Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Neurosci ; 42: 227-247, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-30909783

RESUMEN

Microsatellite mutations involving the expansion of tri-, tetra-, penta-, or hexanucleotide repeats cause more than 40 different neurological disorders. Although, traditionally, the position of the repeat within or outside of an open reading frame has been used to focus research on disease mechanisms involving protein loss of function, protein gain of function, or RNA gain of function, the discoveries of bidirectional transcription and repeat-associated non-ATG (RAN) have blurred these distinctions. Here we review what is known about RAN proteins in disease, the mechanisms by which they are produced, and the novel therapeutic opportunities they provide.


Asunto(s)
Expansión de las Repeticiones de ADN/genética , Proteínas del Tejido Nervioso/genética , Enfermedades del Sistema Nervioso/genética , Biosíntesis de Proteínas , Codón Iniciador/genética , Estrés del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/fisiología , Mutación con Ganancia de Función , Código Genético , Humanos , Mutación con Pérdida de Función , Repeticiones de Microsatélite/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Transcripción Genética
2.
Ann Neurol ; 93(2): 398-416, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36151701

RESUMEN

OBJECTIVE: The mechanistic target of rapamycin (mTOR) kinase is one of the master coordinators of cellular stress responses, regulating metabolism, autophagy, and apoptosis. We recently reported that staufen1 (STAU1), a stress granule (SG) protein, was overabundant in fibroblast cell lines from patients with spinocerebellar ataxia type 2 (SCA2), amyotrophic lateral sclerosis, frontotemporal degeneration, Huntington's, Alzheimer's, and Parkinson's diseases as well as animal models, and patient tissues. STAU1 overabundance is associated with mTOR hyperactivation and links SG formation with autophagy. Our objective was to determine the mechanism of mTOR regulation by STAU1. METHODS: We determined STAU1 abundance with disease- and chemical-induced cellular stressors in patient cells and animal models. We also used RNA-binding assays to contextualize STAU1 interaction with MTOR mRNA. RESULTS: STAU1 and mTOR were overabundant in bacterial artificial chromosome (BAC)-C9ORF72, ATXN2Q127 , and Thy1-TDP-43 transgenic mouse models. Reducing STAU1 levels in these mice normalized mTOR levels and activity and autophagy-related marker proteins. We also saw increased STAU1 levels in HEK293 cells transfected to express C9ORF72-relevant dipeptide repeats (DPRs). Conversely, DPR accumulations were not observed in cells treated by STAU1 RNA interference (RNAi). Overexpression of STAU1 in HEK293 cells increased mTOR levels through direct MTOR mRNA interaction, activating downstream targets and impairing autophagic flux. Targeting mTOR by rapamycin or RNAi normalized STAU1 abundance in an SCA2 cellular model. INTERPRETATION: STAU1 interaction with mTOR drives its hyperactivation and inhibits autophagic flux in multiple models of neurodegeneration. Staufen, therefore, constitutes a novel target to modulate mTOR activity and autophagy, and for the treatment of neurodegenerative diseases. ANN NEUROL 2023;93:398-416.


Asunto(s)
Ataxias Espinocerebelosas , Serina-Treonina Quinasas TOR , Humanos , Ratones , Animales , Proteína C9orf72 , Células HEK293 , Serina-Treonina Quinasas TOR/metabolismo , Ratones Transgénicos , Autofagia , ARN Mensajero , Sirolimus , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ARN/metabolismo
3.
Brain ; 146(10): 4217-4232, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37143315

RESUMEN

Myotonic dystrophy type 1 is a dominantly inherited multisystemic disease caused by CTG tandem repeat expansions in the DMPK 3' untranslated region. These expanded repeats are transcribed and produce toxic CUG RNAs that sequester and inhibit activities of the MBNL family of developmental RNA processing factors. Although myotonic dystrophy is classified as a muscular dystrophy, the brain is also severely affected by an unusual cohort of symptoms, including hypersomnia, executive dysfunction, as well as early onsets of tau/MAPT pathology and cerebral atrophy. To address the molecular and cellular events that lead to these pathological outcomes, we recently generated a mouse Dmpk CTG expansion knock-in model and identified choroid plexus epithelial cells as particularly affected by the expression of toxic CUG expansion RNAs. To determine if toxic CUG RNAs perturb choroid plexus functions, alternative splicing analysis was performed on lateral and hindbrain choroid plexi from Dmpk CTG knock-in mice. Choroid plexus transcriptome-wide changes were evaluated in Mbnl2 knockout mice, a developmental-onset model of myotonic dystrophy brain dysfunction. To determine if transcriptome changes also occurred in the human disease, we obtained post-mortem choroid plexus for RNA-seq from neurologically unaffected (two females, three males; ages 50-70 years) and myotonic dystrophy type 1 (one female, three males; ages 50-70 years) donors. To test that choroid plexus transcriptome alterations resulted in altered CSF composition, we obtained CSF via lumbar puncture from patients with myotonic dystrophy type 1 (five females, five males; ages 35-55 years) and non-myotonic dystrophy patients (three females, four males; ages 26-51 years), and western blot and osmolarity analyses were used to test CSF alterations predicted by choroid plexus transcriptome analysis. We determined that CUG RNA induced toxicity was more robust in the lateral choroid plexus of Dmpk CTG knock-in mice due to comparatively higher Dmpk and lower Mbnl RNA levels. Impaired transitions to adult splicing patterns during choroid plexus development were identified in Mbnl2 knockout mice, including mis-splicing previously found in Dmpk CTG knock-in mice. Whole transcriptome analysis of myotonic dystrophy type 1 choroid plexus revealed disease-associated RNA expression and mis-splicing events. Based on these RNA changes, predicted alterations in ion homeostasis, secretory output and CSF composition were confirmed by analysis of myotonic dystrophy type 1 CSF. Our results implicate choroid plexus spliceopathy and concomitant alterations in CSF homeostasis as an unappreciated contributor to myotonic dystrophy type 1 CNS pathogenesis.


Asunto(s)
Distrofia Miotónica , Humanos , Femenino , Ratones , Animales , Distrofia Miotónica/genética , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Empalme Alternativo , ARN/genética , Ratones Noqueados , Expansión de Repetición de Trinucleótido
4.
Hum Mol Genet ; 30(11): 1020-1029, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-33856033

RESUMEN

Repeat-associated non-ATG (RAN) proteins have been reported in 11 microsatellite expansion disorders but the factors that allow RAN translation to occur and the effects of different repeat motifs and alternative AUG-like initiation codons are unclear. We studied the mechanisms of RAN translation across myotonic dystrophy type 2 (DM2) expansion transcripts with (CCUG) or without (CAGG) efficient alternative AUG-like codons. To better understand how DM2 LPAC and QAGR RAN proteins are expressed, we generated a series of CRISPR/Cas9-edited HEK293T cell lines. We show that LPAC and QAGR RAN protein levels are reduced in protein kinase R (PKR)-/- and PKR-like endoplasmic reticulum kinase (PERK)-/- cells, with more substantial reductions of CAGG-encoded QAGR in PKR-/- cells. Experiments using mutant eIF2α-S51A HEK293T cells show that p-eIF2α is required for QAGR production. In contrast, LPAC levels were only partially reduced in these cells, suggesting that both non-AUG and close-cognate initiation occur across CCUG RNAs. Overexpression of the alternative initiation factor eIF2A increases LPAC and QAGR protein levels but, notably, has a much larger effect on QAGR expressed from CAGG-expansion RNAs that lack efficient close-cognate codons. The effects of eIF2A on increasing LPAC are consistent with previous reports that eIF2A affects CUG-initiation translation. The observation that eIF2A also increases QAGR proteins is novel because CAGG expansion transcripts do not contain CUG or similarly efficient close-cognate AUG-like codons. For QAGR but not LPAC, the eIF2A-dependent increases are not seen when p-eIF2α is blocked. These data highlight the differential regulation of DM2 RAN proteins and eIF2A as a potential therapeutic target for DM2 and other RAN diseases.


Asunto(s)
Factor 2 Eucariótico de Iniciación/genética , Distrofia Miotónica/genética , eIF-2 Quinasa/genética , Sistemas CRISPR-Cas/genética , Expansión de las Repeticiones de ADN/genética , Células HEK293 , Humanos , Repeticiones de Microsatélite/genética , Distrofia Miotónica/fisiopatología , Biosíntesis de Proteínas/genética
5.
Hum Mol Genet ; 29(24): 3900-3918, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33378537

RESUMEN

C9orf72 ALS/FTD patients show remarkable clinical heterogeneity, but the complex biology of the repeat expansion mutation has limited our understanding of the disease. BAC transgenic mice were used to better understand the molecular mechanisms and repeat length effects of C9orf72 ALS/FTD. Genetic analyses of these mice demonstrate that the BAC transgene and not integration site effects cause ALS/FTD phenotypes. Transcriptomic changes in cell proliferation, inflammation and neuronal pathways are found late in disease and alternative splicing changes provide early molecular markers that worsen with disease progression. Isogenic sublines of mice with 800, 500 or 50 G4C2 repeats generated from the single-copy C9-500 line show longer repeats result in earlier onset, increased disease penetrance and increased levels of RNA foci and dipeptide RAN protein aggregates. These data demonstrate G4C2 repeat length is an important driver of disease and identify alternative splicing changes as early biomarkers of C9orf72 ALS/FTD.


Asunto(s)
Empalme Alternativo , Esclerosis Amiotrófica Lateral/patología , Proteína C9orf72/metabolismo , Expansión de las Repeticiones de ADN , Modelos Animales de Enfermedad , Demencia Frontotemporal/patología , Penetrancia , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Proteína C9orf72/genética , Demencia Frontotemporal/etiología , Demencia Frontotemporal/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación , Fenotipo
6.
Hum Genet ; 142(12): 1747-1754, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37957369

RESUMEN

Machado-Joseph disease (MJD/SCA3) is the most frequent dominant ataxia worldwide. It is caused by a (CAG)n expansion. MJD has two major ancestral backgrounds: the Machado lineage, found mainly in Portuguese families; and the Joseph lineage, present in all five continents, probably originating in Asia. MJD has been described in a few African and African-American families, but here we report the first diagnosed in Sudan to our knowledge. The proband presented with gait ataxia at age 24; followed by muscle cramps and spasticity, and dysarthria, by age 26; he was wheel-chair bound at 29 years of age. His brother had gait problems from age 20 years and, by age 21, lost the ability to run, showed dysarthria and muscle cramps. To assess the mutational origin of this family, we genotyped 30 SNPs and 7 STRs flanking the ATXN3_CAG repeat in three siblings and the non-transmitting father. We compared the MJD haplotype segregating in the family with our cohort of MJD families from diverse populations. Unlike all other known families of African origin, the Machado lineage was observed in Sudan, being shared with 86 Portuguese, 2 Spanish and 2 North-American families. The STR-based haplotype of Sudanese patients, however, was distinct, being four steps (2 STR mutations and 2 recombinations) away from the founder haplotype shared by 47 families, all of Portuguese extraction. Based on the phylogenetic network constructed with all MJD families of the Machado lineage, we estimated a common ancestry at 3211 ± 693 years ago.


Asunto(s)
Enfermedad de Machado-Joseph , Masculino , Humanos , Adulto Joven , Adulto , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/diagnóstico , Portugal , Calambre Muscular , Disartria , Filogenia , África Oriental
7.
Proc Natl Acad Sci U S A ; 117(31): 18591-18599, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32690681

RESUMEN

Repeat associated non-AUG (RAN) translation is found in a growing number of microsatellite expansion diseases, but the mechanisms remain unclear. We show that RAN translation is highly regulated by the double-stranded RNA-dependent protein kinase (PKR). In cells, structured CAG, CCUG, CAGG, and G4C2 expansion RNAs activate PKR, which leads to increased levels of multiple RAN proteins. Blocking PKR using PKR-K296R, the TAR RNA binding protein or PKR-KO cells, reduces RAN protein levels. p-PKR is elevated in C9orf72 ALS/FTD human and mouse brains, and inhibiting PKR in C9orf72 BAC transgenic mice using AAV-PKR-K296R or the Food and Drug Administration (FDA)-approved drug metformin, decreases RAN proteins, and improves behavior and pathology. In summary, targeting PKR, including by use of metformin, is a promising therapeutic approach for C9orf72 ALS/FTD and other expansion diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteína C9orf72 , Metformina/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , eIF-2 Quinasa , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Modelos Animales de Enfermedad , Demencia Frontotemporal/metabolismo , Humanos , Ratones , Ratones Transgénicos , Repeticiones de Microsatélite/genética , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
8.
Curr Opin Neurol ; 34(5): 748-755, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34392299

RESUMEN

PURPOSE OF REVIEW: An intronic G4C2 expansion mutation in C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Although there are currently no treatments for this insidious, fatal disease, intense research has led to promising therapeutic strategies, which will be discussed here. RECENT FINDINGS: Therapeutic strategies for C9-ALS/FTD have primarily focused on reducing the toxic effects of mutant expansion RNAs or the dipeptide repeat proteins (DPRs). The pathogenic effects of G4C2 expansion transcripts have been targeted using approaches aimed at promoting their degradation, inhibiting nuclear export or silencing transcription. Other promising strategies include immunotherapy to reduce the DPRs themselves, reducing RAN translation, removing the repeats using DNA or RNA editing and manipulation of downstream disease-altered stress granule pathways. Finally, understanding the molecular triggers that lead to pheno-conversion may lead to opportunities that can delay symptomatic disease onset. SUMMARY: A large body of evidence implicates RAN-translated DPRs as a main driver of C9-ALS/FTD. Promising therapeutic strategies for these devastating diseases are being rapidly developed with several approaches already in or approaching clinical trials.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Proteína C9orf72/genética , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Demencia Frontotemporal/terapia , Humanos , Proteínas , Gránulos de Estrés
9.
Proc Natl Acad Sci U S A ; 115(16): 4234-4239, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29610297

RESUMEN

Expansions of simple sequence repeats, or microsatellites, have been linked to ∼30 neurological-neuromuscular diseases. While these expansions occur in coding and noncoding regions, microsatellite sequence and repeat length diversity is more prominent in introns with eight different trinucleotide to hexanucleotide repeats, causing hereditary diseases such as myotonic dystrophy type 2 (DM2), Fuchs endothelial corneal dystrophy (FECD), and C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Here, we test the hypothesis that these GC-rich intronic microsatellite expansions selectively trigger host intron retention (IR). Using DM2, FECD, and C9-ALS/FTD as examples, we demonstrate that retention is readily detectable in affected tissues and peripheral blood lymphocytes and conclude that IR screening constitutes a rapid and inexpensive biomarker for intronic repeat expansion disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Distrofia Endotelial de Fuchs/genética , Intrones/genética , Distrofia Miotónica/genética , Composición de Base , Biomarcadores , Humanos , Linfocitos/química , Músculo Esquelético/química , Miocardio/química , Especificidad de Órganos , Polimorfismo de Nucleótido Simple , Empalme del ARN , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Análisis de Matrices Tisulares
10.
J Biol Chem ; 293(42): 16127-16141, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30213863

RESUMEN

Microsatellite expansions cause more than 40 neurological disorders, including Huntington's disease, myotonic dystrophy, and C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). These repeat expansion mutations can produce repeat-associated non-ATG (RAN) proteins in all three reading frames, which accumulate in disease-relevant tissues. There has been considerable interest in RAN protein products and their downstream consequences, particularly for the dipeptide proteins found in C9ORF72 ALS/FTD. Understanding how RAN translation occurs, what cellular factors contribute to RAN protein accumulation, and how these proteins contribute to disease should lead to a better understanding of the basic mechanisms of gene expression and human disease.


Asunto(s)
Expansión de las Repeticiones de ADN , Repeticiones de Microsatélite , Enfermedades del Sistema Nervioso/genética , Esclerosis Amiotrófica Lateral/genética , Proteína C9orf72 , Dipéptidos , Demencia Frontotemporal/genética , Expresión Génica , Enfermedad de Huntington/genética , Mutación , Distrofia Miotónica/genética
11.
Lab Invest ; 99(7): 929-942, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30918326

RESUMEN

More than 40 different neurological diseases are caused by microsatellite repeat expansions. Since the discovery of repeat-associated non-AUG (RAN) translation by Zu et al. in 2011, nine expansion disorders have been identified as RAN-positive diseases. RAN proteins are translated from different types of nucleotide repeat expansions and can be produced from both sense and antisense transcripts. In some diseases, RAN proteins have been shown to accumulate in affected brain regions. Here we review the pathological and molecular aspects associated with RAN protein accumulation for each particular disorder, the correlation between disease pathology and the available in vivo models and the common aspects shared by some of the newly discovered RAN proteins.


Asunto(s)
Repeticiones de Microsatélite , Enfermedades del Sistema Nervioso/genética , Deficiencias en la Proteostasis/etiología , Animales , Expresión Génica , Humanos , Deficiencias en la Proteostasis/terapia
12.
Stem Cells ; 33(6): 1829-38, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25702800

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by expanded CTG repeats in the 3'-untranslated region (3' UTR) of the DMPK gene. Correcting the mutation in DM1 stem cells would be an important step toward autologous stem cell therapy. The objective of this study is to demonstrate in vitro genome editing to prevent production of toxic mutant transcripts and reverse phenotypes in DM1 stem cells. Genome editing was performed in DM1 neural stem cells (NSCs) derived from human DM1 induced pluripotent stem (iPS) cells. An editing cassette containing SV40/bGH polyA signals was integrated upstream of the CTG repeats by TALEN-mediated homologous recombination (HR). The expression of mutant CUG repeats transcript was monitored by nuclear RNA foci, the molecular hallmarks of DM1, using RNA fluorescence in situ hybridization. Alternative splicing of microtubule-associated protein tau (MAPT) and muscleblind-like (MBNL) proteins were analyzed to further monitor the phenotype reversal after genome modification. The cassette was successfully inserted into DMPK intron 9 and this genomic modification led to complete disappearance of nuclear RNA foci. MAPT and MBNL 1, 2 aberrant splicing in DM1 NSCs were reversed to normal pattern in genome-modified NSCs. Genome modification by integration of exogenous polyA signals upstream of the DMPK CTG repeat expansion prevents the production of toxic RNA and leads to phenotype reversal in human DM1 iPS-cells derived stem cells. Our data provide proof-of-principle evidence that genome modification may be used to generate genetically modified progenitor cells as a first step toward autologous cell transfer therapy for DM1.


Asunto(s)
Genoma Humano , Células Madre Pluripotentes Inducidas/citología , Proteínas Asociadas a Microtúbulos/metabolismo , Distrofia Miotónica/metabolismo , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Humanos , Distrofia Miotónica/patología , Fenotipo
13.
Proc Natl Acad Sci U S A ; 110(51): E4968-77, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24248382

RESUMEN

The finding that a GGGGCC (G4C2) hexanucleotide repeat expansion in the chromosome 9 ORF 72 (C9ORF72) gene is a common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) links ALS/FTD to a large group of unstable microsatellite diseases. Previously, we showed that microsatellite expansion mutations can be bidirectionally transcribed and that these mutations express unexpected proteins by a unique mechanism, repeat-associated non-ATG (RAN) translation. In this study, we show that C9ORF72 antisense transcripts are elevated in the brains of C9ORF72 expansion-positive [C9(+)] patients, and antisense GGCCCC (G2C4) repeat-expansion RNAs accumulate in nuclear foci in brain. Additionally, sense and antisense foci accumulate in blood and are potential biomarkers of the disease. Furthermore, we show that RAN translation occurs from both sense and antisense expansion transcripts, resulting in the expression of six RAN proteins (antisense: Pro-Arg, Pro-Ala, Gly-Pro; and sense: Gly-Ala, Gly-Arg, Gly-Pro). These proteins accumulate in cytoplasmic aggregates in affected brain regions, including the frontal and motor cortex, hippocampus, and spinal cord neurons, with some brain regions showing dramatic RAN protein accumulation and clustering. The finding that unique antisense G2C4 RNA foci and three unique antisense RAN proteins accumulate in patient tissues indicates that bidirectional transcription of expanded alleles is a fundamental pathologic feature of C9ORF72 ALS/FTD. Additionally, these findings suggest the need to test therapeutic strategies that target both sense and antisense RNAs and RAN proteins in C9ORF72 ALS/FTD, and to more broadly consider the role of antisense expression and RAN translation across microsatellite expansion diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/metabolismo , Demencia Frontotemporal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas/metabolismo , ARN sin Sentido/biosíntesis , Proteína de Unión al GTP ran/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Encéfalo/patología , Proteína C9orf72 , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Femenino , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Células HEK293 , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas/genética , ARN sin Sentido/genética , Secuencias Repetitivas de Ácidos Nucleicos , Proteína de Unión al GTP ran/genética
14.
J Neurosci ; 34(30): 9891-904, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25057192

RESUMEN

Spinocerebellar ataxia type 5 (SCA5), a dominant neurodegenerative disease characterized by profound Purkinje cell loss, is caused by mutations in SPTBN2, a gene that encodes ß-III spectrin. SCA5 is the first neurodegenerative disorder reported to be caused by mutations in a cytoskeletal spectrin gene. We have developed a mouse model to understand the mechanistic basis for this disease and show that expression of mutant but not wild-type ß-III spectrin causes progressive motor deficits and cerebellar degeneration. We show that endogenous ß-III spectrin interacts with the metabotropic glutamate receptor 1α (mGluR1α) and that mice expressing mutant ß-III spectrin have cerebellar dysfunction with altered mGluR1α localization at Purkinje cell dendritic spines, decreased mGluR1-mediated responses, and deficient mGluR1-mediated long-term potentiation. These results indicate that mutant ß-III spectrin causes mislocalization and dysfunction of mGluR1α at dendritic spines and connects SCA5 with other disorders involving glutamatergic dysfunction and synaptic plasticity abnormalities.


Asunto(s)
Modelos Animales de Enfermedad , Mutación/genética , Receptores de Glutamato Metabotrópico/análisis , Receptores de Glutamato Metabotrópico/genética , Espectrina/genética , Ataxias Espinocerebelosas/genética , Animales , Cerebelo/química , Cerebelo/patología , Espinas Dendríticas/química , Espinas Dendríticas/patología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Receptores de Glutamato Metabotrópico/metabolismo , Ataxias Espinocerebelosas/fisiopatología
15.
Hum Mol Genet ; 22(R1): R45-51, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23918658

RESUMEN

Well-established rules of translational initiation have been used as a cornerstone in molecular biology to understand gene expression and to frame fundamental questions on what proteins a cell synthesizes, how proteins work and to predict the consequences of mutations. For a group of neurological diseases caused by the abnormal expansion of short segments of DNA (e.g. CAG•CTG repeats), mutations within or outside of predicted coding and non-coding regions are thought to cause disease by protein gain- or loss-of-function or RNA gain-of-function mechanisms. In contrast to these predictions, the recent discovery of repeat-associated non-ATG (RAN) translation showed expansion mutations can express homopolymeric expansion proteins in all three reading frames without an AUG start codon. This unanticipated, non-canonical type of protein translation is length-and hairpin-dependent, takes place without frameshifting or RNA editing and occurs across a variety of repeat motifs. To date, RAN proteins have been reported in spinocerebellar ataxia type 8 (SCA8), myotonic dystrophy type 1 (DM1), fragile X tremor ataxia syndrome (FXTAS) and C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). In this article, we review what is currently known about RAN translation and recent progress toward understanding its contribution to disease.


Asunto(s)
Proteínas Mutantes/metabolismo , Enfermedades del Sistema Nervioso/genética , Iniciación de la Cadena Peptídica Traduccional , Biosíntesis de Proteínas , Expansión de Repetición de Trinucleótido , Proteína C9orf72 , Codón Iniciador , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Expresión Génica , Proteínas Mutantes/genética , Mutación , Proteína Quinasa de Distrofia Miotónica , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Sistemas de Lectura Abierta , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Edición de ARN
16.
Nat Genet ; 38(7): 758-69, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16804541

RESUMEN

We previously reported that a (CTG)n expansion causes spinocerebellar ataxia type 8 (SCA8), a slowly progressive ataxia with reduced penetrance. We now report a transgenic mouse model in which the full-length human SCA8 mutation is transcribed using its endogenous promoter. (CTG)116 expansion, but not (CTG)11 control lines, develop a progressive neurological phenotype with in vivo imaging showing reduced cerebellar-cortical inhibition. 1C2-positive intranuclear inclusions in cerebellar Purkinje and brainstem neurons in SCA8 expansion mice and human SCA8 autopsy tissue result from translation of a polyglutamine protein, encoded on a previously unidentified antiparallel transcript (ataxin 8, ATXN8) spanning the repeat in the CAG direction. The neurological phenotype in SCA8 BAC expansion but not BAC control lines demonstrates the pathogenicity of the (CTG-CAG)n expansion. Moreover, the expression of noncoding (CUG)n expansion transcripts (ataxin 8 opposite strand, ATXN8OS) and the discovery of intranuclear polyglutamine inclusions suggests SCA8 pathogenesis involves toxic gain-of-function mechanisms at both the protein and RNA levels.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Ataxias Espinocerebelosas/genética , Expansión de Repetición de Trinucleótido , Animales , Secuencia de Bases , Cromosomas Artificiales Bacterianos/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Péptidos/genética , Péptidos/metabolismo , Fenotipo , ARN Largo no Codificante , ARN no Traducido , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología
17.
Nat Genet ; 38(2): 184-90, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16429157

RESUMEN

We have discovered that beta-III spectrin (SPTBN2) mutations cause spinocerebellar ataxia type 5 (SCA5) in an 11-generation American kindred descended from President Lincoln's grandparents and two additional families. Two families have separate in-frame deletions of 39 and 15 bp, and a third family has a mutation in the actin/ARP1 binding region. Beta-III spectrin is highly expressed in Purkinje cells and has been shown to stabilize the glutamate transporter EAAT4 at the surface of the plasma membrane. We found marked differences in EAAT4 and GluRdelta2 by protein blot and cell fractionation in SCA5 autopsy tissue. Cell culture studies demonstrate that wild-type but not mutant beta-III spectrin stabilizes EAAT4 at the plasma membrane. Spectrin mutations are a previously unknown cause of ataxia and neurodegenerative disease that affect membrane proteins involved in glutamate signaling.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas del Tejido Nervioso/genética , Ataxias Espinocerebelosas/clasificación , Ataxias Espinocerebelosas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Estudios de Casos y Controles , Línea Celular , Cerebelo/patología , Niño , Mapeo Cromosómico , Proteínas del Citoesqueleto/química , Transportador 4 de Aminoácidos Excitadores/metabolismo , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Linaje , Espectrina
18.
Hum Mol Genet ; 21(21): 4645-54, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22846424

RESUMEN

Myotonic dystrophy (DM) is a multisystemic disease caused by CTG or CCTG expansion mutations. There is strong evidence that DM1 CUG and DM2 CCUG expansion transcripts sequester muscleblind-like (MBNL) proteins and that loss of MBNL function causes alternative splicing abnormalities that contribute to disease. Because MBNL1 loss is thought to play an important role in disease and localized AAV delivery of MBNL1 partially rescues skeletal muscle pathology in DM mice, there is strong interest in MBNL1 overexpression as a therapeutic strategy. We developed the first transgenic MBNL1 overexpression mouse model (MBNL1-OE) to test the safety and efficacy of multisystemic MBNL1 overexpression. First, we demonstrate that MBNL1 overexpression is generally well-tolerated in skeletal muscle. Second, we show the surprising result that premature shifts in alternative splicing of MBNL1-regulated genes in multiple organ systems are compatible with life and do not cause embryonic lethality. Third, we show for the first time that early and long-term MBNL1 overexpression prevents CUG-induced myotonia, myopathy and alternative splicing abnormalities in DM1 mice. In summary, MBNL1 overexpression may be a valuable strategy for treating the skeletal muscle features of DM.


Asunto(s)
Proteínas de Unión al ADN , Músculo Esquelético , Distrofia Miotónica , Proteínas de Unión al ARN , Empalme Alternativo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Miotónica/genética , Distrofia Miotónica/fisiopatología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(1): 260-5, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21173221

RESUMEN

Trinucleotide expansions cause disease by both protein- and RNA-mediated mechanisms. Unexpectedly, we discovered that CAG expansion constructs express homopolymeric polyglutamine, polyalanine, and polyserine proteins in the absence of an ATG start codon. This repeat-associated non-ATG translation (RAN translation) occurs across long, hairpin-forming repeats in transfected cells or when expansion constructs are integrated into the genome in lentiviral-transduced cells and brains. Additionally, we show that RAN translation across human spinocerebellar ataxia type 8 (SCA8) and myotonic dystrophy type 1 (DM1) CAG expansion transcripts results in the accumulation of SCA8 polyalanine and DM1 polyglutamine expansion proteins in previously established SCA8 and DM1 mouse models and human tissue. These results have implications for understanding fundamental mechanisms of gene expression. Moreover, these toxic, unexpected, homopolymeric proteins now should be considered in pathogenic models of microsatellite disorders.


Asunto(s)
Biosíntesis de Proteínas/genética , Ataxias Espinocerebelosas/genética , Expansión de Repetición de Trinucleótido/genética , Secuencia de Aminoácidos , Northern Blotting , Línea Celular , Clonación Molecular , Codón Iniciador/genética , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Lentivirus , Espectrometría de Masas , Datos de Secuencia Molecular , Mutagénesis , Distrofia Miotónica/genética , Péptidos/genética , Péptidos/metabolismo , Biosíntesis de Proteínas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
bioRxiv ; 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38948725

RESUMEN

Spinocerebellar Ataxia Type 8 (SCA8) is an inherited neurodegenerative disease caused by a bidirectionally expressed CTG●CAG expansion mutation in the ATXN-8 and ATXN8-OS genes. While primarily a motor disorder, psychiatric and cognitive symptoms have been reported. It is difficult to elucidate how the disease alters brain function in areas with little or no degeneration producing both motor and cognitive symptoms. Using transparent polymer skulls and CNS-wide GCaMP6f expression, we studied neocortical networks throughout SCA8 progression using wide-field Ca2+ imaging in a transgenic mouse model of SCA8. We observed that neocortical networks in SCA8+ mice were hyperconnected globally which led to network configurations with increased global efficiency and centrality. At the regional level, significant network changes occurred in nearly all cortical regions, however mainly involved sensory and association cortices. Changes in functional connectivity in anterior motor regions worsened later in the disease. Near perfect decoding of animal genotype was obtained using a generalized linear model based on canonical correlation strengths between activity in cortical regions. The major contributors to decoding were concentrated in the somatosensory, higher visual and retrosplenial cortices and occasionally extended into the motor regions, demonstrating that the areas with the largest network changes are predictive of disease state.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA