Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.593
Filtrar
1.
J Virol ; 98(3): e0192123, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38319104

RESUMO

Hepatitis C virus (HCV) infection progresses to chronicity in the majority of infected individuals. Its high intra-host genetic variability enables HCV to evade the continuous selection pressure exerted by the host, contributing to persistent infection. Utilizing a cell culture-adapted HCV population (p100pop) which exhibits increased replicative capacity in various liver cell lines, this study investigated virus and host determinants that underlie enhanced viral fitness. Characterization of a panel of molecular p100 clones revealed that cell culture adaptive mutations optimize a range of virus-host interactions, resulting in expanded cell tropism, altered dependence on the cellular co-factor micro-RNA 122 and increased rates of virus spread. On the host side, comparative transcriptional profiling of hepatoma cells infected either with p100pop or its progenitor virus revealed that enhanced replicative fitness correlated with activation of endoplasmic reticulum stress signaling and the unfolded protein response. In contrast, infection of primary human hepatocytes with p100pop led to a mild attenuation of virion production which correlated with a greater induction of cell-intrinsic antiviral defense responses. In summary, long-term passage experiments in cells where selective pressure from innate immunity is lacking improves multiple virus-host interactions, enhancing HCV replicative fitness. However, this study further indicates that HCV has evolved to replicate at low levels in primary human hepatocytes to minimize innate immune activation, highlighting that an optimal balance between replicative fitness and innate immune induction is key to establish persistence. IMPORTANCE: Hepatitis C virus (HCV) infection remains a global health burden with 58 million people currently chronically infected. However, the detailed molecular mechanisms that underly persistence are incompletely defined. We utilized a long-term cell culture-adapted HCV, exhibiting enhanced replicative fitness in different human liver cell lines, in order to identify molecular principles by which HCV optimizes its replication fitness. Our experimental data revealed that cell culture adaptive mutations confer changes in the host response and usage of various host factors. The latter allows functional flexibility at different stages of the viral replication cycle. However, increased replicative fitness resulted in an increased activation of the innate immune system, which likely poses boundary for functional variation in authentic hepatocytes, explaining the observed attenuation of the adapted virus population in primary hepatocytes.


Assuntos
Aptidão Genética , Hepacivirus , Hepatócitos , Interações entre Hospedeiro e Microrganismos , Imunidade Inata , Mutação , Humanos , Células Cultivadas , Estresse do Retículo Endoplasmático , Aptidão Genética/genética , Aptidão Genética/imunologia , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/imunologia , Hepacivirus/fisiologia , Hepatite C/imunologia , Hepatite C/virologia , Hepatócitos/imunologia , Hepatócitos/virologia , Interações entre Hospedeiro e Microrganismos/imunologia , MicroRNAs/metabolismo , Inoculações Seriadas , Resposta a Proteínas não Dobradas , Tropismo Viral , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Replicação Viral/genética , Replicação Viral/imunologia
2.
J Virol ; 98(3): e0150223, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38315015

RESUMO

Capsid assembly is critical in the hepatitis B virus (HBV) life cycle, mediated by the viral core protein. Capsid assembly is the target for new anti-viral therapeutics known as capsid assembly modulators (CAMs) of which the CAM-aberrant (CAM-A) class induces aberrant shaped core protein structures and leads to hepatocyte cell death. This study aimed to identify the mechanism of action of CAM-A modulators leading to HBV-infected hepatocyte elimination where CAM-A-mediated hepatitis B surface antigen (HBsAg) reduction was evaluated in a stable HBV replicating cell line and in AAV-HBV-transduced C57BL/6, C57BL/6 SCID, and HBV-infected chimeric mice with humanized livers. Results showed that in vivo treatment with CAM-A modulators induced pronounced reductions in hepatitis B e antigen (HBeAg) and HBsAg, associated with a transient alanine amino transferase (ALT) increase. Both HBsAg and HBeAg reductions and ALT increase were delayed in C57BL/6 SCID and chimeric mice, suggesting that adaptive immune responses may indirectly contribute. However, CD8+ T cell depletion in transduced wild-type mice did not impact antigen reduction, indicating that CD8+ T cell responses are not essential. Transient ALT elevation in AAV-HBV-transduced mice coincided with a transient increase in endoplasmic reticulum stress and apoptosis markers, followed by detection of a proliferation marker. Microarray data revealed antigen presentation pathway (major histocompatibility complex class I molecules) upregulation, overlapping with the apoptosis. Combination treatment with HBV-specific siRNA demonstrated that CAM-A-mediated HBsAg reduction is dependent on de novo core protein translation. To conclude, CAM-A treatment eradicates HBV-infected hepatocytes with high core protein levels through the induction of apoptosis, which can be a promising approach as part of a regimen to achieve functional cure. IMPORTANCE: Treatment with hepatitis B virus (HBV) capsid assembly modulators that induce the formation of aberrant HBV core protein structures (CAM-A) leads to programmed cell death, apoptosis, of HBV-infected hepatocytes and subsequent reduction of HBV antigens, which differentiates CAM-A from other CAMs. The effect is dependent on the de novo synthesis and high levels of core protein.


Assuntos
Antivirais , Apoptose , Regulação Viral da Expressão Gênica , Antígenos do Núcleo do Vírus da Hepatite B , Vírus da Hepatite B , Hepatócitos , Biossíntese de Proteínas , Animais , Camundongos , Antivirais/farmacologia , Antivirais/uso terapêutico , Apoptose/efeitos dos fármacos , Capsídeo/química , Capsídeo/classificação , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Hepatite B/tratamento farmacológico , Hepatite B/imunologia , Hepatite B/metabolismo , Hepatite B/virologia , Antígenos do Núcleo do Vírus da Hepatite B/biossíntese , Antígenos do Núcleo do Vírus da Hepatite B/metabolismo , Antígenos E da Hepatite B/metabolismo , Antígenos de Superfície da Hepatite B/metabolismo , Vírus da Hepatite B/crescimento & desenvolvimento , Vírus da Hepatite B/imunologia , Vírus da Hepatite B/metabolismo , Vírus da Hepatite B/patogenicidade , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Camundongos Endogâmicos C57BL , Camundongos SCID , Replicação Viral , Linhagem Celular , Linfócitos T CD8-Positivos/imunologia , Apresentação de Antígeno
3.
Biochim Biophys Acta Mol Cell Res ; 1871(1): 119602, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37778471

RESUMO

The enzyme L-Dopa Decarboxylase (DDC) synthesizes the catecholamine dopamine and the indolamine serotonin. Apart from its role in the brain as a neurotransmitter biosynthetic enzyme, DDC has been detected also in the liver and other peripheral organs, where it is implicated in cell proliferation, apoptosis, and host-virus interactions. Dengue virus (DENV) suppresses DDC expression at the later stages of infection, during which DENV also inhibits autophagosome-lysosome fusion. As dopamine affects autophagy in neuronal cells, we investigated the possible association of DDC with autophagy in human hepatocytes and examined whether DDC mediates the relationship between DENV infection and autophagy. We performed DDC silencing/overexpression and evaluated autophagic markers upon induction of autophagy, or suppression of autophagosome-lysosome fusion. Our results showed that DDC favored the autophagic process, at least in part, through its biosynthetic function, while knockdown of DDC or inhibition of DDC enzymatic activity prevented autophagy completion. In turn, autophagy induction upregulated DDC, while autophagy reduction by chemical or genetic (ATG14L knockout) ways caused the opposite effect. This study also implicated DDC with the cellular energetic status, as DDC silencing reduced the oxidative phosphorylation activity of the cell. We also report that upon DDC silencing, the repressive effect of DENV on the completion of autophagy was enhanced, and the inhibition of autolysosome formation did not exert an additive effect on viral proliferation. These data unravel a novel role of DDC in the autophagic process and suggest that DENV downregulates DDC expression to inhibit the completion of autophagy, reinforcing the importance of this protein in viral infections.


Assuntos
Autofagia , Vírus da Dengue , Hepatócitos , Humanos , Vírus da Dengue/metabolismo , Dopa Descarboxilase/genética , Dopa Descarboxilase/metabolismo , Dopamina/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia
4.
J Virol ; 97(9): e0085323, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37695055

RESUMO

Rift Valley fever virus (RVFV) causes mild to severe disease in humans and livestock. Outbreaks of RVFV have been reported throughout Africa and have spread outside Africa since 2000, calling for urgent worldwide attention to this emerging virus. RVFV directly infects the liver, and elevated transaminases are a hallmark of severe RVFV infection. However, the specific contribution of viral replication in hepatocytes to pathogenesis of RVFV remains undefined. To address this, we generated a recombinant miRNA-targeted virus, RVFVmiR-122, to limit hepatocellular replication. MicroRNAs are evolutionarily conserved non-coding RNAs that regulate mRNA expression by targeting them for degradation. RVFVmiR-122 includes an insertion of four target sequences of the liver-specific miR-122. In contrast to control RVFVmiR-184, which contains four target sequences of mosquito-specific miR-184, RVFVmiR-122 has restricted replication in vitro in primary mouse hepatocytes. RVFVmiR-122-infected C57BL/6 mice survived acute hepatitis and instead developed late-onset encephalitis. This difference in clinical outcome was eliminated in Mir-122 KO mice, confirming the specificity of the finding. Interestingly, C57BL/6 mice infected with higher doses of RVFVmiR-122 had a higher survival rate which was correlated with faster clearance of virus from the liver, suggesting a role for activation of host immunity in the phenotype. Together, our data demonstrate that miR-122 can specifically restrict the replication of RVFVmiR-122 in liver tissue both in vitro and in vivo, and this restriction alters the clinical course of disease following RVFVmiR-122 infection. IMPORTANCE Rift Valley fever virus (RVFV) is a hemorrhagic fever virus that causes outbreaks in humans and livestock throughout Africa and has spread to continents outside Africa since 2000. However, no commercial vaccine or treatment is currently available for human use against RVFV. Although the liver has been demonstrated as a key target of RVFV, the contribution of viral replication in hepatocytes to overall RVFV pathogenesis is less well defined. In this study we addressed this question by using a recombinant miRNA-targeted virus with restricted replication in hepatocytes. We gained a better understanding of how this individual cell type contributes to the development of disease caused by RVFV. Techniques used in this study provide an innovative tool to the RVFV field that could be applied to study the consequences of limited RVFV replication in other target cells.


Assuntos
Hepatócitos , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Replicação Viral , Animais , Humanos , Camundongos , Hepatócitos/patologia , Hepatócitos/virologia , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/fisiologia
5.
Virology ; 586: 23-34, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37478771

RESUMO

Current therapies control but rarely achieve a cure for hepatitis B virus (HBV) infection. Restoration of the HBV-specific immunity by cell-based therapy represents a potential approach for a cure. In this study, we generated HBV specific CAR T cells based on an antibody 2H5-A14 targeting a preS1 region of the HBV large envelope protein. We show that the A14 CAR T cell is capable of killing hepatocytes infected by HBV with high specificity; adoptive transfer of A14 CAR T cells to HBV infected humanized FRG mice resulted in reductions of all serum and intrahepatic virological markers to levels below the detection limit. A14 CAR T cells treatment increased the levels of human IFN-γ, GM-CSF, and IL-8/CXCL-8 in the mice. These results show that A14 CAR T cells may be further developed for curative therapy against HBV infection by eliminating HBV-infected hepatocytes and inducing production of pro-inflammatory and antiviral cytokines.


Assuntos
Vírus da Hepatite B , Hepatite B , Imunoterapia Adotiva , Humanos , Animais , Camundongos , Vírus da Hepatite B/fisiologia , Hepatite B/terapia , Fígado/virologia , Transdução Genética , Lentivirus/genética , Vetores Genéticos , Células T de Memória/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Inflamação/metabolismo , Citocinas/imunologia , Hepatócitos/virologia
6.
Nature ; 617(7961): 555-563, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36996873

RESUMO

An outbreak of acute hepatitis of unknown aetiology in children was reported in Scotland1 in April 2022 and has now been identified in 35 countries2. Several recent studies have suggested an association with human adenovirus with this outbreak, a virus not commonly associated with hepatitis. Here we report a detailed case-control investigation and find an association between adeno-associated virus 2 (AAV2) infection and host genetics in disease susceptibility. Using next-generation sequencing, PCR with reverse transcription, serology and in situ hybridization, we detected recent infection with AAV2 in plasma and liver samples in 26 out of 32 (81%) cases of hepatitis compared with 5 out of 74 (7%) of samples from unaffected individuals. Furthermore, AAV2 was detected within ballooned hepatocytes alongside a prominent T cell infiltrate in liver biopsy samples. In keeping with a CD4+ T-cell-mediated immune pathology, the human leukocyte antigen (HLA) class II HLA-DRB1*04:01 allele was identified in 25 out of 27 cases (93%) compared with a background frequency of 10 out of 64 (16%; P = 5.49 × 10-12). In summary, we report an outbreak of acute paediatric hepatitis associated with AAV2 infection (most likely acquired as a co-infection with human adenovirus that is usually required as a 'helper virus' to support AAV2 replication) and disease susceptibility related to HLA class II status.


Assuntos
Infecções por Adenovirus Humanos , Dependovirus , Hepatite , Criança , Humanos , Doença Aguda/epidemiologia , Infecções por Adenovirus Humanos/epidemiologia , Infecções por Adenovirus Humanos/genética , Infecções por Adenovirus Humanos/virologia , Alelos , Estudos de Casos e Controles , Linfócitos T CD4-Positivos/imunologia , Coinfecção/epidemiologia , Coinfecção/virologia , Dependovirus/isolamento & purificação , Predisposição Genética para Doença , Vírus Auxiliares/isolamento & purificação , Hepatite/epidemiologia , Hepatite/genética , Hepatite/virologia , Hepatócitos/virologia , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Fígado/virologia
7.
Microbiol Spectr ; 11(1): e0123522, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36519846

RESUMO

Hepatitis B virus (HBV) infection targets host restriction factors that inhibit its replication and survival. Previous studies have shown that barriers to autointegration factor1 (BANF1) inhibited the replication of herpes simplex virus and vaccinia virus by binding to phosphate backbone of dsDNA. To date, no reports are available for the interplay between BANF1 and HBV. In this study, we elucidated the mechanisms by which HBV inhibit BANF1. First, the effect of HBV on BANF1 was observed in Huh-7, Hep G2, and Hep G2.2.15 cells. Huh-7 cells were transfected with pHBV1.3 or HBx plasmids. The results showed that there was a decreased expression of BANF1 in Hep G2.2.15 cells (P ≤ 0.005) or in HBV/HBx expressing Huh-7 cells (P ≤ 0.005), whereas BANF1 overexpression decreased viral replication (P ≤ 0.05). To study whether phosphorylation/dephosphorylation of BANF1 was responsible for antiviral activity, mutants were created, and it was found that inhibition due to mutants was less significant compared to BANF1 wild type. Previous studies have shown that HBV, at least in part, could regulate the expression of host miRNAs via HBx. It was found that miR-203 expression was high in Hep G2.2.15 cells (P ≤ 0.005) compared to Hep G2 cells. Next, the effect of HBx on miR-203 expression was studied and result showed that HBx upregulated miR-203 expression (P ≤ 0.005). Overexpression of miR-203 downregulated BANF1 expression (P ≤ 0.05) and viral titer was upregulated (P ≤ 0.05), while inhibition of miR-203, reversed these changes. In conclusion, BANF1 downregulated HBV, whereas HBV inhibited BANF1, at least in part, via HBx-mediated miR-203 upregulation in hepatic cells. IMPORTANCE In this study, for the first time, we found that BANF1 inhibited HBV replication and restricted the viral load. However, as previously reported for other viruses, the results in this study showed that BAF1 phosphorylation/dephosphorylation is not involved in its antiviral activity against HBV. HBV infection inhibited the intracellular expression of BANF1, via HBx-mediated upregulation of miR-203 expression. Overexpression of miR-203 downregulated BANF1 and increased the viral titer, while inhibition of miR-203 reversed these changes. This study helped us to understand the molecular mechanisms by which HBV survives and replicates in the host cells.


Assuntos
Hepatite B , MicroRNAs , Transativadores , Proteínas Virais Reguladoras e Acessórias , Humanos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Hepatite B/genética , Hepatite B/metabolismo , Hepatite B/virologia , Vírus da Hepatite B/genética , Hepatócitos/metabolismo , Hepatócitos/virologia , MicroRNAs/genética , MicroRNAs/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(34): e2202653119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35969792

RESUMO

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and is the leading cause of enterically transmitted viral hepatitis worldwide. Ribavirin (RBV) is currently the only treatment option for many patients; however, cases of treatment failures or posttreatment relapses have been frequently reported. RBV therapy was shown to be associated with an increase in HEV genome heterogeneity and the emergence of distinct HEV variants. In this study, we analyzed the impact of eight patient-derived open reading frame 2 (ORF2) single-nucleotide variants (SNVs), which occurred under RBV treatment, on the replication cycle and pathogenesis of HEV. The parental HEV strain and seven ORF2 variants showed comparable levels of RNA replication in human hepatoma cells and primary human hepatocytes. However, a P79S ORF2 variant demonstrated reduced RNA copy numbers released in the supernatant and an impairment in the production of infectious particles. Biophysical and biochemical characterization revealed that this SNV caused defective, smaller HEV particles with a loss of infectiousness. Furthermore, the P79S variant displayed an altered subcellular distribution of the ORF2 protein and was able to interfere with antibody-mediated neutralization of HEV in a competition assay. In conclusion, an SNV in the HEV ORF2 could be identified that resulted in altered virus particles that were noninfectious in vitro and in vivo, but could potentially serve as immune decoys. These findings provide insights in understanding the biology of circulating HEV variants and may guide development of personalized antiviral strategies in the future.


Assuntos
Vírus da Hepatite E , Ribavirina , Proteínas Virais , Linhagem Celular Tumoral , Vírus da Hepatite E/genética , Vírus da Hepatite E/fisiologia , Hepatócitos/virologia , Humanos , Recidiva Local de Neoplasia/genética , Nucleotídeos , RNA Viral , Ribavirina/farmacologia , Proteínas Virais/genética , Replicação Viral
9.
J Virol ; 96(18): e0084922, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36037476

RESUMO

The existing cell culture-based methods to study hepatitis B virus (HBV) have limitations and do not allow for viral long-term passage. The aim of this study was to develop a robust in vitro long-term viral passage system with optimized cell culture conditions and a viral isolate with the ability to spread and passage. An HBV genotype A clinical isolate was subjected to multiple rounds of UV treatment and passaged in an optimized primary human hepatocyte (PHH)/human fibroblast coculture system. The passaged UV-treated virus was sequenced and further characterized. In addition, a panel of mutant viruses containing different combinations of mutations observed in this virus was investigated. The clinical isolate was passaged for 20 rounds with 21 days per round in an optimized PHH/human fibroblast coculture system while subject to UV mutagenesis. This passaged UV-mutated isolate harbored four mutations: G225A (sR24K) in the S gene, A2062T in the core gene, and two mutations G1764A and C1766T (xV131I) in the basal core promoter (BCP) region. In vitro characterization of the four mutations suggested that the two BCP mutations G1764A and C1766T contributed to the increased viral replication and viral infectivity. A robust in vitro long-term HBV viral passage system has been established by passaging a UV-treated clinical isolate in an optimized PHH/fibroblast coculture system. The two BCP mutations played a key role in the virus's ability to passage. This passage system can be used for studying the entire life cycle of HBV and has the potential for in vitro drug-resistance selection upon further optimization. IMPORTANCE The existing cell culture-based methods to study HBV have limitations and do not allow for viral long-term passage. In this study, an HBV genotype A clinical isolate was subjected to multiple rounds of UV treatment and passaged in an optimized PHH/human fibroblast coculture system. This passaged UV-mutated isolate carried four mutations across the HBV genome, and in vitro characterization of the four mutations suggested that the two basal core promoter (BCP) mutations G1764A and C1766T played a key role in the virus's ability to passage. In summary, we have developed a robust in vitro long-term HBV viral passage system by passaging an UV-treated HBV genotype A clinical isolate in an optimized PHH/human fibroblast coculture system. This passage system can be used for studying the entire life cycle of HBV and has the potential for in vitro drug-resistance selection upon further optimization.


Assuntos
Técnicas de Cocultura , Vírus da Hepatite B , Hepatite B , Virologia , DNA Viral/genética , Fibroblastos/virologia , Genótipo , Hepatite B/virologia , Vírus da Hepatite B/genética , Hepatócitos/virologia , Humanos , Mutagênese , Mutação , Virologia/métodos , Replicação Viral
10.
J Biol Chem ; 298(7): 102097, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35660020

RESUMO

Hepatitis B virus (HBV) infection is a major global health problem with no established cure. Dedicator of cytokinesis 11 (DOCK11), known as a guanine nucleotide exchange factor (GEF) for Cdc42, is reported to be essential for the maintenance of HBV. However, potential therapeutic strategies targeting DOCK11 have not yet been explored. We have previously developed an in vitro virus method as a more efficient tool for the analysis of proteomics and evolutionary protein engineering. In this study, using the in vitro virus method, we screened and identified a novel antiasialoglycoprotein receptor (ASGR) antibody, ASGR3-10M, and a DOCK11-binding peptide, DCS8-42A, for potential use in HBV infection. We further constructed a fusion protein (10M-D42AN) consisting of ASGR3-10M, DCS8-42A, a fusogenic peptide, and a nuclear localization signal to deliver the peptide inside hepatocytes. We show using immunofluorescence staining that 10M-D42AN was endocytosed into early endosomes and released into the cytoplasm and nucleus. Since DCS8-42A shares homology with activated cdc42-associated kinase 1 (Ack1), which promotes EGFR endocytosis required for HBV infection, we also found that 10M-D42AN inhibited endocytosis of EGFR and Ack1. Furthermore, we show 10M-D42AN suppressed the function of DOCK11 in the host DNA repair system required for covalently closed circular DNA synthesis and suppressed HBV proliferation in mice. In conclusion, this study realizes a novel hepatocyte-specific drug delivery system using an anti-ASGR antibody, a fusogenic peptide, and DOCK11-binding peptide to provide a novel treatment for HBV.


Assuntos
Sistemas de Liberação de Medicamentos , Fatores de Troca do Nucleotídeo Guanina , Vírus da Hepatite B , Hepatite B , Anticorpos de Cadeia Única , Animais , DNA Circular/genética , Receptores ErbB/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hepatite B/tratamento farmacológico , Vírus da Hepatite B/patogenicidade , Vírus da Hepatite B/fisiologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Camundongos , Peptídeos/metabolismo , Anticorpos de Cadeia Única/metabolismo , Replicação Viral/genética
11.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(2): 285-290, 2022 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-35332731

RESUMO

Objective: To explore the internal mechanism of hepatocellular carcinoma (HCC) induced by chronic hepatitis B virus (HBV) infection. Methods: L02, HepG2 and Huh7 cells stably overexpressing HBV preS1 antigen were analyzed by flow cytometry, qRT-PCR and tumorigenesis in nude mice to evaluate the effect of preS1 antigen in HBV-related hepatocarcinogenesis. Results: Our results showed that the expression of cancer stem cell (CSCs) related factors and cell surface markers in preS1 overexpressing cells were up-regulated, and the tumorigenicity of these cells was enhanced in nude mice. In addition, preS1 overexpression could down-regulate the expression of major histocompatibility complex Ⅰ (MHC-Ⅰ). The expression of MHC-Ⅰ on the cell surface could be restored by adding interferon gamma (IFN-γ) in the process of cell culturing and the tumorigenicity of cells in nude mice could thus be reduced. Conclusion: Based on the above results, we believe that preS1 is a carcinogen of HBV and that it promotes the formation of liver cancer through down regulating MHC-Ⅰ on the surface of hepatocytes.


Assuntos
Carcinoma Hepatocelular , Genes MHC Classe I , Antígenos de Superfície da Hepatite B , Hepatite B Crônica , Neoplasias Hepáticas , Precursores de Proteínas , Animais , Carcinogênese , Carcinoma Hepatocelular/virologia , Antígenos de Superfície da Hepatite B/genética , Vírus da Hepatite B/genética , Hepatite B Crônica/complicações , Hepatócitos/virologia , Neoplasias Hepáticas/virologia , Camundongos , Camundongos Nus , Precursores de Proteínas/genética
12.
J Virol ; 96(7): e0199521, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35297672

RESUMO

C-type lectin domain-containing proteins (CTLDcps) shape host responses to pathogens and infectious disease outcomes. Previously, we identified the murine CTLDcp Cd302 as restriction factor, limiting hepatitis C virus (HCV) infection of murine hepatocytes. In this study, we investigated in detail the human orthologue's ability to restrict HCV infection in human liver cells. CD302 overexpression in Huh-7.5 cells potently inhibited infection of diverse HCV chimeras representing seven genotypes. Transcriptional profiling revealed abundant CD302 mRNA expression in human hepatocytes, the natural cellular target of HCV. Knockdown of endogenously expressed CD302 modestly enhanced HCV infection of Huh-7.5 cells and primary human hepatocytes. Functional analysis of naturally occurring CD302 transcript variants and engineered CD302 mutants showed that the C-type lectin-like domain (CTLD) is essential for HCV restriction, whereas the cytoplasmic domain (CPD) is dispensable. Coding single nucleotide polymorphisms occurring in human populations and mapping to different domains of CD302 did not influence the capacity of CD302 to restrict HCV. Assessment of the anti-HCV phenotype at different life cycle stages indicated that CD302 preferentially targets the viral entry step. In contrast to the murine orthologue, overexpression of human CD302 did not modulate downstream expression of nuclear receptor-controlled genes. Ectopic CD302 expression restricted infection of liver tropic hepatitis E virus (HEV), while it did not affect infection rates of two respiratory viruses, including respiratory syncytial virus (RSV) and the alpha coronavirus HVCoV-229E. Together, these findings suggest that CD302 contributes to liver cell-intrinsic defense against HCV and might mediate broader antiviral defenses against additional hepatotropic viruses. IMPORTANCE The liver represents an immunoprivileged organ characterized by enhanced resistance to immune responses. However, the importance of liver cell-endogenous, noncytolytic innate immune responses in pathogen control is not well defined. Although the role of myeloid cell-expressed CTLDcps in host responses to viruses has been characterized in detail, we have little information about their potential functions in the liver and their relevance for immune responses in this organ. Human hepatocytes endogenously express the CTLDcp CD302. Here, we provide evidence that CD302 limits HCV infection of human liver cells, likely by inhibiting a viral cell entry step. We confirm that the dominant liver-expressed transcript variant, as well as naturally occurring coding variants of CD302, maintain the capacity to restrict HCV. We further show that the CTLD of the protein is critical for the anti-HCV activity and that overexpressed CD302 limits HEV infection. Thus, CD302 likely contributes to human liver-intrinsic antiviral defenses.


Assuntos
Hepacivirus , Hepatite C , Lectinas Tipo C , Receptores de Superfície Celular , Antivirais/metabolismo , Hepacivirus/fisiologia , Hepatite C/imunologia , Hepatócitos/imunologia , Hepatócitos/virologia , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Replicação Viral
13.
Microbiol Spectr ; 10(1): e0132821, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35171034

RESUMO

Chronic infection of hepatitis B virus (HBV) remains a major health burden worldwide. While the immune response has been recognized to play crucial roles in HBV pathogenesis, the direct cytopathic effects of HBV infection and replication on host hepatocytes and the HBV-host interactions are only partially defined due to limited culture systems. Here, based on our recently developed 5 chemical-cultured primary human hepatocytes (5C-PHHs) model that supports long-term HBV infection, we performed multiplexed quantitative analysis of temporal changes of host proteome and transcriptome on PHHs infected by HBV for up to 4 weeks. We showed that metabolic-, complement-, cytoskeleton-, mitochondrial-, and oxidation-related pathways were modulated at transcriptional or posttranscriptional levels during long-term HBV infection, which led to cytopathic effects and could be partially rescued by early, rather than late, nucleot(s)ide analog (NA) administration and could be significantly relieved by blocking viral antigens with RNA interference (RNAi). Overexpression screening of the dysregulated proteins identified a series of host factors that may contribute to pro- or anti-HBV responses of the infected hepatocytes. In conclusion, our results suggest that long-term HBV infection in primary human hepatocytes leads to cytopathic effects through remodeling the proteome and transcriptome and early antiviral treatment may reduce the extent of such effects, indicating a role of virological factors in HBV pathogenesis and a potential benefit of early administration of antiviral treatment. IMPORTANCE Global temporal quantitative proteomic and transcriptomic analysis using long-term hepatitis B virus (HBV)-infected primary human hepatocytes uncovered extensive remodeling of the host proteome and transcriptome and revealed cytopathic effects of long-term viral replication. Metabolic-, complement-, cytoskeleton-, mitochondrial-, and oxidation-related pathways were modulated at transcriptional or posttranscriptional levels, which could be partially rescued by early, rather than late, NA therapy and could be relieved by blocking viral antigens with RNAi. Overexpression screening identified a series of pro- or anti-HBV host factors. These data have deepened the understanding of the mechanisms of viral pathogenesis and HBV-host interactions in hepatocytes, with implications for therapeutic intervention.


Assuntos
Antivirais/farmacologia , Efeito Citopatogênico Viral , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/fisiologia , Hepatite B/tratamento farmacológico , Hepatócitos/virologia , Técnicas de Cultura de Células , Guanina/análogos & derivados , Guanina/farmacologia , Hepatite B/genética , Hepatite B/imunologia , Hepatite B/virologia , Vírus da Hepatite B/genética , Hepatócitos/imunologia , Humanos , Modelos Biológicos , Transcriptoma/efeitos dos fármacos , Replicação Viral
14.
Viruses ; 14(2)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35215828

RESUMO

Treatment options for human cytomegalovirus (CMV) remain limited and are associated with significant adverse effects and the selection of resistant CMV strains in transplant recipients and congenitally infected infants. Although most approved drugs target and inhibit the CMV DNA polymerase, additional agents with distinct mechanisms of action are needed for the treatment and prevention of CMV. In a large high throughput screen using our CMV-luciferase reporter Towne, we identified several unique inhibitors of CMV replication. Here, we synthesize and test in vitro 13 analogs of the original NCGC2955 hit (1). Analogs with no activity against the CMV-luciferase at 10 µM and 30 µM (2-6, 10-14) were removed from further analysis. Three analogs (7-9) inhibited CMV replication in infected human foreskin fibroblasts. The EC50 of (1) was 1.7 ± 0.6 µM and 1.99 ± 0.15 µM, based on luciferase and plaque assay, respectively. Compounds 7, 8, and 9 showed similar activities: the EC50 values of 7 were 0.21 ± 0.06 µM (luciferase) and 0.55 ± 0.06 (plaque), of 8: 0.28 ± 0.06 µM and 0.42 ± 0.07, and of 9: 0.30 ± 0.05 µM (luciferase) and 0.35 ± 0.07 (plaque). The CC50 for 7, 8, and 9 in non-infected human foreskin fibroblasts was > 500µM, yielding a selectivity index of >1500. Compounds 1, 7, and 8 were also tested in CMV-infected primary human hepatocytes and showed a dose-response against CMV by luciferase activity and viral protein expression. None of the active compounds inhibited herpes simplex virus 1 or 2. Compounds 7 and 8 inhibited mouse CMV replication in vitro. Both inhibited CMV at late stages of replication; 7 reduced virus yield at all late time points, although not to the same degree as letermovir. Finally, the activity of analog 8 was additive with newly identified CMV inhibitors (MLS8969, NFU1827, MSL8554, and MSL8091) and with ganciclovir. Further structural activity development should provide promising anti-CMV agents for use in clinical studies.


Assuntos
Antivirais/síntese química , Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Animais , Células Cultivadas , Citomegalovirus/fisiologia , Ganciclovir/farmacologia , Hepatócitos/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Muromegalovirus/efeitos dos fármacos , Relação Estrutura-Atividade , Carga Viral , Replicação Viral/efeitos dos fármacos
15.
Viruses ; 14(2)2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35215970

RESUMO

During viral evolution and adaptation, many viruses have utilized host cellular factors and machinery as their partners. HBx, as a multifunctional viral protein encoded by the hepatitis B virus (HBV), promotes HBV replication and greatly contributes to the development of HBV-associated hepatocellular carcinoma (HCC). HBx interacts with several host factors in order to regulate HBV replication and evolve carcinogenesis. The cellular FADD-like IL-1ß-converting enzyme (FLICE)-like inhibitory protein (c-FLIP) is a major factor that functions in a variety of cellular pathways and specifically in apoptosis. It has been shown that the interaction between HBx and c-FLIP determines HBV fate. In this review, we provide a comprehensive and detailed overview of the interplay between c-FLIP and HBV in various environmental circumstances. We describe strategies adapted by HBV to establish its chronic infection. We also summarize the conventional roles of c-FLIP and highlight the functional outcome of the interaction between c-FLIP and HBV or other viruses in viral replication and the innate immune system.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Vírus da Hepatite B/fisiologia , Interações Hospedeiro-Patógeno , Sistema Imunitário/metabolismo , Replicação Viral , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Carcinoma Hepatocelular/virologia , Hepatócitos/virologia , Humanos , Neoplasias Hepáticas/virologia , Proteínas Virais Reguladoras e Acessórias
16.
J Immunol ; 208(3): 672-684, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35022275

RESUMO

Hepatitis B virus (HBV)/hepatitis C virus (HCV) coinfection accelerates liver fibrosis progression compared with HBV or HCV monoinfection. Octamer binding transcription factor 4 (OCT4) and Nanog are direct targets of the profibrogenic TGF-ß1 signaling cascade. We leveraged a coculture model to monitor the effects of HBV and HCV coinfection on fibrogenesis in both sodium taurocholate cotransporting polypeptide-transfected Huh7.5.1 hepatoma cells and LX2 hepatic stellate cells (HSCs). We used CRISPR-Cas9 to knock out OCT4 and Nanog to evaluate their effects on HBV-, HCV-, or TGF-ß1-induced liver fibrogenesis. HBV/HCV coinfection and HBx, HBV preS2, HCV Core, and HCV NS2/3 overexpression increased TGF-ß1 mRNA levels in sodium taurocholate cotransporting polypeptide-Huh7.5.1 cells compared with controls. HBV/HCV coinfection further enhanced profibrogenic gene expression relative to HBV or HCV monoinfection. Coculture of HBV and HCV monoinfected or HBV/HCV coinfected hepatocytes with LX2 cells significantly increased profibrotic gene expression and LX2 cell invasion and migration. OCT4 and Nanog guide RNA independently suppressed HBV-, HCV-, HBV/HCV-, and TGF-ß1-induced α-SMA, TIMP-1, and Col1A1 expression and reduced Huh7.5.1, LX2, primary hepatocyte, and primary human HSC migratory capacity. OCT4/Nanog protein expression also correlated positively with fibrosis stage in liver biopsies from patients with chronic HBV or HCV infection. In conclusion, HBV and HCV independently and cooperatively promote liver fibrogenesis through a TGF-ß1-induced OCT4/Nanog-dependent pathway.


Assuntos
Hepatite B/patologia , Hepatite C/patologia , Cirrose Hepática/patologia , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Actinas/biossíntese , Adulto , Sistemas CRISPR-Cas/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Coinfecção/patologia , Cadeia alfa 1 do Colágeno Tipo I/biossíntese , Feminino , Técnicas de Inativação de Genes , Hepacivirus/metabolismo , Células Estreladas do Fígado/patologia , Células Estreladas do Fígado/virologia , Vírus da Hepatite B/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Fígado/patologia , Cirrose Hepática/virologia , Masculino , Proteína Homeobox Nanog/genética , Fator 3 de Transcrição de Octâmero/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Inibidor Tecidual de Metaloproteinase-1/biossíntese
17.
Nat Commun ; 13(1): 105, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013224

RESUMO

Zika virus (ZIKV) infection can be associated with neurological pathologies, such as microcephaly in newborns and Guillain-Barre syndrome in adults. Effective therapeutics are currently not available. As such, a comprehensive understanding of virus-host interactions may guide the development of medications for ZIKV. Here we report a human genome-wide overexpression screen to identify host factors that regulate ZIKV infection and find TMEM120A as a ZIKV restriction factor. TMEM120A overexpression significantly inhibits ZIKV replication, while TMEM120A knockdown increases ZIKV infection in cell lines. Moreover, Tmem120a knockout in mice facilitates ZIKV infection in primary mouse embryonic fibroblasts (MEF) cells. Mechanistically, the antiviral activity of TMEM120A is dependent on STING, as TMEM120A interacts with STING, promotes the translocation of STING from the endoplasmic reticulum (ER) to ER-Golgi intermediate compartment (ERGIC) and enhances the phosphorylation of downstream TBK1 and IRF3, resulting in the expression of multiple antiviral cytokines and interferon-stimulated genes. In summary, our gain-of-function screening identifies TMEM120A as a key activator of the antiviral signaling of STING.


Assuntos
Interações Hospedeiro-Patógeno/genética , Canais Iônicos/genética , Proteínas de Membrana/genética , Infecção por Zika virus/genética , Zika virus/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Feminino , Regulação da Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Hepatócitos/imunologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Canais Iônicos/deficiência , Canais Iônicos/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Zika virus/crescimento & desenvolvimento , Zika virus/patogenicidade , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
18.
Signal Transduct Target Ther ; 7(1): 23, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35078968
19.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055105

RESUMO

Hepatitis C virus (HCV) relies on cellular lipid metabolism for its replication, and actively modulates lipogenesis and lipid trafficking in infected hepatocytes. This translates into an intracellular accumulation of triglycerides leading to liver steatosis, cirrhosis and hepatocellular carcinoma, which are hallmarks of HCV pathogenesis. While the interaction of HCV with hepatocyte metabolic pathways is patent, how viral proteins are able to redirect central carbon metabolism towards lipogenesis is unclear. Here, we report that the HCV protein NS5A activates the glucokinase (GCK) isoenzyme of hexokinases through its D2 domain (NS5A-D2). GCK is the first rate-limiting enzyme of glycolysis in normal hepatocytes whose expression is replaced by the hexokinase 2 (HK2) isoenzyme in hepatocellular carcinoma cell lines. We took advantage of a unique cellular model specifically engineered to re-express GCK instead of HK2 in the Huh7 cell line to evaluate the consequences of NS5A-D2 expression on central carbon and lipid metabolism. NS5A-D2 increased glucose consumption but decreased glycogen storage. This was accompanied by an altered mitochondrial respiration, an accumulation of intracellular triglycerides and an increased production of very-low density lipoproteins. Altogether, our results show that NS5A-D2 can reprogram central carbon metabolism towards a more energetic and glycolytic phenotype compatible with HCV needs for replication.


Assuntos
Glucoquinase/metabolismo , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Glicogênio/metabolismo , Glicólise , Interações Hospedeiro-Patógeno , Humanos , Metabolismo dos Lipídeos , Lipogênese , Mitocôndrias/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , RNA Polimerase Dependente de RNA/química , Proteínas não Estruturais Virais/química
20.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35058348

RESUMO

Novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants pose a challenge to controlling the COVID-19 pandemic. Previous studies indicate that clinical samples collected from individuals infected with the Delta variant may contain higher levels of RNA than previous variants, but the relationship between levels of viral RNA and infectious virus for individual variants is unknown. We measured infectious viral titer (using a microfocus-forming assay) and total and subgenomic viral RNA levels (using RT-PCR) in a set of 162 clinical samples containing SARS-CoV-2 Alpha, Delta, and Epsilon variants that were collected in identical swab kits from outpatient test sites and processed soon after collection. We observed a high degree of variation in the relationship between viral titers and RNA levels. Despite this, the overall infectivity differed among the three variants. Both Delta and Epsilon had significantly higher infectivity than Alpha, as measured by the number of infectious units per quantity of viral E gene RNA (5.9- and 3.0-fold increase; P < 0.0001, P = 0.014, respectively) or subgenomic E RNA (14.3- and 6.9-fold increase; P < 0.0001, P = 0.004, respectively). In addition to higher viral RNA levels reported for the Delta variant, the infectivity (amount of replication competent virus per viral genome copy) may be increased compared to Alpha. Measuring the relationship between live virus and viral RNA is an important step in assessing the infectivity of novel SARS-CoV-2 variants. An increase in the infectivity for Delta may further explain increased spread, suggesting a need for increased measures to prevent viral transmission.


Assuntos
COVID-19/epidemiologia , Regulação Viral da Expressão Gênica , Genoma Viral , RNA Viral/genética , SARS-CoV-2/genética , SARS-CoV-2/patogenicidade , Animais , COVID-19/patologia , COVID-19/transmissão , COVID-19/virologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteínas do Envelope de Coronavírus/genética , Proteínas do Envelope de Coronavírus/metabolismo , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , RNA Viral/metabolismo , SARS-CoV-2/classificação , SARS-CoV-2/metabolismo , Células Vero , Carga Viral , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...