Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.597
Filtrar
1.
Mol Plant Pathol ; 25(10): e70012, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39350560

RESUMEN

Autophagy, an intracellular degradation process, has emerged as a crucial innate immune response against various plant pathogens, including viruses. Tomato spotted wilt orthotospovirus (TSWV) is a highly destructive plant pathogen that infects over 1000 plant species and poses a significant threat to global food security. However, the role of autophagy in defence against the TSWV pathogen, and whether the virus counteracts this defence, remains unknown. In this study, we report that autophagy plays an important role in antiviral defence against TSWV infection; however, this autophagy-mediated defence is counteracted by the viral effector NSs. Transcriptome profiling revealed the up-regulation of autophagy-related genes (ATGs) upon TSWV infection. Blocking autophagy induction by chemical treatment or knockout/down of ATG5/ATG7 significantly enhanced TSWV accumulation. Notably, the TSWV nucleocapsid (N) protein, a major component of the viral replication unit, strongly induced autophagy. However, the TSWV nonstructural protein NSs was able to effectively suppress N-induced autophagy in a dose-dependent manner. Further investigation revealed that NSs inhibited ATG6-mediated autophagy induction. These findings provide new insights into the defence role of autophagy against TSWV, a representative segmented negative-strand RNA virus, as well as the tospoviral pathogen counterdefence mechanism.


Asunto(s)
Autofagia , Enfermedades de las Plantas , Tospovirus , Tospovirus/fisiología , Tospovirus/patogenicidad , Enfermedades de las Plantas/virología , Enfermedades de las Plantas/inmunología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Solanum lycopersicum/virología , Solanum lycopersicum/inmunología , Solanum lycopersicum/genética , Nicotiana/virología , Nicotiana/inmunología , Nicotiana/genética
2.
J Med Virol ; 96(10): e70007, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39400381

RESUMEN

Nuclear factor κB (NF-κB) plays a crucial role in various cellular processes, including inflammatory and immune responses. Its activation is tightly regulated by the IKK (IκB kinase) complex. Upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the virus is initially recognized by the innate immune system and typically activates the NF-κB pathway, leading to a severe inflammatory response. However, the influence of viral proteins upon pro-inflammatory pathway is complicated. Here, we demonstrated that the viral protein nsp3 of SARS-CoV-2 exhibits an unusual function, which attenuated the NF-κB-mediated inflammatory response against SARS-CoV-2 infection in a unique manner. nsp3 interacted with the essential NF-κB modulator NEMO/IKKγ and promoted its polyubiquitylation via the E3 ubiquitin ligase CBL (Cbl Proto-Oncogene). Consequently, polyubiquitylated NEMO undergoes proteasome-dependent degradation, which disrupts NF-κB activation. Moreover, we found that the SARS unique domain (SUD) in nsp3 of SARS-CoV-2 is essential for inducing NEMO degradation, whereas this function is absent in SUD of SARS-CoV. The reduced activation of pro-inflammatory response at an early stage could mask the host immune response and faciliate excessive viral replication. Conversely, this finding may partially explain why SARS-CoV-2 causes a less inflammatory reaction than SARS-CoV, resulting in more mild or moderate COVID-19 cases and greater transmissibility. Given that NEMO is important for NF-κB activation, we propose that inhibiting polyubiquitylation and degradation of NEMO upon SARS-CoV-2 infection is a novel strategy to modulate the host inflammatory response.


Asunto(s)
COVID-19 , Quinasa I-kappa B , FN-kappa B , SARS-CoV-2 , Humanos , FN-kappa B/metabolismo , SARS-CoV-2/inmunología , COVID-19/inmunología , COVID-19/virología , COVID-19/metabolismo , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/genética , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Transducción de Señal , Células HEK293 , Ubiquitinación , Proto-Oncogenes Mas , Dominios Proteicos , Inmunidad Innata , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética
3.
Cell Mol Life Sci ; 81(1): 433, 2024 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-39395053

RESUMEN

Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an enveloped, positive-sense RNA virus that emerged in 2012, causing sporadic cases and localized outbreaks of severe respiratory illness with high fatality rates. A characteristic feature of the immune response to MERS-CoV infection is low type I IFN induction, despite its importance in viral clearance. The non-structural proteins (nsps) of other coronaviruses have been shown to block IFN production. However, the role of nsp5 from MERS-CoV in IFN induction of human respiratory cells is unclear. In this study, we elucidated the role of MERS-CoV-nsp5, the viral main protease, in modulating the host's antiviral responses in human bronchial epithelial BEAS 2b cells. We found that overexpression of MERS-CoV-nsp5 had a dose-dependent inhibitory effect on IFN-ß promoter activation and cytokine production induced by HMW-poly(I:C). It also suppressed IFN-ß promoter activation triggered by overexpression of key components in the RIG-I-like receptor (RLR) pathway, including RIG-I, MAVS, IKK-ε and IRF3. Moreover, the overexpression of MERS-CoV-nsp5 did not impair expression or phosphorylation of IRF3, but suppressed the nuclear translocation of IRF3. Further investigation revealed that MERS-CoV-nsp5 specifically interacted with IRF3. Using docking and molecular dynamic (MD) simulations, we also found that amino acids on MERS-CoV-nsp5, IRF3, and KPNA4 may participate in protein-protein interactions. Additionally, we uncovered protein conformations that mask the nuclear localization signal (NLS) regions of IRF3 and KPNA4 when interacting with MERS-CoV-nsp5, suggesting a mechanism by which this viral protein blocks IRF3 nuclear translocation. Of note, the IFN-ß expression was restored after administration of protease inhibitors targeting nsp5, indicating this suppression of IFN-ß production was dependent on the enzyme activity of nsp5. Collectively, our findings elucidate a mechanism by which MERS-CoV-nsp5 disrupts the host's innate antiviral immunity and thus provides insights into viral pathogenesis.


Asunto(s)
Células Epiteliales , Factor 3 Regulador del Interferón , Coronavirus del Síndrome Respiratorio de Oriente Medio , Proteínas no Estructurales Virales , Humanos , Factor 3 Regulador del Interferón/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Interferón Tipo I/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Interferón beta/metabolismo , Transducción de Señal/efectos de los fármacos , Poli I-C/farmacología , Regiones Promotoras Genéticas/genética , alfa Carioferinas/metabolismo , alfa Carioferinas/genética , Transporte Activo de Núcleo Celular , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/genética
4.
Nat Commun ; 15(1): 8932, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39414822

RESUMEN

Antibody-dependent enhancement (ADE) is a potential concern for the development of Zika virus (ZIKV) vaccines. Cross-reactive but poorly neutralizing antibodies, usually targeting viral pre-membrane or envelope (E) proteins, can potentially enhance dengue virus (DENV) infection. Although E domain III (EDIII) contains ZIKV-specific epitopes, its immunogenicity is poor. Here, we show that dimeric EDIII, fused to human IgG1 Fc fragment (EDIII-Fc) and encoded by circular RNA (circRNA), induces better germinal center reactions and higher neutralizing antibodies compared to circRNAs encoding monomeric or trimeric EDIII. Two doses of circRNAs encoding EDIII-Fc and ZIKV nonstructural protein NS1, another protective antigen, prevent lethal ZIKV infection in neonates born to immunized C57BL/6 mice and in interferon-α/ß receptor knockout adult C57BL/6 mice. Importantly, a single-dose optimized circRNA vaccine with improved antigen expression confers potent and durable protection without inducing obvious DENV ADE in mice, laying the groundwork for developing flavivirus vaccines based on circRNAs encoding EDIII-Fc and NS1.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Ratones Endogámicos C57BL , ARN Circular , Infección por el Virus Zika , Virus Zika , Animales , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Virus Zika/genética , Ratones , Anticuerpos Neutralizantes/inmunología , ARN Circular/genética , ARN Circular/inmunología , Anticuerpos Antivirales/inmunología , Virus del Dengue/inmunología , Virus del Dengue/genética , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Humanos , Ratones Noqueados , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Dengue/prevención & control , Dengue/inmunología , Femenino , Acrecentamiento Dependiente de Anticuerpo , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Receptor de Interferón alfa y beta/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/genética
5.
PLoS Pathog ; 20(9): e1012480, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39226332

RESUMEN

Norovirus infection is characterised by a rapid onset of disease and the development of debilitating symptoms including projectile vomiting and diffuse diarrhoea. Vaccines and antivirals are sorely lacking and developments in these areas are hampered by the lack of an adequate cell culture system to investigate human norovirus replication and pathogenesis. Herein, we describe how the model norovirus, Mouse norovirus (MNV), produces a viral protein, NS3, with the functional capacity to attenuate host protein translation which invokes the activation of cell death via apoptosis. We show that this function of NS3 is conserved between human and mouse viruses and map the protein domain attributable to this function. Our study highlights a critical viral protein that mediates crucial activities during replication, potentially identifying NS3 as a worthy target for antiviral drug development.


Asunto(s)
Infecciones por Caliciviridae , Macrófagos , Norovirus , Norovirus/fisiología , Animales , Ratones , Infecciones por Caliciviridae/virología , Macrófagos/virología , Macrófagos/metabolismo , Humanos , Biosíntesis de Proteínas , Replicación Viral/fisiología , Muerte Celular/fisiología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Apoptosis
6.
Antimicrob Agents Chemother ; 68(10): e0056224, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39225484

RESUMEN

We have synthesized a novel and highly selective severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease peptide mimetic inhibitor mimicking the replicase 1ab recognition sequence -Val-Leu-Gln- and utilizing a cysteine selective acyloxymethyl ketone as the electrophilic warhead to target the active site Cys145. Utilizing a constrained cyclic peptide that locks the conformation between the P3 (Val) and P2 (Leu) residues, we identified a highly selective inhibitor that fills the P2 pocket occupied by the leucine residue sidechain of PF-00835231 and the dimethyl-3-azabicyclo-hexane motif in nirmatrelvir (PF-07321332). This strategy resulted in potent and highly selective Mpro inhibitors without inhibiting essential host cathepsin cysteine or serine proteases. The lead prototype compound 1 (MPro IC50 = 230 ± 18 nM) also inhibits the replication of multiple SARS-CoV-2 variants in vitro, including SARS-CoV-2 variants of concern, and can synergize at lower concentrations with the viral RNA polymerase inhibitor, remdesivir, to inhibit replication. It also reduces SARS-CoV-2 replication in SARS-CoV-2 Omicron-infected Syrian golden hamsters without obvious toxicities, demonstrating in vivo efficacy. This novel lead structure provides the basis for optimization of improved agents targeting evolving SARS-CoV-2 drug resistance that can selectively act on Mpro versus host proteases and are less likely to have off-target effects due to non-specific targeting. Developing inhibitors against the active site of the main protease (Mpro), which is highly conserved across coronaviruses, is expected to impart a higher genetic barrier to evolving SARS-CoV-2 drug resistance. Drugs that selectively inhibit the viral Mpro are less likely to have off-target effects warranting efforts to improve this therapy.


Asunto(s)
Adenosina Monofosfato , Antivirales , Proteasas 3C de Coronavirus , Inhibidores de Proteasas , SARS-CoV-2 , Replicación Viral , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacología , Antivirales/química , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Replicación Viral/efectos de los fármacos , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Humanos , COVID-19/virología , Diseño de Fármacos , Alanina/análogos & derivados , Alanina/farmacología , Chlorocebus aethiops , Betacoronavirus/efectos de los fármacos , Células Vero , Tratamiento Farmacológico de COVID-19 , Cricetinae , Mesocricetus , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Dominio Catalítico , Lactamas , Leucina , Nitrilos , Prolina
7.
J Biomed Sci ; 31(1): 86, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39232783

RESUMEN

BACKGROUND: While dengue NS1 antigen has been shown to be associated with disease pathogenesis in some studies, it has not been linked in other studies, with the reasons remaining unclear. NS1 antigen levels in acute dengue are often associated with increased disease severity, but there has been a wide variation in results based on past dengue infection and infecting dengue virus (DENV) serotype. As NS1 engages with many host lipids, we hypothesize that the type of NS1-lipid interactions alters its pathogenicity. METHODS: Primary human monocyte derived macrophages (MDMs) were co-cultured with NS1 alone or with HDL, LDL, LPS and/or platelet activating factor (PAF) from individuals with a history of past dengue fever (DF = 8) or dengue haemorrhagic fever (DHF = 8). IL-1ß levels were measured in culture supernatants, and gene expression analysis carried out in MDMs. Monocyte subpopulations were assessed by flow cytometry. Hierarchical cluster analysis with Euclidean distance calculations were used to differentiate clusters. Differentially expressed variables were extracted and a classifier model was developed to differentiate between past DF and DHF. RESULTS: Significantly higher levels of IL-1ß were seen in culture supernatants when NS1 was co-cultured with LDL (p = 0.01, median = 45.69 pg/ml), but lower levels when NS1 was co-cultured with HDL (p = 0.05, median = 4.617 pg/ml). MDMs of those with past DHF produced higher levels of IL-1ß when NS1 was co-cultured with PAF (p = 0.02). MDMs of individuals with past DHF, were significantly more likely to down-regulate RPLP2 gene expression when macrophages were co-cultured with either PAF alone, or NS1 combined with PAF, or NS1 combined with LDL. When NS1 was co-cultured with PAF, HDL or LDL two clusters were detected based on IL10 expression, but these did not differentiate those with past DF or DHF. CONCLUSIONS: As RPLP2 is important in DENV replication, regulating cellular stress responses and immune responses and IL-10 is associated with severe disease, it would be important to further explore how differential expression of RPLP2 and IL-10 could lead to disease pathogenesis based on NS1 and lipid interactions.


Asunto(s)
Virus del Dengue , Dengue , Macrófagos , Proteínas no Estructurales Virales , Humanos , Proteínas no Estructurales Virales/metabolismo , Dengue/virología , Dengue/inmunología , Macrófagos/metabolismo , Masculino , Adulto , Femenino , Interleucina-1beta/metabolismo , Lípidos
8.
Int J Biol Macromol ; 279(Pt 2): 135274, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39226976

RESUMEN

Stress granules (SGs) are cytoplasmic aggregates of proteins and mRNA that form in response to diverse environmental stressors, including viral infections. Several viruses possess the ability to block the formation of stress granules by targeting the SGs marker protein G3BP. However, the molecular functions and mechanisms underlying the regulation of SGs formation by Getah virus (GETV) remain unclear. In this study, we found that GETV infection triggered the formation of Nsp3-G3BP aggregates, which differed in composition from SGs. Further studies revealed that the presence of these aggregates was dependent on the activation of the PKR/eIF2α signaling pathway. Interestingly, we found that Nsp3 HVD domain blocked the formation of SGs by binding to G3BP NTF2 domain. Moreover, knockout of G3BP in NCI-H1299 cells had no effect on GETV replication, while overexpression of G3BP to form the genuine SGs significantly inhibited GETV replication. Overall, our study elucidates a novel role GETV Nsp3 to change the composition of SG as well as cellular stress response.


Asunto(s)
ADN Helicasas , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Gránulos de Estrés , Proteínas no Estructurales Virales , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Humanos , Gránulos de Estrés/metabolismo , ARN Helicasas/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral , Transducción de Señal , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Gránulos Citoplasmáticos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética
9.
Viruses ; 16(8)2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39205277

RESUMEN

West Nile virus (WNV) nonstructural protein 5 (NS5) possesses multiple enzymatic domains essential for viral RNA replication. During infection, NS5 predominantly localizes to unique replication organelles (ROs) at the rough endoplasmic reticulum (RER), known as vesicle packets (VPs) and convoluted membranes (CMs), with a portion of NS5 accumulating in the nucleus. NS5 is a soluble protein that must be in the VP, where its enzymatic activities are required for viral RNA synthesis. However, the mechanistic processes behind the recruitment of NS5 from the cytoplasm to the RER membrane remain unclear. Here, we utilize high-resolution confocal microscopy and sucrose density gradient ultracentrifugation to investigate whether the association of NS5 with other NS proteins contributes to its membrane recruitment and retention. We demonstrate that NS1 or NS3 partially influences the NS5 association with the membrane. We further demonstrate that processed NS5 is predominantly in the cytoplasm and nucleus, indicating that the processing of NS5 from the viral polyprotein does not contribute to its membrane localization. These observations suggest that other host or viral factors, such as the enwrapment of NS5 by the RO, may also be necessary for the complete membrane retention of NS5. Therefore, studies on the inhibitors that disrupt the membrane localization of WNV NS5 are warranted for antiviral drug development.


Asunto(s)
Proteínas no Estructurales Virales , Virus del Nilo Occidental , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Virus del Nilo Occidental/enzimología , Virus del Nilo Occidental/fisiología , Humanos , Animales , Replicación Viral , ARN Helicasas/metabolismo , ARN Helicasas/genética , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Chlorocebus aethiops , Citoplasma/metabolismo , Células Vero , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Fiebre del Nilo Occidental/virología , Línea Celular , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-box
10.
PLoS Pathog ; 20(8): e1012387, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39213434

RESUMEN

Infection of Rift Valley fever virus (RVFV), a highly pathogenic mosquito-borne zoonotic virus, triggers severe inflammatory pathogenesis but the underlying mechanism of inflammation activation is currently unclear. Here, we report that the non-structural protein NSs of RVFV triggers mitochondrial damage to activate the NLRP3 inflammasome leading to viral pathogenesis in vivo. It is found that the host transcription inhibition effect of NSs causes rapid down-regulation of myeloid cell leukemia-1(MCL-1), a pro-survival member of the Bcl-2 (B-cell lymphoma protein 2) protein family. MCL-1 down-regulation led to BAK activation in the mitochondria, which triggered mtROS production and release of oxidized mitochondrial DNA (ox-mtDNA) into the cytosol. Cytosolic ox-mtDNA binds and activates the NLRP3 inflammasome triggering NLRP3-GSDMD pyroptosis in RVFV infected cells. A NSs mutant virus (RVFV-NSsRM) that is compromised in inducing transcription inhibition did not trigger MCL-1 down-regulation nor NLRP3-GSDMD pyroptosis. RVFV infection of the Nlrp3-/- mouse model demonstrated that the RVFV-triggered NLRP3 pyroptosis contributed to RVFV inflammatory pathogenesis and fatal infection in vivo. Infection with the RVFV-NSsRM mutant virus similarly showed alleviated inflammatory pathogenesis and reduced fatality rate. Taken together, these results revealed a mechanism by which a virulence factor activates the mitochondrial MCL-1-BAK axis through inducing host transcription inhibition to trigger NLRP3-dependent inflammatory pathogenesis.


Asunto(s)
Mitocondrias , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Animales , Humanos , Ratones , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/virología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Virus de la Fiebre del Valle del Rift , Proteínas no Estructurales Virales
11.
J Virol ; 98(9): e0063524, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39158346

RESUMEN

Flavivirus infection capitalizes on cellular lipid metabolism to remodel the cellular intima, creating a specialized lipid environment conducive to viral replication, assembly, and release. The Japanese encephalitis virus (JEV), a member of the Flavivirus genus, is responsible for significant morbidity and mortality in both humans and animals. Currently, there are no effective antiviral drugs available to combat JEV infection. In this study, we embarked on a quest to identify anti-JEV compounds within a lipid compound library. Our research led to the discovery of two novel compounds, isobavachalcone (IBC) and corosolic acid (CA), which exhibit dose-dependent inhibition of JEV proliferation. Time-of-addition assays indicated that IBC and CA predominantly target the late stage of the viral replication cycle. Mechanistically, JEV nonstructural proteins 1 and 2A (NS1 and NS2A) impede 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK) activation by obstructing the liver kinase B1 (LKB1)-AMPK interaction, resulting in decreased p-AMPK expression and a consequent upsurge in lipid synthesis. In contrast, IBC and CA may stimulate AMPK by binding to its active allosteric site, thereby inhibiting lipid synthesis essential for JEV replication and ultimately curtailing viral infection. Most importantly, in vivo experiments demonstrated that IBC and CA protected mice from JEV-induced mortality, significantly reducing viral loads in the brain and mitigating histopathological alterations. Overall, IBC and CA demonstrate significant potential as effective anti-JEV agents by precisely targeting AMPK-associated signaling pathways. These findings open new therapeutic avenues for addressing infections caused by Flaviviruses. IMPORTANCE: This study is the inaugural utilization of a lipid compound library in antiviral drug screening. Two lipid compounds, isobavachalcone (IBC) and corosolic acid (CA), emerged from the screening, exhibiting substantial inhibitory effects on the Japanese encephalitis virus (JEV) proliferation in vitro. In vivo experiments underscored their efficacy, with IBC and CA reducing viral loads in the brain and mitigating JEV-induced histopathological changes, effectively shielding mice from fatal JEV infection. Intriguingly, IBC and CA may activate 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK) by binding to its active site, curtailing the synthesis of lipid substances, and thus suppressing JEV proliferation. This indicates AMPK as a potential antiviral target. Remarkably, IBC and CA demonstrated suppression of multiple viruses, including Flaviviruses (JEV and Zika virus), porcine herpesvirus (pseudorabies virus), and coronaviruses (porcine deltacoronavirus and porcine epidemic diarrhea virus), suggesting their potential as broad-spectrum antiviral agents. These findings shed new light on the potential applications of these compounds in antiviral research.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Antivirales , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Metabolismo de los Lípidos , Replicación Viral , Animales , Metabolismo de los Lípidos/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/fisiología , Ratones , Antivirales/farmacología , Humanos , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/virología , Proteínas Quinasas Activadas por AMP/metabolismo , Chalconas/farmacología , Triterpenos/farmacología , Proteínas no Estructurales Virales/metabolismo , Infecciones por Flavivirus/tratamiento farmacológico , Infecciones por Flavivirus/virología , Infecciones por Flavivirus/metabolismo , Flavivirus/efectos de los fármacos , Línea Celular
12.
J Biol Chem ; 300(9): 107724, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39214299

RESUMEN

Single-stranded, positive-sense RNA ((+)RNA) viruses replicate their genomes in virus-induced intracellular membrane compartments. (+)RNA viruses dedicate a significant part of their small genomes (a few thousands to a few tens of thousands of bases) to the generation of these compartments by encoding membrane-interacting proteins and/or protein domains. Noroviruses are a very diverse genus of (+)RNA viruses including human and animal pathogens. Human noroviruses are the major cause of acute gastroenteritis worldwide, with genogroup II genotype 4 (GII.4) noroviruses accounting for the vast majority of infections. Three viral proteins encoded in the N terminus of the viral replication polyprotein direct intracellular membrane rearrangements associated with norovirus replication. Of these three, nonstructural protein 4 (NS4) seems to be the most important, although its exact functions in replication organelle formation are unknown. Here, we produce, purify, and characterize GII.4 NS4. AlphaFold modeling combined with experimental data refines and corrects our previous crude structural model of NS4. Using simple artificial liposomes, we report an extensive characterization of the membrane properties of NS4. We find that NS4 self-assembles and thereby bridges liposomes together. Cryo-EM, NMR, and membrane flotation show formation of several distinct NS4 assemblies, at least two of them bridging pairs of membranes together in different fashions. Noroviruses belong to (+)RNA viruses whose replication compartment is extruded from the target endomembrane and generates double-membrane vesicles. Our data establish that the 21-kDa GII.4 human norovirus NS4 can, in the absence of any other factor, recapitulate in tubo several features, including membrane apposition, that occur in such processes.


Asunto(s)
Norovirus , Proteínas no Estructurales Virales , Norovirus/metabolismo , Norovirus/química , Norovirus/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Humanos , Multimerización de Proteína , Liposomas/metabolismo , Liposomas/química , Replicación Viral
13.
Viruses ; 16(8)2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39205189

RESUMEN

Adeno-associated viruses (AAVs) are small, non-enveloped viruses that package a single-stranded (ss)DNA genome of 4.7 kilobases (kb) within their T = 1 icosahedral capsid. AAVs are replication-deficient viruses that require a helper virus to complete their life cycle. Recombinant (r)AAVs have been utilized as gene delivery vectors for decades in gene therapy applications. So far, six rAAV-based gene medicines have been approved by the US FDA. The 4.7 kb ssDNA genome of AAV encodes nine proteins, including three viral structural/capsid proteins, VP1, VP2, and VP3; four large nonstructural proteins (replication-related proteins), Rep78/68 and Rep52/40; and two small nonstructural proteins. The two nonstructured proteins are viral accessory proteins, namely the assembly associated protein (AAP) and membrane-associated accessory protein (MAAP). Although the accessory proteins are conserved within AAV serotypes, their functions are largely obscure. In this review, we focus on the expression strategy and functional properties of the small nonstructural proteins of AAVs.


Asunto(s)
Dependovirus , Vectores Genéticos , Proteínas no Estructurales Virales , Dependovirus/genética , Humanos , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Vectores Genéticos/genética , Replicación Viral , Animales , Terapia Genética/métodos , Genoma Viral
14.
PLoS Pathog ; 20(8): e1012388, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39102425

RESUMEN

Enteroviruses are a vast genus of positive-sense RNA viruses that cause diseases ranging from common cold to poliomyelitis and viral myocarditis. They encode a membrane-bound AAA+ ATPase, 2C, that has been suggested to serve several roles in virus replication, e.g. as an RNA helicase and capsid assembly factor. Here, we report the reconstitution of full-length, poliovirus 2C's association with membranes. We show that the N-terminal membrane-binding domain of 2C contains a conserved glycine, which is suggested by structure predictions to divide the domain into two amphipathic helix regions, which we name AH1 and AH2. AH2 is the main mediator of 2C oligomerization, and is necessary and sufficient for its membrane binding. AH1 is the main mediator of a novel function of 2C: clustering of membranes. Cryo-electron tomography reveal that several 2C copies mediate this function by localizing to vesicle-vesicle interfaces. 2C-mediated clustering is partially outcompeted by RNA, suggesting a way by which 2C can switch from an early role in coalescing replication organelles and lipid droplets, to a later role where 2C assists RNA replication and particle assembly. 2C is sufficient to recruit RNA to membranes, with a preference for double-stranded RNA (the replicating form of the viral genome). Finally, the in vitro reconstitution revealed that full-length, membrane-bound 2C has ATPase activity and ATP-independent, single-strand ribonuclease activity, but no detectable helicase activity. Together, this study suggests novel roles for 2C in membrane clustering, RNA membrane recruitment and cleavage, and calls into question a role of 2C as an RNA helicase. The reconstitution of functional, 2C-decorated vesicles provides a platform for further biochemical studies into this protein and its roles in enterovirus replication.


Asunto(s)
ARN Viral , Proteínas Virales , Replicación Viral , ARN Viral/metabolismo , ARN Viral/genética , Humanos , Replicación Viral/fisiología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Poliovirus/metabolismo , Poliovirus/fisiología , Membrana Celular/metabolismo , Enterovirus/fisiología , Adenosina Trifosfatasas/metabolismo , Proteínas Portadoras , Proteínas no Estructurales Virales
15.
Microb Pathog ; 195: 106894, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39214424

RESUMEN

Dengue virus (DENV) infection is a worldwide public health concern infecting approximately 400 million individuals and about 40,000 mortalities yearly. Despite this, no licensed or readily available antiviral medication is currently available specifically for DENV infection, and therapy is typically symptomatic. Therefore, the objective of the study was to investigate the antiviral activity of Beta vulgaris L. phytoconstituents against DENV-2 targeting NS3 protein. The antiviral activity of phytochemicals was examined through virtual ligand-based screening, antiviral inhibition and dosage response assays, western blotting analysis and MD simulations. We conducted toxicological, and pharmacokinetic analysis to assess plant-based natural compound's efficacy, safety, and non-toxic doses. Molecular docking and MD simulation results revealed that the nonstructural protein-3 (NS3) might prove as a funamental target for Betanin and Glycine Betaine against Dengue virus. Betanin and Glycine betaine were initially studied for their non-toxic doses in HeLa, CHO, and Vero cells via MTT assay. HeLa cells were transiently transfected with cloned vector pcDNA3.1/Zeo(+)/DENV-2 NS3 along with non-toxic doses (80 µM-10 µM) of selected phytochemicals. The dose-response assay illustrated downregulated expression of DENV-2 NS3 gene after administration of Betanin (IC50 = 4.35 µM) and Glycine Betaine (IC50 = 4.49 µM). Dose response analysis of Betanin (80 µM-10 µM) depicted the significant inhibition of NS3 protein expression as well. These results suggested downregulated expression of DENV-2 NS3 at mRNA and protein level portraying the DENV replication inhibition. Based on our study findings, NS3 protease is depicted as distinctive DENV-2 inhibitor target. We will channel our study further into in vitro characterization employing the mechanistic study to understand the role of host factors in anti-flavi therapeutic.


Asunto(s)
Antivirales , Betaína , Virus del Dengue , Simulación del Acoplamiento Molecular , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , Humanos , Antivirales/farmacología , Células HeLa , Animales , Chlorocebus aethiops , Células Vero , Betaína/farmacología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Betacianinas/farmacología , Células CHO , Cricetulus , Fitoquímicos/farmacología , Simulación de Dinámica Molecular , Replicación Viral/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Dengue/tratamiento farmacológico , Dengue/virología , Proteasas Virales
16.
Vet Microbiol ; 296: 110173, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38971119

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is the most economically significant disease caused by porcine reproductive and respiratory syndrome virus (PRRSV). Type I interferon (IFN) induces a large number of interferon-stimulated genes (ISGs) expression to inhibit PRRSV infection. To survive in the host, PRRSV has evolved multiple strategies to antagonize host innate immune response. Previous studies have reported that PRRSV N protein decreases the expression of TRIM25 and TRIM25-mediated RIG-I ubiquitination to suppress IFN-ß production. However, whether other PRRSV proteins inhibit the antiviral function of TRIM25 is less well understood. In this study, we first found that PRRSV NSP1α decreased ISGylation of TRIM25. Meanwhile, NSP1α significantly suppressed TRIM25-mediated IFN-ß production to promote PRRSV replication. Further studies demonstrated that PRRSV NSP1α reduced the protein level of TRIM25 in proteasome system but did not regulate the transcription level of TRIM25. In addition, the function of NSP1α in TRIM25 degradation did not rely on its papain-like cysteine protease activity. Taken together, PRRSV NSP1α antagonizes the antiviral response of TRIM25 by mediating TRIM25 degradation to promote PRRSV replication. Our data identify TRIM25 as a natural target of PRRSV NSP1α and reveal a novel mechanism that PRRSV induces TRIM25 degradation and inhibits host antiviral immune response.


Asunto(s)
Inmunidad Innata , Virus del Síndrome Respiratorio y Reproductivo Porcino , Complejo de la Endopetidasa Proteasomal , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas no Estructurales Virales , Replicación Viral , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Animales , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Porcinos , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Interferón beta/genética , Interferón beta/metabolismo , Interferón beta/inmunología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Línea Celular , Ubiquitinación , Humanos , Células HEK293 , Interacciones Huésped-Patógeno/inmunología
17.
Int J Biol Macromol ; 276(Pt 1): 133706, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38981557

RESUMEN

Main proteases (Mpros) are a class of conserved cysteine hydrolases among coronaviruses and play a crucial role in viral replication. Therefore, Mpros are ideal targets for the development of pan-coronavirus drugs. X77, previously developed against SARS-CoV Mpro, was repurposed as a non-covalent tight binder inhibitor against SARS-CoV-2 Mpro during COVID-19 pandemic. Many novel inhibitors with favorable efficacy have been discovered using X77 as a reference, suggesting that X77 could be a valuable scaffold for drug design. However, the broad-spectrum performance of X77 and underlying mechanism remain less understood. Here, we reported the crystal structures of Mpros from SARS-CoV-2, SARS-CoV, and MERS-CoV, and several Mpro mutants from SARS-CoV-2 variants bound to X77. A detailed analysis of these structures revealed key structural determinants essential for interaction and elucidated the binding modes of X77 with different coronaviral Mpros. The potencies of X77 against these investigated Mpros were further evaluated through molecular dynamic simulation and binding free energy calculation. These data provide molecular insights into broad-spectrum inhibition against coronaviral Mpros by X77 and the similarities and differences of X77 when bound to various Mpros, which will promote X77-based design of novel antivirals with broad-spectrum efficacy against different coronaviruses and SARS-CoV-2 variants.


Asunto(s)
Proteasas 3C de Coronavirus , Simulación de Dinámica Molecular , SARS-CoV-2 , SARS-CoV-2/enzimología , SARS-CoV-2/efectos de los fármacos , Cristalografía por Rayos X , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , Antivirales/química , Antivirales/farmacología , Humanos , Unión Proteica , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , COVID-19/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Betacoronavirus/enzimología , Betacoronavirus/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/enzimología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Sitios de Unión , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/tratamiento farmacológico , Pandemias , Neumonía Viral/virología , Neumonía Viral/tratamiento farmacológico
18.
Virol Sin ; 39(4): 619-631, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38969340

RESUMEN

A unique feature of coronaviruses is their utilization of self-encoded nonstructural protein 16 (nsp16), 2'-O-methyltransferase (2'-O-MTase), to cap their RNAs through ribose 2'-O-methylation modification. This process is crucial for maintaining viral genome stability, facilitating efficient translation, and enabling immune escape. Despite considerable advances in the ultrastructure of SARS-CoV-2 nsp16/nsp10, insights into its molecular mechanism have so far been limited. In this study, we systematically characterized the 2'-O-MTase activity of nsp16 in SARS-CoV-2, focusing on its dependence on nsp10 stimulation. We observed cross-reactivity between nsp16 and nsp10 in various coronaviruses due to a conserved interaction interface. However, a single residue substitution (K58T) in SARS-CoV-2 nsp10 restricted the functional activation of MERS-CoV nsp16. Furthermore, the cofactor nsp10 effectively enhanced the binding of nsp16 to the substrate RNA and the methyl donor S-adenosyl-l-methionine (SAM). Mechanistically, His-80, Lys-93, and Gly-94 of nsp10 interacted with Asp-102, Ser-105, and Asp-106 of nsp16, respectively, thereby effectively stabilizing the SAM binding pocket. Lys-43 of nsp10 interacted with Lys-38 and Gly-39 of nsp16 to dynamically regulate the RNA binding pocket and facilitate precise binding of RNA to the nsp16/nsp10 complex. By assessing the conformational epitopes of nsp16/nsp10 complex, we further determined the critical residues involved in 2'-O-MTase activity. Additionally, we utilized an in vitro biochemical platform to screen potential inhibitors targeting 2'-O-MTase activity. Overall, our results significantly enhance the understanding of viral 2'-O methylation process and mechanism, providing valuable targets for antiviral drug development.


Asunto(s)
Metiltransferasas , SARS-CoV-2 , Proteínas no Estructurales Virales , SARS-CoV-2/enzimología , SARS-CoV-2/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/química , Metiltransferasas/metabolismo , Metiltransferasas/genética , Metiltransferasas/química , Humanos , ARN Viral/genética , ARN Viral/metabolismo , COVID-19/virología , Unión Proteica , S-Adenosilmetionina/metabolismo , Metilación , Betacoronavirus/enzimología , Betacoronavirus/genética , Modelos Moleculares , Coronavirus del Síndrome Respiratorio de Oriente Medio/enzimología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Proteínas Reguladoras y Accesorias Virales
19.
Sci Rep ; 14(1): 17645, 2024 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085250

RESUMEN

The Middle East has witnessed a greater spread of infectious Dengue viruses, with serotype 2 (DENV-2) being the most prevalent form. Through this work, multi-epitope peptide vaccines against DENV-2 that target E and nonstructural (NS1) proteins were generated through an immunoinformatic approach. MHC class I and II and LBL epitopes among NS1 and envelope E proteins sequences were predicted and their antigenicity, toxicity, and allergenicity were investigated. Studies of the population coverage denoted the high prevalence of NS1 and envelope-E epitopes among different countries where DENV-2 endemic. Further, both the CTL and HTL epitopes retrieved from NS1 epitopes exhibited high conservancies' percentages with other DENV serotypes (1, 3, and 4). Three vaccine constructs were created and the expected immune responses for the constructs were estimated using C-IMMSIM and HADDOCK (against TLR 2,3,4,5, and 7). Molecular dynamics simulation for vaccine construct 2 with TLR4 denoted high binding affinity and stability of the construct with the receptor which might foretell favorable in vivo interaction and immune responses.


Asunto(s)
Vacunas contra el Dengue , Virus del Dengue , Dengue , Serogrupo , Vacunas de Subunidad , Proteínas no Estructurales Virales , Virus del Dengue/inmunología , Vacunas de Subunidad/inmunología , Vacunas contra el Dengue/inmunología , Humanos , Dengue/prevención & control , Dengue/inmunología , Dengue/virología , Proteínas no Estructurales Virales/inmunología , Biología Computacional/métodos , Epítopos de Linfocito T/inmunología , Proteínas del Envoltorio Viral/inmunología , Simulación de Dinámica Molecular , Epítopos/inmunología , Epítopos/química , Vacunas de Subunidades Proteicas
20.
Protein Pept Lett ; 31(7): 532-543, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39039677

RESUMEN

BACKGROUND: Peptide drugs are advantageous because they are subject to rational design and exhibit highly diverse structures and broad biological activities. The NS2B-NS3 protein is a particularly promising flavivirus therapeutic target, with extensive research on the development of inhibitors as therapeutic candidates, and was used as a model in this work to determine the mechanism by which GA-Hecate inhibits ZIKV replication. OBJECTIVE: The present study aimed to evaluate the potential of GA-Hecate, a new antiviral developed by our group, against the Brazilian Zika virus and to evaluate the mechanism of action of this compound on the flavivirus NS2B-NS3 protein. METHODS: Solid-phase peptide Synthesis, High-Performance Liquid Chromatography, and Mass Spectrometry were used to obtain, purify, and characterize the synthesized compound. Real-time and enzymatic assays were used to determine the antiviral potential of GA-Hecate against ZIKV. RESULTS: The RT-qPCR results showed that GA-Hecate decreased the number of ZIKV RNA copies in the virucidal, pre-treatment, and post-entry assays, with 5- to 6-fold fewer RNA copies at the higher nontoxic concentration in Vero cells (HNTC: 10 µM) than in the control cells. Enzymatic and kinetic assays indicated that GA-Hecate acts as a competitive ZIKV NS2B-NS3 protease inhibitor with an IC50 of 32 nM and has activity against the yellow fever virus protease. CONCLUSION: The results highlight the antiviral potential of the GA-Hecate bioconjugate and open the door for the development of new antivirals.


Asunto(s)
Antivirales , Proteínas no Estructurales Virales , Replicación Viral , Virus Zika , Virus Zika/efectos de los fármacos , Antivirales/farmacología , Antivirales/química , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Chlorocebus aethiops , Células Vero , Replicación Viral/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Péptidos/farmacología , Péptidos/química , ARN Helicasas/metabolismo , ARN Helicasas/antagonistas & inhibidores , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología , Humanos , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-box
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA