Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.368
Filtrar
1.
Nat Commun ; 15(1): 7486, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39209804

RESUMO

Chronic liver disease and cancer are global health challenges. The role of the circadian clock as a regulator of liver physiology and disease is well established in rodents, however, the identity and epigenetic regulation of rhythmically expressed genes in human disease is less well studied. Here we unravel the rhythmic transcriptome and epigenome of human hepatocytes using male human liver chimeric mice. We identify a large number of rhythmically expressed protein coding genes in human hepatocytes of male chimeric mice, which includes key transcription factors, chromatin modifiers, and critical enzymes. We show that hepatitis C virus (HCV) infection, a major cause of liver disease and cancer, perturbs the transcriptome by altering the rhythmicity of the expression of more than 1000 genes, and affects the epigenome, leading to an activation of critical pathways mediating metabolic alterations, fibrosis, and cancer. HCV-perturbed rhythmic pathways remain dysregulated in patients with advanced liver disease. Collectively, these data support a role for virus-induced perturbation of the hepatic rhythmic transcriptome and pathways in cancer development and may provide opportunities for cancer prevention and biomarkers to predict HCC risk.


Assuntos
Ritmo Circadiano , Hepacivirus , Hepatite C , Hepatócitos , Fígado , Transcriptoma , Humanos , Fígado/metabolismo , Fígado/virologia , Animais , Masculino , Hepatócitos/metabolismo , Hepatócitos/virologia , Camundongos , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatite C/genética , Hepatite C/metabolismo , Hepatite C/virologia , Ritmo Circadiano/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Neoplasias Hepáticas/metabolismo , Relógios Circadianos/genética , Epigênese Genética
2.
Viruses ; 16(8)2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39205194

RESUMO

The hepatitis C virus (HCV) co-opts many cellular factors-including proteins and microRNAs-to complete its life cycle. A cellular RNA-binding protein, poly(rC)-binding protein 2 (PCBP2), was previously shown to bind to the hepatitis C virus (HCV) genome; however, its precise role in the viral life cycle remained unclear. Herein, using the HCV cell culture (HCVcc) system and assays that isolate each step of the viral life cycle, we found that PCBP2 does not have a direct role in viral entry, translation, genome stability, or HCV RNA replication. Rather, our data suggest that PCBP2 depletion only impacts viral RNAs that can undergo genome packaging. Taken together, our data suggest that endogenous PCBP2 modulates the early steps of genome packaging, and therefore only has an indirect effect on viral translation and RNA replication, likely by increasing the translating/replicating pool of viral RNAs to the detriment of virion assembly.


Assuntos
Genoma Viral , Hepacivirus , RNA Viral , Proteínas de Ligação a RNA , Replicação Viral , Hepacivirus/genética , Hepacivirus/fisiologia , Humanos , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , RNA Viral/genética , RNA Viral/metabolismo , Empacotamento do Genoma Viral , Biossíntese de Proteínas , Montagem de Vírus , Linhagem Celular , Hepatite C/virologia , Hepatite C/metabolismo
3.
PLoS One ; 19(6): e0306345, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38935609

RESUMO

Chronic liver diseases are caused by hepatic viral infection, chemicals, and metabolic stress. The protein Grb2-associated binder 1 (Gab1) binds to various growth factor receptors, and triggers cell differentiation/survival signaling pathways. To identify signaling molecules involved in the progression of liver diseases, we performed reverse-phase protein microarray (RPMA)-based screening of hepatocytes isolated from humanized mice after acute HCV infection. Acute viral infection in humanized liver mice significantly decreased the level of hepatocyte p-Gab1. Moreover, hepatoma cells upon HCV infection decreased Gab1 mRNA at later times of infection (D3 to D5) and p-Gab1 level was inversely related to the production of TGF-ß. In contrast, the level of p-Gab1 was increased in CCL4-induced fibrotic liver. Hepatoma cells showed elevation of p-Gab1, along with an increase in STAT3 and ERK activation, upon treatment with HGF (ligand of HGF receptor/c-Met) and CCL4. In Gab1 knockdown hepatoma cells, cell proliferative signaling activity was reduced but the level of activated caspase-3 was increased. These findings suggest that hepatocyte Gab1 expression may play a role in promoting liver fibrosis progression by triggering ERK activation and inhibiting apoptosis. It implies that the Gab1-mediated signaling pathway would be a promising therapeutic target to treat chronic liver diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose , Proliferação de Células , Fator de Crescimento de Hepatócito , Hepatócitos , Cirrose Hepática , Proteínas Proto-Oncogênicas c-met , Transdução de Sinais , Animais , Hepatócitos/metabolismo , Hepatócitos/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-met/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Linhagem Celular Tumoral , Hepatite C/metabolismo , Hepatite C/patologia , Hepatite C/complicações
4.
Cells ; 13(12)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38920664

RESUMO

Hepatitis C virus (HCV) is an oncogenic virus that causes chronic liver disease in more than 80% of patients. During the last decade, efficient direct-acting antivirals were introduced into clinical practice. However, clearance of the virus does not reduce the risk of end-stage liver diseases to the level observed in patients who have never been infected. So, investigation of HCV pathogenesis is still warranted. Virus-induced changes in cell metabolism contribute to the development of HCV-associated liver pathologies. Here, we studied the impact of the virus on the metabolism of polyamines and proline as well as on the urea cycle, which plays a crucial role in liver function. It was found that HCV strongly suppresses the expression of arginase, a key enzyme of the urea cycle, leading to the accumulation of arginine, and up-regulates proline oxidase with a concomitant decrease in proline concentrations. The addition of exogenous proline moderately suppressed viral replication. HCV up-regulated transcription but suppressed protein levels of polyamine-metabolizing enzymes. This resulted in a decrease in polyamine content in infected cells. Finally, compounds targeting polyamine metabolism demonstrated pronounced antiviral activity, pointing to spermine and spermidine as compounds affecting HCV replication. These data expand our understanding of HCV's imprint on cell metabolism.


Assuntos
Hepacivirus , Poliaminas , Prolina , Ureia , Replicação Viral , Prolina/metabolismo , Humanos , Hepacivirus/fisiologia , Hepacivirus/efeitos dos fármacos , Poliaminas/metabolismo , Ureia/metabolismo , Ureia/farmacologia , Replicação Viral/efeitos dos fármacos , Arginase/metabolismo , Antivirais/farmacologia , Antivirais/metabolismo , Hepatite C/metabolismo , Hepatite C/virologia , Linhagem Celular Tumoral , Prolina Oxidase/metabolismo
5.
Viruses ; 16(5)2024 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-38793620

RESUMO

Hepatitis C virus (HCV) infects the human liver, and its chronic infection is one of the major causes of Hepatocellular carcinoma. Translation of HCV RNA is mediated by an Internal Ribosome Entry Site (IRES) element located in the 5'UTR of viral RNA. Several RNA Binding proteins of the host interact with the HCV IRES and modulate its function. Here, we demonstrate that PSPC1 (Paraspeckle Component 1), an essential paraspeckle component, upon HCV infection is relocalized and interacts with HCV IRES to prevent viral RNA translation. Competition UV-crosslinking experiments showed that PSPC1 interacts explicitly with the SLIV region of the HCV IRES, which is known to play a vital role in ribosomal loading to the HCV IRES via interaction with Ribosomal protein S5 (RPS5). Partial silencing of PSPC1 increased viral RNA translation and, consequently, HCV replication, suggesting a negative regulation by PSPC1. Interestingly, the silencing of PSPC1 protein leads to an increased interaction of RPS5 at the SLIV region, leading to an overall increase in the viral RNA in polysomes. Overall, our results showed how the host counters viral infection by relocalizing nuclear protein to the cytoplasm as a survival strategy.


Assuntos
Hepacivirus , Sítios Internos de Entrada Ribossomal , Biossíntese de Proteínas , RNA Viral , Proteínas de Ligação a RNA , Proteínas Ribossômicas , Humanos , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatite C/virologia , Hepatite C/metabolismo , Interações Hospedeiro-Patógeno , Ligação Proteica , Proteínas Ribossômicas/metabolismo , RNA Viral/metabolismo , RNA Viral/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Replicação Viral
6.
J Immunol Res ; 2024: 6343757, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38715844

RESUMO

This study aims to explore the influence of coinfection with HCV and HIV on hepatic fibrosis. A coculture system was set up to actively replicate both viruses, incorporating CD4 T lymphocytes (Jurkat), hepatic stellate cells (LX-2), and hepatocytes (Huh7.5). LX-2 cells' susceptibility to HIV infection was assessed through measurements of HIV receptor expression, exposure to cell-free virus, and cell-to-cell contact with HIV-infected Jurkat cells. The study evaluated profibrotic parameters, including programed cell death, ROS imbalance, cytokines (IL-6, TGF-ß, and TNF-α), and extracellular matrix components (collagen, α-SMA, and MMP-9). The impact of HCV infection on LX-2/HIV-Jurkat was examined using soluble factors released from HCV-infected hepatocytes. Despite LX-2 cells being nonsusceptible to direct HIV infection, bystander effects were observed, leading to increased oxidative stress and dysregulated profibrotic cytokine release. Coculture with HIV-infected Jurkat cells intensified hepatic fibrosis, redox imbalance, expression of profibrotic cytokines, and extracellular matrix production. Conversely, HCV-infected Huh7.5 cells exhibited elevated profibrotic gene transcriptions but without measurable effects on the LX-2/HIV-Jurkat coculture. This study highlights how HIV-infected lymphocytes worsen hepatic fibrosis during HCV/HIV coinfection. They increase oxidative stress, profibrotic cytokine levels, and extracellular matrix production in hepatic stellate cells through direct contact and soluble factors. These insights offer valuable potential therapies for coinfected individuals.


Assuntos
Efeito Espectador , Técnicas de Cocultura , Coinfecção , Citocinas , Infecções por HIV , Hepacivirus , Células Estreladas do Fígado , Hepatite C , Cirrose Hepática , Humanos , Células Estreladas do Fígado/metabolismo , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Infecções por HIV/imunologia , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , Hepatite C/complicações , Hepatite C/imunologia , Células Jurkat , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/virologia , Cirrose Hepática/etiologia , Citocinas/metabolismo , Hepatócitos/metabolismo , Hepatócitos/virologia , HIV/fisiologia , Estresse Oxidativo , Comunicação Celular , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Matriz Extracelular/metabolismo
7.
Gastroenterology ; 167(3): 522-537, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38636680

RESUMO

BACKGROUND & AIMS: High expression of phosphatidylinositol 4-kinase III alpha (PI4KIIIα) correlates with poor survival rates in patients with hepatocellular carcinoma. In addition, hepatitis C virus (HCV) infections activate PI4KIIIα and contribute to hepatocellular carcinoma progression. We aimed at mechanistically understanding the impact of PI4KIIIα on the progression of liver cancer and the potential contribution of HCV in this process. METHODS: Several hepatic cell culture and mouse models were used to study the functional importance of PI4KIIIα on liver pathogenesis. Antibody arrays, gene silencing, and PI4KIIIα-specific inhibitor were applied to identify the involved signaling pathways. The contribution of HCV was examined by using HCV infection or overexpression of its nonstructural protein. RESULTS: High PI4KIIIα expression and/or activity induced cytoskeletal rearrangements via increased phosphorylation of paxillin and cofilin. This led to morphologic alterations and higher migratory and invasive properties of liver cancer cells. We further identified the liver-specific lipid kinase phosphatidylinositol 3-kinase C2 domain-containing subunit gamma (PIK3C2γ) working downstream of PI4KIIIα in regulation of the cytoskeleton. PIK3C2γ generates plasma membrane phosphatidylinositol 3,4-bisphosphate-enriched, invadopodia-like structures that regulate cytoskeletal reorganization by promoting Akt2 phosphorylation. CONCLUSIONS: PI4KIIIα regulates cytoskeleton organization via PIK3C2γ/Akt2/paxillin-cofilin to favor migration and invasion of liver cancer cells. These findings provide mechanistic insight into the contribution of PI4KIIIα and HCV to the progression of liver cancer and identify promising targets for therapeutic intervention.


Assuntos
Fatores de Despolimerização de Actina , Carcinoma Hepatocelular , Movimento Celular , Citoesqueleto , Neoplasias Hepáticas , Invasividade Neoplásica , Paxilina , Transdução de Sinais , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Humanos , Animais , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Paxilina/metabolismo , Camundongos , Fatores de Despolimerização de Actina/metabolismo , Fatores de Despolimerização de Actina/genética , Fosforilação , Hepacivirus , Linhagem Celular Tumoral , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Células Hep G2 , Hepatite C/patologia , Hepatite C/metabolismo , Hepatite C/virologia , Interferência de RNA
8.
Exp Mol Med ; 56(5): 1080-1106, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38689093

RESUMO

Recent progress in the investigation of microRNA (miRNA) biogenesis and the miRNA processing machinery has revealed previously unknown roles of posttranscriptional regulation in gene expression. The molecular mechanistic interplay between miRNAs and their regulatory factors, RNA-binding proteins (RBPs) and exoribonucleases, has been revealed to play a critical role in tumorigenesis. Moreover, recent studies have shown that the proliferation of hepatocellular carcinoma (HCC)-causing hepatitis C virus (HCV) is also characterized by close crosstalk of a multitude of host RBPs and exoribonucleases with miR-122 and its RNA genome, suggesting the importance of the mechanistic interplay among these factors during the proliferation of HCV. This review primarily aims to comprehensively describe the well-established roles and discuss the recently discovered understanding of miRNA regulators, RBPs and exoribonucleases, in relation to various cancers and the proliferation of a representative cancer-causing RNA virus, HCV. These have also opened the door to the emerging potential for treating cancers as well as HCV infection by targeting miRNAs or their respective cellular modulators.


Assuntos
Exorribonucleases , Regulação Neoplásica da Expressão Gênica , Hepacivirus , MicroRNAs , Neoplasias , Proteínas de Ligação a RNA , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Neoplasias/genética , Neoplasias/metabolismo , Exorribonucleases/metabolismo , Exorribonucleases/genética , Animais , Hepacivirus/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Hepatite C/metabolismo , Hepatite C/genética , Hepatite C/virologia
9.
J Biol Chem ; 300(5): 107286, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38636657

RESUMO

Hepatitis C virus (HCV) infection is tightly connected to the lipid metabolism with lipid droplets (LDs) serving as assembly sites for progeny virions. A previous LD proteome analysis identified annexin A3 (ANXA3) as an important HCV host factor that is enriched at LDs in infected cells and required for HCV morphogenesis. To further characterize ANXA3 function in HCV, we performed proximity labeling using ANXA3-BioID2 as bait in HCV-infected cells. Two of the top proteins identified proximal to ANXA3 during HCV infection were the La-related protein 1 (LARP1) and the ADP ribosylation factor-like protein 8B (ARL8B), both of which have been previously described to act in HCV particle production. In follow-up experiments, ARL8B functioned as a pro-viral HCV host factor without localizing to LDs and thus likely independent of ANXA3. In contrast, LARP1 interacts with HCV core protein in an RNA-dependent manner and is translocated to LDs by core protein. Knockdown of LARP1 decreased HCV spreading without altering HCV RNA replication or viral titers. Unexpectedly, entry of HCV particles and E1/E2-pseudotyped lentiviral particles was reduced by LARP1 depletion, whereas particle production was not altered. Using a recombinant vesicular stomatitis virus (VSV)ΔG entry assay, we showed that LARP1 depletion also decreased entry of VSV with VSV, MERS, and CHIKV glycoproteins. Therefore, our data expand the role of LARP1 as an HCV host factor that is most prominently involved in the early steps of infection, likely contributing to endocytosis of viral particles through the pleiotropic effect LARP1 has on the cellular translatome.


Assuntos
Anexina A3 , Hepacivirus , Hepatite C , Antígeno SS-B , Internalização do Vírus , Humanos , Anexina A3/metabolismo , Anexina A3/genética , Autoantígenos/metabolismo , Autoantígenos/genética , Células HEK293 , Hepacivirus/metabolismo , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , Hepatite C/genética , Interações Hospedeiro-Patógeno , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/virologia , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/genética , Proteínas do Core Viral/metabolismo , Proteínas do Core Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética
10.
J Virol ; 98(3): e0163823, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38353536

RESUMO

Reverse genetics systems have played a central role in developing recombinant viruses for a wide spectrum of virus research. The circular polymerase extension reaction (CPER) method has been applied to studying positive-strand RNA viruses, allowing researchers to bypass molecular cloning of viral cDNA clones and thus leading to the rapid generation of recombinant viruses. However, thus far, the CPER protocol has only been established using cap-dependent RNA viruses. Here, we demonstrate that a modified version of the CPER method can be successfully applied to positive-strand RNA viruses that use cap-independent, internal ribosomal entry site (IRES)-mediated translation. As a proof-of-concept, we employed mammalian viruses with different types (classes I, II, and III) of IRES to optimize the CPER method. Using the hepatitis C virus (HCV, class III), we found that inclusion in the CPER assembly of an RNA polymerase I promoter and terminator, instead of those from polymerase II, allowed greater viral production. This approach was also successful in generating recombinant bovine viral diarrhea virus (class III) following transfection of MDBK/293T co-cultures to overcome low transfection efficiency. In addition, we successfully generated the recombinant viruses from clinical specimens. Our modified CPER could be used for producing hepatitis A virus (HAV, type I) as well as de novo generation of encephalomyocarditis virus (type II). Finally, we generated recombinant HCV and HAV reporter viruses that exhibited replication comparable to that of the wild-type parental viruses. The recombinant HAV reporter virus helped evaluate antivirals. Taking the findings together, this study offers methodological advances in virology. IMPORTANCE: The lack of versatility of reverse genetics systems remains a bottleneck in viral research. Especially when (re-)emerging viruses reach pandemic levels, rapid characterization and establishment of effective countermeasures using recombinant viruses are beneficial in disease control. Indeed, numerous studies have attempted to establish and improve the methods. The circular polymerase extension reaction (CPER) method has overcome major obstacles in generating recombinant viruses. However, this method has not yet been examined for positive-strand RNA viruses that use cap-independent, internal ribosome entry site-mediated translation. Here, we engineered a suitable gene cassette to expand the CPER method for all positive-strand RNA viruses. Furthermore, we overcame the difficulty of generating recombinant viruses because of low transfection efficiency. Using this modified method, we also successfully generated reporter viruses and recombinant viruses from a field sample without virus isolation. Taking these findings together, our adapted methodology is an innovative technology that could help advance virologic research.


Assuntos
Hepatite C , Biossíntese de Proteínas , Genética Reversa , Animais , Hepatite C/metabolismo , Sítios Internos de Entrada Ribossomal/genética , Mamíferos/genética , Vírus de RNA de Cadeia Positiva/genética , Vírus de RNA de Cadeia Positiva/metabolismo , Genética Reversa/métodos , RNA Viral/genética
11.
PLoS Pathog ; 19(12): e1011887, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38157366

RESUMO

The multi-step process of hepatitis C virus (HCV) entry is facilitated by various host factors, including epidermal growth factor receptor (EGFR) and the tight junction proteins claudin-1 (CLDN1) and occludin (OCLN), which are thought to function at later stages of the HCV entry process. Using single particle imaging of HCV infection of polarized hepatoma spheroids, we observed that EGFR performs multiple functions in HCV entry, both phosphorylation-dependent and -independent. We previously observed, and in this study confirmed, that EGFR is not required for HCV migration to the tight junction. EGFR is required for the recruitment of clathrin to HCV in a phosphorylation-independent manner. EGFR phosphorylation is required for virion internalization at a stage following the recruitment of clathrin. HCV entry activates the RAF-MEK-ERK signaling pathway downstream of EGFR phosphorylation. This signaling pathway regulates the sorting and maturation of internalized HCV into APPL1- and EEA1-associated early endosomes, which form the site of virion uncoating. The tight junction proteins, CLDN1 and OCLN, function at two distinct stages of HCV entry. Despite its appreciated function as a "late receptor" in HCV entry, CLDN1 is required for efficient HCV virion accumulation at the tight junction. Huh-7.5 cells lacking CLDN1 accumulate HCV virions primarily at the initial basolateral surface. OCLN is required for the late stages of virion internalization. This study produced further insight into the unusually complex HCV endocytic process.


Assuntos
Claudina-1 , Hepacivirus , Hepatite C , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Clatrina , Claudina-1/genética , Claudina-1/metabolismo , Receptores ErbB , Hepacivirus/fisiologia , Hepatite C/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Ocludina/metabolismo , Internalização do Vírus
12.
Front Biosci (Landmark Ed) ; 28(10): 239, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37919063

RESUMO

BACKGROUND: Hepatitis C virus (HCV) infection is a global health threat to the public, and vaccines against it are not yet available. The HCV envelope glycoprotein E2 is a key target for anti-HCV vaccines. The majority of previous studies have focused on the hypervariable region and the glycosylation sites of the_HCV structural protein. This study aims to investigate a conserved domain of HCV E2 glycoprotein and explore its potential to induce an immune response against HCV. METHODS: HCV E2 conserved domain (encompassing amino acids 505-702) was prepared in Escherichia coli (E. coli). Peripheral blood mononuclear cells (PBMCs) were isolated from patients with HCV or healthy controls. Interferon-gamma (IFN-γ) enzyme-linked immunosorbent spot assay was conducted to examine the HCV E2-specific immune response as reflected by IFN-γ-secreting cells/106 PBMCs. RESULTS: HCV E2 conserved domain was highly conserved among 25 HCV subtypes, and its recombinant soluble production in E. coli was recognized by anti-HCV E2 monoclonal antibodies. This study characterized in vitro direct interaction between bacterially expressed HCV E2 conserved domain and human CD81 (hCD81). Furthermore, the recombinant HCV E2_conserved domain markedly induced the production of IFN-γ by PBMCs from patients with HCV. Its stimulated specific immune response was significantly different from non-specific peptide controls or PBMCs isolated from healthy controls. CONCLUSIONS: HCV E2 conserved domain directly binds hCD81 and activates the production of IFN-γ in the PBMCs of patients with HCV. Therefore, the conserved domain of HCV E2 glycoprotein may be a new candidate for developing an HCV vaccine.


Assuntos
Hepatite C , Vacinas , Humanos , Escherichia coli/genética , Hepacivirus/fisiologia , Hepatite C/metabolismo , Interferon gama/metabolismo , Leucócitos Mononucleares/metabolismo , Tetraspanina 28/química , Tetraspanina 28/metabolismo
13.
Nucleic Acids Res ; 51(22): 12397-12413, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-37941151

RESUMO

Hepatitis C virus (HCV) requires two cellular factors, microRNA-122 (miR-122) and poly(C) binding protein 2 (PCBP2), for optimal replication. These host factors compete for binding to the 5' end of the single-stranded RNA genome to regulate the viral replication cycle. To understand how they interact with the RNA, we measured binding affinities of both factors for an RNA probe representing the 5' 45 nucleotides of the HCV genome (HCV45). Isothermal titration calorimetry revealed two, unequal miR-122 binding sites in HCV45, high-affinity (S1) and low-affinity (S2), differing roughly 100-fold in binding affinity. PCBP2 binds a site overlapping S2 with affinity similar to miR-122 binding to S2. PCBP2 circularizes the genome by also binding to the 3' UTR, bridging the 5' and 3' ends of the genome. By competing with PCBP2 for binding at S2, miR-122 disrupts PCBP2-mediated genome circularization. We show that the viral RNA-dependent RNA polymerase, NS5B, also binds to HCV45, and that the binding affinity of NS5B is increased in the presence of miR-122, suggesting miR-122 promotes recruitment of the polymerase. We propose that competition between miR-122 and PCBP2 for HCV45 functions as a translation-to-replication switch, determining whether the RNA genome templates protein synthesis or RNA replication.


Assuntos
Hepacivirus , Hepatite C , MicroRNAs , Humanos , Regiões 5' não Traduzidas , Proteínas de Transporte/genética , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Replicação Viral/genética
14.
EMBO Rep ; 24(11): e56614, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37789674

RESUMO

ATPase family AAA domain-containing protein 1 (ATAD1) maintains mitochondrial homeostasis by removing mislocalized tail-anchored (TA) proteins from the mitochondrial outer membrane (MOM). Hepatitis C virus (HCV) infection induces mitochondrial fragmentation, and viral NS5B protein is a TA protein. Here, we investigate whether ATAD1 plays a role in regulating HCV infection. We find that HCV infection has no effect on ATAD1 expression, but knockout of ATAD1 significantly enhances HCV infection; this enhancement is suppressed by ATAD1 complementation. NS5B partially localizes to mitochondria, dependent on its transmembrane domain (TMD), and induces mitochondrial fragmentation, which is further enhanced by ATAD1 knockout. ATAD1 interacts with NS5B, dependent on its three internal domains (TMD, pore-loop 1, and pore-loop 2), and induces the proteasomal degradation of NS5B. In addition, we provide evidence that ATAD1 augments the antiviral function of MAVS upon HCV infection. Taken together, we show that the mitochondrial quality control exerted by ATAD1 can be extended to a novel antiviral function through the extraction of the viral TA-protein NS5B from the mitochondrial outer membrane.


Assuntos
Hepacivirus , Hepatite C , Humanos , Hepacivirus/metabolismo , Proteínas Virais/metabolismo , Hepatite C/metabolismo , Mitocôndrias/metabolismo , Antivirais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
15.
Front Biosci (Schol Ed) ; 15(3): 10, 2023 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-37806953

RESUMO

Currently, the issue relating to the discussion raised in this article appears to be for what purposes the hepatitis C virus (HCV) modulates cellular processes, such as antiviral defense, metabolism, apoptosis, and mitochondrial dynamics, by inhibiting the activity or expression of mitochondrial proteins and a number of cellular proteins. Additionally, to what pathological changes do these alterations lead? Thus, the aim of this review is to propose potential protein mitochondrial targets of HCV for the future development of new drugs aimed at inhibiting its interaction with cellular proteins. Considering current analyses in the literature, promising targets for the acute and chronic phases of HCV are proposed which include mitochondrial antiviral signaling (MAVS) (antiviral response protein), Parkin (mitophagy protein), Drp1 (mitochondrial fission protein), subunits 1 and 4 of the electron transport chain (ETC) complex (oxidative phosphorylation proteins), among others. This review illustrates how viral strategies for modulating cellular processes involving HCV proteins differ in the acute and chronic phases and, as a result, the complications that arise.


Assuntos
Hepatite C , Mitocôndrias , Humanos , Mitocôndrias/metabolismo , Mitofagia/fisiologia , Proteínas Mitocondriais/metabolismo , Hepatite Crônica/metabolismo , Antivirais/metabolismo , Hepatite C/metabolismo
16.
mBio ; 14(5): e0154923, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37671888

RESUMO

IMPORTANCE: Assessments of viral stability on surfaces or in body fluids under different environmental conditions and/or temperatures are often performed, as they are key to understanding the routes and parameters of viral transmission and to providing clues on the epidemiology of infections. However, for most viruses, the mechanisms of inactivation vs stability of viral particles remain poorly defined. Although they are structurally diverse, with different compositions, sizes, and shapes, enveloped viruses are generally less stable than non-enveloped viruses, pointing out the role of envelopes themselves in virus lability. In this report, we investigated the properties of hepatitis C virus (HCV) particles with regards to their stability. We found that, compared to alternative enveloped viruses such as Dengue virus (DENV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), hepatitis delta virus (HDV), and Crimean-Congo hemorrhagic fever virus (CCHFV) that infect the liver, HCV particles are intrinsically labile. We determined the mechanisms that drastically alter their specific infectivity through oxidation of their lipids, and we highlighted that they are protected from lipid oxidation by secreted cellular proteins, which can protect their membrane fusion capacity and overall infectivity.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Hepatite C , Humanos , Hepacivirus , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Hepatite C/metabolismo
17.
PLoS Pathog ; 19(8): e1011552, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37540723

RESUMO

Host protein HuR translocation from nucleus to cytoplasm following infection is crucial for the life cycle of several RNA viruses including hepatitis C virus (HCV), a major causative agent of hepatocellular carcinoma. HuR assists the assembly of replication-complex on the viral-3'UTR, and its depletion hampers viral replication. Although cytoplasmic HuR is crucial for HCV replication, little is known about how the virus orchestrates the mobilization of HuR into the cytoplasm from the nucleus. We show that two viral proteins, NS3 and NS5A, act co-ordinately to alter the equilibrium of the nucleo-cytoplasmic movement of HuR. NS3 activates protein kinase C (PKC)-δ, which in-turn phosphorylates HuR on S318 residue, triggering its export to the cytoplasm. NS5A inactivates AMP-activated kinase (AMPK) resulting in diminished nuclear import of HuR through blockade of AMPK-mediated phosphorylation and acetylation of importin-α1. Cytoplasmic retention or entry of HuR can be reversed by an AMPK activator or a PKC-δ inhibitor. Our findings suggest that efforts should be made to develop inhibitors of PKC-δ and activators of AMPK, either separately or in combination, to inhibit HCV infection.


Assuntos
Hepacivirus , Hepatite C , Humanos , Hepacivirus/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Proteína Semelhante a ELAV 1/genética , Proteína Semelhante a ELAV 1/metabolismo , Citoplasma/metabolismo , Hepatite C/metabolismo , Linhagem Celular Tumoral , Replicação Viral , Proteínas não Estruturais Virais/metabolismo
18.
PLoS Pathog ; 19(8): e1011591, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37585449

RESUMO

Hepatitis C virus (HCV) is a pathogen characterized not only by its persistent infection leading to the development of cirrhosis and hepatocellular carcinoma (HCC), but also by metabolic disorders such as lipid and iron dysregulation. Elevated iron load is commonly observed in the livers of patients with chronic hepatitis C, and hepatic iron overload is a highly profibrogenic and carcinogenic factor that increases the risk of HCC. However, the underlying mechanisms of elevated iron accumulation in HCV-infected livers remain to be fully elucidated. Here, we observed iron accumulation in cells and liver tissues under HCV infection and in mice expressing viral proteins from recombinant adenoviruses. We established two molecular mechanisms that contribute to increased iron load in cells caused by HCV infection. One is the transcriptional induction of hepcidin, the key hormone for modulating iron homeostasis. The transcription factor cAMP-responsive element-binding protein hepatocyte specific (CREBH), which was activated by HCV infection, not only directly recognizes the hepcidin promoter but also induces bone morphogenetic protein 6 (BMP6) expression, resulting in an activated BMP-SMAD pathway that enhances hepcidin promoter activity. The other is post-translational regulation of the iron-exporting membrane protein ferroportin 1 (FPN1), which is cleaved between residues Cys284 and Ala285 in the intracytoplasmic loop region of the central portion mediated by HCV NS3-4A serine protease. We propose that host transcriptional activation triggered by endoplasmic reticulum stress and FPN1 cleavage by viral protease work in concert to impair iron efflux, leading to iron accumulation in HCV-infected cells.


Assuntos
Carcinoma Hepatocelular , Hepatite C , Neoplasias Hepáticas , Animais , Camundongos , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Ferro/metabolismo , Ativação Transcricional , Regulação para Cima
19.
Mol Biol (Mosk) ; 57(3): 427-439, 2023.
Artigo em Russo | MEDLINE | ID: mdl-37326046

RESUMO

The key role of histone deacetylases (HDAC) in the regulation of the cellular response to infection with the hepatitis C virus (HCV) was first demonstrated in 2008. When studying the metabolism of iron in the liver tissues of patients with chronic hepatitis C, the authors found that the expression of the hepcidin gene (HAMP), a hormone regulator of iron export, is markedly reduced in hepatocytes under conditions of oxidative stress caused by viral infection. HDAC were involved in the regulation of hepcidin expression through the control of acetylation level of histones and transcription factors, primarily STAT3, associated with the HAMP promoter. The purpose of this review was to summarize current data on the functioning of the HCV-HDAC3-STAT3-HAMP regulatory circuit as an example of a well-characterized interaction between the virus and the epigenetic apparatus of the host cell.


Assuntos
Hepatite C , Hepcidinas , Humanos , Hepcidinas/genética , Hepcidinas/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatite C/genética , Hepatite C/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Ferro/metabolismo , Replicação Viral
20.
J Virol ; 97(7): e0018023, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37338368

RESUMO

Although most of the early events of the hepatitis C virus (HCV) life cycle are well characterized, our understanding of HCV egress is still unclear. Some reports implicate the conventional endoplasmic reticulum (ER)-Golgi route, while some propose noncanonical secretory routes. Initially, the envelopment of HCV nucleocapsid occurs by budding into the ER lumen. Subsequently, the HCV particle exit from the ER is assumed to be mediated by coat protein complex II (COPII) vesicles. COPII vesicle biogenesis also involves the recruitment of cargo to the site of vesicle biogenesis via interaction with COPII inner coat proteins. We investigated the modulation and the specific role of the individual components of the early secretory pathway in HCV egress. We observed that HCV inhibits cellular protein secretion and triggers the reorganization of the ER exit sites and ER-Golgi intermediate compartments (ERGIC). Gene-specific knockdown of the components of this pathway such as SEC16A, TFG, ERGIC-53, and COPII coat proteins demonstrated the functional significance of these components and the distinct role played by these proteins in various aspects of the HCV life cycle. SEC16A is essential for multiple steps in the HCV life cycle, whereas TFG is specifically involved in HCV egress and ERGIC-53 is crucial for HCV entry. Overall, our study establishes that the components of the early secretory pathway are essential for HCV propagation and emphasize the importance of the ER-Golgi secretory route in this process. Surprisingly, these components are also required for the early stages of the HCV life cycle due to their role in overall intracellular trafficking and homeostasis of the cellular endomembrane system. IMPORTANCE The virus life cycle involves entry into the host, replication of the genome, assembly of infectious progeny, and their subsequent release. Different aspects of the HCV life cycle, including entry, genome replication, and assembly, are well characterized; however, our understanding of the HCV release is still not clear and subject to debate due to varied findings. Here, we attempted to address this controversy and enhance our understanding of HCV egress by evaluating the role of the different components of the early secretory pathway in the HCV life cycle. To our surprise, we found that the components of the early secretory pathway are not only essential for HCV release but also contribute to many other earlier events of the HCV life cycle. This study emphasizes the importance of the early secretory pathway for the establishment of productive HCV infection in hepatocytes.


Assuntos
Retículo Endoplasmático , Hepatite C , Humanos , Animais , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Via Secretória , Hepacivirus/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Transporte Proteico , Hepatite C/metabolismo , Estágios do Ciclo de Vida , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...