Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Pediatr Blood Cancer ; 68(5): e28926, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33533561

RESUMEN

BACKGROUND: Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is a progressive autosomal recessive disorder characterized by cachexia, gastrointestinal (GI) dysmotility, ptosis, peripheral neuropathy, and brain magnetic resonance imaging (MRI) white matter changes. Bi-allelic TYMP mutations lead to deficient thymidine phosphorylase (TP) activity, toxic accumulation of plasma nucleosides (thymidine and deoxyuridine), nucleotide pool imbalances, and mitochondrial DNA (mtDNA) instability. Death is mainly due to GI complications: intestinal perforation, peritonitis, and/or liver failure. Based on our previous observations in three patients with MNGIE that platelet infusions resulted in a transient 40% reduction of plasma nucleoside levels, in 2005 we performed the first hematopoietic stem cell transplantation (HSCT) worldwide as a life-long source of TP in a patient with MNGIE. PROCEDURE: HSCT was performed in a total of six patients with MNGIE. The multiple factors involved in the prognosis of this cohort were analyzed and compared to the literature experience. RESULTS: Cell source was bone marrow in five patients and peripheral stem cells in one, all from fully human leukocyte antigen (HLA)-matched related donors, including four who were TYMP mutation carriers. Four of six (66%) survived compared to the 37% survival rate in the literature. Reduced intensity conditioning regimen contributed to secondary graft failure in two patients. Fifteen years post HSCT, the first transplanted patient is seemingly cured. Severe GI symptoms before transplantation were mostly irreversible and were poor prognostic factors. CONCLUSIONS: Allogenic HSCT could constitute a curative therapeutic option for carefully selected, young, presymptomatic, or mildly affected patients. Timing, donor selection, and optimal conditioning protocol are major determinants of outcome. HSCT is inadvisable in patients with advanced MNGIE disease.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Seudoobstrucción Intestinal/terapia , Distrofia Muscular Oculofaríngea/terapia , Oftalmoplejía/congénito , Adolescente , Adulto , Niño , Estudios de Cohortes , Femenino , Humanos , Masculino , Oftalmoplejía/terapia , Linaje , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
2.
EBioMedicine ; 62: 103133, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33232869

RESUMEN

BACKGROUND: Preclinical studies have shown that gene therapy is a feasible approach to treat mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). However, the genetic murine model of the disease (Tymp/Upp1 double knockout, dKO) has a limited functional phenotype beyond the metabolic imbalances, and so the studies showing efficacy of gene therapy have relied almost exclusively on demonstrating correction of the biochemical phenotype. Chronic oral administration of thymidine (dThd) and deoxyuridine (dUrd) to dKO mice deteriorates the phenotype of the animals, providing a better model to test therapy approaches. METHODS: dKO mice were treated with both dThd and dUrd in drinking water from weaning until the end of the study. At 8 - 11 weeks of age, mice were treated with several doses of adeno-associated virus (AAV) serotype 8 vector carrying the human TYMP coding sequence under the control of different liver-specific promoters (TBG, AAT, or HLP). The biochemical profile and functional phenotype were studied over the life of the animals. FINDINGS: Nucleoside exposure resulted in 30-fold higher plasma nucleoside levels in dKO mice compared with non-exposed wild type mice. AAV-treatment provided elevated TP activity in liver and lowered systemic nucleoside levels in exposed dKO mice. Exposed dKO mice had enlarged brain ventricles (assessed by magnetic resonance imaging) and motor impairment (rotarod test); both were prevented by AAV treatment. Among all promoters tested, AAT showed the best efficacy. INTERPRETATION: Our results show that AAV-mediated gene therapy restores the biochemical homeostasis in the murine model of MNGIE and, for the first time, demonstrate that this treatment improves the functional phenotype. FUNDING: This work was funded in part by the Spanish Instituto de Salud Carlos III, and the Generalitat de Catalunya. The disclosed funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Seudoobstrucción Intestinal/genética , Seudoobstrucción Intestinal/terapia , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/terapia , Nucleósidos/farmacología , Oftalmoplejía/congénito , Animales , Terapia Combinada , Modelos Animales de Enfermedad , Activación Enzimática , Dosificación de Gen , Expresión Génica , Terapia Genética/métodos , Humanos , Hígado/metabolismo , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/terapia , Oftalmoplejía/genética , Oftalmoplejía/terapia , Fenotipo , Timidina Fosforilasa/genética , Resultado del Tratamiento
3.
Muscle Nerve ; 60(6): 724-731, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31531865

RESUMEN

INTRODUCTION: There is currently little evidence regarding oculopharyngeal muscular dystrophy (OPMD) disease burden reported by patients. In this study we aim to elicit direct patient input regarding OPMD disease burden. METHODS: We conducted semistructured interviews with 25 participants with genetically confirmed OPMD and a wide range of disease duration (15 ± 8 years). Using the Framework Technique, themes and categories were then extracted. RESULTS: Analyses revealed 7 themes (physical impact, mental impact, social impact, perception of progression, treatment perceptions, coping strategies, and access to disease information), encompassing 27 categories of OPMD disease burden. The most frequent categories were related to dysphagia, coping strategies for dysphagia, and impaired mobility. DISCUSSION: This study demonstrates the importance of considering, when providing clinical care, the broad range of coping strategies patients use to deal with OPMD symptoms, especially dysphagia, to properly assess limitations and monitor real disease progression.


Asunto(s)
Distrofia Muscular Oculofaríngea/fisiopatología , Distrofia Muscular Oculofaríngea/psicología , Acceso a la Información , Adaptación Psicológica , Adolescente , Adulto , Anciano , Actitud Frente a la Salud , Trastorno Dismórfico Corporal , Niño , Costo de Enfermedad , Trastornos de Deglución/etiología , Trastornos de Deglución/fisiopatología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Limitación de la Movilidad , Debilidad Muscular/etiología , Debilidad Muscular/fisiopatología , Distrofia Muscular Oculofaríngea/complicaciones , Distrofia Muscular Oculofaríngea/terapia , Dolor/etiología , Dolor/fisiopatología , Medición de Resultados Informados por el Paciente , Distrés Psicológico , Investigación Cualitativa , Participación Social , Trastornos de la Voz/etiología , Trastornos de la Voz/fisiopatología , Rendimiento Laboral , Adulto Joven
4.
Hum Mol Genet ; 28(19): 3301-3308, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31294444

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is a rare autosomal dominant late-onset muscular dystrophy affecting approximately 1:100 000 individuals in Europe. OPMD is mainly characterized by progressive eyelid drooping (ptosis) and dysphagia although muscles of the limbs can also be affected late in life. This muscle disease is due to a trinucleotide repeat expansion in the polyA-binding protein nuclear-1 gene. Patients express a protein with an 11-18 alanine tract that is misfolded and prone to form intranuclear inclusions, which are the hallmark of the disease. Other features of OPMD include muscle fibrosis and atrophy in affected muscles. Currently, no pharmacological treatments are available, and OPMD patients can only be referred to surgeons for cricopharyngeal myotomy or corrective surgery of extraocular muscles to ease ptosis. We recently tested a two-AAV `silence' and `replace' vector-based gene therapy treatment in a mouse model of OPMD. We demonstrate here that this gene therapy approach can revert already established insoluble aggregates and partially rescues the muscle from atrophy, which are both crucially important since in most cases OPMD patients already have an established disease when diagnosed. This strategy also prevents the formation of muscle fibrosis and stabilizes the muscle strength to the level of healthy muscles. Furthermore, we show here that similar results can be obtained using a single AAV vector incorporating both the `silence' and `replace' cassettes. These results further support the application of a gene therapy approach as a novel treatment for OPMD in humans.


Asunto(s)
Dependovirus/genética , Cuerpos de Inclusión Intranucleares/metabolismo , Distrofia Muscular Oculofaríngea/terapia , Proteína I de Unión a Poli(A)/genética , Proteína I de Unión a Poli(A)/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Humanos , Ratones , Ratones Transgénicos , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Expansión de Repetición de Trinucleótido
5.
Neurol Sci ; 40(4): 661-669, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30847674

RESUMEN

Recent advances in pathophysiological and genetic mechanisms of some neuromuscular diseases and a rapid progress in new pharmacological technologies led to an accelerated development of innovative treatments, generating an unexpected therapeutic revolution. In part 1, we report already commercially available drugs, just approved drugs and new therapeutic promises in the treatment of peripheral neuropathies. Hereditary transthyretin amyloidosis (hATTR) is a devastating disease due to amyloid accumulation in peripheral nerves, heart and autonomic system. The first specific drug approved for hATTR was tafamidis, a TTR tetramer stabilizer. In 2018, the positive results of two phase 3 trials have been reported leading to start of regulatory approval route for inotersen, an antisense oligonucleotide and patisiran, the first-ever RNA interference (RNAi) therapeutic. System biology targeting approach has indicated baclofen, naltrexone and sorbitol in combination (PXT3003) as candidate drugs for Charcot-Marie-Tooth disease type 1A. This hypothesis was confirmed in experimental models and in phase 2 and 3 clinical trials. Givosiran, another RNAi therapeutic, targeting 5-aminolevulinic acid synthase, has been positively tested in acute intermittent porphyria in phase 1/2 and ongoing phase 3 trials. Although allogenic hematopoietic stem cell transplantation resulted recently a long-term therapy in mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), a new strategy is liver transplantation which is able to revert the severe biochemical and clinical imbalance of the disease. Recently, a gene therapy has been tested in a MNGIE murine model, indicating that it may become a new therapeutic option.


Asunto(s)
Neuropatías Amiloides Familiares/terapia , Enfermedad de Charcot-Marie-Tooth/terapia , Seudoobstrucción Intestinal/terapia , Distrofia Muscular Oculofaríngea/terapia , Porfiria Intermitente Aguda/terapia , Tratamiento con ARN de Interferencia/métodos , Humanos , Oftalmoplejía/congénito
7.
J Gastrointestin Liver Dis ; 27(3): 321-325, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30240477

RESUMEN

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is a rare autosomal recessive disease caused by a mutation in the TYMP gene encoding thymidine phosphorylase. MNGIE causes gastrointestinal and neurological symptoms in homozygous individuals and is often misdiagnosed as anorexia nervosa, inflammatory bowel disease, or celiac disease. We present the case of a 26-year-old female with MNGIE, who was initially diagnosed with anorexia nervosa and Crohn's disease. The diagnosis of MNGIE was established by biochemical confirmation of elevated serum and urine thymidine and deoxyuridine levels after multiple examinations and several years of disease progression and ineffective treatment. Subsequent molecular genetic testing demonstrated a homozygous TYMP gene mutation. MNGIE should be considered in patients with unexplained malnutrition, intestinal dysmotility, and atypical neurological symptoms.


Asunto(s)
Anorexia Nerviosa/diagnóstico , Enfermedad de Crohn/diagnóstico , Seudoobstrucción Intestinal/diagnóstico , Desnutrición/etiología , Distrofia Muscular Oculofaríngea/diagnóstico , Adulto , Biopsia , Análisis Mutacional de ADN , Diagnóstico Diferencial , Femenino , Predisposición Genética a la Enfermedad , Humanos , Seudoobstrucción Intestinal/complicaciones , Seudoobstrucción Intestinal/genética , Seudoobstrucción Intestinal/terapia , Imagen por Resonancia Magnética , Desnutrición/diagnóstico , Desnutrición/fisiopatología , Desnutrición/terapia , Distrofia Muscular Oculofaríngea/complicaciones , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/terapia , Mutación , Evaluación Nutricional , Estado Nutricional , Oftalmoplejía/congénito , Nutrición Parenteral , Fenotipo , Valor Predictivo de las Pruebas , Timidina Fosforilasa/genética , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
8.
Hum Gene Ther ; 29(6): 708-718, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29284302

RESUMEN

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is caused by mutations in TYMP, the gene encoding the enzyme thymidine phosphorylase (TP). TP dysfunction results in systemic accumulation of the noxious TP substrates thymidine and deoxyuridine. Gene therapy using either a lentiviral vector or adeno-associated vector (AAV) has proven to be a feasible strategy, as both vectors restore biochemical homeostasis in a murine model of the disease. This study shows that the effect of an AAV containing the TYMP coding sequence transcriptionally targeted to the liver persists long term in mice. Although the vector copy number was diluted and AAV-mediated liver TP activity eventually reduced or lost after 21 months at the lowest vector doses, the effect was sustained (with a negligible decrease in TP activity) and fully effective on nucleoside homeostasis for at least 21 months at a dose of 2 × 1012 vg/kg. Macroscopic visual inspection of the animals' organs at completion of the study showed no adverse effects associated with the treatment. These results further support the feasibility of gene therapy for MNGIE.


Asunto(s)
Dependovirus/genética , Terapia Genética , Seudoobstrucción Intestinal/genética , Seudoobstrucción Intestinal/terapia , Hígado/patología , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/terapia , Animales , Carcinogénesis/patología , Desoxiuridina/sangre , Femenino , Dosificación de Gen , Vectores Genéticos/metabolismo , Seudoobstrucción Intestinal/sangre , Estimación de Kaplan-Meier , Masculino , Ratones , Mitocondrias Hepáticas/metabolismo , Distrofia Muscular Oculofaríngea/sangre , Oftalmoplejía/congénito , Timidina/sangre , Timidina Fosforilasa/genética , Factores de Tiempo , Transgenes
9.
Nat Commun ; 8: 14848, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28361972

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant, late-onset muscle disorder characterized by ptosis, swallowing difficulties, proximal limb weakness and nuclear aggregates in skeletal muscles. OPMD is caused by a trinucleotide repeat expansion in the PABPN1 gene that results in an N-terminal expanded polyalanine tract in polyA-binding protein nuclear 1 (PABPN1). Here we show that the treatment of a mouse model of OPMD with an adeno-associated virus-based gene therapy combining complete knockdown of endogenous PABPN1 and its replacement by a wild-type PABPN1 substantially reduces the amount of insoluble aggregates, decreases muscle fibrosis, reverts muscle strength to the level of healthy muscles and normalizes the muscle transcriptome. The efficacy of the combined treatment is further confirmed in cells derived from OPMD patients. These results pave the way towards a gene replacement approach for OPMD treatment.


Asunto(s)
Terapia Genética/métodos , Fuerza Muscular/genética , Distrofia Muscular Oculofaríngea/terapia , Mioblastos Esqueléticos/metabolismo , Proteína I de Unión a Poli(A)/genética , Transcriptoma/genética , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen/métodos , Células HEK293 , Humanos , Técnicas In Vitro , Ratones , Ratones Transgénicos , Distrofia Muscular Oculofaríngea/fisiopatología , Expansión de Repetición de Trinucleótido
10.
Hum Gene Ther ; 26(5): 286-92, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25860803

RESUMEN

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.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Genética , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Técnicas In Vitro , Cuerpos de Inclusión Intranucleares , Distrofia Muscular Oculofaríngea/tratamiento farmacológico , Distrofia Muscular Oculofaríngea/cirugía , Mutación , Proteína I de Unión a Poli(A)/genética
11.
Muscle Nerve ; 52(4): 554-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25677583

RESUMEN

INTRODUCTION: Mild ophthalmoparesis can be seen in oculopharyngeal muscular dystrophy (OPMD). METHODS: Orthoptic analysis included assessment of phoria/tropia, eye excursion, saccades, pursuit, stereoacuity, and Hess-Lancaster screen test. Video-oculography included fixation, horizontal and vertical saccades, and pursuit. RESULTS: Orthoptic abnormalities were: tropia (4 of 6); abnormal eye excursion (4 of 6, 78% involved lateral or superior rectus muscles); abnormal horizontal or vertical saccades (2 of 6); abnormal pursuit (0 of 6); abnormal stereoacuity (2 of 6); and pathological Hess-Lancaster screen (4 of 6). Video-oculographic abnormalities were present for: fixation (1 of 6); saccade latency (1 of 6); horizontal pursuit (3 of 6); and vertical pursuit (0 of 6). For horizontal saccades, mean velocity, peak velocity, and gain were pathological in 5 of 6, 5 of 6 (61% of pathological mean and peak velocities involved abducting eye movements), and 3 of 6, respectively. For vertical saccades, mean velocity, peak velocity, and gain were pathological in 4 of 6, 4 of 6 (53% involved upward movements), and 3 of 6, respectively. CONCLUSION: The data indicate preferential involvement of lateral and (to a lesser degree) superior rectus muscles in OPMD.


Asunto(s)
Electrooculografía , Distrofia Muscular Oculofaríngea/diagnóstico , Distrofia Muscular Oculofaríngea/terapia , Ortóptica/métodos , Grabación en Video , Anciano , Movimientos Oculares/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Distrofia Muscular Oculofaríngea/fisiopatología , Estimulación Luminosa , Tiempo de Reacción/fisiología
12.
Mol Ther ; 22(1): 219-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23831596

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset autosomal dominant genetic disease mainly characterized by ptosis and dysphagia. We conducted a phase I/IIa clinical study (ClinicalTrials.gov NCT00773227) using autologous myoblast transplantation following myotomy in adult OPMD patients. This study included 12 patients with clinical diagnosis of OPMD, indication for cricopharyngeal myotomy, and confirmed genetic diagnosis. The feasibility and safety end points of both autologous myoblast transplantation and the surgical procedure were assessed by videoendoscopy in addition to physical examinations. Potential therapeutic benefit was also assessed through videoendoscopy and videofluoroscopy of swallowing, quality of life score, dysphagia grade, and a drink test. Patients were injected with a median of 178 million myoblasts following myotomy. Short and long-term (2 years) safety and tolerability were observed in all the patients, with no adverse effects. There was an improvement in the quality of life score for all 12 patients, and no functional degradation in swallowing was observed for 10 patients. A cell dose-dependant improvement in swallowing was even observed in this study. This trial supports the hypothesis that a local injection of autologous myoblasts in the pharyngeal muscles is a safe and efficient procedure for OPMD patients.


Asunto(s)
Distrofia Muscular Oculofaríngea/terapia , Mioblastos Esqueléticos/trasplante , Anciano , Esfínter Esofágico Superior/metabolismo , Esfínter Esofágico Superior/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Distrofia Muscular Oculofaríngea/diagnóstico , Distrofia Muscular Oculofaríngea/genética , Músculos Faríngeos/metabolismo , Músculos Faríngeos/fisiopatología , Músculos Faríngeos/cirugía , Trasplante Autólogo , Resultado del Tratamiento
14.
Acta Otorrinolaringol Esp ; 63(6): 482-4, 2012.
Artículo en Inglés, Español | MEDLINE | ID: mdl-21683316

RESUMEN

Oculopharyngeal muscular dystrophy is an infrequent, not widely known entity. Of genetic origin, it usually shows up in the 5th or 6th decade of life. Most cases are referred directly to the gastroenterologist by their general practitioner and not to the otolaryngologist, so it is essential to be aware of this disease to suspect it. We report a case diagnosed and treated in our hospital and we review the literature.


Asunto(s)
Distrofia Muscular Oculofaríngea , Anciano , Femenino , Humanos , Distrofia Muscular Oculofaríngea/diagnóstico , Distrofia Muscular Oculofaríngea/terapia
15.
Dysphagia ; 27(2): 216-20, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21805106

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is a rare autosomal dominant, progressive degenerative muscle disorder featuring dysphagia with limited therapeutic options. The aim of this study was to evaluate the safety and efficacy of repeated endoscopic dilatation for OPMD over a 15-year period. All patients seen at our Regional Swallowing Clinic with OPMD confirmed by genetic analysis were included. Cricopharyngeal dilatation was performed as an outpatient procedure using a wire-guided 18-mm (54 Fr) Savary-Gilliard bougie with the patient under sedation. Patients were offered repeat endoscopic dilatation when symptoms recurred. Symptom severity prior to initial dilatation and at follow-up was evaluated using the Sydney Swallow Questionnaire (SSQ). Nine patients (7 female, 2 male) were included for analysis. Median total treatment period was 13 years (range = 3-15), median number of dilatations per patient was 7.2 (range = 1-16), and median interval between treatments was 15 months (range = 4.5-45). All patients recorded sustained symptom improvement. Mean SSQ score (out of 1,700) was 1,108.11 (SD ± 272.85) prior to first dilatation and 297.78 (SD ± 189.14) at last follow-up, representing a 73% decrease (95% CI = 52-94) in degree of dysphagia symptoms (paired t-test, P = 0.0001). All mean scores for individual questions also showed significant improvement (P < 0.05). No adverse events were reported with all patients maintaining oral feeding at last follow-up. Repeated cricopharyngeal dilatation is a safe, effective, well-tolerated, and long-lasting treatment for dysphagia in OPMD.


Asunto(s)
Trastornos de Deglución/terapia , Distrofia Muscular Oculofaríngea/terapia , Anciano , Trastornos de Deglución/etiología , Dilatación , Endoscopía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Distrofia Muscular Oculofaríngea/complicaciones , Faringe , Índice de Severidad de la Enfermedad
17.
Hum Mol Genet ; 18(10): 1849-59, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19258344

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is a late onset disorder characterized by progressive weakening of specific muscles. It is caused by short expansions of the N-terminal polyalanine tract in the poly(A) binding protein nuclear 1 (PABPN1), and it belongs to the group of protein aggregation diseases, such as Huntington's, Parkinson's and Alzheimer diseases. Mutant PABPN1 forms nuclear aggregates in diseased muscles in both patients and animal models. Intrabodies are antibodies that are modified to be expressed intracellularly and target specific antigens in subcellular locations. They are commonly generated by artificially linking the variable domains of antibody heavy and light chains. However, natural single-chain antibodies are produced in Camelids and, when engineered, combined the advantages of being single-chain, small sized and very stable. Here, we determine the in vivo efficiency of Llama intrabodies against PABPN1, using the established Drosophila model of OPMD. Among six anti-PABPN1 intrabodies expressed in muscle nuclei, we identify one as a strong suppressor of OPMD muscle degeneration in Drosophila, leading to nearly complete rescue. Expression of this intrabody affects PABPN1 aggregation and restores muscle gene expression. This approach promotes the identification of intrabodies with high therapeutic value and highlights the potential of natural single-chain intrabodies in treating protein aggregation diseases.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Modelos Animales de Enfermedad , Drosophila , Distrofia Muscular Oculofaríngea/terapia , Proteína II de Unión a Poli(A)/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Camélidos del Nuevo Mundo , Drosophila/genética , Drosophila/metabolismo , Femenino , Terapia Genética , Humanos , Inmunoterapia , Masculino , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Distrofia Muscular Oculofaríngea/prevención & control , Proteína II de Unión a Poli(A)/metabolismo , Transporte de Proteínas
18.
Hum Mol Genet ; 17(14): 2108-17, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18397876

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is caused by polyalanine expansion in nuclear protein PABPN1 [poly(A) binding protein nuclear 1] and characterized by muscle degeneration. Druggable modifiers of proteotoxicity in degenerative diseases, notably the longevity modulators sirtuins, may constitute useful therapeutic targets. However, the modifiers of mutant PABPN1 are unknown. Here, we report that longevity and cell metabolism modifiers modulate mutant PABPN1 toxicity in the muscle cell. Using PABPN1 nematodes that show muscle cell degeneration and abnormal motility, we found that increased dosage of the sirtuin and deacetylase sir-2.1/SIRT1 exacerbated muscle pathology, an effect dependent on the transcription factor daf-16/FoxO and fuel sensor aak-2/AMPK (AMP-activated protein kinase), while null mutants of sir-2.1, daf-16 and aak-2 were protective. Consistently, the Sir2 inhibitor sirtinol was protective, whereas the Sir2 and AMPK activator resveratrol was detrimental. Furthermore, rescue by sirtinol was dependent on daf-16 and not aak-2, whereas aggravation by resveratrol was dependent on aak-2 and not daf-16. Finally, the survival of mammalian cells expressing mutant PABPN1 was promoted by sirtinol and decreased by resveratrol. Altogether, our data identify Sir2 and AMPK inhibition as therapeutic strategies for muscle protection in OPMD, extending the value of druggable proteins in cell maintenance networks to polyalanine diseases.


Asunto(s)
Caenorhabditis elegans/metabolismo , Distrofia Muscular Oculofaríngea/terapia , Factores de Iniciación de Péptidos/metabolismo , Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Sirtuinas/metabolismo , Proteínas Quinasas Activadas por AMP , Acetilación , Animales , Animales Modificados Genéticamente , Benzamidas/farmacología , Células COS , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Supervivencia Celular , Chlorocebus aethiops , Factores de Transcripción Forkhead , Regulación de la Expresión Génica , Genes Reporteros , Histonas/metabolismo , Humanos , Complejos Multienzimáticos , Músculos/metabolismo , Músculos/fisiopatología , Distrofia Muscular Oculofaríngea/metabolismo , Distrofia Muscular Oculofaríngea/fisiopatología , Naftoles/farmacología , Factores de Iniciación de Péptidos/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/genética , Resveratrol , Sirtuinas/antagonistas & inhibidores , Sirtuinas/genética , Sirtuinas/farmacología , Estilbenos/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Biochim Biophys Acta ; 1772(2): 173-85, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17110089

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disorder characterized by progressive eyelid drooping, swallowing difficulties and proximal limb weakness. OPMD is caused by a small expansion of a short polyalanine tract in the poly (A) binding protein nuclear 1 protein (PABPN1). The mechanism by which the polyalanine expansion mutation in PABPN1 causes disease is unclear. PABPN1 is a nuclear multi-functional protein which is involved in pre-mRNA polyadenylation, transcription regulation, and mRNA nucleocytoplasmic transport. The distinct pathological hallmark of OPMD is the presence of filamentous intranuclear inclusions (INIs) in patient's skeletal muscle cells. The exact relationship between mutant PABPN1 intranuclear aggregates and pathology is not clear. OPMD is a unique disease sharing common pathogenic features with other polyalanine disorders, as well as with polyglutamine and dystrophic disorders. This chapter aims to review the rapidly growing body of knowledge concerning OPMD. First, we outline the background of OPMD. Second, we compare OPMD with other trinucleotide repeat disorders. Third, we discuss the recent advances in the understanding of the molecular mechanisms underlying OPMD pathogenesis. Finally, we review recent therapeutic strategies for OPMD.


Asunto(s)
Distrofia Muscular Oculofaríngea/etiología , Distrofia Muscular Oculofaríngea/terapia , Animales , Humanos , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patología
20.
Neuromuscul Disord ; 16(11): 770-81, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17005403

RESUMEN

Cultures of myoblasts isolated from cricopharyngeal muscles from patients with oculopharyngeal muscular dystrophy (OPMD) have been performed to study the effect of the expanded (GCG)8-13 repeat, located on the poly(A) binding protein nuclear-1 (PABPN1), on satellite cell phenotype. Cell cultures exhibited a reduced myogenicity, as well as a rapid decrease in proliferative lifespan, as compared to controls. The incorporation of BrdU decreased during the proliferative lifespan, due to a progressive accumulation of non-dividing cells. A lower fusion index was also observed, but myoblasts were able to form large myotubes when OPMD cultures were purified, although a rapid loss of myogenicity during successive passages was also observed. Myoblasts isolated from unaffected muscles did not show the defects observed in cricopharyngeal muscle cultures. The PABPN1 was predominantly located in nuclei of myoblasts and in both the nuclei and cytoplasm of myotubes in OPMD cultures. In vivo analysis of OPMD muscles showed that the number of satellite cells was slightly higher than that observed in age matched controls. Mutation of the PABPN1 in OPMD provokes premature senescence in dividing myoblasts, that may be due to intranuclear toxic aggregates. These results suggest that myoblast autografts, isolated from unaffected muscles, and injected into the dystrophic pharyngeal muscles, may be a useful therapeutic strategy to restore muscular function. Its tolerance and feasibility has been preclinically demonstrated in the dog.


Asunto(s)
Proliferación Celular , Esfínter Esofágico Superior/patología , Distrofia Muscular Oculofaríngea/patología , Distrofia Muscular Oculofaríngea/terapia , Mioblastos/trasplante , Proteína II de Unión a Poli(A)/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biopsia , Trasplante de Células/métodos , Células Cultivadas , Senescencia Celular , ADN/metabolismo , Perros , Regulación de la Expresión Génica/genética , Humanos , Persona de Mediana Edad , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Mioblastos/metabolismo , Mioblastos/patología , Fenotipo , Proteína II de Unión a Poli(A)/metabolismo , Células Satélite del Músculo Esquelético/patología , Trasplante Autólogo , Repeticiones de Trinucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA