Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(30): e2201927119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858426

RESUMEN

Hepatitis B virus (HBV) DNA replication takes place inside the viral core particle and is dependent on autophagy. Here we show that HBV core particles are associated with autophagosomes and phagophores in cells that productively replicate HBV. These autophagic membrane-associated core particles contain almost entirely the hypophosphorylated core protein and are DNA replication competent. As the hyperphosphorylated core protein can be localized to phagophores and the dephosphorylation of the core protein is associated with the packaging of viral pregenomic RNA (pgRNA), these results are in support of the model that phagophores can serve as the sites for the packaging of pgRNA. In contrast, in cells that replicate HBV, the precore protein derivatives, which are related to the core protein, are associated with autophagosomes but not with phagophores via a pathway that is independent of its signal peptide. Interestingly, when the core protein is expressed by itself, it is associated with phagophores but not with autophagosomes. These observations indicate that autophagic membranes are differentially involved in the trafficking of precore and core proteins. HBV induces the fusion of autophagosomes and multivesicular bodies and the silencing of Rab11, a regulator of this fusion, is associated with the reduction of release of mature HBV particles. Our studies thus indicate that autophagic membranes participate in the assembly of HBV nucleocapsids, the trafficking of HBV precore and core proteins, and likely also the egress of HBV particles.


Asunto(s)
Autofagosomas , Virus de la Hepatitis B , Nucleocápside , Empaquetamiento del Genoma Viral , Replicación Viral , Autofagosomas/fisiología , ADN Viral/metabolismo , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Nucleocápside/genética , Nucleocápside/fisiología , Transporte de Proteínas , ARN Viral/metabolismo , Replicación Viral/genética
2.
PLoS Pathog ; 17(3): e1009480, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33784371

RESUMEN

Dengue virus (DENV) causes dengue fever and severe hemorrhagic fever in humans and is primarily transmitted by Aedes aegypti and A. albopictus mosquitoes. The incidence of DENV infection has been gradually increasing in recent years due to global urbanization and international travel. Understanding the virulence determinants in host and vector transmissibility of emerging epidemic DENV will be critical to combat potential outbreaks. The DENV serotype 2 (DENV-2), which caused a widespread outbreak in Taiwan in 2015 (TW2015), is of the Cosmopolitan genotype and is phylogenetically related to the virus strain linked to another large outbreak in Indonesia in 2015. We found that the TW2015 virus was highly virulent in type I and type II interferon-deficient mice, with robust replication in spleen, lung, and intestine. The TW2015 virus also had high transmissibility to Aedes mosquitoes and could be effectively spread in a continuous mosquitoes-mouse-mosquitoes-mouse transmission cycle. By making 16681-based mutants carrying different segments of the TW2015 virus, we identified the structural pre-membrane (prM) and envelope (E) genes as key virulence determinants in the host, with involvement in the high transmissibility of the TW2015 virus in mosquitoes. The transmission mouse model will make a useful platform for evaluation of DENV with high epidemic potential and development of new strategies against dengue outbreaks.


Asunto(s)
Culicidae/virología , Virus del Dengue/genética , Virus del Dengue/patogenicidad , Dengue/virología , Insectos Vectores/virología , Virulencia/fisiología , Animales , Modelos Animales de Enfermedad , Genotipo , Ratones
3.
Recent Results Cancer Res ; 217: 47-70, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33200361

RESUMEN

Hepatitis B virus (HBV) is a major cause of hepatocellular carcinoma (HCC). There are approximately 250 million people in the world that are chronically infected by this virus, resulting in nearly 1 million deaths every year. Many of these patients die from severe liver diseases, including HCC. HBV may induce HCC through the induction of chronic liver inflammation, which can cause oxidative stress and DNA damage. However, many studies also indicated that HBV could induce HCC via the alteration of hepatocellular physiology that may involve genetic and epigenetic changes of the host DNA, the alteration of cellular signaling pathways, and the inhibition of DNA repair mechanisms. This alteration of cellular physiology can lead to the accumulation of DNA damages and the promotion of cell cycles and predispose hepatocytes to oncogenic transformation.


Asunto(s)
Carcinoma Hepatocelular , Virus de la Hepatitis B , Hepatitis B , Neoplasias Hepáticas , Carcinoma Hepatocelular/virología , Reparación del ADN , Hepatitis B/complicaciones , Hepatitis B/genética , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Humanos , Neoplasias Hepáticas/virología , Oncogenes
4.
Int J Mol Sci ; 22(8)2021 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-33921710

RESUMEN

MicroRNAs (miRNAs) play versatile roles in multiple biological processes. However, little is known about miRNA's involvement in flavivirus persistent infection. Here, we used an miRNA array analysis of Japanese encephalitis virus (JEV)-infected cells to search for persistent infection-associated miRNAs in comparison to acute infection. Among all differentially expressed miRNAs, the miR-125b-5p is the most significantly increased one. The high level of miR-125b-5p in persistently JEV-infected cells was confirmed by Northern analysis and real-time quantitative polymerase chain reaction. As soon as the cells established a persistent infection, a significantly high expression of miR-125b-5p was readily observed. Transfecting excess quantities of a miR-125b-5p mimic into acutely infected cells reduced genome replication and virus titers. Host targets of miR125b-5p were analyzed by target prediction algorithms, and six candidates were confirmed by a dual-luciferase reporter assay. These genes were upregulated in the acutely infected cells and sharply declined in the persistently infected cells. The transfection of the miR125b-5p mimic reduced the expression levels of Stat3, Map2k7, and Triap1. Our studies indicated that miR-125b-5p targets both viral and host sequences, suggesting its role in coordinating viral replication and host antiviral responses. This is the first report to characterize the potential roles of miR-125b-5p in persistent JEV infections.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Encefalitis Japonesa/genética , MicroARNs/metabolismo , Animales , Línea Celular , Cricetinae , Virus de la Encefalitis Japonesa (Especie)/genética , Humanos , MicroARNs/genética , Replicación Viral/genética , Replicación Viral/fisiología
5.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728260

RESUMEN

Retinoid (vitamin A) is an essential diet constituent that governs a broad range of biological processes. Its biologically active metabolite, all-trans retinoic acid (ATRA), exhibits a potent antiviral property by enhancing both innate and adaptive antiviral immunity against a variety of viral pathogens, such as, but not limited to, HIV, respiratory syncytial virus (RSV), herpes simplex virus (HSV), and measles. Even though the hepatocyte is highly enriched with retinoid and its metabolite ATRA, it supports the establishment of efficient hepatitis C virus (HCV) replication. Here, we demonstrate the hepatocyte-specific cell-intrinsic mechanism by which ATRA exerts either a proviral or antiviral effect, depending on how it engages cellular retinoic acid binding proteins (CRABPs). We found that the engagement of CRABP1 by ATRA potently supported viral infection by promoting the accumulation of lipid droplets (LDs), which robustly enhanced the formation of a replication complex on the LD-associated endoplasmic reticulum (ER) membrane. In contrast, ATRA binding to CRABP2 potently inhibited HCV via suppression of LD accumulation. However, this antiviral effect of CRABP2 was abrogated due to the functional and quantitative predominance of CRABP1 in the hepatocytes. In summary, our study demonstrates that CRABPs serve as an on-off switch that modulates the efficiency of the HCV life cycle and elucidates how HCV evades the antiviral properties of ATRA via the exploitation of CRABP1 functionality.IMPORTANCE ATRA, a biologically active metabolite of vitamin A, exerts pleiotropic biological effects, including the activation of both innate and adaptive immunity, thereby serving as a potent antimicrobial compound against numerous viral pathogens. Despite the enrichment of hepatocytes with vitamin A, HCV still establishes an efficient viral life cycle. Here, we discovered that the hepatocellular response to ATRA creates either a proviral or an antiviral environment depending on its engagement with CRABP1 or -2, respectively. CRABP1 supports the robust replication of HCV, while CRABP2 potently inhibits the efficiency of viral replication. Our biochemical, genetic, and microscopic analyses reveal that the pro- and antiviral effects of CRABPs are mediated by modulation of LD abundance, where HCV establishes the platform for viral replication and assembly on the LD-associated ER membrane. This study uncovered a cell-intrinsic mechanism by which HCV exploits the proviral function of CRABP1 to establish an efficient viral life cycle.


Asunto(s)
Hepacivirus/metabolismo , Hepatitis C/metabolismo , Gotas Lipídicas/metabolismo , Receptores de Ácido Retinoico/metabolismo , Antivirales/farmacología , Línea Celular , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Hepatitis C/patología , Humanos , Gotas Lipídicas/virología , Tretinoina/farmacología
6.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28381578

RESUMEN

The NS2A protein of dengue virus (DENV) has eight predicted transmembrane segments (pTMS1 to -8) and participates in RNA replication, virion assembly, and host antiviral response. However, the roles of specific amino acid residues within the pTMS regions of NS2A during the viral life cycle are not clear. Here, we explore the function of DENV NS2A by introducing a series of alanine substitutions into the N-terminal half (pTMS1 to -4) of the protein in the context of a DENV infectious clone or subgenomic replicon. Six NS2A mutants (NM5, -7, -9, and -17 to -19) around pTMS1 and -2 displayed a novel phenotype showing a >1,000-fold reduction in virus yield, an absence of plaque formation despite wild-type-like replicon activity, and infectious-virus-like particle yields. HEK-293 cells infected with the six NS2A mutant viruses failed to cause a virus-induced cytopathic effect (CPE) by MitoCapture staining, cell proliferation, and lactate dehydrogenase release assays. Sequencing analyses of pseudorevertant viruses derived from lethal-mutant viruses revealed two consensus reversion mutations, leucine to phenylalanine at codon 181 (L181F) within pTMS7 of NS2A and isoleucine to threonine at codon 114 (I114T) within NS2B. The introduction of an NS2A-L181F mutation into the lethal (NM15, -16, -25, and -33) and CPE-defective (NM7, -9, and -19) mutants substantially rescued virus infectivity and virus-induced CPE, respectively, whereas the NS2B-L114T mutation rescued the NM16, -25, and -33 mutants. In conclusion, the results revealed the essential roles of the N-terminal half of NS2A in RNA replication and virus-induced CPE. Intramolecular interactions between pTMSs of NS2A and intermolecular interactions between the NS2A and NS2B proteins were also implicated.IMPORTANCE The characterization of the N-terminal (current study) and C-terminal halves of DENV NS2A is the most comprehensive mutagenesis study to date to investigate the function of NS2A during the flaviviral life cycle. A novel region responsible for virus-induced cytopathic effect (CPE) within pTMS1 and -2 of DENV NS2A was identified. Revertant genetics studies implied unexpected relationships between various pTMSs of DENV NS2A and NS2B. These results provide comprehensive information regarding the functions of DENV NS2A and the specific amino acids and transmembrane segments responsible for these functions. The positions and properties of the rescuing mutations were also revealed, providing important clues regarding the manner in which intramolecular or intermolecular interactions between the pTMSs of NS2A and NS2B regulate virus replication, assembly/secretion, and virus-induced CPE. These results expand the understanding of flavivirus replication. The knowledge may also facilitate studies of pathogenesis and novel vaccine and antiflaviviral drug development.


Asunto(s)
Efecto Citopatogénico Viral , Virus del Dengue/genética , Mutagénesis , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Alanina/metabolismo , Sustitución de Aminoácidos , Proliferación Celular/genética , Virus del Dengue/química , Virus del Dengue/fisiología , Células HEK293 , Humanos , L-Lactato Deshidrogenasa/metabolismo , Leucina/genética , Mutación , Fenilalanina/genética , ARN Viral/metabolismo , Análisis de Secuencia , Proteínas no Estructurales Virales/química , Ensamble de Virus , Replicación Viral/genética
7.
J Virol ; 89(22): 11406-19, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26339052

RESUMEN

UNLABELLED: The risk of liver cancer in patients infected with the hepatitis B virus (HBV) and their clinical response to interferon alpha therapy vary based on the HBV genotype. The mechanisms underlying these differences in HBV pathogenesis remain unclear. In HepG2 cells transfected with a mutant HBV(G2335A) expression plasmid that does not transcribe the 2.2-kb doubly spliced RNA (2.2DS-RNA) expressed by wild-type HBV genotype A, the level of HBV pregenomic RNA (pgRNA) was higher than that in cells transfected with an HBV genotype A expression plasmid. By using cotransfection with HBV genotype D and 2.2DS-RNA expression plasmids, we found that a reduction of pgRNA was observed in the cells even in the presence of small amounts of the 2.2DS-RNA plasmid. Moreover, ectopic expression of 2.2DS-RNA in the HBV-producing cell line 1.3ES2 reduced the expression of pgRNA. Further analysis showed that exogenously transcribed 2.2DS-RNA inhibited a reconstituted transcription in vitro. In Huh7 cells ectopically expressing 2.2DS-RNA, RNA immunoprecipitation revealed that 2.2DS-RNA interacted with the TATA-binding protein (TBP) and that nucleotides 432 to 832 of 2.2DS-RNA were required for efficient TBP binding. Immunofluorescence experiments showed that 2.2DS-RNA colocalized with cytoplasmic TBP and the stress granule components, G3BP and poly(A)-binding protein 1 (PABP1), in Huh7 cells. In conclusion, our study reveals that 2.2DS-RNA acts as a repressor of HBV transcription through an interaction with TBP that induces stress granule formation. The expression of 2.2DS-RNA may be one of the viral factors involved in viral replication, which may underlie differences in clinical outcomes of liver disease and responses to interferon alpha therapy between patients infected with different HBV genotypes. IMPORTANCE: Patients infected with certain genotypes of HBV have a lower risk of hepatocellular carcinoma and exhibit a more favorable response to antiviral therapy than patients infected with other HBV genotypes. Using cultured human hepatoma cells as a model of HBV infection, we found that the expression of 2.2DS-RNA caused a decrease in HBV replication. In cultured cells, the ectopic expression of 2.2DS-RNA obviously reduced the intracellular levels of HBV mRNAs. Our analysis of the 2.2DS-RNA-mediated suppression of viral RNA expression showed that 2.2DS-RNA inhibited transcription via binding to the TATA-binding protein and stress granule proteins. Our findings suggest that the 2.2DS-RNA acts as a suppressive noncoding RNA that modulates HBV replication, which may in turn influence the development of chronic hepatitis B.


Asunto(s)
Virus de la Hepatitis B/genética , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína de Unión a TATA-Box/metabolismo , Replicación Viral/genética , Células 3T3 , Animales , Sitios de Unión/genética , Carcinoma Hepatocelular/epidemiología , Carcinoma Hepatocelular/virología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , ADN Helicasas , Regulación Viral de la Expresión Génica , Células HEK293 , Células Hep G2 , Antígenos de Superficie de la Hepatitis B/metabolismo , Antígenos e de la Hepatitis B/metabolismo , Hepatitis B Crónica/virología , Humanos , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/virología , Ratones , Plásmidos/genética , Proteína I de Unión a Poli(A)/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , ARN Helicasas , Precursores del ARN/biosíntesis , Proteínas con Motivos de Reconocimiento de ARN , Empalme del ARN/genética , ARN Bicatenario/biosíntesis , ARN Viral/genética , Transcripción Genética/genética
8.
EBioMedicine ; 94: 104723, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37487418

RESUMEN

BACKGROUND: Dengue virus outbreaks are increasing in number and severity worldwide. Viral transmission is assumed to require a minimum time period of viral replication within the mosquito midgut. It is unknown if alternative transmission periods not requiring replication are possible. METHODS: We used a mouse model of dengue virus transmission to investigate the potential of mechanical transmission of dengue virus. We investigated minimal viral titres necessary for development of symptoms in bitten mice and used resulting parameters to inform a new model of dengue virus transmission within a susceptible population. FINDINGS: Naïve mice bitten by mosquitoes immediately after they took partial blood meals from dengue infected mice showed symptoms of dengue virus, followed by mortality. Incorporation of mechanical transmission into mathematical models of dengue virus transmission suggest that this supplemental transmission route could result in larger outbreaks which peak sooner. INTERPRETATION: The potential of dengue transmission routes independent of midgut viral replication has implications for vector control strategies that target mosquito lifespan and suggest the possibility of similar mechanical transmission routes in other disease-carrying mosquitoes. FUNDING: This study was funded by grants from the National Health Research Institutes, Taiwan (04D2-MMMOST02), the Human Frontier Science Program (RGP0033/2021), the National Institutes of Health (1R01AI143698-01A1, R01AI151004 and DP2AI152071) and the Ministry of Science and Technology, Taiwan (MOST104-2321-B-400-016).


Asunto(s)
Aedes , Virus del Dengue , Dengue , Humanos , Animales , Ratones , Dengue/epidemiología , Brotes de Enfermedades , Mosquitos Vectores
9.
Virulence ; 13(1): 258-296, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35100095

RESUMEN

Hepatitis B virus (HBV) is a hepatotropic virus and an important human pathogen. There are an estimated 296 million people in the world that are chronically infected by this virus, and many of them will develop severe liver diseases including hepatitis, cirrhosis and hepatocellular carcinoma (HCC). HBV is a small DNA virus that replicates via the reverse transcription pathway. In this review, we summarize the molecular pathways that govern the replication of HBV and its interactions with host cells. We also discuss viral and non-viral factors that are associated with HBV-induced carcinogenesis and pathogenesis, as well as the role of host immune responses in HBV persistence and liver pathogenesis.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Virus de la Hepatitis B/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Virulencia
10.
J Biomed Sci ; 18: 96, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22208719

RESUMEN

BACKGROUND: The covalently closed-circular DNA (cccDNA) of hepatitis B virus (HBV) is associated with viral persistence in HBV-infected hepatocytes. However, the regulation of cccDNA and its transcription in the host cells at different growth stages is not well understood. METHODS: We took advantages of a stably HBV-producing cell line, 1.3ES2, and examine the dynamic changes of HBV cccDNA, viral transcripts, and viral replication intermediates in different cellular growth stages. RESULTS: In this study, we showed that cccDNA increased suddenly in the initial proliferation phase of cell growth, probably attributable to its nuclear replenishment by intracellular nucleocapsids. The amount of cccDNA then decreased dramatically in the cells during their exponential proliferation similar to the loss of extrachromosomal plasmid DNA during cell division, after which it accumulated gradually while the host cells grew to confluency. We found that cccDNA was reduced in dividing cells and could be removed when proliferating cells were subjected to long term of lamivudine (3TC) treatment. The amounts of viral replicative intermediates were rapidly reduced in these proliferating cells and were significantly increased after cells reaching confluency. The expression levels of viral transcripts were increased in parallel with the elevated expression of hepatic transcription factors (HNF4α, CEBPα, PPARα, etc.) during cell growth confluency. The HBV transcripts were transcribed from both integrated viral genome and cccDNA, however the transcriptional abilities of cccDNA was less efficient then that from integrated viral genome in all cell growth stages. We also noted increases in the accumulation of intracellular viral particles and the secretion of mature virions as the cells reached confluency and ceased to grow. CONCLUSIONS: Based on the dynamics of HBV replication, we propose that HBV replication is modulated differently in the different stages of cell growth, and can be divided into three phases (initial proliferation phase, exponential proliferation phase and growth confluency phase) according to the cell growth curve. The regulation of cccDNA in different cell growth phase and its importance regarding HBV replication are discussed.


Asunto(s)
ADN Circular/metabolismo , ADN Viral/metabolismo , Virus de la Hepatitis B/genética , Transcripción Genética , Proliferación Celular , Replicación del ADN , Virus de la Hepatitis B/metabolismo , Plásmidos , Replicación Viral/genética
11.
Curr Opin Virol ; 51: 158-163, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34717215

RESUMEN

Hepatitis B virus (HBV) e antigen (HBeAg) was discovered in the sera of HBV patients nearly 50 years ago. It is not essential for HBV to infect or replicate in hepatocytes. Earlier clinical studies suggested that this antigen might play an important role for HBV to establish persistence in babies after its mother-to-child transmission. Subsequent clinical studies also suggested that HBeAg might have immunomodulatory activities. In recent years, a large body of information on how HBeAg might modulate host immunity was published. In this review, we summarize recent research progresses on the immunomodulatory activities of HBeAg and discuss how these activities of HBeAg may promote HBV persistence.


Asunto(s)
Antígenos e de la Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/virología , Inmunomodulación , Hepatitis B Crónica/inmunología , Humanos , Transmisión Vertical de Enfermedad Infecciosa
12.
Trends Microbiol ; 26(1): 33-42, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28823759

RESUMEN

Hepatitis B virus (HBV) chronically infects 250 million people worldwide, resulting in nearly one million deaths annually. Studies in recent years have significantly improved our knowledge on the mechanisms of HBV persistence. HBV uses multiple pathways to harness host innate immunity to enhance its replication. It can also take advantage of the developing immune system and the not-yet-stabilized gut microbiota of young children to facilitate its persistence, and use maternal viral e antigen to educate immunity of the offspring to support its persistence after vertical transmission. The knowledge gained from these recent studies paves the way for the development of new therapies for the treatment of chronic HBV infection, which has so far been very challenging.


Asunto(s)
Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/fisiología , Inmunidad Innata , Factores de Edad , Niño , Microbioma Gastrointestinal/inmunología , Antígenos e de la Hepatitis B , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/terapia , Interacciones Huésped-Patógeno/inmunología , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Estadios del Ciclo de Vida , Herencia Materna/inmunología , Microbiota , Carga Viral
13.
PLoS Negl Trop Dis ; 12(4): e0006417, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29668683

RESUMEN

Zika virus (ZIKV) is primarily transmitted by Aedes mosquitoes in the subgenus Stegomyia but can also be transmitted sexually and vertically in humans. STAT1 is an important downstream factor that mediates type I and II interferon signaling. In the current study, we showed that mice with STAT1 knockout (Stat1-/-) were highly susceptible to ZIKV infection. As low as 5 plaque-forming units of ZIKV could cause viremia and death in Stat1-/- mice. ZIKV replication was initially detected in the spleen but subsequently spread to the brain with concomitant reduction of the virus in the spleen in the infected mice. Furthermore, ZIKV could be transmitted from mosquitoes to Stat1-/- mice back to mosquitoes and then to naïve Stat1-/- mice. The 50% mosquito infectious dose of viremic Stat1-/- mouse blood was close to 810 focus-forming units (ffu)/ml. Our further studies indicated that the activation of macrophages and conventional dendritic cells were likely critical for the resolution of ZIKV infection. The newly developed mouse and mosquito transmission models for ZIKV infection will be useful for the evaluation of antiviral drugs targeting the virus, vector, and host.


Asunto(s)
Aedes/virología , Modelos Animales de Enfermedad , Mosquitos Vectores/virología , Factor de Transcripción STAT1/genética , Infección por el Virus Zika/transmisión , Virus Zika/fisiología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Viremia , Infección por el Virus Zika/virología
14.
Virus Res ; 124(1-2): 139-50, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17134784

RESUMEN

Defective interfering (DI) RNAs are deletion mutants of viral genomes that are known in many cases to contribute to persistent infection and modification of viral pathogenesis. Cell type also plays a critical role in the establishment of viral persistence. In this study we have identified for the first time the generation of DI RNAs of Japanese encephalitis virus in C6/36 mosquito cells. A persistent infection was established by replacing growth medium on surviving cells and continued cell passaging. Persistent infection was demonstrated by a continual release of infectious virus, fluorescent antibody staining, and Northern analysis. A population of DI RNAs of approximately 8.2-9.7 kb, not detectable in acutely infected cells, became apparent in the persistently infected cells by 25 days postinfection. Sequence analyses revealed a population of DI RNAs that contained in-frame deletions of 1.3-2.8 kb covering the region of the E gene and some flanking C or prM and NS1 gene sequences. Transcripts from one cDNA clone of a DI RNA replicated in uninfected mosquito cells as demonstrated by RT-PCR. DI RNA-containing virions in supernatant fluids from persistently infected mosquito cells could be used to establish persistent infection in BHK-21 cells. The correlation of DI RNA presence with cell survival suggests that DI RNAs are contributing mechanistically to the establishment of persistent infection in both the mosquito and mammalian cells.


Asunto(s)
Culicidae/virología , Virus Defectuosos/genética , Virus de la Encefalitis Japonesa (Especie)/fisiología , ARN Viral/biosíntesis , ARN Viral/genética , Animales , Northern Blotting , Línea Celular , Supervivencia Celular , Células , Cricetinae , Culicidae/genética , Dípteros , Virus de la Encefalitis Japonesa (Especie)/genética , Inmunohistoquímica , Infecciones , Glicoproteínas de Membrana/genética , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Eliminación de Secuencia , Proteínas del Envoltorio Viral/genética , Proteínas no Estructurales Virales/genética
15.
PLoS One ; 10(6): e0131743, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121644

RESUMEN

Hepatitis B virus reactivation is an important medical issue in cancer patients who undergo systemic chemotherapy. Up to half of CHB carriers receiving chemotherapy develop hepatitis and among these cases a notable proportion are associated with HBV reactivation. However, the molecular mechanism(s) through which various chemotherapeutic agents induce HBV reactivation is not yet fully understood. In this study, we investigated the role of the cell cycle regulator p21 (Waf1/Cip1) in the modulation of HBV replication when a common chemotherapeutic agent, doxorubicin, is present. We showed that p21 expression was increased by doxorubicin treatment. This elevation in p21 expression enhanced the expression of CCAAT/enhancer-binding protein α (C/EBPα); such an increase is likely to promote the binding of C/EBPα to the HBV promoter, which will contribute to the activation of HBV replication. Our current study thus reveals the mechanism underlying doxorubicin modulation of HBV replication and provides an increased understanding of HBV reactivation in CHB patients who are receiving systemic chemotherapy.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Proteína alfa Potenciadora de Unión a CCAAT/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Doxorrubicina/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , ARN Viral , Elementos de Respuesta
16.
PLoS One ; 10(3): e0119625, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25785443

RESUMEN

Hepatitis B virus (HBV) is the smallest DNA virus and the major cause of acute and chronic hepatitis. The 3.2 kb HBV viral genome generates four major species of unspliced viral transcript as well as several alternatively spliced RNAs. A 2.2 kb singly-spliced RNA is the most abundant spliced RNA and is widely expressed among all HBV genotypes. The expression of the singly-spliced RNA, as well as that of its encoded protein HBSP, is strongly associated with hepatopathology during HBV infection. Here, we report a novel inhibitory role of a p21.5 protein, which is encoded by a 2.2 kb singly-spliced RNA, in the modulation of HBV replication. We show that overexpression of the singly-spliced RNA is able to efficiently inhibit HBV replication. Furthermore, a mutation in the ATG start codon of the precore region completely abolishes the inhibitory effect of the singly-spliced RNA, indicating that a viral protein (p21.5) derived from the singly-spliced RNA is the mediator of the inhibition. Furthermore, p21.5 is able to form a homodimer that interacts with core dimers forming hybrid viral assembly components. Sucrose gradient fractionation revealed that co-expression of p21.5 resulted in a spread distribution pattern of core proteins ranging from low to high sucrose densities. When compared with p22, p21.5 is almost ten times more efficient at destabilizing HBV nucleocapsid assembly in Huh7 cells overexpressing either p21.5 or p22 protein. Moreover, in vivo expression of p21.5 protein by tail vein injection was found to decrease the amount of nucleocapsid in the livers of HBV-expressing BALB/c mice. In conclusion, our study reveals that the HBV 2.2 kb singly-spliced RNA encodes a 21.5 kDa viral protein that significantly interferes with the assembly of nucleocapsids during HBV nucleocapsid formation. These findings provide a possible strategy for elimination of HBV particles inside cells.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Nucleocápside/antagonistas & inhibidores , Empalme del ARN , Proteínas Virales/genética , Animales , Línea Celular Tumoral , Dimerización , Genotipo , Células HEK293 , Células Hep G2 , Hepatitis B/patología , Hepatitis B/virología , Antígenos de Superficie de la Hepatitis B/sangre , Virus de la Hepatitis B/metabolismo , Hepatocitos/patología , Hepatocitos/virología , Humanos , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Nucleocápside/biosíntesis , Nucleocápside/genética , Nucleocápside/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Replicación Viral/genética
17.
PLoS One ; 7(1): e30360, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22276183

RESUMEN

Several studies have demonstrated that cytokine-mediated noncytopathic suppression of hepatitis B virus (HBV) replication may provide an alternative therapeutic strategy for the treatment of chronic hepatitis B infection. In our previous study, we showed that transforming growth factor-beta1 (TGF-ß1) could effectively suppress HBV replication at physiological concentrations. Here, we provide more evidence that TGF-ß1 specifically diminishes HBV core promoter activity, which subsequently results in a reduction in the level of viral pregenomic RNA (pgRNA), core protein (HBc), nucleocapsid, and consequently suppresses HBV replication. The hepatocyte nuclear factor 4alpha (HNF-4α) binding element(s) within the HBV core promoter region was characterized to be responsive for the inhibitory effect of TGF-ß1 on HBV regulation. Furthermore, we found that TGF-ß1 treatment significantly repressed HNF-4α expression at both mRNA and protein levels. We demonstrated that RNAi-mediated depletion of HNF-4α was sufficient to reduce HBc synthesis as TGF-ß1 did. Prevention of HNF-4α degradation by treating with proteasome inhibitor MG132 also prevented the inhibitory effect of TGF-ß1. Finally, we confirmed that HBV replication could be rescued by ectopic expression of HNF-4α in TGF-ß1-treated cells. Our data clarify the mechanism by which TGF-ß1 suppresses HBV replication, primarily through modulating the expression of HNF-4α gene.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Replicación Viral/efectos de los fármacos , Northern Blotting , Western Blotting , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Ensayo de Cambio de Movilidad Electroforética , Células Hep G2 , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Humanos , Leupeptinas/farmacología , Nucleocápside/genética , Nucleocápside/metabolismo , Regiones Promotoras Genéticas/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA