Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 50(22): 12979-12996, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36533443

RESUMEN

Aggregation of the microtubule-associated protein tau characterizes tauopathies, including Alzheimer's disease and frontotemporal lobar degeneration (FTLD-Tau). Gene expression regulation of tau is complex and incompletely understood. Here we report that the human tau gene (MAPT) generates two circular RNAs (circRNAs) through backsplicing of exon 12 to either exon 7 (12→7 circRNA) or exon 10 (12→10 circRNA). Both circRNAs lack stop codons. The 12→7 circRNA contains one start codon and is translated in a rolling circle, generating a protein consisting of multimers of the microtubule-binding repeats R1-R4. For the 12→10 circRNA, a start codon can be introduced by two FTLD-Tau mutations, generating a protein consisting of multimers of the microtubule-binding repeats R2-R4, suggesting that mutations causing FTLD may act in part through tau circRNAs. Adenosine to inosine RNA editing dramatically increases translation of circRNAs and, in the 12→10 circRNA, RNA editing generates a translational start codon by changing AUA to AUI. Circular tau proteins self-aggregate and promote aggregation of linear tau proteins. Our data indicate that adenosine to inosine RNA editing initiates translation of human circular tau RNAs, which may contribute to tauopathies.


Asunto(s)
Tauopatías , Proteínas tau , Humanos , Adenosina/metabolismo , Codón Iniciador , Inosina/metabolismo , ARN/genética , ARN/metabolismo , Edición de ARN , ARN Circular/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/genética , Tauopatías/metabolismo
2.
Infect Immun ; 90(3): e0058621, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35099275

RESUMEN

The mechanisms through which oral commensal bacteria mitigates uncontrolled inflammatory responses of the oral mucosa remain unknown. Here, we show that representative oral bacterial species normally associated with oral health [S. gordonii (Sg), V. parvula (Vp), A. naeslundii (An), C. sputigena (Cs), and N. mucosa (Nm)] enhanced differential chemokine responses in oral epithelial cells (OECs), with some bacteria (An, Vp, and Nm) inducing higher chemokine levels (CXCL1, CXCL8) than others (Sg, Cs). Although all bacterial species (except Cs) increased CCL20 mRNA levels consistent with protein elevations in cell lysates, only An, Vp, and Nm induced higher CCL20 secretion, similar to the effect of the oral pathogen F. nucleatum (Fn). In contrast, most CCL20 remained associated with OECs exposed to Sg and negligible amounts released into the cell supernatants. Consistently, Sg attenuated An-induced CCL20. MiR-4516 and miR-663a were identified as Sg-specifically induced miRNAs modulating validated targets of chemokine-associated pathways. Cell transfection with miR-4516 and miR-663a decreased An- and Fn-induced CCL20. MiRNA upregulation and attenuation of An-induced CCL20 by Sg were reversed by catalase. Up-regulation of both miRNAs was specifically enhanced by oral streptococci H2O2-producers. These findings suggest that CCL20 levels produced by OECs in response to bacterial challenge are regulated by Sg-induced miR-4516 and miR-663a in a mechanism that involves hydrogen peroxide. This type of molecular mechanism could partly explain the central role of specific oral streptococcal species in balancing inflammatory and antimicrobial responses given the critical role of CCL20 in innate (antimicrobial) and adaptive immunity (modulates Th17 responses).


Asunto(s)
MicroARNs , Streptococcus gordonii , Bacterias/genética , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Células Epiteliales/microbiología , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Mucosa Bucal
3.
Bioessays ; 39(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28505386

RESUMEN

C/D box snoRNAs (SNORDs) are an abundantly expressed class of short, non-coding RNAs that have been long known to perform 2'-O-methylation of rRNAs. However, approximately half of human SNORDs have no predictable rRNA targets, and numerous SNORDs have been associated with diseases that show no defects in rRNAs, among them Prader-Willi syndrome, Duplication 15q syndrome and cancer. This apparent discrepancy has been addressed by recent studies showing that SNORDs can act to regulate pre-mRNA alternative splicing, mRNA abundance, activate enzymes, and be processed into shorter ncRNAs resembling miRNAs and piRNAs. Furthermore, recent biochemical studies have shown that a given SNORD can form both methylating and non-methylating ribonucleoprotein complexes, providing an indication of the likely physical basis for such diverse new functions. Thus, SNORDs are more structurally and functionally diverse than previously thought, and their role in gene expression is under-appreciated. The action of SNORDs in non-methylating complexes can be substituted with oligonucleotides, allowing devising therapies for diseases like Prader-Willi syndrome.


Asunto(s)
Regulación de la Expresión Génica , ARN Nucleolar Pequeño/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Humanos , Metilación , Síndrome de Prader-Willi/tratamiento farmacológico , Síndrome de Prader-Willi/metabolismo , Precursores del ARN/metabolismo , ARN Ribosómico/metabolismo , Levaduras/genética , Levaduras/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(12): E1625-34, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26957605

RESUMEN

C/D box small nucleolar RNAs (SNORDs) are small noncoding RNAs, and their best-understood function is to target the methyltransferase fibrillarin to rRNA (for example, SNORD27 performs 2'-O-methylation of A27 in 18S rRNA). Unexpectedly, we found a subset of SNORDs, including SNORD27, in soluble nuclear extract made under native conditions, where fibrillarin was not detected, indicating that a fraction of the SNORD27 RNA likely forms a protein complex different from canonical snoRNAs found in the insoluble nuclear fraction. As part of this previously unidentified complex,SNORD27 regulates the alternative splicing of the transcription factor E2F7p re-mRNA through direct RNA-RNA interaction without methylating the RNA, likely by competing with U1 small nuclear ribonucleoprotein (snRNP). Furthermore, knockdown of SNORD27 activates previously "silent" exons in several other genes through base complementarity across the entire SNORD27 sequence, not just the antisense boxes. Thus, some SNORDs likely function in both rRNA and pre-mRNA processing, which increases the repertoire of splicing regulators and links both processes.


Asunto(s)
Empalme Alternativo , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , ARN Ribosómico/metabolismo , ARN Nucleolar Pequeño/fisiología , Emparejamiento Base , Secuencia de Bases , Ciclo Celular , División Celular , Fraccionamiento Celular/métodos , Núcleo Celular/química , Proteínas Cromosómicas no Histona/análisis , Factor de Transcripción E2F7/genética , Exones/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Metilación , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/genética , Biogénesis de Organelos , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Ribosomas/metabolismo , Solubilidad , Empalmosomas/metabolismo
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 2753-2760, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29729314

RESUMEN

The microtubule-associated protein Tau, generated by the MAPT gene is involved in dozens of neurodegenerative conditions ("tauopathies"), including Alzheimer's disease (AD) and frontotemporal lobar degeneration/frontotemporal dementia (FTLD/FTD). The pre-mRNA of MAPT is well studied and its aberrant pre-mRNA splicing is associated with frontotemporal dementia. Using a PCR screen of RNA from human brain tissues, we found that the MAPT locus generates circular RNAs through a backsplicing mechanism from exon 12 to either exon 10 or 7. MAPT circular RNAs are localized in the cytosol and contain open reading frames encoding Tau protein fragments. The MAPT exon 10 is alternatively spliced and proteins involved in its regulation, such as CLK2, SRSF7/9G8, PP1 (protein phosphatase 1) and NIPP1 (nuclear inhibitor of PP1) reduce the abundance of the circular MAPT exon 12 → 10 backsplice RNA after being transfected into cultured HEK293 cells. In summary, we report the identification of new bona fide human brain RNAs produced from the MAPT locus. These may be a component of normal human brain Tau regulation and, since the circular RNAs could generate high molecular weight proteins with multiple microtubule binding sites, they could contribute to taupathies.


Asunto(s)
Demencia Frontotemporal/genética , Precursores del ARN/genética , ARN/genética , Tauopatías/genética , Proteínas tau/genética , Anciano , Anciano de 80 o más Años , Empalme Alternativo , Encéfalo/metabolismo , Encéfalo/patología , Exones/genética , Femenino , Demencia Frontotemporal/patología , Células HEK293 , Humanos , Masculino , Mutación , ARN Circular , Tauopatías/patología
6.
Hum Genet ; 136(9): 1079-1091, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28664341

RESUMEN

The central nervous system-specific serotonin receptor 2C (5HT2C) controls key physiological functions, such as food intake, anxiety, and motoneuron activity. Its deregulation is involved in depression, suicidal behavior, and spasticity, making it the target for antipsychotic drugs, appetite controlling substances, and possibly anti-spasm agents. Through alternative pre-mRNA splicing and RNA editing, the 5HT2C gene generates at least 33 mRNA isoforms encoding 25 proteins. The 5HT2C is a G-protein coupled receptor that signals through phospholipase C, influencing the expression of immediate/early genes like c-fos. Most 5HT2C isoforms show constitutive activity, i.e., signal without ligand binding. The constitutive activity of 5HT2C is decreased by pre-mRNA editing as well as alternative pre-mRNA splicing, which generates a truncated isoform that switches off 5HT2C receptor activity through heterodimerization; showing that RNA processing regulates the constitutive activity of the 5HT2C system. RNA processing events influencing the constitutive activity target exon Vb that forms a stable double stranded RNA structure with its downstream intron. This structure can be targeted by small molecules and oligonucleotides that change exon Vb alternative splicing and influence 5HT2C signaling in mouse models, leading to a reduction in food intake. Thus, the 5HT2C system is a candidate for RNA therapy in multiple models of CNS disorders.


Asunto(s)
Empalme Alternativo , Exones , Multimerización de Proteína , Precursores del ARN , Receptores de Serotonina , Animales , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/terapia , Humanos , Ratones , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , Receptores de Serotonina/biosíntesis , Receptores de Serotonina/genética
7.
J Biol Chem ; 290(28): 17269-81, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-26013829

RESUMEN

Transformer 2ß1 (Tra2ß1) is a splicing effector protein composed of a core RNA recognition motif flanked by two arginine-serine-rich (RS) domains, RS1 and RS2. Although Tra2ß1-dependent splicing is regulated by phosphorylation, very little is known about how protein kinases phosphorylate these two RS domains. We now show that the serine-arginine protein kinase-1 (SRPK1) is a regulator of Tra2ß1 and promotes exon inclusion in the survival motor neuron gene 2 (SMN2). To understand how SRPK1 phosphorylates this splicing factor, we performed mass spectrometric and kinetic experiments. We found that SRPK1 specifically phosphorylates 21 serines in RS1, a process facilitated by a docking groove in the kinase domain. Although SRPK1 readily phosphorylates RS2 in a splice variant lacking the N-terminal RS domain (Tra2ß3), RS1 blocks phosphorylation of these serines in the full-length Tra2ß1. Thus, RS2 serves two new functions. First, RS2 positively regulates binding of the central RNA recognition motif to an exonic splicing enhancer sequence, a phenomenon reversed by SRPK1 phosphorylation on RS1. Second, RS2 enhances ligand exchange in the SRPK1 active site allowing highly efficient Tra2ß1 phosphorylation. These studies demonstrate that SRPK1 is a regulator of Tra2ß1 splicing function and that the individual RS domains engage in considerable cross-talk, assuming novel functions with regard to RNA binding, splicing, and SRPK1 catalysis.


Asunto(s)
Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Sitios de Unión , Exones , Células HEK293 , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , ARN/genética , ARN/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/química , Factores de Empalme Serina-Arginina , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
8.
Bioorg Med Chem Lett ; 26(3): 965-968, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26725024

RESUMEN

Phosphorylation and dephosphorylation of splicing factors play a key role in pre-mRNA splicing events, and cantharidin and norcantharidin analogs inhibit protein phosphatase-1 (PP1) and change alternative pre-mRNA splicing. Targeted inhibitors capable of selectively inhibiting PP-1 could promote exon 7 inclusion in the survival-of-motorneuron-2 gene (SMN2) and shift the proportion of SMN2 protein from a dysfunctional to a functional form. As a prelude to the development of norcantharidin-tethered oligonucleotide inhibitors, the synthesis a norcantharidin-tethered guanosine was developed in which a suitable tether prevented the undesired cyclization of norcantharidin monoamides to imides and possessed a secondary amine terminus suited to the synthesis of oligonucleotides analogs. Application of this methodology led to the synthesis of a diastereomeric mixture of norcantharidin-tethered guanosines, namely bisammonium (1R,2S,3R,4S)- and (1S,2R,3S,4R)-3-((4-(2-(((((2R,3R,4R,5R)-5-(2-amino-6-oxo-1,6-dihydro-9H-purin-9-yl)-2-(hydroxymethyl)-4-methoxytetrahydrofuran-3-yl)oxy)oxidophosphoryl)oxy)ethyl)-phenethyl)(methyl)carbamoyl)-7-oxabicyclo[2.2.1]heptane-2-carboxylate, which showed activity in an assay for SMN2 pre-mRNA splicing.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/química , Inhibidores Enzimáticos/síntesis química , Guanosina/análogos & derivados , Proteína Fosfatasa 1/antagonistas & inhibidores , Empalme Alternativo , Animales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Guanosina/síntesis química , Guanosina/metabolismo , Células HEK293 , Humanos , Ratones , Proteína Fosfatasa 1/metabolismo , ARN Mensajero/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
9.
Nucleic Acids Res ; 42(8): 4947-61, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24623796

RESUMEN

Sudemycin E is an analog of the pre-messenger RNA splicing modulator FR901464 and its derivative spliceostatin A. Sudemycin E causes the death of cancer cells through an unknown mechanism. We found that similar to spliceostatin A, sudemycin E binds to the U2 small nuclear ribonucleoprotein (snRNP) component SF3B1. Native chromatin immunoprecipitations showed that U2 snRNPs physically interact with nucleosomes. Sudemycin E induces a dissociation of the U2 snRNPs and decreases their interaction with nucleosomes. To determine the effect on gene expression, we performed genome-wide array analysis. Sudemycin E first causes a rapid change in alternative pre-messenger RNA splicing, which is later followed by changes in overall gene expression and arrest in the G2 phase of the cell cycle. The changes in alternative exon usage correlate with a loss of the H3K36me3 modification in chromatin encoding these exons. We propose that sudemycin E interferes with the ability of U2 snRNP to maintain an H3K36me3 modification in actively transcribed genes. Thus, in addition to the reversible changes in alternative splicing, sudemycin E causes changes in chromatin modifications that result in chromatin condensation, which is a likely contributing factor to cancer cell death.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Antineoplásicos/farmacología , Cromatina/efectos de los fármacos , Compuestos Epoxi/farmacología , Compuestos de Espiro/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Línea Celular Tumoral , Células Cultivadas , Cromatina/química , Citotoxinas/toxicidad , Compuestos Epoxi/metabolismo , Compuestos Epoxi/toxicidad , Expresión Génica/efectos de los fármacos , Células HEK293 , Histonas/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Compuestos de Espiro/metabolismo , Compuestos de Espiro/toxicidad
10.
Bioessays ; 35(1): 46-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23180440

RESUMEN

Recent experimental evidence suggests that most of the genome is transcribed into non-coding RNAs. The initial transcripts undergo further processing generating shorter, metabolically stable RNAs with diverse functions. Small nucleolar RNAs (snoRNAs) are non-coding RNAs that modify rRNAs, tRNAs, and snRNAs that were considered stable. We review evidence that snoRNAs undergo further processing. High-throughput sequencing and RNase protection experiments showed widespread expression of snoRNA fragments, known as snoRNA-derived RNAs (sdRNAs). Some sdRNAs resemble miRNAs, these can associate with argonaute proteins and influence translation. Other sdRNAs are longer, form complexes with hnRNPs and influence gene expression. C/D box snoRNA fragmentation patterns are conserved across multiple cell types, suggesting a processing event, rather than degradation. The loss of expression from genetic loci that generate canonical snoRNAs and processed snoRNAs results in diseases, such as Prader-Willi Syndrome, indicating possible physiological roles for processed snoRNAs. We propose that processed snoRNAs acquire new roles in gene expression and represent a new class of regulatory RNAs distinct from canonical snoRNAs.


Asunto(s)
Regulación de la Expresión Génica , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Proteínas Argonautas , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Síndrome de Prader-Willi/genética , Biosíntesis de Proteínas/genética , ARN Ribosómico/genética , ARN de Transferencia/genética
11.
Nucleic Acids Res ; 41(6): 3819-32, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23393189

RESUMEN

The serotonin receptor 2C plays a central role in mood and appetite control. It undergoes pre-mRNA editing as well as alternative splicing. The RNA editing suggests that the pre-mRNA forms a stable secondary structure in vivo. To identify substances that promote alternative exons inclusion, we set up a high-throughput screen and identified pyrvinium pamoate as a drug-promoting exon inclusion without editing. Circular dichroism spectroscopy indicates that pyrvinium pamoate binds directly to the pre-mRNA and changes its structure. SHAPE (selective 2'-hydroxyl acylation analysed by primer extension) assays show that part of the regulated 5'-splice site forms intramolecular base pairs that are removed by this structural change, which likely allows splice site recognition and exon inclusion. Genome-wide analyses show that pyrvinium pamoate regulates >300 alternative exons that form secondary structures enriched in A-U base pairs. Our data demonstrate that alternative splicing of structured pre-mRNAs can be regulated by small molecules that directly bind to the RNA, which is reminiscent to an RNA riboswitch.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Compuestos de Pirvinio/farmacología , ARN Mensajero/efectos de los fármacos , Receptor de Serotonina 5-HT2C/genética , Secuencia de Bases , Exones , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Filogenia , Compuestos de Pirvinio/metabolismo , Edición de ARN , Precursores del ARN/metabolismo , ARN Bicatenario/química , ARN Bicatenario/efectos de los fármacos , ARN Mensajero/química , ARN Mensajero/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Empalmosomas/metabolismo
12.
Biochim Biophys Acta ; 1832(8): 1129-35, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23545117

RESUMEN

Mutations in the RNA binding protein FUS (fused in sarcoma) have been linked to a subset of familial amyotrophic lateral sclerosis (ALS) cases. The mutations are clustered in the C-terminal nuclear localization sequence (NLS). Various FUS mutants accumulate in the cytoplasm whereas wild-type (WT) FUS is mainly nuclear. Here we investigate the effect of one ALS causing mutant (FUS-ΔNLS, also known as R495X) on pre-mRNA splicing and RNA expression using genome wide exon-junction arrays. Using a non-neuronal stable cell line with inducible FUS expression, we detected early changes in RNA composition. In particular, mutant FUS-ΔNLS increased calcium/calmodulin-dependent protein kinase II inhibitor 2 (CAMK2N2) at both mRNA and protein levels, whereas WT-FUS had no effect. Chromatin immunoprecipitation experiments showed that FUS-ΔNLS accumulated at the CAMK2N2 promoter region, whereas promoter occupation by WT-FUS remained constant. Given the loss of FUS-ΔNLS in the nucleus through the mutation-induced translocation, this increase of promoter occupancy is surprising. It indicates that, despite the obvious cytoplasmic accumulation, FUS-ΔNLS can act through a nuclear gain of function mechanism.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Mutación , Proteína FUS de Unión a ARN/genética , Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Línea Celular , Núcleo Celular/genética , Cromatina/genética , Cromatina/metabolismo , Citoplasma/genética , Exones , Estudio de Asociación del Genoma Completo/métodos , Células HEK293 , Humanos , Regiones Promotoras Genéticas , Precursores del ARN/genética , Empalme del ARN/genética , Proteína FUS de Unión a ARN/biosíntesis
13.
EMBO J ; 29(7): 1235-47, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20186123

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by loss of motor neurons in patients with null mutations in the SMN1 gene. An almost identical SMN2 gene is unable to compensate for this deficiency because a single C-to-T transition at position +6 in exon-7 causes skipping of the exon by a mechanism not yet fully elucidated. We observed that the C-to-T transition in SMN2 creates a putative binding site for the RNA-binding protein Sam68. RNA pull-down assays and UV-crosslink experiments showed that Sam68 binds to this sequence. In vivo splicing assays showed that Sam68 triggers SMN2 exon-7 skipping. Moreover, mutations in the Sam68-binding site of SMN2 or in the RNA-binding domain of Sam68 completely abrogated its effect on exon-7 skipping. Retroviral infection of dominant-negative mutants of Sam68 that interfere with its RNA-binding activity, or with its binding to the splicing repressor hnRNP A1, enhanced exon-7 inclusion in endogenous SMN2 and rescued SMN protein expression in fibroblasts of SMA patients. Our results thus indicate that Sam68 is a novel crucial regulator of SMN2 splicing.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Proteínas de Unión al ADN/metabolismo , Atrofia Muscular Espinal/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular , Secuencia de Consenso , Proteínas de Unión al ADN/genética , Exones , Fibroblastos/metabolismo , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Atrofia Muscular Espinal/genética , Mutación , Unión Proteica , ARN/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
14.
Mol Carcinog ; 53(11): 883-92, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23765422

RESUMEN

The ubiquitously expressed splicing factor YT521 (YTHDC1) is characterized by alternatively spliced isoforms with regulatory impact on cancer-associated gene expression. Our recent findings account for the prognostic significance of YT521 in endometrial cancer. In this study, we investigated the hypoxia-dependency of YT521 expression as well as its differential isoform activities on oncological important target genes. YT521's potential regulatory influence on splicing was investigated by a minigene assay for the specific target gene CD44. Functional splicing analysis was performed by YT521 knock-down or overexpression, respectively. In addition, YT521 expression was determined under hypoxia. The two protein-generating YT521 mRNA isoforms 1 and 2 caused a comparable, specific induction of CD44v alternative splicing (P < 0.01). In a number of oncological target genes, YT521 upregulation significantly altered BRCA2 expression pattern, while YT521 knock-down created a significant regulatory impact on PGR expression, respectively. Hypoxia induced a specific switch towards the processing of two non-protein-coding mRNA variants, of which one is described for the first time in this study. The presented study underlines the comparable regulatory potential of both YT521 isoforms 1 and 2, on the investigated target genes in vivo and in vitro. Hypoxia induces a specific switch in YT521 expression pattern towards the two non-protein coding mRNA variants, the already characterized isoform 3 and the newly discovered exon 8-skipping isoform. The altered YT521 alternative splicing is functionally coupled with nonsense-mediated decay and can be interpreted as regulated unproductive splicing and transcription with consecutive impact on the processing of specific cancer-associated genes, such as BRCA2 and PGR.


Asunto(s)
Neoplasias/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Empalme Alternativo , Proteína BRCA2/biosíntesis , Hipoxia de la Célula/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Células MCF-7 , Proteínas Nucleares/biosíntesis , Isoformas de Proteínas/genética , Interferencia de ARN , Factores de Empalme de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño
15.
Nucleic Acids Res ; 40(9): 4025-39, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22210893

RESUMEN

Alternative pre-mRNA processing is a central element of eukaryotic gene regulation. The cell frequently alters the use of alternative exons in response to physiological stimuli. Ceramides are lipid-signaling molecules composed of sphingosine and a fatty acid. Previously, water-insoluble ceramides were shown to change alternative splicing and decrease SR-protein phosphorylation by activating protein phosphatase-1 (PP1). To gain further mechanistical insight into ceramide-mediated alternative splicing, we analyzed the effect of C6 pyridinium ceramide (PyrCer) on alternative splice site selection. PyrCer is a water-soluble ceramide analog that is under investigation as a cancer drug. We found that PyrCer binds to the PP1 catalytic subunit and inhibits the dephosphorylation of several splicing regulatory proteins containing the evolutionarily conserved RVxF PP1-binding motif (including PSF/SFPQ, Tra2-beta1 and SF2/ASF). In contrast to natural ceramides, PyrCer promotes phosphorylation of splicing factors. Exons that are regulated by PyrCer have in common suboptimal splice sites, are unusually short and share two 4-nt motifs, GAAR and CAAG. They are dependent on PSF/SFPQ, whose phosphorylation is regulated by PyrCer. Our results indicate that lipids can influence pre-mRNA processing by regulating the phosphorylation status of specific regulatory factors, which is mediated by protein phosphatase activity.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Ceramidas/farmacología , Inhibidores Enzimáticos/farmacología , Proteína Fosfatasa 1/antagonistas & inhibidores , Compuestos de Piridinio/farmacología , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Sitios de Unión , Exones , Células HEK293 , Células HeLa , Humanos , Fosforilación/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo
16.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167036, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38286213

RESUMEN

At least 53 mutations in the microtubule associated protein tau gene (MAPT) have been identified that cause frontotemporal dementia. 47 of these mutations are localized between exons 7 and 13. They could thus affect the formation of circular RNAs (circRNAs) from the MAPT gene that occurs through backsplicing from exon 12 to either exon 10 or exon 7. We analyzed representative mutants and found that five FTDP-17 mutations increase the formation of 12➔7 circRNA and three different mutations increase the amount of 12➔10 circRNA. CircRNAs are translated after undergoing adenosine to inosine RNA editing, catalyzed by ADAR enzymes. We found that the interferon induced ADAR1-p150 isoform has the strongest effect on circTau RNA translation. ADAR1-p150 activity had a stronger effect on circTau RNA expression and strongly decreased 12➔7 circRNA, but unexpectedly increased 12➔10 circRNA. In both cases, ADAR-activity strongly promoted translation of circTau RNAs. Unexpectedly, we found that the 12➔7 circTau protein interacts with eukaryotic initiation factor 4B (eIF4B), which is reduced by four FTDP-17 mutations located in the second microtubule domain. These are the first studies of the effect of human mutations on circular RNA formation and translation. They show that point mutations influence circRNA expression levels, likely through changes in pre-mRNA structures. The effect of the mutations is surpassed by editing of the circular RNAs, leading to their translation. Thus, circular RNAs and their editing status should be considered when analyzing FTDP-17 mutations.


Asunto(s)
Demencia Frontotemporal , Humanos , Demencia Frontotemporal/genética , Mutación , ARN/genética , ARN Circular/genética , Proteínas tau/genética
17.
J Biol Chem ; 287(37): 31185-94, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22782896

RESUMEN

The glycine receptor-deficient mutant mouse spastic carries a full-length long interspersed nuclear element (LINE1) retrotransposon in intron 6 of the glycine receptor ß subunit gene, Glrb(spa). The mutation arose in the C57BL/6J strain and is associated with skipping of exon 6 or a combination of the exons 5 and 6, thus resulting in a translational frameshift within the coding regions of the GlyR ß subunit. The effect of the Glrb(spa) LINE1 insertion on pre-mRNA splicing was studied using a minigene approach. Sequence comparison as well as motif prediction and mutational analysis revealed that in addition to the LINE1 insertion the inactivation of an exonic splicing enhancer (ESE) within exon 6 is required for skipping of exon 6. Reconstitution of the ESE by substitution of a single residue was sufficient to prevent exon skipping. In addition to the ESE, two regions within the 5' and 3' UTR of the LINE1 were shown to be critical determinants for exon skipping, indicating that LINE1 acts as efficient modifier of subtle endogenous splicing phenotypes. Thus, the spastic allele of the murine glycine receptor ß subunit gene is a two-hit mutation, where the hypomorphic alteration in an ESE is amplified by the insertion of a LINE1 element in the adjacent intron. Conversely, the LINE1 effect on splicing may be modulated by individual polymorphisms, depending on the insertional environment within the host genome.


Asunto(s)
Alelos , Elementos de Nucleótido Esparcido Largo/fisiología , Polimorfismo Genético , Precursores del ARN/genética , Empalme del ARN/genética , Receptores de Glicina/genética , Regiones no Traducidas 3'/fisiología , Regiones no Traducidas 5'/fisiología , Animales , Exones/fisiología , Células HEK293 , Humanos , Intrones/fisiología , Ratones , Ratones Mutantes , Mutación , Precursores del ARN/metabolismo , Receptores de Glicina/metabolismo
18.
Exp Brain Res ; 230(4): 387-94, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23494383

RESUMEN

The serotonin receptor 2C (HTR2C) gene encodes a G protein-coupled receptor that is exclusively expressed in neurons. Here, we report that the 5' untranslated region of the receptor pre-mRNA as well as its hosted miRNAs is widely expressed in non-neuronal cell lines. Alternative splicing of HTR2C is regulated by MBII-52. MBII-52 and the neighboring MBII-85 cluster are absent in people with Prader-Willi syndrome, which likely causes the disease. We show that MBII-52 and MBII-85 increase expression of the HTR2C 5' UTR and influence expression of the hosted miRNAs. The data indicate that the transcriptional unit expressing HTR2C is more complex than previously recognized and likely deregulated in Prader-Willi syndrome.


Asunto(s)
Regiones no Traducidas 5'/genética , Empalme Alternativo/fisiología , Regulación de la Expresión Génica , MicroARNs/metabolismo , Precursores del ARN/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Humanos , Ratones , MicroARNs/genética , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Precursores del ARN/genética , Receptor de Serotonina 5-HT2C/genética , Serotonina/metabolismo
19.
Nucleic Acids Res ; 39(22): 9720-30, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21880592

RESUMEN

We describe a new method that allows cloning of double-stranded RNAs (dsRNAs) that are generated in RNase protection experiments. We demonstrate that the mouse C/D box snoRNA MBII-85 (SNORD116) is processed into at least five shorter RNAs using processing sites near known functional elements of C/D box snoRNAs. Surprisingly, the majority of cloned RNAs from RNase protection experiments were derived from endogenous cellular RNA, indicating widespread antisense expression. The cloned dsRNAs could be mapped to genome areas that show RNA expression on both DNA strands and partially overlapped with experimentally determined argonaute-binding sites. The data suggest a conserved processing pattern for some C/D box snoRNAs and abundant expression of longer, non-coding RNAs in the cell that can potentially form dsRNAs.


Asunto(s)
Clonación Molecular/métodos , Procesamiento Postranscripcional del ARN , ARN sin Sentido/metabolismo , ARN Bicatenario/metabolismo , ARN Nucleolar Pequeño/metabolismo , Animales , Proteínas Argonautas/metabolismo , Sitios de Unión , Ratones , Ensayos de Protección de Nucleasas , ARN Bicatenario/genética , ARN Nucleolar Pequeño/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasas
20.
bioRxiv ; 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37786725

RESUMEN

At least 53 mutations in the microtubule associated protein tau gene (MAPT) have been identified that cause frontotemporal dementia. 47 of these mutations are localized between exons 7 and 13. They could thus affect the formation of circular RNAs (circRNAs) from the MAPT gene that occur through backsplicing from exon 12 to either exon 10 or exon 7. We analyzed representative mutants and found that five FTDP-17 mutations increase the formation of 12➔7 circRNA and three different mutations increase the amount of 12➔10 circRNA. CircRNAs are translated after undergoing adenosine to inosine RNA editing, catalyzed by ADAR enzymes. We found that the interferon induced ADAR1-p150 isoform has the strongest effect on circTau RNA translation. ADAR1-p150 activity had a stronger effect on circTau RNA expression and strongly decreased 12➔7 circRNA, but unexpectedly increased 12➔10 circRNA. In both cases, ADAR-activity strongly promoted translation of circTau RNAs. Unexpectedly, we found that the 12➔7 circTau protein interacts with eukaryotic initiation factor 4B (eIF4B), which is reduced by four FTDP-17 mutations located in the second microtubule domain. These are the first studies of the effect of human mutations on circular RNA formation and translation. They show that point mutations influence circRNA expression levels, likely through changes in the secondary pre-mRNA structures. The effect of the mutations is surpassed by editing of the circular RNAs, leading to their translation. Thus, circular RNAs and their editing status should be considered when analyzing FTDP-17 mutations. Highlights: 47/53 known FTDP-17 mutations are located in regions that could influence generation of circular RNAs from the MAPT geneCircular Tau RNAs are translated after adenosine to inosine RNA editing, most effectively caused by ADAR1-p150FTDP-17 mutations influence both circTau RNA and circTau protein expression levelsCircTau protein expression levels do not correlate with circTau RNA expression levelsCircTau proteins bind to eukaryotic initiation factor 4B, which is antagonized by FTDP-17 mutations in exon 10.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA