Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 185(21): 3980-3991.e18, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36182704

RESUMEN

Simian arteriviruses are endemic in some African primates and can cause fatal hemorrhagic fevers when they cross into primate hosts of new species. We find that CD163 acts as an intracellular receptor for simian hemorrhagic fever virus (SHFV; a simian arterivirus), a rare mode of virus entry that is shared with other hemorrhagic fever-causing viruses (e.g., Ebola and Lassa viruses). Further, SHFV enters and replicates in human monocytes, indicating full functionality of all of the human cellular proteins required for viral replication. Thus, simian arteriviruses in nature may not require major adaptations to the human host. Given that at least three distinct simian arteriviruses have caused fatal infections in captive macaques after host-switching, and that humans are immunologically naive to this family of viruses, development of serology tests for human surveillance should be a priority.


Asunto(s)
Arterivirus , Fiebres Hemorrágicas Virales , Animales , Arterivirus/fisiología , Fiebres Hemorrágicas Virales/veterinaria , Fiebres Hemorrágicas Virales/virología , Humanos , Macaca , Primates , Zoonosis Virales , Internalización del Virus , Replicación Viral
2.
Cell ; 139(7): 1243-54, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064371

RESUMEN

Influenza viruses exploit host cell machinery to replicate, resulting in epidemics of respiratory illness. In turn, the host expresses antiviral restriction factors to defend against infection. To find host cell modifiers of influenza A H1N1 viral infection, we used a functional genomic screen and identified over 120 influenza A virus-dependency factors with roles in endosomal acidification, vesicular trafficking, mitochondrial metabolism, and RNA splicing. We discovered that the interferon-inducible transmembrane proteins IFITM1, 2, and 3 restrict an early step in influenza A viral replication. The IFITM proteins confer basal resistance to influenza A virus but are also inducible by interferons type I and II and are critical for interferon's virustatic actions. Further characterization revealed that the IFITM proteins inhibit the early replication of flaviviruses, including dengue virus and West Nile virus. Collectively this work identifies a family of antiviral restriction factors that mediate cellular innate immunity to at least three major human pathogens.


Asunto(s)
Infecciones por Flavivirus/inmunología , Gripe Humana/inmunología , Proteínas de la Membrana/inmunología , Animales , Antígenos de Diferenciación , Línea Celular Tumoral , Virus del Dengue/inmunología , Humanos , Inmunidad Innata , Virus de la Influenza A/inmunología , Interferones/inmunología , Ratones , Proteínas de Unión al ARN/inmunología , Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología
3.
Proc Natl Acad Sci U S A ; 114(27): 7112-7117, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630320

RESUMEN

CCR5 (R5)-tropic, but not CXCR4 (X4)-tropic, HIV-1 is associated with primary HIV-1 infection and transmission. Recent studies have shown that IFN-induced transmembrane (IFITM) proteins, including IFITM1, IFITM2, and IFITM3, restrict a broad range of viruses. Here, we demonstrate that an IFITM2 isoform (Δ20 IFITM2) lacking 20 amino acids at the N terminus differentially restricts X4 and R5 HIV-1. Δ20 IFITM2 suppresses replication of X4 HIV-1 strains by inhibiting their entry. High levels of Δ20 IFITM2 expression could be detected in CD4+ T cells and in monocytes. Infection of X4 viruses in monocyte-derived macrophages and dendritic cells is enhanced upon depletion of IFITM2 isoforms. Furthermore, we also show that coreceptor use is the determining factor for differential HIV-1 restriction of Δ20 IFITM2. When we replace the C terminus of CCR5 with the C terminus of CXCR4, R5 viruses become more susceptible to Δ20 IFITM2-mediated restriction. In contrast to previous studies, our research reveals that neither X4 nor R5 HIV-1 is suppressed by IFITM2 and IFITM3. The multifactor gatekeeping model has been proposed to explain restriction of X4 viruses in the early stage of HIV-1 diseases. Our findings indicate that Δ20 IFITM2 may serve as a major contributor to this gatekeeping mechanism.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/clasificación , Proteínas de la Membrana/metabolismo , Inmunidad Adaptativa , Alelos , Membrana Celular/metabolismo , Epítopos/inmunología , Frecuencia de los Genes , Células HEK293 , Infecciones por VIH/virología , Humanos , Inmunidad Innata , Células Jurkat , Isoformas de Proteínas , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Virión
4.
J Virol ; 92(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29212931

RESUMEN

The recent outbreak of Zika virus (ZIKV), a reemerging flavivirus, and its associated neurological disorders, such as Guillain-Barré (GB) syndrome and microcephaly, have generated an urgent need to develop effective ZIKV vaccines and therapeutic agents. Here, we used human endothelial cells and astrocytes, both of which represent key cell types for ZIKV infection, to identify potential inhibitors of ZIKV replication. Because several pathways, including the AMP-activated protein kinase (AMPK), protein kinase A (PKA), and mitogen-activated protein kinase (MAPK) signaling pathways, have been reported to play important roles in flavivirus replication, we tested inhibitors and agonists of these pathways for their effects on ZIKV replication. We identified the PKA inhibitor PKI 14-22 (PKI) to be a potent inhibitor of ZIKV replication. PKI effectively suppressed the replication of ZIKV from both the African and Asian/American lineages with a high efficiency and minimal cytotoxicity. While ZIKV infection does not induce PKA activation, endogenous PKA activity is essential for supporting ZIKV replication. Interestingly, in addition to PKA, PKI also inhibited another unknown target(s) to block ZIKV replication. PKI inhibited ZIKV replication at the postentry stage by preferentially affecting negative-sense RNA synthesis as well as viral protein translation. Together, these results have identified a potential inhibitor of ZIKV replication which could be further explored for future therapeutic application.IMPORTANCE There is an urgent need to develop effective vaccines and therapeutic agents against Zika virus (ZIKV) infection, a reemerging flavivirus associated with neurological disorders, including Guillain-Barré (GB) syndrome and microcephaly. By screening for inhibitors of several cellular pathways, we have identified the PKA inhibitor PKI 14-22 (PKI) to be a potent inhibitor of ZIKV replication. We show that PKI effectively suppresses the replication of all ZIKV strains tested with minimal cytotoxicity to human endothelial cells and astrocytes, two key cell types for ZIKV infection. Furthermore, we show that PKI inhibits ZIKV negative-sense RNA synthesis and viral protein translation. This study has identified a potent inhibitor of ZIKV infection which could be further explored for future therapeutic application.


Asunto(s)
Astrocitos/efectos de los fármacos , Proteínas Portadoras/farmacología , Células Endoteliales/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Fragmentos de Péptidos/farmacología , Virus Zika/efectos de los fármacos , Animales , Astrocitos/virología , Chlorocebus aethiops , Células Endoteliales/virología , Humanos , Sistema de Señalización de MAP Quinasas , Células Vero , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico
5.
J Med Virol ; 91(2): 179-189, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30192399

RESUMEN

After recent outbreaks, Zika virus (ZIKV) was linked to severe neurological diseases including Guillain-Barré syndrome in adults and microcephaly in newborns. The severities of pathological manifestations have been associated with different ZIKV strains. To better understand the tropism of ZIKV, we infected 10 human and four nonhuman cell lines (types) with two African (IbH30656 and MR766) and two Asian (PRVABC59 and H/FP/2013) ZIKV strains. Cell susceptibility to ZIKV infection was determined by examining viral titers, synthesis of viral proteins, and replication of positive and negative strands of viral genome. Among nonhuman cell lines, only Vero cells were efficiently infected by ZIKV. Among human cell lines, all were permissive to ZIKV infection. However, 293T and HeLa cells showed differential susceptibility towards African strains. In 293T cells, the NS1 protein was expressed at the high level by African strains but was almost not expressed by Asian strains though there was no obvious difference in viral genome replication, suggesting that the differential susceptibility might be controlled at the stage of viral protein translation. This study provides comprehensive results of the permissiveness of different cell types to both African and Asian ZIKV strains, which might help clarify their different pathogenesis.


Asunto(s)
Tropismo Viral , Replicación Viral , Virus Zika/crecimiento & desarrollo , Animales , Línea Celular , Humanos , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/virología
6.
J Biol Phys ; 41(2): 135-49, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25515930

RESUMEN

A biomimetic minimalist model membrane was used to study the mechanism and kinetics of cell-free in vitro HIV-1 Gag budding from a giant unilamellar vesicle (GUV). Real-time interaction of Gag, RNA, and lipid, leading to the formation of mini-vesicles, was measured using confocal microscopy. Gag forms resolution-limited punctae on the GUV lipid membrane. Introduction of the Gag and urea to a GUV solution containing RNA led to the budding of mini-vesicles on the inside surface of the GUV. The GUV diameter showed a linear decrease in time due to bud formation. Both bud formation and decrease in GUV size were proportional to Gag concentration. In the absence of RNA, addition of urea to GUVs incubated with Gag also resulted in subvesicle formation. These observations suggest the possibility that clustering of GAG proteins leads to membrane invagination even in the absence of host cell proteins. The method presented here is promising, and allows for systematic study of the dynamics of assembly of immature HIV and help classify the hierarchy of factors that impact the Gag protein initiated assembly of retroviruses such as HIV.


Asunto(s)
VIH-1/fisiología , Liposomas Unilamelares/metabolismo , Liberación del Virus , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/virología , VIH-1/efectos de los fármacos , VIH-1/metabolismo , Cinética , ARN Viral/metabolismo , Urea/farmacología , Liberación del Virus/efectos de los fármacos
7.
J Biol Chem ; 288(45): 32184-32193, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24067232

RESUMEN

The interferon-induced transmembrane (IFITM) proteins are a family of small membrane proteins that inhibit the cellular entry of several genera of viruses. These proteins had been predicted to adopt a two-pass, type III transmembrane topology with an intracellular loop, two transmembrane helices (TM1 and TM2), and extracellular N and C termini. Recent work, however, supports an intramembrane topology for the helices with cytosolic orientation of both termini. Here we determined the topology of murine Ifitm3. We found that the N terminus of Ifitm3 could be stained by antibodies at the cell surface but that this conformation was cell type-dependent and represented a minority of the total plasma membrane pool. In contrast, the C terminus was readily accessible to antibodies at the cell surface and extracellular C termini comprised most or all of those present at the plasma membrane. The addition of a C-terminal KDEL endoplasmic reticulum retention motif to Ifitm3 resulted in sequestration of Ifitm3 in the ER, demonstrating an ER-luminal orientation of the C terminus. C-terminal, but not N-terminal, epitope tags were also degraded within lysosomes, consistent with their luminal orientation. Furthermore, epitope-tagged Ifitm3 TM2 functioned as a signal anchor sequence when expressed in isolation. Collectively, our results demonstrate a type II transmembrane topology for Ifitm3 and will provide insight into its interaction with potential targets and cofactors.


Asunto(s)
Proteínas de la Membrana/metabolismo , Secuencias de Aminoácidos , Animales , Retículo Endoplásmico/química , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Lisosomas/química , Lisosomas/genética , Lisosomas/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Estructura Terciaria de Proteína
8.
J Virol ; 87(15): 8451-64, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720721

RESUMEN

We show that interferon-induced transmembrane protein 1 (IFITM-1), IFITM-2, and IFITM-3 exhibit a broad spectrum of antiviral activity against several members of the Bunyaviridae family, including Rift Valley fever virus (RVFV), La Crosse virus, Andes virus, and Hantaan virus, all of which can cause severe disease in humans and animals. We found that RVFV was restricted by IFITM-2 and -3 but not by IFITM-1, whereas the remaining viruses were equally restricted by all IFITMs. Indeed, at low doses of alpha interferon (IFN-α), IFITM-2 and -3 mediated more than half of the antiviral activity of IFN-α against RVFV. IFITM-2 and -3 restricted RVFV infection mostly by preventing virus membrane fusion with endosomes, while they had no effect on virion attachment to cells, endocytosis, or viral replication kinetics. We found that large fractions of IFITM-2 and IFITM-3 occupy vesicular compartments that are distinct from the vesicles coated by IFITM-1. In addition, although overexpression of all IFITMs expanded vesicular and acidified compartments within cells, there were marked phenotypic differences among the vesicular compartments occupied by IFITMs. Collectively, our data provide new insights into the possible mechanisms by which the IFITM family members restrict distinct viruses.


Asunto(s)
Antígenos de Diferenciación/inmunología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Línea Celular , Virus Hantaan/inmunología , Virus Hantaan/fisiología , Orthohantavirus/inmunología , Orthohantavirus/fisiología , Humanos , Interferón-alfa/inmunología , Virus La Crosse/inmunología , Virus La Crosse/fisiología
9.
PLoS Pathog ; 8(9): e1002909, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22969429

RESUMEN

Interferon-induced transmembrane (IFITM) proteins are a family of viral restriction factors that inhibit the entry processes of several pathogenic viruses, including influenza A virus (IAV), in vitro. Here we report that IAV-infected knockout mice lacking the Ifitm locus on chromosome 7 exhibited accelerated disease progression, greater mortality, and higher pulmonary and systemic viral burdens as compared to wild type controls. We further observed that the phenotype of Ifitm3-specific knockout mice was indistinguishable from that of mice lacking the entire Ifitm locus. Ifitm3 was expressed by IAV target cells including alveolar type II pneumocytes and tracheal/bronchial respiratory epithelial cells. Robust Ifitm3 expression was also observed in several tissues in the absence of infection. Among murine Ifitm promoters, only that of Ifitm3 could be induced by type I and II interferons. Ifitm3 could also be upregulated by the gp130 cytokines IL-6 and oncostatin M on cells expressing appropriate receptors, suggesting that multiple cytokine signals could contribute to Ifitm3 expression in a cell or tissue-specific manner. Collectively, these findings establish a central role for Ifitm3 in limiting acute influenza in vivo, and provide further insight into Ifitm3 expression and regulation.


Asunto(s)
Virus de la Influenza A/patogenicidad , Proteínas de la Membrana/fisiología , Infecciones por Orthomyxoviridae/genética , Células 3T3 , Enfermedad Aguda , Animales , Células Cultivadas , Embrión de Pollo , Resistencia a la Enfermedad/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/prevención & control , Índice de Severidad de la Enfermedad
10.
PLoS Pathog ; 7(1): e1001258, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21253575

RESUMEN

Interferon-inducible transmembrane proteins 1, 2, and 3 (IFITM1, 2, and 3) are recently identified viral restriction factors that inhibit infection mediated by the influenza A virus (IAV) hemagglutinin (HA) protein. Here we show that IFITM proteins restricted infection mediated by the entry glycoproteins (GP(1,2)) of Marburg and Ebola filoviruses (MARV, EBOV). Consistent with these observations, interferon-ß specifically restricted filovirus and IAV entry processes. IFITM proteins also inhibited replication of infectious MARV and EBOV. We observed distinct patterns of IFITM-mediated restriction: compared with IAV, the entry processes of MARV and EBOV were less restricted by IFITM3, but more restricted by IFITM1. Moreover, murine Ifitm5 and 6 did not restrict IAV, but efficiently inhibited filovirus entry. We further demonstrate that replication of infectious SARS coronavirus (SARS-CoV) and entry mediated by the SARS-CoV spike (S) protein are restricted by IFITM proteins. The profile of IFITM-mediated restriction of SARS-CoV was more similar to that of filoviruses than to IAV. Trypsin treatment of receptor-associated SARS-CoV pseudovirions, which bypasses their dependence on lysosomal cathepsin L, also bypassed IFITM-mediated restriction. However, IFITM proteins did not reduce cellular cathepsin activity or limit access of virions to acidic intracellular compartments. Our data indicate that IFITM-mediated restriction is localized to a late stage in the endocytic pathway. They further show that IFITM proteins differentially restrict the entry of a broad range of enveloped viruses, and modulate cellular tropism independently of viral receptor expression.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Filoviridae/patogenicidad , Virus de la Influenza A/patogenicidad , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Virosis/virología , Internalización del Virus , Animales , Antígenos de Diferenciación/inmunología , Línea Celular Tumoral , Chlorocebus aethiops , Endotelio Vascular , Femenino , Filoviridae/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Ratones , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/crecimiento & desarrollo , Células Vero , Virosis/inmunología , Virosis/metabolismo , Replicación Viral
11.
J Virol ; 82(10): 4834-43, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18321971

RESUMEN

Enveloped viruses use multiple mechanisms to inhibit infection of a target cell by more than one virion. These mechanisms may be of particular importance for the evolution of segmented viruses, because superinfection exclusion may limit the frequency of reassortment of viral genes. Here, we show that cellular expression of influenza A virus neuraminidase (NA), but not hemagglutinin (HA) or the M2 proton pump, inhibits entry of HA-pseudotyped retroviruses. Cells infected with H1N1 or H3N2 influenza A virus were similarly refractory to HA-mediated infection and to superinfection with a second influenza A virus. Both HA-mediated entry and viral superinfection were rescued by the neuraminidase inhibitors oseltamivir carboxylate and zanamivir. These inhibitors also prevented the removal of alpha-2,3- and alpha-2,6-linked sialic acid observed in cells expressing NA or infected with influenza A viruses. Our data indicate that NA alone among viral proteins limits influenza A virus superinfection.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Neuraminidasa/fisiología , Interferencia Viral , Proteínas Virales/fisiología , Animales , Antivirales/farmacología , Línea Celular , Perros , Hemaglutininas/genética , Hemaglutininas/fisiología , Humanos , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/genética , Oseltamivir/farmacología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/fisiología , Zanamivir/farmacología
12.
Sci Rep ; 7(1): 14883, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093521

RESUMEN

Zika virus (ZIKV) infection is associated with microcephaly in fetuses, but the pathogenesis of ZIKV-related microcephaly is not well understood. Here we show that ZIKV infects the subventricular zone in human fetal brain tissues and that the tissue tropism broadens with the progression of gestation. Our research demonstrates also that intermediate progenitor cells (IPCs) are the main target cells for ZIKV. Post-mitotic committed neurons become susceptible to ZIKV infection as well at later stages of gestation. Furthermore, activation of microglial cells, DNA fragmentation, and apoptosis of infected or uninfected cells could be found in ZIKV-infected brain tissues. Our studies identify IPCs as the main target cells for ZIKV. They also suggest that immune activation after ZIKV infection may play an important role in the pathogenesis of ZIKV-related microcephaly.


Asunto(s)
Encéfalo/virología , Feto/virología , Neuronas/virología , Células Madre/virología , Infección por el Virus Zika/patología , Virus Zika , Encéfalo/embriología , Encéfalo/patología , Femenino , Feto/patología , Humanos , Inmunidad Innata , Microcefalia/etiología , Mitosis , Embarazo , Técnicas de Cultivo de Tejidos , Infección por el Virus Zika/inmunología
13.
Annu Rev Virol ; 1: 261-283, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25599080

RESUMEN

Animal cells use a wide variety of mechanisms to slow or prevent replication of viruses. These mechanisms are usually mediated by antiviral proteins whose expression and activities can be constitutive but are frequently amplified by interferon induction. Among these interferon-stimulated proteins, members of the IFITM (interferon-induced transmembrane) family are unique because they prevent infection before a virus can traverse the lipid bilayer of the cell. At least three human IFITM proteins-IFITM1, IFITM2, and IFITM3-have antiviral activities. These activities limit infection in cultured cells by many viruses, including dengue virus, Ebola virus, influenza A virus, severe acute respiratory syndrome coronavirus, and West Nile virus. Murine Ifitm3 controls influenza A virus infection in vivo, and polymorphisms in human IFITM3 correlate with the severity of both seasonal and highly pathogenic avian influenza virus. Here we review the discovery and characterization of the IFITM proteins, describe the spectrum of their antiviral activities, and discuss potential mechanisms underlying these effects.

14.
PLoS One ; 9(5): e96579, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24827144

RESUMEN

Type I interferons (IFN-α and ß) induce dynamic host defense mechanisms to inhibit viral infections. It has been recently recognized that the interferon-inducible transmembrane proteins (IFITM) 1, 2 and 3 can block entry of a broad spectrum of RNA viruses. However, no study to date has focused on the role of IFITM proteins in DNA virus restriction. Here, we demonstrate that IFN-α or -ß treatment of keratinocytes substantially decreases human papillomavirus 16 (HPV16) infection while robustly inducing IFITM1, 2 and 3 expression. However, IFITM1, 2 and 3 overexpression did not inhibit HPV16 infection; rather, IFITM1 and IFITM3 modestly enhanced HPV16 infection in various cell types including primary keratinocytes. Moreover, IFITM1, 2 and 3 did not inhibit infection by two other DNA viruses, human cytomegalovirus (HCMV) and adenovirus type 5 (Ad5). Taken together, we reveal that the entry of several DNA viruses, including HPV, HCMV, and Ad5 is not affected by IFITM1, 2 and 3 expression. These results imply that HPV, and other DNA viruses, may bypass IFITM restriction during intracellular trafficking.


Asunto(s)
Adenoviridae/inmunología , Antígenos de Diferenciación/inmunología , Citomegalovirus/inmunología , Papillomavirus Humano 16/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Antígenos de Diferenciación/genética , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Interferón-alfa/farmacología , Interferón beta/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/metabolismo , Queratinocitos/virología , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética
15.
PLoS One ; 9(10): e110096, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25314048

RESUMEN

The IFITM3 polymorphism rs12252-C, which encodes an IFITM3 isoform (Δ21 IFITM3) lacking 21 amino acids at the amino terminus, has been controversially associated with poor clinical outcomes in patients with H1N1 influenza A virus (IAV) infections. In vitro studies have shown that Δ21 IFITM3 loses its ability to restrict H1N1 IAV. Subsequent research has also revealed that tyrosine 20 is the key determinant for IFITM3 endocytic trafficking, which is essential for the efficient anti-viral activity of IFITM3. In contrast to previous studies, we demonstrated that both Δ21 IFITM3 and an IFITM3 variant (Y20A IFITM3), in which tyrosine 20 is substituted with alanine, strongly restricted entry mediated by IAV H1, H3, H5, and H7 proteins. Δ21 IFITM3 also efficiently suppressed replication of H1N1 and, to a lesser extent, H3N2 IAV. Δ21 IFITM3 and Y20A IFITM3 had broader subcellular distributions than full-length IFITM3 but an abundant amount of both IFITM3 variants still localized to late endosomes and lysosomes. Our data indicate that tyrosine 20 partially regulates the subcellular localization of IFITM3 but is not functionally essential for IFITM3-mediated IAV restriction. They also suggested that mechanisms, other than viral entry restriction, might contribute to variations in clinical outcomes of H1N1 influenza associated with rs12252-C.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/genética , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética , Animales , Chlorocebus aethiops , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Gripe Humana/virología , Proteínas de la Membrana/metabolismo , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Proteínas de Unión al ARN/metabolismo , Células Vero , Internalización del Virus , Replicación Viral
16.
Cell Host Microbe ; 13(4): 452-64, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23601107

RESUMEN

Vesicle-membrane-protein-associated protein A (VAPA) and oxysterol-binding protein (OSBP) regulate intracellular cholesterol homeostasis, which is required for many virus infections. During entry, viruses or virus-containing vesicles can fuse with endosomal membranes to mediate the cytosolic release of virions, and alterations in endosomal cholesterol can inhibit this invasion step. We show that the antiviral effector protein interferon-inducible transmembrane protein 3 (IFITM3) interacts with VAPA and prevents its association with OSBP, thereby disrupting intracellular cholesterol homeostasis and inhibiting viral entry. By altering VAPA-OSBP function, IFITM3 induces a marked accumulation of cholesterol in multivesicular bodies and late endosomes, which inhibits the fusion of intraluminal virion-containing vesicles with endosomal membranes and thereby blocks virus release into the cytosol. Consequently, ectopic expression or depletion of the VAPA gene profoundly affects IFITM3-mediated inhibition of viral entry. Thus, IFITM3 disrupts intracellular cholesterol homeostasis to block viral entry, further underscoring the importance of cholesterol in virus infection.


Asunto(s)
Colesterol/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Virión/fisiología , Internalización del Virus , Línea Celular , Línea Celular Tumoral , Citosol/metabolismo , Citosol/virología , Endosomas/metabolismo , Endosomas/virología , Células HEK293 , Homeostasis/fisiología , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Proteínas R-SNARE/metabolismo , Receptores de Esteroides/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Vesiculovirus/metabolismo , Vesiculovirus/fisiología , Virión/metabolismo
17.
PLoS One ; 8(4): e60838, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573288

RESUMEN

Mannose-binding lectin (MBL) is a key soluble effector of the innate immune system that recognizes pathogen-specific surface glycans. Surprisingly, low-producing MBL genetic variants that may predispose children and immunocompromised individuals to infectious diseases are more common than would be expected in human populations. Since certain immune defense molecules, such as immunoglobulins, can be exploited by invasive pathogens, we hypothesized that MBL might also enhance infections in some circumstances. Consequently, the low and intermediate MBL levels commonly found in human populations might be the result of balancing selection. Using model infection systems with pseudotyped and authentic glycosylated viruses, we demonstrated that MBL indeed enhances infection of Ebola, Hendra, Nipah and West Nile viruses in low complement conditions. Mechanistic studies with Ebola virus (EBOV) glycoprotein pseudotyped lentiviruses confirmed that MBL binds to N-linked glycan epitopes on viral surfaces in a specific manner via the MBL carbohydrate recognition domain, which is necessary for enhanced infection. MBL mediates lipid-raft-dependent macropinocytosis of EBOV via a pathway that appears to require less actin or early endosomal processing compared with the filovirus canonical endocytic pathway. Using a validated RNA interference screen, we identified C1QBP (gC1qR) as a candidate surface receptor that mediates MBL-dependent enhancement of EBOV infection. We also identified dectin-2 (CLEC6A) as a potentially novel candidate attachment factor for EBOV. Our findings support the concept of an innate immune haplotype that represents critical interactions between MBL and complement component C4 genes and that may modify susceptibility or resistance to certain glycosylated pathogens. Therefore, higher levels of native or exogenous MBL could be deleterious in the setting of relative hypocomplementemia which can occur genetically or because of immunodepletion during active infections. Our findings confirm our hypothesis that the pressure of infectious diseases may have contributed in part to evolutionary selection of MBL mutant haplotypes.


Asunto(s)
Ebolavirus/fisiología , Infecciones por Filoviridae/metabolismo , Lectina de Unión a Manosa/metabolismo , Receptores Mitogénicos/metabolismo , Internalización del Virus , Animales , Chlorocebus aethiops , Proteínas del Sistema Complemento/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Glicoproteínas de Membrana/metabolismo , Pinocitosis , Células Vero , Proteínas del Envoltorio Viral/metabolismo
18.
PLoS One ; 7(3): e34508, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22479637

RESUMEN

Interferon-inducible transmembrane (IFITM) proteins restrict the entry processes of several pathogenic viruses, including the flaviviruses West Nile virus and dengue virus (DENV). DENV infects cells directly or via antibody-dependent enhancement (ADE) in Fc-receptor-bearing cells, a process thought to contribute to severe disease in a secondary infection. Here we investigated whether ADE-mediated DENV infection bypasses IFITM-mediated restriction or whether IFITM proteins can be protective in a secondary infection. We observed that IFITM proteins restricted ADE-mediated and direct infection with comparable efficiencies in a myelogenous leukemia cell line. Our data suggest that IFITM proteins can contribute to control of secondary DENV infections.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo , Virus del Dengue/inmunología , Dengue/inmunología , Interferones/inmunología , Proteínas de la Membrana/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Dengue/virología , Humanos , Ratones
20.
Cell Host Microbe ; 5(5): 439-49, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19454348

RESUMEN

The ubiquitin ligase TRIM25 mediates Lysine 63-linked ubiquitination of the N-terminal CARD domains of the viral RNA sensor RIG-I to facilitate type I interferon (IFN) production and antiviral immunity. Here, we report that the influenza A virus nonstructural protein 1 (NS1) specifically inhibits TRIM25-mediated RIG-I CARD ubiquitination, thereby suppressing RIG-I signal transduction. A novel domain in NS1 comprising E96/E97 residues mediates its interaction with the coiled-coil domain of TRIM25, thus blocking TRIM25 multimerization and RIG-I CARD domain ubiquitination. Furthermore, a recombinant influenza A virus expressing an E96A/E97A NS1 mutant is defective in blocking TRIM25-mediated antiviral IFN response and loses virulence in mice. Our findings reveal a mechanism by which influenza virus inhibits host IFN response and also emphasize the vital role of TRIM25 in modulating antiviral defenses.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Interacciones Huésped-Patógeno , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Femenino , Humanos , Virus de la Influenza A/química , Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/virología , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos BALB C , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores Inmunológicos , Transducción de Señal , Factores de Transcripción/química , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA