Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 110
Filtre
1.
Emerg Microbes Infect ; 12(1): 2207678, 2023 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-2316508

Résumé

SUMMARY: Intranasal infection of newly-weaned Syrian hamsters by SARS-CoV-2 Omicron variants can lead to brain inflammation and neuron degeneration with detectable low level of viral load and sparse expression of viral nucleoprotein.


Sujets)
COVID-19 , Encéphalite , Animaux , Cricetinae , SARS-CoV-2 , Mesocricetus , Encéphale
2.
Fundamental Research ; 2023.
Article Dans Anglais | ScienceDirect | ID: covidwho-2311541

Résumé

Combinatorial drug therapies are generally more effective than monotherapies in treating viral infections. However, it is critical for dose optimization to maximize the efficacy and minimize side effects. Although various strategies have been deviseenchmark functions is available at Github repositoryd to accelerate the optimization process, their efficiencies were limited by the high noises and suboptimal reproducibility of biological assays. With conventional methods, variances among the replications are used to evaluate the errors of the readouts alone rather than actively participating in the optimization. Herein, we present the Regression Modeling Enabled by Monte Carlo Method (ReMEMC) algorithm for rapid identification of effective combinational therapies. ReMEMC transforms the sample variations into probability distributions of the regression coefficients and predictions. In silico simulations revealed that ReMEMC outperformed conventional regression methods in benchmark problems, and demonstrated its superior robustness against experimental noises. Using COVID-19 as a model disease, ReMEMC successfully identified an optimal 3-drug combination among 10 anti-SARS-CoV-2 drug compounds within two rounds of experiments. The optimal combination showed 2-log and 3-log higher load reduction than non-optimized combinations and monotherapy, respectively. Further workflow refinement allowed identification of personalized drug combinational therapies within 5 days. The strategy may serve as an efficient and universal tool for dose combination optimization.

3.
Proc Natl Acad Sci U S A ; 120(17): e2300376120, 2023 04 25.
Article Dans Anglais | MEDLINE | ID: covidwho-2298706

Résumé

The high transmissibility of SARS-CoV-2 Omicron subvariants was generally ascribed to immune escape. It remained unclear whether the emerging variants have gradually acquired replicative fitness in human respiratory epithelial cells. We sought to evaluate the replicative fitness of BA.5 and earlier variants in physiologically active respiratory organoids. BA.5 exhibited a dramatically increased replicative capacity and infectivity than B.1.1.529 and an ancestral strain wildtype (WT) in human nasal and airway organoids. BA.5 spike pseudovirus showed a significantly higher entry efficiency than that carrying WT or B.1.1.529 spike. Notably, we observed prominent syncytium formation in BA.5-infected nasal and airway organoids, albeit elusive in WT- and B.1.1.529-infected organoids. BA.5 spike-triggered syncytium formation was verified by lentiviral overexpression of spike in nasal organoids. Moreover, BA.5 replicated modestly in alveolar organoids, with a significantly lower titer than B.1.1.529 and WT. Collectively, the higher entry efficiency and fusogenic activity of BA.5 spike potentiated viral spread through syncytium formation in the human airway epithelium, leading to enhanced replicative fitness and immune evasion, whereas the attenuated replicative capacity of BA.5 in the alveolar organoids may account for its benign clinical manifestation.


Sujets)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/génétique , COVID-19/génétique , Nez , Organoïdes , Glycoprotéine de spicule des coronavirus/génétique , Anticorps neutralisants , Anticorps antiviraux
4.
Int J Infect Dis ; 132: 40-49, 2023 Jul.
Article Dans Anglais | MEDLINE | ID: covidwho-2298611

Résumé

OBJECTIVES: We sought to identify the predictors of delayed viral clearance in patients with cancer with asymptomatic COVID-19 when the SARS-CoV-2 Omicron variants prevailed in Hong Kong. METHODS: All patients with cancer who were attending radiation therapy for head and neck malignancies or systemic anticancer therapy saved their deep throat saliva or nasopharyngeal swabs at least twice weekly for SARS-CoV-2 screening between January 1 and April 30, 2022. The multivariate analyses identified predictors of delayed viral clearance (or slow recovery), defined as >21 days for the cycle threshold values rising to ≥30 or undetectable in two consecutive samples saved within 72 hours. Three machine learning algorithms evaluated the prediction performance of the predictors. RESULTS: A total of 200 (15%) of 1309 patients tested positive for SARS-CoV-2. Age >65 years (P = 0.036), male sex (P = 0.003), high Charlson comorbidity index (P = 0.042), lung cancer (P = 0.018), immune checkpoint inhibitor (P = 0.036), and receipt of one or no dose of COVID-19 vaccine (P = 0.003) were significant predictors. The three machine learning algorithms revealed that the mean ± SD area-under-the-curve values predicting delayed viral clearance with the cut-off cycle threshold value ≥30 was 0.72 ± 0.11. CONCLUSION: We identified subgroups with delayed viral clearance that may benefit from targeted interventions.


Sujets)
COVID-19 , Tumeurs du poumon , Humains , Mâle , Sujet âgé , SARS-CoV-2 , COVID-19/diagnostic , Vaccins contre la COVID-19
5.
Nat Commun ; 14(1): 2081, 2023 04 12.
Article Dans Anglais | MEDLINE | ID: covidwho-2294153

Résumé

Current available vaccines for COVID-19 are effective in reducing severe diseases and deaths caused by SARS-CoV-2 infection but less optimal in preventing infection. Next-generation vaccines which are able to induce mucosal immunity in the upper respiratory to prevent or reduce infections caused by highly transmissible variants of SARS-CoV-2 are urgently needed. We have developed an intranasal vaccine candidate based on a live attenuated influenza virus (LAIV) with a deleted NS1 gene that encodes cell surface expression of the receptor-binding-domain (RBD) of the SARS-CoV-2 spike protein, designated DelNS1-RBD4N-DAF. Immune responses and protection against virus challenge following intranasal administration of DelNS1-RBD4N-DAF vaccines were analyzed in mice and compared with intramuscular injection of the BioNTech BNT162b2 mRNA vaccine in hamsters. DelNS1-RBD4N-DAF LAIVs induced high levels of neutralizing antibodies against various SARS-CoV-2 variants in mice and hamsters and stimulated robust T cell responses in mice. Notably, vaccination with DelNS1-RBD4N-DAF LAIVs, but not BNT162b2 mRNA, prevented replication of SARS-CoV-2 variants, including Delta and Omicron BA.2, in the respiratory tissues of animals. The DelNS1-RBD4N-DAF LAIV system warrants further evaluation in humans for the control of SARS-CoV-2 transmission and, more significantly, for creating dual function vaccines against both influenza and COVID-19 for use in annual vaccination strategies.


Sujets)
COVID-19 , Vaccins antigrippaux , Orthomyxoviridae , Animaux , Cricetinae , Humains , SARS-CoV-2/génétique , Administration par voie nasale , Vaccins contre la COVID-19 , COVID-19/prévention et contrôle , Glycoprotéine de spicule des coronavirus/génétique , Anticorps neutralisants , Vaccin BNT162 , Anticorps antiviraux
6.
EBioMedicine ; 89: 104485, 2023 Mar.
Article Dans Anglais | MEDLINE | ID: covidwho-2254674

Résumé

BACKGROUND: Obesity is a worldwide epidemic and is considered a risk factor of severe manifestation of Coronavirus Disease 2019 (COVID-19). The pathogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses to infection, re-infection, and vaccination in individuals with obesity remain incompletely understood. METHODS: Using the diet-induced obese (DIO) mouse model, we studied SARS-CoV-2 Alpha- and Omicron BA.1-induced disease manifestations and host immune responses to infection, re-infection, and COVID-19 mRNA vaccination. FINDINGS: Unlike in lean mice, Omicron BA.1 and Alpha replicated to comparable levels in the lungs of DIO mice and resulted in similar degree of tissue damages. Importantly, both T cell and B cell mediated adaptive immune responses to SARS-CoV-2 infection or COVID-19 mRNA vaccination are impaired in DIO mice, leading to higher propensity of re-infection and lower vaccine efficacy. However, despite the absence of neutralizing antibody, vaccinated DIO mice are protected from lung damage upon Omicron challenge, accompanied with significantly more IFN-α and IFN-ß production in the lung tissue. Lung RNAseq and subsequent experiments indicated that COVID-19 mRNA vaccination in DIO mice boosted antiviral innate immune response, including the expression of IFN-α, when compared to the nonvaccinated controls. INTERPRETATION: Our findings suggested that COVID-19 mRNA vaccination enhances host innate antiviral responses in obesity which protect the DIO mice to a certain degree when adaptive immunity is suboptimal. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Sujets)
Vaccins contre la COVID-19 , COVID-19 , Animaux , Humains , Souris , SARS-CoV-2 , Souris obèse , Réinfection , Régime alimentaire , Obésité , Anticorps neutralisants , Interféron alpha , ARN messager , Antiviraux , Anticorps antiviraux
7.
Viruses ; 15(3)2023 03 06.
Article Dans Anglais | MEDLINE | ID: covidwho-2253490

Résumé

The emergence of new immune-evasive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and subvariants outpaces the development of vaccines specific against the dominant circulating strains. In terms of the only accepted immune correlate of protection, the inactivated whole-virion vaccine using wild-type SARS-CoV-2 spike induces a much lower serum neutralizing antibody titre against the Omicron subvariants. Since the inactivated vaccine given intramuscularly is one of the most commonly used coronavirus disease 2019 (COVID-19) vaccines in developing regions, we tested the hypothesis that intranasal boosting after intramuscular priming would provide a broader level of protection. Here, we showed that one or two intranasal boosts with the Fc-linked trimeric spike receptor-binding domain from wild-type SARS-CoV-2 can induce significantly higher serum neutralizing antibodies against wild-type SARS-CoV-2 and the Omicron subvariants, including BA.5.2 and XBB.1, with a lower titre in the bronchoalveolar lavage of vaccinated Balb/c mice than vaccination with four intramuscular doses of inactivated whole virion vaccine. The intranasally vaccinated K18-hACE2-transgenic mice also had a significantly lower nasal turbinate viral load, suggesting a better protection of the upper airway, which is the predilected site of infection by Omicron subvariants. This intramuscular priming and intranasal boosting approach that achieves broader cross-protection against Omicron variants and subvariants may lengthen the interval required for changing the vaccine immunogen from months to years.


Sujets)
COVID-19 , Cornets , Souris , Animaux , SARS-CoV-2/génétique , Charge virale , COVID-19/prévention et contrôle , Souris transgéniques , Anticorps neutralisants , Vaccins contre la COVID-19 , Souris de lignée BALB C , Anticorps antiviraux , Glycoprotéine de spicule des coronavirus/génétique
9.
Cell Rep Med ; 3(9): 100743, 2022 09 20.
Article Dans Anglais | MEDLINE | ID: covidwho-2254238

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron BA.2 was a dominant circulating SARS-CoV-2 variant worldwide. Recent reports hint that BA.2 is similarly potent regarding antibody evasion but may be more transmissible than BA.1. The pathogenicity of BA.2 remains unclear and is of critical public health significance. Here we investigated the virological features and pathogenicity of BA.2 with in vitro and in vivo models. We show that BA.2 is less dependent on transmembrane protease serine 2 (TMPRSS2) for virus entry in comparison with BA.1 in vitro. In K18-hACE2 mice, BA.2 replicates more efficiently than BA.1 in the nasal turbinates and replicates marginally less efficiently in the lungs, leading to decreased body weight loss and improved survival. Our study indicates that BA.2 is similarly attenuated in lungs compared with BA.1 but is potentially more transmissible because of its better replication at the nasal turbinates.


Sujets)
COVID-19 , SARS-CoV-2 , Animaux , Humains , Souris , SARS-CoV-2/génétique , Sérine , Virulence
10.
Sci Adv ; 9(3): eadd3867, 2023 Jan 20.
Article Dans Anglais | MEDLINE | ID: covidwho-2239636

Résumé

Successful severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection requires proteolytic cleavage of the viral spike protein. While the role of the host transmembrane protease serine 2 in SARS-CoV-2 infection is widely recognized, the involvement of other proteases capable of facilitating SARS-CoV-2 entry remains incompletely explored. Here, we show that multiple members from the membrane-type matrix metalloproteinase (MT-MMP) and a disintegrin and metalloproteinase families can mediate SARS-CoV-2 entry. Inhibition of MT-MMPs significantly reduces SARS-CoV-2 replication in vitro and in vivo. Mechanistically, we show that MT-MMPs can cleave SARS-CoV-2 spike and angiotensin-converting enzyme 2 and facilitate spike-mediated fusion. We further demonstrate that Omicron BA.1 has an increased efficiency on MT-MMP usage, while an altered efficiency on transmembrane serine protease usage for virus entry compared with that of ancestral SARS-CoV-2. These results reveal additional protease determinants for SARS-CoV-2 infection and enhance our understanding on the biology of coronavirus entry.


Sujets)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/métabolisme , Peptide hydrolases/métabolisme , Protéolyse , Metalloproteases/métabolisme , Pénétration virale
11.
Clin Infect Dis ; 2022 Jun 28.
Article Dans Anglais | MEDLINE | ID: covidwho-2236091

Résumé

BACKGROUND: Early antiviral therapy was effective in the treatment of COVID-19. We assessed the efficacy and safety of combined interferon beta-1b and remdesivir treatment in hospitalized COVID-19 patients. METHODS: We conducted a multicentre, prospective open-label, randomized-controlled trial involving high-risk adults hospitalized for COVID-19. Patients were randomly assigned to a 5-day interferon beta-1b 16 million units daily and remdesivir 200mg loading on day 1 followed by 100mg daily on day 2 to 5 (combination-group), or to remdesivir only of similar regimen (control-group) (1:1). The primary end-point was the time to complete alleviation of symptoms (NEWS2 = 0). RESULTS: Two-hundred and twelve patients were enrolled. The median days of starting treatment from symptom-onset was 3 days. The median age was 65 years and 159 patients (75%) had chronic disease. The baseline demographics were similar. There was no mortality. For the primary-endpoint, the combination-group was significantly quicker to NEWS2 = 0 (4 versus 6.5 days; hazard-ratio [HR],6.59; 95% confidence-interval [CI],6.1-7.09; p < 0.0001) when compared to the control-group. For the secondary endpoints, the combination-group was quicker to negative NPS VL (6 versus 8 days; HR,8.16; 95% CI,7.79-8.52; p < 0.0001) and develop seropositive IgG (8 versus 10 days; HR,10.78; 95% CI,9.98-11.58; p < 0.0001). All adverse events resolved upon follow-up. Combination group (HR,4.1 95%CI,1.9-8.6, p < 0.0001), was the most significant independent factor associated with NEWS2 = 0 on day 4. CONCLUSIONS: Early treatment with interferon beta-1b and remdesivir was safe and better than remdesivir only in alleviating symptoms, shorten viral shedding and hospitalization with earlier seropositivity in high-risk COVID-19 patients.

12.
Viruses ; 15(2)2023 01 26.
Article Dans Anglais | MEDLINE | ID: covidwho-2216969

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease 2019 (COVID-19) pandemic that has caused disastrous effects on the society and human health globally. SARS-CoV-2 is a sarbecovirus in the Coronaviridae family with a positive-sense single-stranded RNA genome. It mainly replicates in the cytoplasm and viral components including RNAs and proteins can be sensed by pattern recognition receptors including toll-like receptors (TLRs), RIG-I-like receptors (RLRs), and NOD-like receptors (NLRs) that regulate the host innate and adaptive immune responses. On the other hand, the SARS-CoV-2 genome encodes multiple proteins that can antagonize the host immune response to facilitate viral replication. In this review, we discuss the current knowledge on host sensors and viral countermeasures against host innate immune response to provide insights on virus-host interactions and novel approaches to modulate host inflammation and antiviral responses.


Sujets)
COVID-19 , Virus du SRAS , Humains , SARS-CoV-2/génétique , Cytoplasme , Interactions hôte-microbes , ARN
13.
Infect Control Hosp Epidemiol ; 41(11): 1258-1265, 2020 11.
Article Dans Anglais | MEDLINE | ID: covidwho-2096345

Résumé

BACKGROUND: The role of severe respiratory coronavirus virus 2 (SARS-CoV-2)-laden aerosols in the transmission of coronavirus disease 2019 (COVID-19) remains uncertain. Discordant findings of SARS-CoV-2 RNA in air samples were noted in early reports. METHODS: Sampling of air close to 6 asymptomatic and symptomatic COVID-19 patients with and without surgical masks was performed with sampling devices using sterile gelatin filters. Frequently touched environmental surfaces near 21 patients were swabbed before daily environmental disinfection. The correlation between the viral loads of patients' clinical samples and environmental samples was analyzed. RESULTS: All air samples were negative for SARS-CoV-2 RNA in the 6 patients singly isolated inside airborne infection isolation rooms (AIIRs) with 12 air changes per hour. Of 377 environmental samples near 21 patients, 19 (5.0%) were positive by reverse-transcription polymerase chain reaction (RT-PCR) assay, with a median viral load of 9.2 × 102 copies/mL (range, 1.1 × 102 to 9.4 × 104 copies/mL). The contamination rate was highest on patients' mobile phones (6 of 77, 7.8%), followed by bed rails (4 of 74, 5.4%) and toilet door handles (4 of 76, 5.3%). We detected a significant correlation between viral load ranges in clinical samples and positivity rate of environmental samples (P < .001). CONCLUSION: SARS-CoV-2 RNA was not detectable by air samplers, which suggests that the airborne route is not the predominant mode of transmission of SARS-CoV-2. Wearing a surgical mask, appropriate hand hygiene, and thorough environmental disinfection are sufficient infection control measures for COVID-19 patients isolated singly in AIIRs. However, this conclusion may not apply during aerosol-generating procedures or in cohort wards with large numbers of COVID-19 patients.


Sujets)
Microbiologie de l'air , Betacoronavirus/isolement et purification , Infections à coronavirus/transmission , Matières contaminées/virologie , Prévention des infections/méthodes , Chambre de patient , Pneumopathie virale/transmission , Adolescent , Adulte , Aérosols , COVID-19 , Infections à coronavirus/diagnostic , Infections à coronavirus/prévention et contrôle , Femelle , Hospitalisation , Humains , Mâle , Adulte d'âge moyen , Pandémies/prévention et contrôle , Pneumopathie virale/diagnostic , Pneumopathie virale/prévention et contrôle , SARS-CoV-2 , Charge virale
14.
Cell Rep Med ; 3(10): 100774, 2022 10 18.
Article Dans Anglais | MEDLINE | ID: covidwho-2050073

Résumé

"Pan-coronavirus" antivirals targeting conserved viral components can be designed. Here, we show that the rationally engineered H84T-banana lectin (H84T-BanLec), which specifically recognizes high mannose found on viral proteins but seldom on healthy human cells, potently inhibits Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (including Omicron), and other human-pathogenic coronaviruses at nanomolar concentrations. H84T-BanLec protects against MERS-CoV and SARS-CoV-2 infection in vivo. Importantly, intranasally and intraperitoneally administered H84T-BanLec are comparably effective. Mechanistic assays show that H84T-BanLec targets virus entry. High-speed atomic force microscopy depicts real-time multimolecular associations of H84T-BanLec dimers with the SARS-CoV-2 spike trimer. Single-molecule force spectroscopy demonstrates binding of H84T-BanLec to multiple SARS-CoV-2 spike mannose sites with high affinity and that H84T-BanLec competes with SARS-CoV-2 spike for binding to cellular ACE2. Modeling experiments identify distinct high-mannose glycans in spike recognized by H84T-BanLec. The multiple H84T-BanLec binding sites on spike likely account for the drug compound's broad-spectrum antiviral activity and the lack of resistant mutants.


Sujets)
COVID-19 , Coronavirus du syndrome respiratoire du Moyen-Orient , Humains , SARS-CoV-2 , Lectines/pharmacologie , Mannose/pharmacologie , Angiotensin-converting enzyme 2 , Glycoprotéine de spicule des coronavirus/pharmacologie , Antiviraux/pharmacologie
15.
Emerg Microbes Infect ; 11(1): 2275-2287, 2022 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-2004930

Résumé

SARS-CoV-2 B.1.1.529.1 (Omicron BA.1) emerged in November 2021 and quickly became the predominant circulating SARS-CoV-2 variant globally. Omicron BA.1 contains more than 30 mutations in the spike protein, which contribute to its altered virological features when compared to the ancestral SARS-CoV-2 or previous SARS-CoV-2 variants. Recent studies by us and others demonstrated that Omicron BA.1 is less dependent on transmembrane serine protease 2 (TMPRSS2), less efficient in spike cleavage, less fusogenic, and adopts an altered propensity to utilize the plasma membrane and endosomal pathways for virus entry. Ongoing studies suggest that these virological features of Omicron BA.1 are in part retained by the subsequent Omicron sublineages. However, the exact spike determinants that contribute to these altered features of Omicron remain incompletely understood. In this study, we investigated the spike determinants for the observed virological characteristics of Omicron. By screening for the individual changes on Omicron BA.1 and BA.2 spike, we identify that 69-70 deletion, E484A, and H655Y contribute to the reduced TMPRSS2 usage while 25-27 deletion, S375F, and T376A result in less efficient spike cleavage. Among the shared spike mutations of BA.1 and BA.2, S375F and H655Y reduce spike-mediated fusogenicity. Interestingly, the H655Y change consistently reduces serine protease usage while increases the use of endosomal proteases. In keeping with these findings, the H655Y substitution alone reduces plasma membrane entry and facilitates endosomal entry when compared to SARS-CoV-2 WT. Overall, our study identifies key changes in Omicron spike that contributes to our understanding on the virological determinant and pathogenicity of Omicron.


Sujets)
COVID-19 , SARS-CoV-2 , Humains , Mutation , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme
16.
EBioMedicine ; 80: 104035, 2022 06.
Article Dans Anglais | MEDLINE | ID: covidwho-2004029
17.
Emerg Microbes Infect ; 11(1): 2093-2101, 2022 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-1978182

Résumé

The replication and pathogenicity of SARS-CoV-2 Omicron BA.2 are comparable to that of BA.1 in experimental animal models. However, BA.2 has rapidly emerged to overtake BA.1 to become the predominant circulating SARS-CoV-2 variant worldwide. Here, we compared the replication fitness of BA.1 and BA.2 in cell culture and in the Syrian hamster model of COVID-19. Using a reverse genetics approach, we found that the BA.1-specific spike mutation G496S compromises its replication fitness, which may contribute to BA.1 being outcompeted by BA.2 in the real world. Additionally, the BA.1-unique G496S substitution confers differentiated sensitivity to therapeutic monoclonal antibodies, which partially recapitulates the immunoevasive phenotype of BA.1 and BA.2. In summary, our study identified G496S as an important determinant during the evolutionary trajectory of SARS-CoV-2.


Sujets)
COVID-19 , SARS-CoV-2 , Animaux , Anticorps monoclonaux , Cricetinae , Humains , Mesocricetus , Mutation faux-sens , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique
18.
Nature ; 609(7928): 785-792, 2022 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1972633

Résumé

Highly pathogenic coronaviruses, including severe acute respiratory syndrome coronavirus 2 (refs. 1,2) (SARS-CoV-2), Middle East respiratory syndrome coronavirus3 (MERS-CoV) and SARS-CoV-1 (ref. 4), vary in their transmissibility and pathogenicity. However, infection by all three viruses results in substantial apoptosis in cell culture5-7 and in patient tissues8-10, suggesting a potential link between apoptosis and pathogenesis of coronaviruses. Here we show that caspase-6, a cysteine-aspartic protease of the apoptosis cascade, serves as an important host factor for efficient coronavirus replication. We demonstrate that caspase-6 cleaves coronavirus nucleocapsid proteins, generating fragments that serve as interferon antagonists, thus facilitating virus replication. Inhibition of caspase-6 substantially attenuates lung pathology and body weight loss in golden Syrian hamsters infected with SARS-CoV-2 and improves the survival of mice expressing human DPP4 that are infected with mouse-adapted MERS-CoV. Our study reveals how coronaviruses exploit a component of the host apoptosis cascade to facilitate virus replication.


Sujets)
Acide aspartique , Caspase-6 , Infections à coronavirus , Coronavirus , Cystéine , Interactions hôte-pathogène , Réplication virale , Animaux , Apoptose , Acide aspartique/métabolisme , Caspase-6/métabolisme , Coronavirus/croissance et développement , Coronavirus/pathogénicité , Infections à coronavirus/enzymologie , Infections à coronavirus/virologie , Protéines de la nucléocapside des coronavirus/immunologie , Protéines de la nucléocapside des coronavirus/métabolisme , Cricetinae , Cystéine/métabolisme , Dipeptidyl peptidase 4/génétique , Dipeptidyl peptidase 4/métabolisme , Humains , Interférons/antagonistes et inhibiteurs , Interférons/immunologie , Poumon/anatomopathologie , Mesocricetus , Souris , Coronavirus du syndrome respiratoire du Moyen-Orient , Virus du SRAS , SARS-CoV-2 , Taux de survie , Perte de poids
19.
Signal Transduct Target Ther ; 7(1): 266, 2022 08 03.
Article Dans Anglais | MEDLINE | ID: covidwho-1972575

Résumé

Defective interfering genes (DIGs) are short viral genomes and interfere with wild-type viral replication. Here, we demonstrate that the new designed SARS-CoV-2 DIG (CD3600) can significantly inhibit the replication of SARS-CoV-2 including Alpha, Delta, Kappa and Omicron variants in human HK-2 cells and influenza DIG (PAD4) can significantly inhibit influenza virus replication in human A549 cells. One dose of influenza DIGs prophylactically protects 90% mice from lethal challenge of A(H1N1)pdm09 virus and CD3600 inhibits SARS-CoV-2 replication in hamster lungs when DIGs are administrated to lungs one day before viral challenge. To further investigate the gene delivery vector in the respiratory tract, a peptidic TAT2-P1&LAH4, which can package genes to form small spherical nanoparticles with high endosomal escape ability, is demonstrated to dramatically increase gene expression in the lung airway. TAT2-P1&LAH4, with the dual-functional TAT2-P1 (gene-delivery and antiviral), can deliver CD3600 to significantly inhibit the replication of Delta and Omicron SARS-CoV-2 in hamster lungs. This peptide-based nanoparticle system can effectively transfect genes in lungs and deliver DIGs to inhibit SARS-CoV-2 variants and influenza virus in vivo, which provides the new insight into the drug delivery system for gene therapy against respiratory viruses.


Sujets)
COVID-19 , Sous-type H1N1 du virus de la grippe A , Virus de la grippe A , Grippe humaine , Nanoparticules , Animaux , COVID-19/génétique , Cricetinae , Humains , Sous-type H1N1 du virus de la grippe A/génétique , Grippe humaine/prévention et contrôle , Souris , Peptides/génétique , Peptides/pharmacologie , SARS-CoV-2/génétique
20.
Cell Discov ; 8(1): 57, 2022 Jun 17.
Article Dans Anglais | MEDLINE | ID: covidwho-1967594

Résumé

The airways and alveoli of the human respiratory tract are lined by two distinct types of epithelium, which are the primary targets of respiratory viruses. We previously established long-term expanding human lung epithelial organoids from lung tissues and developed a 'proximal' differentiation protocol to generate mucociliary airway organoids. However, a respiratory organoid system with bipotential of the airway and alveolar differentiation remains elusive. Here we defined a 'distal' differentiation approach to generate alveolar organoids from the same source for the derivation of airway organoids. The alveolar organoids consisting of type I and type II alveolar epithelial cells (AT1 and AT2, respectively) functionally simulate the alveolar epithelium. AT2 cells maintained in lung organoids serve as progenitor cells from which alveolar organoids derive. Moreover, alveolar organoids sustain a productive SARS-CoV-2 infection, albeit a lower replicative fitness was observed compared to that in airway organoids. We further optimized 2-dimensional (2D) airway organoids. Upon differentiation under a slightly acidic pH, the 2D airway organoids exhibit enhanced viral replication, representing an optimal in vitro correlate of respiratory epithelium for modeling the high infectivity of SARS-CoV-2. Notably, the higher infectivity and replicative fitness of the Omicron variant than an ancestral strain were accurately recapitulated in these optimized airway organoids. In conclusion, we have established a bipotential organoid culture system able to reproducibly expand the entire human respiratory epithelium in vitro for modeling respiratory diseases, including COVID-19.

SÉLECTION CITATIONS
Détails de la recherche