Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtre
1.
J Med Virol ; 95(6): e28845, 2023 06.
Article Dans Anglais | MEDLINE | ID: covidwho-20241588

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological pathogen of coronavirus disease 2019 (COVID-19), a highly contagious disease, spreading quickly and threatening global public health. The symptoms of COVID-19 vary from mild reactions to severe respiratory distress or even fatal outcomes probably due to the different status of host immunity against the virus. Here in the study, we unveiled plasma proteomic signatures and transcriptional patterns of peripheral blood mononuclear cells (PBMCs) using blood samples of 10 COVID-19 patients with different severity. Through systemic analysis, α-defensin-1 (DEFA1) was identified to be elevated in both plasma and PBMCs, and correlated with disease severity and stages. In vitro study demonstrated that DEFA1 was secreted from immunocytes and suppressed SARS-CoV-2 infection of both original and mutated strains with dose dependency. By using sequencing data, we discovered that DEFA1 was activated in monocytes through NF-κB signaling pathway after infection, and secreted into circulation to perturb SARS-CoV-2 infection by interfering protein kinase C expression. It worked mainly during virus replication instead of entry in host cells. Together, the anti-SARS-CoV-2 mechanism of DEFA1 has unveiled a corner of how innate immunity is against SARS-CoV-2 and explored its clinical potential in disease prognosis and therapeutic intervention.


Sujets)
COVID-19 , Défensines-alpha , Humains , SARS-CoV-2 , Défensines-alpha/génétique , Monocytes , Agranulocytes , , Protéomique
2.
Int J Biol Macromol ; 229: 413-421, 2023 Feb 28.
Article Dans Anglais | MEDLINE | ID: covidwho-2165363

Résumé

Fucoidan is a highly sulfated polysaccharide with a wide range of bioactivities, including anti-pathogenic activity. However, the relationship between structure and activity of fucoidan in inhibiting pathogen infections remains unclear. Here, different-molecular-weight fucoidans were prepared by photocatalytic degradation followed by membrane ultrafiltration, and their chemical structures and anti-pathogenic microbiota activity were compared. Results showed that photocatalytic degradation could effectively degrade fucoidan while its structure block and sulfate groups were not destroyed obviously. Fucoidan (90.8 kDa) of 5 mg/mL could inhibit the growth of S. aureus, S. typhimurium and E. coli, but its degradation products, Dfuc1 (19.2 kDa) and Dfuc2 (5.5 kDa), demonstrated lower inhibitory effect. In addition, compared to Dfuc1 and Dfuc2, fucoidan showed stronger capability to prevent the adhesion of S. aureus, L. monocytogenes, V. parahaemolyticus and S. typhimurium to HT-29 cells. Moreover, the inhibitory effect against SARS-CoV-2 and the binding activity to S protein were also positively correlated to molecular weight. These results indicate that natural fucoidan with higher molecular weight are more effective to inhibit these pathogenic bacteria and SARS-CoV-2, providing a better understanding of the relationship between structure and activity of fucoidan against pathogenic microbiota.


Sujets)
COVID-19 , Laminaria , Humains , Laminaria/composition chimique , SARS-CoV-2 , Masse moléculaire , Escherichia coli , Staphylococcus aureus , Polyosides/composition chimique , Bactéries , Sulfates/métabolisme
3.
mSphere ; 7(4): e0021122, 2022 08 31.
Article Dans Anglais | MEDLINE | ID: covidwho-1938010

Résumé

The innate interferon (IFN) response constitutes the first line of host defense against viral infections. It has been shown that IFN-I/III treatment could effectively contain severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication in vitro. However, how SARS-CoV-2 survives through the innate antiviral mechanism remains to be explored. Our study uncovered that human angiotensin-converting enzyme 2 (ACE2), identified as a primary receptor for SARS-CoV-2 entry, can disturb the IFN-I signaling pathway during SARS-CoV-2 infection in human lung cells. We identified that ACE2 was significantly upregulated by SARS-CoV-2 and Sendai virus (SeV) infection, and exogenous expression of ACE2 suppressed IFN-I production in a dose-dependent manner. Mechanistically, ACE2 disrupted poly (I:C)-mediated inhibition of SARS-CoV2 replication by antagonizing IFN-I production by blocking IRF3 phosphorylation and nuclear translocation. Moreover, ACE2 quenched the IFN-mediated antiviral immune response by degrading endogenous STAT2 protein, inhibiting STAT2 phosphorylation and nuclear translocation. Interestingly, IFN-inducible short ACE2 (dACE2 or MIRb-ACE2) can also be induced by virus infection and inhibits the IFN signaling. Thus, our findings provide mechanistic insight into the distinctive role of ACE2 in promoting SARS-CoV-2 infection and enlighten us that the development of interventional strategies might be further optimized to interrupt ACE2-mediated suppression of IFN-I and its signaling pathway. IMPORTANCE Efficient antiviral immune responses against SARS-CoV-2 infection play a key role in controlling the coronavirus diseases 2019 (COVID-19) caused by this virus. Although SARS-CoV-2 has developed strategies to counteract the IFN-I signaling through the virus-derived proteins, our knowledge of how SARS-CoV-2 survives through the innate antiviral mechanism remains poor. We herein discovered the distinctive role of ACE2 as a restraining factor of the IFN-I signaling in facilitating SARS-CoV-2 infection in human lung cells. Both full-length ACE2 and truncated dACE2 can antagonize IFN-mediated antiviral response. These findings are key to understanding the counteraction between SARS-CoV-2 pathogenicity and the host antiviral defenses.


Sujets)
Angiotensin-converting enzyme 2 , COVID-19 , Interféron de type I , Transduction du signal , Angiotensin-converting enzyme 2/métabolisme , COVID-19/immunologie , Humains , Interféron de type I/immunologie , ARN viral , SARS-CoV-2
4.
Front Immunol ; 13: 851642, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-1933645

Résumé

The rapid evolution of highly infectious pathogens is a major threat to global public health. In the front line of defense against bacteria, fungi, and viruses, antimicrobial peptides (AMPs) are naturally produced by all living organisms and offer new possibilities for next-generation antibiotic development. However, the low yields and difficulties in the extraction and purification of AMPs have hindered their industry and scientific research applications. To overcome these barriers, we enabled high expression of bomidin, a commercial recombinant AMP based upon bovine myeloid antimicrobial peptide-27. This novel AMP, which can be expressed in Escherichia coli by adding methionine to the bomidin sequence, can be produced in bulk and is more biologically active than chemically synthesized AMPs. We verified the function of bomidin against a variety of bacteria and enveloped viruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), herpes simplex virus (HSV), dengue virus (DENV), and chikungunya virus (CHIKV). Furthermore, based on the molecular modeling of bomidin and membrane lipids, we elucidated the possible mechanism by which bomidin disrupts bacterial and viral membranes. Thus, we obtained a novel AMP with an optimized, efficient heterologous expression system for potential therapeutic application against a wide range of life-threatening pathogens.


Sujets)
COVID-19 , Virus , AMP , Animaux , Peptides antimicrobiens , Antiviraux/pharmacologie , Bovins , SARS-CoV-2
5.
J Med Virol ; 94(10): 4809-4819, 2022 10.
Article Dans Anglais | MEDLINE | ID: covidwho-1898903

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the seventh member of the coronavirus family that can infect humans. Recently, more contagious and pathogenic variants of SARS-CoV-2 have been continuously emerging. Clinical candidates with high efficacy and ready availability are still in urgent need. To identify potent anti-SARS-CoV-2 repurposing drugs, we evaluated the antiviral efficacy of 18 selective estrogen receptor modulators (SERMs) against SARS-CoV-2 infection. Six SERMs exhibited excellent anti-SARS-CoV-2 effects in Vero E6 cells and three human cell lines. Clomifene citrate, tamoxifen, toremifene citrate, and bazedoxifene acetate reduced the weight loss of hamsters challenged with SARS-CoV-2, and reduced hamster pulmonary viral load and interleukin-6 expression when assayed at 4 days postinfection. In particular, bazedoxifene acetate was identified to act on the penetration stage of the postattachment step via altering cholesterol distribution and endosome acidification. And, bazedoxifene acetate inhibited pseudoviruses infection of original SARS-CoV-2, Delta variant, Omicron variant, and SARS-CoV. These results offer critical information supporting bazedoxifene acetate as a promising agent against coronaviruses.


Sujets)
, SARS-CoV-2 , Antiviraux/pharmacologie , Humains , Indoles , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie
6.
Eur J Immunol ; 52(8): 1308-1320, 2022 08.
Article Dans Anglais | MEDLINE | ID: covidwho-1825936

Résumé

Human nasal mucosa is susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and serves as a reservoir for viral replication before spreading to other organs (e.g. the lung and brain) and transmission to other individuals. Chronic rhinosinusitis (CRS) is a common respiratory tract disease and there is evidence suggesting that susceptibility to SARS-CoV-2 infection differs between the two known subtypes, eosinophilic CRS and non-ECRS (NECRS). However, the mechanism of SARS-CoV-2 infection in the human nasal mucosa and its association with CRS has not been experimentally validated. In this study, we investigated whether the human nasal mucosa is susceptible to SARS-CoV-2 infection and how different endotypes of CRS impact on viral infection and progression. Primary human nasal mucosa tissue culture revealed highly efficient SARS-CoV-2 viral infection and production, with particularly high susceptibility in the NECRS group. The gene expression differences suggested that human nasal mucosa is highly susceptible to SARS-CoV-2 infection, presumably due to an increase in ACE2-expressing cells and a deficiency in antiviral immune response, especially for NECRS. Importantly, patients with NECRS may be at a particularly high risk of viral infection and transmission, and therefore, close monitoring should be considered.


Sujets)
COVID-19 , Rhinite , Sinusite , Maladie chronique , Humains , Muqueuse nasale/métabolisme , Rhinite/complications , Rhinite/métabolisme , SARS-CoV-2 , Sinusite/complications , Sinusite/métabolisme
7.
J Virol ; 96(4): e0157821, 2022 02 23.
Article Dans Anglais | MEDLINE | ID: covidwho-1759290

Résumé

The ongoing SARS-CoV-2 pandemic poses a severe global threat to public health, as do influenza viruses and other coronaviruses. Here, we present chimpanzee adenovirus 68 (AdC68)-based vaccines designed to universally target coronaviruses and influenza. Our design is centered on an immunogen generated by fusing the SARS-CoV-2 receptor-binding domain (RBD) to the conserved stalk of H7N9 hemagglutinin (HA). Remarkably, the constructed vaccine effectively induced both SARS-CoV-2-targeting antibodies and anti-influenza antibodies in mice, consequently affording protection from lethal SARS-CoV-2 and H7N9 challenges as well as effective H3N2 control. We propose our AdC68-vectored coronavirus-influenza vaccine as a universal approach toward curbing respiratory virus-causing pandemics. IMPORTANCE The COVID-19 pandemic exemplifies the severe public health threats of respiratory virus infection and influenza A viruses. The currently envisioned strategy for the prevention of respiratory virus-causing diseases requires the comprehensive administration of vaccines tailored for individual viruses. Here, we present an alternative strategy by designing chimpanzee adenovirus 68-based vaccines which target both the SARS-CoV-2 receptor-binding-domain and the conserved stalk of influenza hemagglutinin. When tested in mice, this strategy attained potent neutralizing antibodies against wild-type SARS-CoV-2 and its emerging variants, enabling an effective protection against lethal SARS-CoV-2 challenge. Notably, it also provided complete protection from lethal H7N9 challenge and efficient control of H3N2-induced morbidity. Our study opens a new avenue to universally curb respiratory virus infection by vaccination.


Sujets)
COVID-19/prévention et contrôle , Vaccin ChAdOx1 nCoV-19 , Sous-type H7N9 du virus de la grippe A/immunologie , Vaccins antigrippaux , Infections à Orthomyxoviridae/prévention et contrôle , SARS-CoV-2/immunologie , Animaux , COVID-19/épidémiologie , COVID-19/génétique , COVID-19/immunologie , Vaccin ChAdOx1 nCoV-19/génétique , Vaccin ChAdOx1 nCoV-19/immunologie , Vaccin ChAdOx1 nCoV-19/pharmacologie , Femelle , Cellules HEK293 , Humains , Sous-type H7N9 du virus de la grippe A/génétique , Vaccins antigrippaux/génétique , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/pharmacologie , Souris , Souris de lignée BALB C , Souris de lignée ICR , Souris transgéniques , Infections à Orthomyxoviridae/épidémiologie , Infections à Orthomyxoviridae/génétique , Infections à Orthomyxoviridae/immunologie , Pandémies , SARS-CoV-2/génétique
8.
Sci China Life Sci ; 65(6): 1181-1197, 2022 06.
Article Dans Anglais | MEDLINE | ID: covidwho-1596898

Résumé

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global crisis. Clinical candidates with high efficacy, ready availability, and that do not develop resistance are in urgent need. Despite that screening to repurpose clinically approved drugs has provided a variety of hits shown to be effective against SARS-CoV-2 infection in cell culture, there are few confirmed antiviral candidates in vivo. In this study, 94 compounds showing high antiviral activity against SARS-CoV-2 in Vero E6 cells were identified from 2,580 FDA-approved small-molecule drugs. Among them, 24 compounds with low cytotoxicity were selected, and of these, 17 compounds also effectively suppressed SARS-CoV-2 infection in HeLa cells transduced with human ACE2. Six compounds disturb multiple processes of the SARS-CoV-2 life cycle. Their prophylactic efficacies were determined in vivo using Syrian hamsters challenged with SARS-CoV-2 infection. Seven compounds reduced weight loss and promoted weight regain of hamsters infected not only with the original strain but also the D614G variant. Except for cisatracurium, six compounds reduced hamster pulmonary viral load, and IL-6 and TNF-α mRNA when assayed at 4 d postinfection. In particular, sertraline, salinomycin, and gilteritinib showed similar protective effects as remdesivir in vivo and did not induce antiviral drug resistance after 10 serial passages of SARS-CoV-2 in vitro, suggesting promising application for COVID-19 treatment.


Sujets)
, SARS-CoV-2 , Animaux , Antiviraux/usage thérapeutique , Cricetinae , Cellules HeLa , Humains
9.
Carbohydr Polym ; 280: 119006, 2022 Mar 15.
Article Dans Anglais | MEDLINE | ID: covidwho-1588175

Résumé

Caulerpa lentillifera (Bryopsidophyceae, Chlorophyta) is an edible seaweed attracting great attention for its expansion of farming scale and increasing consumption in these years. In the present study, a sulfated polysaccharide (CLSP-2) was isolated and separated from C. lentillifera, and its chemical structure was elucidated by a series of chemical and spectroscopic methods. Among these methods, mild acid hydrolysis and photocatalytic degradation were applied to release mono- and oligo-saccharide fragments which were further identified by HPLC-MSn analysis, affording the information of the sugar sequences and the sulfate substitution in CLSP-2. Results indicated that the backbone of CLSP-2 was constructed of →6)-ß-Manp-(1→ with sulfated branches at C2, which were comprised of prevalent →3)-ß-Galp4S-(1→, →3)-ß-Galp2,4S-(1→, and minor Xyl. In addition, the virus neutralization assay revealed that CLSP-2 could effectively protect HeLa cells against SARS-CoV-2 infection with an IC50 of 48.48 µg/mL. Hence, the present study suggests CLSP-2 as a promising agent against SARS-CoV-2.


Sujets)
COVID-19/virologie , Caulerpa/composition chimique , Polyosides/composition chimique , Polyosides/pharmacologie , Antiviraux/composition chimique , Antiviraux/pharmacologie , Chromatographie en phase liquide à haute performance/méthodes , Cellules HeLa , Humains , Hydrolyse , Spectroscopie par résonance magnétique/méthodes , Spectrométrie de masse/méthodes , Masse moléculaire , Polyosides/analyse , SARS-CoV-2 , Algue marine/composition chimique , Spectroscopie infrarouge à transformée de Fourier/méthodes , Sulfates/composition chimique
10.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Article Dans Anglais | MEDLINE | ID: covidwho-1555255

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), binds to host receptor angiotensin-converting enzyme 2 (ACE2) through its spike (S) glycoprotein, which mediates membrane fusion and viral entry. However, the expression of ACE2 is extremely low in a variety of human tissues, especially in the airways. Thus, other coreceptors and/or cofactors on the surface of host cells may contribute to SARS-CoV-2 infection. Here, we identified nonmuscle myosin heavy chain IIA (MYH9) as an important host factor for SARS-CoV-2 infection of human pulmonary cells by using APEX2 proximity-labeling techniques. Genetic ablation of MYH9 significantly reduced SARS-CoV-2 pseudovirus infection in wild type (WT) A549 and Calu-3 cells, and overexpression of MYH9 enhanced the pseudovirus infection in WT A549 and H1299 cells. MYH9 was colocalized with the SARS-CoV-2 S and directly interacted with SARS-CoV-2 S through the S2 subunit and S1-NTD (N-terminal domain) by its C-terminal domain (designated as PRA). Further experiments suggested that endosomal or myosin inhibitors effectively block the viral entry of SARS-CoV-2 into PRA-A549 cells, while transmembrane protease serine 2 (TMPRSS2) and cathepsin B and L (CatB/L) inhibitors do not, indicating that MYH9 promotes SARS-CoV-2 endocytosis and bypasses TMPRSS2 and CatB/L pathway. Finally, we demonstrated that loss of MYH9 reduces authentic SARS-CoV-2 infection in Calu-3, ACE2-A549, and ACE2-H1299 cells. Together, our results suggest that MYH9 is a candidate host factor for SARS-CoV-2, which mediates the virus entering host cells by endocytosis in an ACE2-dependent manner, and may serve as a potential target for future clinical intervention strategies.


Sujets)
COVID-19/virologie , Chaînes lourdes de myosine/métabolisme , SARS-CoV-2/physiologie , Angiotensin-converting enzyme 2/métabolisme , Lignée cellulaire , Membrane cellulaire/métabolisme , Humains , Poumon/métabolisme , Coronavirus du syndrome respiratoire du Moyen-Orient/physiologie , Chaînes lourdes de myosine/composition chimique , Chaînes lourdes de myosine/génétique , Liaison aux protéines , Domaines protéiques , Virus du SRAS/physiologie , Glycoprotéine de spicule des coronavirus/métabolisme , Pénétration virale
11.
J Med Chem ; 64(23): 17486-17495, 2021 12 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1531976

Résumé

The pandemic of acute respiratory disease in 2019 caused by highly pathogenic and infectious SARS-CoV-2 has seriously endangered human public safety. The 6-HB (HR1-HR2 complex) formation occurring in the process of spike protein-mediated membrane fusion could serve as a conserved and potential target for the design of fusion inhibitors. Based on the HR2 domain of 6-HB, we designed and synthesized 32 stapled peptides using an all-hydrocarbon peptide stapling strategy. Owing to the improved proteolytic stability and higher helical contents, the optimized stapled peptides termed SCH2-1-20 and SCH2-1-27 showed better inhibitory activities against pseudo and authentic SARS-CoV-2 compared to the linear counterpart. Of note, SCH2-1-20 and SCH2-1-27 were proved to interfere with S protein-mediated membrane fusion. Structural modeling indicated similar binding modes between SCH2-1-20 and the linear peptide. These optimized stapled peptides could serve as potent fusion inhibitors in treating and preventing SARS-CoV-2, and the corresponding SAR could facilitate further optimization.


Sujets)
Glycoprotéine de spicule des coronavirus , Fusion membranaire , Pandémies , Liaison aux protéines
12.
Biol Direct ; 16(1): 20, 2021 10 21.
Article Dans Anglais | MEDLINE | ID: covidwho-1477450

Résumé

SARS-CoV-2 infection could cause severe acute respiratory syndrome, largely attributed to dysregulated immune activation and extensive lung tissue damage. However, the underlying mechanisms are not fully understood. Here, we reported that viral infection could induce syncytia formation within cells expressing ACE2 and the SARS-CoV-2 spike protein, leading to the production of micronuclei with an average rate of about 4 per syncytium (> 93%). Remarkably, these micronuclei were manifested with a high level of activation of both DNA damage response and cGAS-STING signaling, as indicated by micronucleus translocation of γH2Ax and cGAS, and upregulation of their respective downstream target genes. Since activation of these signaling pathways were known to be associated with cellular catastrophe and aberrant immune activation, these findings help explain the pathological effects of SARS-CoV-2 infection at cellular and molecular levels, and provide novel potential targets for COVID-19 therapy.


Sujets)
COVID-19/génétique , COVID-19/métabolisme , Altération de l'ADN , Protéines membranaires/métabolisme , Nucleotidyltransferases/métabolisme , SARS-CoV-2/pathogénicité , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , COVID-19/virologie , Cellules géantes/métabolisme , Cellules géantes/virologie , Cellules HeLa , Humains , Tests de micronucleus , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , Transduction du signal , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme
13.
Cell Res ; 31(12): 1230-1243, 2021 12.
Article Dans Anglais | MEDLINE | ID: covidwho-1475291

Résumé

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the ongoing global pandemic that poses substantial challenges to public health worldwide. A subset of COVID-19 patients experience systemic inflammatory response, known as cytokine storm, which may lead to death. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is an important mediator of inflammation and cell death. Here, we examined the interaction of RIPK1-mediated innate immunity with SARS-CoV-2 infection. We found evidence of RIPK1 activation in human COVID-19 lung pathological samples, and cultured human lung organoids and ACE2 transgenic mice infected by SARS-CoV-2. Inhibition of RIPK1 using multiple small-molecule inhibitors reduced the viral load of SARS-CoV-2 in human lung organoids. Furthermore, therapeutic dosing of the RIPK1 inhibitor Nec-1s reduced mortality and lung viral load, and blocked the CNS manifestation of SARS-CoV-2 in ACE2 transgenic mice. Mechanistically, we found that the RNA-dependent RNA polymerase of SARS-CoV-2, NSP12, a highly conserved central component of coronaviral replication and transcription machinery, promoted the activation of RIPK1. Furthermore, NSP12 323L variant, encoded by the SARS-CoV-2 C14408T variant first detected in Lombardy, Italy, that carries a Pro323Leu amino acid substitution in NSP12, showed increased ability to activate RIPK1. Inhibition of RIPK1 downregulated the transcriptional induction of proinflammatory cytokines and host factors including ACE2 and EGFR that promote viral entry into cells. Our results suggest that SARS-CoV-2 may have an unexpected and unusual ability to hijack the RIPK1-mediated host defense response to promote its own propagation and that inhibition of RIPK1 may provide a therapeutic option for the treatment of COVID-19.


Sujets)
COVID-19/anatomopathologie , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , SARS-CoV-2/physiologie , Angiotensin-converting enzyme 2/génétique , Animaux , COVID-19/mortalité , COVID-19/virologie , ARN polymérase ARN-dépendante de coronavirus/génétique , ARN polymérase ARN-dépendante de coronavirus/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Humains , Imidazoles/pharmacologie , Imidazoles/usage thérapeutique , Indoles/pharmacologie , Indoles/usage thérapeutique , Poumon/anatomopathologie , Poumon/virologie , Souris , Souris transgéniques , Mutation , Receptor-Interacting Protein Serine-Threonine Kinases/antagonistes et inhibiteurs , SARS-CoV-2/isolement et purification , SARS-CoV-2/métabolisme , Taux de survie , Transcriptome/effets des médicaments et des substances chimiques , Charge virale/effets des médicaments et des substances chimiques , Pénétration virale ,
14.
Emerg Microbes Infect ; 10(1): 1555-1573, 2021 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-1324547

Résumé

To curb the pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), multiple platforms have been employed toward a safe and highly effective vaccine. Here, we develop a novel cell-based vaccine candidate, namely K562-S, by utilizing human cell K562 as a cellular carrier to display Spike (S) protein of SARS-CoV-2 on the membrane. Analogous to the traditional inactivated vaccine, K562-S cells can be propagated to a large scale by culturing and completely lose their viability after exposure to X-ray irradiation or formalin. We in turn demonstrated high immunogenicity of formalin-inactivated K562-S vaccine in both mouse and non-human primates and its protective efficacy in mice. In mice, immunization with inactivated K562-S vaccines can elicit potent neutralizing antibody (nAb) responses persisting longer than 5 months. We consequently showed in a hACE2 mouse model of SARS-CoV-2 infection that a two-shot vaccination with adjuvanted K562-S rendered greater than 3 log reduction in viral lung load and concomitant ameliorated lung pathology. Of importance, the administration of the same regimen in non-human primates was able to induce a neutralizing antibody titer averaging three-fold higher relative to human convalescent serum. These results together support the promise of K562-based, S-protein-expressing vaccines as a novel vaccination approach against SARS-CoV-2. Importantly, with a powerful capacity to carry external genes for cell-based vectors, this platform could rapidly generate two- and multiple-valent vaccines by incorporating SARS-CoV-2 mutants, SARS-CoV, or MERS-CoV.


Sujets)
Anticorps neutralisants/sang , Anticorps antiviraux/sang , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Immunogénicité des vaccins , SARS-CoV-2/immunologie , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Animaux , Animal génétiquement modifié , Vaccins contre la COVID-19/administration et posologie , Femelle , Cellules HEK293 , Humains , Cellules K562 , Macaca mulatta , Souris , Souris de lignée C57BL , Souris de lignée ICR , Primates , Organismes exempts d'organismes pathogènes spécifiques , Glycoprotéine de spicule des coronavirus/administration et posologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Vaccination/méthodes , Vaccins inactivés/administration et posologie , Vaccins inactivés/immunologie
15.
Cell Death Differ ; 28(9): 2765-2777, 2021 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1195611

Résumé

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus is highly contagious and causes lymphocytopenia, but the underlying mechanisms are poorly understood. We demonstrate here that heterotypic cell-in-cell structures with lymphocytes inside multinucleate syncytia are prevalent in the lung tissues of coronavirus disease 2019 (COVID-19) patients. These unique cellular structures are a direct result of SARS-CoV-2 infection, as the expression of the SARS-CoV-2 spike glycoprotein is sufficient to induce a rapid (~45.1 nm/s) membrane fusion to produce syncytium, which could readily internalize multiple lines of lymphocytes to form typical cell-in-cell structures, remarkably leading to the death of internalized cells. This membrane fusion is dictated by a bi-arginine motif within the polybasic S1/S2 cleavage site, which is frequently present in the surface glycoprotein of most highly contagious viruses. Moreover, candidate anti-viral drugs could efficiently inhibit spike glycoprotein processing, membrane fusion, and cell-in-cell formation. Together, we delineate a molecular and cellular rationale for SARS-CoV-2 pathogenesis and identify novel targets for COVID-19 therapy.


Sujets)
COVID-19/virologie , Cellules géantes/virologie , Lymphocytes/virologie , SARS-CoV-2/métabolisme , SARS-CoV-2/pathogénicité , Glycoprotéine de spicule des coronavirus/métabolisme , COVID-19/anatomopathologie , Lignée cellulaire , Lignée cellulaire tumorale , Cellules géantes/anatomopathologie , Cellules HEK293 , Cellules HeLa , Humains , Cellules Jurkat , Cellules K562 , Lymphocytes/anatomopathologie , Pénétration virale , Réplication virale/génétique
17.
Academic Journal of Second Military Medical University ; 41(4):365-370, 2020.
Article Dans Chinois | GIM | ID: covidwho-829836

Résumé

Objective To isolate the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from nasal/throat swabs of coronavirus disease 19 (COVID-19) patients. Methods Three nasal/throat swab samples from COVID-19 patients in Shanghai were treated with TPCK trypsin and were used to treat Vero E6 cells inoculated in 96-well plates. When most of the cells showed obvious cytopathy, the cell culture supernatants were collected. We then detected the viral nucleic acid by fluorescent quantitative real-time polymerase chain reaction, and amplified the gene fragment of the virus receptor binding domain (RBD) by reverse transcription polymerase chain reaction. After amplification and culture, the virus was used to infect the Vero E6 cells inoculated in 96-well plates. The cytopathy was observed and the virus protein was detected by immunofluorescence. Results The Vero E6 cells that cultured with two of three nasal/pharyngeal swab samples showed obvious cytopathic effect and newly synthesized viral nucleic acid was detected in the supernatants of the cell culture. The amplified RBD sequence was completely consistent with the corresponding fragment of SARS-CoV-2 isolated earlier. Virus-infected Vero E6 cells showed cytopathies rapidly and could react with the monoclonal antibody against nucleocapsid protein (N protein) and spike protein (S protein) of SARS-CoV-2, and convalescence sera of COVID-19 patients. Conclusion Two SARS-CoV-2 strains were successfully isolated from two nasal/throat swab samples of COVID-19 patients in Shanghai, which provides evidence for the mechanism research on the infection and pathogenesis of SARS-CoV-2 as well as the development of drugs and vaccines against SARS-CoV-2.

18.
Academic Journal of Second Military Medical University ; 41(4):359-364, 2020.
Article Dans Chinois | CAB Abstracts | ID: covidwho-829835

Résumé

Objective: To establish a method for preparing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudoparticles (SARS-CoV-2 pps).

19.
Chinese Journal of Zoonoses ; 36(5):341-348, 2020.
Article Dans Chinois | GIM | ID: covidwho-828697

Résumé

The first pneumonia case with unknown etiology appeared in Wuhan on December 1, 2019, and the pathogen of which was identified as a new species of coronavirus. The World Health Organization (WHO) was tentatively named the virus as 2019 novel coronavirus (2019-nCoV) on January 31, 2020. Then the caused pneumonia was named as novel coronavirus pneumonia (NCP) by National Health Commission on February 8, 2020, which was changed to coronavirus disease 2019 (COVID-19) under agreed guidelines of WHO three days later on February 11, 2020. One same day, the International Committee on Taxonomy of Viruses designated the 2019-nCoV as severe acute respiratory syndrome coronavirus 2 (2019-nCoV). WHO characterizes COVID-19 as a pandemic on March 11, 2020. After the outbreak of COVID-19, researchers at home and abroad make great efforts in doing epidemiology investigation, establishing detection methods, improving clinical diagnosis and treatment plan, exploring pathogenesis and developing anti-viral drugs and vaccines, effectively preventing the spread of the domestic epidemic and winning time for controlling the epidemic outside China. Based on the axis of time, this documentary article records the important events in research progress of COVID-19 in genomics, virus origins, epidemiology, detection, pathogenesis, clinical diagnosis and treatment.

20.
Food Funct ; 11(9): 7415-7420, 2020 Sep 23.
Article Dans Anglais | MEDLINE | ID: covidwho-786676

Résumé

Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread around the world at an unprecedented rate. In the present study, 4 marine sulfated polysaccharides were screened for their inhibitory activity against SARS-CoV-2, including sea cucumber sulfated polysaccharide (SCSP), fucoidan from brown algae, iota-carrageenan from red algae, and chondroitin sulfate C from sharks (CS). Of them, SCSP, fucoidan, and carrageenan showed significant antiviral activities at concentrations of 3.90-500 µg mL-1. SCSP exhibited the strongest inhibitory activity with IC50 of 9.10 µg mL-1. Furthermore, a test using pseudotype virus with S glycoprotein confirmed that SCSP could bind to the S glycoprotein to prevent SARS-CoV-2 host cell entry. The three antiviral polysaccharides could be employed to treat and prevent COVID-19.


Sujets)
Antiviraux/pharmacologie , Betacoronavirus/effets des médicaments et des substances chimiques , Phaeophyta/composition chimique , Polyosides/pharmacologie , Rhodophyta/composition chimique , Concombres de mer/composition chimique , Animaux , Antiviraux/composition chimique , Betacoronavirus/physiologie , COVID-19 , Infections à coronavirus/virologie , Humains , Pandémies , Pneumopathie virale/virologie , Polyosides/composition chimique , SARS-CoV-2 , Requins , Sulfates/composition chimique , Pénétration virale/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
Détails de la recherche