[go: up one dir, main page]

WO2022121110A1 - Mechanism based on gastrointestinal symptoms caused by sars-cov-2 and application therefor - Google Patents

Mechanism based on gastrointestinal symptoms caused by sars-cov-2 and application therefor Download PDF

Info

Publication number
WO2022121110A1
WO2022121110A1 PCT/CN2021/075675 CN2021075675W WO2022121110A1 WO 2022121110 A1 WO2022121110 A1 WO 2022121110A1 CN 2021075675 W CN2021075675 W CN 2021075675W WO 2022121110 A1 WO2022121110 A1 WO 2022121110A1
Authority
WO
WIPO (PCT)
Prior art keywords
cov
sars
vegf
gastrointestinal
mice
Prior art date
Application number
PCT/CN2021/075675
Other languages
French (fr)
Chinese (zh)
Inventor
何欢欢
单鸿
曾发敏
何建忠
邓昭华
李颍雯
Original Assignee
中山大学附属第五医院
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 中山大学附属第五医院 filed Critical 中山大学附属第五医院
Publication of WO2022121110A1 publication Critical patent/WO2022121110A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61DVETERINARY INSTRUMENTS, IMPLEMENTS, TOOLS, OR METHODS
    • A61D7/00Devices or methods for introducing solid, liquid, or gaseous remedies or other materials into or onto the bodies of animals

Definitions

  • the present invention relates to the field of medical technology, and more particularly, to the mechanism and application of gastrointestinal symptoms caused by SARS-CoV-2.
  • COVID-19 is a severe acute respiratory illness caused by the SARS-CoV-2 virus that has caused a worldwide pandemic since its outbreak in late 2019.
  • SARS-CoV-2 infection the common feature is typical respiratory symptoms, and the airway is also identified as a target organ.
  • SARS-CoV-2 infection has been further studied, clinical reports of cases with gastrointestinal symptoms, which typically manifest as diarrhea, anorexia, nausea, and vomiting, continue to increase.
  • some patients infected with SARS-CoV-2 exhibited only digestive symptoms at the time of onset or even during the course of the disease, indicating that the gastrointestinal tract is highly susceptible to SARS-CoV-2.
  • the technical problem to be solved by the present invention is to overcome the defect that the mechanism of gastrointestinal symptoms caused by SARS-CoV-2 in the prior art is not clear. Gastrointestinal symptoms in patients with -19 provide an explanatory mechanism and a therapeutic strategy.
  • Angiotensin-converting enzyme maintains circulatory homeostasis by regulating angiogenesis, thrombosis, and vascular remodeling, and it has been reported that ACE2 is the major binding receptor for SARS-CoV-2.
  • ACE2 is expressed in many organs, including the digestive and vascular systems. Notably, human ACE2 is most highly expressed in the small intestine.
  • SARS-CoV-2 replicates in intestinal epithelial cells using ACE2 as an entry receptor using human intestinal organoids and a colon epithelial cancer cell line (Caco-2), respectively.
  • SARS-CoV-2 can also bind to ACE2 in other vertebrates, such as mice and rats, without being infected, raising the question of whether SARS-CoV-2 binding to ACE2 is possible in these organisms cause any signaling.
  • the present invention first provides a method of inhibiting SARS-CoV-2-induced damage to the vascular barrier in gastrointestinal tissue, comprising administering to the gastrointestinal tissue a vascular rejection targeting signal.
  • VEGF vascular permeability
  • SARS-CoV- 2 Spike The protein can regulate the phosphorylation of downstream ERK by binding to intestinal epithelial ACE2, thereby promoting the secretion of VEGF.
  • Secreted VEGF stimulated lower VE-cad expression or higher VE-cad phosphorylation in vascular endothelial cells. Increase vascular permeability.
  • ERK inhibitors can reduce the phosphorylation of pERK and the production of VEGF, thereby restoring the expression of VE-cad in vascular endothelial cells, indicating that ERK inhibitors can be used as reference drugs.
  • VEGF antagonists inhibit the binding of VEGF to VEGF receptors, thereby inhibiting the increase of vascular endothelial permeability.
  • the repulsive targeting signal includes an ERK inhibitor and/or a VEGF antagonist
  • the ERK inhibitor refers to a substance that can bind to ERK and block the biological activity of ERK
  • the VEGF antagonist refers to Substances that bind to VEGF and block the biological activity of VEGF.
  • the ERK inhibitors include clinical stage inhibitors SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996, MK-8353, CC-90003, LTT462, etc., and those in preclinical stage or biological activity evaluation stage
  • the VEGF antagonists include bevacizumab, ramucirumab, ranibizumab, aflibercept, conbercept and the like.
  • the above-mentioned ERK inhibitor is SCH772984, and the VEGF antagonist is bevacizumab.
  • the duodenum is the most sensitive to the SARS-CoV-2 Spike protein, so preferably, the above-mentioned gastrointestinal tract tissue is the duodenum.
  • the above study of the present invention also highlights the sensitivity of the duodenum to SARS-CoV-2 Spike-induced increased vascular permeability and its potential pathways, providing a potential therapeutic target for gastrointestinal symptoms in COVID-19 patients.
  • the present invention also provides a method for screening or evaluating an agent that inhibits SARS-CoV-2-induced damage to the vascular barrier in the gastrointestinal tract, specifically: determining the effect of the agent on the VEGF-mediated increase in the vascular permeability of the gastrointestinal tract. ability.
  • the present invention studies the mechanism of gastrointestinal symptoms caused by SARS-CoV-2, namely SARS-CoV-2 Spike protein directly binds to ACE2 on intestinal epithelial cells, activates the downstream ERK/VEGF signaling pathway, and induces increased vascular permeability.
  • SARS-CoV-2 Spike protein directly binds to ACE2 on intestinal epithelial cells, activates the upstream molecules of ERK Ras, Raf and MEK in intestinal epithelial cells, further activates ERK, promotes the secretion of VEGF from intestinal epithelial cells, and makes intestinal endothelial cells VE-cad Decreased expression or increased phosphorylation results in increased intestinal endothelial vascular permeability.
  • the ability to increase vascular permeability refers to determining the ability of the agent to inhibit ERK, MEK, Ras, Raf, VEGF.
  • the effect of the agent on the increase in vascular permeability of the gastrointestinal tract mediated by VEGF is determined by the following steps:
  • the endothelial cells in the candidate drug group can detect higher VE-cad expression or lower VE-cad phosphorylation level than the endothelial cells in the placebo group, indicating that the candidate drug can Inhibits VEGF-mediated increase in gastrointestinal vascular permeability.
  • the placebo described in step (2) is selected from PBS or DMEM.
  • said agent when in vivo, affects VEGF-mediated increase in gastrointestinal vascular permeability as determined by the following steps:
  • a lower expression level of VEGF can be detected in the gastrointestinal tract tissue of the animals after (3) treatment than in the gastrointestinal tract tissue of the animals after treatment (2), indicating that the agent can Inhibits VEGF-mediated increase in gastrointestinal vascular permeability.
  • the placebo described in step (2) is selected from PBS or DMEM.
  • the gastrointestinal tract tissue in step (4) is the duodenum.
  • the construction method of the SARS-CoV-2 gastrointestinal inflammation animal model in step (1) includes the following steps:
  • mice were intraperitoneally injected with SARS-CoV-2 Spike protein;
  • SARS-CoV-2 Spike protein in the present invention Before intraperitoneal injection of SARS-CoV-2 Spike protein in the present invention, acid enema treatment is performed on the epithelium of the gastrointestinal tract, and then SARS-CoV-2 Spike protein stimulation is more conducive to the binding of SARS-CoV-2 Spike protein; the animal model obtained in this way is consistent with the clinical symptoms of COVID-19 patients reported in the present, so the animal model constructed by the method of the present invention can replicate clinical symptoms Gastrointestinal manifestations in patients with COVID-19.
  • the SARS-CoV-2 described in step S3 The dosage of Spike protein was 5.5 nM/kg mouse body weight.
  • step S4 refers to 6-24 hours.
  • mice were fed normally. After 16-24 hours, the candidate drugs were injected into the mice through the abdominal cavity respectively to obtain the mice in the candidate drug group; the placebo was injected into the mice through the abdominal cavity respectively to obtain the mice in the placebo group. ;
  • mice in the candidate drug group and the placebo group were injected with SARS-CoV-2 by intraperitoneal injection.
  • Spike protein mice in the control group were intraperitoneally injected with the same amount of IgG-Fc protein;
  • mice in the control group were taken respectively.
  • the gastrointestinal tissues of the mice in the placebo group were able to detect higher
  • the gastrointestinal tissue of the candidate drug group could detect a lower VEGF expression level, indicating that the candidate drug can inhibit the VEGF-mediated gastrointestinal vascular permeability Increase.
  • the concentration of acetic acid in the above step (2) is 1-2% v/v, and the volume is 500 ⁇ L to 1000 ⁇ L.
  • the SARS-CoV-2 described in the above step (4) The dosage of Spike protein was 5.5nM/kg mouse body weight.
  • the present invention also provides a method for treating or preventing gastrointestinal symptoms caused by SARS-CoV-2, characterized by comprising the following steps:
  • the repulsive guidance signal includes an ERK inhibitor and/or a VEGF antagonist
  • the ERK inhibitor refers to a substance that can bind to ERK and block the biological activity of ERK
  • the VEGF antagonist refers to a substance that can interact with VEGF Substances that bind to and block the biological activity of VEGF.
  • the ERK inhibitors include clinical stage inhibitors SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996, MK-8353, CC-90003, LTT462, etc., which are in the preclinical stage or the biological activity evaluation stage.
  • Inhibitors FR180204, VTX-11e, BL-EI-001, etc.; the VEGF antagonists include bevacizumab, ramucirumab, ranibizumab, aflibercept, conbercept and the like.
  • the above-mentioned ERK inhibitor is SCH772984
  • the VEGF antagonist is the anti-VEGF drug bevacizumab.
  • the present invention also provides a medicament for treating or preventing or alleviating gastrointestinal symptoms caused by SARS-CoV-2, the medicament having at least one of the following functions:
  • the drugs include, but are not limited to: ERK inhibitors and/or VEGF antagonists.
  • the ERK inhibitors include clinical stage inhibitors SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996, MK-8353, CC-90003, LTT462, etc., which are in the preclinical stage or the biological activity evaluation stage.
  • Inhibitors FR180204, VTX-11e, BL-EI-001, etc.; the VEGF antagonists include bevacizumab, ramucirumab, ranibizumab, aflibercept, conbercept and the like.
  • the present invention has the following beneficial effects:
  • the present study found that the SARS-CoV-2 Spike protein can combine with human and mouse ACE2 to induce increased vascular permeability.
  • Spike protein activates the Ras-Raf-MEK-ERK pathway by binding to ACE2 of intestinal epithelial cells, and promotes the secretion of VEGF from epithelial cells, which does not exist in endothelial cells.
  • ERK or VEGF blockade rescued Spike protein-enhanced vascular permeability in vivo and alleviated gastrointestinal symptoms.
  • Figure 1a shows the co-localization of ACE2 and SARS-CoV-2 Spike protein observed in the gastrointestinal tissue of COVID-19 patients;
  • Figure 1b shows the local inflammation in H&E stained gastrointestinal tissue;
  • Figure 2a shows the construction process of the animal model
  • Figure 2b shows the HE staining images of different parts of the experimental group (Spike-Fc) mice and the control group (Control-Fc) mice
  • Figure 2c and Figure 2d show the experimental group (Spike-Fc) mice.
  • Figure 3c shows that the HUVECs of experimental group (Spike-Fc) and control group (Control-Fc) were determined by WB
  • Figure 4a shows the pull-down results of Spike-Fc protein and human ACE2 in Caco-2 cells and 239T cells, respectively, where Input represents total cell lysate, and IP:Fc represents the protein pulled down by Fc peptide;
  • Figure 4b shows Spike-Fc Fluorescent staining results of Fc protein and human ACE2 in 239T cells;
  • Figure 5a shows the detection of cohesin ZO-1, VE-cad, pVE-cad (Y658) and pVE-cad (Y731) in the supernatant of the co-culture system of Caco-2-HUVECs incubated with Spike-Fc or Control-Fc by WB.
  • Figure 5b and Figure 5c show VE-cad and IHC plot of pVE-cad(Y731), the scale bar is 100 ⁇ m, and the scatter plot shows the expression levels of VE-cad and pVE-cad(Y731) in the duodenum, jejunum, colon and rectum (data are presented as mean ⁇ SD represents, p value is by t test, ns represents no significance, * represents p ⁇ 0.05, ** represents p ⁇ 0.01); Figure 5d and Figure 5e show VE-cad in duodenum of healthy and COVID-19 patients and the detection of pVE-cad (Y731) (data are represented by mean ⁇ SD,
  • Figure 6a shows the RT-PCR analysis of VEGF transcript levels in the duodenum, jejunum, colon and rectum of experimental (Spike-Fc) and control (Control-Fc) mice (data are expressed as mean ⁇ SD, The p value was tested by t-test, * represents p ⁇ 0.05);
  • Figure 6b shows the ELISA analysis of VEGF in the duodenum, jejunum, colon and rectum of experimental (Spike-Fc) and control (Control-Fc) mice The scatter plot shows the protein concentration of VEGF in intestinal tissue (data are represented by mean ⁇ SD, p value is by t test, ns represents no significance); The amount of VEGF secreted by Cntrol-Fc-treated Caco-2 cells;
  • Figure 6d shows the amount of VEGF in plasma of mice treated with Spike-Fc or Control-Fc was analyzed by ELISA; scatter plot shows the protein of VEGF in plasma Concentration (data are represented by mean ⁇ SD,
  • Figure 7 shows immunoblot analysis of ERK or pERK in Caco-2 cells treated with Spike-Fc or Control-Fc; histograms show quantification of protein expression relative to the amount of ⁇ -actin by densitometric scanning. Data are represented by mean ⁇ SD, p value is by paired t test, * represents p ⁇ 0.05;
  • Figure 8a shows the detection of Caco-2 siRNA (with three siRNA knockdown sequences 01, 02 and 03) by western blot. Compared with the control sequence (NC), all three sequences showed obvious knockdown effect, and the endogenous ACE2 After knockdown, the expressions of Ras, C-Raf, pMEK, pERK and p-P90RSK were significantly increased;
  • Figure 8b shows that after stimulating HUVCE cells with Control-Fc or Spike-Fc protein for 1 h, the expression of ERK and pERK proteins was detected by western blotting happening;
  • Figure 9a shows the expression levels of each protein in the Ras-Raf-MEK-ERK signaling pathway after Spike-Fc, Control-Fc and SCH772984 treatment of Caco-2 cells, the histogram shows the protein expression relative to ⁇ -actin was quantified by protein grayscale The experimental data were repeated three times, the data were represented by the mean ⁇ SD, the p value was tested by paired t test, * represents p ⁇ 0.05, ns represents no significance;
  • Figure 9b shows that Spike-Fc, Control-Fc and SCH772984 were compared with Caco After the -2 cells were co-cultured, the amount of VEGF was determined by ELISA, the data were expressed as mean ⁇ SD, the p value was expressed by t test, * represents p ⁇ 0.05;
  • Figure 9c shows the experimental group (Spike-Fc) and the control group (Control-Fc) Fc) IHC map of ERK and pERK in intestinal tissues (duodenum, jejun
  • Figure 10a shows the expression and localization of VE-cad in Caco-2 cells treated with Spike-Fc or Control-Fc or SCH772984 or Bevacizumab
  • Figure 10c shows the experimental group (Spike-Fc) and the control group (Control-Fc) intraperitoneal injection of SCH772984 or Bevacizumab model mice intestinal tissue (12 IHC plots of VE-cad and pVE-cad (Y731) in the denum, jejunum, ileum, colon and
  • Figure 11a shows the use of ELISA to analyze the expression of VEGF in the intestinal tissues (duodenum, jejunum, colon and rectum) of model mice injected with SCH772984 or Bevacizumab in the experimental group (Spike-Fc) and the control group (Control-Fc) by intraperitoneal injection.
  • FIG. 11b shows the experimental group (Spike-Fc) and the control group (Control-Fc) ) or immunoblot analysis of ERK or pERK in intestinal tissues (duodenum, jejunum, colon, and rectum) of model mice injected with SCH772984 or Bevacizumab intraperitoneally;
  • the amount of ⁇ -actin, the experimental data was repeated three times, the data were expressed as mean ⁇ SD, and the p value was expressed by paired t test, *represents p ⁇ 0.05, **represents p ⁇ 0.01, and ***represents p ⁇ 0.0001;
  • Figure 12b shows the evens permeated per unit weight (g) of intestinal tissue (duodenum, jejunum, colon and rectum) The OD value of blue dye;
  • Figure 12c shows the experimental group (Spike-Fc) and the control group (Control-Fc) or the intestinal tissue (duodenum, jejunum, colon and rectum) of model mice injected with SCH772984 or Bevacizumab intraperitoneally ) of H&E staining; yellow arrows represent inflammatory infiltrates; red stars represent edema, and no inflammation, moderate inflammation, and severe inflammation were assessed in the intestinal tissue; his
  • Figure 13 shows the model mechanism of SARS-CoV-2 Spike protein-mediated vascular hyperpermeability and intestinal tissue inflammation.
  • the clinical data of 17 patients are shown in Supplementary Table 1. Ethically approved by the Ethics Committee of the Fifth affiliated Hospital of Sun Yat-Sen University (No. 2020L029-1), all patients signed informed consent.
  • Human umbilical vein endothelial cells were purchased from ScienCell (Cat. No. 8000, Cat. No. 5000) in ECM medium (ScienCell, Cat. No. 5000) supplemented with 10% fetal bovine serum. 1001) in culture.
  • Human colorectal adenocarcinoma cells (Caco-2) were purchased from (Guangzhou IGE Biotechnology Company, Guangzhou) and cultured in Dulbecco's modified Eagle's medium (Gibco) supplemented with 10% fetal bovine serum, 50 U /mL penicillin and 50 mg/mL streptomycin (Gibco, catalog number 15140-122).
  • a murine endothelial cell line (C166) was obtained from the American Biological Resource Center (ATCC, Manassas, USA), supplemented with 10% fetal bovine serum, 50 U/mL penicillin, and 50 mg of Dulbecco's modified Eagle's medium ( Gibco). Cells were cultured in a humidified 37°C incubator with 5% CO . The following studies were approved by the Fifth affiliated Hospital of Sun Yat-Sen University. For all animal experiments, the permission of the Experimental Animal Ethics Committee of the Fifth affiliated Hospital of Sun Yat-sen University was obtained. Statistical analysis: Statistical analysis was performed using SPSS v13.0 software.
  • SARS-CoV-2 Spike Protein preferentially induces duodenal interstitial edema
  • Tissue sections Clinical specimens from COVID-19 patients were formalin-fixed, paraffin-embedded and sectioned (4 ⁇ m).
  • Sections need to be placed in 10% goat serum prepared in PBST, incubated at room temperature for 1 hour, and then added with primary antibodies (anti-ACE2, Santa Cruz, sc390851, 1:100; anti-SARS-Cov-2 Spike, Sino Biological, 40150-R007, 1:500), the sections were incubated at 4°C overnight. The next day, wash 3 times with PBST and add fluorescent secondary antibodies (AlexaFluor® 647-conjugated goat anti-rabbit IgG, bs-0296G-AF647, Bioss, 1:100; Dylight-550 goat anti-rabbit IgG secondary antibody BA1135, 1:200 ) at room temperature for 1 h.
  • primary antibodies anti-ACE2, Santa Cruz, sc390851, 1:100; anti-SARS-Cov-2 Spike, Sino Biological, 40150-R007, 1:500
  • RESULTS Using double immunofluorescence staining, co-localization of ACE2 and SARS-CoV-2 Spike protein was observed in the duodenum in gastrointestinal tissues obtained by endoscopy from patients with COVID-19, whereas in the duodenum Spike proteins were detectable in the denum (Fig. 1a). H&E staining further showed marked edema of the mucosal lamina limba interstitium, local inflammation with plasma cell and lymphocyte infiltration (Fig. 1b).
  • interstitial edema was significantly associated with disease type, acid reflux, total bilirubin, glutamate-pyruvate aminotransferase (ALT) and aspartate aminotransferase (AST) (Table 2), suggesting progression of the disease prediction features.
  • mice 8-9 weeks old C57BL/6J mice were randomly divided into two groups (experimental group and control group); (2) all mice were fasted for 24 hours; (3) after fasting for 24 hours, the abdominal cavity of each mouse was Inject 100 ⁇ L of 1% pentobarbital for anesthesia; (4) After 5 minutes of anesthesia, apply petroleum jelly on the surface of the hose, and then gently insert it from the anus to a depth of 4 cm; (5) Connect a syringe to one end of the hose and inject 500 ⁇ L of 1% acetic acid, and the control group was injected with an equal volume of PBS; (6) the mice were inverted for 1 min, and then lavaged with 500 ⁇ L of PBS twice to wash off the injected acetic acid; (7) The mice were put back into the cage and supplemented with food; (8) 16-24 hours later, the S protein (SARS-CoV-2 Spike protein) 5 ⁇ g/mice (or 5.5 nmol/kg) was intraperitoneally injected, and the control
  • SARS-CoV-2 gastrointestinal inflammation model mice and control groups (Mouse IgG1-Fc protein) was injected into the tail vein of TRITC-dextran, and after half an hour, the peritoneal cavity was washed with 2.5 mL of PBS; after taking the ascites, the ascites was centrifuged at 1500 rpm for 10 min, and the excitation and emission wavelengths were 540 nm and 590 using a microplate reader, respectively. Fluorescence was measured at nm; duodenal, jejunum, colon and rectal tissues were taken after the mice were sacrificed, embedded and dehydrated, and then sliced and stained with H&E.
  • mice were intravenously injected with TRITC-dextran, followed by quantitative detection of dextran leaking into the abdominal cavity. Greater leakage was observed in mice treated with Spike-Fc compared to controls (Control-Fc) (Fig. 3a), suggesting that Spike proteins can cause impairment of the intestinal vascular barrier.
  • ACE2 has been reported to be expressed in various cell types including endothelial cells, so here we first investigated whether Spike proteins could mediate permeability by directly affecting the endothelium.
  • HAVEC human umbilical vein endothelial cells
  • Spike protein did not significantly affect the permeability of HUVEC cells (Fig. 3b).
  • ACE2 is the main binding receptor of SARS-CoV-2.
  • ACE2 is expressed in a variety of organs, including the digestive and vascular systems.
  • Caco-2 colon epithelial cancer cell line
  • SARS-CoV-2 replicates in intestinal epithelial cells using ACE2 as an entry receptor
  • the in vivo permeability experiments described above give Therefore, it is reasonable to speculate that Spike protein induces endothelial permeability by affecting intestinal epithelial cells.
  • HUVEC cells were incubated with conditioned medium of Spike-Fc-treated intestinal epithelial cells Caco-2, as determined by permeability, using Spike-Fc (0.25 mg/mL) for Caco-2 (5 ⁇ 10 5 ), the control group was treated with IgG-Fc (0.25mg/mL) for 24h, and then the cultured Caco-2 cell supernatant was filtered through a 0.22 ⁇ m filter.
  • HUVEC cells (1 x 105 ) were plated in a monolayer in the upper chamber of a 24-well plate overnight.
  • the wells were pipetted into the same 24-well plate, medium with 1% FBS was added until the HUVECs reached confluency for 6 h, and then the filtered Caco-2 supernatant was replaced to the bottom of the Transwell chamber and co-cultured with HUVEC for 12 h.
  • the upper chamber medium was removed, and tetramethylrhodamine isothiocyanate-Dextran (T1162, Sigma) (2 mg/mL) was added to the upper wells. After 3 hours, the Dextran-added cells were collected. culture medium, and fluorescence was measured at excitation and emission wavelengths of 540 nm and 590 nm, respectively, using a microplate reader.
  • Recombinant SARS-CoV-2 Spike protein (RBD, Fc tag) was purchased from Sino Bioloical.
  • Caco-2 cells and cells were treated with Spike-Fc (0.25 mg/mL) or control, respectively IgG-Fc (0.25 mg/mL) was incubated at 37 °C for 1 h, and then the protein in the lysate was pulled down with Protein G Sepharose for immunoblotting.
  • Cell lysates were analyzed by western blot and quantitative PCR, and cell culture supernatants (24 h) were filtered through 0.22 ⁇ m filters (MILLEX GP) and stored at -80°C for Elisa and co-culture experiments.
  • ERK, pERK, VE-cad and pVE-cad(Y731) proteins were assessed for tissue samples.
  • HE staining was also performed on human/mouse tissue sections to assess tissue morphological characteristics and distribution of target proteins.
  • Tight junctions and adherent junctions are the basic components of the intestinal vascular barrier. Changes in tight junction proteins such as ZO-1 were first analyzed, but no significant changes were found. However, the expression of a key adhesion protein, VE-cadherin (VE-cad), was reduced, accompanied by an increase in its phosphorylation at Tyr731 but not at 658 (pVE-cad) (Fig. 5a). Then, we measured the expression and phosphorylation levels of VE-cad in vivo. Different parts of the gastrointestinal tract of mice treated with Spike protein were analyzed by western blot and immunohistochemistry (IHC). The results showed a persistent decrease in VE-cad in the duodenum (Fig.
  • SARS-CoV-2 Spike protein through ERK/VEGF pathway mediates vascular permeability
  • RNA from tissues was isolated using TRIzol by conventional RNA extraction protocols.
  • Total RNA from cells was isolated with Total RNA Kit I.
  • the isolated RNA was reverse transcribed into cDNA (Vazyme, Nanjing, China).
  • qRT-PCR was performed using a real-time PCR system (Bio-Rad, America) and ChamQ Universal SYBR qPCR master mix (Vazyme, Nanjing, China). Primers were designed and synthesized by Guangzhou IGE Biotechnology Company. GAPDH was used as an internal control and all reactions were repeated three times. Relative RNA expression was calculated using the 2 - ⁇ Ct method.
  • VEGF concentration of VEGF produced from cells, tissues and serum was detected by ELISA.
  • Quantikine ELISA human VEGF immunoassay catalog. No. DVE00, R&D
  • Determination of VEGF concentration for mouse samples, use mouse VEGF according to the manufacturer's instructions Simplestep ELISA kit (cat. No. ab209882, Abcam) to measure VEGF levels in tissue homogenates (prepared in cold PBS using an electric homogenizer) and serum.
  • VEGF known as a potent vascular permeability factor
  • SARS-CoV-2 infection significantly increases VEGF expression in human lung epithelial cells.
  • enterocytes which may be responsible for gastrointestinal symptoms in COVID-19 patients. The above experimental data verified our expectation.
  • Caco-2 cells were attached to a six-well plate; (2) Lipofectamine was used LTX (Invitrogen, ThermoFisher Scientific Corporation) transfection reagent, and the negative control siRNA and ACE2 siRNA were transfected into Caco-2 cells; (3) After 48h, use RIPA buffer containing protease/phosphatase inhibitor mixture to lyse Caco-2, and extract the protein; (4) Western blot was used to detect the knockdown efficiency and the expression of related proteins.
  • LTX Invitrogen, ThermoFisher Scientific Corporation
  • Caco-2 was transfected with negative control siRNA and ACE2 siRNA for 48 hours using Lipofectamine LTX (Invitrogen, ThermoFisher Scientific), then Caco-2 was lysed using RIPA buffer containing a protease/phosphatase inhibitor cocktail in preparation for a western blot.
  • Caco-2 (5 ⁇ 10 5 ) was treated with Spike-Fc (0.25mg/mL), control group was treated with IgG-Fc (0.25mg/mL), Spike-Fc with SCH772984 (1uM), bevacizumab Bevacizumab (25ug/ml) cells were treated for 24h, and then the cultured Caco-2 cell supernatant was filtered through a 0.22 ⁇ m filter. A monolayer of HUVEC ( 1 x 105) was plated in the upper chamber of a 24-well plate overnight.
  • the wells were pipetted into the same 24-well plate, medium with 1% FBS was added until the HUVEC cells reached confluency for 6 h, and then the filtered Caco-2 cell supernatant was added. The solution was changed to the bottom of the Transwell chamber and co-cultured with HUVEC cells for 12 h.
  • the upper chamber medium was removed, and tetramethylrhodamine isothiocyanate-Dextran (T1162, Sigma) (2 mg/mL) was added to the upper well. After 3 hours, the collection was added with Dextran of medium, and measured fluorescence using a microplate reader at excitation and emission wavelengths of 540 nm and 590 nm, respectively.
  • mice 8-9 weeks old C57BL/6J mice were randomly divided into six groups: control group, placebo group I, placebo group II, Spike protein group, SCH772984 group and bevacizumab group; (2) All mice were fasted for 24 hours; (3) After fasting for 24 hours, each mouse was anesthetized by intraperitoneal injection of 100 ⁇ L of 1% pentobarbital; (4) After 5 minutes of anesthesia, the surface of the hose was smeared with Vaseline, and then injected from the anus.
  • the insertion depth is 4cm; (5) Connect a syringe to one end of the hose, inject 500 ⁇ L of 1% acetic acid, and the control group is injected with an equal volume of PBS; (6) Invert the mouse for 1 min, and then use 500 ⁇ L of PBS for Lavage, lavage twice, in order to wash off the injected acetic acid; (7) Put the mice back in the cage and supplement with food; (8) After 16-24 hours, the mice in the SCH772984 group were injected intraperitoneally at a dose of 50 mg/kg, The bevacizumab group was injected intraperitoneally at a dose of 5 mg/kg, the placebo group I mice were intraperitoneally injected with the solvent of SCH772984, the placebo group II mice were intraperitoneally injected with the bevacizumab solvent, and the control group did not.
  • the SCH772984 group, the bevacizumab group, the placebo group I, and the placebo group II were given intraperitoneal injection of S protein (SARS-CoV-2 Spike protein) 5 ⁇ g / animal (or 5.5 ⁇ g) respectively.
  • the control group was injected with the same type control Mouse IgG1-Fc protein; (10) 1 ⁇ 2h, the mice in the SCH772984 group were again injected intraperitoneally at a dose of 50 mg/kg, the bevacizumab group was injected at a dose of 5 mg/kg, and the placebo group I was injected with SCH772984 (11) TRITC-dextran was injected into the tail vein after 4-6 hours, and 2.5ml of PBS was washed in the abdominal cavity after half an hour; (12) after taking the ascites, the ascites was rotated at 1500rpm.
  • mice were sacrificed, and the duodenum, colon, jejunum, ileum, and rectum were taken, and some tissues were embedded, dehydrated, and sliced. After HE staining; and immunofluorescence staining experiments were performed to investigate the co-localization of ACE2 and SARS-CoV-2. The content of VEGF in some fresh tissues was detected by Elisa kit.
  • HUVECs were seeded into 15 mm glass bottom cell culture dishes (2.5 ⁇ 10 5 ) and co-cultured with the supernatant of Caco-2 (treated with Spike-Fc and IgG-Fc, respectively) for 24 h. It was then fixed with 4% paraformaldehyde. Samples were stained sequentially with the following antibodies or fluorescent dyes: VE-Cadherin (Cat.No.44-1145G, ThermoFisher, 1:1,000), Dylight-488 Goat Anti-mouse IgG secondary antibody (Cat.No. BA1126, BOSTER, 1:200), Antifade Mounting Medium with DAPI (Cat. No. Ab104139, Abcam). The results were acquired with a confocal microscope, and the sample pictures were taken by a Zeiss 880 with a 60x objective lens, and image processing was performed using ImageJ software.
  • VE-Cadherin Cat.No.44-1145G, ThermoFisher, 1:1,000
  • mice were fasted for 24 hours in advance; (2) 100ul of 1% pentobarbital was injected intraperitoneally into the mice; (3) After the mice were anesthetized for about 5 minutes, apply Vaseline to the surface of the tube, and the tube was removed from the anus.
  • mice in the SCH772984 group were injected intraperitoneally according to the dosage of SCH772984: 50 mg/kg, and the mice in the bevacizumab group were intraperitoneally injected with the dosage of 5 mg/kg , the mice in the placebo group I were intraperitoneally injected with the solvent of SCH772984, and the mice in the placebo group II were injected with the solvent of bevacizumab, and the control group was not treated; (8) SCH772984 group, bevacizumab group and Placebo group I and placebo group II were intraperitoneally injected with S protein (SARS)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to the field of medical technology. Mechanistic studies have found that the SARS-CoV-2 Spike protein activates an Ras-Raf-MEK-ERK pathway by means of binding to ACE2 receptors of intestinal epithelial cells, thereby promoting the secretion of VEGF by intestinal epithelial cells. However, the pathway is not altered in endothelial cells. An ERK or VEGF blocker can remedy Spike protein-enhanced vascular permeability and alleviate gastrointestinal symptoms in mice. The results indicate that in gastrointestinal tracts, especially in duodenum in which ACE2 is highly expressed, the SARS-CoV-2 Spike protein can directly bind to ACE2 in the intestinal epithelial cells, activate a downstream ERK/VEGF signal pathway, and induce an increase in vascular permeability, thereby leading to inflammation-related gastrointestinal symptoms. The present study provides an explanatory mechanism and a treatment strategy for gastrointestinal symptoms in patients having COVID-19.

Description

基于SARS-CoV-2引起的胃肠道症状的机制及其应用Mechanisms and applications of gastrointestinal symptoms based on SARS-CoV-2 技术领域technical field
本发明涉及医药技术领域,更具体地,涉及基于SARS-CoV-2引起的胃肠道症状的机制及其应用。The present invention relates to the field of medical technology, and more particularly, to the mechanism and application of gastrointestinal symptoms caused by SARS-CoV-2.
背景技术Background technique
COVID-19是由SARS-CoV-2病毒引起的严重的急性呼吸系统疾病,自2019年底爆发以来已经在全世界范围内引起了大流行。SARS-CoV-2感染后,共同特征是典型的呼吸道症状,人们也将气道认定为靶器官。然而,随着SARS-CoV-2感染研究深入,出现胃肠道症状的病例的临床报告不断增多,这些症状通常表现为腹泻、厌食、恶心和呕吐。此外,一些感染SARS-CoV-2的患者在发病时甚至在病程中仅表现出消化系统症状,说明胃肠道对SARS-CoV-2是高度易感的。COVID-19 is a severe acute respiratory illness caused by the SARS-CoV-2 virus that has caused a worldwide pandemic since its outbreak in late 2019. After SARS-CoV-2 infection, the common feature is typical respiratory symptoms, and the airway is also identified as a target organ. However, as SARS-CoV-2 infection has been further studied, clinical reports of cases with gastrointestinal symptoms, which typically manifest as diarrhea, anorexia, nausea, and vomiting, continue to increase. In addition, some patients infected with SARS-CoV-2 exhibited only digestive symptoms at the time of onset or even during the course of the disease, indicating that the gastrointestinal tract is highly susceptible to SARS-CoV-2.
对这些表现出胃肠道症状的病人进行检测,病理上腹部CT显示肠壁增厚。值得注意的是,在ICU患者中尤其能观察到小肠壁增厚。然而,COVID-19患者胃肠道易感部位的系统特征以及胃肠道症状发生的机制尚不清楚。COVID-19患者胃肠道症状常表现为炎症细胞浸润和间质水肿,提示肠道血管通透性受损。虽然呼吸系统是SARS-CoV-2的主要靶点,但血管系统也会受到破坏,如临床出现的心肌梗死、肠系膜缺血和肠内皮炎等血管相关并发症,这表明内皮细胞可能是病毒的直接目标。事实上,病毒直接感染内皮细胞的证据最近已经出现。然而,胃肠道炎症和血管通透性改变是内皮感染的结果,还是由周围细胞对病毒的反应引起的继发性影响,目前尚不清楚。In these patients with gastrointestinal symptoms, pathological upper abdominal CT showed thickening of the bowel wall. Notably, thickening of the small bowel wall was especially observed in ICU patients. However, the systemic characteristics of susceptible sites in the gastrointestinal tract of patients with COVID-19 and the mechanisms underlying the development of gastrointestinal symptoms remain unclear. Gastrointestinal symptoms in COVID-19 patients often manifest as inflammatory cell infiltration and interstitial edema, suggesting impaired intestinal vascular permeability. While the respiratory system is the primary target of SARS-CoV-2, the vascular system is also compromised, with clinically occurring vascular-related complications such as myocardial infarction, mesenteric ischemia, and intestinal endotheliitis, suggesting that endothelial cells may be the source of the virus direct target. In fact, evidence for direct viral infection of endothelial cells has recently emerged. However, whether gastrointestinal inflammation and altered vascular permeability are the result of endothelial infection or secondary effects arising from the response of surrounding cells to the virus is unclear.
技术问题technical problem
本发明所要解决的技术问题是克服现有技术基于SARS-CoV-2引起的胃肠道症状的机制不明确的缺陷,研究了基于SARS-CoV-2引起的胃肠道症状的机制,为COVID-19患者出现的胃肠道症状提供了一种解释机制和治疗策略。The technical problem to be solved by the present invention is to overcome the defect that the mechanism of gastrointestinal symptoms caused by SARS-CoV-2 in the prior art is not clear. Gastrointestinal symptoms in patients with -19 provide an explanatory mechanism and a therapeutic strategy.
血管紧张素转换酶(ACE2)通过调节血管生成、血栓形成和血管重塑来维持循环系统的稳态,已有报道表明ACE2是SARS-CoV-2的主要结合受体。ACE2在许多器官中表达,包括消化系统和血管系统。值得注意的是,人类ACE2在小肠中表达最高。有两项研究分别利用人小肠类器官和结肠上皮癌细胞株(Caco-2)证明,SARS-CoV-2以ACE2作为进入受体在肠上皮细胞中复制。SARS-CoV-2也可以与其他脊椎动物的ACE2结合,如小鼠和大鼠,而不被感染,这就提出了一个问题:在这些生物体内,SARS-CoV-2与ACE2的结合是否可能导致任何信号传导。Angiotensin-converting enzyme (ACE2) maintains circulatory homeostasis by regulating angiogenesis, thrombosis, and vascular remodeling, and it has been reported that ACE2 is the major binding receptor for SARS-CoV-2. ACE2 is expressed in many organs, including the digestive and vascular systems. Notably, human ACE2 is most highly expressed in the small intestine. Two studies have demonstrated that SARS-CoV-2 replicates in intestinal epithelial cells using ACE2 as an entry receptor using human intestinal organoids and a colon epithelial cancer cell line (Caco-2), respectively. SARS-CoV-2 can also bind to ACE2 in other vertebrates, such as mice and rats, without being infected, raising the question of whether SARS-CoV-2 binding to ACE2 is possible in these organisms cause any signaling.
在这里,我们研究了SARS-CoV-2 Spike / ACE2介导的胃肠道及其相关病理和临床症状的信号通路,这可能为感染COVID-19患者的肠血管屏障损伤的修复和胃肠道症状的缓解提供治疗靶点。Here, we investigated SARS-CoV-2 Spike/ACE2-mediated signaling pathways in the gastrointestinal tract and its associated pathology and clinical symptoms, which may provide insights into the repair of intestinal vascular barrier damage and the gastrointestinal tract in patients infected with COVID-19 Relief of symptoms provides a therapeutic target.
技术解决方案technical solutions
本发明的目的通过以下技术方案实现:The object of the present invention is achieved through the following technical solutions:
本发明首先提供了抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的方法,包括给胃肠道组织施用血管的排斥性导向信号。The present invention first provides a method of inhibiting SARS-CoV-2-induced damage to the vascular barrier in gastrointestinal tissue, comprising administering to the gastrointestinal tissue a vascular rejection targeting signal.
在这里,我们研究了SARS-CoV-2 Spike / ACE2介导的胃肠道及其相关的病理和临床结果的信号通路,我们已经确定了SARS-CoV-2 Spike蛋白引起肠道血管通透性和胃肠道炎症的可能途径和机制。我们的研究数据表明,通过与ACE2结合,Spike蛋白通过激活肠上皮细胞中的Ras-Raf-MEK-ERK途径诱导VEGF的产生,从而导致VE-cad介导的血管通透性过高,最终导致组织炎症和水肿。阻断ERK或VEGF能够减轻Spike刺激引起的胃肠道症状。Here, we investigate SARS-CoV-2 Spike/ACE2-mediated signaling pathways in the gastrointestinal tract and its associated pathology and clinical outcomes, and we have identified SARS-CoV-2 Possible pathways and mechanisms of Spike protein-induced intestinal vascular permeability and gastrointestinal inflammation. Our data suggest that by binding to ACE2, Spike protein induces VEGF production by activating the Ras-Raf-MEK-ERK pathway in intestinal epithelial cells, resulting in VE-cad-mediated vascular hyperpermeability, ultimately leading to Tissue inflammation and edema. Blockade of ERK or VEGF was able to alleviate gastrointestinal symptoms induced by Spike stimulation.
我们在新冠病人肠道组织切片染色中发现有水肿和炎性细胞浸润等现象,推测了血管通透性发生了改变,VEGF是影响血管通透性的重要因素,于是我们验证了SARS-CoV-2的Spike 蛋白能够通过结合肠上皮ACE2调控下游ERK的磷酸化增加,进而促进VEGF的分泌。分泌的VEGF刺激了血管内皮细胞更低VE-cad表达量,或更高VE-cad磷酸化水平。使血管通透性增加。针对这一过程,使用ERK抑制剂可以降低pERK的磷酸化,并能降低VEGF的产生,进而恢复血管内皮细胞VE-cad表达量,说明ERK抑制剂可以作为参考药物。而VEGF拮抗剂抑制VEGF与VEGF受体结合,从而抑制了血管内皮通透性的增加。We found edema and inflammatory cell infiltration in the staining of intestinal tissue sections of patients with COVID-19, and speculated that vascular permeability had changed, and VEGF was an important factor affecting vascular permeability, so we verified SARS-CoV- 2 Spike The protein can regulate the phosphorylation of downstream ERK by binding to intestinal epithelial ACE2, thereby promoting the secretion of VEGF. Secreted VEGF stimulated lower VE-cad expression or higher VE-cad phosphorylation in vascular endothelial cells. Increase vascular permeability. For this process, the use of ERK inhibitors can reduce the phosphorylation of pERK and the production of VEGF, thereby restoring the expression of VE-cad in vascular endothelial cells, indicating that ERK inhibitors can be used as reference drugs. VEGF antagonists inhibit the binding of VEGF to VEGF receptors, thereby inhibiting the increase of vascular endothelial permeability.
因此,优选的,所述排斥性导向信号包括ERK抑制剂和/或VEGF拮抗剂,所述ERK抑制剂是指能与ERK结合并阻断ERK生物活性的物质,所述VEGF拮抗剂是指能与VEGF结合并阻断VEGF生物活性的物质。Therefore, preferably, the repulsive targeting signal includes an ERK inhibitor and/or a VEGF antagonist, the ERK inhibitor refers to a substance that can bind to ERK and block the biological activity of ERK, and the VEGF antagonist refers to Substances that bind to VEGF and block the biological activity of VEGF.
具体的,所述ERK抑制剂包括处于临床阶段的抑制剂SCH772984、GDC-0994、Ulixertinib、KO-947、LY3214996、MK-8353、CC-90003、LTT462等,以及处于临床前阶段或者生物活性评价阶段的抑制剂FR180204、VTX-11e、BL-EI-001等;所述VEGF拮抗剂包括贝伐珠单抗、雷莫芦单抗、雷珠单抗、阿柏西普、康柏西普等。Specifically, the ERK inhibitors include clinical stage inhibitors SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996, MK-8353, CC-90003, LTT462, etc., and those in preclinical stage or biological activity evaluation stage The VEGF antagonists include bevacizumab, ramucirumab, ranibizumab, aflibercept, conbercept and the like.
更具体的,上述ERK抑制剂为SCH772984,VEGF拮抗剂为贝伐珠单抗。More specifically, the above-mentioned ERK inhibitor is SCH772984, and the VEGF antagonist is bevacizumab.
本发明在研究过程中发现胃肠道中,十二指肠对于SARS-CoV-2 Spike蛋白敏感性最强,因此优选的,上述胃肠道组织为十二指肠。本发明上述研究也突出了十二指肠对SARS-CoV-2 Spike诱导的血管通透性增加的敏感性及其潜在途径,为COVID-19患者胃肠道症状提供了潜在的治疗靶标。In the research process of the present invention, it is found that in the gastrointestinal tract, the duodenum is the most sensitive to the SARS-CoV-2 Spike protein, so preferably, the above-mentioned gastrointestinal tract tissue is the duodenum. The above study of the present invention also highlights the sensitivity of the duodenum to SARS-CoV-2 Spike-induced increased vascular permeability and its potential pathways, providing a potential therapeutic target for gastrointestinal symptoms in COVID-19 patients.
本发明还提供筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,具体是:确定所述药剂影响VEGF介导的胃肠道血管通透性增加的能力。The present invention also provides a method for screening or evaluating an agent that inhibits SARS-CoV-2-induced damage to the vascular barrier in the gastrointestinal tract, specifically: determining the effect of the agent on the VEGF-mediated increase in the vascular permeability of the gastrointestinal tract. ability.
本发明研究了SARS-CoV-2引起的胃肠道症状的机制,即SARS-CoV-2 Spike蛋白直接与肠上皮细胞上的ACE2结合,激活下游ERK/VEGF信号通路,诱导血管通透性增高。具体的,SARS-CoV-2 Spike蛋白直接与肠上皮细胞上的ACE2结合,在肠上皮细胞中激活了ERK的上游分子Ras,Raf和MEK,进一步激活了ERK,促进肠上皮细胞VEGF分泌,使得肠道内皮细胞VE-cad的表达降低或其磷酸化增加,导致肠道内皮血管通透性增高。The present invention studies the mechanism of gastrointestinal symptoms caused by SARS-CoV-2, namely SARS-CoV-2 Spike protein directly binds to ACE2 on intestinal epithelial cells, activates the downstream ERK/VEGF signaling pathway, and induces increased vascular permeability. Specifically, SARS-CoV-2 Spike protein directly binds to ACE2 on intestinal epithelial cells, activates the upstream molecules of ERK Ras, Raf and MEK in intestinal epithelial cells, further activates ERK, promotes the secretion of VEGF from intestinal epithelial cells, and makes intestinal endothelial cells VE-cad Decreased expression or increased phosphorylation results in increased intestinal endothelial vascular permeability.
由上述机理可知,抑制ERK、MEK、Ras、Raf、VEGF,就可以缓解SARS-CoV-2引起的胃肠道血管屏障受损程度,因此优选的,确定所述药剂影响VEGF介导的胃肠道血管通透性增加的能力是指确定所述药剂抑制ERK、MEK、Ras、Raf、VEGF的能力。It can be seen from the above mechanism that inhibiting ERK, MEK, Ras, Raf, and VEGF can alleviate the degree of damage to the gastrointestinal vascular barrier caused by SARS-CoV-2. Therefore, it is preferable to determine that the agent affects the VEGF-mediated gastrointestinal tract The ability to increase vascular permeability refers to determining the ability of the agent to inhibit ERK, MEK, Ras, Raf, VEGF.
作为一种优选的实施方式,体外实验时,所述药剂影响VEGF介导的胃肠道血管通透性增加通过以下步骤确定:As a preferred embodiment, in an in vitro experiment, the effect of the agent on the increase in vascular permeability of the gastrointestinal tract mediated by VEGF is determined by the following steps:
(1)准备候选药剂组,所述候选药剂组由接触SARS-CoV-2 Spike蛋白的肠道上皮细胞的分泌物、内皮细胞与候选药剂组成;(1) Prepare a candidate agent group consisting of exposure to SARS-CoV-2 Intestinal epithelial cell secretions, endothelial cells and candidate agents of Spike protein;
(2)准备安慰剂组,所述第安慰剂组由接触SARS-CoV-2 Spike蛋白的肠道上皮细胞的分泌物、内皮细胞与安慰剂组成;(2) Prepare a placebo group consisting of exposure to SARS-CoV-2 Intestinal epithelial secretions of Spike protein, endothelial cells and placebo composition;
(3)其中,候选药剂组中内皮细胞与安慰剂组中内皮细胞相比,能够检测到更高的VE-cad的表达量或更低的VE-cad磷酸化水平,表明所述候选药剂能抑制VEGF介导的胃肠道血管通透性增加。(3) Among them, the endothelial cells in the candidate drug group can detect higher VE-cad expression or lower VE-cad phosphorylation level than the endothelial cells in the placebo group, indicating that the candidate drug can Inhibits VEGF-mediated increase in gastrointestinal vascular permeability.
优选的,步骤(2)所述的安慰剂选自PBS或DMEM。Preferably, the placebo described in step (2) is selected from PBS or DMEM.
作为另外一种实施方式,当在体内时,所述药剂影响VEGF介导的胃肠道血管通透性增加通过以下步骤确定:As another embodiment, when in vivo, said agent affects VEGF-mediated increase in gastrointestinal vascular permeability as determined by the following steps:
(1)构建SARS-CoV-2胃肠炎症动物模型;(1) Construct an animal model of SARS-CoV-2 gastrointestinal inflammation;
(2)对(1)的模型动物腹腔注射或者不注射安慰剂;(2) Intraperitoneal injection or no placebo injection to the model animals of (1);
(3)对(1)的模型动物腹腔注射候选药剂;(3) Inject the candidate drug into the model animal of (1) by intraperitoneal injection;
(4)其中,(3)处理后的动物胃肠道组织中和(2)处理后的动物的胃肠道组织中相比,能够检测到更低的VEGF的表达量,表明所述药剂能抑制VEGF介导的胃肠道血管通透性增加。(4) wherein, a lower expression level of VEGF can be detected in the gastrointestinal tract tissue of the animals after (3) treatment than in the gastrointestinal tract tissue of the animals after treatment (2), indicating that the agent can Inhibits VEGF-mediated increase in gastrointestinal vascular permeability.
优选的,步骤(2)所述的安慰剂选自PBS或DMEM。Preferably, the placebo described in step (2) is selected from PBS or DMEM.
优选的,步骤(4)所述胃肠道组织为十二指肠。Preferably, the gastrointestinal tract tissue in step (4) is the duodenum.
优选的,步骤(1)所述SARS-CoV-2胃肠炎症动物模型的构建方法包括以下步骤:Preferably, the construction method of the SARS-CoV-2 gastrointestinal inflammation animal model in step (1) includes the following steps:
S1、小鼠禁食16~24h后进行麻醉;S1. Mice were anesthetized after fasting for 16-24 hours;
S2、从小鼠肛门注入乙酸,倒立小鼠,并对小鼠胃肠道进行灌洗;S2. Inject acetic acid from the anus of the mouse, turn the mouse upside down, and lavage the gastrointestinal tract of the mouse;
S3、一段时间后小鼠腹腔注射SARS-CoV-2 Spike蛋白;S3. After a period of time, mice were intraperitoneally injected with SARS-CoV-2 Spike protein;
S4、数小时后即成功得到SARS-CoV-2胃肠炎症动物模型。S4. The animal model of SARS-CoV-2 gastrointestinal inflammation was successfully obtained after a few hours.
本发明腹腔注射SARS-CoV-2 Spike蛋白之前,先对胃肠道上皮进行酸灌肠处理,再用SARS-CoV-2 Spike蛋白刺激则更有利于SARS-CoV-2 Spike蛋白的结合;经过这种方式获得的动物模型与现有报道的COVID-19患者的临床症状一致,因此本发明方法构建的动物模型可以复制临床COVID-19患者的胃肠道表现。Before intraperitoneal injection of SARS-CoV-2 Spike protein in the present invention, acid enema treatment is performed on the epithelium of the gastrointestinal tract, and then SARS-CoV-2 Spike protein stimulation is more conducive to the binding of SARS-CoV-2 Spike protein; the animal model obtained in this way is consistent with the clinical symptoms of COVID-19 patients reported in the present, so the animal model constructed by the method of the present invention can replicate clinical symptoms Gastrointestinal manifestations in patients with COVID-19.
优选地,上述动物模型的构建方法中,步骤S3所述SARS-CoV-2 Spike蛋白的用量为5.5 nM/kg小鼠体重。Preferably, in the construction method of the above-mentioned animal model, the SARS-CoV-2 described in step S3 The dosage of Spike protein was 5.5 nM/kg mouse body weight.
同样,还对腹腔注射SARS-CoV-2 Spike蛋白的作用时间进行了优化,发现6h后即可以达到临床COVID-19患者的胃肠道损伤表现,且24h与6h的临床COVID-19患者的胃肠道损伤表现差异不明显。因此,优选地,上述动物模型的构建方法中,其特征在于,步骤S4所述数小时是指6~24h。Similarly, the action time of intraperitoneal injection of SARS-CoV-2 Spike protein was optimized, and it was found that the clinical manifestations of gastrointestinal damage in patients with COVID-19 could be achieved after 6 hours. There was no significant difference in the performance of intestinal injury. Therefore, preferably, in the above-mentioned method for constructing an animal model, it is characterized in that the several hours in step S4 refers to 6-24 hours.
更具体的,体内实验时,所述药剂影响VEGF介导的胃肠道血管通透性增加通过以下步骤确定:More specifically, in vivo experiments, the effect of the agent on the VEGF-mediated increase in gastrointestinal vascular permeability was determined by the following steps:
(1)小鼠禁食16~24h后进行麻醉;(1) Mice were anesthetized after fasting for 16-24 hours;
(2)从小鼠肛门注入乙酸,倒立小鼠,并对小鼠胃肠道进行灌洗;(2) Inject acetic acid from the anus of the mouse, turn the mouse upside down, and lavage the gastrointestinal tract of the mouse;
(3)小鼠正常喂食,16~24h后,将候选药剂通过腹腔分别注射到小鼠体内,得到候选药剂组小鼠;将安慰剂通过腹腔分别注射到小鼠体内,得到安慰剂组小鼠;(3) The mice were fed normally. After 16-24 hours, the candidate drugs were injected into the mice through the abdominal cavity respectively to obtain the mice in the candidate drug group; the placebo was injected into the mice through the abdominal cavity respectively to obtain the mice in the placebo group. ;
(4)1~2h后候选药剂组和安慰剂组小鼠腹腔分别注射SARS-CoV-2 Spike蛋白,对照组小鼠腹腔注射等量的IgG-Fc蛋白;(4) After 1-2 hours, the mice in the candidate drug group and the placebo group were injected with SARS-CoV-2 by intraperitoneal injection. Spike protein, mice in the control group were intraperitoneally injected with the same amount of IgG-Fc protein;
(5)1~2h后,再次对候选药剂组小鼠腹腔注射候选药剂,对安慰剂组小鼠腹腔注射安慰剂;(5) After 1-2 hours, the candidate drug was injected intraperitoneally to the mice in the candidate drug group again, and the placebo was intraperitoneally injected to the mice in the placebo group;
(6)分别取对照组小鼠、候选药剂组小鼠和安慰剂组小鼠胃肠道组织,其中,安慰剂组小鼠胃肠组织与对照组小鼠相比,能够检测到更高的VEGF表达量,而候选药剂组小鼠胃肠组织与安慰剂组小鼠相比,能够检测到更低的VEGF表达量,表明所述候选药剂能抑制VEGF介导的胃肠道血管通透性增加。(6) The gastrointestinal tissues of the mice in the control group, the mice in the candidate drug group and the mice in the placebo group were taken respectively. Compared with the mice in the control group, the gastrointestinal tissues of the mice in the placebo group were able to detect higher Compared with the mice in the placebo group, the gastrointestinal tissue of the candidate drug group could detect a lower VEGF expression level, indicating that the candidate drug can inhibit the VEGF-mediated gastrointestinal vascular permeability Increase.
作为一种优选的实施方式,体内实验时,上述步骤(2)乙酸的浓度为1~2% v/v,体积为500μL~1000μL。As a preferred embodiment, in the in vivo experiment, the concentration of acetic acid in the above step (2) is 1-2% v/v, and the volume is 500 μL to 1000 μL.
作为一种优选的实施方式,体内实验时,上述步骤(4)所述SARS-CoV-2 Spike蛋白的用量为5.5nM/kg小鼠体重。As a preferred embodiment, in the in vivo experiment, the SARS-CoV-2 described in the above step (4) The dosage of Spike protein was 5.5nM/kg mouse body weight.
本发明还提供治疗或预防SARS-CoV-2引起的胃肠道症状的方法,其特征在于,包括以下步骤:The present invention also provides a method for treating or preventing gastrointestinal symptoms caused by SARS-CoV-2, characterized by comprising the following steps:
(1)鉴定患有所述SARS-CoV-2引起的胃肠道症状的对象,以及,(1) identify subjects with gastrointestinal symptoms caused by said SARS-CoV-2, and,
(2)给对象胃肠道施用与ACE2和/或ERK和/或VEGF结合的排斥性导向信号。(2) administering to the gastrointestinal tract of a subject a repulsive targeting signal that binds to ACE2 and/or ERK and/or VEGF.
具体的,所述排斥性导向信号包括ERK抑制剂和/或VEGF拮抗剂,所述ERK抑制剂是指能与ERK结合并阻断ERK生物活性的物质,所述VEGF拮抗剂是指能与VEGF结合并阻断VEGF生物活性的物质。Specifically, the repulsive guidance signal includes an ERK inhibitor and/or a VEGF antagonist, the ERK inhibitor refers to a substance that can bind to ERK and block the biological activity of ERK, and the VEGF antagonist refers to a substance that can interact with VEGF Substances that bind to and block the biological activity of VEGF.
具体的,所述ERK抑制剂包括处于临床阶段的抑制剂SCH772984、GDC-0994、Ulixertinib、KO-947、LY3214996、MK-8353、CC-90003、LTT462等,处于临床前阶段或者生物活性评价阶段的抑制剂FR180204、VTX-11e、BL-EI-001等;所述VEGF拮抗剂包括贝伐珠单抗、雷莫芦单抗、雷珠单抗、阿柏西普、康柏西普等。Specifically, the ERK inhibitors include clinical stage inhibitors SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996, MK-8353, CC-90003, LTT462, etc., which are in the preclinical stage or the biological activity evaluation stage. Inhibitors FR180204, VTX-11e, BL-EI-001, etc.; the VEGF antagonists include bevacizumab, ramucirumab, ranibizumab, aflibercept, conbercept and the like.
更具体的,上述ERK抑制剂为SCH772984,VEGF拮抗剂为抗VEGF药物贝伐珠单抗。More specifically, the above-mentioned ERK inhibitor is SCH772984, and the VEGF antagonist is the anti-VEGF drug bevacizumab.
本发明还提供用于治疗或预防或缓解SARS-CoV-2引起的胃肠道症状的药物,所述药物具有以下至少一种功能:The present invention also provides a medicament for treating or preventing or alleviating gastrointestinal symptoms caused by SARS-CoV-2, the medicament having at least one of the following functions:
(1)抑制肠道上皮细胞VEGF的表达;(1) Inhibit the expression of VEGF in intestinal epithelial cells;
(2)抑制肠道上皮细胞ERK的表达;(2) Inhibit the expression of ERK in intestinal epithelial cells;
(3)挽救SARS-CoV-2刺突蛋白引起的血管内皮检测到更低的VE-cad表达量或其更高的磷酸化水平。(3) Lower VE-cad expression or higher phosphorylation levels were detected in the vascular endothelium caused by rescue of the SARS-CoV-2 spike protein.
优选的,所述药物包括但不限于:ERK抑制剂和/或VEGF拮抗剂。Preferably, the drugs include, but are not limited to: ERK inhibitors and/or VEGF antagonists.
具体的,所述ERK抑制剂包括处于临床阶段的抑制剂SCH772984、GDC-0994、Ulixertinib、KO-947、LY3214996、MK-8353、CC-90003、LTT462等,处于临床前阶段或者生物活性评价阶段的抑制剂FR180204、VTX-11e、BL-EI-001等;所述VEGF拮抗剂包括贝伐珠单抗、雷莫芦单抗、雷珠单抗、阿柏西普、康柏西普等。Specifically, the ERK inhibitors include clinical stage inhibitors SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996, MK-8353, CC-90003, LTT462, etc., which are in the preclinical stage or the biological activity evaluation stage. Inhibitors FR180204, VTX-11e, BL-EI-001, etc.; the VEGF antagonists include bevacizumab, ramucirumab, ranibizumab, aflibercept, conbercept and the like.
越来越多的证据表明,SARS-CoV-2可以感染肠道和肺中的内皮细胞,这引发了一个问题,即感染时的内皮表现是由于直接靶向内皮细胞还是靶向其他细胞的继发作用。在这项研究中,我们阐明了Spike引发的信号传导并不会通过影响内皮通透性来损害血管完整性,而是通过调节血管周围肠上皮细胞的VEGF途径而损害血管完整性。VEGF是急性呼吸窘迫综合征(ARDS)的关键因素,在COVID-19患者的血清中猛增。与这项研究部分相符,我们发现被Spike感染的局部组织中VEGF升高,这可能是导致胃肠道血管屏障损伤的原因。Growing evidence that SARS-CoV-2 can infect endothelial cells in the gut and lung raises the question of whether endothelial manifestations upon infection are due to direct targeting of endothelial cells or secondary cells targeting other cells. effect. In this study, we elucidated that Spike-triggered signaling does not impair vascular integrity by affecting endothelial permeability, but rather by modulating the VEGF pathway in perivascular intestinal epithelial cells. VEGF, a key factor in acute respiratory distress syndrome (ARDS), surged in the serum of COVID-19 patients. Consistent in part with this study, we found elevated VEGF in local tissue infected with Spike, which may be responsible for damage to the vascular barrier in the gastrointestinal tract.
我们的研究提供了第一个证据,即肠上皮细胞中的ACE2 / ERK / VEGF轴在SARS-CoV-2诱导的肠道炎症中起关键作用。Our study provides the first evidence that the ACE2/ERK/VEGF axis in intestinal epithelial cells plays a critical role in SARS-CoV-2-induced intestinal inflammation.
有益效果beneficial effect
与现有技术相比,本发明具有以下有益效果:Compared with the prior art, the present invention has the following beneficial effects:
本发明研究发现,SARS-CoV-2 Spike蛋白可与人和小鼠ACE2结合,诱导血管通透性增加。通过对内皮和上皮细胞的共培养发现,Spike蛋白通过结合肠上皮细胞的ACE2,激活了Ras- Raf-MEK-ERK通路,促进了上皮细胞分泌VEGF,而该途径在内皮细胞中不存在。ERK或VEGF阻断剂在体内可挽救Spike蛋白增强的血管通透性,减轻胃肠道症状。这些结果表明,在胃肠道,特别是十二指肠,SARS-CoV-2 Spike蛋白可直接与肠上皮细胞的ACE2结合,激活下游ERK/VEGF信号通路,诱导血管通透性升高。本研究为COVID-19患者出现的胃肠道症状提供了一种机制解释和合理的治疗策略。The present study found that the SARS-CoV-2 Spike protein can combine with human and mouse ACE2 to induce increased vascular permeability. Through co-culture of endothelial and epithelial cells, it was found that Spike protein activates the Ras-Raf-MEK-ERK pathway by binding to ACE2 of intestinal epithelial cells, and promotes the secretion of VEGF from epithelial cells, which does not exist in endothelial cells. ERK or VEGF blockade rescued Spike protein-enhanced vascular permeability in vivo and alleviated gastrointestinal symptoms. These results suggest that in the gastrointestinal tract, especially the duodenum, the SARS-CoV-2 Spike protein can directly bind to ACE2 in intestinal epithelial cells, activate the downstream ERK/VEGF signaling pathway, and induce increased vascular permeability. This study provides a mechanistic explanation and a rational treatment strategy for gastrointestinal symptoms in patients with COVID-19.
附图说明Description of drawings
图1a显示COVID-19患者胃肠道组织观察到ACE2和SARS-CoV-2 Spike蛋白的共定位;图1b显示 H&E染色胃肠道组织局部炎症情况;Figure 1a shows the co-localization of ACE2 and SARS-CoV-2 Spike protein observed in the gastrointestinal tissue of COVID-19 patients; Figure 1b shows the local inflammation in H&E stained gastrointestinal tissue;
图2 a显示动物模型构建流程;图2b显示实验组(Spike-Fc)小鼠和对照组(Control-Fc)小鼠不同部位的HE切片染色图;图2c和图2d显示实验组(Spike-Fc)小鼠和对照组(Control-Fc)小鼠胃肠道不同部位的免疫荧光染色图;Figure 2a shows the construction process of the animal model; Figure 2b shows the HE staining images of different parts of the experimental group (Spike-Fc) mice and the control group (Control-Fc) mice; Figure 2c and Figure 2d show the experimental group (Spike-Fc) mice. Fc) Immunofluorescence staining of different parts of the gastrointestinal tract of mice and control (Control-Fc) mice;
图3a显示实验组(Spike-Fc)小鼠和对照组(Control-Fc)小鼠(n=9)血管屏障功能评价(采用TRITC-葡聚糖示踪剂);数据用平均值±SD表示,P值用t检验,*代表p<0.05;图3b显示了实验组(Spike-Fc)和对照组(Control-Fc)对HUVECs细胞的渗透性的影响(采用TRITC-葡聚糖示踪剂),n=9,数据用平均值±SD表示,P值用t检验,ns代表没有显著性;图3c显示采用WB测定了实验组(Spike-Fc)和对照组(Control-Fc)的HUVECs细胞中粘附蛋白VE-cad和pVE-cad(Y731)的表达,柱状图代表蛋白质表达相对于β-actin定量的比例,实验数据经过三次平均,数据用平均值±SD表示,p值用配对t检验,ns代表没有显著性;图3d显示体外渗透实验过程;图3e显示通过右旋糖酐的透性来评价HUVEC屏障的通透性,数据用平均值±SD表示,n=8,p值用t检验,***代表p<0.0001;Figure 3a shows the evaluation of vascular barrier function (using TRITC-dextran tracer) in experimental (Spike-Fc) and control (Control-Fc) mice (n=9); data are expressed as mean ± SD , P value is by t test, * represents p<0.05; Figure 3b shows the effect of experimental group (Spike-Fc) and control group (Control-Fc) on the permeability of HUVECs cells (using TRITC-dextran tracer) ), n=9, data are expressed as mean ± SD, P value is expressed by t test, ns means no significance; Figure 3c shows that the HUVECs of experimental group (Spike-Fc) and control group (Control-Fc) were determined by WB The expression of adhesion proteins VE-cad and pVE-cad (Y731) in cells, the histogram represents the ratio of protein expression relative to β-actin quantification, the experimental data were averaged three times, the data are expressed as mean ± SD, and the p value is paired with t-test, ns represents no significance; Figure 3d shows the process of in vitro permeation experiments; Figure 3e shows the permeability of HUVEC barriers evaluated by the permeability of dextran, data are expressed as mean ± SD, n=8, p-values are expressed by t Test, *** represents p<0.0001;
图4a显示Spike-Fc蛋白与人ACE2分别在Caco-2 细胞和239T细胞中的下拉结果,其中,Input代表总细胞裂解液,IP:Fc代表用Fc肽段下拉的蛋白;图4b显示Spike-Fc蛋白与人ACE2在239T细胞中的荧光染色结果;Figure 4a shows the pull-down results of Spike-Fc protein and human ACE2 in Caco-2 cells and 239T cells, respectively, where Input represents total cell lysate, and IP:Fc represents the protein pulled down by Fc peptide; Figure 4b shows Spike-Fc Fluorescent staining results of Fc protein and human ACE2 in 239T cells;
图5a显示采用WB检测与Spike-Fc或者Control-Fc孵育的Caco-2-HUVECs共培养体系上清液中粘连蛋白ZO-1、VE-cad、pVE-cad(Y658)和pVE-cad(Y731)的表达;柱状图显示通过密度扫描定量了蛋白表达相对于β-actin的量,实验数据经过三次平均,数据用平均值±SD表示,p值用配对t检验,*代表p<0.05,ns代表没有显著性;图5b和图5c显示了实验组(Spike-Fc)和对照组(IgG-Fc)小鼠肠道组织(十二指肠、空回肠、结肠和直肠)中VE-cad和 pVE-cad(Y731)的IHC图,标尺为100μm,散点图显示了十二指肠、空回肠、结肠和直肠中VE-cad和pVE-cad(Y731)的表达水平(数据用平均值±SD表示,p值用t检验,ns代表没有显著性,*代表p<0.05,**代表p<0.01);图5d和图5e显示健康人和COVID-19患者十二指肠中VE-cad和pVE-cad(Y731)的检测(数据用平均值±SD表示,p值用t检验,ns代表没有显著性,**代表p<0.01,***代表p<0.0001);Figure 5a shows the detection of cohesin ZO-1, VE-cad, pVE-cad (Y658) and pVE-cad (Y731) in the supernatant of the co-culture system of Caco-2-HUVECs incubated with Spike-Fc or Control-Fc by WB. ) expression; the histogram shows that the protein expression relative to the amount of β-actin was quantified by densitometric scanning, the experimental data were averaged three times, the data were expressed as mean ± SD, the p value was tested by paired t test, * represents p < 0.05, ns Representatives are not significant; Figure 5b and Figure 5c show VE-cad and IHC plot of pVE-cad(Y731), the scale bar is 100 μm, and the scatter plot shows the expression levels of VE-cad and pVE-cad(Y731) in the duodenum, jejunum, colon and rectum (data are presented as mean ± SD represents, p value is by t test, ns represents no significance, * represents p<0.05, ** represents p<0.01); Figure 5d and Figure 5e show VE-cad in duodenum of healthy and COVID-19 patients and the detection of pVE-cad (Y731) (data are represented by mean ± SD, p value is represented by t test, ns represents no significance, ** represents p<0.01, *** represents p<0.0001);
图6a显示采用RT-PCR分析实验组(Spike-Fc)和对照组(Control-Fc)小鼠十二指肠、空回肠、结肠和直肠中VEGF的转录水平(数据用平均值±SD表示,p值用t检验,*代表p<0.05);图6b显示采用ELISA分析了实验组(Spike-Fc)和对照组(Control-Fc)小鼠十二指肠、空回肠、结肠和直肠中VEGF的含量,散点图显示了肠道组织中VEGF的蛋白浓度(数据用平均值±SD表示,p值用t检验,ns代表没有显著性);图6c显示采用ELISA分析了用Spike-Fc或Cntrol-Fc处理的Caco-2细胞分泌的VEGF的量;图6d显示采用ELISA分析了用Spike-Fc或Control-Fc处理的小鼠血浆中VEGF的量;散点图显示了血浆中VEGF的蛋白浓度(数据用平均值±SD表示,p值用t检验,**代表p<0.01);Figure 6a shows the RT-PCR analysis of VEGF transcript levels in the duodenum, jejunum, colon and rectum of experimental (Spike-Fc) and control (Control-Fc) mice (data are expressed as mean ± SD, The p value was tested by t-test, * represents p<0.05); Figure 6b shows the ELISA analysis of VEGF in the duodenum, jejunum, colon and rectum of experimental (Spike-Fc) and control (Control-Fc) mice The scatter plot shows the protein concentration of VEGF in intestinal tissue (data are represented by mean ± SD, p value is by t test, ns represents no significance); The amount of VEGF secreted by Cntrol-Fc-treated Caco-2 cells; Figure 6d shows the amount of VEGF in plasma of mice treated with Spike-Fc or Control-Fc was analyzed by ELISA; scatter plot shows the protein of VEGF in plasma Concentration (data are represented by mean ± SD, p value by t test, ** represents p<0.01);
图7显示用Spike-Fc或Control-Fc处理的Caco-2细胞中ERK或者pERK的免疫印迹分析;柱状图显示通过密度扫描定量了蛋白表达相对于β-actin的量,实验数据经过三次平均,数据用平均值±SD表示,p值用配对t检验,*代表p<0.05;Figure 7 shows immunoblot analysis of ERK or pERK in Caco-2 cells treated with Spike-Fc or Control-Fc; histograms show quantification of protein expression relative to the amount of β-actin by densitometric scanning. Data are represented by mean ± SD, p value is by paired t test, * represents p < 0.05;
图8a显示了通过western blot检测Caco-2的siRNA(有01、02和03三条siRNA敲降序列),与对照序列(NC)相比,三条序列均显示明显的敲降效果,内源性ACE2敲减后,Ras、C-Raf、pMEK、pERK和p-P90RSK的表达显著增加;图8b显示用Control-Fc或者Spike-Fc蛋白分别刺激HUVCE细胞1h后,western blot检测ERK和pERK蛋白的表达情况;Figure 8a shows the detection of Caco-2 siRNA (with three siRNA knockdown sequences 01, 02 and 03) by western blot. Compared with the control sequence (NC), all three sequences showed obvious knockdown effect, and the endogenous ACE2 After knockdown, the expressions of Ras, C-Raf, pMEK, pERK and p-P90RSK were significantly increased; Figure 8b shows that after stimulating HUVCE cells with Control-Fc or Spike-Fc protein for 1 h, the expression of ERK and pERK proteins was detected by western blotting Happening;
图9a显示Spike-Fc,Control-Fc和SCH772984处理Caco-2细胞后Ras-Raf-MEK-ERK信号通路中各蛋白的表达水平,柱状图显示通过蛋白灰度定量了蛋白表达相对于β-actin的量,实验数据重复三次,数据用平均值±SD表示,p值用配对t检验,*代表p<0.05,ns代表没有显著性;图9b显示Spike-Fc,Control-Fc和SCH772984分别与Caco-2细胞共培养后,用ELISA测定VEGF的量,数据用平均值±SD表示,p值用t检验,*代表p<0.05;图9c显示实验组(Spike-Fc)和对照组(Control-Fc)腹腔注射了SCH772984的模型小鼠肠道组织(十二指肠、空回肠、结肠和直肠)中ERK和pERK的IHC图,标尺为100μm,散点图显示了十二指肠、空回肠、结肠和直肠中ERK和pERK的表达水平(数据用平均值±SD表示,p值用t检验,ns代表没有显著性,*代表p<0.05,**代表p<0.01);图9d和图9e显示健康人和COVID-19患者十二指肠中ERK和pERK的检测,(数据用平均值±SD表示,p值用t检验,ns代表没有显著性,**代表p<0.01;Figure 9a shows the expression levels of each protein in the Ras-Raf-MEK-ERK signaling pathway after Spike-Fc, Control-Fc and SCH772984 treatment of Caco-2 cells, the histogram shows the protein expression relative to β-actin was quantified by protein grayscale The experimental data were repeated three times, the data were represented by the mean ± SD, the p value was tested by paired t test, * represents p<0.05, ns represents no significance; Figure 9b shows that Spike-Fc, Control-Fc and SCH772984 were compared with Caco After the -2 cells were co-cultured, the amount of VEGF was determined by ELISA, the data were expressed as mean ± SD, the p value was expressed by t test, * represents p < 0.05; Figure 9c shows the experimental group (Spike-Fc) and the control group (Control-Fc) Fc) IHC map of ERK and pERK in intestinal tissues (duodenum, jejunum, ileum, colon and rectum) of model mice injected with SCH772984 intraperitoneally, the scale bar is 100 μm, and the scatter plot shows the duodenum, jejunum and ileum , expression levels of ERK and pERK in colon and rectum (data are represented by mean ± SD, p values are represented by t-test, ns represents no significance, * represents p<0.05, ** represents p<0.01); Figure 9d and Figure 9 9e shows the detection of ERK and pERK in the duodenum of healthy people and patients with COVID-19, (data are expressed as mean ± SD, p-values are expressed by t-test, ns means no significance, ** means p<0.01;
图10a显示了用Spike-Fc或Control-Fc或SCH772984或者Bevacizumab处理的Caco-2细胞中VE-cad的表达和定位,图10b显示共培养体系的血管屏障(采用TRITC-葡聚糖示踪剂),共培养体系由HUVEC细胞和用Spike-Fc或Control-Fc或SCH772984或者Bevacizumab处理的Caco-2细胞上清液共培养;数据用平均值±SD表示,n=8,p值用t检验,ns代表没有显著性,***代表p<0.0001;图10c显示了实验组(Spike-Fc)和对照组(Control-Fc)腹腔注射了SCH772984或者Bevacizumab的模型小鼠肠道组织(十二指肠、空回肠、结肠和直肠)中VE-cad和pVE-cad(Y731)的IHC图,标尺为100μm,散点图显示了十二指肠、空回肠、结肠和直肠中VE-cad和pVE-cad(Y731)的表达水平(数据用平均值±SD表示,n=4,p值用t检验,ns代表没有显著性,*代表p<0.05,**代表p<0.01,***代表p<0.0001);Figure 10a shows the expression and localization of VE-cad in Caco-2 cells treated with Spike-Fc or Control-Fc or SCH772984 or Bevacizumab, and Figure 10b shows the vascular barrier of the co-culture system (using TRITC-dextran tracer ), the co-culture system was co-cultured with HUVEC cells and the supernatant of Caco-2 cells treated with Spike-Fc or Control-Fc or SCH772984 or Bevacizumab; data are expressed as mean ± SD, n=8, p-values were tested by t test , ns means no significance, *** means p<0.0001; Figure 10c shows the experimental group (Spike-Fc) and the control group (Control-Fc) intraperitoneal injection of SCH772984 or Bevacizumab model mice intestinal tissue (12 IHC plots of VE-cad and pVE-cad (Y731) in the denum, jejunum, ileum, colon and rectum), the scale bar is 100 μm, and the scatter plot shows VE-cad and pVE-cad in the duodenum, jejunum and ileum, colon and rectum. The expression level of pVE-cad (Y731) (data are represented by mean ± SD, n=4, p value is by t test, ns means no significance, * means p<0.05, ** means p<0.01, *** for p<0.0001);
图11a显示采用ELISA分析实验组(Spike-Fc)和对照组(Control-Fc)腹腔注射了SCH772984或者Bevacizumab的模型小鼠肠道组织(十二指肠、空回肠、结肠和直肠)中VEGF的量,数据用平均值±SD表示,n=4,p值用t检验,ns代表没有显著性,*代表p<0.005;图11b显示用实验组(Spike-Fc)和对照组(Control-Fc)或者腹腔注射了SCH772984或者Bevacizumab的模型小鼠肠道组织(十二指肠、空回肠、结肠和直肠)中ERK或者pERK的免疫印迹分析;柱状图显示通过蛋白灰度定量了蛋白表达相对于β-actin的量,实验数据重复三次,数据用平均值±SD表示,p值用配对t检验,*代表p<0.05,**代表p<0.01,***代表p<0.0001;Figure 11a shows the use of ELISA to analyze the expression of VEGF in the intestinal tissues (duodenum, jejunum, colon and rectum) of model mice injected with SCH772984 or Bevacizumab in the experimental group (Spike-Fc) and the control group (Control-Fc) by intraperitoneal injection. Amount, data are represented by mean ± SD, n=4, p value is by t test, ns represents no significance, * represents p<0.005; Figure 11b shows the experimental group (Spike-Fc) and the control group (Control-Fc) ) or immunoblot analysis of ERK or pERK in intestinal tissues (duodenum, jejunum, colon, and rectum) of model mice injected with SCH772984 or Bevacizumab intraperitoneally; The amount of β-actin, the experimental data was repeated three times, the data were expressed as mean ± SD, and the p value was expressed by paired t test, *represents p<0.05, **represents p<0.01, and ***represents p<0.0001;
图12a显示酸刺激后,小鼠腹腔分别注射Spike-Fc(5.5nM/kg)、Control-Fc(5.5nM/kg)、SCH772984(50mg/kg)或者Bevacizumab(5mg/kg)后肠道组织(十二指肠、空回肠、结肠和直肠)的Evans Blue染料渗出实验,数据用平均值±SD表示,n=6,p值用t检验,ns代表没有显著性,*代表p<0.05,**代表p<0.01,***代表p<0.0001;图12b显示肠道组织(十二指肠、空回肠、结肠和直肠)每单位重量(g)渗透的evens blue染料的OD值;图12c显示实验组(Spike-Fc)和对照组(Control-Fc)或者腹腔注射了SCH772984或者Bevacizumab的模型小鼠肠道组织(十二指肠、空回肠、结肠和直肠)的H&E染色;黄色箭头代表炎性浸润;红星代表水肿,评估了肠道组织中无炎症、中等炎症和重度炎症;柱状图显示评估消化道阴性、轻度和中度炎症的比例,数据以均数±SD表示,每组老鼠4只;Figure 12a shows that after acid stimulation, the intestinal tissue ( Evans of duodenum, jejunum, colon and rectum) Blue dye exudation experiment, data are expressed as mean ± SD, n=6, p value is by t test, ns means no significance, * means p<0.05, ** means p<0.01, *** means p<0.0001 ; Figure 12b shows the evens permeated per unit weight (g) of intestinal tissue (duodenum, jejunum, colon and rectum) The OD value of blue dye; Figure 12c shows the experimental group (Spike-Fc) and the control group (Control-Fc) or the intestinal tissue (duodenum, jejunum, colon and rectum) of model mice injected with SCH772984 or Bevacizumab intraperitoneally ) of H&E staining; yellow arrows represent inflammatory infiltrates; red stars represent edema, and no inflammation, moderate inflammation, and severe inflammation were assessed in the intestinal tissue; histograms show the proportion of negative, mild, and moderate inflammation assessed in the digestive tract, and the data are shown in Mean ± SD, 4 mice in each group;
图13显示了SARS-CoV-2 Spike蛋白介导的血管高通透性和肠道组织炎症模型机制。Figure 13 shows the model mechanism of SARS-CoV-2 Spike protein-mediated vascular hyperpermeability and intestinal tissue inflammation.
本发明的最佳实施方式BEST MODE FOR CARRYING OUT THE INVENTION
下面对本发明的具体实施方式作进一步说明。在此需要说明的是,对于这些实施方式的说明用于帮助理解本发明,但并不构成对本发明的限定。此外,下面所描述的本发明各个实施方式中所涉及的技术特征只要彼此之间未构成冲突就可以相互组合。下述实验例中所使用的试验方法如无特殊说明,均为常规方法;所使用的材料、试剂等,如无特殊说明,为可从商业途径得到的试剂和材料。The specific embodiments of the present invention will be further described below. It should be noted here that the descriptions of these embodiments are used to help the understanding of the present invention, but do not constitute a limitation of the present invention. In addition, the technical features involved in the various embodiments of the present invention described below can be combined with each other as long as they do not conflict with each other. The test methods used in the following experimental examples are conventional methods unless otherwise specified; the materials, reagents, etc. used are commercially available reagents and materials unless otherwise specified.
患者信息和数据收集:根据中国疾病预防控制中心(CDC)的指南,使用内窥镜检查收集了COVID-19患者(n=17)的临床标本,包括十二指肠和直肠。17例患者的临床资料见附表1。经中山大学附属第五医院伦理委员会伦理批准(No. 2020L029-1),所有患者均签署知情同意书。Patient information and data collection: Clinical specimens, including duodenum and rectum, were collected from patients with COVID-19 (n=17) using endoscopy according to the guidelines of the Chinese Center for Disease Control and Prevention (CDC). The clinical data of 17 patients are shown in Supplementary Table 1. Ethically approved by the Ethics Committee of the Fifth Affiliated Hospital of Sun Yat-Sen University (No. 2020L029-1), all patients signed informed consent.
Figure 579220dest_path_image001
Figure 579220dest_path_image001
细胞系和细胞培养:人脐静脉内皮细胞(HUVEC)购自ScienCell(Cat.No.8000, Cat.No.5000),在添加有10%胎牛血清的ECM培养基(ScienCell,Cat.No.1001)中培养。人结直肠腺癌细胞(Caco-2)购自(广州 IGE生物技术公司,广州),在Dulbecco改良的Eagle 培养基(Gibco)中培养,该培养基添加了10%的胎牛血清,50 U/mL青霉素和50 mg/mL 链霉素(Gibco,目录编号15140-122)。从美国生物资源中心(ATCC,马纳萨斯,美国)获得鼠类内皮细胞系(C166),在添加了10%胎牛血清,50 U/mL青霉素和50 mg的Dulbecco 改良的Eagle 培养基(Gibco)中进行培养。将细胞放在含5%CO 2的37°C潮湿培养箱中培养。下述研究得到中山大学附属第五医院的批准。对于所有动物实验,均获得中山大学附属第五医院实验动物伦理委员会的许可。统计分析:使用SPSS v13.0软件进行统计分析。计算平均值及误差进行统计分析,所有数据均使用GraphPad Prism软件(5.0版)进行分析。当仅分析两组时,使用t检验评估组之间的差异;比较定量WB时,通过配对t检验评估组之间的差异。进行皮尔逊相关分析以分析蛋白质表达之间的相关性,认为P<0.05具有统计学意义。 Cell lines and cell culture: Human umbilical vein endothelial cells (HUVEC) were purchased from ScienCell (Cat. No. 8000, Cat. No. 5000) in ECM medium (ScienCell, Cat. No. 5000) supplemented with 10% fetal bovine serum. 1001) in culture. Human colorectal adenocarcinoma cells (Caco-2) were purchased from (Guangzhou IGE Biotechnology Company, Guangzhou) and cultured in Dulbecco's modified Eagle's medium (Gibco) supplemented with 10% fetal bovine serum, 50 U /mL penicillin and 50 mg/mL streptomycin (Gibco, catalog number 15140-122). A murine endothelial cell line (C166) was obtained from the American Biological Resource Center (ATCC, Manassas, USA), supplemented with 10% fetal bovine serum, 50 U/mL penicillin, and 50 mg of Dulbecco's modified Eagle's medium ( Gibco). Cells were cultured in a humidified 37°C incubator with 5% CO . The following studies were approved by the Fifth Affiliated Hospital of Sun Yat-Sen University. For all animal experiments, the permission of the Experimental Animal Ethics Committee of the Fifth Affiliated Hospital of Sun Yat-sen University was obtained. Statistical analysis: Statistical analysis was performed using SPSS v13.0 software. Mean values and errors were calculated for statistical analysis, and all data were analyzed using GraphPad Prism software (version 5.0). Differences between groups were assessed using t-tests when only two groups were analyzed; differences between groups were assessed by paired t-tests when comparing quantitative WB. Pearson correlation analysis was performed to analyze the correlation between protein expressions, and P<0.05 was considered to be statistically significant.
一、one, SARS-CoV-2 SpikeSARS-CoV-2 Spike 蛋白优先诱导十二指肠间质水肿Protein preferentially induces duodenal interstitial edema
1、H&E染色和免疫荧光染色1. H&E staining and immunofluorescence staining
(1)组织切片:COVID-19患者的临床标本样品进行福尔马林固定,石蜡包埋和切片(4µm)。(1) Tissue sections: Clinical specimens from COVID-19 patients were formalin-fixed, paraffin-embedded and sectioned (4 µm).
(2)免疫荧光染色(2) Immunofluorescence staining
需将切片放入由PBST配制的 10%山羊血清中,在室温下孵育1小时,然后添加一抗(anti-ACE2,Santa Cruz,sc390851,1:100;anti-SARS-Cov-2 Spike,Sino Biological, 40150-R007,1:500),将切片放入4℃孵育过夜。次日,用 PBST洗涤3次添加荧光二抗(AlexaFluor®647缀合的山羊抗兔 IgG,bs-0296G-AF647,Bioss,1:100;Dylight-550山羊抗兔IgG二抗BA1135,1:200)室温孵育1h。然后用PBST洗涤3次后,用4',6-二脒基-2-苯基吲哚(DAPI)对细胞核复染色。使用激光扫描共聚焦显微镜(LSM880,卡尔·蔡司显微成像)对载玻片成像。Sections need to be placed in 10% goat serum prepared in PBST, incubated at room temperature for 1 hour, and then added with primary antibodies (anti-ACE2, Santa Cruz, sc390851, 1:100; anti-SARS-Cov-2 Spike, Sino Biological, 40150-R007, 1:500), the sections were incubated at 4°C overnight. The next day, wash 3 times with PBST and add fluorescent secondary antibodies (AlexaFluor® 647-conjugated goat anti-rabbit IgG, bs-0296G-AF647, Bioss, 1:100; Dylight-550 goat anti-rabbit IgG secondary antibody BA1135, 1:200 ) at room temperature for 1 h. Nuclei were then counterstained with 4',6-diamidino-2-phenylindole (DAPI) after 3 washes with PBST. Slides were imaged using a laser scanning confocal microscope (LSM880, Carl Zeiss Microscopy).
结果:从COVID-19患者通过内镜检查取得的胃肠道组织中,使用双重免疫荧光染色,在十二指肠中观察到ACE2和SARS-CoV-2 Spike蛋白的共定位,而在十二指肠中可检测到Spike蛋白(图 1a)。H&E染色进一步显示粘膜固有层间质明显水肿,浆细胞和淋巴细胞浸润的局部炎症情况(图 1b)。重要的是,间质性水肿与疾病类型,胃酸倒流,总胆红素,谷氨酸-丙酮酸转氨酶(ALT)和天冬氨酸转氨酶(AST)显著相关(表2),提示该疾病进展的预测特征。RESULTS: Using double immunofluorescence staining, co-localization of ACE2 and SARS-CoV-2 Spike protein was observed in the duodenum in gastrointestinal tissues obtained by endoscopy from patients with COVID-19, whereas in the duodenum Spike proteins were detectable in the denum (Fig. 1a). H&E staining further showed marked edema of the mucosal lamina propria interstitium, local inflammation with plasma cell and lymphocyte infiltration (Fig. 1b). Importantly, interstitial edema was significantly associated with disease type, acid reflux, total bilirubin, glutamate-pyruvate aminotransferase (ALT) and aspartate aminotransferase (AST) (Table 2), suggesting progression of the disease prediction features.
Figure 675483dest_path_image002
Figure 675483dest_path_image002
二、two, SARS-CoV-2SARS-CoV-2 胃肠炎症动物模型的构建Construction of an animal model of gastrointestinal inflammation
所有动物实验程序均按照《实验动物的护理和使用指南》(NIH出版物第 80-23号,1996 年修订)和中山大学第五附属医院制定的《动物实验机构伦理准则》进行。将54只C57BL/6J雌性和雄性小鼠(7-8周龄)放在明/暗周期(14/10 h),恒温(26°C),可以随意获取食物和水的环境。All animal experimental procedures were performed in accordance with the Guide for the Care and Use of Laboratory Animals (NIH Publication No. 80-23, revised in 1996) and the "Ethical Guidelines for Animal Experiment Institutions" formulated by the Fifth Affiliated Hospital of Sun Yat-sen University. Fifty-four C57BL/6J female and male mice (7-8 weeks old) were placed in a light/dark cycle (14/10 h), constant temperature (26°C) environment with ad libitum access to food and water.
1、SARS-CoV-2胃肠炎症动物模型构建过程,包括以下步骤:1. The construction process of SARS-CoV-2 gastrointestinal inflammation animal model, including the following steps:
(1)将8~9周的C57BL/6J小鼠随机分为两组(实验组和对照组);(2)所有小鼠禁食24h;(3)禁食24h后,每只小鼠腹腔注射1%的戊巴比妥100 μL进行麻醉;(4)麻醉5min后,将软管表面涂抹凡士林,后从肛门轻轻插入,插入深度为4cm;(5)软管一端接上注射器,注入500 μL的1%乙酸,对照组注入等体积的PBS;(6)将老鼠倒立1min,后用500 μL的PBS进行灌洗,灌洗两次,目的是洗掉注入的乙酸;(7)将老鼠放回笼,补充食物;(8)16~24h后,腹腔注射S蛋白(SARS-CoV-2 Spike蛋白)5μg/只(或者5.5 nmol/kg),对照组注射同型对照Mouse IgG1-Fc 蛋白;(9)6h后,处死老鼠,取十二指肠、结肠、空回肠和直肠组织,包埋脱水,切片后HE染色;并进行免疫荧光染色实验,考察ACE2和SARS-CoV-2的共定位。(1) 8-9 weeks old C57BL/6J mice were randomly divided into two groups (experimental group and control group); (2) all mice were fasted for 24 hours; (3) after fasting for 24 hours, the abdominal cavity of each mouse was Inject 100 μL of 1% pentobarbital for anesthesia; (4) After 5 minutes of anesthesia, apply petroleum jelly on the surface of the hose, and then gently insert it from the anus to a depth of 4 cm; (5) Connect a syringe to one end of the hose and inject 500 μL of 1% acetic acid, and the control group was injected with an equal volume of PBS; (6) the mice were inverted for 1 min, and then lavaged with 500 μL of PBS twice to wash off the injected acetic acid; (7) The mice were put back into the cage and supplemented with food; (8) 16-24 hours later, the S protein (SARS-CoV-2 Spike protein) 5μg/mice (or 5.5 nmol/kg) was intraperitoneally injected, and the control group was injected with the isotype control Mouse IgG1-Fc protein; (9) After 6 hours, the mice were sacrificed, and the duodenum, colon, jejunum and rectum tissues were taken, embedded and dehydrated, and sliced for HE staining; and immunofluorescence staining was performed to investigate the co-localization of ACE2 and SARS-CoV-2. .
2、免疫共沉淀2. Co-immunoprecipitation
使用Dynabeads Protein G免疫沉淀试剂盒(ThermoFisher),将Spike与相互作用蛋白进行了共免疫沉淀。Spike was co-immunoprecipitated with interacting proteins using the Dynabeads Protein G Immunoprecipitation Kit (ThermoFisher).
结果:从图2b和表3中可以看出,Spike-Fc引起的炎症主要在十二指肠,空回肠、结肠或者直肠未见明显变化,从图2c和图2d中可以看出:Spike-Fc和ACE2在十二指肠实现了共定位,空回肠、结肠或者直肠未见明显变化。这些现象与现有报道的COVID-19患者的临床症状一致,说明上述方法构建的SARS-CoV-2胃肠炎症动物模型可用于复制临床COVID-19患者的胃肠道表现。Results: As can be seen from Figure 2b and Table 3, the inflammation caused by Spike-Fc was mainly in the duodenum, and there was no significant change in the jejunum, colon or rectum. It can be seen from Figure 2c and Figure 2d: Spike-Fc Fc and ACE2 achieved co-localization in the duodenum, and no significant changes were found in the jejunum, colon or rectum. These phenomena are consistent with existing reported clinical symptoms of COVID-19 patients, indicating that the SARS-CoV-2 gastrointestinal inflammation animal model constructed by the above method can be used to replicate the gastrointestinal manifestations of clinical COVID-19 patients.
表3 SARS-CoV-2 Spike蛋白作用于小鼠肠道组织后组织炎症变化及间质水肿变化Table 3 Changes of tissue inflammation and interstitial edema after SARS-CoV-2 Spike protein acts on mouse intestinal tissue
Figure 294683dest_path_image003
Figure 294683dest_path_image003
三、three, SARS-CoV-2 SpikeSARS-CoV-2 Spike 破坏肠血管屏障disrupt the intestinal vascular barrier
1、体内渗透性测定1. In vivo permeability measurement
实验过程:对前述方法SARS-CoV-2胃肠炎症模型小鼠和对照组(Mouse IgG1-Fc 蛋白)尾静脉注射TRITC-dextran,半小时后用2.5mL的PBS腹腔冲洗;取腹水后,腹水离心1500rpm,10min,并使用酶标仪在激发和发射波长分别为540 nm和590 nm处测量荧光;老鼠处死后取十二指肠、空肠、结肠和直肠组织,包埋脱水,切片后H&E染色。Experimental procedure: SARS-CoV-2 gastrointestinal inflammation model mice and control groups (Mouse IgG1-Fc protein) was injected into the tail vein of TRITC-dextran, and after half an hour, the peritoneal cavity was washed with 2.5 mL of PBS; after taking the ascites, the ascites was centrifuged at 1500 rpm for 10 min, and the excitation and emission wavelengths were 540 nm and 590 using a microplate reader, respectively. Fluorescence was measured at nm; duodenal, jejunum, colon and rectal tissues were taken after the mice were sacrificed, embedded and dehydrated, and then sliced and stained with H&E.
结果:Spike蛋白递送后,给小鼠静脉注射TRITC-dextran,随后定量检测漏到腹腔中的dextran。与对照组(Control-Fc)相比,在用Spike-Fc处理的小鼠中观察到更强的渗漏(图 3a),表明Spike蛋白可引起肠血管屏障受损。Results: Following Spike protein delivery, mice were intravenously injected with TRITC-dextran, followed by quantitative detection of dextran leaking into the abdominal cavity. Greater leakage was observed in mice treated with Spike-Fc compared to controls (Control-Fc) (Fig. 3a), suggesting that Spike proteins can cause impairment of the intestinal vascular barrier.
2、体外渗透性测定2. In vitro permeability assay
据报道ACE2可在包括内皮细胞在内的多种类型的细胞表达,因此这里首先研究了Spike蛋白是否可以通过直接影响内皮而介导通透性。我们将人脐静脉内皮细胞(HUVEC)与 Spike-Fc孵育,并通过dextran渗透性测定评估渗透性,实验过程如下:ACE2 has been reported to be expressed in various cell types including endothelial cells, so here we first investigated whether Spike proteins could mediate permeability by directly affecting the endothelium. We incubated human umbilical vein endothelial cells (HUVEC) with Spike-Fc and assessed permeability by dextran permeability assay as follows:
(1)以每个孔2.5×10 5的内皮细胞密度种于transwell小室(0.4 μm,Corning)的上室,使细胞呈单层分布;(2)下室中加入700ul含5%血清的ECM培养基,37℃,5%CO 2培养24小时;(3)然后在transwell小室的上室加入200ul终浓度为2mg/ml的70-KDa TRITC-dextran;(4)5小时后移除上室,分别从下室中取100ul培养基种至96孔板;(5)用多功能微孔板检测仪检测(激发波长540nm,吸收波长590nm)。 (1) The endothelial cells were seeded in the upper chamber of a transwell chamber (0.4 μm, Corning) at a density of 2.5×10 5 per well, so that the cells were distributed in a monolayer; (2) 700ul ECM containing 5% serum was added to the lower chamber Culture medium, 37°C, 5% CO 2 for 24 hours; (3) Then add 200ul of 70-KDa TRITC-dextran with a final concentration of 2 mg/ml to the upper chamber of the transwell chamber; (4) Remove the upper chamber after 5 hours , respectively, take 100ul of culture medium from the lower chamber and seed it into a 96-well plate; (5) detect with a multi-function microplate detector (excitation wavelength 540nm, absorption wavelength 590nm).
结果:Spike蛋白并未对HUVEC细胞的渗透性产生显著影响(图3b)。根据已有报道表明ACE2是SARS-CoV-2的主要结合受体。ACE2在多种器官中表达,包括消化系统和血管系统。也有两项研究利用人小肠样类器官和结肠上皮癌细胞株(Caco-2)证明,SARS-CoV-2以ACE2作为进入受体在肠上皮细胞中复制,同时根据上述体内渗透性实验给出了Spike可引起肠血管屏障受损的结论,因此,这里可以合理的推测Spike蛋白通过影响肠上皮细胞诱导内皮通透性。Results: Spike protein did not significantly affect the permeability of HUVEC cells (Fig. 3b). According to previous reports, ACE2 is the main binding receptor of SARS-CoV-2. ACE2 is expressed in a variety of organs, including the digestive and vascular systems. There are also two studies using human intestinal-like organoids and a colon epithelial cancer cell line (Caco-2) to demonstrate that SARS-CoV-2 replicates in intestinal epithelial cells using ACE2 as an entry receptor, while the in vivo permeability experiments described above give Therefore, it is reasonable to speculate that Spike protein induces endothelial permeability by affecting intestinal epithelial cells.
接下来,将HUVEC细胞与Spike-Fc处理的肠上皮细胞Caco-2的条件培养基一起孵育,通过渗透性测定,过程为:用Spike-Fc(0.25mg/mL)对Caco-2(5×10 5)进行处理,对照组用IgG-Fc(0.25mg/mL),时间为24h,然后用0.22 μm滤网过滤培养的Caco-2细胞上清液。将HUVEC细胞(1×10 5)单层铺在24孔板的上室过夜。在细胞粘附所需的孵育时间后,将移孔放入相同的24孔板中,添加1%FBS的培养基直至HUVEC达到融合6 h,然后,将经过滤的Caco-2上清液换到Transwell室的底部,并与HUVEC共同培养12 h。为了体外检测HUVEC的渗透性,去除上腔室培养基,并向上孔加入异硫氰酸四甲基罗丹明-Dextran(T1162,Sigma)(2 mg/mL),3小时后,收集加入Dextran的培养基,并使用酶标仪在激发和发射波长分别为540 nm和590 nm处测量荧光。 Next, HUVEC cells were incubated with conditioned medium of Spike-Fc-treated intestinal epithelial cells Caco-2, as determined by permeability, using Spike-Fc (0.25 mg/mL) for Caco-2 (5× 10 5 ), the control group was treated with IgG-Fc (0.25mg/mL) for 24h, and then the cultured Caco-2 cell supernatant was filtered through a 0.22 μm filter. HUVEC cells (1 x 105 ) were plated in a monolayer in the upper chamber of a 24-well plate overnight. After the incubation time required for cell adhesion, the wells were pipetted into the same 24-well plate, medium with 1% FBS was added until the HUVECs reached confluency for 6 h, and then the filtered Caco-2 supernatant was replaced to the bottom of the Transwell chamber and co-cultured with HUVEC for 12 h. To test the permeability of HUVECs in vitro, the upper chamber medium was removed, and tetramethylrhodamine isothiocyanate-Dextran (T1162, Sigma) (2 mg/mL) was added to the upper wells. After 3 hours, the Dextran-added cells were collected. culture medium, and fluorescence was measured at excitation and emission wavelengths of 540 nm and 590 nm, respectively, using a microplate reader.
结果:检测到暴露于Spike蛋白刺激的Caco-2上清液的内皮细胞有明显渗透现象(图3e)。RESULTS: Significant infiltration of endothelial cells exposed to Spike protein-stimulated Caco-2 supernatants was detected (Fig. 3e).
3、SARS-CoV-2 Spike 蛋白结合实验3. SARS-CoV-2 Spike protein binding assay
重组SARS-CoV-2 Spike蛋白(RBD,Fc标签)购自Sino Bioloical。为了进行体外结合测定,将Caco-2细胞和细胞分别与Spike-Fc(0.25mg / mL)或对照 IgG-Fc(0.25mg / mL)于37℃孵育1h,然后用Protein G Sepharose下拉裂解液中的蛋白,进行免疫印迹。在Caco-2 中与Spike-Fc(0.25mg / mL)或对照IgG-Fc(0.25mg / mL)于37℃孵育1h或24h,进行下游分子分析。用western Blot和定量PCR分析细胞裂解液,将细胞培养的上清液(24h)用 0.22μm滤膜(MILLEX GP)过滤,并在-80℃下保存以用于Elisa和共培养实验。Recombinant SARS-CoV-2 Spike protein (RBD, Fc tag) was purchased from Sino Bioloical. For in vitro binding assays, Caco-2 cells and cells were treated with Spike-Fc (0.25 mg/mL) or control, respectively IgG-Fc (0.25 mg/mL) was incubated at 37 °C for 1 h, and then the protein in the lysate was pulled down with Protein G Sepharose for immunoblotting. in Caco-2 Neutralized with Spike-Fc (0.25mg /mL) or control IgG-Fc (0.25mg /mL) at 37°C for 1 h or 24 h for downstream molecular analysis. Cell lysates were analyzed by western blot and quantitative PCR, and cell culture supernatants (24 h) were filtered through 0.22 μm filters (MILLEX GP) and stored at -80°C for Elisa and co-culture experiments.
结果:Spike-Fc蛋白与人ACE2的结合通过下拉测定法(图 4a)和免疫荧光染色(图4b)得到了证实。上述实验数据表明:Spike蛋白可以与肠上皮细胞上的ACE2结合,并可能通过旁分泌途径破坏肠道血管屏障系统。Results: The binding of Spike-Fc protein to human ACE2 was confirmed by pull-down assay (Fig. 4a) and immunofluorescence staining (Fig. 4b). The above experimental data indicate that Spike protein can bind to ACE2 on intestinal epithelial cells and may disrupt the intestinal vascular barrier system through the paracrine pathway.
四、Four, SARS-CoV-2 SpikeSARS-CoV-2 Spike 破坏肠道血管屏障的机制探究Mechanisms of disruption of the intestinal vascular barrier
1、免疫组织化学1. Immunohistochemistry
对于组织样品,进行免疫组织化学以确定ERK,pERK,VE-cad 和 pVE-cad(Y731)蛋白在十二指肠、空回肠、结肠和直肠中的表达水平。随后,还对人/小鼠组织切片进行了HE染色,以评估组织形态特征和靶蛋白的分布。For tissue samples, immunohistochemistry was performed to determine the expression levels of ERK, pERK, VE-cad and pVE-cad(Y731) proteins in the duodenum, jejunum, colon and rectum. Subsequently, HE staining was also performed on human/mouse tissue sections to assess tissue morphological characteristics and distribution of target proteins.
结果:紧密连接和粘附连接是肠血管屏障的基本组成部分。首先分析了紧密连接蛋白(例如ZO-1)的变化,但没有发现明显改变。但是,一种关键的粘附蛋白VE-cadherin(VE-cad)的表达降低了,并伴随着其在Tyr731位点而非658位点的磷酸化(pVE-cad)增加(图5a)。 然后,我们在体内测量VE-cad的表达和磷酸化水平。用Spike蛋白处理过的小鼠的胃肠道的不同部分进行了蛋白质印迹和免疫组化(IHC)分析。结果显示十二指肠中VE-cad持续减少(图5b),pVE-cad持续增加(图5c)。另外,通过实验我们还证实了COVID-19患者的组织也具有这种变化(图5d,图5e)。以上结果表明,Spike通过抑制VE-cad表达促进内皮通透性,同时增加其磷酸化。Results: Tight junctions and adherent junctions are the basic components of the intestinal vascular barrier. Changes in tight junction proteins such as ZO-1 were first analyzed, but no significant changes were found. However, the expression of a key adhesion protein, VE-cadherin (VE-cad), was reduced, accompanied by an increase in its phosphorylation at Tyr731 but not at 658 (pVE-cad) (Fig. 5a). Then, we measured the expression and phosphorylation levels of VE-cad in vivo. Different parts of the gastrointestinal tract of mice treated with Spike protein were analyzed by western blot and immunohistochemistry (IHC). The results showed a persistent decrease in VE-cad in the duodenum (Fig. 5b) and a persistent increase in pVE-cad (Fig. 5c). In addition, we also confirmed this change in the tissues of COVID-19 patients through experiments (Fig. 5d, Fig. 5e). The above results suggest that Spike promotes endothelial permeability by inhibiting VE-cad expression while increasing its phosphorylation.
五、five, SARS-CoV-2 SpikeSARS-CoV-2 Spike 蛋白通过protein through ERK/VEGFERK/VEGF 途径介导血管通透性pathway mediates vascular permeability
1、RNA分离和qRT-PCR1. RNA isolation and qRT-PCR
立即从固定在液氮中的SARS-CoV-2胃肠炎症模型小鼠(包括对照组和实验组)中切除组织。通过常规RNA提取方案,使用TRIzol分离组织的总RNA。用总RNA试剂盒I分离细胞的总RNA。将分离的RNA反转录为cDNA(Vazyme,Nanjing,China)。使用实时PCR 系统(Bio-Rad,America)和ChamQ Universal SYBR qPCR预混液(Vazyme,Nanjing, China)进行qRT-PCR。引物是由广州IGE生物技术公司设计和合成的。GAPDH用作内部对照,所有反应均重复三次。使用 2 - ΔΔ Ct 方法计算相对RNA表达。 Tissues were immediately excised from SARS-CoV-2 gastrointestinal inflammation model mice (including control and experimental groups) fixed in liquid nitrogen. Total RNA from tissues was isolated using TRIzol by conventional RNA extraction protocols. Total RNA from cells was isolated with Total RNA Kit I. The isolated RNA was reverse transcribed into cDNA (Vazyme, Nanjing, China). qRT-PCR was performed using a real-time PCR system (Bio-Rad, America) and ChamQ Universal SYBR qPCR master mix (Vazyme, Nanjing, China). Primers were designed and synthesized by Guangzhou IGE Biotechnology Company. GAPDH was used as an internal control and all reactions were repeated three times. Relative RNA expression was calculated using the 2 - ΔΔCt method.
2、ELISA2. ELISA
通过ELISA检测从细胞,组织和血清产生的VEGF的浓度。对于细胞培养的上清液,根据制造商的说明书通过Quantikine ELISA人VEGF免疫测定法(cat. No. DVE00, R&D)测定 VEGF浓度。对于小鼠样品,按照制造商的说明,使用小鼠VEGF Simplestep ELISA试剂盒(cat. No. ab209882, Abcam)测定组织匀浆(在冷PBS中使用电动匀浆器制备)和血清中的VEGF水平。The concentration of VEGF produced from cells, tissues and serum was detected by ELISA. For cell culture supernatants, Quantikine ELISA human VEGF immunoassay (cat. No. DVE00, R&D) Determination of VEGF concentration. For mouse samples, use mouse VEGF according to the manufacturer's instructions Simplestep ELISA kit (cat. No. ab209882, Abcam) to measure VEGF levels in tissue homogenates (prepared in cold PBS using an electric homogenizer) and serum.
结果:在Spike处理的小鼠十二指肠组织中,无论是mRNA还是蛋白水平,VEGF的表达均显著升高(图6a,6b)。值得注意的是,这些小鼠血清中的VEGF 水平没有改变(图 6d)。因此这里的结论与体外共培养结果一致,即在Spike刺激下产生更多VEGF的是上皮细胞,而不是内皮细胞(图6c)。Results: In the duodenal tissue of Spike-treated mice, the expression of VEGF was significantly increased at both mRNA and protein levels (Fig. 6a, 6b). Notably, VEGF levels in the serum of these mice were not altered (Fig. 6d). The conclusion here is therefore consistent with the in vitro co-culture results that it is epithelial cells, but not endothelial cells, that produce more VEGF upon Spike stimulation (Fig. 6c).
被称为有效的血管通透性因子的VEGF在COVID-19患者的血液样本中被上调。使用细胞模型的研究表明,SARS-CoV-2感染可显著增加人肺上皮细胞中VEGF的表达。基于这些发现,我们推测VEGF可以在肠细胞中被病毒诱导,这可能是COVID-19患者产生胃肠道症状的原因。上述的实验数据验证了我们的预想。VEGF, known as a potent vascular permeability factor, was upregulated in blood samples from COVID-19 patients. Studies using cellular models have shown that SARS-CoV-2 infection significantly increases VEGF expression in human lung epithelial cells. Based on these findings, we speculate that VEGF can be induced by viruses in enterocytes, which may be responsible for gastrointestinal symptoms in COVID-19 patients. The above experimental data verified our expectation.
3、敲低ACE2的Caco-2细胞构建,包括以下步骤:3. Construction of Caco-2 cells knocking down ACE2, including the following steps:
(1)Caco-2细胞接六孔板;(2)用Lipofectamine LTX(Invitrogen公司,ThermoFisher Scientific公司)转染试剂,将阴性对照siRNA和ACE2 siRNA转染Caco-2细胞;(3)48h后使用含有蛋白酶/磷酸酶抑制剂混合物的RIPA缓冲液裂解Caco-2,提取蛋白;(4)Western blot检测敲降效率和相关蛋白的表达。(1) Caco-2 cells were attached to a six-well plate; (2) Lipofectamine was used LTX (Invitrogen, ThermoFisher Scientific Corporation) transfection reagent, and the negative control siRNA and ACE2 siRNA were transfected into Caco-2 cells; (3) After 48h, use RIPA buffer containing protease/phosphatase inhibitor mixture to lyse Caco-2, and extract the protein; (4) Western blot was used to detect the knockdown efficiency and the expression of related proteins.
用阴性对照siRNA和ACE2 siRNA转染Caco-2 48小时,使用Lipofectamine LTX (Invitrogen公司,ThermoFisher Scientific公司),然后使用含有蛋白酶/磷酸酶抑制剂混合物的RIPA缓冲液裂解Caco-2,准备western blot。Caco-2 was transfected with negative control siRNA and ACE2 siRNA for 48 hours using Lipofectamine LTX (Invitrogen, ThermoFisher Scientific), then Caco-2 was lysed using RIPA buffer containing a protease/phosphatase inhibitor cocktail in preparation for a western blot.
到目前为止,我们已经证明了肠上皮是VEGF的产生者,它在Spike诱导的通透性中起着关键作用。据报道,激活ERK信号通路可导致人结肠细胞中VEGF的过度表达。我们在用Spike蛋白处理的Caco-2细胞中验证了该通路。免疫印迹检测到了可靠的ERK激活,包括总ERK和磷酸化ERK(pERK)的升高(图7)。So far, we have shown that the intestinal epithelium is a producer of VEGF, which plays a key role in Spike-induced permeability. Activation of the ERK signaling pathway has been reported to lead to overexpression of VEGF in human colon cells. We validated this pathway in Caco-2 cells treated with Spike protein. Reliable ERK activation was detected by immunoblotting, including increases in total ERK and phosphorylated ERK (pERK) (Figure 7).
我们在敲低ACE2的Caco-2细胞中检测了ERK上游分子Ras,Raf和MEK。以确保其在ACE2的下游发挥作用,从而明确了该途径(图 8a)。接下来,我们在Spike处理的Caco-2细胞中检测了该途径中的每个分子,并确认了每个分子的改变(图9a)。此外,可以通过使用ERK抑制剂SCH772984来抑制这种变化(图9a)。证明了SCH772984完全逆转了Spike处理的Caco-2细胞中过表达的VEGF(图9b)。同时,Spike处理过的HUVEC细胞中的ERK途径保持不变(图8b)。重要的是,在动物组织(图9c)和COVID-19患者的活检标本(图9d,9e)中也证实了ERK和pERK的改变。综上,这些发现定义了ERK/VEGF途径在Spike介导的通透性中的关键作用。We detected ERK upstream molecules Ras, Raf and MEK in ACE2 knockdown Caco-2 cells. to ensure that it functions downstream of ACE2, thus clarifying the pathway (Figure 8a). Next, we examined each molecule in this pathway in Spike-treated Caco-2 cells and confirmed alterations in each (Fig. 9a). Furthermore, this change could be suppressed by using the ERK inhibitor SCH772984 (Fig. 9a). demonstrated that SCH772984 completely reversed overexpressed VEGF in Spike-treated Caco-2 cells (Figure 9b). Meanwhile, the ERK pathway in Spike-treated HUVEC cells remained unchanged (Fig. 8b). Importantly, alterations in ERK and pERK were also demonstrated in animal tissues (Fig. 9c) and biopsy specimens from COVID-19 patients (Figs. 9d, 9e). Taken together, these findings define a critical role for the ERK/VEGF pathway in Spike-mediated permeability.
六、阻断6. Blocking ERK/VEGFERK/VEGF 通路可拯救Pathways can be rescued SARS-CoV-2 SpikeSARS-CoV-2 Spike 诱导的血管通透性过高Induced vascular hyperpermeability
1、体外细胞渗透性测定1. In vitro cell permeability assay
用Spike-Fc(0.25mg/mL)对Caco-2(5×10 5)进行处理,对照组用IgG-Fc(0.25mg/mL),Spike-Fc与SCH772984(1uM)、贝伐珠单抗Bevacizumab(25ug/ml)细胞分别处理24h,然后用0.22 μm滤网过滤培养的Caco-2细胞上清液。将HUVEC(1×10 5)单层铺在24孔板的上室过夜。在细胞粘附所需的孵育时间后,将移孔放入相同的24孔板中,添加1%FBS的培养基直至HUVEC细胞达到融合6 h,然后,将经过滤的Caco-2细胞上清液换成Transwell室的底部,并与 HUVEC细胞共同培养12 h。为了体外检测HUVEC细胞的渗透性,去除上腔室培养基,并向上孔加入异硫氰酸四甲基罗丹明-Dextran(T1162,Sigma)(2 mg/mL),3小时后,收集加入Dextran的培养基,并使用酶标仪在激发和发射波长分别为540nm和590nm处测量荧光。 Caco-2 (5×10 5 ) was treated with Spike-Fc (0.25mg/mL), control group was treated with IgG-Fc (0.25mg/mL), Spike-Fc with SCH772984 (1uM), bevacizumab Bevacizumab (25ug/ml) cells were treated for 24h, and then the cultured Caco-2 cell supernatant was filtered through a 0.22 μm filter. A monolayer of HUVEC ( 1 x 105) was plated in the upper chamber of a 24-well plate overnight. After the incubation time required for cell adhesion, the wells were pipetted into the same 24-well plate, medium with 1% FBS was added until the HUVEC cells reached confluency for 6 h, and then the filtered Caco-2 cell supernatant was added. The solution was changed to the bottom of the Transwell chamber and co-cultured with HUVEC cells for 12 h. To test the permeability of HUVEC cells in vitro, the upper chamber medium was removed, and tetramethylrhodamine isothiocyanate-Dextran (T1162, Sigma) (2 mg/mL) was added to the upper well. After 3 hours, the collection was added with Dextran of medium, and measured fluorescence using a microplate reader at excitation and emission wavelengths of 540 nm and 590 nm, respectively.
2、体内动物实验,包括以下步骤:2. In vivo animal experiments, including the following steps:
(1)将8~9周的C57BL/6J小鼠随机分为六组:对照组、安慰剂组I、安慰剂组II、Spike蛋白组、SCH772984组和贝伐珠单抗组;(2)所有小鼠禁食24h;(3)禁食24h后,每只小鼠腹腔注射1%的戊巴比妥100 μL进行麻醉;(4)麻醉5min后,将软管表面涂抹凡士林,后从肛门轻轻插入,插入深度为4cm;(5)软管一端接上注射器,注入500 μL的1%乙酸,对照组注入等体积的PBS;(6)将老鼠倒立1min,后用500 μL的PBS进行灌洗,灌洗两次,目的是洗掉注入的乙酸;(7)将老鼠放回笼,补充食物;(8)16~24h后,对SCH772984组小鼠按照50mg/kg的用量进行腹腔注射,对贝伐珠单抗组按照5mg/kg的用量进行腹腔注射,对安慰剂组I小鼠腹腔注射SCH772984的溶剂,对安慰剂组II小鼠腹腔注射贝伐珠单抗的溶剂,对照组不做处理;(9)1~2h后,SCH772984组、贝伐珠单抗组和安慰剂组I、安慰剂组II分别腹腔注射S蛋白(SARS-CoV-2 Spike蛋白)5μg/只(或者5.5 nmol/kg),对照组注射同型对照Mouse IgG1-Fc 蛋白;(10)1~2h再次对SCH772984组小鼠按照50mg/kg的用量进行腹腔注射,对贝伐珠单抗组按照5mg/kg的用量进行腹腔注射,安慰剂组I注射SCH772984的溶剂,安慰剂组II注射贝伐珠单抗的溶剂;(11)4~6h后尾静脉注射TRITC-dextran,半小时后2.5mlPBS腹腔冲洗;(12)取腹水后,将腹水以1500rpm转速离心10min,并使用酶标仪在激发和发射波长分别为540nm和590 nm处测量荧光;(13)处死老鼠,取十二指肠、结肠、空回肠和直肠组织,部分组织包埋脱水,切片后HE染色;并进行免疫荧光染色实验,考察ACE2和SARS-CoV-2的共定位。部分新鲜组织用Elisa试剂盒检测组织中VEGF的含量。(1) 8-9 weeks old C57BL/6J mice were randomly divided into six groups: control group, placebo group I, placebo group II, Spike protein group, SCH772984 group and bevacizumab group; (2) All mice were fasted for 24 hours; (3) After fasting for 24 hours, each mouse was anesthetized by intraperitoneal injection of 100 μL of 1% pentobarbital; (4) After 5 minutes of anesthesia, the surface of the hose was smeared with Vaseline, and then injected from the anus. Gently insert, the insertion depth is 4cm; (5) Connect a syringe to one end of the hose, inject 500 μL of 1% acetic acid, and the control group is injected with an equal volume of PBS; (6) Invert the mouse for 1 min, and then use 500 μL of PBS for Lavage, lavage twice, in order to wash off the injected acetic acid; (7) Put the mice back in the cage and supplement with food; (8) After 16-24 hours, the mice in the SCH772984 group were injected intraperitoneally at a dose of 50 mg/kg, The bevacizumab group was injected intraperitoneally at a dose of 5 mg/kg, the placebo group I mice were intraperitoneally injected with the solvent of SCH772984, the placebo group II mice were intraperitoneally injected with the bevacizumab solvent, and the control group did not. (9) After 1-2 hours, the SCH772984 group, the bevacizumab group, the placebo group I, and the placebo group II were given intraperitoneal injection of S protein (SARS-CoV-2 Spike protein) 5 μg / animal (or 5.5 μg) respectively. nmol/kg), the control group was injected with the same type control Mouse IgG1-Fc protein; (10) 1~2h, the mice in the SCH772984 group were again injected intraperitoneally at a dose of 50 mg/kg, the bevacizumab group was injected at a dose of 5 mg/kg, and the placebo group I was injected with SCH772984 (11) TRITC-dextran was injected into the tail vein after 4-6 hours, and 2.5ml of PBS was washed in the abdominal cavity after half an hour; (12) after taking the ascites, the ascites was rotated at 1500rpm. Centrifuge for 10 min, and use a microplate reader to measure the fluorescence at excitation and emission wavelengths of 540 nm and 590 nm, respectively; (13) The mice were sacrificed, and the duodenum, colon, jejunum, ileum, and rectum were taken, and some tissues were embedded, dehydrated, and sliced. After HE staining; and immunofluorescence staining experiments were performed to investigate the co-localization of ACE2 and SARS-CoV-2. The content of VEGF in some fresh tissues was detected by Elisa kit.
3、免疫荧光分析3. Immunofluorescence analysis
为了检测内皮的粘附连接,将HUVECs接种到15mm玻璃底部细胞培养皿(2.5×10 5)中,用Caco-2的上清液(分别用Spike-Fc和IgG-Fc处理)共培养24h,然后用4%多聚甲醛固定。依次用以下抗体或荧光染料对样本进行染色:VE-Cadherin(Cat.No.44-1145G,ThermoFisher,1:1,000),Dylight-488 Goat Anti-mouse IgG secondary antibody(Cat.No. BA1126,BOSTER,1:200),Antifade Mounting Medium with DAPI(Cat.No. Ab104139, Abcam)。用共聚焦显微镜对结果进行采集,样本图片是由Zeiss 880用60倍物镜拍摄的,并使用ImageJ 软件进行图像处理。 To detect endothelial adherent junctions, HUVECs were seeded into 15 mm glass bottom cell culture dishes (2.5×10 5 ) and co-cultured with the supernatant of Caco-2 (treated with Spike-Fc and IgG-Fc, respectively) for 24 h. It was then fixed with 4% paraformaldehyde. Samples were stained sequentially with the following antibodies or fluorescent dyes: VE-Cadherin (Cat.No.44-1145G, ThermoFisher, 1:1,000), Dylight-488 Goat Anti-mouse IgG secondary antibody (Cat.No. BA1126, BOSTER, 1:200), Antifade Mounting Medium with DAPI (Cat. No. Ab104139, Abcam). The results were acquired with a confocal microscope, and the sample pictures were taken by a Zeiss 880 with a 60x objective lens, and image processing was performed using ImageJ software.
结果:为了评估在血管屏障功能中阻断 ERK/VEGF通路的功效,我们引入了ERK抑制剂SCH772984和抗VEGF药物贝伐珠单抗(Bevacizumab),该药物通常被用作治疗癌症血管通透性过高的一线药物,分别进行肠上皮细胞-内皮细胞共培养。SCH772984和贝伐珠单抗均显著抑制VE-cad的内化(图10a)并挽救了SARS-CoV-2 Spike 蛋白诱导的渗透(图10b)。为了在体内证实这些发现,对Spike处理的动物给予了SCH772984或贝伐珠单抗。结果表明,VE-Cad得以恢复(图10c),伴随任一药物治疗时VEGF 的生成减少(图11a),最有可能是通过十二指肠组织中ERK / pERK的下调(图11b)。这些数据表明,阻断ERK/VEGF通路可以恢复被Spike蛋白破坏的血管屏障功能。Results: To assess the efficacy of blocking the ERK/VEGF pathway in vascular barrier function, we introduced the ERK inhibitor SCH772984 and the anti-VEGF drug Bevacizumab, which is commonly used to treat cancer vascular permeability For high first-line drugs, enteroepithelial-endothelial cell co-cultures were performed separately. Both SCH772984 and bevacizumab significantly inhibited the internalization of VE-cad (Fig. 10a) and rescued the SARS-CoV-2 Spike Protein-induced osmosis (Fig. 10b). To confirm these findings in vivo, Spike-treated animals were administered SCH772984 or bevacizumab. The results showed that VE-Cad was restored (Fig. 10c), accompanied by a decrease in VEGF production with either drug treatment (Fig. 11a), most likely through ERK/ Downregulation of pERK (Fig. 11b). These data suggest that blocking the ERK/VEGF pathway restores the vascular barrier function disrupted by the Spike protein.
七、seven, SCH772984SCH772984 或贝伐珠单抗均可缓解or bevacizumab SARS-CoV-2 SpikeSARS-CoV-2 Spike 蛋白诱导的炎症protein-induced inflammation
1、Evans Blue试验,包括以下步骤:1. Evans Blue test, including the following steps:
(1)提前将小鼠禁食24h;(2)鼠腹腔注射100ul的1%的戊巴比妥;(3)5min左右等小鼠麻醉后,将软管表面涂抹凡士林,将软管从肛门轻轻插入,深入长度为4cm;(4)软管一端接注射器,注入500ul的1%乙酸;(5)注入后将老鼠倒立1min,后用1ml的PBS进行灌洗,灌洗两次;(6)将老鼠放回笼,并补充食物;(7)14h后,对SCH772984组小鼠按照SCH772984:50mg/kg的用量进行腹腔注射,对贝伐珠单抗组按照5mg/kg的用量进行腹腔注射,对安慰剂组I小鼠腹腔注射SCH772984的溶剂,对安慰剂组II小鼠腹腔注射贝伐珠单抗的溶剂,对照组不做处理;(8)SCH772984组、贝伐珠单抗组和安慰剂组I、安慰剂组II分别腹腔注射S蛋白(SARS-CoV-2 Spike蛋白)5μg/只(或者5.5 nmol/kg),对照组注射同型对照Mouse IgG1-Fc 蛋白;(9)2h后再次对SCH772984组小鼠按照50mg/kg的用量进行腹腔注射,对贝伐珠单抗组按照5mg/kg的用量进行腹腔注射,安慰剂组I注射SCH772984的溶剂,安慰剂组II注射贝伐珠单抗的溶剂;(10)4h后尾静脉注射evens blue dye,注射量为25mg/kg,半小时后处死小鼠;(11)老鼠处死后取十二指肠、空回肠、结肠和直肠组织,拍照;(12)拍照后对小鼠组织进行称重,按照1ml/100mg的量来加入甲酰胺,50℃,培养箱摇动放置48h;(13)8000g,离心10min后,取100ul的上清液在620nm处测OD值。(1) The mice were fasted for 24 hours in advance; (2) 100ul of 1% pentobarbital was injected intraperitoneally into the mice; (3) After the mice were anesthetized for about 5 minutes, apply Vaseline to the surface of the tube, and the tube was removed from the anus. Gently insert it with a depth of 4cm; (4) The hose is connected to a syringe at one end, and 500ul of 1% acetic acid is injected; (5) After injection, the mouse is inverted for 1 min, and then lavaged with 1ml of PBS, and lavaged twice; ( 6) Put the mice back into the cage and supplement with food; (7) After 14 hours, the mice in the SCH772984 group were injected intraperitoneally according to the dosage of SCH772984: 50 mg/kg, and the mice in the bevacizumab group were intraperitoneally injected with the dosage of 5 mg/kg , the mice in the placebo group I were intraperitoneally injected with the solvent of SCH772984, and the mice in the placebo group II were injected with the solvent of bevacizumab, and the control group was not treated; (8) SCH772984 group, bevacizumab group and Placebo group I and placebo group II were intraperitoneally injected with S protein (SARS-CoV-2 Spike protein) 5 μg/mouse (or 5.5 nmol/kg), respectively, and the control group was injected with the isotype control Mouse IgG1-Fc protein; (9) 2 hours later The mice in the SCH772984 group were again injected intraperitoneally at a dose of 50 mg/kg, the bevacizumab group was injected at a dose of 5 mg/kg, the placebo group I was injected with the solvent of SCH772984, and the placebo group II was injected with bevacizumab The solvent of monoclonal antibody; (10) 4 hours later, evens blue dye was injected into the tail vein, and the injection amount was 25 mg/kg, and the mice were sacrificed after half an hour; (11) duodenum, jejunum, ileum, colon and rectum were collected after the mice were sacrificed. , take pictures; (12) Weigh the mouse tissue after taking pictures, add formamide in an amount of 1ml/100mg, 50°C, shake in an incubator for 48h; (13) 8000g, centrifuge for 10min, take 100ul of supernatant The OD value of the solution was measured at 620 nm.
结果:为了研究在临床前环境中靶向ERK/VEGF途径改善SARS-CoV-2诱导的通透性和炎症的潜力,首先通Evans Blue试验确定了SCH772984或贝伐珠单抗治疗后胃肠道渗透性变化。从SCH772984或贝伐珠单抗治疗组渗入十二指肠组织的Evans Blue染料的量几乎与对照组相同(图12a左侧)。如预期的那样,在空回肠,结肠或直肠中均未检测到明显的影响(图12b)。病理上,经SCH772984或贝伐珠单抗治疗,十二指肠炎症减轻,反映了更强的屏障(图12c)。上述结果表明,ERK/VEGF的抑制可以挽救SARS-CoV-2 Spike蛋白诱导的胃肠道受损的血管屏障和继发性炎症,从而突出了一种治疗COVID-19患者胃肠道症状的新策略。Results: To investigate the potential of targeting the ERK/VEGF pathway in a preclinical setting to improve SARS-CoV-2-induced permeability and inflammation, the gastrointestinal tract following SCH772984 or bevacizumab treatment was first identified by Evans Blue assay. Permeability changes. Evans infiltrated into duodenal tissue from SCH772984 or bevacizumab treatment groups The amount of Blue dye was almost the same as that of the control group (Fig. 12a left). As expected, no significant effects were detected in either the jejunum, colon or rectum (Fig. 12b). Pathologically, duodenal inflammation was reduced with SCH772984 or bevacizumab treatment, reflecting a stronger barrier (Fig. 12c). The above results suggest that ERK/VEGF inhibition rescues the damaged vascular barrier and secondary inflammation in the gastrointestinal tract induced by the SARS-CoV-2 Spike protein, thereby highlighting a novel approach for the treatment of gastrointestinal symptoms in patients with COVID-19. Strategy.

Claims (19)

  1. 抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的方法,其特征在于,包括给胃肠道组织施用血管的排斥性导向信号。A method of inhibiting SARS-CoV-2-induced damage to the vascular barrier in gastrointestinal tissue, comprising administering to the gastrointestinal tissue a vascular rejection targeting signal.
  2. 根据权利要求1所述的抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的方法,其特征在于,所述排斥性导向信号包括ERK抑制剂和/或VEGF拮抗剂,所述ERK抑制剂是指能与ERK结合并阻断ERK生物活性的物质,所述VEGF拮抗剂是指能与VEGF结合并阻断VEGF生物活性的物质。The method for inhibiting vascular barrier damage in gastrointestinal tract tissue caused by SARS-CoV-2 according to claim 1, wherein the repulsive guidance signal comprises an ERK inhibitor and/or a VEGF antagonist, and the An ERK inhibitor refers to a substance that can bind to ERK and block the biological activity of ERK, and the VEGF antagonist refers to a substance that can bind to VEGF and block the biological activity of VEGF.
  3. 根据权利要求2所述的抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的方法,其特征在于,所述ERK抑制剂包括但不限于SCH772984、GDC-0994、Ulixertinib、KO-947、LY3214996、MK-8353、CC-90003、LTT462、FR180204、VTX-11e、BL-EI-001。The method for inhibiting vascular barrier damage in gastrointestinal tract tissue caused by SARS-CoV-2 according to claim 2, wherein the ERK inhibitors include but are not limited to SCH772984, GDC-0994, Ulixertinib, KO- 947, LY3214996, MK-8353, CC-90003, LTT462, FR180204, VTX-11e, BL-EI-001.
  4. 根据权利要求2所述的抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的方法,其特征在于,所述VEGF拮抗剂包括但不限于贝伐珠单抗、雷莫芦单抗、雷珠单抗、阿柏西普、康柏西普。The method for inhibiting vascular barrier damage in gastrointestinal tissue caused by SARS-CoV-2 according to claim 2, wherein the VEGF antagonists include but are not limited to bevacizumab, ramucirumab Antibiotic, ranibizumab, aflibercept, conbercept.
  5. 根据权利要求1至4任一项所述的抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的方法,其特征在于,所述胃肠道组织为十二指肠。The method for inhibiting damage to the vascular barrier in gastrointestinal tract tissue caused by SARS-CoV-2 according to any one of claims 1 to 4, wherein the gastrointestinal tract tissue is duodenum.
  6. 筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,确定所述药剂影响VEGF介导的胃肠道血管通透性增加的能力。A method of screening or evaluating an agent that inhibits SARS-CoV-2-induced impairment of the vascular barrier in gastrointestinal tissue, characterized by determining the ability of the agent to affect VEGF-mediated increase in vascular permeability of the gastrointestinal tract.
  7. 根据权利要求6所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,确定所述药剂影响VEGF介导的胃肠道血管通透性增加的能力是指确定所述药剂抑制ERK、MEK、Ras、Raf、VEGF的能力。The method for screening or evaluating an agent that inhibits vascular barrier damage in gastrointestinal tract tissue caused by SARS-CoV-2 according to claim 6, wherein the agent is determined to affect VEGF-mediated vascular access in the gastrointestinal tract The ability to increase permeability refers to determining the ability of the agent to inhibit ERK, MEK, Ras, Raf, VEGF.
  8. 根据权利要求7所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,所述药剂影响VEGF介导的胃肠道血管通透性增加通过以下步骤确定:The method for screening or evaluating an agent that inhibits SARS-CoV-2-induced damage to the vascular barrier in gastrointestinal tissue according to claim 7, wherein the agent affects VEGF-mediated vascular permeability of the gastrointestinal tract Increased sexuality is determined by the following steps:
    (1)准备候选药剂组,所述候选药剂组由接触SARS-CoV-2 Spike蛋白的肠道上皮细胞的分泌物、内皮细胞与候选药剂组成;(1) Prepare a candidate agent group, which consists of secretions of intestinal epithelial cells exposed to SARS-CoV-2 Spike protein, endothelial cells and candidate agents;
    (2)准备安慰剂组,所述第安慰剂组由接触SARS-CoV-2 Spike蛋白的肠道上皮细胞的分泌物、内皮细胞与安慰剂组成;(2) Prepare a placebo group, which consists of secretions of intestinal epithelial cells exposed to SARS-CoV-2 Spike protein, endothelial cells and placebo;
    (3)其中,候选药剂组中内皮细胞与安慰剂组中内皮细胞相比,能够检测到更高的VE-cad的表达量或更低的VE-cad磷酸化水平,表明所述候选药剂能抑制VEGF介导的胃肠道血管通透性增加。(3) Among them, the endothelial cells in the candidate drug group can detect higher VE-cad expression or lower VE-cad phosphorylation level than the endothelial cells in the placebo group, indicating that the candidate drug can Inhibits VEGF-mediated increase in gastrointestinal vascular permeability.
  9. 根据权利要求7所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,所述药剂影响VEGF介导的胃肠道血管通透性增加通过以下步骤确定:The method for screening or evaluating an agent that inhibits SARS-CoV-2-induced damage to the vascular barrier in gastrointestinal tissue according to claim 7, wherein the agent affects VEGF-mediated vascular permeability of the gastrointestinal tract Increased sexuality is determined by the following steps:
    (1)构建SARS-CoV-2胃肠炎症动物模型;(1) Construct an animal model of SARS-CoV-2 gastrointestinal inflammation;
    (2)对(1)的模型动物腹腔注射或者不注射安慰剂;(2) Intraperitoneal injection or no placebo injection to the model animals of (1);
    (3)对(1)的模型动物腹腔注射候选药剂;(3) Inject the candidate drug into the model animal of (1) by intraperitoneal injection;
    (4)其中,(3)处理后的动物胃肠道组织中和(2)处理后的动物的胃肠道组织中相比,能够检测到更低的VEGF的表达量,表明所述药剂能抑制VEGF介导的胃肠道血管通透性增加。(4) wherein, a lower expression level of VEGF can be detected in the gastrointestinal tract tissue of the animals after (3) treatment than in the gastrointestinal tract tissue of the animals after treatment (2), indicating that the agent can Inhibits VEGF-mediated increase in gastrointestinal vascular permeability.
  10. 根据权利要求9所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,步骤(1)所述SARS-CoV-2胃肠炎症动物模型的构建方法包括以下步骤:The method for screening or evaluating agents for inhibiting vascular barrier damage in gastrointestinal tract tissue caused by SARS-CoV-2 according to claim 9, wherein the SARS-CoV-2 gastrointestinal inflammation in step (1) The construction method of the animal model includes the following steps:
    S1、小鼠禁食16~24h后进行麻醉;S1. Mice were anesthetized after fasting for 16-24 hours;
    S2、从小鼠肛门注入乙酸,倒立小鼠,并对小鼠胃肠道进行灌洗;S2. Inject acetic acid from the anus of the mouse, turn the mouse upside down, and lavage the gastrointestinal tract of the mouse;
    S3、一段时间后小鼠腹腔注射SARS-CoV-2 Spike蛋白;S3. After a period of time, mice were intraperitoneally injected with SARS-CoV-2 Spike protein;
    S4、数小时后即成功得到SARS-CoV-2胃肠炎症动物模型。S4. The animal model of SARS-CoV-2 gastrointestinal inflammation was successfully obtained after a few hours.
  11. 根据权利要求10所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,所述药剂影响VEGF介导的胃肠道血管通透性增加通过以下步骤确定:The method for screening or evaluating an agent that inhibits SARS-CoV-2-induced vascular barrier damage in gastrointestinal tissue according to claim 10, wherein the agent affects VEGF-mediated gastrointestinal vascular permeability Increased sexuality is determined by the following steps:
    (1)小鼠禁食16~24h后进行麻醉;(1) Mice were anesthetized after fasting for 16-24 hours;
    (2)从小鼠肛门注入乙酸,倒立小鼠,并对小鼠胃肠道进行灌洗;(2) Inject acetic acid from the anus of the mouse, turn the mouse upside down, and lavage the gastrointestinal tract of the mouse;
    (3)小鼠正常喂食,16~24h后,将候选药剂通过腹腔分别注射到小鼠体内,得到候选药剂组小鼠;将安慰剂通过腹腔分别注射到小鼠体内,得到安慰剂组小鼠;(3) The mice were fed normally. After 16-24 hours, the candidate drugs were injected into the mice through the abdominal cavity respectively to obtain the mice in the candidate drug group; the placebo was injected into the mice through the abdominal cavity respectively to obtain the mice in the placebo group. ;
    (4)1~2h后候选药剂组和安慰剂组小鼠腹腔分别注射SARS-CoV-2 Spike蛋白,对照组小鼠腹腔注射等量的IgG-Fc蛋白;(4) After 1-2 hours, the mice in the candidate agent group and the placebo group were intraperitoneally injected with SARS-CoV-2 Spike protein respectively, and the mice in the control group were intraperitoneally injected with the same amount of IgG-Fc protein;
    (5)1~2h后,再次对候选药剂组小鼠腹腔注射候选药剂,对安慰剂组小鼠腹腔注射安慰剂;(5) After 1-2 hours, the candidate drug was injected intraperitoneally to the mice in the candidate drug group again, and the placebo was intraperitoneally injected to the mice in the placebo group;
    (6)分别取对照组小鼠、候选药剂组小鼠和安慰剂组小鼠胃肠道组织,其中,安慰剂组小鼠胃肠组织与对照组小鼠相比,能够检测到更高的VEGF表达量,而候选药剂组小鼠胃肠组织与安慰剂组小鼠相比,能够检测到更低的VEGF表达量,表明所述候选药剂能抑制VEGF介导的胃肠道血管通透性增加。(6) The gastrointestinal tissues of the mice in the control group, the mice in the candidate drug group and the mice in the placebo group were taken respectively. Compared with the mice in the control group, the gastrointestinal tissues of the mice in the placebo group were able to detect higher Compared with the mice in the placebo group, the gastrointestinal tissue of the candidate drug group could detect a lower VEGF expression level, indicating that the candidate drug can inhibit the VEGF-mediated gastrointestinal vascular permeability Increase.
  12. 根据权利要求11所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,步骤(2)乙酸的浓度为1~2% v/v,体积为500μL~1000μL。The method for screening or evaluating agents for inhibiting vascular barrier damage in gastrointestinal tract tissue caused by SARS-CoV-2 according to claim 11, wherein the concentration of acetic acid in step (2) is 1-2% v/ v, the volume is 500 μL to 1000 μL.
  13. 根据权利要求12所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,步骤(4)所述SARS-CoV-2 Spike蛋白的用量为5.5nM/kg小鼠体重。The method for screening or evaluating a drug that inhibits vascular barrier damage in gastrointestinal tract tissue caused by SARS-CoV-2 according to claim 12, wherein the step (4) of the SARS-CoV-2 Spike protein The dosage was 5.5nM/kg mouse body weight.
  14. 根据权利要求6至13任一项所述的筛选或评估抑制SARS-CoV-2引起的胃肠道组织中血管屏障受损的药剂的方法,其特征在于,所述胃肠道组织为十二指肠。The method for screening or evaluating an agent that inhibits damage to the vascular barrier in gastrointestinal tract tissue caused by SARS-CoV-2 according to any one of claims 6 to 13, wherein the gastrointestinal tract tissue is twelve the colon.
  15. 治疗或预防SARS-CoV-2引起的胃肠道症状的方法,其特征在于,包括以下步骤:A method for treating or preventing gastrointestinal symptoms caused by SARS-CoV-2, characterized by comprising the following steps:
    (1)鉴定患有所述SARS-CoV-2引起的胃肠道症状的对象;(1) Identifying subjects with gastrointestinal symptoms caused by said SARS-CoV-2;
    (2)给对象胃肠道施用与ACE2和/或ERK和/或VEGF结合的排斥性导向信号。(2) administering to the gastrointestinal tract of a subject a repulsive targeting signal that binds to ACE2 and/or ERK and/or VEGF.
  16. 根据权利要求15所述的治疗或预防SARS-CoV-2引起的胃肠道症状的方法,其特征在于,所述排斥性导向信号包括ERK抑制剂和/或VEGF拮抗剂,所述ERK抑制剂是指能与ERK结合并阻断ERK生物活性的物质,所述VEGF拮抗剂是指能与VEGF结合并阻断VEGF生物活性的物质。The method for treating or preventing gastrointestinal symptoms caused by SARS-CoV-2 according to claim 15, wherein the rejection-directed signal comprises an ERK inhibitor and/or a VEGF antagonist, and the ERK inhibitor It refers to a substance that can bind to ERK and block the biological activity of ERK, and the VEGF antagonist refers to a substance that can bind to VEGF and block the biological activity of VEGF.
  17. 根据权利要求16所述的治疗或预防SARS-CoV-2引起的胃肠道症状的方法,其特征在于,所述ERK抑制剂包括但不限于SCH772984、GDC-0994、Ulixertinib、KO-947、LY3214996、MK-8353、CC-90003、LTT462、FR180204、VTX-11e、BL-EI-001。The method for treating or preventing gastrointestinal symptoms caused by SARS-CoV-2 according to claim 16, wherein the ERK inhibitors include but are not limited to SCH772984, GDC-0994, Ulixertinib, KO-947, LY3214996 , MK-8353, CC-90003, LTT462, FR180204, VTX-11e, BL-EI-001.
  18. 根据权利要求16所述的治疗或预防SARS-CoV-2引起的胃肠道症状的方法,其特征在于,所述VEGF拮抗剂包括但不限于贝伐珠单抗、雷莫芦单抗、雷珠单抗、阿柏西普、康柏西普。The method for treating or preventing gastrointestinal symptoms caused by SARS-CoV-2 according to claim 16, wherein the VEGF antagonists include but are not limited to bevacizumab, ramucirumab, Zizumab, Aflibercept, Conbercept.
  19. 用于治疗或预防或缓解SARS-CoV-2引起的胃肠道症状的药物,其特征在于,所述药物具有以下至少一种功能:A drug for treating or preventing or alleviating gastrointestinal symptoms caused by SARS-CoV-2, characterized in that the drug has at least one of the following functions:
    (1)抑制肠道上皮细胞VEGF的表达;(1) Inhibit the expression of VEGF in intestinal epithelial cells;
    (2)抑制肠道上皮细胞ERK的表达;(2) Inhibit the expression of ERK in intestinal epithelial cells;
    (3)挽救SARS-CoV-2 Spike蛋白引起的血管内皮VE-cad的表达抑制或VE-cad磷酸化水平上调。(3) Rescue SARS-CoV-2 Spike protein-induced inhibition of endothelial VE-cad expression or up-regulation of VE-cad phosphorylation.
PCT/CN2021/075675 2020-12-11 2021-02-06 Mechanism based on gastrointestinal symptoms caused by sars-cov-2 and application therefor WO2022121110A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011449038.9A CN112603581A (en) 2020-12-11 2020-12-11 Method for constructing SARS-CoV-2 gastrointestinal inflammation animal model
CN202011449038.9 2020-12-11

Publications (1)

Publication Number Publication Date
WO2022121110A1 true WO2022121110A1 (en) 2022-06-16

Family

ID=75232983

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/075675 WO2022121110A1 (en) 2020-12-11 2021-02-06 Mechanism based on gastrointestinal symptoms caused by sars-cov-2 and application therefor

Country Status (2)

Country Link
CN (2) CN112603581A (en)
WO (1) WO2022121110A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116369249A (en) * 2023-01-13 2023-07-04 四川轻化工大学 Construction method of zebra fish enteritis model

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101069747A (en) * 2007-04-28 2007-11-14 浙江大学 Method for setting acetic-acid gastric ulcer disease recurrence model
CN101600451A (en) * 2006-12-11 2009-12-09 犹他大学研究基金会 Compositions and methods for treating pathological angiogenesis and vascular permeability
CN111818915A (en) * 2018-01-31 2020-10-23 德西费拉制药有限责任公司 Combination therapy for treating gastrointestinal stromal tumors

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008044599A1 (en) * 2006-10-06 2008-04-17 Proteus Science Co., Ltd. Polypeptide having activity of targeting intestine having inflammation, and use thereof
CN107950472A (en) * 2017-11-27 2018-04-24 大理大学 A kind of construction method of Animal Model of Ulcerative Colitis and application

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101600451A (en) * 2006-12-11 2009-12-09 犹他大学研究基金会 Compositions and methods for treating pathological angiogenesis and vascular permeability
CN101069747A (en) * 2007-04-28 2007-11-14 浙江大学 Method for setting acetic-acid gastric ulcer disease recurrence model
CN111818915A (en) * 2018-01-31 2020-10-23 德西费拉制药有限责任公司 Combination therapy for treating gastrointestinal stromal tumors

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BARKER HARLAN, PARKKILA SEPPO: "Bioinformatic characterization of angiotensin-converting enzyme 2, the entry receptor for SARS-CoV-2", PLOS ONE, vol. 15, no. 10, 28 October 2020 (2020-10-28), pages 1 - 29, XP055941508, DOI: 10.1371/journal.pone.0240647 *
GHASEMNEJAD-BERENJI MORTEZA, PASHAPOUR SARVIN: "SARS-CoV-2 and the Possible Role of Raf/MEK/ERK Pathway in Viral Survival: Is This a Potential Therapeutic Strategy for COVID-19?", PHARMACOLOGY, vol. 106, no. 1-2, 1 January 2021 (2021-01-01), CH , pages 119 - 122, XP055941823, ISSN: 0031-7012, DOI: 10.1159/000511280 *
LIN LU, JIANG XIAYANG, ZHANG ZHENLING, HUANG SIWEN, ZHANG ZHENYI, FANG ZHAOXIONG, GU ZHIQIANG, GAO LIANGQING, SHI HONGGANG, MAI LE: "Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection", GUT MICROBIOTA, vol. 69, no. 6, 1 June 2020 (2020-06-01), UK , pages 997 - 1001, XP055941502, ISSN: 0017-5749, DOI: 10.1136/gutjnl-2020-321013 *
XIAO, GAOCHUN ET AL.: "Role of Pi3k/Akt and Mek/Erk Signaling Pathway in Tumor Vascular Endothelial Cell Migration", CHONGQING MEDICINE, vol. 44, no. 11, 30 April 2015 (2015-04-30), pages 1452 - 1456, XP055941827 *
YIN XI-XI, ZHENG XIANG-RONG, PENG WANG, WU MAO-LAN, MAO XIAO-YUAN: "Vascular Endothelial Growth Factor (VEGF) as a Vital Target for Brain Inflammation during the COVID-19 Outbreak", ACS CHEMICAL NEUROSCIENCE, vol. 11, no. 12, 17 June 2020 (2020-06-17), US , pages 1704 - 1705, XP055941475, ISSN: 1948-7193, DOI: 10.1021/acschemneuro.0c00294 *
ZHENG, QI ET AL.: "Correlation between Serum Vascular Endothelial Cadherin and Septic Shock", CHINESE JOURNAL OF NOSOCOMIOLOGY, vol. 30, no. 5, 10 March 2020 (2020-03-10), pages 667 - 670, XP055941830 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116369249A (en) * 2023-01-13 2023-07-04 四川轻化工大学 Construction method of zebra fish enteritis model
CN116369249B (en) * 2023-01-13 2024-05-17 四川轻化工大学 Method for constructing a zebrafish enteritis model

Also Published As

Publication number Publication date
CN112603581A (en) 2021-04-06
CN113440303A (en) 2021-09-28

Similar Documents

Publication Publication Date Title
Sethi et al. Semaphorin 3B–associated membranous nephropathy is a distinct type of disease predominantly present in pediatric patients
Ibrahim et al. PIK3R3 regulates ZO-1 expression through the NF-kB pathway in inflammatory bowel disease
Nan et al. Tumor-stroma TGF-β1-THBS2 feedback circuit drives pancreatic ductal adenocarcinoma progression via integrin αvβ3/CD36-mediated activation of the MAPK pathway
Ning et al. Hepatocyte nuclear factor 4α suppresses the development of hepatocellular carcinoma
Kato et al. MUC1 contributes to goblet cell metaplasia and MUC5AC expression in response to cigarette smoke in vivo
Chen et al. H. pylori‐induced NF‐κB‐PIEZO1‐YAP1‐CTGF axis drives gastric cancer progression and cancer‐associated fibroblast‐mediated tumour microenvironment remodelling
Di Malta et al. TFEB and TFE3 drive kidney cystogenesis and tumorigenesis
Guo et al. Exosomal CagA from Helicobacter pylori aggravates intestinal epithelium barrier dysfunction in chronic colitis by facilitating Claudin-2 expression
Royal et al. A modified cholera toxin B subunit containing an ER retention motif enhances colon epithelial repair via an unfolded protein response
Ling et al. Irf7 regulates the expression of Srg3 and ferroptosis axis aggravated sepsis-induced acute lung injury
Li et al. Inhibition of endothelial Slit2/Robo1 signaling by thalidomide restrains angiogenesis by blocking the PI3K/Akt pathway
Zhang et al. C-MYC-induced upregulation of LINC01503 promotes progression of non-small cell lung cancer.
Liu et al. Depletion of VPS35 attenuates metastasis of hepatocellular carcinoma by restraining the Wnt/PCP signaling pathway
Yang et al. Genomic DNA activates the AIM2 inflammasome and STING pathways to induce inflammation in lacrimal gland myoepithelial cells
Chen et al. LncRNA MSC-AS1 regulates SNIP1 SUMOylation-mediated EMT by binding to SENP1 to promote intestinal fibrosis in Crohn's disease
WO2022121110A1 (en) Mechanism based on gastrointestinal symptoms caused by sars-cov-2 and application therefor
Zhang et al. Coronin 6 promotes hepatocellular carcinoma progression by enhancing canonical Wnt/beta-catenin signaling pathway
Layoun et al. Pirfenidone mitigates TGF-β1-mediated fibrosis in an idiopathic inflammatory myositis-associated interstitial lung disease model
Li et al. GATA6 promotes fibrotic repair of tracheal injury through NLRP3 inflammasome-mediated epithelial pyroptosis
IL314583A (en) Novel use of melanocortin-1 receptor agonist
JP6341859B2 (en) Cancer markers and their uses
Lin et al. Unveiling the role of Pafah1b3 in liver fibrosis: A novel mechanism revealed
Du et al. Blocking GATA6 Alleviates Pyroptosis and Inhibits Abdominal Wall Endometriosis Lesion Growth Through Inactivating the PI3K/AKT Pathway
Chen et al. Denosumab induces neoplastic stromal cell apoptosis via p62 downregulation dependent on autophagy pathway in giant cell tumour of Bone
Moore The Effects of Chromatin Remodeling and Pseudokinase Activity on Liver Pathophysiology

Legal Events

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

Ref document number: 21901827

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21901827

Country of ref document: EP

Kind code of ref document: A1