WO2015172659A1 - Utilisations de l'il -17 dans l'amélioration de la fonction d'immuno-suppression des cellules souches mésenchymateuses - Google Patents

Utilisations de l'il -17 dans l'amélioration de la fonction d'immuno-suppression des cellules souches mésenchymateuses 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
English (en)
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/fr

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.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne des utilisations de l'IL-17 dans l'amélioration d'une fonction d'immuno-suppression des cellules souches mésenchymateuses. En particulier, la présente invention concerne l'utilisation d'une interleukine -17, d'un dérivé de l'interleukine-17 ou d'un agoniste de ceux-ci pour la préparation d'une préparation ou d'une trousse pour améliorer une fonction d'immuno-suppression des cellules souches mésenchymateuses; pour réguler à la hausse l'expression de facteurs immunosuppresseurs dans les cellules souches mésenchymateuses; pour améliorer la stabilité d'ARNm des facteurs immunosuppresseurs; pour réduire le taux d'expression de protéine de liaison AUF1 à l'ARN; pour inhiber la prolifération des cellules T; et pour traiter l'hépatite ou une lésion du foie.
PCT/CN2015/077862 2014-05-14 2015-04-29 Utilisations de l'il -17 dans l'amélioration de la fonction d'immuno-suppression des cellules souches mésenchymateuses WO2015172659A1 (fr)

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 (fr) 2015-11-19

Family

ID=54479314

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/077862 WO2015172659A1 (fr) 2014-05-14 2015-04-29 Utilisations de l'il -17 dans l'amélioration de la fonction d'immuno-suppression des cellules souches mésenchymateuses

Country Status (2)

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

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022241090A1 (fr) * 2021-05-13 2022-11-17 Primegen Us, Inc. Méthodes et compositions pour le traitement d'une hépatopathie

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105671000A (zh) * 2016-03-02 2016-06-15 深圳爱生再生医学科技有限公司 重组间充质干细胞及其制备方法和应用
CN110612110A (zh) * 2017-03-17 2019-12-24 森迪生物科学公司 免疫调节细胞回路
CN108424914B (zh) * 2018-05-02 2020-05-12 中国医科大学附属盛京医院 抑制人恶性脑胶质瘤的靶向auf1基因抑制剂及其应用
CN109628406B (zh) * 2019-01-03 2021-02-26 北京贝来生物科技有限公司 一种治疗自身免疫疾病的间充质干细胞及其制备方法和应用
CN112402454A (zh) * 2019-08-22 2021-02-26 中国科学院上海营养与健康研究所 预激活间充质干细胞在治疗皮肤创伤的免疫生物学方面的应用
CN117866905A (zh) * 2024-03-12 2024-04-12 北京贝来药业有限公司 基于纳米抗体基因修饰的干细胞及其制备方法和产品

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 (fr) * 2012-12-14 2014-06-19 Rutgers, The State University Of New Jersey Procédés permettant la modulation de l'effet immunorégulateur des cellules souches

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 (fr) * 2012-12-14 2014-06-19 Rutgers, The State University Of New Jersey Procédés permettant la modulation de l'effet immunorégulateur des cellules souches

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 (fr) * 2021-05-13 2022-11-17 Primegen Us, Inc. Méthodes et compositions pour le traitement d'une hépatopathie

Also Published As

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

Similar Documents

Publication Publication Date Title
WO2015172659A1 (fr) Utilisations de l'il -17 dans l'amélioration de la fonction d'immuno-suppression des cellules souches mésenchymateuses
KR102188605B1 (ko) 줄기세포의 면역조절성 효과의 조절 방법
Haak et al. IL-17A and IL-17F do not contribute vitally to autoimmune neuro-inflammation in mice
Wang et al. Interleukin-25 mediates transcriptional control of PD-L1 via STAT3 in multipotent human mesenchymal stromal cells (hMSCs) to suppress Th17 responses
Schulze-Tanzil et al. Interleukin-10 and articular cartilage: experimental therapeutical approaches in cartilage disorders
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
US9855330B2 (en) Granulysin in immunotherapy
CN109810947B (zh) 一种抑制Th17细胞活化的间充质干细胞及其制备方法和应用
WO2012089736A1 (fr) ARNsi DIRIGÉ CONTRE CB1-B ET OPTIONNELLEMENT IL2 ET IL12 À UTILISER DANS LE TRAITEMENT DU CANCER
JP7033066B2 (ja) 免疫寛容応答を生成する組成物及び方法
US20180200301A1 (en) Low-Oxygen-Treated Mesenchymal Stem Cell and Use Thereof
CN112843222B (zh) Ankrd22蛋白在制备治疗或延缓自身免疫性疾病的产品中的应用
CN111979199A (zh) 用于治疗宫腔粘连的宫血干细胞和外泌体
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
EP2687224A1 (fr) Médicament pour le traitement des plaies
RU2769474C2 (ru) Способы лечения рассеянного склероза с использованием аутологичных т-клеток
WO2020150394A1 (fr) Régulation de réponse à un corps étranger
WO2015090223A1 (fr) Protéine et son utilisation pour traiter la sclérose en plaques
KR101673318B1 (ko) 은나노 물질로 처리된 중간엽 줄기세포 또는 그 배양액을 유효성분으로 포함하는 상처 치료용 세포치료제 조성물
TWI782355B (zh) 經細胞介白素預刺激之人類臍帶間質幹細胞於治療類風濕性關節炎之組成及用途
US20160206699A1 (en) Use of interleukin 10 mrna transfected macrophages in anti-inflammatory therapies
CN110590929B (zh) Tdgf-1截短体小分子多肽在抗肝纤维化中的应用
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 (zh) 预激活间充质干细胞在治疗皮肤创伤的免疫生物学方面的应用

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