Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
3.
Cell Biosci ; 12(1): 36, 2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35317858

RESUMEN

BACKGROUND: SARS-CoV-2 is the causative agent of COVID-19. Overproduction and release of proinflammatory cytokines are the underlying cause of severe COVID-19. Treatment of this condition with JAK inhibitors is a double-edged sword, which might result in the suppression of proinflammatory cytokine storm and the concurrent enhancement of viral infection, since JAK signaling is essential for host antiviral response. Improving the current JAK inhibitor therapy requires a detailed molecular analysis on how SARS-CoV-2 modulates interferon (IFN)-induced activation of JAK-STAT signaling. RESULTS: In this study, we focused on the molecular mechanism by which SARS-CoV-2 NSP13 helicase suppresses IFN signaling. Expression of SARS-CoV-2 NSP13 alleviated transcriptional activity driven by type I and type II IFN-responsive enhancer elements. It also prevented nuclear translocation of STAT1 and STAT2. The suppression of NSP13 on IFN signaling occurred at the step of STAT1 phosphorylation. Nucleic acid binding-defective mutant K345A K347A and NTPase-deficient mutant E375A of NSP13 were found to have largely lost the ability to suppress IFN-ß-induced STAT1 phosphorylation and transcriptional activation, indicating the requirement of the helicase activity for NSP13-mediated inhibition of STAT1 phosphorylation. NSP13 did not interact with JAK1 nor prevent STAT1-JAK1 complex formation. Mechanistically, NSP13 interacted with STAT1 to prevent JAK1 kinase from phosphorylating STAT1. CONCLUSION: SARS-CoV-2 NSP13 helicase broadly suppresses IFN signaling by targeting JAK1 phosphorylation of STAT1.

4.
Int J Biol Sci ; 17(6): 1547-1554, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33907518

RESUMEN

Suppression of type I interferon (IFN) response is one pathological outcome of the infection of highly pathogenic human coronaviruses. To effect this, severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2 encode multiple IFN antagonists. In this study, we reported on the IFN antagonism of SARS-CoV-2 main protease NSP5. NSP5 proteins of both SARS-CoV and SARS-CoV-2 counteracted Sendai virus-induced IFN production. NSP5 variants G15S and K90R commonly seen in circulating strains of SARS-CoV-2 retained the IFN-antagonizing property. The suppressive effect of NSP5 on IFN-ß gene transcription induced by RIG-I, MAVS, TBK1 and IKKϵ suggested that NSP5 likely acts at a step downstream of IRF3 phosphorylation in the cytoplasm. NSP5 did not influence steady-state expression or phosphorylation of IRF3, suggesting that IRF3, regardless of its phosphorylation state, might not be the substrate of NSP5 protease. However, nuclear translocation of phosphorylated IRF3 was severely compromised in NSP5-expressing cells. Taken together, our work revealed a new mechanism by which NSP5 proteins encoded by SARS-CoV and SARS-CoV-2 antagonize IFN production by retaining phosphorylated IRF3 in the cytoplasm. Our findings have implications in rational design and development of antiviral agents against SARS-CoV-2.


Asunto(s)
Núcleo Celular/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , SARS-CoV-2/enzimología , Animales , COVID-19/virología , Chlorocebus aethiops , Humanos , Fosforilación , Transporte de Proteínas , Células Vero
5.
Cell ; 184(8): 2212-2228.e12, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33713620

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can cause acute respiratory disease and multiorgan failure. Finding human host factors that are essential for SARS-CoV-2 infection could facilitate the formulation of treatment strategies. Using a human kidney cell line-HK-2-that is highly susceptible to SARS-CoV-2, we performed a genome-wide RNAi screen and identified virus dependency factors (VDFs), which play regulatory roles in biological pathways linked to clinical manifestations of SARS-CoV-2 infection. We found a role for a secretory form of SARS-CoV-2 receptor, soluble angiotensin converting enzyme 2 (sACE2), in SARS-CoV-2 infection. Further investigation revealed that SARS-CoV-2 exploits receptor-mediated endocytosis through interaction between its spike with sACE2 or sACE2-vasopressin via AT1 or AVPR1B, respectively. Our identification of VDFs and the regulatory effect of sACE2 on SARS-CoV-2 infection shed insight into pathogenesis and cell entry mechanisms of SARS-CoV-2 as well as potential treatment strategies for COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/inmunología , Interacciones Microbiota-Huesped/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vasopresinas/inmunología , Internalización del Virus , COVID-19/inmunología , COVID-19/virología , Línea Celular , Humanos , Unión Proteica
6.
Cancer Cell Int ; 20: 403, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32855620

RESUMEN

BACKGROUND: Long non-coding RNA (lncRNA) BM742401 is a tumor suppressor in gastric cancer and chronic lymphocytic leukemia. As the promoter and coding region of BM742401 are fully embedded in a CpG island, we hypothesized that BM742401 is a tumor suppressor lncRNA epigenetically silenced by promoter DNA methylation in multiple myeloma. METHODS: Methylation-specific PCR and quantitative bisulfite pyrosequencing were performed to detect the methylation of BM742401 in normal plasma cells, myeloma cell lines and primary myeloma samples. The expression of BM742401 was measured by qRT-PCR. The function of BM742401 in multiple myeloma cells was analyzed by lentivirus transduction followed by migration assay. RESULTS: BM742401 methylation was detected in 10 (66.7%) myeloma cell lines but not normal plasma cells, and inversely correlated with expression of BM742401. In primary samples, BM742401 methylation was detected in 3 (12.5%) monoclonal gammopathy of undetermined significance, 9 (15.8%) myeloma at diagnosis and 8 (17.0%) myeloma at relapse/progression. Moreover, BM742401 methylation at diagnosis was associated with inferior overall survival (median OS: 25 vs. 39 months; P = 0.0496). In myeloma cell line JJN-3, stable overexpression of BM742401 by lentivirus transduction resulted in reduced cell migration (P = 0.0001) but not impacting cell death or proliferation. CONCLUSIONS: This is the first report of tumor-specific methylation-mediated silencing of BM742401 in myeloma, which is likely an early event in myelomagenesis with adverse impact on overall survival. Moreover, BM742401 is a tumor suppressor lncRNA by inhibiting myeloma cell migration, hence implicated in myeloma plasma cell homing, metastasis and disease progression.

7.
J Immunol ; 205(6): 1564-1579, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32747502

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic human coronavirus causing severe disease and mortality. MERS-CoV infection failed to elicit robust IFN response, suggesting that the virus might have evolved strategies to evade host innate immune surveillance. In this study, we identified and characterized type I IFN antagonism of MERS-CoV open reading frame (ORF) 8b accessory protein. ORF8b was abundantly expressed in MERS-CoV-infected Huh-7 cells. When ectopically expressed, ORF8b inhibited IRF3-mediated IFN-ß expression induced by Sendai virus and poly(I:C). ORF8b was found to act at a step upstream of IRF3 to impede the interaction between IRF3 kinase IKKε and chaperone protein HSP70, which is required for the activation of IKKε and IRF3. An infection study using recombinant wild-type and ORF8b-deficient MERS-CoV further confirmed the suppressive role of ORF8b in type I IFN induction and its disruption of the colocalization of HSP70 with IKKε. Ectopic expression of HSP70 relieved suppression of IFN-ß expression by ORF8b in an IKKε-dependent manner. Enhancement of IFN-ß induction in cells infected with ORF8b-deficient virus was erased when HSP70 was depleted. Taken together, HSP70 chaperone is important for IKKε activation, and MERS-CoV ORF8b suppresses type I IFN expression by competing with IKKε for interaction with HSP70.


Asunto(s)
Activación Enzimática/inmunología , Quinasa I-kappa B/inmunología , Interferón Tipo I/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Proteínas Virales/inmunología , Betacoronavirus , COVID-19 , Línea Celular , Infecciones por Coronavirus , Proteínas HSP70 de Choque Térmico/inmunología , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Interferón Tipo I/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Pandemias , Neumonía Viral , SARS-CoV-2 , Proteínas Virales/metabolismo
8.
FASEB J ; 33(8): 8865-8877, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31034780

RESUMEN

Severe acute respiratory syndrome coronavirus (SARS-CoV) is capable of inducing a storm of proinflammatory cytokines. In this study, we show that the SARS-CoV open reading frame 3a (ORF3a) accessory protein activates the NLRP3 inflammasome by promoting TNF receptor-associated factor 3 (TRAF3)-mediated ubiquitination of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). SARS-CoV and its ORF3a protein were found to be potent activators of pro-IL-1ß gene transcription and protein maturation, the 2 signals required for activation of the NLRP3 inflammasome. ORF3a induced pro-IL-1ß transcription through activation of NF-κB, which was mediated by TRAF3-dependent ubiquitination and processing of p105. ORF3a-induced elevation of IL-1ß secretion was independent of its ion channel activity or absent in melanoma 2 but required NLRP3, ASC, and TRAF3. ORF3a interacted with TRAF3 and ASC, colocalized with them in discrete punctate structures in the cytoplasm, and facilitated ASC speck formation. TRAF3-dependent K63-linked ubiquitination of ASC was more pronounced in SARS-CoV-infected cells or when ORF3a was expressed. Taken together, our findings reveal a new mechanism by which SARS-CoV ORF3a protein activates NF-κB and the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of p105 and ASC.-Siu, K.-L., Yuen, K.-S., Castaño-Rodriguez, C., Ye, Z.-W., Yeung, M.-L., Fung, S.-Y., Yuan, S., Chan, C.-P., Yuen, K.-Y., Enjuanes, L., Jin, D.-Y. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ubiquitinación , Proteínas Estructurales Virales/metabolismo , Células A549 , Animales , Chlorocebus aethiops , Células HEK293 , Humanos , Inflamasomas/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Células Vero
9.
EMBO Rep ; 19(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30104205

RESUMEN

Mouse p202 is a disease locus for lupus and a dominant-negative inhibitor of AIM2 inflammasome activation. A human homolog of p202 has not been identified so far. Here, we report a novel transcript isoform of human IFI16-designated IFI16-ß, which has a domain architecture similar to that of mouse p202. Like p202, IFI16-ß contains two HIN domains, but lacks the pyrin domain. IFI16-ß is ubiquitously expressed in various human tissues and cells. Its mRNA levels are also elevated in leukocytes of patients with lupus, virus-infected cells, and cells treated with interferon-ß or phorbol ester. IFI16-ß co-localizes with AIM2 in the cytoplasm, whereas IFI16-α is predominantly found in the nucleus. IFI16-ß interacts with AIM2 to impede the formation of a functional AIM2-ASC complex. In addition, IFI16-ß sequesters cytoplasmic dsDNA and renders it unavailable for AIM2 sensing. Enforced expression of IFI16-ß inhibits the activation of AIM2 inflammasome, whereas knockdown of IFI16-ß augments interleukin-1ß secretion triggered by dsDNA but not dsRNA Thus, cytoplasm-localized IFI16-ß is functionally equivalent to mouse p202 that exerts an inhibitory effect on AIM2 inflammasome.


Asunto(s)
Proteínas de Unión al ADN/genética , Inflamasomas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animales , Núcleo Celular/genética , ADN/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-1beta/genética , Ratones , Isoformas de Proteínas/genética , ARN Bicatenario/genética , ARN Mensajero/genética
10.
Emerg Microbes Infect ; 5: e39, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27094905

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) infection has claimed hundreds of lives and has become a global threat since its emergence in Saudi Arabia in 2012. The ability of MERS-CoV to evade the host innate antiviral response may contribute to its severe pathogenesis. Many MERS-CoV-encoded proteins were identified to have interferon (IFN)-antagonizing properties, which correlates well with the reduced IFN levels observed in infected patients and ex vivo models. In this study, we fully characterized the IFN-antagonizing property of the MERS-CoV M protein. Expression of MERS-CoV M protein suppressed type I IFN expression in response to Sendai virus infection or poly(I:C) induction. This suppressive effect was found to be specific for the activation of IFN regulatory factor 3 (IRF3) but not nuclear factor-κB. MERS-CoV M protein interacted with TRAF3 and disrupted TRAF3-TBK1 association leading to reduced IRF3 activation. M proteins from MERS-CoV and SARS-CoV have three highly similar conserved N-terminal transmembrane domains and a C-terminal region. Using chimeric and truncation mutants, the N-terminal transmembrane domains of the MERS-CoV M protein were found to be sufficient for its inhibitory effect on IFN expression, whereas the C-terminal domain was unable to induce this suppression. Collectively, our findings suggest a common and conserved mechanism through which highly pathogenic MERS-CoV and SARS-CoV harness their M proteins to suppress type I IFN expression at the level of TBK1-dependent phosphorylation and activation of IRF3 resulting in evasion of the host innate antiviral response.


Asunto(s)
Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Fosfotransferasas , Proteínas Serina-Treonina Quinasas/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Proteínas de la Matriz Viral/fisiología , Proteínas M de Coronavirus , Proteína 58 DEAD Box/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Evasión Inmune , Inmunidad Innata , Factor 3 Regulador del Interferón/inmunología , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Arabia Saudita , Virus Sendai/genética , Virus Sendai/inmunología , Alineación de Secuencia , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/inmunología , Proteínas de la Matriz Viral/genética
11.
Int J Biochem Cell Biol ; 61: 53-62, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25681686

RESUMEN

Although expression quantitative trait locus, eQTL, serves as an explicit indicator of gene-gene associations, challenges remain to disentangle the mechanisms by which genetic variations alter gene expression. Here we combined eQTL and molecular analyses to identify an association between two seemingly non-associated genes in brain expression data from BXD inbred mice, namely Ptpn21 and Nrg3. Using biotinylated receptor tracking and immunoprecipitation analyses, we determined that PTPN21 de-phosphorylates the upstream receptor tyrosine kinase ErbB4 leading to the up-regulation of its downstream signaling. Conversely, kinase-dead ErbB4 (K751R) or phosphatase-dead PTPN21 (C1108S) mutants impede PTPN21-dependent signaling. Furthermore, PTPN21 also induced Elk-1 activation in embryonic cortical neurons and a novel Elk-1 binding motif was identified in a region located 1919bp upstream of the NRG3 initiation codon. This enables PTPN21 to promote NRG3 expression through Elk-1, which provides a biochemical mechanism for the PTPN21-NRG3 association identified by eQTL. Biologically, PTPN21 positively influences cortical neuronal survival and, similar to Elk-1, it also enhances neuritic length. Our combined approaches show for the first time, a link between NRG3 and PTPN21 within a signaling cascade. This may explain why these two seemingly unrelated genes have previously been identified as risk genes for schizophrenia.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuritas/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Receptor ErbB-4/metabolismo , Animales , Supervivencia Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Neurregulinas/biosíntesis , Neurregulinas/genética , Neurregulinas/metabolismo , Neuronas/citología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Sitios de Carácter Cuantitativo , Receptor ErbB-4/genética , Transducción de Señal , Transfección
12.
Sci Rep ; 5: 7897, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600293

RESUMEN

Ferredoxins are iron-sulfur proteins that play important roles in electron transport and redox homeostasis. Yeast Apd1p is a novel member of the family of thioredoxin-like ferredoxins. In this study, we characterized the hydroxyurea (HU)-hypersensitive phenotype of apd1Δ cells. HU is an inhibitor of DNA synthesis, a cellular stressor and an anticancer agent. Although the loss of APD1 did not influence cell proliferation or cell cycle progression, it resulted in HU sensitivity. This sensitivity was reverted in the presence of antioxidant N-acetyl-cysteine, implicating a role for intracellular redox. Mutation of the iron-binding motifs in Apd1p abrogated its ability to rescue HU sensitivity in apd1Δ cells. The iron-binding activity of Apd1p was verified by a color assay. By mass spectrometry two irons were found to be incorporated into one Apd1p protein molecule. Surprisingly, ribonucleotide reductase genes were not induced in apd1Δ cells and the HU sensitivity was unaffected when dNTP production was boosted. A suppressor screen was performed and the expression of stress-regulated transcription factor Yap1p was found to effectively rescue the HU sensitivity in apd1Δ cells. Taken together, our work identified Apd1p as a new ferredoxin which serves critical roles in cellular defense against HU.


Asunto(s)
Replicación del ADN/genética , Ferredoxinas/genética , Hidroxiurea/farmacología , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Acetilcisteína/química , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Replicación del ADN/efectos de los fármacos , Ferredoxinas/química , Hierro/química , Oxidación-Reducción , Fenotipo , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo
13.
Cell Mol Immunol ; 11(2): 141-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24509444

RESUMEN

Coronaviruses have developed various measures to evade innate immunity. We have previously shown that severe acute respiratory syndrome (SARS) coronavirus M protein suppresses type I interferon (IFN) production by impeding the formation of functional TRAF3-containing complex. In this study, we demonstrate that the IFN-antagonizing activity is specific to SARS coronavirus M protein and is mediated through its first transmembrane domain (TM1) located at the N terminus. M protein from human coronavirus HKU1 does not inhibit IFN production. Whereas N-linked glycosylation of SARS coronavirus M protein has no influence on IFN antagonism, TM1 is indispensable for the suppression of IFN production. TM1 targets SARS coronavirus M protein and heterologous proteins to the Golgi apparatus, yet Golgi localization is required but not sufficient for IFN antagonism. Mechanistically, TM1 is capable of binding with RIG-I, TRAF3, TBK1 and IKKε, and preventing the interaction of TRAF3 with its downstream effectors. Our work defines the molecular architecture of SARS coronavirus M protein required for suppression of innate antiviral response.


Asunto(s)
Coronavirus/inmunología , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Síndrome Respiratorio Agudo Grave/inmunología , Proteínas de la Matriz Viral/metabolismo , Proteínas M de Coronavirus , Células HEK293 , Células HeLa , Humanos , Quinasa I-kappa B/metabolismo , Evasión Inmune , Inmunidad Innata , Terapia de Inmunosupresión , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Mutación/genética , Unión Proteica/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Señales de Clasificación de Proteína/genética , Estructura Terciaria de Proteína/genética , Síndrome Respiratorio Agudo Grave/virología , Factor 3 Asociado a Receptor de TNF/metabolismo , Proteínas de la Matriz Viral/genética
14.
J Virol ; 88(9): 4866-76, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24522921

RESUMEN

UNLABELLED: Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging pathogen that causes severe disease in human. MERS-CoV is closely related to bat coronaviruses HKU4 and HKU5. Evasion of the innate antiviral response might contribute significantly to MERS-CoV pathogenesis, but the mechanism is poorly understood. In this study, we characterized MERS-CoV 4a protein as a novel immunosuppressive factor that antagonizes type I interferon production. MERS-CoV 4a protein contains a double-stranded RNA-binding domain capable of interacting with poly(I · C). Expression of MERS-CoV 4a protein suppressed the interferon production induced by poly(I · C) or Sendai virus. RNA binding of MERS-CoV 4a protein was required for IFN antagonism, a property shared by 4a protein of bat coronavirus HKU5 but not by the counterpart in bat coronavirus HKU4. MERS-CoV 4a protein interacted with PACT in an RNA-dependent manner but not with RIG-I or MDA5. It inhibited PACT-induced activation of RIG-I and MDA5 but did not affect the activity of downstream effectors such as RIG-I, MDA5, MAVS, TBK1, and IRF3. Taken together, our findings suggest a new mechanism through which MERS-CoV employs a viral double-stranded RNA-binding protein to circumvent the innate antiviral response by perturbing the function of cellular double-stranded RNA-binding protein PACT. PACT targeting might be a common strategy used by different viruses, including Ebola virus and herpes simplex virus 1, to counteract innate immunity. IMPORTANCE: Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging and highly lethal human pathogen. Why MERS-CoV causes severe disease in human is unclear, and one possibility is that MERS-CoV is particularly efficient in counteracting host immunity, including the sensing of virus invasion. It will therefore be critical to clarify how MERS-CoV cripples the host proteins that sense viruses and to compare MERS-CoV with its ancestral viruses in bats in the counteraction of virus sensing. This work not only provides a new understanding of the abilities of MERS-CoV and closely related bat viruses to subvert virus sensing but also might prove useful in revealing new strategies for the development of vaccines and antivirals.


Asunto(s)
Coronavirus/inmunología , ARN Helicasas DEAD-box/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Interferones/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Proteína 58 DEAD Box , Humanos , Evasión Inmune , Helicasa Inducida por Interferón IFIH1 , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores Inmunológicos
15.
Cell Biosci ; 4(1): 3, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24410900

RESUMEN

BACKGROUND: Whereas severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) is associated with severe disease, human coronavirus HKU1 (HCoV-HKU1) commonly circulates in the human populations causing generally milder illness. Spike (S) protein of SARS-CoV activates the unfolded protein response (UPR). It is not understood whether HCoV-HKU1 S protein has similar activity. In addition, the UPR-activating domain in SARS-CoV S protein remains to be identified. RESULTS: In this study we compared S proteins of SARS-CoV and HCoV-HKU1 for their ability to activate the UPR. Both S proteins were found in the endoplasmic reticulum. Transmembrane serine protease TMPRSS2 catalyzed the cleavage of SARS-CoV S protein, but not the counterpart in HCoV-HKU1. Both S proteins showed a similar pattern of UPR-activating activity. Through PERK kinase they activated the transcription of UPR effector genes such as Grp78, Grp94 and CHOP. N-linked glycosylation was not required for the activation of the UPR by S proteins. S1 subunit of SARS-CoV but not its counterpart in HCoV-HKU1 was capable of activating the UPR. A central region (amino acids 201-400) of SARS-CoV S1 was required for this activity. CONCLUSIONS: SARS-CoV and HCoV-HKU1 S proteins use distinct UPR-activating domains to exert the same modulatory effects on UPR signaling.

16.
Vaccine ; 30(16): 2605-10, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22342549

RESUMEN

Recipients of influenza A (H1N1) vaccine in 1976 had an increased risk for the neurologic disorder Guillain-Barré syndrome (GBS). Anti-ganglioside antibodies, which might be associated with the development of GBS, were previously reported to be induced in mice immunized with an H1N1 vaccine of 1976 or another influenza vaccine. In this study we analyzed anti-ganglioside antibodies in human subjects infected with or vaccinated against 2009 pandemic H1N1, including eight patients diagnosed to have post-vaccination GBS. Antibodies against GM1 or another ganglioside were not detected in any subject or in vaccinated mice. Our results did not support the induction of anti-ganglioside antibodies by influenza viruses or vaccines.


Asunto(s)
Autoanticuerpos/sangre , Gangliósidos/inmunología , Síndrome de Guillain-Barré/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Pandemias , Animales , Autoanticuerpos/inmunología , Femenino , Gangliósidos/sangre , Síndrome de Guillain-Barré/epidemiología , Síndrome de Guillain-Barré/etiología , Pruebas de Inhibición de Hemaglutinación , Hong Kong/epidemiología , Humanos , Vacunas contra la Influenza/efectos adversos , Gripe Humana/epidemiología , Gripe Humana/prevención & control , Ratones , Ratones Endogámicos C57BL , Vacunación , Vacunas de Productos Inactivados
17.
J Immunol ; 187(12): 6473-82, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22079989

RESUMEN

TNFR-associated factor (TRAF) 3 is an important adaptor that transmits upstream activation signals to protein kinases that phosphorylate transcription factors to induce the production of type I IFNs, the important effectors in innate antiviral immune response. MIP-T3 interacts specifically with TRAF3, but its function in innate IFN response remains unclear. In this study, we demonstrated a negative regulatory role of MIP-T3 in type I IFN production. Overexpression of MIP-T3 inhibited RIG-I-, MDA5-, VISA-, TBK1-, and IKKε-induced transcriptional activity mediated by IFN-stimulated response elements and IFN-ß promoter. MIP-T3 interacted with TRAF3 and perturbed in a dose-dependent manner the formation of functional complexes of TRAF3 with VISA, TBK1, IKKε, and IFN regulatory factor 3. Consistent with this finding, retinoic acid-inducible gene I- and TBK1-induced phosphorylation of IFN regulatory factor 3 was significantly diminished when MIP-T3 was overexpressed. Depletion of MIP-T3 facilitated Sendai virus-induced activation of IFN production and attenuated the replication of vesicular stomatitis virus. In addition, MIP-T3 was found to be dissociated from TRAF3 during the course of Sendai virus infection. Our findings suggest that MIP-T3 functions as a negative regulator of innate IFN response by preventing TRAF3 from forming protein complexes with critical downstream transducers and effectors.


Asunto(s)
Regulación hacia Abajo/inmunología , Inmunidad Innata , Interferón Tipo I/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/fisiología , Animales , Chlorocebus aethiops , Regulación hacia Abajo/genética , Células HEK293 , Humanos , Inmunidad Innata/genética , Interferón Tipo I/biosíntesis , Interferón beta/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Regiones Promotoras Genéticas/inmunología , Unión Proteica/inmunología , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/virología , Virus Sendai/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor 3 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 3 Asociado a Receptor de TNF/metabolismo , Células Vero
18.
Cell Host Microbe ; 9(4): 299-309, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21501829

RESUMEN

RIG-I, a virus sensor that triggers innate antiviral response, is a DExD/H box RNA helicase bearing structural similarity with Dicer, an RNase III-type nuclease that mediates RNA interference. Dicer requires double-stranded RNA-binding protein partners, such as PACT, for optimal activity. Here we show that PACT physically binds to the C-terminal repression domain of RIG-I and potently stimulates RIG-I-induced type I interferon production. PACT potentiates the activation of RIG-I by poly(I:C) of intermediate length. PACT also cooperates with RIG-I to sustain the activation of antiviral defense. Depletion of PACT substantially attenuates viral induction of interferons. The activation of RIG-I by PACT does not require double-stranded RNA-dependent protein kinase or Dicer, but is mediated by a direct interaction that leads to stimulation of its ATPase activity. Our findings reveal PACT as an important component in initiating and sustaining the RIG-I-dependent antiviral response.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Proteínas de Unión al ARN/metabolismo , Virus/inmunología , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/biosíntesis , Ratones , Plásmidos/genética , Poli I-C/inmunología , Interferencia de ARN , ARN Bicatenario/metabolismo , ARN Interferente Pequeño , Receptores Inmunológicos , Transducción de Señal/fisiología , Células Vero , Virus/metabolismo , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/fisiología
19.
Proc Natl Acad Sci U S A ; 108(7): 2729-34, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282621

RESUMEN

Hydrogen peroxide is thought to regulate cellular processes by direct oxidation of numerous cellular proteins, whereas antioxidants, most notably thiol peroxidases, are thought to reduce peroxides and inhibit H(2)O(2) response. However, thiol peroxidases have also been implicated in activation of transcription factors and signaling. It remains unclear if these enzymes stimulate or inhibit redox regulation and whether this regulation is widespread or limited to a few cellular components. Herein, we found that Saccharomyces cerevisiae cells lacking all eight thiol peroxidases were viable and withstood redox stresses. They transcriptionally responded to various redox treatments, but were unable to activate and repress gene expression in response to H(2)O(2). Further studies involving redox transcription factors suggested that thiol peroxidases are major regulators of global gene expression in response to H(2)O(2). The data suggest that thiol peroxidases sense and transfer oxidative signals to the signaling proteins and regulate transcription, whereas a direct interaction between H(2)O(2) and other cellular proteins plays a secondary role.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Peroxidasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Transducción de Señal/efectos de los fármacos , Secuencia de Bases , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/genética , Peroxidasas/deficiencia , Fenotipo , Proteínas Ribosómicas/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Análisis de Secuencia de ADN , Transducción de Señal/fisiología
20.
PLoS Genet ; 5(10): e1000697, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19851444

RESUMEN

Peroxiredoxins are a family of antioxidant enzymes critically involved in cellular defense and signaling. Particularly, yeast peroxiredoxin Tsa1p is thought to play a role in the maintenance of genome integrity, but the underlying mechanism is not understood. In this study, we took a genetic approach to investigate the cause of genome instability in tsa1Delta cells. Strong genetic interactions of TSA1 with DNA damage checkpoint components DUN1, SML1, and CRT1 were found when mutant cells were analyzed for either sensitivity to DNA damage or rate of spontaneous base substitutions. An elevation in intracellular dNTP production was observed in tsa1Delta cells. This was associated with constitutive activation of the DNA damage checkpoint as indicated by phosphorylation of Rad9/Rad53p, reduced steady-state amount of Sml1p, and induction of RNR and HUG1 genes. In addition, defects in the DNA damage checkpoint did not modulate intracellular level of reactive oxygen species, but suppressed the mutator phenotype of tsa1Delta cells. On the contrary, overexpression of RNR1 exacerbated this phenotype by increasing dNTP levels. Taken together, our findings uncover a new role of TSA1 in preventing the overproduction of dNTPs, which is a root cause of genome instability.


Asunto(s)
Daño del ADN , Inestabilidad Genómica , Nucleótidos/metabolismo , Peroxidasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Reparación del ADN , Mutación , Peroxidasas/genética , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...