CN114209843A - Inhibitors/activators associated with RELT signaling pathway and uses thereof - Google Patents

Inhibitors/activators associated with RELT signaling pathway and uses thereof Download PDF

Info

Publication number
CN114209843A
CN114209843A CN202210084094.XA CN202210084094A CN114209843A CN 114209843 A CN114209843 A CN 114209843A CN 202210084094 A CN202210084094 A CN 202210084094A CN 114209843 A CN114209843 A CN 114209843A
Authority
CN
China
Prior art keywords
relt
inhibitor
activator
immune
teff
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210084094.XA
Other languages
Chinese (zh)
Inventor
王从义
孙飞
荣善杰
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tongji Medical College of Huazhong University of Science and Technology
Original Assignee
Tongji Medical College of Huazhong University of Science and Technology
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 Tongji Medical College of Huazhong University of Science and Technology filed Critical Tongji Medical College of Huazhong University of Science and Technology
Priority to CN202210084094.XA priority Critical patent/CN114209843A/en
Publication of CN114209843A publication Critical patent/CN114209843A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • A61P5/16Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4 for decreasing, blocking or antagonising the activity of the thyroid hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics

Abstract

The invention discloses an inhibitor/activator related to a RELT signal pathway and application thereof, belonging to the technical field of immunotherapy. The inhibitor/activator is a substance that affects RELT function and/or RELT expression, wherein the inhibitor reduces Teff activity and enhances Treg function by blocking a RELT-related signaling pathway, and the activator promotes Teff activity and attenuates Treg function by activating a RELT-related signaling pathway. The invention lays a foundation for the targeted prevention and/or treatment of autoimmune diseases, or immune activation, and the treatment of other tumor-related diseases.

Description

Inhibitors/activators associated with RELT signaling pathway and uses thereof
Technical Field
The invention relates to an inhibitor/activator related to a RELT signaling pathway, belongs to the technical field of immunotherapy, and particularly relates to an inhibitor/activator related to a RELT signaling pathway and application thereof.
Background
From the perspective of cellular function, CD4+T cells can be divided into two broad categories, namely effector T cells: effect T cells, Teff for short: after receiving antigen stimulation, the medicine can proliferate, differentiate and release cytokines, such as interleukins, interferons and the like, exert immune effect by enhancing the interaction of various cells, and resist or eliminate exogenous and endogenous pathogenic factors (as recorded in PMID: 24009159); and regulatory T cells: regulation T cells, Treg for short: a subset of T cells with significant immunosuppressive effects can be contacted or non-contacted to limit the effector functions of other immune cells and avoid over-activation of the immune system (as described in PMID: 30714682). The balance of Teff/Treg is the key to ensure a moderate immune response and to maintain immune homeostasis. As the main antigen presenting cells, dendritic cells: dendritic cells, abbreviated DC, are believed to regulate CD4+The core role of Th cell responses (as described in PMID: 32433540). DCs, in addition to providing TCR ligand pMHC complexes and co-stimulatory molecules, also modulate CD4 through a variety of signals+Poles of T cellsThe state of the cells is changed, the sub-population ratio of Teff/Treg is directly influenced, and the cells play an important role in the immune homeostasis control of Teff/Treg.
RELT: receptor Expressed on lymphoma Tissues belong to the TNF Receptor superfamily: TNF Receptor Superfamily, also known as TNFRSF19L, is expressed primarily in immune cells. Like other members of the TNF receptor superfamily, RELT exists in both membrane bound and free states, and the transition from the membrane bound to the free state is achieved by extracellular domain shedding. Amino acid 137 of RELT extracellular membrane-proximal part can be cleaved and released to become soluble sRLT, which is mediated by matrix metalloprotease and disintegrin family members ADAM17, ADAM9, ADAM10, ADAM19, MMP7, Elastase, protease 3 (as described in PMID:30354964, PMID:23557269, PMID: 27192566). Studies have found a significant increase in serum free Soluble RELT (Soluble RELT) in tumor patients, suggesting that RELT may be a potential early diagnostic marker for tumors (as described in PMID: 30873760). However, the immune regulation mechanisms of RELT membrane-bound and free forms are still not well understood, and the specific roles that they mediate immune cell conversation, regulate immune system homeostasis and play in immune-related diseases remain to be resolved.
Type 1 diabetes is an autoimmune metabolic disease characterized by an absolute deficiency of insulin (as described in PMID: 29916386) due to a disturbance in the body's immune system that attacks the beta cells of the islets of Langerhans, leading to massive beta cell death. Numerous studies have shown that an imbalance in Treg and Teff homeostasis is a key feature in the development and progression of type 1 diabetes (as documented by PMID: 28770318). In the pathological process of T1D, Teff cells are the main cell population that damages beta cells, inducing an autoimmune response. Teff can directly secrete inflammatory cytokines such as IFN-gamma, TNF-alpha and IL-1 beta and other soluble mediators such as nitric oxide to induce beta cell death, and can also activate CD8+T cells release granzyme, perforin or kill beta cells via FAS-mediated cytolysis (PMID: 24054837). Tregs are normally able to maintain immune homeostasis by inhibiting Teff proliferation, differentiation and cytokine secretion (as described in PMID: 29155454); in patients with T1D, Teff/Treg is disbalanced in function, with its Teff being overactivated,The proportion of Treg cells decreases and Treg cells often exhibit a degree of dysfunction, thereby exacerbating disease progression.
Malignant tumors, as the second largest killer threatening human health, remain a great challenge for modern medical research. Studies have shown that the tumor microenvironment: the Tumor immunity is shortened to TME, the number of Treg cells in the Tumor immunity is obviously increased, the infiltration of Teff is obviously reduced, cells of myeloid origin such as DC and macrophage present unique tolerant phenotype, and the immune escape of the Tumor is promoted together (as recorded in PMID: 25728990). Treg cells are recruited to tumor tissues via chemokine receptor-chemokine interactions, such as the CCR4-CCL17/22, CCR8-CCL1, CCR10-CCL28, and CXCR3-CCL9/10/11 axes; treg cells subsequently suppress the anti-tumor immune response of Teff cells by competitively binding IL-2, secreting suppressive cytokines, regulating tryptophan and adenosine metabolism, etc. (as described in PMID: 31102428). The imbalance in Teff/Treg immune homeostasis in the Tumor Microenvironment (TME) is closely associated with the sustained progression and low survival of various tumors. Immunotherapy of tumors as an innovative therapeutic approach, specifically blocks inhibitory or amplifies stimulatory signals through molecular targeting, thereby correcting the steady state imbalance of Teff/tregs, which has become a leading hotspot in the tumor field.
In addition, Teff/Treg immune homeostasis imbalance is an important mechanism for the development of immune-related diseases such as rheumatoid arthritis, systemic lupus erythematosus, sjogren's syndrome, multiple sclerosis, hashimoto's thyroiditis, primary myxoedema, hyperthyroidism, autoimmune hemolytic anemia, ulcerative colitis, atrophic gastritis, and autoimmune glomerulonephritis.
In view of the above, there is an urgent need in numerous clinical scenarios to treat immune-related diseases by modulating Teff/Treg immune homeostasis.
Disclosure of Invention
In order to solve the technical problems, the invention discloses an inhibitor/activator related to a RELT signal pathway and application thereof. It lays the foundation for the target prevention and/or treatment of autoimmune diseases, or immune activation, and the treatment of other tumor-related diseases.
In order to achieve the above technical objects, the present invention discloses an inhibitor/activator, which is a substance affecting RELT function and/or RELT expression and used for regulating Teff/Treg immune homeostasis, wherein the inhibitor reduces Teff activity and enhances Treg function by blocking a RELT-related signaling pathway, and the activator promotes Teff activity and attenuates Treg function by activating a RELT-related signaling pathway. Immune homeostasis here refers in particular to the balance of cell number, cell activity, differentiation, etc., of Teff/Treg.
Further, the inhibitor is selected from any one or more of siRNA, shRNA, miRNA or neutralizing antibody which takes RELT as a target, sRLT recombinant protein and other chemical inhibitors. Preferably, the inhibitor is selected from sRELT recombinant proteins and/or neutralizing antibodies targeting RELT. Wherein, the sRLT recombinant protein is obtained by expressing and purifying an RELT extracellular segment ligand binding domain in vitro. The neutralizing antibody taking RELT as a target is obtained by immunizing a rabbit with RELT, sorting antigen-specific B cells in a flow mode, sequencing BCR, recombining and expressing in vitro, and performing ELISA high-throughput screening, WB detection and functional test verification.
Further, the activator is a mimic activator of RELT, and the mimic activator of RELT is selected from any one or more of Fc fusion protein of RELT, RELT agonist antibody, recombinant plasmid for over-expressing RELT, viral vector for over-expressing RELT, immune cell for over-expressing RELT, or other chemical agonist. Preferably, the activator is selected from the group consisting of an Fc-RELT fusion protein and/or a recombinant plasmid overexpressing RELT, a viral vector overexpressing RELT, an immune cell overexpressing RELT, and the like. The Fc-RELT fusion protein is obtained by fusing a RELT extracellular domain ligand binding domain with an Fc segment of IgG2, and can activate a reverse signal of RELT ligand receptor action so as to promote downstream immune activation. The over-expression RELT is obtained by constructing a CD 4T cell specific RELT over-expression mouse model, and the RELT mediated immune activation effect can be detected in vivo.
The invention also aims to disclose and protect the application of the inhibitor/activator in preparing the medicine for preventing and/or treating autoimmune diseases. In particular, T cell activation is inhibited by inhibiting RELT-related signaling pathways.
Further, the autoimmune disease includes type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, sjogren's syndrome, multiple sclerosis, hashimoto's thyroiditis, primary myxedema, hyperthyroidism, autoimmune hemolytic anemia, ulcerative colitis, atrophic gastritis, autoimmune glomerulonephritis.
Further, the present invention preferably relates to the use of the above inhibitor/activator for the preparation of a medicament for the prevention and/or treatment of type 1 diabetes.
Further, the present invention preferably relates to the use of the above inhibitor/activator for the preparation of a medicament for the prevention and/or treatment of rheumatoid arthritis.
The invention also discloses the application of the inhibitor/activator in preparing a medicine for immune activation and/or treating tumor-related diseases. In particular, by activating RELT-related signaling pathways to promote immune activation and anti-tumor responses.
The fourth object of the present invention is to disclose the use of the above inhibitor/activator for the preparation of a pharmaceutical composition for modulating a signal transduction pathway associated with RELT.
The fifth purpose of the invention is to disclose and protect a pharmaceutical composition, which is used for regulating and controlling a signal transduction pathway related to RELT.
Further, the pharmaceutical composition comprises the inhibitor/activator, and a pharmaceutically acceptable carrier.
Has the advantages that: the invention firstly uses recombinant protein technology, flow cytometry and related technology to clarify the adjustable Teff/Treg immune homeostasis of RELT and mediate the occurrence and development of immune-related diseases, and secondly proves that the inhibition of RELT function can treat and prevent type 1 diabetes, and the enhancement of RELT function can promote immune activation and anti-tumor response. Therefore, the RELT can be a brand-new target for preventing and treating diseases related to immune disorder, and a new choice and a wide application prospect are provided for preparing a therapeutic drug or a preparation aiming at various causes and few side effects.
Drawings
FIG. 1 is a schematic diagram of the construction of sRLT recombinant proteins and Fc-RELT fusion proteins. Wherein FIG. 1A is the construction of sRLT recombinant protein, and FIG. 1B is the construction of Fc-RELT fusion protein.
Fig. 2 is a schematic representation of sRELT recombinant protein inhibiting Th1 differentiation and enhancing Treg stability.
FIG. 3 is a schematic diagram showing the effect of sRLT recombinant protein on the prevention and treatment of spontaneous diabetes in NOD mice. Wherein, fig. 3A is a graph of incidence of intervention type 1 diabetes after administration of NOD mice BSA or sRELT, respectively, fig. 3B is activation marker molecular flow staining of CD 4T cells, fig. 3C is mouse spleen Treg cell flow staining, and fig. 3D is mouse spleen Teff cell flow staining.
Fig. 4 is a schematic diagram of a process for preparing a RELT neutralizing antibody.
FIG. 5 is a preliminary evaluation graph of the immunosuppressive effects of RELT neutralizing antibodies. Wherein, fig. 5A is a flow test of effectiveness of the RELT antiserum in rabbits after four immunizations, and fig. 5B is a functional test of the RELT antiserum in inhibiting CD 4T cell activation.
Fig. 6 is a schematic diagram of the promotion of T cell activation by RELT overexpression. Wherein, fig. 6A is the proportion of Treg cells in CD 4T cell-specific RELT overexpressing mice, and fig. 6B is the proportion of Teff cells in CD 4T cell-specific RELT overexpressing mice.
FIG. 7 shows Fc-RELT fusion protein pairs
Figure BDA0003481817630000051
Schematic representation of the effects of T cell activation, proliferation and differentiation. Among them, FIG. 7A shows Fc-RELT promotion
Figure BDA0003481817630000061
T cell activation, FIG. 7B shows that Fc-RELT promotes polarization of Th1 cells, and FIG. 7C shows that Fc-RELT inhibits differentiation of Treg cells.
FIG. 8 is a schematic diagram of Fc-RELT fusion protein mediating the progression of spontaneous diabetes in NOD mice. Wherein, FIG. 8A is a curve showing the incidence rate of type 1 diabetes after NOD mouse isotype IgG or Fc-RELT stem cells are respectively administered, FIG. 8B is the activation marker molecular flow staining of CD 4T cells, FIG. 8C is the flow staining of mouse spleen Treg cells, and FIG. 8D is the flow staining of mouse spleen Teff cells.
FIG. 9 is a schematic representation of the tumor tissue growth process after administration of Fc-RELT fusion protein.
FIG. 10 is a schematic representation of Fc-RELT fusion proteins promoting T cell activation in tumor tissue. Wherein, FIG. 10A shows the activation status of CD 4T cells and the proportion of Treg cells infiltrating into the tumor tissue, and FIG. 10B shows the activation status of CD 8T cells and IFN-g positive effect CD8+Proportion of T cells.
Detailed Description
The present invention will now be described more fully hereinafter with reference to the accompanying drawings, in which some, but not all embodiments of the invention are shown. Based on the embodiments of the present invention, those skilled in the art may make various changes, modifications, substitutions and alterations without creative efforts, and still fall within the protection scope of the present invention.
The experimental methods used in the following experimental examples are all conventional methods unless otherwise specified; the materials, reagents and the like used are, unless otherwise specified, commercially available reagents and materials. Throughout the specification, unless otherwise specifically noted, terms used herein should be understood as having meanings as commonly used in the art. Accordingly, unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In case of conflict, the present definition takes precedence.
The medicaments or pharmaceutical compositions of the present invention may be formulated for administration by a variety of routes including, but not limited to, oral, subcutaneous, intramuscular, intraperitoneal, intravenous and airway injection and topical administration.
The pharmaceutical composition can be delivered to a subject in need thereof by any of several routes known to those skilled in the art. As non-limiting examples, the composition may be delivered orally, intravenously, intraperitoneally, by infusion, e.g., bolus infusion, subcutaneously, enterally, rectally, intranasally, inhalationally, buccally, sublingually, intramuscularly, transdermally, intradermally, topically, intraocularly, vaginally, rectally, or by intracranial injection, or any combination thereof.
RELT inhibitors/mimic activators can be administered subcutaneously or intravenously for type 1 diabetes or neoplastic disease. In addition, administration of RELT inhibitors/mimic activators may be combined with other pharmaceutical or non-pharmaceutical therapies. For example, in the treatment of type 1 diabetes in combination with insulin, insulin analogs, metformin, glucosidase inhibitors, GLP-1, low dose interleukin 2 and other immunosuppressive agents such as tacrolimus and the like; in the treatment of neoplastic diseases, radiotherapy, chemotherapy and other immune checkpoint blockade therapies such as anti-PD1, anti-CTLA4, etc. are combined.
In general, the RELT inhibitors/mimic activators of the invention are provided in amounts of 10 milligrams per kilogram body weight to 100 milligrams per kilogram body weight. For example, for the treatment of type 1 diabetes, RELT inhibitors can be administered at 10-100 mg/kg by intravenous or subcutaneous injection; for the treatment of neoplastic diseases, 10-100 mg/kg of RELT mimic activator can be administered by intravenous or subcutaneous injection.
The RELT inhibitor/mimetic activators of the present invention can be administered to a subject in combination with a pharmaceutically acceptable carrier or excipient. The pharmaceutically acceptable carrier or excipient may be any material that, when combined with the RELT inhibitor/mimetic activator of the present invention, allows the ingredient to retain biological activity. Examples include, but are not limited to, any standard physiologically acceptable carrier, such as phosphate buffered saline solution, water, emulsions, and various types of wetting agents. Preferred diluents for aerosol or parenteral administration are phosphate buffered saline or normal saline. Compositions comprising such carriers are formulated by well-known conventional methods.
The following examples are not intended to limit the scope of the present invention, but also discuss the mechanism associated with the present invention.
Mice were treated according to the "rules for administration of animal care for medical experimentation". NOD mice, C57BL/6 mice, were maintained in SPF-grade animal facilities.
The following examples are mainly applied to mice, but may also include other subjects such as humans.
Example 1
The sRLT recombinant protein has the functions of preventing and treating the spontaneous diabetes of NOD mice: the specific exploration process is as follows:
NOD mice at 8 weeks of age were given sRELT (n ═ 6) or BSA (n ═ 6) (10mg per kg body weight, approximately 100ug per mouse), intraperitoneally, three times a week for a total of two weeks, and then discontinued. Blood glucose was measured weekly, with blood glucose levels above 12.8mM twice in a row being considered to develop type 1 diabetes; when the incidence rate of one group of mice reaches 80%, the two groups of mice are euthanized, spleens are taken, single cell suspensions are prepared by grinding, and related indexes of immune activation are detected. As shown in fig. 3A, BSA control mice developed initial disease at 12 weeks of age, with 80% incidence at 21 weeks of age; the sRELT-treated mice started to develop disease at 17 weeks of age, with a prevalence rate of only 20% at 21 weeks of age, demonstrating that sRELT has prophylactic and therapeutic effects on diabetes mellitus spontaneously in NOD mice. As shown in FIG. 3B, activated CD44 in mice+CD62L-There was no substantial difference in T cell ratios. However, as can be seen from FIG. 3C, after sRELT treatment, Foxp3 was present in mice+The proportion of Treg cells was increased, and as can be seen from FIG. 3D, IFN-g was present in the mice after sRLT treatment+The proportion of Teff cells was reduced, suggesting that the sRELT recombinant protein can regulate the immune homeostasis of Teff/Treg.
As can be seen from FIG. 1A, the sRELT recombinant protein was obtained by in vitro expression and purification of the RELT extracellular domain ligand binding domain.
Fig. 2 is a schematic diagram of sRELT recombinant protein inhibiting Th1 differentiation and enhancing Treg stability, and it can be known from fig. 2 that sRELT can inhibit Th1 differentiation without affecting Treg differentiation, and meanwhile, sRELT can also enhance Treg stability. The sRLT recombinant protein can therefore modulate Teff/Treg immune homeostasis.
Example 2
RELT antisera can inhibit T cell activation: the specific exploration process is as follows:
CD 4T cell specificity RELT transgenic mice for experiments are euthanized, spleens are taken, single cell suspension is prepared through grinding, and magnetic bead sorting is carried out to obtain RELT over-expression CD 4T cells, wherein preferably the CD 4T cells of the Meitian whirly mice are sorted into magnetic beads and incubated with PBS blank samples shown in figure 5A, FITC labeled anti-rabbit secondary antibodies or rabbit anti-RELT serum and FITC labeled anti-rabbit secondary antibodies respectively, and flow detection proves that antibodies in the rabbit anti-RELT serum can identify and bind to the CD 4T cells. To test the ability of rabbit anti-RELT serum to specifically inhibit CD 4T cell activation, we coated cell culture plates with TCR (α CD 33 ug/ml + α CD 283 ug/ml), TCR + RELT (α CD 33 ug/ml + α CD 283 ug/ml + Fc-RELT) as shown in fig. 5B, inoculated sorted CD 4T cells for 12 hours, one of which was added to 10 microliters of rabbit anti-RELT serum (TCR + RELT + α RELT), and tested for expression of the T cell activation markers CD25 and CD 69. The results show that the addition of rabbit anti-RELT serum significantly inhibits T cell activation.
Fig. 4 shows a flow chart of preparing RELT-neutralizing antibodies.
Example 3
CD 4T cell-specific RELT transgenic mice display an immune-activated phenotype: the specific exploration process is as follows:
the CD 4T cell specific RELT transgenic mice (RELT) for experiments at 10-12 weeks of ageCD4-TG) And a wild type control mouse (Ctrl) is euthanized, the spleen is taken, and single cell suspension is prepared by grinding and related indexes of immune activation are detected. Shown in FIG. 6A, RELTCD4-TGFoxp3 in mice+Treg cell proportion was significantly reduced, as shown in FIG. 6B, IFN-g+The Teff cell ratio was significantly increased, suggesting that CD 4T cell-specific RELT transgenic mice exhibit an immune-activated phenotype.
Example 4
The Fc-RELT fusion protein promotes disease progression in NOD mice in spontaneous diabetes: the specific exploration process is as follows:
NOD mice at 8 weeks of age were administered Fc-RELT (n ═ 12) or control IgG (n ═ 13) (10mg per kg body weight, approximately 100ug per mouse), intraperitoneally, three times a week for a total of two weeks, and then discontinued. Blood glucose was measured weekly, with blood glucose levels above 12.8mM twice in a row being considered to develop type 1 diabetes; when the incidence rate of one group of mice reaches 80%, the two groups of mice are euthanized, spleens are taken, single cell suspensions are prepared by grinding, and related indexes of immune activation are detected. As shown in fig. 8A, IgG control mice began to develop disease at 12 weeks of age, with a 30% incidence rate at 20 weeks of age;
the onset of the mice in the Fc-RELT treatment group is started at 11 weeks of age, the onset time is advanced, and the incidence rate reaches 80% at 20 weeks of age, which proves that the Fc-RELT has the promotion effect on the spontaneous diabetes of NOD mice. As can be seen from FIG. 8B, the activated CD44 was found in mice treated with Fc-RELT+CD62L-The T cell ratio increased, as can be seen from FIG. 8C, Foxp3+Treg cell proportion decreased, and as can be seen from FIG. 8D, IFN-g+The proportion of Teff cells was increased, suggesting that Fc-RELT fusion proteins may disrupt the immune homeostasis of Teff/Treg.
Wherein, the Fc-RELT fusion protein is obtained by fusing a RELT extracellular domain ligand binding domain with the Fc segment of IgG2 as shown in FIG. 1B.
FIG. 7 shows Fc-RELT fusion protein pairs
Figure BDA0003481817630000101
Schematic representation of the effects of T cell activation, proliferation and differentiation. As can be seen from FIG. 7, Fc-RELT promotes
Figure BDA0003481817630000102
T cell activation, Th1 cell polarization, and at the same time, inhibition of Treg cell differentiation. Therefore, the Fc-RELT fusion protein can regulate Teff/Treg immune homeostasis.
Example 5
Fc-RELT fusion protein inhibits tumor progression: the specific exploration process is as follows:
the mice were inoculated subcutaneously with 106 prostate cancer cells (PCa), given Fc-RELT (n-7) or control IgG (n-7) (10mg per kg body weight, approximately 100ug per mouse), and injected intraperitoneally three times a week until one group of tumor masses reached the ethical lower limit (approximately 1-2 cc)). Subsequently, the mice were euthanized and the tumor mass was detached, and as a result, as shown in fig. 9, it was found that the tumor tissue was significantly reduced after the Fc-RELT treatment; the tumor tissue was further minced, digested with collagenase and the immune cells were extracted from the lymphocyte isolate and subjected to flow analysis. The results are shown in FIG. 10, where FIG. 10A shows an enhanced level of intratumoral activation of CD 4T cells after Fc-RELT treatment, Foxp3+The proportion of Treg cells is reduced; FIG. 10B shows increased levels of activation of CD 8T cells, IFN-g+Increased proportion of effector CD 8T cells. The above results indicate that Fc-RELT can inhibit tumor progression by modulating the immune microenvironment.
In conclusion, sRELT recombinant proteins and/or neutralizing antibodies targeting RELT can inhibit RELT function and thus treat and prevent type 1 diabetes, and Fc-RELT fusion proteins and/or over-expressed RELT substances can enhance RELT function and further promote immune activation and anti-tumor response.
Although the examples mainly explore type 1 diabetes, for immune-related diseases such as rheumatoid arthritis, systemic lupus erythematosus, sjogren's syndrome, multiple sclerosis, hashimoto's thyroiditis, primary myxoedema, hyperthyroidism, autoimmune hemolytic anemia, ulcerative colitis, atrophic gastritis, autoimmune glomerulonephritis, Teff/Treg immune homeostasis imbalance is also an important mechanism for the development of these diseases, and inhibitors/activators that regulate Teff/Treg immune homeostasis by controlling the RELT signaling pathway can also be used for the prevention and/or treatment of the above autoimmune diseases.
Therefore, the inhibitor/activator related to the RELT signal pathway and used for regulating and controlling the immune homeostasis of Teff/Treg can become a brand-new target for preventing and treating diseases related to immune disorder, and provides a new choice and a wide application prospect for preparing a therapeutic drug or preparation aiming at various etiologies and having fewer side effects.
The above examples are merely preferred examples and are not intended to limit the embodiments of the present invention. In addition to the above embodiments, the present invention has other embodiments. All technical solutions formed by adopting equivalent substitutions or equivalent transformations fall within the protection scope of the claims of the present invention.

Claims (10)

1. An inhibitor/activator, wherein the inhibitor/activator is a substance that affects RELT function and/or RELT expression and is used to modulate Teff/Treg immune homeostasis, wherein the inhibitor decreases Teff activity and enhances Treg function by blocking a RELT-associated signaling pathway, and the activator promotes Teff activity and attenuates Treg function by activating a RELT-associated signaling pathway.
2. The inhibitor/activator according to claim 1, wherein the inhibitor is selected from any one or more of siRNA, shRNA, miRNA, or neutralizing antibody targeting RELT, or sRELT recombinant protein, other chemical inhibitor.
3. The inhibitor/activator according to claim 1, wherein the activator is a mimic activator of RELT selected from any one or more of Fc fusion protein of RELT, agonist antibody of RELT, recombinant plasmid overexpressing RELT, viral vector overexpressing RELT, immune cell overexpressing RELT, or other chemical agonist.
4. Use of an inhibitor/activator according to any one of claims 1 to 3 for the preparation of a medicament for the prevention and/or treatment of autoimmune diseases.
5. The use according to claim 4, wherein the autoimmune disease comprises type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, sjogren's syndrome, multiple sclerosis, hashimoto's thyroiditis, primary myxoedema, hyperthyroidism, autoimmune hemolytic anemia, ulcerative colitis, atrophic gastritis, autoimmune glomerulonephritis.
6. The use according to claim 5, wherein the autoimmune disease is type 1 diabetes.
7. Use of the inhibitor/activator of any one of claims 1 to 3 for the preparation of a medicament for the immune activation and/or treatment of a tumor-related disease.
8. Use of the inhibitor/activator of claims 1-3 for the preparation of a pharmaceutical composition for modulating a signal transduction pathway associated with RELT.
9. A pharmaceutical composition for modulating a signaling pathway associated with RELT.
10. The pharmaceutical composition according to claim 9, wherein the pharmaceutical composition comprises the inhibitor/activator according to any one of claims 1 to 3, and further comprises a pharmaceutically acceptable carrier.
CN202210084094.XA 2022-01-21 2022-01-21 Inhibitors/activators associated with RELT signaling pathway and uses thereof Pending CN114209843A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210084094.XA CN114209843A (en) 2022-01-21 2022-01-21 Inhibitors/activators associated with RELT signaling pathway and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210084094.XA CN114209843A (en) 2022-01-21 2022-01-21 Inhibitors/activators associated with RELT signaling pathway and uses thereof

Publications (1)

Publication Number Publication Date
CN114209843A true CN114209843A (en) 2022-03-22

Family

ID=80708721

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210084094.XA Pending CN114209843A (en) 2022-01-21 2022-01-21 Inhibitors/activators associated with RELT signaling pathway and uses thereof

Country Status (1)

Country Link
CN (1) CN114209843A (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101432306A (en) * 2006-02-13 2009-05-13 健泰科生物技术公司 Methods and compositions for targeting RELT
US20210260113A1 (en) * 2018-07-17 2021-08-26 Gero LLC Devices, methods, compositions and systems for the treatment of aging and age-related disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101432306A (en) * 2006-02-13 2009-05-13 健泰科生物技术公司 Methods and compositions for targeting RELT
US20210260113A1 (en) * 2018-07-17 2021-08-26 Gero LLC Devices, methods, compositions and systems for the treatment of aging and age-related disorders

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHOI.BK,ET AL: "RELT negatively regulates the early phase of the T cell response in mice", 《EUROPEAN JOURNAL OF IMMUNOLOGY》 *
SICA,ET AL: "RELT, a new member of the tumor necrosis factor receptor superfamily, is selectively expressed in hematopoietic tissues and activates transcription factor NF-kB", 《BLOOD》 *
朱医高等: "肿瘤坏死因子受体超家族成员在免疫系统和疾病中的研究", 《中国生物化学与分子生物学报》 *

Similar Documents

Publication Publication Date Title
Sanjabi et al. Opposing effects of TGF-β and IL-15 cytokines control the number of short-lived effector CD8+ T cells
Pallandre et al. Role of STAT3 in CD4+ CD25+ FOXP3+ regulatory lymphocyte generation: implications in graft-versus-host disease and antitumor immunity
Van Belle et al. Interleukin-21 receptor-mediated signals control autoreactive T cell infiltration in pancreatic islets
Deng et al. Targeting tumors with IL-21 reshapes the tumor microenvironment by proliferating PD-1intTim-3–CD8+ T cells
US10501537B2 (en) Methods for treating cancer
US10786532B2 (en) Method of providing cellular therapy using modified natural killer cells or T lymphocytes
Gkountidi et al. MHC Class II antigen presentation by lymphatic endothelial cells in tumors promotes intratumoral regulatory T cell–suppressive functions
Soto et al. MHC-class I-restricted CD4 T cells: a nanomolar affinity TCR has improved anti-tumor efficacy in vivo compared to the micromolar wild-type TCR
Xia et al. IL-2 augments the therapeutic efficacy of adoptively transferred B cells which directly kill tumor cells via the CXCR4/CXCL12 and perforin pathways
WO2012075291A1 (en) Compositions and methods for treating foxp3+ treg related diseases
US20220381772A1 (en) Systems and methods for evaluating nk cells
Yang et al. Adoptive therapy with amyloid-β specific regulatory T cells alleviates Alzheimer's disease
Han et al. Intratumoral electroporation of plasmid encoded IL12 and membrane-anchored anti-CD3 increases systemic tumor immunity
Okamoto et al. A genetic method specifically delineates Th1-type Treg cells and their roles in tumor immunity
EP2356222A2 (en) Foxp3 natural killer t-cells and the treatment of immune related diseases
Lu et al. Unique features of pancreatic-resident regulatory T cells in autoimmune type 1 diabetes
Qian et al. NKG2D ligand RAE1ε induces generation and enhances the inhibitor function of myeloid‐derived suppressor cells in mice
Kim et al. Soluble γc receptor attenuates anti‐tumor responses of CD8+ T cells in T cell immunotherapy
US20210369837A1 (en) Methods of treating tim-3 elevation
Wei et al. Combination therapy of HIFα inhibitors and Treg depletion strengthen the anti‐tumor immunity in mice
CN114209843A (en) Inhibitors/activators associated with RELT signaling pathway and uses thereof
CN111733154B (en) Magnetic field treated immune cells and uses thereof
Cunha et al. Infiltration of Tumors Is Regulated by T cell–Intrinsic Nitric Oxide Synthesis
Cao et al. CTRP4/interleukin-6 receptor signaling ameliorates autoimmune encephalomyelitis by suppressing Th17 cell differentiation
Hanke et al. PIAS 1 and STAT‐3 impair the tumoricidal potential of IFN‐γ‐stimulated mouse dendritic cells generated with IL‐15

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination