Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 31(7): 1994-2004, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-36805083

RESUMEN

Gene therapy with an adeno-associated virus serotype 8 (AAV8) vector (AAV8-LSPhGAA) could eliminate the need for enzyme replacement therapy (ERT) by creating a liver depot for acid α-glucosidase (GAA) production. We report initial safety and bioactivity of the first dose (1.6 × 1012 vector genomes/kg) cohort (n = 3) in a 52-week open-label, single-dose, dose-escalation study (NCT03533673) in patients with late-onset Pompe disease (LOPD). Subjects discontinued biweekly ERT after week 26 based on the detection of elevated serum GAA activity and the absence of clinically significant declines per protocol. Prednisone (60 mg/day) was administered as immunoprophylaxis through week 4, followed by an 11-week taper. All subjects demonstrated sustained serum GAA activities from 101% to 235% of baseline trough activity 2 weeks following the preceding ERT dose. There were no treatment-related serious adverse events. No subject had anti-capsid T cell responses that decreased transgene expression. Muscle biopsy at week 24 revealed unchanged muscle glycogen content in two of three subjects. At week 52, muscle GAA activity for the cohort was significantly increased (p < 0.05). Overall, these initial data support the safety and bioactivity of AAV8-LSPhGAA, the safety of withdrawing ERT, successful immunoprophylaxis, and justify continued clinical development of AAV8-LSPhGAA therapy in Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Humanos , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo , Anticuerpos/genética , Terapia de Reemplazo Enzimático/métodos , Terapia Genética/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Hígado/metabolismo
2.
Hum Gene Ther ; 33(9-10): 492-498, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35102744

RESUMEN

Pompe disease is an autosomal recessive lysosomal storage disorder caused by deficiency of acid α-glucosidase (GAA), resulting in skeletal muscle weakness and cardiomyopathy. Muscle weakness progresses despite currently available therapy, which has prompted the development of gene therapy with adeno-associated virus (AAV) type 2 vectors cross-packaged as AAV8 (2/8). Preclinical studies of gene therapy demonstrated that the minimum effective dose (MED) for biochemical correction with AAV2/8-LSPhGAA was ∼2 × 1011 vector genomes (vg)/kg body weight. The current study examined the transduction of AAV2/8-LSPeGFP vector in adult GAA-KO mice with Pompe disease, and correlated that degree of transduction with the biochemical correction achieved by the same dose of AAV2/8-LSPhGAA. The MED was found to be ∼2 × 1011 vg/kg, with all hepatocytes variably transducing at this dose. At this dose, liver GAA significantly increased, while liver glycogen significantly decreased. The 2 × 1011 vg/kg dose was sufficient to significantly decrease diaphragm glycogen. However, the heart, diaphragm, and quadriceps all required a fourfold higher dose to achieve correction of GAA deficiency in association with significant clearance of stored glycogen, which correlated with increased serum GAA activity. These data indicate that AAV2/8-LSPeGFP transduced all hepatocytes when the 2 × 1011 vg/kg dose was administered, which correlated with partial biochemical correction from the equivalent dose of AAV2/8-LSPhGAA. Altogether, these data support the conclusion that substantial transduction of the liver is required to achieve biochemical correction from AAV2/8-LSPhGAA.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Animales , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Glucógeno , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Ratones , Ratones Noqueados , Músculo Esquelético , alfa-Glucosidasas/genética
3.
Hum Gene Ther ; 33(9-10): 479-491, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35081735

RESUMEN

Pompe disease is an autosomal recessive lysosomal storage disorder caused by deficiency of acid α-glucosidase (GAA), resulting in skeletal muscle weakness and cardiomyopathy that progresses despite currently available therapy in some patients. The development of gene therapy with adeno-associated virus (AAV) vectors revealed a sex-dependent decrease in efficacy in female mice with Pompe disease. This study evaluated the effect of testosterone on gene therapy with an AAV2/8 vector containing a liver-specific promoter to drive expression of GAA (AAV2/8-LSPhGAA) in female GAA-knockout (KO) mice that were implanted with pellets containing testosterone propionate before vector administration. Six weeks after treatment, neuromuscular function and muscle strength were improved as demonstrated by increased Rotarod and wirehang latency for female mice treated with testosterone and vector, in comparison with vector alone. Biochemical correction improved after the addition of testosterone as demonstrated by increased GAA activity and decreased glycogen content in the skeletal muscles of female mice treated with testosterone and vector, in comparison with vector alone. An alternative androgen, oxandrolone, was evaluated similarly to reveal increased GAA in the diaphragm and extensor digitorum longus of female GAA-KO mice after oxandrolone administration; however, glycogen content was unchanged by oxandrolone treatment. The efficacy of androgen hormone treatment in females correlated with increased mannose-6-phosphate receptor in skeletal muscle. These data confirmed the benefits of brief treatment with an androgen hormone in mice with Pompe disease during gene therapy.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Andrógenos/metabolismo , Animales , Dependovirus/genética , Dependovirus/metabolismo , Femenino , Terapia Genética/métodos , Vectores Genéticos/genética , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Humanos , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Oxandrolona/metabolismo , Testosterona/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/uso terapéutico
4.
Commun Biol ; 4(1): 524, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953320

RESUMEN

In Pompe disease, the deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA) causes skeletal and cardiac muscle weakness, respiratory failure, and premature death. While enzyme replacement therapy using recombinant human GAA (rhGAA) can significantly improve patient outcomes, detailed disease mechanisms and incomplete therapeutic effects require further studies. Here we report a three-dimensional primary human skeletal muscle ("myobundle") model of infantile-onset Pompe disease (IOPD) that recapitulates hallmark pathological features including reduced GAA enzyme activity, elevated glycogen content and lysosome abundance, and increased sensitivity of muscle contractile function to metabolic stress. In vitro treatment of IOPD myobundles with rhGAA or adeno-associated virus (AAV)-mediated hGAA expression yields increased GAA activity and robust glycogen clearance, but no improvements in stress-induced functional deficits. We also apply RNA sequencing analysis to the quadriceps of untreated and AAV-treated GAA-/- mice and wild-type controls to establish a Pompe disease-specific transcriptional signature and reveal novel disease pathways. The mouse-derived signature is enriched in the transcriptomic profile of IOPD vs. healthy myobundles and partially reversed by in vitro rhGAA treatment, further confirming the utility of the human myobundle model for studies of Pompe disease and therapy.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Contracción Muscular , Músculo Esquelético/citología , Miocardio/citología , Ingeniería de Tejidos/métodos , alfa-Glucosidasas/metabolismo , Animales , Dependovirus/genética , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , alfa-Glucosidasas/administración & dosificación , alfa-Glucosidasas/genética
5.
Mol Ther Methods Clin Dev ; 17: 133-142, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31909086

RESUMEN

Pompe disease is caused by the deficiency of lysosomal acid α-glucosidase (GAA). It is expected that gene therapy to replace GAA with adeno-associated virus (AAV) vectors will be less effective early in life because of the rapid loss of vector genomes. AAV2/8-LSPhGAA (3 × 1010 vector genomes [vg]/mouse) was administered to infant (2-week-old) or adult (2-month-old) GAA knockout mice. AAV vector transduction in adult mice significantly corrected GAA deficiency in the heart (p < 0.0001), diaphragm (p < 0.01), and quadriceps (p < 0.001) for >50 weeks. However, in infant mice, the same treatment only partially corrected GAA deficiency in the heart (p < 0.05), diaphragm (p < 0.05), and quadriceps (p < 0.05). The clearance of glycogen was much more efficient in adult mice compared with infant mice. Improved wire hang test latency was observed for treated adults (p < 0.05), but not for infant mice. Abnormal ventilation was corrected in both infant and adult mice. Vector-treated female mice demonstrated functional improvement, despite a lower degree of biochemical correction compared with male mice. The relative vector dose for infants was approximately 3-fold higher than adults, when normalized to body weight at the time of vector administration. Given these data, the dose requirement to achieve similar efficacy will be higher for the treatment of young patients.

6.
Mol Genet Metab ; 129(2): 67-72, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31839530

RESUMEN

This 24-week, Phase I/II, double-blind, randomized, placebo-controlled study investigated the safety and efficacy of extended-release albuterol in late-onset Pompe disease stably treated with enzyme replacement therapy at the standard dose for 4.9 (1.0-9.4) years and with no contraindications to intake of albuterol. Twelve of 13 participants completed the study. No serious adverse events were related to albuterol, and transient minor drug-related adverse events included muscle spasms and tremors. For the albuterol group, forced vital capacity in the supine position increased by 10% (p < .005), and forced expiratory volume in one second increased by 8% (p < .05); the six-minute walk test increased 25 m (p < .05; excluding one participant unable to complete muscle function testing); the Gross Motor Function Measure increased by 8% (p < .005) with the greatest increases in the Standing (18%; p < .05) and Walking, Running, and Jumping (11%; p < .005) subtests. No significant improvements would be expected in patients with late-onset Pompe disease who were stably treated with enzyme replacement therapy. The placebo group demonstrated no significant increases in performance on any measure. These data support a potential benefit of extended-release albuterol as adjunctive therapy in carefully selected patients with late-onset Pompe disease based on ability to take albuterol on enzyme replacement therapy (NCT01885936).


Asunto(s)
Albuterol/administración & dosificación , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedades de Inicio Tardío/tratamiento farmacológico , Músculo Esquelético/efectos de los fármacos , Adulto , Método Doble Ciego , Terapia de Reemplazo Enzimático , Femenino , Volumen Espiratorio Forzado , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/fisiología , Resultado del Tratamiento , Capacidad Vital , Prueba de Paso
7.
Mol Genet Metab ; 129(2): 73-79, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31645300

RESUMEN

Pompe disease is caused by the deficiency of lysosomal acid α-glucosidase (GAA) leading to progressive myopathy. Enzyme replacement therapy (ERT) with recombinant human (rh) GAA has limitations, including inefficient uptake of rhGAA in skeletal muscle linked to low cation-independent mannose-6-phosphate receptor (CI-MPR) expression. PURPOSE: To test the hypothesis that antihypertensive agents causing muscle hypertrophy by increasing insulin-like growth factor 1 expression can increase CI-MPR-mediated uptake of recombinant enzyme with therapeutic effects in skeletal muscle. METHODS: Three such agents were evaluated in mice with Pompe disease (carvedilol, losartan, and propranolol), either with or without concurrent ERT. RESULTS: Carvedilol, a selective ß-blocker, increased muscle strength but reduced biochemical correction from ERT. Administration of drugs alone had minimal effect, with the exception of losartan that increased glycogen storage and mortality either by itself or in combination with ERT. CONCLUSION: The ß-blocker carvedilol had beneficial effects during ERT in mice with Pompe disease, in comparison with propranolol or losartan. Caution is warranted when prescribing antihypertensive drugs in Pompe disease.


Asunto(s)
Antihipertensivos/uso terapéutico , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Músculo Esquelético/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/patología , alfa-Glucosidasas/genética
9.
Hum Gene Ther ; 30(7): 855-864, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30803275

RESUMEN

Gene therapy for Pompe disease with adeno-associated virus (AAV) vectors has advanced into early phase clinical trials; however, the paucity of cation-independent mannose-6-phosphate receptor (CI-MPR) in skeletal muscle, where it is needed to take up acid α-glucosidase (GAA), has impeded the efficacy of Pompe disease gene therapy. Long-acting selective ß2 receptor agonists previously enhanced the CI-MPR expression in muscle. In this study we have evaluated the selective ß2 agonist salmeterol in GAA knockout mice in combination with an AAV vector expressing human GAA specifically in the liver. Quadriceps glycogen content was significantly decreased by administration of the AAV vector with salmeterol, in comparison with the AAV vector alone (p < 0.01). Importantly, glycogen content of the quadriceps was reduced to its lowest level by the combination of AAV vector and salmeterol administration. Rotarod testing revealed significant improvement following treatment, in comparison with untreated mice, and salmeterol improved wirehang performance. Salmeterol treatment decreased abnormalities of autophagy in the quadriceps, as shown be lower LC3 and p62. Vector administration reduced the abnormal vacuolization and accumulation of nuclei in skeletal muscle. Thus, salmeterol could be further developed as adjunctive therapy to improve the efficacy of liver depot gene therapy for Pompe disease.


Asunto(s)
Terapia Genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Xinafoato de Salmeterol/farmacología , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Activación Enzimática , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Ratones , Ratones Noqueados , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Transducción Genética
10.
Mol Ther ; 26(9): 2304-2314, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30025991

RESUMEN

This 52-week, phase I/II double-blind, randomized, placebo-controlled study investigated the novel use of clenbuterol in late-onset Pompe disease (LOPD) stably treated with ERT. Eleven of thirteen participants completed the study. No serious adverse events were related to clenbuterol, and transient minor adverse events included mild elevations of creatine kinase, muscle spasms, and tremors. At week 52, the 6-min walk test distance increased by a mean of 16 m (p = 0.08), or a mean of 3% of predicted performance (p = 0.03), and the maximum inspiratory pressure increased 8% (p = 0.003) for the clenbuterol group. The quick motor function test score improved by a mean of seven points (p = 0.007); and the gait, stairs, gower, chair test improved by a mean of two points (p = 0.004). Clenbuterol decreased glycogen content in the vastus lateralis by 50% at week 52. Transcriptome analysis revealed more normal muscle gene expression for 38 of 44 genes related to Pompe disease following clenbuterol. The placebo group demonstrated no significant changes over the course of the study. This study provides initial evidence for safety and efficacy of adjunctive clenbuterol in patients with LOPD (NCT01942590).


Asunto(s)
Clenbuterol/uso terapéutico , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Adulto , Anciano , Método Doble Ciego , Femenino , Glucógeno/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Cuádriceps/efectos de los fármacos , Músculo Cuádriceps/metabolismo
11.
Mol Ther Methods Clin Dev ; 4: 126-136, 2017 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-28344998

RESUMEN

Pompe disease results from acid α-glucosidase (GAA) deficiency, and enzyme replacement therapy (ERT) with recombinant human (rh) GAA has clinical benefits, although its limitations include the short half-life of GAA and the formation of antibody responses. The present study compared the efficacy of ERT against gene transfer with an adeno-associated viral (AAV) vector containing a liver-specific promoter. GAA knockout (KO) mice were administered either a weekly injection of rhGAA (20 mg/kg) or a single injection of AAV2/8-LSPhGAA (8 × 1011 vector genomes [vg]/kg). Both treatments significantly reduced glycogen content of the heart and diaphragm. Although ERT triggered anti-GAA antibody formation, there was no detectable antibody response following AAV vector administration. The efficacy of three lower dosages of AAV2/8-LSPhGAA was evaluated in GAA-KO mice, either alone or in combination with ERT. The minimum effective dose (MED) identified was 8 × 1010 vg/kg to reduce glycogen content in the heart and diaphragm of GAA-KO mice. A 3-fold higher dose was required to suppress antibody responses to ERT. Efficacy from liver gene therapy was slightly greater in male mice than in female mice. Vector dose correlated inversely with anti-GAA antibody formation, whereas higher vector doses suppressed previously formed anti-GAA antibodies as late as 25 weeks after the start of ERT and achieved biochemical correction of glycogen accumulation. In conclusion, we identified the MED for effective AAV2/8-LSPhGAA-mediated tolerogenic gene therapy in Pompe disease mice.

12.
Mol Genet Metab ; 118(1): 35-40, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27017193

RESUMEN

Enzyme replacement therapy (ERT) with recombinant human (rh) acid α-glucosidase (GAA) has prolonged the survival of patients. However, the paucity of cation-independent mannose-6-phosphate receptor (CI-MPR) in skeletal muscle, where it is needed to take up rhGAA, correlated with a poor response to ERT by muscle in Pompe disease. Clenbuterol, a selective ß2 receptor agonist, enhanced the CI-MPR expression in striated muscle through Igf-1 mediated muscle hypertrophy, which correlated with increased CI-MPR (also the Igf-2 receptor) expression. In this study we have evaluated 4 new drugs in GAA knockout (KO) mice in combination with an adeno-associated virus (AAV) vector encoding human GAA, 3 alternative ß2 agonists and dehydroepiandrosterone (DHEA). Mice were injected with AAV2/9-CBhGAA (1E+11 vector particles) at a dose that was not effective at clearing glycogen storage from the heart. Heart GAA activity was significantly increased by either salmeterol (p<0.01) or DHEA (p<0.05), in comparison with untreated mice. Furthermore, glycogen content was reduced in the heart by treatment with DHEA (p<0.001), salmeterol (p<0.05), formoterol (p<0.01), or clenbuterol (p<0.01) in combination with the AAV vector, in comparison with untreated GAA-KO mice. Wirehang testing revealed that salmeterol and the AAV vector significantly increased performance, in comparison with the AAV vector alone (p<0.001). Similarly, salmeterol with the vector increased performance significantly more than any of the other drugs. The most effective individual drugs had no significant effect in absence of vector, in comparison with untreated mice. Thus, salmeterol should be further developed as adjunctive therapy in combination with either ERT or gene therapy for Pompe disease.


Asunto(s)
Terapia Genética/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Miocardio/metabolismo , Xinafoato de Salmeterol/administración & dosificación , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo , Animales , Clenbuterol/administración & dosificación , Deshidroepiandrosterona/administración & dosificación , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático , Vectores Genéticos/administración & dosificación , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Humanos , Ratones , Ratones Noqueados
13.
J Biol Chem ; 291(14): 7450-64, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26839314

RESUMEN

Toll-like receptor 4 (TLR4) promotes vascular inflammatory disorders such as neointimal hyperplasia and atherosclerosis. TLR4 triggers NFκB signaling through the ubiquitin ligase TRAF6 (tumor necrosis factor receptor-associated factor 6). TRAF6 activity can be impeded by deubiquitinating enzymes like ubiquitin-specific protease 20 (USP20), which can reverse TRAF6 autoubiquitination, and by association with the multifunctional adaptor protein ß-arrestin2. Although ß-arrestin2 effects on TRAF6 suggest an anti-inflammatory role, physiologic ß-arrestin2 promotes inflammation in atherosclerosis and neointimal hyperplasia. We hypothesized that anti- and proinflammatory dimensions of ß-arrestin2 activity could be dictated by ß-arrestin2's ubiquitination status, which has been linked with its ability to scaffold and localize activated ERK1/2 to signalosomes. With purified proteins and in intact cells, our protein interaction studies showed that TRAF6/USP20 association and subsequent USP20-mediated TRAF6 deubiquitination were ß-arrestin2-dependent. Generation of transgenic mice with smooth muscle cell-specific expression of either USP20 or its catalytically inactive mutant revealed anti-inflammatory effects of USP20in vivoandin vitro Carotid endothelial denudation showed that antagonizing smooth muscle cell USP20 activity increased NFκB activation and neointimal hyperplasia. We found that ß-arrestin2 ubiquitination was promoted by TLR4 and reversed by USP20. The association of USP20 with ß-arrestin2 was augmented when ß-arrestin2 ubiquitination was prevented and reduced when ß-arrestin2 ubiquitination was rendered constitutive. Constitutive ß-arrestin2 ubiquitination also augmented NFκB activation. We infer that pro- and anti-inflammatory activities of ß-arrestin2 are determined by ß-arrestin2 ubiquitination and that changes in USP20 expression and/or activity can therefore regulate inflammatory responses, at least in part, by defining the ubiquitination status of ß-arrestin2.


Asunto(s)
Arrestinas/metabolismo , Endopeptidasas/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Ubiquitinación/fisiología , Animales , Arrestinas/genética , Línea Celular , Endopeptidasas/genética , Ratones , Ratones Noqueados , FN-kappa B/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 4/genética , Ubiquitina Tiolesterasa , beta-Arrestinas
14.
Mol Genet Metab ; 117(2): 114-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26454691

RESUMEN

UNLABELLED: Enzyme replacement therapy (ERT) with recombinant human acid α-glucosidase (rhGAA) fails to completely reverse muscle weakness in Pompe disease. ß2-agonists enhanced ERT by increasing receptor-mediated uptake of rhGAA in skeletal muscles. PURPOSE: To test the hypothesis that a ß-blocker might reduce the efficacy of ERT, because the action of ß-blockers opposes those of ß2-agonists. METHODS: Mice with Pompe disease were treated with propranolol (a ß-blocker) or clenbuterol in combination with ERT, or with ERT alone. RESULTS: Propranolol-treated mice had decreased weight gain (p<0.01), in comparison with clenbuterol-treated mice. Left ventricular mass was decreased (and comparable to wild-type) in ERT only and clenbuterol-treated groups of mice, and unchanged in propranolol-treated mice. GAA activity increased following either clenbuterol or propranolol in skeletal muscles. However, muscle glycogen was reduced only in clenbuterol-treated mice, not in propranolol-treated mice. Cell-based experiments confirmed that propranolol reduces uptake of rhGAA into Pompe fibroblasts and also demonstrated that the drug induces intracellular accumulation of glycoproteins at higher doses. CONCLUSION: Propranolol, a commonly prescribed ß-blocker, reduced weight, increased left ventricular mass and decreased glycogen clearance in skeletal muscle following ERT. ß-Blockers might therefore decrease the efficacy from ERT in patients with Pompe disease.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Propranolol/farmacología , alfa-Glucosidasas/uso terapéutico , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Células Cultivadas , Antagonismo de Drogas , Evaluación Preclínica de Medicamentos , Fibroblastos/metabolismo , Glucógeno/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Humanos , Ratones Noqueados , Propranolol/uso terapéutico , alfa-Glucosidasas/farmacología
15.
Hum Gene Ther ; 26(11): 743-50, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26417913

RESUMEN

Pompe disease (glycogen storage disease type II; acid maltase deficiency) is a devastating myopathy resulting from acid α-glucosidase (GAA) deficiency in striated and smooth muscle. Despite the availability of enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA), the limitations of ERT have prompted the preclinical development of gene therapy. Gene therapy has the advantage of continuously producing GAA, in contrast to ERT, which requires frequent injections of rhGAA. An adeno-associated viral (AAV) vector containing a muscle-specific promoter, AAV-MHCK7hGAApA, achieved high GAA expression in heart and skeletal muscle in mice with Pompe disease. However, elevated GAA activity was not sufficient to completely clear accumulated glycogen in skeletal muscle. The process of glycogen clearance from lysosomes might require improved trafficking of GAA to the lysosomes in skeletal muscle, previously achieved with the ß(2)-agonist clenbuterol that enhanced glycogen clearance in skeletal muscle without increasing GAA activity. Glycogen clearance was clearly enhanced by treatment with a nondepleting anti-CD4 monoclonal antibody (anti-CD4 mAb) along with muscle-specific GAA expression in cardiac muscle, but that treatment was not effective in skeletal muscle. Furthermore, anti-CD4 mAb treatment along with clenbuterol achieved synergistic therapeutic efficacy in both cardiac and skeletal muscle. This triple therapy increased both muscle strength and weight gain. Overall, triple therapy to enhance GAA trafficking and to suppress immune responses significantly improved the efficacy of muscle-targeted gene therapy in murine Pompe disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Clenbuterol/uso terapéutico , Terapia Genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , alfa-Glucosidasas/uso terapéutico , Agonistas Adrenérgicos beta/uso terapéutico , Animales , Antígenos CD4/metabolismo , Terapia Combinada , Dependovirus/genética , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Tolerancia Inmunológica , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Recombinantes/uso terapéutico , alfa-Glucosidasas/genética
16.
Hum Gene Ther ; 26(1): 26-35, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25382056

RESUMEN

Enzyme replacement therapy (ERT) is the standard-of-care treatment of Pompe disease, a lysosomal storage disorder caused by deficiency of acid α-glucosidase (GAA). One limitation of ERT with recombinant human (rh) GAA is antibody formation against GAA. Similarly, in adeno-associated virus (AAV) vector-mediated gene transfer for Pompe disease, development of antibodies against the GAA transgene product and the AAV vector prevents therapeutic efficacy and vector readministration, respectively. Here a nondepleting anti-CD4 monoclonal antibody (mAb) was administrated intravenously prior to administration of an AAV2/9 vector encoding GAA to suppress anti-GAA responses, leading to a substantial reduction of anti-GAA immunoglobulins, including IgG1, IgG2a, IgG2b, IgG2c, and IgG3. Transduction efficiency in liver with a subsequent AAV2/8 vector was massively improved by the administration of anti-CD4 mAb with the initial AAV2/9 vector, indicating a spread of benefit derived from control of the immune response to the first AAV2/9 vector. Anti-CD4 mAb along with AAV2/9-CBhGAApA significantly increased GAA activity in heart and skeletal muscles along with a significant reduction of glycogen accumulation. Taken together, these data demonstrated that the addition of nondepleting anti-CD4 mAb with gene therapy controls humoral immune responses to both vector and transgene, resulting in clear therapeutic benefit in mice with Pompe disease.


Asunto(s)
Anticuerpos/genética , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , alfa-Glucosidasas/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Antígenos CD4/inmunología , Proteínas de la Cápside/inmunología , Línea Celular , Reacciones Cruzadas/inmunología , Dependovirus/inmunología , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Factores Sexuales , Transducción Genética , Transgenes , alfa-Glucosidasas/inmunología , alfa-Glucosidasas/metabolismo
17.
EMBO Rep ; 14(2): 164-71, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23208550

RESUMEN

ß-arrestin 1 and 2 (also known as arrestin 2 and 3) are homologous adaptor proteins that regulate seven-transmembrane receptor trafficking and signalling. Other proteins with predicted 'arrestin-like' structural domains but lacking sequence homology have been indicated to function like ß-arrestin in receptor regulation. We demonstrate that ß-arrestin2 is the primary adaptor that rapidly binds agonist-activated ß(2) adrenergic receptors (ß(2)ARs) and promotes clathrin-dependent internalization, E3 ligase Nedd4 recruitment and ubiquitin-dependent lysosomal degradation of the receptor. The arrestin-domain-containing (ARRDC) proteins 2, 3 and 4 are secondary adaptors recruited to internalized ß(2)AR-Nedd4 complexes on endosomes and do not affect the adaptor roles of ß-arrestin2. Rather, the role of ARRDC proteins is to traffic Nedd4-ß(2)AR complexes to a subpopulation of early endosomes.


Asunto(s)
Arrestinas/fisiología , Receptores Adrenérgicos beta 2/metabolismo , Ubiquitinación , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Animales , Células COS , Chlorocebus aethiops , Endocitosis , Células HEK293 , Humanos , Isoproterenol/farmacología , Microscopía Fluorescente , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteolisis , beta-Arrestina 1 , beta-Arrestinas
18.
J Cell Biol ; 199(5): 817-30, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23166351

RESUMEN

Lysosomal degradation of ubiquitinated ß(2)-adrenergic receptors (ß(2)ARs) serves as a major mechanism of long-term desensitization in response to prolonged agonist stimulation. Surprisingly, the ßAR antagonist carvedilol also induced ubiquitination and lysosomal trafficking of both endogenously expressed ß(2)ARs in vascular smooth muscle cells (VSMCs) and overexpressed Flag-ß(2)ARs in HEK-293 cells. Carvedilol prevented ß(2)AR recycling, blocked recruitment of Nedd4 E3 ligase, and promoted the dissociation of the deubiquitinases USP20 and USP33. Using proteomics approaches (liquid chromatography-tandem mass spectrometry), we identified that the E3 ligase MARCH2 interacted with carvedilol-bound ß(2)AR. The association of MARCH2 with internalized ß(2)ARs was stabilized by carvedilol and did not involve ß-arrestin. Small interfering RNA-mediated down-regulation of MARCH2 ablated carvedilol-induced ubiquitination, endocytosis, and degradation of endogenous ß(2)ARs in VSMCs. These findings strongly suggest that specific ligands recruit distinct E3 ligase machineries to activated cell surface receptors and direct their intracellular itinerary. In response to ß blocker therapy with carvedilol, MARCH2 E3 ligase activity regulates cell surface ß(2)AR expression and, consequently, its signaling.


Asunto(s)
Carbazoles/farmacología , Proteínas Portadoras/metabolismo , Endocitosis , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Propanolaminas/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Animales , Proteínas Portadoras/genética , Carvedilol , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Ratas , Receptores Adrenérgicos beta 2/genética , Ubiquitina-Proteína Ligasas
19.
PLoS One ; 7(8): e42868, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22900059

RESUMEN

Protein ubiquitination, which is highly selective, regulates many important biological processes including cellular differentiation and pathogenesis in eukaryotic cells. Here, we integrated pharmacological, molecular and proteomic approaches to explore the role of ubiquitination in Magnaporthe oryzae, the leading fungal disease of rice world-wide. Inhibition of ubiquitin-mediated proteolysis using the 26S proteasome inhibitor, Bortezomib, significantly attenuated conidia germination, appressorium formation and pathogenicity in M. oryzae. Gene expression analysis revealed that many genes associated with protein ubiquitination were developmentally regulated during conidia germination. Only a few, including a polyubiquitin encoding gene, MGG_01282, were more abundantly expressed during appressorium formation and under nitrogen starvation. Targeted gene deletion of MGG_01282, in addition to a significant reduction in protein ubiquitination as determined by immuno blot assays, resulted in pleiotropic effects on M. oryzae including reduced growth and sporulation, abnormal conidia morphology, reduced germination and appressorium formation, and the inability to cause disease. Mutants were also defective in sexual development and were female sterile. Using mass spectrometry, we identified 63 candidate polyubiquitinated proteins under nitrogen starvation, which included overrepresentation of proteins involved in translation, transport and protein modification. Our study suggests that ubiquitination of target proteins plays an important role in nutrient assimilation, development and pathogenicity of M. oryzae.


Asunto(s)
Magnaporthe/crecimiento & desarrollo , Magnaporthe/genética , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Poliubiquitina/genética , Ácidos Borónicos/farmacología , Bortezomib , Análisis por Conglomerados , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Perfilación de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Germinación/genética , Magnaporthe/patogenicidad , Mutación , Nitrógeno/metabolismo , Poliubiquitina/metabolismo , Inhibidores de Proteasoma/farmacología , Proteolisis/efectos de los fármacos , Proteómica , Pirazinas/farmacología , Ubiquitinación
20.
Reprod Biol Endocrinol ; 7: 66, 2009 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-19552818

RESUMEN

BACKGROUND: Activin A increases production of follicle stimulating hormone (FSH) by inducing transcription of its beta subunit (FSHB). This induction has been studied here in LbetaT2 gonadotropes using transient expression of ovine FSHBLuc (-4741 bp of ovine FSHB promoter plus exon/intron 1 linked to Luc). Several sequences between -169/-58 bp of the ovine FSHB proximal promoter are necessary for induction by activin A in LbetaT2 cells, but deletions between -4741/-752 bp decrease induction > 70% suggesting the existence of other important 5' sequences. Induction disappears if a minimal T81 thymidine kinase promoter replaces the ovine FSHB TATA box and 3' exon/intron. The study reported here was designed to determine if sequences outside -169/-58 bp are important for induction of ovine FSHB by activin A. METHODS: Progressively longer deletions of ovine FSHBLuc were created between -4741/-195 bp. Deletions internal to this region were created also, but replaced with substitute DNA. The ovine FSHB TATA box region (-40/+3 bp) was replaced by thymidine kinase and rat prolactin minimal promoters, and substitutions were made in 3' intron/exon sequences. All constructs were tested for basal and activin A-induced expression in LbetaT2 cells. RESULTS: Successive 5' deletions progressively lowered fold-induction by activin A from 9.5 to zero, but progressively increased basal expression. Replacing deletions with substitute DNA showed no changes in basal expression or fold-induction. Induction by activin A was supported by the minimal rat prolactin promoter (TATA box) but not the thymidine kinase promoter (no TATA box). Replacement mutations in the 3' region did not decrease induction by activin A. CONCLUSION: The data show that specific ovine FSHB sequences 5' to -175 bp or 3' of the transcription start site are not required for induction by activin A. A minimal TATA box promoter supports induction by activin A, but the sequence between the TATA box and transcription start site seems unimportant.


Asunto(s)
Activinas/farmacología , Hormona Folículo Estimulante de Subunidad beta/genética , Regiones Promotoras Genéticas/genética , TATA Box , Animales , Secuencia de Bases , Hormona Folículo Estimulante de Subunidad beta/biosíntesis , Gonadotrofos/metabolismo , Ratones , Ratas , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...