Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nature ; 600(7890): 701-706, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34673755

RESUMO

Following severe adverse reactions to the AstraZeneca ChAdOx1-S-nCoV-19 vaccine1,2, European health authorities recommended that patients under the age of 55 years who received one dose of ChAdOx1-S-nCoV-19 receive a second dose of the Pfizer BNT162b2 vaccine as a booster. However, the effectiveness and the immunogenicity of this vaccination regimen have not been formally tested. Here we show that the heterologous ChAdOx1-S-nCoV-19 and BNT162b2 combination confers better protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection than the homologous BNT162b2 and BNT162b2 combination in a real-world observational study of healthcare workers (n = 13,121). To understand the underlying mechanism, we conducted a longitudinal survey of the anti-spike immunity conferred by each vaccine combination. Both combinations induced strong anti-spike antibody responses, but sera from heterologous vaccinated individuals displayed a stronger neutralizing activity regardless of the SARS-CoV-2 variant. This enhanced neutralizing potential correlated with increased frequencies of switched and activated memory B cells that recognize the SARS-CoV-2 receptor binding domain. The ChAdOx1-S-nCoV-19 vaccine induced a weaker IgG response but a stronger T cell response than the BNT162b2 vaccine after the priming dose, which could explain the complementarity of both vaccines when used in combination. The heterologous vaccination regimen could therefore be particularly suitable for immunocompromised individuals.


Assuntos
Vacina BNT162/administração & dosagem , Vacina BNT162/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , ChAdOx1 nCoV-19/administração & dosagem , ChAdOx1 nCoV-19/imunologia , SARS-CoV-2/imunologia , Vacinação/estatística & dados numéricos , Adulto , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Feminino , França/epidemiologia , Hospitais Universitários , Humanos , Memória Imunológica/imunologia , Incidência , Masculino , Células B de Memória/imunologia , Células T de Memória/imunologia , Pessoa de Meia-Idade , Glicoproteína da Espícula de Coronavírus/imunologia
2.
Traffic ; 23(1): 63-80, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34729868

RESUMO

Lipid droplets (LDs) are involved in viral infections, but exactly how remains unclear. Here, we study the hepatitis C virus (HCV) whose core capsid protein binds to LDs but is also involved in the assembly of virions at the endoplasmic reticulum (ER) bilayer. We found that the amphipathic helix-containing domain of core, D2, senses triglycerides (TGs) rather than LDs per se. In the absence of LDs, D2 can bind to the ER membrane but only if TG molecules are present in the bilayer. Accordingly, the pharmacological inhibition of the diacylglycerol O-acyltransferase enzymes, mediating TG synthesis in the ER, inhibits D2 association with the bilayer. We found that TG molecules enable D2 to fold into alpha helices. Sequence analysis reveals that D2 resembles the apoE lipid-binding region. Our data support that TG in LDs promotes the folding of core, which subsequently relocalizes to contiguous ER regions. During this motion, core may carry TG molecules to these regions where HCV lipoviroparticles likely assemble. Consistent with this model, the inhibition of Arf1/COPI, which decreases LD surface accessibility to proteins and ER-LD material exchange, severely impedes the assembly of virions. Altogether, our data uncover a critical function of TG in the folding of core and HCV replication and reveals, more broadly, how TG accumulation in the ER may provoke the binding of soluble amphipathic helix-containing proteins to the ER bilayer.


Assuntos
Retículo Endoplasmático , Hepatite C , Retículo Endoplasmático/metabolismo , Hepacivirus/fisiologia , Hepatite C/metabolismo , Humanos , Gotículas Lipídicas/metabolismo , Triglicerídeos/metabolismo , Proteínas do Core Viral/metabolismo
3.
J Biol Chem ; 296: 100111, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33229438

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a ß-coronavirus, is the causative agent of the COVID-19 pandemic. Like for other coronaviruses, its particles are composed of four structural proteins: spike (S), envelope (E), membrane (M), and nucleoprotein (N) proteins. The involvement of each of these proteins and their interactions are critical for assembly and production of ß-coronavirus particles. Here, we sought to characterize the interplay of SARS-CoV-2 structural proteins during the viral assembly process. By combining biochemical and imaging assays in infected versus transfected cells, we show that E and M regulate intracellular trafficking of S as well as its intracellular processing. Indeed, the imaging data reveal that S is relocalized at endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) or Golgi compartments upon coexpression of E or M, as observed in SARS-CoV-2-infected cells, which prevents syncytia formation. We show that a C-terminal retrieval motif in the cytoplasmic tail of S is required for its M-mediated retention in the ERGIC, whereas E induces S retention by modulating the cell secretory pathway. We also highlight that E and M induce a specific maturation of N-glycosylation of S, independently of the regulation of its localization, with a profile that is observed both in infected cells and in purified viral particles. Finally, we show that E, M, and N are required for optimal production of virus-like-particles. Altogether, these results highlight how E and M proteins may influence the properties of S proteins and promote the assembly of SARS-CoV-2 viral particles.


Assuntos
Proteínas do Envelope de Coronavírus/genética , Proteínas do Nucleocapsídeo/genética , SARS-CoV-2/crescimento & desenvolvimento , Glicoproteína da Espícula de Coronavírus/genética , Proteínas da Matriz Viral/genética , Vírion/crescimento & desenvolvimento , Montagem de Vírus/fisiologia , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteínas do Envelope de Coronavírus/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Retículo Endoplasmático/virologia , Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Complexo de Golgi/virologia , Células HEK293 , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas do Nucleocapsídeo/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Proteínas da Matriz Viral/metabolismo , Vírion/genética , Vírion/metabolismo , Internalização do Vírus , Liberação de Vírus/fisiologia
4.
PLoS Pathog ; 13(12): e1006774, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29253880

RESUMO

Viroporins are small transmembrane proteins with ion channel activities modulating properties of intracellular membranes that have diverse proviral functions. Hepatitis C virus (HCV) encodes a viroporin, p7, acting during assembly, envelopment and secretion of viral particles (VP). HCV p7 is released from the viral polyprotein through cleavage at E2-p7 and p7-NS2 junctions by signal peptidase, but also exists as an E2p7 precursor, of poorly defined properties. Here, we found that ectopic p7 expression in HCVcc-infected cells reduced secretion of particle-associated E2 glycoproteins. Using biochemical assays, we show that p7 dose-dependently slows down the ER-to-Golgi traffic, leading to intracellular retention of E2, which suggested that timely E2p7 cleavage and p7 liberation are critical events to control E2 levels. By studying HCV mutants with accelerated E2p7 processing, we demonstrate that E2p7 cleavage controls E2 intracellular expression and secretion levels of nucleocapsid-free subviral particles and infectious virions. In addition, our imaging data reveal that, following p7 liberation, the amino-terminus of p7 is exposed towards the cytosol and coordinates the encounter between NS5A and NS2-based assembly sites loaded with E1E2 glycoproteins, which subsequently leads to nucleocapsid envelopment. We identify punctual mutants at p7 membrane interface that, by abrogating NS2/NS5A interaction, are defective for transmission of infectivity owing to decreased secretion of core and RNA and to increased secretion of non/partially-enveloped particles. Altogether, our results indicate that the retarded E2p7 precursor cleavage is essential to regulate the intracellular and secreted levels of E2 through p7-mediated modulation of the cell secretory pathway and to unmask critical novel assembly functions located at p7 amino-terminus.


Assuntos
Hepacivirus/fisiologia , Hepacivirus/patogenicidade , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Células HEK293 , Hepacivirus/genética , Hepatite C/etiologia , Hepatite C/virologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Modelos Biológicos , Mutação , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/fisiologia , Proteínas Virais/química , Proteínas Virais/genética , Virulência/genética , Virulência/fisiologia , Montagem de Vírus/genética , Montagem de Vírus/fisiologia
5.
Gastroenterology ; 152(4): 895-907.e14, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27932311

RESUMO

BACKGROUND & AIMS: Daclatasvir is a direct-acting antiviral agent and potent inhibitor of NS5A, which is involved in replication of the hepatitis C virus (HCV) genome, presumably via membranous web shaping, and assembly of new virions, likely via transfer of the HCV RNA genome to viral particle assembly sites. Daclatasvir inhibits the formation of new membranous web structures and, ultimately, of replication complex vesicles, but also inhibits an early assembly step. We investigated the relationship between daclatasvir-induced clustering of HCV proteins, intracellular localization of viral RNAs, and inhibition of viral particle assembly. METHODS: Cell-culture-derived HCV particles were produced from Huh7.5 hepatocarcinoma cells in presence of daclatasvir for short time periods. Infectivity and production of physical particles were quantified and producer cells were subjected to subcellular fractionation. Intracellular colocalization between core, E2, NS5A, NS4B proteins, and viral RNAs was quantitatively analyzed by confocal microscopy and by structured illumination microscopy. RESULTS: Short exposure of HCV-infected cells to daclatasvir reduced viral assembly and induced clustering of structural proteins with non-structural HCV proteins, including core, E2, NS4B, and NS5A. These clustered structures appeared to be inactive assembly platforms, likely owing to loss of functional connection with replication complexes. Daclatasvir greatly reduced delivery of viral genomes to these core clusters without altering HCV RNA colocalization with NS5A. In contrast, daclatasvir neither induced clustered structures nor inhibited HCV assembly in cells infected with a daclatasvir-resistant mutant (NS5A-Y93H), indicating that daclatasvir targets a mutual, specific function of NS5A inhibiting both processes. CONCLUSIONS: In addition to inhibiting replication complex biogenesis, daclatasvir prevents viral assembly by blocking transfer of the viral genome to assembly sites. This leads to clustering of HCV proteins because viral particles and replication complex vesicles cannot form or egress. This dual mode of action of daclatasvir could explain its efficacy in blocking HCV replication in cultured cells and in treatment of patients with HCV infection.


Assuntos
Antivirais/farmacologia , Genoma Viral , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Imidazóis/farmacologia , Transporte de RNA/efeitos dos fármacos , RNA Viral/metabolismo , Carbamatos , Linhagem Celular Tumoral , Hepacivirus/fisiologia , Humanos , Transporte Proteico/efeitos dos fármacos , Pirrolidinas , Valina/análogos & derivados , Proteínas do Core Viral/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos
6.
J Virol ; 91(6)2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28053108

RESUMO

The assembly of hepatitis C virus (HCV), a complicated process in which many viral and cellular factors are involved, has not been thoroughly deciphered. NS3 is a multifunctional protein that contains an N-terminal amphipathic α helix (designated helix α0), which is crucial for the membrane association and stability of NS3 protein, followed by a serine protease domain and a C-terminal helicase/NTPase domain. NS3 participates in HCV assembly likely through its C-terminal helicase domain, in which all reported adaptive mutations enhancing virion assembly reside. In this study, we determined that the N-terminal helix α0 of NS3 may contribute to HCV assembly. We identified a single mutation from methionine to threonine at amino acid position 21 (M21T) in helix α0, which significantly promoted viral production while having no apparent effect on the membrane association and protease activity of NS3. Subsequent analyses demonstrated that the M21T mutation did not affect HCV genome replication but rather promoted virion assembly. Further study revealed a shift in the subcellular localization of core protein from lipid droplets (LD) to the endoplasmic reticulum (ER). Finally, we showed that the M21T mutation increased the colocalization of core proteins and viral envelope proteins, leading to a more efficient envelopment of viral nucleocapsids. Collectively, the results of our study revealed a new function of NS3 helix α0 and aid understanding of the role of NS3 in HCV virion morphogenesis.IMPORTANCE HCV NS3 protein possesses the protease activity in its N-terminal domain and the helicase activity in its C-terminal domain. The role of NS3 in virus assembly has been mainly attributed to its helicase domain, because all adaptive mutations enhancing progeny virus production are found to be within this domain. Our study identified, for the first time to our knowledge, an adaptive mutation within the N-terminal helix α0 domain of NS3 that significantly enhanced virus assembly while having no effect on viral genome replication. The mechanistic studies suggested that this mutation promoted the relocation of core proteins from LD to the ER, leading to a more efficient envelopment of viral nucleocapsids. Our results revealed a possible new function of helix α0 in the HCV life cycle and provided new clues to understanding the molecular mechanisms for the action of NS3 in HCV assembly.


Assuntos
Retículo Endoplasmático/virologia , Hepacivirus/fisiologia , Mutação Puntual , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus , Liberação de Vírus , Linhagem Celular , Hepacivirus/genética , Hepatócitos/virologia , Humanos , Metionina/genética , Metionina/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Treonina/genética , Treonina/metabolismo , Proteínas não Estruturais Virais/genética
7.
Emerg Microbes Infect ; 13(1): 2348508, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38661085

RESUMO

The Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne bunyavirus that causes high mortality in humans. This enveloped virus harbors two surface glycoproteins (GP), Gn and Gc, that are released by processing of a glycoprotein precursor complex whose maturation takes place in the ER and is completed through the secretion pathway. Here, we characterized the trafficking network exploited by CCHFV GPs during viral assembly, envelopment, and/or egress. We identified membrane trafficking motifs in the cytoplasmic domains (CD) of CCHFV GPs and addressed how they impact these late stages of the viral life cycle using infection and biochemical assays, and confocal microscopy in virus-producing cells. We found that several of the identified CD motifs modulate GP transport through the retrograde trafficking network, impacting envelopment and secretion of infectious particles. Finally, we identified PACS-2 as a crucial host factor contributing to CCHFV GPs trafficking required for assembly and release of viral particles.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Transporte Proteico , Montagem de Vírus , Humanos , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Animais , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Domínios Proteicos , Motivos de Aminoácidos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Chlorocebus aethiops , Células HEK293 , Células Vero
8.
Nat Commun ; 15(1): 4542, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38806525

RESUMO

The Crimean-Congo hemorrhagic fever virus (CCHFV) is an emerging pathogen of the Orthonairovirus genus that can cause severe and often lethal hemorrhagic diseases in humans. CCHFV has a broad tropism and can infect a variety of species and tissues. Here, by using gene silencing, blocking antibodies or soluble receptor fragments, we identify the low-density lipoprotein receptor (LDL-R) as a CCHFV entry factor. The LDL-R facilitates binding of CCHFV particles but does not allow entry of Hazara virus (HAZV), another member of the genus. In addition, we show that apolipoprotein E (apoE), an exchangeable protein that mediates LDL/LDL-R interaction, is incorporated on CCHFV particles, though not on HAZV particles, and enhances their specific infectivity by promoting an LDL-R dependent entry. Finally, we show that molecules that decrease LDL-R from the surface of target cells could inhibit CCHFV infection. Our study highlights that CCHFV takes advantage of a lipoprotein receptor and recruits its natural ligand to promote entry into cells.


Assuntos
Apolipoproteínas E , Vírus da Febre Hemorrágica da Crimeia-Congo , Receptores de LDL , Internalização do Vírus , Humanos , Receptores de LDL/metabolismo , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética , Vírus da Febre Hemorrágica da Crimeia-Congo/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Animais , Células HEK293 , Chlorocebus aethiops , Febre Hemorrágica da Crimeia/virologia , Febre Hemorrágica da Crimeia/metabolismo , Vírion/metabolismo , Células Vero
9.
PLoS Pathog ; 7(7): e1002144, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21814513

RESUMO

Hepatitis C virus (HCV) assembly remains a poorly understood process. Lipid droplets (LDs) are thought to act as platforms for the assembly of viral components. The JFH1 HCV strain replicates and assembles in association with LD-associated membranes, around which viral core protein is predominantly detected. In contrast, despite its intrinsic capacity to localize to LDs when expressed individually, we found that the core protein of the high-titer Jc1 recombinant virus was hardly detected on LDs of cell culture-grown HCV (HCVcc)-infected cells, but was mainly localized at endoplasmic reticulum (ER) membranes where it colocalized with the HCV envelope glycoproteins. Furthermore, high-titer cell culture-adapted JFH1 virus, obtained after long-term culture in Huh7.5 cells, exhibited an ER-localized core in contrast to non-adapted JFH1 virus, strengthening the hypothesis that ER localization of core is required for efficient HCV assembly. Our results further indicate that p7 and NS2 are HCV strain-specific factors that govern the recruitment of core protein from LDs to ER assembly sites. Indeed, using expression constructs and HCVcc recombinant genomes, we found that p7 is sufficient to induce core localization at the ER, independently of its ion-channel activity. Importantly, the combined expression of JFH1 or Jc1 p7 and NS2 induced the same differential core subcellular localization detected in JFH1- vs. Jc1-infected cells. Finally, results obtained by expressing p7-NS2 chimeras between either virus type indicated that compatibilities between the p7 and the first NS2 trans-membrane domains is required to induce core-ER localization and assembly of extra- and intra-cellular infectious viral particles. In conclusion, we identified p7 and NS2 as key determinants governing the subcellular localization of HCV core to LDs vs. ER and required for initiation of the early steps of virus assembly.


Assuntos
Retículo Endoplasmático/metabolismo , Hepacivirus/fisiologia , Proteínas Virais/metabolismo , Montagem de Vírus/fisiologia , Linhagem Celular , Retículo Endoplasmático/genética , Retículo Endoplasmático/virologia , Humanos , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética
10.
mBio ; 14(5): e0154923, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37671888

RESUMO

IMPORTANCE: Assessments of viral stability on surfaces or in body fluids under different environmental conditions and/or temperatures are often performed, as they are key to understanding the routes and parameters of viral transmission and to providing clues on the epidemiology of infections. However, for most viruses, the mechanisms of inactivation vs stability of viral particles remain poorly defined. Although they are structurally diverse, with different compositions, sizes, and shapes, enveloped viruses are generally less stable than non-enveloped viruses, pointing out the role of envelopes themselves in virus lability. In this report, we investigated the properties of hepatitis C virus (HCV) particles with regards to their stability. We found that, compared to alternative enveloped viruses such as Dengue virus (DENV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), hepatitis delta virus (HDV), and Crimean-Congo hemorrhagic fever virus (CCHFV) that infect the liver, HCV particles are intrinsically labile. We determined the mechanisms that drastically alter their specific infectivity through oxidation of their lipids, and we highlighted that they are protected from lipid oxidation by secreted cellular proteins, which can protect their membrane fusion capacity and overall infectivity.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Hepatite C , Humanos , Hepacivirus , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Hepatite C/metabolismo
11.
mBio ; 13(2): e0292321, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35258330

RESUMO

Nup98, an essential component of the nuclear pore that also participates in annulate lamella pore structures localized in the cytosol, is involved in hepatitis C virus (HCV) assembly. Here, we combined confocal microscopy and biochemical assays to study the interplay between Nup98, core (i.e., the HCV capsid protein), and viral genomes. Our results show that in HCV-infected cells, core protein is necessary and sufficient to induce relocalization of Nup98 from annulate lamellae to lipid droplet-apposed areas in which core/NS5A and HCV genomic RNA [(+)RNA] are clustered to promote viral assembly. Furthermore, we found that Nup98 interacts with HCV RNA and that upon Nup98 downregulation, the viral (+)RNA genome was specifically excluded from areas that contain active translating ribosomes and the core and NS5A proteins. Altogether, these results indicate that Nup98 is recruited by HCV core from annulate lamellae to viral assembly sites to locally increase the concentration of (+)RNA genome, which may favor its encapsidation into nascent virions. IMPORTANCE Nup98 is an essential component of the nuclear pore that also participates in annulate lamella pore structures localized in the cytosol. Nup98 is involved in HCV assembly, though its role remains elusive. Here, we show that Nup98 is retrieved from annulate lamellae during HCV infection. We demonstrate that Nup98 interacts with viral genome within infected cells and that these interactions are essential to maintain viral (+)RNAs in subcellular regions promoting viral replication, assembly, and translation. Importantly, we also show that HCV core nucleocapsid protein is the viral component responsible for the retrieval of Nup98 protein from annulate lamellae, hence allowing an enrichment of Nup98 complexed with viral (+)RNAs in core protein-containing areas. Altogether, our results indicate that Nup98 is recruited from annulate lamellae to viral assembly sites by HCV core protein to promote viral assembly, which highlights a novel virus-induced subversion mechanism of nuclear pore complex components.


Assuntos
Hepatite C , Proteínas do Core Viral , Hepacivirus/genética , Humanos , Complexo de Proteínas Formadoras de Poros Nucleares/genética , RNA Viral/genética , RNA Viral/metabolismo , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo , Montagem de Vírus/fisiologia
12.
Vet Med Sci ; 8(1): 14-20, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34704394

RESUMO

Although there are several reports in the literature of SARS-CoV-2 infection in cats, few SARS-CoV-2 sequences from infected cats have been published. In this study, SARS-CoV-2 infection was evaluated in two cats by clinical observation, molecular biology (qPCR and NGS), and serology (microsphere immunoassay and seroneutralization). Following the observation of symptomatic SARS-CoV-2 infection in two cats, infection status was confirmed by RT-qPCR and, in one cat, serological analysis for antibodies against N-protein and S-protein, as well as neutralizing antibodies. Comparative analysis of five SARS-CoV-2 sequence fragments obtained from one of the cats showed that this infection was not with one of the three recently emerged variants of SARS-CoV-2. This study provides additional information on the clinical, molecular, and serological aspects of SARS-CoV-2 infection in cats.


Assuntos
COVID-19 , Doenças do Gato , Animais , COVID-19/veterinária , Doenças do Gato/epidemiologia , Gatos , França/epidemiologia , Pandemias , SARS-CoV-2
13.
Viruses ; 13(9)2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34578341

RESUMO

Despite the probable zoonotic origin of SARS-CoV-2, only limited research efforts have been made to understand the role of companion animals in SARS-CoV-2 epidemiology. According to recent serological prevalence studies, human-to-companion animal transmission is quite frequent, which led us to consider that the risk of SARS-CoV-2 transmission from animal to human, albeit negligible in the present context, may have been underestimated. In this study, we provide the results of a prospective survey that was conducted to evaluate the SARS-CoV-2 isolation rate by qRT-PCR in dogs and cats with different exposure risks and clinical statuses. From April 2020 to April 2021, we analyzed 367 samples and investigated the presence of SARS-CoV-2 RNA using qRT-PCR. Only four animals tested positive, all of them being cats. Three cats were asymptomatic and one presented a coryza-like syndrome. We describe in detail the infection in two cats and the associated clinical characteristics. Importantly, we obtained SARS-CoV-2 genomes from one infected animal and characterized them as Alpha variants. This represents the first identification of the SARS-CoV-2 Alpha variant in an infected animal in France.


Assuntos
COVID-19/veterinária , Doenças do Gato/virologia , Doenças do Cão/virologia , Animais , COVID-19/epidemiologia , COVID-19/transmissão , COVID-19/virologia , Doenças do Gato/epidemiologia , Gatos , Doenças do Cão/epidemiologia , Cães , França/epidemiologia , Humanos , Masculino , Animais de Estimação/virologia , Prevalência , Estudos Prospectivos , RNA Viral , Reação em Cadeia da Polimerase em Tempo Real/veterinária , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Análise de Sequência de RNA , Eliminação de Partículas Virais
14.
Elife ; 102021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34190687

RESUMO

Cell entry of enveloped viruses relies on the fusion between the viral and plasma or endosomal membranes, through a mechanism that is triggered by a cellular signal. Here we used a combination of computational and experimental approaches to unravel the main determinants of hepatitis B virus (HBV) membrane fusion process. We discovered that ERp57 is a host factor critically involved in triggering HBV fusion and infection. Then, through modeling approaches, we uncovered a putative allosteric cross-strand disulfide (CSD) bond in the HBV S glycoprotein and we demonstrate that its stabilization could prevent membrane fusion. Finally, we identified and characterized a potential fusion peptide in the preS1 domain of the HBV L glycoprotein. These results underscore a membrane fusion mechanism that could be triggered by ERp57, allowing a thiol/disulfide exchange reaction to occur and regulate isomerization of a critical CSD, which ultimately leads to the exposition of the fusion peptide.


Assuntos
Antígenos de Superfície da Hepatite B/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Precursores de Proteínas/metabolismo , Proteínas do Envelope Viral/metabolismo , Ligação Viral , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Regulação Viral da Expressão Gênica , Vírus da Hepatite B , Hepatócitos , Humanos , Masculino , Fusão de Membrana , Camundongos , Isomerases de Dissulfetos de Proteínas/genética , Proteínas do Envelope Viral/genética
15.
One Health ; 11: 100192, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33169106

RESUMO

In a survey of household cats and dogs of laboratory-confirmed COVID-19 patients, we found a high seroprevalence of SARS-CoV-2 antibodies, ranging from 21% to 53%, depending on the positivity criteria chosen. Seropositivity was significantly greater among pets from COVID-19+ households compared to those with owners of unknown status. Our results highlight the potential role of pets in the spread of the epidemic.

16.
Cell Mol Immunol ; 18(2): 318-327, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33408342

RESUMO

Understanding the immune responses elicited by SARS-CoV-2 infection is critical in terms of protection against reinfection and, thus, for public health policy and vaccine development for COVID-19. In this study, using either live SARS-CoV-2 particles or retroviruses pseudotyped with the SARS-CoV-2 S viral surface protein (Spike), we studied the neutralizing antibody (nAb) response in serum samples from a cohort of 140 SARS-CoV-2 qPCR-confirmed infections, including patients with mild symptoms and also more severe forms, including those that required intensive care. We show that nAb titers correlated strongly with disease severity and with anti-spike IgG levels. Indeed, patients from intensive care units exhibited high nAb titers; conversely, patients with milder disease symptoms had heterogeneous nAb titers, and asymptomatic or exclusive outpatient-care patients had no or low nAbs. We found that nAb activity in SARS-CoV-2-infected patients displayed a relatively rapid decline after recovery compared to individuals infected with other coronaviruses. Moreover, we found an absence of cross-neutralization between endemic coronaviruses and SARS-CoV-2, indicating that previous infection by human coronaviruses may not generate protective nAbs against SARS-CoV-2. Finally, we found that the D614G mutation in the spike protein, which has recently been identified as the current major variant in Europe, does not allow neutralization escape. Altogether, our results contribute to our understanding of the immune correlates of SARS-CoV-2-induced disease, and rapid evaluation of the role of the humoral response in the pathogenesis of SARS-CoV-2 is warranted.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Teste Sorológico para COVID-19 , COVID-19/imunologia , COVID-19/patologia , SARS-CoV-2/imunologia , Índice de Gravidade de Doença , Adulto , Idoso , Idoso de 80 Anos ou mais , Substituição de Aminoácidos , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/sangue , COVID-19/virologia , Feminino , Humanos , Cinética , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia
17.
Viruses ; 12(4)2020 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-32290553

RESUMO

Hepatitis C virus (HCV) infection is a major public health issue leading to chronic liver diseases. HCV particles are unique owing to their particular lipid composition, namely the incorporation of neutral lipids and apolipoproteins. The mechanism of association between HCV virion components and these lipoproteins factors remains poorly understood as well as its impact in subsequent steps of the viral life cycle, such as entry into cells. It was proposed that the lipoprotein biogenesis pathway is involved in HCV morphogenesis; yet, recent evidence indicated that HCV particles can mature and evolve biochemically in the extracellular medium after egress. In addition, several viral, cellular and blood components have been shown to influence and regulate this specific association. Finally, this specific structure and composition of HCV particles was found to influence entry into cells as well as their stability and sensitivity to neutralizing antibodies. Due to its specific particle composition, studying the association of HCV particles with lipoproteins remains an important goal towards the rational design of a protective vaccine.


Assuntos
Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , Interações Hospedeiro-Patógeno , Lipoproteínas/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Hepatite C/imunologia , Humanos , Metabolismo dos Lipídeos , Lipoproteínas/biossíntese , Transporte Proteico , Transdução de Sinais , Vírion , Montagem de Vírus , Internalização do Vírus
18.
Viruses ; 11(1)2019 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-30621318

RESUMO

Hepatitis C infection is the leading cause of liver diseases worldwide and a major health concern that affects an estimated 3% of the global population. Novel therapies available since 2014 and 2017 are very efficient and the WHO considers HCV eradication possible by the year 2030. These treatments are based on the so-called direct acting antivirals (DAAs) that have been developed through research efforts by academia and industry since the 1990s. After a brief overview of the HCV life cycle, we describe here the functions of the different targets of current DAAs, the mode of action of these DAAs and potential future inhibitors.


Assuntos
Antivirais/farmacologia , Hepacivirus/fisiologia , Hepatite C/tratamento farmacológico , Hepacivirus/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Inibidores de Proteases/farmacologia
19.
Nat Commun ; 10(1): 2098, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31068585

RESUMO

Hepatitis D virus (HDV) doesn't encode envelope proteins for packaging of its ribonucleoprotein (RNP) and typically relies on the surface glycoproteins (GPs) from hepatitis B virus (HBV) for virion assembly, envelopment and cellular transmission. HDV RNA genome can efficiently replicate in different tissues and species, raising the possibility that it evolved, and/or is still able to transmit, independently of HBV. Here we show that alternative, HBV-unrelated viruses can act as helper viruses for HDV. In vitro, envelope GPs from several virus genera, including vesiculovirus, flavivirus and hepacivirus, can package HDV RNPs, allowing efficient egress of HDV particles in the extracellular milieu of co-infected cells and subsequent entry into cells expressing the relevant receptors. Furthermore, HCV can propagate HDV infection in the liver of co-infected humanized mice for several months. Further work is necessary to evaluate whether HDV is currently transmitted by HBV-unrelated viruses in humans.


Assuntos
Coinfecção/transmissão , Hepatite D/transmissão , Vírus Delta da Hepatite/fisiologia , Montagem de Vírus , Animais , Linhagem Celular Tumoral , Coinfecção/virologia , Flavivirus/metabolismo , Hepacivirus/metabolismo , Hepacivirus/patogenicidade , Hepatite D/virologia , Vírus Delta da Hepatite/isolamento & purificação , Vírus Delta da Hepatite/patogenicidade , Hepatócitos/transplante , Hepatócitos/virologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Cultura Primária de Células , RNA Viral/isolamento & purificação , Ribonucleoproteínas/metabolismo , Vesiculovirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
20.
Virol J ; 3: 70, 2006 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-16948862

RESUMO

There is an urgent need to develop novel approaches to vaccination against the emerging, highly pathogenic avian influenza viruses. Here, we engineered influenza viral-like particles (Flu-VLPs) derived from retroviral core particles that mimic the properties of the viral surface of two highly pathogenic influenza viruses of either H7N1 or H5N1 antigenic subtype. We demonstrate that, upon recovery of viral RNAs from a field strain, one can easily generate expression vectors that encode the HA, NA and M2 surface proteins of either virus and prepare high-titre Flu-VLPs. We characterise these Flu-VLPs incorporating the HA, NA and M2 proteins and we show that they induce high-titre neutralising antibodies in mice.


Assuntos
Virus da Influenza A Subtipo H5N1/imunologia , Vírion/isolamento & purificação , Animais , Aves , Influenza Aviária/virologia , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa