Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 19(8): 828-837, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29988089

RESUMEN

Memory T cells are critical for the immune response to recurring infections. Their instantaneous reactivity to pathogens is empowered by the persistent expression of cytokine-encoding mRNAs. How the translation of proteins from pre-formed cytokine-encoding mRNAs is prevented in the absence of infection has remained unclear. Here we found that protein production in memory T cells was blocked via a 3' untranslated region (3' UTR)-mediated process. Germline deletion of AU-rich elements (AREs) in the Ifng-3' UTR led to chronic cytokine production in memory T cells. This aberrant protein production did not result from increased expression and/or half-life of the mRNA. Instead, AREs blocked the recruitment of cytokine-encoding mRNA to ribosomes; this block depended on the ARE-binding protein ZFP36L2. Thus, AREs mediate repression of translation in mouse and human memory T cells by preventing undesirable protein production from pre-formed cytokine-encoding mRNAs in the absence of infection.


Asunto(s)
Regiones no Traducidas 3'/genética , Elementos Ricos en Adenilato y Uridilato/genética , Interferón gamma/genética , ARN Mensajero/genética , Linfocitos T/inmunología , Animales , Células Cultivadas , Represión Epigenética , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Extensión de la Cadena Peptídica de Translación , Ribosomas/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo
2.
Nat Immunol ; 15(1): 72-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24241692

RESUMEN

IFNL3, which encodes interferon-λ3 (IFN-λ3), has received considerable attention in the hepatitis C virus (HCV) field, as many independent genome-wide association studies have identified a strong association between polymorphisms near IFNL3 and clearance of HCV. However, the mechanism underlying this association has remained elusive. In this study, we report the identification of a functional polymorphism (rs4803217) in the 3' untranslated region (UTR) of IFNL3 mRNA that dictated transcript stability. We found that this polymorphism influenced AU-rich element (ARE)-mediated decay (AMD) of IFNL3 mRNA, as well as the binding of HCV-induced microRNAs during infection. Together these pathways mediated robust repression of the unfavorable IFNL3 polymorphism. Our data reveal a previously unknown mechanism by which HCV attenuates the antiviral response and indicate new potential therapeutic targets for HCV treatment.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Interleucinas/genética , MicroARNs/genética , Polimorfismo de Nucleótido Simple , Estabilidad del ARN/genética , Regiones no Traducidas 3'/genética , Secuencia de Bases , Línea Celular Tumoral , Citometría de Flujo , Genotipo , Células Hep G2 , Hepacivirus/fisiología , Hepatitis C/genética , Hepatitis C/virología , Interacciones Huésped-Patógeno , Humanos , Interferones , Interleucinas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico
3.
Proc Natl Acad Sci U S A ; 116(8): 2935-2944, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30718402

RESUMEN

Human antigen R (HuR) is a key regulator of cellular mRNAs containing adenylate/uridylate-rich elements (AU-rich elements; AREs). These are a major class of cis elements within 3' untranslated regions, targeting these mRNAs for rapid degradation. HuR contains three RNA recognition motifs (RRMs): a tandem RRM1 and 2, followed by a flexible linker and a C-terminal RRM3. While RRM1 and 2 are structurally characterized, little is known about RRM3. Here we present a 1.9-Å-resolution crystal structure of RRM3 bound to different ARE motifs. This structure together with biophysical methods and cell-culture assays revealed the mechanism of RRM3 ARE recognition and dimerization. While multiple RNA motifs can be bound, recognition of the canonical AUUUA pentameric motif is possible by binding to two registers. Additionally, RRM3 forms homodimers to increase its RNA binding affinity. Finally, although HuR stabilizes ARE-containing RNAs, we found that RRM3 counteracts this effect, as shown in a cell-based ARE reporter assay and by qPCR with native HuR mRNA targets containing multiple AUUUA motifs, possibly by competing with RRM12.


Asunto(s)
Proteínas ELAV/química , Proteína 1 Similar a ELAV/química , Motivo de Reconocimiento de ARN/genética , Proteínas de Unión al ARN/química , Regiones no Traducidas 3' , Elementos Ricos en Adenilato y Uridilato/genética , Cristalografía por Rayos X , Dimerización , Proteína 1 Similar a ELAV/genética , Humanos , Espectroscopía de Resonancia Magnética , Proteínas de Unión al ARN/genética , Ribonucleósido Difosfato Reductasa/química , Proteínas Supresoras de Tumor/química
4.
Immunology ; 164(1): 57-72, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33884612

RESUMEN

As part of the adaptive immune system, T cells are vital for the eradication of infected and malignantly transformed cells. To perform their protective function, T cells produce effector molecules that are either directly cytotoxic, such as granzymes, perforin, interferon-γ and tumour necrosis factor α, or attract and stimulate (immune) cells, such as interleukin-2. As these molecules can also induce immunopathology, tight control of their production is required. Indeed, inflammatory cytokine production is regulated on multiple levels. Firstly, locus accessibility and transcription factor availability and activity determine the amount of mRNA produced. Secondly, post-transcriptional mechanisms, influencing mRNA splicing/codon usage, stability, decay, localization and translation rate subsequently determine the amount of protein that is produced. In the immune suppressive environments of tumours, T cells gradually lose the capacity to produce effector molecules, resulting in tumour immune escape. Recently, the role of post-transcriptional regulation in fine-tuning T-cell effector function has become more appreciated. Furthermore, several groups have shown that exhausted or dysfunctional T cells from cancer patients or murine models possess mRNA for inflammatory mediators, but fail to produce effector molecules, hinting that post-transcriptional events also play a role in hampering tumour-infiltrating lymphocyte effector function. Here, the post-transcriptional regulatory events governing T-cell cytokine production are reviewed, with a specific focus on the importance of post-transcriptional regulation in anti-tumour responses. Furthermore, potential approaches to circumvent tumour-mediated dampening of T-cell effector function through the (dis)engagement of post-transcriptional events are explored, such as CRISPR/Cas9-mediated genome editing or chimeric antigen receptors.


Asunto(s)
Inmunoterapia/tendencias , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Procesamiento Postranscripcional del ARN/inmunología , Linfocitos T/inmunología , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Edición Génica , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Microambiente Tumoral
6.
J Autoimmun ; 111: 102436, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32220507

RESUMEN

Low grade, chronic inflammation is a critical risk factor for immunologic dysfunction including autoimmune diseases. However, the multiplicity of complex mechanisms and lack of relevant murine models limit our understanding of the precise role of chronic inflammation. To address these hurdles, we took advantage of multi-omics data and a unique murine model with a low but chronic expression of IFN-γ, generated by replacement of the AU-rich element (ARE) in the 3' UTR region of IFN-γ mRNA with random nucleotides. Herein, we demonstrate that low but differential expression of IFN-γ in mice by homozygous or heterozygous ARE replacement triggers distinctive gut microbial alterations, of which alteration is female-biased with autoimmune-associated microbiota. Metabolomics data indicates that gut microbiota-dependent metabolites have more robust sex-differences than microbiome profiling, particularly those involved in fatty acid oxidation and nuclear receptor signaling. More importantly, homozygous ARE-Del mice have dramatic changes in tryptophan metabolism, bile acid and long-chain lipid metabolism, which interact with gut microbiota and nuclear receptor signaling similarly with sex-dependent metabolites. Consistent with these findings, nuclear receptor signaling, encompassing molecules such as PPARs, FXR, and LXRs, was detectable as a top canonical pathway in comparison of blood and tissue-specific gene expression between female homozygous vs heterozygous ARE-Del mice. Further analysis implies that dysregulated autophagy in macrophages is critical for breaking self-tolerance and gut homeostasis, while pathways interact with nuclear receptor signaling to regulate inflammatory responses. Overall, pathway-based integration of multi-omics data provides systemic and cellular insights about how chronic inflammation driven by IFN-γ results in the development of autoimmune diseases with specific etiopathological features.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Disbiosis/inmunología , Inflamación/inmunología , Interferón gamma/metabolismo , Macrófagos/inmunología , Regiones no Traducidas 3'/genética , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Autofagia , Enfermedad Crónica , Femenino , Microbioma Gastrointestinal/inmunología , Interferón gamma/genética , Masculino , Ratones , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/metabolismo , Sexismo , Transducción de Señal
7.
J Mol Cell Cardiol ; 129: 247-256, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30880251

RESUMEN

Inflammation has recently gained tremendous attention as a key contributor in several chronic diseases. While physiological inflammation is essential to counter a wide variety of damaging stimuli and to improve wound healing, dysregulated inflammation such as in the myocardium and vasculature can promote cardiovascular diseases. Given the high severity, prevalence, and economic burden of these diseases, understanding the factors involved in the regulation of physiological inflammation is essential. Like other complex biological phenomena, RNA-based processes are emerging as major regulators of inflammatory responses. Among such processes are cis-regulatory elements in the mRNA of inflammatory genes, noncoding RNAs directing the production or localization of inflammatory cytokines/chemokines, or pathogenic RNA driving inflammatory responses. In this review, we describe several specific RNA-based molecular mechanisms by which physiological inflammation pertaining to cardiovascular diseases is regulated. These include the role of AU-rich element-containing mRNAs, long non-coding RNAs, microRNAs, and viral RNAs.


Asunto(s)
Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patología , Inflamación/genética , Inflamación/patología , ARN/metabolismo , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Humanos , Modelos Biológicos , ARN/genética , ARN Viral/genética , ARN Viral/metabolismo
8.
RNA ; 23(8): 1209-1223, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28455422

RESUMEN

ZFP36L2 (L2) destabilizes AU-rich element (ARE)-containing transcripts and has been implicated in female fertility. We have shown that only one of three putative AREs within the 3' UTR of murine luteinizing hormone receptor mRNA, ARE2197 (UAUUUAU), is capable of interacting with L2. To assess whether structural elements of ARE2197 could explain this unique binding ability, we performed whole-transcript SHAPE-MaP (selective 2' hydroxyl acylation by primer extension-mutational profiling) of the full-length mLHR mRNA. The data revealed that the functional ARE2197 is located in a hairpin loop structure and most nucleotides are highly reactive. In contrast, each of the nonbinding AREs, 2301 and 2444, contains only a pentamer AUUUA; and in ARE2301 much of the ARE sequence is poorly accessible. Because the functional mARE was also found to be conserved in humans at the sequence level (ARE 2223), we decided to investigate whether binding and structure are also preserved. Similar to mouse, only one ARE in hLHR mRNA is capable of binding to L2; and it is also located in a hairpin structure, based on our SHAPE-MaP data. To investigate the role of secondary structure in the binding, we mutated specific nucleotides in both functional AREs. Mutations in the flexible stem region proximal to the loop that enforce strong base-pairing, drastically reduced L2 binding affinity; this confirms that the structural context is critical for L2 recognition of hARE2223. Collectively, our results suggest that a combination of minimal ARE sequence, placement of the ARE in a hairpin loop, and stem flexibility mediate high-affinity L2 binding to hLHR mRNA.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , ARN Mensajero/metabolismo , Receptores de HL/metabolismo , Tristetraprolina/metabolismo , Animales , Emparejamiento Base , Secuencia de Bases , Humanos , Ratones , Mutación/genética , Conformación de Ácido Nucleico , ARN Mensajero/química , ARN Mensajero/genética , Receptores de HL/genética , Alineación de Secuencia , Tristetraprolina/química , Tristetraprolina/genética
9.
Biochim Biophys Acta ; 1861(11): 1816-1827, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27616329

RESUMEN

Cardiolipin and phosphatidic acid-binding protein (CLPABP) is a pleckstrin homology domain-containing protein and is localized on the surface of mitochondria of cultured cells as a large protein-RNA complex. To analyze the physiological functions of CLPABP, we established and characterized a CLPABP knockout (KO) mouse. Although expression levels of CLPABP transcripts in the developmental organs were high, CLPABP KO mice were normal at birth and grew normally when young. However, old male mice presented a fatty phenotype, similar to that seen in metabolic syndrome, in parallel with elevated male- and age-dependent CLPABP gene expression. One of the reasons for this obesity in CLPABP KO mice is dependence on increases in leptin concentration in plasma. The leptin transcripts were also upregulated in the adipose tissue of KO mice compared with wild-type (WT) mice. To understand the difference in levels of the transcriptional product, we focused on the effect of CLPABP on the stability of mRNA involving an AU-rich element (ARE) in its 3'UTR dependence on the RNA stabilizer, human antigen R (HuR), which is one of the CLPABP-binding proteins. Increase in stability of ARE-containing mRNAs of leptin by HuR was antagonized by the expression of CLPABP in cultured cells. Depletion of CLPABP disturbed the normal subcellular localization of HuR to stress granules, and overexpression of CLPABP induced instability of leptin mRNA by inhibiting HuR function. Consequently, leptin levels in old male mice might be regulated by CLPABP expression, which might lead to body weight control.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Envejecimiento/genética , Proteínas ELAV/metabolismo , Leptina/genética , Proteínas Ligadas a Lípidos/metabolismo , Obesidad/genética , Estabilidad del ARN/genética , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Eliminación de Gen , Regulación de la Expresión Génica , Leptina/metabolismo , Proteínas Ligadas a Lípidos/genética , Masculino , Metaboloma , Ratones Noqueados , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/genética , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Fracciones Subcelulares/metabolismo , Transcripción Genética
10.
PLoS Pathog ; 11(1): e1004597, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25569678

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the infectious cause of several AIDS-related cancers, including the endothelial cell (EC) neoplasm Kaposi's sarcoma (KS). KSHV-infected ECs secrete abundant host-derived pro-inflammatory molecules and angiogenic factors that contribute to tumorigenesis. The precise contributions of viral gene products to this secretory phenotype remain to be elucidated, but there is emerging evidence for post-transcriptional regulation. The Kaposin B (KapB) protein is thought to contribute to the secretory phenotype in infected cells by binding and activating the stress-responsive kinase MK2, thereby selectively blocking decay of AU-rich mRNAs (ARE-mRNAs) encoding pro-inflammatory cytokines and angiogenic factors. Processing bodies (PBs) are cytoplasmic ribonucleoprotein foci in which ARE-mRNAs normally undergo rapid 5' to 3' decay. Here, we demonstrate that PB dispersion is a feature of latent KSHV infection, which is dependent on kaposin protein expression. KapB is sufficient to disperse PBs, and KapB-mediated ARE-mRNA stabilization could be partially reversed by treatments that restore PBs. Using a combination of genetic and chemical approaches we provide evidence that KapB-mediated PB dispersion is dependent on activation of a non-canonical Rho-GTPase signaling axis involving MK2, hsp27, p115RhoGEF and RhoA. PB dispersion in latently infected cells is likewise dependent on p115RhoGEF. In addition to PB dispersion, KapB-mediated RhoA activation in primary ECs caused actin stress fiber formation, increased cell motility and angiogenesis; these effects were dependent on the activity of the RhoA substrate kinases ROCK1/2. By contrast, KapB-mediated PB dispersion occurred in a ROCK1/2-independent manner. Taken together, these observations position KapB as a key contributor to viral reprogramming of ECs, capable of eliciting many of the phenotypes characteristic of KS tumor cells, and strongly contributing to the post-transcriptional control of EC gene expression and secretion.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Citoesqueleto/metabolismo , Herpesvirus Humano 8/fisiología , Estabilidad del ARN/fisiología , Elementos Ricos en Adenilato y Uridilato/genética , Células Cultivadas , Células HEK293 , Proteínas de Choque Térmico HSP27/metabolismo , Células HeLa , Proteínas de Choque Térmico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Chaperonas Moleculares , Proteínas Serina-Treonina Quinasas/metabolismo , Procesamiento Postranscripcional del ARN , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Sarcoma de Kaposi/virología , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(17): E1740-8, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733888

RESUMEN

AU-rich element mRNA-binding proteins (AUBPs) are key regulators of development, but how they are controlled and what functional roles they play depends on cellular context. Here, we show that Brf1 (zfp36l1), an AUBP from the Zfp36 protein family, operates downstream of FGF/Erk MAP kinase signaling to regulate pluripotency and cell fate decision making in mouse embryonic stem cells (mESCs). FGF/Erk MAP kinase signaling up-regulates Brf1, which disrupts the expression of core pluripotency-associated genes and attenuates mESC self-renewal without inducing differentiation. These regulatory effects are mediated by rapid and direct destabilization of Brf1 targets, such as Nanog mRNA. Enhancing Brf1 expression does not compromise mESC pluripotency but does preferentially regulate mesendoderm commitment during differentiation, accelerating the expression of primitive streak markers. Together, these studies demonstrate that FGF signals use targeted mRNA degradation by Brf1 to enable rapid posttranscriptional control of gene expression in mESCs.


Asunto(s)
Diferenciación Celular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Factor 1 de Respuesta al Butirato , Proliferación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Endodermo/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Semivida , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Sistema de Señalización de MAP Quinasas , Mesodermo/citología , Ratones , Proteína Homeótica Nanog , Células Madre Pluripotentes/metabolismo , Unión Proteica/genética , ARN Mensajero/metabolismo , Elementos de Respuesta/genética , Tristetraprolina/genética , Tristetraprolina/metabolismo
12.
PLoS Genet ; 9(9): e1003747, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086143

RESUMEN

Interleukin 17A (IL-17), a mediator implicated in chronic and severe inflammatory diseases, enhances the production of pro-inflammatory mediators by attenuating decay of the encoding mRNAs. The decay of many of these mRNAs depends on proteins (AUBps) that target AU-rich elements in the 3'-untranslated region of mRNAs and facilitate either mRNA decay or stabilization. Here we show that AUBps and the target mRNA assemble in a novel ribonucleoprotein complex in the presence of microRNA16 (miR16), which leads to the degradation of the target mRNA. Notably, IL-17 attenuates miR16 expression and promotes the binding of stabilizing AUBps over that of destabilizing AUBps, reducing mRNA decay. These findings indicate that miR16 independently of a seed sequence, directs the competition between degrading and stabilizing AUBps for target mRNAs. Since AUBps affect expression of about 8% of the human transcriptome and miR16 is ubiquitously expressed, IL-17 may in addition to inflammation affect many other cellular processes.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Inflamación/genética , Interleucina-17/metabolismo , MicroARNs/genética , Regiones no Traducidas 3'/genética , Hidrolasas de Éster Carboxílico/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Inflamación/patología , Interleucina-17/genética , Unión Proteica , Estabilidad del ARN/genética , ARN Mensajero/genética
13.
Proc Natl Acad Sci U S A ; 110(9): 3351-6, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23401530

RESUMEN

Although the p53 network has been intensively studied, genetic analyses long hinted at the existence of components that remained elusive. Recent studies have shown regulation of p53 at the mRNA level mediated via both the 5' and the 3' untranslated regions and affecting the stability and translation efficiency of the p53 mRNA. Here, we provide evidence of a feedback loop between p53 and the poly(A)-specific ribonuclease (PARN), in which PARN deadenylase keeps p53 levels low in nonstress conditions by destabilizing p53 mRNA, and the UV-induced increase in p53 activates PARN deadenylase, regulating gene expression during DNA damage response in a transactivation-independent manner. This model is innovative because it provides insights into p53 function and the mechanisms behind the regulation of mRNA 3' end processing in different cellular conditions.


Asunto(s)
Retroalimentación Fisiológica , Procesamiento de Término de ARN 3'/genética , Proteína p53 Supresora de Tumor/genética , Regiones no Traducidas 3'/genética , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Línea Celular Tumoral , Exorribonucleasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Modelos Genéticos , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/genética , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Proc Natl Acad Sci U S A ; 110(18): E1669-75, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589852

RESUMEN

Herpes simplex virus 1 (HSV-1) encodes an endoribonuclease that is responsible for the shutoff of host protein synthesis [virion host shutoff (VHS)-RNase]. The VHS-RNase released into cells during infection targets differentially four classes of mRNAs. Thus, (a) VHS-RNase degrades stable cellular mRNAs and α (immediate early) viral mRNAs; (b) it stabilizes host stress response mRNAs after deadenylation and subsequent cleavage near the adenylate-uridylate (AU)-rich elements; (c) it does not effectively degrade viral ß or γ mRNAs; and (d) it selectively spares from degradation a small number of cellular mRNAs. Current evidence suggests that several viral and at least one host protein (tristetraprolin) regulate its activity. Thus, virion protein (VP) 16 and VP22 neutralize the RNase activity at late times after infection. By binding to AU-rich elements via its interaction with tristetraprolin, the RNase deadenylates and cleaves the mRNAs in proximity to the AU-rich elements. In this report we show that another virion protein, UL47, brought into the cell during infection, attenuates the VHS-RNase activity with respect to stable host and viral α mRNAs and effectively blocks the degradation of ß and γ mRNAs, but it has no effect on the processing of AU-rich mRNAs. The properties of UL47 suggest that it, along with the α protein infected cell protein 27, attenuates degradation of mRNAs by the VHS-RNase through interaction with the enzyme in polyribosomes. Mutants lacking both VHS-RNase and UL47 overexpress α genes and delay the expression of ß and γ genes, suggesting that overexpression of α genes inhibits the downstream expression of early and late genes.


Asunto(s)
Herpesvirus Humano 1/metabolismo , Estabilidad del ARN , Ribonucleasas/metabolismo , Proteínas Virales de Fusión/metabolismo , Proteínas Virales/metabolismo , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Extractos Celulares , ADN Viral/metabolismo , Eliminación de Gen , Células HEK293 , Herpes Simple/metabolismo , Herpes Simple/virología , Humanos , Proteínas Inmediatas-Precoces , Modelos Biológicos , Mutación/genética , ARN Viral/metabolismo , Reproducibilidad de los Resultados , Transfección
15.
J Biol Chem ; 289(51): 35042-60, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25342740

RESUMEN

Members of the mammalian tristetraprolin family of CCCH tandem zinc finger proteins can bind to certain AU-rich elements (AREs) in mRNAs, leading to their deadenylation and destabilization. Mammals express three or four members of this family, but Drosophila melanogaster and other insects appear to contain a single gene, Tis11. We found that recombinant Drosophila Tis11 protein could bind to ARE-containing RNA oligonucleotides with low nanomolar affinity. Remarkably, co-expression in mammalian cells with "target" RNAs demonstrated that Tis11 could promote destabilization of ARE-containing mRNAs and that this was partially dependent on a conserved C-terminal sequence resembling the mammalian NOT1 binding domain. Drosophila Tis11 promoted both deadenylation and decay of a target transcript in this heterologous cell system. We used chromosome deletion/duplication and P element insertion to produce two types of Tis11 deficiency in adult flies, both of which were viable and fertile. To address the hypothesis that Tis11 deficiency would lead to the abnormal accumulation of potential target transcripts, we analyzed gene expression in adult flies by deep mRNA sequencing. We identified 69 transcripts from 56 genes that were significantly up-regulated more than 1.5-fold in both types of Tis11-deficient flies. Ten of the up-regulated transcripts encoded probable proteases, but many other functional classes of proteins were represented. Many of the up-regulated transcripts contained potential binding sites for tristetraprolin family member proteins that were conserved in other Drosophila species. Tis11 is thus an ARE-binding, mRNA-destabilizing protein that may play a role in post-transcriptional gene expression in Drosophila and other insects.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación de la Expresión Génica , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Elementos Ricos en Adenilato y Uridilato/genética , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Sondas de Oligonucleótidos/química , Sondas de Oligonucleótidos/genética , Sondas de Oligonucleótidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN/métodos , Homología de Secuencia de Aminoácido
16.
RNA Biol ; 12(12): 1289-300, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26512911

RESUMEN

We describe here a platform for high-throughput protein expression and interaction analysis aimed at identifying the RNA-interacting domainome. This approach combines the selection of a phage library displaying "filtered" open reading frames with next-generation DNA sequencing. The method was validated using an RNA bait corresponding to the AU-rich element of α-prothymosin, an RNA motif that promotes mRNA stability and translation through its interaction with the RNA-binding protein ELAVL1. With this strategy, we not only confirmed known RNA-binding proteins that specifically interact with the target RNA (such as ELAVL1/HuR and RBM38) but also identified proteins not previously known to be ARE-binding (R3HDM2 and RALY). We propose this technology as a novel approach for studying the RNA-binding proteome.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Sistemas de Lectura Abierta/genética , Dominios y Motivos de Interacción de Proteínas/genética , Precursores de Proteínas/genética , Proteínas de Unión al ARN/metabolismo , Timosina/análogos & derivados , Células HEK293 , Humanos , Unión Proteica , Precursores de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Timosina/genética , Timosina/metabolismo
17.
PLoS Genet ; 8(9): e1002977, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028373

RESUMEN

TNF expression of macrophages is under stringent translational control that depends on the p38 MAPK/MK2 pathway and the AU-rich element (ARE) in the TNF mRNA. Here, we elucidate the molecular mechanism of phosphorylation-regulated translation of TNF. We demonstrate that translation of the TNF-precursor at the ER requires expression of the ARE-binding and -stabilizing factor human antigen R (HuR) together with either activity of the p38 MAPK/MK2 pathway or the absence of the ARE-binding and -destabilizing factor tristetraprolin (TTP). We show that phosphorylation of TTP by MK2 decreases its affinity to the ARE, inhibits its ability to replace HuR, and permits HuR-mediated initiation of translation of TNF mRNA. Since translation of TTP's own mRNA is also regulated by this mechanism, an intrinsic feedback control of the inflammatory response is ensured. The phosphorylation-regulated TTP/HuR exchange at target mRNAs provides a reversible switch between unstable/non-translatable and stable/efficiently translated mRNAs.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Tristetraprolina , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas ELAV/metabolismo , Humanos , Macrófagos/metabolismo , Fosforilación , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Biol Chem ; 288(33): 23680-6, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23836903

RESUMEN

Macrophage inhibitory cytokine-1 (MIC-1), a secreted cytokine, is a direct target of p53 and known to play a role in cell proliferation, apoptosis, cell metastasis, and angiogenesis through autocrine and paracrine signaling. Previous studies have shown that serum levels of MIC-1 closely parallel cancer progression and are being explored as a diagnostic tool. MIC-1 has also shown potential as a therapeutic agent as it has exhibited several anti-carcinogenic activities. Thus, MIC-1 displays two opposing effects: tumor suppression versus promotion. However, it remains unclear whether MIC-1 is regulated by a mechanism other than transcription and how MIC-1 exerts its tumor suppression. In this study, we show that overexpression of RNA-binding protein RNPC1 can increase, whereas knockdown or knock-out of RNPC1 decreases, MIC-1 transcript and protein levels. Additionally, we demonstrate that RNPC1 can bind to MIC-1 mRNA via an AU-rich element within MIC-1 3'-UTR and then enhances MIC-1 mRNA stability. Finally, to explore the functional significance of MIC-1, we showed that knockdown of MIC-1 can decrease RNPC1-induced cell growth suppression. Altogether, we uncover a novel mechanism by which MIC-1 can be regulated through RNPC1 via mRNA stability.


Asunto(s)
Regulación de la Expresión Génica , Factor 15 de Diferenciación de Crecimiento/genética , Estabilidad del ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regiones no Traducidas 3'/genética , Elementos Ricos en Adenilato y Uridilato/genética , Animales , Secuencia de Bases , Línea Celular , Proliferación Celular , Técnicas de Silenciamiento del Gen , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
19.
Biochim Biophys Acta ; 1829(6-7): 680-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23246978

RESUMEN

AUF1 is a family of four proteins generated by alternative pre-mRNA splicing that form high affinity complexes with AU-rich, mRNA-destabilizing sequences located within the 3' untranslated regions of many labile mRNAs. While AUF1 binding is most frequently associated with accelerated mRNA decay, emerging examples have demonstrated roles as a mRNA stabilizer or even translational regulator for specific transcripts. In this review, we summarize recent advances in our understanding of mRNA recognition by AUF1 and the biochemical and functional consequences of these interactions. In addition, unique properties of individual AUF1 isoforms and the roles of these proteins in modulating expression of genes associated with inflammatory, neoplastic, and cardiac diseases are discussed. Finally, we describe mechanisms that regulate AUF1 expression in cells, and current knowledge of regulatory switches that modulate the cellular levels and/or activities of AUF1 isoforms through distinct protein post-translational modifications. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo D/genética , Estabilidad del ARN/genética , ARN Mensajero/genética , Empalme Alternativo/genética , Regulación de la Expresión Génica , Ribonucleoproteína Nuclear Heterogénea D0 , Humanos , Procesamiento Proteico-Postraduccional , Precursores del ARN/genética
20.
Biochim Biophys Acta ; 1829(6-7): 666-79, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23428348

RESUMEN

Changes in mRNA stability and translation are critical control points in the regulation of gene expression, particularly genes encoding growth factors, inflammatory mediators, and proto-oncogenes. Adenosine and uridine (AU)-rich elements (ARE), often located in the 3' untranslated regions (3'UTR) of mRNAs, are known to target transcripts for rapid decay. They are also involved in the regulation of mRNA stability and translation in response to extracellular cues. This review focuses on one of the best characterized ARE binding proteins, tristetraprolin (TTP), the founding member of a small family of CCCH tandem zinc finger proteins. In this survey, we have reviewed the current status of TTP interactions with mRNA and proteins, and discussed current thinking about TTP's mechanism of action to promote mRNA decay. We also review the proposed regulation of TTP's functions by phosphorylation. Finally, we have discussed emerging evidence for TTP operating as a translational regulator. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato/genética , Biosíntesis de Proteínas , Estabilidad del ARN/genética , ARN Mensajero/genética , Tristetraprolina/genética , Regiones no Traducidas 3' , Secuencia de Bases , Línea Celular , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Estabilidad del ARN/efectos de los fármacos , Tristetraprolina/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Dedos de Zinc/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA