Nothing Special   »   [go: up one dir, main page]

CN115804772B - Application of iron death inhibitor in antiviral infection - Google Patents

Application of iron death inhibitor in antiviral infection Download PDF

Info

Publication number
CN115804772B
CN115804772B CN202211742287.6A CN202211742287A CN115804772B CN 115804772 B CN115804772 B CN 115804772B CN 202211742287 A CN202211742287 A CN 202211742287A CN 115804772 B CN115804772 B CN 115804772B
Authority
CN
China
Prior art keywords
cells
hbv
iron death
liproxstatin
cell
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.)
Active
Application number
CN202211742287.6A
Other languages
Chinese (zh)
Other versions
CN115804772A (en
Inventor
韩秋菊
李浩浩
杨莎莎
张建
李蓉晖
赵华俊
霍琛鑫
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.)
Shandong University
Original Assignee
Shandong University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shandong University filed Critical Shandong University
Priority to CN202211742287.6A priority Critical patent/CN115804772B/en
Publication of CN115804772A publication Critical patent/CN115804772A/en
Application granted granted Critical
Publication of CN115804772B publication Critical patent/CN115804772B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the technical field of medicines, and discloses a pharmaceutical composition for resisting viral infection and/or reducing the content of viruses, which comprises Liproxstatin-1 or pharmaceutically acceptable salt thereof and pharmaceutical excipients. The pharmaceutical composition has wide application prospect in the field of treating chronic infection.

Description

Application of iron death inhibitor in antiviral infection
Technical Field
The invention relates to the field of biological medicine, and relates to an application of an iron death inhibitor Liproxstatin-1 in antiviral infection.
Background
Chronic infections are metabolically adapted by enhancing lipid uptake or storage, which in turn is related to their dysfunction. Excessive lipid uptake leads to deregulation of lipid metabolism and increased production of Reactive Oxygen Species (ROS) leading to lipid peroxidation. Lipid peroxidation can produce large amounts of deleterious lipid peroxides that disrupt cell membranes and thereby trigger cell-mediated iron death [1], where GPX4 is considered an iron death core regulatory molecule. Cytotoxic cytokine production by cd8+ T cells, which are lipid peroxidized and iron-dead, is reduced, resulting in a greatly impaired anti-tumor capacity [2-4]. However, studies of chronic infections leading to cd8+ cell iron death have not been reported.
Currently known iron death inducers can be broadly divided into four types, depending on the mechanism of action: 1. inhibit type I of the Xc-system (CLASS I FIN); 2. inhibit or degrade GPX4, form II (CLASS II FIN); 3. type III (CLASS III FIN) consuming coenzyme Q10; 4. type IV (CLASS IV FIN) of lipid peroxidation induced by iron or polyunsaturated fatty acid (PUFA) overload. All four types have higher specificity for induction of iron death, namely: during induction of iron death, markers of other types of cell death are not activated substantially. The mechanism of action of different types of iron death inducers is completely different and has no reference significance.
In the prior researches, currently commonly used iron death inhibitors mainly comprise Liproxstatin-1, ferrostatin-1, alpha-Toc, vitE, DFO and the like, wherein Liproxstatin-1 and Ferrostatin-1 are aromatic amine antioxidants, and lipid peroxidation is inhibited by scavenging ROS; alpha-Toc and VitE are lipophilic natural antioxidants, and play an antioxidant role mainly by breaking the chain reaction of autoxidation; DFO iron chelators inhibit the production of hydroxyl radicals by blocking the fenton reaction. Common iron death inducers include targeting System Xc-, erastin affecting GSH synthesis, sulfasalazine, sorafenib, and the like; GPX4 is targeted, RSL3 with reduced antioxidant capacity is adopted; targeting iron ions, dihydroartemisinin which promotes accumulation of cellular ROS, and the like.
It has been found that iron death inhibitors have different biological effects in different cells, tissues or models. For example, iron death inhibitor Ferrostatin-1 can reverse the increase of the lipid peroxidation of CD8+ T cells caused by externally added fatty acid, improve cytokine production [2], and Ferrostatin-1 can also reduce the lipid peroxidation of tumor infiltrating T cells, thereby exerting stronger anti-tumor capability [5]; however, another study found that iron death inhibitor Ferrostatin-1 alone did not improve the killing function of cd8+ T cells [6].
It was found that the biological effects of different inhibitors are different. For example, ferrostatin-1 can inhibit sterol synthesis in macrophages infected with pathogenic yeast histoplasma capsulatum, not only can reduce death of infected macrophages, but also can inhibit growth of pseudomonas capsulata; while another arylamine antioxidant Liproxstatin-1 does not exert the above effects, and is not effective in preventing fungal growth or reducing macrophage death [7]. In addition, liproxstatin-1 was able to inhibit lipid peroxidation, restore GSH and GPX4 expression, and thereby significantly inhibit iron death from occurring in RSL 3-treated oligodendrocytes, whereas DFO effects were not apparent as other iron death inhibitors [8].
The effect of iron death inhibitors/inducers on different immune cells also varies. For example, in macrophages, iron death inducer RSL3 can induce iron death in M2 type macrophages, but cannot induce iron death in M1 type macrophages [9]. In the tumor microenvironment, iron death inducing agents Erastin and RSL3 at certain concentrations are effective in inducing iron death in tumor cells, but not in CD4+ T cells and CD8+ T cells [10]. In LCMV infected mouse models, feeding vitamin E significantly increases the absolute cell number of cd8+ T cells without affecting the absolute cell number of cd4+ T cells; however, in GPX4 knockout infected mice, vitamin E fed was able to restore the absolute cell number of CD4+ T cells, but not of CD8+ T cells [1].
Hepatitis B Virus (HBV) is a pathogen causing hepatitis B (abbreviated as hepatitis B), belongs to the family of hepadnaviridae, and comprises two genera of orthohepadnaviridae and avian hepadnaviridae, and causes infection of human bodies. The degree of liver cell damage is related to the intensity of immune response of organism, and the mechanism of HBV causing immune pathological damage is that the immune response of organism is low due to virus, drug resistance is generated by virus variation, immune pathological damage is mediated by antibody, immune pathological damage is mediated by cell, and the like. Common drugs include interferon, lamivudine, adefovir dipivoxil, telbivudine, entecavir, tenofovir disoproxil, and the like. Because the current first-line antiviral drugs cannot meet the clinical cure demands of most patients, there is a need to continue developing new antiviral drugs and researching new therapeutic schemes to improve the clinical cure of HB in a limited course of treatment, and the current new antiviral drugs are mainly divided into direct antiviral Drugs (DAAs) targeting the viral life cycle and indirect antiviral drugs participating in immune regulation. DAAs mainly comprise small interfering RNA, antisense oligonucleotides, gene editing silencing, capsid assembly regulator, cytostatic agent, HBsAg release inhibitor; indirect antiviral drugs involved in immunomodulation mainly include TLR7/8 innate immune modulators, therapeutic vaccines, monoclonal antibodies, immune checkpoint inhibitors, and the like.
In HBV chronic infection, there are serious obstacles to the body's innate immune response and acquired immune response. Replication of HBV within hepatocytes is not perceived by the intracellular innate immune system, resulting in impairment of antigen presenting function of DCs [11]. In addition, NK cells and HBV-specific cd4+, cd8+ T cells are severely dysfunctional and depleted, HBV is able to produce and secrete a large amount of viral antigen, gradually changing and depleting the function of HBV-specific T cells, resulting in decreased production of antiviral cytokines such as IFN- γ and TNF- α, ultimately resulting in HBV that is long-lasting in the body and is not readily cleared [12,13]. Current research consensus is that cd4+ and cd8+ T cell mediated immune responses determine HBV treatment outcome. However, even with the current standard therapies, the deficiency of the immune response makes it difficult for patients to restore HBV-specific immune cell function to normal levels, and thus does not effectively control infection.
The mechanism of virus infection is extremely complex, clinical symptoms caused by different virus infections are quite different, the immunological mechanism and principle of different treatment strategies are quite different, whether the inhibitor with a single mechanism can inhibit the virus infection is difficult to expect, and the treatment effects are also quite different. For example, the IAP antagonist APG-1387 for the research of a novel anti-HBV drug can induce the apoptosis of liver cells expressing HBV antigen and can enhance the specific T cell response of HBV [14]; anti-HBV selectively activates IL-2 pathway in cd8+ T cells, reversing the deficiency of cd8+ T cells without significant effect on other immune cells in the IL-2 immunotherapy drug studied AB359 [15]; in anti-HBV treatment, the TLR8 agonist GS-9688 can improve the cytolytic and non-cytolytic effect functions of NK cells, increase the number of HBV specific CD8+ T cells and the production of cytokines of partial patients, increase the number of follicular helper T cells and reduce the number of tregs and monocyte MDSCs [16]; mitochondrial antioxidants MitoQ and MitoTempo were able to significantly increase the effector function of HBV-specific CD8+ T cells, whereas only very weak effects were observed on the total T cell population [17]. In view of this, the mechanism by which different antiviral drugs regulate the antiviral immune response is complex.
1Matsushita M,Freigang S,Schneider C,Conrad M,Bornkamm GW,Kopf M.T cell lipid peroxidation induces ferroptosis and prevents immunity to infection.J Exp Med 2015;212:555–568.
2Ma X,Xiao L,Liu L,Ye L,Su P,Bi E,et al.CD36-mediated ferroptosis dampens intratumoral CD8+T cell effector function and impairs their antitumor ability.Cell Metab 2021;33:1001-1012.e5.
3Xu S,Chaudhary O,Rodríguez-Morales P,Sun X,Chen D,Zappasodi R,et al.Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+T cells in tumors.Immunity 2021;54:1561-1577.e7.
4Yang WS,SriRamaratnam R,Welsch ME,Shimada K,Skouta R,Viswanathan VS,et al.Regulation of Ferroptotic Cancer Cell Death by GPX4.Cell2014;156:317-331.
5Xiao L,Ma X,Ye L,Su P,Xiong W,Bi E,et al.IL-9/STAT3/fatty acid oxidation-mediated lipid peroxidation contributes to Tc9 cell longevity and enhanced antitumor activity.J Clin Invest;132:e153247.
6Drijvers JM,Gillis JE,Muijlwijk T,Nguyen TH,Gaudiano EF,Harris IS,et al.Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+T Cells.Cancer Immunol Res2021;9:184-199.
7Horwath MC,Bell-Horwath TR,Lescano V,Krishnan K,Merino EJ,Deepe GS.Antifungal Activity of the Lipophilic Antioxidant Ferrostatin-1.Chembiochem2017;18:2069-2078.
8Fan B-Y,Pang Y-L,Li W-X,Zhao C-X,ZhangY,Wang X,et al.Liprpxstatin-1is an effective inhibitor of oligodendrpcyte ferroptosis induced by inhibition of glutathione peroxidase 4.Neural Regen Res2020;16:561-566.
9Kapralov AA,Yang Q,Dar HH,Tyurina YY,Anthonymuthu TS,Kim R,et al.Redpx Lipid Reprogramming Commands Susceptibility of Macrophages and Micrpglia to Ferroptotic Death.Nat Chem Biol2020;16:278-290.
10Wang W,Green M,Choi JE,Gijón M,Kennedy PD,Johnson JK,etal.CD8+T cells regulate tumor ferroptosis during cancer immunptherapy.Nature2019;569:270-274.
11Papatheodoridis GV,Manolakopoulos S,Dusheiko G,Archimandritis AJ.Therapeutic strategies in the management of patients with chronic hepatitis B virus infection.Lancet Infect Dis2008;8:167-178.
12Guidotti LG,Chisari FV.Noncytolytic control of viralinfections by the innate and adaptive immune response.Annu Rev Immunol200l;19:65-91.
13Iannacone M,Guidotti LG.Immunobiology and pathogenesis of hepatitis B virus infection.Nat Rev Immunol2022;22∶19-32.
14Aseentage Pharma Group Inc.A Phase I Study of the Safety,Pharmacokinetic and Pharmacodynamic Properties of APG-1387in Patients With Chronic Hepatitis B.clinicaltrials.gov;2021.https://clinicaltrials.gov/ct2/show/NCT03585322(accessed 9Nov2022).
15The Liver Meeting|AASLD.https://www.aasld.org/the-liver-meeting(accessed11Nov2022).
16Amin OE,Colbeck EJ,Daffis S,Khan S,Ramakrishnan D,Pattabiraman D,et al.Therapeutic Potential of TLR8 Agonist GS-9688(Selgantolimod)in Chronic Hepatitis B:Remodeling of Antiviral and Regulatory Mediatprs.Hepatology2021;74:55-71.
17FisicaroP,Barili V,Montanini B,Acerbi G,Ferracin M,Guerrieri F,et al.Targeting mitpchondrial dysfunction can restpre antiviral activity of exhausted HBV-specific CD8 T cells in chronic hepatitis B.Nat Med 2017;23:327-336.
Liproxstatin-1 (Lip-1) is a potent inhibitor of iron death that can be used in vivo and in vitro. Lip-1 is easy to penetrate into the lipid bilayer and stays in the lipid bilayer, so that the active site of the Lip-1 is in close directional contact with the lipid peroxidation site, and CH3OO is started to extract hydrogen atoms from the aromatic amine site. After scavenging the lipid peroxide, the Lip-1 free radical formed by Lip-1 can be reduced by other antioxidants in the body (e.g. ubiquinone). Lip-1 inhibits iron death more effectively than other iron death inhibitors, such as deferoxamine, vitamin E, and ferrostatin-1. Lip-1 has been demonstrated to protect mouse myocardium from ischemia/reperfusion injury by reducing VDAC1 levels and restoring GPX4 levels; can inhibit RSL3 induced human renal proximal tubular epithelial cell death and GPX4 deficiency induced acute renal failure.
The mechanism of action of iron death inhibitor Liproxstatin-1 in antiviral infections has not been known so far, and no report has been made of Liproxstatin-1 in antiviral infections.
Disclosure of Invention
In order to overcome the technical problems in the prior art, the invention provides a new application of an iron death inhibitor Liproxstatin-1, in particular to an application in improving the effector function of CD8+T cells.
The invention is realized by the following technical scheme.
Use of an iron death inhibitor Liproxstatin-1 or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for increasing cd8+ T cell effector function or for combating viral infection.
Preferably, the method comprises the steps of,
The CD8+ T cells are CD8+ T cells and antigen specific CD8+ T cells in a chronic hepatitis B model.
As a preferred technical scheme of the invention: the use is to reduce the lipid peroxidation level of cd8+ T cells and antigen specific cd8+ T cells.
As a preferred technical scheme of the invention: the use is to increase the production of cd8+ T cells and antigen specific cd8+ T cell cytotoxic cytokines.
As a preferred technical scheme of the invention: the application is to inhibit iron death of CD8+ T cells and antigen-specific CD8+ T cells.
As a preferred technical scheme of the invention: the application is to improve the antiviral capacity of CD8+ T cells.
Preferably, the virus is HBV virus.
The invention also provides a medicine for resisting viral infection, the active ingredient of which is iron death inhibitor Liproxstatin-1 or pharmaceutically acceptable salt thereof. The virus is HBV.
Drawings
Fig. 1A: effect on cd8+ T cell lipid peroxidation in CHB patients after co-incubation with iron death inhibitor Liproxstatin-1.
Fig. 1B: antigen specific cd8+ T cell loop gate strategy was labeled using MHC tetramers.
Fig. 1C: effect of antigen-specific cd8+ T lipid peroxidation following co-incubation with iron death inhibitor Liproxstatin-1.
Fig. 1D: effect on cd8+ T cells IFN- γ and TNF- α production following co-incubation with iron death inhibitor Liproxstatin-1.
Fig. 1E: effects of antigen-specific CD8+ T cells IFN-gamma and TNF-alpha following co-incubation with iron death inhibitor Liproxstatin-1.
Fig. 1F: effects on cd8+ T cell GPX 4-related expression following co-incubation with iron death inhibitor Liproxstatin-1.
Fig. 2A: treatment protocol diagram of mice.
Fig. 2B: effect on lipid peroxidation of cd8+ T cells in the liver following treatment with iron death inhibitor Liproxstatin-1.
Fig. 2C: effects on intra-hepatic cd8+ T cell IFN- γ and TNF- α production following treatment with iron death inhibitor Liproxstatin-1.
Fig. 2D: effect on HBsAg in mice during treatment with iron death inhibitor Liproxstatin-1. Fig. 2E: effect on mouse HBVDNA following treatment with iron death inhibitor Liproxstatin-1;
FIG. 2F is the effect on mouse body weight following treatment with iron death inhibitor Liproxstatin-1.
Fig. 2G: effects on liver pathology in mice following treatment with iron death inhibitor Liproxstatin-1.
Detailed Description
The invention is further described below, the embodiments presented in this description are only exemplary and do not limit the scope of the invention. It will be understood by those skilled in the art that the details and forms of the invention may be modified or substituted without departing from the spirit and scope of the invention.
Definition of terms
As used herein, the term "cd8+ T cell" refers to a cytotoxic T lymphocyte, a killer T cell, that can participate in immunization by secretion of various cytokines.
As used herein, the term "iron death" is a novel mode of cell death first proposed by the Brent R Stockwell doctor's laboratory in 2012. Iron death is different from pyro-death, apoptosis, necrosis and autophagic cell death. This process of iron death is manifested by the accumulation of lethal ROS and lipid peroxidation products (LPOs) during iron metabolism.
As used herein, the term "Lip-1", i.e., iron death inhibitor Liproxstatin-1, is a potent iron death inhibitor. By inhibiting the generation of lipid free radicals, lipid peroxidation and iron death are inhibited.
As used herein, the term "Med" is a cell culture medium, i.e., a primary cell culture medium.
As used herein, the term "Pep" is an HBV antigenic peptide, the sequence of which is HBV-core (aa 18-27: FLPSDFFPSV).
As used herein, the term "MHC tetramer" is a combination of soluble MHC proteins and polypeptides, and is a simple and effective method for identifying, isolating and studying antigen-specific T cells from a variety of biological samples.
As used herein, the term "IFN- γ", i.e. interferon γ, is an important cytokine capable of performing an immunomodulatory function. It was found due to its antiviral activity. IFN-gamma plays a key role in host defense through antiviral, antiproliferative and immunomodulatory functions. Can reflect immune cell effector functions.
As used herein, the term "TNF- α," tumor necrosis factor α, is involved in normal inflammatory and immune responses, and may synergistically regulate the production of other cytokines, cell survival and death to coordinate tissue homeostasis. Cytotoxicity to various tumor cells is an important factor in mediating the immune response to viral infection. Can reflect immune cell effector functions.
As used herein, the term "GPX4", glutathione peroxidase-4, plays a key role in degrading deleterious lipid peroxides to maintain homeostasis as an iron death core regulatory molecule.
This patent is described in further detail below in conjunction with the drawings and specific experiments. Unless otherwise indicated, all reagents, instruments, devices and methods used in this patent are those commonly available in the art.
Example 1
In vitro experiments.
Isolation of PBMCs: PBMCs were isolated from fresh heparinized blood using Ficoll-Hypaque density gradient.
Specific T cell treatment: isolated PBMCs from CHB patients were cultured in RPMI1640 containing 10% fetal bovine serum. HBV-specific CD8+ T cells were further analyzed by stimulating PBMCs with HBV peptide (1 μM) for 10 days in the presence or absence of iron death inhibitor Liproxstatin-1 (50 nM). Sorting and activation of cd8+ T cells: CD8+ T cells were isolated from PBMC using a negative selection kit (Biolegend) and stimulated with the addition of human T activating antibodies CD3/CD28 (10. Mu.g/ml, biolegend) and IL-2 (10 ng/ml). Cd8+ T cells were stimulated with HBV peptide (1 μm) for 10 days in the presence or absence of iron death inhibitor Liproxstatin-1.
Lipid peroxidation measurement: the experiments were performed according to the manufacturer's protocol. Briefly, cells were incubated with lipid peroxidation sensor C11 BODIPY 581/591 (Invitrogen) in cell culture medium for 30 minutes in a 37℃incubator containing 5% carbon dioxide. After incubation, cells were washed and flow cytometry detection was performed with FACSCELESTA (BD) within 2 hours after staining.
MHC tetramer staining: to 25. Mu.L of peripheral blood mononuclear cell suspension (2X 10 7/mL) was added 1. Mu. LMHC tetrameric dye (HelixGen) and incubated for 60 minutes in ice protected from light. After incubation, FACSBuffer (pbs+2% calf serum+0.1% sodium azide) was added for washing and then resuspended in 200 μl of fixative (pbs+1% paraformaldehyde) for fixation.
Production assay of T cell cytokines: cell activation medium (100 IU/mL IL-2+1. Mu.g/mL ionomycin+50 ng/mL PMA+10. Mu.g/mL brefeldin A) was prepared, 1mL activation medium was added to 1X 10 6 PBMC cells, and incubated in 24-well plates for 4 hours. After labeling CD3-Percp-Cy5.5 (Biolegend), CD8-BV421 (Biolegend) and MHC tetramer-APC, the samples were subjected to fixation-permeabilization blocking, labeling with IFN-. Gamma. -PE (Biolegend) and TNF-. Alpha. -PE-CF594 (Biolegend), and washing. GPX4 markers: the permeabilized blocked cells were blocked by adding 1/400 dilution of recombinant Anti-Glutathione Peroxidase antibody (Abcam) for 2h at 4deg.C, and goat serum was added to block, goat Anti-rabbit IgG-FITC (Abclonal) was used as secondary antibody at a dilution of 1/2000.
As shown in FIG. 1A, lip-1 significantly reduced the lipid peroxidation level of CD8+ T cells in PBMC of CHB patients.
As shown in fig. 1B, the main cell population was circled in the sorted cd8+ T cells, after diagonal deblocking, the single cell population was circled, and finally the tetramer positive cell population, i.e., HBV-specific cd8+ T cells, was circled.
As shown in FIG. 1C, lip-1 significantly reduced the lipid peroxidation level of HBV-specific CD8+ T cells in PBMC of CHB patients.
As shown in FIG. 1D, lip-1 significantly increased cytokine production (IFN-. Gamma.and TNF-. Alpha.) by CD8+ T cells in PBMC of CHB patients.
As shown in FIG. 1E, lip-1 was able to increase the production of cytokines (IFN-. Gamma.and TNF-. Alpha.) by HBV-specific CD8+ T cells in PBMC of CHB patients.
As shown in FIG. 1F, lip-1 significantly increased the level of GPX4 in CD8+ T cells and HBV-specific CD8+ T cells in PBMC of CHB patients.
(Wherein "ns" represents no significant difference, "" represents p <0.05, "" represents p < 0.01)
Example 2
In vivo experiments.
Construction of HBV-carrier mouse model: taking C57BL/6J mice of 5-6 weeks old, injecting 8 mu gpAAV/HBV1.2 plasmid into tail vein under high pressure, taking peripheral blood after 5-6 weeks to separate serum, and detecting the HBsAg level in the serum, wherein the serum HBsAg concentration is higher than 500ng/mL, which is HBV-carrier mice successfully molded.
Treatment strategy: mice with successful molding were intraperitoneally injected with 30mg/kg of iron death inhibitor Liproxstatin-1 daily, and body weight changes were detected every two days for 15 days of treatment.
Mice were bled to isolate serum: fixing the mice in the fixer under the aseptic state, and exposing tails of the mice; gently wiping the tail of the mice with 70% alcohol for disinfection; the tail end of the mouse was cut at about 2mm, and the tail was massaged from the tail root to the tail tip, and the blood flowing out of the tail tip was collected. Standing at room temperature for 30min, centrifuging at 3000rpm for 15min, collecting supernatant, and storing at-80deg.C.
Obtaining liver mononuclear cells: dissecting and separating the liver of the mouse, grinding and then collecting the liver into a 15mL centrifuge tube; 100g, centrifuging for 1 min, and collecting a supernatant; centrifuging 400g for 5 min, and discarding supernatant to obtain precipitate; 4ml of 40% Percoll solution (cytiva) was added to resuspend the cells; centrifuging at 800g for 25 min, discarding supernatant to retain precipitate; adding erythrocyte lysate to resuspend cells, standing at 4 ℃ for 10 minutes, stopping erythrocyte lysis, and filtering to separate tubes.
Serum HBsAg levels in mice peripheral blood were measured by CLIA method: diluting the serum sample according to experimental requirements; add 50. Mu.L of sample or standard (0, 0.05, 0.8, 10, 85, 250 ng/mL) to the corresponding coated wells, respectively; adding 50 mu L of enzyme conjugate into each hole, gently shaking and mixing, pasting a sealing plate film, and incubating for 1h at 37 ℃; after the incubation is finished, the sealing plate film is torn off, liquid in the holes is forcibly thrown off, the coated plate is washed by HBsAg washing liquid, the washing is repeated for 5 times, and finally, the coated plate is patted to be dry as much as possible; mixing the luminous substrate A and the luminous substrate B according to the proportion of 1:1, adding 50 mu L of the mixed luminous substrate into the coating hole, gently shaking, and standing for 10min at room temperature in a dark place. And detecting the luminous intensity by using a Synergy 2 multifunctional enzyme-labeled instrument, making a standard curve, and calculating the concentration of HBsAg in the sample. Serum HBV DNA detection: to 200. Mu.L of the sample, 450. Mu.L of DNA extract I and 4. Mu.L of internal standard solution were added, and the mixture was shaken and mixed for 15 seconds, followed by instantaneous centrifugation to remove water droplets on the lid and the tube wall. Treating at 100deg.C for 10+ -1 min, and centrifuging at 12000rpm for 5min. The corresponding amount of PCR reaction liquid and Taq enzyme (HBV PCR reaction liquid 27 mu L/tube+Taq enzyme 3 mu L/tube) are taken according to the proportion, and are fully mixed and packaged into PCR reaction empty tubes suitable for each instrument according to 30 mu L/tube for standby. And respectively adding the extracted sample nucleic acid to be detected, HBV negative quality control product, HBV strong positive quality control product, HBV critical positive quality control product and positive quantitative reference product into the HBV reaction tube by using a suction nozzle with a filter element, centrifuging for a plurality of seconds at 8000rpm, and transferring to an amplification detection area. The PCR reaction conditions were as follows (DA 7600):
TABLE 1 PCR reaction conditions for detecting peripheral blood serum DNA
As shown in FIG. 2A, HBV-carrier mice after 5 weeks of successful molding were intraperitoneally injected with 30mg/kg of iron death inhibitor Liproxstatin-1 per day, and body weights were measured every two days for 15 days of treatment.
As shown in fig. 2B, the lipid peroxidation level of cd8+ T cells in the liver of HBV-carrier mice was significantly higher than that of wild-type mice, and Lip-1 treatment significantly reduced the lipid peroxidation level of HBV-carrier mice.
As shown in fig. 2C, lip-1 treatment significantly improved cytokine production by cd8+ T cells (IFN- γ and TNF- α) in the liver of HBV-carrier mice.
As shown in fig. 2D, lip-1 treatment was able to reduce HBsAg levels in HBV-carrier mouse serum.
As shown in fig. 2E, lip-1 treatment was able to reduce the level of HBVDNA in HBV-carrier mouse serum.
As shown in fig. 2F, lip-1 treatment reduced HBV-carrier mice weight at day 5 and gradually recovered to normal at day 10.
As shown in fig. 2G, lip-1 treatment did not show damage to the liver of mice.
(Wherein "ns" represents no significant difference, "" represents p <0.05, "" represents p <0.01, "" represents p < 0.0001)
The present study also explored the effect of other iron death inhibitors on antigen-specific cd8+ T cells of HBV infection, found that edaravone, ferrostatin-1, vitamins E, UAMC-3203 and defetoxamine, etc. all had no significant activating/stimulating effect, nor observed therapeutic effect that could significantly benefit in vivo in anti-HBV infection studies.
While the invention has been described with reference to specific embodiments thereof, it will be understood by those skilled in the art that the invention is not limited thereto, and that various changes and modifications may be made without departing from the spirit of the invention.

Claims (2)

1. A method of making antigen-specific cd8+ T cells, comprising the steps of:
Step 1) isolated PBMCs from patients with chronic hepatitis B are incubated in RPMI 1640 containing 10% fetal bovine serum, and the PBMCs are stimulated with HBV peptide in the presence of iron death inhibitor Liproxstatin-1 for 10 days;
step 2) CD8+ T cells are isolated from PBMC and stimulated with the addition of human T activating antibodies CD3/CD28 and IL-2; stimulating cd8+ T cells with HBV peptide in the presence of iron death inhibitor Liproxstatin-1 for 10 days;
The HBV peptide is FLPSDFFPSV.
2. The method according to claim 1, wherein the concentration of Liproxstatin-1 used in step 1) and step 2) is 50nM.
CN202211742287.6A 2022-11-16 2022-11-16 Application of iron death inhibitor in antiviral infection Active CN115804772B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211742287.6A CN115804772B (en) 2022-11-16 2022-11-16 Application of iron death inhibitor in antiviral infection

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211742287.6A CN115804772B (en) 2022-11-16 2022-11-16 Application of iron death inhibitor in antiviral infection

Publications (2)

Publication Number Publication Date
CN115804772A CN115804772A (en) 2023-03-17
CN115804772B true CN115804772B (en) 2024-04-26

Family

ID=85487214

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211742287.6A Active CN115804772B (en) 2022-11-16 2022-11-16 Application of iron death inhibitor in antiviral infection

Country Status (1)

Country Link
CN (1) CN115804772B (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112638375A (en) * 2018-06-15 2021-04-09 旗舰创业创新五公司 Increasing immune activity through modulation of post-cellular signaling factors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112638375A (en) * 2018-06-15 2021-04-09 旗舰创业创新五公司 Increasing immune activity through modulation of post-cellular signaling factors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Inhibiting ACSL1-Related Ferroptosis Restrains Murine Coronavirus Infection;Huawei Xia等;Viruses.;20211128;第13卷(第12期);论文编号2383 *

Also Published As

Publication number Publication date
CN115804772A (en) 2023-03-17

Similar Documents

Publication Publication Date Title
Hunter et al. Production of gamma interferon by natural killer cells from Toxoplasma gondii-infected SCID mice: regulation by interleukin-10, interleukin-12, and tumor necrosis factor alpha
KR100518903B1 (en) Immune response modifier compounds for treatment of the th2 mediated and related diseases
Wang et al. The in vitro and in vivo antiviral effects of salidroside from Rhodiola rosea L. against coxsackievirus B3
AU2014244744B2 (en) Pharmaceutical composition for inhibiting immune response through inducing differentiation into regulator T cells and promoting proliferation of regulator T cells
US20190388369A1 (en) Use of N-acetylcysteine Amide in the Treatment Of Disease and Injury
JP2013522302A (en) Pharmacogenomics and therapeutic response guide treatment of infectious diseases using yeast-based immunotherapy
JP2014523878A (en) Yeast-based compositions and methods for the treatment or prevention of hepatitis D virus infection
TWI732156B (en) Bisdiazabicyclic compounds for the treatment and/or prevention of diseases or disorders related to hepatitis virus
US20060079510A1 (en) Use of PARP-1 inhibitors for protecting tumorcidal lymphocytes from apoptosis
CA2649290C (en) Allogeneic cell therapy for treatment of opportunistic infection
AU2017267172A1 (en) A specific trifluoroethyl quinoline analogue for use in the treatment of APDS
JP2004532803A (en) Methods and compositions for promoting monocyte maturation
CN115804772B (en) Application of iron death inhibitor in antiviral infection
WO2017147894A1 (en) Composition for enhancing capacity to kill abnormal cells and use thereof
US11771759B2 (en) Immunopotentiator, immunotherapeutic pharmaceutical composition and its preparation and use
Pérez-Casanova et al. Morphine-mediated deterioration of oxidative stress leads to rapid disease progression in SIV/SHIV-infected macaques
WO2022213870A1 (en) Drug and method for inhibiting cd4+treg cells by means of oral administration
CN114949189A (en) Application of nano tumor specific antigen and ICD (acute transient adhesion) -generated tumor cell combination in preparation of therapeutic tumor vaccine
US20040081686A1 (en) Use of particle vectors in immunomodulation
US20220226281A1 (en) Compounds for use in anti-cancer immunotherapy
US20050239844A1 (en) Composition fo preventing secretion of immunoglobulin e-dependent histamine releasing factor
Liang et al. 1Prevention of immune dysfunction, vitamin E deficiency, and loss of Cryptosporidium resistance during murine retrovirus infection by T cell receptor peptide immunization
WO2014059147A1 (en) Methods and composition for treatment of th2-mediated and th17-mediated diseases
KleinJan et al. Dendritic cells in rhinitis
KR20090009963A (en) Use of thymosin alpha 1 for the treatment of immunological diseases

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
GR01 Patent grant
GR01 Patent grant