Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 135
Filtrer
1.
PLoS Pathog ; 20(6): e1012262, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38924060

RÉSUMÉ

Viral haemorrhagic fevers (VHF) pose a significant threat to human health. In recent years, VHF outbreaks caused by Ebola, Marburg and Lassa viruses have caused substantial morbidity and mortality in West and Central Africa. In 2022, an Ebola disease outbreak in Uganda caused by Sudan virus resulted in 164 cases with 55 deaths. In 2023, a Marburg disease outbreak was confirmed in Equatorial Guinea and Tanzania resulting in over 49 confirmed or suspected cases; 41 of which were fatal. There are no clearly defined correlates of protection against these VHF, impeding targeted vaccine development. Any vaccine developed should therefore induce strong and preferably long-lasting humoral and cellular immunity against these viruses. Ideally this immunity should also cross-protect against viral variants, which are known to circulate in animal reservoirs and cause human disease. We have utilized two viral vectored vaccine platforms, an adenovirus (ChAdOx1) and Modified Vaccinia Ankara (MVA), to develop a multi-pathogen vaccine regime against three filoviruses (Ebola virus, Sudan virus, Marburg virus) and an arenavirus (Lassa virus). These platform technologies have consistently demonstrated the capability to induce robust cellular and humoral antigen-specific immunity in humans, most recently in the rollout of the licensed ChAdOx1-nCoV19/AZD1222. Here, we show that our multi-pathogen vaccines elicit strong cellular and humoral immunity, induce a diverse range of chemokines and cytokines, and most importantly, confers protection after lethal Ebola virus, Sudan virus and Marburg virus challenges in a small animal model.

2.
Virus Res ; 346: 199409, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38815869

RÉSUMÉ

Crimean-Congo Haemorrhagic Fever Virus (CCHFV) is spread by infected ticks or direct contact with blood, tissues and fluids from infected patients or livestock. Infection with CCHFV causes severe haemorrhagic fever in humans which is fatal in up to 83 % of cases. CCHFV is listed as a priority pathogen by the World Health Organization (WHO) and there are currently no widely-approved vaccines. Defining a serological correlate of protection against CCHFV infection would support the development of vaccines by providing a 'target threshold' for pre-clinical and clinical immunogenicity studies to achieve in subjects and potentially obviate the need for in vivo protection studies. We therefore sought to establish titratable protection against CCHFV using pooled human convalescent plasma, in a mouse model. Convalescent plasma collected from seven individuals with a known previous CCHFV virus infection were characterised using binding antibody and neutralisation assays. All plasma recognised nucleoprotein and the Gc glycoprotein, but some had a lower Gn glycoprotein response by ELISA. Pooled plasma and two individual donations from convalescent donors were administered intraperitoneally to A129 mice 24 h prior to intradermal challenge with CCHFV (strain IbAr10200). A partial protective effect was observed with all three convalescent plasmas characterised by longer survival post-challenge and reduced clinical score. These protective responses were titratable. Further characterisation of the serological reactivities within these samples will establish their value as reference materials to support assay harmonisation and accelerate vaccine development for CCHFV.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Modèles animaux de maladie humaine , Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Animaux , Fièvre hémorragique de Crimée-Congo/immunologie , Fièvre hémorragique de Crimée-Congo/prévention et contrôle , Souris , Virus de la fièvre hémorragique de Crimée-Congo/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Humains , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Femelle , Tests de neutralisation , Plasma sanguin/immunologie , Mâle
3.
J Virol ; 98(6): e0052424, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38757972

RÉSUMÉ

Ebola virus glycoprotein (EBOV GP) is one of the most heavily O-glycosylated viral glycoproteins, yet we still lack a fundamental understanding of the structure of its large O-glycosylated mucin-like domain and to what degree the host O-glycosylation capacity influences EBOV replication. Using tandem mass spectrometry, we identified 47 O-glycosites on EBOV GP and found similar glycosylation signatures on virus-like particle- and cell lysate-derived GP. Furthermore, we performed quantitative differential O-glycoproteomics on proteins produced in wild-type HEK293 cells and cell lines ablated for the three key initiators of O-linked glycosylation, GalNAc-T1, -T2, and -T3. The data show that 12 out of the 47 O-glycosylated sites were regulated, predominantly by GalNAc-T1. Using the glycoengineered cell lines for authentic EBOV propagation, we demonstrate the importance of O-linked glycan initiation and elongation for the production of viral particles and the titers of progeny virus. The mapped O-glycan positions and structures allowed to generate molecular dynamics simulations probing the largely unknown spatial arrangements of the mucin-like domain. The data highlight targeting GALNT1 or C1GALT1C1 as a possible way to modulate O-glycan density on EBOV GP for novel vaccine designs and tailored intervention approaches.IMPORTANCEEbola virus glycoprotein acquires its extensive glycan shield in the host cell, where it is decorated with N-linked glycans and mucin-type O-linked glycans. The latter is initiated by a family of polypeptide GalNAc-transferases that have different preferences for optimal peptide substrates resulting in a spectrum of both very selective and redundant substrates for each isoform. In this work, we map the exact locations of O-glycans on Ebola virus glycoprotein and identify subsets of sites preferentially initiated by one of the three key isoforms of GalNAc-Ts, demonstrating that each enzyme contributes to the glycan shield integrity. We further show that altering host O-glycosylation capacity has detrimental effects on Ebola virus replication, with both isoform-specific initiation and elongation playing a role. The combined structural and functional data highlight glycoengineered cell lines as useful tools for investigating molecular mechanisms imposed by specific glycans and for steering the immune responses in future vaccine designs.


Sujet(s)
Ebolavirus , Polyosides , Réplication virale , Ebolavirus/physiologie , Ebolavirus/métabolisme , Humains , Cellules HEK293 , Glycosylation , Polyosides/métabolisme , Protéines de l'enveloppe virale/métabolisme , Fièvre hémorragique à virus Ebola/virologie , Fièvre hémorragique à virus Ebola/métabolisme , N-acetylgalactosaminyltransferase/métabolisme , N-acetylgalactosaminyltransferase/génétique , Glycoprotéines/métabolisme ,
4.
J Gen Virol ; 105(4)2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38687001

RÉSUMÉ

Nairoviridae is a family for negative-sense RNA viruses with genomes of about 17.2-21.1 kb. These viruses are maintained in and/or transmitted by arthropods among birds, reptiles and mammals. Norwaviruses and orthonairoviruses can cause febrile illness in humans. Several orthonairoviruses can infect mammals, causing mild, severe and sometimes, fatal diseases. Nairovirids produce enveloped virions containing two or three single-stranded RNA segments with open reading frames that encode a nucleoprotein (N), sometimes a glycoprotein precursor (GPC), and a large (L) protein containing an RNA-directed RNA polymerase (RdRP) domain. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) report on the family Nairoviridae, which is available at www.ictv.global/report/nairoviridae.


Sujet(s)
Génome viral , Animaux , Humains , Cadres ouverts de lecture , Protéines virales/génétique , Nairovirus/génétique , Nairovirus/classification , Nairovirus/isolement et purification , ARN viral/génétique , Phylogenèse , Virion/ultrastructure , RNA replicase/génétique
5.
Nat Microbiol ; 9(6): 1499-1512, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38548922

RÉSUMÉ

Climate change and population densities accelerated transmission of highly pathogenic viruses to humans, including the Crimean-Congo haemorrhagic fever virus (CCHFV). Here we report that the Low Density Lipoprotein Receptor (LDLR) is a critical receptor for CCHFV cell entry, playing a vital role in CCHFV infection in cell culture and blood vessel organoids. The interaction between CCHFV and LDLR is highly specific, with other members of the LDLR protein family failing to bind to or neutralize the virus. Biosensor experiments demonstrate that LDLR specifically binds the surface glycoproteins of CCHFV. Importantly, mice lacking LDLR exhibit a delay in CCHFV-induced disease. Furthermore, we identified the presence of Apolipoprotein E (ApoE) on CCHFV particles. Our findings highlight the essential role of LDLR in CCHFV infection, irrespective of ApoE presence, when the virus is produced in tick cells. This discovery holds profound implications for the development of future therapies against CCHFV.


Sujet(s)
Apolipoprotéines E , Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Récepteurs aux lipoprotéines LDL , Pénétration virale , Animaux , Humains , Souris , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Virus de la fièvre hémorragique de Crimée-Congo/physiologie , Fièvre hémorragique de Crimée-Congo/virologie , Fièvre hémorragique de Crimée-Congo/métabolisme , Souris knockout , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs viraux/métabolisme , Tiques/virologie , Tiques/métabolisme
6.
Antiviral Res ; 225: 105844, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38428749

RÉSUMÉ

The Third International Conference on Crimean-Congo Hemorrhagic Fever (CCHF) was held in Thessaloniki, Greece, September 19-21, 2023, bringing together a diverse group of international partners, including public health professionals, clinicians, ecologists, epidemiologists, immunologists, and virologists. The conference was attended by 118 participants representing 24 countries and the World Health Organization (WHO). Meeting sessions covered the epidemiology of CCHF in humans; Crimean-Congo hemorrhagic fever virus (CCHFV) in ticks; wild and domestic animal hosts; molecular virology; pathogenesis and animal models; immune response related to therapeutics; and CCHF prevention in humans. The concluding session focused on recent WHO recommendations regarding disease prevention, control strategies, and innovations against CCHFV outbreaks. This meeting report summarizes lectures by the invited speakers and highlights advances in the field.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Tiques , Animaux , Humains , Fièvre hémorragique de Crimée-Congo/épidémiologie , Grèce , Épidémies de maladies
7.
Microorganisms ; 12(3)2024 Feb 22.
Article de Anglais | MEDLINE | ID: mdl-38543494

RÉSUMÉ

While having already killed more than 7 million of people worldwide in 4 years, SARS-CoV-2, the etiological agent of COVID-19, is still circulating and evolving. Understanding the pathogenesis of the virus is of capital importance. It was shown that in vitro and in vivo infection with SARS-CoV-2 can lead to cell cycle arrest but the effect of the cell cycle arrest on the virus infection and the associated mechanisms are still unclear. By stopping cells in the G1 phase as well as targeting several pathways involved using inhibitors and small interfering RNAs, we were able to determine that the cell cycle arrest in the late G1 is beneficial for SARS-CoV-2 replication. This cell cycle arrest is independent of p53 but is dependent on the CDC25A-CDK2/cyclin E pathway. These data give a new understanding in SARS-CoV-2 pathogenesis and highlight some possible targets for the development of novel therapeutic approaches.

8.
Mol Ther ; 32(2): 540-555, 2024 Feb 07.
Article de Anglais | MEDLINE | ID: mdl-38213030

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific neutralizing antibodies (NAbs) lack cross-reactivity between SARS-CoV species and variants and fail to mediate long-term protection against infection. The maintained protection against severe disease and death by vaccination suggests a role for cross-reactive T cells. We generated vaccines containing sequences from the spike or receptor binding domain, the membrane and/or nucleoprotein that induced only T cells, or T cells and NAbs, to understand their individual roles. In three models with homologous or heterologous challenge, high levels of vaccine-induced SARS-CoV-2 NAbs protected against neither infection nor mild histological disease but conferred rapid viral control limiting the histological damage. With no or low levels of NAbs, vaccine-primed T cells, in mice mainly CD8+ T cells, partially controlled viral replication and promoted NAb recall responses. T cells failed to protect against histological damage, presumably because of viral spread and subsequent T cell-mediated killing. Neither vaccine- nor infection-induced NAbs seem to provide long-lasting protective immunity against SARS-CoV-2. Thus, a more realistic approach for universal SARS-CoV-2 vaccines should be to aim for broadly cross-reactive NAbs in combination with long-lasting highly cross-reactive T cells. Long-lived cross-reactive T cells are likely key to prevent severe disease and fatalities during current and future pandemics.


Sujet(s)
Anticorps neutralisants , Vaccins contre la COVID-19 , COVID-19 , Animaux , Humains , Souris , Anticorps antiviraux , Lymphocytes T CD8+ , COVID-19/prévention et contrôle , Vaccins contre la COVID-19/effets indésirables , SARS-CoV-2 , Vaccins antiviraux
9.
Nat Commun ; 14(1): 6785, 2023 10 25.
Article de Anglais | MEDLINE | ID: mdl-37880247

RÉSUMÉ

Marburg and Ebola filoviruses are two of the deadliest infectious agents and several outbreaks have occurred in the last decades. Although several receptors and co-receptors have been reported for Ebola virus, key host factors remain to be elucidated. In this study, using a haploid cell screening platform, we identify the guanine nucleotide exchange factor CCZ1 as a key host factor in the early stage of filovirus replication. The critical role of CCZ1 for filovirus infections is validated in 3D primary human hepatocyte cultures and human blood-vessel organoids, both critical target sites for Ebola and Marburg virus tropism. Mechanistically, CCZ1 controls early to late endosomal trafficking of these viruses. In addition, we report that CCZ1 has a role in the endosomal trafficking of endocytosis-dependent SARS-CoV-2 infections, but not in infections by Lassa virus, which enters endo-lysosomal trafficking at the late endosome stage. Thus, we have identified an essential host pathway for filovirus infections in cell lines and engineered human target tissues. Inhibition of CCZ1 nearly completely abolishes Marburg and Ebola infections. Thus, targeting CCZ1 could potentially serve as a promising drug target for controlling infections caused by various viruses, such as SARS-CoV-2, Marburg, and Ebola.


Sujet(s)
Ebolavirus , Fièvre hémorragique à virus Ebola , Maladie de Marbourg , Marburgvirus , Protéines du transport vésiculaire , Animaux , Humains , Ebolavirus/métabolisme , Lysosomes , Maladie de Marbourg/génétique , Maladie de Marbourg/métabolisme , Marburgvirus/métabolisme , Protéines du transport vésiculaire/métabolisme
10.
Proc Natl Acad Sci U S A ; 120(37): e2304722120, 2023 09 12.
Article de Anglais | MEDLINE | ID: mdl-37669378

RÉSUMÉ

Crimean-Congo hemorrhagic fever (CCHF) caused by CCHF virus (CCHFV) is one of the epidemic-prone diseases prioritized by the World Health Organisation as public health emergency with an urgent need for accelerated research. The trajectory of host response against CCHFV is multifarious and remains unknown. Here, we reported the temporal spectrum of pathogenesis following the CCHFV infection using genome-wide blood transcriptomics analysis followed by advanced systems biology analysis, temporal immune-pathogenic alterations, and context-specific progressive and postinfection genome-scale metabolic models (GSMM) on samples collected during the acute (T0), early convalescent (T1), and convalescent-phase (T2). The interplay between the retinoic acid-inducible gene-I-like/nucleotide-binding oligomerization domain-like receptor and tumor necrosis factor signaling governed the trajectory of antiviral immune responses. The rearrangement of intracellular metabolic fluxes toward the amino acid metabolism and metabolic shift toward oxidative phosphorylation and fatty acid oxidation during acute CCHFV infection determine the pathogenicity. The upregulation of the tricarboxylic acid cycle during CCHFV infection, compared to the noninfected healthy control and between the severity groups, indicated an increased energy demand and cellular stress. The upregulation of glycolysis and pyruvate metabolism potentiated energy generation through alternative pathways associated with the severity of the infection. The downregulation of metabolic processes at the convalescent phase identified by blood cell transcriptomics and single-cell type proteomics of five immune cells (CD4+ and CD8+ T cells, CD14+ monocytes, B cells, and NK cells) potentially leads to metabolic rewiring through the recovery due to hyperactivity during the acute phase leading to post-viral fatigue syndrome.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Humains , Lymphocytes T CD8+ , Régulation positive , Métabolome
11.
Vaccine ; 41(32): 4743-4751, 2023 07 19.
Article de Anglais | MEDLINE | ID: mdl-37353452

RÉSUMÉ

Targeting the site of infection is a promising strategy for improving vaccine effectivity. To date, licensed COVID-19 vaccines have been administered intramuscularly despite the fact that SARS-CoV-2 is a respiratory virus. Here, we aim to induce local protective mucosal immune responses with an inhaled subunit vaccine candidate, ISR52, based on the SARS-CoV-2 Spike S1 protein. When tested in a lethal challenge hACE2 transgenic SARS-CoV-2 mouse model, intranasal and intratracheal administration of ISR52 provided superior protection against severe infection, compared to the subcutaneous injection of the vaccine. Interestingly for a protein-based vaccine, inhaled ISR52 elicited both CD4 and CD8 T-cell Spike-specific responses that were maintained for at least 6 months in wild-type mice. Induced IgG and IgA responses cross-reacting with several SARS- CoV-2 variants of concern were detected in the lung and in serum and protected animals displayed neutralizing antibodies. Based on our results, we are developing ISR52 as a dry powder formulation for inhalation, that does not require cold-chain distribution or the use of needle administration, for evaluation in a Phase I/II clinical trial.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/immunologie , Administration par inhalation , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Animaux , Souris , Réactions croisées , COVID-19/prévention et contrôle , Souris transgéniques , Anticorps antiviraux/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Poudres , Femelle
12.
Life Sci Alliance ; 6(7)2023 07.
Article de Anglais | MEDLINE | ID: mdl-37072184

RÉSUMÉ

Viruses with an RNA genome are often the cause of zoonotic infections. In order to identify novel pro-viral host cell factors, we screened a haploid insertion-mutagenized mouse embryonic cell library for clones that are resistant to Rift Valley fever virus (RVFV). This screen returned the low-density lipoprotein receptor-related protein 1 (LRP1) as a top hit, a plasma membrane protein involved in a wide variety of cell activities. Inactivation of LRP1 in human cells reduced RVFV RNA levels already at the attachment and entry stages of infection. Moreover, the role of LRP1 in promoting RVFV infection was dependent on physiological levels of cholesterol and on endocytosis. In the human cell line HuH-7, LRP1 also promoted early infection stages of sandfly fever Sicilian virus and La Crosse virus, but had a minor effect on late infection by vesicular stomatitis virus, whereas encephalomyocarditis virus was entirely LRP1-independent. Moreover, siRNA experiments in human Calu-3 cells demonstrated that also SARS-CoV-2 infection benefitted from LRP1. Thus, we identified LRP1 as a host factor that supports infection by a spectrum of RNA viruses.


Sujet(s)
COVID-19 , Virus de la fièvre de la vallée du Rift , Animaux , Humains , Souris , Protéine-1 apparentée au récepteur des LDL/génétique , Protéine-1 apparentée au récepteur des LDL/métabolisme , SARS-CoV-2/génétique , Virus de la fièvre de la vallée du Rift/génétique , Virus de la fièvre de la vallée du Rift/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Lipoprotéines LDL/métabolisme
14.
Mol Ther ; 31(2): 387-397, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36184852

RÉSUMÉ

Crimean-Congo hemorrhagic fever virus (CCHFV) is widely distributed throughout Africa, the Middle East, Southern Asia, and Southern and Eastern Europe. Spread by Hyalomma ticks or by contact with infected animals, CCHF begins non-specifically but can rapidly progress to severe, sometimes fatal, disease. Due to the non-specific early symptoms and often unrecognized infections, patients often present to healthcare systems exhibiting later stages of disease, when treatment is limited to supportive care. Consequently, simple vaccines are critically needed to protect populations at risk of CCHFV infection. Currently, there are no widely approved vaccines for CCHFV. We have previously reported significant efficacy of a three-dose DNA-based vaccination regimen for CCHFV in cynomolgus macaques (Macaca fasicularis). Here, we show that in cynomolgus macaques, plasmid-expressed CCHFV nucleoprotein (NP) and glycoprotein precursor (GPC) antigens elicit primarily humoral and cellular immunity, respectively. We found that a two-dose vaccination regimen with plasmids expressing the NP and GPC provides significant protection against CCHFV infection. Studies investigating vaccinations with either antigen alone showed that plasmid-expressed NPs could also confer protection. Cumulatively, our data show that this vaccine confers robust protection against CCHFV and suggest that both humoral and cellular immunity contribute to optimal vaccine-mediated protection.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Vaccins à ADN , Animaux , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Fièvre hémorragique de Crimée-Congo/prévention et contrôle , Fièvre hémorragique de Crimée-Congo/diagnostic , Macaca , Vaccination
15.
Nat Commun ; 13(1): 7926, 2022 12 24.
Article de Anglais | MEDLINE | ID: mdl-36566234

RÉSUMÉ

Recent waves of COVID-19 correlate with the emergence of the Delta and the Omicron variant. We report that the Spike trimer acts as a highly dynamic molecular caliper, thereby forming up to three tight bonds through its RBDs with ACE2 expressed on the cell surface. The Spike of both Delta and Omicron (B.1.1.529) Variant enhance and markedly prolong viral attachment to the host cell receptor ACE2, as opposed to the early Wuhan-1 isolate. Delta Spike shows rapid binding of all three Spike RBDs to three different ACE2 molecules with considerably increased bond lifetime when compared to the reference strain, thereby significantly amplifying avidity. Intriguingly, Omicron (B.1.1.529) Spike displays less multivalent bindings to ACE2 molecules, yet with a ten time longer bond lifetime than Delta. Delta and Omicron (B.1.1.529) Spike variants enhance and prolong viral attachment to the host, which likely not only increases the rate of viral uptake, but also enhances the resistance of the variants against host-cell detachment by shear forces such as airflow, mucus or blood flow. We uncover distinct binding mechanisms and strategies at single-molecule resolution, employed by circulating SARS-CoV-2 variants to enhance infectivity and viral transmission.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , SARS-CoV-2 , Imagerie de molécules uniques , Glycoprotéine de spicule des coronavirus , Humains , Angiotensin-converting enzyme 2/métabolisme , COVID-19/virologie , SARS-CoV-2/métabolisme , SARS-CoV-2/physiologie , Glycoprotéine de spicule des coronavirus/métabolisme , Attachement viral
16.
EMBO Mol Med ; 14(10): e15821, 2022 10 10.
Article de Anglais | MEDLINE | ID: mdl-35986481

RÉSUMÉ

New variants in the SARS-CoV-2 pandemic are more contagious (Alpha/Delta), evade neutralizing antibodies (Beta), or both (Omicron). This poses a challenge in vaccine development according to WHO. We designed a more universal SARS-CoV-2 DNA vaccine containing receptor-binding domain loops from the huCoV-19/WH01, the Alpha, and the Beta variants, combined with the membrane and nucleoproteins. The vaccine induced spike antibodies crossreactive between huCoV-19/WH01, Beta, and Delta spike proteins that neutralized huCoV-19/WH01, Beta, Delta, and Omicron virus in vitro. The vaccine primed nucleoprotein-specific T cells, unlike spike-specific T cells, recognized Bat-CoV sequences. The vaccine protected mice carrying the human ACE2 receptor against lethal infection with the SARS-CoV-2 Beta variant. Interestingly, priming of cross-reactive nucleoprotein-specific T cells alone was 60% protective, verifying observations from humans that T cells protect against lethal disease. This SARS-CoV vaccine induces a uniquely broad and functional immunity that adds to currently used vaccines.


Sujet(s)
COVID-19 , Vaccins à ADN , Vaccins antiviraux , Angiotensin-converting enzyme 2/génétique , Animaux , Anticorps neutralisants , Anticorps antiviraux , COVID-19/prévention et contrôle , Vaccins contre la COVID-19 , Humains , Souris , Nucléoprotéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/génétique , Lymphocytes T , Vaccins à ADN/génétique , Protéines de l'enveloppe virale/composition chimique , Protéines de l'enveloppe virale/génétique , Vaccins antiviraux/génétique
17.
EMBO Mol Med ; 14(8): e15230, 2022 08 08.
Article de Anglais | MEDLINE | ID: mdl-35781796

RÉSUMÉ

The recent emergence of multiple SARS-CoV-2 variants has caused considerable concern due to both reduced vaccine efficacy and escape from neutralizing antibody therapeutics. It is, therefore, paramount to develop therapeutic strategies that inhibit all known and future SARS-CoV-2 variants. Here, we report that all SARS-CoV-2 variants analyzed, including variants of concern (VOC) Alpha, Beta, Gamma, Delta, and Omicron, exhibit enhanced binding affinity to clinical grade and phase 2 tested recombinant human soluble ACE2 (APN01). Importantly, soluble ACE2 neutralized infection of VeroE6 cells and human lung epithelial cells by all current VOC strains with markedly enhanced potency when compared to reference SARS-CoV-2 isolates. Effective inhibition of infections with SARS-CoV-2 variants was validated and confirmed in two independent laboratories. These data show that SARS-CoV-2 variants that have emerged around the world, including current VOC and several variants of interest, can be inhibited by soluble ACE2, providing proof of principle of a pan-SARS-CoV-2 therapeutic.


Sujet(s)
Angiotensin-converting enzyme 2 , Traitements médicamenteux de la COVID-19 , Humains , Peptidyl-Dipeptidase A/génétique , Peptidyl-Dipeptidase A/métabolisme , SARS-CoV-2
18.
Pathogens ; 11(5)2022 May 22.
Article de Anglais | MEDLINE | ID: mdl-35631123

RÉSUMÉ

Bluetongue virus (BTV), an arbovirus of ruminants, is a causative agent of numerous epidemics around the world. Due to the emergence of novel reassortant BTV strains and new outbreaks, there is an unmet need for efficacious antivirals. In this study, we used an improved haploid screening platform to identify the relevant host factors for BTV infection. Our screening tool identified and validated the host factor Niemann-Pick C1 (NPC1), a lysosomal membrane protein that is involved in lysosomal cholesterol transport, as a critical factor in BTV infection. This finding prompted us to investigate the possibility of testing imipramine, an antidepressant drug known to inhibit NPC1 function by interfering with intracellular cholesterol trafficking. In this study, we evaluated the sensitivity of BTV to imipramine using in vitro assays. Our results demonstrate that imipramine pretreatment inhibited in vitro replication and progeny release of BTV-4, BTV-8, and BTV-16. Collectively, our findings highlight the importance of NPC1 for BTV infection and recommend the reprofiling of imipramine as a potential antiviral drug against BTV.

19.
Cell Metab ; 34(6): 857-873.e9, 2022 06 07.
Article de Anglais | MEDLINE | ID: mdl-35561674

RÉSUMÉ

It is not well understood why diabetic individuals are more prone to develop severe COVID-19. To this, we here established a human kidney organoid model promoting early hallmarks of diabetic kidney disease development. Upon SARS-CoV-2 infection, diabetic-like kidney organoids exhibited higher viral loads compared with their control counterparts. Genetic deletion of the angiotensin-converting enzyme 2 (ACE2) in kidney organoids under control or diabetic-like conditions prevented viral detection. Moreover, cells isolated from kidney biopsies from diabetic patients exhibited altered mitochondrial respiration and enhanced glycolysis, resulting in higher SARS-CoV-2 infections compared with non-diabetic cells. Conversely, the exposure of patient cells to dichloroacetate (DCA), an inhibitor of aerobic glycolysis, resulted in reduced SARS-CoV-2 infections. Our results provide insights into the identification of diabetic-induced metabolic programming in the kidney as a critical event increasing SARS-CoV-2 infection susceptibility, opening the door to the identification of new interventions in COVID-19 pathogenesis targeting energy metabolism.


Sujet(s)
Angiotensin-converting enzyme 2/métabolisme , COVID-19 , Diabète , Néphropathies diabétiques , Humains , Rein/métabolisme , Organoïdes , Peptidyl-Dipeptidase A/génétique , Peptidyl-Dipeptidase A/métabolisme , SARS-CoV-2
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...