Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.002
Filtrar
1.
Hematology ; 29(1): 2392028, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39222052

RESUMEN

OBJECTIVE: To assess the fibrinogen function in patients with hepatitis B-related cirrhosis and explore the relationship between dysfibrinogenemia and bleeding and thrombotic events. METHODS: Medical records and laboratory data of the patients with hepatitis B-related cirrhosis were collected. Patients were categorized into three groups based on the Child-Pugh score. Fibrinogen activity and antigen, fibrinogen-bound sialic acid (FSA), fibrinogen polymerization and fibrinolysis kinetic analysis, thrombin-antithrombin complex (TAT) and plasmin-α2-antiplasmin complex (PAP) were detected. RESULTS: Eighty patients with seventeen, thirty-eight and twenty-five in Child-Pugh A, B and C, respectively, were included. Seventeen patients experienced bleeding events and eight patients had thrombotic events. Fibrinogen activity and antigen levels were reduced with the severity of cirrhosis. Twenty-two patients exhibited dysfibrinogenemia. The FSA levels in patients with non-dysfibrinogenemia and those with dysfibrinogenemia were increased to 1.25 and 1.37 times of healthy controls, negatively correlated with fibrinogen activity (ρ = -0.393, p = 0.006). Compared to healthy controls, the amount of clot formation was reduced (p < 0.001), the polymerization was delayed (p < 0.001) and the rate of fibrinolysis was reduced (p < 0.001). The TAT levels were significantly increased in the Child-Pugh C patients compared to the Child-Pugh B patients (p = 0.032) while the PAP levels were comparable among 3 groups (p = 0.361). CONCLUSION: Sialylation of fibrinogen is one of the main causes of modifications of fibrinogen in patients with hepatitis B-related cirrhosis. The polymerization and fibrinolysis functions of fibrinogen are impaired. The degree of impaired fibrinolysis function is more severe than that of polymerization function, and may be partly related to the occurrence of thrombotic events.


Asunto(s)
Fibrinógeno , Fibrinólisis , Hepatitis B , Cirrosis Hepática , Humanos , Masculino , Femenino , Fibrinógeno/metabolismo , Fibrinógeno/análisis , Persona de Mediana Edad , Cirrosis Hepática/sangre , Cirrosis Hepática/metabolismo , Cirrosis Hepática/complicaciones , Hepatitis B/complicaciones , Hepatitis B/sangre , Hepatitis B/metabolismo , Adulto , Anciano
2.
PLoS Pathog ; 20(9): e1012485, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39259704

RESUMEN

Hepatitis B virus (HBV) exploits the endosomal sorting complexes required for transport (ESCRT)/multivesicular body (MVB) pathway for virion budding. In addition to enveloped virions, HBV-replicating cells nonlytically release non-enveloped (naked) capsids independent of the integral ESCRT machinery, but the exact secretory mechanism remains elusive. Here, we provide more detailed information about the existence and characteristics of naked capsid, as well as the viral and host regulations of naked capsid egress. HBV capsid/core protein has two highly conserved Lysine residues (K7/K96) that potentially undergo various types of posttranslational modifications for subsequent biological events. Mutagenesis study revealed that the K96 residue is critical for naked capsid egress, and the intracellular egress-competent capsids are associated with ubiquitinated host proteins. Consistent with a previous report, the ESCRT-III-binding protein Alix and its Bro1 domain are required for naked capsid secretion through binding to intracellular capsid, and we further found that the ubiquitinated Alix binds to wild type capsid but not K96R mutant. Moreover, screening of NEDD4 E3 ubiquitin ligase family members revealed that AIP4 stimulates the release of naked capsid, which relies on AIP4 protein integrity and E3 ligase activity. We further demonstrated that AIP4 interacts with Alix and promotes its ubiquitination, and AIP4 is essential for Alix-mediated naked capsid secretion. However, the Bro1 domain of Alix is non-ubiquitinated, indicating that Alix ubiquitination is not absolutely required for AIP4-induced naked capsid secretion. Taken together, our study sheds new light on the mechanism of HBV naked capsid egress in viral life cycle.


Asunto(s)
Cápside , Virus de la Hepatitis B , Ubiquitina-Proteína Ligasas Nedd4 , Ubiquitina-Proteína Ligasas , Liberación del Virus , Humanos , Proteínas de Unión al Calcio , Cápside/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/fisiología , Virus de la Hepatitis B/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Liberación del Virus/fisiología
3.
Int J Biol Sci ; 20(11): 4178-4189, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247820

RESUMEN

Currently, chronic hepatitis B virus infection is still one of the most serious public health problems in the world. Though current strategies are effective in controlling infection and slowing down the disease process, it remains a big challenge to achieve a functional cure for chronic hepatitis B in a majority of patients due to the inability to clear the cccDNA pool. The mammalian target of rapamycin (mTOR) integrates nutrition, energy, growth factors, and other extracellular signals, participating in gene transcription, protein translation, ribosome synthesis, and other biological processes. Additionally, mTOR plays an extremely important role in cell growth, apoptosis, autophagy, and metabolism. More and more evidence show that HBV infection can activate the mTOR pathway, suggesting that HBV uses or hijacks the mTOR pathway to facilitate its own replication. Therefore, mTOR signaling pathway may be a key target for controlling HBV infection. However, the role of the central cytokine mTOR in the pathogenesis of HBV infection has not yet been systematically addressed. Notably, mTOR is commonly activated in hepatocellular carcinoma, which can progress from chronic hepatitis B. This review systematically summarizes the role of mTOR in the life cycle of HBV and its impact on the clinical progression of HBV infection.


Asunto(s)
Carcinoma Hepatocelular , Virus de la Hepatitis B , Neoplasias Hepáticas , Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Virus de la Hepatitis B/fisiología , Hepatitis B/metabolismo , Animales , Hepatitis B Crónica/metabolismo
4.
Nat Commun ; 15(1): 7856, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251623

RESUMEN

Despite recent advances in systemic therapy for hepatocellular carcinoma (HCC), the prognosis of hepatitis B virus (HBV)-induced HCC patients remains poor. By screening a sgRNA library targeting human deubiquitinases, we find that ubiquitin-specific peptidase 26 (USP26) deficiency impairs HBV-positive HCC cell proliferation. Genetically engineered murine models with Usp26 knockout confirm that Usp26 drives HCC tumorigenesis. Mechanistically, we find that the HBV-encoded protein HBx binds to the promoter and induces the production of USP26, which is an X-linked gene exclusively expressed in the testis. HBx consequently promotes the association of USP26 with SIRT1 to synergistically stabilize SIRT1 by deubiquitination, which promotes cell proliferation and impedes cell apoptosis to accelerate HCC tumorigenesis. In patients with HBV-positive HCC, USP26 is robustly induced, and its levels correlate with SIRT1 levels and poor prognosis. Collectively, our study highlights a causative link between HBV infection, deubiquitinase induction and development of HCC, identifying a druggable target, USP26.


Asunto(s)
Carcinoma Hepatocelular , Proliferación Celular , Epigénesis Genética , Virus de la Hepatitis B , Neoplasias Hepáticas , Sirtuina 1 , Transactivadores , Proteínas Reguladoras y Accesorias Virales , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Humanos , Animales , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Virus de la Hepatitis B/genética , Ratones , Sirtuina 1/metabolismo , Sirtuina 1/genética , Transactivadores/metabolismo , Transactivadores/genética , Masculino , Proliferación Celular/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Carcinogénesis/genética , Hepatitis B/virología , Hepatitis B/complicaciones , Hepatitis B/genética , Hepatitis B/metabolismo , Línea Celular Tumoral , Ratones Noqueados , Regulación Neoplásica de la Expresión Génica , Enzimas Desubicuitinizantes/metabolismo , Enzimas Desubicuitinizantes/genética , Apoptosis/genética , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Regiones Promotoras Genéticas/genética
5.
Lipids Health Dis ; 23(1): 268, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39182089

RESUMEN

BACKGROUND: The progression of tumours is related to abnormal phospholipid metabolism. This study is anticipated to present a fresh perspective for disease therapy targets of hepatocarcinoma caused by hepatitis B virus in the future by screening feature genes related to phospholipid metabolism. METHODS: This study analysed GSE121248 to pinpoint differentially expressed genes (DEGs). By examining the overlap between the metabolism-related genes and DEGs, the research focused on the genes involved in phospholipid metabolism. To find feature genes, functional enrichment studies were carried out and a network diagram was proposed. These findings were validated via data base of The Cancer Genome Atlas (TCGA). Further analyses included immune infiltration studies and metabolomics. Finally, the relationships between differentially abundant metabolites and feature genes were confirmed by molecular docking, providing a thorough comprehension of the molecular mechanisms. RESULTS: The seven genes with the highest degree of connection (PTGS2, IGF1, SPP1, BCHE, NR1I2, NAMPT, and FABP1) were identified as feature genes. In the TCGA database, the seven feature genes also had certain diagnostic efficiency. Immune infiltration analysis revealed that feature genes regulate the infiltration of various immune cells. Metabolomics successfully identified the different metabolites of the phospholipid metabolism pathway between patients and normal individuals. The docking study indicated that different metabolites may play essential roles in causing disease by targeting feature genes. CONCLUSIONS: In this study, for the first time, it reveals the possible involvement of genes linked to phospholipid metabolism-related genes using bioinformatics analysis. Identifying genes and probable therapeutic targets could provide clues for the further treatment of disease.


Asunto(s)
Carcinoma Hepatocelular , Virus de la Hepatitis B , Hepatitis B , Neoplasias Hepáticas , Simulación del Acoplamiento Molecular , Fosfolípidos , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/metabolismo , Hepatitis B/genética , Hepatitis B/complicaciones , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/genética , Fosfolípidos/metabolismo , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Metabolómica/métodos , Perfilación de la Expresión Génica
6.
Cell Mol Life Sci ; 81(1): 345, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39133305

RESUMEN

BACKGROUND AND AIMS: Hepatitis B virus (HBV)-associated liver cirrhosis (LC), a common condition with high incidence and mortality rates, is often associated with diabetes mellitus (DM). However, the molecular mechanisms underlying impaired glucose regulation during HBV-associated LC remain unclear. METHODS: Data from 63 patients with LC and 62 patients with LC-associated DM were analysed. Co-culture of NK cells and islet ß cell lines were used to study the glucose regulation mechanism. A mouse model of LC was used to verify the effect of S100A8/A9 on the glucose regulation. RESULTS: Higher levels of interferon (IFN)-γ derived from natural killer (NK) cells and lower levels of insulin emerged in the peripheral blood of patients with both LC and DM compared with those from patients with LC only. IFN-γ derived from NK cells facilitated ß cell necroptosis and impaired insulin production. Furthermore, S100A8/A9 elevation in patients with both LC and DM was found to upregulate IFN-γ production in NK cells. Consistently, in the mouse model for LC, mice treated with carbon tetrachloride (CCL4) and S100A8/A9 exhibited increased blood glucose, impaired insulin production, increased IFN-γ, and increased ß cells necroptosis compared with those treated with CCL4. Mechanistically, S100A8/A9 activated the p38 MAPK pathway to increase IFN-γ production in NK cells. These effects were diminished after blocking RAGE. CONCLUSION: Together, the data indicate that IFN-γ produced by NK cells induces ß cell necroptosis via the S100A8/A9-RAGE-p38 MAPK axis in patients with LC and DM. Reduced levels of S100A8/A9, NK cells, and IFN-γ could be valuable for the treatment of LC with DM. Accumulation of S100A8/A9 in patients with LC may indicate the emergence of DM.


Asunto(s)
Calgranulina A , Calgranulina B , Virus de la Hepatitis B , Células Secretoras de Insulina , Interferón gamma , Células Asesinas Naturales , Cirrosis Hepática , Necroptosis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Humanos , Animales , Interferón gamma/metabolismo , Calgranulina B/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/virología , Cirrosis Hepática/inmunología , Ratones , Masculino , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/virología , Calgranulina A/metabolismo , Ratones Endogámicos C57BL , Femenino , Persona de Mediana Edad , Hepatitis B/complicaciones , Hepatitis B/patología , Hepatitis B/metabolismo , Modelos Animales de Enfermedad , Tetracloruro de Carbono
7.
Sci Rep ; 14(1): 19917, 2024 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-39198597

RESUMEN

Small extracellular vesicles (sEVs) have the ability to transfer genetic material between cells, but their role in mediating HBV infection and regulating M1 macrophages to promote immune evasion remains unclear. In this study, we utilized PMA + LPS + IFN-γ to induce THP-1 into M1 macrophages. We then extracted sEVs from HepG2.2.15 cell and treated the M1 macrophages with these sEVs. QPCR detection revealed the presence of HBV-DNA in the M1 macrophages. Additionally, RT-qPCR and WB analysis demonstrated a significantly decreased in the expression of TLR4, NLRP3, pro-caspase-1, caspase-1p20, IL-1ß and IL-18 in the M1 macrophages (P < 0.05). Furthermore, RT-qPCR results displayed high expression levels of that miR-146a and FEN-1 in the sEVs derived from HepG2.2.15 cells (P < 0.01). RT -qPCR and WB analysis showed that these sEVs enhanced the expression of FEN-1 or miR-146a in the M1 macrophages through miR-146a or FEN-1 (P < 0.05), while simultaneously reducing the expression of TLR4, NLRP3, caspase-1p20, IL-1ß and IL-18 in the M1 macrophages (P < 0.05). In summary, our findings indicate that sEVs loaded with HBV inhibit the inflammatory function of M1 macrophages and promote immune escape. Additionally, miR-146a and FEN-1 present in the sEVs play a crucial role in this process.


Asunto(s)
Vesículas Extracelulares , Virus de la Hepatitis B , Hepatitis B , Evasión Inmune , Macrófagos , MicroARNs , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/inmunología , Virus de la Hepatitis B/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , MicroARNs/genética , MicroARNs/metabolismo , Células Hep G2 , Hepatitis B/virología , Hepatitis B/inmunología , Hepatitis B/metabolismo , Receptor Toll-Like 4/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Interleucina-18/metabolismo , Células THP-1
8.
Viruses ; 16(7)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39066215

RESUMEN

In sub-Saharan Africa (SSA), the (sub)genotypes A1, D3, and E of the hepatitis B virus (HBV) prevail. Individuals infected with subgenotype A1 have a 4.5-fold increased risk of HCC compared to those infected with other (sub)genotypes. The effect of (sub)genotypes on protein expression and host signalling has not been studied. Mass spectrometry was used to analyse the proteome of Huh7 cells transfected with replication-competent clones. Proteomic analysis revealed significantly differentially expressed proteins between SSA (sub)genotypes. Different (sub)genotypes have the propensity to dysregulate specific host signalling pathways. Subgenotype A1 resulted in dysregulation within the Ras pathway. Ras-associated protein, RhoC, was significantly upregulated in cells transfected with subgenotype A1 compared to those transfected with other (sub)genotypes, on both a proteomic (>1.5-fold) and mRNA level (p < 0.05). Two of the main cellular signalling pathways involving RHOC, MAPK and PI3K/Akt/mTOR, regulate cell growth, motility, and survival. Downstream signalling products of these pathways have been shown to increase MMP2 and MMP9 expression. An extracellular MMP2 and MMP9 ELISA revealed a non-significant increase in MMP2 and MMP9 in the cells transfected with A1 compared to the other (sub)genotypes (p < 0.05). The upregulated Ras-associated proteins have been implicated as oncoproteins in various cancers and could contribute to the increased hepatocarcinogenic potential of A1.


Asunto(s)
Genotipo , Virus de la Hepatitis B , Proteómica , Humanos , Virus de la Hepatitis B/genética , Línea Celular Tumoral , Transducción de Señal , África del Sur del Sahara , Proteoma , Proteína rhoC de Unión a GTP/metabolismo , Proteína rhoC de Unión a GTP/genética , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Transfección , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Hepatitis B/virología , Hepatitis B/metabolismo , Hepatitis B/genética
9.
Sci Rep ; 14(1): 13981, 2024 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886481

RESUMEN

Occult hepatitis B virus infection (OBI) is characterized by the presence of HBV DNA in the absence of detectable HBsAg. OBI is an important risk factor for cirrhosis and hepatocellular carcinoma, but its pathogenesis has not been fully elucidated. Mutations in the HBV preS/S genes can lead to impaired secretion of either HBsAg or S-protein resulting in the accumulation of defective viruses or S protein in cells. In our previous work, the M133S mutation was present in the HBV S gene of maintenance hemodialysis (MHD) patients with OBI. In this study, we investigated the potential role of amino acid substitutions in S proteins in S protein production and secretion through the construction of mutant S gene plasmids, structural prediction, transcriptome sequencing analysis, and in vitro functional studies. Protein structure prediction showed that the S protein M133S mutant exhibited hydrophilic modifications, with greater aggregation and accumulation of the entire structure within the membrane phospholipid bilayer. Differential gene enrichment analysis of transcriptome sequencing data showed that differentially expressed genes were mainly concentrated in protein processing in the endoplasmic reticulum (ER). The expression of heat shock family proteins and ER chaperone molecules was significantly increased in the wild-type and mutant groups, whereas the expression of mitochondria-associated proteins was decreased. Immunofluorescence staining and protein blotting showed that the endoplasmic reticulum-associated protein PDI, the autophagy marker LC3, and the lysosome-associated protein LAMP2 co-localized with the S proteins in the wild-type and mutant strains, and their expression was increased. The mitochondria-associated TOMM20 protein was also co-expressed with the S protein, but expression was significantly reduced in the mutant. The M133S mutation in the S gene is expressed as a defective and misfolded protein that accumulates in the endoplasmic reticulum causing secretion-impaired endoplasmic reticulum stress, which in turn triggers mitochondrial autophagy and recruits lysosomes to fuse with the autophagosome, leading to mitochondrial clearance. This study preliminarily demonstrated that the mutation of M133S in the S gene can cause OBI and is associated with disease progression, providing a theoretical basis for the diagnosis and treatment of OBI.


Asunto(s)
Estrés del Retículo Endoplásmico , Antígenos de Superficie de la Hepatitis B , Virus de la Hepatitis B , Hepatitis B , Mitofagia , Diálisis Renal , Humanos , Mitofagia/genética , Hepatitis B/virología , Hepatitis B/genética , Hepatitis B/metabolismo , Hepatitis B/complicaciones , Virus de la Hepatitis B/genética , Estrés del Retículo Endoplásmico/genética , Antígenos de Superficie de la Hepatitis B/genética , Antígenos de Superficie de la Hepatitis B/metabolismo , Masculino , Mutación , Femenino , Persona de Mediana Edad , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Mitocondrias/metabolismo , Mitocondrias/genética , Sustitución de Aminoácidos , Adulto
10.
Cell Commun Signal ; 22(1): 280, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773448

RESUMEN

INTRODUCTION: Hepatitis B Virus (HBV) is widely recognized as a "metabolic virus" that disrupts hepatic metabolic homeostasis, rendering it one of the foremost risk factors for hepatocellular carcinoma (HCC). Except for antiviral therapy, the fundamental principles underlying HBV- and HBV+ HCC have remained unchanged, limiting HCC treatment options. OBJECTIVES: In this study, we aim to identify the distinctive metabolic profile of HBV-associated HCC, with the promise of identifying novel metabolic targets that confer survival advantages and ultimately impede cancer progression. METHODS: We employed a comprehensive methodology to evaluate metabolic alterations systematically. Initially, we analyzed transcriptomic and proteomic data obtained from a public database, subsequently validating these findings within our test cohort at both the proteomic and transcriptomic levels. Additionally, we conducted a comprehensive analysis of tissue metabolomics profiles, lipidomics, and the activity of the MAPK and AKT signaling pathway to corroborate the abovementioned changes. RESULTS: Our multi-omics approach revealed distinct metabolic dysfunctions associated with HBV-associated HCC. Specifically, we observed upregulated steroid hormone biosynthesis, primary bile acid metabolism, and sphingolipid metabolism in HBV-associated HCC patients' serum. Notably, metabolites involved in primary bile acid and sphingolipids can activate the MAPK/mTOR pathway. Tissue metabolomics and lipidomics analyses further validated the serum metabolic alterations, particularly alterations in lipid composition and accumulation of unsaturated fatty acids. CONCLUSION: Our findings emphasize the pivotal role of HBV in HCC metabolism, elucidating the activation of a unique MAPK/mTOR signaling axis by primary bile acids and sphingolipids. Moreover, the hyperactive MAPK/mTOR signaling axis transduction leads to significant reprogramming in lipid metabolism within HCC cells, further triggering the activation of the MAPK/mTOR pathway in turn, thereby establishing a self-feeding circle driven by primary bile acids and sphingolipids.


Asunto(s)
Carcinoma Hepatocelular , Virus de la Hepatitis B , Neoplasias Hepáticas , Serina-Treonina Quinasas TOR , Humanos , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Serina-Treonina Quinasas TOR/metabolismo , Virus de la Hepatitis B/fisiología , Metabolismo de los Lípidos , Masculino , Lípidos/sangre , Transducción de Señal , Sistema de Señalización de MAP Quinasas , Hepatitis B/complicaciones , Hepatitis B/virología , Hepatitis B/metabolismo , Persona de Mediana Edad , Femenino
11.
Clin Mol Hepatol ; 30(3): 539-560, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38741238

RESUMEN

BACKGROUND/AIMS: The major histocompatibility class II (MHC II) transactivator, known as CIITA, is induced by Interferon gamma (IFN-γ) and plays a well-established role in regulating the expression of class II MHC molecules in antigen-presenting cells. METHODS: Primary human hepatocytes (PHH) were isolated via therapeutic hepatectomy from two donors. The hepatocellular carcinoma (HCC) cell lines HepG2 and Huh7 were used for the mechanistic study, and HBV infection was performed in HepG2-NTCP cells. HBV DNA replication intermediates and secreted antigen levels were measured using Southern blotting and ELISA, respectively. RESULTS: We identified a non-canonical function of CIITA in the inhibition of hepatitis B virus (HBV) replication in both HCC cells and patient-derived PHH. Notably, in vivo experiments demonstrated that HBV DNA and secreted antigen levels were significantly decreased in mice injected with the CIITA construct. Mechanistically, CIITA inhibited HBV transcription and replication by suppressing the activity of HBV-specific enhancers/promoters. Indeed, CIITA exerts antiviral activity in hepatocytes through ERK1/2-mediated down-regulation of the expression of hepatocyte nuclear factor 1α (HNF1α) and HNF4α, which are essential factors for virus replication. In addition, silencing of CIITA significantly abolished the IFN-γ-mediated anti-HBV activity, suggesting that CIITA mediates the anti-HBV activity of IFN-γ to some extent. HBV X protein (HBx) counteracts the antiviral activity of CIITA via direct binding and impairing its function. CONCLUSION: Our findings reveal a novel antiviral mechanism of CIITA that involves the modulation of the ERK pathway to restrict HBV transcription. Additionally, our results suggest the possibility of a new immune avoidance mechanism involving HBx.


Asunto(s)
Virus de la Hepatitis B , Hepatocitos , Proteínas Nucleares , Transactivadores , Replicación Viral , Virus de la Hepatitis B/fisiología , Humanos , Transactivadores/metabolismo , Transactivadores/genética , Animales , Ratones , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Hepatocitos/metabolismo , Hepatocitos/citología , Hepatocitos/virología , Células Hep G2 , Hepatitis B/metabolismo , Interferón gamma/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , ADN Viral/metabolismo , Proteínas Reguladoras y Accesorias Virales
12.
Autophagy ; 20(9): 2109-2111, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38752371

RESUMEN

CALCOCO2/NDP52 recognizes LGALS8 (galectin 8)-coated invading bacteria and initiates anti-bacterial autophagy by recruiting RB1CC1/FIP200 and TBKBP1/SINTBAD-AZI2/NAP1. Whether CALCOCO2 exerts similar functions against viral infection is unknown. In our recent study we show that CALCOCO2 targets envelope proteins of hepatitis B virus (HBV) to the lysosome for degradation, resulting in inhibition of viral replication. In contrast to anti-bacterial autophagy, lysosomal degradation of HBV does not require either LGALS8 or ATG5, and CALCOCO2 mutants abolishing the formation of the RB1CC1-CALCOCO2-TBKBP1-AZI2 complex maintain their inhibitory function on the virus. CALCOCO2-mediated inhibition depends on RAB9, which is a key factor in the alternative autophagy pathway. CALCOCO2 forms a complex with RAB9 only in the presence of viral envelope proteins and links HBV to the RAB9-dependent lysosomal degradation pathway. These findings reveal a new mechanism by which CALCOCO2 triggers antiviral responses against HBV infection and suggest direct roles for autophagy receptors in other lysosomal degradation pathways than canonical autophagy.


Asunto(s)
Autofagia , Virus de la Hepatitis B , Hepatitis B , Lisosomas , Proteínas de Unión al GTP rab , Lisosomas/metabolismo , Humanos , Virus de la Hepatitis B/fisiología , Virus de la Hepatitis B/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Hepatitis B/virología , Hepatitis B/metabolismo , Antivirales/farmacología , Antivirales/metabolismo , Proteínas Nucleares/metabolismo
13.
J Virol ; 98(6): e0046824, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38780244

RESUMEN

The antiviral role of the tripartite motif-containing (TRIM) protein family , a member of the E3-ubiquitin ligase family, has recently been actively studied. Hepatitis B virus (HBV) infection is a major contributor to liver diseases; however, the host factors regulated by cytokine-inducible TRIM21 to suppress HBV remain unclear. In this study, we showed the antiviral efficacy of TRIM21 against HBV in hepatoma cell lines, primary human hepatocytes isolated from patient liver tissues, and mouse model. Using TRIM21 knock-out cells, we confirmed that the antiviral effects of interferon-gamma, which suppress HBV replication, are diminished when TRIM21 is deficient. Northern blot analysis confirmed a reduction of HBV RNA levels by TRIM21. Using Luciferase reporter assay, we also discovered that TRIM21 decreases the activity of HBV enhancers, which play a crucial role in covalently closed circular DNA transcription. The participation of the RING domain and PRY-SPRY domain in the anti-HBV effect of TRIM21 was demonstrated through experiments using deletion mutants. We identified a novel interaction between TRIM21 and hepatocyte nuclear factor 4α (HNF4α) through co-immunoprecipitation assay. More specifically, ubiquitination assay revealed that TRIM21 promotes ubiquitin-mediated proteasomal degradation of HNF4α. HNF1α transcription is down-regulated as a result of the degradation of HNF4α, an activator for the HNF1α promoter. Therefore, the reduction of key HBV enhancer activators, HNF4α and HNF1α, by TRIM21 resulted in a decline in HBV transcription, ultimately leading to the inhibition of HBV replication.IMPORTANCEDespite extensive research efforts, a definitive cure for chronic hepatitis B remains elusive, emphasizing the persistent importance of this viral infection as a substantial public health concern. Although the risks associated with hepatitis B virus (HBV) infection are well known, host factors capable of suppressing HBV are largely uncharacterized. This study elucidates that tripartite motif-containing protein 21 (TRIM21) suppresses HBV transcription and consequently inhibits HBV replication by downregulating the hepatocyte nuclear factors, which are host factors associated with the HBV enhancers. Our findings demonstrate a novel anti-HBV mechanism of TRIM21 in interferon-gamma-induced anti-HBV activity. These findings may contribute to new strategies to block HBV.


Asunto(s)
Virus de la Hepatitis B , Factor Nuclear 4 del Hepatocito , Hepatocitos , Interferón gamma , Ribonucleoproteínas , Replicación Viral , Humanos , Virus de la Hepatitis B/fisiología , Animales , Ratones , Interferón gamma/farmacología , Interferón gamma/metabolismo , Hepatocitos/virología , Hepatocitos/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/genética , Hepatitis B/virología , Hepatitis B/metabolismo , Células Hep G2 , Línea Celular Tumoral
14.
Viruses ; 16(5)2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38793626

RESUMEN

HBV infection is challenging to cure due to the persistence of viral covalently closed circular viral DNA (cccDNA). The dedicator of cytokinesis 11 (DOCK11) is recognized as a guanine nucleotide exchange factor (GEF) for CDC42 that has been reported to be required for HBV persistence. DOCK11 is expressed in both the cytoplasm and nucleus of human hepatocytes and is functionally associated with retrograde trafficking proteins Arf-GAP with GTPase domain, ankyrin repeat, and pleckstrin homology domain-containing protein 2 (AGAP2), and ADP-ribosylation factor 1 (ARF1), together with the HBV capsid, in the trans-Golgi network (TGN). This opens an alternative retrograde trafficking route for HBV from early endosomes (EEs) to the TGN and then to the endoplasmic reticulum (ER), thereby avoiding lysosomal degradation. DOCK11 also facilitates the association of cccDNA with H3K4me3 and RNA Pol II for activating cccDNA transcription. In addition, DOCK11 plays a crucial role in the host DNA repair system, being essential for cccDNA synthesis. This function can be inhibited by 10M-D42AN, a novel DOCK11-binding peptide, leading to the suppression of HBV replication both in vitro and in vivo. Treatment with a combination of 10M-D42AN and entecavir may represent a promising therapeutic strategy for patients with chronic hepatitis B (CHB). Consequently, DOCK11 may be seen as a potential candidate molecule in the development of molecularly targeted drugs against CHB.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido , Virus de la Hepatitis B , Hepatocitos , Humanos , Virus de la Hepatitis B/fisiología , Virus de la Hepatitis B/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Hepatocitos/virología , Hepatocitos/metabolismo , Internalización del Virus , Replicación Viral , Hepatitis B/virología , Hepatitis B/metabolismo , ADN Viral/metabolismo , ADN Viral/genética , Animales
15.
Microbiol Spectr ; 12(5): e0378823, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38567974

RESUMEN

The key to a curative treatment of hepatitis B virus (HBV) infection is the eradication of the intranuclear episomal covalently closed circular DNA (cccDNA), the stable persistence reservoir of HBV. Currently, established therapies can only limit HBV replication but fail to tackle the cccDNA. Thus, novel therapeutic approaches toward curative treatment are urgently needed. Recent publications indicated a strong association between the HBV core protein SUMOylation and the association with promyelocytic leukemia nuclear bodies (PML-NBs) on relaxed circular DNA to cccDNA conversion. We propose that interference with the cellular SUMOylation system and PML-NB integrity using arsenic trioxide provides a useful tool in the treatment of HBV infection. Our study showed a significant reduction in HBV-infected cells, core protein levels, HBV mRNA, and total DNA. Additionally, a reduction, albeit to a limited extent, of HBV cccDNA could be observed. Furthermore, this interference was also applied for the treatment of an established HBV infection, characterized by a stably present nuclear pool of cccDNA. Arsenic trioxide (ATO) treatment not only changed the amount of expressed HBV core protein but also induced a distinct relocalization to an extranuclear phenotype during infection. Moreover, ATO treatment resulted in the redistribution of transfected HBV core protein away from PML-NBs, a phenotype similar to that previously observed with SUMOylation-deficient HBV core. Taken together, these findings revealed the inhibition of HBV replication by ATO treatment during several steps of the viral replication cycle, including viral entry into the nucleus as well as cccDNA formation and maintenance. We propose ATO as a novel prospective treatment option for further pre-clinical and clinical studies against HBV infection. IMPORTANCE: The main challenge for the achievement of a functional cure for hepatitis B virus (HBV) is the covalently closed circular DNA (cccDNA), the highly stable persistence reservoir of HBV, which is maintained by further rounds of infection with newly generated progeny viruses or by intracellular recycling of mature nucleocapsids. Eradication of the cccDNA is considered to be the holy grail for HBV curative treatment; however, current therapeutic approaches fail to directly tackle this HBV persistence reservoir. The molecular effect of arsenic trioxide (ATO) on HBV infection, protein expression, and cccDNA formation and maintenance, however, has not been characterized and understood until now. In this study, we reveal ATO treatment as a novel and innovative therapeutic approach against HBV infections, repressing viral gene expression and replication as well as the stable cccDNA pool at low micromolar concentrations by affecting the cellular function of promyelocytic leukemia nuclear bodies.


Asunto(s)
Trióxido de Arsénico , Núcleo Celular , ADN Circular , ADN Viral , Virus de la Hepatitis B , Hepatitis B , Sumoilación , Replicación Viral , Trióxido de Arsénico/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Replicación Viral/efectos de los fármacos , Hepatitis B/virología , Hepatitis B/tratamiento farmacológico , Hepatitis B/metabolismo , Sumoilación/efectos de los fármacos , ADN Circular/genética , ADN Circular/metabolismo , Núcleo Celular/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Antivirales/farmacología , Proteínas del Núcleo Viral/metabolismo , Proteínas del Núcleo Viral/genética , Células Hep G2
16.
J Med Virol ; 96(3): e29546, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38516804

RESUMEN

Tapasin, a crucial molecular chaperone involved viral antigen processing and presentation, plays an important role in antivirus immunity. However, its impact on T cell differentiation in the context of virus clearance remains unclear. In this study, we employed induced pluripotent stem cells to differentiate into hepatocyte-like cell, which were subsequently inserted to the inverted colloidal crystal scaffolds, thus establishing a hepatocyte organoid (HO). By inoculating hepatitis B virus (HBV) particles in the system, we successfully engineered a robust in vitro HBV infection model for at least 3 weeks. Furthermore, we aimed to explore the effects of lentivirus-mediated short hairpin RNA (shRNA) targeting human Tapasin on the differentiation and antiviral function of CD8+ T cells. Specifically, we transfected dendritic cells (DCs) with Tapasin-shRNA and cocultured with T cells. The results demonstrated that Tapasin-shRNA transfected DCs effectively suppressed T cell proliferation and impeded HBV-specific cytotoxic T lymphocyte responses. Our investigation also revealed the role of mTOR pathway activation in reducing autophagy activity within CD8+ T cells. Expressions of autophagy-related proteins, beclin-1, LC3II/LC3I were decreased and PI3K/AKT/mTOR activity was increased in Tapasin-shRNA group. Collectively, our findings elucidate that shRNA targeting the Tapasin gene within DCs inhibits T cell differentiation by reducing autophagy activity to hamper viral clearance in the HBV-infected HO.


Asunto(s)
Células Dendríticas , Hepatitis B , Proteínas de Transporte de Membrana , Humanos , Autofagia/genética , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/metabolismo , Regulación hacia Abajo , Hepatitis B/metabolismo , Antígenos del Núcleo de la Hepatitis B/genética , Virus de la Hepatitis B , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Interferente Pequeño/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Organoides/metabolismo , Organoides/virología
17.
Virus Res ; 344: 199366, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38548137

RESUMEN

Gamma-aminobutyric acid (GABA) signals in various non-neuronal cells including hepatocytes and some immune cells. Studies, including ours, show that type A GABA receptors (GABAARs)-mediated signaling occurs in macrophages regulating tissue-specific functions. Our recent study reveals that activation of GABAARs in liver macrophages promotes their M2-like polarization and increases HBV replication in mice. This short article briefly summarizes the GABA signaling system in macrophages and discusses potential mechanisms by which GABA signaling promotes HBV replication.


Asunto(s)
Hepatitis B , Macrófagos , Receptores de GABA-A , Transducción de Señal , Replicación Viral , Ácido gamma-Aminobutírico , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Ácido gamma-Aminobutírico/metabolismo , Hepatitis B/virología , Hepatitis B/metabolismo , Virus de la Hepatitis B/fisiología , Virus de la Hepatitis B/genética , Hígado/virología , Hígado/metabolismo , Macrófagos/virología , Receptores de GABA-A/metabolismo , Receptores de GABA-A/genética
18.
Mol Cancer Res ; 22(7): 642-655, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38546386

RESUMEN

Hepatitis B viral (HBV) persistent infection plays a significant role in hepatocellular carcinoma (HCC) tumorigenesis. Many studies have revealed the pivotal roles of N6-methyladenosine (m6A) in multiple cancers, while the regulatory mechanism in stemness maintenance of HBV persistent infection-related HCC remains elusive. Here, we demonstrated that the level of m6A modification was downregulated by HBV in HBV-positive HCC, through enhanced stability of ALKBH5 mRNA. More specifically, we also identified that ALKBH5 mRNA was functionally required for the stemness maintenance and self-renewal in the HBV-positive HCC, but dispensable in HBV-negative HCC. Mechanistically, ALKBH5 demethylated the m6A modification in the 3' untranslated region of the oncogenic gene SNAI2 to prevent the recognition of YTHDF2 therewith stabilize SNAI2 transcripts, contributing to cancer stem cell traits in HBV-positive HCC. Moreover, the expression of SNAI2 reversed the suppression of stemness properties by knocking down ALKBH5. In addition, ALKBH5/SNAI2 axis accelerates tumor immune evasion through activated ligand of immune checkpoint CD155. Our study unveiled that the ALKBH5 induces m6A demethylation of the SNAI2 as a key regulator in HBV-related HCC, and identifies the function of ALKBH5/SNAI2/YTHDF2 axis in promoting the stem-like cells phenotype and immune escape during HBV infection. IMPLICATIONS: HBV promotes HCC stemness maintenance through elevate m6A modification of SNAI2 in an ALKBH5-YTHDF2-dependent manner and increases the expression of the ligand of immune checkpoint CD155.


Asunto(s)
Adenosina , Desmetilasa de ARN, Homólogo 5 de AlkB , Carcinoma Hepatocelular , Virus de la Hepatitis B , Neoplasias Hepáticas , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/virología , Ratones , Animales , Desmetilación , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción de la Familia Snail/genética , Escape del Tumor/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Masculino , Hepatitis B/virología , Hepatitis B/complicaciones , Hepatitis B/genética , Hepatitis B/metabolismo , Proteínas de Unión al ARN
19.
J Biol Chem ; 300(3): 105724, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38325742

RESUMEN

Mammalian cells have evolved strategies to regulate gene expression when oxygen is limited. Hypoxia-inducible factors (HIF) are the major transcriptional regulators of host gene expression. We previously reported that HIFs bind and activate hepatitis B virus (HBV) DNA transcription under low oxygen conditions; however, the global cellular response to low oxygen is mediated by a family of oxygenases that work in concert with HIFs. Recent studies have identified a role for chromatin modifiers in sensing cellular oxygen and orchestrating transcriptional responses, but their role in the HBV life cycle is as yet undefined. We demonstrated that histone lysine demethylase 4 (KDM4) can restrict HBV, and pharmacological or oxygen-mediated inhibition of the demethylase increases viral RNAs derived from both episomal and integrated copies of the viral genome. Sequencing studies demonstrated that KDM4 is a major regulator of the hepatic transcriptome, which defines hepatocellular permissivity to HBV infection. We propose a model where HBV exploits cellular oxygen sensors to replicate and persist in the liver. Understanding oxygen-dependent pathways that regulate HBV infection will facilitate the development of physiologically relevant cell-based models that support efficient HBV replication.


Asunto(s)
Virus de la Hepatitis B , Histona Demetilasas con Dominio de Jumonji , Oxígeno , Replicación Viral , Humanos , ADN Viral/genética , Genoma Viral/genética , Hepatitis B/enzimología , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Hígado/enzimología , Hígado/metabolismo , Hígado/virología , Oxígeno/metabolismo , Plásmidos/genética , Transcriptoma , Replicación Viral/genética
20.
Org Biomol Chem ; 22(11): 2218-2225, 2024 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-38358380

RESUMEN

Chronic infection with hepatitis B virus (HBV) is a major cause of cirrhosis and liver cancer. Capsid assembly modulators can induce error-prone assembly of HBV core proteins to prevent the formation of infectious virions, representing promising candidates for treating chronic HBV infections. To explore novel capsid assembly modulators from unexplored mirror-image libraries of natural products, we have investigated the synthetic process of the HBV core protein for preparing the mirror-image target protein. In this report, the chemical synthesis of full-length HBV core protein (Cp183) containing an arginine-rich nucleic acid-binding domain at the C-terminus is presented. Sequential ligations using four peptide segments enabled the synthesis of Cp183 via convergent and C-to-N direction approaches. After refolding under appropriate conditions, followed by the addition of nucleic acid, the synthetic Cp183 assembled into capsid-like particles.


Asunto(s)
Hepatitis B , Ácidos Nucleicos , Humanos , Cápside/química , Proteínas de la Cápside/metabolismo , Virus de la Hepatitis B , Hepatitis B/metabolismo , Proteínas del Núcleo Viral/análisis , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/metabolismo , Replicación Viral , Antivirales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA