Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 13(11): e14434, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34606154

RESUMO

Pompe disease is a metabolic myopathy due to acid alpha-glucosidase deficiency. In addition to glycogen storage, secondary dysregulation of cellular functions, such as autophagy and oxidative stress, contributes to the disease pathophysiology. We have tested whether oxidative stress impacts on enzyme replacement therapy with recombinant human alpha-glucosidase (rhGAA), currently the standard of care for Pompe disease patients, and whether correction of oxidative stress may be beneficial for rhGAA therapy. We found elevated oxidative stress levels in tissues from the Pompe disease murine model and in patients' cells. In cells, stress levels inversely correlated with the ability of rhGAA to correct the enzymatic deficiency. Antioxidants (N-acetylcysteine, idebenone, resveratrol, edaravone) improved alpha-glucosidase activity in rhGAA-treated cells, enhanced enzyme processing, and improved mannose-6-phosphate receptor localization. When co-administered with rhGAA, antioxidants improved alpha-glucosidase activity in tissues from the Pompe disease mouse model. These results indicate that oxidative stress impacts on the efficacy of enzyme replacement therapy in Pompe disease and that manipulation of secondary abnormalities may represent a strategy to improve the efficacy of therapies for this disorder.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Animais , Terapia de Reposição de Enzimas , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Humanos , Camundongos , Estresse Oxidativo , alfa-Glucosidases/metabolismo , alfa-Glucosidases/uso terapêutico
2.
J Enzyme Inhib Med Chem ; 36(1): 2068-2079, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34565280

RESUMO

Pompe disease is an inherited metabolic disorder due to the deficiency of the lysosomal acid α-glucosidase (GAA). The only approved treatment is enzyme replacement therapy with the recombinant enzyme (rhGAA). Further approaches like pharmacological chaperone therapy, based on the stabilising effect induced by small molecules on the target enzyme, could be a promising strategy. However, most known chaperones could be limited by their potential inhibitory effects on patient's enzymes. Here we report on the discovery of novel chaperones for rhGAA, L- and D-carnitine, and the related compound acetyl-D-carnitine. These drugs stabilise the enzyme at pH and temperature without inhibiting the activity and acted synergistically with active-site directed pharmacological chaperones. Remarkably, they enhanced by 4-fold the acid α-glucosidase activity in fibroblasts from three Pompe patients with added rhGAA. This synergistic effect of L-carnitine and rhGAA has the potential to be translated into improved therapeutic efficacy of ERT in Pompe disease.


Assuntos
Carnitina/farmacologia , Inibidores de Glicosídeo Hidrolases/farmacologia , Lisossomos/efeitos dos fármacos , Chaperonas Moleculares/farmacologia , alfa-Glucosidases/metabolismo , Regulação Alostérica/efeitos dos fármacos , Carnitina/química , Relação Dose-Resposta a Droga , Inibidores de Glicosídeo Hidrolases/química , Humanos , Lisossomos/enzimologia , Chaperonas Moleculares/química , Estrutura Molecular , Relação Estrutura-Atividade
3.
J Med Chem ; 60(23): 9462-9469, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29112434

RESUMO

The highly stereocontrolled de novo synthesis of l-NBDNJ (the unnatural enantiomer of the iminosugar drug Miglustat) and a preliminary evaluation of its chaperoning potential are herein reported. l-NBDNJ is able to enhance lysosomal α-glucosidase levels in Pompe disease fibroblasts, either when administered singularly or when coincubated with the recombinant human α-glucosidase. In addition, differently from its d-enantiomer, l-NBDNJ does not act as a glycosidase inhibitor.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , alfa-Glucosidases/metabolismo , 1-Desoxinojirimicina/síntese química , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacologia , Regulação Alostérica/efeitos dos fármacos , Linhagem Celular , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Doença de Depósito de Glicogênio Tipo II/enzimologia , Doença de Depósito de Glicogênio Tipo II/metabolismo , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Lisossomos/metabolismo , Modelos Moleculares , Estereoisomerismo
4.
Mol Ther ; 22(11): 2004-12, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25052852

RESUMO

Enzyme replacement therapy is currently the only approved treatment for Pompe disease, due to acid α-glucosidase deficiency. Clinical efficacy of this approach is variable, and more effective therapies are needed. We showed in preclinical studies that chaperones stabilize the recombinant enzyme used for enzyme replacement therapy. Here, we evaluated the effects of a combination of enzyme therapy and a chaperone on α-glucosidase activity in Pompe disease patients. α-Glucosidase activity was analyzed by tandem-mass spectrometry in dried blood spots from patients treated with enzyme replacement therapy, either alone or in combination with the chaperone N-butyldeoxynojirimycin given at the time of the enzyme infusion. Thirteen patients with different presentations (3 infantile-onset, 10 late-onset) were enrolled. In 11 patients, the combination treatment resulted in α-glucosidase activities greater than 1.85-fold the activities with enzyme replacement therapy alone. In the whole patient population, α-glucosidase activity was significantly increased at 12 hours (2.19-fold, P = 0.002), 24 hours (6.07-fold, P = 0.001), and 36 hours (3.95-fold, P = 0.003). The areas under the curve were also significantly increased (6.78-fold, P = 0.002). These results suggest improved stability of recombinant α-glucosidase in blood in the presence of the chaperone.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Doença de Depósito de Glicogênio Tipo II/sangue , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Inibidores de Glicosídeo Hidrolases/farmacologia , alfa-Glucosidases/farmacologia , 1-Desoxinojirimicina/farmacologia , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Teste em Amostras de Sangue Seco , Sinergismo Farmacológico , Terapia de Reposição de Enzimas/métodos , Estabilidade Enzimática , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem , alfa-Glucosidases/sangue , alfa-Glucosidases/uso terapêutico
6.
Mol Genet Metab ; 107(3): 267-75, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22963910

RESUMO

Anderson-Fabry disease is an X-linked lysosomal storage disorder resulting from the deficiency of the hydrolytic enzyme alpha galactosidase A, with consequent accumulation of globotrioasoyl ceramide in cells and tissues of the body, resulting in a multi-system pathology including end organ failure. In the classical phenotype, cardiac failure, renal failure and stroke result in a reduced median life expectancy. The current causal treatment for Fabry disease is the enzyme replacement therapy (ERT): two different products, Replagal (agalsidase alfa) and Fabrazyme (agalsidase beta), have been commercially available in Europe for almost 10 years and they are both indicated for long-term treatment. In fact, clinical trials, observational studies and registry data have provided many evidences for safety and efficacy of ERT in improving symptoms of pain, gastrointestinal disturbances, hypohidrosis, left ventricular mass index, glomerular filtration rate and quality of life. Few data are available on comparison of the two treatments and on the clinical course of the disease. This article reviews the published evidence for clinical efficacy of the two available enzyme preparations.


Assuntos
Doença de Fabry/tratamento farmacológico , Insuficiência Cardíaca/prevenção & controle , Isoenzimas/uso terapêutico , Insuficiência Renal/prevenção & controle , Acidente Vascular Cerebral/prevenção & controle , alfa-Galactosidase/uso terapêutico , Adulto , Ensaios Clínicos como Assunto , Terapia de Reposição de Enzimas , Doença de Fabry/complicações , Doença de Fabry/metabolismo , Doença de Fabry/patologia , Feminino , Galactosidases/deficiência , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Isoenzimas/farmacologia , Masculino , Proteínas Recombinantes , Insuficiência Renal/etiologia , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Triexosilceramidas/metabolismo , alfa-Galactosidase/farmacologia
7.
Mol Ther ; 20(12): 2201-11, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22990675

RESUMO

Pompe disease (PD) is a metabolic myopathy due to the deficiency of the lysosomal enzyme α-glucosidase (GAA). The only approved treatment for this disorder, enzyme replacement with recombinant human GAA (rhGAA), has shown limited therapeutic efficacy in some PD patients. Pharmacological chaperone therapy (PCT), either alone or in combination with enzyme replacement, has been proposed as an alternative therapeutic strategy. However, the chaperones identified so far also are active site-directed molecules and potential inhibitors of target enzymes. We demonstrated that N-acetylcysteine (NAC) is a novel allosteric chaperone for GAA. NAC improved the stability of rhGAA as a function of pH and temperature without disrupting its catalytic activity. A computational analysis of NAC-GAA interactions confirmed that NAC does not interact with GAA catalytic domain. NAC enhanced the residual activity of mutated GAA in cultured PD fibroblasts and in COS7 cells overexpressing mutated GAA. NAC also enhanced rhGAA efficacy in PD fibroblasts. In cells incubated with NAC and rhGAA, GAA activities were 3.7-8.7-fold higher than those obtained in cells treated with rhGAA alone. In a PD mouse model the combination of NAC and rhGAA resulted in better correction of enzyme activity in liver, heart, diaphragm and gastrocnemia, compared to rhGAA alone.


Assuntos
Acetilcisteína/uso terapêutico , alfa-Glucosidases/metabolismo , alfa-Glucosidases/uso terapêutico , Acetilcisteína/farmacocinética , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Estabilidade Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Imunofluorescência , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Humanos , Camundongos , Microscopia Confocal , Chaperonas Moleculares/farmacologia , Chaperonas Moleculares/uso terapêutico , alfa-Glucosidases/química
8.
J Nephrol ; 25(4): 582-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22307442

RESUMO

Fabry disease is an X-linked lysosomal disease caused by mutations of the alpha-galactosidase A (GLA) gene at chromosome subband Xq22.1. To date, more than 600 genetic mutations have been identified to determine the nature and frequency of the molecular lesions causing the classical and milder variant phenotypes and for precise carrier detection. We report here a Fabry family (mother, son and daughter) where the alpha-galactosidase A defect was associated with a glucose-6-phosphate dehydrogenase (G6PD) deficiency. Mutation analysis revealed for the GLA gene the presence of a new mutation, i.e., a small deletion (c.452delA) on exon 3 and for the G6PD gene the presence of 2 mutations, p.V68M (G6PD Asahi, G6PD A+) and p.N126D (G6PD A+) on exon 3 and exon 4, respectively.


Assuntos
Doença de Fabry/genética , Deficiência de Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/genética , Deleção de Sequência , alfa-Galactosidase/genética , Adolescente , Análise Mutacional de DNA , Terapia de Reposição de Enzimas , Éxons , Doença de Fabry/diagnóstico , Doença de Fabry/tratamento farmacológico , Doença de Fabry/enzimologia , Favismo/genética , Feminino , Predisposição Genética para Doença , Deficiência de Glucosefosfato Desidrogenase/diagnóstico , Deficiência de Glucosefosfato Desidrogenase/enzimologia , Humanos , Isoenzimas/uso terapêutico , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Adulto Jovem , alfa-Galactosidase/uso terapêutico
9.
J Inherit Metab Dis ; 35(3): 513-20, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22187137

RESUMO

Fabry disease (FD) is an X-linked inherited disease due to alpha-galactosidase A (alpha-Gal A) deficiency and characterized by lysosomal storage of globotriaosylceramide (Gb3) and related neutral glycosphingolipids. Storage of these substrates results in multisystem manifestations, including renal failure, cardiomyopathy, premature myocardial infarctions, stroke, chronic neuronopathic pain, gastrointestinal disturbances, and skin angiokeratoma. Enzyme replacement therapy (ERT) with recombinant human alpha-galactosidase A (rh-alpha-Gal A) is now available for the treatment of FD and in most patients results in clinical improvement or stabilization. However, ERT efficacy may vary in different tissues and its long-term effects remain to be defined. As a strategy to improve the efficacy of ERT, we tested the combination of rh-alpha-Gal A with the chaperone molecule 1-deoxynojirimycin (DGJ) in cultured FD fibroblasts with negligible residual enzyme activity. Compared to the effects of rh-alpha-Gal A alone, co-administration of DGJ and rh-alpha-Gal A resulted in better correction (4.8 to 16.9-fold) of intracellular alpha-Gal A activity, and increased amounts of the enzyme within the lysosomal compartment. The clearance of lyso-Gb3, one of the substrates stored in FD and a potent inhibitor of alpha-Gal A, was also significantly improved with the co-administration of DGJ and rh-alpha-Gal A. This study provides additional evidence for a synergistic effect between ERT and pharmacological chaperone therapy and supports the idea that the efficacy of combination protocols may be superior to ERT alone.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Doença de Fabry/enzimologia , Doença de Fabry/metabolismo , Fibroblastos/metabolismo , Proteínas Recombinantes/metabolismo , alfa-Galactosidase/metabolismo , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacologia , Estudos de Casos e Controles , Éxons , Genótipo , Humanos , Lisossomos/metabolismo , Masculino , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Mutação , Triexosilceramidas/química
10.
Hum Mutat ; 30(12): 1683-92, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19862843

RESUMO

Pompe disease is a lysosomal storage disorder (LSD) caused by mutations in the gene that encodes acid alpha-glucosidase (GAA). Recently, small molecule pharmacological chaperones have been shown to increase protein stability and cellular levels for mutant lysosomal enzymes and have emerged as a new therapeutic strategy for the treatment of LSDs. In this study, we characterized the pharmacological chaperone 1-deoxynojirimycin (DNJ) on 76 different mutant forms of GAA identified in Pompe disease. DNJ significantly increased enzyme activity and protein levels for 16 different GAA mutants in patient-derived fibroblasts and in transiently transfected COS-7 cells. Additionally, DNJ increased the processing of these GAA mutants to their mature lysosomal forms, suggesting facilitated trafficking through the secretory pathway. Immunofluorescence microscopy studies showed increased colocalization of GAA with the lysosomal marker LAMP2 after incubation with DNJ, confirming increased lysosomal trafficking. Lastly, a GAA structural model was constructed based on the related eukaryotic glucosidase maltase-glucoamylase. The mutated residues identified in responsive forms of GAA are located throughout most of the structural domains, with half of these residues located in two short regions within the catalytic domain. Taken together, these data support further evaluation of DNJ as a potential treatment for Pompe disease in patients that express responsive forms of GAA.


Assuntos
1-Desoxinojirimicina/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Proteínas Mutantes/metabolismo , alfa-Glucosidases/metabolismo , Adolescente , Adulto , Animais , Células COS , Chlorocebus aethiops , Estabilidade Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Doença de Depósito de Glicogênio Tipo II/enzimologia , Humanos , Lactente , Modelos Moleculares , Estrutura Secundária de Proteína , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , alfa-Glucosidases/química
11.
Mol Ther ; 17(6): 964-71, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19293774

RESUMO

In spite of the progress in the treatment of lysosomal storage diseases (LSDs), in some of these disorders the available therapies show limited efficacy and a need exists to identify novel therapeutic strategies. We studied the combination of enzyme replacement and enzyme enhancement by pharmacological chaperones in Pompe disease (PD), a metabolic myopathy caused by the deficiency of the lysosomal acid alpha-glucosidase. We showed that coincubation of Pompe fibroblasts with recombinant human alpha-glucosidase and the chaperone N-butyldeoxynojirimycin (NB-DNJ) resulted in more efficient correction of enzyme activity. The chaperone improved alpha-glucosidase delivery to lysosomes, enhanced enzyme maturation, and increased enzyme stability. Improved enzyme correction was also found in vivo in a mouse model of PD treated with coadministration of single infusions of recombinant human alpha-glucosidase and oral NB-DNJ. The enhancing effect of chaperones on recombinant enzymes was also observed in fibroblasts from another lysosomal disease, Fabry disease, treated with recombinant alpha-galactosidase A and the specific chaperone 1-deoxygalactonojirimycin (DGJ). These results have important clinical implications, as they demonstrate synergy between pharmacological chaperones and enzyme replacement. A synergistic effect of these treatments may result particularly useful in patients responding poorly to therapy and in tissues in which sufficient enzyme levels are difficult to obtain.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Inibidores Enzimáticos/uso terapêutico , Fibroblastos/efeitos dos fármacos , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , 1-Desoxinojirimicina/farmacologia , 1-Desoxinojirimicina/uso terapêutico , Animais , Transporte Biológico/efeitos dos fármacos , Western Blotting , Linhagem Celular , Estabilidade de Medicamentos , Inibidores Enzimáticos/farmacologia , Fibroblastos/patologia , Humanos , Lisossomos/metabolismo , Camundongos , Microscopia Confocal , alfa-Galactosidase/administração & dosagem , alfa-Galactosidase/metabolismo , alfa-Galactosidase/farmacologia , alfa-Galactosidase/uso terapêutico
12.
Pathogenetics ; 1(1): 6, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19046416

RESUMO

BACKGROUND: Pompe disease (PD) is a metabolic myopathy caused by alpha-glucosidase (GAA) deficiency and characterized by generalized glycogen storage. Heterogeneous GAA gene mutations result in wide phenotypic variability, ranging from the severe classic infantile presentation to the milder intermediate and late-onset forms. Enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA), the only treatment available for PD, intriguingly shows variable efficacy in different PD patients. To investigate the mechanisms underlying the variable response to ERT, we studied cell morphology of PD fibroblasts, the distribution and trafficking of the cation-independent mannose-6-phosphate receptor (CI-MPR) that mediates rhGAA uptake, and rhGAA uptake itself. RESULTS: We observed abnormalities of cell morphology in PD cells. Electron microscopy analysis showed accumulation of multivesicular bodies and expansion of the Golgi apparatus, and immunolocalization and western blot analysis of LC3 showed activation of autophagy. Immunofluorescence analysis showed abnormal intracellular distribution of CI-MPR in PD fibroblasts, increased co-localization with LC3 and reduced availability of the receptor at the plasma membrane. The recycling of CI-MPR from the plasma membrane to the trans-Golgi network was also impaired. All these abnormalities were more prominent in severe and intermediate PD fibroblasts, correlating with disease severity. In severe and intermediate PD cells rhGAA uptake and processing were less efficient and correction of GAA activity was reduced. CONCLUSION: These results indicate a role for disrupted CI-MPR trafficking in the variable response to ERT in PD and have implications for ERT efficacy and optimization of treatment protocols.

13.
Mol Ther ; 15(3): 508-14, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17213836

RESUMO

We investigated the use of pharmacological chaperones for the therapy of Pompe disease, a metabolic myopathy due to mutations of the gene encoding the lysosomal hydrolase alpha-glucosidase (GAA) and characterized by generalized glycogen storage in cardiac and skeletal muscle. We studied the effects of two imino sugars, deoxynojirimycin (DNJ) and N-butyldeoxynojirimycin (NB-DNJ), on residual GAA activity in fibroblasts from eight patients with different forms of Pompe disease (two classic infantile, two non-classic infantile onset, four late-onset forms), and with different mutations of the GAA gene. We demonstrated a significant increase of GAA activity (1.3-7.5-fold) after imino sugar treatment in fibroblasts from patients carrying the mutations L552P (three patients) and G549R (one patient). GAA enhancement was confirmed in HEK293T cells where the same mutations were overexpressed. No increase of GAA activity was observed for the other mutations. Western blot analysis showed that imino sugars increase the amount of mature GAA molecular forms. Immunofluorescence studies in HEK293T cells overexpressing the L552P mutation showed an improved trafficking of the mutant enzyme to lysosomes after imino sugar treatment. These results provide a rationale for an alternative treatment, other than enzyme replacement, to Pompe disease.


Assuntos
Doença de Depósito de Glicogênio Tipo II/enzimologia , Imino Açúcares/farmacologia , alfa-Glucosidases/metabolismo , Adulto , Linhagem Celular , Criança , Pré-Escolar , Ativação Enzimática/efeitos dos fármacos , Feminino , Fibroblastos , Genótipo , Doença de Depósito de Glicogênio Tipo II/genética , Humanos , Mutação/genética , Fenótipo , alfa-Glucosidases/genética
14.
Mol Ther ; 15(3): 508-514, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28182897

RESUMO

We investigated the use of pharmacological chaperones for the therapy of Pompe disease, a metabolic myopathy due to mutations of the gene encoding the lysosomal hydrolase α-glucosidase (GAA) and characterized by generalized glycogen storage in cardiac and skeletal muscle. We studied the effects of two imino sugars, deoxynojirimycin (DNJ) and N-butyldeoxynojirimycin (NB-DNJ), on residual GAA activity in fibroblasts from eight patients with different forms of Pompe disease (two classic infantile, two non-classic infantile onset, four late-onset forms), and with different mutations of the GAA gene. We demonstrated a significant increase of GAA activity (1.3-7.5-fold) after imino sugar treatment in fibroblasts from patients carrying the mutations L552P (three patients) and G549R (one patient). GAA enhancement was confirmed in HEK293T cells where the same mutations were overexpressed. No increase of GAA activity was observed for the other mutations. Western blot analysis showed that imino sugars increase the amount of mature GAA molecular forms. Immunofluorescence studies in HEK293T cells overexpressing the L552P mutation showed an improved trafficking of the mutant enzyme to lysosomes after imino sugar treatment. These results provide a rationale for an alternative treatment, other than enzyme replacement, to Pompe disease.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...