Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 187(11): 2735-2745.e12, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38723628

RESUMEN

Hepatitis B virus (HBV) is a small double-stranded DNA virus that chronically infects 296 million people. Over half of its compact genome encodes proteins in two overlapping reading frames, and during evolution, multiple selective pressures can act on shared nucleotides. This study combines an RNA-based HBV cell culture system with deep mutational scanning (DMS) to uncouple cis- and trans-acting sequence requirements in the HBV genome. The results support a leaky ribosome scanning model for polymerase translation, provide a fitness map of the HBV polymerase at single-nucleotide resolution, and identify conserved prolines adjacent to the HBV polymerase termination codon that stall ribosomes. Further experiments indicated that stalled ribosomes tether the nascent polymerase to its template RNA, ensuring cis-preferential RNA packaging and reverse transcription of the HBV genome.


Asunto(s)
Virus de la Hepatitis B , Transcripción Reversa , Humanos , Genoma Viral/genética , Virus de la Hepatitis B/genética , Mutación , Ribosomas/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Línea Celular
2.
Annu Rev Immunol ; 32: 513-45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24555472

RESUMEN

Interferon-stimulated gene (ISG) products take on a number of diverse roles. Collectively, they are highly effective at resisting and controlling pathogens. In this review, we begin by introducing interferon (IFN) and the JAK-STAT signaling pathway to highlight features that impact ISG production. Next, we describe ways in which ISGs both enhance innate pathogen-sensing capabilities and negatively regulate signaling through the JAK-STAT pathway. Several ISGs that directly inhibit virus infection are described with an emphasis on those that impact early and late stages of the virus life cycle. Finally, we describe ongoing efforts to identify and characterize antiviral ISGs, and we provide a forward-looking perspective on the ISG landscape.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad/fisiología , Interferones/metabolismo , Animales , Humanos , Quinasas Janus/metabolismo , Receptores de Interferón/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
3.
Cell ; 184(1): 120-132.e14, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33382968

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has claimed the lives of over one million people worldwide. The causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a member of the Coronaviridae family of viruses that can cause respiratory infections of varying severity. The cellular host factors and pathways co-opted during SARS-CoV-2 and related coronavirus life cycles remain ill defined. To address this gap, we performed genome-scale CRISPR knockout screens during infection by SARS-CoV-2 and three seasonal coronaviruses (HCoV-OC43, HCoV-NL63, and HCoV-229E). These screens uncovered host factors and pathways with pan-coronavirus and virus-specific functional roles, including major dependency on glycosaminoglycan biosynthesis, sterol regulatory element-binding protein (SREBP) signaling, bone morphogenetic protein (BMP) signaling, and glycosylphosphatidylinositol biosynthesis, as well as a requirement for several poorly characterized proteins. We identified an absolute requirement for the VMP1, TMEM41, and TMEM64 (VTT) domain-containing protein transmembrane protein 41B (TMEM41B) for infection by SARS-CoV-2 and three seasonal coronaviruses. This human coronavirus host factor compendium represents a rich resource to develop new therapeutic strategies for acute COVID-19 and potential future coronavirus pandemics.


Asunto(s)
Infecciones por Coronavirus/genética , Estudio de Asociación del Genoma Completo , SARS-CoV-2/fisiología , Células A549 , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Coronavirus Humano 229E/fisiología , Infecciones por Coronavirus/virología , Coronavirus Humano NL63/fisiología , Coronavirus Humano OC43/fisiología , Técnicas de Inactivación de Genes , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Mapeo de Interacción de Proteínas
4.
Cell ; 184(1): 133-148.e20, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33338421

RESUMEN

Flaviviruses pose a constant threat to human health. These RNA viruses are transmitted by the bite of infected mosquitoes and ticks and regularly cause outbreaks. To identify host factors required for flavivirus infection, we performed full-genome loss of function CRISPR-Cas9 screens. Based on these results, we focused our efforts on characterizing the roles that TMEM41B and VMP1 play in the virus replication cycle. Our mechanistic studies on TMEM41B revealed that all members of the Flaviviridae family that we tested require TMEM41B. We tested 12 additional virus families and found that SARS-CoV-2 of the Coronaviridae also required TMEM41B for infection. Remarkably, single nucleotide polymorphisms present at nearly 20% in East Asian populations reduce flavivirus infection. Based on our mechanistic studies, we propose that TMEM41B is recruited to flavivirus RNA replication complexes to facilitate membrane curvature, which creates a protected environment for viral genome replication.


Asunto(s)
Infecciones por Flavivirus/genética , Flavivirus/fisiología , Proteínas de la Membrana/metabolismo , Animales , Pueblo Asiatico/genética , Autofagia , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Sistemas CRISPR-Cas , Línea Celular , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/metabolismo , Infecciones por Flavivirus/virología , Técnicas de Inactivación de Genes , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple , SARS-CoV-2/fisiología , Replicación Viral , Virus de la Fiebre Amarilla/fisiología , Virus Zika/fisiología
5.
Cell ; 172(3): 423-438.e25, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29249360

RESUMEN

Stem cells are highly resistant to viral infection compared to their differentiated progeny; however, the mechanism is mysterious. Here, we analyzed gene expression in mammalian stem cells and cells at various stages of differentiation. We find that, conserved across species, stem cells express a subset of genes previously classified as interferon (IFN) stimulated genes (ISGs) but that expression is intrinsic, as stem cells are refractory to interferon. This intrinsic ISG expression varies in a cell-type-specific manner, and many ISGs decrease upon differentiation, at which time cells become IFN responsive, allowing induction of a broad spectrum of ISGs by IFN signaling. Importantly, we show that intrinsically expressed ISGs protect stem cells against viral infection. We demonstrate the in vivo importance of intrinsic ISG expression for protecting stem cells and their differentiation potential during viral infection. These findings have intriguing implications for understanding stem cell biology and the evolution of pathogen resistance.


Asunto(s)
Inmunidad Innata , Células Madre Pluripotentes/inmunología , Virosis/inmunología , Animales , Células Cultivadas , Femenino , Células HEK293 , Humanos , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Células Madre Pluripotentes/virología , Especificidad de la Especie
6.
Nat Immunol ; 20(4): 493-502, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833792

RESUMEN

Interferon-stimulated genes (ISGs) form the backbone of the innate immune system and are important for limiting intra- and intercellular viral replication and spread. We conducted a mass-spectrometry-based survey to understand the fundamental organization of the innate immune system and to explore the molecular functions of individual ISGs. We identified interactions between 104 ISGs and 1,401 cellular binding partners engaging in 2,734 high-confidence interactions. 90% of these interactions are unreported so far, and our survey therefore illuminates a far wider activity spectrum of ISGs than is currently known. Integration of the resulting ISG-interaction network with published datasets and functional studies allowed us to identify regulators of immunity and processes related to the immune system. Given the extraordinary robustness of the innate immune system, this ISG network may serve as a blueprint for therapeutic targeting of cellular systems to efficiently fight viral infections.


Asunto(s)
Inmunidad Innata , Interferones/fisiología , Mapeo de Interacción de Proteínas , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Expresión Génica , Glicoproteínas/metabolismo , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/genética , Espectrometría de Masas , Receptores CCR4/metabolismo , Receptores de Péptidos/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virales/metabolismo
7.
Immunity ; 46(3): 379-392, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28329704

RESUMEN

Type III interferons (IFN-λs) signal through a heterodimeric receptor complex composed of the IFN-λR1 subunit, specific for IFN-λs, and interleukin-10Rß (IL-10Rß), which is shared by multiple cytokines in the IL-10 superfamily. Low affinity of IL-10Rß for cytokines has impeded efforts aimed at crystallizing cytokine-receptor complexes. We used yeast surface display to engineer a higher-affinity IFN-λ variant, H11, which enabled crystallization of the ternary complex. The structure revealed that IL-10Rß uses a network of tyrosine residues as hydrophobic anchor points to engage IL-10 family cytokines that present complementary hydrophobic binding patches, explaining its role as both a cross-reactive but cytokine-specific receptor. H11 elicited increased anti-proliferative and antiviral activities in vitro and in vivo. In contrast, engineered higher-affinity type I IFNs did not increase antiviral potency over wild-type type I IFNs. Our findings provide insight into cytokine recognition by the IL-10R family and highlight the plasticity of type III interferon signaling and its therapeutic potential.


Asunto(s)
Interferones/inmunología , Receptores de Interferón/inmunología , Receptores de Interleucina-10/inmunología , Animales , Línea Celular , Cristalografía por Rayos X , Citometría de Flujo , Humanos , Ratones , Reacción en Cadena de la Polimerasa , Resonancia por Plasmón de Superficie
8.
EMBO Rep ; 23(12): e55218, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36256515

RESUMEN

Co-opting host cell protein synthesis is a hallmark of many virus infections. In response, certain host defense proteins limit mRNA translation globally, albeit at the cost of the host cell's own protein synthesis. Here, we describe an interferon-stimulated helicase, DDX60, that decreases translation from viral internal ribosome entry sites (IRESs). DDX60 acts selectively on type II IRESs of encephalomyocarditis virus (EMCV) and foot and mouth disease virus (FMDV), but not by other IRES types or by 5' cap. Correspondingly, DDX60 reduces EMCV and FMDV (type II IRES) replication, but not that of poliovirus or bovine enterovirus 1 (BEV-1; type I IRES). Furthermore, replacing the IRES of poliovirus with a type II IRES is sufficient for DDX60 to inhibit viral replication. Finally, DDX60 selectively modulates the amount of translating ribosomes on viral and in vitro transcribed type II IRES mRNAs, but not 5' capped mRNA. Our study identifies a novel facet in the repertoire of interferon-stimulated effector genes, the selective downregulation of translation from viral type II IRES elements.


Asunto(s)
Interferones , Sitios Internos de Entrada al Ribosoma
9.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33723056

RESUMEN

Human adenosine deaminase acting on RNA 1 (ADAR1) catalyzes adenosine-to-inosine deamination reactions on double-stranded RNA molecules to regulate cellular responses to endogenous and exogenous RNA. Defective ADAR1 editing leads to disorders such as Aicardi-Goutières syndrome, an autoinflammatory disease that manifests in the brain and skin, and dyschromatosis symmetrica hereditaria, a skin pigmentation disorder. Two ADAR1 protein isoforms, p150 (150 kDa) and p110 (110 kDa), are expressed and can edit RNA, but the contribution of each isoform to the editing landscape remains unclear, largely because of the challenges in expressing p150 without p110. In this study, we demonstrate that p110 is coexpressed with p150 from the canonical p150-encoding mRNA due to leaky ribosome scanning downstream of the p150 start codon. The presence of a strong Kozak consensus context surrounding the p110 start codon suggests the p150 mRNA is optimized to leak p110 alongside expression of p150. To reduce leaky scanning and translation initiation at the p110 start codon, we introduced synonymous mutations in the coding region between the p150 and p110 start codons. Cells expressing p150 constructs with these mutations produced significantly reduced levels of p110. Editing analysis of total RNA from ADAR1 knockout cells reconstituted separately with modified p150 and p110 revealed that more than half of the A-to-I edit sites are selectively edited by p150, and the other half are edited by either p150 or p110. This method of isoform-selective editing analysis, making use of the modified p150, has the potential to be adapted for other cellular contexts.


Asunto(s)
Adenosina Desaminasa/genética , Regulación de la Expresión Génica , Isoformas de Proteínas/genética , Edición de ARN , Proteínas de Unión al ARN/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Susceptibilidad a Enfermedades , Técnicas de Inactivación de Genes , Predisposición Genética a la Enfermedad , Humanos , Malformaciones del Sistema Nervioso/genética , Trastornos de la Pigmentación/congénito , Trastornos de la Pigmentación/genética
10.
Proc Natl Acad Sci U S A ; 117(3): 1678-1688, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31915293

RESUMEN

Primary human hepatocytes (PHHs) are an essential tool for modeling drug metabolism and liver disease. However, variable plating efficiencies, short lifespan in culture, and resistance to genetic manipulation have limited their use. Here, we show that the pyrrolizidine alkaloid retrorsine improves PHH repopulation of chimeric mice on average 10-fold and rescues the ability of even poorly plateable donor hepatocytes to provide cells for subsequent ex vivo cultures. These mouse-passaged (mp) PHH cultures overcome the marked donor-to-donor variability of cryopreserved PHH and remain functional for months as demonstrated by metabolic assays and infection with hepatitis B virus and Plasmodium falciparum mpPHH can be efficiently genetically modified in culture, mobilized, and then recultured as spheroids or retransplanted to create highly humanized mice that carry a genetically altered hepatocyte graft. Together, these advances provide flexible tools for the study of human liver disease and evaluation of hepatocyte-targeted gene therapy approaches.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatopatías/genética , Alcaloides de Pirrolicidina/farmacología , Animales , Trasplante de Células , Quimera , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Hepatitis B , Virus de la Hepatitis B , Hepatocitos/trasplante , Proteínas de Homeodominio/genética , Humanos , Hidrolasas/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Hígado/patología , Hepatopatías/patología , Malaria , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Plasmodium falciparum
11.
J Virol ; 93(18)2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31118263

RESUMEN

Given the unprecedented scale of the recent Ebola and Zika viral epidemics, it is crucial to understand the biology of host factors with broad antiviral action in order to develop novel therapeutic approaches. Here, we look into one such factor: zinc finger antiviral protein (ZAP) inhibits a variety of RNA and DNA viruses. Alternative splicing results in two isoforms that differ at their C termini: ZAPL (long) encodes a poly(ADP-ribose) polymerase (PARP)-like domain that is missing in ZAPS (short). Previously, it has been shown that ZAPL is more antiviral than ZAPS, while the latter is more induced by interferon (IFN). In this study, we discovered and confirmed the expression of two additional splice variants of human ZAP: ZAPXL (extralong) and ZAPM (medium). We also found two haplotypes of human ZAP. Since ZAPL and ZAPS have differential activities, we hypothesize that all four ZAP isoforms have evolved to mediate distinct antiviral and/or cellular functions. By taking a gene-knockout-and-reconstitution approach, we have characterized the antiviral, translational inhibition, and IFN activation activities of individual ZAP isoforms. Our work demonstrates that ZAPL and ZAPXL are more active against alphaviruses and hepatitis B virus (HBV) than ZAPS and ZAPM and elucidates the effects of splice variants on the action of a broad-spectrum antiviral factor.IMPORTANCE ZAP is an IFN-induced host factor that can inhibit a wide range of viruses, and there is great interest in fully characterizing its antiviral mechanism. This is the first study that defines the antiviral capacities of individual ZAP isoforms in the absence of endogenous ZAP expression and, hence, cross talk with other isoforms. Our data demonstrate that ZAP is expressed as four different forms: ZAPS, ZAPM, ZAPL, and ZAPXL. The longer ZAP isoforms better inhibit alphaviruses and HBV, while all isoforms equally inhibit Ebola virus transcription and replication. In addition, there is no difference in the abilities of ZAP isoforms to enhance the induction of type I IFN expression. Our results show that the full spectrum of ZAP activities can change depending on the virus target and the relative levels of basal expression and induction by IFN or infection.


Asunto(s)
Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Células A549 , Alphavirus/genética , Empalme Alternativo , Línea Celular , Células HEK293 , Haplotipos , Células HeLa , Virus de la Hepatitis B/genética , Humanos , Isoformas de Proteínas , Empalme del ARN/genética , ARN Viral/genética , Replicación Viral/efectos de los fármacos , Dedos de Zinc
12.
PLoS Pathog ; 13(1): e1006145, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28060952

RESUMEN

The host factor and interferon (IFN)-stimulated gene (ISG) product, zinc-finger antiviral protein (ZAP), inhibits a number of diverse viruses by usurping and intersecting with multiple cellular pathways. To elucidate its antiviral mechanism, we perform a loss-of-function genome-wide RNAi screen to identify cellular cofactors required for ZAP antiviral activity against the prototype alphavirus, Sindbis virus (SINV). In order to exclude off-target effects, we carry out stringent confirmatory assays to verify the top hits. Important ZAP-liaising partners identified include proteins involved in membrane ion permeability, type I IFN signaling, and post-translational protein modification. The factor contributing most to the antiviral function of ZAP is TRIM25, an E3 ubiquitin and ISG15 ligase. We demonstrate here that TRIM25 interacts with ZAP through the SPRY domain, and TRIM25 mutants lacking the RING or coiled coil domain fail to stimulate ZAP's antiviral activity, suggesting that both TRIM25 ligase activity and its ability to form oligomers are critical for its cofactor function. TRIM25 increases the modification of both the short and long ZAP isoforms by K48- and K63-linked polyubiquitin, although ubiquitination of ZAP does not directly affect its antiviral activity. However, TRIM25 is critical for ZAP's ability to inhibit translation of the incoming SINV genome. Taken together, these data uncover TRIM25 as a bona fide ZAP cofactor that leads to increased ZAP modification enhancing its translational inhibition activity.


Asunto(s)
Infecciones por Alphavirus/prevención & control , Antivirales/metabolismo , Proteínas de Unión al ARN/metabolismo , Virus Sindbis/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Cricetinae , Células HEK293 , Humanos , Interferón Tipo I/metabolismo , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
13.
Trends Immunol ; 36(3): 124-38, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25704559

RESUMEN

Over half a century has passed since interferons (IFNs) were discovered and shown to inhibit virus infection in cultured cells. Since then, researchers have steadily brought to light the molecular details of IFN signaling, catalogued their pleiotropic effects on cells, and harnessed their therapeutic potential for a variety of maladies. While advances have been plentiful, several fundamental questions have yet to be answered and much complexity remains to be unraveled. We explore the current knowledge surrounding four main questions: are type I IFN subtypes differentially produced in response to distinct pathogens? How are IFN subtypes distinguished by cells? What are the mechanisms and consequences of viral antagonism? Lastly, how can the IFN response be harnessed to improve vaccine efficacy?


Asunto(s)
Inmunidad Innata , Interferón Tipo I/inmunología , Interleucinas/inmunología , Linfocitos/inmunología , Virus/inmunología , Inmunidad Adaptativa , Evolución Biológica , Linaje de la Célula/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Evasión Inmune , Interferón Tipo I/clasificación , Interferón Tipo I/genética , Interleucinas/clasificación , Interleucinas/genética , Linfocitos/virología , Filogenia , Receptores de Reconocimiento de Patrones/genética , Receptores de Reconocimiento de Patrones/inmunología , Transducción de Señal , Virus/patogenicidad
14.
J Hepatol ; 66(2): 288-296, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27650283

RESUMEN

BACKGROUND & AIMS: As important virological markers, serum hepatitis B surface antigen (HBsAg) and hepatitis B virus (HBV) DNA levels show large fluctuations among chronic hepatitis B patients. The aim of this study was to reveal the potential impact and mechanisms of amino acid substitutions in small hepatitis B surface proteins (SHBs) on serum HBsAg and HBV DNA levels. METHODS: Serum samples from 230 untreated chronic hepatitis B patients with genotype C HBV were analyzed in terms of HBV DNA levels, serological markers of HBV infection and SHBs sequences. In vitro functional analysis of the identified SHBs mutants was performed. RESULTS: Among 230 SHBs sequences, there were 39 (16.96%) sequences with no mutation detected (wild-type) and 191 (83.04%) with single or multiple mutations. SHBs consist of 226 amino acids, of which 104 (46.02%) had mutations in our study. Some mutations (e.g., sE2G, sL21S, sR24K, sT47A/K, sC69stop (sC69∗), sL95W, sL98V, and sG145R) negatively correlated with serum HBsAg levels. HBsAg and HBV DNA levels from this group of patients had a positive correlation (r=0.61, p<0.001). In vitro analysis showed that these mutations reduced extracellular HBsAg and HBV DNA levels by restricting virion secretion and antibody binding capacity. Virion secretion could be rescued for sE2G, sC69∗, and sG145R by co-expression of wild-type HBsAg. CONCLUSION: The serum HBsAg levels were lower in untreated CHB patients with novel SHBs mutations outside the major antigenic region than those without mutations. Underlying mechanisms include impairment of virion secretion and lower binding affinity to antibodies used for HBsAg measurements. LAY SUMMARY: The hepatitis B surface antigen (HBsAg) is a major viral protein of the hepatitis B virus (HBV) secreted into patient blood serum and its quantification value serves as an important marker for the evaluation of chronic HBV infection and antiviral response. We found a few new amino acid substitutions in HBsAg associated with lower serum HBsAg and HBV DNA levels. These different substitutions might impair virion secretion, change the ability of HBsAg to bind to antibodies, or impact HBV replication. These could all result in decreased detectable levels of serum HBsAg. The factors affecting circulating HBsAg level and HBsAg detection are varied and caution is needed when interpreting clinical significance of serum HBsAg levels. Clinical trial number: NCT01088009.


Asunto(s)
ADN Viral , Antígenos de Superficie de la Hepatitis B , Virus de la Hepatitis B , Hepatitis B Crónica , Adulto , Sustitución de Aminoácidos , ADN Viral/análisis , ADN Viral/sangre , Femenino , Antígenos de Superficie de la Hepatitis B/sangre , Antígenos de Superficie de la Hepatitis B/genética , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/aislamiento & purificación , Hepatitis B Crónica/diagnóstico , Hepatitis B Crónica/virología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Proteínas Virales/genética , Virión/genética , Virión/aislamiento & purificación , Replicación Viral
15.
PLoS Pathog ; 11(11): e1005297, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26588073

RESUMEN

The hepatitis C virus (HCV) p7 protein is required for infectious virus production via its role in assembly and ion channel activity. Although NMR structures of p7 have been reported, the location of secondary structural elements and orientation of the p7 transmembrane domains differ among models. Furthermore, the p7 structure-function relationship remains unclear. Here, extensive mutagenesis, coupled with infectious virus production phenotyping and molecular modeling, demonstrates that the N-terminal helical region plays a previously underappreciated yet critical functional role, especially with respect to E2/p7 cleavage efficiency. Interrogation of specific N-terminal helix residues identified as having p7-specific defects and predicted to point toward the channel pore, in a context of independent E2/p7 cleavage, further supports p7 as a structurally plastic, minimalist ion channel. Together, our findings indicate that the p7 N-terminal helical region is critical for E2/p7 processing, protein-protein interactions, ion channel activity, and infectious HCV production.


Asunto(s)
Hepacivirus/metabolismo , Canales Iónicos/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Modelos Moleculares , Ensamble de Virus , Replicación Viral
16.
Nucleic Acids Res ; 43(11): 5647-63, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-25969444

RESUMEN

Target-site selection by retroviral integrase (IN) proteins profoundly affects viral pathogenesis. We describe the solution nuclear magnetic resonance structure of the Moloney murine leukemia virus IN (M-MLV) C-terminal domain (CTD) and a structural homology model of the catalytic core domain (CCD). In solution, the isolated MLV IN CTD adopts an SH3 domain fold flanked by a C-terminal unstructured tail. We generated a concordant MLV IN CCD structural model using SWISS-MODEL, MMM-tree and I-TASSER. Using the X-ray crystal structure of the prototype foamy virus IN target capture complex together with our MLV domain structures, residues within the CCD α2 helical region and the CTD ß1-ß2 loop were predicted to bind target DNA. The role of these residues was analyzed in vivo through point mutants and motif interchanges. Viable viruses with substitutions at the IN CCD α2 helical region and the CTD ß1-ß2 loop were tested for effects on integration target site selection. Next-generation sequencing and analysis of integration target sequences indicate that the CCD α2 helical region, in particular P187, interacts with the sequences distal to the scissile bonds whereas the CTD ß1-ß2 loop binds to residues proximal to it. These findings validate our structural model and disclose IN-DNA interactions relevant to target site selection.


Asunto(s)
Integrasas/química , Virus de la Leucemia Murina de Moloney/enzimología , Proteínas Virales/química , Dominio Catalítico , ADN/química , ADN/metabolismo , Integrasas/genética , Integrasas/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN , Proteínas Virales/metabolismo
17.
Nucleic Acids Res ; 42(9): 5917-28, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24623816

RESUMEN

We report alterations to the murine leukemia virus (MLV) integrase (IN) protein that successfully result in decreasing its integration frequency at transcription start sites and CpG islands, thereby reducing the potential for insertional activation. The host bromo and extraterminal (BET) proteins Brd2, 3 and 4 interact with the MLV IN protein primarily through the BET protein ET domain. Using solution NMR, protein interaction studies, and next generation sequencing, we show that the C-terminal tail peptide region of MLV IN is important for the interaction with BET proteins and that disruption of this interaction through truncation mutations affects the global targeting profile of MLV vectors. The use of the unstructured tails of gammaretroviral INs to direct association with complexes at active promoters parallels that used by histones and RNA polymerase II. Viruses bearing MLV IN C-terminal truncations can provide new avenues to improve the safety profile of gammaretroviral vectors for human gene therapy.


Asunto(s)
Integrasas/química , Virus de la Leucemia Murina/genética , Proteínas de Unión al ARN/química , Proteínas Virales/química , Integración Viral , Secuencia de Aminoácidos , Sitios de Unión , Islas de CpG , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Análisis de Secuencia de ADN , Eliminación de Secuencia , Factores de Transcripción , Sitio de Iniciación de la Transcripción
18.
Proc Natl Acad Sci U S A ; 110(23): 9487-92, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23661057

RESUMEN

The p12 protein of murine leukemia virus (MuLV) group-specific antigen (Gag) is associated with the preintegration complex, and mutants of p12 (PM14) show defects in nuclear entry or retention. Here we show that p12 proteins engineered to encode peptide sequences derived from known viral tethering proteins can direct chromatin binding during the early phase of viral replication and rescue a lethal p12-PM14 mutant. Peptides studied included segments of Kaposi sarcoma herpesvirus latency-associated nuclear antigen (LANA)(1-23), human papillomavirus 8 E2, and prototype foamy virus chromatin-binding sequences. Amino acid substitutions in Kaposi sarcoma herpesvirus LANA and prototype foamy virus chromatin-binding sequences that blocked nucleosome association failed to rescue MuLV p12-PM14. Rescue by a larger LANA peptide, LANA(1-32), required second-site mutations that are predicted to reduce peptide binding affinity to chromosomes, suggesting that excessively high binding affinity interfered with Gag/p12 function. This is supported by confocal microscopy of chimeric p12-GFP fusion constructs showing the reverted proteins had weaker association to condensed mitotic chromosomes. Analysis of the integration-site selection of these chimeric viruses showed no significant change in integration profile compared with wild-type MuLV, suggesting release of the tethered p12 post mitosis, before viral integration.


Asunto(s)
ADN Viral/genética , Productos del Gen gag/genética , Virus de la Leucemia Murina/genética , Integración Viral/genética , Replicación Viral/genética , Animales , Antígenos Virales/metabolismo , Secuencia de Bases , Western Blotting , Línea Celular , Cromatina/metabolismo , Clonación Molecular , Perros , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoprecipitación , Microscopía Confocal , Datos de Secuencia Molecular , Mutación/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Ingeniería de Proteínas , Análisis de Secuencia de ADN , Transactivadores/metabolismo
19.
Sci Adv ; 9(15): eadg6265, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37043562

RESUMEN

Hepatitis B virus (HBV) chronically infects an estimated 300 million people, and standard treatments are rarely curative. Infection increases the risk of liver cirrhosis and hepatocellular carcinoma, and consequently, nearly 1 million people die each year from chronic hepatitis B. Tools and approaches that bring insights into HBV biology and facilitate the discovery and evaluation of antiviral drugs are in demand. Here, we describe a method to initiate the replication of HBV, a DNA virus, using synthetic RNA. This approach eliminates contaminating background signals from input virus or plasmid DNA that plagues existing systems and can be used to study multiple stages of HBV replication. We further demonstrate that this method can be uniquely applied to identify sequence variants that confer resistance to antiviral drugs.


Asunto(s)
Hepatitis B Crónica , Neoplasias Hepáticas , Humanos , Virus de la Hepatitis B/genética , Antivirales/farmacología , Antivirales/uso terapéutico , ARN , Hepatitis B Crónica/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/tratamiento farmacológico , Replicación Viral
20.
Curr Opin Virol ; 52: 71-77, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34896863

RESUMEN

Flaviviruses are zoonotic pathogens transmitted by the bite of infected mosquitos and ticks and represent a constant burden to human health. Here we review recent literature aimed at uncovering how flaviviruses interact with the cells that they infect. A better understanding of these interactions may ultimately lead to novel therapeutic targets. We highlight several studies that employed low-biased methods to discover new protein-protein, protein-RNA, and genetic interactions, and spotlight recent work characterizing the host protein, TMEM41B, which has been shown to be critical for infection by diverse flaviviruses and coronaviruses.


Asunto(s)
Infecciones por Flavivirus , Flavivirus , Animales , Antivirales/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Flavivirus/genética , Flavivirus/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Provirus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA