Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Virol ; : e0085024, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38953378

RESUMEN

Viruses are obligate parasites that depend on the cellular machinery for their propagation. Several viruses also incorporate cellular proteins that facilitate viral spread. Defining these cellular proteins is critical to decipher viral life cycles and delineate novel therapeutic strategies. While numerous studies have explored the importance of host proteins in coronavirus spread, information about their presence in mature virions is limited. In this study, we developed a protocol to highly enrich mature HCoV-OC43 virions and characterize them by proteomics. Recognizing that cells release extracellular vesicles whose content is modulated by viruses, and given our ability to separate virions from these vesicles, we also analyzed their protein content in both uninfected and infected cells. We uncovered 69 unique cellular proteins associated with virions including 31 high-confidence hits. These proteins primarily regulate RNA metabolism, enzymatic activities, vesicular transport, cell adhesion, metabolite interconversion, and translation. We further discovered that the virus had a profound impact on exosome composition, incorporating 47 novel cellular proteins (11 high confidence) and excluding 92 others (61 high confidence) in virus-associated extracellular vesicles compared to uninfected cells. Moreover, a dsiRNA screen revealed that 11 of 18 select targets significantly impacted viral yields, including proteins found in virions or extracellular vesicles. Overall, this study provides new and important insights into the incorporation of numerous host proteins into HCoV-OC43 virions, their biological significance, and the ability of the virus to modulate extracellular vesicles. IMPORTANCE: In recent years, coronaviruses have dominated global attention, making it crucial to develop methods to control them and prevent future pandemics. Besides viral proteins, host proteins play a significant role in viral propagation and offer potential therapeutic targets. Targeting host proteins is advantageous because they are less likely to mutate and develop resistance compared to viral proteins, a common issue with many antiviral treatments. In this study, we examined the protein content of the less virulent biosafety level 2 HCoV-OC43 virus as a stand-in for the more virulent SARS-CoV-2. Our findings reveal that several cellular proteins incorporated into the virion regulate viral spread. In addition, we report that the virus extensively modulates the content of extracellular vesicles, enhancing viral dissemination. This underscores the critical interplay between the virus, host proteins, and extracellular vesicles.

2.
FASEB J ; 37(2): e22765, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36607642

RESUMEN

The first line of defense against respiratory viruses relies on the antiviral and proinflammatory cytokine response initiated in infected respiratory epithelial cells. The cytokine response not only restricts virus replication and spreading, but also orchestrates the subsequent immune response. The epithelial Dual Oxidase 2 (DUOX2) has recently emerged as a regulator of the interferon antiviral response. Here, we investigated the role of DUOX2 in the inflammatory cytokine response using a model of A549 cells deficient in DUOX2 generated using Crispr-Cas9 and infected by Sendai virus. We found that the absence of DUOX2 selectively reduced the induction of a restricted panel of 14 cytokines and chemokines secreted in response to Sendai virus by 20 to 89%. The secreted factors produced by epithelial cells upon virus infection promoted the migration, adhesion, and degranulation of primary human neutrophils, in part through the DUOX2-dependent secretion of TNF and chemokines. In contrast, DUOX2 expression did not impact neutrophil viability or NETosis, thereby highlighting a selective impact of DUOX2 in neutrophil functions. Overall, this study unveils previously unrecognized roles of epithelial DUOX2 in the epithelial-immune cells crosstalk during respiratory virus infection.


Asunto(s)
Neutrófilos , Virus , Humanos , Oxidasas Duales/genética , Oxidasas Duales/metabolismo , Células Epiteliales/metabolismo , Citocinas/metabolismo , Antivirales/farmacología , Quimiocinas/metabolismo
3.
J Biol Chem ; 295(12): 3808-3825, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32029478

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal disease, characterized by the selective loss of motor neurons leading to paralysis. Mutations in the gene encoding superoxide dismutase 1 (SOD1) are the second most common cause of familial ALS, and considerable evidence suggests that these mutations result in an increase in toxicity due to protein misfolding. We previously demonstrated in the SOD1G93A rat model that misfolded SOD1 exists as distinct conformers and forms deposits on mitochondrial subpopulations. Here, using SOD1G93A rats and conformation-restricted antibodies specific for misfolded SOD1 (B8H10 and AMF7-63), we identified the interactomes of the mitochondrial pools of misfolded SOD1. This strategy identified binding proteins that uniquely interacted with either AMF7-63 or B8H10-reactive SOD1 conformers as well as a high proportion of interactors common to both conformers. Of this latter set, we identified the E3 ubiquitin ligase TNF receptor-associated factor 6 (TRAF6) as a SOD1 interactor, and we determined that exposure of the SOD1 functional loops facilitates this interaction. Of note, this conformational change was not universally fulfilled by all SOD1 variants and differentiated TRAF6 interacting from TRAF6 noninteracting SOD1 variants. Functionally, TRAF6 stimulated polyubiquitination and aggregation of the interacting SOD1 variants. TRAF6 E3 ubiquitin ligase activity was required for the former but was dispensable for the latter, indicating that TRAF6-mediated polyubiquitination and aggregation of the SOD1 variants are independent events. We propose that the interaction between misfolded SOD1 and TRAF6 may be relevant to the etiology of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Superóxido Dismutasa-1/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Anticuerpos/inmunología , Línea Celular , Modelos Animales de Enfermedad , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , Agregado de Proteínas , Pliegue de Proteína , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Transgénicas , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/inmunología , Factor 6 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 6 Asociado a Receptor de TNF/genética , Ubiquitinación
4.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32321802

RESUMEN

Influenza A virus (IAV) increases the presentation of class I human leukocyte antigen (HLA) proteins that limit antiviral responses mediated by natural killer (NK) cells, but molecular mechanisms for these processes have not yet been fully elucidated. We observed that infection with A/Fort Monmouth/1/1947(H1N1) IAV significantly increased the presentation of HLA-B, -C, and -E on lung epithelial cells. Virus entry was not sufficient to induce HLA upregulation because UV-inactivated virus had no effect. Aberrant internally deleted viral RNAs (vRNAs) known as mini viral RNAs (mvRNAs) and defective interfering RNAs (DI RNAs) expressed from an IAV minireplicon were sufficient for inducing HLA upregulation. These defective RNAs bind to retinoic acid-inducible gene I (RIG-I) and initiate mitochondrial antiviral signaling (MAVS) protein-dependent antiviral interferon (IFN) responses. Indeed, MAVS was required for HLA upregulation in response to IAV infection or ectopic mvRNA/DI RNA expression. The effect was partially due to paracrine signaling, as we observed that IAV infection or mvRNA/DI RNA-expression stimulated production of IFN-ß and IFN-λ1 and conditioned media from these cells elicited a modest increase in HLA surface levels in naive epithelial cells. HLA upregulation in response to aberrant viral RNAs could be prevented by the Janus kinase (JAK) inhibitor ruxolitinib. While HLA upregulation would seem to be advantageous to the virus, it is kept in check by the viral nonstructural 1 (NS1) protein; we determined that NS1 limits cell-intrinsic and paracrine mechanisms of HLA upregulation. Taken together, our findings indicate that aberrant IAV RNAs stimulate HLA presentation, which may aid viral evasion of innate immunity.IMPORTANCE Human leukocyte antigens (HLAs) are cell surface proteins that regulate innate and adaptive immune responses to viral infection by engaging with receptors on immune cells. Many viruses have evolved ways to evade host immune responses by modulating HLA expression and/or processing. Here, we provide evidence that aberrant RNA products of influenza virus genome replication can trigger retinoic acid-inducible gene I (RIG-I)/mitochondrial antiviral signaling (MAVS)-dependent remodeling of the cell surface, increasing surface presentation of HLA proteins known to inhibit the activation of an immune cell known as a natural killer (NK) cell. While this HLA upregulation would seem to be advantageous to the virus, it is kept in check by the viral nonstructural 1 (NS1) protein, which limits RIG-I activation and interferon production by the infected cell.


Asunto(s)
Genes MHC Clase I/genética , Antígenos HLA/metabolismo , Subtipo H1N1 del Virus de la Influenza A/genética , Células A549 , Proteínas Adaptadoras Transductoras de Señales/genética , Proteína 58 DEAD Box/genética , Bases de Datos Genéticas , Células Epiteliales/virología , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata , Virus de la Influenza A/genética , Gripe Humana/genética , Células Asesinas Naturales/metabolismo , Pulmón/virología , ARN Viral/genética , Transducción de Señal , Activación Transcripcional , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
5.
Proc Natl Acad Sci U S A ; 115(4): E601-E609, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29317535

RESUMEN

Cytokine signaling through the JAK/STAT pathway controls multiple cellular responses including growth, survival, differentiation, and pathogen resistance. An expansion in the gene regulatory repertoire controlled by JAK/STAT signaling occurs through the interaction of STATs with IRF transcription factors to form ISGF3, a complex that contains STAT1, STAT2, and IRF9 and regulates expression of IFN-stimulated genes. ISGF3 function depends on selective interaction between IRF9, through its IRF-association domain (IAD), with the coiled-coil domain (CCD) of STAT2. Here, we report the crystal structures of the IRF9-IAD alone and in a complex with STAT2-CCD. Despite similarity in the overall structure among respective paralogs, the surface features of the IRF9-IAD and STAT2-CCD have diverged to enable specific interaction between these family members. We derive a model for the ISGF3 complex bound to an ISRE DNA element and demonstrate that the observed interface between STAT2 and IRF9 is required for ISGF3 function in cells.


Asunto(s)
Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor de Transcripción STAT2/metabolismo , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Quinasas Janus/metabolismo , Ratones , Mutación Puntual , Dominios Proteicos , Factor de Transcripción STAT2/genética , Transducción de Señal
6.
BMC Cancer ; 17(1): 356, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28532474

RESUMEN

BACKGROUND: IKKε is an oncogenic kinase that was found amplified and overexpressed in a substantial percentage of human breast cancer cell lines and primary tumors using genomic and gene expression analyses. Molecular studies have provided the rational for a key implication of IKKε in breast cancer cells proliferation and invasiveness through the phosphorylation of several substrates. METHODS: Here, we performed immunohistochemical detection of IKKε expression on tissue microarrays constituted of 154 characterized human breast cancer tumors. We further determined the association with multiple clinicopathological parameters and 5-years overall, disease-free and distant disease free survival. RESULTS: We observed expression of IKKε in 60.4% of the breast cancer tumors. IKKε expression status showed no association with a panel of markers used for molecular classification of the tumors, including ER/PR/HER2 status, or with the molecular subtypes. However, IKKε expression was inversely associated with lymph node metastasis status (p = 0.0032). Additionally, we identified a novel association between IKKε and EGFR expression (p = 0.0011). CONCLUSIONS: The unexpected observation of an inverse association between IKKε and lymph node metastasis advocates for larger scale immunohistochemical profiling of primary breast tumors to clarify the role of IKKε in metastasis. This study suggests that breast cancer tumors expressing EGFR and IKKε may be potential targets for drugs aiming at inhibiting IKKε activity or expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Receptores ErbB/metabolismo , Quinasa I-kappa B/metabolismo , Adulto , Anticuerpos Monoclonales/química , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/secundario , Supervivencia sin Enfermedad , Femenino , Humanos , Quinasa I-kappa B/inmunología , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Células MCF-7 , Persona de Mediana Edad , Pronóstico
7.
PLoS Pathog ; 9(6): e1003416, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785285

RESUMEN

To identify new regulators of antiviral innate immunity, we completed the first genome-wide gene silencing screen assessing the transcriptional response at the interferon-ß (IFNB1) promoter following Sendai virus (SeV) infection. We now report a novel link between WNT signaling pathway and the modulation of retinoic acid-inducible gene I (RIG-I)-like receptor (RLR)-dependent innate immune responses. Here we show that secretion of WNT2B and WNT9B and stabilization of ß-catenin (CTNNB1) upon virus infection negatively regulate expression of representative inducible genes IFNB1, IFIT1 and TNF in a CTNNB1-dependent effector mechanism. The antiviral response is drastically reduced by glycogen synthase kinase 3 (GSK3) inhibitors but restored in CTNNB1 knockdown cells. The findings confirm a novel regulation of antiviral innate immunity by a canonical-like WNT/CTNNB1 signaling pathway. The study identifies novel avenues for broad-spectrum antiviral targets and preventing immune-mediated diseases upon viral infection.


Asunto(s)
Glicoproteínas/inmunología , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/inmunología , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Proteínas Wnt/inmunología , Vía de Señalización Wnt/inmunología , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Estudio de Asociación del Genoma Completo , Glicoproteínas/metabolismo , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Interferencia de ARN , Proteínas de Unión al ARN , Receptores Inmunológicos , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/patología , Virus Sendai/metabolismo , Proteínas Wnt/metabolismo
8.
Clin Sci (Lond) ; 128(6): 337-47, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25456319

RESUMEN

Determining the role of NADPH oxidases in the context of virus infection is an emerging area of research and our knowledge is still sparse. The expression of various isoforms of NOX/DUOX (NADPH oxidase/dual oxidase) in the epithelial cells (ECs) lining the respiratory tract renders them primary sites from which to orchestrate the host defence against respiratory viruses. Accumulating evidence reveals distinct facets of the involvement of NOX/DUOX in host antiviral and pro-inflammatory responses and in the control of the epithelial barrier integrity, with individual isoforms mediating co-operative, but surprisingly also opposing, functions. Although in vivo studies in mice are in line with some of these observations, a complete understanding of the specific functions of epithelial NOX/DUOX awaits lung epithelial-specific conditional knockout mice. The goal of the present review is to summarize our current knowledge of the role of individual NOX/DUOX isoforms expressed in the lung epithelium in the context of respiratory virus infections so as to highlight potential opportunities for therapeutic intervention.


Asunto(s)
Pulmón/inmunología , NADPH Oxidasas/inmunología , Infecciones del Sistema Respiratorio/inmunología , Virosis/inmunología , Animales , Oxidasas Duales , Células Epiteliales/inmunología , Humanos , Inmunidad Mucosa , Mediadores de Inflamación/metabolismo , Isoenzimas/inmunología , Mucosa Respiratoria/inmunología
9.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119722, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38615720

RESUMEN

This review explores the nuanced role of reactive oxygen species (ROS) in cell fate, challenging the traditional view that equates ROS with cellular damage. Through significant technological advancements in detecting localized redox states and identifying oxidized cysteines, a paradigm shift has emerged: from ROS as merely damaging agents to crucial players in redox signaling. We delve into the intricacies of redox mechanisms, which, although confined, exert profound influences on cellular physiological responses. Our analysis extends to both the positive and negative impacts of these mechanisms on cell death processes, including uncontrolled and programmed pathways. By unraveling these complex interactions, we argue against the oversimplified notion of a 'stress response', advocating for a more nuanced understanding of redox signaling. This review underscores the importance of localized redox states in determining cell fate, highlighting the sophistication and subtlety of ROS functions beyond mere damage.


Asunto(s)
Oxidación-Reducción , Especies Reactivas de Oxígeno , Transducción de Señal , Especies Reactivas de Oxígeno/metabolismo , Humanos , Estrés Oxidativo , Animales , Apoptosis
10.
J Immunol ; 186(8): 4618-30, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21398606

RESUMEN

HIV type 1 infection is associated with a rapid depletion of Th17 cells from the GALT. The chemokine receptor CCR6 is a marker for Th17 lineage polarization and HIV permissiveness in memory CD4(+) T cells. CCR6(+) T cells have the potential to migrate into the GALT via the gut-homing integrin α(4)ß(7), a newly identified HIV-gp120 binding receptor. In this study, we investigated whether memory T cells coexpressing CCR6 and integrin ß(7) are selective HIV targets and whether retinoic acid (RA)-induced imprinting for gut-homing selectively increases CCR6(+) T cell permissiveness to infection. We demonstrated that ß(7)(-)R6(+) and ß(7)(+)R6(+) compared with ß(7)(-)R6(-) and ß(7)(+)R6(-) T cells were highly permissive to HIV, produced Th17 cytokines, and their frequency was decreased in the peripheral blood of HIV-infected subjects. RA upregulated integrin α(4) and ß(7) coexpression in both CCR6(+) and CCR6(-) T cells, but increased HIV permissiveness selectively in CCR6(+) T cells via entry (CCR5 upregulation) and postentry mechanisms. In conclusion, these results demonstrate that CCR6, but not the integrin ß(7), is a discriminative marker for memory T cells imprinted with a transcriptional program favorable to HIV replication. Nevertheless, given the ability of integrin ß(7) to regulate cell migration into the GALT and bind HIV-gp120, CCR6(+) T cells coexpressing integrin ß(7) and CCR5 might have an extraordinary ability to disseminate HIV from the portal sites of entry. Understanding the molecular mechanisms of memory CCR6(+) T cell differentiation is critical for the design of new therapeutic strategies that should interfere with viral permissiveness but not Th17 lineage commitment and gut-homing potential in CCR6(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , VIH-1/inmunología , Cadenas beta de Integrinas/inmunología , Receptores CCR6/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Diferenciación Celular/inmunología , Células Cultivadas , ADN Viral/genética , Citometría de Flujo , Células HEK293 , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Memoria Inmunológica/inmunología , Integrina alfa4/inmunología , Integrina alfa4/metabolismo , Cadenas beta de Integrinas/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Reacción en Cadena de la Polimerasa , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Receptores CCR6/metabolismo , Tretinoina/farmacología , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
11.
Redox Biol ; 59: 102583, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36567215

RESUMEN

Protein disulfide isomerases (PDIs) catalyze redox reactions that reduce, oxidize, or isomerize disulfide bonds and act as chaperones of proteins as they fold. The characteristic features of PDIs are the presence of one or more catalytic thioredoxin (TRX)-like domains harboring typical CXXC catalytic motifs responsible for redox reactions, as well as non-catalytic TRX-like domain. As increasing attention is paid to oxidative post-translational modifications of cysteines (Cys ox-PTMs) with the recognition that they control cellular signaling, strategies to identify sites of Cys ox-PTM by redox proteomics have been optimized. Exploration of an available Cys redoxome dataset supported by modeled structure provided arguments for the existence of an additional non-catalytic thiol-disulfide motif, distinct from those contained in the TRX type patterns, typical of PDIAs. Further structural analysis of PDIA3 and 6 allows us to consider the possibility that this hypothesis could be extended to other members of PDI. These elements invite future studies to decipher the exact role of these non-catalytic thiol-disulfide motifs in the functions of PDIs. Strategies that would allow to validate this hypothesis are discussed.


Asunto(s)
Proteína Disulfuro Isomerasas , Compuestos de Sulfhidrilo , Proteína Disulfuro Isomerasas/metabolismo , Disulfuros/química , Tiorredoxinas/metabolismo , Proteómica , Oxidación-Reducción , Cisteína/metabolismo
12.
J Hepatol ; 56(1): 70-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21835140

RESUMEN

BACKGROUND & AIMS: Innate sensing of viral infection activates a global defense response including type I interferon (IFN) and IFN-stimulated genes (ISGs) expression. We previously reported that HCV NS3/4A protease, an essential protein in viral polyprotein processing, can abrogate antiviral signaling pathways and effectors' response when ectopically expressed in human hepatocytes by cleaving antiviral adaptor CARDIF. However, whether HCV mediates evasion of innate immunity in patients with chronic infection remains unclear. METHODS: In this study, paired liver biopsies and corresponding purified hepatocytes of chronic hepatitis C patients and controls were subjected to transcriptional analysis of selected innate immune genes and to CARDIF protein detection. RESULTS: We report that an antiviral response is largely supported by infected hepatocytes as demonstrated by upregulation of the representative antiviral genes ISG15, ISG56, and OASL as well as chemokines genes CXCL9, CXCL10, and CXCL11 measured in both HCV-derived liver biopsies and hepatocytes; that the mRNA levels of these indicator ISGs correlate inversely with HCV RNA level; and more importantly that expression of the early responsive IRF3-dependent genes type I IFNß, type III IL28A/IL29, and chemokine CCL5 are severely compromised and associated to a global decrease of CARDIF adaptor in infected hepatocytes. CONCLUSIONS: Altogether the data argue for a strong viral strategy that counteracts the host's early antiviral response of hepatocytes from chronic patients without impairing ISGs induced via classical IFN pathway.


Asunto(s)
Hepatitis C Crónica/inmunología , Inmunidad Innata , Hígado/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Quimiocinas/genética , Femenino , Hepatitis C Crónica/genética , Hepatitis C Crónica/metabolismo , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Inmunidad Innata/genética , Factores Reguladores del Interferón/genética , Interferones/genética , Hígado/metabolismo , Hígado/virología , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Regulación hacia Arriba
13.
J Virol ; 85(17): 8689-701, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21697465

RESUMEN

We recently provided evidence that the ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 (HSV-1 and -2) protect cells against tumor necrosis factor alpha- and Fas ligand-induced apoptosis by interacting with caspase 8. Double-stranded RNA (dsRNA) is a viral intermediate known to initiate innate antiviral responses. Poly(I · C), a synthetic analogue of viral dsRNA, rapidly triggers caspase 8 activation and apoptosis in HeLa cells. Here, we report that HeLa cells after HSV-1 and HSV-2 infection were quickly protected from apoptosis caused by either extracellular poly(I · C) combined with cycloheximide or transfected poly(I · C). Cells infected with the HSV-1 R1 deletion mutant ICP6Δ were killed by poly(I · C), indicating that HSV-1 R1 plays a key role in antiapoptotic responses to poly(I · C). Individually expressed HSV R1s counteracted caspase 8 activation by poly(I · C). In addition to their binding to caspase 8, HSV R1s also interacted constitutively with receptor-interacting protein 1 (RIP1) when expressed either individually or with other viral proteins during HSV infection. R1(1-834)-green fluorescent protein (GFP), an HSV-2 R1 deletion mutant protein devoid of antiapoptotic activity, did not interact with caspase 8 and RIP1, suggesting that these interactions are required for protection against poly(I · C). HSV-2 R1 inhibited the interaction between the Toll/interleukin-1 receptor domain-containing adaptor-inducing beta interferon (IFN-ß) (TRIF) and RIP1, an interaction that is essential for apoptosis triggered by extracellular poly(I · C) plus cycloheximide or TRIF overexpression. TRIF silencing reduced poly(I · C)-triggered caspase 8 activation in mock- and ICP6Δ-infected cells, confirming that TRIF is involved in poly(I · C)-induced apoptosis. Thus, by interacting with caspase 8 and RIP1, HSV R1s impair the apoptotic host defense mechanism prompted by dsRNA.


Asunto(s)
Apoptosis , Inhibidores de Caspasas , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 2/enzimología , Poli I-C/toxicidad , Ribonucleótido Reductasas/metabolismo , Células HeLa , Humanos , Proteínas de Complejo Poro Nuclear/antagonistas & inhibidores , Unión Proteica , Subunidades de Proteína/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores
14.
PLoS Pathog ; 6(6): e1000930, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20532218

RESUMEN

The innate immune response is essential to the host defense against viruses, through restriction of virus replication and coordination of the adaptive immune response. Induction of antiviral genes is a tightly regulated process initiated mainly through sensing of invading virus nucleic acids in the cytoplasm by RIG-I like helicases, RIG-I or Mda5, which transmit the signal through a common mitochondria-associated adaptor, MAVS. Although major breakthroughs have recently been made, much remains unknown about the mechanisms that translate virus recognition into antiviral genes expression. Beside the reputed detrimental role, reactive oxygen species (ROS) act as modulators of cellular signaling and gene regulation. NADPH oxidase (NOX) enzymes are a main source of deliberate cellular ROS production. Here, we found that NOX2 and ROS are required for the host cell to trigger an efficient RIG-I-mediated IRF-3 activation and downstream antiviral IFNbeta and IFIT1 gene expression. Additionally, we provide evidence that NOX2 is critical for the expression of the central mitochondria-associated adaptor MAVS. Taken together these data reveal a new facet to the regulation of the innate host defense against viruses through the identification of an unrecognized role of NOX2 and ROS.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Bronquios/inmunología , ARN Helicasas DEAD-box/metabolismo , Regulación de la Expresión Génica , Neoplasias Pulmonares/inmunología , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Western Blotting , Bronquios/citología , Bronquios/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Luciferasas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/virología , Glicoproteínas de Membrana/genética , NADPH Oxidasa 2 , NADPH Oxidasas/genética , ARN Mensajero/genética , Proteínas de Unión al ARN , Receptores Inmunológicos , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sendai/fisiología , Transducción de Señal
15.
Am J Physiol Lung Cell Mol Physiol ; 301(6): L945-55, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21908587

RESUMEN

Chronic infection and inflammation have been associated with progressive airway epithelial damage in patients with cystic fibrosis (CF). However, the effect of inflammatory products on the repair capacity of respiratory epithelia is unclear. Our objective was to study the regulation of repair mechanisms by tumor necrosis factor-α (TNF-α), a major component of inflammation in CF, in a model of mechanical wounding, in two bronchial cell lines, non-CF NuLi and CF CuFi. We observed that TNF-α enhanced the NuLi and CuFi repair rates. Chronic exposure (24-48 h) to TNF-α augmented this stimulation as well as the migration rate during repair. The cellular mechanisms involved in this stimulation were then evaluated. First, we discerned that TNF-α induced metalloproteinase-9 release, epidermal growth factor (EGF) shedding, and subsequent EGF receptor transactivation. Second, TNF-α-induced stimulation of the NuLi and CuFi wound-closure rates was prevented by GM6001 (metalloproteinase inhibitor), EGF antibody (to titrate secreted EGF), and EGF receptor tyrosine kinase inhibitors. Furthermore, we recently reported a relationship between the EGF response and K(+) channel function, both controlling bronchial repair. We now show that TNF-α enhances KvLQT1 and K(ATP) currents, while their inhibition abolishes TNF-α-induced repair stimulation. These results indicate that the effect of TNF-α is mediated, at least in part, through EGF receptor transactivation and K(+) channel stimulation. In contrast, cell proliferation during repair was slowed by TNF-α, suggesting that TNF-α could exert contrasting actions on repair mechanisms of CF airway epithelia. Finally, the stimulatory effect of TNF-α on airway wound repair was confirmed on primary airway epithelial cells, from non-CF and CF patients.


Asunto(s)
Fibrosis Quística/patología , Células Epiteliales/patología , Factor de Necrosis Tumoral alfa/farmacología , Bronquiolos/metabolismo , Bronquiolos/patología , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibrosis Quística/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/fisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Receptores ErbB/agonistas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Potenciales de la Membrana , Fosforilación , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/metabolismo , Cultivo Primario de Células , Compuestos de Amonio Cuaternario/farmacología , Activación Transcripcional , Factor de Necrosis Tumoral alfa/fisiología
16.
J Virol ; 84(14): 7267-77, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410276

RESUMEN

Respiratory syncytial virus (RSV) is the etiological agent of acute respiratory diseases, such as bronchiolitis and pneumonia. The exacerbated production of proinflammatory cytokines and chemokines in the airways in response to RSV is an important pillar in the development of these pathologies. As such, a keen understanding of the mechanisms that modulate the inflammatory response during RSV infection is of pivotal importance to developing effective treatment. The NF-kappaB transcription factor is a major regulator of proinflammatory cytokine and chemokine genes. However, RSV-mediated activation of NF-kappaB is far from characterized. We recently demonstrated that aside from the well-characterized IkappaBalpha phosphorylation and degradation, the phosphorylation of p65 at Ser536 is an essential event regulating the RSV-mediated NF-kappaB-dependent promoter transactivation. In the present study, using small interfering RNA and pharmacological inhibitors, we now demonstrate that RSV sensing by the RIG-I cytoplasmic receptor triggers a signaling cascade involving the MAVS and TRAF6 adaptors that ultimately leads to p65ser536 phosphorylation by the IKKbeta kinase. In a previous study, we highlighted a critical role of the NOX2-containing NADPH oxidase enzyme as an upstream regulator of both the IkappaBalphaSer32 and p65Ser536 in human airway epithelial cells. Here, we demonstrate that inhibition of NOX2 significantly decreases IKKbeta activation. Taken together, our data identify a new RIG-I/MAVS/TRAF6/IKKbeta/p65Ser536 pathway placed under the control of NOX2, thus characterizing a novel regulatory pathway involved in NF-kappaB-driven proinflammatory response in the context of RSV infection.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Quinasa I-kappa B/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Virus Sincitiales Respiratorios/fisiología , Serina/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Humanos , Quinasa I-kappa B/genética , Glicoproteínas de Membrana/genética , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Receptores Inmunológicos , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/genética , Factor de Transcripción ReIA/genética , Replicación Viral
17.
Med Sci (Paris) ; 27(6-7): 619-25, 2011.
Artículo en Francés | MEDLINE | ID: mdl-21718646

RESUMEN

The IKKε kinase, an atypical member of the IKK family of kinases, was recently identified as an oncogene overexpressed in over 30% of breast cancers. Besides its role in the regulation of the NF-κB transcription factor, which is well recognized for its implication in the development of breast cancers, IKKε was shown to phosphorylate numerous targets. Analysis of the phosphorylation of some of these substrates in the context of breast cancer highlighted new oncogenic signaling pathways that constitute potential targets for new therapies. Interestingly, IKKε is involved in the development of resistance to Tamoxifène. Thus, IKKε is a promising therapeutic target for newly developed breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas I-kappa B/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/etiología , Neoplasias de la Mama/genética , Transformación Celular Neoplásica , Enzima Desubiquitinante CYLD , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas I-kappa B/antagonistas & inhibidores , Proteínas I-kappa B/química , Proteínas I-kappa B/deficiencia , Ratones , Modelos Biológicos , Proteínas de Neoplasias/antagonistas & inhibidores , Oncogenes , Fosforilación , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/deficiencia , Transducción de Señal , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/metabolismo
18.
Trends Microbiol ; 29(10): 871-873, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34373192

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution is expected, given the nature of virus replication. Selection and establishment of variants in the human population depend on viral fitness and on molecular and immunological selection pressures. Here we discuss how mechanisms of replication and recombination may contribute to the emergence of current and future variants of SARS-CoV-2.


Asunto(s)
COVID-19/virología , SARS-CoV-2/genética , Genoma Viral , Humanos , Recombinación Genética , SARS-CoV-2/fisiología , Replicación Viral
19.
Sci Signal ; 14(680)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33906974

RESUMEN

Protein function is regulated by posttranslational modifications (PTMs), among which reversible oxidation of cysteine residues has emerged as a key regulatory mechanism of cellular responses. Given the redox regulation of virus-host interactions, the identification of oxidized cysteine sites in cells is essential to understand the underlying mechanisms involved. Here, we present a proteome-wide identification of reversibly oxidized cysteine sites in oxidant-treated cells using a maleimide-based bioswitch method coupled to mass spectrometry analysis. We identified 2720 unique oxidized cysteine sites within 1473 proteins with distinct abundances, locations, and functions. Oxidized cysteine sites were found in numerous signaling pathways, many relevant to virus-host interactions. We focused on the oxidation of STING, the central adaptor of the innate immune type I interferon pathway, which is stimulated in response to the detection of cytosolic DNA by cGAS. We demonstrated the reversible oxidation of Cys148 and Cys206 of STING in cells. Molecular analyses led us to establish a model in which Cys148 oxidation is constitutive, whereas Cys206 oxidation is inducible by oxidative stress or by the natural ligand of STING, 2'3'-cGAMP. Our data suggest that the oxidation of Cys206 prevented hyperactivation of STING by causing a conformational change associated with the formation of inactive polymers containing intermolecular disulfide bonds. This finding should aid the design of therapies targeting STING that are relevant to autoinflammatory disorders, immunotherapies, and vaccines.


Asunto(s)
Cisteína , Proteínas de la Membrana/metabolismo , Proteómica , Cisteína/metabolismo , Humanos , Oxidación-Reducción , Procesamiento Proteico-Postraduccional , Proteoma/genética , Proteoma/metabolismo
20.
PLoS One ; 16(7): e0253022, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34252093

RESUMEN

Influenza and RSV are human viruses responsible for outbreaks in hospitals, long-term care facilities and nursing homes. The present study assessed an air treatment using ozone at two relative humidity conditions (RHs) in order to reduce the infectivity of airborne influenza. Bovine pulmonary surfactant (BPS) and synthetic tracheal mucus (STM) were used as aerosols protectants to better reflect the human aerosol composition. Residual ozone concentration inside the aerosol chamber was also measured. RSV's sensitivity resulted in testing its resistance to aerosolization and sampling processes instead of ozone exposure. The results showed that without supplement and with STM, a reduction in influenza A infectivity of four orders of magnitude was obtained with an exposure to 1.70 ± 0.19 ppm of ozone at 76% RH for 80 min. Consequently, ozone could be considered as a virucidal disinfectant for airborne influenza A. RSV did not withstand the aerosolization and sampling processes required for the use of the experimental setup. Therefore, ozone exposure could not be performed for this virus. Nonetheless, this study provides great insight for the efficacy of ozone as an air treatment for the control of nosocomial influenza A outbreaks.


Asunto(s)
Virus de la Influenza A/efectos de los fármacos , Ozono/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Inactivación de Virus/efectos de los fármacos , Aerosoles , Microbiología del Aire , Infección Hospitalaria/prevención & control , Desinfección/métodos , Humanos , Gripe Humana/prevención & control , Ozono/administración & dosificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones por Virus Sincitial Respiratorio/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA