Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Pathog ; 9(4): e1003298, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23633948

RESUMEN

The RIG-I like receptor pathway is stimulated during RNA virus infection by interaction between cytosolic RIG-I and viral RNA structures that contain short hairpin dsRNA and 5' triphosphate (5'ppp) terminal structure. In the present study, an RNA agonist of RIG-I was synthesized in vitro and shown to stimulate RIG-I-dependent antiviral responses at concentrations in the picomolar range. In human lung epithelial A549 cells, 5'pppRNA specifically stimulated multiple parameters of the innate antiviral response, including IRF3, IRF7 and STAT1 activation, and induction of inflammatory and interferon stimulated genes - hallmarks of a fully functional antiviral response. Evaluation of the magnitude and duration of gene expression by transcriptional profiling identified a robust, sustained and diversified antiviral and inflammatory response characterized by enhanced pathogen recognition and interferon (IFN) signaling. Bioinformatics analysis further identified a transcriptional signature uniquely induced by 5'pppRNA, and not by IFNα-2b, that included a constellation of IRF7 and NF-kB target genes capable of mobilizing multiple arms of the innate and adaptive immune response. Treatment of primary PBMCs or lung epithelial A549 cells with 5'pppRNA provided significant protection against a spectrum of RNA and DNA viruses. In C57Bl/6 mice, intravenous administration of 5'pppRNA protected animals from a lethal challenge with H1N1 Influenza, reduced virus titers in mouse lungs and protected animals from virus-induced pneumonia. Strikingly, the RIG-I-specific transcriptional response afforded partial protection from influenza challenge, even in the absence of type I interferon signaling. This systems approach provides transcriptional, biochemical, and in vivo analysis of the antiviral efficacy of 5'pppRNA and highlights the therapeutic potential associated with the use of RIG-I agonists as broad spectrum antiviral agents.


Asunto(s)
Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , ARN Viral/farmacología , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/metabolismo , Animales , Antivirales/uso terapéutico , Línea Celular , Activación Enzimática , Humanos , Inmunidad Innata , Inflamación , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Interferencia de ARN , ARN Viral/genética , ARN Viral/metabolismo , ARN Viral/uso terapéutico , Receptores de Ácido Retinoico/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
2.
Immunity ; 33(6): 833-5, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21168773
3.
Adv Exp Med Biol ; 809: 49-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25302365

RESUMEN

The A20 protein has emerged as an important negative regulator of Toll like receptor (TLR) and retinoic acid-inducible gene 1 (RIG-I)-mediated anti-viral signaling. A20 functions both as a RING-type E3 ubiquitin ligase and as a de-ubiquitinating enzyme. Nuclear factor kappa B (NF-kappaB) and interferon regulatory factor (IRF) pathways are targeted by A20 through mechanisms that appear to be both overlapping and distinct, resulting in the downregulation of interferon alpha/beta (IFNalpha/beta) production. This review specifically details the impact of A20 on the cytosolic RIG-I/MDA5 pathway, a process that is less understood than that of NF-kappaB but is essential for the regulation of the innate immune response to viral infection.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Inmunidad Innata/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Virus/inmunología , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , FN-kappa B/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitinación
4.
J Virol ; 86(2): 726-37, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22072751

RESUMEN

X-linked inhibitor of apoptosis (XIAP) is a potent antagonist of caspase 3-, 7-, and 9-dependent apoptotic activities that functions as an E3 ubiquitin ligase, and it targets caspases for degradation. In this study, we demonstrate that Sendai virus (SeV) infection results in the IKKε- or TBK1-mediated phosphorylation of XIAP in vivo at Ser430, resulting in Lys(48)-linked autoubiquitination at Lys322/328 residues, followed by the subsequent proteasomal degradation of XIAP. Interestingly, IKKε expression and XIAP turnover increases SeV-triggered mitochondrion-dependent apoptosis via the release of caspase 3, whereas TBK1 expression does not increase apoptosis. Interestingly, phosphorylation also regulates XIAP interaction with the transcription factor IRF3, suggesting a role in IRF3-Bax-mediated apoptosis. Our findings reveal a novel function of IKKε as a regulator of the virus-induced triggering of apoptosis via the phosphorylation-dependent turnover of XIAP.


Asunto(s)
Apoptosis , Quinasa I-kappa B/metabolismo , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/fisiopatología , Virus Sendai/fisiología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Secuencias de Aminoácidos , Línea Celular , Humanos , Quinasa I-kappa B/genética , Fosforilación , Infecciones por Respirovirus/virología , Virus Sendai/genética , Proteína Inhibidora de la Apoptosis Ligada a X/química , Proteína Inhibidora de la Apoptosis Ligada a X/genética
5.
Sarcoidosis Vasc Diffuse Lung Dis ; 40(1): e2023011, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36975051

RESUMEN

Efzofitimod is a first-in-class biologic based on a naturally occurring splice variant of histidyl-tRNA synthetase (HARS) that downregulates immune responses via selective modulation of neuropilin-2 (NRP2). Preclinical data found high expression of NRP2 in sarcoidosis granulomas. Treatment with efzofitimod reduced the granulomatous inflammation induced by P. acnes in an animal model of sarcoidosis. A dose escalating trial of efzofitimod in sarcoidosis with chronic symptomatic pulmonary disease found that treatment with efzofitimod was associated with improved quality of life with a trend towards reduced glucocorticoid use and stable to improved pulmonary function. These studies have led to a large Phase 3 trial of efzofitimod in symptomatic pulmonary sarcoidosis.

6.
Sci Transl Med ; 15(694): eadf1128, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37134152

RESUMEN

Although blocking the binding of vascular endothelial growth factor (VEGF) to neuropilin-2 (NRP2) on tumor cells is a potential strategy to treat aggressive carcinomas, a lack of effective reagents that can be used clinically has hampered this potential therapy. Here, we describe the generation of a fully humanized, high-affinity monoclonal antibody (aNRP2-10) that specifically inhibits the binding of VEGF to NRP2, conferring antitumor activity without causing toxicity. Using triple-negative breast cancer as a model, we demonstrated that aNRP2-10 could be used to isolate cancer stem cells (CSCs) from heterogeneous tumor populations and inhibit CSC function and epithelial-to-mesenchymal transition. aNRP2-10 sensitized cell lines, organoids, and xenografts to chemotherapy and inhibited metastasis by promoting the differentiation of CSCs to a state that is more responsive to chemotherapy and less prone to metastasis. These data provide justification for the initiation of clinical trials designed to improve the response of patients with aggressive tumors to chemotherapy using this monoclonal antibody.


Asunto(s)
Neuropilina-2 , Neoplasias de la Mama Triple Negativas , Humanos , Neuropilina-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Unión Proteica , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Línea Celular Tumoral , Neuropilina-1/metabolismo
7.
Nucleic Acid Ther ; 32(6): 473-485, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36355073

RESUMEN

Nucleic acid-based phosphorothioate containing antisense oligonucleotides (PS-ASOs) have the potential to activate cellular innate immune responses, and the level of activation can vary quite dramatically with sequence. Minimizing the degree of proinflammatory effect is one of the main selection criteria for compounds intended to move into clinical trials. While a recently developed human peripheral blood mononuclear cell (hPBMC)-based assay showed excellent ability to detect innate immune active PS-ASOs, which can then be discarded from the developmental process, this assay is highly resource intensive and easily affected by subject variability. This compelled us to develop a more convenient high-throughput assay. In this study, we describe a new in vitro assay, utilizing a cultured human Bjab cell line, which was developed and validated to identify PS-ASOs that may cause innate immune activation. The assay was calibrated to replicate results from the hPBMC assay. The Bjab assay was designed to be high throughput and more convenient by using RT-qPCR readout of mRNA of the chemokine Ccl22. The Bjab assay was also shown to be highly reproducible and to provide a large dynamic range in determining the immune potential of PS-ASOs through comparison to known benchmark PS-ASO controls that were previously shown to be safe or inflammatory in clinical trials. In addition, we demonstrate that Bjab cells can be used to provide mechanistic information on PS-ASO TLR9-dependent innate immune activation.


Asunto(s)
Linfoma de Burkitt , Oligonucleótidos Antisentido , Humanos , Oligonucleótidos Antisentido/genética , Linfoma de Burkitt/genética , Linfoma de Burkitt/terapia , Leucocitos Mononucleares , Receptor Toll-Like 9/genética
8.
Nucleic Acid Ther ; 32(6): 457-472, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35976085

RESUMEN

A human peripheral blood mononuclear cell (PBMC)-based assay was developed to identify antisense oligonucleotide (ASO) with the potential to activate a cellular innate immune response outside of an acceptable level. The development of this assay was initiated when ISIS 353512 targeting the messenger ribonucleic acid for human C-reactive protein (CRP) was tested in a phase I clinical trial, in which healthy human volunteers unexpectedly experienced increases in interleukin-6 (IL-6) and CRP. This level of immune stimulation was not anticipated following rodent and nonhuman primate safety studies in which no evidence of exaggerated proinflammatory effects were observed. The IL-6 increase induced by ISIS 353512 was caused by activation of B cells. The IL-6 induction was inhibited by chloroquine pretreatment of PBMCs and the nature of ASOs suggested that the response is mediated by a Toll-like receptor (TLR), in all likelihood TLR9. While assessing the inter PBMC donor variability, two classes of human PBMC responders to ISIS 353512 were identified (discriminator and nondiscriminators). The discriminator donor PBMCs were shown to produce low level of IL-6 after 24 h in culture, in the absence of ASO treatment. The PBMC assay using discriminator donors was shown to be reproducible, allowing to assess reliably the immune potential of ASOs by comparison to known benchmark ASO controls that were previously shown to be either safe or inflammatory in clinical trials. Clinical Trial registration numbers: NCT00048321 NCT00330330 NCT00519727.


Asunto(s)
Leucocitos Mononucleares , Oligonucleótidos Antisentido , Humanos , Oligonucleótidos Antisentido/genética , Voluntarios Sanos , Interleucina-6/genética
9.
Nucleic Acid Ther ; 30(2): 94-103, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32043907

RESUMEN

Inotersen, a 2'-O-methoxyethyl (2'-MOE) phosphorothioate antisense oligonucleotide, reduced disease progression and improved quality of life in patients with hereditary transthyretin amyloidosis with polyneuropathy (hATTR-PN) in the NEURO-TTR and NEURO-TTR open-label extension (OLE) trials. However, 300 mg/week inotersen treatment was associated with platelet count reductions in several patients. Mean platelet counts in patients in the NEURO-TTR-inotersen group remained ≥140 × 109/L in 50% and ≥100 × 109/L in 80% of the subjects. However, grade 4 thrombocytopenia (<25 × 109/L) occurred in three subjects in NEURO-TTR trial, and one of these suffered a fatal intracranial hemorrhage. The two others were treated successfully with corticosteroids and discontinuation of inotersen. Investigations in a subset of subjects in NEURO-TTR (n = 17 placebo; n = 31 inotersen) and OLE (n = 33) trials ruled out direct myelotoxicity, consumptive coagulopathy, and heparin-induced thrombocytopenia. Antiplatelet immunoglobulin G (IgG) antibodies were detected at baseline in 5 of 31 (16%) inotersen-treated subjects in NEURO-TTR, 4 of whom eventually developed grade 1 or 2 thrombocytopenia while on the drug. In addition, 24 subjects in the same group developed treatment-emergent antiplatelet IgG antibodies, of which 2 developed grade 2, and 3 developed grade 4 thrombocytopenia. Antiplatelet IgG antibodies in two of the three grade 4 thrombocytopenia subjects targeted GPIIb/IIIa. Plasma cytokines previously implicated in immune dysregulation, such as interleukin (IL)-23 and a proliferation-inducing ligand (APRIL) were often above the normal range at baseline. Collectively, these findings suggest an underlying immunologic dysregulation predisposing some individuals to immune-mediated thrombocytopenia during inotersen treatment.


Asunto(s)
Neuropatías Amiloides Familiares/tratamiento farmacológico , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos/administración & dosificación , Trombocitopenia/sangre , Adulto , Anciano , Neuropatías Amiloides Familiares/genética , Neuropatías Amiloides Familiares/inmunología , Neuropatías Amiloides Familiares/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Enfermedades del Sistema Inmune/inducido químicamente , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/patología , Inmunoglobulina G , Hemorragias Intracraneales/inducido químicamente , Hemorragias Intracraneales/inmunología , Hemorragias Intracraneales/patología , Masculino , Persona de Mediana Edad , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Oligonucleótidos/efectos adversos , Oligonucleótidos Antisentido/efectos adversos , Calidad de Vida , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología , Trombocitopenia/patología
10.
Trends Mol Med ; 12(2): 53-6, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16406812

RESUMEN

Intracellular viral infection is detected by the cytoplasmic RNA helicase RIG-I, which has an essential role in initiating the host antiviral response. The adaptor molecule that connects RIG-I sensing of incoming viral RNA to downstream signaling and gene activation has recently been elucidated by four independent research groups, and has been ascribed four different names: MAVS, IPS-1, VISA and Cardif. The fact that MAVS/IPS-1/VISA/Cardif localizes to the mitochondrial membrane suggests a link between viral infection, mitochondrial function and development of innate immunity. Furthermore, the hepatitis C virus NS3/4A protease specifically cleaves MAVS/IPS-1/VISA/Cardif as part of its immune-evasion strategy. These studies highlight a novel role for the mitochondria and for caspase activation and recruitment domain (CARD)-containing proteins in coordinating immune and apoptotic responses.


Asunto(s)
Inmunidad Innata/inmunología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , FN-kappa B/metabolismo , Animales , Apoptosis , Caspasas/metabolismo , Humanos
11.
Nucleic Acid Ther ; 27(5): 272-284, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28605247

RESUMEN

Antisense oligonucleotides (ASOs) are widely accepted therapeutic agents that suppress RNA transcription. While the majority of ASOs are well tolerated in vivo, few sequences trigger inflammatory responses in absence of conventional CpG motifs. In this study, we identified non-CpG oligodeoxy-nucleotide (ODN) capable of triggering an inflammatory response resulting in B cell and macrophage activation in a MyD88- and TLR9-dependent manner. In addition, we found the receptor for advance glycation end product (RAGE) receptor to be involved in the initiation of inflammatory response to suboptimal concentrations of both CpG- and non-CpG-containing ODNs. In contrast, dosing RAGE KO mice with high doses of CpG or non-CpG ODNs lead to a stronger inflammatory response than observed in wild-type mice. Together, our data provide a previously uncharacterized in vivo mechanism contingent on ODN-administered dose, where TLR9 governs the primary response and RAGE plays a distinct and cooperative function in providing a pivotal role in balancing the immune response.


Asunto(s)
Inmunidad Celular/inmunología , Inflamación/inmunología , Oligonucleótidos Antisentido/uso terapéutico , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Linfocitos B/inmunología , Citocinas/sangre , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/metabolismo , Cultivo Primario de Células , ARN/genética , ARN/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética , Análisis de la Célula Individual , Receptor Toll-Like 9/genética , Transcripción Genética
12.
Cell Host Microbe ; 12(2): 211-22, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22901541

RESUMEN

The RIG-I/Mda5 sensors recognize viral intracellular RNA and trigger host antiviral responses. RIG-I signals through the adaptor protein MAVS, which engages various TRAF family members and results in type I interferon (IFNs) and proinflammatory cytokine production via activation of IRFs and NF-κB, respectively. Both the IRF and NF-κB pathways also require the adaptor protein NEMO. We determined that the RIG-I pathway is differentially regulated by the linear ubiquitin assembly complex (LUBAC), which consists of the E3 ligases HOIL-1L, HOIP, and the accessory protein SHARPIN. LUBAC downregulated virus-mediated IFN induction by targeting NEMO for linear ubiquitination. Linear ubiquitinated NEMO associated with TRAF3 and disrupted the MAVS-TRAF3 complex, which inhibited IFN activation while stimulating NF-κB-dependent signaling. In SHARPIN-deficient MEFs, vesicular stomatitis virus replication was decreased due to increased IFN production. Linear ubiquitination thus switches NEMO from a positive to a negative regulator of RIG-I signaling, resulting in an attenuated IFN response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa I-kappa B/metabolismo , Interferones/inmunología , Factor 3 Asociado a Receptor de TNF/metabolismo , Estomatitis Vesicular/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Regulación hacia Abajo , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Ratones , Ratones Noqueados , Unión Proteica , Factor 3 Asociado a Receptor de TNF/genética , Ubiquitinación , Estomatitis Vesicular/genética , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/fisiología
13.
Curr Opin Immunol ; 23(5): 564-72, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21865020

RESUMEN

Sensing of RNA virus infection by the RIG-I-like receptors (RLRs) engages a complex signaling cascade that utilizes the mitochondrial antiviral signaling (MAVS) adapter protein to orchestrate the innate host response to pathogen, ultimately leading to the induction of antiviral and inflammatory responses mediated by type I interferon (IFN) and NF-κB pathways. MAVS is localized to the outer mitochondrial membrane, and has been associated with peroxisomes, the endoplasmic reticulum and autophagosomes, where it coordinates signaling events downstream of RLRs. MAVS not only plays a pivotal role in the induction of antiviral and inflammatory pathways but is also involved in the coordination of apoptotic and metabolic functions. This review summarizes recent findings related to the MAVS adapter and its essential role in the innate immune response to RNA viruses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Inmunidad Innata , Mitocondrias/inmunología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , Transducción de Señal/inmunología , Factores de Transcripción/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/inmunología , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Humanos , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1 , Mitocondrias/metabolismo , Membranas Mitocondriales/inmunología , Membranas Mitocondriales/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Peroxisomas/inmunología , Peroxisomas/metabolismo , ARN Helicasas/inmunología , ARN Helicasas/metabolismo , Infecciones por Virus ARN/virología , ARN Viral/inmunología , Transactivadores , Factores de Transcripción/metabolismo
14.
Cell Res ; 21(6): 895-910, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21200404

RESUMEN

Recognition of viral RNA structures by the cytosolic sensor retinoic acid-inducible gene-I (RIG-I) results in the activation of signaling cascades that culminate with the generation of the type I interferon (IFN) antiviral response. Onset of antiviral and inflammatory responses to viral pathogens necessitates the regulated spatiotemporal recruitment of signaling adapters, kinases and transcriptional proteins to the mitochondrial antiviral signaling protein (MAVS). We previously demonstrated that the serine/threonine kinase IKKε is recruited to the C-terminal region of MAVS following Sendai or vesicular stomatitis virus (VSV) infection, mediated by Lys63-linked polyubiquitination of MAVS at Lys500, resulting in inhibition of downstream IFN signaling (Paz et al, Mol Cell Biol, 2009). In this study, we demonstrate that C-terminus of MAVS harbors a novel TRAF3-binding site in the aa450-468 region of MAVS. A consensus TRAF-interacting motif (TIM), 455-PEENEY-460, within this site is required for TRAF3 binding and activation of IFN antiviral response genes, whereas mutation of the TIM eliminates TRAF3 binding and the downstream IFN response. Reconstitution of MAVS(-/-) mouse embryo fibroblasts with a construct expressing a TIM-mutated version of MAVS failed to restore the antiviral response or block VSV replication, whereas wild-type MAVS reconstituted antiviral inhibition of VSV replication. Furthermore, recruitment of IKKε to an adjacent C-terminal site (aa 468-540) in MAVS via Lys500 ubiquitination decreased TRAF3 binding and protein stability, thus contributing to IKKε-mediated shutdown of the IFN response. This study demonstrates that MAVS harbors a functional C-terminal TRAF3-binding site that participates in positive and negative regulation of the IFN antiviral response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Retroalimentación Fisiológica , Inmunidad Innata , Interferón Tipo I/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Quinasa I-kappa B/metabolismo , Interferón Tipo I/inmunología , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Estomatitis Vesicular/inmunología , Vesiculovirus/inmunología
15.
Mol Cell Biol ; 29(12): 3401-12, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19380491

RESUMEN

Induction of the antiviral interferon response is initiated upon recognition of viral RNA structures by the RIG-I or Mda-5 DEX(D/H) helicases. A complex signaling cascade then converges at the mitochondrial adapter MAVS, culminating in the activation of the IRF and NF-kappaB transcription factors and the induction of interferon gene expression. We have previously shown that MAVS recruits IkappaB kinase epsilon (IKKepsilon) but not TBK-1 to the mitochondria following viral infection. Here we map the interaction of MAVS and IKKepsilon to the C-terminal region of MAVS and demonstrate that this interaction is ubiquitin dependent. MAVS is ubiquitinated following Sendai virus infection, and K63-linked ubiquitination of lysine 500 (K500) of MAVS mediates recruitment of IKKepsilon to the mitochondria. Real-time PCR analysis reveals that a K500R mutant of MAVS increases the mRNA level of several interferon-stimulated genes and correlates with increased NF-kappaB activation. Thus, recruitment of IKKepsilon to the mitochondria upon MAVS K500 ubiquitination plays a modulatory role in the cascade leading to NF-kappaB activation and expression of inflammatory and antiviral genes. These results provide further support for the differential role of IKKepsilon and TBK-1 in the RIG-I/Mda5 pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa I-kappa B/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Células HeLa , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Interferón beta/metabolismo , Lisina/química , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virus Sendai/patogenicidad , Transducción de Señal
16.
J Biol Chem ; 284(33): 21797-21809, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19546225

RESUMEN

The mitochondria-bound adapter MAVS participates in IFN induction by recruitment of downstream partners such as members of the TRAF family, leading to activation of NF-kappaB, and the IRF3 pathways. A yeast two-hybrid search for MAVS-interacting proteins yielded the Polo-box domain (PBD) of the mitotic Polo-like kinase PLK1. We showed that PBD associates with two different domains of MAVS in both dependent and independent phosphorylation events. The phosphodependent association requires the phosphopeptide binding ability of PBD. It takes place downstream of the proline-rich domain of MAVS, within an STP motif, characteristic of the binding of PLK1 to its targets, where the central Thr234 residue is phosphorylated. Its phosphoindependent association takes place at the C terminus of MAVS. PLK1 strongly inhibits the ability of MAVS to activate the IRF3 and NF-kappaB pathways and to induce IFN. Reciprocally, depletion of PLK1 can increase IFN induction in response to RIG-I/SeV or RIG-I/poly(I)-poly(C) treatments. This inhibition is dependent on the phosphoindependent association of PBD at the C terminus of MAVS where it disrupts the association of MAVS with its downstream partner TRAF3. IFN induction was strongly inhibited in cells arrested in G2/M by nocodazole, which provokes increased expression of endogenous PLK1. Interestingly, depletion of PLK1 from these nocodazole-treated cells could restore, at least partially, IFN induction. Altogether, these data demonstrate a new function for PLK1 as a regulator of IFN induction and provide the basis for the development of inhibitors preventing the PLK1/MAVS association to sustain innate immunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Interferones/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Modelos Biológicos , FN-kappa B/metabolismo , Nocodazol/farmacología , Fosfopéptidos/química , Fosforilación , Prolina/química , Estructura Terciaria de Proteína , Treonina/química , Técnicas del Sistema de Dos Híbridos , Quinasa Tipo Polo 1
17.
J Immunol ; 176(11): 7051-61, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16709867

RESUMEN

Primary effusion lymphoma (PEL) is a herpesvirus-8-associated lymphoproliferative disease characterized by migration of tumor cells to serous body cavities. PEL cells originate from postgerminal center B cells and share a remarkable alteration in B cell transcription factor expression and/or activation with classical Hodgkin's disease cells. Comparative analysis of gene expression by cDNA microarray of BCBL-1 cells (PEL), L-428 (classical Hodgkin's disease), and BJAB cells revealed a subset of genes that were differentially expressed in BCBL-1 cells. Among these, four genes involved in cell migration and chemotaxis were strongly up-regulated in PEL cells: leukotriene A4 (LTA4) hydrolase (LTA4H), IL-16, thrombospondin-1 (TSP-1), and selectin-P ligand (PSGL-1). Up-regulation of LTA4H was investigated at the transcriptional level. Full-length LTA4H promoter exhibited 50% higher activity in BCBL-1 cells than in BJAB or L-428 cells. Deletion analysis of the LTA4H promoter revealed a positive cis-regulatory element active only in BCBL-1 cells in the promoter proximal region located between -76 and -40 bp. Formation of a specific DNA-protein complex in this region was confirmed by EMSA. Coculture of ionophore-stimulated primary neutrophils with BCBL-1 cells leads to an increased production of LTB4 compared with coculture with BJAB and L-428 cells as measured by enzyme immunoassay, demonstrating the functional significance of LTA4H up-regulation.


Asunto(s)
Epóxido Hidrolasas/biosíntesis , Epóxido Hidrolasas/genética , Leucotrieno B4/biosíntesis , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Regulación hacia Arriba , Línea Celular Tumoral , Activación Enzimática/genética , Epóxido Hidrolasas/aislamiento & purificación , Epóxido Hidrolasas/fisiología , Perfilación de la Expresión Génica , Enfermedad de Hodgkin/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Interleucina-16/fisiología , Linfoma de Células B/metabolismo , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Trombospondina 1/biosíntesis , Trombospondina 1/genética , Trombospondina 1/fisiología , Transcripción Genética , Regulación hacia Arriba/genética
18.
J Virol ; 80(12): 6072-83, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731946

RESUMEN

Intracellular RNA virus infection is detected by the cytoplasmic RNA helicase RIG-I that plays an essential role in signaling to the host antiviral response. Recently, the adapter molecule that links RIG-I sensing of incoming viral RNA to downstream signaling and gene activation events was characterized by four different groups; MAVS/IPS-1-1/VISA/Cardif contains an amino-terminal CARD domain and a carboxyl-terminal mitochondrial transmembrane sequence that localizes to the mitochondrial membrane. Furthermore, the hepatitis C virus NS3-4A protease complex specifically targets MAVS/IPS-1/VISA/Cardif for cleavage as part of its immune evasion strategy. With a novel search program written in python, we also identified an uncharacterized protein, KIAA1271 (K1271), containing a single CARD-like domain at the N terminus and a Leu-Val-rich C terminus that is identical to that of MAVS/IPS-1/VISA/Cardif. Using a combination of biochemical analysis, subcellular fractionation, and confocal microscopy, we now demonstrate that NS3-4A cleavage of MAVS/IPS-1/VISA/Cardif/K1271 results in its dissociation from the mitochondrial membrane and disrupts signaling to the antiviral immune response. Furthermore, virus-induced IKKepsilon kinase, but not TBK1, colocalized strongly with MAVS at the mitochondrial membrane, and the localization of both molecules was disrupted by NS3-4A expression. Mutation of the critical cysteine 508 to alanine was sufficient to maintain mitochondrial localization of MAVS/IPS-1/VISA/Cardif and IKKepsilon in the presence of NS3-4A. These observations provide an outline of the mechanism by which hepatitis C virus evades the interferon antiviral response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Hepacivirus , Quinasa I-kappa B/metabolismo , Proteínas Mitocondriales/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas no Estructurales Virales/metabolismo , Hepacivirus/química , Humanos , Membranas Intracelulares/metabolismo , Mitocondrias/ultraestructura , Péptido Hidrolasas/metabolismo , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA