Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 47(6): 2884-2905, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30698797

RESUMEN

Circular RNAs (circRNAs) perform diverse functions, including the regulation of transcription, translation, peptide synthesis, macromolecular sequestration and trafficking. Inverted Alu repeats capable of forming RNA:RNA duplexes that bring splice sites together for backsplicing are known to facilitate circRNA generation. However, higher limits of circRNAs produced by a single Alu-rich gene are currently not predictable due to limitations of amplification and analyses. Here, using a tailored approach, we report a surprising diversity of exon-containing circRNAs generated by the Alu-rich Survival Motor Neuron (SMN) genes that code for SMN, an essential multifunctional protein in humans. We show that expression of the vast repertoire of SMN circRNAs is universal. Several of the identified circRNAs harbor novel exons derived from both intronic and intergenic sequences. A comparison with mouse Smn circRNAs underscored a clear impact of primate-specific Alu elements on shaping the overall repertoire of human SMN circRNAs. We show the role of DHX9, an RNA helicase, in splicing regulation of several SMN exons that are preferentially incorporated into circRNAs. Our results suggest self- and cross-regulation of biogenesis of various SMN circRNAs. These findings bring a novel perspective towards a better understanding of SMN gene function.


Asunto(s)
Empalme Alternativo/fisiología , ARN/genética , Proteínas del Complejo SMN/genética , Región de Flanqueo 5' , Elementos Alu/genética , Células Cultivadas , Biología Computacional , Exones , Células HEK293 , Células HeLa , Humanos , ARN Circular , ARN Mensajero , Proteínas del Complejo SMN/fisiología
2.
J Cell Sci ; 129(4): 804-16, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26743087

RESUMEN

Disconnection between membrane signalling and actin networks can have catastrophic effects depending on cell size and polarity. The survival motor neuron (SMN) protein is ubiquitously involved in assembly of spliceosomal small nuclear ribonucleoprotein particles. Other SMN functions could, however, affect cellular activities driving asymmetrical cell surface expansions. Genes able to mitigate SMN deficiency operate within pathways in which SMN can act, such as mRNA translation, actin network and endocytosis. Here, we found that SMN accumulates at membrane protrusions during the dynamic rearrangement of the actin filaments. In addition to localization data, we show that SMN interacts with caveolin-1, which mediates anchoring of translation machinery components. Importantly, SMN deficiency depletes the plasma membrane of ribosomes, and this correlates with the failure of fibroblasts to extend membrane protrusions. These findings strongly support a relationship between SMN and membrane dynamics. We propose that SMN could assembly translational platforms associated with and governed by the plasma membrane. This activity could be crucial in cells that have an exacerbated interdependence of membrane remodelling and local protein synthesis.


Asunto(s)
Membrana Celular/metabolismo , Proteínas del Complejo SMN/fisiología , Citoesqueleto de Actina/metabolismo , Caveolina 1/metabolismo , Membrana Celular/ultraestructura , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Células Cultivadas , Humanos , Biosíntesis de Proteínas , Transporte de Proteínas , Ribosomas/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(30): 9382-7, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26170331

RESUMEN

The mechanisms by which poikilothermic organisms ensure that biological processes are robust to temperature changes are largely unknown. Temperature compensation, the ability of circadian rhythms to maintain a relatively constant period over the broad range of temperatures resulting from seasonal fluctuations in environmental conditions, is a defining property of circadian networks. Temperature affects the alternative splicing (AS) of several clock genes in fungi, plants, and flies, but the splicing factors that modulate these effects to ensure clock accuracy throughout the year remain to be identified. Here we show that GEMIN2, a spliceosomal small nuclear ribonucleoprotein assembly factor conserved from yeast to humans, modulates low temperature effects on a large subset of pre-mRNA splicing events. In particular, GEMIN2 controls the AS of several clock genes and attenuates the effects of temperature on the circadian period in Arabidopsis thaliana. We conclude that GEMIN2 is a key component of a posttranscriptional regulatory mechanism that ensures the appropriate acclimation of plants to daily and seasonal changes in temperature conditions.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Regulación de la Expresión Génica de las Plantas , Proteínas del Complejo SMN/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Arabidopsis/fisiología , Proteínas de Arabidopsis/fisiología , Ritmo Circadiano , Análisis por Conglomerados , Evolución Molecular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Prueba de Complementación Genética , Estudio de Asociación del Genoma Completo , Humanos , Intrones , Datos de Secuencia Molecular , Mutación , Proteínas del Tejido Nervioso/genética , Hojas de la Planta/fisiología , ARN Nuclear Pequeño/genética , Proteínas de Unión al ARN/genética , Proteínas del Complejo SMN/genética , Homología de Secuencia de Aminoácido , Empalmosomas/fisiología , Temperatura , Transcripción Genética
4.
PLoS One ; 8(12): e82871, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358231

RESUMEN

Nuclear bodies are large sub-nuclear structures composed of RNA and protein molecules. The Survival of Motor Neuron (SMN) protein localizes to Cajal bodies (CBs) and nuclear gems. Diminished cellular concentration of SMN is associated with the neurodegenerative disease Spinal Muscular Atrophy (SMA). How nuclear body architecture and its structural components influence neuronal differentiation remains elusive. In this study, we analyzed the effects of SMN and two of its interaction partners in cellular models of neuronal differentiation. The nuclear 23 kDa isoform of Fibroblast Growth Factor - 2 (FGF-2(23)) is one of these interacting proteins - and was previously observed to influence nuclear bodies by destabilizing nuclear gems and mobilizing SMN from Cajal bodies (CBs). Here we demonstrate that FGF-2(23) blocks SMN-promoted neurite outgrowth, and also show that SMN disrupts FGF-2(23)-dependent transcription. Our results indicate that FGF-2(23) and SMN form an inactive complex that interferes with neuronal differentiation by mutually antagonizing nuclear functions. Coilin is another nuclear SMN binding partner and a marker protein for Cajal bodies (CBs). In addition, coilin is essential for CB function in maturation of small nuclear ribonucleoprotein particles (snRNPs). The role of coilin outside of Cajal bodies and its putative impacts in tissue differentiation are poorly defined. The present study shows that protein levels of nucleoplasmic coilin outside of CBs decrease during neuronal differentiation. Overexpression of coilin has an inhibitory effect on neurite outgrowth. Furthermore, we find that nucleoplasmic coilin inhibits neurite outgrowth independent of SMN binding revealing a new function for coilin in neuronal differentiation.


Asunto(s)
Diferenciación Celular/genética , Núcleo Celular/metabolismo , Cuerpos Enrollados/metabolismo , Neurogénesis/genética , Neuronas/fisiología , Proteínas del Complejo SMN/fisiología , Animales , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Neuritas/fisiología , Proteínas Nucleares/metabolismo , Células PC12 , Unión Proteica , Ratas , Proteínas del Complejo SMN/metabolismo
5.
J Cell Biol ; 203(5): 835-47, 2013 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-24297751

RESUMEN

The neural crest, an embryonic stem cell population, initially resides within the dorsal neural tube but subsequently undergoes an epithelial-to-mesenchymal transition (EMT) to commence migration. Although neural crest and cancer EMTs are morphologically similar, little is known regarding conservation of their underlying molecular mechanisms. We report that Sip1, which is involved in cancer EMT, plays a critical role in promoting the neural crest cell transition to a mesenchymal state. Sip1 transcripts are expressed in premigratory/migrating crest cells. After Sip1 loss, the neural crest specifier gene FoxD3 was abnormally retained in the dorsal neuroepithelium, whereas Sox10, which is normally required for emigration, was diminished. Subsequently, clumps of adherent neural crest cells remained adjacent to the neural tube and aberrantly expressed E-cadherin while lacking N-cadherin. These findings demonstrate two distinct phases of neural crest EMT, detachment and mesenchymalization, with the latter involving a novel requirement for Sip1 in regulation of cadherin expression during completion of neural crest EMT.


Asunto(s)
Proteínas Aviares/fisiología , Transición Epitelial-Mesenquimal , Cresta Neural/metabolismo , Proteínas del Complejo SMN/fisiología , Animales , Proteínas Aviares/análisis , Proteínas Aviares/metabolismo , Embrión de Pollo , Técnicas de Silenciamiento del Gen , Cresta Neural/citología , Neurulación , Proteínas del Complejo SMN/análisis , Proteínas del Complejo SMN/metabolismo
7.
Uirusu ; 61(1): 73-80, 2011 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-21972558

RESUMEN

Reverse transcription of retroviral RNA into double stranded DNA is a characteristic feature of rertoviruses including human immunodeficiency virus type I (HIV-1). There has been accumulating evidence for the involvement of retroviral integrase (IN) in the reverse transcription of viral RNA. Here, we summarized recent our studies demonstrating direct functional roles of IN and its binding partner of host factor, Gemin2 in the reverse transcription. We established new in vitro cell-free assay to mimic natural reverse transcription and found that HIV-1 IN and host factor, Gemin2 synergistically stimulate reverse transcriptase (RT) activity. Analysis of intracellular stability and multimer formation of IN suggest that that high-ordered structures, especially tetramer formation of IN is critical for the function. In addition, Gemin2 might have a role to keep the higher-order structure of IN. Thus, we provide new aspects of reverse transcription of HIV-1 through IN and host factors in addition to RT.


Asunto(s)
Genoma Viral/genética , Integrasa de VIH/fisiología , Transcriptasa Inversa del VIH/fisiología , VIH-1/enzimología , VIH-1/genética , Transcripción Reversa , Proteínas del Complejo SMN/fisiología , Sistema Libre de Células , ADN , ADN Viral , ARN Viral/genética , Replicación Viral/genética
8.
Biochim Biophys Acta ; 1814(9): 1134-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21609790

RESUMEN

The survival motor neuron (SMN) protein plays an essential role in the assembly of uridine-rich small nuclear ribonuclear protein complexes. Phosphorylation of SMN can regulate its function, stability, and sub-cellular localization. This study shows that protein kinase A (PKA) phosphorylates SMN both in vitro and in vivo. Bioinformatic analysis predicts 12 potential PKA phosphorylation sites in human SMN. Mass spectrometric analysis of a tryptic digest of SMN after PKA phosphorylation identified five distinct phosphorylation sites in SMN (serines 4, 5, 8, 187 and threonine 85). Mutagenesis of this subset of PKA-phosphorylated sites in SMN affects association of SMN with Gemin2 and Gemin8. This result indicates that phosphorylation of SMN by PKA may play a role in regulation of the in vivo function of SMN.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas del Complejo SMN/química , Secuencia de Aminoácidos , Biología Computacional , Células HEK293 , Humanos , Datos de Secuencia Molecular , Fosforilación , Proteínas del Complejo SMN/fisiología
9.
PLoS Genet ; 6(10): e1001172, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21124729

RESUMEN

Spinal Muscular Atrophy (SMA) is caused by diminished function of the Survival of Motor Neuron (SMN) protein, but the molecular pathways critical for SMA pathology remain elusive. We have used genetic approaches in invertebrate models to identify conserved SMN loss of function modifier genes. Drosophila melanogaster and Caenorhabditis elegans each have a single gene encoding a protein orthologous to human SMN; diminished function of these invertebrate genes causes lethality and neuromuscular defects. To find genes that modulate SMN function defects across species, two approaches were used. First, a genome-wide RNAi screen for C. elegans SMN modifier genes was undertaken, yielding four genes. Second, we tested the conservation of modifier gene function across species; genes identified in one invertebrate model were tested for function in the other invertebrate model. Drosophila orthologs of two genes, which were identified originally in C. elegans, modified Drosophila SMN loss of function defects. C. elegans orthologs of twelve genes, which were originally identified in a previous Drosophila screen, modified C. elegans SMN loss of function defects. Bioinformatic analysis of the conserved, cross-species, modifier genes suggests that conserved cellular pathways, specifically endocytosis and mRNA regulation, act as critical genetic modifiers of SMN loss of function defects across species.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Invertebrados/genética , Interferencia de ARN , Proteínas del Complejo SMN/genética , Análisis de Varianza , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Evolución Molecular , Femenino , Genoma de los Helmintos/genética , Genoma de los Insectos/genética , Humanos , Invertebrados/crecimiento & desarrollo , Masculino , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , Mutación , Pupa/genética , Pupa/crecimiento & desarrollo , Proteínas del Complejo SMN/fisiología
11.
Proc Natl Acad Sci U S A ; 106(31): 12747-52, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19617559

RESUMEN

Nuclear bodies are distinct subnuclear structures. The survival of motoneuron (SMN) gene is mutated or deleted in patients with the neurodegenerative disease spinal muscular atrophy (SMA). The gene product SMN is a marker protein for one class of nuclear bodies denoted as nuclear gems. SMN has also been found in Cajal bodies, which co-localize with gems in many cell types. Interestingly, SMA patients display a reduced number of gems. Little is known about the regulation of nuclear body formation and stabilization. We have previously shown that a nuclear isoform of the fibroblast growth factor-2 (FGF-2(23)) binds directly to SMN. In this study, we analyzed the consequences of FGF-2(23) binding to SMN with regard to nuclear body formation. On a molecular level, we showed that FGF-2(23) competed with Gemin2 (a component of the SMN complex that is necessary for gem stabilization) for binding to SMN. Down-regulation of Gemin2 by siRNA caused destabilization of SMN-positive nuclear bodies. This process is reflected in both cellular and in vivo systems by a negative regulatory function of FGF-2 in nuclear body formation: in HEK293 cells, FGF-2(23) decreased the number of SMN-positive nuclear bodies. The same effect could be observed in motoneurons of FGF-2 transgenic mice. This study demonstrates the functional role of a growth factor in the regulation of structural entities of the nucleus.


Asunto(s)
Cuerpos Enrollados/fisiología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Gemini de los Cuerpos Enrollados/fisiología , Animales , Humanos , Inmunoprecipitación , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/fisiología , Proteínas de Unión al ARN/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Proteínas del Complejo SMN/análisis , Proteínas del Complejo SMN/fisiología
12.
Results Probl Cell Differ ; 48: 289-326, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19343312

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease that results from loss of function of the SMN1 gene, encoding the ubiquitously expressed survival of motor neuron (SMN) protein, a protein best known for its housekeeping role in the SMN-Gemin multiprotein complex involved in spliceosomal small nuclear ribonucleoprotein (snRNP) assembly. However, numerous studies reveal that SMN has many interaction partners, including mRNA binding proteins and actin regulators, suggesting its diverse role as a molecular chaperone involved in mRNA metabolism. This review focuses on studies suggesting an important role of SMN in regulating the assembly, localization, or stability of axonal messenger ribonucleoprotein (mRNP) complexes. Various animal models for SMA are discussed, and phenotypes described that indicate a predominant function for SMN in neuronal development and synapse formation. These models have begun to be used to test different therapeutic strategies that have the potential to restore SMN function. Further work to elucidate SMN mechanisms within motor neurons and other cell types involved in neuromuscular circuitry hold promise for the potential treatment of SMA.


Asunto(s)
Axones/fisiología , Modelos Biológicos , Atrofia Muscular Espinal , ARN Mensajero , Proteínas del Complejo SMN/fisiología , Proteína 1 para la Supervivencia de la Neurona Motora/fisiología , Actinas/metabolismo , Animales , Caenorhabditis elegans , Modelos Animales de Enfermedad , Drosophila , Humanos , Ratones , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , ARN Mensajero/metabolismo , Proteínas del Complejo SMN/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Pez Cebra
13.
Nucleic Acids Res ; 37(2): 582-90, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19066202

RESUMEN

In eukaryotic cells translation initiation occurs through two alternative mechanisms, a cap-dependent operating in the majority of mRNAs, and a 5'-end-independent driven by internal ribosome entry site (IRES) elements, specific for a subset of mRNAs. IRES elements recruit the translation machinery to an internal position in the mRNA through a mechanism involving the IRES structure and several trans-acting factors. Here, we identified Gemin5 protein bound to the foot-and-mouth disease virus (FMDV) and hepatitis C virus (HCV) IRES using two independent approaches, riboproteomic analysis and immunoprecipitation of photocrosslinked factors. Functional analysis performed in Gemin5 shRNA-depleted cells, or in in vitro translation reactions, revealed an unanticipated role of Gemin5 in translation control as a down-regulator of cap-dependent and IRES-driven translation initiation. Consistent with this, pull-down assays showed that Gemin5 forms part of two distinct complexes, a specific IRES-ribonucleoprotein complex and an IRES-independent protein complex containing eIF4E. Thus, beyond its role in snRNPs biogenesis, Gemin5 also functions as a modulator of translation activity.


Asunto(s)
Iniciación de la Cadena Peptídica Traduccional , ARN Mensajero/química , ARN Viral/química , Secuencias Reguladoras de Ácido Ribonucleico , Proteínas del Complejo SMN/fisiología , Línea Celular , Regulación hacia Abajo , Humanos , Péptidos/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas del Complejo SMN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...