CN116355844B - Establishment and application of SARS-CoV-2 antigen induced cytokine storm model - Google Patents

Establishment and application of SARS-CoV-2 antigen induced cytokine storm model Download PDF

Info

Publication number
CN116355844B
CN116355844B CN202310628453.8A CN202310628453A CN116355844B CN 116355844 B CN116355844 B CN 116355844B CN 202310628453 A CN202310628453 A CN 202310628453A CN 116355844 B CN116355844 B CN 116355844B
Authority
CN
China
Prior art keywords
cov
sars
cytokine storm
cells
model
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
CN202310628453.8A
Other languages
Chinese (zh)
Other versions
CN116355844A (en
Inventor
牛超
崔久嵬
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.)
First Hospital Jinlin University
Original Assignee
First Hospital Jinlin 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 First Hospital Jinlin University filed Critical First Hospital Jinlin University
Priority to CN202310628453.8A priority Critical patent/CN116355844B/en
Publication of CN116355844A publication Critical patent/CN116355844A/en
Application granted granted Critical
Publication of CN116355844B publication Critical patent/CN116355844B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • General Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention discloses a method for establishing and applying SARS-CoV-2 antigen induced cytokine storm model, concretely, the method includes adding SARS-CoV-2 antigen and interleukin into immune cell culture system. The invention provides a model of SARS-CoV-2 antigen induced cytokine storm, which has simple operation and lower requirements on laboratory level and hardware, is suitable for developing effective medicaments for resisting SARS-CoV-2 induced cytokine storm by various scientific research institutions and enterprises, is also suitable for clinically screening effective therapeutic medicaments for SARS-CoV-2 infected patients, and has good application prospect.

Description

Establishment and application of SARS-CoV-2 antigen induced cytokine storm model
Technical Field
The invention belongs to the field of biological medicine, and in particular relates to establishment and application of a SARS-CoV-2 antigen induced cytokine storm model.
Background
Cytokine storm (Cytokine storm), a syndrome of Cytokine release (Cytokine release syndrome, CRS), is an excessive response of the body to infection. When the body is infected by some viruses or the immune function is abnormal, the cytokine balance in the body is broken, the proinflammatory cytokines are continuously and largely produced, and more immune cells are continuously activated to be accumulated at the inflammation part. Excessive immune cells and a variety of pro-inflammatory cytokines can cause tissue congestion, edema, fever, injury, and may also cause other secondary infections and even lead to "systemic inflammatory response syndrome" (sepsis), leading to death in patients due to multiple organ failure.
The main cause of death of moderately severe SARS-CoV-2 infected patients is the occurrence and development of cytokine storm, and the development of drugs capable of alleviating cytokine storm is not easy. The virus-induced cytokine storm model is the most direct and effective choice for studying SARS-CoV-2. However, the virus research has a certain risk, especially the SARS-CoV-2 virus has strong infectivity, and has extremely high requirements on laboratory grade and personnel qualification, so the selection of the model is limited to a certain extent. There are few reports of in vivo studies in mice currently using the cytokine storm caused by SARS-CoV-2 infection directly. The non-clinical pharmacodynamics experimental method of the cytokine storm therapeutic drug comprises the steps of establishing a cytokine storm model of virus induction, LPS induction, TLR agonist induction and antigen induction on mice, and selecting primates such as rhesus monkeys for experiment more closely to the animal model of human. In vitro models, RAW264.7 cell line or primary macrophages extracted from mice, peripheral blood mononuclear cells, etc. can be selected for testing, and the induction method is also generally to stimulate monocytes or macrophages by using LPS or a TLR agonist. Typical immune cells include macrophages, dendritic cells, lymphocytes, peripheral blood mononuclear cells, and the like.
In addition, the research of SARS-CoV-2 requires a P3 laboratory, but the institutions with the P3 laboratory in the universities and enterprises of China are not more, which results in difficult direct research of SARS-CoV-2 and difficult establishment of a cytokine release syndrome model caused by SARS-CoV-2. Even if a corresponding model is established, the method is difficult to widely popularize and use due to the lack of a P3 laboratory or high cost. Due to the lack of a simple, efficient, low cost model for the massive release of SARS-CoV-2-induced cytokines, the development of drugs against SARS-CoV-2-induced cytokine release has been slow. In addition, researchers have constructed an inflammation model using alternative viruses or LPS and the like, but a model induced by non-SARS-CoV-2 cannot truly respond to the real situation that SARS-CoV-2 causes the body to release a large amount of cytokines.
Disclosure of Invention
In order to solve the above-mentioned shortcomings, the present invention aims to establish an in vitro model for large scale release of SARS-CoV-2 antigen induced cytokine without P3 laboratory, and in order to achieve the purpose, the present invention adopts the following technical scheme:
in a first aspect, the present invention provides a method for constructing a model for inducing a cytokine storm by SARS-CoV-2 antigen, said method comprising adding SARS-CoV-2 antigen and interleukin into a culture system of immune cells.
It should be noted that the terms "comprising" and/or "including" are used to indicate that various aspects and embodiments of the disclosure "comprise" a particular feature or features. It should be understood that this/these terms may also encompass aspects and/or embodiments that "consist essentially of, or" consist of, the relevant feature or features.
As a preferred embodiment, the cell culture medium used in the immune cell culture system is a serum-free medium added to plasma.
As a preferred embodiment, the plasma is human plasma.
As a preferred embodiment, the plasma is autologous plasma, more preferably, the autologous plasma is present at a concentration of 10% in the cell culture medium.
As an alternative embodiment, the plasma may be replaced with serum, including autologous serum or fetal bovine serum.
SARS-CoV-2 is a large enveloped positive-stranded RNA virus, and the SARS-CoV-2 genome is packaged within a spiral capsid surrounded by an envelope. At least 3 structural proteins are associated with the viral envelope: spike protein (S), envelope protein (E) and membrane protein (M).
In the present invention, the SARS-CoV-2 antigen can be selected from the group consisting of: spike protein (S), envelope protein (E), membrane protein (M), nucleoprotein (N) and/or hemagglutinin esterase protein (HE).
As a preferred embodiment, the SARS-CoV-2 antigen comprises a viral antigen that is capable of acting through TLR 4.
As a preferred embodiment, the viral antigen capable of acting through TLR4 comprises spike protein.
As a preferred embodiment, the concentration of SARS-CoV-2 antigen is 0.3125, 0.625, 1.25, 2.5, 5, 10 or 20 nM, more preferably the concentration of SARS-CoV-2 antigen is 10 nM.
"Interleukin" refers to a cytokine protein produced by lymphocytes, including but not limited to cytokines commonly known as IL-2, IL-3, IL-4, IL-5, IL-10, IL-12, IL-13, IL-18, and mixtures thereof. The interleukins may be derived from natural sources or recombinantly produced.
As a preferred embodiment, the interleukin is IL-2.
As a preferred embodiment, the IL-2 concentration is 0.1-100 ng/mL, more preferably, the IL-2 concentration is 10 ng/mL.
As used herein, the term immune cell refers to any cell that plays a role in an immune response. Immune cells are derived from hematopoietic stem cells and include, but are not limited to, lymphocytes, such as T cells, NK cells, and B cells; myeloid cells such as monocytes, macrophages, dendritic cells, eosinophils, neutrophils, basophils and mast cells.
The term "lymphocyte" refers to all immature, mature, undifferentiated and differentiated lymphocyte populations, including tissue specific and specialized varieties. As non-limiting examples, it encompasses NK cells, T cells, NKT cells, and B cells.
As used herein, the term T cell includes naive T cells, CD4 + T cells, CD8 + T cells, memory T cells, activated T cells, anergic T cells, tolerating T cells, chimeric T cells, antigen-specific T cells, and regulatory T cells.
As a non-limiting example, the term "B cell (B cell or B cell)" refers to a pre-B cell, a progenitor B cell, an early pre-B cell, a late pre-B cell, a large pre-B cell, a small pre-B cell, an immature B cell, a mature B cell, a naive B cell, a plasma B cell, an activated B cell, an anergic B cell, a tolerogenic B cell, a chimeric B cell, an antigen-specific B cell, a memory B cell, a B-1 cell, a B-2 cell.
As a preferred embodiment, the immune cells are peripheral blood mononuclear cells.
As used herein, "peripheral blood mononuclear cells" refers to any cell with a single nucleus in peripheral blood, including lymphocytes and monocytes.
In a specific embodiment of the invention, the peripheral blood mononuclear cells are human peripheral blood mononuclear cells.
As a preferred embodiment, the concentration of the peripheral blood mononuclear cells is (0.5-3). Times.10 6 In the specific embodiment of the invention, the concentration of the peripheral blood mononuclear cells is 1X 10 6 cells/mL.
As a preferred embodiment, the method further comprises culturing the culture system with SARS-CoV-2 antigen and interleukin immune cells in a cell culture incubator.
As a preferred embodiment, the time of the culture is 4 to 24 hours, more preferably 12 to 16 hours. In a specific embodiment of the invention, the incubation time is 16 hours.
The cell culture vessel used in the culture system of the present invention is not particularly limited as long as it is capable of culturing cells, and examples thereof include culture flasks, tissue culture flasks, dishes, plates, tissue culture dishes, multi-well dishes, microplates, microwell plates, multi-well plates, culture dishes, tubes, culture bags, and roller bottles.
As a preferred embodiment, the cell culture vessel is a multi-well plate including, but not limited to, 6-well plate, 12-well plate, 24-well plate, 48-well plate, 96-well plate, 384-well plate. In a specific embodiment of the invention, the cell culture vessel is a 24-well plate.
In the present invention, the culture conditions can be appropriately set. For example, the culture temperature is not particularly limited, and may be about 30 to 40℃and preferably about 37 ℃. CO 2 The concentration may be about 1 to 10%, preferably about 2 to 5%. The oxygen partial pressure may be 1 to 10%.
"cytokine", as used herein, is a widely and loosely-varying small protein (about 5-20 kDa) that is important in cell signaling. They are released by the cells and affect the behavior of other cells. Cytokines may also be involved in autocrine signaling. Cytokines include chemokines, interferons, interleukins, lymphokines, tumor necrosis factors, and growth factors. In specific embodiments of the invention, the cytokine comprises IL-1β, IL-6 or IL-8.
In a second aspect, the invention provides a model of SARS-CoV-2 antigen induced cytokine storm, said model being prepared by the method of the first aspect of the invention.
In a third aspect, the present invention provides the use of the model of SARS-CoV-2 antigen induced cytokine storm described in the second aspect of the present invention for screening an anti-cytokine storm drug using the model of SARS-CoV-2 antigen induced cytokine storm described in the second aspect of the present invention.
The invention has the advantages and beneficial effects that:
the invention discloses that the phenomenon that SARS-CoV-2 induces a large amount of human cytokines to be released is simulated by using SARS-CoV-2 antigen and interleukin to stimulate immune cells for the first time. The invention discloses a model of SARS-CoV-2 antigen induced cytokine storm, which has simple operation and lower requirements on laboratory level and hardware, is suitable for developing effective medicaments for resisting SARS-CoV-2 induced cytokine storm by various scientific research institutions and enterprises, is also suitable for clinically screening effective therapeutic medicaments for SARS-CoV-2 infected patients, and has good application prospect.
Drawings
FIG. 1 is a graph of experimental results of detecting the secretion of a large amount of cytokines by PBMCs stimulated by spike proteins and/or IL-2, wherein, graph A is a statistical graph of IL-1β detection results, graph B is a statistical graph of IL-6 detection results, and graph C is a statistical graph of IL-8 detection results;
FIG. 2 is a graph of experimental results for detecting cytokines in models established by different methods, wherein graph A is a statistical graph of IL-1. Beta. Detection results, graph B is a statistical graph of IL-6 detection results, and graph C is a statistical graph of IL-8 detection results;
FIG. 3 is a graph of experimental results of detecting the inhibition of cytokine storm-related inflammatory factor release by different drugs using the model of the present invention, wherein graph A is a statistical graph of IL-1β detection results, graph B is a statistical graph of IL-6 detection results, and graph C is a statistical graph of IL-8 detection results.
Detailed Description
Embodiments of the present invention will be described in detail below with reference to examples, but it will be understood by those skilled in the art that the following examples are only for illustrating the present invention and should not be construed as limiting the scope of the present invention. The specific conditions are not noted in the examples and are carried out according to conventional conditions or conditions recommended by the manufacturer. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention.
Experimental materials
Example construction of in vitro model of SARS-CoV-2 antigen-induced cytokine storm
Experimental method
1. Isolation of PBMCs:
1. the heparin anticoagulation tube directly collects peripheral blood (3-100 mL), extracts 0.2-1 mL blood in a biosafety cabinet for detecting mononuclear cells, T cells and NK cells in the peripheral blood by flow cytometry, distributes the rest blood into two 15 mL or 50 mL sterile centrifuge tubes on average, and centrifugates for 10 min at room temperature with 700-900 g.
2. The plasma was transferred to a new 15 mL or 50 mL sterile centrifuge tube using a disposable sterile pipette and placed in a constant temperature incubator at 56 ℃ for 30 min to inactivate complement.
3. Adding normal saline or PBS buffer solution with the same volume as the blood sampling amount into the centrifuged blood cells, uniformly mixing, slowly moving the diluted cell suspension into a 15 mL or 50 mL centrifuge tube containing lymphocyte separation liquid by adopting a disposable sterile pipette according to the proportion of 1-2:1 (volume ratio), slightly moving the centrifuge tube into the centrifuge, and centrifuging for 30 min at the room temperature at 350-450 g.
4. The centrifuge tubes were slowly removed and gently placed in a biosafety cabinet, and the middle peripheral blood mononuclear cell (Pheripheral blood mononuclear cells, PBMCs) layers were aspirated with disposable sterile pipettes and placed into new 15 mL or 50 mL sterile centrifuge tubes, each of which contained a white film layer that was not expected to exceed 10% of the tube capacity.
5. Adding PBS or physiological saline into a sterile centrifuge tube, mixing, and centrifuging at 400-500-g and room temperature for 8 min. The wash was repeated once.
2. Establishing an in vitro model
1. Modulation of PBMCs to 1X 10 6 cell/mL 10% autologous plasma serum-free medium.
2. And PBMCs were seeded at 400. Mu.L/well in 24-well plates and PBS, spike protein (0, 0.3125, 0.625, 1.25, 2.5, 5, 10, 20 nM, most preferably 10 nM), IL-2 (within 100 ng/mL, most preferably 10 ng/mL), IL-2 (10 ng/mL) and spike protein (10 nM) were added, respectively, at 37℃in 5% CO 2 After incubation for 16 hours in an incubator, collecting supernatant, and temporarily storing the supernatant in a refrigerator at 4 ℃ in the detection day; can be stored for a long time and packaged, and stored in a refrigerator at-80deg.C.
3. In vitro models of SARS-CoV-2 antigen-induced cytokine storm include: (1) a PBS group; (2) spike group (10 nM); (3) IL-2 group (10 ng/mL); (4) IL-2 (10 ng/mL) in combination with spike protein (10 nM). Wherein the (1), (2) and (3) groups are used as controls, and the (4) group is an in vitro experimental model of novel coronavirus antigen induced cytokine storm.
4. ELISA or CBA detects cytokines IL-1 beta, IL-6, IL-8 and the like related to cytokine release syndrome.
3. Application (drug screening for inhibiting SARS-CoV-2 induced cytokine storm)
1. Can be used as an in vitro model for drug screening, and the experimental group comprises: (1) a PBS group; (2) spike group (10 nM); (3) IL-2 group (10 ng/mL); (4) IL-2 in combination with spike protein groups; (5) drug screening group: based on the group (4), different drugs (such as celecoxib and TLR4 inhibitor) are added, and the control effect of the different drugs on SARS-CoV-2 induced cytokine release is detected.
2. At 37 ℃,5% CO 2 After 16 hours incubation in the incubator, the supernatant was collected. The detection on the same day can be stored in a refrigerator at 4 ℃; can be stored for a long time and packaged, and stored in a refrigerator at-80deg.C.
3. ELISA or CBA detects cytokines IL-1 beta, IL-6, IL-8 and the like related to cytokine release syndrome.
4. Based on the expression of cytokines, the anti-inflammatory effects of different drugs and the anti-inflammatory effects of different doses of drugs were analyzed.
Experimental results
1. Isolation of donor Peripheral Blood Mononuclear Cells (PBMCs), and adjustment of PBMCs to a density of 1X 10 with serum-free Medium containing 10% autologous plasma 6 cells/mL, 400. Mu.L/well were inoculated into 24-well plates, and PBS, spike protein (10 nM), IL-2 (10 ng/mL), IL-2-associated spike protein were then added, respectively, at 37℃with 5% CO 2 After 16 hours incubation in the incubator, the supernatants were collected and CBA assayed for the concentration of various cytokines.
As shown in FIG. 1, the use of both spike protein and IL-2 alone stimulated Peripheral Blood Mononuclear Cells (PBMCs) to secrete IL-1 beta, IL-6 and IL-8; however, the spike protein in combination with IL-2 (10 ng/mL) was able to stimulate the secretion of more IL-1β, IL-6 and IL-8 by PBMCs. IL-1 beta, IL-6 and IL-8 are key cytokines in SARS-CoV-2 induced cytokine storm. The results in fig. 1 show that: SARS-CoV-2 spike protein, combined with lower concentrations of IL-2 to stimulate PBMCs, is able to mimic SARS-CoV-2-induced cytokine storm in vitro.
2. Isolation of donor Peripheral Blood Mononuclear Cells (PBMCs), and adjustment of PBMCs to a density of 1X 10 with serum-free Medium containing 10% autologous plasma 6 cells/mL, 400. Mu.L/well were inoculated into 24-well plates, and PBS, spike protein (10 nM), IL-2 (10 ng/mL), IL-2-associated spike protein, IL-15 (10 ng/mL), IL-15-associated spike protein were then added, respectively, at 37℃with 5% CO 2 After 16 hours incubation in the incubator, the supernatants were collected and CBA detected the concentrations of IL-1. Beta., IL-6 and IL-8.
As shown in FIG. 2, IL-2 is capable of stimulating PBMCs to secrete large amounts of IL-1β, IL-6 and IL-8 in concert with spike proteins; IL-15 in combination with the spike protein stimulated PBMCs failed to produce significant amounts of IL-1β, IL-6 and IL-8 compared to the spike protein alone stimulated group.
IL-15 was selected in this study to create an in vitro model for SARS-CoV-2 drug screening because previous studies reported that IL-15, like IL-2, was able to function via IL-2 receptors β and γ chain, and that IL-15 was also significantly upregulated in SARS-CoV-2-induced cytokine storm, but this study found that IL-15 in combination with spike protein did not have significant effect in stimulating peripheral blood mononuclear cells to secrete IL-1 β, IL-6 and IL-8. The results show that the invention has good technical effect by selecting IL-2.
3. Isolation of donor Peripheral Blood Mononuclear Cells (PBMCs), and adjustment of PBMCs to a density of 1X 10 with serum-free Medium containing 10% autologous plasma 6 cells/mL, 400. Mu.L/well were seeded in 24 well plates, and PBS, spike protein (10 nM), IL-2 (10 ng/mL), IL-2 associated spike protein, IL-2 associated spike protein+TLR 4 inhibitor (1. Mu.M TAK-242), IL-2 associated spike protein+celecoxib (100. Mu.M), were then added, respectively, at 37℃with 5% CO 2 After 16 hours incubation in the incubator, the supernatants were collected and CBA detected the concentrations of IL-1. Beta., IL-6 and IL-8.
As shown in fig. 3, the invention model is used for drug screening, and the inhibition effect of the TLR4 inhibitor and celecoxib on the release of inflammatory factors related to cytokine storm is detected, so that the TLR4 inhibitor and celecoxib can effectively inhibit the synergistic spike protein of IL-2 to stimulate the PBMCs to release a large amount of IL-1 beta, IL-6 and IL-8. The research results are consistent with the reported research results that the TLR4 inhibitor and celecoxib inhibitor can reduce the cytokine release caused by SARS-CoV-2, which shows that the model of the invention can be used as a drug screening for clinical treatment of novel coronary patients.
Although specific embodiments of the invention have been described in detail, those skilled in the art will appreciate that: many modifications and variations of the details are possible in light of the above teachings, and such variations are within the scope of the invention. The full scope of the invention is given by the appended claims and any equivalents thereof.

Claims (2)

1. A method for constructing a model of SARS-CoV-2 antigen induced cytokine storm is characterized in that the method comprises adding 10nM SARS-CoV-2 spike protein and 10ng/mL IL-2 into a culture system of human peripheral blood mononuclear cells.
2. The method of claim 1, wherein the cell culture medium used in the culture system is a serum-free medium comprising human plasma.
CN202310628453.8A 2023-05-31 2023-05-31 Establishment and application of SARS-CoV-2 antigen induced cytokine storm model Active CN116355844B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310628453.8A CN116355844B (en) 2023-05-31 2023-05-31 Establishment and application of SARS-CoV-2 antigen induced cytokine storm model

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310628453.8A CN116355844B (en) 2023-05-31 2023-05-31 Establishment and application of SARS-CoV-2 antigen induced cytokine storm model

Publications (2)

Publication Number Publication Date
CN116355844A CN116355844A (en) 2023-06-30
CN116355844B true CN116355844B (en) 2023-08-18

Family

ID=86905333

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310628453.8A Active CN116355844B (en) 2023-05-31 2023-05-31 Establishment and application of SARS-CoV-2 antigen induced cytokine storm model

Country Status (1)

Country Link
CN (1) CN116355844B (en)

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111053909A (en) * 2020-02-27 2020-04-24 南通大学 Application of 2019-nCoV3CL hydrolase inhibitor and IL-6 monoclonal antibody in preparation of medicine for treating neocoronary pneumonia
CN112656798A (en) * 2020-12-28 2021-04-16 复旦大学附属肿瘤医院 Application of CDK7 targeted inhibitor in preparation of drug for treating cytokine release syndrome
CN113559107A (en) * 2020-04-28 2021-10-29 深圳埃格林医药有限公司 Application of progestogen in preparing medicine for inhibiting cell factor storm
CA3186973A1 (en) * 2020-06-24 2021-12-30 Bioniz, Llc Antagonistic peptide targeting il-2, il-9, and il-15 signaling for the treatment of cytokine-release syndrome and cytokine storm associated disorders
WO2022027110A1 (en) * 2020-08-03 2022-02-10 Bosnalijek, Farmaceutska I Hemijska Industrija, Dioničko Društvo Combination of tridecactide and methenkephalin for treating conditions involving cytokine storm
WO2022092389A1 (en) * 2020-11-02 2022-05-05 아레즈 주식회사 Composition for preventing or treating sars-cov-2, comprising pegylated albumin-bound steroidal ginsenoside derivative nanoparticles
EP3995829A1 (en) * 2020-11-09 2022-05-11 MediaPharma S.r.l. Lgals3bp as a diagnostic, prognostic, and therapeutic biomarker in covid-19 and other cytokine storm conditions
CN115044535A (en) * 2022-05-27 2022-09-13 华中科技大学同济医学院附属协和医院 In-vitro five-cell-factor storm model and preparation method and application thereof
CN115119509A (en) * 2020-02-18 2022-09-27 塞里维瑞疗法公司 Improved cell-permeable nuclear transport inhibitor synthetic peptide for inhibiting cytokine storm or inflammatory disease and use thereof
CN115461069A (en) * 2020-04-13 2022-12-09 遗传工程与生物技术中心 Peptides for the treatment of cytokine storm syndrome
CN115484953A (en) * 2020-03-27 2022-12-16 健艾仕生物医药有限公司 Methods of treating cytokine storm syndrome and related diseases

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115119509A (en) * 2020-02-18 2022-09-27 塞里维瑞疗法公司 Improved cell-permeable nuclear transport inhibitor synthetic peptide for inhibiting cytokine storm or inflammatory disease and use thereof
CN111053909A (en) * 2020-02-27 2020-04-24 南通大学 Application of 2019-nCoV3CL hydrolase inhibitor and IL-6 monoclonal antibody in preparation of medicine for treating neocoronary pneumonia
CN115484953A (en) * 2020-03-27 2022-12-16 健艾仕生物医药有限公司 Methods of treating cytokine storm syndrome and related diseases
CN115461069A (en) * 2020-04-13 2022-12-09 遗传工程与生物技术中心 Peptides for the treatment of cytokine storm syndrome
CN113559107A (en) * 2020-04-28 2021-10-29 深圳埃格林医药有限公司 Application of progestogen in preparing medicine for inhibiting cell factor storm
CA3186973A1 (en) * 2020-06-24 2021-12-30 Bioniz, Llc Antagonistic peptide targeting il-2, il-9, and il-15 signaling for the treatment of cytokine-release syndrome and cytokine storm associated disorders
WO2022027110A1 (en) * 2020-08-03 2022-02-10 Bosnalijek, Farmaceutska I Hemijska Industrija, Dioničko Društvo Combination of tridecactide and methenkephalin for treating conditions involving cytokine storm
WO2022092389A1 (en) * 2020-11-02 2022-05-05 아레즈 주식회사 Composition for preventing or treating sars-cov-2, comprising pegylated albumin-bound steroidal ginsenoside derivative nanoparticles
EP3995829A1 (en) * 2020-11-09 2022-05-11 MediaPharma S.r.l. Lgals3bp as a diagnostic, prognostic, and therapeutic biomarker in covid-19 and other cytokine storm conditions
CN112656798A (en) * 2020-12-28 2021-04-16 复旦大学附属肿瘤医院 Application of CDK7 targeted inhibitor in preparation of drug for treating cytokine release syndrome
CN115044535A (en) * 2022-05-27 2022-09-13 华中科技大学同济医学院附属协和医院 In-vitro five-cell-factor storm model and preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
丁芳.新型冠状病毒通过CCL2-CCR2介导巨噬细胞炎症反应的研究.《中国优秀硕士学位论文全文数据库医药卫生科技辑》.2023,(第02期),第E059-408页. *

Also Published As

Publication number Publication date
CN116355844A (en) 2023-06-30

Similar Documents

Publication Publication Date Title
Scharton et al. Natural killer cells are a source of interferon gamma that drives differentiation of CD4+ T cell subsets and induces early resistance to Leishmania major in mice.
Plevy et al. A role for TNF-alpha and mucosal T helper-1 cytokines in the pathogenesis of Crohn's disease.
Maldonado-López et al. Cytokines regulate the capacity of CD8α+ and CD8α− dendritic cells to prime Th1/Th2 cells in vivo
Delgado et al. Production of tumor necrosis factor-alpha, interleukin 1-beta, interleukin 2, and interleukin 6 by rat leukocyte subpopulations after exposure to substance P
CN105754941B (en) In-vitro induced amplification culture method for peripheral blood NK cells
JPH02109976A (en) Preparation of lak active limphoid cell
CN111454903B (en) Immune cell in vitro culture, induction, activation and cryopreservation method and cell bank establishment thereof
BONDER et al. Interleukin‐4 regulation of human monocyte and macrophage interleukin‐10 and interleukin‐12 production. Role of a functional interleukin‐2 receptor γ‐chain
Wei et al. IL-10-producing B cells attenuate cardiac inflammation by regulating Th1 and Th17 cells in acute viral myocarditis induced by coxsackie virus B3
CN111593022B (en) vMIP-II induced CD8+ T cell dephosphorylation to Tcm and its application in medicine
CN116355844B (en) Establishment and application of SARS-CoV-2 antigen induced cytokine storm model
Stevenson et al. Characterization of biological response modifier release by human monocytes cultured in suspension in serum-free medium
Donati et al. Uremic serum effects on peripheral blood mononuclear cell and purified T lymphocyte responses
Claman et al. The response of cultured human thymus cells to phytohemagglutinin
CN114507640B (en) Culture method and application of CIK cells with high proliferation capacity and high cytotoxicity
CN106267425A (en) AIDS immunoadsorption therapy instrument
CN106039448B (en) AIDS cell adsorbing therapy instrument
Van Dam et al. In vitro stimulation of goat peripheral blood lymphocytes: Optimization and kinetics of the response to mitogens and to allogeneic lymphocytes
CN115521914A (en) Human primary natural killer cell in-vitro amplification system and method
Chiba et al. RANTES and eotaxin enhance CD11b and CD18 expression on eosinophils from allergic patients with eosinophilia in the application of whole blood flow cytometry analysis
CN111154721B (en) NK cell amplification method
CN108690830B (en) Method for efficiently amplifying NKT cells
Zheng et al. Effects of lamivudine on the function of dendritic cells derived from patients with chronic hepatitis B virus infection
Gieni et al. Limiting dilution analysis of CD4 T-cell cytokine production in mice administered native versus polymerized ovalbumin: directed induction of T-helper type-1-like activation.
Swanson et al. Cortisol upregulates HIV p24 antigen production in cultured human monocyte-derived macrophages

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