Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cell ; 150(4): 710-24, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22901804

RESUMEN

The muscleblind-like (Mbnl) family of RNA-binding proteins plays important roles in muscle and eye development and in myotonic dystrophy (DM), in which expanded CUG or CCUG repeats functionally deplete Mbnl proteins. We identified transcriptome-wide functional and biophysical targets of Mbnl proteins in brain, heart, muscle, and myoblasts by using RNA-seq and CLIP-seq approaches. This analysis identified several hundred splicing events whose regulation depended on Mbnl function in a pattern indicating functional interchangeability between Mbnl1 and Mbnl2. A nucleotide resolution RNA map associated repression or activation of exon splicing with Mbnl binding near either 3' splice site or near the downstream 5' splice site, respectively. Transcriptomic analysis of subcellular compartments uncovered a global role for Mbnls in regulating localization of mRNAs in both mouse and Drosophila cells, and Mbnl-dependent translation and protein secretion were observed for a subset of mRNAs with Mbnl-dependent localization. These findings hold several new implications for DM pathogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Distrofia Miotónica/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcriptoma , Regiones no Traducidas 3' , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila , Drosophila melanogaster/metabolismo , Exones , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Mioblastos/metabolismo , Distrofia Miotónica/genética , Proteínas Nucleares , Especificidad de Órganos , Sitios de Empalme de ARN , Proteínas de Unión al ARN/genética
2.
EMBO Rep ; 12(7): 735-42, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21637295

RESUMEN

We describe a new mechanism by which CTG tract expansion affects myotonic dystrophy (DM1). Changes to the levels of a panel of RNAs involved in muscle development and function that are downregulated in DM1 are due to aberrant localization of the transcription factor SHARP (SMART/HDAC1-associated repressor protein). Mislocalization of SHARP in DM1 is consistent with increased CRM1-mediated export of SHARP to the cytoplasm. A direct link between CTG repeat expression and SHARP mislocalization is demonstrated as expression of expanded CTG repeats in normal cells recapitulates cytoplasmic SHARP localization. These results demonstrate a role for the inactivation of SHARP transcription in DM1 biology.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Homeodominio/metabolismo , Distrofia Miotónica/fisiopatología , Proteínas Nucleares/metabolismo , ARN/metabolismo , Antibióticos Antineoplásicos/farmacología , Citoplasma/metabolismo , Proteínas de Unión al ADN , Ácidos Grasos Insaturados/farmacología , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Mioblastos/metabolismo , Distrofia Miotónica/genética , Proteínas Nucleares/genética , Transporte de Proteínas/efectos de los fármacos , Empalme del ARN/genética , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido/genética
3.
Exp Cell Res ; 318(1): 1-7, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21871450

RESUMEN

Lamin A is a component of the nuclear envelope that is synthesized as a precursor prelamin A molecule and then processed into mature lamin A through sequential steps of posttranslational modifications and proteolytic cleavages. Remarkably, over 400 distinct point mutations have been so far identified throughout the LMNA gene, which result in the development of at least ten distinct human disorders, collectively known as laminopathies, among which is the premature aging disease Hutchinson-Gilford progeria syndrome (HGPS). The majority of HGPS cases are associated with a single point mutation in the LMNA gene that causes the production of a permanently farnesylated mutant lamin A protein termed progerin. The mechanism by which progerin leads to premature aging and the classical HGPS disease phenotype as well as the relationship between this disorder and the onset of analogous symptoms during the lifespan of a normal individual are not well understood. Yet, recent studies have provided critical insights on the cellular processes that are affected by accumulation of progerin and have suggested that cellular alterations in the lamin A processing pathway leading to the accumulation of farnesylated prelamin A intermediates may play a role in the aging process in the general population. In this review we provide a short background on lamin A and its maturation pathway and discuss the current knowledge of how progerin or alterations in the prelamin A processing pathway are thought to influence cell function and contribute to human aging.


Asunto(s)
Envejecimiento , Lamina Tipo A/metabolismo , Prenilación , Animales , Humanos , Lamina Tipo A/genética , Proteínas Nucleares/metabolismo , Progeria/genética , Progeria/metabolismo , Precursores de Proteínas/metabolismo
4.
iScience ; 26(5): 106732, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37216102

RESUMEN

Patients with myotonic dystrophy type I (DM1) demonstrate visuospatial dysfunction and impaired performance in tasks requiring recognition or memory of figures and objects. In DM1, CUG expansion RNAs inactivate the muscleblind-like (MBNL) proteins. We show that constitutive Mbnl2 inactivation in Mbnl2ΔE2/ΔE2 mice selectively impairs object recognition memory in the novel object recognition test. When exploring the context of a novel arena in which the objects are later encountered, the Mbnl2ΔE2/ΔE2 dorsal hippocampus responds with a lack of enrichment for learning and memory-related pathways, mounting instead transcriptome alterations predicted to impair growth and neuron viability. In Mbnl2ΔE2/ΔE2 mice, saturation effects may prevent deployment of a functionally relevant transcriptome response during novel context exploration. Post-novel context exploration alterations in genes implicated in tauopathy and dementia are observed in the Mbnl2ΔE2/ΔE2 dorsal hippocampus. Thus, MBNL2 inactivation in patients with DM1 may alter novel context processing in the dorsal hippocampus and impair object recognition memory.

5.
J Biol Chem ; 286(44): 38427-38438, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21900255

RESUMEN

To understand the role of the splice regulator muscleblind 1 (MBNL1) in the development of RNA splice defects in myotonic dystrophy I (DM1), we purified RNA-independent MBNL1 complexes from normal human myoblasts and examined the behavior of these complexes in DM1 myoblasts. Antibodies recognizing MBNL1 variants (MBNL1(CUG)), which can sequester in the toxic CUG RNA foci that develop in DM1 nuclei, were used to purify MBNL1(CUG) complexes from normal myoblasts. In normal myoblasts, MBNL1(CUG) bind 10 proteins involved in remodeling ribonucleoprotein complexes including hnRNP H, H2, H3, F, A2/B1, K, L, DDX5, DDX17, and DHX9. Of these proteins, only MBNL1(CUG) colocalizes extensively with DM1 CUG foci (>80% of foci) with its partners being present in <10% of foci. Importantly, the stoichiometry of MBNL1(CUG) complexes is altered in DM1 myoblasts, demonstrating an increase in the steady state levels of nine of its partner proteins. These changes are recapitulated by the expression of expanded CUG repeat RNA in Cos7 cells. Altered stoichiometry of MBNL1(CUG) complexes results from aberrant protein synthesis or stability and is unlinked to PKCα function. Modeling these changes in normal myoblasts demonstrates that increased levels of hnRNP H, H2, H3, F, and DDX5 independently dysregulate splicing in overlapping RNA subsets. Thus expression of expanded CUG repeats alters the stoichiometry of MBNL1(CUG) complexes to allow both the reinforcement and expansion of RNA processing defects.


Asunto(s)
Distrofia Miotónica/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Inmunoprecipitación , Espectrometría de Masas/métodos , Músculo Esquelético/metabolismo , Distrofia Miotónica/genética , Mapeo de Interacción de Proteínas/métodos , ARN Interferente Pequeño/metabolismo , Fracciones Subcelulares
6.
Exp Cell Res ; 317(3): 319-29, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20974128

RESUMEN

Lamin A is a component of the nuclear lamina that plays a major role in the structural organization and function of the nucleus. Lamin A is synthesized as a prelamin A precursor which undergoes four sequential post-translational modifications to generate mature lamin A. Significantly, a large number of point mutations in the LMNA gene cause a range of distinct human disorders collectively known as laminopathies. The mechanisms by which mutations in lamin A affect cell function and cause disease are unclear. Interestingly, recent studies have suggested that alterations in the normal lamin A pathway can contribute to cellular dysfunction. Specifically, we and others have shown, at the cellular level, that in the absence of mutations or altered splicing events, increased expression of wild-type prelamin A results in a growth defective phenotype that resembles that of cells expressing the mutant form of lamin A, termed progerin, associated with Hutchinson-Gilford Progeria syndrome (HGPS). Remarkably, the phenotypes of cells expressing elevated levels of wild-type prelamin A can be reversed by either treatment with farnesyltransferase inhibitors or overexpression of ZMPSTE24, a critical prelamin A processing enzyme, suggesting that minor increases in the steady-state levels of one or more prelamin A intermediates is sufficient to induce cellular toxicity. Here, to investigate the molecular basis of the lamin A pathway toxicity, we characterized the phenotypic changes occurring in cells expressing distinct prelamin A variants mimicking specific prelamin A processing intermediates. This analysis demonstrates that distinct prelamin A variants differentially affect cell growth, nuclear membrane morphology, nuclear distribution of lamin A and the fundamental process of transcription. Expression of prelamin A variants that are constitutively farnesylated induced the formation of lamin A aggregates and dramatic changes in nuclear membrane morphology, which led to reduced levels of the basal transcription factor TATA-binding protein (TBP) and global transcription, and severely limited cell growth. Expression of a prelamin A variant that cannot be farnesylated, although did not appreciably influence cell growth, resulted in the formation of lamin A nucleoplasmic foci and caused, in a minor subpopulation of cells, changes in nuclear morphology that were accompanied by reduced levels of TBP and transcription. In contrast, expression of mature lamin A did not affect any of these parameters. These data demonstrate that accumulation of any partially processed prelamin A protein alters cellular homeostasis to some degree, even though the most dramatic effects are caused by variants with a permanently farnesylated carboxyl-terminal tail.


Asunto(s)
Fibroblastos/metabolismo , Homeostasis , Diploidia , Fibroblastos/fisiología , Variación Genética , Humanos , Lamina Tipo A/genética , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progeria , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
7.
J Biol Chem ; 285(33): 25426-37, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20519504

RESUMEN

The insulin receptor exists as two isoforms, IR-A and IR-B, which result from alternative splicing of exon 11 in the primary transcript. These two isoforms show a cell-specific distribution, and their relative proportions also vary during development, aging, and in different disease states. We have previously demonstrated that both intron 10 and the alternatively spliced exon 11 contain regulatory sequences that affect insulin receptor splicing both positively and negatively and that these sequences bind the serine/arginine-rich (SR) proteins SRp20 and SF2/ASF and the CELF protein CUG-BP1. In this study, we describe a new intronic splicing element within intron 11 that is highly conserved across species. Using minigenes carrying deletion mutations within intron 11, we demonstrated that this sequence functions as an intronic splicing enhancer. We subsequently used RNA affinity chromatography to identify Mbnl1 as a splicing factor that recognizes this enhancer. By ribonucleoprotein immunoprecipitation, we also established that Mbnl1 binds specifically to the INSR (insulin receptor gene) RNA. Overexpression or knockdown of Mbnl1 in hepatoma and embryonic kidney cells altered the levels of exon 11 inclusion. Finally, we showed that deletion of the intronic enhancer eliminates the ability of Mbnl1 to promote exon inclusion. Collectively, these findings demonstrate a role for Mbnl1 in controlling insulin receptor exon 11 inclusion via binding to a downstream intronic enhancer element.


Asunto(s)
Exones/genética , Intrones/genética , Proteínas de Unión al ARN/metabolismo , Receptor de Insulina/genética , Animales , Western Blotting , Línea Celular , Elementos de Facilitación Genéticos , Evolución Molecular , Células Hep G2 , Humanos , Unión Proteica/genética , Unión Proteica/fisiología , Empalme del ARN/genética , Empalme del ARN/fisiología , ARN Interferente Pequeño , Proteínas de Unión al ARN/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Commun Biol ; 4(1): 1342, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34848815

RESUMEN

Myotonic Dystrophy Type I (DM1) patients demonstrate widespread and variable brain structural alterations whose etiology is unclear. We demonstrate that inactivation of the Muscleblind-like proteins, Mbnl1 and Mbnl2, initiates brain structural defects. 2D FSE T2w MRIs on 4-month-old Mbnl1+/-/Mbnl2-/- mice demonstrate whole-brain volume reductions, ventriculomegaly and regional gray and white matter volume reductions. Comparative MRIs on 2-month-old Mbnl1-/-, Mbnl2-/- and Mbnl1-/-/Mbnl2+/- brains show genotype-specific reductions in white and gray matter volumes. In both cohorts, white matter volume reductions predominate, with Mbnl2 loss leading to more widespread alterations than Mbnl1 loss. Hippocampal volumes are susceptible to changes in either Mbnl1 or Mbnl2 levels, where both single gene and dual depletions result in comparable volume losses. In contrast, the cortex, inter/midbrain, cerebellum and hindbrain regions show both gene and dose-specific volume decreases. Our results provide a molecular explanation for phenotype intensification in congenital DM1 and the variability in the brain structural alterations reported in DM1.


Asunto(s)
Encéfalo/patología , Proteínas de Unión al ADN/genética , Genotipo , Proteínas de Unión al ARN/genética , Animales , Proteínas de Unión al ADN/metabolismo , Femenino , Ratones , Proteínas de Unión al ARN/metabolismo
9.
ScientificWorldJournal ; 9: 1449-62, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20024518

RESUMEN

Syndromes of accelerated aging could provide an entry point for identifying and dissecting the cellular pathways that are involved in the development of age-related pathologies in the general population. However, their usefulness for aging research has been controversial, as it has been argued that these diseases do not faithfully reflect the process of natural aging. Here we review recent findings on the molecular basis of two progeroid diseases, Werner syndrome (WS) and Hutchinson-Gilford progeria syndrome (HGPS), and highlight functional connections to cellular processes that may contribute to normal aging.


Asunto(s)
Envejecimiento/metabolismo , Nucleoproteínas/metabolismo , Progeria/metabolismo , Telómero/metabolismo , Síndrome de Werner/metabolismo , Investigación Biomédica , Cromatina/metabolismo , Cromatina/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Lamina Tipo A , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Nucleoproteínas/fisiología , Precursores de Proteínas/metabolismo , Precursores de Proteínas/fisiología , Telómero/fisiología
10.
Nucleic Acids Res ; 34(17): 4752-66, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16971462

RESUMEN

High levels of rRNA synthesis by RNA polymerase I are important for cell growth and proliferation. In vitro studies have indicated that the formation of a stable complex between the HMG box factor [Upstream binding factor (UBF)] and SL1 at the rRNA gene promoter is necessary to direct multiple rounds of Pol I transcription initiation. The recruitment of SL1 to the promoter occurs through protein interactions with UBF and is regulated by phosphorylation of UBF. Here we show that the protein kinase CK2 co-immunoprecipitates with the Pol I complex and is associated with the rRNA gene promoter. Inhibition of CK2 kinase activity reduces Pol I transcription in cultured cells and in vitro. Significantly, CK2 regulates the interaction between UBF and SL1 by counteracting the inhibitory effect of HMG boxes five and six through the phosphorylation of specific serines located at the C-terminus of UBF. Transcription reactions with immobilized templates indicate that phosphorylation of CK2 phosphoacceptor sites in the C-terminal domain of UBF is important for promoting multiple rounds of Pol I transcription. These data demonstrate that CK2 is recruited to the rRNA gene promoter and directly regulates Pol I transcription re-initiation by stabilizing the association between UBF and SL1.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , ARN Polimerasa I/metabolismo , Transcripción Genética , Sitios de Unión , Línea Celular , Genes de ARNr , Humanos , Mutación , Fosforilación , Proteínas del Complejo de Iniciación de Transcripción Pol1/química , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Factores de Transcripción/metabolismo
11.
Mol Cell Biol ; 37(3)2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27849570

RESUMEN

The Werner syndrome protein (WRN) suppresses the loss of telomeres replicated by lagging-strand synthesis by a yet to be defined mechanism. Here, we show that whereas either WRN or the Bloom syndrome helicase (BLM) stimulates DNA polymerase δ progression across telomeric G-rich repeats, only WRN promotes sequential strand displacement synthesis and FEN1 cleavage, a critical step in Okazaki fragment maturation, at these sequences. Helicase activity, as well as the conserved winged-helix (WH) motif and the helicase and RNase D C-terminal (HRDC) domain play important but distinct roles in this process. Remarkably, WRN also influences the formation of FEN1 cleavage products during strand displacement on a nontelomeric substrate, suggesting that WRN recruitment and cooperative interaction with FEN1 during lagging-strand synthesis may serve to regulate sequential strand displacement and flap cleavage at other genomic sites. These findings define a biochemical context for the physiological role of WRN in maintaining genetic stability.


Asunto(s)
ADN Polimerasa III/metabolismo , Replicación del ADN , Endonucleasas de ADN Solapado/metabolismo , Helicasa del Síndrome de Werner/metabolismo , Secuencias de Aminoácidos , ADN/biosíntesis , Células HeLa , Homeostasis , Humanos , Polimerizacion , Dominios Proteicos , RecQ Helicasas/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos/genética , Especificidad por Sustrato , Telómero/metabolismo , Helicasa del Síndrome de Werner/química
12.
F1000Res ; 5: 2536, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27803806

RESUMEN

Lamins are major components of the nuclear lamina, a network of proteins that supports the nuclear envelope in metazoan cells. Over the past decade, biochemical studies have provided support for the view that lamins are not passive bystanders providing mechanical stability to the nucleus but play an active role in the organization of the genome and the function of fundamental nuclear processes. It has also become apparent that lamins are critical for human health, as a large number of mutations identified in the gene that encodes for A-type lamins are associated with tissue-specific and systemic genetic diseases, including the accelerated aging disorder known as Hutchinson-Gilford progeria syndrome. Recent years have witnessed great advances in our understanding of the role of lamins in the nucleus and the functional consequences of disease-associated A-type lamin mutations. Many of these findings have been presented in comprehensive reviews. In this mini-review, we discuss recent breakthroughs in the role of lamins in health and disease and what lies ahead in lamin research.

13.
Sci Rep ; 6: 30999, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27484195

RESUMEN

Myotonic dystrophy type I (DM1) exhibits distinctive disease specific phenotypes and the accelerated onset of a spectrum of age-associated pathologies. In DM1, dominant effects of expanded CUG repeats result in part from the inactivation of the muscleblind-like (MBNL) proteins. To test the role of MBNL3, we deleted Mbnl3 exon 2 (Mbnl3(ΔE2)) in mice and examined the onset of age-associated diseases over 4 to 13 months of age. Accelerated onset of glucose intolerance with elevated insulin levels, cardiac systole deficits, left ventricle hypertrophy, a predictor of a later onset of heart failure and the development of subcapsular and cortical cataracts is observed in Mbnl3(ΔE2) mice. Retention of embryonic splice isoforms in adult organs, a prominent defect in DM1, is not observed in multiple RNAs including the Insulin Receptor (Insr), Cardiac Troponin T (Tnnt2), Lim Domain Binding 3 (Ldb3) RNAs in Mbnl3(ΔE2) mice. Although rare DM1-like splice errors underlying the observed phenotypes cannot be excluded, our data in conjunction with the reported absence of alternative splice errors in embryonic muscles of a similar Mbnl3(ΔE2) mouse by RNA-seq studies, suggest that mechanisms distinct from the adult retention of embryonic splice patterns may make important contributions to the onset of age-associated pathologies in DM1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Empalme Alternativo , Proteínas Portadoras/fisiología , Proteínas con Dominio LIM/genética , Músculo Esquelético/patología , Distrofia Miotónica/patología , Animales , Exones , Regulación del Desarrollo de la Expresión Génica , Intolerancia a la Glucosa , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofia Miotónica/etiología , Distrofia Miotónica/metabolismo , Unión Proteica , Proteínas de Unión al ARN
14.
EBioMedicine ; 2(9): 1034-47, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26501102

RESUMEN

Loss of Muscleblind-like 1 (Mbnl1) is known to alter Clc-1 splicing to result in myotonia. Mbnl1(ΔE3/ΔE3)/Mbnl3(ΔE2) mice, depleted of Mbnl1 and Mbnl3, demonstrate a profound enhancement of myotonia and an increase in the number of muscle fibers with very low Clc-1 currents, where gClmax values approach ~ 1 mS/cm(2), with the absence of a further enhancement in Clc-1 splice errors, alterations in polyA site selection or Clc-1 localization. Significantly, Mbnl1(ΔE3/ΔE3)/Mbnl3(ΔE2) muscles demonstrate an aberrant accumulation of Clc-1 RNA on monosomes and on the first polysomes. Mbnl1 and Mbnl3 bind Clc-1 RNA and both proteins bind Hsp70 and eEF1A, with these associations being reduced in the presence of RNA. Thus binding of Mbnl1 and Mbnl3 to Clc-1 mRNA engaged with ribosomes can facilitate an increase in the local concentration of Hsp70 and eEF1A to assist Clc-1 translation. Dual depletion of Mbnl1 and Mbnl3 therefore initiates both Clc-1 splice errors and translation defects to synergistically enhance myotonia. As the HSA(LR) model for myotonic dystrophy (DM1) shows similar Clc-1 defects, this study demonstrates that both splice errors and translation defects are required for DM1 pathology to manifest. RESEARCH IN CONTEXT: Research in context: Myotonic Dystrophy type 1 (DM1) is a dominant disorder resulting from the expression of expanded CUG repeat RNA, which aberrantly sequesters and inactivates the muscleblind-like (MBNL) family of proteins. In mice, inactivation of Mbnl1 is known to alter Clc-1 splicing to result in myotonia. We demonstrate that concurrent depletion of Mbnl1 and Mbnl3 results in a synergistic enhancement of myotonia, with an increase in muscle fibers showing low chloride currents. The observed synergism results from the aberrant accumulation of Clc-1 mRNA on monosomes and the first polysomes. This translation error reflects the ability of Mbnl1 and Mbnl3 to act as adaptors that recruit Hsp70 and eEF1A to the Clc-1 mRNA engaged with ribosomes, to facilitate translation. Thus our study demonstrates that Clc-1 RNA translation defects work coordinately with Clc-1 splice errors to synergistically enhance myotonia in mice lacking Mbnl1 and Mbnl3.


Asunto(s)
Proteínas Portadoras/genética , Canales de Cloruro/genética , Proteínas de Unión al ADN/genética , Miotonía/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Empalme Alternativo , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Canales de Cloruro/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Immunoblotting , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miotonía/metabolismo , Miotonía/fisiopatología , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Distrofia Miotónica/fisiopatología , Factor 1 de Elongación Peptídica/metabolismo , Unión Proteica , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribosomas/genética , Ribosomas/metabolismo
15.
Sci Rep ; 5: 9042, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25761764

RESUMEN

Cardiac dysfunction is a prominent cause of mortality in myotonic dystrophy I (DM1), a disease where expanded CUG repeats bind and disable the muscleblind-like family of splice regulators. Deletion of muscleblind-like 1 (Mbnl1(ΔE2/ΔE2)) in 129 sv mice results in QRS, QTc widening, bundle block and STc narrowing at 2-4 months of age. With time, cardiac function deteriorates further and at 6 months, decreased R wave amplitudes, sinus node dysfunction, cardiac hypertrophy, interstitial fibrosis, multi-focal myocardial fiber death and calcification manifest. Sudden death, where no end point illness is overt, is observed at a median age of 6.5 and 4.8 months in ~67% and ~86% of male and female Mbnl1(ΔE2/ΔE2) mice, respectively. Mbnl1 depletion results in the persistence of embryonic splice isoforms in a network of cardiac RNAs, some of which have been previously implicated in DM1, regulating sodium and calcium currents, Scn5a, Junctin, Junctate, Atp2a1, Atp11a, Cacna1s, Ryr2, intra and inter cellular transport, Clta, Stx2, Tjp1, cell survival, Capn3, Sirt2, Csda, sarcomere and cytoskeleton organization and function, Trim55, Mapt, Pdlim3, Pdlim5, Sorbs1, Sorbs2, Fhod1, Spag9 and structural components of the sarcomere, Myom1, Tnnt2, Zasp. Thus this study supports a key role for Mbnl1 loss in the initiation of DM1 cardiac disease.


Asunto(s)
Empalme Alternativo , Eliminación de Gen , Distrofia Miotónica/genética , Isoformas de ARN , Proteínas de Unión al ARN/genética , Animales , Arritmia Sinusal , Calcinosis , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Fibrosis , Expresión Génica , Orden Génico , Marcación de Gen , Sitios Genéticos , Longevidad/genética , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Distrofia Miotónica/fisiopatología , Fenotipo
16.
Physiol Genomics ; 12(2): 147-57, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12454205

RESUMEN

DMPK is a serine/threonine kinase implicated in the human disease myotonic muscular dystrophy (DM). Skeletal muscle Na channels exhibit late reopenings in Dmpk-deficient mice and peak current density is reduced, implicating DMPK in regulation of membrane excitability. Since complete heart block and sudden cardiac death occur in the disease, we tested the hypothesis that cardiac Na channels also exhibit abnormal gating in Dmpk-deficient mice. We made whole cell and cell-attached patch clamp recordings of ventricular cardiomyocytes enzymatically isolated from wild-type, Dmpk+/-, and Dmpk-/- mice. Recordings from membrane patches containing one or a few Na channels revealed multiple Na channel reopenings occurring after the macroscopic Na current had subsided in both Dmpk+/- and Dmpk-/- muscle, but only rare reopenings in wild-type muscle (>3-fold difference, P < 0.05). This resulted in a plateau of non-inactivating Na current in Dmpk-deficient muscle. The magnitude of this plateau current was independent on the magnitude of the test potential from -40 to 0 mV and was also independent of gene dose. Macroscopic Na current density was similar in wild-type and Dmpk-deficient muscle, as was steady-state Na channel gating. Decay of macroscopic currents was slowed in Dmpk-/- muscle, but not in Dmpk+/- or wild-type muscle. Entry into, and recovery from, inactivation were similar at multiple test potentials in wild-type and Dmpk-deficient muscle. Resting membrane potential was depolarized, and action potential duration was significantly prolonged in Dmpk-deficient muscle. Thus in cardiac muscle, Dmpk deficiency results in multiple late reopenings of Na channels similar to those seen in Dmpk-deficient skeletal muscle. This is reflected in a plateau of non-inactivating macroscopic Na current and prolongation of cardiac action potentials.


Asunto(s)
Activación del Canal Iónico/genética , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Canales de Sodio/genética , Potenciales de Acción/genética , Animales , Animales Congénicos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Miocitos Cardíacos/química , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica , Técnicas de Placa-Clamp , Canales de Sodio/metabolismo , Canales de Sodio/fisiología
17.
Neuromuscul Disord ; 14(8-9): 497-506, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15336691

RESUMEN

Myotonic dystrophy1 (DM1) is an autosomal dominant, multi-system disorder resulting from a CTG repeat expansion located in the 3' untranslated region of DMPK and immediately in the 5' of SIX5. Skeletal muscle, heart and smooth muscle are prominently affected in DM1. Endocrine abnormalities, gonadal atrophy, brain, skin, skeletal, immune and respiratory defects are also features of the disorder. Both DMPK and SIX5 levels are decreased in DM1 patients. Importantly, expression of mutant mRNAs encoding expanded CUG repeats has been shown to alter the activity of CUG repeat binding proteins in DM1. Mouse models have demonstrated that decreased levels of Dmpk, Six5 and the expression of expanded CUG repeats independently contribute to the development of DM1 pathology. However, an important gap in these studies is a lack of clear understanding of the expression pattern of Dmpk. We demonstrate that Dmpk mRNA is expressed in a range of adult mouse tissues that show pathology in DM1 including skeletal muscle, heart, smooth muscle, bone, testis, pituitary, brain, eye, skin, thymus and lung. Thus DMPK loss or CUG repeat expansion could contribute to DM1 pathology to these tissues. Dmpk mRNA is not detected in the ovary, pancreas or kidney. Significantly, Dmpk mRNA is expressed in the intestinal epithelium, cartilage and liver, which have not been reported to show consistent abnormalities in Dmpk(-/-) mice or in transgenic animals expressing CUG repeats. Taken together these data suggest that Dmpk loss or CUG repeat expression per se may not be sufficient to initiate pathology and are consistent with the hypothesis that coexpression of specific CUG repeat binding proteins with the mutant Dmpk mRNA or deregulation of genes such as Six5 that flank the CTG repeat tract may be necessary for DM1 to manifest.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hibridación in Situ/métodos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Northern Blotting/métodos , Huesos/metabolismo , Encéfalo/metabolismo , Cartílago/metabolismo , Femenino , Corazón , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculos/metabolismo , Proteína Quinasa de Distrofia Miotónica , ARN Mensajero/metabolismo , Factores Sexuales , Piel/metabolismo , Testículo/metabolismo , Timo/metabolismo
18.
J Interv Card Electrophysiol ; 7(2): 127-35, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12397222

RESUMEN

Myotonic dystrophy (DM) is an autosomal dominant multisystem disorder, caused by expansion of a CTG trinucleotide repeat in the 3' untranslated region of the myotonic dystrophy protein kinase gene (DMPK) on chromosome 19q13. Cardiac involvement in DM includes conduction abnormalities and functional deficits. Three hypotheses of molecular mechanisms for DM pathophysiology are; first, partial loss of myotonic dystrophy protein kinase (DMPK); second, decreased transcription of a neighboring homeodomain-encoding gene, Six5 (or DMAHP), and third, transdominant effects of the RNA and regulation of splicing associated with expression of expanded CUG repeats. However, the precise pathogenetic mechanism remains unresolved. We previously reported that dosage of Dm15, the mouse homologue of DMPK, strongly associates with the cardiac conduction abnormalities. For further distinction of the molecular mechanisms underlying the cardiac phenotype of DM, in the present study, we characterized the cardiac conduction findings of mice with targeted disruption of Six5 gene. Six5 heterozygous mice (adult and young) and their age matched wild type littermates were studied using in vivo electrophysiologic techniques, echocardiography, heart rate variability and exercise tolerance testing. No PR prolongation was detected, however, prolonged QRS duration and delayed infraHisian conduction were significant in adult Six5 heterozygous mice. By echocardiography, left ventricular (LV) end-diastolic dimension was enlarged in adult Six5 heterozygous mice, although neither fractioning shortening nor LV wall thickness showed significant differences. Six5 loss may partly contribute to conduction abnormalities in myotonic dystrophy, particularly infraHisian conduction delay, one of the initial phenotypes of adult-onset cardiac conduction abnormalities in DM patients.


Asunto(s)
Eliminación de Gen , Sistema de Conducción Cardíaco/anomalías , Proteínas de Homeodominio/genética , Animales , Ecocardiografía , Electrocardiografía , Electrofisiología , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca , Heterocigoto , Ratones , Ratones Endogámicos , Monitoreo Fisiológico/métodos , Actividad Motora/fisiología , Resistencia Física , Telemetría
19.
Aging Cell ; 13(2): 367-78, 2014 04.
Artículo en Inglés | MEDLINE | ID: mdl-24757718

RESUMEN

The Werner syndrome protein (WRN) is a nuclear protein required for cell growth and proliferation. Loss-of-function mutations in the Werner syndrome gene are associated with the premature onset of age-related diseases. How loss of WRN limits cell proliferation and induces replicative senescence is poorly understood. Here, we show that WRN depletion leads to a striking metabolic shift that coordinately weakens the pathways that generate reducing equivalents for detoxification of reactive oxygen species and increases mitochondrial respiration. In cancer cells, this metabolic shift counteracts the Warburg effect, a defining characteristic of many malignant cells, resulting in altered redox balance and accumulation of oxidative DNA damage that inhibits cell proliferation and induces a senescence-like phenotype. Consistent with these findings, supplementation with antioxidant rescues at least in part cell proliferation and decreases senescence in WRN-knockdown cancer cells. These results demonstrate that WRN plays a critical role in cancer cell proliferation by contributing to the Warburg effect and preventing metabolic stress.


Asunto(s)
Regulación hacia Abajo/genética , Exodesoxirribonucleasas/genética , Homeostasis , Neoplasias/metabolismo , Neoplasias/patología , RecQ Helicasas/genética , Animales , Antioxidantes/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Daño del ADN , Regulación hacia Abajo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Exodesoxirribonucleasas/metabolismo , Técnicas de Silenciamiento del Gen , Glutatión/metabolismo , Glutatión/farmacología , Homeostasis/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sustancias Macromoleculares/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Niacinamida/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , RecQ Helicasas/metabolismo , Síndrome de Werner/genética , Helicasa del Síndrome de Werner
20.
PLoS One ; 7(11): e48825, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166594

RESUMEN

Myotonic dystrophy (DM1) is a highly variable, multi-system disorder resulting from the expansion of an untranslated CTG tract in DMPK. In DM1 expanded CUG repeat RNAs form hairpin secondary structures that bind and aberrantly sequester the RNA splice regulator, MBNL1. RNA splice defects resulting as a consequence of MBNL1 depletion have been shown to play a key role in the development of DM1 pathology. In patient populations, both the number and severity of DM1 symptoms increase broadly as a function of CTG tract length. However significant variability in the DM1 phenotype is observed in patients encoding similar CTG repeat numbers. Here we demonstrate that a gradual decrease in MBNL1 levels results both in the expansion of the repertoire of splice defects and an increase in the severity of the splice alterations. Thus, MBNL1 loss does not have an all or none outcome but rather shows a graded effect on the number and severity of the ensuing splice defects. Our results suggest that once a critical threshold is reached, relatively small dose variations of free MBNL1 levels, which may reflect modest changes in the size of the CUG tract or the extent of hairpin secondary structure formation, can significantly alter the number and severity of splice abnormalities and thus contribute to the phenotype variability observed in DM1 patients.


Asunto(s)
Distrofia Miotónica/genética , Fenotipo , Empalme del ARN/genética , Proteínas de Unión al ARN/metabolismo , Western Blotting , Semivida , Humanos , Músculo Esquelético/metabolismo , Distrofia Miotónica/patología , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Expansión de Repetición de Trinucleótido/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA