Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Hum Mol Genet ; 22(4): 704-16, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23139243

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by the expansion of CTG repeats in the 3' untranslated region of the DMPK gene. Several missplicing events and transcriptional alterations have been described in DM1 patients. A large number of these defects have been reproduced in animal models expressing CTG repeats alone. Recent studies have also reported miRNA dysregulation in DM1 patients. In this work, a Drosophila model was used to investigate miRNA transcriptome alterations in the muscle, specifically triggered by CTG expansions. Twenty miRNAs were differentially expressed in CTG-expressing flies. Of these, 19 were down-regulated, whereas 1 was up-regulated. This trend was confirmed for those miRNAs conserved between Drosophila and humans (miR-1, miR-7 and miR-10) in muscle biopsies from DM1 patients. Consistently, at least seven target transcripts of these miRNAs were up-regulated in DM1 skeletal muscles. The mechanisms involved in dysregulation of miR-7 included a reduction of its primary precursor both in CTG-expressing flies and in DM1 patients. Additionally, a regulatory role for Muscleblind (Mbl) was also suggested for miR-1 and miR-7, as these miRNAs were down-regulated in flies where Mbl had been silenced. Finally, the physiological relevance of miRNA dysregulation was demonstrated for miR-10, since over-expression of this miRNA in Drosophila extended the lifespan of CTG-expressing flies. Taken together, our results contribute to our understanding of the origin and the role of miRNA alterations in DM1.


Asunto(s)
MicroARNs/genética , Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido , Animales , Secuencia de Bases , Células Cultivadas , Regulación hacia Abajo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Esperanza de Vida , Masculino , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcriptoma
2.
Nature ; 457(7227): 322-6, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18971929

RESUMEN

The nephron is the basic structural and functional unit of the vertebrate kidney. It is composed of a glomerulus, the site of ultrafiltration, and a renal tubule, along which the filtrate is modified. Although widely regarded as a vertebrate adaptation, 'nephron-like' features can be found in the excretory systems of many invertebrates, raising the possibility that components of the vertebrate excretory system were inherited from their invertebrate ancestors. Here we show that the insect nephrocyte has remarkable anatomical, molecular and functional similarity to the glomerular podocyte, a cell in the vertebrate kidney that forms the main size-selective barrier as blood is ultrafiltered to make urine. In particular, both cell types possess a specialized filtration diaphragm, known as the slit diaphragm in podocytes or the nephrocyte diaphragm in nephrocytes. We find that fly (Drosophila melanogaster) orthologues of the major constituents of the slit diaphragm, including nephrin, NEPH1 (also known as KIRREL), CD2AP, ZO-1 (TJP1) and podocin, are expressed in the nephrocyte and form a complex of interacting proteins that closely mirrors the vertebrate slit diaphragm complex. Furthermore, we find that the nephrocyte diaphragm is completely lost in flies lacking the orthologues of nephrin or NEPH1-a phenotype resembling loss of the slit diaphragm in the absence of either nephrin (as in human congenital nephrotic syndrome of the Finnish type, NPHS1) or NEPH1. These changes markedly impair filtration function in the nephrocyte. The similarities we describe between invertebrate nephrocytes and vertebrate podocytes provide evidence suggesting that the two cell types are evolutionarily related, and establish the nephrocyte as a simple model in which to study podocyte biology and podocyte-associated diseases.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Podocitos/citología , Podocitos/fisiología , Animales , Línea Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/fisiología , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Podocitos/metabolismo
3.
Proc Natl Acad Sci U S A ; 108(29): 11866-71, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730182

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by the expansion of noncoding CTG repeats in the dystrophia myotonica-protein kinase gene. Mutant transcripts form CUG hairpins that sequester RNA-binding factors into nuclear foci, including Muscleblind-like-1 protein (MBNL1), which regulate alternative splicing and gene expression. To identify molecules that target toxic CUG transcripts in vivo, we performed a positional scanning combinatorial peptide library screen using a Drosophila model of DM1. The screen identified a D-amino acid hexapeptide (ABP1) that reduced CUG foci formation and suppressed CUG-induced lethality and muscle degeneration when administered orally. Transgenic expression of natural, L-amino acid ABP1 analogues reduced CUG-induced toxicity in fly eyes and muscles. Furthermore, ABP1 reversed muscle histopathology and splicing misregulation of MBNL1 targets in DM1 model mice. In vitro, ABP1 bound to CUG hairpins and induced a switch to a single-stranded conformation. Our findings demonstrate that ABP1 shows antimyotonic dystrophy activity by targeting the core of CUG toxicity.


Asunto(s)
Distrofia Miotónica/genética , Oligopéptidos/metabolismo , Conformación Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido/genética , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila , Descubrimiento de Drogas , Ratones , Músculos/metabolismo , Proteína Quinasa de Distrofia Miotónica , Oligopéptidos/genética , Biblioteca de Péptidos , Proteínas de Unión al ARN/genética
4.
Nat Commun ; 9(1): 2032, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29795225

RESUMEN

Modification of SMN2 exon 7 (E7) splicing is a validated therapeutic strategy against spinal muscular atrophy (SMA). However, a target-based approach to identify small-molecule E7 splicing modifiers has not been attempted, which could reveal novel therapies with improved mechanistic insight. Here, we chose as a target the stem-loop RNA structure TSL2, which overlaps with the 5' splicing site of E7. A small-molecule TSL2-binding compound, homocarbonyltopsentin (PK4C9), was identified that increases E7 splicing to therapeutic levels and rescues downstream molecular alterations in SMA cells. High-resolution NMR combined with molecular modelling revealed that PK4C9 binds to pentaloop conformations of TSL2 and promotes a shift to triloop conformations that display enhanced E7 splicing. Collectively, our study validates TSL2 as a target for small-molecule drug discovery in SMA, identifies a novel mechanism of action for an E7 splicing modifier, and sets a precedent for other splicing-mediated diseases where RNA structure could be similarly targeted.


Asunto(s)
Imidazoles/farmacología , Indoles/farmacología , Atrofia Muscular Espinal/tratamiento farmacológico , ARN Mensajero/metabolismo , Empalme Alternativo , Animales , Animales Modificados Genéticamente , Drosophila , Evaluación Preclínica de Medicamentos , Exones/genética , Células HeLa , Humanos , Imidazoles/química , Imidazoles/uso terapéutico , Indoles/química , Indoles/uso terapéutico , Terapia Molecular Dirigida/métodos , Atrofia Muscular Espinal/genética , Fenotipo , Sitios de Empalme de ARN , ARN Mensajero/química , ARN Mensajero/genética , Elementos Reguladores de la Transcripción/efectos de los fármacos , Proteína 2 para la Supervivencia de la Neurona Motora/genética
5.
Hereditas ; 143(2006): 117-22, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17362344

RESUMEN

Motile mesodermal cells contribute several cell types to developing embryos. In Drosophila, blood cell precursors or prohemocytes, are first detected in the procephalic mesoderm by the expression of the GATA transcription factor Serpent. Once specified, a subset of prohemocytes migrate posteriorly to populate most of the embryo and further differentiate as plasmatocytes. Similarly, Drosophila nephrogenesis involves integration of posterior mesodermal cells into the Malpighian tubule primordia where these cells differentiate as stellate cells. Here we investigated the possibility that the immunoglobulin-domain protein Hibris and the GATA factor Serpent were co-expressed in motile mesodermal cells by using the hibris expression reporter P[w(+)]36.1 and antibody staining. We show that P[w(+)]36.1 reproduces the endogenous expression of hibris in several embryonic tissue types and organs, including mesectoderm, early mesoderm, pharyngeal musculature, hindgut, anal plates, posterior spiracles, and antennomaxillary complex. We find that both migrating prohemocytes and posterior mesodermal cells, before their integration into the Malpighian tubule primordia, simultaneously express the hibris reporter and Serpent. We also show that hibris function is not essential for prohemocyte migration out of the procephalic mesoderm NOR maintenance of Serpent expression in prohemocytes.


Asunto(s)
Movimiento Celular , Proteínas de Drosophila/genética , Drosophila/embriología , Factores de Transcripción GATA/genética , Hematopoyesis , Túbulos de Malpighi/embriología , Proteínas de la Membrana/genética , Animales , Drosophila/citología , Drosophila/genética , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/metabolismo , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Organogénesis
6.
Dis Model Mech ; 6(1): 184-96, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23118342

RESUMEN

Myotonic dystrophy type 1 (DM1) is a genetic disease caused by the pathological expansion of a CTG trinucleotide repeat in the 3' UTR of the DMPK gene. In the DMPK transcripts, the CUG expansions sequester RNA-binding proteins into nuclear foci, including transcription factors and alternative splicing regulators such as MBNL1. MBNL1 sequestration has been associated with key features of DM1. However, the basis behind a number of molecular and histological alterations in DM1 remain unclear. To help identify new pathogenic components of the disease, we carried out a genetic screen using a Drosophila model of DM1 that expresses 480 interrupted CTG repeats, i(CTG)480, and a collection of 1215 transgenic RNA interference (RNAi) fly lines. Of the 34 modifiers identified, two RNA-binding proteins, TBPH (homolog of human TAR DNA-binding protein 43 or TDP-43) and BSF (Bicoid stability factor; homolog of human LRPPRC), were of particular interest. These factors modified i(CTG)480 phenotypes in the fly eye and wing, and TBPH silencing also suppressed CTG-induced defects in the flight muscles. In Drosophila flight muscle, TBPH, BSF and the fly ortholog of MBNL1, Muscleblind (Mbl), were detected in sarcomeric bands. Expression of i(CTG)480 resulted in changes in the sarcomeric patterns of these proteins, which could be restored by coexpression with human MBNL1. Epistasis studies showed that Mbl silencing was sufficient to induce a subcellular redistribution of TBPH and BSF proteins in the muscle, which mimicked the effect of i(CTG)480 expression. These results provide the first description of TBPH and BSF as targets of Mbl-mediated CTG toxicity, and they suggest an important role of these proteins in DM1 muscle pathology.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Epistasis Genética , Femenino , Técnicas de Silenciamiento del Gen , Genes de Insecto , Humanos , Músculos/metabolismo , Músculos/patología , Distrofia Miotónica/patología , Proteínas Nucleares/antagonistas & inhibidores , Interferencia de ARN , Sarcómeros/metabolismo , Sarcómeros/patología , Expansión de Repetición de Trinucleótido
7.
Biol Rev Camb Philos Soc ; 86(4): 947-58, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21489124

RESUMEN

Regulated use of exons in pre-mRNAs, a process known as alternative splicing, strongly contributes to proteome diversity. Alternative splicing is finely regulated by factors that bind specific sequences within the precursor mRNAs. Members of the Muscleblind (Mbl) family of splicing factors control critical exon use changes during the development of specific tissues, particularly heart and skeletal muscle. Muscleblind homologs are only found in metazoans from Nematoda to mammals. Splicing targets and recognition mechanisms are also conserved through evolution. In this recognition, Muscleblind CCCH-type zinc finger domains bind to intronic motifs in pre-mRNA targets in which the protein can either activate or repress splicing of nearby exons, depending on the localization of the binding motifs relative to the regulated alternative exon. In humans, the Muscleblind-like 1 (MBNL1) proteins play a critical role in hereditary diseases caused by microsatellite expansions, particularly myotonic dystrophy type 1 (DM1), in which depletion of MBNL1 activity through sequestration explains most misregulated alternative splicing events, at least in murine models. Because of the involvement of these proteins in human diseases, further understanding of the molecular mechanisms by which MBNL1 regulates splicing will help design therapies to revert pathological splicing alterations. Here we summarize the most relevant findings on this family of proteins in recent years, focusing on recently described functional motifs, transcriptional regulation of Muscleblind, regulatory activity on splicing, and involvement in human diseases.


Asunto(s)
Empalme Alternativo/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Humanos , Familia de Multigenes , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Unión Proteica , ARN/genética , ARN/metabolismo
8.
PLoS One ; 3(2): e1595, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18270582

RESUMEN

Non-coding CUG repeat expansions interfere with the activity of human Muscleblind-like (MBNL) proteins contributing to myotonic dystrophy 1 (DM1). To understand this toxic RNA gain-of-function mechanism we developed a Drosophila model expressing 60 pure and 480 interrupted CUG repeats in the context of a non-translatable RNA. These flies reproduced aspects of the DM1 pathology, most notably nuclear accumulation of CUG transcripts, muscle degeneration, splicing misregulation, and diminished Muscleblind function in vivo. Reduced Muscleblind activity was evident from the sensitivity of CUG-induced phenotypes to a decrease in muscleblind genetic dosage and rescue by MBNL1 expression, and further supported by the co-localization of Muscleblind and CUG repeat RNA in ribonuclear foci. Targeted expression of CUG repeats to the developing eye and brain mushroom bodies was toxic leading to rough eyes and semilethality, respectively. These phenotypes were utilized to identify genetic and chemical modifiers of the CUG-induced toxicity. 15 genetic modifiers of the rough eye phenotype were isolated. These genes identify putative cellular processes unknown to be altered by CUG repeat RNA, and they include mRNA export factor Aly, apoptosis inhibitor Thread, chromatin remodelling factor Nurf-38, and extracellular matrix structural component Viking. Ten chemical compounds suppressed the semilethal phenotype. These compounds significantly improved viability of CUG expressing flies and included non-steroidal anti-inflammatory agents (ketoprofen), muscarinic, cholinergic and histamine receptor inhibitors (orphenadrine), and drugs that can affect sodium and calcium metabolism such as clenbuterol and spironolactone. These findings provide new insights into the DM1 phenotype, and suggest novel candidates for DM1 treatments.


Asunto(s)
Expansión de las Repeticiones de ADN , Repeticiones de Trinucleótidos/efectos de los fármacos , Repeticiones de Trinucleótidos/fisiología , Animales , Encéfalo , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Ojo , Dosificación de Gen , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN
9.
PLoS One ; 3(2): e1613, 2008 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-18286170

RESUMEN

BACKGROUND: Muscleblind-like proteins (MBNL) have been involved in a developmental switch in the use of defined cassette exons. Such transition fails in the CTG repeat expansion disease myotonic dystrophy due, in part, to sequestration of MBNL proteins by CUG repeat RNA. Four protein isoforms (MblA-D) are coded by the unique Drosophila muscleblind gene. METHODOLOGY/PRINCIPAL FINDINGS: We used evolutionary, genetic and cell culture approaches to study muscleblind (mbl) function in flies. The evolutionary study showed that the MblC protein isoform was readily conserved from nematods to Drosophila, which suggests that it performs the most ancestral muscleblind functions. Overexpression of MblC in the fly eye precursors led to an externally rough eye morphology. This phenotype was used in a genetic screen to identify five dominant suppressors and 13 dominant enhancers including Drosophila CUG-BP1 homolog aret, exon junction complex components tsunagi and Aly, and pro-apoptotic genes Traf1 and reaper. We further investigated Muscleblind implication in apoptosis and splicing regulation. We found missplicing of troponin T in muscleblind mutant pupae and confirmed Muscleblind ability to regulate mouse fast skeletal muscle Troponin T (TnnT3) minigene splicing in human HEK cells. MblC overexpression in the wing imaginal disc activated apoptosis in a spatially restricted manner. Bioinformatics analysis identified a conserved FKRP motif, weakly resembling a sumoylation target site, in the MblC-specific sequence. Site-directed mutagenesis of the motif revealed no change in activity of mutant MblC on TnnT3 minigene splicing or aberrant binding to CUG repeat RNA, but altered the ability of the protein to form perinuclear aggregates and enhanced cell death-inducing activity of MblC overexpression. CONCLUSIONS/SIGNIFICANCE: Taken together our genetic approach identify cellular processes influenced by Muscleblind function, whereas in vivo and cell culture experiments define Drosophila troponin T as a new Muscleblind target, reveal a potential involvement of MblC in programmed cell death and recognize the FKRP motif as a putative regulator of MblC function and/or subcellular location in the cell.


Asunto(s)
Empalme Alternativo , Apoptosis , Proteínas de Drosophila/fisiología , Proteínas de Unión al ARN/fisiología , Troponina T/genética , Secuencias de Aminoácidos , Animales , Drosophila , Expansión de Repetición de Trinucleótido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA