Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 378
Filter
1.
Genes Dis ; 11(6): 101142, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39281837

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a leading cause of liver cirrhosis and hepatocellular carcinoma, representing a global health problem for which a functional cure is difficult to achieve. The HBV core protein (HBc) is essential for multiple steps in the viral life cycle. It is the building block of the nucleocapsid in which viral DNA reverse transcription occurs, and its mediation role in viral-host cell interactions is critical to HBV infection persistence. However, systematic studies targeting HBc-interacting proteins remain lacking. Here, we combined HBc with the APEX2 to systematically identify HBc-related host proteins in living cells. Using functional screening, we confirmed that proteasome activator subunit 1 (PSME1) is a potent HBV-associated host factor. PSME1 expression was up-regulated upon HBV infection, and the protein level of HBc decreased after PSME1 knockdown. Mechanistically, the interaction between PSME1 and HBc inhibited the degradation of HBc by the 26S proteasome, thereby improving the stability of the HBc protein. Furthermore, PSME1 silencing inhibits HBV transcription in the HBV infection system. Our findings reveal an important mechanism by which PSME1 regulates HBc proteins and may facilitate the development of new antiviral therapies targeting PSME1 function.

2.
Nat Commun ; 15(1): 6964, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138195

ABSTRACT

Though RNAi and RNA-splicing machineries are involved in regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, their precise roles in coronavirus disease 2019 (COVID-19) pathogenesis remain unclear. Herein, we show that decreased RNAi component (Dicer and XPO5) and splicing factor (SRSF3 and hnRNPA3) expression correlate with increased COVID-19 severity. SARS-CoV-2 N protein induces the autophagic degradation of Dicer, XPO5, SRSF3, and hnRNPA3, inhibiting miRNA biogenesis and RNA splicing and triggering DNA damage, proteotoxic stress, and pneumonia. Dicer, XPO5, SRSF3, and hnRNPA3 knockdown increases, while their overexpression decreases, N protein-induced pneumonia's severity. Older mice show lower expression of Dicer, XPO5, SRSF3, and hnRNPA3 in their lung tissues and exhibit more severe N protein-induced pneumonia than younger mice. PJ34, a poly(ADP-ribose) polymerase inhibitor, or anastrozole, an aromatase inhibitor, ameliorates N protein- or SARS-CoV-2-induced pneumonia by restoring Dicer, XPO5, SRSF3, and hnRNPA3 expression. These findings will aid in developing improved treatments for SARS-CoV-2-associated pneumonia.


Subject(s)
COVID-19 , Karyopherins , Ribonuclease III , SARS-CoV-2 , Serine-Arginine Splicing Factors , Animals , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Humans , Ribonuclease III/metabolism , Ribonuclease III/genetics , SARS-CoV-2/genetics , COVID-19/metabolism , COVID-19/virology , COVID-19/genetics , Mice , Karyopherins/metabolism , Karyopherins/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Down-Regulation , Lung/metabolism , Lung/pathology , Lung/virology , Male , Female , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Splicing , Autophagy/genetics , DNA Damage , Heterogeneous-Nuclear Ribonucleoprotein Group A-B
4.
J Hepatol ; 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38960374

ABSTRACT

BACKGROUND & AIMS: Sodium taurocholate cotransporting polypeptide (NTCP) has been identified as the cellular receptor for HBV. However, hepatocytes expressing NTCP exhibit varying susceptibilities to HBV infection. This study aimed to investigate whether other host factors modulate the process of HBV infection. METHODS: Liver biopsy samples obtained from children with hepatitis B were used for single-cell sequencing and susceptibility analysis. Primary human hepatocytes, HepG2-NTCP cells, and human liver chimeric mice were used to analyze the effect of candidate host factors on HBV infection. RESULTS: Single-cell sequencing and susceptibility analysis revealed a positive correlation between neuropilin-1 (NRP1) expression and HBV infection. In the HBV-infected cell model, NRP1 overexpression before HBV inoculation significantly enhanced viral attachment and internalization, and promoted viral infection in the presence of NTCP. Mechanistic studies indicated that NRP1 formed a complex with LHBs (large hepatitis B surface proteins) and NTCP. The NRP1 b domain mediated its interaction with conserved arginine residues at positions 88 and 92 in the preS1 domain of LHBs. This NRP1-preS1 interaction subsequently promoted the binding of preS1 to NTCP, facilitating viral infection. Moreover, disruption of the NRP1-preS1 interaction by the NRP1 antagonist EG00229 significantly attenuated the binding affinity between NTCP and preS1, thereby inhibiting HBV infection both in vitro and in vivo. CONCLUSIONS: Our findings indicate that NRP1 is a novel host factor for HBV infection, which interacts with preS1 and NTCP to modulate HBV entry into hepatocytes. IMPACT AND IMPLICATIONS: HBV infection is a global public health problem, but the understanding of the early infection process of HBV remains limited. Through single-cell sequencing, we identified a novel host factor, NRP1, which modulates HBV entry by interacting with HBV preS1 and NTCP. Moreover, antagonists targeting NRP1 can inhibit HBV infection both in vitro and in vivo. This study could further advance our comprehension of the early infection process of HBV.

5.
J Med Virol ; 96(7): e29805, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39011773

ABSTRACT

Heterogeneous nuclear protein U (HNRNPU) plays a pivotal role in innate immunity by facilitating chromatin opening to activate immune genes during host defense against viral infection. However, the mechanism by which HNRNPU is involved in Hepatitis B virus (HBV) transcription regulation through mediating antiviral immunity remains unknown. Our study revealed a significant decrease in HNRNPU levels during HBV transcription, which depends on HBx-DDB1-mediated degradation. Overexpression of HNRNPU suppressed HBV transcription, while its knockdown effectively promoted viral transcription, indicating HNRNPU as a novel host restriction factor for HBV transcription. Mechanistically, HNRNPU inhibits HBV transcription by activating innate immunity through primarily the positive regulation of the interferon-stimulating factor 2'-5'-oligoadenylate synthetase 3, which mediates an ribonuclease L-dependent mechanism to enhance innate immune responses. This study offers new insights into the host immune regulation of HBV transcription and proposes potential targets for therapeutic intervention against HBV infection.


Subject(s)
2',5'-Oligoadenylate Synthetase , Hepatitis B virus , Immunity, Innate , Transcription, Genetic , Humans , Hepatitis B virus/immunology , Hepatitis B virus/genetics , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Hep G2 Cells , Hepatitis B/immunology , Hepatitis B/virology , Hepatitis B/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/immunology , Trans-Activators
6.
Virol Sin ; 39(4): 655-666, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852920

ABSTRACT

The landscape of hepatitis B virus (HBV) integration in the plasma cell-free DNA (cfDNA) of HBV-infected patients with different stages of liver diseases [chronic hepatitis B (CHB), liver cirrhosis (LC), and hepatocellular carcinoma (HCC)] remains unclear. In this study, we developed an improved strategy for detecting HBV DNA integration in plasma cfDNA, based on DNA probe capture and next-generation sequencing. Using this optimized strategy, we successfully detected HBV integration events in chimeric artificial DNA samples and HBV-infected HepG2-NTCP cells at day one post infection, with high sensitivity and accuracy. The characteristics of HBV integration events in the HBV-infected HepG2-NTCP cells and plasma cfDNA from HBV-infected individuals (CHB, LC, and HCC) were further investigated. A total of 112 and 333 integration breakpoints were detected in the HepG2-NTCP cells and 22 out of 25 (88%) clinical HBV-infected samples, respectively. In vivo analysis showed that the normalized number of support unique sequences (nnsus) in HCC was significantly higher than in CHB or LC patients (P values â€‹< â€‹0.05). All integration breakpoints are randomly distributed on human chromosomes and are enriched in the HBV genome around nt 1800. The majority of integration breakpoints (61.86%) are located in the gene-coding region. Both non-homologous end-joining (NHEJ) and microhomology-mediated end-joining (MMEJ) interactions occurred during HBV integration across the three different stages of liver diseases. Our study provides evidence that HBV DNA integration can be detected in the plasma cfDNA of HBV-infected patients, including those with CHB, LC, or HCC, using this optimized strategy.


Subject(s)
Carcinoma, Hepatocellular , Cell-Free Nucleic Acids , DNA, Viral , Hepatitis B virus , Hepatitis B, Chronic , High-Throughput Nucleotide Sequencing , Liver Neoplasms , Virus Integration , Humans , Hepatitis B virus/genetics , Hepatitis B virus/isolation & purification , DNA, Viral/genetics , DNA, Viral/blood , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/isolation & purification , Hep G2 Cells , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/blood , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/blood , Liver Neoplasms/virology , Liver Neoplasms/blood , Liver Cirrhosis/virology , Liver Cirrhosis/blood , Liver Cirrhosis/diagnosis , Male , Middle Aged , Adult , Female
8.
STAR Protoc ; 5(2): 102998, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38573862

ABSTRACT

We recently identified a class of small cytosolic double-stranded DNA (scDNA) approximately 20-40 bp in size in human and mouse cells. Here, we present a protocol for scDNA isolation from cultured murine cells. We describe steps for cytosolic compartment separation, DNA isolation in the cytosolic fraction using phenol-chloroform extraction, and ethanol precipitation. We then detail procedures for denaturing purified cytosolic DNA through urea polyacrylamide gel electrophoresis and obtaining scDNA in the cytosolic DNA fraction via gel purification. For complete details on the use and execution of this protocol, please refer to Liu et al.1.


Subject(s)
Cytosol , DNA , Animals , Mice , DNA/isolation & purification , Cytosol/metabolism , Cytosol/chemistry , Cells, Cultured , Electrophoresis, Polyacrylamide Gel/methods
9.
JCI Insight ; 9(7)2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38587080

ABSTRACT

BACKGROUNDAs Omicron is prompted to replicate in the upper airway, neutralizing antibodies (NAbs) delivered through inhalation might inhibit early-stage infection in the respiratory tract. Thus, elucidating the prophylactic efficacy of NAbs via nasal spray addresses an important clinical need.METHODSThe applicable potential of a nasal spray cocktail containing 2 NAbs was characterized by testing its neutralizing potency, synergetic neutralizing mechanism, emergency protective and therapeutic efficacy in a hamster model, and pharmacokinetics/pharmacodynamic (PK/PD) in human nasal cavity.RESULTSThe 2 NAbs displayed broad neutralizing efficacy against Omicron, and they could structurally compensate each other in blocking the Spike-ACE2 interaction. When administrated through the intranasal mucosal route, this cocktail demonstrated profound efficacy in the emergency prevention in hamsters challenged with authentic Omicron BA.1. The investigator-initiated trial in healthy volunteers confirmed the safety and the PK/PD of the NAb cocktail delivered via nasal spray. Nasal samples from the participants receiving 4 administrations over a course of 16 hours demonstrated potent neutralization against Omicron BA.5 in an ex vivo pseudovirus neutralization assay.CONCLUSIONThese results demonstrate that the NAb cocktail nasal spray provides a good basis for clinical prophylactic efficacy against Omicron infections.TRIAL REGISTRATIONwww.chictr.org.cn, ChiCTR2200066525.FUNDINGThe National Science and Technology Major Project (2017ZX10202203), the National Key Research and Development Program of China (2018YFA0507100), Guangzhou National Laboratory (SRPG22-015), Lingang Laboratory (LG202101-01-07), Science and Technology Commission of Shanghai Municipality (YDZX20213100001556), and the Emergency Project from the Science & Technology Commission of Chongqing (cstc2021jscx-fyzxX0001).


Subject(s)
Antibodies, Neutralizing , Nasal Sprays , Animals , Cricetinae , Humans , China , Trachea , Healthy Volunteers
10.
Adv Sci (Weinh) ; 11(24): e2306810, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38647380

ABSTRACT

Persistent transcription of HBV covalently closed circular DNA (cccDNA) is critical for chronic HBV infection. Silencing cccDNA transcription through epigenetic mechanisms offers an effective strategy to control HBV. Long non-coding RNAs (lncRNAs), as important epigenetic regulators, have an unclear role in cccDNA transcription regulation. In this study, lncRNA sequencing (lncRNA seq) is conducted on five pairs of HBV-positive and HBV-negative liver tissue. Through analysis, HOXA-AS2 (HOXA cluster antisense RNA 2) is identified as a significantly upregulated lncRNA in HBV-infected livers. Further experiments demonstrate that HBV DNA polymerase (DNA pol) induces HOXA-AS2 after establishing persistent high-level HBV replication. Functional studies reveal that HOXA-AS2 physically binds to cccDNA and significantly inhibits its transcription. Mechanistically, HOXA-AS2 recruits the MTA1-HDAC1/2 deacetylase complex to cccDNA minichromosome by physically interacting with metastasis associated 1 (MTA1) subunit, resulting in reduced acetylation of histone H3 at lysine 9 (H3K9ac) and lysine 27 (H3K27ac) associated with cccDNA and subsequently suppressing cccDNA transcription. Altogether, the study reveals a mechanism to self-limit HBV replication, wherein the upregulation of lncRNA HOXA-AS2, induced by HBV DNA pol, can epigenetically suppress cccDNA transcription.


Subject(s)
DNA, Circular , Epigenesis, Genetic , Hepatitis B virus , RNA, Long Noncoding , Repressor Proteins , Trans-Activators , Humans , Hepatitis B virus/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Epigenesis, Genetic/genetics , DNA, Circular/genetics , DNA, Circular/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Transcription, Genetic/genetics , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology
11.
J Med Virol ; 96(2): e29439, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38294104

ABSTRACT

Hepatitis B virus (HBV) infection is a serious global health problem. After the viruses infect the human body, the host can respond to the virus infection by coordinating various cellular responses, in which mitochondria play an important role. Evidence has shown that mitochondrial proteins are involved in host antiviral responses. In this study, we found that the overexpression of TIM22 and TIM29, the members of the inner membrane translocase TIM22 complex, significantly reduced the level of intracellular HBV DNA and RNA and secreted HBV surface antigens and E antigen. The effects of TIM22 and TIM29 on HBV replication and transcription is attributed to the reduction of core promoter activity mediated by the increased expression of SRSF1 which acts as a suppressor of HBV replication. This study provides new evidence for the critical role of mitochondria in the resistance of HBV infection and new targets for the development of treatment against HBV infection.


Subject(s)
Hepatitis B virus , Hepatitis B , Mitochondrial Precursor Protein Import Complex Proteins , Serine-Arginine Splicing Factors , Humans , Hepatitis B e Antigens/genetics , Hepatitis B e Antigens/metabolism , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/physiology , Serine-Arginine Splicing Factors/metabolism , Virus Replication , Mitochondrial Precursor Protein Import Complex Proteins/metabolism
12.
J Exp Clin Cancer Res ; 43(1): 35, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38287371

ABSTRACT

BACKGROUND: Hepatocellular Carcinoma (HCC) is a matter of great global public health importance; however, its current therapeutic effectiveness is deemed inadequate, and the range of therapeutic targets is limited. The aim of this study was to identify early growth response 1 (EGR1) as a transcription factor target in HCC and to explore its role and assess the potential of gene therapy utilizing EGR1 for the management of HCC. METHODS: In this study, both in vitro and in vivo assays were employed to examine the impact of EGR1 on the growth of HCC. The mouse HCC model and human organoid assay were utilized to assess the potential of EGR1 as a gene therapy for HCC. Additionally, the molecular mechanism underlying the regulation of gene expression and the suppression of HCC growth by EGR1 was investigated. RESULTS: The results of our investigation revealed a notable decrease in the expression of EGR1 in HCC. The decrease in EGR1 expression promoted the multiplication of HCC cells and the growth of xenografted tumors. On the other hand, the excessive expression of EGR1 hindered the proliferation of HCC cells and repressed the development of xenografted tumors. Furthermore, the efficacy of EGR1 gene therapy was validated using in vivo mouse HCC models and in vitro human hepatoma organoid models, thereby providing additional substantiation for the anti-cancer role of EGR1 in HCC. The mechanistic analysis demonstrated that EGR1 interacted with the promoter region of phosphofructokinase-1, liver type (PFKL), leading to the repression of PFKL gene expression and consequent inhibition of PFKL-mediated aerobic glycolysis. Moreover, the sensitivity of HCC cells and xenografted tumors to sorafenib was found to be increased by EGR1. CONCLUSION: Our findings suggest that EGR1 possesses therapeutic potential as a tumor suppressor gene in HCC, and that EGR1 gene therapy may offer benefits for HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Regulation, Neoplastic , Glycolysis , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Liver Neoplasms/metabolism , Sorafenib/pharmacology
13.
Adv Sci (Weinh) ; 11(5): e2305374, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38059827

ABSTRACT

Solute carrier family 27 member 5, a key enzyme in fatty acid transport and bile acid metabolism in the liver, is frequently expressed in low quantities in patients with hepatocellular carcinoma, resulting in poor prognosis. However, it is unclear whether SLC27A5 plays non-canonical functions and regulates HCC progression. Here, an unexpected non-canonical role of SLC27A5 is reported: regulating the alternative splicing of mRNA to inhibit the metastasis of HCC independently of its metabolic enzyme activity. Mechanistically, SLC27A5 interacts with IGF2BP3 to prevent its translocation into the nucleus, thereby inhibiting its binding to target mRNA and modulating PIP4K2A pre-mRNA splicing. Loss of SLC27A5 results in elevated levels of the PIP4K2A-S isoform, thus positively regulating phosphoinositide 3-kinase signaling via enhanced p85 stability in HCC. SLC27A5 restoration by AAV-Slc27a5 or IGF2BP3 RNA decoy oligonucleotides exerts an inhibitory effect on HCC metastasis with reduced expression of the PIP4K2A-S isoform. Therefore, PIP4K2A-S may be a novel target for treating HCC with SLC27A5 deficiency.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Phosphotransferases (Alcohol Group Acceptor) , RNA Splicing , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Fatty Acid Transport Proteins , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Isoforms/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Adv Sci (Weinh) ; 11(2): e2304408, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37957540

ABSTRACT

Although the dysregulation of bile acid (BA) composition has been associated with fibrosis progression, its precise roles in liver fibrosis is poorly understood. This study demonstrates that solute carrier family 27 member 5 (SLC27A5), an enzyme involved in BAs metabolism, is substantially downregulated in the liver tissues of patients with cirrhosis and fibrosis mouse models. The downregulation of SLC27A5 depends on RUNX family transcription factor 2 (RUNX2), which serves as a transcriptional repressor. The findings reveal that experimental SLC27A5 knockout (Slc27a5-/- ) mice display spontaneous liver fibrosis after 24 months. The loss of SLC27A5 aggravates liver fibrosis induced by carbon tetrachloride (CCI4 ) and thioacetamide (TAA). Mechanistically, SLC27A5 deficiency results in the accumulation of unconjugated BA, particularly cholic acid (CA), in the liver. This accumulation leads to the activation of hepatic stellate cells (HSCs) by upregulated expression of early growth response protein 3 (EGR3). The re-expression of hepatic SLC27A5 by an adeno-associated virus or the reduction of CA levels in the liver using A4250, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, ameliorates liver fibrosis in Slc27a5-/- mice. In conclusion, SLC27A5 deficiency in mice drives hepatic fibrosis through CA-induced activation of HSCs, highlighting its significant implications for liver fibrosis treatment.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Animals , Humans , Mice , Bile Acids and Salts , Cholic Acid/adverse effects , Cholic Acid/metabolism , Disease Models, Animal , Fatty Acid Transport Proteins/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/pathology
15.
Biochim Biophys Acta Mol Cell Res ; 1871(2): 119642, 2024 02.
Article in English | MEDLINE | ID: mdl-37996058

ABSTRACT

Liver cancer is ranked as the sixth most prevalent from of malignancy globally and stands as the third primary contributor to cancer-related mortality. Metastasis is the main reason for liver cancer treatment failure and patient deaths. Speckle-type POZ protein (SPOP) serves as a crucial substrate junction protein within the cullin-RING E3 ligase complex, acting as a significant tumor suppressor in liver cancer. Nevertheless, the precise molecular mechanism underlying the role of SPOP in liver cancer metastasis remain elusive. In the current study, we identified cAMP response element binding 5 (CREB5) as a novel SPOP substrate in liver cancer. SPOP facilitates non-degradative K63-polyubiquitination of CREB5 on K432 site, consequently hindering its capacity to activate receptor tyrosine kinase MET. Moreover, liver cancer-associated SPOP mutant S119N disrupts the SPOP-CREB5 interactions and impairs the ubiquitination of CREB5.This disruption ultimately leads to the activation of the MET signaling pathway and enhances metastatic properties of hepatoma cells both in vitro and in vivo. In conclusion, our findings highlight the functional significance of the SPOP-CREB5-MET axis in liver cancer metastasis.


Subject(s)
Liver Neoplasms , Humans , Ubiquitination , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Cell Nucleus , Cell Line , Signal Transduction , Nuclear Proteins/genetics , Repressor Proteins/genetics , Cyclic AMP Response Element-Binding Protein A
16.
Virol Sin ; 39(1): 9-23, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38110037

ABSTRACT

The achievement of a functional cure for chronic hepatitis B (CHB) remains limited to a minority of patients treated with currently approved drugs. The primary objective in developing new anti-HBV drugs is to enhance the functional cure rates for CHB. A critical prerequisite for the functional cure of CHB is a substantial reduction, or even eradication of covalently closed circular DNA (cccDNA). Within this context, the changes in cccDNA levels during treatment become as a pivotal concern. We have previously analyzed the factors influencing cccDNA dynamics and introduced a preliminary classification of hepatitis B treatment strategies based on these dynamics. In this review, we employ a systems thinking perspective to elucidate the fundamental aspects of the HBV replication cycle and to rationalize the classification of treatment strategies according to their impact on the dynamic equilibrium of cccDNA. Building upon this foundation, we categorize current anti-HBV strategies into two distinct groups and advocate for their combined use to significantly reduce cccDNA levels within a well-defined timeframe.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Humans , Hepatitis B, Chronic/drug therapy , Hepatitis B virus/genetics , DNA, Circular/genetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA, Viral/genetics , Virus Replication/genetics
17.
Infect Drug Resist ; 16: 7413-7425, 2023.
Article in English | MEDLINE | ID: mdl-38077299

ABSTRACT

Purpose: The objective of this study was to investigate the prevalence and molecular characteristics of Vibrio parahaemolyticus isolates from fecal samples of patients in Nantong, China. Methods: From 2018 to 2021, a total of 106 clinical cases and samples of V. parahaemolyticus infection were collected. The virulence genes, serotypes and antibiotic resistance of these isolates were analyzed. Additionally, pulsed-field gel electrophoresis (PFGE) was used to analyze the homogeneity of the isolates. Results: Outbreaks of V. parahaemolyticus infection were concentrated in the summer, with seafood consumption being the primary contributing factor, followed by meat and meat products. tlh+tdh+trh- was confirmed as the most frequently detected virulence genotype among the clinical isolates. 16 serotypes were identified, and O3:K6 was the dominant serotype in Nantong. The antimicrobial susceptibility testing revealed the highest resistance rate to cefazolin (99.1%, 104/106), followed by ampicillin (64.2%, 68/106) and tetracycline (29.2%, 31/106). Fourteen resistant phenotypes were identified, with ampicillin-cefazolin being the most prevalent. The multiple antibiotic resistance (MAR) index ranged from 0.07 to 0.36. PFGE typing clustered isolates with similarity greater than 85% into ten genetic clusters (A-J). Conclusion: Clinical isolates generally exhibited pathogenicity and drug resistance, with some isolates displaying high homology. Clusters C, E, and G were the predominant circulating clusters in this area, posing a potential risk of recurrent outbreaks, which demanded our vigilance.

18.
Autophagy ; : 1-3, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38013411

ABSTRACT

The CGAS (cyclic GMP-AMP synthase)-STING1 (stimulator of interferon response cGAMP interactor 1) pathway is an important innate immune pathway that induces proinflammatory cytokine production following stimulation with dsDNA > 45 bp. We recently identified a class of ~ 20-40 bp small cytosolic dsDNA (scDNA) that blocks CGAS-STING1 activation. In this punctum, we discuss the mechanism underlying the inhibition of CGAS-STING1 activation via scDNA. scDNA binds to CGAS but cannot activate its enzymatic activity. It competes with dsDNA > 45 bp for binding with CGAS to inhibit CGAS-STING1 activation. Moreover, scDNA activates macroautophagy/autophagy and induces the autophagic degradation of STING1 and long dsDNA. Autophagy then increases scDNA levels, driving a feedback loop that accelerates the degradation of STING1 and long cytosolic dsDNA. These findings reveal that mutual communication between scDNA and autophagy inhibits CGAS-STING1 activation following stimulation with dsDNA > 45 bp.

19.
J Med Virol ; 95(12): e29254, 2023 12.
Article in English | MEDLINE | ID: mdl-38018242

ABSTRACT

Hepatitis B virus (HBV) infection remains a significant public health burden worldwide. The persistence of covalently closed circular DNA (cccDNA) within the nucleus of infected hepatocytes is responsible for the failure of antiviral treatments. The ubiquitin proteasome system (UPS) has emerged as a promising antiviral target, as it can regulate HBV replication by promoting critical protein degradation in steps of viral life cycle. Speckle-type POZ protein (SPOP) is a critical adaptor for Cul3-RBX1 E3 ubiquitin ligase complex, but the effect of SPOP on HBV replication is less known. Here, we identified SPOP as a novel host antiviral factor against HBV infection. SPOP overexpression significantly inhibited the transcriptional activity of HBV cccDNA without affecting cccDNA level in HBV-infected HepG2-NTCP and primary human hepatocyte cells. Mechanism studies showed that SPOP interacted with hepatocyte nuclear factor 1α (HNF1α), and induced HNF1α degradation through host UPS pathway. Moreover, the antiviral role of SPOP was also confirmed in vivo. Together, our findings reveal that SPOP is a novel host factor which inhibits HBV transcription and replication by ubiquitination and degradation of HNF1α, providing a potential therapeutic strategy for the treatment of HBV infection.


Subject(s)
Hepatitis B virus , Hepatitis B , Humans , Antiviral Agents/pharmacology , DNA, Circular , DNA, Viral/genetics , Hepatitis B/genetics , Hepatitis B virus/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Ubiquitination , Virus Replication
20.
J Med Virol ; 95(10): e29189, 2023 10.
Article in English | MEDLINE | ID: mdl-37855689

ABSTRACT

Infectious diseases remain a major global issue in public health. It is important to develop rapid, sensitive, and accurate diagnostic methods to detect pathogens and their mutations. Cas12f1 is an exceptionally compact RNA-guided nuclease and have the potential to fulfill the clinical needs. Based on the interaction between crRNA-SSDNA binary sequence and Cas12f1, here, we addressed the essential features that determine the recognition ability of CRISPR-Cas12f1 single-nucleotide polymorphism (SNP), such as the length of spacer region and the base pairing region that determines the trans-cleavage of ssDNA. A fine-tuning spacer design strategy is also proposed to enhance the SNP recognition capability of CRISPR-Cas12f1. The optimized spacer confers the Cas12f1 system a strong SNP identification capability for viral or bacterial drug-resistance mutations, with a specificity ratio ranging from 19.63 to 110.20 and an admirable sensitivity up to 100  copy/µL. Together, the spacer screening and CRISPR-Cas12f1 based SNP identification method, is sensitive and versatile, and will have a wide application prospect in pathogen DNA mutation diagnosis and other mutation profiling.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Polymorphism, Single Nucleotide , Humans , RNA/genetics , DNA, Single-Stranded/genetics , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL