Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
PLoS One ; 19(6): e0298965, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38829854

RESUMEN

Familial Dysautonomia (FD) is a rare disease caused by ELP1 exon 20 skipping. Here we clarify the role of RNA Polymerase II (RNAPII) and chromatin on this splicing event. A slow RNAPII mutant and chromatin-modifying chemicals that reduce the rate of RNAPII elongation induce exon skipping whereas chemicals that create a more relaxed chromatin exon inclusion. In the brain of a mouse transgenic for the human FD-ELP1 we observed on this gene an age-dependent decrease in the RNAPII density profile that was most pronounced on the alternative exon, a robust increase in the repressive marks H3K27me3 and H3K9me3 and a decrease of H3K27Ac, together with a progressive reduction in ELP1 exon 20 inclusion level. In HEK 293T cells, selective drug-induced demethylation of H3K27 increased RNAPII elongation on ELP1 and SMN2, promoted the inclusion of the corresponding alternative exons, and, by RNA-sequencing analysis, induced changes in several alternative splicing events. These data suggest a co-transcriptional model of splicing regulation in which age-dependent changes in H3K27me3/Ac modify the rate of RNAPII elongation and affect processing of ELP1 alternative exon 20.


Asunto(s)
Empalme Alternativo , Cromatina , Disautonomía Familiar , Exones , ARN Polimerasa II , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Humanos , Exones/genética , Animales , Cromatina/metabolismo , Cromatina/genética , Ratones , Células HEK293 , Histonas/metabolismo , Ratones Transgénicos , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo , Cinética , Empalme del ARN , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
2.
Am J Hum Genet ; 109(8): 1534-1548, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35905737

RESUMEN

Familial dysautonomia (FD) is a currently untreatable, neurodegenerative disease caused by a splicing mutation (c.2204+6T>C) that causes skipping of exon 20 of the elongator complex protein 1 (ELP1) pre-mRNA. Here, we used adeno-associated virus serotype 9 (AAV9-U1-FD) to deliver an exon-specific U1 (ExSpeU1) small nuclear RNA, designed to cause inclusion of ELP1 exon 20 only in those cells expressing the target pre-mRNA, in a phenotypic mouse model of FD. Postnatal systemic and intracerebral ventricular treatment in these mice increased the inclusion of ELP1 exon 20. This also augmented the production of functional protein in several tissues including brain, dorsal root, and trigeminal ganglia. Crucially, the treatment rescued most of the FD mouse mortality before one month of age (89% vs 52%). There were notable improvements in ataxic gait as well as renal (serum creatinine) and cardiac (ejection fraction) functions. RNA-seq analyses of dorsal root ganglia from treated mice and human cells overexpressing FD-ExSpeU1 revealed only minimal global changes in gene expression and splicing. Overall then, our data prove that AAV9-U1-FD is highly specific and will likely be a safe and effective therapeutic strategy for this debilitating disease.


Asunto(s)
Disautonomía Familiar , Enfermedades Neurodegenerativas , Animales , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Exones/genética , Humanos , Ratones , Enfermedades Neurodegenerativas/genética , Precursores del ARN/genética , Empalme del ARN/genética , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo
3.
Mol Med ; 27(1): 157, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34906067

RESUMEN

BACKGROUND: Aberrant splicing is a common outcome in the presence of exonic or intronic variants that might hamper the intricate network of interactions defining an exon in a specific gene context. Therefore, the evaluation of the functional, and potentially pathological, role of nucleotide changes remains one of the major challenges in the modern genomic era. This aspect has also to be taken into account during the pre-clinical evaluation of innovative therapeutic approaches in animal models of human diseases. This is of particular relevance when developing therapeutics acting on splicing, an intriguing and expanding research area for several disorders. Here, we addressed species-specific splicing mechanisms triggered by the OTC c.386G>A mutation, relatively frequent in humans, leading to Ornithine TransCarbamylase Deficiency (OTCD) in patients and spfash mice, and its differential susceptibility to RNA therapeutics based on engineered U1snRNA. METHODS: Creation and co-expression of engineered U1snRNAs with human and mouse minigenes, either wild-type or harbouring different nucleotide changes, in human (HepG2) and mouse (Hepa1-6) hepatoma cells followed by analysis of splicing pattern. RNA pulldown studies to evaluate binding of specific splicing factors. RESULTS: Comparative nucleotide analysis suggested a role for the intronic +10-11 nucleotides, and pull-down assays showed that they confer preferential binding to the TIA1 splicing factor in the mouse context, where TIA1 overexpression further increases correct splicing. Consistently, the splicing profile of the human minigene with mouse +10-11 nucleotides overlapped that of mouse minigene, and restored responsiveness to TIA1 overexpression and to compensatory U1snRNA. Swapping the human +10-11 nucleotides into the mouse context had opposite effects. Moreover, the interplay between the authentic and the adjacent cryptic 5'ss in the human OTC dictates pathogenic mechanisms of several OTCD-causing 5'ss mutations, and only the c.386+5G>A change, abrogating the cryptic 5'ss, was rescuable by engineered U1snRNA. CONCLUSIONS: Subtle intronic variations explain species-specific OTC splicing patterns driven by the c.386G>A mutation, and the responsiveness to engineered U1snRNAs, which suggests careful elucidation of molecular mechanisms before proposing translation of tailored therapeutics from animal models to humans.


Asunto(s)
Ornitina Carbamoiltransferasa/genética , Empalme del ARN , Animales , Línea Celular Tumoral , Humanos , Intrones , Ratones , Mutación , ARN/uso terapéutico , Ribonucleoproteína Nuclear Pequeña U1/genética
4.
Hum Mutat ; 41(12): 2143-2154, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32935393

RESUMEN

In cystic fibrosis (CF), the correction of splicing defects represents an interesting therapeutic approach to restore normal CFTR function. In this study, we focused on 10 common mutations/variants 711+3A>G/C, 711+5G>A, TG13T3, TG13T5, TG12T5, 1863C>T, 1898+3A>G, 2789+5G>A, and 3120G>A that induce skipping of the corresponding CFTR exons 5, 10, 13, 16, and 18. To rescue the splicing defects we tested, in a minigene assay, a panel of modified U1 small nuclear RNAs (snRNAs), named Exon Specific U1s (ExSpeU1s), that was engineered to bind to intronic sequences downstream of each defective exon. Using this approach, we show that all 10 splicing mutations analyzed are efficiently corrected by specific ExSpeU1s. Using complementary DNA-splicing competent minigenes, we also show that the ExspeU1-mediated splicing correction at the RNA level recovered the full-length CFTR protein for 1863C>T, 1898+3A>G, 2789+5G>A variants. In addition, detailed mutagenesis experiments performed on exon 13 led us to identify a novel intronic regulatory element involved in the ExSpeU1-mediated splicing rescue. These results provide a common strategy based on modified U1 snRNAs to correct exon skipping in a group of disease-causing CFTR mutations.


Asunto(s)
Fibrosis Quística/genética , Exones/genética , Mutación/genética , ARN Nuclear Pequeño/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células HeLa , Humanos , Intrones/genética , Conformación de Ácido Nucleico , Empalme del ARN/genética , ARN Nuclear Pequeño/química
5.
Int J Mol Sci ; 21(6)2020 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-32244944

RESUMEN

The elucidation of aberrant splicing mechanisms, frequently associated with disease has led to the development of RNA therapeutics based on the U1snRNA, which is involved in 5' splice site (5'ss) recognition. Studies in cellular models have demonstrated that engineered U1snRNAs can rescue different splicing mutation types. However, the assessment of their correction potential in vivo is limited by the scarcity of animal models with the targetable splicing defects. Here, we challenged the U1snRNA in the FAH5961SB mouse model of hepatic fumarylacetoacetate hydrolase (FAH) deficiency (Hereditary Tyrosinemia type I, HT1) due to the FAH c.706G>A splicing mutation. Through minigene expression studies we selected a compensatory U1snRNA (U1F) that was able to rescue this mutation. Intriguingly, adeno-associated virus-mediated delivery of U1F (AAV8-U1F), but not of U1wt, partially rescued FAH splicing in mouse hepatocytes. Consistently, FAH protein was detectable only in the liver of AAV8-U1F treated mice, which displayed a slightly prolonged survival. Moreover, RNA sequencing revealed the negligible impact of the U1F on the splicing profile and overall gene expression, thus pointing toward gene specificity. These data provide early in vivo proof-of-principle of the correction potential of compensatory U1snRNAs in HTI and encourage further optimization on a therapeutic perspective, and translation to other splicing-defective forms of metabolic diseases.


Asunto(s)
Hidrolasas/genética , Empalme del ARN/genética , ARN Nuclear Pequeño/genética , Tirosinemias/enzimología , Tirosinemias/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
6.
Nucleic Acids Res ; 47(14): 7618-7632, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31127278

RESUMEN

Spinal Muscular Atrophy results from loss-of-function mutations in SMN1 but correcting aberrant splicing of SMN2 offers hope of a cure. However, current splice therapy requires repeated infusions and is expensive. We previously rescued SMA mice by promoting the inclusion of a defective exon in SMN2 with germline expression of Exon-Specific U1 snRNAs (ExspeU1). Here we tested viral delivery of SMN2 ExspeU1s encoded by adeno-associated virus AAV9. Strikingly the virus increased SMN2 exon 7 inclusion and SMN protein levels and rescued the phenotype of mild and severe SMA mice. In the severe mouse, the treatment improved the neuromuscular function and increased the life span from 10 to 219 days. ExspeU1 expression persisted for 1 month and was effective at around one five-hundredth of the concentration of the endogenous U1snRNA. RNA-seq analysis revealed our potential drug rescues aberrant SMA expression and splicing profiles, which are mostly related to DNA damage, cell-cycle control and acute phase response. Vastly overexpressing ExspeU1 more than 100-fold above the therapeutic level in human cells did not significantly alter global gene expression or splicing. These results indicate that AAV-mediated delivery of a modified U1snRNP particle may be a novel therapeutic option against SMA.


Asunto(s)
Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , Distrofia Muscular Animal/terapia , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Exones/genética , Células HEK293 , Humanos , Ratones Noqueados , Atrofia Muscular Espinal/genética , Distrofia Muscular Animal/genética , Mutación , Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U1/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
7.
RNA ; 24(9): 1158-1171, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29895677

RESUMEN

MicroRNAs are found throughout the genome and are processed by the microprocessor complex (MPC) from longer precursors. Some precursor miRNAs overlap intron:exon junctions. These splice site overlapping microRNAs (SO-miRNAs) are mostly located in coding genes. It has been intimated, in the rarer examples of SO-miRNAs in noncoding RNAs, that the competition between the spliceosome and the MPC modulates alternative splicing. However, the effect of this overlap on coding transcripts is unknown. Unexpectedly, we show that neither Drosha silencing nor SF3b1 silencing changed the inclusion ratio of SO-miRNA exons. Two SO-miRNAs, located in genes that code for basal membrane proteins, are known to inhibit proliferation in primary keratinocytes. These SO-miRNAs were up-regulated during differentiation and the host mRNAs were down-regulated, but again there was no change in inclusion ratio of the SO-miRNA exons. Interestingly, Drosha silencing increased nascent RNA density, on chromatin, downstream from SO-miRNA exons. Overall our data suggest a novel mechanism for regulating gene expression in which MPC-dependent cleavage of SO-miRNA exons could cause premature transcriptional termination of coding genes rather than affecting alternative splicing.


Asunto(s)
Queratinocitos/citología , MicroARNs/genética , Fosfoproteínas/genética , Sitios de Empalme de ARN , Factores de Empalme de ARN/genética , Ribonucleasa III/genética , Empalme Alternativo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Cromatina/genética , Regulación hacia Abajo , Exones , Silenciador del Gen , Humanos , Queratinocitos/metabolismo , Empalmosomas/metabolismo , Regulación hacia Arriba
8.
Hum Mol Genet ; 27(14): 2466-2476, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29701768

RESUMEN

Familial dysautonomia (FD) is a rare genetic disease with no treatment, caused by an intronic point mutation (c.2204+6T>C) that negatively affects the definition of exon 20 in the elongator complex protein 1 gene (ELP1 also known as IKBKAP). This substitution modifies the 5' splice site and, in combination with regulatory splicing factors, induces different levels of exon 20 skipping, in various tissues. Here, we evaluated the therapeutic potential of a novel class of U1 snRNA molecules, exon-specific U1s (ExSpeU1s), in correcting ELP1 exon 20 recognition. Lentivirus-mediated expression of ELP1-ExSpeU1 in FD fibroblasts improved ELP1 splicing and protein levels. We next focused on a transgenic mouse model that recapitulates the same tissue-specific mis-splicing seen in FD patients. Intraperitoneal delivery of ELP1-ExSpeU1s-adeno-associated virus particles successfully increased the production of full-length human ELP1 transcript and protein. This splice-switching class of molecules is the first to specifically correct the ELP1 exon 20 splicing defect. Our data provide proof of principle of ExSpeU1s-adeno-associated virus particles as a novel therapeutic strategy for FD.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/terapia , Terapia Genética , ARN Nuclear Pequeño/genética , Empalme Alternativo/genética , Animales , Proteínas Portadoras/uso terapéutico , Dependovirus/genética , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Disautonomía Familiar/fisiopatología , Exones/genética , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Intrones/genética , Ratones , Ratones Transgénicos , Empalme del ARN/genética , ARN Nuclear Pequeño/uso terapéutico , Factores de Elongación Transcripcional
9.
Exp Dermatol ; 27(6): 656-662, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29512856

RESUMEN

In our recent cDNA microarray experiment, three SR-rich splicing factors-SFRS18, PPIG and LUC7L3-were shown to exert altered responsiveness upon T-lymphokine stimulation of psoriatic non-involved and healthy epidermis samples. We have also demonstrated that double silencing LUC7L3 and SFRS18 efficiently decreased production of the psoriasis-associated EDA+ fibronectin isoform. These findings prompted the further investigation of signalling pathways affected by LUC7L3 and SFRS18. To detect gene expression and splicing pattern alterations upon double silencing of LUC7L3 and SFRS18 in an HPV-immortalised keratinocyte cell culture, paired-end RNA sequencing was carried out. Marked changes in exon usage were revealed, in contrast to the modest alterations detected in gene expression, providing a closer delineation of the potential targets of the examined splicing factors. The most prominent gene expression change was detected for IFI6, an interferon-inducible gene highly expressed in psoriasis. Interacting partners of IFI6 and certain psoriasis-associated transcripts also exhibited significantly increased expression upon silencing. In addition to elevated abundance of the EDA+ fibronectin interactor ITGA5, we confirmed decreased EDA domain inclusion, which agrees well with our prior experimental data. Furthermore, differential exon usage was established for the transcription element CREB1, along with HERC6 and CUL1, which are implicated in ubiquitination. Although immortalised keratinocytes express low levels of TINCR, a long non-coding RNA involved in terminal differentiation of keratinocytes, splicing alterations were successfully demonstrated for this RNA as well. We believe that the targeted investigation of mRNA maturation disturbances may help us gain deeper insight into the molecular pathogenesis of psoriasis.


Asunto(s)
Exones/genética , Proteínas Mitocondriales/genética , Psoriasis/genética , Proteínas de Unión al ARN/genética , Transducción de Señal/genética , Línea Celular , Proteínas Cullin/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Ectodisplasinas/genética , Expresión Génica , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Integrinas , Queratinocitos , Proteínas Nucleares , ARN Largo no Codificante/genética , Análisis de Secuencia de ARN , Factores de Empalme Serina-Arginina , Ubiquitina-Proteína Ligasas/genética
10.
Mol Ther Nucleic Acids ; 5(10): e370, 2016 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-27701399

RESUMEN

In cellular models we have demonstrated that a unique U1snRNA targeting an intronic region downstream of a defective exon (Exon-specific U1snRNA, ExSpeU1) can rescue multiple exon-skipping mutations, a relevant cause of genetic disease. Here, we explored in mice the ExSpeU1 U1fix9 toward two model Hemophilia B-causing mutations at the 5' (c.519A > G) or 3' (c.392-8T > G) splice sites of F9 exon 5. Hydrodynamic injection of wt-BALB/C mice with plasmids expressing the wt and mutant (hFIX-2G5'ss and hFIX-8G3'ss) splicing-competent human factor IX (hFIX) cassettes resulted in the expression of hFIX transcripts lacking exon 5 in liver, and in low plasma levels of inactive hFIX. Coinjection of U1fix9, but not of U1wt, restored exon inclusion of variants and in the intrinsically weak FIXwt context. This resulted in appreciable circulating hFIX levels (mean ± SD; hFIX-2G5'ss, 1.0 ± 0.5 µg/ml; hFIX-8G3'ss, 1.2 ± 0.3 µg/ml; and hFIXwt, 1.9 ± 0.6 µg/ml), leading to a striking shortening (from ~100 seconds of untreated mice to ~80 seconds) of FIX-dependent coagulation times, indicating a hFIX with normal specific activity. This is the first proof-of-concept in vivo that a unique ExSpeU1 can efficiently rescue gene expression impaired by distinct exon-skipping variants, which extends the applicability of ExSpeU1s to panels of mutations and thus cohort of patients.

11.
PLoS Genet ; 12(5): e1006082, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27227676

RESUMEN

Mutations that result in amino acid changes can affect both pre-mRNA splicing and protein function. Understanding the combined effect is essential for correct diagnosis and for establishing the most appropriate therapeutic strategy at the molecular level. We have identified a series of disease-causing splicing mutations in coagulation factor IX (FIX) exon 5 that are completely recovered by a modified U1snRNP particle, through an SRSF2-dependent enhancement mechanism. We discovered that synonymous mutations and missense substitutions associated to a partial FIX secretion defect represent targets for this therapy as the resulting spliced-corrected proteins maintains normal FIX coagulant specific activity. Thus, splicing and protein alterations contribute to define at the molecular level the disease-causing effect of a number of exonic mutations in coagulation FIX exon 5. In addition, our results have a significant impact in the development of splicing-switching therapies in particular for mutations that affect both splicing and protein function where increasing the amount of a correctly spliced protein can circumvent the basic functional defects.


Asunto(s)
Trastornos de la Coagulación Sanguínea/genética , Factor IX/genética , Empalme del ARN/genética , Trastornos de la Coagulación Sanguínea/patología , Técnicas de Cultivo de Célula , Exones/genética , Vectores Genéticos , Humanos , Mutación , Precursores del ARN/genética , Sitios de Empalme de ARN/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Factores de Empalme Serina-Arginina/genética , Transfección
12.
Nat Commun ; 7: 11168, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-27041075

RESUMEN

Modified U1 snRNAs bound to intronic sequences downstream of the 5' splice site correct exon skipping caused by different types of mutations. Here we evaluate the therapeutic activity and structural requirements of these exon-specific U1 snRNA (ExSpeU1) particles. In a severe spinal muscular atrophy, mouse model, ExSpeU1, introduced by germline transgenesis, increases SMN2 exon 7 inclusion, SMN protein production and extends life span. In vitro, RNA mutant analysis and silencing experiments show that while U1A protein is dispensable, the 70K and stem loop IV elements mediate most of the splicing rescue activity through improvement of exon and intron definition. Our findings indicate that precise engineering of the U1 core spliceosomal RNA particle has therapeutic potential in pathologies associated with exon-skipping mutations.


Asunto(s)
Atrofia Muscular Espinal/genética , Empalme del ARN , ARN Nuclear Pequeño/fisiología , Animales , Animales Modificados Genéticamente , Terapia Genética , Ratones , Atrofia Muscular Espinal/patología , Conformación de Ácido Nucleico , Fenotipo , Sitios de Empalme de ARN , ARN Nuclear Pequeño/química , Empalmosomas/química , Empalmosomas/genética , Empalmosomas/fisiología
13.
Sci Rep ; 5: 11746, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26123042

RESUMEN

Spinal muscular atrophy (SMA) is a primary genetic cause of infant mortality due to mutations in the Survival Motor Neuron (SMN) 1 gene. No cure is available. Antisense oligonucleotides (ASOs) aimed at increasing SMN levels from the paralogous SMN2 gene represent a possible therapeutic strategy. Here, we tested in SMA human induced pluripotent stem cells (iPSCs) and iPSC-differentiated motor neurons, three different RNA approaches based on morpholino antisense targeting of the ISSN-1, exon-specific U1 small nuclear RNA (ExSpeU1), and Transcription Activator-Like Effector-Transcription Factor (TALE-TF). All strategies act modulating SMN2 RNA: ASO affects exon 7 splicing, TALE-TF increase SMN2 RNA acting on the promoter, while ExSpeU1 improves pre-mRNA processing. These approaches induced up-regulation of full-length SMN mRNA and differentially affected the Delta-7 isoform: ASO reduced this isoform, while ExSpeU1 and TALE-TF increased it. All approaches upregulate the SMN protein and significantly improve the in vitro SMA motor neurons survival. Thus, these findings demonstrate that therapeutic tools that act on SMN2 RNA are able to rescue the SMA disease phenotype. Our data confirm the feasibility of SMA iPSCs as in vitro disease models and we propose novel RNA approaches as potential therapeutic strategies for treating SMA and other genetic neurological disorders.


Asunto(s)
Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/terapia , Secuencia de Bases , Supervivencia Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Terapia Genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Morfolinos/genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Fenotipo , Regiones Promotoras Genéticas , ARN Nuclear Pequeño/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Factores de Transcripción/genética
14.
J Physiol ; 593(17): 3829-48, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26108457

RESUMEN

TMEM16F is a membrane protein with possible dual function as an ion channel and a phospholipid scramblase. The properties of ion channels associated with TMEM16F and the link between ion channel and scramblase activity are a matter of debate. We studied the properties of four isoforms of TMEM16F generated by alternative splicing. Upregulation of three TMEM16F isoforms or silencing of endogenous TMEM16F increased and decreased, respectively, both scramblase and channel activities. Introduction of an activating mutation in TMEM16F sequence caused a marked increase in phosphatidylserine scrambling and in ion transport indicating direct involvement of the protein in both functions. TMEM16F, also known as ANO6, is a membrane protein that has been associated with phospholipid scramblase and ion channel activity. However, the characteristics of TMEM16F-dependent channels, particularly the ion selectivity, are a matter of debate. Furthermore, the direct involvement of TMEM16F in phospholipid scrambling has been questioned. We studied the properties of different TMEM16F variants generated by alternative splicing. Using whole-cell patch-clamp recordings, we found that V1, V2 and V5 variants generated membrane currents activated by very high (micromolar) intracellular Ca(2+) concentrations and positive membrane potentials. These variants showed different degrees of Ca(2+) sensitivity and kinetics of activation but similar ion permeability, characterized by a slight selectivity for Cl(-) over Na(+) . A fourth variant (V3) showing a unique carboxy-terminus was devoid of activity, in agreement with its intracellular localization. We also measured scramblase activity using the binding of annexin V to detect phosphatidylserine on the cell surface. V1, V2 and V5 variants were associated with calcium-dependent phosphatidylserine externalization. Interestingly, introduction of an activating mutation, D409G, produced a marked increase in the apparent Ca(2+) sensitivity of TMEM16F-dependent channels. In parallel, this mutation also enhanced the extent of phosphatidylserine externalization that occurred even under resting conditions. These results support the conclusion that TMEM16F proteins are directly involved in dual activity, as a phospholipid scramblase and as an ion channel.


Asunto(s)
Canales Iónicos , Proteínas de Transferencia de Fosfolípidos , Anoctaminas , Calcio/metabolismo , Línea Celular , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Proteínas de Transferencia de Fosfolípidos/genética , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteínas de Transferencia de Fosfolípidos/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , ARN Mensajero/metabolismo
15.
Hum Mutat ; 36(5): 504-12, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25665175

RESUMEN

The c.891C>T synonymous transition in SPINK5 induces exon 11 (E11) skipping and causes Netherton syndrome (NS). Using a specific RNA-protein interaction assay followed by mass spectrometry analysis along with silencing and overexpression of splicing factors, we showed that this mutation affects an exonic bifunctional splicing regulatory element composed by two partially overlapping silencer and enhancer sequences, recognized by hnRNPA1 and Tra2ß splicing factors, respectively. The C-to-T substitution concomitantly increases hnRNPA1 and weakens Tra2ß-binding sites, leading to pathological E11 skipping. In hybrid minigenes, exon-specific U1 small nuclear RNAs (ExSpe U1s) that target by complementarity intronic sequences downstream of the donor splice site rescued the E11 skipping defect caused by the c.891C>T mutation. ExSpe U1 lentiviral-mediated transduction of primary NS keratinocytes from a patient bearing the mutation recovered the correct full-length SPINK5 mRNA and the corresponding functional lympho-epithelial Kazal-type related inhibitor protein in a dose-dependent manner. This study documents the reliability of a mutation-specific, ExSpe U1-based, splicing therapy for a relatively large subset of European NS patients. Usage of ExSpe U1 may represent a general approach for correction of splicing defects affecting skin disease genes.


Asunto(s)
Empalme Alternativo , Exones , Mutación , Proteínas Inhibidoras de Proteinasas Secretoras/genética , ARN Nuclear Pequeño/genética , Secuencias Reguladoras de Ácidos Nucleicos , Línea Celular , Expresión Génica , Silenciador del Gen , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Queratinocitos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Síndrome de Netherton/genética , Síndrome de Netherton/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Inhibidoras de Proteinasas Secretoras/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Inhibidor de Serinpeptidasas Tipo Kazal-5 , Factores de Empalme Serina-Arginina
16.
Am J Hum Genet ; 96(1): 93-103, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25557785

RESUMEN

Exon-specific U1 snRNAs (ExSpe U1s) are modified U1 snRNAs that interact with intronic sequences downstream of the 5' splice site (ss) by complementarity. This process restores exon skipping caused by different types of mutation. We have investigated the molecular mechanism and activity of these molecules in spinal muscular atrophy (SMA), a genetic neuromuscular disease where a silent exonic transition on the survival motor neuron 2 (SMN2) leads to exon 7 (E7) skipping. By using different cellular models, we show that a single chromosome-integrated copy of ExSpe U1 induced a significant correction of endogenous SMN2 E7 splicing and resulted in the restoration of the corresponding SMN protein levels. Interestingly, the analysis of pre-mRNA transcript abundance and decay showed that ExSpe U1s promote E7 inclusion and stabilizes the SMN pre-mRNA intermediate. This selective effect on pre-mRNA stability resulted in higher levels of SMN mRNAs in comparison with those after treatment with an antisense oligonucleotide (AON) that targets corresponding intronic sequences. In mice harboring the SMN2 transgene, AAV-mediated delivery of ExSpe U1 increased E7 inclusion in brain, heart, liver, kidney, and skeletal muscle. The positive effect of ExSpe U1s on SMN pre-mRNA processing highlights their therapeutic potential in SMA and in other pathologies caused by exon-skipping mutations.


Asunto(s)
Precursores del ARN/genética , Sitios de Empalme de ARN , Empalme del ARN , ARN Nuclear Pequeño/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Modelos Animales de Enfermedad , Exones , Femenino , Sitios Genéticos , Células HEK293 , Humanos , Intrones , Lentivirus/genética , Masculino , Ratones , Ratones Transgénicos , Atrofia Muscular Espinal/genética , Conformación de Ácido Nucleico , ARN Nuclear Pequeño/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Transducción Genética
17.
Hepatology ; 61(4): 1382-91, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25421123

RESUMEN

UNLABELLED: ATP8B1 deficiency is a severe autosomal recessive liver disease resulting from mutations in the ATP8B1 gene characterized by a continuous phenotypical spectrum from intermittent (benign recurrent intrahepatic cholestasis; BRIC) to progressive familial intrahepatic cholestasis (PFIC). Current therapeutic options are insufficient, and elucidating the molecular consequences of mutations could lead to personalized mutation-specific therapies. We investigated the effect on pre-messenger RNA splicing of 14 ATP8B1 mutations at exon-intron boundaries using an in vitro minigene system. Eleven mutations, mostly associated with a PFIC phenotype, resulted in aberrant splicing and a complete absence of correctly spliced product. In contrast, three mutations led to partially correct splicing and were associated with a BRIC phenotype. These findings indicate an inverse correlation between the level of correctly spliced product and disease severity. Expression of modified U1 small nuclear RNAs (snRNA) complementary to the splice donor sites strongly improved or completely rescued splicing for several ATP8B1 mutations located at donor, as well as acceptor, splice sites. In one case, we also evaluated exon-specific U1 snRNAs that, by targeting nonconserved intronic sequences, might reduce possible off-target events. Although very effective in correcting exon skipping, they also induced retention of the short downstream intron. CONCLUSION: We systematically characterized the molecular consequences of 14 ATP8B1 mutations at exon-intron boundaries associated with ATP8B1 deficiency and found that the majority resulted in total exon skipping. The amount of correctly spliced product inversely correlated with disease severity. Compensatory modified U1 snRNAs, complementary to mutated donor splice sites, were able to improve exon definition very efficiently and could be a novel therapeutic strategy in ATP8B1 deficiency as well as other genetic diseases.


Asunto(s)
Adenosina Trifosfatasas/genética , Mutación , Empalme del ARN/genética , ARN Nuclear Pequeño/fisiología , Células Cultivadas , Humanos
18.
Wiley Interdiscip Rev RNA ; 5(5): 647-58, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24788135

RESUMEN

The spliceosome and the microprocessor complex (MPC) are two important processing machineries that act on precursor (pre)-mRNA. Both cleave the pre-mRNA to generate spliced mature transcripts and microRNAs (miRNAs), respectively. While spliceosomes identify in a complex manner correct splice sites, MPCs typically target RNA hairpins (pri-miRNA hairpins). In addition, pre-mRNA transcripts can contain pri-miRNA-like hairpins that are cleaved by the MPC without generating miRNAs. Recent evidence indicates that the position of hairpins on pre-mRNA, their distance from splice sites, and the relative efficiency of cropping and splicing contribute to determine the fate of a pre-mRNA. Depending on these factors, a pre-mRNA can be preferentially used to generate a miRNA, a constitutively or even an alternative spliced transcript. For example, competition between splicing and cropping on splice-site-overlapping miRNAs (SO miRNAs) results in alternative spliced isoforms and influences miRNA biogenesis. In several cases, the outcome of a pre-mRNA transcript and its final handling as miRNA or mRNA substrate can be frequently closely connected to the functional relationships between diverse pre-mRNA processing events. These events are influenced by both gene context and physiopathological conditions.


Asunto(s)
MicroARNs/genética , Precursores del ARN/genética , Empalme del ARN/genética , ARN Mensajero/genética , Empalmosomas/genética , Empalme Alternativo/genética , Humanos , Secuencias Invertidas Repetidas/genética , MicroARNs/biosíntesis , MicroARNs/metabolismo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/biosíntesis , Empalmosomas/metabolismo
19.
Biochim Biophys Acta ; 1842(7): 1052-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24704046

RESUMEN

The pathological role of mutations that affect not conserved splicing regulatory sequences can be difficult to determine. In a patient with Fanconi anemia, we identified two unpredictable splicing mutations that act on either sides of FANCA exon 8. In patients-derived cells and in minigene splicing assay, we showed that both an apparently benign intronic c.710-5T>C transition and the nonsense c.790C>T substitution induce almost complete exon 8 skipping. Site-directed mutagenesis experiments indicated that the c.710-5T>C transition affects a polypyrimidine tract where most of the thymidines cannot be compensated by cytidines. The c.790C>T mutation located in position -3 relative to the donor site induce exon 8 skipping in an NMD-independent manner and complementation experiments with modified U1 snRNAs showed that U1 snRNP is only partially involved in the splicing defect. Our results highlight the importance of performing splicing functional assay for correct identification of disease-causing mechanism of genomic variants and provide mechanistic insights on how these two FANCA mutations affect exon 8 definition.


Asunto(s)
Codón sin Sentido , Exones , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Sitios de Empalme de ARN/genética , Empalme del ARN , Secuencia de Bases , Línea Celular Tumoral , Células HeLa , Humanos , Intrones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida/métodos , Ribonucleoproteínas Nucleares Pequeñas
20.
Nucleic Acids Res ; 41(18): 8680-91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23863840

RESUMEN

We have explored the functional relationships between spliceosome and Microprocessor complex activities in a novel class of microRNAs (miRNAs), named Splice site Overlapping (SO) miRNAs, whose pri-miRNA hairpins overlap splice sites. We focused on the evolutionarily conserved SO miR-34b, and we identified two indispensable elements for recognition of its 3' splice site: a branch point located in the hairpin and a downstream purine-rich exonic splicing enhancer. In minigene systems, splicing inhibition owing to exonic splicing enhancer deletion or AG 3'ss mutation increases miR-34b levels. Moreover, small interfering-mediated silencing of Drosha and/or DGCR8 improves splicing efficiency and abolishes miR-34b production. Thus, the processing of this 3' SO miRNA is regulated in an antagonistic manner by the Microprocessor and the spliceosome owing to competition between these two machineries for the nascent transcript. We propose that this novel mechanism is commonly used to regulate the relative amount of SO miRNA and messenger RNA produced from primary transcripts.


Asunto(s)
Empalme Alternativo , MicroARNs/biosíntesis , MicroARNs/química , Sitios de Empalme de ARN , Animales , Secuencia de Bases , Células HeLa , Humanos , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Proteínas/antagonistas & inhibidores , Proteínas/genética , Purinas/análisis , Interferencia de ARN , Proteínas de Unión al ARN , Secuencias Reguladoras de Ácido Ribonucleico , Ribonucleasa III/antagonistas & inhibidores , Ribonucleasa III/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA