Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
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
2.
Proc Natl Acad Sci U S A ; 114(9): E1698-E1706, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28193861

RESUMEN

Many invasive bacteria establish pathogen-containing vacuoles (PVs) as intracellular niches for microbial growth. Immunity to these infections is dependent on the ability of host cells to recognize PVs as targets for host defense. The delivery of several host defense proteins to PVs is controlled by IFN-inducible guanylate binding proteins (GBPs), which themselves dock to PVs through poorly characterized mechanisms. Here, we demonstrate that GBPs detect the presence of bacterial protein secretion systems as "patterns of pathogenesis" associated with PVs. We report that the delivery of GBP2 to Legionella-containing vacuoles is dependent on the bacterial Dot/Icm secretion system, whereas the delivery of GBP2 to Yersinia-containing vacuoles (YCVs) requires hypersecretion of Yersinia translocon proteins. We show that the presence of bacterial secretion systems directs cytosolic carbohydrate-binding protein Galectin-3 to PVs and that the delivery of GBP1 and GBP2 to Legionella-containing vacuoles or YCVs is substantially diminished in Galectin-3-deficient cells. Our results illustrate that insertion of bacterial secretion systems into PV membranes stimulates Galectin-3-dependent recruitment of antimicrobial GBPs to PVs as part of a coordinated host defense program.


Asunto(s)
Antiinfecciosos/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Proteínas de Unión al GTP/metabolismo , Galectina 3/metabolismo , Vacuolas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Legionella/metabolismo , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Receptores de Superficie Celular/metabolismo
3.
Nature ; 484(7395): 519-23, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446628

RESUMEN

The 2009 H1N1 influenza pandemic showed the speed with which a novel respiratory virus can spread and the ability of a generally mild infection to induce severe morbidity and mortality in a subset of the population. Recent in vitro studies show that the interferon-inducible transmembrane (IFITM) protein family members potently restrict the replication of multiple pathogenic viruses. Both the magnitude and breadth of the IFITM proteins' in vitro effects suggest that they are critical for intrinsic resistance to such viruses, including influenza viruses. Using a knockout mouse model, we now test this hypothesis directly and find that IFITM3 is essential for defending the host against influenza A virus in vivo. Mice lacking Ifitm3 display fulminant viral pneumonia when challenged with a normally low-pathogenicity influenza virus, mirroring the destruction inflicted by the highly pathogenic 1918 'Spanish' influenza. Similar increased viral replication is seen in vitro, with protection rescued by the re-introduction of Ifitm3. To test the role of IFITM3 in human influenza virus infection, we assessed the IFITM3 alleles of individuals hospitalized with seasonal or pandemic influenza H1N1/09 viruses. We find that a statistically significant number of hospitalized subjects show enrichment for a minor IFITM3 allele (SNP rs12252-C) that alters a splice acceptor site, and functional assays show the minor CC genotype IFITM3 has reduced influenza virus restriction in vitro. Together these data reveal that the action of a single intrinsic immune effector, IFITM3, profoundly alters the course of influenza virus infection in mouse and humans.


Asunto(s)
Virus de la Influenza A/patogenicidad , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Proteínas de Unión al ARN/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Citocinas/inmunología , Inglaterra/epidemiología , Eliminación de Gen , Humanos , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/epidemiología , Gripe Humana/mortalidad , Gripe Humana/virología , Leucocitos/inmunología , Pulmón/patología , Pulmón/virología , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/etiología , Neumonía Viral/patología , Neumonía Viral/prevención & control , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Escocia/epidemiología , Replicación Viral
4.
Proc Natl Acad Sci U S A ; 112(41): E5628-37, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26417105

RESUMEN

Many microbes create and maintain pathogen-containing vacuoles (PVs) as an intracellular niche permissive for microbial growth and survival. The destruction of PVs by IFNγ-inducible guanylate binding protein (GBP) and immunity-related GTPase (IRG) host proteins is central to a successful immune response directed against numerous PV-resident pathogens. However, the mechanism by which IRGs and GBPs cooperatively detect and destroy PVs is unclear. We find that host cell priming with IFNγ prompts IRG-dependent association of Toxoplasma- and Chlamydia-containing vacuoles with ubiquitin through regulated translocation of the E3 ubiquitin ligase tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6). This initial ubiquitin labeling elicits p62-mediated escort and deposition of GBPs to PVs, thereby conferring cell-autonomous immunity. Hypervirulent strains of Toxoplasma gondii evade this process via specific rhoptry protein kinases that inhibit IRG function, resulting in blockage of downstream PV ubiquitination and GBP delivery. Our results define a ubiquitin-centered mechanism by which host cells deliver GBPs to PVs and explain how hypervirulent parasites evade GBP-mediated immunity.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , Proteínas de Unión al GTP/inmunología , Evasión Inmune , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Ubiquitina/inmunología , Vacuolas/inmunología , Animales , Proteínas de Unión al GTP/genética , Inmunidad Innata , Ratones , Ratones Noqueados , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Toxoplasmosis/genética , Toxoplasmosis/patología , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Vacuolas/metabolismo , Vacuolas/microbiología
5.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28784930

RESUMEN

Gram-negative bacterial pathogens utilize virulence-associated secretion systems to inject, or translocate, effector proteins into host cells to manipulate cellular processes and promote bacterial replication. However, translocated bacterial products are sensed by nucleotide binding domain and leucine-rich repeat-containing proteins (NLRs), which trigger the formation of a multiprotein complex called the inflammasome, leading to secretion of interleukin-1 (IL-1) family cytokines, pyroptosis, and control of pathogen replication. Pathogenic Yersinia bacteria inject effector proteins termed Yops, as well as pore-forming proteins that comprise the translocon itself, into target cells. The Yersinia translocation regulatory protein YopK promotes bacterial virulence by limiting hyperinjection of the translocon proteins YopD and YopB into cells, thereby limiting cellular detection of Yersinia virulence activity. How hyperinjection of translocon proteins leads to inflammasome activation is currently unknown. We found that translocated YopB and YopD colocalized with the late endosomal/lysosomal protein LAMP1 and that the frequency of YopD and LAMP1 association correlated with the level of caspase-1 activation in individual cells. We also observed colocalization between YopD and Galectin-3, an indicator of endosomal membrane damage. Intriguingly, YopK limited the colocalization of Galectin-3 with YopD, suggesting that YopK limits the induction or sensing of endosomal membrane damage by components of the type III secretion system (T3SS) translocon. Furthermore, guanylate binding proteins (GBPs) encoded on chromosome 3 (GbpChr3 ), which respond to pathogen-induced damage or alteration of host membranes, were necessary for inflammasome activation in response to hyperinjected YopB/-D. Our findings indicate that lysosomal damage by Yersinia translocon proteins promotes inflammasome activation and implicate GBPs as key regulators of this process.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Unión al GTP/genética , Inflamasomas/inmunología , Sistemas de Secreción Tipo III/metabolismo , Yersinia pseudotuberculosis/inmunología , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Caspasa 1/metabolismo , Línea Celular , Citocinas/biosíntesis , Citocinas/inmunología , Proteínas de Unión al GTP/metabolismo , Galectina 3/metabolismo , Inflamasomas/genética , Inflamasomas/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Transporte de Proteínas , Virulencia , Yersinia pseudotuberculosis/fisiología
6.
J Virol ; 87(14): 7837-52, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23658454

RESUMEN

The interferon-induced transmembrane protein 3 (IFITM3) gene is an interferon-stimulated gene that inhibits the replication of multiple pathogenic viruses in vitro and in vivo. IFITM3 is a member of a large protein superfamily, whose members share a functionally undefined area of high amino acid conservation, the CD225 domain. We performed mutational analyses of IFITM3 and identified multiple residues within the CD225 domain, consisting of the first intramembrane domain (intramembrane domain 1 [IM1]) and a conserved intracellular loop (CIL), that are required for restriction of both influenza A virus (IAV) and dengue virus (DENV) infection in vitro. Two phenylalanines within IM1 (F75 and F78) also mediate a physical association between IFITM proteins, and the loss of this interaction decreases IFITM3-mediated restriction. By extension, similar IM1-mediated associations may contribute to the functions of additional members of the CD225 domain family. IFITM3's distal N-terminal domain is also needed for full antiviral activity, including a tyrosine (Y20), whose alteration results in mislocalization of a portion of IFITM3 to the cell periphery and surface. Comparative analyses demonstrate that similar molecular determinants are needed for IFITM3's restriction of both IAV and DENV. However, a portion of the CIL including Y99 and R87 is preferentially needed for inhibition of the orthomyxovirus. Several IFITM3 proteins engineered with rare single-nucleotide polymorphisms demonstrated reduced expression or mislocalization, and these events were associated with enhanced viral replication in vitro, suggesting that possessing such alleles may impact an individual's risk for viral infection. On the basis of this and other data, we propose a model for IFITM3-mediated restriction.


Asunto(s)
Virus del Dengue/fisiología , Virus de la Influenza A/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Animales , Técnicas de Cultivo de Célula , Clonación Molecular , Secuencia Conservada/genética , Análisis Mutacional de ADN , ADN Complementario/genética , Perros , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Células de Riñón Canino Madin Darby , Espectrometría de Masas , Microscopía Confocal , Modelos Biológicos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Estructura Terciaria de Proteína/genética , Replicación Viral/genética
7.
PLoS Pathog ; 7(10): e1002337, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046135

RESUMEN

To replicate, viruses must gain access to the host cell's resources. Interferon (IFN) regulates the actions of a large complement of interferon effector genes (IEGs) that prevent viral replication. The interferon inducible transmembrane protein family members, IFITM1, 2 and 3, are IEGs required for inhibition of influenza A virus, dengue virus, and West Nile virus replication in vitro. Here we report that IFN prevents emergence of viral genomes from the endosomal pathway, and that IFITM3 is both necessary and sufficient for this function. Notably, viral pseudoparticles were inhibited from transferring their contents into the host cell cytosol by IFN, and IFITM3 was required and sufficient for this action. We further demonstrate that IFN expands Rab7 and LAMP1-containing structures, and that IFITM3 overexpression is sufficient for this phenotype. Moreover, IFITM3 partially resides in late endosomal and lysosomal structures, placing it in the path of invading viruses. Collectively our data are consistent with the prediction that viruses that fuse in the late endosomes or lysosomes are vulnerable to IFITM3's actions, while viruses that enter at the cell surface or in the early endosomes may avoid inhibition. Multiple viruses enter host cells through the late endocytic pathway, and many of these invaders are attenuated by IFN. Therefore these findings are likely to have significance for the intrinsic immune system's neutralization of a diverse array of threats.


Asunto(s)
Citosol/virología , Virus de la Influenza A/efectos de los fármacos , Gripe Humana/inmunología , Interferón gamma/farmacología , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Internalización del Virus/efectos de los fármacos , Animales , Pollos , Citosol/efectos de los fármacos , Citosol/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Interferón gamma/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Replicación Viral
8.
mBio ; 8(6)2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29233899

RESUMEN

Dynamin-like guanylate binding proteins (GBPs) are gamma interferon (IFN-γ)-inducible host defense proteins that can associate with cytosol-invading bacterial pathogens. Mouse GBPs promote the lytic destruction of targeted bacteria in the host cell cytosol, but the antimicrobial function of human GBPs and the mechanism by which these proteins associate with cytosolic bacteria are poorly understood. Here, we demonstrate that human GBP1 is unique among the seven human GBP paralogs in its ability to associate with at least two cytosolic Gram-negative bacteria, Burkholderia thailandensis and Shigella flexneri Rough lipopolysaccharide (LPS) mutants of S. flexneri colocalize with GBP1 less frequently than wild-type S. flexneri does, suggesting that host recognition of O antigen promotes GBP1 targeting to Gram-negative bacteria. The targeting of GBP1 to cytosolic bacteria, via a unique triple-arginine motif present in its C terminus, promotes the corecruitment of four additional GBP paralogs (GBP2, GBP3, GBP4, and GBP6). GBP1-decorated Shigella organisms replicate but fail to form actin tails, leading to their intracellular aggregation. Consequentially, the wild type but not the triple-arginine GBP1 mutant restricts S. flexneri cell-to-cell spread. Furthermore, human-adapted S. flexneri, through the action of one its secreted effectors, IpaH9.8, is more resistant to GBP1 targeting than the non-human-adapted bacillus B. thailandensis These studies reveal that human GBP1 uniquely functions as an intracellular "glue trap," inhibiting the cytosolic movement of normally actin-propelled Gram-negative bacteria. In response to this powerful human defense program, S. flexneri has evolved an effective counterdefense to restrict GBP1 recruitment.IMPORTANCE Several pathogenic bacterial species evolved to invade, reside in, and replicate inside the cytosol of their host cells. One adaptation common to most cytosolic bacterial pathogens is the ability to coopt the host's actin polymerization machinery in order to generate force for intracellular movement. This actin-based motility enables Gram-negative bacteria, such as Shigella species, to propel themselves into neighboring cells, thereby spreading from host cell to host cell without exiting the intracellular environment. Here, we show that the human protein GBP1 acts as a cytosolic "glue trap," capturing cytosolic Gram-negative bacteria through a unique protein motif and preventing disseminated infections in cell culture models. To escape from this GBP1-mediated host defense, Shigella employs a virulence factor that prevents or dislodges the association of GBP1 with cytosolic bacteria. Thus, therapeutic strategies to restore GBP1 binding to Shigella may lead to novel treatment options for shigellosis in the future.


Asunto(s)
Actinas/metabolismo , Secuencias de Aminoácidos , Arginina/química , Citosol/microbiología , Proteínas de Unión al GTP/química , Shigella flexneri/fisiología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Burkholderia/fisiología , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Células HeLa , Humanos , Lipopolisacáridos/genética , Mutación , Antígenos O/metabolismo , Shigella flexneri/patogenicidad , Ubiquitinación , Factores de Virulencia
9.
mBio ; 8(5)2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974614

RESUMEN

The Gram-negative bacterial cell wall component lipopolysaccharide (LPS) is recognized by the noncanonical inflammasome protein caspase-11 in the cytosol of infected host cells and thereby prompts an inflammatory immune response linked to sepsis. Host guanylate binding proteins (GBPs) promote infection-induced caspase-11 activation in tissue culture models, and yet their in vivo role in LPS-mediated sepsis has remained unexplored. LPS can be released from lysed bacteria as "free" LPS aggregates or actively secreted by live bacteria as a component of outer membrane vesicles (OMVs). Here, we report that GBPs control inflammation and sepsis in mice injected with either free LPS or purified OMVs derived from Gram-negative Escherichia coli In agreement with our observations from in vivo experiments, we demonstrate that macrophages lacking GBP2 expression fail to induce pyroptotic cell death and proinflammatory interleukin-1ß (IL-1ß) and IL-18 secretion when exposed to OMVs. We propose that in order to activate caspase-11 in vivo, GBPs control the processing of bacterium-derived OMVs by macrophages as well as the processing of circulating free LPS by as-yet-undetermined cell types.IMPORTANCE The bacterial cell wall component LPS is a strong inducer of inflammation and is responsible for much of the toxicity of Gram-negative bacteria. Bacteria shed some of their cell wall and its associated LPS in the form of outer membrane vesicles (OMVs). Recent work demonstrated that secreted OMVs deliver LPS into the host cell cytosol by an unknown mechanism, resulting in the activation of the proinflammatory LPS sensor caspase-11. Here, we show that activation of cytosolic caspase-11 by OMVs requires additional host factors, the so-called guanylate binding proteins (GBPs). The discovery of GBPs as regulators of OMV-mediated inflammation paves the way toward a mechanistic understanding of the host response toward bacterial OMVs and may lead to effective strategies to ameliorate inflammation induced by bacterial infections.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Escherichia coli/patogenicidad , Proteínas de Unión al GTP/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Animales , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/genética , Caspasas/metabolismo , Caspasas Iniciadoras , Células Cultivadas , Citosol/metabolismo , Activación Enzimática , Inflamación , Interleucina-18/biosíntesis , Interleucina-1beta/biosíntesis , Lipopolisacáridos/inmunología , Ratones , Piroptosis , Vesículas Secretoras/metabolismo
10.
Cell Rep ; 13(8): 1717-31, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26586435

RESUMEN

Direct visualization of HIV-1 replication would improve our understanding of the viral life cycle. We adapted established technology and reagents to develop an imaging approach, ViewHIV, which allows evaluation of early HIV-1 replication intermediates, from reverse transcription to integration. These methods permit the simultaneous evaluation of both the capsid protein (CA) and viral DNA genome (vDNA) components of HIV-1 in both the cytosol and nuclei of single cells. ViewHIV is relatively rapid, uses readily available reagents in combination with standard confocal microscopy, and can be done with virtually any HIV-1 strain and permissive cell lines or primary cells. Using ViewHIV, we find that CA enters the nucleus and associates with vDNA in both transformed and primary cells. We also find that CA's interaction with the host polyadenylation factor, CPSF6, enhances nuclear entry and potentiates HIV-1's depth of nuclear invasion, potentially aiding the virus's integration into gene-dense regions.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , VIH-1/genética , Integración Viral/genética , Replicación Viral/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citosol/metabolismo , Replicación del ADN/genética , Genoma Viral/genética , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Células HeLa , Humanos , Transcripción Reversa/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA