Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hepatology ; 77(3): E60-E61, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36104065
2.
Front Immunol ; 13: 752215, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35222366

RESUMEN

The four isoforms of the RNA-binding protein hnRNPD/AUF1 have been proposed to limit the use of inflammatory mRNAs in innate immune cells. Mice engineered to lack AUF1s in all tissues are sensitive to acute inflammatory assaults; however, they also manifest complex degenerations obscuring assessment of AUF1s' roles in innate immune cells. Here, we restricted a debilitating AUF1 mutation to the mouse myeloid lineage and performed disease-oriented phenotypic analyses to assess the requirement of AUF1s in variable contexts of innate immune reactivity. Contrary to the whole-body mutants, the myeloid mutants of AUF1s did not show differences in their susceptibility to cytokine storms occurring during endotoxemia; neither in type-I cell-mediated reactions driving intestinal inflammation by chemical irritants. Instead, they were resistant to allergic airway inflammation and displayed reductions in inflammatory infiltrates and an altered T-helper balance. The ex-vivo analysis of macrophages revealed that the loss of AUF1s had a minimal effect on their proinflammatory gene expression. Moreover, AUF1s were dispensable for the classical polarization of cultured macrophages by LPS & IFNγ correlating with the unchanged response of mutant mice to systemic and intestinal inflammation. Notably, AUF1s were also dispensable for the alternative polarization of macrophages by IL4, TGFß and IL10, known to be engaged in allergic reactions. In contrast, they were required to switch proinflammatory macrophages towards a pro-angiogenic phenotype induced by adenosine receptor signals. Congruent to this, the myeloid mutants of AUF1 displayed lower levels of vascular remodeling factors in exudates from allergen exposed lungs; were unable to support the growth and inflammatory infiltration of transplanted melanoma tumors; and failed to vascularize inert grafts unless supplemented with angiogenic factors. Mechanistically, adenosine receptor signals enhanced the association of AUF1s with the Vegfa, Il12b, and Tnf mRNAs to differentially regulate and facilitate the pro-angiogenic switch. Our data collectively demonstrates that AUF1s do not act as general anti-inflammatory factors in innate immune cells but have more specialized roles in regulons allowing specific innate immune cell transitions to support tissue infiltration and remodeling processes.


Asunto(s)
Hipersensibilidad , Neoplasias , Adenosina/metabolismo , Animales , Hipersensibilidad/metabolismo , Inflamación , Pulmón/metabolismo , Macrófagos , Ratones , Células Mieloides/metabolismo , Neoplasias/metabolismo , ARN Mensajero/genética
3.
Mamm Genome ; 33(1): 120-122, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34328547

RESUMEN

Improving reproducibility and replicability in preclinical research is a widely discussed and pertinent topic, especially regarding ethical responsibility in animal research. INFRAFRONTIER, the European Research Infrastructure for the generation, phenotyping, archiving, and distribution of model mammalian genomes, is addressing this issue by developing internal quality principles for its different service areas, that provides a quality framework for its operational activities. This article introduces the INFRAFRONTIER Quality Principles in Systemic Phenotyping of genetically altered mouse models. A total of 11 key principles are included, ranging from general requirements for compliance with guidelines on animal testing, to the need for well-trained personnel and more specific standards such as the exchange of reference lines. Recently established requirements such as the provision of FAIR (Findable, Accessible, Interoperable, Reusable) data are also addressed. For each quality principle, we have outlined the specific context, requirements, further recommendations, and key references.


Asunto(s)
Genoma , Mamíferos , Animales , Modelos Animales de Enfermedad , Ratones , Reproducibilidad de los Resultados
4.
Hepatology ; 75(4): 881-897, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34519101

RESUMEN

BACKGROUND AND AIMS: NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS: Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS: HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.


Asunto(s)
Carcinoma Hepatocelular , Proteína 1 Similar a ELAV , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Animales , Carcinoma Hepatocelular/patología , Proteína 1 Similar a ELAV/metabolismo , Homeostasis , Inflamación/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología , ARN , Triglicéridos/metabolismo
5.
Oncogene ; 40(36): 5518-5532, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34294847

RESUMEN

In response to oncogenic signals, Alternative Splicing (AS) regulators such as SR and hnRNP proteins show altered expression levels, subnuclear distribution and/or post-translational modification status, but the link between signals and these changes remains unknown. Here, we report that a cytosolic scaffold protein, IQGAP1, performs this task in response to heat-induced signals. We show that in gastric cancer cells, a nuclear pool of IQGAP1 acts as a tethering module for a group of spliceosome components, including hnRNPM, a splicing factor critical for the response of the spliceosome to heat-shock. IQGAP1 controls hnRNPM's sumoylation, subnuclear localisation and the relevant response of the AS machinery to heat-induced stress. Genome-wide analyses reveal that IQGAP1 and hnRNPM co-regulate the AS of a cell cycle-related RNA regulon in gastric cancer cells, thus favouring the accelerated proliferation phenotype of gastric cancer cells. Overall, we reveal a missing link between stress signals and AS regulation.


Asunto(s)
Neoplasias Gástricas , Empalme Alternativo , Estudio de Asociación del Genoma Completo , Humanos , Estómago , Proteínas Activadoras de ras GTPasa
6.
Nat Immunol ; 21(6): 626-635, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32424362

RESUMEN

The inflammasome NLRP6 plays a crucial role in regulating inflammation and host defense against microorganisms in the intestine. However, the molecular mechanisms by which NLRP6 function is inhibited to prevent excessive inflammation remain unclear. Here, we demonstrate that the deubiquitinase Cyld prevents excessive interleukin 18 (IL-18) production in the colonic mucosa by deubiquitinating NLRP6. We show that deubiquitination inhibited the NLRP6-ASC inflammasome complex and regulated the maturation of IL-18. Cyld deficiency in mice resulted in elevated levels of active IL-18 and severe colonic inflammation following Citrobacter rodentium infection. Further, in patients with ulcerative colitis, the concentration of active IL-18 was inversely correlated with CYLD expression. Thus, we have identified a novel regulatory mechanism that inhibits the NLRP6-IL-18 pathway in intestinal inflammation.


Asunto(s)
Enzima Desubiquitinante CYLD/metabolismo , Enterocolitis/etiología , Enterocolitis/metabolismo , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Citrobacter rodentium , Enzima Desubiquitinante CYLD/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Enterocolitis/patología , Expresión Génica , Humanos , Interleucina-18/antagonistas & inhibidores , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Unión Proteica/inmunología , Ubiquitinación
7.
J Clin Invest ; 130(6): 3188-3204, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32125284

RESUMEN

As there is growing evidence for the tumor microenvironment's role in tumorigenesis, we investigated the role of fibroblast-expressed kinases in triple-negative breast cancer (TNBC). Using a high-throughput kinome screen combined with 3D invasion assays, we identified fibroblast-expressed PIK3Cδ (f-PIK3Cδ) as a key regulator of cancer progression. Although PIK3Cδ was expressed in primary fibroblasts derived from TNBC patients, it was barely detectable in breast cancer (BC) cell lines. Genetic and pharmacological gain- and loss-of-function experiments verified the contribution of f-PIK3Cδ in TNBC cell invasion. Integrated secretomics and transcriptomics analyses revealed a paracrine mechanism via which f-PIK3Cδ confers its protumorigenic effects. Inhibition of f-PIK3Cδ promoted the secretion of factors, including PLGF and BDNF, that led to upregulation of NR4A1 in TNBC cells, where it acts as a tumor suppressor. Inhibition of PIK3Cδ in an orthotopic BC mouse model reduced tumor growth only after inoculation with fibroblasts, indicating a role of f-PIK3Cδ in cancer progression. Similar results were observed in the MMTV-PyMT transgenic BC mouse model, along with a decrease in tumor metastasis, emphasizing the potential immune-independent effects of PIK3Cδ inhibition. Finally, analysis of BC patient cohorts and TCGA data sets identified f-PIK3Cδ (protein and mRNA levels) as an independent prognostic factor for overall and disease-free survival, highlighting it as a therapeutic target for TNBC.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/biosíntesis , Fibroblastos/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Mama Triple Negativas/enzimología , Animales , Fosfatidilinositol 3-Quinasa Clase I/genética , Femenino , Fibroblastos/patología , Xenoinjertos , Humanos , Ratones , Ratones Transgénicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
8.
J Autoimmun ; 104: 102334, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604649

RESUMEN

AU-rich elements (AREs) comprise one of the most widely studied families of regulatory RNA structures met in RNAs engaged in complex immunological reactions. A multitude of genetic, molecular, holistic and functional studies have been utilized for the analyses of the AREs and their interactions to proteins that bind to them. Data stemming from these studies brought forth a world of RNA-related check-points against infection, chronic inflammation, tumor associated immunity, and autoimmunity; and the interest to capitalize the interactions of AREs for clinical management and therapy. They also provided lessons on the cellular capabilities of post-transcriptional control. Originally thought as transcript-restricted regulators of turnover and translation, ARE-binding proteins do in fact harbor great versatility and interactivity across nuclear and cytoplasmic compartments; and act as functional coordinators of immune-cellular programs. Harnessing these deterministic functions requires extensive knowledge of their synergies or antagonisms at a cell-specific level; but holds great promise since it can provide the efficacy of combinatorial therapies with single agents.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/efectos de los fármacos , Enfermedades Autoinmunes/inmunología , Regulación de la Expresión Génica/inmunología , Proteínas de Unión al ARN/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/terapia , Enfermedad Crónica , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Proteínas de Unión al ARN/genética
9.
Nat Commun ; 10(1): 4171, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519904

RESUMEN

The master posttranscriptional regulator HuR promotes muscle fiber formation in cultured muscle cells. However, its impact on muscle physiology and function in vivo is still unclear. Here, we show that muscle-specific HuR knockout (muHuR-KO) mice have high exercise endurance that is associated with enhanced oxygen consumption and carbon dioxide production. muHuR-KO mice exhibit a significant increase in the proportion of oxidative type I fibers in several skeletal muscles. HuR mediates these effects by collaborating with the mRNA decay factor KSRP to destabilize the PGC-1α mRNA. The type I fiber-enriched phenotype of muHuR-KO mice protects against cancer cachexia-induced muscle loss. Therefore, our study uncovers that under normal conditions HuR modulates muscle fiber type specification by promoting the formation of glycolytic type II fibers. We also provide a proof-of-principle that HuR expression can be targeted therapeutically in skeletal muscles to combat cancer-induced muscle wasting.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Neoplasias/complicaciones , Animales , Línea Celular , Línea Celular Tumoral , Estudios Transversales , Proteína 1 Similar a ELAV/genética , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados
10.
Front Immunol ; 9: 2732, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30532756

RESUMEN

HuR is an abundant RNA-binding protein acting as a post-transcriptional regulator of many RNAs including mRNAs encoding inflammatory mediators, cytokines, death signalers and cell cycle regulators. In the context of intestinal pathologies, elevated HuR is considered to enhance the stability and the translation of pro-tumorigenic mRNAs providing the rationale for its pharmacological targeting. However, HuR also possesses specific regulatory functions for innate immunity and cytokine mRNA control which can oppose intestinal inflammation and tumor promotion. Here, we aim to identify contexts of intestinal inflammation where the innate immune and the epithelial functions of HuR converge or diverge. To address this, we use a disease-oriented phenotypic approach using mice lacking HuR either in intestinal epithelia or myeloid-derived immune compartments. These mice were compared for their responses to (a) Chemically induced Colitis; (b) Colitis- associated Cancer (CAC); (c) T-cell mediated enterotoxicity; (d) Citrobacter rodentium-induced colitis; and (e) TNF-driven inflammatory bowel disease. Convergent functions of epithelial and myeloid HuR included their requirement for suppressing inflammation in chemically induced colitis and their redundancies in chronic TNF-driven IBD and microbiota control. In the other contexts however, their functions diversified. Epithelial HuR was required to protect the epithelial barrier from acute inflammatory or infectious degeneration but also to promote tumor growth. In contrast, myeloid HuR was required to suppress the beneficial inflammation for pathogen clearance and tumor suppression. This cellular dichotomy in HuR's functions was validated further in mice engineered to express ubiquitously higher levels of HuR which displayed diminished pathologic and beneficial inflammatory responses, resistance to epithelial damage yet a heightened susceptibility to CAC. Our study demonstrates that epithelial and myeloid HuR affect different cellular dynamics in the intestine that need to be carefully considered for its pharmacological exploitation and points toward potential windows for harnessing HuR functions in intestinal inflammation.


Asunto(s)
Citrobacter rodentium/inmunología , Colitis/inmunología , Proteína 1 Similar a ELAV/inmunología , Infecciones por Enterobacteriaceae/inmunología , Mucosa Intestinal/inmunología , Animales , Colitis/genética , Colitis/microbiología , Colitis/patología , Proteína 1 Similar a ELAV/genética , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Transgénicos
11.
12.
Cell Oncol (Dordr) ; 39(3): 287-93, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27042826

RESUMEN

PURPOSE: CYLD is a tumor suppressor that has been linked to the development of various human malignancies, including colon cancer. The tumor-suppressing function of CYLD is associated with its deubiquitinating activity, which maps to the carboxyl-terminal region of the protein. In the present study we evaluated the role of intestinal epithelial CYLD in colitis-associated cancer using a conditional mouse CYLD inactivation model. METHODS: In order to evaluate the role of CYLD in intestinal epithelial carcinogenesis, mice (IEC-Cyld (Δ9) mice) that carry a mutation that eliminates the deubiquitinating domain of CYLD in intestinal epithelial cells (IEC) were generated by crossing Villin-Cre transgenic mice to previously generated mice carrying a loxP-flanked Cyld exon 9 (Cyld (flx9) mice). RESULTS: We found that IEC-Cyld (Δ9) mice did not present spontaneous intestinal abnormalities up to one year of age. However, upon challenge with a combination of genotoxic (AOM) and pro-inflammatory (DSS) agents we found that the number of adenomas in the IEC-Cyld (Δ9) mice was dramatically increased compared to the control mice. Inactivation of CYLD in intestinal epithelial cells did not affect the classical nuclear factor-kappaB (NF-κB) and c-Jun kinase (JNK) activation pathways under physiological conditions, suggesting that these pathways do not predispose CYLD-deficient intestinal epithelia to colorectal cancer development before the onset of genotoxic and/or pro-inflammatory stress. CONCLUSIONS: Our findings underscore a critical tumor-suppressing role for functional intestinal epithelial CYLD in colitis-associated carcinogenesis. CYLD expression and its associated pathways in intestinal tumors may be exploited for future prognostic and therapeutic purposes.


Asunto(s)
Carcinogénesis/genética , Colitis/complicaciones , Neoplasias Colorrectales/genética , Cisteína Endopeptidasas/genética , Mucosa Intestinal/patología , Animales , Colitis/genética , Enzima Desubiquitinante CYLD , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Nat Immunol ; 16(4): 415-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25706746

RESUMEN

Post-transcriptional regulation of mRNA by the RNA-binding protein HuR (encoded by Elavl1) is required in B cells for the germinal center reaction and for the production of class-switched antibodies in response to thymus-independent antigens. Transcriptome-wide examination of RNA isoforms and their abundance and translation in HuR-deficient B cells, together with direct measurements of HuR-RNA interactions, revealed that HuR-dependent splicing of mRNA affected hundreds of transcripts, including that encoding dihydrolipoamide S-succinyltransferase (Dlst), a subunit of the 2-oxoglutarate dehydrogenase (α-KGDH) complex. In the absence of HuR, defective mitochondrial metabolism resulted in large amounts of reactive oxygen species and B cell death. Our study shows how post-transcriptional processes control the balance of energy metabolism required for the proliferation and differentiation of B cells.


Asunto(s)
Linfocitos B/inmunología , Proteínas ELAV/inmunología , Centro Germinal/inmunología , Inmunidad Humoral , Inmunoglobulinas/biosíntesis , ARN Mensajero/inmunología , Aciltransferasas/genética , Aciltransferasas/inmunología , Empalme Alternativo/inmunología , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Muerte Celular , Diferenciación Celular , Proliferación Celular , Proteínas ELAV/genética , Eritrocitos/inmunología , Centro Germinal/citología , Centro Germinal/efectos de los fármacos , Inmunización , Cambio de Clase de Inmunoglobulina , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/inmunología , ARN Mensajero/genética , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Ovinos
14.
Proc Natl Acad Sci U S A ; 111(36): E3815-24, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25157170

RESUMEN

Precise spatiotemporal control of mRNA translation machinery is essential to the development of highly complex systems like the neocortex. However, spatiotemporal regulation of translation machinery in the developing neocortex remains poorly understood. Here, we show that an RNA-binding protein, Hu antigen R (HuR), regulates both neocorticogenesis and specificity of neocortical translation machinery in a developmental stage-dependent manner in mice. Neocortical absence of HuR alters the phosphorylation states of initiation and elongation factors in the core translation machinery. In addition, HuR regulates the temporally specific positioning of functionally related mRNAs into the active translation sites, the polysomes. HuR also determines the specificity of neocortical polysomes by defining their combinatorial composition of ribosomal proteins and initiation and elongation factors. For some HuR-dependent proteins, the association with polysomes likewise depends on the eukaryotic initiation factor 2 alpha kinase 4, which associates with HuR in prenatal developing neocortices. Finally, we found that deletion of HuR before embryonic day 10 disrupts both neocortical lamination and formation of the main neocortical commissure, the corpus callosum. Our study identifies a crucial role for HuR in neocortical development as a translational gatekeeper for functionally related mRNA subgroups and polysomal protein specificity.


Asunto(s)
Proteínas ELAV/metabolismo , Neocórtex/metabolismo , Polirribosomas/metabolismo , Biosíntesis de Proteínas , Proteínas de Unión al ARN/metabolismo , Animales , Cuerpo Calloso/embriología , Cuerpo Calloso/metabolismo , Proteína 1 Similar a ELAV , Factor 2 Eucariótico de Iniciación/metabolismo , Eliminación de Gen , Técnicas de Inactivación de Genes , Ratones , Mitosis , Modelos Biológicos , Neocórtex/embriología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Neuroepiteliales/metabolismo , Neurogénesis , Neuroglía/metabolismo , Neuronas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Ribosómicas/metabolismo , Factores de Tiempo , Transcripción Genética
15.
Nat Immunol ; 15(6): 492-502, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24840980

RESUMEN

Immunological reactions are propelled by ever-changing signals that alter the translational ability of the RNA in the cells involved. Such alterations are considered to be consequential modifications in the transcriptomic decoding of the genetic blueprint. The identification of RNA-binding protein (RBP) assemblies engaged in the coordinative regulation of state-specific RNAs indicates alternative and exclusive means for determining the activation, plasticity and tolerance of cells of the immune system. Here we review current knowledge about RBP-regulated post-transcriptional events involved in the reactivity of cells of the immune system and the importance of their alteration during chronic inflammatory pathology and autoimmunity.


Asunto(s)
Inmunidad Celular/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico/genética , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Humanos , Tolerancia Inmunológica/genética , Inmunidad Celular/inmunología , Inflamación/genética , Inflamación/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Linfopoyesis/genética , Linfopoyesis/inmunología , Ratones , Proteínas de Unión al ARN/genética , Secuencias Reguladoras de Ácido Ribonucleico/inmunología , Transducción de Señal/inmunología
16.
Mol Cell Biol ; 33(10): 1886-900, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23508105

RESUMEN

p19(ARF) plays an essential role in the senescence of mouse cells, and its expression is lost by methylation or deletion of the ARF locus; otherwise, p53 is inactivated to bypass senescence. ARF expression is tightly regulated, but little is known about its posttranscriptional regulation. Here, we show that an RNA-binding protein, HuR (human antigen R), represses ARF mRNA translation, thereby maintaining the replicative life span of mouse embryonic fibroblasts (MEFs). Loss of HuR results in premature senescence, with concomitant increases in p19(ARF) but not p16(Ink4a) levels, and this senescence is not observed in ARF-null MEFs that retain an intact Ink4a locus. HuR depletion does not alter ARF transcription or stability but enhances ribosome association with ARF mRNA. Under these conditions, ARF mRNA accumulates in nucleoli, where it associates with nucleolin. Furthermore, adipose-specific deletion of the HuR gene results in increased p19(ARF) expression in aged animals, which is accompanied by decreased insulin sensitivity. Together, our findings demonstrate that p19(ARF) is also regulated at the translational level, and this translational regulation restrains the cellular life span and tissue functions in vivo.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas ELAV/fisiología , Fibroblastos/fisiología , Biosíntesis de Proteínas , Regiones no Traducidas 5' , Adipogénesis , Animales , Proliferación Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Resistencia a la Insulina , Ratones , Ratones Noqueados , Células 3T3 NIH , Fosfoproteínas/metabolismo , Unión Proteica , Estabilidad Proteica , Transporte de Proteínas , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/metabolismo , Nucleolina
17.
Artículo en Inglés | MEDLINE | ID: mdl-22761012

RESUMEN

Inflammation involves a continuum of intercellular interactions and cellular responses targeting infectious or tissue damage while maintaining homeostasis. At its core, this continuum encompasses the alternating phenotypes of innate immune cells; each phenotype is typified by the expression of molecules which either support host defence or aid tissue restoration and the resolution of inflammation. The aberrant persistence of any such phenotype can drive chronic inflammatory pathology. For macrophages, these phenotypes arise as changes in cellular plasticity because of adaptation. As such their underlying gene expression programs may not be determined by robust transcriptomic and epigenetic programs but by more flexible means like post-transcriptional mechanisms affecting mRNA use. These mechanisms require the assemblies of RNA-binding proteins (RBPs) and non-coding RNAs onto specific elements on their RNA targets in Ribonucleoprotein particles (RNPs) which control mRNA maturation, turnover and translation. The collection of RNPs within a cell defines the ribonome, that is, a high order system of coordinative post-transcriptional determination. mRNAs involved in the definition of different macrophage activation phenotypes share elements of RBP recognition rendering them amenable to ribonomic regulation. The molecular features of their cognitive RBPs and the pathologies developing in the corresponding mouse mutants support their involvement in inflammatory reactions. We view this information in the context of macrophage activation states to propose that these states can be determined via differential--synergistic or antagonistic--RNP associations. In doing so, we substantiate the need for the use of systems platforms to model RNP hierarchies controlling the continuum of inflammation.


Asunto(s)
Macrófagos/metabolismo , Animales , Antígenos de Superficie/metabolismo , Proteínas ELAV/metabolismo , Proteína 1 Similar a ELAV , Epigénesis Genética , Ribonucleoproteína Nuclear Heterogénea D0 , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos/inmunología , Ratones , MicroARNs/metabolismo , Modelos Moleculares , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN no Traducido/metabolismo , Proteínas de Unión al ARN/metabolismo
18.
J Clin Invest ; 122(1): 48-61, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22201685

RESUMEN

The innate immune response involves a variety of inflammatory reactions that can result in inflammatory disease and cancer if they are not resolved and instead are allowed to persist. The effective activation and resolution of innate immune responses relies on the production and posttranscriptional regulation of mRNAs encoding inflammatory effector proteins. The RNA-binding protein HuR binds to and regulates such mRNAs, but its exact role in inflammation remains unclear. Here we show that HuR maintains inflammatory homeostasis by controlling macrophage plasticity and migration. Mice lacking HuR in myeloid-lineage cells, which include many of the cells of the innate immune system, displayed enhanced sensitivity to endotoxemia, rapid progression of chemical-induced colitis, and severe susceptibility to colitis-associated cancer. The myeloid cell-specific HuR-deficient mice had an exacerbated inflammatory cytokine profile and showed enhanced CCR2-mediated macrophage chemotaxis. At the molecular level, activated macrophages from these mice showed enhancements in the use of inflammatory mRNAs (including Tnf, Tgfb, Il10, Ccr2, and Ccl2) due to a lack of inhibitory effects on their inducible translation and/or stability. Conversely, myeloid overexpression of HuR induced posttranscriptional silencing, reduced inflammatory profiles, and protected mice from colitis and cancer. Our results highlight the role of HuR as a homeostatic coordinator of mRNAs that encode molecules that guide innate inflammatory effects and demonstrate the potential of harnessing the effects of HuR for clinical benefit against pathologic inflammation and cancer.


Asunto(s)
Colitis/prevención & control , Neoplasias Colorrectales/prevención & control , Proteínas ELAV/fisiología , Células Mieloides/fisiología , Animales , Colitis/genética , Colitis/inmunología , Colitis/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Proteínas ELAV/deficiencia , Proteínas ELAV/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endotoxemia/etiología , Endotoxemia/inmunología , Endotoxemia/prevención & control , Inmunidad Innata , Inflamación/etiología , Inflamación/inmunología , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Activación de Macrófagos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/patología , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/inmunología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Nat Immunol ; 12(10): 923-5, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21934669

RESUMEN

ASC has emerged as an adaptor for inflammasome sensors in cells of the innate immune response. New inflammasome-independent roles have been identified for ASC in the control of adaptive immunity; these include the post-transcriptional regulation of cytoskeletal rearrangements.


Asunto(s)
Actinas/química , Proteínas del Citoesqueleto/fisiología , Proteínas Activadoras de GTPasa/fisiología , Inflamasomas/fisiología , Proteínas de Unión al GTP rac/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Factores de Intercambio de Guanina Nucleótido
20.
Mol Biol Cell ; 22(16): 2875-85, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21737689

RESUMEN

Posttranscriptional mechanisms are crucial to regulate spermatogenesis. Accurate protein synthesis during germ cell development relies on RNA binding proteins that control the storage, stability, and translation of mRNAs in a tightly and temporally regulated manner. Here, we focused on the RNA binding protein Embryonic Lethal Abnormal Vision (ELAV) L1/Human antigen R (HuR) known to be a key regulator of posttranscriptional regulation in somatic cells but the function of which during gametogenesis has never been investigated. In this study, we have used conditional loss- and gain-of-function approaches to address this issue in mice. We show that targeted deletion of HuR specifically in germ cells leads to male but not female sterility. Mutant males are azoospermic because of the extensive death of spermatocytes at meiotic divisions and failure of spermatid elongation. The latter defect is also observed upon HuR overexpression. To elucidate further the molecular mechanisms underlying spermatogenesis defects in HuR-deleted and -overexpressing testes, we undertook a target gene approach and discovered that heat shock protein (HSP)A2/HSP70-2, a crucial regulator of spermatogenesis, was down-regulated in both situations. HuR specifically binds hspa2 mRNA and controls its expression at the translational level in germ cells. Our study provides the first genetic evidence of HuR involvement during spermatogenesis and reveals Hspa2 as a target for HuR.


Asunto(s)
Antígenos de Superficie/metabolismo , Meiosis , Proteínas de Unión al ARN/metabolismo , Espermatogénesis , Testículo/citología , Animales , Antígenos de Superficie/genética , Puntos de Control del Ciclo Celular , Diferenciación Celular , Proteínas ELAV , Proteína 1 Similar a ELAV , Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Infertilidad Masculina , Masculino , Ratones , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Espermátides/citología , Espermátides/metabolismo , Espermátides/fisiología , Espermatocitos/citología , Espermatocitos/metabolismo , Espermatocitos/fisiología , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...