Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Biosci Rep ; 32(2): 131-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22115364

ABSTRACT

NF1 (neurofibromatosis type I) is a common genetic disease that affects one in 3500 individuals. The disease is completely penetrant but shows variable phenotypic expression in patients. NF1 is a large gene, and its pre-mRNA undergoes alternative splicing. The NF1 protein, neurofibromin, is involved in diverse signalling cascades. One of the best characterized functions of NF1 is its function as a Ras-GAP (GTPase-activating protein). NF1 exon 23a is an alternative exon that lies within the GAP-related domain of neurofibromin. This exon is predominantly included in most tissues, and it is skipped in CNS (central nervous system) neurons. The isoform in which exon 23a is skipped has 10 times higher Ras-GAP activity than the isoform in which exon 23a is included. Exon 23a inclusion is tightly regulated by at least three different families of RNA-binding proteins: CELF {CUG-BP (cytosine-uridine-guanine-binding protein) and ETR-3 [ELAV (embryonic lethal abnormal vision)-type RNA-binding protein]-like factor}, Hu and TIA-1 (T-cell intracellular antigen 1)/TIAR (T-cell intracellular antigen 1-related protein). The CELF and Hu proteins promote exon 23a skipping, while the TIA-1/TIAR proteins promote its inclusion. The widespread clinical variability that is observed among NF1 patients cannot be explained by NF1 mutations alone and it is believed that modifier genes may have a role in the variability. We suggest that the regulation of alternative splicing may act as a modifier to contribute to the variable expression in NF1 patients.


Subject(s)
Alternative Splicing , Genes, Neurofibromatosis 1 , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , RNA Precursors/genetics , Animals , Humans , Neurofibromatosis 1/metabolism , Neurofibromin 1/metabolism , Phenotype , RNA Precursors/metabolism
2.
Proc Natl Acad Sci U S A ; 108(36): E627-35, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21808035

ABSTRACT

Recent studies have provided strong evidence for a regulatory link among chromatin structure, histone modification, and splicing regulation. However, it is largely unknown how local histone modification patterns surrounding alternative exons are connected to differential alternative splicing outcomes. Here we show that splicing regulator Hu proteins can induce local histone hyperacetylation by association with their target sequences on the pre-mRNA surrounding alternative exons of two different genes. In both primary and mouse embryonic stem cell-derived neurons, histone hyperacetylation leads to an increased local transcriptional elongation rate and decreased inclusion of these exons. Furthermore, we demonstrate that Hu proteins interact with histone deacetylase 2 and inhibit its deacetylation activity. We propose that splicing regulators may actively modulate chromatin structure when recruited to their target RNA sequences cotranscriptionally. This "reaching back" interaction with chromatin provides a means to ensure accurate and efficient regulation of alternative splicing.


Subject(s)
Alternative Splicing/physiology , Chromatin/metabolism , ELAV Proteins/metabolism , Histones/metabolism , Neurons/metabolism , RNA Precursors/metabolism , Acetylation , Animals , Cells, Cultured , Exons/physiology , Histone Deacetylase 2/metabolism , Mice , Neurons/cytology , Transcription, Genetic/physiology
3.
Nucleic Acids Res ; 38(1): 253-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19854948

ABSTRACT

The CUG-BP and ETR-3 like factors (CELF) are a family of six highly conserved RNA-binding proteins that preferentially bind to UG-rich sequences. One of the key functions of these proteins is to mediate alternative splicing in a number of tissues, including brain, heart and muscle. To fully understand the function of CELF proteins, it is important to identify downstream targets of CELF proteins. In this communication, we report that neurofibromatosis type I (NF1) exon 23a is a novel target of CELF protein-mediated splicing regulation in neuron-like cells. NF1 regulates Ras signaling, and the isoform that excludes exon 23a shows 10 times greater ability to down-regulate Ras signaling than the isoform that includes exon 23a. Five of the six CELF proteins strongly suppress the inclusion of NF1 exon 23a. Over-expression or siRNA knockdown of these proteins in cell transfection experiments altered the levels of NF1 exon 23a inclusion. In vitro binding and splicing analyses demonstrate that CELF proteins block splicing through interfering with binding of U2AF(65). These studies, combined with our previous investigations demonstrating a role for Hu proteins and TIA-1/TIAR in controlling NF1 exon 23a inclusion, highlight the complex nature of regulation of this important alternative splicing event.


Subject(s)
Alternative Splicing , CCAAT-Enhancer-Binding Protein-delta/metabolism , Neurofibromin 1/genetics , RNA Precursors/metabolism , RNA, Messenger/metabolism , Animals , Base Sequence , Cell Line, Tumor , Exons , HeLa Cells , Humans , Molecular Sequence Data , Neurofibromin 1/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/chemistry , Rats , Ribonucleoproteins/metabolism , Splicing Factor U2AF
4.
Mol Biol Cell ; 17(12): 5105-14, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17035636

ABSTRACT

Recent advances in genome-wide analysis of alternative splicing indicate that extensive alternative RNA processing is associated with many proteins that play important roles in the nervous system. Although differential splicing and polyadenylation make significant contributions to the complexity of the nervous system, our understanding of the regulatory mechanisms underlying the neuron-specific pathways is very limited. Mammalian neuron-specific embryonic lethal abnormal visual-like Hu proteins (HuB, HuC, and HuD) are a family of RNA-binding proteins implicated in neuronal differentiation and maintenance. It has been established that Hu proteins increase expression of proteins associated with neuronal function by up-regulating mRNA stability and/or translation in the cytoplasm. We report here a novel function of these proteins as RNA processing regulators in the nucleus. We further elucidate the underlying mechanism of this regulation. We show that in neuron-like cells, Hu proteins block the activity of TIA-1/TIAR, two previously identified, ubiquitously expressed proteins that promote the nonneuronal pathway of calcitonin/calcitonin gene-related peptide (CGRP) pre-mRNA processing. These studies define not only the first neuron-specific regulator of the calcitonin/CGRP system but also the first nuclear function of Hu proteins.


Subject(s)
Alternative Splicing/genetics , Cell Nucleus/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , Animals , Base Sequence , CHO Cells , Calcitonin/genetics , Calcitonin Gene-Related Peptide/genetics , Cricetinae , Cricetulus , Enhancer Elements, Genetic/genetics , Exons/genetics , HeLa Cells , Humans , Introns/genetics , Mice , Molecular Sequence Data , Organ Specificity , Poly(A)-Binding Proteins/metabolism , Protein Binding , RNA Precursors/metabolism , Rats , T-Cell Intracellular Antigen-1 , Uridine/chemistry , Uridine/genetics , Uridine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...