Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Cell ; 152(5): 1037-50, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452852

RESUMEN

Although somatic cell reprogramming to generate inducible pluripotent stem cells (iPSCs) is associated with profound epigenetic changes, the roles and mechanisms of epigenetic factors in this process remain poorly understood. Here, we identify Jmjd3 as a potent negative regulator of reprogramming. Jmjd3-deficient MEFs produced significantly more iPSC colonies than did wild-type cells, whereas ectopic expression of Jmjd3 markedly inhibited reprogramming. We show that the inhibitory effects of Jmjd3 are produced through both histone demethylase-dependent and -independent pathways. The latter pathway involves Jmjd3 targeting of PHF20 for ubiquitination and degradation via recruitment of an E3 ligase, Trim26. Importantly, PHF20-deficient MEFs could not be converted to fully reprogrammed iPSCs, even with knockdown of Jmjd3, Ink4a, or p21, indicating that PHF20 is required for reprogramming. Our findings demonstrate, to the best of our knowledge, a previously unrecognized role of Jmjd3 in cellular reprogramming and provide molecular insight into the mechanisms by which the Jmjd3-PHF20 axis controls this process.


Asunto(s)
Reprogramación Celular , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Cinética , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Regulación hacia Arriba
2.
PLoS Genet ; 19(3): e1010701, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36996023

RESUMEN

[This corrects the article DOI: 10.1371/journal.pgen.1004524.].

3.
Immunity ; 45(5): 1093-1107, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27793594

RESUMEN

Type I interferon (IFN) is critical for controlling pathogen infection; however, its regulatory mechanisms in plasmacytoid cells (pDCs) still remain unclear. Here, we have shown that nucleic acid sensors cGAS-, STING-, MDA5-, MAVS-, or transcription factor IRF3-deficient mice produced high amounts of type I IFN-α and IFN-ß (IFN-α/ß) in the serum and were resistant to lethal plasmodium yoelii YM infection. Robust IFN-α/ß production was abolished when gene encoding nucleic acid sensor TLR7, signaling adaptor MyD88, or transcription factor IRF7 was ablated or pDCs were depleted. Further, we identified SOCS1 as a key negative regulator to inhibit MyD88-dependent type I IFN signaling in pDCs. Finally, we have demonstrated that pDCs, cDCs, and macrophages were required for generating IFN-α/ß-induced subsequent protective immunity. Thus, our findings have identified a critical regulatory mechanism of type I IFN signaling in pDCs and stage-specific function of immune cells in generating potent immunity against lethal YM infection.


Asunto(s)
Inmunidad Adaptativa/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Malaria/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , Plasmodium yoelii , Reacción en Cadena de la Polimerasa
4.
EMBO Rep ; 24(12): e57828, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37971847

RESUMEN

Microbial products, such as lipopolysaccharide (LPS), can elicit efficient innate immune responses against invading pathogens. However, priming with LPS can induce a form of innate immune memory, termed innate immune "tolerance", which blunts subsequent NF-κB signaling. Although epigenetic and transcriptional reprogramming has been shown to play a role in innate immune memory, the involvement of post-translational regulation remains unclear. Here, we report that ubiquitin-specific protease 3 (USP3) participates in establishing "tolerance" innate immune memory through non-transcriptional feedback. Upon NF-κB signaling activation, USP3 is stabilized and exits the nucleus. The cytoplasmic USP3 specifically removes the K63-linked polyubiquitin chains on MyD88, thus negatively regulating TLR/IL1ß-induced inflammatory signaling activation. Importantly, cytoplasmic translocation is a prerequisite step for USP3 to deubiquitinate MyD88. Additionally, LPS priming could induce cytoplasmic retention and faster and stronger cytoplasmic translocation of USP3, enabling it to quickly shut down NF-κB signaling upon the second LPS challenge. This work identifies a previously unrecognized post-translational feedback loop in the MyD88-USP3 axis, which is critical for inducing normal "tolerance" innate immune memory.


Asunto(s)
Factor 88 de Diferenciación Mieloide , FN-kappa B , FN-kappa B/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Lipopolisacáridos/farmacología , Transducción de Señal , Inmunidad Innata , Tolerancia Inmunológica
5.
Cell ; 141(3): 483-96, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20434986

RESUMEN

Stringent control of the NF-kappaB and type I interferon signaling pathways is critical to effective host immune responses, yet the molecular mechanisms that negatively regulate these pathways are poorly understood. Here, we show that NLRC5, a member of the highly conserved NOD-like protein family, can inhibit the IKK complex and RIG-I/MDA5 function. NLRC5 inhibited NF-kappaB-dependent responses by interacting with IKKalpha and IKKbeta and blocking their phosphorylation. It also interacted with RIG-I and MDA5, but not with MAVS, to inhibit RLR-mediated type I interferon responses. Consistent with these observations, NLRC5-specific siRNA knockdown not only enhanced the activation of NF-kappaB and its responsive genes, TNF-alpha and IL-6, but also promoted type I interferon signaling and antiviral immunity. Our findings identify NLRC5 as a negative regulator that blocks two central components of the NF-kappaB and type I interferon signaling pathways and suggest an important role for NLRC5 in homeostatic control of innate immunity.


Asunto(s)
Inmunidad Innata , Interferón Tipo I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Animales , Clonación Molecular , ARN Helicasas DEAD-box/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Inflamación , Péptidos y Proteínas de Señalización Intracelular/química , Ligandos , Ratones , Fosforilación , Receptores Toll-Like/metabolismo
6.
Mol Cell ; 68(2): 293-307.e5, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29053956

RESUMEN

Mitochondrial antiviral signaling platform protein (MAVS) acts as a central hub for RIG-I receptor proximal signal propagation. However, key components in the assembly of the MAVS mitochondrial platform that promote RIG-I mitochondrial localization and optimal activation are still largely undefined. Employing pooled RNAi and yeast two-hybrid screenings, we report that the mitochondrial adaptor protein tripartite motif (TRIM)14 provides a docking platform for the assembly of the mitochondrial signaling complex required for maximal activation of RIG-I-mediated signaling, consisting of WHIP and protein phosphatase PPP6C. Following viral infection, the ubiquitin-binding domain in WHIP bridges RIG-I with MAVS by binding to polyUb chains of RIG-I at lysine 164. The ATPase domain in WHIP contributes to stabilization of the RIG-I-dsRNA interaction. Moreover, phosphatase PPP6C is responsible for RIG-I dephosphorylation. Together, our findings define the WHIP-TRIM14-PPP6C mitochondrial signalosome required for RIG-I-mediated innate antiviral immunity.


Asunto(s)
Proteínas Portadoras/inmunología , Proteína 58 DEAD Box/inmunología , Proteínas de Unión al ADN/inmunología , Inmunidad Innata , Mitocondrias/inmunología , Proteínas Mitocondriales/inmunología , Complejos Multiproteicos/inmunología , Fosfoproteínas Fosfatasas/inmunología , Transducción de Señal/inmunología , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Chlorocebus aethiops , Proteína 58 DEAD Box/genética , Proteínas de Unión al ADN/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mitocondrias/genética , Proteínas Mitocondriales/genética , Complejos Multiproteicos/genética , Fosfoproteínas Fosfatasas/genética , Receptores Inmunológicos , Transducción de Señal/genética , Proteínas de Motivos Tripartitos , Células Vero , Virosis/genética , Virosis/inmunología , Virus/genética , Virus/inmunología
7.
Nat Immunol ; 13(4): 387-95, 2012 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-22388039

RESUMEN

Stringent control of the type I interferon signaling pathway is important for maintaining host immune responses and homeostasis, yet the molecular mechanisms responsible for its tight regulation are still poorly understood. Here we report that the pattern-recognition receptor NLRP4 regulated the activation of type I interferon mediated by double-stranded RNA or DNA by targeting the kinase TBK1 for degradation. NLRP4 recruited the E3 ubiquitin ligase DTX4 to TBK1 for Lys48 (K48)-linked polyubiquitination at Lys670, which led to degradation of TBK1. Knockdown of either DTX4 or NLRP4 abrogated K48-linked ubiquitination and degradation of TBK1 and enhanced the phosphorylation of TBK1 and the transcription factor IRF3. Our results identify a previously unrecognized role for NLRP4 in the regulation of type I interferon signaling and provide molecular insight into the mechanisms by which NLRP4-DTX4 targets TBK1 for degradation.


Asunto(s)
Interferón Tipo I/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Línea Celular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata/inmunología , Immunoblotting , Inmunoprecipitación , Interferón Tipo I/inmunología , Fosforilación , Proteínas Serina-Treonina Quinasas/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/inmunología , Transfección , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
8.
Mol Cell ; 64(1): 105-119, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27666593

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is an essential DNA virus sensor that triggers type I interferon (IFN) signaling by producing cGAMP to initiate antiviral immunity. However, post-translational regulation of cGAS remains largely unknown. We report that K48-linked ubiquitination of cGAS is a recognition signal for p62-depdendent selective autophagic degradation. The induction of TRIM14 by type I IFN accelerates cGAS stabilization by recruiting USP14 to cleave the ubiquitin chains of cGAS at lysine (K) 414. Knockout of TRIM14 impairs herpes simplex virus type 1 (HSV-1)-triggered antiviral responses in a cGAS-dependent manner. Due to impaired type I IFN production, Trim14-/- mice are highly susceptible to lethal HSV-1 infection. Taken together, our findings reveal a positive feedback loop of cGAS signaling generated by TRIM14-USP14 and provide insights into the crosstalk between autophagy and type I IFN signaling in innate immunity.


Asunto(s)
Herpes Simple/genética , Inmunidad Innata , Nucleotidiltransferasas/genética , Procesamiento Proteico-Postraduccional , Proteína Sequestosoma-1/genética , Transactivadores/genética , Ubiquitina Tiolesterasa/genética , Animales , Autofagia/efectos de los fármacos , Retroalimentación Fisiológica , Células HEK293 , Herpes Simple/inmunología , Herpes Simple/mortalidad , Herpes Simple/virología , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/farmacología , Péptidos y Proteínas de Señalización Intracelular , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Noqueados , Nucleotidiltransferasas/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína Sequestosoma-1/inmunología , Transducción de Señal , Análisis de Supervivencia , Transactivadores/inmunología , Proteínas de Motivos Tripartitos , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/deficiencia
9.
Mol Cell ; 64(2): 267-281, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27692986

RESUMEN

TBK1 is a component of the type I interferon (IFN) signaling pathway, yet the mechanisms controlling its activity and degradation remain poorly understood. Here we report that USP38 negatively regulates type I IFN signaling by targeting the active form of TBK1 for degradation in vitro and in vivo. USP38 specifically cleaves K33-linked poly-ubiquitin chains from TBK1 at Lys670, and it allows for subsequent K48-linked ubiquitination at the same position mediated by DTX4 and TRIP. Knockdown or knockout of USP38 increases K33-linked ubiquitination, but it abrogates K48-linked ubiquitination and degradation of TBK1, thus enhancing type I IFN signaling. Our findings identify an essential role for USP38 in negatively regulating type I IFN signaling, and they provide insights into the mechanisms by which USP38 regulates TBK1 ubiquitination through the NLRP4 signalosome.


Asunto(s)
Inmunidad Innata , Interferón Tipo I/metabolismo , Macrófagos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Transducción de Señal/inmunología , Proteasas Ubiquitina-Específicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/virología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/inmunología , Factores Eucarióticos de Iniciación/metabolismo , Regulación de la Expresión Génica , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Noqueados , Fosforilación , Poliubiquitina/genética , Poliubiquitina/inmunología , Poliubiquitina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas/genética , Proteínas/inmunología , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/inmunología , Ubiquitinación , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/inmunología
10.
Proc Natl Acad Sci U S A ; 117(28): 16567-16578, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32606244

RESUMEN

Malaria infection induces complex and diverse immune responses. To elucidate the mechanisms underlying host-parasite interaction, we performed a genetic screen during early (24 h) Plasmodium yoelii infection in mice and identified a large number of interacting host and parasite genes/loci after transspecies expression quantitative trait locus (Ts-eQTL) analysis. We next investigated a host E3 ubiquitin ligase gene (March1) that was clustered with interferon (IFN)-stimulated genes (ISGs) based on the similarity of the genome-wide pattern of logarithm of the odds (LOD) scores (GPLS). March1 inhibits MAVS/STING/TRIF-induced type I IFN (IFN-I) signaling in vitro and in vivo. However, in malaria-infected hosts, deficiency of March1 reduces IFN-I production by activating inhibitors such as SOCS1, USP18, and TRIM24 and by altering immune cell populations. March1 deficiency increases CD86+DC (dendritic cell) populations and levels of IFN-γ and interleukin 10 (IL-10) at day 4 post infection, leading to improved host survival. T cell depletion reduces IFN-γ level and reverse the protective effects of March1 deficiency, which can also be achieved by antibody neutralization of IFN-γ. This study reveals functions of MARCH1 (membrane-associated ring-CH-type finger 1) in innate immune responses and provides potential avenues for activating antimalaria immunity and enhancing vaccine efficacy.


Asunto(s)
Malaria/inmunología , Plasmodium yoelii/fisiología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Malaria/enzimología , Malaria/genética , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/inmunología , Ubiquitina-Proteína Ligasas/genética
11.
EMBO J ; 37(3): 351-366, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29288164

RESUMEN

The RIG-I-like receptors (RLRs) are critical for protection against RNA virus infection, and their activities must be stringently controlled to maintain immune homeostasis. Here, we report that leucine-rich repeat containing protein 25 (LRRC25) is a key negative regulator of RLR-mediated type I interferon (IFN) signaling. Upon RNA virus infection, LRRC25 specifically binds to ISG15-associated RIG-I to promote interaction between RIG-I and the autophagic cargo receptor p62 and to mediate RIG-I degradation via selective autophagy. Depletion of either LRRC25 or ISG15 abrogates RIG-I-p62 interaction as well as the autophagic degradation of RIG-I. Collectively, our findings identify a previously unrecognized role of LRRC25 in type I IFN signaling activation by which LRRC25 acts as a secondary receptor to assist RIG-I delivery to autophagosomes for degradation in a p62-dependent manner.


Asunto(s)
Autofagia/inmunología , Proteína 58 DEAD Box/metabolismo , Interferón Tipo I/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Citocinas/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Unión Proteica/inmunología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Inmunológicos , Transducción de Señal/inmunología , Ubiquitinas/metabolismo , Virus de la Estomatitis Vesicular Indiana/inmunología
12.
Immunity ; 36(1): 43-54, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22226633

RESUMEN

Stringent control of NF-κB and mitogen-activated protein kinase (MAPK) signaling is critical during innate immune responses. TGF-ß activated kinase-1 (TAK1) is essential for NF-κB activation in T and B cells but has precisely the opposite activity in myeloid cells. Specific deletion of TAK1 (Map3k7(ΔM/ΔM)) led to development of splenomegaly and lymphomegaly associated with neutrophilia. Compared with wild-type cells, TAK1-deficient neutrophils enhanced the phosphorylation of the kinases IKK, p38, and JNK and the production of interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor-α (TNF-α), and reactive oxygen species (ROS) after lipopolysaccharide (LPS) stimulation. Map3k7(ΔM/ΔM) mice were significantly more susceptible to LPS-induced septic shock and produced higher amounts of IL-1ß, IL-6, and TNF-α in plasma than do wild-type mice. Specific ablation of p38 rescued the phenotype and functional properties of Map3k7(ΔM/ΔM) mice. Our findings identify a previously unrecognized role of TAK1 as a negative regulator of p38 and IKK activation in a cell type-specific manner.


Asunto(s)
Antígeno CD11b , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Neutrófilos/enzimología , Receptores de Quimiocina , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Antígeno CD11b/metabolismo , Proliferación Celular , Regulación hacia Abajo , Eliminación de Gen , Quinasas Quinasa Quinasa PAM/genética , Macrófagos/inmunología , Ratones , Ratones Noqueados , Modelos Inmunológicos , Neutrófilos/citología , Neutrófilos/inmunología , Fenotipo , Receptores de Quimiocina/metabolismo , Transducción de Señal
14.
EMBO J ; 35(8): 866-80, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26988033

RESUMEN

Autophagy, mediated by a number of autophagy-related (ATG) proteins, plays an important role in the bulk degradation of cellular constituents. Beclin-1 (also known as Atg6 in yeast) is a core protein essential for autophagic initiation and other biological processes. The activity of Beclin-1 is tightly regulated by multiple post-translational modifications, including ubiquitination, yet the molecular mechanism underpinning its reversible deubiquitination remains poorly defined. Here, we identified ubiquitin-specific protease 19 (USP19) as a positive regulator of autophagy, but a negative regulator of type I interferon (IFN) signaling.USP19 stabilizes Beclin-1 by removing the K11-linked ubiquitin chains of Beclin-1 at lysine 437. Moreover, we foundthat USP19 negatively regulates type IIFNsignaling pathway, by blockingRIG-I-MAVSinteraction in a Beclin-1-dependent manner. Depletion of eitherUSP19 or Beclin-1 inhibits autophagic flux and promotes type IIFNsignaling as well as cellular antiviral immunity. Our findings reveal novel dual functions of theUSP19-Beclin-1 axis by balancing autophagy and the production of type IIFNs.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Beclina-1 , Línea Celular/virología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Endopeptidasas/genética , Endopeptidasas/inmunología , Células HeLa/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Virus de la Influenza A/patogenicidad , Interferón Tipo I/metabolismo , Lisina/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Estabilidad Proteica , Receptores Inmunológicos , Transducción de Señal/fisiología , Ubiquitinación
15.
PLoS Pathog ; 14(2): e1006886, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29462185

RESUMEN

Melanoma differentiation-associated gene-5 (MDA5) recognizes distinct subsets of viruses including Encephalomyocarditis virus (EMCV) of picornavirus family, but the molecular mechanisms underlying the specificity of the viral recognition of MDA5 in immune cells remain obscure. DHX29 is an RNA helicase required for the translation of 5' structured mRNA of host and many picornaviruses (such as EMCV). We identify that DXH29 as a key RNA co-sensor, plays a significant role for specific recognition and triggering anti-EMCV immunity. We have observed that DHX29 regulates MDA5-, but not RIG-I-, mediated type I interferon signaling by preferentially interacting with structured RNAs and specifically with MDA5 for enhancing MDA5-dsRNA binding affinity. Overall, our results identify a critical role for DHX29 in innate immune response and provide molecular insights into the mechanisms by which DHX29 recognizes 5' structured EMCV RNA and interacts with MDA5 for potent type I interferon signaling and antiviral immunity.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Virus de la Encefalomiocarditis/inmunología , Inmunidad Innata/genética , Helicasa Inducida por Interferón IFIH1/fisiología , ARN Helicasas/fisiología , ARN Viral/inmunología , Animales , Infecciones por Cardiovirus/genética , Células Cultivadas , Chlorocebus aethiops , Virus de la Encefalomiocarditis/genética , Células HEK293 , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , ARN Helicasas/genética , ARN Viral/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Vero
16.
Immunity ; 34(6): 843-53, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21703539

RESUMEN

Tight regulation of NF-κB signaling is essential for innate and adaptive immune responses, yet the molecular mechanisms responsible for its negative regulation are not completely understood. Here, we report that NLRX1, a NOD-like receptor family member, negatively regulates Toll-like receptor-mediated NF-κB activation. NLRX1 interacts with TRAF6 or IκB kinase (IKK) in an activation signal-dependent fashion. Upon LPS stimulation, NLRX1 is rapidly ubiquitinated, disassociates from TRAF6, and then binds to the IKK complex, resulting in inhibition of IKKα and IKKß phosphorylation and NF-κB activation. Knockdown of NLRX1 in various cell types markedly enhances IKK phosphorylation and the production of NF-κB-responsive cytokines after LPS stimulation. We further provide in vivo evidence that NLRX1 knockdown in mice markedly enhances susceptibility to LPS-induced septic shock and plasma IL-6 level. Our study identifies a previously unrecognized role for NLRX1 in the negative regulation of TLR-induced NF-κB activation by dynamically interacting with TRAF6 and the IKK complex.


Asunto(s)
Quinasa I-kappa B/inmunología , Proteínas Mitocondriales/inmunología , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/inmunología , Receptores Toll-Like/inmunología , Animales , Línea Celular , Citocinas/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Lipopolisacáridos/inmunología , Ratones , Proteínas Mitocondriales/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación , Unión Proteica , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitinación
17.
J Neuroinflammation ; 16(1): 53, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30823925

RESUMEN

Development of central nervous system (CNS) is regulated by both intrinsic and peripheral signals. Previous studies have suggested that environmental factors affect neurological activities under both physiological and pathological conditions. Although there is anatomical separation, emerging evidence has indicated the existence of bidirectional interaction between gut microbiota, i.e., (diverse microorganisms colonizing human intestine), and brain. The cross-talk between gut microbiota and brain may have crucial impact during basic neurogenerative processes, in neurodegenerative disorders and tumors of CNS. In this review, we discuss the biological interplay between gut-brain axis, and further explore how this communication may be dysregulated in neurological diseases. Further, we highlight new insights in modification of gut microbiota composition, which may emerge as a promising therapeutic approach to treat CNS disorders.


Asunto(s)
Encéfalo/fisiología , Enfermedades del Sistema Nervioso Central/inmunología , Microbioma Gastrointestinal/inmunología , Fenómenos del Sistema Inmunológico/fisiología , Animales , Enfermedades del Sistema Nervioso Central/fisiopatología , Humanos , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/fisiopatología
18.
Biomed Microdevices ; 21(2): 39, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30949852

RESUMEN

Effective migration of dendritic cells into the lymphatic system organs is the prerequisite for a functional dendritic cell vaccine. We have previously developed a porous silicon microparticle (PSM)-based therapeutic dendritic cell vaccine (Nano-DC vaccine) where PSM serves both as the vehicle for antigen peptides and an adjuvant. Here, we analyzed parameters that determined dendritic cell uptake of PSM particles and Nano-DC vaccine accumulation in lymphatic tissues in a murine model of HER2-positive breast cancer. Our study revealed a positive correlation between sphericity of the PSM particles and their cellular uptake by circulating dendritic cells. In addition, the intravenously administered vaccines accumulated more in the spleens and inguinal lymph nodes, while the intradermally inoculated vaccines got enriched in the popliteal lymph nodes. Furthermore, mice with large tumors received more vaccines in the lymph nodes than those with small to medium size tumors. Information from this study will provide guidance on design and optimization of future therapeutic cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/metabolismo , Células Dendríticas/metabolismo , Nanomedicina , Animales , Transporte Biológico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Células Dendríticas/inmunología , Ratones , Microesferas , Fagocitos/inmunología , Silicio/química , Distribución Tisular , Carga Tumoral/inmunología
19.
PLoS Genet ; 10(7): e1004524, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25079229

RESUMEN

Histone demethylases have emerged as important players in developmental processes. Jumonji domain containing-3 (Jmjd3) has been identified as a key histone demethylase that plays a critical role in the regulation of gene expression; however, the in vivo function of Jmjd3 in embryonic development remains largely unknown. To this end, we generated Jmjd3 global and conditional knockout mice. Global deletion of Jmjd3 induces perinatal lethality associated with defective lung development. Tissue and stage-specific deletion revealed that Jmjd3 is dispensable in the later stage of embryonic lung development. Jmjd3 ablation downregulates the expression of genes critical for lung development and function, including AQP-5 and SP-B. Jmjd3-mediated alterations in gene expression are associated with locus-specific changes in the methylation status of H3K27 and H3K4. Furthermore, Jmjd3 is recruited to the SP-B promoter through interactions with the transcription factor Nkx2.1 and the epigenetic protein Brg1. Taken together, these findings demonstrate that Jmjd3 plays a stage-dependent and locus-specific role in the mouse lung development. Our study provides molecular insights into the mechanisms by which Jmjd3 regulates target gene expression in the embryonic stages of lung development.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Pulmón/metabolismo , Animales , ADN Helicasas/biosíntesis , Histona Demetilasas con Dominio de Jumonji/metabolismo , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Lisina , Ratones , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas , Proteína B Asociada a Surfactante Pulmonar/biosíntesis , Factor Nuclear Tiroideo 1 , Factores de Transcripción/biosíntesis
20.
J Labelled Comp Radiopharm ; 60(9): 385-393, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28423195

RESUMEN

The purpose of our study was to develop and evaluate a novel integrin αv ß3 -specific delivery carrier for transfection of siRNA in malignant tumors. We adopted arginine-glycine-aspartate (RGD) motif as a tissue target for specific recognition of integrin αν ß3 . A chimaeric peptide was synthesized by adding nonamer arginine residues (9-arginine [9R]) at the carboxy terminus of cyclic-RGD dimer, designated as c(RGD)2 -9R, to enable small interfering RNA (siRNA) binding. To test the applicability of the delivery carrier in vivo, c(RGD)2 -9R was labeled with radionuclide of technetium-99m. Biodistribution and γ-camera imaging studies were performed in HepG2 xenograft-bearing nude mice. As results, an optimal 10:1 molar ratio of 99m Tc-c(RGD)2 -9R to siRNA was indicated by the electrophoresis on agarose gels. 99m Tc-c(RGD)2 -9R/siRNA remained stable under a set of conditions in vitro. For in vivo study, tumor radioactivity uptake of 99m Tc-c(RGD)2 -9R/siRNA in nude mice bearing HepG2 xenografts was significantly higher than that of control probe (P < .05). The xenografts were clearly visualized at 4 hours till 6 hours noninvasively after intravenous injection of 99m Tc-c(RGD)2 -9R/siRNA, while the xenografts were not visualized at any time after injection of control probe. It was concluded that c(RGD)2 -9R could be an effective siRNA delivery carrier. Technetium-99m radiolabeled-delivery carrier represents a potential imaging strategy for RNAi-based therapy.


Asunto(s)
Portadores de Fármacos/química , Neoplasias Hepáticas/patología , Péptidos Cíclicos/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Transfección , Animales , Dimerización , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Femenino , Células Hep G2 , Humanos , Integrina alfaVbeta3/metabolismo , Marcaje Isotópico , Neoplasias Hepáticas/diagnóstico por imagen , Ratones , Ratones Desnudos , Cintigrafía , Tecnecio/química , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA