Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.886
Filtrar
1.
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
2.
BMC Infect Dis ; 24(1): 688, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987682

RESUMO

BACKGROUND: Dengue fever has become a significant worldwide health concern, because of its high morbidity rate and the potential for an increase in mortality rates due to lack of adequate treatment. There is an immediate need for the development of effective medication for dengue fever. METHODS: Homology modeling of dengue virus (DENV) non-structural 4B (NS4B) protein was performed by SWISS-MODEL to predict the 3D structure of the protein. Structure validation was conducted using PROSA, PROCHECK, Ramachandran plot, and VERIFY-3D. MOE software was used to find out the in-Silico inhibitory potential of the five triterpenoids against the DENV-NS4B protein. RESULTS: The SWISS-MODEL was employed to predict the three-dimensional protein structure of the NS4B protein. Through molecular docking, it was found that the chosen triterpenoid NS4B protein had a high binding affinity interaction. It was observed that the NS4B protein binding energy for 15-oxoursolic acid, betulinic acid, ursolic acid, lupeol, and 3-o-acetylursolic acid were - 7.18, - 7.02, - 5.71, - 6.67 and - 8.00 kcal/mol, respectively. CONCLUSIONS: NS4B protein could be a promising target which showed good interaction with tested triterpenoids which can be developed as a potential antiviral drug for controlling dengue virus pathogenesis by inhibiting viral replication. However, further investigations are necessary to validate and confirm their efficacy.


Assuntos
Antivirais , Vírus da Dengue , Simulação de Acoplamento Molecular , Triterpenos , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Triterpenos/farmacologia , Triterpenos/química , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/química , Antivirais/farmacologia , Antivirais/química , Ligação Proteica , Humanos , Dengue/virologia , Dengue/tratamento farmacológico , Conformação Proteica , Proteínas de Membrana
3.
Virologie (Montrouge) ; 28(3): 187-197, 2024 Jun 01.
Artigo em Francês | MEDLINE | ID: mdl-38970340

RESUMO

Orthoflaviviruses are enveloped positive-sense RNA viruses comprising numerous human pathogens transmitted by hematophagous arthropods. This includes viruses such as dengue virus, Zika virus, and yellow fever virus. The viral nonstructural protein NS1 plays a central role in the pathogenesis and cycle of these viruses by acting in two different forms: associated with the plasma membrane (NS1m) or secreted outside the cell (NS1s). The versatility of NS1 is evident in its ability to modulate various aspects of the infectious process, from immune evasion to pathogenesis. As an intracellular protein, it disrupts many processes, interfering with signaling pathways and facilitating viral replication in concert with other viral proteins. As a secreted protein, NS1 actively participates in immune evasion, interfering with the host immune system, inhibiting the complement system, facilitating viral dissemination, and disrupting the integrity of endothelial barriers. This review primarily aims to address the role of NS1 in viral pathogenesis associated with orthoflaviviruses.


Assuntos
Proteínas não Estruturais Virais , Replicação Viral , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/fisiologia , Humanos , Animais , Infecções por Flavivirus/virologia , Evasão da Resposta Imune , Flavivirus/fisiologia , Flavivirus/patogenicidade , Zika virus/fisiologia , Zika virus/patogenicidade , Vírus da Dengue/fisiologia
5.
Viruses ; 16(6)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38932107

RESUMO

Rotaviruses (RVs) are 11-segmented, double-stranded (ds) RNA viruses and important causes of acute gastroenteritis in humans and other animal species. Early RV particle assembly is a multi-step process that includes the assortment, packaging and replication of the 11 genome segments in close connection with capsid morphogenesis. This process occurs inside virally induced, cytosolic, membrane-less organelles called viroplasms. While many viral and cellular proteins play roles during early RV assembly, the octameric nonstructural protein 2 (NSP2) has emerged as a master orchestrator of this key stage of the viral replication cycle. NSP2 is critical for viroplasm biogenesis as well as for the selective RNA-RNA interactions that underpin the assortment of 11 viral genome segments. Moreover, NSP2's associated enzymatic activities might serve to maintain nucleotide pools for use during viral genome replication, a process that is concurrent with early particle assembly. The goal of this review article is to summarize the available data about the structures, functions and interactions of RV NSP2 while also drawing attention to important unanswered questions in the field.


Assuntos
Genoma Viral , Rotavirus , Proteínas não Estruturais Virais , Montagem de Vírus , Replicação Viral , Rotavirus/genética , Rotavirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Humanos , Animais , RNA Viral/genética , RNA Viral/metabolismo , Capsídeo/metabolismo , Proteínas de Ligação a RNA
6.
Gigascience ; 132024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38869150

RESUMO

Viral helicases are promising targets for the development of antiviral therapies. Given their vital function of unwinding double-stranded nucleic acids, inhibiting them blocks the viral replication cycle. Previous studies have elucidated key structural details of these helicases, including the location of substrate binding sites, flexible domains, and the discovery of potential inhibitors. Here we present a series of new Galaxy tools and workflows for performing and analyzing molecular dynamics simulations of viral helicases. We first validate them by demonstrating recapitulation of data from previous simulations of Zika (NS3) and SARS-CoV-2 (NSP13) helicases in apo and complex with inhibitors. We further demonstrate the utility and generalizability of these Galaxy workflows by applying them to new cases, proving their usefulness as a widely accessible method for exploring antiviral activity.


Assuntos
Simulação de Dinâmica Molecular , SARS-CoV-2 , SARS-CoV-2/enzimologia , Zika virus/enzimologia , Fluxo de Trabalho , RNA Helicases/química , RNA Helicases/metabolismo , Humanos , DNA Helicases/química , DNA Helicases/metabolismo , Antivirais/química , Antivirais/farmacologia , Proteases Semelhantes à Papaína de Coronavírus/química , Proteases Semelhantes à Papaína de Coronavírus/metabolismo , Sítios de Ligação , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo
7.
Pol J Microbiol ; 73(2): 207-215, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38905281

RESUMO

Chikungunya virus (CHIKV) causes a debilitating fever and joint pain, with no specific antiviral treatment available. Halogenated secondary metabolites from plants are a promising new class of drug candidates against chikungunya, with unique properties that make them effective against the virus. Plants produce these compounds to defend themselves against pests and pathogens, and they are effective against a wide range of viruses, including chikungunya. This study investigated the interactions of halogenated secondary metabolites with nsP2pro, a therapeutic target for CHIKV. A library of sixty-six halogenated plant metabolites screened previously for ADME properties was used. Metabolites without violation of Lipinski's rule were docked with nsP2pro using AutoDock Vina. To find the stability of the pipoxide chlorohydrin-nsP2pro complex, the GROMACS suite was used for MD simulation. The binding free energy of the ligand-protein complex was computed using MMPBSA. Molecular docking studies revealed that halogenated metabolites interact with nsP2pro, suggesting they are possible inhibitors. Pipoxide chlorohydrin showed the greatest affinity to the target. This was further confirmed by the MD simulations, surface accessible area, and MMPBSA studies. Pipoxide chlorohydrin, a halogenated metabolite, was the most potent against nsP2pro in the survey.


Assuntos
Antivirais , Vírus Chikungunya , Simulação de Acoplamento Molecular , Vírus Chikungunya/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/química , Antivirais/metabolismo , Febre de Chikungunya/virologia , Febre de Chikungunya/tratamento farmacológico , Metabolismo Secundário , Simulação de Dinâmica Molecular , Halogenação , Plantas/química , Simulação por Computador , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química
8.
Int J Biol Macromol ; 272(Pt 1): 132855, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38834129

RESUMO

Approximately 3.9 billion individuals are vulnerable to dengue infection, a prevalent cause of tropical diseases worldwide. Currently, no drugs are available for preventing or treating Flavivirus diseases, including Dengue, West Nile, and the more recent Zika virus. The highly conserved Flavivirus NS2B-NS3 protease, crucial for viral replication, is a promising therapeutic target. This study employed in-silico methodologies to identify novel and potentially effective anti-dengue small molecules. A pharmacophore model was constructed using an experimentally validated NS2B-NS3 inhibitor, with the Gunner Henry score confirming the model's validity. The Natural Product Activity and Species Source (NPASS) database was screened using the validated pharmacophore model, yielding a total of 60 hits against the NS2B-NS3 protease. Furthermore, the docking finding reveals that our newly identified compounds from the NPASS database have enhanced binding affinities and established significant interactions with allosteric residues of the target protein. MD simulation and post-MD analysis further validated this finding. The free binding energy was computed in terms of MM-GBSA analysis, with the total binding energy for compound 1 (-57.3 ± 2.8 and - 52.9 ± 1.9 replica 1 and 2) indicating a stronger binding affinity for the target protein. Overall, this computational study identified these compounds as potential hit molecules, and these findings can open up a new avenue to explore and develop inhibitors against Dengue virus infection.


Assuntos
Antivirais , Vírus da Dengue , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteases , Serina Endopeptidases , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/enzimologia , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Antivirais/farmacologia , Antivirais/química , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Avaliação Pré-Clínica de Medicamentos , Ligação Proteica , Proteases Virais
9.
Math Biosci Eng ; 21(5): 5996-6018, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38872567

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS­CoV­2) has been evolving rapidly after causing havoc worldwide in 2020. Since then, it has been very hard to contain the virus owing to its frequently mutating nature. Changes in its genome lead to viral evolution, rendering it more resistant to existing vaccines and drugs. Predicting viral mutations beforehand will help in gearing up against more infectious and virulent versions of the virus in turn decreasing the damage caused by them. In this paper, we have proposed different NMT (neural machine translation) architectures based on RNNs (recurrent neural networks) to predict mutations in the SARS-CoV-2-selected non-structural proteins (NSP), i.e., NSP1, NSP3, NSP5, NSP8, NSP9, NSP13, and NSP15. First, we created and pre-processed the pairs of sequences from two languages using k-means clustering and nearest neighbors for training a neural translation machine. We also provided insights for training NMTs on long biological sequences. In addition, we evaluated and benchmarked our models to demonstrate their efficiency and reliability.


Assuntos
COVID-19 , Genoma Viral , Mutação , Redes Neurais de Computação , SARS-CoV-2 , Proteínas não Estruturais Virais , SARS-CoV-2/genética , Humanos , COVID-19/virologia , COVID-19/transmissão , Proteínas não Estruturais Virais/genética , Algoritmos
10.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38892307

RESUMO

Carnivorous pitcher plants from the genus Nepenthes are renowned for their ethnobotanical uses. This research explores the therapeutic potential of Nepenthes miranda leaf extract against nonstructural protein 9 (Nsp9) of SARS-CoV-2 and in treating human non-small cell lung carcinoma (NSCLC) cell lines. Nsp9, essential for SARS-CoV-2 RNA replication, was expressed and purified, and its interaction with ssDNA was assessed. Initial tests with myricetin and oridonin, known for targeting ssDNA-binding proteins and Nsp9, respectively, did not inhibit the ssDNA-binding activity of Nsp9. Subsequent screenings of various N. miranda extracts identified those using acetone, methanol, and ethanol as particularly effective in disrupting Nsp9's ssDNA-binding activity, as evidenced by electrophoretic mobility shift assays. Molecular docking studies highlighted stigmast-5-en-3-ol and lupenone, major components in the leaf extract of N. miranda, as potential inhibitors. The cytotoxic properties of N. miranda leaf extract were examined across NSCLC lines H1975, A549, and H838, focusing on cell survival, apoptosis, and migration. Results showed a dose-dependent cytotoxic effect in the following order: H1975 > A549 > H838 cells, indicating specificity. Enhanced anticancer effects were observed when the extract was combined with afatinib, suggesting synergistic interactions. Flow cytometry indicated that N. miranda leaf extract could induce G2 cell cycle arrest in H1975 cells, potentially inhibiting cancer cell proliferation. Gas chromatography-mass spectrometry (GC-MS) enabled the tentative identification of the 19 most abundant compounds in the leaf extract of N. miranda. These outcomes underscore the dual utility of N. miranda leaf extract in potentially managing SARS-CoV-2 infection through Nsp9 inhibition and offering anticancer benefits against lung carcinoma. These results significantly broaden the potential medical applications of N. miranda leaf extract, suggesting its use not only in traditional remedies but also as a prospective treatment for pulmonary diseases. Overall, our findings position the leaf extract of N. miranda as a promising source of natural compounds for anticancer therapeutics and antiviral therapies, warranting further investigation into its molecular mechanisms and potential clinical applications.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Simulação de Acoplamento Molecular , Extratos Vegetais , Folhas de Planta , SARS-CoV-2 , Humanos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/metabolismo , Folhas de Planta/química , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Linhagem Celular Tumoral , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/antagonistas & inibidores , Células A549 , Tratamento Farmacológico da COVID-19 , COVID-19/virologia , COVID-19/metabolismo , Apoptose/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/química
11.
Front Immunol ; 15: 1395809, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38938568

RESUMO

Human respiratory viruses are the most prevalent cause of disease in humans, with the highly infectious RSV being the leading cause of infant bronchiolitis and viral pneumonia. Responses to type I IFNs are the primary defense against viral infection. However, RSV proteins have been shown to antagonize type I IFN-mediated antiviral innate immunity, specifically dampening intracellular IFN signaling. Respiratory epithelial cells are the main target for RSV infection. In this study, we found RSV-NS1 interfered with the IFN-α JAK/STAT signaling pathway of epithelial cells. RSV-NS1 expression significantly enhanced IFN-α-mediated phosphorylation of STAT1, but not pSTAT2; and neither STAT1 nor STAT2 total protein levels were affected by RSV-NS1. However, expression of RSV-NS1 significantly reduced ISRE and GAS promoter activity and anti-viral IRG expression. Further mechanistic studies demonstrated RSV-NS1 bound STAT1, with protein modeling indicating a possible interaction site between STAT1 and RSV-NS1. Nuclear translocation of STAT1 was reduced in the presence of RSV-NS1. Additionally, STAT1's interaction with the nuclear transport adapter protein, KPNA1, was also reduced, suggesting a mechanism by which RSV blocks STAT1 nuclear translocation. Indeed, reducing STAT1's access to the nucleus may explain RSV's suppression of IFN JAK/STAT promoter activation and antiviral gene induction. Taken together these results describe a novel mechanism by which RSV controls antiviral IFN-α JAK/STAT responses, which enhances our understanding of RSV's respiratory disease progression.


Assuntos
Interferon-alfa , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Fator de Transcrição STAT1 , Transdução de Sinais , Proteínas não Estruturais Virais , Fator de Transcrição STAT1/metabolismo , Humanos , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Interferon-alfa/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/fisiologia , Proteínas não Estruturais Virais/metabolismo , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Janus Quinases/metabolismo , Núcleo Celular/metabolismo , Fosforilação , Transporte Ativo do Núcleo Celular , Linhagem Celular
12.
Nat Commun ; 15(1): 5426, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926343

RESUMO

Zika and dengue virus nonstructural protein 5 antagonism of STAT2, a critical interferon signaling transcription factor, to suppress the host interferon response is required for viremia and pathogenesis in a vertebrate host. This affects viral species tropism, as mouse STAT2 resistance renders only immunocompromised or humanized STAT2 mice infectable. Here, we explore how STAT2 evolution impacts antagonism. By measuring the susceptibility of 38 diverse STAT2 proteins, we demonstrate that resistance arose numerous times in mammalian evolution. In four species, resistance requires distinct sets of multiple amino acid changes that often individually disrupt STAT2 signaling. This reflects an evolutionary ridge where progressive resistance is balanced by the need to maintain STAT2 function. Furthermore, resistance may come with a fitness cost, as resistance that arose early in lemur evolution was subsequently lost in some lemur lineages. These findings underscore that while it is possible to evolve resistance to antagonism, complex evolutionary trajectories are required to avoid detrimental host fitness consequences.


Assuntos
Evolução Molecular , Fator de Transcrição STAT2 , Proteínas não Estruturais Virais , Fator de Transcrição STAT2/metabolismo , Fator de Transcrição STAT2/genética , Animais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Humanos , Camundongos , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Zika virus/genética , Flavivirus/genética , Flavivirus/fisiologia , Filogenia , Interações Hospedeiro-Patógeno/genética
13.
Viruses ; 16(6)2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38932114

RESUMO

When designing live-attenuated respiratory syncytial virus (RSV) vaccine candidates, attenuating mutations can be developed through biologic selection or reverse-genetic manipulation and may include point mutations, codon and gene deletions, and genome rearrangements. Attenuation typically involves the reduction in virus replication, due to direct effects on viral structural and replicative machinery or viral factors that antagonize host defense or cause disease. However, attenuation must balance reduced replication and immunogenic antigen expression. In the present study, we explored a new approach in order to discover attenuating mutations. Specifically, we used protein structure modeling and computational methods to identify amino acid substitutions in the RSV nonstructural protein 1 (NS1) predicted to cause various levels of structural perturbation. Twelve different mutations predicted to alter the NS1 protein structure were introduced into infectious virus and analyzed in cell culture for effects on viral mRNA and protein expression, interferon and cytokine expression, and caspase activation. We found the use of structure-based machine learning to predict amino acid substitutions that reduce the thermodynamic stability of NS1 resulted in various levels of loss of NS1 function, exemplified by effects including reduced multi-cycle viral replication in cells competent for type I interferon, reduced expression of viral mRNAs and proteins, and increased interferon and apoptosis responses.


Assuntos
Aprendizado de Máquina , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Proteínas não Estruturais Virais , Replicação Viral , Humanos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas contra Vírus Sincicial Respiratório/genética , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/imunologia , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/genética , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Substituição de Aminoácidos , Mutação , Linhagem Celular
14.
Vet Res ; 55(1): 79, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38886840

RESUMO

Porcine deltacoronavirus (PDCoV) is an enteropathogenic coronavirus that has been reported to use various strategies to counter the host antiviral innate immune response. The cGAS-STING signalling pathway plays an important role in antiviral innate immunity. However, it remains unclear whether PDCoV achieves immune evasion by regulating the cGAS-STING pathway. Here, we demonstrated that the nonstructural protein 2 (nsp2) encoded by PDCoV inhibits cGAS-STING-mediated type I and III interferon (IFN) responses via the regulation of porcine STING (pSTING) stability. Mechanistically, ectopically expressed PDCoV nsp2 was found to interact with the N-terminal region of pSTING. Consequently, pSTING was degraded through K48-linked ubiquitination and the proteasomal pathway, leading to the disruption of cGAS-STING signalling. Furthermore, K150 and K236 of pSTING were identified as crucial residues for nsp2-mediated ubiquitination and degradation. In summary, our findings provide a basis for elucidating the immune evasion mechanism of PDCoV and will contribute to the development of targets for anti-coronavirus drugs.


Assuntos
Deltacoronavirus , Proteínas não Estruturais Virais , Animais , Suínos , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Deltacoronavirus/genética , Deltacoronavirus/fisiologia , Doenças dos Suínos/virologia , Doenças dos Suínos/imunologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/metabolismo , Interferon Tipo I/metabolismo , Interferon Tipo I/genética , Imunidade Inata , Células HEK293 , Evasão da Resposta Imune , Ubiquitinação
15.
PLoS Pathog ; 20(6): e1012329, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38900816

RESUMO

Coronavirus (CoV) nonstructural protein 1 (nsp1) is considered a pathogenic factor due to its ability to inhibit host antiviral responses by inducing general shutoff of host protein synthesis. Nsp1 is expressed by α- and ß-CoVs, but its functions and strategies to induce host shutoff are not fully elucidated. We compared the nsp1s from two ß-CoVs (SARS-CoV and SARS-CoV-2) and two α-CoVs (NL63 and 229E) and found that NL63 nsp1 has the strongest shutoff activity. Unlike SARS-CoV nsp1s, which bind to 40S ribosomes and block translation of cellular mRNA, NL63 nsp1 did not inhibit translation of mRNAs transfected into cells. Instead, NL63 nsp1 localized to the nucleus and specifically inhibited transcription of genes under an RNA polymerase II (RNAPII) promoter. Further analysis revealed that NL63 nsp1 induces degradation of the largest subunit of RNAPII, Rpb1. This degradation was detected regardless of the phosphorylation state of Rpb1 and was blocked by the proteasome inhibitor MG132. We also found that Rpb1 was ubiquitinated in NL63-infected cells, and inhibition of ubiquitination by a ubiquitin activating enzyme inhibitor (TAK243) prevented degradation of Rpb1 in virus-infected cells. These data reveal an unrecognized strategy of host shutoff by human α-CoV NL63: targeting host transcription by inducing Rpb1 degradation to prevent host protein expression. Our study indicates that viruses within the same family can use completely distinct mechanisms to regulate host antiviral responses.


Assuntos
Biossíntese de Proteínas , RNA Polimerase II , Proteínas não Estruturais Virais , Humanos , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , RNA Polimerase II/metabolismo , Coronavirus Humano NL63/metabolismo , SARS-CoV-2 , Células HEK293
16.
Int J Mol Sci ; 25(12)2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38928101

RESUMO

In our prior investigations, we elucidated the role of the tryptophan-to-tyrosine substitution at the 61st position in the nonstructural protein NSsW61Y in diminishing the interaction between nonstructural proteins (NSs) and nucleoprotein (NP), impeding viral replication. In this study, we focused on the involvement of NSs in replication via the modulation of autophagosomes. Initially, we examined the impact of NP expression levels, a marker for replication, upon the infection of HeLa cells with severe fever thrombocytopenia syndrome virus (SFTSV), with or without the inhibition of NP binding. Western blot analysis revealed a reduction in NP levels in NSsW61Y-expressing conditions. Furthermore, the expression levels of the canonical autophagosome markers p62 and LC3 decreased in HeLa cells expressing NSsW61Y, revealing the involvement of individual viral proteins on autophagy. Subsequent experiments confirmed that NSsW61Y perturbs autophagy flux, as evidenced by reduced levels of LC3B and p62 upon treatment with chloroquine, an inhibitor of autophagosome-lysosome fusion. LysoTracker staining demonstrated a decrease in lysosomes in cells expressing the NS mutant compared to those expressing wild-type NS. We further explored the mTOR-associated regulatory pathway, a key regulator affected by NS mutant expression. The observed inhibition of replication could be linked to conformational changes in the NSs, impairing their binding to NP and altering mTOR regulation, a crucial upstream signaling component in autophagy. These findings illuminate the intricate interplay between NSsW61Y and the suppression of host autophagy machinery, which is crucial for the generation of autophagosomes to facilitate viral replication.


Assuntos
Autofagossomos , Autofagia , Phlebovirus , Triptofano , Tirosina , Proteínas não Estruturais Virais , Replicação Viral , Humanos , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Replicação Viral/genética , Autofagossomos/metabolismo , Células HeLa , Phlebovirus/genética , Phlebovirus/fisiologia , Phlebovirus/metabolismo , Autofagia/genética , Tirosina/metabolismo , Triptofano/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Mutação , Substituição de Aminoácidos , Febre Grave com Síndrome de Trombocitopenia/metabolismo , Febre Grave com Síndrome de Trombocitopenia/virologia , Febre Grave com Síndrome de Trombocitopenia/genética , Lisossomos/metabolismo , Nucleoproteínas/metabolismo , Nucleoproteínas/genética
18.
PLoS Pathog ; 20(6): e1012355, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38935808

RESUMO

Stress granules (SGs), formed by untranslated messenger ribonucleoproteins (mRNPs) during cellular stress in eukaryotes, have been linked to flavivirus interference without clear understanding. This study reveals the role of Zika virus (ZIKV) NS2B as a scaffold protein mediating interaction between protein phosphatase 1α (PP1α) and eukaryotic initiation factor 2α (eIF2α). This interaction promotes eIF2α dephosphorylation by PP1α, inhibiting SG formation. The NS2B-PP1α complex exhibits remarkable stability, resisting ubiquitin-induced degradation and amplifying eIF2α dephosphorylation, thus promoting ZIKV replication. In contrast, the NS2BV35A mutant, interacting exclusively with eIF2α, fails to inhibit SG formation, resulting in reduced viral replication and diminished impact on brain organoid growth. These findings reveal PP1α's dual role in ZIKV infection, inducing interferon production as an antiviral factor and suppressing SG formation as a viral promoter. Moreover, we found that NS2B also serves as a versatile mechanism employed by flaviviruses to counter host antiviral defenses, primarily by broadly inhibiting SG formation. This research advances our comprehension of the complex interplay in flavivirus-host interactions, offering potential for innovative therapeutic strategies against flavivirus infections.


Assuntos
Fator de Iniciação 2 em Eucariotos , Proteína Fosfatase 1 , Grânulos de Estresse , Proteínas não Estruturais Virais , Replicação Viral , Infecção por Zika virus , Zika virus , Zika virus/fisiologia , Replicação Viral/fisiologia , Humanos , Infecção por Zika virus/virologia , Infecção por Zika virus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Proteína Fosfatase 1/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Grânulos de Estresse/metabolismo , Animais
19.
Virulence ; 15(1): 2359470, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38918890

RESUMO

Influenza A virus (IAV) is the leading cause of highly contagious respiratory infections, which poses a serious threat to public health. The non-structural protein 1 (NS1) is encoded by segment 8 of IAV genome and is expressed in high levels in host cells upon IAV infection. It is the determinant of virulence and has multiple functions by targeting type Ι interferon (IFN-I) and type III interferon (IFN-III) production, disrupting cell apoptosis and autophagy in IAV-infected cells, and regulating the host fitness of influenza viruses. This review will summarize the current research on the NS1 including the structure and related biological functions of the NS1 as well as the interaction between the NS1 and host cells. It is hoped that this will provide some scientific basis for the prevention and control of the influenza virus.


Assuntos
Vírus da Influenza A , Influenza Humana , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Animais , Autofagia , Virulência , Interações Hospedeiro-Patógeno , Apoptose , Interferons/metabolismo , Interferons/imunologia , Interferons/genética
20.
Vet Microbiol ; 295: 110137, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38851153

RESUMO

Porcine deltacoronavirus (PDCoV) is an emergent enteric coronavirus, primarily inducing diarrhea in swine, particularly in nursing piglets, with the additional potential for zoonotic transmission to humans. Despite the significant impact of PDCoV on swine populations, its pathogenic mechanisms remain incompletely understood. Complement component 3 (C3) plays a pivotal role in the prevention of viral infections, however, there are no reports concerning the influence of C3 on the proliferation of PDCoV. In this study, we initially demonstrated that PDCoV is capable of activating the C3 and eliciting inflammatory responses. The overexpression of C3 significantly suppressed PDCoV replication, while inhibition of C3 expression facilitated PDCoV replication. We discovered that nonstructural proteins Nsp7, Nsp14, and M, considerably stimulated C3 expression, particularly Nsp14, through activation of the p38-MAPK-C/EBP-ß pathway. The N7-MTase constitutes a significant functional domain of the non-structural protein Nsp14, which is more obvious to upregulate C3. Furthermore, functional mutants of the N7-MTase domain suggested that the D44 and T135 of N7-Mtase constituted a pivotal amino acid site to promote C3 expression. This provides fresh insights into comprehending how the virus manipulates the host immune response and suggests potential antiviral strategies against PDCoV.


Assuntos
Complemento C3 , Deltacoronavirus , Proteínas não Estruturais Virais , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Complemento C3/genética , Complemento C3/metabolismo , Complemento C3/imunologia , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Suínos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Deltacoronavirus/genética , Doenças dos Suínos/virologia , Doenças dos Suínos/genética , Infecções por Coronavirus/virologia , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/imunologia , Sistema de Sinalização das MAP Quinases , Humanos , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA