Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Dermatol Sci ; 98(1): 2-12, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32192826

RESUMEN

Stevens-Johnson syndrome and toxic epidermal necrolysis (SJS/TEN) are potentially life-threatening, immune-mediated adverse reactions characterized by widespread erythema, epidermal necrosis, and detachment of skin and mucosa. Efforts to grow and develop functional international collaborations and a multidisciplinary interactive network focusing on SJS/TEN as an uncommon but high burden disease will be necessary to improve efforts in prevention, early diagnosis and improved acute and long-term management. SJS/TEN 2019: From Science to Translation was a 1.5-day scientific program held April 26-27, 2019, in Vancouver, Canada. The meeting successfully engaged clinicians, researchers, and patients and conducted many productive discussions on research and patient care needs.


Asunto(s)
Necesidades y Demandas de Servicios de Salud/organización & administración , Grupo de Atención al Paciente/organización & administración , Síndrome de Stevens-Johnson/terapia , Congresos como Asunto , Carga Global de Enfermedades , Salud Global , Humanos , Cooperación Internacional , Farmacogenética/organización & administración , Sistema de Registros/estadística & datos numéricos , Síndrome de Stevens-Johnson/epidemiología , Síndrome de Stevens-Johnson/etiología , Investigación Biomédica Traslacional/organización & administración
3.
FASEB J ; 33(1): 942-952, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30080445

RESUMEN

The role for the NOD-like receptor (NLR) P3 inflammasome in enterovirus infection remains controversial. Available data suggest that the NLRP3 inflammasome is protective against enterovirus A71 but detrimental to the host during coxsackievirus B3 (CVB3) infection. CVB3 is a common etiologic agent associated with myocarditis and pancreatitis. Previous findings on the role of NLRP3 in CVB3 were based primarily on indirect evidence. Here, we utilized NLRP3 knockout mice as well as immune and cardiac cells to investigate the direct interplay between CVB3 infection and NLRP3 activation. We demonstrated that NLRP3 knockout mice exhibited more severe disease phenotype after CVB3 infection (significantly higher virus titers), increased myocardial, and pancreatic damage, as well as markedly impaired cardiac function compared to nontransgenic control mice. We further showed that NLRP3 activity was enhanced during early stage of CVB3 infection, as evidenced by increased gene expression and/or secretion of IL-1ß and caspase-1. Finally, we demonstrated that CVB3 inactivates the NLRP3 inflammasome by degrading NLRP3 and its upstream serine/threonine-protein kinase receptor-interacting protein 1/3 via the proteolytic activity of virus-encoded proteinases. Taken together, our results reveal the functional significance of NLRP3 in host antiviral immunity against CVB3 infection and the mechanisms by which CVB3 has evolved to counteract the host defense response.-Wang, C., Fung, G., Deng, H., Jagdeo, J., Mohamud, Y., Xue, Y. C., Jan, E., Hirota, J. A., Luo, H. NLRP3 deficiency exacerbates enterovirus infection in mice.


Asunto(s)
Infecciones por Enterovirus/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Animales , Caspasa 1/metabolismo , Línea Celular , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/inmunología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis
4.
J Virol ; 92(8)2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29437971

RESUMEN

Enteroviruses encode proteinases that are essential for processing of the translated viral polyprotein. In addition, viral proteinases also target host proteins to manipulate cellular processes and evade innate antiviral responses to promote replication and infection. Although some host protein substrates of enterovirus proteinases have been identified, the full repertoire of targets remains unknown. We used a novel quantitative in vitro proteomics-based approach, termed terminal amine isotopic labeling of substrates (TAILS), to identify with high confidence 72 and 34 new host protein targets of poliovirus and coxsackievirus B3 (CVB3) 3C proteinases (3Cpros) in HeLa cell and cardiomyocyte HL-1 cell lysates, respectively. We validated a subset of candidate substrates that are targets of poliovirus 3Cproin vitro including three common protein targets, phosphoribosylformylglycinamidine synthetase (PFAS), hnRNP K, and hnRNP M, of both proteinases. 3Cpro-targeted substrates were also cleaved in virus-infected cells but not noncleavable mutant proteins designed from the TAILS-identified cleavage sites. Knockdown of TAILS-identified target proteins modulated infection both negatively and positively, suggesting that cleavage by 3Cpro promotes infection. Indeed, expression of a cleavage-resistant mutant form of the endoplasmic reticulum (ER)-Golgi vesicle-tethering protein p115 decreased viral replication and yield. As the first comprehensive study to identify and validate functional enterovirus 3Cpro substrates in vivo, we conclude that N-terminomics by TAILS is an effective strategy to identify host targets of viral proteinases in a nonbiased manner.IMPORTANCE Enteroviruses are positive-strand RNA viruses that encode proteases that cleave the viral polyprotein into the individual mature viral proteins. In addition, viral proteases target host proteins in order to modulate cellular pathways and block antiviral responses in order to facilitate virus infection. Although several host protein targets have been identified, the entire list of proteins that are targeted is not known. In this study, we used a novel unbiased proteomics approach to identify ∼100 novel host targets of the enterovirus 3C protease, thus providing further insights into the network of cellular pathways that are modulated to promote virus infection.


Asunto(s)
Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/metabolismo , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano B/enzimología , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Poliovirus/enzimología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Células HeLa , Humanos , Marcaje Isotópico/métodos , Especificidad por Sustrato/fisiología
5.
J Virol ; 89(14): 7064-78, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25926642

RESUMEN

UNLABELLED: Picornavirus infection involves a dynamic interplay of host and viral protein interactions that modulates cellular processes to facilitate virus infection and evade host antiviral defenses. Here, using a proteomics-based approach known as TAILS to identify protease-generated neo-N-terminal peptides, we identify a novel target of the poliovirus 3C proteinase, the heterogeneous nuclear ribonucleoproteinM(hnRNP M), a nucleocytoplasmic shuttling RNA-binding protein that is primarily known for its role in pre-mRNA splicing. hnRNPMis cleaved in vitro by poliovirus and coxsackievirus B3 (CVB3) 3C proteinases and is targeted in poliovirus- and CVB3-infected HeLa cells and in the hearts of CVB3-infected mice. hnRNPMrelocalizes from the nucleus to the cytoplasm during poliovirus infection. Finally, depletion of hnRNPMusing small interfering RNA knockdown approaches decreases poliovirus and CVB3 infections in HeLa cells and does not affect poliovirus internal ribosome entry site translation and viral RNA stability. We propose that cleavage of and subverting the function of hnRNPMis a general strategy utilized by picornaviruses to facilitate viral infection. IMPORTANCE: Enteroviruses, a member of the picornavirus family, are RNA viruses that cause a range of diseases, including respiratory ailments, dilated cardiomyopathy, and paralysis. Although enteroviruses have been studied for several decades, the molecular basis of infection and the pathogenic mechanisms leading to disease are still poorly understood. Here, we identify hnRNPMas a novel target of a viral proteinase. We demonstrate that the virus subverts the function of hnRNPMand redirects it to a step in the viral life cycle. We propose that cleavage of hnRNPMis a general strategy that picornaviruses use to facilitate infection.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enterovirus Humano B/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo M/metabolismo , Interacciones Huésped-Patógeno , Poliovirus/fisiología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Animales , Enterovirus Humano B/enzimología , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Células HeLa , Corazón/virología , Humanos , Ratones , Miocardio/patología , Poliovirus/enzimología , Proteolisis
6.
PLoS One ; 9(6): e99214, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24919196

RESUMEN

Localized hypoxia in solid tumors activates transcriptional programs that promote the metastatic transformation of cells. Like hypoxia-inducible hyper-vascularization, loss of the retinoblastoma protein (Rb) is a trait common to advanced stages of tumor progression in many metastatic cancers. However, no link between the role of Rb and hypoxia-driven metastatic processes has been established. We demonstrated that Rb is a key mediator of the hypoxic response mediated by HIF1α/ß, the master regulator of the hypoxia response, and its essential co-activator, the thyroid hormone receptor/retinoblastoma-interacting protein (TRIP230). Furthermore, loss of Rb unmasks the full co-activation potential of TRIP230. Using small inhibitory RNA approaches in vivo, we established that Rb attenuates the normal physiological response to hypoxia by HIF1α. Notably, loss of Rb results in hypoxia-dependent biochemical changes that promote acquisition of an invasive phenotype in MCF7 breast cancer cells. In addition, Rb is present in HIF1α-ARNT/HIF1ß transcriptional complexes associated with TRIP230 as determined by co-immuno-precipitation, GST-pull-down and ChIP assays. These results demonstrate that Rb is a negative modulator of hypoxia-regulated transcription by virtue of its direct effects on the HIF1 complex. This work represents the first link between the functional ablation of Rb in tumor cells and HIF1α-dependent transcriptional activation and invasion.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Nucleares/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas del Citoesqueleto , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Células MCF-7 , Proteínas Nucleares/genética , ARN Interferente Pequeño/genética , Proteína de Retinoblastoma/genética
7.
PLoS One ; 8(11): e79546, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260247

RESUMEN

Stress granules (SGs) are dynamic cytosolic aggregates containing messenger ribonucleoproteins and target poly-adenylated (A)-mRNA. A key component of SGs is Ras-GAP SH3 domain binding protein-1 (G3BP1), which in part mediates protein-protein and protein-RNA interactions. SGs are modulated during infection by several viruses, however, the function and significance of this process remains poorly understood. In this study, we investigated the interplay between SGs and Coxsackievirus type B3 (CVB3), a member of the Picornaviridae family. Our studies demonstrated that SGs were formed early during CVB3 infection; however, G3BP1-positive SGs were actively disassembled at 5 hrs post-infection, while poly(A)-positive RNA granules persisted. Furthermore, we confirmed G3BP1 cleavage by 3C(pro) at Q325. We also demonstrated that overexpression of G3BP1-SGs negatively impacted viral replication at the RNA, protein, and viral progeny levels. Using electron microscopy techniques, we showed that G3BP1-positive SGs localized near mitochondrial surfaces. Finally, we provided evidence that the C-terminal cleavage product of G3BP1 inhibited SG formation and promoted CVB3 replication. Taken together, we conclude that CVB3 infection selectively targets G3BP1-SGs by cleaving G3BP1 to produce a dominant-negative fragment that further inhibits G3BP1-SG formation and facilitates viral replication.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por Coxsackievirus/metabolismo , Gránulos Citoplasmáticos/metabolismo , Mitocondrias/metabolismo , Western Blotting , Proteínas Portadoras/genética , Gránulos Citoplasmáticos/ultraestructura , ADN Helicasas , Técnica del Anticuerpo Fluorescente Indirecta , Células HeLa , Humanos , Hibridación in Situ , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Mitocondrias/ultraestructura , Proteínas de Unión a Poli-ADP-Ribosa , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Replicación Viral/fisiología
8.
Autophagy ; 9(10): 1591-603, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23989536

RESUMEN

The adaptor protein, sequestosome 1 (SQSTM1)/p62, plays an essential role in mediating selective autophagy. It serves as an autophagy receptor targeting ubiquitinated proteins to autophagosomes for degradation. In addition, it functions as a scaffold protein to regulate signaling pathways. Here we explored the interplay between coxsackievirus B3 (CVB3) and SQSTM1-mediated selective autophagy. We reported that SQSTM1 was cleaved at glycine 241 following CVB3 infection through the activity of viral protease 2A(pro). The resulting cleavage fragments of SQSTM1 were no longer the substrates of autophagy, and their ability to form protein aggregates was greatly decreased. Although the C-terminal truncation sustained the binding activity of SQSTM1 to microtubule-associated protein 1 light chain (LC3), it failed to interact with ubiquitinated proteins. It was also found that colocalization between the C-terminal fragment of SQSTM1 (SQSTM1-C) and LC3 and ubiquitin within the punctate structures was markedly disrupted. Moreover, we observed that SQSTM1-C retained the ability of SQSTM1 to stabilize antioxidant transcription factor NFE2L2 [nuclear factor (erythroid-derived 2)-like 2]; however, both the N-terminal fragment of SQSTM1 (SQSTM1-N) and SQSTM1-C lost the function of SQSTM1 in activating NFKB (the nuclear factor of kappa light polypeptide gene enhancer in B-cells) pathway. Collectively, our results suggest a novel model by which cleavage of SQSTM1 as a result of CVB3 infection impairs the function of SQSTM1 in selective autophagy and host defense signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia/fisiología , Transducción de Señal/fisiología , Autofagia/genética , Enterovirus Humano B/aislamiento & purificación , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Fagosomas/metabolismo , Proteína Sequestosoma-1 , Factores de Transcripción/metabolismo , Proteínas Ubiquitinadas/metabolismo
9.
FASEB J ; 27(7): 2777-87, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23572232

RESUMEN

Coxsackievirus B3 (CVB3) is a causative agent of viral myocarditis, hepatitis, pancreatitis, and meningitis in humans. The adenosine-uridine (AU)-rich element RNA binding factor 1 (AUF1) is an integral component in the regulation of gene expression. AUF1 destabilizes mRNAs and targets them for degradation by binding to AU-rich elements in the 3' untranslated region (UTR) of mRNAs. The 3'-UTR of the CVB3 genome contains canonical AU-rich sequences, raising the possibility that CVB3 RNA may also be subjected to AUF1-mediated degradation. Here, we reported that CVB3 infection led to cytoplasmic redistribution and cleavage of AUF1. These events are independent of CVB3-induced caspase activation but require viral protein production. Overexpression of viral protease 2A reproduced CVB3-induced cytoplasmic redistribution of AUF1, while in vitro cleavage assay revealed that viral protease 3C contributed to AUF1 cleavage. Furthermore, we showed that knockdown of AUF1 facilitated viral RNA, protein, and progeny production, suggesting an antiviral property for AUF1 against CVB3 infection. Finally, an immunoprecipitation study demonstrated the physical interaction between AUF1 and the 3'-UTR of CVB3, potentially targeting CVB3 genome toward degradation. Together, our results suggest that cleavage of AUF1 may be a strategy employed by CVB3 to enhance the stability of its viral genome.


Asunto(s)
Citoplasma/metabolismo , Enterovirus Humano B/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , ARN Viral/metabolismo , Regiones no Traducidas 3'/genética , Western Blotting , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Citoplasma/virología , Enterovirus Humano B/genética , Enterovirus Humano B/fisiología , Expresión Génica , Genoma Viral/genética , Células HeLa , Ribonucleoproteína Nuclear Heterogénea D0 , Ribonucleoproteína Heterogénea-Nuclear Grupo D/genética , Interacciones Huésped-Patógeno/genética , Humanos , Microscopía Confocal , Modelos Genéticos , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Unión Proteica , Transporte de Proteínas , Proteolisis , Interferencia de ARN , Estabilidad del ARN , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
10.
J Leukoc Biol ; 88(5): 925-35, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20720161

RESUMEN

We demonstrate herein that SHIP negatively regulates the proliferation, differentiation, and survival of FL-DCs from BM precursors, as shown by a more rapid appearance and higher numbers of CD11c(+) DCs from SHIP-/- cultures as well as increased survival of mature FL-DCs in the absence of Flt3L. This increased survival, which is lost with low levels of the PI3K inhibitor, LY, correlates with an enhanced constitutive activation of the Akt pathway. Interestingly, however, these SHIP-/- FL-DCs display a less-mature phenotype after TLR ligand stimulation, as far as MHCII, CD40, and CD86 are concerned. Unexpectedly, SHIP-/- FL-DCs activated with TLR ligands, which use MyD88-independent pathways, are markedly impaired in their ability to stimulate Ag-specific T cell proliferation, and SHIP-/- FL-DCs activated by TLRs, which exclusively use the MyD88-dependent pathway, are as capable as WT FL-DCs. There is also a more pronounced T(H)1 skewing by the SHIP-/- FL-DCs than by WT FL-DCs, which is consistent with our finding that SHIP-/- FL-DCs secrete higher levels of IL-12 and TNF-α in response to LPS or dsRNA than their WT counterparts. These results suggest that SHIP negatively regulates FL-DC generation but positively regulates the maturation and function of FL-DCs induced by TLRs, which operate via MyD88-independent pathways.


Asunto(s)
Células Dendríticas/fisiología , Monoéster Fosfórico Hidrolasas/genética , Animales , Western Blotting , Células Cultivadas , Cruzamientos Genéticos , Cartilla de ADN , Células Dendríticas/citología , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Inositol Polifosfato 5-Fosfatasas , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Monoéster Fosfórico Hidrolasas/deficiencia , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...