably 1-200 ng/ml, more preferably 1-20 ng/ml, optimally 1-10 ng/ml) IFNγ, 0.5-1000 ng/ml (preferably 1-200 ng/ml, more preferably 1-20 ng/ml, optimally 1-10 ng/ml) of TNFα, and 0.2-1000 ng/ml (preferably 0.5-) 200 ng/ml, more preferably 1-20 ng/ml, optimally 1-10 ng/ml) interleukin-17.
在另一优选例中,所述预处理中,使用1-2ng/ml IFNγ、1-2ng/ml TNFα和0.5-2ng/ml白介素-17;或使用10ng/ml IFNγ、10ng/ml TNFα和10ng/ml白介素-17。In another preferred embodiment, 1-2 ng/ml IFNγ, 1-2 ng/ml TNFα, and 0.5-2 ng/ml interleukin-17 are used in the pretreatment; or 10 ng/ml IFNγ, 10 ng/ml TNFα, and 10 ng are used. /ml IL-17.
在另一优选例中,所述的增强的抑制T细胞增殖的能力指I1/I0≥1.5(较佳地≥2,更佳地≥4),其中,I1为所述MSC细胞对T细胞增殖的百分比抑制率;而I0为对照组的同物种的野生型的MSC细胞对T细胞增殖的百分比抑制率。In another preferred embodiment, the enhanced ability to inhibit T cell proliferation refers to I1/I0 ≥ 1.5 (preferably ≥ 2, more preferably ≥ 4), wherein I1 is the proliferation of T cells by the MSC cells. Percent inhibition rate; while I0 is the percent inhibition of T cell proliferation by wild-type MSC cells of the same species in the control group.
在另一优选例中,所述的“基本上由…构成”指MSC细胞占所述细胞群中细胞总数的至少90%,较佳地至少95%,更佳地至少99%。In another preferred embodiment, said "consisting essentially of" means that the MSC cells comprise at least 90%, preferably at least 95%, more preferably at least 99% of the total number of cells in said population of cells.
在另一优选例中,所述MSC细胞群具有以下特征:将所述MSC细胞群施用于动物后,可导致所述动物体内发生选自下组的变化:In another preferred embodiment, the MSC cell population has the characteristic that, after administration of the MSC cell population to an animal, a change in the animal from the group of the following occurs:
(a)血清ALT的水平下降;(a) a decrease in the level of serum ALT;
(b)肝脏中单个核细胞下降;(b) a decrease in mononuclear cells in the liver;
(c)CD4+T细胞数下降;和/或(c) a decrease in the number of CD4 + T cells; and/or
(d)CD8+T细胞数下降。(d) Decreased CD8 + T cell count.
在另一优选例中,In another preferred example,
(a)血清ALT的水平下降50%~80%;(a) serum ALT levels are reduced by 50% to 80%;
(b)肝脏中单个核细胞下降50%~80%;(b) Mononuclear cells in the liver are reduced by 50% to 80%;
(c)CD4+T细胞数下降50%~80%;和/或(c) a decrease in the number of CD4 + T cells by 50% to 80%; and/or
(d)CD8+T细胞数下降50%~75%。(d) The number of CD8 + T cells decreased by 50% to 75%.
在另一优选例中,所述预处理过程包括步骤:In another preferred embodiment, the preprocessing process includes the steps of:
胰酶消化MSC细胞后,先培养至细胞贴壁,然后加入上述浓度的细胞因子,继续培养6-24小时后,再消化收集细胞群。 After trypsinizing the MSC cells, the cells are cultured until the cells are attached, and then the cytokines at the above concentrations are added, and after further culture for 6-24 hours, the cell population is further digested and collected.
在另一优选例中,所述MSC细胞为骨髓来源的MSC细胞、脐带来源MSC细胞、脂肪来源MSC细胞、胎盘来源MSC细胞、和/或牙髓来源MSC细胞等。In another preferred embodiment, the MSC cells are bone marrow-derived MSC cells, umbilical cord-derived MSC cells, adipose-derived MSC cells, placental-derived MSC cells, and/or pulp-derived MSC cells.
本发明的第五方面,提供了一种遗传改造的细胞株,所述细胞株是经基因工程改造从而导致内源的Act1蛋白过表达和/或AUF1蛋白缺失或活性降低。In a fifth aspect of the invention, there is provided a genetically engineered cell strain which is genetically engineered to result in overexpression of an endogenous Actl protein and/or a decrease in AUF1 protein or activity.
在另一优选例中,所述细胞株为哺乳动物MSC细胞株。In another preferred embodiment, the cell strain is a mammalian MSC cell line.
在另一优选例中,所述细胞株用于增强间充质干细胞的免疫抑制功能。In another preferred embodiment, the cell strain is for enhancing the immunosuppressive function of mesenchymal stem cells.
本发明的第六方面,提供了一种分离的蛋白复合物,所述蛋白复合物为Act1蛋白和AUF1蛋白结合的蛋白复合物。In a sixth aspect of the invention, an isolated protein complex is provided, the protein complex being a protein complex bound by an Act1 protein and an AUF1 protein.
在另一优选例中,所述蛋白复合物分子量在80KD~130KD。In another preferred embodiment, the protein complex has a molecular weight of from 80 KD to 130 KD.
本发明的第七方面,提供了本发明第六方面所述蛋白复合物的应用,所述用途为用于筛选药物或化合物,所述药物或化合物促进或抑制Act1蛋白和AUF1蛋白形成所述的复合物。A seventh aspect of the invention provides the use of the protein complex of the sixth aspect of the invention for screening a drug or a compound which promotes or inhibits the formation of the Act1 protein and the AUF1 protein Complex.
在另一优选例中,所述的药物用于:In another preferred embodiment, the medicament is for:
(1)提高间充质干细胞的免疫抑制功能;和/或(1) improving the immunosuppressive function of mesenchymal stem cells; and/or
(2)上调MSC中免疫抑制因子的表达;和/或(2) up-regulating the expression of immunosuppressive factors in MSC; and/or
(3)增强MSC中免疫抑制因子的mRNA的稳定性;和/或(3) enhancing the stability of mRNA of immunosuppressive factors in MSC; and/or
(4)降低RNA结合蛋白AUF1的表达水平;和/或(4) reducing the expression level of the RNA binding protein AUF1; and/or
(5)抑制T细胞的增殖;和/或(5) inhibiting the proliferation of T cells; and/or
(6)治疗肝炎或肝损伤。(6) Treatment of hepatitis or liver damage.
在另一优选例中,当应用所述蛋白复合物筛选药物时,所述应用包括步骤:In another preferred embodiment, when the protein complex is used to screen for a drug, the application comprises the steps of:
(a)将目标物质与MSC细胞共培养;(a) co-cultivating the target substance with MSC cells;
(b)检测培养的细胞中所述蛋白复合物的含量。当细胞中所述蛋白复合物含量较空白对照升高时,则该物质为阳性候选物质。(b) detecting the content of the protein complex in the cultured cells. When the protein complex content in the cell is higher than the blank control, the substance is a positive candidate substance.
在另一优选例中,所述的筛选中还包括阳性对照组,较佳地所述的阳性对照组中添加IL17。(实验表明,IL17能增强两个蛋白的结合形成复合物的能力。)In another preferred embodiment, the screening further comprises a positive control group, preferably IL17 is added to the positive control group. (Experiments have shown that IL17 enhances the ability of two proteins to form a complex.)
在另一优选例中,所述MSC细胞为骨髓来源的MSC细胞、脐带来源MSC细胞、脂肪来源MSC细胞、胎盘来源MSC细胞、和/或牙髓来源MSC细胞。In another preferred embodiment, the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
在另一优选例中,所述肝炎包括自身免疫性肝炎、病毒性肝炎;所述肝损伤包括酒精性肝损伤、药物诱导性肝损伤、肝纤维化、肝硬化。In another preferred embodiment, the hepatitis comprises autoimmune hepatitis, viral hepatitis; the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
在另一优选例中,所述免疫抑制因子包括iNOS、吲哚胺-2,3-双加氧酶、PGE2、TSG6、IL-6、HO-1、IL-10、PD-L1、Galetin和/或B7-H4。In another preferred embodiment, the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
本发明的第八方面,提供了一种试剂盒,所述试剂盒中含有以下组分:In an eighth aspect of the invention, a kit is provided, the kit comprising the following components:
(a)白介素-17或其衍生物;(a) interleukin-17 or a derivative thereof;
(b)IFNγ;和(b) IFNγ; and
(c)TNFα;(c) TNFα;
以及使用说明书,And instructions for use,
其中,所述的组分(a)、(b)和(c)分别位于一个或多个不同的容器或位于同一容器中。Wherein the components (a), (b) and (c) are respectively located in one or more different containers or in the same container.
在另一优选例中,所述的说明书中描述:所述试剂盒用于In another preferred embodiment, the description describes: the kit is for
(1)提高间充质干细胞的免疫抑制功能;和/或(1) improving the immunosuppressive function of mesenchymal stem cells; and/or
(2)上调MSC中免疫抑制因子的表达;和/或(2) up-regulating the expression of immunosuppressive factors in MSC; and/or
(3)增强MSC中免疫抑制因子的mRNA的稳定性;和/或(3) enhancing the stability of mRNA of immunosuppressive factors in MSC; and/or
(4)降低RNA结合蛋白AUF1的表达水平;和/或(4) reducing the expression level of the RNA binding protein AUF1; and/or
(5)抑制T细胞的增殖。 (5) inhibiting the proliferation of T cells.
在另一优选例中,所述MSC细胞为骨髓来源的MSC细胞、脐带来源MSC细胞、脂肪来源MSC细胞、胎盘来源MSC细胞、和/或牙髓来源MSC细胞。In another preferred embodiment, the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
在另一优选例中,所述肝炎包括自身免疫性肝炎、病毒性肝炎;所述肝损伤包括酒精性肝损伤、药物诱导性肝损伤、肝纤维化、肝硬化。In another preferred embodiment, the hepatitis comprises autoimmune hepatitis, viral hepatitis; the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
在另一优选例中,所述免疫抑制因子包括iNOS、吲哚胺-2,3-双加氧酶、PGE2、TSG6、IL-6、HO-1、IL-10、PD-L1、Galetin和/或B7-H4。In another preferred embodiment, the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
本发明的第九方面,提供了一种药盒,所述药盒中包括如权利要求3所述的药物组合物和说明书,所述说明书中记载该药物组合物用于:According to a ninth aspect of the invention, there is provided a kit comprising the pharmaceutical composition according to claim 3 and instructions, wherein the pharmaceutical composition is described for:
(1)提高间充质干细胞的免疫抑制功能;和/或(1) improving the immunosuppressive function of mesenchymal stem cells; and/or
(2)上调MSC中免疫抑制因子的表达;和/或(2) up-regulating the expression of immunosuppressive factors in MSC; and/or
(3)增强免疫抑制因子的mRNA的稳定性;和/或(3) enhancing the stability of mRNA of an immunosuppressive factor; and/or
(4)降低RNA结合蛋白AUF1的表达水平;和/或(4) reducing the expression level of the RNA binding protein AUF1; and/or
(5)抑制T细胞的增殖;和/或(5) inhibiting the proliferation of T cells; and/or
(6)治疗肝炎或肝损伤。(6) Treatment of hepatitis or liver damage.
在另一优选例中,所述MSC细胞为骨髓来源的MSC细胞、脐带来源MSC细胞、脂肪来源MSC细胞、胎盘来源MSC细胞、和/或牙髓来源MSC细胞。In another preferred embodiment, the MSC cells are bone marrow derived MSC cells, umbilical cord derived MSC cells, adipose derived MSC cells, placental derived MSC cells, and/or pulp derived MSC cells.
在另一优选例中,所述肝炎包括自身免疫性肝炎、病毒性肝炎;所述肝损伤包括酒精性肝损伤、药物诱导性肝损伤、肝纤维化、肝硬化。In another preferred embodiment, the hepatitis comprises autoimmune hepatitis, viral hepatitis; the liver damage includes alcoholic liver damage, drug-induced liver damage, liver fibrosis, cirrhosis.
在另一优选例中,所述免疫抑制因子包括iNOS、吲哚胺-2,3-双加氧酶、PGE2、TSG6、IL-6、HO-1、IL-10、PD-L1、Galetin和/或B7-H4。In another preferred embodiment, the immunosuppressive factors include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin, and / or B7-H4.
本发明的第十方面,提供了一种治疗肝炎或肝损伤的方法,所述方法包括步骤:In a tenth aspect of the invention, a method of treating hepatitis or liver damage is provided, the method comprising the steps of:
给需要的对象施加治疗有效量的MSC细胞。A therapeutically effective amount of MSC cells is administered to the subject in need thereof.
在另一优选例中,所述肝损伤为肝硬化。在另一优选例中,所述对象为哺乳动物(如,人)。In another preferred embodiment, the liver damage is cirrhosis. In another preferred embodiment, the subject is a mammal (e.g., a human).
在另一优选例中,所述施加为静脉输注。In another preferred embodiment, the application is an intravenous infusion.
在另一优选例中,所述MSC细胞为本发明第四方面所述的MSC细胞群。In another preferred embodiment, the MSC cell is the MSC cell population of the fourth aspect of the invention.
在另一优选例中,所述MSC细胞为体外经预处理的MSC细胞群,其中所述预处理指用(i)白介素-17或其衍生物、(ii)IFNγ和(iii)TNFα同时、依次或先后进行处理。In another preferred embodiment, the MSC cell is a pre-treated MSC cell population, wherein the pretreatment refers to (i) interleukin-17 or a derivative thereof, (ii) IFNγ, and (iii) TNFα simultaneously. Process in sequence or sequentially.
在另一优选例中,所述所述MSC细胞为体外经预处理的MSC细胞群,其中所述预处理指用IFNγ和TNFα同时、依次或先后进行处理。In another preferred embodiment, the MSC cells are in vitro pretreated MSC cell populations, wherein the pretreatment refers to simultaneous, sequential or sequential treatment with IFNγ and TNFα.
在另一优选例中,所述预处理过程中MSC细胞群的密度为1×104细胞/ml-5×106细胞/ml,优选为5×104细胞/ml-5×105细胞/ml。In another preferred embodiment, the density of the MSC cell population during the pretreatment is 1×10 4 cells/ml-5×10 6 cells/ml, preferably 5×10 4 cells/ml-5×10 5 cells. /ml.
在另一优选例中,所述预处理过程中白介素-17的浓度为0.5-1000ng/ml,优选为1-100ng/ml。In another preferred embodiment, the concentration of interleukin-17 during the pretreatment is from 0.5 to 1000 ng/ml, preferably from 1 to 100 ng/ml.
在另一优选例中,所述预处理过程中IFNγ的浓度为0.5-1000ng/ml,优选为1-100ng/ml。In another preferred embodiment, the concentration of IFNγ during the pretreatment is from 0.5 to 1000 ng/ml, preferably from 1 to 100 ng/ml.
在另一优选例中,所述预处理过程中TNFα的浓度为0.5-1000ng/ml,优选为1-100ng/ml。In another preferred embodiment, the concentration of TNFα in the pretreatment is from 0.5 to 1000 ng/ml, preferably from 1 to 100 ng/ml.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here.
附图说明DRAWINGS
图1包括图1A、图1B、图1C、图1D、图1E、图1F、图1G、和图1H显示IL-17增强MSC的免疫抑制功能。 1 includes FIGS. 1A, 1B, 1C, 1D, 1E, 1F, 1G, and 1H showing that IL-17 enhances the immunosuppressive function of MSCs.
图2包括图2A和图2B显示IL-17增强MSC免疫抑制的特性依赖于iNOS的活性。Figure 2, comprising Figures 2A and 2B, shows that IL-17 enhances the immunosuppressive properties of MSCs depending on the activity of iNOS.
图3显示IL-17增强MSC的免疫抑制功能不依赖于对T细胞凋亡的影响。Figure 3 shows that IL-17 enhances the immunosuppressive function of MSCs independent of the effects on T cell apoptosis.
图4包括图4A、图4B、图4C、图4D、图4E、图4F、和图4G显示IL-17协同炎症细胞因子上调MSC中iNOS的表达。Figure 4, comprising Figures 4A, 4B, 4C, 4D, 4E, 4F, and 4G, shows that IL-17 synergistic inflammatory cytokines up-regulate the expression of iNOS in MSCs.
图5包括图5A、图5B、图5C、和图5D显示IL-17在MSC中发挥的增强免疫抑制和基因表达的作用需要Act1的参与。Figure 5, comprising Figures 5A, 5B, 5C, and 5D, shows that IL-17 plays an important role in enhancing immunosuppression and gene expression in MSCs requiring Act1 involvement.
图6包括图6A、图6B、和图6C显示IL-17、IFNγ和TNFα预处理的MSC能有效地治疗ConA诱导的肝损伤。Figure 6 includes Figures 6A, 6B, and 6C showing that IL-17, IFNy, and TNF[alpha] pretreated MSCs are effective in treating ConA-induced liver damage.
图7包括图7A、图7B、图7C、和图7D显示IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于其对T细胞的免疫抑制功能,而不影响T细胞亚群的比例。Figure 7 includes Figures 7A, 7B, 7C, and 7D showing that the therapeutic effects of IL-17, IFNγ, and TNFα pretreated MSCs on CIH depend on their immunosuppressive function on T cells without affecting T cell subsets. proportion.
图8中的图A-E显示IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于iNOS的表达。Panels A-E in Figure 8 show that the therapeutic effect of IL-17, IFNy and TNF[alpha] pretreated MSCs on CIH is dependent on the expression of iNOS.
图9包括图9A、图9B、图9C、图9D、图9E、和图9F显示IL-17可以逆转RNA结合蛋白AUF1对基因表达的抑制作用。Figure 9 includes Figures 9A, 9B, 9C, 9D, 9E, and 9F showing that IL-17 can reverse the inhibition of gene expression by the RNA binding protein AUF1.
图10中的图A-图D显示IL-17通过调节AUF1的水平来增强iNOS mRNA的稳定性。Panels A-D of Figure 10 show that IL-17 enhances the stability of iNOS mRNA by modulating the level of AUF1.
图11中的图A-图B显示AUF1是介导IL-17信号转导的的关键分子。Panels A-B of Figure 11 show that AUF1 is a key molecule that mediates IL-17 signaling.
图12中的图A-图D显示AUF1是介导IL-17增强MSC治疗CIH效果的关键分子。Panels A-D of Figure 12 show that AUF1 is a key molecule that mediates IL-17 enhancing MSC treatment of CIH.
具体实施方式detailed description
本发明人通过广泛而深入的研究,意外地发现IL-17能显著地提高间充质干细胞的免疫抑制功能,抑制T细胞的增殖;该功能通过降低RNA结合蛋白AUF1的表达水平增强iNOS的mRNA的稳定性并上调MSC中iNOS的表达来实现。体内和体外实验结果证明IL-17预处理的MSC能有效地治疗CONA诱导的肝损伤。在此基础上完成了本发明。Through extensive and intensive research, the present inventors have unexpectedly discovered that IL-17 can significantly enhance the immunosuppressive function of mesenchymal stem cells and inhibit the proliferation of T cells; this function enhances the mRNA of iNOS by decreasing the expression level of the RNA binding protein AUF1. The stability is achieved by up-regulating the expression of iNOS in MSC. In vivo and in vitro experiments demonstrated that IL-17 pretreated MSC can effectively treat CONA-induced liver injury. The present invention has been completed on this basis.
术语the term
术语“治疗”是指基于治愈、缓解、改善、减轻、影响治疗对象疾病、症状、疾病体质(predisposition)的目的而给予需要治疗的对象本发明的白介素-17。The term "treating" refers to the administration of interleukin-17 of the present invention to a subject in need of treatment for the purpose of curing, alleviating, ameliorating, alleviating, affecting the disease, symptoms, and disease predisposition of the subject.
术语“治疗对象”是指鼠、人及其他哺乳动物。The term "therapeutic subject" refers to rats, humans, and other mammals.
术语“治疗有效量”是指能够在治疗对象体内达到治疗目的的白介素-17的量。本领域的普通技术人员应理解,所述“治疗有效量”可随白介素-17的给药途径、所用药物辅料以及与其他药物联合用药情况的不同而有所不同。The term "therapeutically effective amount" refers to an amount of interleukin-17 that is capable of achieving a therapeutic purpose in a subject. It will be understood by one of ordinary skill in the art that the "therapeutically effective amount" may vary depending on the route of administration of interleukin-17, the pharmaceutical excipients used, and the combination with other drugs.
间充质干细胞(mesenchymal stem cells,MSC)Mesenchymal stem cells (MSC)
本发明人的研究成果表明,MSC通过一氧化氮(NO)和趋化因子的联合作用发挥对T细胞的免疫抑制功能。T细胞在其受体(TCR)活化后能分泌大量的炎症细胞因子,包括IFNγ、TNFα、IL-1α和IL-1β,这些炎症细胞因子能够刺激MSC使其分泌大量iNOS和趋化因子,这些趋化因子能够招募T细胞到MSC周围,通过iNOS的代谢产物NO抑制T细胞的增殖。此外,本发明人还发现转录因子C/EBPβ和STAT1在诱导iNOS产生的过程中是关键的。本发明人同时也研究了不同物种来源的MSC在介导对T细胞的免疫抑制中的不同机制。对于小鼠来说,iNOS是MSC抑制T细胞的关键分子,而对人来说,IDO则是MSC抑制T细胞的关键分子。然而,也有其他研究者认为HLA-G5、TGF-β和IL-10是介导人MSC发挥免疫抑制的关键分子。The inventors' research results show that MSC exerts an immunosuppressive function on T cells through the combined action of nitric oxide (NO) and chemokines. T cells secrete a large number of inflammatory cytokines, including IFNγ, TNFα, IL-1α and IL-1β, after activation of their receptor (TCR). These inflammatory cytokines stimulate MSCs to secrete large amounts of iNOS and chemokines. Chemokines are capable of recruiting T cells to the periphery of MSCs and inhibiting T cell proliferation through the metabolite NO of iNOS. Furthermore, the inventors have also found that the transcription factors C/EBPβ and STAT1 are critical in the induction of iNOS production. The present inventors also studied different mechanisms by which MSCs of different species are mediated in immunosuppression of T cells. For mice, iNOS is a key molecule for MSCs to suppress T cells, while for humans, IDO is a key molecule for MSCs to inhibit T cells. However, other researchers believe that HLA-G5, TGF-β and IL-10 are key molecules that mediate the immunosuppression of human MSCs.
免疫抑制因子 Immunosuppressive factor
如本发明所用,“免疫抑制因子”是指可以下调机体过强免疫反应的因子或分子。过强的免疫反应包括B细胞、T细胞、自然杀伤细胞、巨噬细胞、树突状细胞、中性粒细胞等细胞介导的超出机体正常范围的免疫反应,可能导致炎症疾病,如过敏反应、自身免疫性疾病等。As used herein, "immunosuppressive factor" refers to a factor or molecule that can downregulate the body's excessive immune response. Excessive immune response, including B cells, T cells, natural killer cells, macrophages, dendritic cells, neutrophils and other cell-mediated immune responses beyond the normal range of the body, may lead to inflammatory diseases, such as allergic reactions , autoimmune diseases, etc.
本发明中优选地免疫抑制因子包括iNOS、吲哚胺-2,3-双加氧酶、PGE2、TSG6、IL-6、HO-1、IL-10、PD-L1、Galetin和/或B7-H4。Preferred immunosuppressive factors in the present invention include iNOS, indole-2,3-dioxygenase, PGE2, TSG6, IL-6, HO-1, IL-10, PD-L1, Galetin and/or B7- H4.
白介素-17或其衍生物及其制备方法Interleukin-17 or its derivative and preparation method thereof
如本发明所用,“白介素-17”或“IL-17”是指一种蛋白质,该蛋白质(a)具有与Zhengbin Yao等.Human IL-17:a novel cytokine derived from T cells.J.Immunol.155(12),5483-5486(1995);和Submitted(11-DEC-1995)Jacqueline Kennedy,Immunology,DNAX Research Institute中描述的人/鼠白介素-17基本相同的氨基酸序列和(b)具有与天然白介素-17相同的生物学活性。本发明的白介素-17包括,但不限于:人白介素-17、重组人白介素、鼠白介素-17和/或重组鼠白介素-17。在某些实施方式中的白介素-17的氨基酸序列为:As used herein, "interleukin-17" or "IL-17" refers to a protein which has (a) and Zhengbin Yao et al. Human IL-17: a novel cytokine derived from T cells. J. Immunol. 155(12), 5483-5486 (1995); and the basic amino acid sequence of human/mouse interleukin-17 described in Submitted (11-DEC-1995) Jacqueline Kennedy, Immunology, DNAX Research Institute and (b) with natural Interleukin-17 has the same biological activity. The interleukin-17 of the present invention includes, but is not limited to, human interleukin-17, recombinant human interleukin, murine interleukin-17, and/or recombinant murine interleukin-17. In certain embodiments, the amino acid sequence of interleukin-17 is:
人IL-17氨基酸序列:(155aa)Human IL-17 amino acid sequence: (155aa)
  1 MTPGKTSLVS LLLLLSLEAI VKAGITIPRN PGCPNSEDKN FPRTVMVNLN IHNRNTNTNP1 MTPGKTSLVS LLLLLSLEAI VKAGITIPRN PGCPNSEDKN FPRTVMVNLN IHNRNTNTNP
 61 KRSSDYYNRS TSPWNLHRNE DPERYPSVIW EAKCRHLGCI NADGNVDYHM NSVPIQQEIL61 KRSSDYYNRS TSPWNLHRNE DPERYPSVIW EAKCRHLGCI NADGNVDYHM NSVPIQQEIL
121 VLRREPPHCP NSFRLEKILV SVGCTCVTPI VHHVA(SEQ ID NO.:1)121 VLRREPPHCP NSFRLEKILV SVGCTCVTPI VHHVA (SEQ ID NO.: 1)
鼠IL-17氨基酸序列:(158aa)Mouse IL-17 amino acid sequence: (158aa)
  1 MSPGRASSVS LMLLLLLSLA ATVKAAAIIP QSSACPNTEA KDFLQNVKVN LKVFNSLGAK1 MSPGRASSVS LMLLLLLSLA ATVKAAAIIP QSSACPNTEA KDFLQNVKVN LKVFNSLGAK
 61 VSSRRPSDYL NRSTSPWTLH RNEDPDRYPS VIWEAQCRHQ RCVNAEGKLD HHMNSVLIQQ61 VSSRRPSDYL NRSTSPWTLH RNEDPDRYPS VIWEAQCRHQ RCVNAEGKLD HHMNSVLIQQ
121 EILVLKREPE SCPFTFRVEK MLVGVGCTCV ASIVRQAA(SEQ ID NO.:2)121 EILVLKREPE SCPFTFRVEK MLVGVGCTCV ASIVRQAA (SEQ ID NO.: 2)
IL-17信号通路激活后,能启动很多促炎症基因的表达,这一点与一些经典的固有免疫受体的激活类似,包括IL-1R(IL-1受体)、TLR(Toll样受体)等。与IL-1R和TLR类似,IL-17RA与IL-17结合后能通过与接头蛋白Act1和TRAF6相互作用,进而激活NFκB信号通路,促进靶基因的表达。IL-17在激活NFκB通路的同时也能激活MAPK信号通路,MAPK的激活又会导致AP1的活化,从而促进靶基因的转录;另外,MAPK激活还可以通过增强mRNA稳定性从而促进基因表达。对于IL-17来说,增强mRNA稳定性是其促进基因表达的重要环节。其中,Act1是介导IL-17增强mRNA稳定性的重要分子。Activation of the IL-17 signaling pathway activates the expression of many pro-inflammatory genes, similar to the activation of some classical innate immune receptors, including IL-1R (IL-1 receptor) and TLR (Toll-like receptor). Wait. Similar to IL-1R and TLR, IL-17RA binds to IL-17 and interacts with the adaptor proteins Act1 and TRAF6 to activate NFκB signaling pathway and promote target gene expression. IL-17 activates the NFκB pathway and activates the MAPK signaling pathway. Activation of MAPK leads to activation of AP1 and promotes transcription of target genes. In addition, MAPK activation can also promote gene expression by enhancing mRNA stability. For IL-17, enhancing mRNA stability is an important part of its promotion of gene expression. Among them, Act1 is an important molecule that mediates IL-17 to enhance mRNA stability.
炎症微环境中,IL-17主要是由Th17细胞分泌的,另外还可以由其他一些细胞生成,包括γδT细胞、NKT细胞、NK细胞、中性粒细胞和嗜酸性粒细胞等。IL-17作为一种促炎症因子,参与了诸多自身免疫病的发病过程。在本发明中,本发明人发现在MSC存在时,IL-17则可以增强MSC的免疫抑制功能,从而有效地抑制T细胞增殖,说明IL-17在MSC存在时可以抑制免疫反应。In the inflammatory microenvironment, IL-17 is mainly secreted by Th17 cells, and can also be produced by other cells, including γδT cells, NKT cells, NK cells, neutrophils and eosinophils. As a pro-inflammatory factor, IL-17 is involved in the pathogenesis of many autoimmune diseases. In the present invention, the inventors have found that IL-17 can enhance the immunosuppressive function of MSC in the presence of MSC, thereby effectively inhibiting T cell proliferation, indicating that IL-17 can inhibit the immune response in the presence of MSC.
术语“基本相同的氨基酸序列”是指序列相同或由一个或多个氨基酸改变(缺失、增加、取代)引起的不同,但这种改变基本上不降低其生物学活性,即可以通过结合IL-17靶细胞受体而发生生物学功能。任何符合“基本相同”要求的白介素-17均包括在本发明内,无论它是糖基化的(即来源于天然的或来源于真核生物表达系统的)或是非糖基化的(即来源于原核生物表达系统或化学合成的)。The term "substantially identical amino acid sequence" refers to a difference in sequence or caused by one or more amino acid changes (deletion, addition, substitution), but such alteration does not substantially reduce its biological activity, ie, by binding to IL- 17 target cell receptors and biological functions. Any interleukin-17 that meets the "substantially identical" requirement is included in the invention, whether it is glycosylated (ie derived from natural or derived from eukaryotic expression systems) or non-glycosylated (ie source) In prokaryotic expression systems or chemically synthesized).
“白介素-17”还包括PEG化的IL-17以及共价修饰的IL-17蛋白质。例如,可用各种活化的分子量为5,000~100,000的聚乙二醇(PEG)修饰以使IL-17高分子化,延长其半衰期。具体操作可参见Greenwald et al.,Bioorg.Med.Chem.Lett.1994,4,2465;Caliceti et al.,IL Farmaco,1993,48,919;Zalipsky and Lee,《聚乙二醇化学:生物 技术与生物医学应用》,J.M.Harris编,Plenum Press,N.Y.,1992。"Interleukin-17" also includes PEGylated IL-17 and covalently modified IL-17 protein. For example, various activated polyethylene glycols (PEG) having a molecular weight of 5,000 to 100,000 can be used to polymerize IL-17 to prolong its half-life. For specific procedures, see Greenwald et al., Bioorg. Med. Chem. Lett. 1994, 4, 2465; Calettiti et al., IL Farmaco, 1993, 48, 919; Zalipsky and Lee, Polyethylene glycol chemistry: biology Technology and Biomedical Applications, edited by J. M. Harris, Plenum Press, N.Y., 1992.
本发明的白介素-17可用基因重组技术克隆表达的。表达的宿主细胞包括原核细胞、酵母细胞或者高等真核生物细胞。除了原核细胞,真核细胞如丝状真菌(filamentous fungi)或酵母菌(yeast)等同样适用于表达或者克隆本发明的白介素-17。用于表达糖基化的本发明的白介素-17的宿主细胞来源于多细胞有机体。无脊椎动物细胞的例子包括昆虫细胞如Drosophila S2和Spodoptera Sf9,植物细胞。适用的哺乳动物宿主细胞的例子包括中华仓鼠卵巢细胞(CHO),COS细胞。本领域的普通技术人员应知如何选择合适的宿主细胞。The interleukin-17 of the present invention can be cloned and expressed by genetic recombination techniques. Host cells for expression include prokaryotic cells, yeast cells, or higher eukaryotic cells. In addition to prokaryotic cells, eukaryotic cells such as filamentous fungi or yeast are equally suitable for expressing or cloning the interleukin-17 of the present invention. The host cell of the interleukin-17 of the present invention for expressing glycosylation is derived from a multicellular organism. Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, plant cells. Examples of suitable mammalian host cells include Chinese hamster ovary cells (CHO), COS cells. One of ordinary skill in the art will know how to select a suitable host cell.
上述宿主细胞经白介素-17表达载体或克隆载体转染或者转化后可在传统的营养基(nutrient media)中培养,所述营养基经修饰后适于诱导启动子(promoter)、选择性转化体(selecting transformant)或者扩增白介素-17编码基因序列。培养条件如培养基、温度、pH等的选择对本领域的普通技术人员来说则是应知的。如何使细胞培养繁殖力最大化的一般原则、方案以及操作技术可参见Mammalian Cell Biotechnology:a Practical Approach,M.Butler,ed.(IRL Press,1991)and Sambrook et al.,supra。The above host cells are transfected or transformed with an interleukin-17 expression vector or a cloning vector, and can be cultured in a conventional nutrient medium, which is modified to be suitable for inducing a promoter and a selective transformant. (selecting transformant) or amplifying the interleukin-17 encoding gene sequence. Selection of culture conditions such as medium, temperature, pH, etc. will be known to those of ordinary skill in the art. General principles, protocols, and techniques for how to maximize cell culture fertility can be found in Mammalian Cell Biotechnology: a Practical Approach, M. Butler, ed. (IRL Press, 1991) and Sambrook et al., supra.
本发明的白介素-17不但可以通过基因重组直接表达,也可以通过与异种多肽形成融和多肽的方式生产,后者可以是一段位于成熟蛋白质或多肽N端的信号序列,也可以是位于成熟蛋白质或多肽N端的具有特异性切割位点的其他多肽片段。The interleukin-17 of the present invention can be directly expressed not only by genetic recombination, but also by forming a fusion polypeptide with a heterologous polypeptide, which may be a signal sequence located at the N-terminus of the mature protein or polypeptide, or may be located in a mature protein or polypeptide. N-terminal other polypeptide fragments with specific cleavage sites.
IL-17二聚体及其制备方法IL-17 dimer and preparation method thereof
编码本发明IL-17二聚体或融合蛋白的DNA序列,可以全部人工合成。也可用PCR扩增或合成的方法获得IL-17第一单体和/或IL-17第二单体的编码DNA序列,然后将其拼接在一起,形成编码本发明融合蛋白的DNA序列。The DNA sequence encoding the IL-17 dimer or fusion protein of the present invention can be all synthetically synthesized. The coding DNA sequences of the IL-17 first monomer and/or the IL-17 second monomer can also be obtained by PCR amplification or synthesis and then spliced together to form a DNA sequence encoding the fusion protein of the present invention.
为了提高宿主细胞的表达量,可以对IL-17二聚体编码序列进行改造,例如采用宿主细胞偏好的密码子,消除不利于基因转录及翻译的序列。在本发明中,可以采用酵母细胞或哺乳动物细胞偏好的密码子,并采用计算机DNA软件对IL-17二聚体基因进行检测,排除在基因中不利于基因转录及翻译的序列,包括内含子剪切位点,转录终止序列等。In order to increase the expression level of the host cell, the IL-17 dimer coding sequence can be engineered, for example, using host cell-preferred codons to eliminate sequences that are detrimental to gene transcription and translation. In the present invention, the yeast cell or mammalian cell-preferred codon can be used, and the IL-17 dimer gene can be detected by computer DNA software, and the sequence which is not conducive to gene transcription and translation in the gene, including the inclusion, is excluded. Sub-shearing site, transcription termination sequence, and the like.
在获得了编码本发明新融合蛋白的DNA序列之后,将其连入合适的表达载体,再转入合适的宿主细胞。最后,培养转化后的宿主细胞,通过分离纯化得到本发明的新的融合蛋白。After obtaining the DNA sequence encoding the novel fusion protein of the present invention, it is ligated into a suitable expression vector and transferred to a suitable host cell. Finally, the transformed host cells are cultured, and the novel fusion protein of the present invention is obtained by isolation and purification.
如本文所用,术语“载体”包括质粒、粘粒、表达载体、克隆载体、病毒载体等。The term "vector," as used herein, includes plasmids, cosmids, expression vectors, cloning vectors, viral vectors, and the like.
在本发明中,可选用本领域已知的各种载体如市售的载体。比如,选用市售的载体,然后将编码本发明新融合蛋白的核苷酸序列可操作地连于表达调控序列,可以形成蛋白表达载体。In the present invention, various carriers known in the art such as commercially available carriers can be used. For example, a commercially available vector is selected, and then a nucleotide sequence encoding a novel fusion protein of the present invention is operably linked to an expression control sequence to form a protein expression vector.
如本文所用,“可操作地连于”指这样一种状况,即线性DNA序列的某些部分能够影响同一线性DNA序列其他部分的活性。例如,如果信号肽DNA作为前体表达并参与多肽的分泌,那么信号肽(分泌前导序列)DNA就是可操作地连于多肽DNA;如果启动子控制序列的转录,那么它是可操作地连于编码序列;如果核糖体结合位点被置于能使其翻译的位置时,那么它是可操作地连于编码序列。一般,“可操作地连于”意味着相邻近,而对于分泌前导序列则意味着在阅读框中相邻。As used herein, "operably linked" refers to a condition in which portions of a linear DNA sequence are capable of affecting the activity of other portions of the same linear DNA sequence. For example, if a signal peptide DNA is expressed as a precursor and is involved in the secretion of a polypeptide, then the signal peptide (secretion leader sequence) DNA is operably linked to the polypeptide DNA; if the promoter controls the transcription of the sequence, then it is operably linked to A coding sequence; if the ribosome binding site is placed at a position that enables translation, then it is operably linked to the coding sequence. Generally, "operably linked to" means adjacent, and for secretory leader sequences means adjacent in the reading frame.
在本发明中,术语“宿主细胞”包括原核细胞和真核细胞。常用的原核宿主细胞的例子包括大肠杆菌、枯草杆菌等。常用的真核宿主细胞包括酵母细胞,昆虫细胞、和哺乳动物细胞等。较佳地,该宿主细胞是真核细胞,更佳地是哺乳动物细胞。In the present invention, the term "host cell" includes prokaryotic cells and eukaryotic cells. Examples of commonly used prokaryotic host cells include Escherichia coli, Bacillus subtilis and the like. Commonly used eukaryotic host cells include yeast cells, insect cells, and mammalian cells. Preferably, the host cell is a eukaryotic cell, more preferably a mammalian cell.
在获得转化的宿主细胞后,可在适合表达本发明融合蛋白的条件下培养该细胞,从而表达出融合蛋白。然后再分离出表达的融合蛋白。 After obtaining the transformed host cell, the cell can be cultured under conditions suitable for expression of the fusion protein of the present invention to express the fusion protein. The expressed fusion protein is then isolated.
药物组合物和施用方法Pharmaceutical composition and method of administration
本发明的药物组合物包含安全、有效量范围内的本发明IL-17或其衍生物及药理上可以接受的赋形剂或载体。其中“安全、有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有0.001-1000mg的IL-17或其衍生物/剂,较佳地0.05-300mg的IL-17或其衍生物/剂,更佳地,含有0.5-200mg的IL-17或其衍生物/剂。The pharmaceutical compositions of the present invention comprise a safe or effective amount of the IL-17 or derivative thereof of the present invention and a pharmaceutically acceptable excipient or carrier. By "safe, effective amount" it is meant that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. In general, the pharmaceutical composition contains 0.001 to 1000 mg of IL-17 or a derivative/agent thereof, preferably 0.05 to 300 mg of IL-17 or a derivative/agent thereof, more preferably 0.5 to 200 mg of IL-17 or Its derivatives/agents.
本发明的IL-17或其衍生物及其药理上可接受的盐可制成各种制剂,其中包含安全、有效量范围内的本发明IL-17或其衍生物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全、有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。化合物的安全、有效量根据治疗对象的年龄、病情、疗程等具体情况来确定。The IL-17 or a derivative thereof of the present invention and a pharmacologically acceptable salt thereof can be formulated into various preparations comprising the IL-17 or derivative thereof of the present invention or a pharmacologically acceptable amount thereof in a safe and effective amount Salts and pharmaceutically acceptable excipients or carriers. By "safe, effective amount" it is meant that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. The safe and effective amount of the compound is determined according to the specific conditions such as the age, condition, and course of treatment of the subject.
“药理上可以接受的赋形剂或载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能与本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药理上可以接受的赋形剂或载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如
Figure PCTCN2015077862-appb-000001
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
"Pharmacologically acceptable excipient or carrier" means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity . By "compatibility" it is meant herein that the components of the composition are capable of intermixing with the compounds of the invention and with each other without significantly reducing the potency of the compound. Examples of pharmaceutically acceptable excipients or carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants ( Such as stearic acid, magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyol (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifier (such as
Figure PCTCN2015077862-appb-000001
), a wetting agent (such as sodium lauryl sulfate), a coloring agent, a flavoring agent, a stabilizer, an antioxidant, a preservative, a pyrogen-free water, and the like.
施用本发明IL-17或其衍生物时,可以口服、直肠、肠胃外(静脉内、肌肉内或皮下)、局部给药。When IL-17 or a derivative thereof of the present invention is administered, it can be administered orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously) or topically.
本发明IL-17或其衍生物可以单独给药,或者与其他药学上可接受的化合物联合给药。The IL-17 or derivative thereof of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
含有本发明的白介素-17或其衍生物的微胶囊可用于本发明的白介素-17的缓释给药。重组蛋白的微囊缓释给药技术已成功应用于重组人生长激素(rhGH)、重组人干扰素(rhIFN)、白介素-2和MNrgp120(Johnson et al.,Nat.Med.,2:795-799(1996);Yasuda,Biomed.Ther 27:1221-1223(1993);WO 97/03692,WO 96/40072,WO 96/07399;U.S.Pat.No.5654010。The microcapsule containing the interleukin-17 of the present invention or a derivative thereof can be used for the sustained release administration of the interleukin-17 of the present invention. The microcapsule sustained release drug delivery technology of recombinant protein has been successfully applied to recombinant human growth hormone (rhGH), recombinant human interferon (rhIFN), interleukin-2 and MNrgp120 (Johnson et al., Nat. Med., 2:795- 799 (1996); Yasuda, Biomed. Ther 27: 1221-1223 (1993); WO 97/03692, WO 96/40072, WO 96/07399; US Pat. No. 5,654,010.
本发明的白介素-17或其衍生物的缓释制剂可用具有良好生物兼容性和宽泛生物可降解性的乳酸羟基乙酸高聚物(PLGA)制备。PLGA的降解产物,乳酸和羟基乙酸可被人体很快清除。而且,该高聚物的降解能力可随其分子量和组成的不同,从几个月延长到几年(Lewis,“Controlled release of bioactive agents form lactide/glycolide polymer,”in:M.Chasin and R.Langer(Eds.),Biodegradable Polymers as Drug Delivery Systems(Marcel Dekker:New York,1990),pp.1-41))。The sustained release preparation of the interleukin-17 of the present invention or a derivative thereof can be produced by a lactic acid glycolic acid high polymer (PLGA) having good biocompatibility and broad biodegradability. The degradation products of PLGA, lactic acid and glycolic acid can be quickly eliminated by the human body. Moreover, the degradation ability of the polymer can be extended from several months to several years depending on its molecular weight and composition (Lewis, "Controlled release of bioactive agents form lactide/glycolide polymer," in: M. Chasin and R. Langer (Eds.), Biodegradable Polymers as Drug Delivery Systems (Marcel Dekker: New York, 1990), pp. 1-41)).
使用药物组合物时,是将安全有效量的本发明IL-17或其二聚体适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,每次给药剂量通常为0.01~300mg,优选0.5~100mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。When a pharmaceutical composition is used, a safe and effective amount of IL-17 or a dimer thereof of the present invention is applied to a mammal (e.g., a human) in need of treatment, wherein the dose at the time of administration is a pharmaceutically effective effective dose for 60 kg. For a person weighing, the dose is usually from 0.01 to 300 mg, preferably from 0.5 to 100 mg. Of course, specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
本发明的主要优点在于:The main advantages of the invention are:
(1)首次揭示了IL-17能显著地提高间充质干细胞的免疫抑制功能。(1) It was first revealed that IL-17 can significantly enhance the immunosuppressive function of mesenchymal stem cells.
(2)首次发现了IL-17具有抑制T细胞的增殖的能力。(2) It was first discovered that IL-17 has the ability to inhibit the proliferation of T cells.
(3)首次阐明了IL-17通过降低RNA结合蛋白AUF1的表达水平增强iNOS的mRNA的稳定性来上调MSC中iNOS的表达进而实现对间充质干细胞的免疫抑制。(3) It was demonstrated for the first time that IL-17 up-regulates the expression of iNOS in MSC by decreasing the expression level of the RNA binding protein AUF1 to enhance the mRNA stability of iNOS, thereby achieving immunosuppression of mesenchymal stem cells.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按 照常规条件如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。The invention is further illustrated below in conjunction with specific embodiments. It is to be understood that the examples are not intended to limit the scope of the invention. The experimental methods in the following examples that do not specify the specific conditions are usually The conditions are as described in conventional conditions such as Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or in accordance with the conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise stated.
材料和方法Materials and Method
实验材料Experimental Materials
本发明所用的试剂和实验材料均可从市售获得,其中生物材料来源如下。The reagents and experimental materials used in the present invention are commercially available, and the sources of the biological materials are as follows.
重组小鼠IFNγ,TNFα,IL-17A,抗IL-17A抗体购自eBiosciences(La Jolla,CA);重组小鼠IL-2购自R&D Systems(Minneapolis,MN);抗β-actin、GAPDH、iNOS、p65、p-IκBα、p-p65、p-JNK、p-ERK1/2抗体购自Cell Signaling Technology(Danvers,MA);抗Act1抗体购自Santa Cruz Biotechnology(Dallas,TX);L-NMMA、propidium iodide、actinomycin D购自Sigma-Aldrich(St.Louis,MO);Concanavaline A(ConA)购自Vector Labs(Burlingame,CA).Recombinant mouse IFNγ, TNFα, IL-17A, anti-IL-17A antibody was purchased from eBiosciences (La Jolla, CA); recombinant mouse IL-2 was purchased from R&D Systems (Minneapolis, MN); anti-β-actin, GAPDH, iNOS , p65, p-IκBα, p-p65, p-JNK, p-ERK1/2 antibody were purchased from Cell Signaling Technology (Danvers, MA); anti-Act1 antibody was purchased from Santa Cruz Biotechnology (Dallas, TX); L-NMMA, Propidium iodide, actinomycin D was purchased from Sigma-Aldrich (St. Louis, MO); Concanavaline A (ConA) was purchased from Vector Labs (Burlingame, CA).
C57BL/6小鼠购自上海斯莱克实验动物有限公司。动物饲养于上海交通大学医学院实验动物科学部清洁级环境中。C57BL/6 mice were purchased from Shanghai Slack Laboratory Animal Co., Ltd. The animals were raised in a clean environment at the Experimental Animal Science Department of the School of Medicine, Shanghai Jiaotong University.
细胞制备方法Cell preparation method
(1)间充质干细胞的分离与培养:(1) Isolation and culture of mesenchymal stem cells:
将实验用小鼠利用颈椎脱臼的方式处死,然后浸泡于75%酒精中3-5分钟。用手术剪刀剪下小鼠两条后腿,取其胫骨、股骨各两根并浸泡于DMEM完全培养基(含有两倍的青霉素和链霉素)中,用手术剪刀剔除骨头周围的肌肉组织。将已剔除干净的骨头两端剪去,留下空腔,即见骨髓;用10ml的注射器抽取5-10ml DMEM培养基(不含FBS)对准骨头空腔,冲洗骨髓至50ml离心管中。离心350g,5分钟,弃上清,沉淀用DMEM完全培养液5ml重悬,显微镜下计数,按照5×106/cm2的接种密度,转移至T25或T75的细胞培养瓶中,放入培养箱(37℃,5%CO2)培养。24小时后换液去除未贴壁的细胞,每隔两天换液一次,通过不断的换液传代对细胞进行纯化,最后用96孔板稀释法挑选单细胞克隆并进行扩增。对所挑选的克隆进行体外分化能力(成骨和成脂)和表面标志的鉴定,所有实验中用的间充质干细胞都在20代以前使用。所有间充质干细胞的培养均使用DMEM完全培养基。The experimental mice were sacrificed by cervical dislocation and then immersed in 75% alcohol for 3-5 minutes. The two hind legs of the mice were cut with surgical scissors, and two of the tibia and femur were taken and immersed in DMEM complete medium (containing twice as much penicillin and streptomycin), and the muscle tissue around the bone was removed with surgical scissors. Cut the ends of the removed bones to leave the cavity, see the bone marrow; use a 10 ml syringe to draw 5-10 ml of DMEM medium (without FBS) to the bone cavity and rinse the bone marrow into a 50 ml centrifuge tube. After centrifugation at 350 g for 5 minutes, the supernatant was discarded, and the pellet was resuspended in 5 ml of DMEM complete medium, counted under a microscope, transferred to a T25 or T75 cell culture flask at a seeding density of 5 × 10 6 /cm 2 , and placed in a culture. Incubate in a box (37 ° C, 5% CO 2 ). After 24 hours, the cells were removed by liquid exchange, and the cells were changed every two days. The cells were purified by continuous liquid exchange. Finally, single cell clones were selected by 96-well plate dilution method and amplified. The selected clones were characterized for in vitro differentiation (osteogenesis and adipogenesis) and surface markers, and the mesenchymal stem cells used in all experiments were used before 20 generations. All mesenchymal stem cells were cultured using DMEM complete medium.
(2)T cell blasts制备(2) Preparation of T cell blasts
小鼠单个核细胞悬液,计数后按照1×106细胞/ml的密度培养在10cm培养皿中培养,1640完全培养液中加入抗小鼠CD3抗体(1μg/ml)和抗小鼠CD28抗体(1μg/ml)。48小时后,将细胞收集后离心500g,5分钟,收集上清即为Sup-CD3(经过抗CD3和抗CD28抗体刺激48小时的小鼠脾细胞培养上清),同时收集细胞沉淀。细胞沉淀用1640完全培养基重悬,计数后按照1×106cells/ml的密度培养在10cm培养皿中(不含抗CD3和抗CD28抗体),培养液中加入IL-2(200U/ml),继续培养48小时后,细胞成团状生长,即为T cell blasts。Mouse mononuclear cell suspension was counted and cultured in a 10 cm culture dish at a density of 1×10 6 cells/ml. Anti-mouse CD3 antibody (1 μg/ml) and anti-mouse CD28 antibody were added to the 1640 complete medium. (1 μg/ml). After 48 hours, the cells were collected and centrifuged for 500 g for 5 minutes, and the supernatant was collected as Sup-CD3 (small mouse spleen cell culture supernatant stimulated with anti-CD3 and anti-CD28 antibody for 48 hours) while collecting the cell pellet. The cell pellet was resuspended in 1640 complete medium, counted and cultured in a 10 cm culture dish at a density of 1 × 10 6 cells/ml (without anti-CD3 and anti-CD28 antibodies), and IL-2 (200 U/ml was added to the culture solution). After 48 hours of incubation, the cells grow into a mass, which is T cell blasts.
T细胞增殖测定(3H-Tdr掺入法)T cell proliferation assay (3H-Tdr incorporation method)
向用于检测T细胞增殖的96孔板中加入3H-Tdr(0.5μCi每孔),继续在培养箱中培养,6小时后将96孔板放入-80℃冰箱。直到检测当天,将板子取出放入37℃烘箱化开,将细胞摄取的同位素全部转移到玻璃纤维素膜。利用微波炉将膜烘干后加入闪烁液,Wallac MicroBeta液闪计数仪上读取cpm值,检测3H的掺入量。3H-Tdr (0.5 μCi per well) was added to a 96-well plate for detecting T cell proliferation, and cultivation was continued in an incubator, and after 6 hours, the 96-well plate was placed in a -80 °C refrigerator. Until the day of the test, the plate was taken out and placed in an oven at 37 ° C to transfer all the isotope taken up by the cells to the glass cellulose membrane. The membrane was dried using a microwave oven and then added to the scintillation fluid. The cpm value was read on a Wallac MicroBeta liquid scintillation counter to measure the amount of 3H incorporated.
细胞凋亡检测Apoptosis detection
将细胞收集后,PBS洗一遍,每个样品加入Propidum Iodide staining buffer(PBS containing 0.2%saponin,50μg/ml propidium iodide,新鲜加入的RNase A 10μg/ml)400 μl重悬,室温染色30min,流式细胞仪检测DNA含量。.After collecting the cells, wash them with PBS, and add Propidum Iodide staining buffer (PBS containing 0.2% saponin, 50 μg/ml propidium iodide, freshly added RNase A 10 μg/ml) 400 to each sample. The μl was resuspended, stained at room temperature for 30 min, and the DNA content was detected by flow cytometry. .
RNA抽提和基因表达分析RNA extraction and gene expression analysis
参照TIANGEN细胞RNA抽提试剂盒说明书抽提RNA。参考TIANGEN TIANScript cDNA第一链生成试剂盒说明将RNA反转为cDNA。采用TAKARA荧光定量PCR试剂,参考其说明书进行荧光定量PCR。以β-actin作为内参,结果分析处理采用2-△△CT法,所有基因表达都换算为β-actin的倍数。所用引物序列如下:RNA was extracted by following the instructions of the TIANGEN Cell RNA Extraction Kit. Refer to the TIANGEN TIANScript cDNA First Strand Generation Kit for inversion of RNA to cDNA. Fluorescence quantitative PCR was performed using TAKARA fluorescent quantitative PCR reagent with reference to its instructions. Using β-actin as an internal reference, the results were analyzed by 2-△△CT method, and all gene expression was converted to a multiple of β-actin. The primer sequences used are as follows:
AUF1:AUF1:
正向引物5’-CAGCAGAGTGGTTATGGGAAAGTATCC-3’(SEQ ID NO.:3);Forward primer 5'-CAGCAGAGTGGTTATGGGAAAGTATCC-3' (SEQ ID NO.: 3);
反向引物5’-GACAGGAGCCACCTTCAAATGAATC-3’(SEQ ID NO.:4);Reverse primer 5'-GACAGGAGCCACCTTCAAATGAATC-3' (SEQ ID NO.: 4);
β-actin:--actin:
正向引物5′-CCACGAGCGGTTCCGATG-3′(SEQ ID NO.:5);Forward primer 5'-CCACGAGCGGTTCCGATG-3' (SEQ ID NO.: 5);
反向引物5′-GCCACAGGATTCCATACCCA-3′(SEQ ID NO.:6);Reverse primer 5'-GCCACAGGATTCCATACCCA-3' (SEQ ID NO.: 6);
CCL2:CCL2:
正向引物5’-TCTCTCTTCCTCCACCACCATG-3’(SEQ ID NO.:7);Forward primer 5'-TCTCTCTTCCTCCACCACCATG-3' (SEQ ID NO.: 7);
反向引物5’-GCGTTAACTGCATCTGGCTGA-3’(SEQ ID NO.:8);Reverse primer 5'-GCGTTAACTGCATCTGGCTGA-3' (SEQ ID NO.: 8);
CCL5:CCL5:
正向引物5’-TTTCTACACCAGCAGCAAGTGC-3’(SEQ ID NO.:9);Forward primer 5'-TTTCTACACCAGCAGCAAGTGC-3' (SEQ ID NO.: 9);
反向引物5’-CCTTCGTGTGACAAACACGAC-3’(SEQ ID NO.:10);Reverse primer 5'-CCTTCGTGTGACAAACACGAC-3' (SEQ ID NO.: 10);
CXCL9:CXCL9:
正向引物5’-AGTGTGGAGTTCGAGGAACCCT-3’(SEQ ID NO.:11);Forward primer 5'-AGTGTGGAGTTCGAGGAACCCT-3' (SEQ ID NO.: 11);
反向引物5’-TGCAGGAGCATCGTGCATT-3’(SEQ ID NO.:12);Reverse primer 5'-TGCAGGAGCATCGTGCATT-3' (SEQ ID NO.: 12);
CXCL10:CXCL10:
正向引物5’-TAGCTCAGGCTCGTCAGTTCT-3’(SEQ ID NO.:13);Forward primer 5'-TAGCTCAGGCTCGTCAGTTCT-3' (SEQ ID NO.: 13);
反向引物5’-GATGGTGGTTAAGTTCGTGCT-3’(SEQ ID NO.:14);Reverse primer 5'-GATGGTGGTTAAGTTCGTGCT-3' (SEQ ID NO.: 14);
IL-6:IL-6:
正向引物5’-GAGGATACCACTCCCAACAGACC-3’(SEQ ID NO.:15);Forward primer 5'-GAGGATACCACTCCCAACAGACC-3' (SEQ ID NO.: 15);
反向引物5’-AAGTGCATCATCGTTGTTCATACA-3’(SEQ ID NO.:16);Reverse primer 5'-AAGTGCATCATCGTTGTTCATACA-3' (SEQ ID NO.: 16);
IL-17RA:IL-17RA:
正向引物5’-AGTGTTTCCTCTACCCAGCAC-3’(SEQ ID NO.:17);Forward primer 5'-AGTGTTTCCTCTACCCAGCAC-3' (SEQ ID NO.: 17);
反向引物5’-GAAAACCGCCACCGCTTAC-3’(SEQ ID NO.:18);Reverse primer 5'-GAAAACCGCCACCGCTTAC-3' (SEQ ID NO.: 18);
IL-17RC:IL-17RC:
正向引物5’-GCTGCCTGATGGTGACAATGT-3’(SEQ ID NO.:19);Forward primer 5'-GCTGCCTGATGGTGACAATGT-3' (SEQ ID NO.: 19);
反向引物5’-TGGACGCAGGTACAGTAAGAAG-3’(SEQ ID NO.:20);Reverse primer 5'-TGGACGCAGGTACAGTAAGAAG-3' (SEQ ID NO.: 20);
iNOS:iNOS:
正向引物5’-CAGCTGGGCTGTACAAACCTT-3’(SEQ ID NO.:21);Forward primer 5'-CAGCTGGGCTGTACAAACCTT-3' (SEQ ID NO.: 21);
反向引物5’-CATTGGAAGTGAAGCGTTTCG-3’(SEQ ID NO.:22).Reverse primer 5'-CATTGGAAGTGAAGCGTTTCG-3' (SEQ ID NO.: 22).
mRNA半衰期测定mRNA half-life determination
第一天,间充质干细胞传代至5块6孔板:1.5×105cells/孔,每孔加入1.6ml完全培养液,放培养箱过夜使细胞贴壁。On the first day, mesenchymal stem cells were passaged to 5 6-well plates: 1.5 × 10 5 cells/well, 1.6 ml of complete medium was added to each well, and the cells were placed overnight to allow the cells to adhere.
第二天,中午12点向6孔板加入IFNγ+TNFα,IFNγ+TNFα+IL-17细胞因子刺激,终浓度10ng/ml。6hr后,晚上18点向6孔板加入Act.D(5μg/ml),收0、0.5hr、1hr、2hr、3hr和4hr共5个时间点的RNA,每种处理对应的每个时间点各有三个复孔,加入裂解液混匀后放-80℃。The next day, at 12 noon, IFNγ+TNFα was added to the 6-well plate, and IFNγ+TNFα+IL-17 cytokine was stimulated to a final concentration of 10 ng/ml. After 6 hr, Act.D (5 μg/ml) was added to the 6-well plate at 18 o'clock in the evening, and RNA was collected at 0, 0.5 hr, 1 hr, 2 hr, 3 hr, and 4 hr for 5 time points, each time point corresponding to each treatment. There are three duplicate wells, and the lysate is added and mixed, and then placed at -80 °C.
第三天,抽提总RNA,测定RNA浓度。On the third day, total RNA was extracted and the RNA concentration was determined.
第四天,将RNA反转录为cDNA,做实时定量PCR。 On the fourth day, RNA was reverse transcribed into cDNA for real-time quantitative PCR.
第五天,结果分析处理采用2-△△CT法,以β-actin或GAPDH作为内参,每个时间点每个重复的RNA量表示为ratio toβ-actin/GAPDH,以时间0点的平均RNA量作为基准,计算出剩余各时间点各重复孔RNA量相对于0点基准值的百分比。EXCEL计算完毕后,用Graphpad Prism 5软件作mRNA decay的半对数曲线,其中Y轴为各百分比,采用对数表示;X轴为各时间点,线性表示。根据软件拟合出的非线性回归曲线的斜率k值,计算mRNA的半衰期,计算公式为t1/2=ln(2/k).On the fifth day, the results were analyzed by 2-△△CT method with β-actin or GAPDH as internal parameters. The amount of RNA per repeat at each time point was expressed as ratio to β-actin/GAPDH, with average RNA at time 0 Using the amount as a reference, the percentage of the amount of repeating-well RNA in each of the remaining time points relative to the zero-point reference value was calculated. After the EXCEL calculation was completed, Graphpad Prism 5 software was used for the semi-logarithmic curve of mRNA decay, wherein the Y-axis is the percentage and expressed in logarithm; the X-axis is the time point and is linearly represented. The half-life of mRNA was calculated according to the slope k value of the nonlinear regression curve fitted by the software, and the formula was calculated as t1/2=ln(2/k).
RNA干扰RNA interference
质粒转染采用电转方法,使用试剂盒Amaxa Cell Line Nucleofector Kit V,参考其说明书,操作如下。Plasmid transfection was performed by electroporation using the kit Amaxa Cell Line Nucleofector Kit V, with reference to its instructions, as follows.
质粒:shCTRL,shAUF1,shAct1,GFP plasmid(试剂盒自带)。Plasmid: shCTRL, shAUF1, shAct1, GFP plasmid (included in the kit).
取出4个6孔板,每个6孔板中的4个孔各加入DMEM完全培养基1ml,放入37℃培养箱预平衡。Four 6-well plates were taken out, and 4 wells of each of the 6-well plates were added to 1 ml of DMEM complete medium, and pre-equilibrated in a 37 ° C incubator.
取出试剂盒中的Nucleofector Solution和Supplement,平衡至室温,按照Nucleofector Solution:Supplement=4.5:1配制Solution,每个反应100μl。The Nucleofector Solution and Supplement in the kit were taken out, equilibrated to room temperature, and the Solution was prepared according to Nucleofector Solution: Supplement = 4.5:1, 100 μl per reaction.
消化2个10cm dish小鼠间充质干细胞,收集后用PBS洗一遍,然后计数。Two 10 cm dish mouse mesenchymal stem cells were digested, collected, washed once with PBS, and then counted.
各吸取1.5×106个细胞至4个干净无菌的1.5ml离心管中,90g室温离心10min,尽量去除上清。Pipette 1.5×10 6 cells into 4 clean sterile 1.5 ml centrifuge tubes, centrifuge at 90 g for 10 min at room temperature, and remove the supernatant as much as possible.
用100μl的Solution重悬细胞,并向每个离心管中加入相应的质粒2μg,轻轻吹匀。The cells were resuspended in 100 μl of Solution, and 2 μg of the corresponding plasmid was added to each centrifuge tube, which was gently blown.
将上述溶液转移到电转杯中,不要有气泡,盖上盖子。Transfer the above solution to the electric rotor without air bubbles and cover.
打开电转仪,选择U-023程序,将电转杯放入电转仪的holder,按下X按钮启动电转程序。Turn on the electro-rotator, select the U-023 program, place the electric rotor into the holder of the electro-rotator, and press the X button to start the electro-rotation procedure.
程序运行完毕后,立即取出电转杯,迅速向杯中加入500μl的预平衡的DMEM完全培养液。用吸管吹匀后,平均转移到6孔板的4个孔中进行培养。Immediately after the program was run, remove the electric rotor and quickly add 500 μl of pre-equilibrated DMEM complete medium to the cup. After homogenizing with a pipette, the cells were transferred to 4 wells of a 6-well plate on average for culture.
第二天,将孔板取出,荧光显微镜观察转染了GFP质粒的细胞,可以看出80%以上的细胞发出绿色荧光,即转染效率有80%以上。对所有的6孔板进行换液。On the next day, the well plate was taken out, and the cells transfected with the GFP plasmid were observed by fluorescence microscopy. It can be seen that more than 80% of the cells emitted green fluorescence, that is, the transfection efficiency was over 80%. All 6-well plates were exchanged.
(10)第三天,开始用250μg/ml的hygromycin B筛选阳性细胞。(10) On the third day, positive cells were screened with 250 μg/ml of hygromycin B.
(11)筛选2-3周后,得到均一的阳性细胞,取出少部分细胞裂解蛋白,做Western Blot鉴定目的蛋白knockdown的效果。若效果好,则稳转细胞株建立成功,可进行下一步实验。(11) After 2-3 weeks of screening, uniform positive cells were obtained, and a small amount of cell lysed protein was taken out, and Western Blot was used to identify the effect of knockdown of the target protein. If the effect is good, the stable cell line is established successfully, and the next experiment can be carried out.
依此方法建立了AUF1 knockdown和Act1knockdown两株细胞。Two cells, AUF1 knockdown and Act1knockdown, were established according to this method.
Western blotWestern blot
可用细胞刮或者胰酶消化法来收集细胞,PBS洗一遍后,将收集好的细胞,加入一定量的RIPA裂解液,吹打混匀后,冰上放置30分钟,期间震荡3-4次;然后高速13,000rpm,低温离心30min;将上清液转移到一新的离心管中,放冰上待用。采用Bio-Rad公司的Protein Assay Dye Reagent Concentrate(Bradford法)测定蛋白浓度。选用8%分离胶、5%积层胶来制作SDS-聚丙烯酰胺凝胶。将样本按体积比加入5×上样缓冲液,100℃煮10分钟后,室温冷却,上样。各样本约含50-100μg蛋白;每块胶含一道蛋白marker;插好电极,开始电泳,先80V电泳30min,再120V电泳60min。将凝胶取下后,切除积层胶,放到电转移缓冲液中;将NC膜(硝酸纤维素膜)放到转移缓冲液中平衡10分钟左右;从下到上将海绵、滤纸、膜、凝胶、滤纸、海绵依次放好,每放一层都要注意排出气泡,将转移装置连接好,然后接通电源,恒压100V,2小时;转移结束后关闭电源,取出膜,根据蛋白marker和目的蛋白分子量大小剪出相应位置的条带,做好标记。将膜放在塑料盒中,加入适量封闭液,于摇床上室温放置2小时;用1×TBST洗液洗3次,每次3分钟;然后将膜转入到塑料袋中,加入稀释好的第一抗体,4℃摇床过夜。第二天取出膜然后用1×TBST洗3次,每次10分钟, 再加入稀释好的HRP标记二抗,室温摇床孵育1-2小时;取出膜用1×TBST洗3次,每次10分钟;将膜转移到片夹中,在暗室里加ECL显色液避光反应,适时终止反应,进行胶片的曝光、冲片。根据胶片相应位置上条带的粗细和浓淡来判断目标蛋白的含量。扫描胶片并保存。The cells can be collected by cell scraping or trypsinization. After washing with PBS, the collected cells are added with a certain amount of RIPA lysate, mixed by blowing, and placed on ice for 30 minutes, shaking for 3-4 times; Transfer at high speed 13,000 rpm for 30 min at low temperature; transfer the supernatant to a new centrifuge tube and place on ice for later use. Protein concentration was determined using Bio-Rad's Protein Assay Dye Reagent Concentrate (Bradford method). SDS-polyacrylamide gel was prepared by using 8% separating glue and 5% laminated glue. The sample was added to 5× loading buffer by volume ratio, and boiled at 100 ° C for 10 minutes, then cooled at room temperature and loaded. Each sample contains about 50-100μg of protein; each gel contains a protein marker; the electrode is inserted and electrophoresis is started, first 80V electrophoresis for 30min, then 120V electrophoresis for 60min. After the gel is removed, the laminated gel is removed and placed in an electrotransfer buffer; the NC membrane (nitrocellulose membrane) is placed in a transfer buffer for about 10 minutes; from bottom to top, the sponge, filter paper, and membrane are placed. , gel, filter paper, sponge are placed in turn, each layer should pay attention to discharge air bubbles, connect the transfer device, and then turn on the power, constant voltage 100V, 2 hours; after the transfer is finished, turn off the power, remove the membrane, according to the protein Marker and molecular weight of the target protein cut out the strip at the corresponding position and mark it. Place the film in a plastic box, add an appropriate amount of blocking solution, and leave it on the shaker for 2 hours at room temperature; wash 3 times with 1×TBST solution for 3 minutes each time; then transfer the film into a plastic bag and add it to the diluted solution. The first antibody was shaken overnight at 4 °C. The film was taken out the next day and then washed 3 times with 1 x TBST for 10 minutes each time. Add the diluted HRP-labeled secondary antibody and incubate for 1-2 hours at room temperature. Remove the membrane and wash it with 1×TBST for 3 times for 10 minutes each time. Transfer the membrane to the holder and add ECL coloring solution in the dark room. The photoreaction is terminated in a timely manner, and the film is exposed and punched. The content of the target protein is judged based on the thickness and shade of the strip at the corresponding position of the film. Scan the film and save it.
免疫共沉淀Immunoprecipitation
用胰酶消化法来收集细胞,PBS洗一遍后,将收集好的细胞,加入一定量的RIPA裂解液,吹打混匀后,冰上放置30分钟,期间震荡3-4次;然后高速13,000rpm,低温离心30min;将上清液转移到一新的离心管中,放冰上待用。采用Bio-Rad公司的Protein Assay Dye Reagent Concentrate(Bradford法)测定蛋白浓度。将上述蛋白裂解上清与protein G sepharose beads进行孵育以去除非特异性结合,然后高速离心后取上清,向上清中加入1μg Act1抗体或1μg非特异性IgG对照,然后将之与20μl protein G sepharose beads(GE Healthcare)进行孵育,4℃过夜。将beads用RIPA buffer洗4次,然后用20μl 2×SDS loading buffer重悬,在100℃煮10min,然后用Western Blot检测蛋白表达。The cells were collected by trypsinization. After washing with PBS, the collected cells were added with a certain amount of RIPA lysate, mixed by blowing, and placed on ice for 30 minutes, shaking for 3-4 times; then high speed 13,000 rpm Centrifuge at low temperature for 30 min; transfer the supernatant to a new centrifuge tube and let it stand on ice. Protein concentration was determined using Bio-Rad's Protein Assay Dye Reagent Concentrate (Bradford method). The above protein cleavage supernatant was incubated with protein G sepharose beads to remove non-specific binding, and then the supernatant was taken after high-speed centrifugation. 1 μg of Act1 antibody or 1 μg of non-specific IgG control was added to the supernatant, and then 20 μl of protein G sepharose beads was added. Incubate (GE Healthcare) overnight at 4 °C. The beads were washed 4 times with RIPA buffer, then resuspended in 20 μl of 2×SDS loading buffer, boiled at 100 ° C for 10 min, and then protein expression was detected by Western Blot.
CIH模型的建立,细胞治疗和评价Establishment of CIH model, cell therapy and evaluation
(1)间充质干细胞提前用细胞因子预处理12-16小时:WT MSC,iNOS-/-MSC或auf1-/-MSC给予IFNγ+TNFα/IFNγ+TNFα+IL-17(10ng/ml)细胞因子刺激。(1) Mesenchymal stem cells were pretreated with cytokines for 12-16 hours in advance: WT MSC, iNOS-/-MSC or auf1-/-MSC administered IFNγ+TNFα/IFNγ+TNFα+IL-17 (10 ng/ml) cells Factor stimulation.
(2)根据体重,给予小鼠静脉注射15mg/kg的ConA。(2) According to the body weight, mice were intravenously administered with 15 mg/kg of ConA.
(3)给予ConA注射30min后,开始间充质干细胞静脉注射治疗,每只小鼠给予5×105个细胞治疗。每个处理组有5只小鼠。(3) After 30 minutes of ConA injection, intravenous injection of mesenchymal stem cells was started, and each mouse was treated with 5 × 10 5 cells. There were 5 mice per treatment group.
(4)ConA注射8小时后处死小鼠,收集血清,检测血清中ALT的水平,留取一叶肝脏浸泡在10%福尔马林中作病理切片H&E染色;剩余肝脏全部用来分离单个核细胞。(4) Eight hours after ConA injection, the mice were sacrificed, serum was collected, serum ALT levels were measured, and one leaf liver was immersed in 10% formalin for pathological section H&E staining; the remaining livers were all used to isolate mononuclear cells.
(5)小鼠血清中ALT的检测:参考上海伊华公司生产的ALT检测试剂盒说明书进行操作。(5) Detection of ALT in mouse serum: Refer to the instructions of the ALT test kit produced by Shanghai Yihua Company for operation.
MSC在损伤肝脏组织中的定位Localization of MSC in injured liver tissue
将GFP-MSC提前用细胞因子IFNγ+TNFα/IFNγ+TNFα+IL-17(10ng/ml)刺激进行预处理12-16小时;ConA注射30min后,开始各组GFP-MSC静脉注射治疗,每只小鼠给予5×105个细胞治疗。ConA注射8小时后处死小鼠。GFP-MSC was pretreated with cytokine IFNγ+TNFα/IFNγ+TNFα+IL-17 (10ng/ml) for 12-16 hours in advance; 30 minutes after ConA injection, each group received GFP-MSC intravenous injection, each treatment. Mice were treated with 5 x 10 5 cells. Mice were sacrificed 8 hours after ConA injection.
(1)小鼠处死后,立即取一小叶肝脏直接放入OCT包埋,并冻在-80℃。(1) Immediately after the mice were sacrificed, a small leaf liver was directly placed in the OCT and frozen at -80 °C.
(2)冷冻切片机切片,片厚10μm。(2) The slicer was sliced to a thickness of 10 μm.
(3)切片用-20℃预冷的丙酮中固定10min,晾干后浸入PBS中20min。(3) The sections were fixed in acetone pre-cooled at -20 ° C for 10 min, dried and immersed in PBS for 20 min.
(4)PBS洗两遍,加入PBS+3%BSA室温处理1小时。(4) Wash twice with PBS and add PBS + 3% BSA for 1 hour at room temperature.
(5)吸弃多余液体,加入一抗(anti-GFP按1:400稀释),37℃湿盒2小时。(5) Aspirate excess liquid, add primary antibody (anti-GFP diluted 1:400), and wet the box at 37 °C for 2 hours.
(6)PBS洗三次,每次5min,加入二抗(Alexa Fluor 594Donkey anti-Rabbit antibody 1:500)+DAPI(1:500),37℃湿盒2小时。(6) Wash PBS three times for 5 min each time, add secondary antibody (Alexa Fluor 594 Donkey anti-Rabbit antibody 1:500) + DAPI (1:500), and wet the box at 37 °C for 2 hours.
(7)PBS洗三次,每次5min,晾干后封片,荧光显微镜观察并拍照。(7) Wash the PBS three times, each time for 5 min, dry it, seal it, observe it with a fluorescence microscope and take a picture.
细胞表面分子的免疫荧光染色Immunofluorescence staining of cell surface molecules
取细胞1×106重悬于100μl FACS缓冲液中用于染色。加入荧光标记的抗体后(如PE标记抗小鼠IL-17RA抗体,PE标记抗小鼠CD3抗体,PerCP/Cy5.5标记抗小鼠CD4抗体,APC标记抗小鼠CD8抗体,APC标记的抗小鼠CD45抗体,APC标记的抗小鼠CD25抗体,FITC标记的抗小鼠CD3抗体),4℃避光孵育30分钟。用FACS缓冲液洗涤两次。最后将细胞重悬于400μl的FACS buffer,FACS Calibur流式细胞仪检测分析。 The cells were resuspended in 100 μl of FACS buffer for staining at 1 × 10 6 . After addition of fluorescently labeled antibodies (eg PE-labeled anti-mouse IL-17RA antibody, PE-labeled anti-mouse CD3 antibody, PerCP/Cy5.5-labeled anti-mouse CD4 antibody, APC-labeled anti-mouse CD8 antibody, APC-labeled antibody Mouse CD45 antibody, APC-labeled anti-mouse CD25 antibody, FITC-labeled anti-mouse CD3 antibody), incubated for 30 minutes at 4 ° C in the dark. Wash twice with FACS buffer. Finally, the cells were resuspended in 400 μl of FACS buffer and analyzed by FACS Calibur flow cytometry.
细胞内细胞因子或Foxp3的免疫荧光染色Immunofluorescence staining of intracellular cytokines or Foxp3
(1)所有细胞(约5-10×105)用PBS洗一遍后,都用抗小鼠CD16/CD32抗体封闭10min,冰上,每管30μl体系。(1) All cells (about 5-10×10 5 ) were washed once with PBS, and blocked with anti-mouse CD16/CD32 antibody for 10 min, on ice, 30 μl of system per tube.
(2)先进行细胞表面分子的染色,方法参照11.1.(2) First stain the cell surface molecules, refer to 11.1.
(3)FACS缓冲液洗一遍后,每管各加入Fixation/Permeabilization buffer 100μl,放4℃固定过夜。(3) After washing the FACS buffer once, 100 μl of Fixation/Permeabilization buffer was added to each tube, and fixed at 4 ° C overnight.
(4)400×g,离心5min,加入1×Permeabilization buffer 200μl,混匀。(4) 400 × g, centrifuge for 5 min, add 1 × Permeabilization buffer 200 μl, mix.
(5)400×g,离心5min,加入相应的细胞因子抗体或者Foxp3抗体(抗体用1×Permeabilization buffer稀释),每管50μl体系,放冰上避光染色1小时。(5) 400 × g, centrifuge for 5 min, add the corresponding cytokine antibody or Foxp3 antibody (antibody diluted with 1 × Permeabilization buffer), 50 μl of each tube, and stain on ice for 1 hour in the dark.
(6)直接加入1×Permeabilization buffer 200μl,混匀,400×g,离心5min。(6) Directly add 200 μl of 1×Permeabilization buffer, mix well, 400×g, and centrifuge for 5 min.
(7)弃上清后,再加入1×Permeabilization buffer 200μl,混匀,400×g离心5min。(7) After discarding the supernatant, add 2 μM of 1×Permeabilization buffer, mix, and centrifuge at 400×g for 5 min.
(8)弃上清后,再加入FACS buffer 200μl,混匀,400×g,离心5min。细胞沉淀用FACS buffer 200μl重悬,即可用流式细胞仪检测分析。(8) After discarding the supernatant, add 200 μl of FACS buffer, mix well, 400×g, and centrifuge for 5 min. The cell pellet was resuspended in FACS buffer 200 μl and assayed by flow cytometry.
数据处理和统计学分析Data processing and statistical analysis
所示实验结果中每个处理组都设置有三个或三个以上的重复,通过Graphpad Prism 5软件作图,图中所示数据以平均数±标准差表示。数据的统计分析采用Student’s t检验或单因素方差分析。显著性水准定为α=0.05,*代表p<0.05;**代表p<0.01;***代表p<0.001。Three or more replicates were set for each treatment group in the experimental results shown, plotted by Graphpad Prism 5 software, and the data shown is expressed as mean ± standard deviation. Statistical analysis of the data was performed using Student's t test or one-way ANOVA. The significance level was determined as α = 0.05, * represents p < 0.05; ** represents p < 0.01; *** represents p < 0.001.
实施例1 IL-17能够增强MSC的免疫抑制功能Example 1 IL-17 can enhance the immunosuppressive function of MSC
已有研究表明,间充质干细胞的免疫抑制能力不是固有的,而是在炎症细胞因子的诱导下获得的,这些炎症细胞因子包括IFNγ、TNFα、IL-1α和IL-1β。间充质干细胞在炎症细胞因子IFNγ和TNFα的刺激下,分泌大量的iNOS和趋化因子,趋化因子招募T细胞至间充质干细胞的周围,由iNOS的代谢产物NO发挥抑制T细胞增殖的作用。众所周知,IL-17是一种多效的促炎症细胞因子,它在很多感染性疾病、炎症性疾病和自身免疫病的发病过程中发挥了重要作用。本发明人向体外培养的间充质干细胞中加入炎症细胞因子IFNγ和TNFα,或在此基础上再加入IL-17,观察这些细胞因子对间充质干细胞免疫抑制能力的影响。本发明人发现,只有在IFNγ和TNFα存在的情况下,间充质干细胞才获得对T细胞增殖的抑制能力;而IL-17则能够显著增强IFNγ和TNFα所诱导的间充质干细胞的免疫抑制能力,这一作用表现为T细胞增殖明显降低(图1A)。Studies have shown that the immunosuppressive ability of mesenchymal stem cells is not intrinsic, but is obtained under the induction of inflammatory cytokines including IFNγ, TNFα, IL-1α and IL-1β. Mesenchymal stem cells secrete a large amount of iNOS and chemokines under the stimulation of inflammatory cytokines IFNγ and TNFα. Chemokines recruit T cells to the periphery of mesenchymal stem cells, and the NO, a metabolite of iNOS, inhibits T cell proliferation. effect. It is well known that IL-17 is a pleiotropic pro-inflammatory cytokine that plays an important role in the pathogenesis of many infectious diseases, inflammatory diseases and autoimmune diseases. The present inventors added inflammatory cytokines IFNγ and TNFα to mesenchymal stem cells cultured in vitro, or added IL-17 thereto, and observed the effects of these cytokines on the immunosuppressive ability of mesenchymal stem cells. The present inventors have found that mesenchymal stem cells can only inhibit the proliferation of T cells in the presence of IFNγ and TNFα, while IL-17 can significantly enhance the immunosuppression of mesenchymal stem cells induced by IFNγ and TNFα. Capacity, this effect is manifested by a significant decrease in T cell proliferation (Fig. 1A).
以往的研究已经证明,IL-17是通过与细胞表面的IL-17RA和IL-17RC组成的异二聚体结合进而激活下游信号通路的。既然IL-17可以作用于间充质干细胞并显著增强其免疫抑制能力,本发明人检测了间充质干细胞的IL-17RA和IL-17RC的表达情况。结果显示,间充质干细胞可以组成性地表达IL-17RA和IL-17RC(图1B,C)。Previous studies have demonstrated that IL-17 activates downstream signaling pathways by binding to heterodimers composed of IL-17RA and IL-17RC on the cell surface. Since IL-17 can act on mesenchymal stem cells and significantly enhance its immunosuppressive ability, the present inventors examined the expression of IL-17RA and IL-17RC in mesenchymal stem cells. The results showed that mesenchymal stem cells can constitutively express IL-17RA and IL-17RC (Fig. 1B, C).
在炎症反应的不同阶段,炎症细胞因子的水平是不同的,因此本发明人研究了不同剂量的IFNγ和TNFα对IL-17的这一增强免疫抑制的功能是否有影响。本发明人发现,当IFNγ和TNFα的水平仅有1-2ng/ml时,IL-17即可以明显增强间充质干细胞的免疫抑制功能(图1D,4E)。即使当IFNγ和TNFα的水平很高时(10-20ng/ml),IL-17仍可以改善间充质干细胞的免疫抑制功能。本发明人也观察了不同剂量的IL-17对其增强间充质干细胞的免疫抑制能力这一特性的影响。结果显示,大约0.5ng/ml的IL-17即可明显地增强间充质干细胞的免疫抑制功能(图1F)。At different stages of the inflammatory response, the levels of inflammatory cytokines are different, so the inventors investigated whether different doses of IFNy and TNF[alpha] have an effect on the enhanced immunosuppressive function of IL-17. The present inventors have found that IL-17 can significantly enhance the immunosuppressive function of mesenchymal stem cells when the levels of IFNγ and TNFα are only 1-2 ng/ml (Fig. 1D, 4E). Even when the levels of IFNγ and TNFα are high (10-20 ng/ml), IL-17 can improve the immunosuppressive function of mesenchymal stem cells. The present inventors also observed the effect of different doses of IL-17 on the property of enhancing the immunosuppressive ability of mesenchymal stem cells. The results showed that approximately 0.5 ng/ml of IL-17 significantly enhanced the immunosuppressive function of mesenchymal stem cells (Fig. 1F).
为了进一步明确在T细胞介导的免疫反应中IL-17是否参与MSC对T细胞增殖的抑制作用,本发明人将MSC与anti-CD3激活的脾细胞共培养,并向共培养体系中加入anti-IL-17A抗体。结果显示,MSC可以明显地抑制激活的脾细胞的增殖,而anti-IL-17A 抗体可以在很大程度上恢复激活的脾细胞的增殖,逆转MSC的抑制作用(图1G)。当MSC和脾细胞的比例为1:40时,anti-IL-17A的这一逆转作用最为显著;当MSC和脾细胞的比例为1:20时,虽然anti-IL-17A的逆转作用减弱了,但仍有统计学意义。总之,本发明人的研究表明,在T细胞介导的免疫反应中,特别是免疫微环境中存在的炎症细胞因子IFNγ和TNFα水平较低时,IL-17可以显著增强MSC对T细胞的免疫抑制功能。To further clarify whether IL-17 is involved in the inhibition of T cell proliferation by MSCs in a T cell-mediated immune response, the inventors co-cultured MSCs with anti-CD3 activated splenocytes and added anti to the co-culture system. -IL-17A antibody. The results showed that MSC can significantly inhibit the proliferation of activated spleen cells, while anti-IL-17A The antibody can largely restore the proliferation of activated splenocytes, reversing the inhibition of MSC (Fig. 1G). When the ratio of MSC to spleen cells is 1:40, the reversal effect of anti-IL-17A is most significant; when the ratio of MSC to spleen cells is 1:20, although the reversal effect of anti-IL-17A is weakened , but still statistically significant. In conclusion, the present inventors have shown that IL-17 can significantly enhance MSC immunity against T cells in T cell-mediated immune responses, particularly when the levels of inflammatory cytokines IFNγ and TNFα present in the immune microenvironment are low. Suppress function.
为了扩展本发明人的研究,本发明人在脂肪来源的MSC上也检测了IL-17对其免疫抑制功能的影响作用,得到了与骨髓来源的MSC类似的结果(图1H),即IL-17同样可以增强脂肪来源MSC的免疫抑制功能,这提示着IL-17的这一功能可能不依赖于MSC的来源。In order to expand the research of the present inventors, the present inventors also examined the effect of IL-17 on its immunosuppressive function on adipose-derived MSCs, and obtained similar results to bone marrow-derived MSCs (Fig. 1H), namely IL- 17 also enhances the immunosuppressive function of adipose-derived MSCs, suggesting that this function of IL-17 may not depend on the source of MSCs.
图1显示了IL-17增强MSC的免疫抑制功能。Figure 1 shows that IL-17 enhances the immunosuppressive function of MSCs.
(A).首先将MSC用细胞因子IFNγ、TNFα和IL-17(每种因子的浓度均为2ng/ml)的不同组合刺激12小时,然后将之与T cell blasts共培养(MSC与T cell blasts的比例为1:20),12小时后,用3H-Tdr掺入法测定T细胞增殖。(B).运用实时荧光定量PCR的方法检测IL-17RA和IL-17RC在MSC和Raw264.7细胞(阳性对照)上的表达。(C).运用流式细胞术检测MSC表面IL-17RA的表达。(D和E).首先将MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17刺激12小时(其中IFNγ和TNFα的浓度作梯度稀释,IL-17浓度固定为10ng/ml),然后将之与T cell blasts(需加入IL-2方可增殖,且增殖中不产生炎症细胞因子)。(D)或A1.1细胞(T细胞杂交瘤细胞,能自主增殖,且增殖中不产生炎症细胞因子,该细胞的制备方法可参考文献(Nature.1989 Jun22;339(6226):625-6.)(E)共培养,MSC和T细胞比例为1:20,12小时后,用3H-Tdr掺入法测定T细胞增殖。(F).首先将MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17刺激12小时(其中IFNγ和TNFα的浓度固定为2ng/ml,IL-17浓度作梯度稀释),然后将之A1.1细胞共培养,MSC和T细胞比例为1:10,12小时后,用3H-Tdr掺入法测定T细胞增殖。(G).将MSC与anti-CD3和anti-CD28激活的脾细胞按1:40或1:20的比例进行共培养,或在培养基中加入anti-IL-17A,48小时后,用3H-Tdr掺入法测定脾细胞增殖。(H).首先将BMMSC或ADSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17(每种因子的浓度均为10ng/ml)刺激12小时,然后将之与A1.1细胞共培养(MSC与A1.1的比例为1:10),12小时后,用3H-Tdr掺入法测定T细胞增殖。结果表示为平均数±标准差,BMMSC:骨髓来源的MSC;ADSC:脂肪来源的MSC。所示数据代表三次实验的结果。(A). MSCs were first stimulated with different combinations of cytokines IFNγ, TNFα and IL-17 (each concentration of 2 ng/ml) for 12 hours, and then co-cultured with T cell blasts (MSC and T cell). The ratio of blasts was 1:20), and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation. (B). The expression of IL-17RA and IL-17RC on MSC and Raw264.7 cells (positive control) was detected by real-time fluorescent quantitative PCR. (C). Flow cytometry was used to detect the expression of IL-17RA on MSC surface. (D and E). The MSCs were first stimulated with the cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 for 12 hours (the concentration of IFNγ and TNFα was diluted in a gradient, the IL-17 concentration was fixed at 10 ng/ml), and then And T cell blasts (requires the addition of IL-2 to proliferate, and does not produce inflammatory cytokines during proliferation). (D) or A1.1 cells (T cell hybridoma cells, can proliferate independently, and do not produce inflammatory cytokines during proliferation, the preparation method of the cells can be referred to the literature (Nature. 1989 Jun22; 339 (6226): 625-6 (E) Co-culture, the ratio of MSC to T cells was 1:20, and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation. (F). First, MSC was treated with cytokine IFNγ + TNFα or IFNγ. +TNFα+IL-17 was stimulated for 12 hours (the concentration of IFNγ and TNFα was fixed at 2 ng/ml, and the concentration of IL-17 was diluted), then A1.1 cells were co-cultured, and the ratio of MSC to T cells was 1:10. After 12 hours, T cell proliferation was measured by 3H-Tdr incorporation. (G). MSCs were co-cultured with anti-CD3 and anti-CD28-activated splenocytes at a ratio of 1:40 or 1:20, or Anti-IL-17A was added to the culture medium, and 48 hours later, spleen cell proliferation was measured by 3 H-Tdr incorporation method. (H). First, BMMSC or ADSC was treated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-. 17 (10 ng/ml for each factor) was stimulated for 12 hours, then co-cultured with A1.1 cells (1:10 ratio of MSC to A1.1), and after 12 hours, 3 H-Tdr T cell proliferation was determined by incorporation assay. Results are expressed as mean Number ± standard deviation, BMMSC: bone marrow-derived MSC; ADSC: fat-derived MSC. The data shown represents the results of three experiments.
实施例2 IL-17增强MSC免疫抑制的特性依赖于iNOS的活性Example 2 IL-17 enhances the immunosuppressive properties of MSCs depending on the activity of iNOS
以上的结果显示,IL-17可以显著增强MSC对T细胞增殖的免疫抑制功能,接下来本发明人进一步研究了其中的相关机制。之前的研究认为,IL-17可以促进人MSC的增殖。因此,本发明人首先研究了IL-17是否通过影响MSC的增殖进而影响其免疫抑制功能。本发明人将MSC用IFNγ+TNFα或IFNγ+TNFα+IL-17刺激24小时,然后检测MSC的增殖情况。本发明人发现,在受到IFNγ和TNFα刺激的同时,IL-17的加入反而能抑制MSC的增殖(图2A)。因此,IL-17并非通过影响MSC的增殖来增强其免疫抑制能力。The above results show that IL-17 can significantly enhance the immunosuppressive function of MSCs against T cell proliferation, and the inventors further studied the related mechanisms. Previous studies have suggested that IL-17 can promote the proliferation of human MSCs. Therefore, the inventors first studied whether IL-17 affects the immunosuppressive function by affecting the proliferation of MSC. The present inventors stimulated MSCs with IFNγ+TNFα or IFNγ+TNFα+IL-17 for 24 hours, and then detected the proliferation of MSCs. The present inventors have found that the addition of IL-17 inhibits the proliferation of MSCs while being stimulated by IFNγ and TNFα (Fig. 2A). Therefore, IL-17 does not enhance its immunosuppressive ability by affecting the proliferation of MSCs.
本发明人之前的研究表明,iNOS是介导小鼠MSC发挥免疫抑制的重要分子。本发明人检测了IL-17的这一增强MSC免疫抑制的作用是否依赖于iNOS的活性。结果显示,IL-17的作用能够被iNOS抑制剂L-NMMA完全逆转(图2B),表明iNOS在其中发挥了至关重要的作用。Previous studies by the present inventors have shown that iNOS is an important molecule mediating the immunosuppression of mouse MSCs. The present inventors examined whether this effect of IL-17 enhancing MSC immunosuppression is dependent on the activity of iNOS. The results showed that the effect of IL-17 was completely reversed by the iNOS inhibitor L-NMMA (Fig. 2B), indicating that iNOS plays a crucial role in it.
图2显示IL-17增强MSC免疫抑制的特性依赖于iNOS的活性。Figure 2 shows that IL-17 enhances the immunosuppressive properties of MSCs depending on the activity of iNOS.
(A).将MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17(IFNγ+TNFα的浓度作梯度稀释,IL-17的浓度固定为10ng/ml)处理24小时,然后用3H-Tdr掺入法测定T细胞增殖。(B).首先将MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17(每种因子的 浓度均为2ng/ml)刺激12小时,然后将之与A1.1细胞共培养(MSC与A1.1的比例为1:10),或在培养基中加入iNOS抑制剂L-NMMA,12小时后,用3H-Tdr掺入法测定T细胞增殖。所有数据代表三次独立实验的结果。(A). MSC was treated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 (concentration of IFNγ+TNFα was diluted, IL-17 concentration was fixed at 10 ng/ml) for 24 hours, then 3H-Tdr was used. T cell proliferation was determined by incorporation. (B). First, MSC was stimulated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 (each concentration was 2ng/ml) for 12 hours, and then co-cultured with A1.1 cells (MSC and The ratio of A1.1 was 1:10), or the iNOS inhibitor L-NMMA was added to the medium, and 12 hours later, T cell proliferation was measured by 3 H-Tdr incorporation. All data represent the results of three independent experiments.
实施例3 IL-17增强MSC的免疫抑制功能不依赖于对T细胞凋亡的影响Example 3 IL-17 enhances the immunosuppressive function of MSC independent of the effect on T cell apoptosis
以上的研究表明,IL-17可以协同炎症细胞因子IFNγ和TNFα增强MSC的免疫抑制功能,而这一功能主要体现在对T细胞增殖的抑制,并且依赖于iNOS的活性(图1,图2)。另外,MSC在炎症细胞因子IFNγ和TNFα存在的情况下可以诱导淋巴细胞的凋亡,这一过程也依赖于NO。为了确定IL-17的这一增强MSC免疫抑制的作用是否依赖于对T细胞凋亡的影响,本发明人将MSC用IFNγ+TNFα或IFNγ+TNFα+IL-17提前刺激12小时,然后将之与A1.1细胞按1:10的比例进行共培养,12小时后收集A1.1细胞,检测细胞凋亡情况。本发明人发现,A1.1细胞在单独培养情况下,凋亡细胞比例低于0.5%;而当其与未处理的MSC共培养后,凋亡细胞的比例明显增加,比例在3-5%;当其与IFNγ+TNFα预处理的MSC共培养时,凋亡细胞的比例进一步增加,比例在7-10%;A1.1与IFNγ+TNFα+IL-17预处理的MSC共培养的情况和与IFNγ+TNFα预处理的MSC共培养时相似,凋亡细胞的比例仍在7-10%(图3)。另外,这一现象不依赖于IFNγ和TNFα的浓度,不管IFNγ和TNFα的浓度在5ng/ml还是10ng/ml,IFNγ+TNFα+IL-17预处理的MSC都不能进一步促进T细胞的凋亡。因此,IL-17增强MSC的免疫抑制不依赖于对T细胞凋亡的影响。The above studies show that IL-17 can synergize with the inflammatory cytokines IFNγ and TNFα to enhance the immunosuppressive function of MSC, and this function is mainly reflected in the inhibition of T cell proliferation and depends on the activity of iNOS (Fig. 1, Fig. 2). . In addition, MSCs can induce lymphocyte apoptosis in the presence of inflammatory cytokines IFNγ and TNFα, a process that is also dependent on NO. In order to determine whether this effect of IL-17 enhancing MSC immunosuppression is dependent on the effect on T cell apoptosis, the inventors pre-stimulated MSC with IFNγ+TNFα or IFNγ+TNFα+IL-17 for 12 hours, and then Co-culture was performed with A1.1 cells at a ratio of 1:10, and A1.1 cells were collected 12 hours later to detect apoptosis. The present inventors have found that the ratio of apoptotic cells in A1.1 cells is less than 0.5% in the case of single culture; and when co-cultured with untreated MSCs, the proportion of apoptotic cells is significantly increased, the ratio is 3-5%. When co-cultured with IFNγ+TNFα pretreated MSC, the proportion of apoptotic cells was further increased, the ratio was 7-10%; A1.1 was co-cultured with IFNγ+TNFα+IL-17 pretreated MSC and Similar to the IFNγ+TNFα pretreated MSC, the proportion of apoptotic cells was still 7-10% (Fig. 3). In addition, this phenomenon does not depend on the concentration of IFNγ and TNFα. Regardless of the concentration of IFNγ and TNFα at 5 ng/ml or 10 ng/ml, IFNγ+TNFα+IL-17 pretreated MSC could not further promote T cell apoptosis. Thus, IL-17 enhances the immunosuppression of MSCs independent of the effects on T cell apoptosis.
图3显示IL-17增强MSC的免疫抑制功能不依赖于对T细胞凋亡的影响。Figure 3 shows that IL-17 enhances the immunosuppressive function of MSCs independent of the effects on T cell apoptosis.
将MSC提前用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17刺激12小时(其中IFNγ和TNFα的浓度为5ng/ml或10ng/ml,IL-17的浓度始终为10ng/ml),然后按1:10的比例与A1.1细胞共培养,12小时后,收集A1.1细胞,PI染色后,流式细胞仪分析其DNA含量。The MSC was stimulated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 for 12 hours (the concentration of IFNγ and TNFα was 5 ng/ml or 10 ng/ml, the concentration of IL-17 was always 10 ng/ml), and then The ratio of 1:10 was co-cultured with A1.1 cells. After 12 hours, A1.1 cells were collected, and after PI staining, the DNA content was analyzed by flow cytometry.
实施例4 IL-17协同炎症细胞因子上调MSC中iNOS的表达Example 4 IL-17 synergistic inflammatory cytokines up-regulate the expression of iNOS in MSC
之前的研究表明,iNOS和趋化因子是介导MSC免疫抑制功能的核心分子,而且MSC启动表达iNOS和一些趋化因子需要炎症细胞因子IFNγ和TNFα的诱导。本发明人上述结果显示,IL-17增强MSC免疫抑制的特性依赖于iNOS的活性。本发明人用IFNγ+TNFα或IFNγ+TNFα+IL-17来处理MSC,然后用荧光定量PCR和Western Blot的方法来测定iNOS和趋化因子的表达。结果显示,无论在mRNA水平还是蛋白水平上,IL-17都能够协同IFNγ和TNFα明显促进MSC表达iNOS(图4A,B,C,D)。同时,本发明人也检测了一些和MSC发挥免疫抑制相关的趋化因子的mRNA表达,包括CCL2、CCL5、CXCL9、CXCL10,结果发现IL-17并不能协同IFNγ和TNFα改变这些趋化因子的表达(图4E)。由此可见,IL-17增强MSC的免疫抑制特异地依赖于其对iNOS的上调作用。为了进一步验证在T细胞介导的免疫应答中IL-17对MSC的一些基因表达的上调作用,本发明人将激活的脾细胞上清与anti-IL-17A孵育一段时间以中和上清中的IL-17,然后用这些上清来刺激MSC,进而检测MSC一些免疫调节基因的表达。本发明人发现,激活的脾细胞上清能明显地诱导MSC表达iNOS、CCL2、CCL5、CXCL9和CXCL10;中和了IL-17的上清则不能很好地诱导MSC表达iNOS(图4F,G),但趋化因子CCL2、CCL5、CXCL9和CXCL10的表达则不受影响(图4G)。上述结果表明,IL-17能够协同IFNγ和TNFα明显促进MSC表达iNOS,但却不影响趋化因子CCL2、CCL5、CXCL9和CXCL10的表达。Previous studies have shown that iNOS and chemokines are the core molecules that mediate MSC immunosuppressive function, and that MSCs that initiate expression of iNOS and some chemokines require induction of the inflammatory cytokines IFNγ and TNFα. The above results of the present inventors show that the property of IL-17 to enhance MSC immunosuppression depends on the activity of iNOS. The present inventors treated MSCs with IFNγ+TNFα or IFNγ+TNFα+IL-17, and then used real-time PCR and Western Blot to determine the expression of iNOS and chemokines. The results showed that IL-17 was able to significantly promote MSC expression of iNOS in combination with IFNγ and TNFα at both mRNA and protein levels (Fig. 4A, B, C, D). At the same time, the present inventors also detected mRNA expression of chemokines involved in immunosuppression of MSC, including CCL2, CCL5, CXCL9, and CXCL10, and found that IL-17 does not cooperate with IFNγ and TNFα to alter the expression of these chemokines. (Fig. 4E). Thus, IL-17 enhances the immunosuppression of MSCs specifically depending on its up-regulation of iNOS. To further verify the up-regulation of IL-17 on some of the gene expression of MSCs in a T cell-mediated immune response, the inventors incubated the activated spleen cell supernatant with anti-IL-17A for a period of time to neutralize the supernatant. IL-17 is then used to stimulate MSCs to detect the expression of some immunoregulatory genes in MSCs. The present inventors have found that activated spleen cell supernatant can significantly induce MSC expression of iNOS, CCL2, CCL5, CXCL9 and CXCL10; neutralizing the supernatant of IL-17 does not induce MSC to express iNOS well (Fig. 4F, G However, expression of the chemokines CCL2, CCL5, CXCL9 and CXCL10 was not affected (Fig. 4G). The above results indicate that IL-17 can promote MSC expression of iNOS in combination with IFNγ and TNFα, but does not affect the expression of chemokines CCL2, CCL5, CXCL9 and CXCL10.
图4显示IL-17协同炎症细胞因子上调MSC中iNOS的表达。Figure 4 shows that IL-17 synergistic inflammatory cytokines up-regulate the expression of iNOS in MSCs.
(A和E).用IFNγ、TNFα和IL-17(每种细胞因子浓度均为10ng/ml)几种细胞因子的不同组合刺激MSC,12小时后收细胞抽提RNA,用Real-Time PCR的方法检测iNOS、CCL2、CCL5、CXCL9、CXCL10几种mRNA的表达。(B).将MSC用IFNγ+TNFα或 IFNγ+TNFα+IL-17(每种细胞因子浓度均为10ng/ml)处理指定的不同时间,收细胞提取蛋白,Western Blot检测iNOS的蛋白表达。(C).将MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17刺激24小时(其中IFNγ和TNFα的浓度作梯度稀释,IL-17浓度固定为10ng/ml),收集细胞上清,用Greiss试剂测NO的量。(D).用IFNγ、TNFα和IL-17(每种细胞因子浓度均为10ng/ml)几种细胞因子的不同组合刺激MSC,24小时后收细胞提取蛋白,Western Blot检测iNOS的蛋白表达。(F).将MSC用未处理或anti-IL-17A处理过的激活脾细胞上清刺激24小时,收集细胞提取蛋白,Western Blot检测iNOS的蛋白表达。(G).将MSC用未处理或anti-IL-17A处理过的激活脾细胞上清刺激12小时,收集细胞提取RNA,用Real-Time PCR的方法检测iNOS、CCL2、CCL5、CXCL9、CXCL10几种mRNA的表达。(A and E). MSCs were stimulated with different combinations of several cytokines of IFNγ, TNFα and IL-17 (each cytokine concentration was 10 ng/ml), and RNA was extracted from cells after 12 hours, using Real-Time PCR. The method was used to detect the expression of several mRNAs of iNOS, CCL2, CCL5, CXCL9 and CXCL10. (B). MSC with IFNγ+TNFα or IFNγ+TNFα+IL-17 (each cytokine concentration was 10 ng/ml) was treated at different times, and the protein was extracted from the cells, and the protein expression of iNOS was detected by Western Blot. (C). MSCs were stimulated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 for 24 hours (the concentration of IFNγ and TNFα was diluted in a gradient, IL-17 concentration was fixed at 10 ng/ml), and the cell supernatant was collected. The amount of NO was measured using a Greiss reagent. (D). MSCs were stimulated with different combinations of cytokines of IFNγ, TNFα and IL-17 (each cytokine concentration was 10 ng/ml). After 24 hours, the cells were extracted and the protein expression of iNOS was detected by Western Blot. (F). MSCs were stimulated with untreated or anti-IL-17A-treated activated splenocyte supernatant for 24 hours, cell extract proteins were collected, and protein expression of iNOS was detected by Western Blot. (G). MSCs were stimulated with untreated or anti-IL-17A-treated activated splenocyte supernatant for 12 hours, and the cells were harvested for RNA extraction. Real-Time PCR was used to detect iNOS, CCL2, CCL5, CXCL9, and CXCL10. Expression of mRNA.
实施例5 Act1是介导IL-17增强MSC免疫抑制作用和相关基因表达的关键分子Example 5 Act1 is a key molecule mediating IL-17 to enhance MSC immunosuppression and related gene expression
以往的研究已经证明,IL-17是通过与细胞表面的IL-17RA和IL-17RC组成的异二聚体结合进而激活下游信号通路的,而在IL-17与IL-17RA、IL-17RC结合后,接头蛋白Act1与IL-17R的结合是激活IL-17下游信号通路的核心步骤。在下游信号传导中,IL-17主要通过激活MAPK和NFκB通路来实现对靶基因的调节。有报道表明,在Act1基因缺失的MEF(小鼠胚胎成纤维细胞)中,IL-17的功能和信号传导存在明显缺陷。本发明人建立了Act1knockdown稳转细胞及相应的对照细胞,对比研究了两株细胞受到IL-17刺激后MAPK和NFκB信号通路蛋白磷酸化的变化。与Act1-/-MEF相似,Act1knockdown MSC在受到IL-17刺激后,IκBα、ERK、JNK和p65的磷酸化水平都明显降低,可见IL-17信号传导明显受到抑制(图5A)。Previous studies have demonstrated that IL-17 binds to heterodimers composed of IL-17RA and IL-17RC on the cell surface to activate downstream signaling pathways, while IL-17 binds to IL-17RA and IL-17RC. Afterwards, binding of the adaptor protein Act1 to IL-17R is a central step in activating the downstream signaling pathway of IL-17. In downstream signaling, IL-17 regulates target genes primarily by activating the MAPK and NFκB pathways. It has been reported that IL-17 function and signaling have obvious defects in MEF (mouse embryonic fibroblasts) in which Act1 gene is deleted. The present inventors established Act1knockdown stable cells and corresponding control cells, and compared the phosphorylation of MAPK and NFκB signaling pathway proteins in the two cells after IL-17 stimulation. Similar to Act1 -/- MEF, the phosphorylation levels of IκBα, ERK, JNK, and p65 were significantly reduced in Act1knockdown MSCs after stimulation with IL-17, and IL-17 signaling was significantly inhibited (Fig. 5A).
本发明人同时研究了Act1knockdown后,MSC受到IFNγ、TNFα和IL-17刺激后免疫抑制功能和相关免疫调节基因表达的变化。结果表明,Act1knockdown之后,IL-17不能有效增加IFNγ和TNFα诱导MSC表达iNOS的水平(图5B、C),同时,IL-17也不能增强MSC的免疫抑制功能(图5D)。这些可以归因于Act1 knockdown之后所引起的IL-17信号传导的抑制,也进一步佐证了IL-17可以增强MSC免疫抑制功能和基因表达这一发现。The present inventors also studied the changes in immunosuppressive function and related immunoregulatory gene expression of MSCs stimulated by IFNγ, TNFα and IL-17 after Act1knockdown. The results showed that IL-17 could not effectively increase the level of iNOS expression induced by IFNγ and TNFα after Act1knockdown (Fig. 5B, C), and IL-17 could not enhance the immunosuppressive function of MSC (Fig. 5D). These can be attributed to the inhibition of IL-17 signaling caused by Act1 knockdown, and further confirm the discovery that IL-17 can enhance MSC immunosuppressive function and gene expression.
图5显示IL-17在MSC中发挥的增强免疫抑制和基因表达的作用需要Act1的参与。Figure 5 shows that the role of IL-17 in enhancing enhanced immunosuppression and gene expression in MSC requires the involvement of Act1.
(A).将Act1 knockdown细胞(shAct1MSC)和相应的对照细胞(shCTRL MSC)分别用IL-17(10ng/ml)处理指定的时间,收集不同时间点的细胞,抽提蛋白,Western Blot方法检测p-IκBα、p-ERK、p-JNK、p-p65几种蛋白的表达情况。p-代表磷酸化。(B).将shCTRL MSC和shAct1 MSC分别用IFNγ+TNFα或IFNγ+TNFα+IL-17(每种细胞因子浓度均为10ng/ml)处理24小时,收细胞提取蛋白,Western Blot检测iNOS的蛋白表达。(C).用IFNγ、TNFα和IL-17(每种细胞因子浓度均为10ng/ml)几种细胞因子的不同组合刺激shCTRL MSC和shAct1MSC,12小时后收细胞抽提RNA,用Real-TimePCR的方法检测iNOS mRNA的表达。(D).首先将shCTRL MSC或shAct1MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17(每种因子的浓度均为2ng/ml)刺激12小时,然后将之与A1.1细胞共培养(MSC与A1.1的比例为1:10),12小时后,用3H-Tdr掺入法测定T细胞增殖。所有数据代表三次独立实验的结果。(A). Act1 knockdown cells (shAct1MSC) and corresponding control cells (shCTRL MSC) were treated with IL-17 (10 ng/ml) for a specified time, cells at different time points were collected, and proteins were extracted and detected by Western Blot. Expression of several proteins of p-IκBα, p-ERK, p-JNK and p-p65. P- stands for phosphorylation. (B). shCTRL MSC and shAct1 MSC were treated with IFNγ+TNFα or IFNγ+TNFα+IL-17 (each cytokine concentration was 10 ng/ml) for 24 hours, and the protein was extracted and the protein of iNOS was detected by Western Blot. expression. (C). shCTRL MSC and shAct1 MSC were stimulated with different combinations of cytokines of IFNγ, TNFα and IL-17 (each cytokine concentration was 10 ng/ml). After 12 hours, the cells were extracted and RNA was used, and Real-Time PCR was used. The method detects the expression of iNOS mRNA. (D). First, shCTRL MSC or shAct1MSC was stimulated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 (each concentration was 2 ng/ml) for 12 hours, and then co-cultured with A1.1 cells. (The ratio of MSC to A1.1 was 1:10), and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation. All data represent the results of three independent experiments.
实施例6 IL-17、IFNγ和TNFα预处理的MSC能有效地治疗ConA诱导的肝损伤Example 6 IL-17, IFNγ and TNFα pretreated MSCs can effectively treat ConA-induced liver injury
既然IL-17能明显增强IFNγ和TNFα所诱导的MSC的免疫抑制功能。本发明人进而采用了ConA诱导的肝炎(CIH)模型探索IL-17对MSC免疫抑制功能调节在疾病治疗中的作用意义。CIH主要是T细胞介导的急性肝炎模型,它很好地模拟了人急性爆发性肝炎和病毒性肝炎,T细胞的激活增殖和对肝实质细胞的破坏是其重要的致病机理。很多报道认为,通过药物抑制免疫应答或去除特定的T细胞亚群后,ConA诱导的肝损伤均能明显缓解。围绕IL-17能显著增强MSC抑制T细胞增殖的能力这一研究结果,本 发明人给予CIH小鼠输注经过不同预处理的GFP-MSC(从GFP转基因小鼠骨髓中分离得到的MSC),输注后约7.5小时,检测GFP-MSC在肝脏组织中的聚集情况。研究表明MSC能定位到受损肝脏部位(图6A)。进一步的研究中,本发明人将MSC提前用IFNγ+TNFα或IFNγ+TNFα+IL-17刺激12小时,在小鼠接受ConA注射30分钟后,将这些不同处理的MSC输注到小鼠体内,观察并检测小鼠的肝脏损伤程度。结果显示,相比MSC治疗组和IFNγ+TNFα处理过的MSC治疗组,IFNγ+TNFα+IL-17处理过的MSC能更有效地治疗ConA诱导的肝损伤,主要表现在小鼠血清中ALT的含量明显降低(图6B),肝脏充血程度明显减轻(图6C)。至此,本发明人的研究从体内和体外实验证明了IL-17可以显著增强MSC的免疫抑制功能。Since IL-17 can significantly enhance the immunosuppressive function of MSC induced by IFNγ and TNFα. The present inventors further adopted the ConA-induced hepatitis (CIH) model to explore the role of IL-17 in the regulation of MSC immunosuppressive function in the treatment of diseases. CIH is mainly a T cell-mediated acute hepatitis model, which is a good model for human acute fulminant hepatitis and viral hepatitis. The activation and proliferation of T cells and the destruction of hepatocytes are important pathogenic mechanisms. Many reports suggest that ConA-induced liver damage can be significantly alleviated by drugs that suppress the immune response or remove specific T cell subsets. Surrounding IL-17 can significantly enhance the ability of MSCs to inhibit T cell proliferation. The inventors administered CIH mice to different pretreated GFP-MSCs (MSCs isolated from the bone marrow of GFP transgenic mice) and detected GFP-MSC aggregation in liver tissue approximately 7.5 hours after infusion. Studies have shown that MSC can locate damaged liver sites (Figure 6A). In a further study, the inventors stimulated MSCs with IFNγ+TNFα or IFNγ+TNFα+IL-17 for 12 hours in advance, and infused these MSCs into mice after 30 minutes of ConA injection. The degree of liver damage in the mice was observed and examined. The results showed that IFNγ+TNFα+IL-17 treated MSCs were more effective in treating ConA-induced liver injury than MSC-treated and IFNγ+TNFα-treated MSC-treated groups, mainly in ALT of mouse serum. The content was significantly reduced (Fig. 6B), and the degree of hepatic congestion was significantly reduced (Fig. 6C). So far, the inventors' studies have demonstrated that IL-17 can significantly enhance the immunosuppressive function of MSCs from in vivo and in vitro experiments.
图6显示IL-17、IFNγ和TNFα预处理的MSC能有效地治疗ConA诱导的肝损伤。Figure 6 shows that IL-17, IFNγ and TNFα pretreated MSCs are effective in treating ConA-induced liver damage.
(A).给小鼠静脉注射ConA(15mg/Kg)30min后,静脉输入control、IFNγ+TNFα或IFNγ+TNFα+IL-17预处理的GFP-MSC(每只小鼠给予5×105个细胞)。7.5小时后,取一小叶肝脏做冰冻切片,通过免疫荧光的方法检测GFP-MSC在肝脏中的定位。蓝色代表DAPI,红色代表GFP。图中箭头指向GFP-MSC,放大倍数为400×。(B,C).给小鼠静脉注射ConA(15mg/Kg)30min后,静脉输入不同细胞因子预处理的MSC(每组5只小鼠,每只小鼠给予5×105个细胞)。7.5小时后,取血,处死小鼠取肝脏做后续检测。(B).血清ALT的水平。(C).各组小鼠肝脏大体观察。(A). After intravenous injection of ConA (15 mg/Kg) for 30 min, GFP-MSC pretreated with control, IFNγ+TNFα or IFNγ+TNFα+IL-17 was administered intravenously (5×10 5 per mouse) cell). After 7.5 hours, a small leaf liver was taken for frozen section, and the localization of GFP-MSC in the liver was detected by immunofluorescence. Blue represents DAPI and red represents GFP. The arrow in the figure points to GFP-MSC with a magnification of 400×. (B, C). After intravenous injection of ConA (15 mg/Kg) for 30 min, MSCs pretreated with different cytokines were intravenously administered (5 mice per group, 5 × 10 5 cells per mouse). After 7.5 hours, blood was taken and the mice were sacrificed for liver testing. (B). Level of serum ALT. (C). Liver observation of mice in each group.
实施例7 IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于其对T细胞的免疫抑制功能,而不影响T细胞亚群的比例Example 7 The therapeutic effect of IL-17, IFNγ and TNFα pretreated MSCs on CIH depends on their immunosuppressive function on T cells without affecting the proportion of T cell subsets.
基于IL-17、IFNγ和TNFα预处理的MSC对ConA诱导的肝损伤有很好的治疗效果,本发明人进一步分析了此种预处理MSC调控CIH的具体机制。本发明人从小鼠的肝脏组织中分离了其中的单个核细胞,计数后发现,L-17、IFNγ和TNFα预处理的MSC治疗后小鼠肝脏单个核细胞数目显著减少(图7A);流式细胞术分析发现,CD4+和CD8+的T细胞数目明显减少(图7A)。这些结果表明,IL-17、IFNγ和TNFα预处理的MSC治疗肝损伤后,MSC强大的免疫抑制功能使得肝脏组织中T细胞增殖受到抑制,进而T细胞介导的免疫应答和对肝细胞的损伤都明显减弱。MSCs pretreated based on IL-17, IFNγ and TNFα have a good therapeutic effect on ConA-induced liver injury, and the inventors further analyzed the specific mechanism of such pretreatment of MSC to regulate CIH. The present inventors isolated mononuclear cells from the liver tissue of mice, and found that the number of liver mononuclear cells in mice treated with L-17, IFNγ and TNFα pretreated MSCs was significantly reduced (Fig. 7A); Cytological analysis revealed a significant decrease in the number of T cells in CD4 + and CD8 + (Fig. 7A). These results indicate that IL-17, IFNγ and TNFα pretreated MSCs have a strong immunosuppressive function in inhibiting T cell proliferation in liver tissue after liver injury, and thus T cell-mediated immune response and damage to liver cells. Both are significantly weakened.
MSC的免疫调节功能,不仅体现在其对T细胞的免疫抑制,也表现为对T细胞亚群分化的调节。此外,MSC可以诱导Th17细胞向Treg细胞分化,进而起到降低免疫反应的作用。为此,本发明人进而研究在CIH模型中所观察到的IL-17、IFNγ和TNFα预处理的MSC的治疗效果是否也与MSC对T细胞亚群分化的影响相关。本发明人从不同处理组小鼠的肝脏中分离得到单个核细胞,在体外经过PMA和inomycin刺激后,检测了其中T细胞亚群的变化。结果显示,正常小鼠肝脏中,CD4+T细胞和CD8+T细胞的比例约为2.3:1(图7B)。ConA诱导后的肝脏中,CD8+T细胞比例增加了一倍左右,CD4+T细胞和CD8+T细胞的比例约为(0.9~1.1):1;然而不同处理的MSC治疗后,CD4+T细胞和CD8+T细胞的比例变化不大(图7B);CD4+T细胞中,IFNγ+IL-17A-的Th1细胞、IFNγ-IL-17A+的Th17细胞(图7C)以及Treg细胞(图7D)的比例都没有显著差异。因此,IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于其对T细胞增殖的免疫抑制功能,而不影响T细胞亚群的比例。The immunomodulatory function of MSC is not only reflected in its immunosuppression of T cells, but also in the regulation of T cell subset differentiation. In addition, MSC can induce Th17 cells to differentiate into Treg cells, thereby reducing the immune response. To this end, the inventors further investigated whether the therapeutic effects of IL-17, IFNγ, and TNFα-pretreated MSCs observed in the CIH model are also related to the effect of MSCs on T cell subset differentiation. The present inventors isolated mononuclear cells from the livers of mice of different treatment groups, and detected changes in T cell subsets after stimulation with PMA and inomycin in vitro. The results showed that the ratio of CD4 + T cells to CD8 + T cells in the liver of normal mice was about 2.3:1 (Fig. 7B). In the liver after ConA induction, the proportion of CD8 + T cells increased by about one-fold, and the ratio of CD4 + T cells to CD8 + T cells was about (0.9 to 1.1): 1; however, after treatment with different treatments, CD4 + T The ratio of cells to CD8 + T cells did not change much (Fig. 7B); in CD4 + T cells, IFNγ + IL-17A - Th1 cells, IFNγ - IL-17A + Th17 cells (Fig. 7C) and Treg cells (Fig. There is no significant difference in the proportion of 7D). Thus, the therapeutic effect of IL-17, IFNγ, and TNFα pretreated MSCs on CIH is dependent on their immunosuppressive function on T cell proliferation without affecting the proportion of T cell subsets.
图7显示IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于其对T细胞的免疫抑制功能,而不影响T细胞亚群的比例。Figure 7 shows that the therapeutic effect of IL-17, IFNy and TNF[alpha] pretreated MSCs on CIH is dependent on their immunosuppressive function on T cells without affecting the proportion of T cell subsets.
给小鼠静脉注射ConA(15mg/kg)30min后,静脉输入不同细胞因子预处理的MSC(每组5只小鼠,每只小鼠给予5×105个细胞)。8小时后,处死小鼠取肝脏做后续检测。(A).研磨肝脏后分离单个核细胞并计数。流式细胞术分析CD4+和CD8+T细胞的比例并换算为绝对数目。(B).流式细胞术分析肝脏中CD4+和CD8+T细胞的比例。(C和D).将肝脏中单个核细胞用PMA和inomycin刺激5小时后,向培养体系中加入BFA继续培养1小时,然后流式细胞术分析肝脏CD4+T细胞中IFNγ+IL-17A-Th1、IFNγ- IL-17A+Th17、Foxp3+Treg细胞的比例。After intravenous injection of ConA (15 mg/kg) for 30 min, mice were intravenously injected with MSCs pretreated with different cytokines (5 mice per group, 5 × 10 5 cells per mouse). Eight hours later, the mice were sacrificed and the liver was taken for follow-up testing. (A). Mononuclear cells were isolated and counted after grinding the liver. The ratio of CD4 + and CD8 + T cells was analyzed by flow cytometry and converted to an absolute number. (B). Flow cytometry analysis of the proportion of CD4 + and CD8 + T cells in the liver. (C and D). After stimulation of mononuclear cells in the liver with PMA and inomycin for 5 hours, BFA was added to the culture system for further 1 hour, and then IFNγ+IL-17A in liver CD4+ T cells was analyzed by flow cytometry. The ratio of Th1, IFNγ-IL-17A + Th17, Foxp3 + Treg cells.
实施例8 IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于iNOS的表达Example 8 The therapeutic effect of IL-17, IFNγ and TNFα pretreated MSCs on CIH depends on the expression of iNOS
由于在体外实验中IL-17增强MSC的免疫抑制作用依赖iNOS的活性(图2B),本发明人进一步应用体内实验验证iNOS在此过程中的核心作用。为了证明这一点,本发明人用iNOS-/-MSC来替代WT MSC,并以IFNγ+TNFα或IFNγ+TNFα+IL-17不同因子组合刺激细胞,然后用上述经过不同预处理iNOS-/-MSC来治疗CIH小鼠,观察其疗效。结果显示,当MSC缺失iNOS时,即便以IFNγ、TNFα、和IL-17预处理细胞,MSC也不能很好地治疗ConA诱发的肝损伤。主要表现在,相比于IFNγ+TNFα+IL-17预处理的WT MSC,无论是未处理iNOS-/-MSC还是IFNγ+TNFα+IL-17预处理iNOS-/-MSC均不能有效抑制CIH小鼠血清中的ALT,(图8A),小鼠肝脏中单个核细胞浸润也未降低(图8B),CD4+T细胞和CD8+T细胞的数目没有下降(图8C),并且肝脏病理切片仍显示有较多的坏死区域(图8D,8E)。因此,本发明人的研究以体内实验证明iNOS是IL-17增强MSC免疫抑制功能的关键分子。Since IL-17 enhances the immunosuppressive effect of MSCs on iNOS activity in vitro (Fig. 2B), the inventors further applied in vivo experiments to verify the central role of iNOS in this process. To demonstrate this, the inventors used iNOS -/- MSCs instead of WT MSCs and stimulated cells with a combination of different factors, IFNγ+TNFα or IFNγ+TNFα+IL-17, and then used the different pretreatments of iNOS -/- MSCs. To treat CIH mice, observe the efficacy. The results showed that MSCs did not treat ConA-induced liver injury well when MSCs were deficient in iNOS, even if cells were pretreated with IFNγ, TNFα, and IL-17. Mainly, compared with IFNγ+TNFα+IL-17 pretreated WT MSC, both untreated iNOS -/- MSC or IFNγ+TNFα+IL-17 pretreated iNOS -/- MSC could not effectively inhibit CIH In ALT in murine serum (Fig. 8A), mononuclear cell infiltration in mouse liver was also not reduced (Fig. 8B), and the number of CD4 + T cells and CD8 + T cells did not decrease (Fig. 8C), and liver pathological sections were still More necrotic areas were shown (Fig. 8D, 8E). Therefore, the inventors' studies have demonstrated in vivo that iNOS is a key molecule for IL-17 to enhance the immunosuppressive function of MSC.
图8显示IL-17、IFNγ和TNFα预处理的MSC对于CIH的治疗效果依赖于iNOS的表达。Figure 8 shows that the therapeutic effect of IL-17, IFNγ and TNFα pretreated MSCs on CIH is dependent on the expression of iNOS.
给小鼠静脉注射ConA(15mg/Kg)30min后,静脉输入不同细胞因子预处理的WT MSC或iNOS-/-MSC(每组5只小鼠,每只小鼠给予5×105个细胞)。7.5小时后,取血后,处死小鼠取肝脏做后续检测。(A).检测血清ALT水平。(B).研磨肝脏后分离单个核细胞并计数。(C).流式细胞术分析CD4+和CD8+T细胞的比例并换算为绝对数目。(D).取一小叶肝脏做石蜡包埋、H&E染色并拍照,放大倍数为200×。(E).计算(D)图中各组肝脏病理切片中坏死区域的百分比。结果以平均数±标准差表示。After intravenous injection of ConA (15 mg/Kg) for 30 min, WT MSCs or iNOS-/-MSCs pretreated with different cytokines were intravenously administered (5 mice per group, 5 × 10 5 cells per mouse) . After 7.5 hours, after taking blood, the mice were sacrificed and the liver was taken for follow-up testing. (A). Detection of serum ALT levels. (B). Mononuclear cells were isolated and counted after grinding the liver. (C). Flow cytometry analysis of the ratio of CD4 + and CD8 + T cells and conversion to absolute numbers. (D) Take a small leaf liver for paraffin embedding, H&E staining and photographing, the magnification is 200×. (E). Calculate the percentage of necrotic areas in each group of liver pathological sections in (D). Results are expressed as mean ± standard deviation.
实施例9 IL-17可以逆转RNA结合蛋白AUF1对基因表达的抑制作用Example 9 IL-17 can reverse the inhibition of gene expression by the RNA binding protein AUF1
接下来本发明人研究了IL-17协同炎症细胞因子IFNγ和TNFα促进MSC表达iNOS的分子机制。在免疫应答中产生的许多免疫分子的mRNA量的控制是非常关键的,一旦mRNA存在过多积累就会引起免疫细胞的过度激活,进而引发超敏反应。在体内,mRNA的积累可以受多种机制调节,其中最重要的机制之一就是mRNA结合蛋白和mRNA的相互作用加快mRNA的降解。然而,在许多炎症反应中,信号通路的活化往往能促进一些基因的表达,而这一促进作用则需要通过增强mRNA的稳定性来实现。因此,控制mRNA的稳定性对许多免疫反应中基因表达的调控都是至关重要的。IL-17也不例外,IL-17正是通过提高mRNA的稳定性来促进一些炎症分子的表达。尽管如此,IL-17提高mRNA稳定性的作用机制还未完全阐明。mRNA结合蛋白对于调节mRNA的稳定性起到至关重要的作用,而且有报道认为RNA结合蛋白AUF1可以负向调节iNOSmRNA的稳定性,而敲低AUF1可以明显增加iNOS mRNA的表达,因此本发明人着重研究了AUF1这一RNA结合蛋白是否参与了IL-17调控MSC中iNOS的基因表达。Next, the inventors studied the molecular mechanism by which IL-17 synergistic inflammatory cytokines IFNγ and TNFα promote MSC expression of iNOS. The control of the amount of mRNA of many immune molecules produced in an immune response is critical, and once the mRNA is excessively accumulated, it causes excessive activation of immune cells, which in turn triggers a hypersensitivity reaction. In vivo, mRNA accumulation can be regulated by a variety of mechanisms, one of the most important mechanisms is the interaction of mRNA binding protein and mRNA to accelerate the degradation of mRNA. However, in many inflammatory responses, activation of signaling pathways often promotes the expression of some genes, and this promotion needs to be achieved by enhancing the stability of the mRNA. Therefore, controlling the stability of mRNA is critical for the regulation of gene expression in many immune responses. IL-17 is no exception. IL-17 promotes the expression of some inflammatory molecules by increasing the stability of mRNA. Nevertheless, the mechanism of action of IL-17 to improve mRNA stability has not been fully elucidated. The mRNA binding protein plays a crucial role in regulating the stability of mRNA, and it has been reported that the RNA binding protein AUF1 can negatively regulate the stability of iNOS mRNA, and knocking down AUF1 can significantly increase the expression of iNOS mRNA, so the inventors It was investigated whether AUF1, an RNA-binding protein, is involved in IL-17 regulation of iNOS gene expression in MSCs.
本发明人从auf1-/-小鼠骨髓中分离得到MSC,并比较了WT MSC和auf1-/-MSC在受到IFNγ+TNFα或IFNγ+TNFα+IL-17刺激后iNOS的表达情况。结果表明,尽管在WTMSC中IL-17可以协同IFNγ和TNFα上调iNOS mRNA和蛋白的表达,但在auf1-/-MSC中,仅IFNγ和TNFα即可以诱导MSC产生大量的iNOS(图9A,B),IL-17并不能体现出明显的协同作用。这一现象也在AUF1 knockdown MSC中得到了验证。在AUF1 knockdown MSC中IL-17的协同作用也是明显削弱的(图9C,D,E)。不仅在基因表达水平上,本发明人在IL-17增强免疫抑制这一特性上也发现了相似的现象:在WTMSC中,IL-17可以增强IFNγ和TNFα所诱导的免疫抑制;但是在auf1-/-MSC中,仅IFNγ和TNFα即可诱导出最强的免疫抑制,不需要IL-17的补充加入(图9F)。这些实验结果提示:在WT MSC中,IL-17可以逆转AUF1对iNOS mRNA表达的抑制作用;当AUF1 缺失的情况下,IFNγ和TNFα诱导生成的iNOS mRNA失去了AUF1对其表达的抑制作用,表现出高水平的表达,IL-17的促进作用则会明显减弱;AUF1很可能是IL-17发挥增强基因表达作用过程中作用的靶标。The present inventors isolated MSCs from the bone marrow of auf1 -/- mice and compared the expression of iNOS in WT MSCs and auf1 -/- MSCs after stimulation with IFNγ + TNFα or IFNγ + TNFα + IL-17. The results showed that although IL-17 can up-regulate the expression of iNOS mRNA and protein in WTMSC in combination with IFNγ and TNFα, only IFNγ and TNFα can induce MSC to produce large amounts of iNOS in auf1 -/- MSC (Fig. 9A, B). IL-17 does not reflect obvious synergy. This phenomenon was also verified in the AUF1 knockdown MSC. The synergy of IL-17 was also significantly attenuated in the AUF1 knockdown MSC (Fig. 9C, D, E). Not only at the level of gene expression, the inventors found a similar phenomenon in the enhancement of immunosuppression by IL-17: in WTMSC, IL-17 can enhance the immunosuppression induced by IFNγ and TNFα; however, in auf1 - /- In MSC, only IFNγ and TNFα can induce the strongest immunosuppression without the addition of IL-17 (Fig. 9F). These results suggest that IL-17 can reverse the inhibition of iNOS mRNA expression by AUF1 in WT MSCs; when AUF1 is deleted, iNOS mRNA induced by IFNγ and TNFα loses the inhibitory effect of AUF1 on its expression. At a high level of expression, the promotion of IL-17 is significantly attenuated; AUF1 is likely to be a target for IL-17 to play a role in enhancing gene expression.
图9显示IL-17可以逆转RNA结合蛋白AUF1对基因表达的抑制作用。Figure 9 shows that IL-17 can reverse the inhibition of gene expression by the RNA binding protein AUF1.
(A).用IFNγ、TNFα和IL-17(每种细胞因子浓度均为10ng/ml)几种细胞因子的不同组合刺激WT MSC或auf1-/-MSC,12小时后收细胞抽提RNA,用Real-Time PCR的方法检测iNOS mRNA的表达。(B).将WT MSC或auf1-/-MSC用IFNγ+TNFα或IFNγ+TNFα+IL-17(每种细胞因子浓度均为10ng/ml)刺激24小时,收细胞提取蛋白,Western Blot检测iNOS的蛋白表达。(C).在WT MSC上稳定转染Act1shRNA后用Real-Time PCR和Western Blot的方法鉴定knockdown的效率。(D).将Act1knockdown细胞(shAct1MSC)和相应的对照细胞(shCTRL MSC)用IFNγ、TNFα和IL-17(每种细胞因子浓度均为10ng/ml)几种细胞因子的不同组合刺激,12小时后收细胞抽提RNA,用Real-Time PCR的方法检测iNOS mRNA的表达。(E).将shCTRL MSC或shAUF1 MSC用IFNγ+TNFα或IFNγ+TNFα+IL-17(每种细胞因子浓度均为10ng/ml)刺激12或24小时,收细胞提取蛋白,Western Blot检测iNOS的蛋白表达。(F).首先将WTMSC或auf1-/-MSC用细胞因子IFNγ+TNFα或IFNγ+TNFα+IL-17(每种因子的浓度均为2ng/ml)刺激12小时,然后将之与A1.1细胞共培养(MSC与A1.1的比例为1:10),12小时后,用3H-Tdr掺入法测定T细胞增殖。结果表示为平均数±标准差。所示数据代表四次独立实验的结果。(A). WT MSC or auf1-/-MSCs were stimulated with different combinations of cytokines of IFNγ, TNFα and IL-17 (each cytokine concentration was 10 ng/ml), and RNA was extracted after 12 hours. The expression of iNOS mRNA was detected by Real-Time PCR. (B). WT MSC or auf1-/-MSC was stimulated with IFNγ+TNFα or IFNγ+TNFα+IL-17 (each cytokine concentration was 10 ng/ml) for 24 hours, and the cells were extracted and the protein was detected by Western Blot. Protein expression. (C). The efficiency of knockdown was identified by Real-Time PCR and Western Blot after stable transfection of Act1 shRNA on WT MSC. (D). Actlknockdown cells (shAct1MSC) and corresponding control cells (shCTRL MSC) were stimulated with different combinations of several cytokines of IFNγ, TNFα and IL-17 (each cytokine concentration was 10 ng/ml) for 12 hours. The RNA was extracted from the cells, and the expression of iNOS mRNA was detected by Real-Time PCR. (E). shCTRL MSC or shAUF1 MSC was stimulated with IFNγ+TNFα or IFNγ+TNFα+IL-17 (each cytokine concentration was 10 ng/ml) for 12 or 24 hours, and the cells were extracted and Western blot was used to detect iNOS. Protein. (F). First, WTMSC or auf1-/-MSC was stimulated with cytokine IFNγ+TNFα or IFNγ+TNFα+IL-17 (each concentration was 2ng/ml) for 12 hours, and then it was combined with A1.1. The cells were co-cultured (the ratio of MSC to A1.1 was 1:10), and after 12 hours, T cell proliferation was measured by 3 H-Tdr incorporation. Results are expressed as mean ± standard deviation. The data shown represents the results of four independent experiments.
实施例10 IL-17通过调节AUF1的水平来增强iNOS mRNA的稳定性Example 10 IL-17 enhances the stability of iNOS mRNA by modulating the level of AUF1
以上的研究数据表明,IL-17可以协同炎症细胞因子IFNγ和TNFα提高MSC的免疫抑制功能iNOS的表达,但在AUF1缺失的情况下,IL-17则不能体现出这一协同作用,主要由于IFNγ和TNFα即可诱导auf1-/-MSC产生最强的免疫抑制。这些现象可以解释为:在WT MSC中,iNOS的表达需要IFNγ和TNFα的诱导,而这些诱导出的iNOS mRNA在AUF1存在并与之相互作用的情况下就会快速降解,而IL-17的加入则能逆转AUF1的这一降解mRNA的作用,进而促进了iNOS的基因表达;而在AUF1缺失的情况下,IFNγ和TNFα诱导出的iNOS mRNA则显得非常稳定,IL-17的加入则显示不出促进基因表达的作用。为了验证这一解释,本发明人分别用IFNγ+TNFα或IFNγ+TNFα+IL-17刺激WT MSC和auf1-/-MSC,检测iNOS mRNA在两株细胞中的半衰期。结果发现,在WT MSC中,IL-17可以明显增强IFNγ+TNFα诱导的iNOS mRNA的稳定性,iNOSmRNA的半衰期由t1/2=4.3±1.4hr延长到t1/2>10hr(图10A);在auf1-/-MSC中,IFNγ+TNFα诱导的iNOS mRNA的稳定性大大增强,半衰期t1/2>10hr(图10B),而IL-17的加入没有体现出进一步的延长作用。这些结果证实了本发明人给出的解释。对于不受IL-17影响的CCL2和CXCL10,IL-17也相应地不能增强这些mRNA的稳定性。为了进一步验证这些结果,本发明人也在检测了AUF1 knockdown MSC中iNOS mRNA的半衰期,也得到了与auf1-/-MSC相似的结果(图10C)。The above data indicate that IL-17 can synergize with the inflammatory cytokines IFNγ and TNFα to increase the expression of iNOS in the immunosuppressive function of MSC, but in the absence of AUF1, IL-17 does not reflect this synergistic effect, mainly due to IFNγ. And TNFα can induce the strongest immunosuppression of auf1 -/- MSC. These phenomena can be explained by the fact that in WT MSCs, the expression of iNOS requires the induction of IFNγ and TNFα, and these induced iNOS mRNAs rapidly degrade in the presence and interaction of AUF1, and the addition of IL-17 This can reverse the effect of AUF1 on the degradation of mRNA, which promotes the expression of iNOS gene. In the absence of AUF1, the iNOS mRNA induced by IFNγ and TNFα is very stable, and the addition of IL-17 is not shown. Promote the role of gene expression. To test this explanation, the inventors stimulated WT MSCs and auf1 −/− MSCs with IFNγ+TNFα or IFNγ+TNFα+IL-17, respectively, to detect the half-life of iNOS mRNA in both cells. It was found that IL-17 significantly enhanced the stability of iNOS mRNA induced by IFNγ+TNFα in WT MSCs, and the half-life of iNOS mRNA was extended from t 1/2 =4.3±1.4 hr to t 1/2 >10 hr (Fig. 10A). In auf1 -/- MSC, the stability of iNOS mRNA induced by IFNγ+TNFα was greatly enhanced, and the half-life t 1/2 >10 hr (Fig. 10B), while the addition of IL-17 did not show further prolongation. These results confirm the explanation given by the inventors. For CCL2 and CXCL10 that are not affected by IL-17, IL-17 also does not enhance the stability of these mRNAs accordingly. To further verify these results, the inventors also examined the half-life of iNOS mRNA in AUF1 knockdown MSC, and also obtained similar results as auf1 -/- MSC (Fig. 10C).
既然IL-17能逆转AUF1引起的iNOS mRNA降解,那么AUF1很可能是IL-17在发挥作用中一个很关键的靶点,本发明人接着研究了其中的分子机制。本发明人检测了MSC在受到IFNγ+TNFα或IFNγ+TNFα+IL-17刺激后AUF1表达水平的变化。结果显示,MSC在受到IFNγ、TNFα刺激后,IL-17的加入能明显降低AUF1的蛋白水平(图10D)。由此可见,IL-17通过降低AUF1的水平进而增强iNOS mRNA的稳定性;AUF1是MSC中IL-17发挥增强免疫抑制和基因表达的关键分子。Since IL-17 can reverse the degradation of iNOS mRNA caused by AUF1, AUF1 is likely to be a key target for IL-17 to play a role. The inventors then studied the molecular mechanism. The present inventors examined changes in AUF1 expression levels of MSCs after stimulation with IFNγ+TNFα or IFNγ+TNFα+IL-17. The results showed that the addition of IL-17 significantly reduced the protein level of AUF1 after stimulation with IFNγ and TNFα (Fig. 10D). Thus, IL-17 enhances the stability of iNOS mRNA by decreasing the level of AUF1, which is a key molecule that enhances immunosuppression and gene expression in IL-17.
图10显示IL-17通过调节AUF1的水平来增强iNOS mRNA的稳定性。Figure 10 shows that IL-17 enhances the stability of iNOS mRNA by modulating the level of AUF1.
(A,B,C).将WT MSC(A),auf1-/-MSC(B),shCTRL MSC或shAUF1 MSC(C).用IFNγ+TNFα或IFNγ+TNFα+IL-17(每种细胞因子浓度均为10ng/ml)刺激6小时,然后向培养基中加入Act.D(5μg/ml),在加入Act.D后指定的时间点收集细胞抽提RNA,用 Real-Time PCR的方法测定各时间点iNOS、CCL2、CXCL10的mRNA量,并按材料和方法8中的步骤计算mRNA的半衰期。(D).将MSC用IFNγ+TNFα或IFNγ+TNFα+IL-17(每种细胞因子浓度均为10ng/ml)进行处理,在加入细胞因子后的指定时间点收集细胞提取蛋白,用Western Blot检测各时间点AUF1的蛋白表达。(A, B, C). WT MSC (A), auf1-/-MSC (B), shCTRL MSC or shAUF1 MSC (C). IFNγ+TNFα or IFNγ+TNFα+IL-17 (each cytokine) After stimulation for 6 hours at a concentration of 10 ng/ml, Act.D (5 μg/ml) was added to the medium, and the cells were extracted for RNA at a specified time point after the addition of Act.D. The Real-Time PCR method measures the mRNA levels of iNOS, CCL2, and CXCL10 at each time point, and calculates the half-life of mRNA according to the procedures in Materials and Method 8. (D). MSCs were treated with IFNγ+TNFα or IFNγ+TNFα+IL-17 (each cytokine concentration was 10 ng/ml), and the cell extract protein was collected at the specified time point after the addition of cytokines, using Western Blot. Protein expression of AUF1 at each time point was examined.
实施例11 AUF1是介导IL-17信号转导的关键分子Example 11 AUF1 is a key molecule mediating IL-17 signaling
在上述研究中,IL-17不能协同炎症细胞因子IFNγ和TNFα上调auf1-/-MSC免疫调节基因的表达;而相同的现象也在Act1-/-MSC中可以观察到(图5)。在IL-17与IL-17受体结合后激活细胞时,Act1与IL-17受体发生相互作用,从而介导了信号向下游传导,所以Act1是IL-17信号通路中的关键分子之一。本发明人首先对比了AUF1 knockdown MSC(shAUF1 MSC)和其正常对照细胞(shCTRL MSC)受到IL-17刺激后IL-17信号通路中重要信号分子磷酸化的区别。本发明人发现,在AUF1 knockdown的情况下,MSC接受IL-17刺激后,IL-17信号通路的关键分子p65(NFκB通路)和ERK(MAPK通路)的磷酸化都明显减弱(图11A),提示IL-17信号转导受到抑制,AUF1可能参与了IL-17的信号传递。In the above study, IL-17 was unable to up - regulate the expression of the auf1 -/- MSC immunoregulatory gene in combination with the inflammatory cytokines IFNγ and TNFα; the same phenomenon was observed in Act1 -/- MSC (Fig. 5). When IL-17 binds to the IL-17 receptor and activates cells, Act1 interacts with the IL-17 receptor, which mediates the downstream signaling, so Act1 is one of the key molecules in the IL-17 signaling pathway. . The inventors first compared the difference in phosphorylation of important signaling molecules in the IL-17 signaling pathway by AUF1 knockdown MSC (shAUF1 MSC) and its normal control cells (shCTRL MSC) after IL-17 stimulation. The present inventors have found that in the case of AUF1 knockdown, phosphorylation of the key molecules p65 (NFκB pathway) and ERK (MAPK pathway) of the IL-17 signaling pathway is significantly attenuated after siRNA is stimulated by IL-17 (Fig. 11A). It is suggested that IL-17 signal transduction is inhibited, and AUF1 may be involved in IL-17 signaling.
为了进一步确认AUF1是否通过与Act1相互作用来参与IL-17的信号传导,本发明人将MSC用IL-17刺激后收集不同时间点的细胞,通过免疫共沉淀的方法检测了AUF1和Act1是否存在相互作用。结果显示,在静息状态下的MSC中,Act1与AUF1有少量结合,而在IL-17加入后,与Act1结合的AUF1的量明显增多(图11B)。由此可见,AUF1确实通过与Act1的相互作用参与了IL-17的信号转导。In order to further confirm whether AUF1 participates in IL-17 signaling by interacting with Act1, the inventors collected cells at different time points after stimulation with IL-17, and detected the presence of AUF1 and Act1 by co-immunoprecipitation. interaction. The results showed that Act1 had a small amount of binding to AUF1 in the MSC at rest, and the amount of AUF1 bound to Act1 was significantly increased after IL-17 was added (Fig. 11B). Thus, AUF1 does participate in the signal transduction of IL-17 through interaction with Act1.
至此,本发明人得出如下结论:(1)在启动IL-17信号转导过程中,AUF1与Act1相互作用,从而启动了下游NFκB通路和MAPK通路的活化,进而诱导了靶基因的表达。(2)在IL-17与其他炎症细胞因子如IFNγ和TNFα共同存在的情况下,AUF1则成为IL-17调节的靶点,IL-17通过降低AUF1的水平来增强目的基因mRNA的稳定性,从而促进基因表达。So far, the present inventors have reached the following conclusions: (1) AUF1 interacts with Act1 during the initiation of IL-17 signal transduction, thereby initiating activation of the downstream NFκB pathway and the MAPK pathway, thereby inducing expression of the target gene. (2) In the presence of IL-17 and other inflammatory cytokines such as IFNγ and TNFα, AUF1 becomes a target of IL-17 regulation, and IL-17 enhances the stability of mRNA of the target gene by decreasing the level of AUF1. Thereby promoting gene expression.
图11显示AUF1是介导IL-17信号转导的的关键分子。Figure 11 shows that AUF1 is a key molecule that mediates IL-17 signaling.
(A).将AUF1 knockdown细胞(shAUF1 MSC)和相应的对照细胞(shCTRL MSC)分别用IL-17(10ng/ml)处理指定的时间,收集不同时间点的细胞,抽提蛋白,Western Blot方法检测p-p65、p-ERK蛋白的表达情况。p-代表磷酸化。(B).将MSC用IL-17(10ng/ml)进行处理,在加入细胞因子后的指定时间点收集细胞提取蛋白,用IgG(对照)或抗小鼠Act1抗体与蛋白裂解产物进行免疫共沉淀,Western Blot检测Act1和AUF1。(A). AUF1 knockdown cells (shAUF1 MSC) and corresponding control cells (shCTRL MSC) were treated with IL-17 (10 ng/ml) for a specified time, cells at different time points were collected, and proteins were extracted, Western Blot method. The expression of p-p65 and p-ERK protein was detected. P- stands for phosphorylation. (B). MSCs were treated with IL-17 (10 ng/ml), and cell extract proteins were collected at the indicated time points after addition of cytokines, and immunologically co-immunized with IgG (control) or anti-mouse Act1 antibody and protein cleavage products. Precipitation, Western Blot detection of Act1 and AUF1.
实施例12 AUF1是介导IL-17增强MSC治疗CIH效果的关键分子Example 12 AUF1 is a key molecule mediating the effect of IL-17 on MSC treatment of CIH
基于AUF1在IL-17发挥增强MSC免疫抑制功能中起到关键作用(图6)。接下来,本发明人将在CIH模型中验证这一发现。本发明人将WT MSC和auf1-/-MSC分别用IFNγ+TNFα、IFNγ+TNFα+IL-17预处理12小时,然后给CIH小鼠静脉注射这些不同处理的细胞,观察疗效。本发明人发现:与之前的结果相符,IL-17可以明显增强WT MSC对于CIH的疗效,血清ALT水平、肝脏中单个核细胞、CD4+T细胞和CD8+T细胞的数目、肝脏坏死程度都明显下降。IFNγ+TNFα+IL-17预处理的MSC能明显增强WT MSC对CIH的疗效:血清ALT的平均水平由未处理组的8000U/L降低至2000U/L;肝脏中单个核细胞由未处理组的15×105/g liver降低至5×105/g liver;CD4+T细胞由未处理组的3×105/g liver降低至1×105/g liver;CD8+T细胞由未处理组的3.7×105/gliver降低至1×105/g liver。Based on AUF1, IL-17 plays a key role in enhancing MSC immunosuppressive function (Fig. 6). Next, the inventors will verify this finding in the CIH model. The present inventors pretreated WT MSC and auf1 -/- MSC with IFNγ+TNFα, IFNγ+TNFα+IL-17 for 12 hours, respectively, and then intravenously injected these differently treated cells into CIH mice to observe the therapeutic effect. The present inventors found that IL-17 can significantly enhance the efficacy of WT MSCs for CIH, serum ALT levels, mononuclear cells in the liver, number of CD4 + T cells and CD8 + T cells, and degree of liver necrosis, consistent with previous results. Significant decline. IFNγ+TNFα+IL-17 pretreated MSC significantly enhanced the efficacy of WT MSCs on CIH: the mean level of serum ALT decreased from 8000 U/L to 2000 U/L in the untreated group; mononuclear cells in the liver were from the untreated group. 15×10 5 /g liver was reduced to 5×10 5 /g liver; CD4 + T cells were reduced from 3×10 5 /g liver in the untreated group to 1×10 5 /g liver; CD8 + T cells were untreated The group's 3.7 × 10 5 /gliver was reduced to 1 × 10 5 /g liver.
但是对于auf1-/-MSC而言,IFNγ+TNFα预处理后即可以很好地治疗CIH,而IL-17的加入也不能增强疗效(图12)。至此,本发明人在体内验证了AUF1是介导IL-17增强MSC免疫抑制功能并用于治疗CIH的关键分子。 However, for auf1 -/- MSC, IFNγ + TNFα pretreatment was a good treatment for CIH, and the addition of IL-17 did not enhance the efficacy (Figure 12). So far, the present inventors have demonstrated in vivo that AUF1 is a key molecule that mediates IL-17 enhancing MSC immunosuppressive function and is used for the treatment of CIH.
图12显示AUF1是介导IL-17增强MSC治疗CIH效果的关键分子。Figure 12 shows that AUF1 is a key molecule that mediates the effect of IL-17 on MSC treatment of CIH.
给小鼠静脉注射ConA(15mg/kg)30min后,静脉输入不同细胞因子预处理的WT MSC或auf1-/-MSC(每组3-5只小鼠,每只小鼠给予5×105个细胞)。7.5小时后,取血,处死小鼠取肝脏做后续检测。(A).检测血清ALT水平。(B).研磨肝脏后分离单个核细胞并计数。(C).流式细胞术分析CD4+和CD8+T细胞的比例并换算为绝对数目。(D).取一小叶肝脏做石蜡包埋、H&E染色并拍照,放大倍数为200×。(E).计算(D)图中各组肝脏病理切片中坏死区域的百分比。结果以平均数±标准差表示。After intravenous injection of ConA (15 mg/kg) for 30 min, WT MSCs or auf1 -/- MSCs pretreated with different cytokines were intravenously administered (3-5 mice per group, 5 × 10 5 per mouse) cell). After 7.5 hours, blood was taken and the mice were sacrificed for liver testing. (A). Detection of serum ALT levels. (B). Mononuclear cells were isolated and counted after grinding the liver. (C). Flow cytometry analysis of the ratio of CD4 + and CD8 + T cells and conversion to absolute numbers. (D) Take a small leaf liver for paraffin embedding, H&E staining and photographing, the magnification is 200×. (E). Calculate the percentage of necrotic areas in each group of liver pathological sections in (D). Results are expressed as mean ± standard deviation.
实施例13炎症因子预处理MSCs可以有效治疗肝硬化Example 13 inflammatory factor pretreatment of MSCs can effectively treat cirrhosis
给予C57BL/6小鼠每周灌胃四氯化碳(carbon tetrachloride,CCL4),连续8周,以建立肝硬化模型。C57BL/6 mice were given peritoneal carbon tetrachloride (CCL 4 ) weekly for 8 weeks to establish a model of cirrhosis.
在肝硬化模型建立的第8周给予小鼠输注不同炎症因子(10ng/ml IFNγ+10ng/ml TNFα或10ng/ml IFNγ+10ng/ml TNFα+10ng/ml IL-17)处理的间充质干细胞(mesenchymal stem cells,MSCs)和未处理的MSCs,观察不同处理MSCs对肝硬化的治疗效果。Mice treated with different inflammatory factors (10 ng/ml IFNγ+10 ng/ml TNFα or 10 ng/ml IFNγ+10 ng/ml TNFα+10 ng/ml IL-17) were administered to mice at the 8th week of the cirrhosis model. Stem cells (mesensymal stem cells, MSCs) and untreated MSCs were used to observe the therapeutic effects of differently treated MSCs on cirrhosis.
研究发现,给予小鼠诱导肝硬化后,小鼠血清中总胆红素(total bilirubin,TB)、谷丙转氨酶(alanine aminotrans-ferase,ALT)、谷草转氨酶(aspartate amino-transferase,AST)均显著升高,白蛋白(albumin)水平显著降低。其中,与正常组小鼠相比,肝硬化模型组小鼠体内的TB水平达到12-15μmol/L;ALT水平升高至1200-1800U/L;AST水平升高至3500-4500U/L;白蛋白水平降低至20g/L以下。The study found that after cirrhosis was induced in mice, total bilirubin (TB), alanine aminotrans-ferase (ALT), and aspartate amino-transferase (AST) were significantly increased in the serum of mice. Elevated, albumin levels were significantly reduced. Among them, compared with the normal group of mice, the TB level in the cirrhotic model group reached 12-15μmol/L; the ALT level increased to 1200-1800U/L; the AST level increased to 3500-4500U/L; Protein levels are reduced to below 20 g/L.
对肝硬化模型小鼠输注未经处理的MSCs(1×106),可以有效抑制肝硬化小鼠血清中的TB、ALT、AST的水平,并显著提高白蛋白(albumin)的水平。输注了MSCs7天后,肝硬化小鼠血清中TB水平降低了50-60%;ALT水平降低约50%;AST水平降低了60%;白蛋白水平升高了5%。Infusion of untreated MSCs (1×10 6 ) into cirrhotic model mice can effectively inhibit the levels of TB, ALT and AST in the serum of cirrhotic mice and significantly increase the level of albumin. Seven days after infusion of MSCs, serum TB levels in cirrhotic mice were reduced by 50-60%; ALT levels were reduced by approximately 50%; AST levels were reduced by 60%; and albumin levels were increased by 5%.
而将炎症因子IFNγ+TNFα预处理12小时后的MSCs(1×106),输注到肝硬化小鼠体内7天后,肝硬化小鼠血清中TB水平降低了约70%;ALT水平降低了70-80%;AST水平降低了70%左右;白蛋白水平升高了20%。显示了更好的肝硬化治疗效果。因此,炎症因子预处理MSCs可以有效提高MSCs对于肝硬化的治疗作用。After MSCs (1×10 6 ) pretreated with inflammatory factor IFNγ+TNFα for 12 hours, 7 days after infusion into cirrhotic mice, serum TB levels in cirrhotic mice decreased by about 70%; ALT levels decreased. 70-80%; AST levels are reduced by about 70%; albumin levels are increased by 20%. Shows better treatment of cirrhosis. Therefore, inflammatory factors pretreatment of MSCs can effectively improve the therapeutic effect of MSCs on cirrhosis.
IFNγ+TNFα+IL-17预处理MSCs(1×106),输注到肝硬化小鼠体内7天后,肝硬化小鼠血清中TB水平降低了85%;ALT水平降低了约80-85%;AST水平降低了约80-90%;白蛋白水平升高了30%。IL-17的加入,表现出了明显的协同作用,可以更为显著的提高MSCs对肝硬化的治疗作用。IFNγ+TNFα+IL-17 pretreated MSCs (1×10 6 ), and after 7 days of infusion into cirrhotic mice, serum TB levels in cirrhotic mice decreased by 85%; ALT levels decreased by about 80-85%. The AST level was reduced by about 80-90%; the albumin level was increased by 30%. The addition of IL-17 showed a significant synergistic effect and could significantly improve the therapeutic effect of MSCs on cirrhosis.
讨论discuss
IL-17对于MSC免疫抑制的增强作用的发现使得本发明人对炎症细胞因子调控MSC免疫调节能力的研究更深入了一步,同时也为MSC在临床上的应用奠定了基础。在传统意义上,IL-17是公认的促进免疫应答的炎症因子,很多研究表明IL-17是多种炎症疾病和自身免疫病的致病因子,包括类风湿性关节炎、多发性硬化和炎症性肠病等。IL-17在许多自身免疫病病人的血清和组织中呈高现表达,通过使用IL-17中和抗体或敲除IL-17基因,可以明显缓解这些自身免疫病的症状。尽管如此,IL-17并不是在所有的病理情况下都能促进免疫应答。比如,在DSS(葡聚糖硫酸钠)诱导的小鼠肠炎模型中,IL-17中和抗体的应用或敲除IL-17基因反而会加速疾病进程。然而,这一现象相关的分子机制至今还未完全阐明。本发明人的研究表明在MSC存在的情况下,IL-17可以发挥免疫抑制作用,而当本发明人用中和抗体去除MSC-T细胞共培养体系中的IL-17时,MSC的免疫抑制功能会受损。本发明人的这些发现为在病理生理条件下研究IL-17的作用提供了新的思路和方向。The discovery that IL-17 enhances the immunosuppression of MSCs has enabled the present inventors to further study the ability of inflammatory cytokines to regulate MSC immunoregulation, and also laid a foundation for the clinical application of MSCs. In the traditional sense, IL-17 is recognized as an inflammatory factor that promotes immune responses. Many studies have shown that IL-17 is a causative agent of various inflammatory diseases and autoimmune diseases, including rheumatoid arthritis, multiple sclerosis and inflammation. Sexual bowel disease, etc. IL-17 is highly expressed in the serum and tissues of many autoimmune patients, and the symptoms of these autoimmune diseases can be significantly alleviated by using IL-17 neutralizing antibodies or knocking out the IL-17 gene. Nevertheless, IL-17 does not promote an immune response in all pathological conditions. For example, in a DSS (dextran sulfate sodium)-induced mouse enteritis model, the application of IL-17 neutralizing antibodies or knocking out the IL-17 gene may accelerate disease progression. However, the molecular mechanism associated with this phenomenon has not yet been fully elucidated. Studies by the present inventors have shown that IL-17 can exert an immunosuppressive effect in the presence of MSC, and immunosuppression of MSCs when the present inventors neutralize IL-17 in a MSC-T cell co-culture system with a neutralizing antibody. The function will be damaged. These findings by the present inventors provide new ideas and directions for studying the effects of IL-17 under pathophysiological conditions.
ConA诱导小鼠肝损伤(CIH)是一个很经典的模拟人病毒性或自身免疫性急性 肝炎的动物模型。在这一模型中,急性免疫应答在介导肝损伤中发挥了主导作用。多种免疫细胞(T淋巴细胞、巨噬细胞、NK细胞)和免疫分子(细胞因子IFNγ、TNFα、IL-10、IL-22、IL-25等)都参与了CIH的病理过程。抑制免疫应答可以很有效地治疗CIH。在本论文中,本发明人采用了小鼠骨髓来源的MSC,并尝试将IL-17增强MSC免疫抑制这一特性应用到其对CIH的治疗上。结果发现,未处理的MSC对CIH的治疗效果较差。这主要是因为MSC对T细胞的免疫抑制作用依赖炎症细胞因子的刺激,虽然在CIH疾病进展中存在多种细胞因子包括IFNγ和TNFα,但是这些细胞因子只能在很短时间内保持较高的浓度,不足以赋予MSC很强的免疫抑制能力;然而IFNγ、TNFα和IL-17三者预处理的MSC可以体现最佳的治疗效果,这一现象与本发明人在体外实验中得出的结论相符。IL-17对MSC治疗CIH的增强作用依赖于IL-17加入后增加MSC表达大量的iNOS,对于iNOS-/-MSC而言,即使提前用IFNγ、TNFα和IL-17三者预处理,也不能使其有效地治疗CIH。因此,本发明人在体内实验中同样验证了IL-17增强免疫抑制这一发现。ConA-induced liver injury in mice (CIH) is a classic animal model that mimics human viral or autoimmune acute hepatitis. In this model, the acute immune response plays a leading role in mediating liver damage. A variety of immune cells (T lymphocytes, macrophages, NK cells) and immune molecules (cytokines IFNγ, TNFα, IL-10, IL-22, IL-25, etc.) are involved in the pathological process of CIH. Inhibiting an immune response can be a very effective treatment for CIH. In the present paper, the present inventors employed mouse bone marrow-derived MSCs and attempted to apply the IL-17-enhanced MSC immunosuppression property to its treatment of CIH. It was found that untreated MSCs were less effective in treating CIH. This is mainly because the immunosuppressive effect of MSC on T cells is dependent on the stimulation of inflammatory cytokines. Although there are many cytokines including IFNγ and TNFα in the progression of CIH disease, these cytokines can only remain high in a short time. The concentration is not sufficient to confer strong immunosuppressive ability to MSC; however, MSCs pretreated with IFNγ, TNFα and IL-17 can demonstrate the best therapeutic effect, which is consistent with the inventors' conclusions in vitro. Match. The enhancement of IL-17 treatment of CIH by IL-17 depends on the increase of MSC expression of large amount of iNOS after IL-17 addition. For iNOS -/- MSC, even if it is pretreated with IFNγ, TNFα and IL-17 in advance, it cannot Make it effective in treating CIH. Therefore, the inventors also verified the discovery that IL-17 enhances immunosuppression in an in vivo experiment.
本发明人的研究显示,AUF1的敲除可以促进MSC中IFNγ和TNFα诱导的iNOS的表达,提示了AUF1对MSC中基因表达调控的重要性。为了进一步揭示IL-17上调MSC基因表达的分子机制,本发明人检测了IL-17对iNOS mRNA稳定性的影响,结果显示IL-17可以显著增强IFNγ和TNFα诱导的iNOS mRNA稳定性。当AUF1不存在或被敲低后,IL-17不能协同IFNγ和TNFα增强iNOS基因表达,也不能进一步增强iNOS mRNA稳定性。体外免疫抑制实验显示,auf1-/-MSC只需要IFNγ和TNFα存在即可发挥最强的免疫抑制能力,不需要IL-17的补充。本发明人进一步研究了AUF1在IL-17增强MSC中基因表达的具体机制,结果发现MSC在受到IFNγ和TNFα刺激的同时,IL-17的加入可以降低AUF1的表达水平,这一发现揭示AUF1参与了IL-17对MSC相关基因表达的调控,也就是说,IL-17通过降低AUF1的水平进而促进iNOS mRNA的稳定性。免疫共沉淀结果还显示,IL-17刺激后15min,AUF1和Act1的相互作用明显增强。本发明人还研究了AUF1 knockdown MSC中IL-17信号通路蛋白激活的变化,结果显示AUF1被敲低后,IL-17信号通路中的p65、ERK等蛋白的磷酸化明显下降,这提示着AUF1敲低使得IL-17信号转导受损。这些数据表明,AUF1在IL-17增强MSC基因表达中起着双重作用:AUF1对IL-17启动信号传导是必需的;而在IL-17与其他细胞因子协同作用时,AUF1同样又是IL-17的靶标,IL-17通过降低AUF1的水平来促进相关mRNA的稳定性。因此,本发明人首次发现了AUF1在介导IL-17增强免疫抑制基因表达中的重要作用,为进一步阐明IL-17作用的分子机制奠定了基础。The present inventors have shown that knockdown of AUF1 can promote the expression of iNOS induced by IFNγ and TNFα in MSC, suggesting the importance of AUF1 for regulation of gene expression in MSC. To further reveal the molecular mechanism by which IL-17 up-regulates MSC gene expression, the inventors examined the effect of IL-17 on the stability of iNOS mRNA, and showed that IL-17 significantly enhanced the stability of iNOS mRNA induced by IFNγ and TNFα. When AUF1 is absent or knocked down, IL-17 does not synergize IFNγ and TNFα to enhance iNOS gene expression, and does not further enhance iNOS mRNA stability. In vitro immunosuppressive experiments showed that auf1 -/- MSCs only required the presence of IFNγ and TNFα to exert the strongest immunosuppressive ability and did not require IL-17 supplementation. The present inventors further studied the specific mechanism of AUF1 gene expression in IL-17-enhanced MSCs, and found that MSCs stimulated by IFNγ and TNFα, IL-17 addition can reduce the expression level of AUF1, and this discovery reveals AUF1 involvement. IL-17 regulates the expression of MSC-related genes, that is, IL-17 promotes the stability of iNOS mRNA by decreasing the level of AUF1. The results of co-immunoprecipitation also showed that the interaction between AUF1 and Act1 was significantly enhanced 15 min after IL-17 stimulation. The present inventors also studied the changes in IL-17 signaling pathway protein activation in AUF1 knockdown MSCs, and the results showed that phosphorylation of p65, ERK and the like in the IL-17 signaling pathway was significantly decreased after AUF1 was knocked down, suggesting that AUF1 Knockdown caused impaired IL-17 signal transduction. These data indicate that AUF1 plays a dual role in IL-17-enhanced MSC gene expression: AUF1 is required for IL-17 initiation signaling; whereas when IL-17 synergizes with other cytokines, AUF1 is also IL- The target of 17, IL-17, promotes the stability of related mRNA by decreasing the level of AUF1. Therefore, the present inventors have found for the first time that AUF1 plays an important role in mediating IL-17 enhancing the expression of immunosuppressive genes, and lays a foundation for further elucidating the molecular mechanism of IL-17 action.
综上,本发明人的研究第一次揭示了IL-17可以增强MSC的免疫抑制功能。IL-17的这一作用主要是通过逆转RNA结合蛋白AUF1降解mRNA的作用来实现的。本发明人相信IL-17调节MSC免疫抑制的深入研究将有利于MSC更好地应用于临床。In summary, the inventors' research revealed for the first time that IL-17 can enhance the immunosuppressive function of MSC. This effect of IL-17 is mainly achieved by reversing the degradation of mRNA by the RNA binding protein AUF1. The present inventors believe that an in-depth study of IL-17 regulation of MSC immunosuppression will facilitate the better application of MSCs in clinical practice.
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。 All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the In addition, it should be understood that various modifications and changes may be made by those skilled in the art in the form of the appended claims.

Claims (11)

  1. 一种白介素-17、白介素-17的衍生物或其激动剂的用途,其特征在于,用于制备制剂或试剂盒,所述制剂或试剂盒用于:Use of a derivative of interleukin-17, interleukin-17 or an agonist thereof, for use in the preparation of a preparation or kit for:
    (1)提高间充质干细胞的免疫抑制功能;和/或(1) improving the immunosuppressive function of mesenchymal stem cells; and/or
    (2)上调MSC中免疫抑制因子的表达;和/或(2) up-regulating the expression of immunosuppressive factors in MSC; and/or
    (3)增强MSC中免疫抑制因子的mRNA的稳定性;和/或(3) enhancing the stability of mRNA of immunosuppressive factors in MSC; and/or
    (4)降低RNA结合蛋白AUF1的表达水平;和/或(4) reducing the expression level of the RNA binding protein AUF1; and/or
    (5)抑制T细胞的增殖;和/或(5) inhibiting the proliferation of T cells; and/or
    (6)治疗肝炎或肝损伤。(6) Treatment of hepatitis or liver damage.
  2. 如权利要求1所述的用途,其特征在于,所述白介素-17是哺乳动物的白介素-17。The use according to claim 1, wherein the interleukin-17 is a mammalian interleukin-17.
  3. 一种白介素-17拮抗剂的的用途,其特征在于,用于制备制剂或试剂盒,所述制剂或试剂盒用于:Use of an interleukin-17 antagonist for the preparation of a formulation or kit for:
    (1)降低间充质干细胞的免疫抑制功能;和/或(1) reducing the immunosuppressive function of mesenchymal stem cells; and/or
    (2)下调MSC中免疫抑制因子的表达;和/或(2) down-regulating the expression of immunosuppressive factors in MSC; and/or
    (3)降低MSC中免疫抑制因子的mRNA的稳定性;和/或(3) reducing the stability of mRNA of immunosuppressive factors in MSC; and/or
    (4)增强RNA结合蛋白AUF1的表达水平;和/或(4) enhancing the expression level of the RNA binding protein AUF1; and/or
    (5)促进T细胞的增殖。(5) Promote the proliferation of T cells.
  4. 一种组合物,其特征在于,所述组合物包括白介素-17或其衍生物、IFNγ和TNFα。A composition comprising interleukin-17 or a derivative thereof, IFNγ and TNFα.
  5. 一种分离的MSC细胞群,其特征在于,所述MSC细胞群由MSC细胞构成或基本上由MSC细胞构成,并且所述的MSC细胞具有增强的抑制T细胞增殖的能力,An isolated MSC cell population characterized in that the MSC cell population consists of or consists essentially of MSC cells, and the MSC cells have enhanced ability to inhibit T cell proliferation,
    并且,所述的MSC细胞选自下组:And, the MSC cells are selected from the group consisting of:
    (1)体外经预处理的MSC细胞群,其中所述预处理指用(i)白介素-17或其衍生物、(ii)IFNγ和(iii)TNFα同时、依次或先后进行处理;(1) an in vitro pretreated MSC cell population, wherein the pretreatment refers to simultaneous, sequential or sequential treatment with (i) interleukin-17 or a derivative thereof, (ii) IFNγ and (iii) TNFα;
    (2)Act1蛋白过表达和/或AUF1蛋白缺失或活性降低的细胞群;和(2) a cell population in which Act1 protein is overexpressed and/or AUF1 protein is deleted or reduced in activity;
    (3)体外经预处理的MSC细胞群,其中所述预处理指用IFNγ和TNFα同时、依次或先后进行处理;(3) an in vitro pretreated MSC cell population, wherein the pretreatment refers to simultaneous, sequential or sequential treatment with IFNγ and TNFα;
    (4)组(1)、组(2)和组(3)的组合。(4) A combination of group (1), group (2), and group (3).
  6. 一种遗传改造的细胞株,其特征在于,所述细胞株是经基因工程改造从而导致内源的Act1蛋白过表达和/或AUF1蛋白缺失或活性降低。A genetically engineered cell strain characterized in that the cell strain is genetically engineered to result in overexpression of an endogenous Act1 protein and/or a decrease in AUF1 protein or activity.
  7. 一种分离的蛋白复合物,其特征在于,所述蛋白复合物为Act1蛋白和AUF1蛋白结合的蛋白复合物。An isolated protein complex characterized in that the protein complex is a protein complex that binds to the Act1 protein and the AUF1 protein.
  8. 如权利要求7所述蛋白复合物的应用,其特征在于,所述用途为用于筛选药物或化合物,所述药物或化合物促进或抑制Act1蛋白和AUF1蛋白形成所述的复合物。Use of a protein complex according to claim 7, characterized in that the use is for screening a drug or a compound which promotes or inhibits the formation of the complex by the Act1 protein and the AUF1 protein.
  9. 一种试剂盒,其特征在于,所述试剂盒中含有以下组分:A kit characterized in that the kit contains the following components:
    (a)白介素-17或其衍生物;(a) interleukin-17 or a derivative thereof;
    (b)IFNγ;和(b) IFNγ; and
    (c)TNFα;(c) TNFα;
    以及使用说明书,And instructions for use,
    其中,所述的组分(a)、(b)和(c)分别位于一个或多个不同的容器或位于同一容器中。Wherein the components (a), (b) and (c) are respectively located in one or more different containers or in the same container.
  10. 一种药盒,其特征在于,所述药盒中包括如权利要求4所述的药物组合物和说明书,所述说明书中记载该药物组合物用于:A kit comprising the pharmaceutical composition according to claim 4 and instructions for describing the pharmaceutical composition for:
    (1)提高间充质干细胞的免疫抑制功能;和/或(1) improving the immunosuppressive function of mesenchymal stem cells; and/or
    (2)上调MSC中免疫抑制因子的表达;和/或(2) up-regulating the expression of immunosuppressive factors in MSC; and/or
    (3)增强免疫抑制因子的mRNA的稳定性;和/或(3) enhancing the stability of mRNA of an immunosuppressive factor; and/or
    (4)降低RNA结合蛋白AUF1的表达水平;和/或 (4) reducing the expression level of the RNA binding protein AUF1; and/or
    (5)抑制T细胞的增殖;和/或(5) inhibiting the proliferation of T cells; and/or
    (6)治疗肝炎或肝损伤。(6) Treatment of hepatitis or liver damage.
  11. 一种治疗肝炎或肝损伤的方法,其特征在于,所述方法包括步骤:A method of treating hepatitis or liver damage, characterized in that the method comprises the steps of:
    给需要的对象施加治疗有效量的MSC细胞。 A therapeutically effective amount of MSC cells is administered to the subject in need thereof.
PCT/CN2015/077862 2014-05-14 2015-04-29 Uses of il-17 in enhancing immune-suppression function of mesenchymal stem cells WO2015172659A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201410203949.1 2014-05-14
CN201410203949 2014-05-14

Publications (1)

Publication Number Publication Date
WO2015172659A1 true WO2015172659A1 (en) 2015-11-19

Family

ID=54479314

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/077862 WO2015172659A1 (en) 2014-05-14 2015-04-29 Uses of il-17 in enhancing immune-suppression function of mesenchymal stem cells

Country Status (2)

Country Link
CN (1) CN105079792A (en)
WO (1) WO2015172659A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022241090A1 (en) * 2021-05-13 2022-11-17 Primegen Us, Inc. Methods and compositions for treating liver disease

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105671000A (en) * 2016-03-02 2016-06-15 深圳爱生再生医学科技有限公司 Recombinant mesenchymal stem cells, preparation method and application thereof
WO2018170390A1 (en) * 2017-03-17 2018-09-20 Senti Biosciences, Inc. Immunomodulating cell circuits
CN108424914B (en) * 2018-05-02 2020-05-12 中国医科大学附属盛京医院 Targeted AUF1 gene inhibitor for inhibiting human malignant brain glioma and application thereof
CN109628406B (en) * 2019-01-03 2021-02-26 北京贝来生物科技有限公司 Mesenchymal stem cell for treating autoimmune disease and preparation method and application thereof
CN112402454A (en) * 2019-08-22 2021-02-26 中国科学院上海营养与健康研究所 Application of pre-activated mesenchymal stem cells in immunobiology for treating skin wounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070248577A1 (en) * 2002-03-15 2007-10-25 Phillips Catherine A Methods and compositions for administering stem cells
WO2014093948A1 (en) * 2012-12-14 2014-06-19 Rutgers, The State University Of New Jersey Methods modulating immunoregulatory effect of stem cells

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070248577A1 (en) * 2002-03-15 2007-10-25 Phillips Catherine A Methods and compositions for administering stem cells
WO2014093948A1 (en) * 2012-12-14 2014-06-19 Rutgers, The State University Of New Jersey Methods modulating immunoregulatory effect of stem cells

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GABR, M. A. ET AL.: "Interleukin-17 Synergizes with IFN Y or TNF A to Promote Inflammatory Mediator Release and Intercellular Adhesion Molecule- (ICAM-1) Expression in Human Intervertebral Disc Cells", JOURNAL OF ORTHOPAEDIC RESEARCH, vol. 29, no. 1, 31 January 2011 (2011-01-31), pages 1 - 7, XP055236241 *
HAN, X. ET AL.: "Interleukin-17 Enhances Immunosuppression by Mesenchymal Stem Cells", CELL DEATH AND DIFFERENTIATION, vol. 21, 18 July 2014 (2014-07-18), pages 1758 - 1768, XP055236238 *
SHOU, PEISHUN ET AL.: "Mechanism of Immunosuppression of Mesenchymal Stem Cells and the Application in Diseases", CHINESE JOURNAL OF CELL BIOLOGY, vol. 31, no. 1, 31 December 2009 (2009-12-31), pages 15 - 20 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022241090A1 (en) * 2021-05-13 2022-11-17 Primegen Us, Inc. Methods and compositions for treating liver disease

Also Published As

Publication number Publication date
CN105079792A (en) 2015-11-25

Similar Documents

Publication Publication Date Title
WO2015172659A1 (en) Uses of il-17 in enhancing immune-suppression function of mesenchymal stem cells
KR102188605B1 (en) Methods modulating immunoregulatory effect of stem cells
Haak et al. IL-17A and IL-17F do not contribute vitally to autoimmune neuro-inflammation in mice
Liu et al. IL-17 is a potent synergistic factor with GM-CSF in mice in stimulating myelopoiesis, dendritic cell expansion, proliferation, and functional enhancement
CN109810947B (en) Mesenchymal stem cell for inhibiting activation of Th17 cell and preparation method and application thereof
EP2658566A1 (en) Sirna against cbl-b and optionally il2 und il12 for use in the treatment of cancer
US9358285B2 (en) Granulysin in immunotherapy
CN112843222B (en) Application of ANKRD22 protein in preparing product for treating or delaying autoimmune diseases
US20180200301A1 (en) Low-Oxygen-Treated Mesenchymal Stem Cell and Use Thereof
EP3400074B1 (en) Compositions for generating immunotolerant responses
CN111979199A (en) Uterine blood stem cells and exosomes for treating intrauterine adhesions
Hu et al. Human umbilical cord-derived mesenchymal stem cells alleviate acute lung injury caused by severe burn via secreting TSG-6 and inhibiting inflammatory response
US20190381147A1 (en) Pharmaceutical composition containing insulin-like growth factor-2 and use thereof
US20220062383A1 (en) Regulation of a foreign body response
EP2687224A1 (en) Medicament for wound treatment
Cramer et al. Transcriptomic Regulation of Macrophages by Matrix-Bound Nanovesicle-Associated Interleukin-33
WO2023019913A1 (en) Inducer, macrophage and use thereof
WO2015090223A1 (en) Protein and use thereof in treating multiple sclerosis
KR101673318B1 (en) Cell therapy composition for healing wounds comprising mesenchymal stem cell or the culture medium treated with silver nano particle
TWI782355B (en) Composition and use of interleukin stimulated human umbilical cord mesenchymal stem cells for the treatment of rheumatoid arthritis
CN110590929B (en) Application of TDGF-1 truncated body small molecule polypeptide in anti-hepatic fibrosis
Nguyen et al. Cellular and Molecular Mechanisms of Hypertrophic Scarring
Liu et al. VEGF 165 attenuates the Th17/Treg imbalance that exists when transplanting allogeneic skeletal myoblasts to treat acute myocardial infarction
CN112402454A (en) Application of pre-activated mesenchymal stem cells in immunobiology for treating skin wounds
WO2024011235A1 (en) Methods and compositions for improving wound healing

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15792795

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 27.02.2017)

122 Ep: pct application non-entry in european phase

Ref document number: 15792795

Country of ref document: EP

Kind code of ref document: A1