Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 17(8): 1155-68, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27312109

RESUMEN

Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are cytoplasmic sensors crucial for recognizing different species of viral RNAs, which triggers the production of type I interferons (IFNs) and inflammatory cytokines. Here, we identify RING finger protein 123 (RNF123) as a negative regulator of RIG-I and MDA5. Overexpression of RNF123 inhibits IFN-ß production triggered by Sendai virus (SeV) and encephalomyocarditis picornavirus (EMCV). Knockdown or knockout of endogenous RNF123 potentiates IFN-ß production triggered by SeV and EMCV, but not by the sensor of DNA viruses cGAS RNF123 associates with RIG-I and MDA5 in both endogenous and exogenous cases in a viral infection-inducible manner. The SPRY and coiled-coil, but not the RING, domains of RNF123 are required for the inhibitory function. RNF123 interacts with the N-terminal CARD domains of RIG-I/MDA5 and competes with the downstream adaptor VISA/MAVS/IPS-1/Cardif for RIG-I/MDA5 CARD binding. These findings suggest that RNF123 functions as a novel inhibitor of innate antiviral signaling mediated by RIG-I and MDA5, a function that does not depend on its E3 ligase activity.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Resistencia a la Enfermedad , Interacciones Huésped-Patógeno , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferón beta , Ratones , Unión Proteica , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/virología , Receptores Inmunológicos
2.
Sci Rep ; 5: 14770, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26456228

RESUMEN

Host cells orchestrate the production of IFN-ß upon detecting invading viral pathogens. Here, we report that Ring finger protein 166 (RNF166) potentiates RNA virus-triggered IFN-ß production. Overexpression of RNF166 rather than its homologous proteins RNF114, RNF125, and RNF138, enhanced Sendai virus (SeV)-induced activation of the IFN-ß promoter. Knockdown of endogenous RNF166, but not other RNFs, inhibited the IFN-ß production induced by SeV and encephalomyocarditis virus. RNF166 interacted with TRAF3 and TRAF6. SeV-induced ubiquitination of TRAF3 and TRAF6 was suppressed when endogenous RNF166 rather than RNF114/138 was knocked down. These findings suggest that RNF166 positively regulates RNA virus-triggered IFN-ß production by enhancing the ubiquitination of TRAF3 and TRAF6.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Interferón beta/genética , Factor 3 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Virus de la Encefalomiocarditis/fisiología , Células HEK293 , Células HeLa , Humanos , Interferón beta/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Virus Sendai/fisiología , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Dedos de Zinc
3.
J Virol ; 87(18): 10037-46, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23843640

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is a key sensor for recognizing nucleic acids derived from RNA viruses and triggers beta interferon (IFN-ß) production. Because of its important role in antiviral innate immunity, the activity of RIG-I must be tightly controlled. Here, we used yeast two-hybrid screening to identify a SEC14 family member, SEC14L1, as a RIG-I-associated negative regulator. Transfected SEC14L1 interacted with RIG-I, and endogenous SEC14L1 associated with RIG-I in a viral infection-inducible manner. Overexpression of SEC14L1 inhibited transcriptional activity of the IFN-ß promoter induced by RIG-I but not TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3). Knockdown of endogenous SEC14L1 in both HEK293T cells and HT1080 cells potentiated RIG-I and Sendai virus-triggered IFN-ß production as well as attenuated the replication of Newcastle disease virus. SEC14L1 interacted with the N-terminal domain of RIG-I (RIG-I caspase activation and recruitment domain [RIG-I-CARD]) and competed with VISA/MAVS/IPS-1/Cardif for RIG-I-CARD binding. Domain mapping further indicated that the PRELI-MSF1 and CRAL-TRIO domains but not the GOLD domain of SEC14L1 are required for interaction and inhibitory function. These findings suggest that SEC14L1 functions as a novel negative regulator of RIG-I-mediated antiviral signaling by preventing RIG-I interaction with the downstream effector.


Asunto(s)
Proteínas Portadoras/metabolismo , ARN Helicasas DEAD-box/inmunología , Virus de la Enfermedad de Newcastle/inmunología , ARN Viral/inmunología , Virus Sendai/inmunología , Transducción de Señal , Proteínas Portadoras/genética , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Regulación hacia Abajo , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Unión Proteica , Mapeo de Interacción de Proteínas , ARN Viral/metabolismo , Receptores Inmunológicos , Técnicas del Sistema de Dos Híbridos
4.
J Biol Chem ; 286(12): 10568-80, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21233210

RESUMEN

Retinoic acid-inducible gene I (RIG-I) recognizes RNA virus-derived nucleic acids, which leads to the production of type I interferon (IFN) in most cell types. Tight regulation of RIG-I activity is important to prevent ultra-immune responses. In this study, we identified an ARF-like (ARL) family member, ARL16, as a protein that interacts with RIG-I. Overexpression of ARL16, but not its homologous proteins ARL1 and ARF1, inhibited RIG-I-mediated downstream signaling and antiviral activity. Knockdown of endogenous ARL16 by RNAi potentiated Sendai virus-induced IFN-ß expression and vesicular stomatitis virus replication. ARL16 interacted with the C-terminal domain (CTD) of RIG-I to suppress the association between RIG-I and RNA. ARL16 (T37N) and ARL16Δ45-54, which were restricted to the GTP-disassociated form, did not interact with RIG-I and also lost the inhibitory function. Furthermore, we suggest that endogenous ARL16 changes to GTP binding status upon viral infection and binds with the RIG-I CTD to negatively control its signaling activity. These findings suggested a novel innate immune function for an ARL family member, and a GTP-dependent model in which RIG-I is regulated.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , ARN Helicasas DEAD-box/metabolismo , Guanosina Trifosfato/metabolismo , Transducción de Señal/fisiología , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/inmunología , Secuencia de Aminoácidos , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Regulación de la Expresión Génica/fisiología , Guanosina Trifosfato/genética , Guanosina Trifosfato/inmunología , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/fisiología , Interferón beta/biosíntesis , Interferón beta/genética , Interferón beta/inmunología , Ratones , Estructura Terciaria de Proteína , ARN Viral/genética , ARN Viral/inmunología , ARN Viral/metabolismo , Receptores Inmunológicos , Virus Sendai/genética , Virus Sendai/inmunología , Virus Sendai/metabolismo , Eliminación de Secuencia
5.
Cell Res ; 20(2): 211-22, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20101263

RESUMEN

As a critical apoptosis executioner, caspase-3 becomes activated and then enters into the nucleus to exert its function. However, the molecular mechanism of this nuclear entry of active caspase-3 is still unknown. In this study, we revealed that caspase-3 harbors a crm-1-independent nuclear export signal (NES) in its small subunit. Using reverse-caspase-3 as the study model, we found that the function of the NES in caspase-3 was not disturbed by the conformational changes during induced caspase-3 activation. Mutations disrupting the cleavage activity or p3-recognition site resulted in a defect in the nuclear entry of active caspase-3. We provide evidence that the p3-mediated specific cleavage activity of active caspase-3 abrogated the function of the NES. In conclusion, our results demonstrate that during caspase-3 activation, NES is constitutively present. p3-mediated specific cleavage activity abrogates the NES function in caspase-3, thus facilitating the nuclear entry of active caspase-3.


Asunto(s)
Caspasa 3/metabolismo , Núcleo Celular/metabolismo , Señales de Exportación Nuclear/fisiología , Transporte Activo de Núcleo Celular/fisiología , Secuencia de Aminoácidos , Caspasa 3/química , Caspasa 3/genética , Dominio Catalítico/genética , Análisis Mutacional de ADN , Activación Enzimática/genética , Células HeLa , Humanos , Carioferinas/metabolismo , Modelos Biológicos , Señales de Exportación Nuclear/genética , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional/fisiología , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Exportina 1
6.
Nat Immunol ; 10(12): 1300-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19881509

RESUMEN

MAVS is critical in innate antiviral immunity as the sole adaptor for RIG-I-like helicases. MAVS regulation is essential for the prevention of excessive harmful immune responses. Here we identify PCBP2 as a negative regulator in MAVS-mediated signaling. Overexpression of PCBP2 abrogated cellular responses to viral infection, whereas knockdown of PCBP2 exerted the opposite effect. PCBP2 was induced after viral infection, and its interaction with MAVS led to proteasomal degradation of MAVS. PCBP2 recruited the HECT domain-containing E3 ligase AIP4 to polyubiquitinate and degrade MAVS. MAVS was degraded after viral infection in wild-type mouse embryonic fibroblasts but remained stable in AIP4-deficient (Itch(-/-)) mouse embryonic fibroblasts, coupled with greatly exaggerated and prolonged antiviral responses. The PCBP2-AIP4 axis defines a new signaling cascade for MAVS degradation and 'fine tuning' of antiviral innate immunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Línea Celular , Humanos , Ratones , Ratones Noqueados , Virus de la Enfermedad de Newcastle/inmunología , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Virus Sendai/inmunología , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Vesiculovirus/inmunología
7.
Cell Mol Life Sci ; 66(15): 2573-84, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19521662

RESUMEN

Toll-like receptors (TLRs) act as sensors of microbial components and elicit innate immune responses. All TLR signaling pathways activate the nuclear factor-kappaB (NF-kappaB), which controls the expression of inflammatory cytokine genes. Transforming growth factor-beta-activated kinase 1 (TAK1) is a serine/threonine protein kinase that is critically involved in the activation of NF-kappaB by tumor necrosis factor (TNFalpha), interleukin-1beta (IL-1beta) and TLR ligands. In this study, we identified a novel protein, WD40 domain repeat protein 34 (WDR34) as a TAK1-interacting protein in yeast two-hybrid screens. WDR34 interacted with TAK1, TAK1-binding protein 2 (TAB2), TAK1-binding protein 3 (TAB3) and tumor necrosis factor receptor-associated factor 6 (TRAF6) in overexpression and under physiological conditions. Overexpression of WDR34 inhibited IL-1beta-, polyI:C- and lipopolysaccharide (LPS)-induced but not TNFalpha-induced NF-kappaB activation, whereas knockdown of WDR34 by a RNA-interference construct potentiated NF-kappaB activation by these ligands. Our findings suggest that WDR34 is a TAK1-associated inhibitor of the IL-1R/TLR3/TLR4-induced NF-kappaB activation pathway.


Asunto(s)
Proteínas Portadoras/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Interleucina-1/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Línea Celular , Humanos , Interleucina-1beta/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Datos de Secuencia Molecular , FN-kappa B/genética , Proteínas Nucleares/genética , Poli I-C/metabolismo , Receptores de Interleucina-1/genética , Alineación de Secuencia , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo , Distribución Tisular , Receptor Toll-Like 3/genética , Receptor Toll-Like 4/genética , Técnicas del Sistema de Dos Híbridos
8.
Sci China C Life Sci ; 52(6): 528-38, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19557330

RESUMEN

The tumor suppressor p53 is a critical component of the DNA damage response pathway that induces a set of genes responsible for cell cycle arrest, senescence, apoptosis, and DNA repair. The ataxia telangiectasia mutated protein kinase (ATM) responds to DNA-damage stimuli and signals p53 stabilization and activation, thereby facilitating transactivation of p53 inducible genes and maintainence of genome integrity. In this study, we identified a CXXC zinc finger domain containing protein termed CF5 as a critical component in the DNA damage signaling pathway. CF5 induces p53 transcriptional activity and apoptosis in cells expressing wild type p53 but not in p53-deficient cells. Knockdown of CF5 inhibits DNA damage-induced p53 activation as well as cell cycle arrest. Furthermore, CF5 physically interacts with ATM and is required for DNA damage-induced ATM phosphorylation but not its recruitment to chromatin. These findings suggest that CF5 plays a crucial role in ATM-p53 signaling in response to DNA damage.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño del ADN , Regulación de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Dedos de Zinc , Secuencia de Aminoácidos , Animales , Antineoplásicos Fitogénicos/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas Portadoras/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Etopósido/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Alineación de Secuencia , Transducción de Señal/fisiología , Factores de Transcripción , Proteínas Supresoras de Tumor/metabolismo
9.
PLoS One ; 4(6): e5760, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19484123

RESUMEN

RIG-I and MDA5 are cytoplasmic sensors that recognize different species of viral RNAs, leads to activation of the transcription factors IRF3 and NF-kappaB, which collaborate to induce type I interferons. In this study, we identified REUL, a RING-finger protein, as a specific RIG-I-interacting protein. REUL was associated with RIG-I, but not MDA5, through its PRY and SPRY domains. Overexpression of REUL potently potentiated RIG-I-, but not MDA5-mediated downstream signalling and antiviral activity. In contrast, the RING domain deletion mutant of REUL suppressed Sendai virus (SV)-induced, but not cytoplasmic polyI:C-induced activation of IFN-beta promoter. Knockdown of endogenous REUL by RNAi inhibited SV-triggered IFN-beta expression, and also increased VSV replication. Full-length RIG-I, but not the CARD domain deletion mutant of RIG-I, underwent ubiquitination induced by REUL. The Lys 154, 164, and 172 residues of the RIG-I CARD domain were critical for efficient REUL-mediated ubiquitination, as well as the ability of RIG-I to induce activation of IFN-beta promoter. These findings suggest that REUL is an E3 ubiquitin ligase of RIG-I and specifically stimulates RIG-I-mediated innate antiviral activity.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Antivirales/farmacología , Citoplasma/metabolismo , Proteína 58 DEAD Box , Humanos , Interferón beta/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Interferencia de ARN , Receptores Inmunológicos , Virus Sendai/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/fisiología
10.
Proc Natl Acad Sci U S A ; 106(21): 8653-8, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19433799

RESUMEN

We report here the identification and characterization of a protein, ERIS, an endoplasmic reticulum (ER) IFN stimulator, which is a strong type I IFN stimulator and plays a pivotal role in response to both non-self-cytosolic RNA and dsDNA. ERIS (also known as STING or MITA) resided exclusively on ER membrane. The ER retention/retrieval sequence RIR was found to be critical to retain the protein on ER membrane and to maintain its integrity. ERIS was dimerized on innate immune challenges. Coumermycin-induced ERIS dimerization led to strong and fast IFN induction, suggesting that dimerization of ERIS was critical for self-activation and subsequent downstream signaling.


Asunto(s)
Inmunidad Innata/inmunología , Proteínas de la Membrana/inmunología , Multimerización de Proteína/inmunología , Transducción de Señal/inmunología , Animales , Línea Celular , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Regulación hacia Arriba/inmunología
11.
Cell Res ; 18(11): 1096-104, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18711448

RESUMEN

Viral infection causes host cells to produce type I interferons (IFNs), which are critically involved in viral clearance. Previous studies have demonstrated that activation of the transcription factor interferon regulatory factor (IRF)3 is essential for virus-triggered induction of type I IFNs. Here we show that the E3 ubiquitin ligase RBCC protein interacting with PKC1 (RBCK1) catalyzes the ubiquitination and degradation of IRF3. Overexpression of RBCK1 negatively regulates Sendai virus-triggered induction of type I IFNs, while knockdown of RBCK1 has the opposite effect. Plaque assays consistently demonstrate that RBCK1 negatively regulates the cellular antiviral response. Furthermore, viral infection leads to induction of RBCK1 and subsequent degradation of IRF3. These findings suggest that the cellular antiviral response is controlled by a negative feedback regulatory mechanism involving RBCK1-mediated ubiquitination and degradation of IRF3.


Asunto(s)
Regulación de la Expresión Génica , Factor 3 Regulador del Interferón/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Antivirales/metabolismo , Línea Celular , Retroalimentación Fisiológica , Humanos , Interferón Tipo I/inmunología , Virus Sendai/genética , Virus Sendai/crecimiento & desarrollo , Virus Sendai/metabolismo , Transducción de Señal , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ensayo de Placa Viral
12.
Cell Res ; 18(9): 900-10, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18591963

RESUMEN

Here, we report the identification of GIDE, a mitochondrially located E3 ubiquitin ligase. GIDE contains a C-terminal RING finger domain, which is mostly conserved with those of the IAP family members and is required for GIDE's E3 ligase activity. Overexpression of GIDE induces apoptosis via a pathway involving activation of caspases, since caspase inhibitors, XIAP and an inactive mutant of caspase-9 block GIDE-induced apoptosis. GIDE also activates JNK, and blockage of JNK activation inhibits GIDE-induced release of cytochrome c and Smac as well as apoptosis, suggesting that JNK activation precedes release of cytochrome c and Smac and is required for GIDE-induced apoptosis. These pro-apoptotic properties of GIDE require its E3 ligase activity. When somewhat over- or underexpressed, GIDE slows or accelerates cell growth, respectively. These pro-apoptotic or growth inhibition effects of GIDE may account for its absence in tumor cells.


Asunto(s)
Apoptosis , Mitocondrias/enzimología , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Línea Celular , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Datos de Secuencia Molecular , Células 3T3 NIH , Inhibidores de Proteasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Factores de Transcripción/química , Factores de Transcripción/genética , Transfección , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/efectos de los fármacos
13.
Cell Signal ; 20(4): 666-74, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18234474

RESUMEN

Toll-like receptors are involved in host defense against invading pathogens. The two members of this superfamily, IL-1R and TLR4, activate overlapping NF-kappaB activate signaling pathway mediated by TRAF6. In this study, we identified syntenin as a negative regulator of IL-1R and TLR4 mediated NF-kappaB activation. Overexpressed syntenin inhibited IL-1- or LPS-, but not TNF- induced NF-kappaB activation and IL-8 mRNA expression in a dose dependent manner. Syntenin specifically interacted with TRAF6 in human 293 cells, and inhibited TRAF6 induced NF-kappaB and AP-1 activation. Syntenin also associated with TRAF6 under physiological condition, and dissociated from TRAF6 upon IL-1 stimulation. This might be due to a competition between syntenin and IRAK1, as overexpression of IRAK1 disrupted the interaction of syntenin with TRAF6, and rescued syntenin induced reduction of TRAF6 ubiquitination. Moreover, knockdown of syntenin potentiated IL-1- or LPS- triggered NF-kappaB activation and IL-8 mRNA expression. These findings suggest that syntenin is a physiological suppressor of TRAF6 and plays an inhibitory role in IL-1R- and TLR4- mediated NF-kappaB activation pathways.


Asunto(s)
Receptores de Interleucina-1/metabolismo , Transducción de Señal , Sinteninas/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 4/metabolismo , Línea Celular , Retroalimentación Fisiológica , Humanos , Interleucina-1/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-8/metabolismo , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Unión Proteica , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Interleucina-1/genética , Sinteninas/genética , Factor 6 Asociado a Receptor de TNF/genética , Receptor Toll-Like 4/genética , Factor de Transcripción AP-1/metabolismo , Transfección , Ubiquitina/metabolismo
14.
Mol Immunol ; 45(7): 1926-34, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18068231

RESUMEN

Type I interferons (IFNs) are critical mediators of the innate immune system to defend viral infection. Interferon regulatory factor (IRF) 3 and IRF7 are transcription factors that play critical roles in type I IFN production in response to viral infection. It has been shown that the protein kinase I kappaB kinase alpha (IKK alpha) is critically involved in IRF7 activation and IFN-alpha production in Toll-like receptor 7/9 (TLR7/9) signaling cascades. However, overexpression of IKK alpha does not activate the IFN-alpha promoters. Here we show that the protein kinase nuclear factor kappaB-inducing kinase (NIK) confers IKK alpha the ability to activate IRF3/7. Previous studies have shown that NIK phosphorylates IKK alpha at Ser-176 and Ser-180 residues, and mutation of each of the two residues to glutamate, which mimics its phosphorylation, caused constitutive activation of NF-kappaB. However, mutation of the two serine residues has differential effects on IKK alpha-mediated activation of IRF3/7. While IKK alpha(S176E) constitutively activates IRF3/7, IKK alpha(S180E) losses its ability to activate IRF3/7. These findings suggest that IKK alpha-mediated activation of NF-kappaB and IRF3/7 are differentially regulated by NIK, and NIK plays an important role in TLR7/9-mediated IFN-alpha production.


Asunto(s)
Quinasa I-kappa B/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Antivirales , Línea Celular , Regulación de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Interferón Tipo I/genética , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Fosforilación , Fosfoserina/metabolismo , Regiones Promotoras Genéticas/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Quinasa de Factor Nuclear kappa B
15.
Proc Natl Acad Sci U S A ; 104(28): 11706-11, 2007 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-17600090

RESUMEN

Viral infection leads to activation of the transcription factors interferon regulatory factor-3 and NF-kappaB, which collaborate to induce type I IFNs. The RNA helicase proteins RIG-I and MDA5 were recently identified as two cytoplasmic viral RNA sensors that recognize different species of viral RNAs produced during viral replication. In this study, we identified DAK, a functionally unknown dihydroacetone kinase, as a specific MDA5-interacting protein. DAK was associated with MDA5, but not RIG-I, under physiological conditions. Overexpression of DAK inhibited MDA5- but not RIG-I- or TLR3-mediated IFN-beta induction. Overexpression of DAK also inhibited cytoplasmic dsRNA and SeV-induced activation of the IFN-beta promoter, whereas knockdown of endogenous DAK by RNAi activated the IFN-beta promoter, and increased cytoplasmic dsRNA- or SeV-triggered activation of the IFN-beta promoter. In addition, overexpression of DAK inhibited MDA5- but not RIG-I-mediated antiviral activity, whereas DAK RNAi increased cytoplasmic dsRNA-triggered antiviral activity. These findings suggest that DAK is a physiological suppressor of MDA5 and specifically inhibits MDA5- but not RIG-I-mediated innate antiviral signaling.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Virus Sendai , Transducción de Señal , Virus de la Estomatitis Vesicular Indiana , Inactivación de Virus , Animales , Línea Celular , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/virología , Proteína 58 DEAD Box , Humanos , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1 , Ratones , Receptores Inmunológicos , Virus Sendai/inmunología , Transducción de Señal/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología
16.
J Biol Chem ; 282(23): 16776-82, 2007 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-17449468

RESUMEN

Inflammation is a homeostatic mechanism that limits the effects of infectious agents. Tumor necrosis factor (TNF) and interleukin (IL)-1 are two cytokines that induce inflammation through activation of the transcription factor NF-kappaB. Various studies have suggested that two homologous and structurally related adapter proteins TAB2 and TAB3 play redundant roles in TNF- and IL-1-mediated NF-kappaB activation pathways. Both TAB2 and TAB3 contain CUE, coiled-coil, and nuclear protein localization 4 zinc finger (NZF) domains. The NZF domains of TAB2/3 are critical for TAB2/3 to bind to Lys(63)-linked polyubiquitin chains of other adaptor proteins, such as receptor-interacting protein and TRAF6, which are two signaling proteins essential for TNF- and IL-1-induced NF-kappaB activation, respectively. In a search for proteins containing NZF domains conserved with those of TAB2/3, we identified RBCK1, which has been shown to act as an E3 ubiquitin ligase in iron metabolism. Overexpression of RBCK1 negatively regulates TAB2/3-mediated and TNF- and IL-1-induced NF-kappaB activation, whereas knockdown of RBCK1 by RNA interference potentiates TNF- and IL-1-induced NF-kappaB activation. RBCK1 physically interacts with TAB2/3 and facilitates degradation of TAB2/3 through a proteasome-dependent process. Taken together, our findings suggest that RBCK1 is involved in negative regulation of inflammatory signaling triggered by TNF and IL-1 through targeting TAB2/3 for degradation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interleucina-1/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/fisiología , Factores de Transcripción/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Proteínas Adaptadoras Transductoras de Señales/química , Secuencia de Aminoácidos , Secuencia de Bases , Cartilla de ADN , Humanos , Hidrólisis , Péptidos y Proteínas de Señalización Intracelular/química , Datos de Secuencia Molecular , Interferencia de ARN , Transducción de Señal/fisiología , Factores de Transcripción/química , Ubiquitina-Proteína Ligasas
17.
J Cell Sci ; 120(Pt 3): 520-30, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17251381

RESUMEN

Lamin B receptor (LBR), a chromatin and lamin binding protein in the inner nuclear membrane, has been proposed to play a vital role in nuclear envelope (NE) assembly. But the specific role for LBR in NE assembly remains unknown. In the present study, we show that overexpression of LBR causes membrane overproduction, inducing NE invagination and membrane stack formation, and that these processes require the transmembrane domain of LBR. Biochemical analysis shows that the N-terminal domain of LBR directly interacts with importin beta in a Ran sensitive and importin alpha independent manner. Using an in vitro NE assembly assay, we also demonstrate that blocking full length LBR binding sites on importin beta, by the addition of the LBR N-terminal domain inhibits the recruitment of LBR-containing vesicles to importin beta- or Ran-coated beads to form NE structure. Our results suggest that LBR is recruited to chromatin through direct interaction with importin beta to contribute to the fusion of membrane vesicles and formation of the NE.


Asunto(s)
Estructuras de la Membrana Celular/metabolismo , Cromatina/metabolismo , Membrana Nuclear/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , beta Carioferinas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Estructuras de la Membrana Celular/ultraestructura , Escherichia coli/genética , Femenino , Colorantes Fluorescentes , Eliminación de Gen , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Indoles , Microscopía Fluorescente , Datos de Secuencia Molecular , Membrana Nuclear/ultraestructura , Oocitos/química , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/genética , Transformación Genética , Xenopus , beta Carioferinas/química , Receptor de Lamina B
18.
Cell Res ; 16(7): 632-40, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16735997

RESUMEN

We reconstituted bilayer nuclear membranes, multilayer membranes, and organelles from mixtures of Xenopus laevis egg extracts and demembranated Xenopus sperm nuclei. Varying proportions of the cytosolic and vesicular fractions from the eggs were used in the reconstitution mixtures. A cytosol:vesicle ratio of 10:1 promoted reassembly of the normal bilayer nuclear membrane with inserted nuclear pore complexes around the decondensed Xenopus sperm chromatin. A cytosol:vesicle ratio of 5:1 caused decondensed and dispersed sperm chromatin to be either surrounded by or divided by unusual multilayer membrane structures with inlaid pore complexes. A cytosol:vesicle ratio of 2.5:1 promoted reconstitution of mitochondria, endoplasmic reticulum networks, and Golgi apparatus. During reassembly of the endoplasmic reticulum and Golgi apparatus, vesicular fragments of the corresponding organelles fused together and changed their shape to form flattened cisternae, which were then stacked one on top of another.


Asunto(s)
Membrana Nuclear/metabolismo , Oocitos/citología , Orgánulos/metabolismo , Extractos de Tejidos/química , Animales , Femenino , Masculino , Membrana Nuclear/ultraestructura , Oocitos/fisiología , Orgánulos/ultraestructura , Espermatozoides/química , Espermatozoides/ultraestructura , Xenopus laevis
19.
J Immunol ; 176(2): 1072-80, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16393995

RESUMEN

IFN regulatory factor-3 is a transcription factor that is required for the rapid induction of type I IFNs in the innate antiviral response. Two noncanonical IkappaB kinase (IKK) family members, IKKepsilon and TRAF family-associated NF-kappaB activator-binding kinase-1, have been shown to phosphorylate IFN regulatory factor-3 and are critically involved in virus-triggered and TLR3-mediated signaling leading to induction of type I IFNs. In yeast two-hybrid screens for potential IKKepsilon-interacting proteins, we identified Ret finger protein (RFP) as an IKKepsilon-interacting protein. Coimmunoprecipitation experiments indicated that RFP interacted with IKKepsilon and TRAF family-associated NF-kappaB activator-binding kinase-1 as well as the two canonical IKK family members, IKKbeta and IKKalpha. RFP inhibited activation of the IFN-stimulated response element and/or NF-kappaB mediated by the IKK family members and triggered by TNF, IL-1, polyinosinic-polycytidylic acid (ligand for TLR3), and viral infection. Moreover, knockdown of RFP expression by RNA interference-enhanced activation of IFN-stimulated response element and/or NF-kappaB triggered by polyinosinic-polycytidylic acid, TNF, and IL-1. Taken together, our findings suggest that RFP negatively regulates signaling involved in the antiviral response and inflammation by targeting the IKKs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Humanos , Técnicas In Vitro , Factor 3 Regulador del Interferón/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Interferencia de ARN , Virus Sendai/patogenicidad , Transducción de Señal , Técnicas del Sistema de Dos Híbridos
20.
FEBS Lett ; 580(3): 940-7, 2006 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-16427630

RESUMEN

Recently, it has been shown that really interesting new gene (RING)-in between ring finger (IBR)-RING domain-containing proteins, such as Parkin and Parc, are E3 ubiquitin ligases and are involved in regulation of apoptosis. In this report, we show that p53-inducible RING-finger protein (p53RFP), a p53-inducible E3 ubiquitin ligase, induces p53-dependent but caspase-independent apoptosis. p53RFP contains an N-terminal RING-IBR-RING domain and an uncharacterized, evolutionally highly conserved C-terminal domain. p53RFP interacts with E2 ubiquitin-conjugating enzymes UbcH7 and UbcH8 but not with UbcH5, and this interaction is mediated through the RING-IBR-RING domain of p53RFP. Interestingly, the conserved C-terminal domain of p53RFP is required and sufficient for p53RFP-mediated apoptosis, suggesting p53RFP-mediated apoptosis does not require its E3 ubiquitin ligase activity. Together with a recent report showing that p53RFP is involved in ubiquitination and degradation of p21, a p53 downstream protein promoting growth arrest and antagonizing apoptosis, our findings suggest that p53RFP is involved in switching a cell from p53-mediated growth arrest to apoptosis.


Asunto(s)
Apoptosis/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasas/genética , Caspasas/metabolismo , Línea Celular , Humanos , Ratones , Datos de Secuencia Molecular , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Homología de Secuencia de Aminoácido , Transferasas , Proteína p53 Supresora de Tumor/genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA