Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-37808801

RESUMO

The rapid evolution of SARS-CoV-2 variants highlights the need for new therapies to prevent disease spread. SARS-CoV-2, like SARS-CoV-1, uses the human cell surface protein angiotensin-converting enzyme 2 (ACE2) as its native receptor. Here, we design and characterize a mutant ACE2 that enables rapid affinity purification of a dimeric protein by altering the active site to prevent autoproteolytic digestion of a C-terminal His10 epitope tag. In cultured cells, mutant ACE2 competitively inhibits lentiviral vectors pseudotyped with spike from multiple SARS-CoV-2 variants, and infectious SARS-CoV-2. Moreover, the protein can be nebulized and retains virus-binding properties. We developed a system for delivery of aerosolized ACE2 to K18-hACE2 mice and demonstrate protection by our modified ACE2 when delivered as a prophylactic agent. These results show proof-of-concept for an aerosolized delivery method to evaluate anti-SARS-CoV-2 agents in vivo and suggest a new tool in the ongoing fight against SARS-CoV-2 and other ACE2-dependent viruses.

2.
EMBO Rep ; 24(9): e56901, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37497756

RESUMO

Interferons control viral infection by inducing the expression of antiviral effector proteins encoded by interferon-stimulated genes (ISGs). The field has mostly focused on identifying individual antiviral ISG effectors and defining their mechanisms of action. However, fundamental gaps in knowledge about the interferon response remain. For example, it is not known how many ISGs are required to protect cells from a particular virus, though it is theorized that numerous ISGs act in concert to achieve viral inhibition. Here, we used CRISPR-based loss-of-function screens to identify a markedly limited set of ISGs that confer interferon-mediated suppression of a model alphavirus, Venezuelan equine encephalitis virus (VEEV). We show via combinatorial gene targeting that three antiviral effectors-ZAP, IFIT3, and IFIT1-together constitute the majority of interferon-mediated restriction of VEEV, while accounting for < 0.5% of the interferon-induced transcriptome. Together, our data suggest a refined model of the antiviral interferon response in which a small subset of "dominant" ISGs may confer the bulk of the inhibition of a given virus.


Assuntos
Vírus da Encefalite Equina Venezuelana , Vírus , Animais , Cavalos , Interferons , Linhagem Celular , Replicação Viral , Antivirais/farmacologia , Vírus da Encefalite Equina Venezuelana/fisiologia
3.
Biomed Pharmacother ; 162: 114614, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37068330

RESUMO

The continuing heavy toll of the COVID-19 pandemic necessitates development of therapeutic options. We adopted structure-based drug repurposing to screen FDA-approved drugs for inhibitory effects against main protease enzyme (Mpro) substrate-binding pocket of SARS-CoV-2 for non-covalent and covalent binding. Top candidates were screened against infectious SARS-CoV-2 in a cell-based viral replication assay. Promising candidates included atovaquone, mebendazole, ouabain, dronedarone, and entacapone, although atovaquone and mebendazole were the only two candidates with IC50s that fall within their therapeutic plasma concentration. Additionally, we performed Mpro assays on the top hits, which demonstrated inhibition of Mpro by dronedarone (IC50 18 µM), mebendazole (IC50 19 µM) and entacapone (IC50 9 µM). Atovaquone showed only modest Mpro inhibition, and thus we explored other potential mechanisms. Although atovaquone is Dihydroorotate dehydrogenase (DHODH) inhibitor, we did not observe inhibition of DHODH at the respective SARS-CoV-2 IC50. Metabolomic profiling of atovaquone treated cells showed dysregulation of purine metabolism pathway metabolite, where ecto-5'-nucleotidase (NT5E) was downregulated by atovaquone at concentrations equivalent to its antiviral IC50. Atovaquone and mebendazole are promising candidates with SARS-CoV-2 antiviral activity. While mebendazole does appear to target Mpro, atovaquone may inhibit SARS-CoV-2 viral replication by targeting host purine metabolism.


Assuntos
Antivirais , COVID-19 , Humanos , Antivirais/farmacologia , SARS-CoV-2 , Di-Hidro-Orotato Desidrogenase , Reposicionamento de Medicamentos , Dronedarona/farmacologia , Pandemias , Atovaquona/farmacologia , Mebendazol/farmacologia , Purinas/farmacologia , Simulação de Acoplamento Molecular , Inibidores de Proteases/farmacologia , Simulação de Dinâmica Molecular
4.
bioRxiv ; 2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36865157

RESUMO

Interferons control viral infection by inducing the expression of antiviral effector proteins encoded by interferon-stimulated genes (ISGs). The field has mostly focused on identifying individual antiviral ISG effectors and defining their mechanisms of action. However, fundamental gaps in knowledge about the interferon response remain. For example, it is not known how many ISGs are required to protect cells from a particular virus, though it is theorized that numerous ISGs act in concert to achieve viral inhibition. Here, we used CRISPR-based loss-of-function screens to identify a markedly limited set of ISGs that confer interferon-mediated suppression of a model alphavirus, Venezuelan equine encephalitis virus (VEEV). We show via combinatorial gene targeting that three antiviral effectors - ZAP, IFIT3, and IFIT1 - together constitute the majority of interferon-mediated restriction of VEEV, while accounting for less than 0.5% of the interferon-induced transcriptome. Together, our data suggests a refined model of the antiviral interferon response in which a small subset of "dominant" ISGs may confer the bulk of the inhibition of a given virus.

5.
J Virol ; 96(19): e0133222, 2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36106874

RESUMO

Mammalian TRIM7 is an antiviral protein that inhibits multiple human enteroviruses by degrading the viral 2BC protein. Whether TRIM7 is reciprocally targeted by enteroviruses is not known. Here, we report that the 3C protease (3Cpro) from two enteroviruses, coxsackievirus B3 (CVB3) and poliovirus, targets TRIM7 for cleavage. CVB3 3Cpro cleaves TRIM7 at glutamine 24 (Q24), resulting in a truncated TRIM7 that fails to inhibit CVB3 due to dampened E3 ubiquitin ligase activity. TRIM7 Q24 is highly conserved across mammals, except in marsupials, which instead have a naturally occurring histidine (H24) that is not subject to 3Cpro cleavage. Marsupials also express two isoforms of TRIM7, and the two proteins from koalas have distinct antiviral activities. The longer isoform contains an additional exon due to alternate splice site usage. This additional exon contains a unique 3Cpro cleavage site, suggesting that certain enteroviruses may have evolved to target marsupial TRIM7 even if the canonical Q24 is missing. Combined with computational analyses indicating that TRIM7 is rapidly evolving, our data raise the possibility that TRIM7 may be targeted by enterovirus evasion strategies and that evolution of TRIM7 across mammals may have conferred unique antiviral properties. IMPORTANCE Enteroviruses are significant human pathogens that cause viral myocarditis, pancreatitis, and meningitis. Knowing how the host controls these viruses and how the viruses may evade host restriction is important for understanding fundamental concepts in antiviral immunity and for informing potential therapeutic interventions. In this study, we demonstrate that coxsackievirus B3 uses its virally encoded protease to target the host antiviral protein TRIM7 for cleavage, suggesting a potential mechanism of viral immune evasion. We additionally show that TRIM7 has evolved in certain mammalian lineages to express protein variants with distinct antiviral activities and susceptibilities to viral protease-mediated cleavage.


Assuntos
Proteases Virais 3C , Infecções por Enterovirus , Enterovirus , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Proteases Virais 3C/metabolismo , Animais , Enterovirus/enzimologia , Glutamina , Histidina , Interações Hospedeiro-Patógeno , Phascolarctidae/virologia , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
6.
Curr Opin Virol ; 53: 101202, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35124511

RESUMO

The vertebrate interferon (IFN) response controls viral infections by inducing hundreds of interferon-stimulated genes (ISGs), many of which encode 'restriction factors' that uniquely target certain viruses. ISG studies have historically had a human-centric focus, which is justified because these natural defense mechanisms might be leveraged to treat human viral disease. However, certain mammals are reservoirs for zoonotic viruses that can 'spill over' into humans. Additionally, restriction factors have prominent roles in the ongoing evolutionary genetic conflicts between viruses and their hosts. Thus, there is a growing need to understand antiviral IFN/ISG responses in other species, particularly in known reservoirs of zoonotic viruses. This review focuses on functional and evolutionary insight into antiviral IFN responses that have been obtained from studying non-model mammalian species.


Assuntos
Viroses , Vírus , Animais , Antivirais/farmacologia , Humanos , Interferons , Mamíferos , Viroses/genética , Vírus/genética
7.
Nat Microbiol ; 5(11): 1330-1339, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32704094

RESUMO

Zoonotic coronaviruses (CoVs) are substantial threats to global health, as exemplified by the emergence of two severe acute respiratory syndrome CoVs (SARS-CoV and SARS-CoV-2) and Middle East respiratory syndrome CoV (MERS-CoV) within two decades1-3. Host immune responses to CoVs are complex and regulated in part through antiviral interferons. However, interferon-stimulated gene products that inhibit CoVs are not well characterized4. Here, we show that lymphocyte antigen 6 complex, locus E (LY6E) potently restricts infection by multiple CoVs, including SARS-CoV, SARS-CoV-2 and MERS-CoV. Mechanistic studies revealed that LY6E inhibits CoV entry into cells by interfering with spike protein-mediated membrane fusion. Importantly, mice lacking Ly6e in immune cells were highly susceptible to a murine CoV-mouse hepatitis virus. Exacerbated viral pathogenesis in Ly6e knockout mice was accompanied by loss of hepatic immune cells, higher splenic viral burden and reduction in global antiviral gene pathways. Accordingly, we found that constitutive Ly6e directly protects primary B cells from murine CoV infection. Our results show that LY6E is a critical antiviral immune effector that controls CoV infection and pathogenesis. These findings advance our understanding of immune-mediated control of CoV in vitro and in vivo-knowledge that could help inform strategies to combat infection by emerging CoVs.


Assuntos
Antígenos de Superfície/metabolismo , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Coronavirus/fisiologia , Proteínas Ligadas por GPI/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Betacoronavirus/imunologia , Betacoronavirus/fisiologia , COVID-19 , Coronavirus/imunologia , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Pandemias , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , SARS-CoV-2 , Internalização do Vírus
8.
Nat Microbiol ; 3(11): 1214-1223, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30224801

RESUMO

The endoplasmic reticulum (ER) is an architecturally diverse organelle that serves as a membrane source for the replication of multiple viruses. Flaviviruses, including yellow fever virus, West Nile virus, dengue virus and Zika virus, induce unique single-membrane ER invaginations that house the viral replication machinery1. Whether this virus-induced ER remodelling is vulnerable to antiviral pathways is unknown. Here, we show that flavivirus replication at the ER is targeted by the interferon (IFN) response. Through genome-scale CRISPR screening, we uncovered an antiviral mechanism mediated by a functional gene pairing between IFI6 (encoding IFN-α-inducible protein 6), an IFN-stimulated gene cloned over 30 years ago2, and HSPA5, which encodes the ER-resident heat shock protein 70 chaperone BiP. We reveal that IFI6 is an ER-localized integral membrane effector that is stabilized through interactions with BiP. Mechanistically, IFI6 prophylactically protects uninfected cells by preventing the formation of virus-induced ER membrane invaginations. Notably, IFI6 has little effect on other mammalian RNA viruses, including the related Flaviviridae family member hepatitis C virus, which replicates in double-membrane vesicles that protrude outwards from the ER. These findings support a model in which the IFN response is armed with a membrane-targeted effector that discriminately blocks the establishment of virus-specific ER microenvironments that are required for replication.


Assuntos
Antivirais/farmacologia , Retículo Endoplasmático/metabolismo , Interferon-alfa/farmacologia , Proteínas Mitocondriais/metabolismo , Replicação Viral , Febre Amarela/metabolismo , Vírus da Febre Amarela/efeitos dos fármacos , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Chaperona BiP do Retículo Endoplasmático , Técnicas de Inativação de Genes , Estudo de Associação Genômica Ampla , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Mitocondriais/genética , Ligação Proteica , Especificidade da Espécie , Febre Amarela/virologia , Vírus da Febre Amarela/fisiologia
9.
Nat Commun ; 9(1): 3603, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30190477

RESUMO

Interferons (IFNs) contribute to cell-intrinsic antiviral immunity by inducing hundreds of interferon-stimulated genes (ISGs). In a screen to identify antiviral ISGs, we unexpectedly found that LY6E, a member of the LY6/uPAR family, enhanced viral infection. Here, we show that viral enhancement by ectopically expressed LY6E extends to several cellular backgrounds and affects multiple RNA viruses. LY6E does not impair IFN antiviral activity or signaling, but rather promotes viral entry. Using influenza A virus as a model, we narrow the enhancing effect of LY6E to uncoating after endosomal escape. Diverse mammalian orthologs of LY6E also enhance viral infectivity, indicating evolutionary conservation of function. By structure-function analyses, we identify a single amino acid in a predicted loop region that is essential for viral enhancement. Our study suggests that LY6E belongs to a class of IFN-inducible host factors that enhance viral infectivity without suppressing IFN antiviral activity.


Assuntos
Antígenos de Superfície/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Vírus de RNA/patogenicidade , Animais , Antígenos de Superfície/genética , Evolução Biológica , Linhagem Celular , Fibroblastos/virologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica , Humanos , Vírus da Influenza A/patogenicidade , Interferons/genética , Interferons/metabolismo , Leucina , Infecções por Vírus de RNA/metabolismo , Vírus de RNA/fisiologia , Internalização do Vírus , Replicação Viral , Vírus da Febre Amarela/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...