Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 160(6): 1087-98, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25768905

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is a paradigmatic neurodegenerative proteinopathy, in which a mutant protein (in this case, ATAXIN1) accumulates in neurons and exerts toxicity; in SCA1, this process causes progressive deterioration of motor coordination. Seeking to understand how post-translational modification of ATAXIN1 levels influences disease, we discovered that the RNA-binding protein PUMILIO1 (PUM1) not only directly regulates ATAXIN1 but also plays an unexpectedly important role in neuronal function. Loss of Pum1 caused progressive motor dysfunction and SCA1-like neurodegeneration with motor impairment, primarily by increasing Ataxin1 levels. Breeding Pum1(+/-) mice to SCA1 mice (Atxn1(154Q/+)) exacerbated disease progression, whereas breeding them to Atxn1(+/-) mice normalized Ataxin1 levels and largely rescued the Pum1(+/-) phenotype. Thus, both increased wild-type ATAXIN1 levels and PUM1 haploinsufficiency could contribute to human neurodegeneration. These results demonstrate the importance of studying post-transcriptional regulation of disease-driving proteins to reveal factors underlying neurodegenerative disease.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3' , Animales , Antígenos Ly/genética , Ataxina-1 , Ataxinas , Encéfalo/metabolismo , Técnicas de Sustitución del Gen , Haploinsuficiencia , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Mutación , Enfermedades Neurodegenerativas/patología , Conformación de Ácido Nucleico , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN Mensajero/química
2.
Nat Rev Mol Cell Biol ; 18(2): 102-114, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27847391

RESUMEN

A goal of human genetics studies is to determine the mechanisms by which genetic variation produces phenotypic differences that affect human health. Efforts in this respect have previously focused on genetic variants that affect mRNA levels by altering epigenetic and transcriptional regulation. Recent studies show that genetic variants that affect RNA processing are at least equally as common as, and are largely independent from, those variants that affect transcription. We highlight the impact of genetic variation on pre-mRNA splicing and polyadenylation, and on the stability, translation and structure of mRNAs as mechanisms that produce phenotypic traits. These results emphasize the importance of including RNA processing signals in analyses to identify functional variants.


Asunto(s)
MicroARNs , Procesamiento Postranscripcional del ARN , Empalme del ARN , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Semivida , Humanos , Poliadenilación , Precursores del ARN/genética , Estabilidad del ARN
3.
Mol Cell ; 68(3): 473-475, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100050

RESUMEN

Microsatellite expansion diseases are caused by unstable tandem repeats of 3-10 nucleotides that become pathogenic beyond a threshold number of copies. Two groups present different approaches to reduce pathogenesis by targeting deactivated Cas9 to either the DNA (Pinto et al., 2017) or the RNA (Batra et al., 2017) repeats with therapeutic potential for several diseases.


Asunto(s)
Sistemas CRISPR-Cas , ARN , Animales , Caballos , Repeticiones de Microsatélite
4.
Nucleic Acids Res ; 51(3): 1245-1259, 2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36617982

RESUMEN

Loss of gene function can be compensated by paralogs with redundant functions. An example of such compensation are the paralogs of the Muscleblind-Like (MBNL) family of RNA-binding proteins that are sequestered and lose their function in Myotonic Dystrophy Type 1 (DM1). Loss of MBNL1 increases the levels of its paralog MBNL2 in tissues where Mbnl2 expression is low, allowing MBNL2 to functionally compensate for MBNL1 loss. Here, we show that loss of MBNL1 increases the inclusion of Mbnl2 exon 6 and exon 9. We find that inclusion of Mbnl2 exon 6 increases the translocation of MBNL2 to the nucleus, while the inclusion of Mbnl2 exon 9 shifts the reading frame to an alternative C-terminus. We show that the C-terminus lacking exon 9 contains a PEST domain which causes proteasomal degradation. Loss of MBNL1 increases the inclusion of exon 9, resulting in an alternative C-terminus lacking the PEST domain and the increase of MBNL2. We further find that the compensatory mechanism is active in a mouse DM1 model. Together, this study uncovers the compensatory mechanism by which loss of MBNL1 upregulates its paralog MBNL2 and highlights a potential role of the compensatory mechanism in DM1.


Asunto(s)
Empalme Alternativo , Distrofia Miotónica , Proteínas de Unión al ARN , Animales , Ratones , Proteínas de Unión al ADN/genética , Exones , Distrofia Miotónica/genética , Proteínas de Unión al ARN/metabolismo , Activación Transcripcional , Regulación hacia Arriba
5.
Cell ; 136(4): 777-93, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19239895

RESUMEN

Cellular functions depend on numerous protein-coding and noncoding RNAs and the RNA-binding proteins associated with them, which form ribonucleoprotein complexes (RNPs). Mutations that disrupt either the RNA or protein components of RNPs or the factors required for their assembly can be deleterious. Alternative splicing provides cells with an exquisite capacity to fine-tune their transcriptome and proteome in response to cues. Splicing depends on a complex code, numerous RNA-binding proteins, and an enormously intricate network of interactions among them, increasing the opportunity for exposure to mutations and misregulation that cause disease. The discovery of disease-causing mutations in RNAs is yielding a wealth of new therapeutic targets, and the growing understanding of RNA biology and chemistry is providing new RNA-based tools for developing therapeutics.


Asunto(s)
Enfermedad/genética , Empalme Alternativo , Mutación , ARN/uso terapéutico , Empalme del ARN , Terapéutica
6.
Mol Cell ; 62(2): 155-156, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27105111

RESUMEN

Repeat expansions cause dominantly inherited neurological disorders. In this issue of Molecular Cell, Kearse et al. (2016) examine the requirements for RAN translation of the CGG repeats that cause fragile X-associated tremor/ataxia syndrome, revealing similarities and differences with canonical translation.


Asunto(s)
Ataxia , Síndrome del Cromosoma X Frágil , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Humanos , Temblor
7.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769228

RESUMEN

The contractile cells of skeletal muscles, called myofibers, are elongated multinucleated syncytia formed and maintained by the fusion of proliferative myoblasts. Human myofibers can be hundreds of microns in diameter and millimeters in length. Myofibers are non-mitotic, obviating the need for microtubules in cell division. However, microtubules have been adapted to the unique needs of these cells and are critical for myofiber development and function. Microtubules in mature myofibers are highly dynamic, and studies in several experimental systems have demonstrated the requirements for microtubules in the unique features of muscle biology including myoblast fusion, peripheral localization of nuclei, assembly of the sarcomere, transport and signaling. Microtubule-binding proteins have also been adapted to the needs of the skeletal muscle including the expression of skeletal muscle-specific protein isoforms generated by alternative splicing. Here, we will outline the different roles microtubules play in skeletal muscle cells, describe how microtubule abnormalities can lead to muscle disease and discuss the broader implications for microtubule function.


Asunto(s)
Fibras Musculares Esqueléticas , Músculo Esquelético , Humanos , Músculo Esquelético/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Microtúbulos , Diferenciación Celular , Desarrollo de Músculos , Homeostasis
8.
Hum Mol Genet ; 29(10): 1729-1744, 2020 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-32412585

RESUMEN

mRNA processing is highly regulated during development through changes in RNA-binding protein (RBP) activities. CUG-BP, Elav-like family member 1 (CELF1, also called CUGBP1) is an RBP, the expression of which decreases in skeletal muscle soon after birth. CELF1 regulates multiple nuclear and cytoplasmic RNA processing events. In the nucleus, CELF1 regulates networks of postnatal alternative splicing (AS) transitions, while in the cytoplasm, CELF1 regulates mRNA stability and translation. Stabilization and misregulation of CELF1 has been implicated in human diseases including myotonic dystrophy type 1, Alzheimer's disease and multiple cancers. To understand the contribution of nuclear and cytoplasmic CELF1 activity to normal and pathogenic skeletal muscle biology, we generated transgenic mice for doxycycline-inducible and skeletal muscle-specific expression of active CELF1 mutants engineered to be localized predominantly to either the nucleus or the cytoplasm. Adult mice expressing nuclear, but not cytoplasmic, CELF1 are characterized by strong histopathological defects, muscle loss within 10 days and changes in AS. In contrast, mice expressing cytoplasmic CELF1 display changes in protein levels of targets known to be regulated at the level of translation by CELF1, with minimal changes in AS. These changes are in the absence of overt histopathological changes or muscle loss. RNA-sequencing revealed extensive gene expression and AS changes in mice overexpressing nuclear and naturally localized CELF1 protein, with affected genes involved in cytoskeleton dynamics, membrane dynamics, RNA processing and zinc ion binding. These results support a stronger role for nuclear CELF1 functions as compared to cytoplasmic CELF1 functions in skeletal muscle wasting.


Asunto(s)
Proteínas CELF1/genética , Atrofia Muscular/genética , Distrofia Miotónica/genética , Estabilidad del ARN/genética , Empalme Alternativo/genética , Animales , Nucléolo Celular/genética , Citoplasma/genética , Humanos , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/patología , Distrofia Miotónica/patología , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/genética
9.
Opt Express ; 30(25): 44556-44568, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36522878

RESUMEN

We present near-ideal axisymmetric numerically optimized spline concentrators (OSCs) which outperform the compound parabolic concentrator (CPC). By perturbing the profile of the revolved CPC by a variable-offset spline defined in tangent-normal space, we show that ray rejection can be reduced to nearly half of that of the CPC, without increasing concentrator length. The resulting OSCs achieve acceptance efficiencies as high as 99.3% for an acceptance angle of 45°, the highest reported for any finite-length CPC-like light concentrator. A set of design curves is presented which can be used to generate near "best-form" OSCs for any acceptance angle in the range 10° to 45°.

10.
Mol Cell ; 55(4): 592-603, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25087874

RESUMEN

Alternative splicing plays important regulatory roles during periods of physiological change. During development, a large number of genes coordinately express protein isoform transitions regulated by alternative splicing; however, the mechanisms that coordinate splicing and the functional integration of the resultant tissue-specific protein isoforms are typically unknown. Here we show that the conserved Rbfox2 RNA binding protein regulates 30% of the splicing transitions observed during myogenesis and is required for the specific step of myoblast fusion. Integration of Rbfox2-dependent splicing outcomes from RNA-seq with Rbfox2 iCLIP data identified Mef2d and Rock2 as Rbfox2 splicing targets. Restored activities of Mef2d and Rock2 rescued myoblast fusion in Rbfox2-depleted cultures, demonstrating functional cooperation of protein isoforms generated by coordinated alterative splicing. The results demonstrate that coordinated alternative splicing by a single RNA binding protein modulates transcription (Mef2d) and cell signaling (Rock2) programs to drive tissue-specific functions (cell fusion) to promote a developmental transition.


Asunto(s)
Empalme Alternativo/genética , Factores de Transcripción MEF2/genética , Desarrollo de Músculos/genética , Mioblastos/fisiología , Proteínas de Unión al ARN/fisiología , ARN/genética , Quinasas Asociadas a rho/genética , Empalme Alternativo/fisiología , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Secuencia Conservada , Regulación de la Expresión Génica , Células HEK293 , Humanos , Factores de Transcripción MEF2/metabolismo , Ratones , Desarrollo de Músculos/fisiología , Especificidad de Órganos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análisis de Secuencia de ARN , Quinasas Asociadas a rho/metabolismo
11.
Int J Mol Sci ; 23(23)2022 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-36499107

RESUMEN

Myotonic dystrophy (DM) is a highly variable, multisystemic disorder that clinically affects one in 8000 individuals. While research has predominantly focused on the symptoms and pathological mechanisms affecting striated muscle and brain, DM patient surveys have identified a high prevalence for gastrointestinal (GI) symptoms amongst affected individuals. Clinical studies have identified chronic and progressive dysfunction of the esophagus, stomach, liver and gallbladder, small and large intestine, and rectum and anal sphincters. Despite the high incidence of GI dysmotility in DM, little is known regarding the pathological mechanisms leading to GI dysfunction. In this review, we summarize results from clinical and molecular analyses of GI dysfunction in both genetic forms of DM, DM type 1 (DM1) and DM type 2 (DM2). Based on current knowledge of DM primary pathological mechanisms in other affected tissues and GI tissue studies, we suggest that misregulation of alternative splicing in smooth muscle resulting from the dysregulation of RNA binding proteins muscleblind-like and CUGBP-elav-like is likely to contribute to GI dysfunction in DM. We propose that a combinatorial approach using clinical and molecular analysis of DM GI tissues and model organisms that recapitulate DM GI manifestations will provide important insight into defects impacting DM GI motility.


Asunto(s)
Distrofia Miotónica , Humanos , Distrofia Miotónica/complicaciones , Distrofia Miotónica/genética , Empalme Alternativo , Músculo Esquelético/metabolismo , Proteínas de Unión al ARN/metabolismo
13.
Nature ; 525(7569): 384-8, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26331541

RESUMEN

MYC (also known as c-MYC) overexpression or hyperactivation is one of the most common drivers of human cancer. Despite intensive study, the MYC oncogene remains recalcitrant to therapeutic inhibition. MYC is a transcription factor, and many of its pro-tumorigenic functions have been attributed to its ability to regulate gene expression programs. Notably, oncogenic MYC activation has also been shown to increase total RNA and protein production in many tissue and disease contexts. While such increases in RNA and protein production may endow cancer cells with pro-tumour hallmarks, this increase in synthesis may also generate new or heightened burden on MYC-driven cancer cells to process these macromolecules properly. Here we discover that the spliceosome is a new target of oncogenic stress in MYC-driven cancers. We identify BUD31 as a MYC-synthetic lethal gene in human mammary epithelial cells, and demonstrate that BUD31 is a component of the core spliceosome required for its assembly and catalytic activity. Core spliceosomal factors (such as SF3B1 and U2AF1) associated with BUD31 are also required to tolerate oncogenic MYC. Notably, MYC hyperactivation induces an increase in total precursor messenger RNA synthesis, suggesting an increased burden on the core spliceosome to process pre-mRNA. In contrast to normal cells, partial inhibition of the spliceosome in MYC-hyperactivated cells leads to global intron retention, widespread defects in pre-mRNA maturation, and deregulation of many essential cell processes. Notably, genetic or pharmacological inhibition of the spliceosome in vivo impairs survival, tumorigenicity and metastatic proclivity of MYC-dependent breast cancers. Collectively, these data suggest that oncogenic MYC confers a collateral stress on splicing, and that components of the spliceosome may be therapeutic entry points for aggressive MYC-driven cancers.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Genes myc/genética , Empalmosomas/efectos de los fármacos , Empalmosomas/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Intrones/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Precursores del ARN/biosíntesis , Precursores del ARN/genética , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Ribonucleoproteínas/metabolismo , Factor de Empalme U2AF , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nucleic Acids Res ; 47(1): 421-431, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30418605

RESUMEN

RNA splicing is a highly regulated process dependent on sequences near splice sites. Insertions of Alu retrotransposons can disrupt splice sites or bind splicing regulators. We hypothesized that some common inherited polymorphic Alu insertions are responsible for splicing QTLs (sQTL). We focused on intronic Alu variants mapping within 100 bp of an alternatively used exon and screened for those that alter splicing. We identify five loci, 21.7% of those assayed, where the polymorphic Alu alters splicing. While in most cases the Alu promotes exon skipping, at one locus the Alu increases exon inclusion. Of particular interest is an Alu polymorphism in the CD58 gene. Reduced CD58 expression is associated with risk for developing multiple sclerosis. We show that the Alu insertion promotes skipping of CD58 exon 3 and results in a frameshifted transcript, indicating that the Alu may be the causative variant for increased MS risk at this locus. Using RT-PCR analysis at the endogenous locus, we confirm that the Alu variant is a sQTL for CD58. In summary, altered splicing efficiency is a common functional consequence of Alu polymorphisms including at least one instance where the variant is implicated in disease risk. This work broadens our understanding of splicing regulatory sequences around exons.


Asunto(s)
Elementos Alu/genética , Antígenos CD58/genética , Sitios de Carácter Cuantitativo/genética , Empalme del ARN/genética , Empalme Alternativo/genética , Exones/genética , Variación Genética , Humanos , Intrones/genética , Sitios de Empalme de ARN/genética , ARN Mensajero/genética
15.
Hum Mol Genet ; 27(16): 2789-2804, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29771332

RESUMEN

Myotonic dystrophy type 1 (DM1) is a multi-systemic disease resulting in severe muscle weakening and wasting. DM1 is caused by expansion of CTG repeats in the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. We have developed an inducible, skeletal muscle-specific mouse model of DM1 (CUG960) that expresses 960 CUG repeat-expressing animals (CUG960) in the context of human DMPK exons 11-15. CUG960 RNA-expressing mice induced at postnatal day 1, as well as adult-onset animals, show clear, measurable muscle wasting accompanied by severe histological defects including central myonuclei, reduced fiber cross-sectional area, increased percentage of oxidative myofibers, the presence of nuclear RNA foci that colocalize with Mbnl1 protein, and increased Celf1 protein in severely affected muscles. Importantly, muscle loss, histological abnormalities and RNA foci are reversible, demonstrating recovery upon removal of toxic RNA. RNA-seq and protein array analysis indicate that the balance between anabolic and catabolic pathways that normally regulate muscle mass may be disrupted by deregulation of platelet derived growth factor receptor ß signaling and the PI3K/AKT pathways, along with prolonged activation of AMP-activated protein kinase α signaling. Similar changes were detected in DM1 skeletal muscle compared with unaffected controls. The mouse model presented in this paper shows progressive skeletal muscle wasting and has been used to identify potential molecular mechanisms underlying skeletal muscle loss. The reversibility of the phenotype establishes a baseline response for testing therapeutic approaches.


Asunto(s)
Debilidad Muscular/genética , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Animales , Secuencia de Bases , Proteínas CELF1 , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Humanos , Ratones , Debilidad Muscular/patología , Músculo Esquelético/fisiopatología , Distrofia Miotónica/patología , Proteínas de Unión al ARN , Expansión de Repetición de Trinucleótido
16.
Muscle Nerve ; 60(6): 779-789, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31509256

RESUMEN

INTRODUCTION: Myotonic dystrophy type 1 (DM1) is a multisystemic disease caused by expansion of a CTG repeat in the 3' UTR of the Dystrophia Myotonica-Protein Kinase (DMPK) gene. While multiple organs are affected, more than half of mortality is due to muscle wasting. METHODS: It is unclear whether endurance exercise provides beneficial effects in DM1. Here, we show that a 10-week treadmill endurance exercise program leads to beneficial effects in the HSALR mouse model of DM1. RESULTS: Animals that performed treadmill training displayed reduced CUGexp RNA levels, improved splicing abnormalities, an increase in skeletal muscle weight and improved endurance capacity. DISCUSSION: These results indicate that endurance exercise does not have adverse effects in HSALR animals and contributes to beneficial molecular and physiological outcomes.


Asunto(s)
Entrenamiento Aeróbico/métodos , Músculo Esquelético/metabolismo , Distrofia Miotónica/metabolismo , Condicionamiento Físico Animal/métodos , Resistencia Física/fisiología , Actinas/genética , Tejido Adiposo , Empalme Alternativo , Animales , Composición Corporal , Densidad Ósea , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Miotónica/patología , Distrofia Miotónica/fisiopatología , Tamaño de los Órganos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Expansión de Repetición de Trinucleótido
17.
Genome Res ; 25(6): 858-71, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25883322

RESUMEN

RNA binding proteins of the conserved CUGBP1, Elav-like factor (CELF) family contribute to heart and skeletal muscle development and are implicated in myotonic dystrophy (DM). To understand their genome-wide functions, we analyzed the transcriptome dynamics following induction of CELF1 or CELF2 in adult mouse heart and of CELF1 in muscle by RNA-seq, complemented by crosslinking/immunoprecipitation-sequencing (CLIP-seq) analysis of mouse cells and tissues to distinguish direct from indirect regulatory targets. We identified hundreds of mRNAs bound in their 3' UTRs by both CELF1 and the developmentally induced MBNL1 protein, a threefold greater overlap in target messages than expected, including messages involved in development and cell differentiation. The extent of 3' UTR binding by CELF1 and MBNL1 predicted the degree of mRNA repression or stabilization, respectively, following CELF1 induction. However, CELF1's RNA binding specificity in vitro was not detectably altered by coincubation with recombinant MBNL1. These findings support a model in which CELF and MBNL proteins bind independently to mRNAs but functionally compete to specify down-regulation or localization/stabilization, respectively, of hundreds of mRNA targets. Expression of many alternative 3' UTR isoforms was altered following CELF1 induction, with 3' UTR binding associated with down-regulation of isoforms and genes. The splicing of hundreds of alternative exons was oppositely regulated by these proteins, confirming an additional layer of regulatory antagonism previously observed in a handful of cases. The regulatory relationships between CELFs and MBNLs in control of both mRNA abundance and splicing appear to have evolved to enhance developmental transitions in major classes of heart and muscle genes.


Asunto(s)
Proteínas CELF/genética , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Empalme del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Animales , Proteínas CELF/metabolismo , Proteínas CELF1/genética , Proteínas CELF1/metabolismo , Regulación hacia Abajo , Exones , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , Proteínas del Tejido Nervioso/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ARN , Transcriptoma
18.
PLoS Biol ; 13(7): e1002197, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26176594

RESUMEN

Two insults often underlie a variety of eye diseases including glaucoma, optic atrophy, and retinal degeneration--defects in mitochondrial function and aberrant Rhodopsin trafficking. Although mitochondrial defects are often associated with oxidative stress, they have not been linked to Rhodopsin trafficking. In an unbiased forward genetic screen designed to isolate mutations that cause photoreceptor degeneration, we identified mutations in a nuclear-encoded mitochondrial gene, ppr, a homolog of human LRPPRC. We found that ppr is required for protection against light-induced degeneration. Its function is essential to maintain membrane depolarization of the photoreceptors upon repetitive light exposure, and an impaired phototransduction cascade in ppr mutants results in excessive Rhodopsin1 endocytosis. Moreover, loss of ppr results in a reduction in mitochondrial RNAs, reduced electron transport chain activity, and reduced ATP levels. Oxidative stress, however, is not induced. We propose that the reduced ATP level in ppr mutants underlies the phototransduction defect, leading to increased Rhodopsin1 endocytosis during light exposure, causing photoreceptor degeneration independent of oxidative stress. This hypothesis is bolstered by characterization of two other genes isolated in the screen, pyruvate dehydrogenase and citrate synthase. Their loss also causes a light-induced degeneration, excessive Rhodopsin1 endocytosis and reduced ATP without concurrent oxidative stress, unlike many other mutations in mitochondrial genes that are associated with elevated oxidative stress and light-independent photoreceptor demise.


Asunto(s)
Proteínas de Drosophila/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Células Fotorreceptoras de Invertebrados/efectos de la radiación , Enfermedades de la Retina/genética , Adenosina Trifosfato/biosíntesis , Animales , Citrato (si)-Sintasa/genética , Drosophila , Proteínas de Drosophila/metabolismo , Electrorretinografía , Endocitosis , Mitocondrias/genética , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo , Complejo Piruvato Deshidrogenasa/genética , Complejo Piruvato Deshidrogenasa/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Rodopsina/metabolismo , Visión Ocular
19.
Genes Dev ; 24(7): 653-8, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20299448

RESUMEN

Alternative splicing transitions have been identified recently as a conserved component of vertebrate heart remodeling during postnatal development. Here we report that the targeted deletion of Dicer, specifically in adult mouse myocardium, reveals the role of microRNAs (miRNAs) in regulating networks of postnatal splicing transitions and in maintaining adult splicing programs. We demonstrate a direct role for miR-23a/b in the dramatic postnatal down-regulation of CUGBP and ETR-3-like factor (CELF) proteins that regulate nearly half of developmentally regulated splicing transitions in the heart. These findings define a hierarchy in which rapid postnatal up-regulation of specific miRNAs controls expression of alternative splicing regulators and the subsequent splicing transitions of their downstream targets.


Asunto(s)
Empalme Alternativo/fisiología , Corazón/crecimiento & desarrollo , MicroARNs/metabolismo , Animales , Proteínas CELF1 , Línea Celular , ARN Helicasas DEAD-box/genética , Regulación hacia Abajo , Endorribonucleasas/genética , Técnicas de Silenciamiento del Gen , Ratones , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III , Regulación hacia Arriba
20.
Hum Mol Genet ; 24(8): 2360-74, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25575511

RESUMEN

The Rbfox family of RNA-binding proteins is highly conserved with established roles in alternative splicing (AS) regulation. High-throughput studies aimed at understanding transcriptome remodeling have revealed skeletal muscle as displaying one of the largest number of AS events. This finding is consistent with requirements for tissue-specific protein isoforms needed to sustain muscle-specific functions. Rbfox1 is abundant in vertebrate brain, heart and skeletal muscle. Genome-wide genetic approaches have linked the Rbfox1 gene to autism, and a brain-specific knockout mouse revealed a critical role for this splicing regulator in neuronal function. Moreover, a Caenorhabditis elegans Rbfox1 homolog regulates muscle-specific splicing. To determine the role of Rbfox1 in muscle function, we developed a conditional knockout mouse model to specifically delete Rbfox1 in adult tissue. We show that Rbfox1 is required for muscle function but a >70% loss of Rbfox1 in satellite cells does not disrupt muscle regeneration. Deep sequencing identified aberrant splicing of multiple genes including those encoding myofibrillar and cytoskeletal proteins, and proteins that regulate calcium handling. Ultrastructure analysis of Rbfox1(-/-) muscle by electron microscopy revealed abundant tubular aggregates. Immunostaining showed mislocalization of the sarcoplasmic reticulum proteins Serca1 and Ryr1 in a pattern indicative of colocalization with the tubular aggregates. Consistent with mislocalization of Serca1 and Ryr1, calcium handling was drastically altered in Rbfox1(-/-) muscle. Moreover, muscle function was significantly impaired in Rbfox1(-/-) muscle as indicated by decreased force generation. These results demonstrate that Rbfox1 regulates a network of AS events required to maintain multiple aspects of muscle physiology.


Asunto(s)
Empalme Alternativo , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Calcio/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Enfermedades Musculares/genética , Mioblastos/metabolismo , Factores de Empalme de ARN , Proteínas de Unión al ARN/genética , Células Satélite del Músculo Esquelético/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA