Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurol ; 269(8): 4154-4160, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35244767

RESUMO

OBJECTIVE: Dysphagia can be troublesome in sporadic inclusion body myositis (sIBM) and oculopharyngeal muscular dystrophy (OPMD), but no established treatment exists. Cricopharyngeal muscle botulinum toxin injection has at case level been reported to be effective. We evaluated safety and efficacy of botulinum toxin injections in the cricopharyngeal muscle in patients with dysphagia due to sIBM or OPMD. METHODS: Participants were included from our outpatient clinic. Cricopharyngeal constriction was confirmed by laryngoscopy. After EMG confirmation of needle placement in the cricopharyngeal muscle, botulinum toxin A was injected in awake patients. An individualized dose of 5-10 units of botulinum toxin A was applied initially and titrated up a maximum of 3 times. Outcome measures were change in dysphagia questionnaire, timed cold-water swallow test and subjective dysphagia status (worse, unchanged, improved). Due to the need for individualized dosing and a limited number of available patients, an uncontrolled, un-blinded design was used. RESULTS: Thirteen patients, 3 with OPMD, received at least 1 injection. In the dysphagia questionnaire, all but 2 subjects, none with subjective worsening, improved (p < 0.001). Subjectively, seven felt an improvement, 4 no change and 2 a worsening. No overall change was seen the timed cold-water swallow test. No serious adverse events were observed. CONCLUSION: Botulinum toxin injection of the cricopharyngeal muscle in patients with OPMD and sIBM had a beneficial effect on dysphagia in most of the treated patients. Two of 13 patients experienced a temporary worsening not reflected in dysphagia score. Limitations are the un-blinded and un-randomized design and subjective assessments methods. PROSPECTIVE TRIAL REGISTRATION: EudraCT-number: 2014-002210-23.


Assuntos
Toxinas Botulínicas Tipo A , Transtornos de Deglutição , Distrofia Muscular Oculofaríngea , Miosite de Corpos de Inclusão , Fármacos Neuromusculares , Toxinas Botulínicas Tipo A/efeitos adversos , Toxinas Botulínicas Tipo A/uso terapêutico , Transtornos de Deglutição/tratamento farmacológico , Transtornos de Deglutição/etiologia , Humanos , Distrofia Muscular Oculofaríngea/complicações , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Miosite de Corpos de Inclusão/complicações , Miosite de Corpos de Inclusão/tratamento farmacológico , Fármacos Neuromusculares/efeitos adversos , Fármacos Neuromusculares/uso terapêutico , Estudos Prospectivos , Água
2.
PLoS Genet ; 18(1): e1010015, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35025870

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset disorder characterized by progressive weakness and degeneration of specific muscles. OPMD is due to extension of a polyalanine tract in poly(A) binding protein nuclear 1 (PABPN1). Aggregation of the mutant protein in muscle nuclei is a hallmark of the disease. Previous transcriptomic analyses revealed the consistent deregulation of the ubiquitin-proteasome system (UPS) in OPMD animal models and patients, suggesting a role of this deregulation in OPMD pathogenesis. Subsequent studies proposed that UPS contribution to OPMD involved PABPN1 aggregation. Here, we use a Drosophila model of OPMD to address the functional importance of UPS deregulation in OPMD. Through genome-wide and targeted genetic screens we identify a large number of UPS components that are involved in OPMD. Half dosage of UPS genes reduces OPMD muscle defects suggesting a pathological increase of UPS activity in the disease. Quantification of proteasome activity confirms stronger activity in OPMD muscles, associated with degradation of myofibrillar proteins. Importantly, improvement of muscle structure and function in the presence of UPS mutants does not correlate with the levels of PABPN1 aggregation, but is linked to decreased degradation of muscle proteins. Oral treatment with the proteasome inhibitor MG132 is beneficial to the OPMD Drosophila model, improving muscle function although PABPN1 aggregation is enhanced. This functional study reveals the importance of increased UPS activity that underlies muscle atrophy in OPMD. It also provides a proof-of-concept that inhibitors of proteasome activity might be an attractive pharmacological approach for OPMD.


Assuntos
Atrofia Muscular/patologia , Distrofia Muscular Oculofaríngea/patologia , Proteína I de Ligação a Poli(A)/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Modelos Animais de Doenças , Drosophila melanogaster , Regulação da Expressão Gênica , Testes Genéticos , Humanos , Leupeptinas/farmacologia , Leupeptinas/uso terapêutico , Atrofia Muscular/tratamento farmacológico , Atrofia Muscular/metabolismo , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Mutação , Proteína I de Ligação a Poli(A)/química , Estudo de Prova de Conceito , Agregados Proteicos/efeitos dos fármacos
3.
Hum Mol Genet ; 28(10): 1694-1708, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30649389

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a rare late onset genetic disease leading to ptosis, dysphagia and proximal limb muscles at later stages. A short abnormal (GCN) triplet expansion in the polyA-binding protein nuclear 1 (PABPN1) gene leads to PABPN1-containing aggregates in the muscles of OPMD patients. Here we demonstrate that treating mice with guanabenz acetate (GA), an FDA-approved antihypertensive drug, reduces the size and number of nuclear aggregates, improves muscle force, protects myofibers from the pathology-derived turnover and decreases fibrosis. GA targets various cell processes, including the unfolded protein response (UPR), which acts to attenuate endoplasmic reticulum (ER) stress. We demonstrate that GA increases both the phosphorylation of the eukaryotic translation initiation factor 2α subunit and the splicing of Xbp1, key components of the UPR. Altogether these data show that modulation of protein folding regulation is beneficial for OPMD and promote the further development of GA or its derivatives for treatment of OPMD in humans. Furthermore, they support the recent evidences that treating ER stress could be therapeutically relevant in other more common proteinopathies.


Assuntos
Guanabenzo/farmacologia , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Proteína I de Ligação a Poli(A)/genética , Proteína 1 de Ligação a X-Box/genética , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Animais , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fibrose/tratamento farmacológico , Fibrose/genética , Fibrose/patologia , Humanos , Camundongos , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patologia , Fosforilação/efeitos dos fármacos , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/genética , Dobramento de Proteína , Resposta a Proteínas não Dobradas/efeitos dos fármacos
4.
Orbit ; 38(6): 511-513, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30688154

RESUMO

Oculopharyngeal Muscular Dystrophy (OPMD) is a systemic progressive autosomal dominant myopathy which results in ptosis due to levator weakness. Surgical correction can be complicated by corneal exposure and a non-surgical alternative, such as ptosis props, can be uncomfortable in patients with preserved orbicularis function. We describe a case of a 57-year-old gentleman with OPMD, who declined surgical intervention, and self-manages his ptosis with cosmetic glue.


Assuntos
Blefaroptose/tratamento farmacológico , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Músculos Oculomotores/efeitos dos fármacos , Adesivos Teciduais/uso terapêutico , Técnicas Cosméticas , Humanos , Masculino , Pessoa de Meia-Idade
5.
Hum Gene Ther ; 26(5): 286-92, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25860803

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a muscle-specific, late-onset degenerative disorder whereby muscles of the eyes (causing ptosis), throat (leading to dysphagia), and limbs (causing proximal limb weakness) are mostly affected. The disease is characterized by a mutation in the poly(A)-binding protein nuclear-1 (PABPN1) gene, resulting in a short GCG expansion in the polyalanine tract of PABPN1 protein. Accumulation of filamentous intranuclear inclusions in affected skeletal muscle cells constitutes the pathological hallmark of OPMD. This review highlights the current translational research advances in the treatment of OPMD. In vitro and in vivo disease models are described. Conventional and experimental therapeutic approaches are discussed with emphasis on novel molecular therapies including the use of intrabodies, gene therapy, and myoblast transfer therapy.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/terapia , Animais , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Corpos de Inclusão Intranuclear , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Distrofia Muscular Oculofaríngea/cirurgia , Mutação , Proteína I de Ligação a Poli(A)/genética
8.
Muscle Nerve ; 49(4): 601-3, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24259282

RESUMO

INTRODUCTION: Despite multiple studies reporting marked benefit of botulinum toxin (BTX) for treatment of cricopharyngeal dysphagia, little is known about its safety for this indication. We examined the safety of cricopharyngeal BTX for dysphagia in oculopharyngeal muscular dystrophy (OPMD). METHODS: We reviewed records of patients with OPMD who received cricopharyngeal BTX. RESULTS: Twenty-four patients underwent 66 procedures. Overall adverse event frequency was 44%. The most common adverse events were dysphonia (24%) and worsened dysphagia (14%). Logistic regression demonstrated that dose was a significant predictor of worsened dysphagia (P = 0.036) and of the composite event of dysphonia or worsened dysphagia (P = 0.009). There was a nonsignificant trend for dose as a predictor of dysphonia (P = 0.073). 59% of procedures were associated with symptomatic improvement. CONCLUSIONS: While BTX appears to be beneficial for treatment of dysphagia in OPMD, caution is warranted when injecting the cricopharyngeus muscle due to dose-related risk of dysphonia or worsened dysphagia.


Assuntos
Toxinas Botulínicas/administração & dosagem , Toxinas Botulínicas/efeitos adversos , Transtornos de Deglutição/tratamento farmacológico , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Idoso , Transtornos de Deglutição/epidemiologia , Disfonia/induzido quimicamente , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Muscular Oculofaríngea/epidemiologia , Resultado do Tratamento
9.
Cell Death Dis ; 4: e821, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24091664

RESUMO

Expansion of polyalanine tracts causes at least nine inherited human diseases. Among these, a polyalanine tract expansion in the poly (A)-binding protein nuclear 1 (expPABPN1) causes oculopharyngeal muscular dystrophy (OPMD). So far, there is no treatment for OPMD patients. Developing drugs that efficiently sustain muscle protection by activating key cell survival mechanisms is a major challenge in OPMD research. Proteins that belong to the Wnt family are known for their role in both human development and adult tissue homeostasis. A hallmark of the Wnt signaling pathway is the increased expression of its central effector, beta-catenin (ß-catenin) by inhibiting one of its upstream effector, glycogen synthase kinase (GSK)3ß. Here, we explored a pharmacological manipulation of a Wnt signaling pathway using lithium chloride (LiCl), a GSK-3ß inhibitor, and observed the enhanced expression of ß-catenin protein as well as the decreased cell death normally observed in an OPMD cell model of murine myoblast (C2C12) expressing the expanded and pathogenic form of the expPABPN1. Furthermore, this effect was also observed in primary cultures of mouse myoblasts expressing expPABPN1. A similar effect on ß-catenin was also observed when lymphoblastoid cells lines (LCLs) derived from OPMD patients were treated with LiCl. We believe manipulation of the Wnt/ß-catenin signaling pathway may represent an effective route for the development of future therapy for patients with OPMD.


Assuntos
Cloreto de Lítio/farmacologia , Cloreto de Lítio/uso terapêutico , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Distrofia Muscular Oculofaríngea/patologia , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteínas de Fluorescência Verde/metabolismo , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Distrofia Muscular Oculofaríngea/metabolismo , Proteínas Mutantes/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Mioblastos/patologia , Proteína I de Ligação a Poli(A)/genética , Proteína I de Ligação a Poli(A)/metabolismo , Transporte Proteico/efeitos dos fármacos , Expansão das Repetições de Trinucleotídeos/genética , beta Catenina/metabolismo
10.
Drug Discov Today Technol ; 10(1): e103-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24050237

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a late onset disease which affects specific muscles. No pharmacological treatments are currently available for OPMD. In recent years, genetically tractable models of OPMD ­ Drosophila and Caenorhabditis elegans ­ have been generated. Although these models have not yet been used for large-scale primary drug screening, they have been very useful in candidate approaches for the identification of potential therapeutic compounds for OPMD. In this brief review, we summarize the data that validated active molecules for OPMD in animal models including Drosophila, C. elegans and mouse.


Assuntos
Modelos Animais de Doenças , Descoberta de Drogas , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Animais , Humanos
11.
EMBO Mol Med ; 3(1): 35-49, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21204267

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset syndrome characterized by progressive degeneration of specific muscles. OPMD is caused by extension of a polyalanine tract in poly(A) binding protein nuclear 1 (PABPN1). Insoluble nuclear inclusions form in diseased muscles. We have generated a Drosophila model of OPMD that recapitulates the features of the disorder. Here, we show that the antiprion drugs 6-aminophenanthridine (6AP) and guanabenz acetate (GA), which prevent formation of amyloid fibers by prion proteins in cell models, alleviate OPMD phenotypes in Drosophila, including muscle degeneration and nuclear inclusion formation. The large ribosomal RNA and its activity in protein folding were recently identified as a specific cellular target of 6AP and GA. We show that deletions of the ribosomal DNA locus reduce OPMD phenotypes and act synergistically with sub-effective doses of 6AP. In a complementary approach, we demonstrate that ribosomal RNA accelerates in vitro fibril formation of PABPN1 N-terminal domain. These results reveal the conserved role of ribosomal RNA in different protein aggregation disorders and identify 6AP and GA as general anti-aggregation molecules.


Assuntos
Guanabenzo/uso terapêutico , Distrofia Muscular Oculofaríngea/metabolismo , Fenantridinas/uso terapêutico , Proteína II de Ligação a Poli(A)/metabolismo , Animais , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Larva/metabolismo , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Fenótipo , Doenças Priônicas/tratamento farmacológico , Dobramento de Proteína , RNA Ribossômico/metabolismo
12.
Sci Transl Med ; 2(34): 34ra40, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20519718

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is caused by a trinucleotide repeat expansion mutation in the coding region of the gene encoding PABPN1 (polyadenylate-binding protein nuclear 1). Mutant PABPN1 with a polyalanine tract expansion forms aggregates within the nuclei of skeletal muscle fibers. There is currently no effective treatment. We have developed cell and mouse models of OPMD and have identified the aggregation of mutant PABPN1 and apoptosis as therapeutic targets. Here, we show that transglutaminase activity is increased in muscle from OPMD model mice. Elevated transglutaminase 2 expression enhances, whereas TG2 knockdown suppresses, the toxicity and aggregation of mutant PABPN1 in cells. Cystamine protects against the toxicity of mutant PABPN1 and exerts its effect via the inhibition of transglutaminase 2, as cystamine treatment is unable to further reduce the protective effect of transglutaminase 2 knockdown on mutant PABPN1 toxicity in cells. Cystamine also reduces the aggregation and toxicity of mutant PABPN1 in human cells. In a mouse model of OPMD, cystamine treatment reduced the elevated transglutaminase activity, attenuated muscle weakness, reduced aggregate load, and decreased apoptotic markers in muscle. Therefore, inhibitors of transglutaminase 2 should be considered as possible therapeutics for OPMD.


Assuntos
Cistamina/uso terapêutico , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Distrofia Muscular Oculofaríngea/genética , Peptídeos/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Linhagem Celular , Cistamina/farmacologia , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Debilidade Muscular/tratamento farmacológico , Distrofia Muscular Oculofaríngea/enzimologia , Distrofia Muscular Oculofaríngea/patologia , Transglutaminases/antagonistas & inibidores , Transglutaminases/metabolismo , Expansão das Repetições de Trinucleotídeos/efeitos dos fármacos
13.
Trends Mol Med ; 12(6): 279-86, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16650805

RESUMO

Although genetic mutations that are responsible for most of the inherited neuromuscular diseases have been identified, the molecular and cellular mechanisms that cause muscle and nerve depletion are not well understood and therapies are lacking. Histological studies of many neuromuscular diseases indicated that loss of motor-nerve and/or skeletal-muscle function might be due to excessive cell death by apoptosis. Recent studies have confirmed this possibility by showing that pathology in mouse models of amyotrophic lateral sclerosis, congenital muscular dystrophy, oculopharyngeal muscular dystrophy and collagen-VI deficiency, but not Duchenne muscular dystrophy, is significantly ameliorated by genetic or pharmacological interventions that have been designed to inhibit apoptosis. Thus, apoptosis greatly contributes to pathology in mouse models of several neuromuscular diseases, and appropriate anti-apoptosis therapy might therefore be beneficial for the corresponding human diseases.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Antibacterianos/uso terapêutico , Apoptose/genética , Doxiciclina/uso terapêutico , Terapia Genética , Minociclina/uso terapêutico , Distrofias Musculares/terapia , Agrina/genética , Agrina/metabolismo , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/genética , Animais , Antibacterianos/farmacologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética/métodos , Humanos , Laminina/genética , Laminina/metabolismo , Camundongos , Minociclina/farmacologia , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Mutação , Proteína II de Ligação a Poli(A)/genética , Proteína II de Ligação a Poli(A)/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
14.
Int J Biochem Cell Biol ; 38(9): 1457-62, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16530457

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset, autosomal dominant disease caused by the abnormal expansion of a polyalanine tract within the coding region of poly(A) binding protein nuclear 1 (PABPN1). The resultant mutant PABPN1 forms aggregates within the nuclei of skeletal muscle fibres. The mechanism by which the polyalanine expansion mutation in PABN1 causes disease is unclear. However, the mutation is thought to confer a toxic gain-of-function on the protein. Despite controversy over the role of aggregates, it has been consistently shown that agents that reduce aggregate load in cell models of OPMD also reduce levels of cell death. Recently generated animal models of OPMD will help elucidate the mechanism of disease and allow the trial of potential therapeutics. Indeed, administration of known anti-aggregation drugs attenuated muscle weakness in an OPMD mouse model. This suggests that anti-aggregation therapies may be beneficial in OPMD.


Assuntos
Distrofia Muscular Oculofaríngea/tratamento farmacológico , Proteína II de Ligação a Poli(A)/genética , Animais , Modelos Animais de Doenças , Doxiciclina/uso terapêutico , Humanos , Corpos de Inclusão/metabolismo , Distrofia Muscular Oculofaríngea/genética , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Trealose/uso terapêutico , Repetições de Trinucleotídeos
15.
Hum Mol Genet ; 15(1): 105-11, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16319127

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) belongs to the group of protein aggregation disorders and is caused by extensions of the N-terminal polyalanine stretch of the nuclear polyA-binding protein 1 (PABPN1). The presence of PABPN1-containing intranuclear aggregates in skeletal muscle is unique for OPMD and is also observed in transgenic mouse and cell models for OPMD. These models consistently support a direct role for the protein aggregation in OPMD pathogenesis. We have isolated and characterized a diverse panel of single-domain antibody reagents (VHH), recognizing different epitopes in PABPN1. The antibody reagents specifically detect endogenous PABPN1 in cell lysates on western blot and label PABPN1 in cultured cells and muscle sections. When expressed intracellularly as intrabodies in a cellular model for OPMD, aggregation of PABPN1 was prevented in a dose-dependent manner. More importantly yet, these intrabodies could also reduce the presence of already existing aggregates. Given the domain specificity of VHH-mediated aggregation interference, this approach at least allows the definition of the nucleation kernel in aggregation-prone proteins, thus facilitating etiological insight into this and other protein aggregation disorders, and ultimately, it may well provide useful therapeutic agents.


Assuntos
Anticorpos/metabolismo , Corpos de Inclusão/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Proteína II de Ligação a Poli(A)/genética , Animais , Anticorpos/uso terapêutico , Western Blotting , Células COS , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Mapeamento de Epitopos , Células HeLa , Humanos , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patologia
16.
Hum Mol Genet ; 15(1): 23-31, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16311254

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant disease that presents in the fifth or sixth decade with dysphagia, ptosis and proximal limb weakness. OPMD is caused by the abnormal expansion of a polyalanine tract within the coding region of polyA binding protein nuclear 1 (PABPN1). The resultant mutant PABPN1 forms aggregates within the nuclei of skeletal muscle fibres. We have previously described a transgenic mouse model of OPMD that recapitulates the human disease and develops progressive muscle weakness accompanied by the formation of aggregates in skeletal muscle nuclei. The chemical chaperone trehalose has been used effectively to alleviate symptoms in a mouse model of Huntington's disease and is thought to elicit its effect by binding and stabilizing partially folded polyglutamine proteins and inhibiting the formation of aggregates. Here, we show that trehalose reduces aggregate formation and toxicity of mutant PABPN1 in cell models. Furthermore, oral administration of trehalose attenuated muscle weakness, reduced aggregate formation and decreased the number of TUNEL-labelled nuclei in skeletal muscle in an OPMD transgenic mouse model. Thus, anti-aggregation therapy may prove effective in the treatment of human OPMD.


Assuntos
Corpos de Inclusão/efeitos dos fármacos , Músculo Esquelético/patologia , Distrofia Muscular Oculofaríngea/patologia , Proteína II de Ligação a Poli(A)/metabolismo , Trealose/uso terapêutico , Análise de Variância , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Contração Muscular/fisiologia , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Distrofia Muscular Oculofaríngea/genética , Mutação/genética , Proteína II de Ligação a Poli(A)/genética , Trealose/farmacologia
17.
Nat Med ; 11(6): 672-7, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15864313

RESUMO

The muscular dystrophies are a heterogeneous group of disorders for which there are currently no cures. Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant late-onset, progressive disease that generally presents in the fifth or sixth decade with dysphagia, ptosis and proximal limb weakness. OPMD is caused by the abnormal expansion of a (GCG)n trinucleotide repeat in the coding region of the poly-(A) binding protein nuclear 1 (PABPN1) gene. In unaffected individuals, (GCG)6 codes for the first six alanines in a homopolymeric stretch of ten alanines. In most individuals with OPMD this (GCG)6 repeat is expanded to (GCG)8-13, leading to a stretch of 12-17 alanines in mutant PABPN1. PABPN1 with an expanded polyalanine tract forms aggregates consisting of tubular filaments within the nuclei of skeletal muscle fibers. We have developed a transgenic mouse model of OPMD that manifests progressive muscle weakness accompanied by intranuclear aggregates and TUNEL-stained nuclei in skeletal muscle fibers. The onset and severity of these abnormalities were substantially delayed and attenuated by doxycycline treatment, which may exert its therapeutic effect by reducing aggregates and by distinct antiapoptotic properties. Doxycycline may represent a safe and feasible therapeutic for this disease.


Assuntos
Doxiciclina/farmacologia , Distrofia Muscular Oculofaríngea/tratamento farmacológico , Animais , Morte Celular/efeitos dos fármacos , Genótipo , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/patologia , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patologia , Mutação , Proteína II de Ligação a Poli(A)/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...