Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Immunity ; 52(4): 668-682.e7, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294407

RESUMEN

The primary mechanisms supporting immunoregulatory polarization of myeloid cells upon infiltration into tumors remain largely unexplored. Elucidation of these signals could enable better strategies to restore protective anti-tumor immunity. Here, we investigated the role of the intrinsic activation of the PKR-like endoplasmic reticulum (ER) kinase (PERK) in the immunoinhibitory actions of tumor-associated myeloid-derived suppressor cells (tumor-MDSCs). PERK signaling increased in tumor-MDSCs, and its deletion transformed MDSCs into myeloid cells that activated CD8+ T cell-mediated immunity against cancer. Tumor-MDSCs lacking PERK exhibited disrupted NRF2-driven antioxidant capacity and impaired mitochondrial respiratory homeostasis. Moreover, reduced NRF2 signaling in PERK-deficient MDSCs elicited cytosolic mitochondrial DNA elevation and, consequently, STING-dependent expression of anti-tumor type I interferon. Reactivation of NRF2 signaling, conditional deletion of STING, or blockade of type I interferon receptor I restored the immunoinhibitory potential of PERK-ablated MDSCs. Our findings demonstrate the pivotal role of PERK in tumor-MDSC functionality and unveil strategies to reprogram immunosuppressive myelopoiesis in tumors to boost cancer immunotherapy.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Epitelial de Ovario/inmunología , Regulación Neoplásica de la Expresión Génica , Melanoma Experimental/inmunología , Proteínas de la Membrana/inmunología , Neoplasias Cutáneas/inmunología , eIF-2 Quinasa/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Femenino , Humanos , Terapia de Inmunosupresión , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Mitocondrias/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/patología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/inmunología , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Respuesta de Proteína Desplegada/inmunología , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
2.
Proc Natl Acad Sci U S A ; 116(47): 23671-23681, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31690657

RESUMEN

Invariant NKT (iNKT) cells have the unique ability to shape immunity during antitumor immune responses and other forms of sterile and nonsterile inflammation. Recent studies have highlighted a variety of classes of endogenous and pathogen-derived lipid antigens that can trigger iNKT cell activation under sterile and nonsterile conditions. However, the context and mechanisms that drive the presentation of self-lipid antigens in sterile inflammation remain unclear. Here we report that endoplasmic reticulum (ER)-stressed myeloid cells, via signaling events modulated by the protein kinase RNA-like ER kinase (PERK) pathway, increase CD1d-mediated presentation of immunogenic endogenous lipid species, which results in enhanced iNKT cell activation both in vitro and in vivo. In addition, we demonstrate that actin cytoskeletal reorganization during ER stress results in an altered distribution of CD1d on the cell surface, which contributes to enhanced iNKT cell activation. These results define a previously unidentified mechanism that controls iNKT cell activation during sterile inflammation.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Estrés del Retículo Endoplásmico/inmunología , Activación de Linfocitos , Células T Asesinas Naturales/inmunología , Animales , Presentación de Antígeno , Antígenos CD1d/biosíntesis , Antígenos CD1d/inmunología , Autoantígenos/inmunología , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Citoesqueleto/ultraestructura , Endosomas/inmunología , Glicoesfingolípidos/inmunología , Glicoesfingolípidos/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Lípidos/inmunología , Lisosomas/inmunología , Ratones , Ratones Endogámicos C57BL , Células THP-1 , Tapsigargina/farmacología , Respuesta de Proteína Desplegada/inmunología , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/fisiología
3.
RNA ; 23(2): 153-160, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27837013

RESUMEN

While mammalian somatic cells are incapable of mounting an effective RNA interference (RNAi) response to viral infections, plants and invertebrates are able to generate high levels of viral short interfering RNAs (siRNAs) that can control many infections. In Drosophila, the RNAi response is mediated by the Dicer 2 enzyme (dDcr2) acting in concert with two cofactors called Loqs-PD and R2D2. To examine whether a functional RNAi response could be mounted in human somatic cells, we expressed dDcr2, in the presence or absence of Loqs-PD and/or R2D2, in a previously described human cell line, NoDice/ΔPKR, that lacks functional forms of human Dicer (hDcr) and PKR. We observed significant production of ∼21-nt long siRNAs, derived from a cotransfected double stranded RNA (dsRNA) expression vector, that were loaded into the human RNA-induced silencing complex (RISC) and were able to significantly reduce the expression of a cognate indicator gene. Surprisingly, dDcr2 was able to produce siRNAs even in the absence of Loqs-PD, which is thought to be required for dsRNA cleavage by dDcr2. This result may be explained by our finding that dDcr2 is able to bind the human Loqs-PD homolog TRBP when expressed in human cells in the absence of Loqs-PD. We conclude that it is possible to at least partially rescue the ability of mammalian somatic cells to express functional siRNAs using gene products of invertebrate origin.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , ARN Helicasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Animales , Ingeniería Celular , Línea Celular , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Prueba de Complementación Genética , Humanos , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo , Unión Proteica , ARN Helicasas/metabolismo , ARN Interferente Pequeño/biosíntesis , Proteínas de Unión al ARN/metabolismo , Complejo Silenciador Inducido por ARN/biosíntesis , Complejo Silenciador Inducido por ARN/genética , Ribonucleasa III/deficiencia , Ribonucleasa III/metabolismo , Transgenes , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
4.
Biol Cell ; 110(2): 27-32, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29168198

RESUMEN

In humans, the pathogenesis of diabetes is characterised by two major pancreatic ß cell defects: a reduction in ß cell mass and the failure of ß cells to produce enough insulin. Over the past two decades, multiple studies involving cell cultures, animal models and human subjects have established the importance of the protein kinase RNA-like endoplasmic reticulum kinase (PERK) in the adaptive functional capacity of pancreatic ß cells during embryonic development and into adulthood. In this review, we will highlight major findings identifying PERK as a crucial player in ß cell physiology and in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Epífisis/anomalías , Factor 2 Eucariótico de Iniciación/genética , Homeostasis/genética , Células Secretoras de Insulina/enzimología , Osteocondrodisplasias/genética , eIF-2 Quinasa/genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/inmunología , Adulto , Animales , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Epífisis/enzimología , Epífisis/inmunología , Epífisis/patología , Factor 2 Eucariótico de Iniciación/inmunología , Regulación del Desarrollo de la Expresión Génica , Homeostasis/inmunología , Humanos , Recién Nacido , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/inmunología , Osteocondrodisplasias/enzimología , Osteocondrodisplasias/inmunología , Osteocondrodisplasias/patología , Transducción de Señal , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/inmunología
5.
Appl Microbiol Biotechnol ; 103(15): 6153-6167, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31154490

RESUMEN

MoHrip2, identified from Magnaporthe oryzae as an elicitor, can activate plant defense responses either in the form of recombinant protein in vitro or ectopic expressed protein in rice. However, its intrinsic function in the infective interaction of M. oryzae-rice is largely unknown. Here, we found that mohrip2 expression was significantly induced at stages of fungal penetration and colonization. Meanwhile, the induced MoHrip2 mainly accumulated in the rice apoplast by outlining the entire invasive hyphae during infection, and its secretion was via the conventional endoplasmic reticulum (ER)-to-Golgi pathway, demonstrating the nature of MoHrip2 as an apoplastic effector. What's more, the disease facilitating function of MoHrip2 was revealed by the significantly compromised virulence of Δmohrip2 mutants on rice seedlings and even on the wounded rice leaves. Inoculations of these mutant strains on rice leaf sheaths showed a reduction in penetration and subsequent expansion of fungal growth, which is probably due to activated host immunity including the expression of certain defense-related genes and the production of certain phytoalexins. Altogether, these results demonstrated the necessity of MoHrip2 in suppression of host immunity and the full virulence of M. oryzae.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Magnaporthe/patogenicidad , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Factores de Virulencia/metabolismo , eIF-2 Quinasa/metabolismo , Eliminación de Gen , Magnaporthe/enzimología , Oryza/inmunología , Virulencia , Factores de Virulencia/deficiencia , eIF-2 Quinasa/deficiencia
6.
Nature ; 488(7413): 670-4, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22801494

RESUMEN

The inflammasome regulates the release of caspase activation-dependent cytokines, including interleukin (IL)-1ß, IL-18 and high-mobility group box 1 (HMGB1). By studying HMGB1 release mechanisms, here we identify a role for double-stranded RNA-dependent protein kinase (PKR, also known as EIF2AK2) in inflammasome activation. Exposure of macrophages to inflammasome agonists induced PKR autophosphorylation. PKR inactivation by genetic deletion or pharmacological inhibition severely impaired inflammasome activation in response to double-stranded RNA, ATP, monosodium urate, adjuvant aluminium, rotenone, live Escherichia coli, anthrax lethal toxin, DNA transfection and Salmonella typhimurium infection. PKR deficiency significantly inhibited the secretion of IL-1ß, IL-18 and HMGB1 in E. coli-induced peritonitis. PKR physically interacts with several inflammasome components, including NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), NLRP1, NLR family CARD domain-containing protein 4 (NLRC4), absent in melanoma 2 (AIM2), and broadly regulates inflammasome activation. PKR autophosphorylation in a cell-free system with recombinant NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC, also known as PYCARD) and pro-caspase-1 reconstitutes inflammasome activity. These results show a crucial role for PKR in inflammasome activation, and indicate that it should be possible to pharmacologically target this molecule to treat inflammation.


Asunto(s)
Proteína HMGB1/metabolismo , Inflamasomas/metabolismo , eIF-2 Quinasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfato/farmacología , Animales , Antígenos Bacterianos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Toxinas Bacterianas/farmacología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Células Cultivadas , Cristalinas/metabolismo , Escherichia coli/inmunología , Escherichia coli/fisiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Femenino , Proteína HMGB1/sangre , Humanos , Inflamasomas/agonistas , Interleucina-18/sangre , Interleucina-1beta/sangre , Interleucina-6/análisis , Interleucina-6/sangre , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas NLR , Peritonitis/metabolismo , Fosforilación , ARN Bicatenario/inmunología , ARN Bicatenario/farmacología , Rotenona/farmacología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/inmunología , Salmonella typhimurium/fisiología , Transfección , Ácido Úrico/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
7.
J Biol Chem ; 291(29): 15093-107, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226571

RESUMEN

The sensing of double-stranded RNA (dsRNA) in the liver is important for antiviral defenses but can also contribute to sterile inflammation during liver injury. Hepatocytes are often the target of viral infection and are easily injured by inflammatory insults. Here we sought to establish the pathways involved in the production of type I interferons (IFN-I) in response to extracellular poly(I:C), a dsRNA mimetic, in hepatocytes. This was of interest because hepatocytes are long-lived and, unlike most immune cells that readily die after activation with dsRNA, are not viewed as cells with robust antimicrobial capacity. We found that poly(I:C) leads to rapid up-regulation of inducible nitric oxide synthase (iNOS), double-stranded RNA-dependent protein kinase (PKR), and Src. The production of IFN-ß was dependent on iNOS, PKR, and Src and partially dependent on TLR3/Trif. iNOS and Src up-regulation was partially dependent on TLR3/Trif but entirely dependent on PKR. The phosphorylation of TLR3 on tyrosine 759 was shown to increase in parallel to IFN-ß production in an iNOS- and Src-dependent manner, and Src was found to directly interact with TLR3 in the endosomal compartment of poly(I:C)-treated cells. Furthermore, we identified a robust NO/cGMP/PKG-dependent feedforward pathway for the amplification of iNOS expression. These data identify iNOS/NO as an integral component of IFN-ß production in response to dsRNA in hepatocytes in a pathway that involves the coordinated activities of TLR3/Trif and PKR.


Asunto(s)
Hepatocitos/inmunología , Hepatocitos/metabolismo , Interferón beta/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Bicatenario/inmunología , ARN Bicatenario/farmacología , Receptor Toll-Like 3/metabolismo , eIF-2 Quinasa/metabolismo , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Hepatocitos/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Fosforilación/efectos de los fármacos , Poli I-C/farmacología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Tirosina/química , Regulación hacia Arriba/efectos de los fármacos , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
8.
J Virol ; 90(2): 1144-7, 2016 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-26512090

RESUMEN

Murine cytomegalovirus (MCMV) proteins m142 and m143 are essential for viral replication. They bind double-stranded RNA and prevent protein kinase R-induced protein synthesis shutoff. Whether the two viral proteins have additional functions such as their homologs in human cytomegalovirus do remained unknown. We show that MCMV m142 and m143 knockout mutants attain organ titers equivalent to those attained by wild-type MCMV in Pkr knockout mice, suggesting that these viral proteins do not encode additional PKR-independent functions relevant for pathogenesis in vivo.


Asunto(s)
Muromegalovirus/fisiología , Mutación , Proteínas Virales/genética , Replicación Viral , eIF-2 Quinasa/deficiencia , Animales , Ratones Noqueados , Muromegalovirus/genética , Carga Viral
9.
RNA Biol ; 14(11): 1570-1579, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28594311

RESUMEN

While lentiviral expression vectors are widely used in many facets of molecular biology, due to their ability to stably express heterologous genes in both dividing and non-dividing cells, they suffer from the disadvantage that introns inserted into the vector genome are generally rapidly lost by splicing in packaging cell lines. The presence of an intron, if achievable, has the potential to facilitate the expression of transgene cDNAs, as splicing has been extensively shown to facilitate mRNA biogenesis and function. Moreover, if a stable intron could be introduced into a lentiviral vector, this could greatly facilitate the expression of microRNAs (miRNAs), and especially miRNA clusters, as the introduction of pri-miRNA stems into the exonic region of a lentiviral vector can strongly reduce both vector titer and the expression of any miRNA-linked indicator gene due to cleavage of the vector RNA genome by cellular Drosha. Here, we describe a novel lentiviral vector design in which transgenes and/or miRNAs are expressed using an antisense-orientated, inducible promoter driving an expression cassette bearing a functional intron. We demonstrate that this lentiviral vector, called pTREX, is able to express higher levels of both transgenes and pri-miRNA clusters when compared with a closely similar conventional lentiviral vector.


Asunto(s)
Ingeniería Genética/métodos , Intrones , Lentivirus/genética , MicroARNs/genética , Empalme del ARN , Línea Celular , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Exones , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Insulina/genética , Insulina/metabolismo , Lentivirus/metabolismo , MicroARNs/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Regiones Promotoras Genéticas , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Transgenes , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
10.
J Immunol ; 193(3): 1459-67, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24990083

RESUMEN

Pseudomonas aeruginosa secrete N-(3-oxododecanoyl)-homoserine lactone (HSL-C12) as a quorum-sensing molecule to regulate bacterial gene expression. Because HSL-C12 is membrane permeant, multiple cell types in P. aeruginosa-infected airways may be exposed to HSL-C12, especially adjacent to biofilms where local (HSL-C12) may be high. Previous reports showed that HSL-C12 causes both pro- and anti-inflammatory effects. To characterize HSL-C12's pro- and anti-inflammatory effects in host cells, we measured protein synthesis, NF-κB activation, and KC (mouse IL-8) and IL-6 mRNA and protein secretion in wild-type mouse embryonic fibroblasts (MEF). To test the role of the endoplasmic reticulum stress inducer, PERK we compared these responses in PERK(-/-) and PERK-corrected PERK(-/-) MEF. During 4-h treatments of wild-type MEF, HSL-C12 potentially activated NF-κB p65 by preventing the resynthesis of IκB and increased transcription of KC and IL-6 genes (quantitative PCR). HSL-C12 also inhibited secretion of KC and/or IL-6 into the media (ELISA) both in control conditions and also during stimulation by TNF-α. HSL-C12 also activated PERK (as shown by increased phosphorylation of eI-F2α) and inhibited protein synthesis (as measured by incorporation of [(35)S]methionine by MEF). Comparisons of PERK(-/-) and PERK-corrected MEF showed that HSL-C12's effects were explained in part by activation of PERK→phosphorylation of eI-F2α→inhibition of protein synthesis→reduced IκBα production→activation of NF-κB→increased transcription of the KC gene but reduced translation and secretion of KC. HSL-C12 may be an important modulator of early (up to 4 h) inflammatory signaling in P. aeruginosa infections.


Asunto(s)
4-Butirolactona/análogos & derivados , Factor 2 Eucariótico de Iniciación/fisiología , Mediadores de Inflamación/fisiología , Pseudomonas aeruginosa/inmunología , Percepción de Quorum/inmunología , Transducción de Señal/inmunología , eIF-2 Quinasa/fisiología , 4-Butirolactona/fisiología , Animales , Línea Celular , Estrés del Retículo Endoplásmico/inmunología , Ratones , eIF-2 Quinasa/deficiencia
11.
Circulation ; 129(13): 1397-406, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24463368

RESUMEN

BACKGROUND: Double-stranded RNA-dependent protein kinase (PKR) is a eukaryotic initiation factor 2α kinase that inhibits mRNA translation under stress conditions. PKR also mediates inflammatory and apoptotic signaling independently of translational regulation. Congestive heart failure is associated with cardiomyocyte hypertrophy, inflammation, and apoptosis, but the role of PKR in left ventricular hypertrophy and the development of congestive heart failure has not been examined. METHODS AND RESULTS: We observed increased myocardial PKR expression and translocation of PKR into the nucleus in humans and mice with congestive heart failure. To determine the impact of PKR on the development of congestive heart failure, PKR knockout and wild-type mice were exposed to pressure overload produced by transverse aortic constriction. Although heart size increased similarly in wild-type and PKR knockout mice after transverse aortic constriction, PKR knockout mice exhibited very little pulmonary congestion, well-preserved left ventricular ejection fraction and contractility, and significantly less myocardial fibrosis compared with wild-type mice. Bone marrow-derived cells from wild-type mice did not abolish the cardiac protective effect observed in PKR knockout mice, whereas bone marrow-derived cells from PKR knockout mice had no cardiac protective effect in wild-type mice. Mechanistically, PKR knockout attenuated transverse aortic constriction-induced tumor necrosis factor-α expression and leukocyte infiltration and lowered cardiac expression of proapoptotic factors (Bax and caspase-3), so that PKR knockout hearts were more resistant to transverse aortic constriction-induced cardiomyocyte apoptosis. PKR depletion in isolated cardiomyocytes also conferred protection against tumor necrosis factor-α- or lipopolysaccharide-induced apoptosis. CONCLUSION: PKR is a maladaptive factor upregulated in hemodynamic overload that contributes to myocardial inflammation, cardiomyocyte apoptosis, and the development of congestive heart failure.


Asunto(s)
Presión Sanguínea/fisiología , Insuficiencia Cardíaca/prevención & control , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/fisiología , Disfunción Ventricular Izquierda/prevención & control , eIF-2 Quinasa/deficiencia , Adulto , Anciano , Animales , Aorta/fisiopatología , Apoptosis/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/metabolismo , Humanos , Hipertrofia/fisiopatología , Hipertrofia/prevención & control , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Regulación hacia Arriba/fisiología , eIF-2 Quinasa/genética , eIF-2 Quinasa/fisiología
12.
Cerebellum ; 14(4): 386-97, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25592072

RESUMEN

Ethanol-induced neuronal loss is closely related to the pathogenesis of fetal alcohol spectrum disorders. The cerebellum is one of the brain areas that are most sensitive to ethanol. The mechanism underlying ethanol neurotoxicity remains unclear. Our previous in vitro studies have shown that the double-stranded RNA (dsRNA)-activated protein kinase (PKR) regulates neuronal apoptosis upon ethanol exposure and ethanol activates PKR through association with its intracellular activator RAX. However, the role of PKR and its interaction with RAX in vivo have not been investigated. In the current study, by utilizing N-PKR-/- mice, C57BL/6J mice with a deficient RAX-binding domain in PKR, we determined the critical role of RAX/PKR association in PKR-regulated ethanol neurotoxicity in the developing cerebellum. Our data indicate that while N-PKR-/- mice have a similar BAC profile as wild-type mice, ethanol induces less brain/body mass reduction as well as cerebellar neuronal loss. In addition, ethanol promotes interleukin-1ß (IL-1ß) secretion, and IL-1ß is a master cytokine regulating inflammatory response. Importantly, ethanol-promoted IL-1ß secretion is inhibited in the developing cerebellum of N-PKR-/- mice. Thus, RAX/PKR interaction and PKR activation regulate ethanol neurotoxicity in the developing cerebellum, which may involve ethanol-induced neuroinflammation. Further, PKR could be a possible target for pharmacological intervention to prevent or treat fetal alcohol spectrum disorder (FASD).


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Cerebelo , Etanol/toxicidad , Síndromes de Neurotoxicidad/etiología , eIF-2 Quinasa/deficiencia , Factores de Edad , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Células Cultivadas , Cerebelo/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Cerebelo/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Síndromes de Neurotoxicidad/patología , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/genética , Sincalida/farmacología , Factores de Tiempo , eIF-2 Quinasa/genética
13.
Glia ; 62(5): 680-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24481666

RESUMEN

The immune-mediated central nervous system (CNS) demyelinating disorder multiple sclerosis (MS) is the most common neurological disease in young adults. One important goal of MS research is to identify strategies that will preserve oligodendrocytes (OLs) in MS lesions. During active myelination and remyelination, OLs synthesize large quantities of membrane proteins in the endoplasmic reticulum (ER), which may result in ER stress. During ER stress, pancreatic ER kinase (PERK) phosphorylates eukaryotic translation initiation factor 2α (elF2α), which activates the integrated stress response (ISR), resulting in a stress-resistant state. Previous studies have shown that PERK activity is increased in OLs within the demyelinating lesions of experimental autoimmune encephalomyelitis (EAE), a model of MS. Moreover, our laboratory has shown that PERK protects OLs from the adverse effects of interferon-γ, a key mediator of the CNS inflammatory response. Here, we have examined the role of PERK signaling in OLs during development and in response to EAE. We generated OL-specific PERK knockout (OL-PERK(ko/ko) ) mice that exhibited a lower level of phosphorylated elF2α in the CNS, indicating that the ISR is impaired in the OLs of these mice. Unexpectedly, OL-PERK(ko/ko) mice develop normally and show no myelination defects. Nevertheless, EAE is exacerbated in these mice, which is correlated with increased OL loss, demyelination, and axonal degeneration. These data indicate that although not needed for developmental myelination, PERK signaling provides protection to OLs against inflammatory demyelination and suggest that the ISR in OLs could be a valuable target for future MS therapeutics.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Eliminación de Gen , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , eIF-2 Quinasa/deficiencia , Animales , Enfermedades Desmielinizantes/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/genética , Transducción de Señal/fisiología , eIF-2 Quinasa/genética
14.
Eur J Immunol ; 43(5): 1147-52, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23401008

RESUMEN

Inflammasomes are multi-protein platforms that drive the activation of caspase-1 leading to the processing and secretion of biologically active IL-1ß and IL-18. Different inflammasomes including NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), NLR caspase-recruitment domain-containing 4 (NLRC4) and absent in melanoma 2 (AIM2) are activated and assembled in response to distinct microbial or endogenous stimuli. However, the mechanisms by which upstream stimuli trigger inflammasome activation remain poorly understood. Double-stranded RNA-activated protein kinase (PKR), a protein kinase activated by viral infection, has been recently shown to be required for the activation of the inflammasomes. Using macrophages from two different mouse strains deficient in PKR, we found that PKR is important for the induction of the inducible nitric oxide synthase (iNOS). However, PKR was dispensable for caspase-1 activation, processing of pro-IL-1ß/IL-18 and secretion of IL-1ß induced by stimuli that trigger the activation of NLRP3, NLRC4 and AIM2. These results indicate that PKR is not required for inflammasome activation in macrophages.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/inmunología , eIF-2 Quinasa/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Células Cultivadas , Proteínas de Unión al ADN , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Precursores de Proteínas/inmunología , Precursores de Proteínas/metabolismo , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
15.
J Virol ; 87(2): 756-66, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23115276

RESUMEN

ADAR1, an interferon (IFN)-inducible double-stranded (ds) RNA-specific adenosine deaminase, downregulates host innate responses, including activation of the dsRNA-dependent protein kinase (PKR) and induction of IFN-ß mRNA. Conversely, PKR amplifies IFN-ß induction by measles virus (MV) and inhibits virus protein synthesis. Formation of stress granules (SGs), cytoplasmic aggregates of stalled translation complexes and RNA-binding proteins, is a host response to virus infection mediated by translation initiation factor eIF2α phosphorylation. We examined the roles of PKR and ADAR1 in SG formation using HeLa cells stably deficient in either PKR (PKR(kd)) or ADAR1 (ADAR1(kd)) compared to control (CON(kd)) cells. Infection with either wild-type (WT) MV or an isogenic mutant lacking C protein expression (C(ko)) comparably induced formation of SG in ADAR1(kd) cells, whereas only the C(ko) mutant was an efficient inducer in control cells. Both ADAR1 and PKR colocalized with SG following infection. MV-induced; SG formation was PKR dependent but impaired by ADAR1. Complementation of ADAR1(kd) cells by expression of either p150 WT isoform or the p150 Zα (Y177A) Z-DNA-binding mutant of ADAR1 restored suppression of host responses, including SG formation and PKR activation. In contrast, neither the p110 WT isoform nor the p150 catalytic (H910A, E912A) mutant of ADAR1 complemented the ADAR1(kd) phenotype. These results further establish ADAR1 as a suppressor of host innate responses, including activation of PKR and the subsequent SG response.


Asunto(s)
Adenosina Desaminasa/metabolismo , Gránulos Citoplasmáticos/metabolismo , Interacciones Huésped-Patógeno , Virus del Sarampión/patogenicidad , eIF-2 Quinasa/metabolismo , Adenosina Desaminasa/deficiencia , Prueba de Complementación Genética , Células HeLa , Humanos , Virus del Sarampión/genética , Proteínas de Unión al ARN , eIF-2 Quinasa/deficiencia
16.
J Biol Chem ; 287(19): 15580-9, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22427667

RESUMEN

Inflammation plays an important pathogenic role in a number of metabolic diseases such as obesity, type 2 diabetes, and atherosclerosis. The activation of inflammation in these diseases depends at least in part on the combined actions of TLR4 signaling and endoplasmic reticulum stress, which by acting in concert can boost the inflammatory response. Defining the mechanisms involved in this phenomenon may unveil potential targets for the treatment of metabolic/inflammatory diseases. Here we used LPS to induce endoplasmic reticulum stress in the human monocyte cell-line, THP-1. The unfolded protein response, produced after LPS, was dependent on CD14 activity but not on RNA-dependent protein kinase and could be inhibited by an exogenous chemical chaperone. The induction of the endoplasmic reticulum resident chaperones, GRP94 and GRP78, by LPS was of a much lower magnitude than the effect of LPS on TLR4 and MD-2 expression. In face of this apparent insufficiency of chaperone expression, we induced the expression of GRP94 and GRP78 by glucose deprivation. This approach completely reverted endoplasmic reticulum stress. The inhibition of either GRP94 or GRP78 with siRNA was sufficient to rescue the protective effect of glucose deprivation on LPS-induced endoplasmic reticulum stress. Thus, insufficient LPS-induced chaperone expression links TLR4 signaling to endoplasmic reticulum stress.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Chaperonas Moleculares/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glucosa/farmacología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Immunoblotting , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fosforilación/efectos de los fármacos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/fisiología , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
17.
Sci Immunol ; 7(68): eabi6763, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35148201

RESUMEN

Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αß) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.


Asunto(s)
Inmunidad Innata/inmunología , Interleucinas/inmunología , eIF-2 Quinasa/inmunología , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , eIF-2 Quinasa/deficiencia
18.
Cell Metab ; 4(6): 491-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17141632

RESUMEN

Mutations in PERK (EIF2AK3) result in permanent neonatal diabetes as well as several other anomalies that underlie the human Wolcott-Rallison syndrome, and these anomalies are mirrored in Perk knockout mice. To identify the cause of diabetes in PERK-deficient mice, we generated a series of tissue- and cell-specific knockouts of the Perk gene and performed a developmental analysis of the progression to overt diabetes. We discovered that PERK is specifically required in the insulin-secreting beta cells during the fetal and early neonatal period as a prerequisite for postnatal glucose homeostasis. However, PERK expression in beta cells is not required at the adult stage to maintain beta cell functions and glucose homeostasis. We show that PERK-deficient mice exhibit severe defects in fetal/neonatal beta cell proliferation and differentiation, resulting in low beta cell mass, defects in proinsulin trafficking, and abrogation of insulin secretion that culminate in permanent neonatal diabetes.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus/enzimología , Glucosa/metabolismo , Homeostasis , Células Secretoras de Insulina/enzimología , eIF-2 Quinasa/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Proliferación Celular , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Feto/enzimología , Feto/patología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Glucosa/genética , Humanos , Recién Nacido , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados , Proinsulina/genética , Proinsulina/metabolismo , eIF-2 Quinasa/deficiencia
19.
J Biol Chem ; 285(27): 20993-1003, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20442408

RESUMEN

Endoplasmic reticulum oxidation 1 (ERO1) is a conserved eukaryotic flavin adenine nucleotide-containing enzyme that promotes disulfide bond formation by accepting electrons from reduced protein disulfide isomerase (PDI) and passing them on to molecular oxygen. Although disulfide bond formation is an essential process, recent experiments suggest a surprisingly broad tolerance to genetic manipulations that attenuate the rate of disulfide bond formation and that a hyperoxidizing ER may place stressed cells at a disadvantage. In this study, we report on the development of a high throughput in vitro assay for mammalian ERO1alpha activity and its application to identify small molecule inhibitors. The inhibitor EN460 (IC(50), 1.9 mum) interacts selectively with the reduced, active form of ERO1alpha and prevents its reoxidation. Despite rapid and promiscuous reactivity with thiolates, EN460 exhibits selectivity for ERO1. This selectivity is explained by the rapid reversibility of the reaction of EN460 with unstructured thiols, in contrast to the formation of a stable bond with ERO1alpha followed by displacement of bound flavin adenine dinucleotide from the active site of the enzyme. Modest concentrations of EN460 and a functionally related inhibitor, QM295, promote signaling in the unfolded protein response and precondition cells against severe ER stress. Together, these observations point to the feasibility of targeting the enzymatic activity of ERO1alpha with small molecule inhibitors.


Asunto(s)
Fibroblastos/fisiología , Glicoproteínas/genética , Animales , Supervivencia Celular , Fibroblastos/citología , Fluorescencia , Glutatión Transferasa/genética , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/metabolismo , Glicoproteínas/fisiología , Cinética , Ratones , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo , Oxidorreductasas , Consumo de Oxígeno , Desnaturalización Proteica , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Espectrometría de Fluorescencia , Espectrofotometría , eIF-2 Quinasa/deficiencia
20.
J Virol ; 84(10): 5043-51, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20219905

RESUMEN

The relevance of translational control in the gene expression and oncotropism of the autonomous parvoviruses was investigated with MVMp, the prototype strain of minute virus of mice (MVM), infecting normal and transformed rodent and human cells of different tissue origins. Mouse embryo fibroblasts (MEFs) and NIH 3T3 fibroblasts were resistant to MVMp infection, but 3T3 fibroblasts derived from double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) knockout mice (PKR(o/o)) behaved in a manner that was highly permissive to productive MVMp replication. NIH 3T3 resistance correlated with significant phosphorylation of eukaryotic translation initiation factor 2 (eIF2) occurring at early time points after infection. Permissive PKR(o/o) cells were converted to MVMp-restrictive cells after reintroduction of the PKR gene by transfection. Conversely, regulated expression of the vaccinia virus E3 protein, a PKR inhibitor, in MEFs prevented eIF2alpha phosphorylation and increased MVMp protein synthesis. In vitro-synthesized genome-length R1 mRNA of MVMp was a potent activator of PKR. Virus-resistant primary MEFs and NIH 3T3 cells responded to MVMp infection with significant increases in eIF2alpha phosphorylation. In contrast, virus-permissive mouse (PKR(o/o), BHK21, and A9) and human transformed (NB324K fibroblast, U373 glioma, and HepG2 hepatoma) cells consistently showed no significant increase in the level of eIF2alpha phosphorylation following MVMp infection. The synthesis of the viral NS1 protein was inversely correlated with the steady-state PKR levels. Our results show that the PKR-mediated antiviral response is an important mechanism for control of productive MVMp infection, and its impairment in human transformed cells allowed efficient MVMp gene expression. PKR translational control may therefore contribute to the oncolysis of MVMp and other autonomous parvoviruses.


Asunto(s)
Virus Diminuto del Ratón/inmunología , Virus Diminuto del Ratón/patogenicidad , Biosíntesis de Proteínas , Proteínas Virales/biosíntesis , Replicación Viral , eIF-2 Quinasa/inmunología , eIF-2 Quinasa/metabolismo , Animales , Línea Celular , Factor 2 Eucariótico de Iniciación/metabolismo , Fibroblastos/virología , Prueba de Complementación Genética , Hepatocitos/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/virología , Tropismo Viral , eIF-2 Quinasa/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA