Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23.202
Filtrar
1.
J Med Virol ; 96(7): e29752, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38949191

RESUMO

Antiviral signaling, immune response and cell metabolism are dysregulated by SARS-CoV-2, the causative agent of COVID-19. Here, we show that SARS-CoV-2 accessory proteins ORF3a, ORF9b, ORF9c and ORF10 induce a significant mitochondrial and metabolic reprogramming in A549 lung epithelial cells. While ORF9b, ORF9c and ORF10 induced largely overlapping transcriptomes, ORF3a induced a distinct transcriptome, including the downregulation of numerous genes with critical roles in mitochondrial function and morphology. On the other hand, all four ORFs altered mitochondrial dynamics and function, but only ORF3a and ORF9c induced a marked alteration in mitochondrial cristae structure. Genome-Scale Metabolic Models identified both metabolic flux reprogramming features both shared across all accessory proteins and specific for each accessory protein. Notably, a downregulated amino acid metabolism was observed in ORF9b, ORF9c and ORF10, while an upregulated lipid metabolism was distinctly induced by ORF3a. These findings reveal metabolic dependencies and vulnerabilities prompted by SARS-CoV-2 accessory proteins that may be exploited to identify new targets for intervention.


Assuntos
COVID-19 , Mitocôndrias , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Mitocôndrias/metabolismo , COVID-19/metabolismo , COVID-19/virologia , COVID-19/patologia , Células A549 , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Virais Reguladoras e Acessórias/genética , Transcriptoma , Fases de Leitura Aberta , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Viroporinas
2.
Int J Mol Sci ; 25(11)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38891850

RESUMO

Clostridioides difficile is a causative agent of antibiotic-associated diarrhea as well as pseudomembranous colitis. So far, all known bacteriophages infecting these bacteria are temperate, which means that instead of prompt lysis of host cells, they can integrate into the host genome or replicate episomally. While C. difficile phages are capable of spontaneous induction and entering the lytic pathway, very little is known about the regulation of their maintenance in the state of lysogeny. In this study, we investigated the properties of a putative major repressor of the recently characterized C. difficile phiCDKH01 bacteriophage. A candidate protein belongs to the XRE family and controls the transcription of genes encoding putative phage antirepressors, known to be involved in the regulation of lytic development. Hence, the putative major phage repressor is likely to be responsible for maintenance of the lysogeny.


Assuntos
Bacteriófagos , Clostridioides difficile , Lisogenia , Clostridioides difficile/virologia , Bacteriófagos/genética , Bacteriófagos/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Genoma Viral
3.
Int J Mol Sci ; 25(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38892197

RESUMO

Viral pathogens pose a substantial threat to public health and necessitate the development of effective remediation and antiviral strategies. This short communication aimed to investigate the antiviral efficacy of disinfectants on the surface proteins of human pathogenic viruses. Using in silico modeling, the ligand-binding energies (LBEs) of selected disinfectants were predicted and combined with their environmental impacts and costs through an eco-pharmaco-economic analysis (EPEA). The results revealed that the binding affinities of chemical disinfectants to viral proteins varied significantly (p < 0.005). Rutin demonstrated promising broad-spectrum antiviral efficacy with an LBE of -8.49 ± 0.92 kcal/mol across all tested proteins. Additionally, rutin showed a superior eco-pharmaco-economic profile compared to the other chemicals, effectively balancing high antiviral effectiveness, moderate environmental impact, and affordability. These findings highlight rutin as a key phytochemical for use in remediating viral contaminants.


Assuntos
Antivirais , Desinfetantes , Rutina , Desinfetantes/farmacologia , Desinfetantes/química , Antivirais/farmacologia , Antivirais/química , Rutina/química , Rutina/farmacologia , Humanos , Simulação por Computador , Vírus/efeitos dos fármacos , Proteínas Virais/química , Proteínas Virais/metabolismo , Simulação de Acoplamento Molecular , Proteínas de Membrana/metabolismo , Proteínas de Membrana/química , Ligação Proteica
4.
Emerg Microbes Infect ; 13(1): 2369193, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38873898

RESUMO

The global outbreak of Mpox, caused by the monkeypox virus (MPXV), has attracted international attention and become another major infectious disease event after COVID-19. The mRNA cap N7 methyltransferase (RNMT) of MPXV methylates the N7 position of the added guanosine to the 5'-cap structure of mRNAs and plays a vital role in evading host antiviral immunity. MPXV RNMT is composed of the large subunit E1 and the small subunit E12. How E1 and E12 of MPXV assembly remains unclear. Here, we report the crystal structures of E12, the MTase domain of E1 with E12 (E1CTD-E12) complex, and the E1CTD-E12-SAM ternary complex, revealing the detailed conformations of critical residues and the structural changes upon E12 binding to E1. Functional studies suggest that E1CTD N-terminal extension (Asp545-Arg562) and the small subunit E12 play an essential role in the binding process of SAM. Structural comparison of the AlphaFold2-predicted E1, E1CTD-E12 complex, and the homologous D1-D12 complex of vaccinia virus (VACV) indicates an allosteric activating effect of E1 in MPXV. Our findings provide the structural basis for the MTase activity stimulation of the E1-E12 complex and suggest a potential interface for screening the anti-poxvirus inhibitors.


Assuntos
Metiltransferases , Monkeypox virus , Metiltransferases/química , Metiltransferases/metabolismo , Metiltransferases/genética , Monkeypox virus/genética , Monkeypox virus/enzimologia , Monkeypox virus/química , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo , Cristalografia por Raios X , Capuzes de RNA/metabolismo , Capuzes de RNA/química , Modelos Moleculares , Humanos , Conformação Proteica , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/química
5.
PLoS Pathog ; 20(6): e1012311, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38885273

RESUMO

The majority of rod-shaped and some filamentous plant viruses encode a cysteine-rich protein (CRP) that functions in viral virulence; however, the roles of these CRPs in viral infection remain largely unknown. Here, we used barley stripe mosaic virus (BSMV) as a model to investigate the essential role of its CRP in virus morphogenesis. The CRP protein γb directly interacts with BSMV coat protein (CP), the mutations either on the His-85 site in γb predicted to generate a potential CCCH motif or on the His-13 site in CP exposed to the surface of the virions abolish the zinc-binding activity and their interaction. Immunogold-labeling assays show that γb binds to the surface of rod-shaped BSMV virions in a Zn2+-dependent manner, which enhances the RNA binding activity of CP and facilitates virion assembly and stability, suggesting that the Zn2+-dependent physical association of γb with the virion is crucial for BSMV morphogenesis. Intriguingly, the tightly binding of diverse CRPs to their rod-shaped virions is a general feature employed by the members in the families Virgaviridae (excluding the genus Tobamovirus) and Benyviridae. Together, these results reveal a hitherto unknown role of CRPs in the assembly and stability of virus particles, and expand our understanding of the molecular mechanism underlying virus morphogenesis.


Assuntos
Vírion , Zinco , Zinco/metabolismo , Vírion/metabolismo , Proteínas do Capsídeo/metabolismo , Montagem de Vírus/fisiologia , Vírus de Plantas/metabolismo , Vírus de Plantas/fisiologia , Doenças das Plantas/virologia , Cisteína/metabolismo , Proteínas Virais/metabolismo , Morfogênese
6.
Adv Virus Res ; 119: 1-38, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38897707

RESUMO

The ubiquitination process is a reversible posttranslational modification involved in many essential cellular functions, such as innate immunity, cell signaling, trafficking, protein stability, and protein degradation. Viruses can use the ubiquitin system to efficiently enter host cells, replicate and evade host immunity, ultimately enhancing viral pathogenesis. Emerging evidence indicates that enveloped viruses can carry free (unanchored) ubiquitin or covalently ubiquitinated viral structural proteins that can increase the efficiency of viral entry into host cells. Furthermore, viruses continuously evolve and adapt to take advantage of the host ubiquitin machinery, highlighting its importance during virus infection. This review discusses the battle between viruses and hosts, focusing on how viruses hijack the ubiquitination process at different steps of the replication cycle, with a specific emphasis on viral entry. We discuss how ubiquitination of viral proteins may affect tropism and explore emerging therapeutics strategies targeting the ubiquitin system for antiviral drug discovery.


Assuntos
Ubiquitinação , Internalização do Vírus , Replicação Viral , Humanos , Ubiquitina/metabolismo , Vírus/metabolismo , Interações Hospedeiro-Patógeno , Proteínas Virais/metabolismo , Proteínas Virais/genética , Viroses/virologia , Viroses/imunologia , Viroses/metabolismo , Animais , Processamento de Proteína Pós-Traducional
7.
World J Microbiol Biotechnol ; 40(8): 256, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38926173

RESUMO

The analysis of transcriptional activity of the bacteriophage T5 hol/endo operon conducted in the paper revealed a strong constitutive promoter recognized by E. coli RNA polymerase and a transcription initiation point of the operon. It was also shown that the only translational start codon for holin was a non-canonical TTG. Translation initiation regions (TIRs) of both genes of the operon (hol and endo) were further analyzed using chimeric constructs, in which parts of the hol/endo regulatory regions were fused with the gene of a reporter protein (EGFP). It was found that TIR of hol was 20 times less effective than that of endo. As it turned out, the level of EGFP production was influenced by the composition of the constructs and the type of the hol start codon. Apparently, the translational suppression of holin's accumulation and posttranslational activation of endolysin by Ca2+ are the main factors ensuring the proper timing of the host cell lysis by bacteriophage T5. The approach based on the use of chimeric constructs proposed in the paper can be recommended for studying other native or artificial operons of any complexity: analyzing the impacts of separate DNA regions, as well as their coupled effect, on the processes of transcription and translation of recombinant protein(s).


Assuntos
Endopeptidases , Escherichia coli , Óperon , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Transcrição Gênica , Proteínas Virais , Endopeptidases/genética , Endopeptidases/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Escherichia coli/genética , Escherichia coli/virologia , Regulação Viral da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Códon de Iniciação/genética , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , DNA Viral/genética , Bacteriófagos/genética
8.
Biotechnol J ; 19(6): e2300736, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38900041

RESUMO

During plant-pathogen interaction, plant exhibits a strong defense system utilizing diverse groups of proteins to suppress the infection and subsequent establishment of the pathogen. However, in response, pathogens trigger an anti-silencing mechanism to overcome the host defense machinery. Among plant viruses, geminiviruses are the second largest virus family with a worldwide distribution and continue to be production constraints to food, feed, and fiber crops. These viruses are spread by a diverse group of insects, predominantly by whiteflies, and are characterized by a single-stranded DNA (ssDNA) genome coding for four to eight proteins that facilitate viral infection. The most effective means to managing these viruses is through an integrated disease management strategy that includes virus-resistant cultivars, vector management, and cultural practices. Dynamic changes in this virus family enable the species to manipulate their genome organization to respond to external changes in the environment. Therefore, the evolutionary nature of geminiviruses leads to new and novel approaches for developing virus-resistant cultivars and it is essential to study molecular ecology and evolution of geminiviruses. This review summarizes the multifunctionality of each geminivirus-encoded protein. These protein-based interactions trigger the abrupt changes in the host methyl cycle and signaling pathways that turn over protein normal production and impair the plant antiviral defense system. Studying these geminivirus interactions localized at cytoplasm-nucleus could reveal a more clear picture of host-pathogen relation. Data collected from this antagonistic relationship among geminivirus, vector, and its host, will provide extensive knowledge on their virulence mode and diversity with climate change.


Assuntos
Geminiviridae , Interações Hospedeiro-Patógeno , Doenças das Plantas , Proteínas Virais , Geminiviridae/genética , Geminiviridae/patogenicidade , Proteínas Virais/genética , Proteínas Virais/metabolismo , Doenças das Plantas/virologia , Animais , Plantas/virologia
9.
Annu Rev Immunol ; 42(1): 551-584, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38941604

RESUMO

Poxviruses have evolved a wide array of mechanisms to evade the immune response, and we provide an overview of the different immunomodulatory strategies. Poxviruses prevent the recognition of viral DNA that triggers the immune responses and inhibit signaling pathways within the infected cell. A unique feature of poxviruses is the production of secreted proteins that mimic cytokines and cytokine receptors, acting as decoy receptors to neutralize the activity of cytokines and chemokines. The capacity of these proteins to evade cellular immune responses by inhibiting cytokine activation is complemented by poxviruses' strategies to block natural killer cells and cytotoxic T cells, often through interfering with antigen presentation pathways. Mechanisms that target complement activation are also encoded by poxviruses. Virus-encoded proteins that target immune molecules and pathways play a major role in immune modulation, and their contribution to viral pathogenesis, facilitating virus replication or preventing immunopathology, is discussed.


Assuntos
Evasão da Resposta Imune , Infecções por Poxviridae , Poxviridae , Humanos , Poxviridae/imunologia , Poxviridae/fisiologia , Animais , Infecções por Poxviridae/imunologia , Citocinas/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Proteínas Virais/imunologia , Apresentação de Antígeno/imunologia , Interações Hospedeiro-Patógeno/imunologia
10.
Viruses ; 16(6)2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38932128

RESUMO

This study was conducted to efficiently produce virus-like particles (VLPs) of enterovirus 71 (EV71), a causative virus of hand, foot, and mouth disease (HFMD). The expression level of the P1 precursor, a structural protein of EV71, was modified to increase VLP production, and the optimal expression level and duration of the 3CD protein for P1 cleavage were determined. The expression level and duration of 3CD were controlled by the p10 promoter, which was weakened by repeated burst sequence (BS) applications, as well as the OpIE2 promoter, which was weakened by the insertion of random untranslated region sequences of various lengths. The cleavage and production efficiency of the P1 precursor were compared based on the expression time and level of 3CD, revealing that the p10-BS5 promoter with four repeated BSs was the most effective. When P1 and 3CD were expressed using the hyperexpression vector and the p10-BS5 promoter, high levels of structural protein production and normal HFMD-VLP formation were observed, respectively. This study suggests that the production efficiency of HFMD-VLPs can be significantly enhanced by increasing the expression of the P1 precursor and controlling the amount and duration of 3CD expression.


Assuntos
Enterovirus Humano A , Regiões Promotoras Genéticas , Enterovirus Humano A/genética , Enterovirus Humano A/fisiologia , Animais , Proteínas Virais/genética , Proteínas Virais/metabolismo , Humanos , Doença de Mão, Pé e Boca/virologia , Linhagem Celular , Células Sf9 , Vetores Genéticos/genética
11.
Viruses ; 16(6)2024 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-38932139

RESUMO

The viral interferon regulatory factors (vIRFs) of KSHV are known to dysregulate cell signaling pathways to promote viral oncogenesis and to block antiviral immune responses to facilitate infection. However, it remains unknown to what extent each vIRF plays a role in gene regulation. To address this, we performed a comparative analysis of the protein structures and gene regulation of the four vIRFs. Our structure prediction analysis revealed that despite their low amino acid sequence similarity, vIRFs exhibit high structural homology in both their DNA-binding domain (DBD) and IRF association domain. However, despite this shared structural homology, we demonstrate that each vIRF regulates a distinct set of KSHV gene promoters and human genes in epithelial cells. We also found that the DBD of vIRF1 is essential in regulating the expression of its target genes. We propose that the structurally similar vIRFs evolved to possess specialized transcriptional functions to regulate specific genes.


Assuntos
Células Epiteliais , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8 , Fatores Reguladores de Interferon , Proteínas Virais , Humanos , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Células Epiteliais/virologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Regiões Promotoras Genéticas , Transcrição Gênica , Genoma Viral , Linhagem Celular
12.
Viruses ; 16(6)2024 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-38932138

RESUMO

Viruses exploit the host cell machinery to enable infection and propagation. This review discusses the complex landscape of DNA virus-host interactions, focusing primarily on herpesviruses and adenoviruses, which replicate in the nucleus of infected cells, and vaccinia virus, which replicates in the cytoplasm. We discuss experimental approaches used to discover and validate interactions of host proteins with viral genomes and how these interactions impact processes that occur during infection, including the host DNA damage response and viral genome replication, repair, and transcription. We highlight the current state of knowledge regarding virus-host protein interactions and also outline emerging areas and future directions for research.


Assuntos
DNA Viral , Genoma Viral , Interações Hospedeiro-Patógeno , Replicação Viral , Humanos , DNA Viral/genética , DNA Viral/metabolismo , Vírus de DNA/genética , Animais , Proteínas Virais/metabolismo , Proteínas Virais/genética , Herpesviridae/genética , Herpesviridae/metabolismo , Herpesviridae/fisiologia , Vaccinia virus/genética
13.
Viruses ; 16(6)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38932171

RESUMO

Proteins of the Bcl-2 family regulate cellular fate via multiple mechanisms including apoptosis, autophagy, senescence, metabolism, inflammation, redox homeostasis, and calcium flux. There are several regulated cell death (RCD) pathways, including apoptosis and autophagy, that use distinct molecular mechanisms to elicit the death response. However, the same proteins/genes may be deployed in multiple biochemical pathways. In apoptosis, Bcl-2 proteins control the integrity of the mitochondrial outer membrane (MOM) by regulating the formation of pores in the MOM and apoptotic cell death. A number of prosurvival genes populate the genomes of viruses including those of the pro-survival Bcl-2 family. Viral Bcl-2 proteins are sequence and structural homologs of their cellular counterparts and interact with cellular proteins in apoptotic and autophagic pathways, potentially allowing them to modulate these pathways and determine cellular fate.


Assuntos
Apoptose , Autofagia , Vírus de DNA , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Virais , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Vírus de DNA/genética , Vírus de DNA/fisiologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Animais , Membranas Mitocondriais/metabolismo
14.
Viruses ; 16(6)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38932190

RESUMO

Human coronavirus 229E (HCoV-229E) is associated with upper respiratory tract infections and generally causes mild respiratory symptoms. HCoV-229E infection can cause cell death, but the molecular pathways that lead to virus-induced cell death as well as the interplay between viral proteins and cellular cell death effectors remain poorly characterized for HCoV-229E. Studying how HCoV-229E and other common cold coronaviruses interact with and affect cell death pathways may help to understand its pathogenesis and compare it to that of highly pathogenic coronaviruses. Here, we report that the main protease (Mpro) of HCoV-229E can cleave gasdermin D (GSDMD) at two different sites (Q29 and Q193) within its active N-terminal domain to generate fragments that are now unable to cause pyroptosis, a form of lytic cell death normally executed by this protein. Despite GSDMD cleavage by HCoV-229E Mpro, we show that HCoV-229E infection still leads to lytic cell death. We demonstrate that during virus infection caspase-3 cleaves and activates gasdermin E (GSDME), another key executioner of pyroptosis. Accordingly, GSDME knockout cells show a significant decrease in lytic cell death upon virus infection. Finally, we show that HCoV-229E infection leads to increased lytic cell death levels in cells expressing a GSDMD mutant uncleavable by Mpro (GSDMD Q29A+Q193A). We conclude that GSDMD is inactivated by Mpro during HCoV-229E infection, preventing GSDMD-mediated cell death, and point to the caspase-3/GSDME axis as an important player in the execution of virus-induced cell death. In the context of similar reported findings for highly pathogenic coronaviruses, our results suggest that these mechanisms do not contribute to differences in pathogenicity among coronaviruses. Nonetheless, understanding the interactions of common cold-associated coronaviruses and their proteins with the programmed cell death machineries may lead to new clues for coronavirus control strategies.


Assuntos
Morte Celular , Coronavirus Humano 229E , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Ligação a Fosfato , Piroptose , Humanos , Proteínas de Ligação a Fosfato/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Coronavirus Humano 229E/fisiologia , Coronavirus Humano 229E/genética , Infecções por Coronavirus/virologia , Infecções por Coronavirus/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Linhagem Celular , Interações Hospedeiro-Patógeno , Células HEK293 , Gasderminas
15.
Viruses ; 16(6)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38932209

RESUMO

A proteomics analysis of purified rabies virus (RABV) revealed 47 entrapped host proteins within the viral particles. Out of these, 11 proteins were highly disordered. Our study was particularly focused on five of the RABV-entrapped mouse proteins with the highest levels of disorder: Neuromodulin, Chmp4b, DnaJB6, Vps37B, and Wasl. We extensively utilized bioinformatics tools, such as FuzDrop, D2P2, UniProt, RIDAO, STRING, AlphaFold, and ELM, for a comprehensive analysis of the intrinsic disorder propensity of these proteins. Our analysis suggested that these disordered host proteins might play a significant role in facilitating the rabies virus pathogenicity, immune system evasion, and the development of antiviral drug resistance. Our study highlighted the complex interaction of the virus with its host, with a focus on how the intrinsic disorder can play a crucial role in virus pathogenic processes, and suggested that these intrinsically disordered proteins (IDPs) and disorder-related host interactions can also be a potential target for therapeutic strategies.


Assuntos
Proteínas Intrinsicamente Desordenadas , Vírus da Raiva , Vírion , Vírus da Raiva/fisiologia , Animais , Camundongos , Proteínas Intrinsicamente Desordenadas/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Vírion/metabolismo , Proteômica , Interações Hospedeiro-Patógeno , Raiva/virologia , Biologia Computacional/métodos , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/química
16.
Viruses ; 16(6)2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38932241

RESUMO

African swine fever (ASF) is an acute, hemorrhagic, highly contagious disease in pigs caused by African swine fever virus (ASFV). Our previous study identified that the ASFV MGF300-2R protein functions as a virulence factor and found that MGF300-2R degrades IKKß via selective autophagy. However, the E3 ubiquitin ligase responsible for IKKß ubiquitination during autophagic degradation still remains unknown. In order to solve this problem, we first pulled down 328 proteins interacting with MGF300-2R through immunoprecipitation-mass spectrometry. Next, we analyzed and confirmed the interaction between the E3 ubiquitin ligase TRIM21 and MGF300-2R and demonstrated the catalytic role of TRIM21 in IKKß ubiquitination. Finally, we indicated that the degradation of IKKß by MGF300-2R was dependent on TRIM21. In summary, our results indicate TRIM21 is the E3 ubiquitin ligase involved in the degradation of IKKß by MGF300-2R, thereby augmenting our understanding of the functions of MGF300-2R and offering insights into the rational design of live attenuated vaccines and antiviral strategies against ASF.


Assuntos
Vírus da Febre Suína Africana , Quinase I-kappa B , Ribonucleoproteínas , Ubiquitina-Proteína Ligases , Ubiquitinação , Proteínas Virais , Animais , Vírus da Febre Suína Africana/metabolismo , Vírus da Febre Suína Africana/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Suínos , Quinase I-kappa B/metabolismo , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Febre Suína Africana/virologia , Febre Suína Africana/metabolismo , Humanos , Células HEK293 , Interações Hospedeiro-Patógeno , Fatores de Virulência/metabolismo , Autofagia , Ligação Proteica
17.
Emerg Microbes Infect ; 13(1): 2368221, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38932432

RESUMO

A positive-sense (+) single-stranded RNA (ssRNA) virus (e.g. enterovirus A71, EV-A71) depends on viral polypeptide translation for initiation of virus replication after entry. We reported that EV-A71 hijacks Hsp27 to induce hnRNP A1 cytosol redistribution to initiate viral protein translation, but the underlying mechanism is still elusive. Here, we show that phosphorylation-deficient Hsp27-3A (Hsp27S15/78/82A) and Hsp27S78A fail to translocate into the nucleus and induce hnRNP A1 cytosol redistribution, while Hsp27S15A and Hsp27S82A display similar effects to the wild type Hsp27. Furthermore, we demonstrate that the viral 2A protease (2Apro) activity is a key factor in regulating Hsp27/hnRNP A1 relocalization. Hsp27S78A dramatically decreases the IRES activity and viral replication, which are partially reduced by Hsp27S82A. However, Hsp27S15A displays the same activity as the wild-type Hsp27. Peptide S78 potently suppresses EV-A71 protein translation and reproduction through blockage of EV-A71-induced Hsp27 phosphorylation and Hsp27/hnRNP A1 relocalization. A point mutation (S78A) on S78 impairs its inhibitory functions on Hsp27/hnRNP A1 relocalization and viral replication. Taken together, we demonstrate the importance of Ser78 phosphorylation of Hsp27 regulated by virus infection in nuclear translocation, hnRNP A1 cytosol relocation, and viral replication, suggesting a new path (such as peptide S78) for target-based antiviral strategy.


Assuntos
Enterovirus Humano A , Proteínas de Choque Térmico HSP27 , Ribonucleoproteína Nuclear Heterogênea A1 , Replicação Viral , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/fisiologia , Enterovirus Humano A/genética , Fosforilação , Humanos , Replicação Viral/efeitos dos fármacos , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1/genética , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP27/genética , Infecções por Enterovirus/virologia , Infecções por Enterovirus/metabolismo , Antivirais/farmacologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Serina/metabolismo , Células HeLa , Biossíntese de Proteínas , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/genética , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/genética , Proteínas de Choque Térmico
18.
Signal Transduct Target Ther ; 9(1): 159, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38937432

RESUMO

The ORF9b protein, derived from the nucleocapsid's open-reading frame in both SARS-CoV and SARS-CoV-2, serves as an accessory protein crucial for viral immune evasion by inhibiting the innate immune response. Despite its significance, the precise regulatory mechanisms underlying its function remain elusive. In the present study, we unveil that the ORF9b protein of SARS-CoV-2, including emerging mutant strains like Delta and Omicron, can undergo ubiquitination at the K67 site and subsequent degradation via the proteasome pathway, despite certain mutations present among these strains. Moreover, our investigation further uncovers the pivotal role of the translocase of the outer mitochondrial membrane 70 (TOM70) as a substrate receptor, bridging ORF9b with heat shock protein 90 alpha (HSP90α) and Cullin 5 (CUL5) to form a complex. Within this complex, CUL5 triggers the ubiquitination and degradation of ORF9b, acting as a host antiviral factor, while HSP90α functions to stabilize it. Notably, treatment with HSP90 inhibitors such as GA or 17-AAG accelerates the degradation of ORF9b, leading to a pronounced inhibition of SARS-CoV-2 replication. Single-cell sequencing data revealed an up-regulation of HSP90α in lung epithelial cells from COVID-19 patients, suggesting a potential mechanism by which SARS-CoV-2 may exploit HSP90α to evade the host immunity. Our study identifies the CUL5-TOM70-HSP90α complex as a critical regulator of ORF9b protein stability, shedding light on the intricate host-virus immune response dynamics and offering promising avenues for drug development against SARS-CoV-2 in clinical settings.


Assuntos
COVID-19 , Proteínas Culina , Proteínas de Choque Térmico HSP90 , SARS-CoV-2 , Ubiquitinação , Replicação Viral , Humanos , Proteínas Culina/genética , Proteínas Culina/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , COVID-19/virologia , COVID-19/genética , COVID-19/metabolismo , COVID-19/imunologia , Ubiquitinação/genética , Células HEK293 , Benzoquinonas/farmacologia , Estabilidade Proteica , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Lactamas Macrocíclicas
19.
Front Cell Infect Microbiol ; 14: 1394713, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38836054

RESUMO

The rabies virus enters the nervous system by interacting with several molecular targets on host cells to modify behavior and trigger receptor-mediated endocytosis of the virion by poorly understood mechanisms. The rabies virus glycoprotein (RVG) interacts with the muscle acetylcholine receptor and the neuronal α4ß2 subtype of the nicotinic acetylcholine receptor (nAChR) family by the putative neurotoxin-like motif. Given that the neurotoxin-like motif is highly homologous to the α7 nAChR subtype selective snake toxin α-bungarotoxin (αBTX), other nAChR subtypes are likely involved. The purpose of this study is to determine the activity of the RVG neurotoxin-like motif on nAChR subtypes that are expressed in brain regions involved in rabid animal behavior. nAChRs were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to collect concentration-response data to measure the functional effects. The RVG peptide preferentially and completely inhibits α7 nAChR ACh-induced currents by a competitive antagonist mechanism. Tested heteromeric nAChRs are also inhibited, but to a lesser extent than the α7 subtype. Residues of the RVG peptide with high sequence homology to αBTX and other neurotoxins were substituted with alanine. Altered RVG neurotoxin-like peptides showed that residues phenylalanine 192, arginine 196, and arginine 199 are important determinants of RVG peptide apparent potency on α7 nAChRs, while serine 195 is not. The evaluation of the rabies ectodomain reaffirmed the observations made with the RVG peptide, illustrating a significant inhibitory impact on α7 nAChR with potency in the nanomolar range. In a mammalian cell culture model of neurons, we confirm that the RVG peptide binds preferentially to cells expressing the α7 nAChR. Defining the activity of the RVG peptide on nAChRs expands our understanding of basic mechanisms in host-pathogen interactions that result in neurological disorders.


Assuntos
Glicoproteínas , Vírus da Raiva , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7 , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Vírus da Raiva/fisiologia , Vírus da Raiva/metabolismo , Humanos , Glicoproteínas/metabolismo , Glicoproteínas/genética , Oócitos/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Interações Hospedeiro-Patógeno , Ligação Proteica , Raiva/metabolismo , Raiva/virologia , Acetilcolina/metabolismo , Acetilcolina/farmacologia , Neurotoxinas/metabolismo , Neurotoxinas/farmacologia
20.
Molecules ; 29(11)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38893337

RESUMO

mRNA vaccines are entering a period of rapid development. However, their synthesis is still plagued by challenges related to mRNA impurities and fragments (incomplete mRNA). Most impurities of mRNA products transcribed in vitro are mRNA fragments. Only full-length mRNA transcripts containing both a 5'-cap and a 3'-poly(A) structure are viable for in vivo expression. Therefore, RNA fragments are the primary product-related impurities that significantly hinder mRNA efficacy and must be effectively controlled; these species are believed to originate from either mRNA hydrolysis or premature transcriptional termination. In the manufacturing of commercial mRNA vaccines, T7 RNA polymerase-catalyzed in vitro transcription (IVT) synthesis is a well-established method for synthesizing long RNA transcripts. This study identified a pivotal domain on the T7 RNA polymerase that is associated with erroneous mRNA release. By leveraging the advantageous properties of a T7 RNA polymerase mutant and precisely optimized IVT process parameters, we successfully achieved an mRNA integrity exceeding 91%, thereby further unlocking the immense potential of mRNA therapeutics.


Assuntos
RNA Polimerases Dirigidas por DNA , RNA Mensageiro , Transcrição Gênica , Proteínas Virais , RNA Mensageiro/genética , RNA Polimerases Dirigidas por DNA/metabolismo , RNA Polimerases Dirigidas por DNA/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vacinas de mRNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...