Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Acad Radiol ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38556431

ABSTRACT

RATIONALE AND OBJECTIVES: The role of Programmed death-ligand 1 (PD-L1) expression is crucial in guiding immunotherapy selection. This study aims to develop and evaluate a radiomic model, leveraging Computed Tomography (CT) imaging, with the objective of predicting PD-L1 expression status in patients afflicted with bladder cancer. MATERIALS AND METHODS: The study encompassed 183 subjects diagnosed with histologically confirmed bladder cancer, among which the PD-L1(+) cohort constituted 60.1% of the total population. Stratified random sampling was utilized at a 7:3 ratio. We employed five diverse machine learning algorithms-Decision Tree, Random Forest, Linear Support Vector Classification, Support Vector Machine, and Logistic Regression-to establish radiomic models on the training dataset. These models endeavored to predict PD-L1 expression status premised on radiomic features derived from region-of-interest segmentation. Subsequent to this, the predictive performance of these models was examined on a validation set employing the receiver operating characteristic (ROC) curve. The DeLong test was utilized to contrast ROC curves, thereby pinpointing the model with superior predictive accuracy. RESULTS: 16 features were chosen for the model construction. All five models revealed strong performance in the training set (AUC, 0.920-1) and commendable predictive ability in the validation set (AUC, 0.753-0.766). As per the DeLong test, no statistically significant disparities were observed among any of the models (P > 0.05) in the validation set. Additional verification through the calibration curve and decision curve analysis indicated that the Logistic Regression model exhibited extraordinary precision and practicality. CONCLUSION: Our machine learning model, grounded on radiomic features, demonstrated its proficiency in accurately distinguishing bladder cancer patients with high PD-L1 expression. Future research, incorporating more exhaustive datasets, could potentially augment the predictive efficiency of radiomic algorithms, thereby advancing their clinical utility.

3.
J Med Virol ; 95(7): e28969, 2023 07.
Article in English | MEDLINE | ID: mdl-37485644

ABSTRACT

Despite the extensive use of effective vaccines and antiviral drugs, chronic hepatitis B virus (HBV) infection continues to pose a serious threat to global public health. Therapies with novel mechanisms of action against HBV are being explored for achieving a functional cure. In this study, five murine models of HBV replication were used to investigate the inhibitory effect of RNA binding motif protein 24 (RBM24) on HBV replication. The findings revealed that RBM24 serves as a host restriction factor and suppresses HBV replication in vivo. The transient overexpression of RBM24 in hydrodynamics-based mouse models of HBV replication driven by the CMV or HBV promoters suppressed HBV replication. Additionally, the ectopic expression of RBM24 decreased viral accumulation and the levels of HBV covalently closed circular DNA (cccDNA) in an rcccDNA mouse model. The liver-directed transduction of adeno-associated viruses (AAV)-RBM24 mediated the stable hepatic expression of RBM24 in pAAV-HBV1.2 and HBV/tg mouse models, and markedly reduced the levels of HBV cccDNA and other viral indicators. Altogether, these findings revealed that RBM24 inhibits the replication of HBV in vivo, and RBM24 may be a potential therapeutic target for combating HBV infections.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Mice , Animals , Hepatitis B virus , Virus Replication , DNA, Circular , RNA-Binding Motifs , DNA, Viral/genetics , DNA, Viral/metabolism
4.
Oncol Lett ; 26(1): 305, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37323818

ABSTRACT

This retrospective clinical study described the treatment efficacy and safety of stereotactic body radiotherapy (SBRT) for patients of hepatocellular carcinoma (HCC) and liver metastasis tumors. The therapeutic effect and prognosis of patients with liver cancer treated with stereotactic body radiation therapy (SBRT) at the Fudan University Shanghai Cancer Center (Shanghai, China) between July 2011 and December 2020 were retrospectively analyzed. Overall survival (OS), local control (LC) rates and progression-free survival (PFS) were evaluated using Kaplan-Meier analysis and the log-rank test. Local progression was defined as tumor growth after SBRT on dynamic computed tomography follow-up. Treatment-related toxicities were assessed according to the Common Terminology Criteria for Adverse Events version 4. A total of 36 patients with liver cancer were enrolled in the present study. The prescribed dosages (14 Gy in 3 fractions or 16 Gy in 3 fractions) were applied for SBRT treatments. The median follow-up time was 21.4 months. The median OS time was 20.4 [95% confidence interval (CI): 6.6-34.2] months, and the 2-year OS rates for the total population, HCC group and liver metastasis group were 47.5, 73.3 and 34.2%, respectively. The median PFS time was 17.3 (95% CI: 11.8-22.8) months and the 2-year PFS rates for the total population, HCC group and liver metastasis group were 36.3, 44.0 and 31.4%, respectively. The 2-year LC rates for the total population, HCC group and liver metastasis group were 83.4, 85.7 and 81.6%, respectively. The most common grade IV toxicity for the HCC group was liver function impairment (15.4%), followed by thrombocytopenia (7.7%). There were no grade III/IV radiation pneumonia or digestive discomfort. The present study aimed to explore a safe, effective and non-invasive treatment method for liver tumors. At the same time, the innovation of the present study is to find a safe and effective prescription dose of SBRT in the absence of consensus on guidelines.

5.
J Virol ; 97(5): e0058023, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37166302

ABSTRACT

Hepatitis B virus (HBV) infection affects hepatic metabolism. Serum metabolomics studies have suggested that HBV possibly hijacks the glycerol-3-phosphate (G3P) shuttle. In this study, the two glycerol-3-phosphate dehydrogenases (GPD1 and GPD2) in the G3P shuttle were analyzed for determining their role in HBV replication and the findings revealed that GPD2 and not GPD1 inhibited HBV replication. The knockdown of GPD2 expression upregulated HBV replication, while GPD2 overexpression reduced HBV replication. Moreover, the overexpression of GPD2 significantly reduced HBV replication in hydrodynamic injection-based mouse models. Mechanistically, this inhibitory effect is related to the GPD2-mediated degradation of HBx protein by recruiting the E3 ubiquitin ligase TRIM28 and not to the alterations in G3P metabolism. In conclusion, this study revealed GPD2, a key enzyme in the G3P shuttle, as a host restriction factor in HBV replication. IMPORTANCE The glycerol-3-phosphate (G3P) shuttle is important for the delivery of cytosolic reducing equivalents into mitochondria for oxidative phosphorylation. The study analyzed two key components of the G3P shuttle and identified GPD2 as a restriction factor in HBV replication. The findings revealed a novel mechanism of GPD2-mediated inhibition of HBV replication via the recruitment of TRIM28 for degrading HBx, and the HBx-GPD2 interaction could be another potential therapeutic target for anti-HBV drug development.


Subject(s)
Glycerolphosphate Dehydrogenase , Hepatitis B , Tripartite Motif-Containing Protein 28 , Viral Regulatory and Accessory Proteins , Animals , Mice , Glycerol/metabolism , Glycerolphosphate Dehydrogenase/metabolism , Hepatitis B/metabolism , Hepatitis B virus/physiology , Mitochondria/enzymology , Phosphates/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication
6.
Cancer Med ; 12(6): 6477-6487, 2023 03.
Article in English | MEDLINE | ID: mdl-37012831

ABSTRACT

PURPOSE: This single-center retrospective clinical study aimed to evaluate the efficacy and feasibility of chemoradiotherapy with paclitaxel liposome plus cisplatin for locally advanced esophageal squamous cell carcinoma (ESCC). METHODS: Patients with locally advanced ESCC treated with paclitaxel-liposome-based chemoradiotherapy between 2016 and 2019 were retrospectively analyzed. Overall survival (OS) and progression-free survival (PFS) were evaluated using Kaplan-Meier analysis. RESULTS: Thirty-nine patients with locally advanced ESCC were included in this study. The median follow-up time was 31.5 months. The median OS time was 38.3 (95% confidence interval [CI]: 32.1-45.1) months, and the 1-, 2-, and 3-year OS rates were 84.6%, 64.1%, and 56.2%, respectively. The median PFS time was 32.1 (95% CI: 25.4-39.0) months, and the 1-, 2-, and 3-year PFS rates were 71.8%, 43.6%, and 43.6%, respectively. The most common Grade IV toxicity was neutropenia (30.8%) followed by lymphopenia (20.5%). There were no cases of Grade III/IV radiation pneumonia, and four patients (10.3%) had Grade III/IV esophagitis. CONCLUSION: Chemoradiotherapy using paclitaxel liposome and cisplatin is a well-tolerated and effective treatment regimen for locally advanced ESCC.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/drug therapy , Cisplatin/therapeutic use , Esophageal Neoplasms/drug therapy , Retrospective Studies , Liposomes , Disease-Free Survival , Paclitaxel , Chemoradiotherapy/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects
7.
J Med Virol ; 95(3): e28610, 2023 03.
Article in English | MEDLINE | ID: mdl-36840407

ABSTRACT

Tick-borne encephalitis virus (TBEV) is the causative agent of a potentially fatal neurological infection in humans. Investigating virus-host interaction is important for understanding the pathogenesis of TBEV and developing effective antiviral drugs against this virus. Here, we report that mammalian ste20-like kinase 3 (MST3) is involved in the regulation of TBEV infection. The knockdown or knockout of MST3, but not other mammalian ste20-like kinase family members, inhibited TBEV replication. The knockdown of MST3 also significantly reduced TBEV replication in mouse primary astrocytes. Life cycle analysis indicated that MST3 remarkably impaired virion assembly efficiency and specific infectivity by respectively 59% and 95% in MST3-knockout cells. We further found that MST3 interacts with the viral proteins NS2A and prM; and MST3 enhances the interaction of NS2A-NS4A. Thus, MST3-NS2A complex plays a major role in recruiting prM-E heterodimers and NS4A and mediates the virion assembly. Additionally, we found that MST3 was biotinylated and combined with other proteins (e.g., ATG5, Sec24A, and SNX4) that are associated with the cellular membrane required for TBEV infection. Overall, our study revealed a novel function for MST3 in TBEV infection and identified as a novel host factor supporting TBEV assembly.


Subject(s)
Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Animals , Mice , Humans , Encephalitis Viruses, Tick-Borne/genetics , Viral Proteins/metabolism , Mammals/metabolism , Vesicular Transport Proteins
8.
Antiviral Res ; 209: 105478, 2023 01.
Article in English | MEDLINE | ID: mdl-36464077

ABSTRACT

SARS-CoV-2 is a betacoronavirus with single-stranded positive-sense RNA, which is a serious global threat to human health. Understanding the molecular mechanism of viral replication is crucial for the development of antiviral drugs. The synthesis of viral polyproteins is a crucial step in viral progression. The synthesis of viral polyproteins in coronaviruses is regulated by the 5'-untranslated region (UTR); however, the detailed regulatory mechanism needs further investigation. The present study demonstrated that the RNA binding protein, RBM24, interacts with the RNA genome of SARS-CoV-2 via its RNA recognition submotifs (RNPs). The findings revealed that RBM24 recognizes and binds to the GUGUG element at stem-loop 4 (SL4) in the 5'-UTR of SARS-CoV-2. The interaction between RBM24 and 5'-UTR prevents 80S ribosome assembly, which in turn inhibits polyproteins translation and the replication of SARS-CoV-2. Notably, other RNA viruses, including SARS-CoV, MERS-CoV, Ebolavirus, rhinovirus, West Nile virus, Zika virus, Japanese encephalitis virus, yellow fever virus, hepatitis C virus, and human immunodeficiency virus-1 also contain one or several G(U/C/A)GUG sequences in the 5'-UTR, which is also targeted by RBM24. In conclusion, the present study demonstrated that RBM24 functions by interacting with the 5'-UTR of SARS-CoV-2 RNA, and elucidated that RBM24 could be a host restriction factor for SARS-CoV-2 and other RNA viruses.


Subject(s)
COVID-19 , RNA Viruses , Zika Virus Infection , Zika Virus , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Viral/metabolism , 5' Untranslated Regions , Virus Replication/genetics , Zika Virus/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Nat Commun ; 13(1): 7629, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494373

ABSTRACT

The ongoing COVID-19 pandemic has demonstrated that viral diseases represent an enormous public health and economic threat to mankind and that individuals with compromised immune systems are at greater risk of complications and death from viral diseases. The development of broad-spectrum antivirals is an important part of pandemic preparedness. Here, we have engineer a series of designer cells which we term autonomous, intelligent, virus-inducible immune-like (ALICE) cells as sense-and-destroy antiviral system. After developing a destabilized STING-based sensor to detect viruses from seven different genera, we have used a synthetic signal transduction system to link viral detection to the expression of multiple antiviral effector molecules, including antiviral cytokines, a CRISPR-Cas9 module for viral degradation and the secretion of a neutralizing antibody. We perform a proof-of-concept study using multiple iterations of our ALICE system in vitro, followed by in vivo functionality testing in mice. We show that dual output ALICESaCas9+Ab system delivered by an AAV-vector inhibited viral infection in herpetic simplex keratitis (HSK) mouse model. Our work demonstrates that viral detection and antiviral countermeasures can be paired for intelligent sense-and-destroy applications as a flexible and innovative method against virus infection.


Subject(s)
COVID-19 , Virus Diseases , Viruses , Humans , Mice , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication , Pandemics
10.
J Virol ; 96(13): e0058522, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35862693

ABSTRACT

The biogenesis of covalently closed circular DNA (cccDNA) from relaxed circular DNA (rcDNA) is essential for chronic hepatitis B virus (HBV) infection. Different host DNA repair proteins are involved in the conversion of rcDNA to cccDNA. Here, we reported that the DNA repair factor poly(ADP-ribose) polymerase 1 (PARP1) is engaged in HBV cccDNA formation. PARP1 depletion remarkably impaired HBV replication and cccDNA synthesis. Inhibition of PARP1 poly (ADP-ribosylation) activity by olaparib suppressed cccDNA synthesis both in vitro and in vivo. Specifically, the early stage of cccDNA reservoir establishment was more sensitive to olaparib, suggesting that PARP1 participated in de novo cccDNA formation. Furthermore, PARP1 was activated by recognizing the rcDNA-like lesions directly and combined with other DNA repair proteins. The results presented proposed that the DNA damage-sensing protein PARP1 and poly(ADP-ribosylation) modification play a key role in cccDNA formation, which might be the target for developing the anti-HBV drug. IMPORTANCE The biogenesis and eradication of HBV cccDNA have been a research priority in recent years. In this study, we identified the DNA repair factor PARP1 as a host factor required for the HBV de novo cccDNA formation. HBV infection caused PARylation through PARP1 in Huh7-NTCP cells, primary human hepatocytes, and human-liver chimeric mice. We found that PARP1 could directly bind to the rcDNA lesions and was activated, PARylating other DNA repair proteins. We address the importance of PARP1-mediated PARylation in HBV cccDNA formation, which is a potential therapeutic target for chronic hepatitis B.


Subject(s)
DNA, Circular , Hepatitis B , Poly (ADP-Ribose) Polymerase-1 , Animals , DNA Repair , DNA, Circular/genetics , DNA, Circular/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Hepatitis B/virology , Hepatitis B virus/genetics , Humans , Mice , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Proviruses/genetics
11.
Environ Sci Pollut Res Int ; 29(45): 68422-68431, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35543791

ABSTRACT

Cadmium (Cd) is a poisonous element for human health. This study was conducted to explore whether H2S can alleviate the toxic effects of Cd on ginger. Specifically, ginger plants were grown in soil and treated with 7.5 mg·l-1 CdCl2, after which water (T1), 0.8 mM NaHS (T2), or 0.8 mM NaHS and 0.15 mM HT (T3) were added to the soil. The application of NaHS increased the activities of antioxidant enzymes (APX, GR, MDHAR, and DHAR) during the early treatment stage. It also inhibited the decrease in Pn, Gs, and Ls under Cd stress conditions while also limiting the increase in Ci. An analysis of the expression of Cd uptake-related genes indicated that NaHS upregulated the expression of ZoNramp1, which encodes a metal transporter, in roots as well as ZoPCS1, which encodes a phytochelatin synthase. In contrast, NaHS downregulated ZoHMA2 expression in the rhizomes and roots under Cd stress conditions.


Subject(s)
Hydrogen Sulfide , Zingiber officinale , Antioxidants/metabolism , Cadmium/metabolism , Cadmium/toxicity , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Soil , Sulfides , Water
12.
J Cancer ; 13(3): 965-974, 2022.
Article in English | MEDLINE | ID: mdl-35154462

ABSTRACT

Purpose: This study aimed to develop and validate a recurrence prediction of glioma patients through a radiomics feature training and validation model. Patients and methods: In this study, the prediction model was developed in a training cohort that consisted of 88 patients from January 2014 to July 2017 with pathologically confirmed gliomas. Their pre-radiotherapy and recurrence brain magnetic resonance imaging (MRI) images were collected, and the radiomics features were extracted. Clinical factors including age, gender, WHO grade, Isocitrate dehydrogenases (IDH) mutation status and treatment after surgery were collected. The least absolute shrinkage and selection operator (LASSO) regression model was conducted for data dimension reduction, feature selection, and radiomics feature analysis. Internal validation was assessed. An independent validation cohort contained 41 consecutive patients from August 2017 to December 2018. Furthermore, multivariable logistic regression analysis was used to develop the predicting model by combining the radiomics signature and independent clinical factors. Results: In total, 129 patients were included, among which 40 patients had recurrence. The median follow-up time was 27.4 (range, 2.6-79.2) months. We compared the tumor regions radiomics difference between the recurrence and non-recurrence patients. The radiomics signature was associated with the event of recurrence (P < 0.001 for both training and validation cohorts, respectively). The training model showed good discrimination with a C-index of 0.7578 (95%CI: 0.6549-9.8608) through internal validation on T1 contrast-enhanced magnetic resonance imaging, and a consistent trend in calibration. In the validation cohort, the model also showed good discrimination (C-index, 0.6925, 95%CI: 0.5145-0.8705) and good calibration. In the other two sequences of MRI (T1WI, T2WI), the validation model also showed positive results. Meanwhile, radiomics feature and clinical factors were significantly prognostic for recurrence (P value <0.05, respectively). Conclusion: We identified the radiomics feature derived from brain MRI that presented potential in predicting recurrence in glioma patients. This could be beneficial to risk stratification for patients. Further investigation is necessary to include expanded sample size investigation and external multicenter validation.

13.
Antiviral Res ; 198: 105249, 2022 02.
Article in English | MEDLINE | ID: mdl-35041910

ABSTRACT

The binding of HBV polymerase (Pol) and the epsilon stem loop (ε) on the 5' terminal region of pgRNA is required for pgRNA packaging and HBV replication. Previous research has demonstrated that RNA binding motif protein 24 (RBM24) is involved in pgRNA packaging by mediating the interaction between HBV polymerase (Pol) and the ε element. Here, we demonstrate that RBM38 interacts with ε, pol, RBM24 and HBV core which mediate pgRNA packaging. RBM38 directly binds to the lower bulge of ε via RNA recognition submotifs (RNPs) and interacts with HBV Pol in an RNA-independent manner. RBM38 interacts with RBM24 and forms heterogeneous oligomers, which mediate Pol-ε binding and the formation of the Pol-RBM38/RBM24-ε complex. More important, RBM38 also binds to the HBV core via the C-terminal region (ARD domain), which facilitates the combination of Pol-ε with the HBV core protein. In conclusion, RBM38 facilitates the Pol-ε interaction and mediates Pol-ε in combining with the HBV core, triggering pgRNA packaging for reverse transcription and DNA synthesis. This study provides new insights into pgRNA encapsidation.


Subject(s)
Hepatitis B virus , RNA, Viral , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Nucleocapsid/metabolism , RNA , RNA, Viral/metabolism , RNA-Binding Motifs , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
14.
Protein Cell ; 12(4): 261-278, 2021 04.
Article in English | MEDLINE | ID: mdl-32772249

ABSTRACT

TANK-binding kinase 1 (TBK1), a core kinase of antiviral pathways, activates the production of interferons (IFNs). It has been reported that deacetylation activates TBK1; however, the precise mechanism still remains to be uncovered. We show here that during the early stage of viral infection, the acetylation of TBK1 was increased, and the acetylation of TBK1 at Lys241 enhanced the recruitment of IRF3 to TBK1. HDAC3 directly deacetylated TBK1 at Lys241 and Lys692, which resulted in the activation of TBK1. Deacetylation at Lys241 and Lys692 was critical for the kinase activity and dimerization of TBK1 respectively. Using knockout cell lines and transgenic mice, we confirmed that a HDAC3 null mutant exhibited enhanced susceptibility to viral challenge via impaired production of type I IFNs. Furthermore, activated TBK1 phosphorylated HDAC3, which promoted the deacetylation activity of HDAC3 and formed a feedback loop. In this study, we illustrated the roles the acetylated and deacetylated forms of TBK1 play in antiviral innate responses and clarified the post-translational modulations involved in the interaction between TBK1 and HDAC3.


Subject(s)
Histone Deacetylases/immunology , Protein Serine-Threonine Kinases/immunology , Virus Diseases/immunology , Animals , Chlorocebus aethiops , HEK293 Cells , Histone Deacetylases/genetics , Humans , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , RAW 264.7 Cells , THP-1 Cells , Vero Cells , Virus Diseases/genetics
15.
Virol Sin ; 36(3): 501-509, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33165771

ABSTRACT

Hepatitis B virus (HBV) belongs to Hepadnaviridae family and mainly infects hepatocytes, which can cause acute or chronic hepatitis. Currently, two types of antiviral drugs are approved for chronic infection clinically: interferons and nucleos(t)ide analogues. However, the clinical cure for chronic infection is still rare, and it is a huge challenge for all researchers to develop high-efficiency, safe, non-tolerant, and low-toxicity anti-HBV drugs. Antazoline hydrochloride is a first-generation antihistamine with anticholinergic properties, and it is commonly used to relieve nasal congestion and in eye drops. Recently, an in vitro high-throughput evaluation system was constructed to screen nearly 800 compounds from the Food and Drug Administration (FDA)-approved Drug Library. We found that arbidol hydrochloride and antazoline hydrochloride can effectively reduce HBV DNA in the extracellular supernatant in a dose-dependent manner, with EC50 of 4.321 µmol/L and 2.910 µmol/L in HepAD38 cells, respectively. Moreover, the antiviral effects and potential mechanism of action of antazoline hydrochloride were studied in different HBV replication systems. The results indicate that antazoline hydrochloride also has a significant inhibitory effect on HBV DNA in the extracellular supernatant of Huh7 cells, with an EC50 of 2.349 µmol/L. These findings provide new ideas for screening and research related to HBV agents.


Subject(s)
Antazoline , Drug Repositioning , Hepatitis B , Antazoline/pharmacology , Antazoline/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA , DNA, Viral/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/drug therapy , Humans , Virus Replication/drug effects
16.
Antiviral Res ; 172: 104619, 2019 12.
Article in English | MEDLINE | ID: mdl-31600533

ABSTRACT

Hepatitis B virus (HBV) infection remains an important public health problem worldwide. Covalently closed circular DNA (cccDNA) exhibits as an individual minichromosome and is the molecular basis of HBV infection persistence and antiviral treatment failure. In the current study, we demonstrated that histone deacetylase 11 (HDAC11) inhibits HBV transcription and replication in HBV-transfected Huh7 cells. By using an HBV in vitro infection system, HDAC11 was found to affect the transcriptional activity of cccDNA but did not affect cccDNA production. Chromatin immunoprecipitation (ChIP) assays were utilized to analyze the epigenetic modifications of cccDNA. The results show that HDAC11 specifically reduced the acetylation level of cccDNA-bound histone H3 but did not affect that of histone H4. Furthermore, HDAC11 overexpression decreased the levels of cccDNA-bound acetylated H3K9 (H3K9ac) and H3K27 (H3K27ac). In conclusion, HDAC11 restricts HBV replication through epigenetic repression of cccDNA transcription. These findings reveal the novel role of HDAC11 in HBV infection, further broadening our knowledge regarding the functions of HDAC11 and the roles of HDACs in the epigenetic regulation of HBV cccDNA.


Subject(s)
Epigenetic Repression , Hepatitis B virus/genetics , Histone Deacetylases/metabolism , Virus Replication/genetics , Cell Line , DNA, Circular/metabolism , DNA, Viral/metabolism , Epigenesis, Genetic , Hepatitis B/metabolism , Hepatitis B/virology , Histones/metabolism , Humans , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...