Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
Plus de filtres










Base de données
Gamme d'année
2.
Proc Natl Acad Sci U S A ; 121(18): e2319566121, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38648490

RÉSUMÉ

Respiratory virus infections in humans cause a broad-spectrum of diseases that result in substantial morbidity and mortality annually worldwide. To reduce the global burden of respiratory viral diseases, preventative and therapeutic interventions that are accessible and effective are urgently needed, especially in countries that are disproportionately affected. Repurposing generic medicine has the potential to bring new treatments for infectious diseases to patients efficiently and equitably. In this study, we found that intranasal delivery of neomycin, a generic aminoglycoside antibiotic, induces the expression of interferon-stimulated genes (ISGs) in the nasal mucosa that is independent of the commensal microbiota. Prophylactic or therapeutic administration of neomycin provided significant protection against upper respiratory infection and lethal disease in a mouse model of COVID-19. Furthermore, neomycin treatment protected Mx1 congenic mice from upper and lower respiratory infections with a highly virulent strain of influenza A virus. In Syrian hamsters, neomycin treatment potently mitigated contact transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In healthy humans, intranasal application of neomycin-containing Neosporin ointment was well tolerated and effective at inducing ISG expression in the nose in a subset of participants. These findings suggest that neomycin has the potential to be harnessed as a host-directed antiviral strategy for the prevention and treatment of respiratory viral infections.


Sujet(s)
Administration par voie nasale , Antiviraux , Néomycine , SARS-CoV-2 , Animaux , Néomycine/pharmacologie , Néomycine/administration et posologie , Souris , Humains , Antiviraux/pharmacologie , Antiviraux/administration et posologie , SARS-CoV-2/immunologie , SARS-CoV-2/effets des médicaments et des substances chimiques , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/virologie , Infections de l'appareil respiratoire/immunologie , Infections de l'appareil respiratoire/traitement médicamenteux , Infections de l'appareil respiratoire/virologie , Infections de l'appareil respiratoire/prévention et contrôle , Muqueuse nasale/immunologie , Muqueuse nasale/virologie , Muqueuse nasale/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Traitements médicamenteux de la COVID-19 , Mesocricetus , Femelle , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/immunologie
3.
Sci Transl Med ; 15(709): eabq0603, 2023 08 16.
Article de Anglais | MEDLINE | ID: mdl-37585505

RÉSUMÉ

An inhalable platform for messenger RNA (mRNA) therapeutics would enable minimally invasive and lung-targeted delivery for a host of pulmonary diseases. Development of lung-targeted mRNA therapeutics has been limited by poor transfection efficiency and risk of vehicle-induced pathology. Here, we report an inhalable polymer-based vehicle for delivery of therapeutic mRNAs to the lung. We optimized biodegradable poly(amine-co-ester) (PACE) polyplexes for mRNA delivery using end-group modifications and polyethylene glycol. These polyplexes achieved high transfection of mRNA throughout the lung, particularly in epithelial and antigen-presenting cells. We applied this technology to develop a mucosal vaccine for severe acute respiratory syndrome coronavirus 2 and found that intranasal vaccination with spike protein-encoding mRNA polyplexes induced potent cellular and humoral adaptive immunity and protected susceptible mice from lethal viral challenge. Together, these results demonstrate the translational potential of PACE polyplexes for therapeutic delivery of mRNA to the lungs.


Sujet(s)
COVID-19 , Nanoparticules , Animaux , Souris , Polymères , ARN messager/génétique , COVID-19/prévention et contrôle , Poumon , Vaccination
4.
Science ; 378(6622): eabo2523, 2022 11 25.
Article de Anglais | MEDLINE | ID: mdl-36302057

RÉSUMÉ

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the need for vaccines that not only prevent disease but also prevent transmission. Parenteral vaccines induce robust systemic immunity but poor immunity at the respiratory mucosa. We developed a vaccine strategy that we call "prime and spike," which leverages existing immunity generated by primary vaccination (prime) to elicit mucosal immune memory within the respiratory tract by using unadjuvanted intranasal spike boosters (spike). We show that prime and spike induces robust resident memory B and T cell responses, induces immunoglobulin A at the respiratory mucosa, boosts systemic immunity, and completely protects mice with partial immunity from lethal SARS-CoV-2 infection. Using divergent spike proteins, prime and spike enables the induction of cross-reactive immunity against sarbecoviruses.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Immunité muqueuse , Mémoire immunologique , Cellules B mémoire , Cellules T mémoire , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , Souris , Administration par voie nasale , Anticorps antiviraux , COVID-19/prévention et contrôle , COVID-19/transmission , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Vaccination/méthodes , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/immunologie , Immunoglobuline A , Cellules B mémoire/immunologie , Cellules T mémoire/immunologie
5.
Cell ; 185(14): 2452-2468.e16, 2022 07 07.
Article de Anglais | MEDLINE | ID: mdl-35768006

RÉSUMÉ

COVID survivors frequently experience lingering neurological symptoms that resemble cancer-therapy-related cognitive impairment, a syndrome for which white matter microglial reactivity and consequent neural dysregulation is central. Here, we explored the neurobiological effects of respiratory SARS-CoV-2 infection and found white-matter-selective microglial reactivity in mice and humans. Following mild respiratory COVID in mice, persistently impaired hippocampal neurogenesis, decreased oligodendrocytes, and myelin loss were evident together with elevated CSF cytokines/chemokines including CCL11. Systemic CCL11 administration specifically caused hippocampal microglial reactivity and impaired neurogenesis. Concordantly, humans with lasting cognitive symptoms post-COVID exhibit elevated CCL11 levels. Compared with SARS-CoV-2, mild respiratory influenza in mice caused similar patterns of white-matter-selective microglial reactivity, oligodendrocyte loss, impaired neurogenesis, and elevated CCL11 at early time points, but after influenza, only elevated CCL11 and hippocampal pathology persisted. These findings illustrate similar neuropathophysiology after cancer therapy and respiratory SARS-CoV-2 infection which may contribute to cognitive impairment following even mild COVID.


Sujet(s)
COVID-19 , Grippe humaine , Tumeurs , Animaux , Humains , Grippe humaine/anatomopathologie , Souris , Microglie/anatomopathologie , Gaine de myéline , Tumeurs/anatomopathologie , SARS-CoV-2
6.
Nature ; 606(7914): 585-593, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35483404

RÉSUMÉ

Severe COVID-19 is characterized by persistent lung inflammation, inflammatory cytokine production, viral RNA and a sustained interferon (IFN) response, all of which are recapitulated and required for pathology in the SARS-CoV-2-infected MISTRG6-hACE2 humanized mouse model of COVID-19, which has a human immune system1-20. Blocking either viral replication with remdesivir21-23 or the downstream IFN-stimulated cascade with anti-IFNAR2 antibodies in vivo in the chronic stages of disease attenuates the overactive immune inflammatory response, especially inflammatory macrophages. Here we show that SARS-CoV-2 infection and replication in lung-resident human macrophages is a critical driver of disease. In response to infection mediated by CD16 and ACE2 receptors, human macrophages activate inflammasomes, release interleukin 1 (IL-1) and IL-18, and undergo pyroptosis, thereby contributing to the hyperinflammatory state of the lungs. Inflammasome activation and the accompanying inflammatory response are necessary for lung inflammation, as inhibition of the NLRP3 inflammasome pathway reverses chronic lung pathology. Notably, this blockade of inflammasome activation leads to the release of infectious virus by the infected macrophages. Thus, inflammasomes oppose host infection by SARS-CoV-2 through the production of inflammatory cytokines and suicide by pyroptosis to prevent a productive viral cycle.


Sujet(s)
COVID-19 , Inflammasomes , Macrophages , SARS-CoV-2 , Angiotensin-converting enzyme 2 , Animaux , COVID-19/anatomopathologie , COVID-19/physiopathologie , COVID-19/virologie , Humains , Inflammasomes/métabolisme , Interleukine-1 , Interleukine-18 , Poumon/anatomopathologie , Poumon/virologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Macrophages/virologie , Souris , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/virologie , Pyroptose , Récepteurs du fragment Fc des IgG , SARS-CoV-2/métabolisme , SARS-CoV-2/pathogénicité
7.
Nat Biotechnol ; 40(5): 681-691, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35228707

RÉSUMÉ

As the biomedical community produces datasets that are increasingly complex and high dimensional, there is a need for more sophisticated computational tools to extract biological insights. We present Multiscale PHATE, a method that sweeps through all levels of data granularity to learn abstracted biological features directly predictive of disease outcome. Built on a coarse-graining process called diffusion condensation, Multiscale PHATE learns a data topology that can be analyzed at coarse resolutions for high-level summarizations of data and at fine resolutions for detailed representations of subsets. We apply Multiscale PHATE to a coronavirus disease 2019 (COVID-19) dataset with 54 million cells from 168 hospitalized patients and find that patients who die show CD16hiCD66blo neutrophil and IFN-γ+ granzyme B+ Th17 cell responses. We also show that population groupings from Multiscale PHATE directly fed into a classifier predict disease outcome more accurately than naive featurizations of the data. Multiscale PHATE is broadly generalizable to different data types, including flow cytometry, single-cell RNA sequencing (scRNA-seq), single-cell sequencing assay for transposase-accessible chromatin (scATAC-seq), and clinical variables.


Sujet(s)
COVID-19 , Analyse sur cellule unique , Chromatine , Humains , Analyse sur cellule unique/méthodes , Transposases ,
8.
bioRxiv ; 2022 Mar 23.
Article de Anglais | MEDLINE | ID: mdl-35350207

RÉSUMÉ

An inhalable platform for mRNA therapeutics would enable minimally invasive and lung targeted delivery for a host of pulmonary diseases. Development of lung targeted mRNA therapeutics has been limited by poor transfection efficiency and risk of vehicle-induced pathology. Here we report an inhalable polymer-based vehicle for delivery of therapeutic mRNAs to the lung. We optimized biodegradable poly(amine-co-ester) polyplexes for mRNA delivery using end group modifications and polyethylene glycol. Our polyplexes achieved high transfection of mRNA throughout the lung, particularly in epithelial and antigen-presenting cells. We applied this technology to develop a mucosal vaccine for SARS-CoV-2. Intranasal vaccination with spike protein mRNA polyplexes induced potent cellular and humoral adaptive immunity and protected K18-hACE2 mice from lethal viral challenge. One-sentence summary: Inhaled polymer nanoparticles (NPs) achieve high mRNA expression in the lung and induce protective immunity against SARS-CoV-2.

9.
bioRxiv ; 2022 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-35118464

RÉSUMÉ

As the SARS-CoV-2 pandemic enters its third year, vaccines that not only prevent disease, but also prevent transmission are needed to help reduce global disease burden. Currently approved parenteral vaccines induce robust systemic immunity, but poor immunity at the respiratory mucosa. Here we describe the development of a novel vaccine strategy, Prime and Spike, based on unadjuvanted intranasal spike boosting that leverages existing immunity generated by primary vaccination to elicit mucosal immune memory within the respiratory tract. We show that Prime and Spike induces robust T resident memory cells, B resident memory cells and IgA at the respiratory mucosa, boosts systemic immunity, and completely protects mice with partial immunity from lethal SARS-CoV-2 infection. Using divergent spike proteins, Prime and Spike enables induction of cross-reactive immunity against sarbecoviruses without invoking original antigenic sin. ONE-SENTENCE SUMMARY: Broad sarbecovirus protective mucosal immunity is generated by unadjuvanted intranasal spike boost in preclinical model.

10.
bioRxiv ; 2022 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-34611663

RÉSUMÉ

Severe COVID-19 is characterized by persistent lung inflammation, inflammatory cytokine production, viral RNA, and sustained interferon (IFN) response all of which are recapitulated and required for pathology in the SARS-CoV-2 infected MISTRG6-hACE2 humanized mouse model of COVID-19 with a human immune system 1-20 . Blocking either viral replication with Remdesivir 21-23 or the downstream IFN stimulated cascade with anti-IFNAR2 in vivo in the chronic stages of disease attenuated the overactive immune-inflammatory response, especially inflammatory macrophages. Here, we show SARS-CoV-2 infection and replication in lung-resident human macrophages is a critical driver of disease. In response to infection mediated by CD16 and ACE2 receptors, human macrophages activate inflammasomes, release IL-1 and IL-18 and undergo pyroptosis thereby contributing to the hyperinflammatory state of the lungs. Inflammasome activation and its accompanying inflammatory response is necessary for lung inflammation, as inhibition of the NLRP3 inflammasome pathway reverses chronic lung pathology. Remarkably, this same blockade of inflammasome activation leads to the release of infectious virus by the infected macrophages. Thus, inflammasomes oppose host infection by SARS-CoV-2 by production of inflammatory cytokines and suicide by pyroptosis to prevent a productive viral cycle.

11.
J Exp Med ; 219(1)2022 01 03.
Article de Anglais | MEDLINE | ID: mdl-34757384

RÉSUMÉ

As SARS-CoV-2 continues to cause morbidity and mortality around the world, there is an urgent need for the development of effective medical countermeasures. Here, we assessed the antiviral capacity of a minimal RIG-I agonist, stem-loop RNA 14 (SLR14), in viral control, disease prevention, post-infection therapy, and cross-variant protection in mouse models of SARS-CoV-2 infection. A single dose of SLR14 prevented viral infection in the lower respiratory tract and development of severe disease in a type I interferon (IFN-I)-dependent manner. SLR14 demonstrated remarkable prophylactic protective capacity against lethal SARS-CoV-2 infection and retained considerable efficacy as a therapeutic agent. In immunodeficient mice carrying chronic SARS-CoV-2 infection, SLR14 elicited near-sterilizing innate immunity in the absence of the adaptive immune system. In the context of infection with variants of concern (VOCs), SLR14 conferred broad protection against emerging VOCs. These findings demonstrate the therapeutic potential of SLR14 as a host-directed, broad-spectrum antiviral for early post-exposure treatment and treatment of chronically infected immunosuppressed patients.


Sujet(s)
Antiviraux/pharmacologie , Traitements médicamenteux de la COVID-19 , ARN/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , Animaux , COVID-19/métabolisme , Modèles animaux de maladie humaine , Immunité innée/effets des médicaments et des substances chimiques , Interféron de type I/métabolisme , Souris , Souris de lignée BALB C
12.
Sci Immunol ; 7(68): eabl5652, 2022 Feb 04.
Article de Anglais | MEDLINE | ID: mdl-34914544

RÉSUMÉ

T follicular helper (TFH) cells are the conventional drivers of protective, germinal center (GC)­based antiviral antibody responses. However, loss of TFH cells and GCs has been observed in patients with severe COVID-19. As T cell­B cell interactions and immunoglobulin class switching still occur in these patients, noncanonical pathways of antibody production may be operative during SARS-CoV-2 infection. We found that both TFH-dependent and -independent antibodies were induced against SARS-CoV-2 infection, SARS-CoV-2 vaccination, and influenza A virus infection. Although TFH-independent antibodies to SARS-CoV-2 had evidence of reduced somatic hypermutation, they were still high affinity, durable, and reactive against diverse spike-derived epitopes and were capable of neutralizing both homologous SARS-CoV-2 and the B.1.351 (beta) variant of concern. We found by epitope mapping and B cell receptor sequencing that TFH cells focused the B cell response, and therefore, in the absence of TFH cells, a more diverse clonal repertoire was maintained. These data support an alternative pathway for the induction of B cell responses during viral infection that enables effective, neutralizing antibody production to complement traditional GC-derived antibodies that might compensate for GCs damaged by viral inflammation.


Sujet(s)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , Lymphocytes T auxiliaires folliculaires/immunologie , Séquence d'acides aminés , Animaux , Production d'anticorps/immunologie , Lymphocytes B/immunologie , Vaccins contre la COVID-19/immunologie , Centre germinatif/immunologie , Humains , Activation des lymphocytes/immunologie , Souris , Lymphocytes T auxiliaires
13.
Nat Biotechnol ; 40(6): 906-920, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-34921308

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) is an infectious disease that can present as an uncontrolled, hyperactive immune response, causing severe immunological injury. Existing rodent models do not recapitulate the sustained immunopathology of patients with severe disease. Here we describe a humanized mouse model of COVID-19 that uses adeno-associated virus to deliver human ACE2 to the lungs of humanized MISTRG6 mice. This model recapitulates innate and adaptive human immune responses to severe acute respiratory syndrome coronavirus 2 infection up to 28 days after infection, with key features of chronic COVID-19, including weight loss, persistent viral RNA, lung pathology with fibrosis, a human inflammatory macrophage response, a persistent interferon-stimulated gene signature and T cell lymphopenia. We used this model to study two therapeutics on immunopathology, patient-derived antibodies and steroids and found that the same inflammatory macrophages crucial to containing early infection later drove immunopathology. This model will enable evaluation of COVID-19 disease mechanisms and treatments.


Sujet(s)
COVID-19 , Animaux , Antiviraux , Modèles animaux de maladie humaine , Humains , Interférons , Poumon/anatomopathologie , Souris
14.
J Infect Dis ; 225(3): 374-384, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-34718647

RÉSUMÉ

BACKGROUND: The underlying immunologic deficiencies enabling severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection are currently unknown. We describe deep longitudinal immune profiling of a transplant recipient hospitalized twice for coronavirus disease 2019 (COVID-19). METHODS: A 66-year-old male renal transplant recipient was hospitalized with COVID-19 March 2020 then readmitted to the hospital with COVID-19 233 days after initial diagnosis. Virologic and immunologic investigations were performed on samples from the primary and secondary infections. RESULTS: Whole viral genome sequencing and phylogenetic analysis revealed that viruses causing both infections were caused by distinct genetic lineages without evidence of immune escape mutations. Longitudinal comparison of cellular and humoral responses during primary SARS-CoV-2 infection revealed that this patient responded to the primary infection with low neutralization titer anti-SARS-CoV-2 antibodies that were likely present at the time of reinfection. CONCLUSIONS: The development of neutralizing antibodies and humoral memory responses in this patient failed to confer protection against reinfection, suggesting that they were below a neutralizing titer threshold or that additional factors may be required for efficient prevention of SARS-CoV-2 reinfection. Development of poorly neutralizing antibodies may have been due to profound and relatively specific reduction in naive CD4 T-cell pools. Seropositivity alone may not be a perfect correlate of protection in immunocompromised patients.


Sujet(s)
COVID-19 , Réinfection , Receveurs de transplantation , Sujet âgé , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , Humains , Mâle , Transplantation d'organe , Phylogenèse , Réinfection/immunologie , Réinfection/virologie , SARS-CoV-2/génétique
15.
Sci Immunol ; 6(64): eabl4509, 2021 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-34623900

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused more than 160 million infections and more than 3 million deaths worldwide. Although effective vaccines are currently being deployed, the adaptive immune determinants that promote viral clearance and confer protection remain poorly defined. Using mouse models of SARS-CoV-2, we demonstrate that both humoral and cellular adaptive immunity contribute to viral clearance in the setting of primary infection. Furthermore, we find that either convalescent mice or mice that receive mRNA vaccination are protected from both homologous infection and infection with a variant of concern, B.1.351. In addition, we find that this protection is largely mediated by antibody response and not cellular immunity. These results highlight the in vivo protective capacity of antibodies generated to both vaccine and natural infection.


Sujet(s)
Vaccins contre la COVID-19/pharmacologie , COVID-19/immunologie , COVID-19/prévention et contrôle , Immunité cellulaire , Immunité humorale , SARS-CoV-2/immunologie , Animaux , COVID-19/génétique , Vaccins contre la COVID-19/immunologie , Chlorocebus aethiops , Modèles animaux de maladie humaine , Femelle , Mâle , Souris , Souris knockout , Cellules Vero
17.
Sci Signal ; 14(690)2021 07 06.
Article de Anglais | MEDLINE | ID: mdl-34230210

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) has poorer clinical outcomes in males than in females, and immune responses underlie these sex-related differences. Because immune responses are, in part, regulated by metabolites, we examined the serum metabolomes of COVID-19 patients. In male patients, kynurenic acid (KA) and a high KA-to-kynurenine (K) ratio (KA:K) positively correlated with age and with inflammatory cytokines and chemokines and negatively correlated with T cell responses. Males that clinically deteriorated had a higher KA:K than those that stabilized. KA inhibits glutamate release, and glutamate abundance was lower in patients that clinically deteriorated and correlated with immune responses. Analysis of data from the Genotype-Tissue Expression (GTEx) project revealed that the expression of the gene encoding the enzyme that produces KA, kynurenine aminotransferase, correlated with cytokine abundance and activation of immune responses in older males. This study reveals that KA has a sex-specific link to immune responses and clinical outcomes in COVID-19, suggesting a positive feedback between metabolites and immune responses in males.


Sujet(s)
COVID-19/immunologie , Acide kynurénique/immunologie , SARS-CoV-2 , Adulte , Sujet âgé , COVID-19/sang , Études cas-témoins , Syndrome de libération de cytokines/sang , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/immunologie , Cytokines/sang , Cytokines/immunologie , Femelle , Humains , Acide kynurénique/sang , Modèles logistiques , Mâle , Voies et réseaux métaboliques/immunologie , Métabolomique , Adulte d'âge moyen , Analyse multifactorielle , Indice de gravité de la maladie , Facteurs sexuels , Transduction du signal/immunologie , Tryptophane/métabolisme
18.
bioRxiv ; 2021 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-34159330

RÉSUMÉ

As SARS-CoV-2 continues to cause morbidity and mortality around the world, there is an urgent need for the development of effective medical countermeasures. Here, we assessed the antiviral capacity of a minimal RIG-I agonist, stem-loop RNA 14 (SLR14), in viral control, disease prevention, post-infection therapy, and cross-variant protection in mouse models of SARS-CoV-2 infection. A single dose of SLR14 prevented viral replication in the lower respiratory tract and development of severe disease in a type I interferon (IFN-I) dependent manner. SLR14 demonstrated remarkable protective capacity against lethal SARS-CoV-2 infection when used prophylactically and retained considerable efficacy as a therapeutic agent. In immunodeficient mice carrying chronic SARS-CoV-2 infection, SLR14 elicited near-sterilizing innate immunity by inducing IFN-I responses in the absence of the adaptive immune system. In the context of infection with variants of concern (VOC), SLR14 conferred broad protection and uncovered an IFN-I resistance gradient across emerging VOC. These findings demonstrate the therapeutic potential of SLR14 as a host-directed, broad-spectrum antiviral for early post-exposure treatment and for treatment of chronically infected immunosuppressed patients.

19.
PLoS Pathog ; 17(6): e1009683, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-34166473

RÉSUMÉ

COVID-19 is a global crisis of unimagined dimensions. Currently, Remedesivir is only fully licensed FDA therapeutic. A major target of the vaccine effort is the SARS-CoV-2 spike-hACE2 interaction, and assessment of efficacy relies on time consuming neutralization assay. Here, we developed a cell fusion assay based upon spike-hACE2 interaction. The system was tested by transient co-transfection of 293T cells, which demonstrated good correlation with standard spike pseudotyping for inhibition by sera and biologics. Then established stable cell lines were very well behaved and gave even better correlation with pseudotyping results, after a short, overnight co-incubation. Results with the stable cell fusion assay also correlated well with those of a live virus assay. In summary we have established a rapid, reliable, and reproducible cell fusion assay that will serve to complement the other neutralization assays currently in use, is easy to implement in most laboratories, and may serve as the basis for high throughput screens to identify inhibitors of SARS-CoV-2 virus-cell binding and entry.


Sujet(s)
Angiotensin-converting enzyme 2/métabolisme , Dosage biologique/méthodes , COVID-19/virologie , Récepteurs du coronavirus/métabolisme , SARS-CoV-2/physiologie , Glycoprotéine de spicule des coronavirus/métabolisme , Angiotensin-converting enzyme 2/génétique , COVID-19/sang , Fusion cellulaire , Cellules HEK293 , Humains , Récepteurs du coronavirus/génétique , Glycoprotéine de spicule des coronavirus/génétique , Transfection , Attachement viral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...