Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.945
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(34): e2403235121, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39145933

RESUMO

The ZIKA virus (ZIKV) evades the host immune response by degrading STAT2 through its NS5 protein, thereby inhibiting type I interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism underlying this process has remained elusive. In this study, we performed a genome-wide CRISPR/Cas9 screen, revealing that ZSWIM8 as the substrate receptor of Cullin3-RING E3 ligase is required for NS5-mediated STAT2 degradation. Genetic depletion of ZSWIM8 and CUL3 substantially impeded NS5-mediated STAT2 degradation. Biochemical analysis illuminated that NS5 enhances the interaction between STAT2 and the ZSWIM8-CUL3 E3 ligase complex, thereby facilitating STAT2 ubiquitination. Moreover, ZSWIM8 knockout endowed A549 and Huh7 cells with partial resistance to ZIKV infection and protected cells from the cytopathic effects induced by ZIKV, which was attributed to the restoration of STAT2 levels and the activation of IFN signaling. Subsequent studies in a physiologically relevant model, utilizing human neural progenitor cells, demonstrated that ZSWIM8 depletion reduced ZIKV infection, resulting from enhanced IFN signaling attributed to the sustained levels of STAT2. Our findings shed light on the role of ZIKV NS5, serving as the scaffold protein, reprograms the ZSWIM8-CUL3 E3 ligase complex to orchestrate STAT2 proteasome-dependent degradation, thereby facilitating evasion of IFN antiviral signaling. Our study provides unique insights into ZIKV-host interactions and holds promise for the development of antivirals and prophylactic vaccines.


Assuntos
Proteínas Culina , Interferon Tipo I , Proteólise , Fator de Transcrição STAT2 , Transdução de Sinais , Ubiquitina-Proteína Ligases , Ubiquitinação , Proteínas não Estruturais Virais , Infecção por Zika virus , Zika virus , Humanos , Fator de Transcrição STAT2/metabolismo , Zika virus/imunologia , Zika virus/fisiologia , Zika virus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Interferon Tipo I/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Proteínas Culina/metabolismo , Células A549 , Células HEK293 , Sistemas CRISPR-Cas
2.
Trop Biomed ; 41(2): 166-175, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-39154269

RESUMO

Nsp1 in SARS-CoV-2 is a key protein that increases the virus's pathogenicity and virulence by binding to the host ribosome and blocks the 40S ribosomal subunit channel, which effectively impedes the mRNA translation as well as crippling the host immune system. Previous studies revealed that the N-terminal in Nsp1 is part and parcel of Nsp1 efficiency, and mutations in its core residues have weakened the protein's. This knowledge persuades us to carry out the in silico screening on plant compounds of Piper sarmentosum Roxb. against the five target residues which are Glu36, Glu37, Arg99, Arg124 and Lys125. Potential compounds were tested for their druggability. As a result, we identified five out of 112 compounds including stigmasterol, N-feruloyltyramine, beta-Sitosterol, 13-(1,3-benzodioxol-5-yl)- N-(2methylpropyl) trideca-2,4,12-trienamide and N-(2-methylpropyl) octadeca-2-4dienamide in Piper sarmentosum Roxb. as potential inhibitors for Nsp1. These compounds formed at least a hydrophobic, hydrogen bonding or π-cation interactions with the protein. Furthermore, SwissADME analysis and the number of bindings to the target residues suggest that N-feruloyltyramine is the ideal inhibitor candidate against SARS-CoV-2 at its N-terminal of Nsp1. Lastly, the interaction with N-feruloyltyramine increased flexibility in the loop regions of N-terminal Nsp1, especially residues 54 to 70, with residue 59 showing the highest fluctuation, potentially affecting the protein's stability and function due to the correlation between RMSF and protein function.


Assuntos
Simulação de Acoplamento Molecular , Piper , SARS-CoV-2 , Proteínas não Estruturais Virais , Piper/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , SARS-CoV-2/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/química , Humanos
3.
Protein Sci ; 33(9): e5103, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39145418

RESUMO

Since the emergence of SARS-CoV-2, mutations in all subunits of the RNA-dependent RNA polymerase (RdRp) of the virus have been repeatedly reported. Although RdRp represents a primary target for antiviral drugs, experimental studies exploring the phenotypic effect of these mutations have been limited. This study focuses on the phenotypic effects of substitutions in the three RdRp subunits: nsp7, nsp8, and nsp12, selected based on their occurrence rate and potential impact. We employed nano-differential scanning fluorimetry and microscale thermophoresis to examine the impact of these mutations on protein stability and RdRp complex assembly. We observed diverse impacts; notably, a single mutation in nsp8 significantly increased its stability as evidenced by a 13°C increase in melting temperature, whereas certain mutations in nsp7 and nsp8 reduced their binding affinity to nsp12 during RdRp complex formation. Using a fluorometric enzymatic assay, we assessed the overall effect on RNA polymerase activity. We found that most of the examined mutations altered the polymerase activity, often as a direct result of changes in stability or affinity to the other components of the RdRp complex. Intriguingly, a combination of nsp8 A21V and nsp12 P323L mutations resulted in a 50% increase in polymerase activity. To our knowledge, this is the first biochemical study to demonstrate the impact of amino acid mutations across all components constituting the RdRp complex in emerging SARS-CoV-2 subvariants.


Assuntos
RNA-Polimerase RNA-Dependente de Coronavírus , Mutação , SARS-CoV-2 , Proteínas não Estruturais Virais , SARS-CoV-2/genética , SARS-CoV-2/enzimologia , RNA-Polimerase RNA-Dependente de Coronavírus/genética , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , RNA-Polimerase RNA-Dependente de Coronavírus/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Humanos , COVID-19/virologia , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Estabilidade Proteica , Ligação Proteica
4.
Virology ; 598: 110195, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39089050

RESUMO

Rotavirus A is a leading cause of non-bacterial gastroenteritis in humans and domesticated animals. Despite the vast diversity of bovine Rotavirus A strains documented in South Asian countries, there are very few whole genomes available for phylogenetic study. A cross-sectional study identified a high prevalence of the G6P[11] genotype of bovine Rotavirus A circulating in the commercial cattle population in Bangladesh. Next-generation sequencing and downstream phylogenetic analysis unveiled all 11 complete gene segments of this strain (BD_ROTA_CVASU), classifying it under the genomic constellation G6P[11]-I2-R2-C2-M2-A13-N2-T6-E2-H3, which belongs to a classical DS-1-like genomic backbone. We found strong evidence of intragenic recombination between human and bovine strains in the Non-structural protein 4 (NSP4) gene, which encodes a multifunctional enterotoxin. Our analyses highlight frequent zoonotic transmissions of rotaviruses in diverse human-animal interfaces, which might have contributed to the evolution and pathogenesis of this dominant genotype circulating in the commercial cattle population in Bangladesh.


Assuntos
Doenças dos Bovinos , Genoma Viral , Genótipo , Filogenia , Recombinação Genética , Infecções por Rotavirus , Rotavirus , Toxinas Biológicas , Proteínas não Estruturais Virais , Animais , Bovinos , Rotavirus/genética , Rotavirus/classificação , Rotavirus/isolamento & purificação , Bangladesh/epidemiologia , Proteínas não Estruturais Virais/genética , Humanos , Infecções por Rotavirus/virologia , Infecções por Rotavirus/veterinária , Infecções por Rotavirus/epidemiologia , Doenças dos Bovinos/virologia , Doenças dos Bovinos/epidemiologia , Estudos Transversais , Toxinas Biológicas/genética , Glicoproteínas/genética
5.
Emerg Microbes Infect ; 13(1): 2387910, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39087696

RESUMO

Nuclear export of the viral ribonucleoprotein (vRNP) is a critical step in the influenza A virus (IAV) life cycle and may be an effective target for the development of anti-IAV drugs. The host factor ras-related nuclear protein (RAN) is known to participate in the life cycle of several viruses, but its role in influenza virus replication remains unknown. In the present study, we aimed to determine the function of RAN in influenza virus replication using different cell lines and subtype strains. We found that RAN is essential for the nuclear export of vRNP, as it enhances the binding affinity of XPO1 toward the viral nuclear export protein NS2. Depletion of RAN constrained the vRNP complex in the nucleus and attenuated the replication of various subtypes of influenza virus. Using in silico compound screening, we identified that bepotastine could dissociate the RAN-XPO1-vRNP trimeric complex and exhibit potent antiviral activity against influenza virus both in vitro and in vivo. This study demonstrates the important role of RAN in IAV replication and suggests its potential use as an antiviral target.


Assuntos
Transporte Ativo do Núcleo Celular , Antivirais , Proteína Exportina 1 , Vírus da Influenza A , Carioferinas , Replicação Viral , Proteína ran de Ligação ao GTP , Replicação Viral/efeitos dos fármacos , Humanos , Proteína ran de Ligação ao GTP/metabolismo , Proteína ran de Ligação ao GTP/genética , Antivirais/farmacologia , Animais , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/fisiologia , Carioferinas/metabolismo , Carioferinas/antagonistas & inibidores , Cães , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Células Madin Darby de Rim Canino , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Camundongos , Piperidinas/farmacologia , Influenza Humana/virologia , Células A549 , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Células HEK293 , Linhagem Celular , Núcleo Celular/metabolismo , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/genética
6.
Sci Rep ; 14(1): 18883, 2024 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-39143088

RESUMO

NS1 is an elusive dengue protein, involved in viral replication, assembly, pathogenesis, and immune evasion. Its levels in blood plasm are positively related to disease severity like thrombocytopenia, hemorrhage, and vascular leakage. Despite its pathogenic roles, NS1 is being used in various vaccine formulations due to its sequence conservancy, ability to produce protective antibodies and low risk for inducing antibody-dependent enhancement. In this study, we have used bioinformatics tools and reported literature to develop an NS1 variant (dNS1). Molecular docking studies were performed to evaluate the receptor-binding ability of the NS1 and dNS1 with TLR4. NS1 and dNS1 (153 to 312 amino acid region) genes were cloned, expressed and protein was purified followed by refolding. Docking studies showed the binding of NS1 and dNS1 with the TLR4 receptor which suggests that N and C-terminal sequences of NS1 are not critical for receptor binding. Antibodies against NS1 and dNS1 were raised in rabbits and binding affinity of anti-dNS1 anti-NS1 sera was evaluated against both NS1 and dNS1. Similar results were observed through western blotting which highlight that N and C-terminal deletion of NS1 does not compromise the immunogenic potential of dNS1 hence, supports its use in future vaccine formulations as a substitute for NS1.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Simulação de Acoplamento Molecular , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Vírus da Dengue/imunologia , Vírus da Dengue/genética , Animais , Vacinas contra Dengue/imunologia , Vacinas contra Dengue/genética , Desenvolvimento de Vacinas , Coelhos , Humanos , Dengue/prevenção & controle , Dengue/imunologia , Dengue/virologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Ligação Proteica , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/imunologia , Deleção de Sequência
7.
Methods Mol Biol ; 2824: 397-408, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39039426

RESUMO

The NSs protein is a major virulence factor in bunyaviruses, crucial for viral pathogenesis. However, assessing NSs protein function can be challenging due to its inhibition of cellular RNA polymerase II, impacting NSs protein expression from plasmid DNA. The recombinant Rift Valley fever virus (RVFV) MP-12 strain (rMP-12), a highly attenuated vaccine strain, can be safely manipulated under biosafety level 2 conditions. Leveraging a reverse genetics system, we can engineer rMP-12 variants expressing heterologous NSs genes, enabling functional testing in cultured cells. Human macrophages hold a central role in viral pathogenesis, making them an ideal model for assessing NSs protein functions. Consequently, we can comprehensively compare and analyze the functional significance of various NSs proteins in human macrophages using rMP-12 NSs variants. In this chapter, we provide a detailed overview of the preparation process for rMP-12 NSs variants and introduce two distinct human macrophage models: THP-1 cells and primary macrophages. This research framework promises valuable insights into the virulence mechanisms of RVFV and other bunyaviruses and the potential for vaccine development.


Assuntos
Macrófagos , Vírus da Febre do Vale do Rift , Proteínas não Estruturais Virais , Humanos , Macrófagos/virologia , Macrófagos/imunologia , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/patogenicidade , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Células THP-1
8.
Front Immunol ; 15: 1440407, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39072326

RESUMO

Introduction: Bluetongue (BT), caused by bluetongue virus (BTV), is an important arthropod-borne livestock disease listed by the World Organization for Animal Health. Live-attenuated and inactivated vaccines have permitted to control BT but they do not simultaneously protect against the myriad of BTV serotypes. Recently, we identified the highly conserved BTV nonstructural protein NS1 and the N-terminal region of NS2 as antigens capable of conferring multiserotype protection against BTV. Methods: Here, we designed Modified Vaccinia Ankara (MVA) viral vectors that expressed BTV-4 proteins VP2 or VP7 along with NS1 and NS2-Nt as well as MVAs that expressed proteins VP2, VP7 or NS1 and NS2-Nt. Results: Immunization of IFNAR(-/-) mice with two doses of MVA-NS1-2A-NS2-Nt protected mice from BTV-4M infection by the induction of an antigen-specific T cell immune response. Despite rMVA expressing VP7 alone were not protective in the IFNAR(-/-) mouse model, inclusion of VP7 in the vaccine formulation amplified the cell-mediated response induced by NS1 and NS2-Nt. Expression of VP2 elicited protective non-cross-reactive neutralizing antibodies (nAbs) in immunized animals and improved the protection observed in the MVA-NS1-2A-NS2-Nt immunized mice when these three BTV antigens were co-expressed. Moreover, vaccines candidates co-expressing VP2 or VP7 along with NS1 and NS2-Nt provided multiserotype protection. We assessed protective efficacy of both vaccine candidates in sheep against virulent challenge with BTV-4M. Discussion: Immunization with MVA-VP7-NS1-2A-NS2-Nt partially dumped viral replication and clinical disease whereas administration of MVA-VP2-NS1-2A-NS2-Nt promoted a complete protection, preventing viraemia and the pathology produced by BTV infection.


Assuntos
Vírus Bluetongue , Bluetongue , Proteínas do Capsídeo , Vetores Genéticos , Receptor de Interferon alfa e beta , Vaccinia virus , Proteínas não Estruturais Virais , Vacinas Virais , Animais , Vírus Bluetongue/imunologia , Vírus Bluetongue/genética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/genética , Bluetongue/prevenção & controle , Bluetongue/imunologia , Bluetongue/virologia , Camundongos , Vacinas Virais/imunologia , Vacinas Virais/genética , Vaccinia virus/genética , Vaccinia virus/imunologia , Receptor de Interferon alfa e beta/genética , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/genética , Camundongos Knockout , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Feminino
9.
Front Immunol ; 15: 1419321, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39081319

RESUMO

Similar to other RNA viruses, grass carp reovirus, the causative agent of the hemorrhagic disease, replicates in cytoplasmic viral inclusion bodies (VIBs), orchestrated by host proteins and lipids. The host pathways that facilitate the formation and function of GCRV VIBs are poorly understood. This work demonstrates that GCRV manipulates grass carp oxysterol binding protein 1 (named as gcOSBP1) and vesicle-associated membrane protein-associated protein A/B (named as gcVAP-A/B), 3 components of cholesterol transport pathway, to generate VIBs. By siRNA-mediated knockdown, we demonstrate that gcOSBP1 is an essential host factor for GCRV replication. We reveal that the nonstructural proteins NS80 and NS38 of GCRV interact with gcOSBP1, and that the gcOSBP1 is recruited by NS38 and NS80 for promoting the generation of VIBs. gcOSBP1 increases the expression of gcVAP-A/B and promotes the accumulation of intracellular cholesterol. gcOSBP1 also interacts with gcVAP-A/B for forming gcOSBP1-gcVAP-A/B complexes, which contribute to enhance the accumulation of intracellular cholesterol and gcOSBP1-mediated generation of VIBs. Inhibiting cholesterol accumulation by lovastatin can completely abolish the effects of gcOSBP1 and/or gcVAP-A/B in promoting GCRV infection, suggesting that cholesterol accumulation is vital for gcOSBP1- and/or gcVAP-A/B-mediated GCRV replication. Thus, our results, which highlight that gcOSBP1 functions in the replication of GCRV via its interaction with essential viral proteins for forming VIBs and with host gcVAP-A/B, provide key molecular targets for obtaining anti-hemorrhagic disease grass carp via gene editing technology.


Assuntos
Carpas , Colesterol , Corpos de Inclusão Viral , Receptores de Esteroides , Reoviridae , Replicação Viral , Animais , Reoviridae/fisiologia , Carpas/virologia , Carpas/metabolismo , Corpos de Inclusão Viral/metabolismo , Colesterol/metabolismo , Receptores de Esteroides/metabolismo , Doenças dos Peixes/virologia , Doenças dos Peixes/metabolismo , Doenças dos Peixes/imunologia , Interações Hospedeiro-Patógeno , Infecções por Reoviridae/veterinária , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/virologia , Proteínas de Peixes/metabolismo , Proteínas de Peixes/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética
10.
mSphere ; 9(7): e0040624, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38980068

RESUMO

Dengue virus (DENV) infection is known to affect host cell metabolism, but the molecular players involved are still poorly known. Using a proteomics approach, we identified six DENV proteins associated with mitochondria isolated from infected hepatocytes, and most of the peptides identified were from NS3. We also found an at least twofold decrease of several electron transport system (ETS) host proteins. Thus, we investigated whether NS3 could modulate the ETS function by incubating recombinant DENV NS3 constructs in mitochondria isolated from mouse liver. We found that NS3pro (NS3 protease domain), but not the correspondent catalytically inactive mutant (NS3proS135A), impairs complex I (CI)-dependent NADH:ubiquinone oxidoreductase activity, but not the activities of complexes II, III, IV, or V. Accordingly, using high-resolution respirometry, we found that both NS3pro and full-length NS3 decrease the respiratory rates associated with malate/pyruvate oxidation in mitochondria. The NS3-induced impairment in mitochondrial respiration occurs without altering either leak respiration or mitochondria's capacity to maintain membrane potential, suggesting that NS3 does not deeply affect mitochondrial integrity. Remarkably, CI activity is also inhibited in DENV-infected cells, supporting that the NS3 effects observed in isolated mitochondria may be relevant in the context of the infection. Finally, in silico analyses revealed the presence of potential NS3 cleavage sites in 17 subunits of mouse CI and 16 subunits of human CI, most of them located on the CI surface, suggesting that CI is prone to undergo proteolysis by NS3. Our findings suggest that DENV NS3 can modulate mitochondrial bioenergetics by directly affecting CI function. IMPORTANCE: Dengue virus (DENV) infection is a major public health problem worldwide, affecting about 400 million people yearly. Despite its importance, many molecular aspects of dengue pathogenesis remain poorly known. For several years, our group has been investigating DENV-induced metabolic alterations in the host cells, focusing on the bioenergetics of mitochondrial respiration. The results of the present study reveal that the DENV non-structural protein 3 (NS3) is found in the mitochondria of infected cells, impairing mitochondrial respiration by directly targeting one of the components of the electron transport system, the respiratory complex I (CI). NS3 acts as the viral protease during the DENV replication cycle, and its proteolytic activity seems necessary for inhibiting CI function. Our findings uncover new nuances of DENV-induced metabolic alterations, highlighting NS3 as an important player in the modulation of mitochondria function during infection.


Assuntos
Vírus da Dengue , Complexo I de Transporte de Elétrons , Mitocôndrias , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Animais , Vírus da Dengue/fisiologia , Vírus da Dengue/genética , Camundongos , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/genética , Humanos , Mitocôndrias/metabolismo , Hepatócitos/virologia , Hepatócitos/metabolismo , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Dengue/virologia , Dengue/metabolismo , Respiração Celular , Proteômica , Proteases Virais
11.
Sci Rep ; 14(1): 16363, 2024 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-39013947

RESUMO

Resistance-associated substitutions (RASs) of hepatitis C virus (HCV) affect the efficacy of direct-acting antivirals (DAAs). In this study, we aimed to clarify the susceptibility of the coexistence of nonstructural (NS) 5A Q24K/L28M/R30Q (or R30E)/A92K RASs, which were observed in patients with DAAs re-treatment failure and to consider new therapeutic agents. We used a subgenomic replicon system in which HCV genotype 1B strain 1B-4 was electroporated into OR6c cells derived from HuH-7 cells (Wild-type [WT]). We converted WT genes to NS5A Q24K/L28M/R30Q/A92K or Q24/L28K/R30E/A92K. Compared with the WT, the Q24K/L28M/R30Q/A92K RASs was 36,000-fold resistant to daclatasvir, 440,000-fold resistant to ledipasvir, 6300-fold resistant to velpatasvir, 3100-fold resistant to elbasvir, and 1.8-fold resistant to pibrentasvir. Compared with the WT, the Q24K/L28M/R30E/A92K RASs was 640,000-fold resistant to daclatasvir and ledipasvir, 150,000-fold resistant to velpatasvir, 44,000-fold resistant to elbasvir, and 1500-fold resistant to pibrentasvir. The Q24K/L28M/R30E/A92K RASs was 816.3 times more resistant to pibrentasvir than the Q24K/L28M/R30Q/A92K RASs. Furthermore, a combination of pibrentasvir and sofosbuvir showed therapeutic efficacy against these RASs. Combination regimens may eradicate HCV with NS5A Q24K/L28M/R30E/A92K RASs.


Assuntos
Antivirais , Benzimidazóis , Farmacorresistência Viral , Hepacivirus , Imidazóis , Proteínas não Estruturais Virais , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/antagonistas & inibidores , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Farmacorresistência Viral/genética , Farmacorresistência Viral/efeitos dos fármacos , Benzimidazóis/farmacologia , Imidazóis/farmacologia , Carbamatos/farmacologia , Fluorenos/farmacologia , Sofosbuvir/farmacologia , Pirrolidinas/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Valina/análogos & derivados , Valina/farmacologia , Genótipo , Replicon/efeitos dos fármacos , Replicon/genética , Sulfonamidas/farmacologia , Benzofuranos/farmacologia , Pirazinas/farmacologia , Benzopiranos , RNA Polimerase Dependente de RNA
12.
Front Immunol ; 15: 1424307, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39011043

RESUMO

Introduction: Bluetongue (BT) poses a significant threat to the livestock industry, affecting various animal species and resulting in substantial economic losses. The existence of numerous BT virus (BTV) serotypes has hindered control efforts, highlighting the need for broad-spectrum vaccines. Methodology: In this study, we evaluated the conserved amino acid sequences within key non-structural (NS) proteins of BTV and identified numerous highly conserved murine- and bovine-specific MHC class I-restricted (MHC-I) CD8+ and MHC-II-restricted CD4+ epitopes. We then screened these conserved epitopes for antigenicity, allergenicity, toxicity, and solubility. Using these epitopes, we developed in silico-based broad-spectrum multiepitope vaccines with Toll-like receptor (TLR-4) agonists. The predicted proinflammatory cytokine response was assessed in silico using the C-IMMSIM server. Structural modeling and refinement were achieved using Robetta and GalaxyWEB servers. Finally, we assessed the stability of the docking complexes through extensive 100-nanosecond molecular dynamics simulations before considering the vaccines for codon optimization and in silico cloning. Results: We found many epitopes that meet these criteria within NS1 and NS2 proteins and developed in silico broad-spectrum vaccines. The immune simulation studies revealed that these vaccines induce high levels of IFN-γ and IL-2 in the vaccinated groups. Protein-protein docking analysis demonstrated promising epitopes with strong binding affinities to TLR-4. The docked complexes were stable, with minimal Root Mean Square Deviation and Root Mean Square Fluctuation values. Finally, the in silico-cloned plasmids have high % of GC content with > 0.8 codon adaptation index, suggesting they are suitable for expressing the protein vaccines in prokaryotic system. Discussion: These next-generation vaccine designs are promising and warrant further investigation in wet lab experiments to assess their immunogenicity, safety, and efficacy for practical application in livestock. Our findings offer a robust framework for developing a comprehensive, broad-spectrum vaccine, potentially revolutionizing BT control and prevention strategies in the livestock industry.


Assuntos
Vírus Bluetongue , Biologia Computacional , Epitopos de Linfócito T , Proteínas não Estruturais Virais , Vacinas Virais , Animais , Vírus Bluetongue/imunologia , Epitopos de Linfócito T/imunologia , Vacinas Virais/imunologia , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/genética , Camundongos , Biologia Computacional/métodos , Sorogrupo , Bovinos , Bluetongue/prevenção & controle , Bluetongue/imunologia , Bluetongue/virologia , Sequência Conservada
13.
PLoS Pathog ; 20(7): e1011959, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39008516

RESUMO

An essential aspect of positive-sense RNA virus replication is anchoring the replication complex (RC) to cellular membranes. Positive-sense RNA viruses employ diverse strategies, including co-translational membrane targeting through signal peptides and co-opting cellular membrane trafficking components. Often, N-terminal nonstructural proteins play a crucial role in linking the RC to membranes, facilitating the early association of the replication machinery. Astroviruses utilize a polyprotein strategy to synthesize nonstructural proteins, relying on subsequent processing to form replication-competent complexes. This study provides evidence for the perinuclear ER membrane association of RCs in five distinct human astrovirus strains. Using tagged recombinant classical human astrovirus 1 and neurotropic MLB2 strains, we establish that the N-terminal domain guides the ER membrane association. We identified di-arginine motifs responsible for the perinuclear ER retention and formation of functional RCs through mutational analysis of the N-terminal domain in replicon and reverse genetics systems. In addition, we demonstrate the association of key components of the astrovirus replication complex: double-stranded RNA, RNA-dependent RNA polymerase, protease, and N-terminal protein. Our findings highlight the intricate virus-ER interaction mechanism employed by astroviruses, potentially leading to the development of novel antiviral intervention strategies.


Assuntos
Retículo Endoplasmático , Mamastrovirus , Proteínas não Estruturais Virais , Replicação Viral , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Replicação Viral/fisiologia , Humanos , Mamastrovirus/metabolismo , Mamastrovirus/genética , Infecções por Astroviridae/virologia , Infecções por Astroviridae/metabolismo , Membranas Intracelulares/metabolismo , Membranas Intracelulares/virologia
14.
Nat Commun ; 15(1): 6080, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39030239

RESUMO

Dengue fever represents a significant medical and socio-economic burden in (sub)tropical regions, yet antivirals for treatment or prophylaxis are lacking. JNJ-A07 was described as highly active against the different genotypes within each serotype of the disease-causing dengue virus (DENV). Based on clustering of resistance mutations it has been assumed to target DENV non-structural protein 4B (NS4B). Using a photoaffinity labeling compound with high structural similarity to JNJ-A07, here we demonstrate binding to NS4B and its precursor NS4A-2K-NS4B. Consistently, we report recruitment of the compound to intracellular sites enriched for these proteins. We further specify the mechanism-of-action of JNJ-A07, which has virtually no effect on viral polyprotein cleavage, but targets the interaction between the NS2B/NS3 protease/helicase complex and the NS4A-2K-NS4B cleavage intermediate. This interaction is functionally linked to de novo formation of vesicle packets (VPs), the sites of DENV RNA replication. JNJ-A07 blocks VPs biogenesis with little effect on established ones. A similar mechanism-of-action was found for another NS4B inhibitor, NITD-688. In summary, we unravel the antiviral mechanism of these NS4B-targeting molecules and show how DENV employs a short-lived cleavage intermediate to carry out an early step of the viral life cycle.


Assuntos
Antivirais , Vírus da Dengue , Dengue , Proteínas não Estruturais Virais , Replicação Viral , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Humanos , Dengue/virologia , Dengue/tratamento farmacológico , Sorogrupo , RNA Helicases/metabolismo , RNA Helicases/antagonistas & inibidores , RNA Helicases/genética , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Ligação Proteica , Animais , Organelas/metabolismo , Organelas/efeitos dos fármacos , Proteases Virais , Aminofenóis , Proteínas de Membrana , Indóis , RNA Helicases DEAD-box , Nucleosídeo-Trifosfatase , Butiratos
15.
Virus Res ; 347: 199431, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38969013

RESUMO

Usutu virus (USUV) is an emerging flavivirus that can infect birds and mammals. In humans, in severe cases, it may cause neuroinvasive disease. The innate immune system, and in particular the interferon response, functions as the important first line of defense against invading pathogens such as USUV. Many, if not all, viruses have developed mechanisms to suppress and/or evade the interferon response in order to facilitate their replication. The ability of USUV to antagonize the interferon response has so far remained largely unexplored. Using dual-luciferase reporter assays we observed that multiple of the USUV nonstructural (NS) proteins were involved in suppressing IFN-ß production and signaling. In particular NS4A was very effective at suppressing IFN-ß production. We found that NS4A interacted with the mitochondrial antiviral signaling protein (MAVS) and thereby blocked its interaction with melanoma differentiation-associated protein 5 (MDA5), resulting in reduced IFN-ß production. The TM1 domain of NS4A was found to be essential for binding to MAVS. By screening a panel of flavivirus NS4A proteins we found that the interaction of NS4A with MAVS is conserved among flaviviruses. The increased understanding of the role of NS4A in flavivirus immune evasion could aid the development of vaccines and therapeutic strategies.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Flavivirus , Helicase IFIH1 Induzida por Interferon , Interferon beta , Transdução de Sinais , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Flavivirus/imunologia , Flavivirus/genética , Flavivirus/fisiologia , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/imunologia , Células HEK293 , Evasão da Resposta Imune , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/virologia , Interações Hospedeiro-Patógeno/imunologia , Ligação Proteica , Imunidade Inata , Animais
16.
Proc Natl Acad Sci U S A ; 121(29): e2317977121, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38990941

RESUMO

In a recent characterization of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) variability present in 30 diagnostic samples from patients of the first COVID-19 pandemic wave, 41 amino acid substitutions were documented in the RNA-dependent RNA polymerase (RdRp) nsp12. Eight substitutions were selected in this work to determine whether they had an impact on the RdRp activity of the SARS-CoV-2 nsp12-nsp8-nsp7 replication complex. Three of these substitutions were found around the polymerase central cavity, in the template entry channel (D499G and M668V), and within the motif B (V560A), and they showed polymerization rates similar to the wild type RdRp. The remaining five mutations (P323L, L372F, L372P, V373A, and L527H) were placed near the nsp12-nsp8F contact surface; residues L372, V373, and L527 participated in a large hydrophobic cluster involving contacts between two helices in the nsp12 fingers and the long α-helix of nsp8F. The presence of any of these five amino acid substitutions resulted in important alterations in the RNA polymerization activity. Comparative primer elongation assays showed different behavior depending on the hydrophobicity of their side chains. The substitution of L by the bulkier F side chain at position 372 slightly promoted RdRp activity. However, this activity was dramatically reduced with the L372P, and L527H mutations, and to a lesser extent with V373A, all of which weaken the hydrophobic interactions within the cluster. Additional mutations, specifically designed to disrupt the nsp12-nsp8F interactions (nsp12-V330S, nsp12-V341S, and nsp8-R111A/D112A), also resulted in an impaired RdRp activity, further illustrating the importance of this contact interface in the regulation of RNA synthesis.


Assuntos
Mutação Puntual , RNA Viral , SARS-CoV-2 , Proteínas não Estruturais Virais , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , RNA Viral/genética , RNA Viral/metabolismo , Humanos , RNA-Polimerase RNA-Dependente de Coronavírus/genética , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Polimerização , COVID-19/virologia , Substituição de Aminoácidos , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Modelos Moleculares
18.
ACS Chem Biol ; 19(7): 1648-1660, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38954741

RESUMO

Hepatitis C virus (HCV) is a positive-stranded RNA virus that mainly causes chronic hepatitis, cirrhosis and hepatocellular carcinoma. Recently we confirmed m5C modifications within NS5A gene of HCV RNA genome. However, the roles of the m5C modification and its interaction with host proteins in regulating HCV's life cycle, remain unexplored. Here, we demonstrate that HCV infection enhances the expression of the host m5C reader YBX1 through the transcription factor MAX. YBX1 acts as an m5C reader, recognizing the m5C-modified NS5A C7525 site in the HCV RNA genome and significantly enhancing HCV RNA stability. This m5C-modification is also required for YBX1 colocalization with lipid droplets and HCV Core protein. Moreover, YBX1 facilitates HCV RNA replication, as well as viral assembly/budding. The tryptophan residue at position 65 (W65) of YBX1 is critical for these functions. Knockout of YBX1 or the application of YBX1 inhibitor SU056 suppresses HCV RNA replication and viral protein translation. To our knowledge, this is the first report demonstrating that the interaction between host m5C reader YBX1 and HCV RNA m5C methylation facilitates viral replication. Therefore, hepatic-YBX1 knockdown holds promise as a potential host-directed strategy for HCV therapy.


Assuntos
Hepacivirus , RNA Viral , Replicação Viral , Proteína 1 de Ligação a Y-Box , Hepacivirus/fisiologia , Hepacivirus/efeitos dos fármacos , Proteína 1 de Ligação a Y-Box/metabolismo , Humanos , Replicação Viral/efeitos dos fármacos , RNA Viral/metabolismo , RNA Viral/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Replicação do RNA , RNA Polimerase Dependente de RNA
19.
J Virol ; 98(8): e0085824, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39078257

RESUMO

Japanese encephalitis virus (JEV) is an arthropod-borne, plus-strand flavivirus causing viral encephalitis in humans with a high case fatality rate. The JEV non-structural protein 5 (NS5) with the RNA-dependent RNA polymerase activity interacts with the viral and host proteins to constitute the replication complex. We have identified the multifunctional protein Nucleolin (NCL) as one of the several NS5-interacting host proteins. We demonstrate the interaction and colocalization of JEV NS5 with NCL in the virus-infected HeLa cells. The siRNA-mediated knockdown of NCL indicated that it was required for efficient viral replication. Importantly, JEV grew to higher titers in cells over-expressing exogenous NCL, demonstrating its pro-viral role. We demonstrated that NS5 interacted with the RRM and GAR domains of NCL. We show that the NCL-binding aptamer AS1411 containing the G-quadruplex (GQ) structure and the GQ ligand BRACO-19 caused significant inhibition of JEV replication. The antiviral effect of AS1411 and BRACO-19 could be overcome in HeLa cells by the overexpression of exogenous NCL. We demonstrated that the synthetic RNAs derived from the 3'-NCR of JEV genomic RNA containing the GQ sequence could bind NCL in vitro. The replication complex binding to the 3'-NCR is required for the viral RNA synthesis. It is likely that NCL present in the replication complex destabilizes the GQ structures in the genomic RNA, thus facilitating the movement of the replication complex resulting in efficient virus replication.IMPORTANCEJapanese encephalitis virus (JEV) is endemic in most parts of South-East Asia and the Western Pacific region, causing epidemics of encephalitis with a high case fatality rate. While a tissue culture-derived JEV vaccine is available, no antiviral therapy exists. The JEV NS5 protein has RNA-dependent RNA polymerase activity. Together with several host and viral proteins, it constitutes the replication complex necessary for virus replication. Understanding the interaction of NS5 with the host proteins could help design novel antivirals. We identified Nucleolin (NCL) as a crucial host protein interactor of JEV NS5 having a pro-viral role in virus replication. The NS5-interacting NCL binds to the G-quadruplex (GQ) structure sequence in the 3'-NCR of JEV RNA. This may smoothen the movement of the replication complex along the genomic RNA, thereby facilitating the virus replication. This study is the first report on how NCL, a host protein, helps in JEV replication through GQ-binding.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Nucleolina , Fosfoproteínas , Proteínas de Ligação a RNA , Proteínas não Estruturais Virais , Replicação Viral , Humanos , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Células HeLa , Ligação Proteica , Encefalite Japonesa/virologia , Encefalite Japonesa/metabolismo , Interações Hospedeiro-Patógeno , Quadruplex G , Animais
20.
J Phys Chem B ; 128(29): 7033-7042, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39007765

RESUMO

Experimental evidence has established that SARS-CoV-2 NSP1 acts as a factor that restricts cellular gene expression and impedes mRNA translation within the ribosome's 40S subunit. However, the precise molecular mechanisms underlying this phenomenon have remained elusive. To elucidate this issue, we employed a combination of all-atom steered molecular dynamics and coarse-grained alchemical simulations to explore the binding affinity of mRNA to the 40S ribosome, both in the presence and absence of SARS-CoV-2 NSP1. Our investigations revealed that the binding of SARS-CoV-2 NSP1 to the 40S ribosome leads to a significant enhancement in the binding affinity of mRNA. This observation, which aligns with experimental findings, strongly suggests that SARS-CoV-2 NSP1 has the capability to inhibit mRNA translation. Furthermore, we identified electrostatic interactions between mRNA and the 40S ribosome as the primary driving force behind mRNA translation. Notably, water molecules were found to play a pivotal role in stabilizing the mRNA-40S ribosome complex, underscoring their significance in this process. We successfully pinpointed the specific SARS-CoV-2 NSP1 residues that play a critical role in triggering the translation arrest.


Assuntos
Simulação de Dinâmica Molecular , Biossíntese de Proteínas , RNA Mensageiro , Subunidades Ribossômicas Menores de Eucariotos , SARS-CoV-2 , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/química , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/química , SARS-CoV-2/metabolismo , SARS-CoV-2/química , SARS-CoV-2/genética , Ligação Proteica , Humanos , Eletricidade Estática
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA