Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 6393, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34737297

RESUMEN

Pompe disease (PD) is a severe neuromuscular disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). PD is currently treated with enzyme replacement therapy (ERT) with intravenous infusions of recombinant human GAA (rhGAA). Although the introduction of ERT represents a breakthrough in the management of PD, the approach suffers from several shortcomings. Here, we developed a mouse model of PD to compare the efficacy of hepatic gene transfer with adeno-associated virus (AAV) vectors expressing secretable GAA with long-term ERT. Liver expression of GAA results in enhanced pharmacokinetics and uptake of the enzyme in peripheral tissues compared to ERT. Combination of gene transfer with pharmacological chaperones boosts GAA bioavailability, resulting in improved rescue of the PD phenotype. Scale-up of hepatic gene transfer to non-human primates also successfully results in enzyme secretion in blood and uptake in key target tissues, supporting the ongoing clinical translation of the approach.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , alfa-Glucosidasas/metabolismo , Animales , Autofagia , Terapia de Reemplazo Enzimático , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Hígado/enzimología , Masculino , Ratones , alfa-Glucosidasas/genética
2.
EBioMedicine ; 61: 103052, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33039711

RESUMEN

BACKGROUND: Pompe disease (PD) is a neuromuscular disorder caused by deficiency of acidalpha-glucosidase (GAA), leading to motor and respiratory dysfunctions. Available Gaa knock-out (KO) mouse models do not accurately mimic PD, particularly its highly impaired respiratory phenotype. METHODS: Here we developed a new mouse model of PD crossing Gaa KOB6;129 with DBA2/J mice. We subsequently treated Gaa KODBA2/J mice with adeno-associated virus (AAV) vectors expressing a secretable form of GAA (secGAA). FINDINGS: Male Gaa KODBA2/J mice present most of the key features of the human disease, including early lethality, severe respiratory impairment, cardiac hypertrophy and muscle weakness. Transcriptome analyses of Gaa KODBA2/J, compared to the parental Gaa KOB6;129 mice, revealed a profoundly impaired gene signature in the spinal cord and a similarly deregulated gene expression in skeletal muscle. Muscle and spinal cord transcriptome changes, biochemical defects, respiratory and muscle function in the Gaa KODBA2/J model were significantly improved upon gene therapy with AAV vectors expressing secGAA. INTERPRETATION: These data show that the genetic background impacts on the severity of respiratory function and neuroglial spinal cord defects in the Gaa KO mouse model of PD. Our findings have implications for PD prognosis and treatment, show novel molecular pathophysiology mechanisms of the disease and provide a unique model to study PD respiratory defects, which majorly affect patients. FUNDING: This work was supported by Genethon, the French Muscular Dystrophy Association (AFM), the European Commission (grant nos. 667751, 617432, and 797144), and Spark Therapeutics.


Asunto(s)
Terapia Genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Fenotipo , Médula Espinal/metabolismo , alfa-Glucosidasas/genética , Alelos , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Homocigoto , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Fuerza Muscular/genética , Músculo Esquelético , Pronóstico , Médula Espinal/fisiopatología , Transducción Genética , Resultado del Tratamiento , alfa-Glucosidasas/metabolismo
3.
Mol Ther ; 28(9): 2056-2072, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32526204

RESUMEN

Pompe disease is a neuromuscular disorder caused by disease-associated variants in the gene encoding for the lysosomal enzyme acid α-glucosidase (GAA), which converts lysosomal glycogen to glucose. We previously reported full rescue of Pompe disease in symptomatic 4-month-old Gaa knockout (Gaa-/-) mice by adeno-associated virus (AAV) vector-mediated liver gene transfer of an engineered secretable form of GAA (secGAA). Here, we showed that hepatic expression of secGAA rescues the phenotype of 4-month-old Gaa-/- mice at vector doses at which the native form of GAA has little to no therapeutic effect. Based on these results, we then treated severely affected 9-month-old Gaa-/- mice with an AAV vector expressing secGAA and followed the animals for 9 months thereafter. AAV-treated Gaa-/- mice showed complete reversal of the Pompe phenotype, with rescue of glycogen accumulation in most tissues, including the central nervous system, and normalization of muscle strength. Transcriptomic profiling of skeletal muscle showed rescue of most altered pathways, including those involved in mitochondrial defects, a finding supported by structural and biochemical analyses, which also showed restoration of lysosomal function. Together, these results provide insight into the reversibility of advanced Pompe disease in the Gaa-/- mouse model via liver gene transfer of secGAA.


Asunto(s)
Terapia Genética/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Hígado/metabolismo , Vías Secretoras/genética , Transfección/métodos , alfa-Glucosidasas/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Lisosomas/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Fenotipo , Transducción de Señal/genética , Transcriptoma , Resultado del Tratamiento , alfa-Glucosidasas/genética
4.
Mol Ther Methods Clin Dev ; 12: 85-101, 2019 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-30581888

RESUMEN

Hepatocyte-restricted, AAV-mediated gene transfer is being used to provide sustained, tolerogenic transgene expression in gene therapy. However, given the episomal status of the AAV genome, this approach cannot be applied to pediatric disorders when hepatocyte proliferation may result in significant loss of therapeutic efficacy over time. In addition, many multi-systemic diseases require widespread expression of the therapeutic transgene that, when provided with ubiquitous or tissue-specific non-hepatic promoters, often results in anti-transgene immunity. Here we have developed tandem promoter monocistronic expression cassettes that, packaged in a single AAV, provide combined hepatic and extra-hepatic tissue-specific transgene expression and prevent anti-transgene immunity. We validated our approach in infantile Pompe disease, a prototype disease caused by lack of the ubiquitous enzyme acid-alpha-glucosidase (GAA), presenting multi-systemic manifestations and detrimental anti-GAA immunity. We showed that the use of efficient tandem promoters prevents immune responses to GAA following systemic AAV gene transfer in immunocompetent Gaa-/- mice. Then we demonstrated that neonatal gene therapy with either AAV8 or AAV9 in Gaa-/- mice resulted in persistent therapeutic efficacy when using a tandem liver-muscle promoter (LiMP) that provided high and persistent transgene expression in non-dividing extra-hepatic tissues. In conclusion, the tandem promoter design overcomes important limitations of AAV-mediated gene transfer and can be beneficial when treating pediatric conditions requiring persistent multi-systemic transgene expression and prevention of anti-transgene immunity.

5.
Sci Transl Med ; 9(418)2017 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-29187643

RESUMEN

Glycogen storage disease type II or Pompe disease is a severe neuromuscular disorder caused by mutations in the lysosomal enzyme, acid α-glucosidase (GAA), which result in pathological accumulation of glycogen throughout the body. Enzyme replacement therapy is available for Pompe disease; however, it has limited efficacy, has high immunogenicity, and fails to correct pathological glycogen accumulation in nervous tissue and skeletal muscle. Using bioinformatics analysis and protein engineering, we developed transgenes encoding GAA that could be expressed and secreted by hepatocytes. Then, we used adeno-associated virus (AAV) vectors optimized for hepatic expression to deliver the GAA transgenes to Gaa knockout (Gaa-/-) mice, a model of Pompe disease. Therapeutic gene transfer to the liver rescued glycogen accumulation in muscle and the central nervous system, and ameliorated cardiac hypertrophy as well as muscle and respiratory dysfunction in the Gaa-/- mice; mouse survival was also increased. Secretable GAA showed improved therapeutic efficacy and lower immunogenicity compared to nonengineered GAA. Scale-up to nonhuman primates, and modeling of GAA expression in primary human hepatocytes using hepatotropic AAV vectors, demonstrated the therapeutic potential of AAV vector-mediated liver expression of secretable GAA for treating pathological glycogen accumulation in multiple tissues in Pompe disease.


Asunto(s)
Dependovirus/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Hígado/metabolismo , Animales , Terapia Genética , Vectores Genéticos , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/fisiología
6.
PLoS One ; 11(9): e0163958, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27685806

RESUMEN

BACKGROUND: Angioedema without wheals (AE) is a symptom characterised by localised episodes of oedema presumably caused by kinin release from kininogen cleavage. It can result from a hereditary deficiency in C1 Inhibitor (C1Inh), but it can present with normal level of C1Inh. These forms are typically difficult to diagnose although enhanced kinin production is suspected or demonstrated in some cases. OBJECTIVES: We wanted to investigate bradykinin overproduction in all AE condition with normal C1Inh, excluding cases with enhanced kinin catabolism, and to propose this parameter as a disease biomarker. METHODS: We retrospectively investigated high molecular weight kininogen (HK) cleavage pattern, using gel electrophoresis and immunorevelation. Plasma samples were drawn using the same standardised procedure from blood donors or AE patients with normal C1Inh conditions, normal kinin catabolism, and without prophylaxis. RESULTS: Circulating native HK plasma concentrations were similar in the healthy men (interquartile range: 98-175µg/mL, n = 51) and in healthy women (90-176µg/mL, n = 74), while HK cleavage was lower (p<0.001) in men (0-5%) than women (3-9%). Patients exhibited lower native HK concentration (p<10-4; 21-117µg/mL, n = 31 for men; 0-84µg/mL, n = 41 for women) and higher HK cleavage (p<10-4; 10-30% and 14-89%, respectively) than healthy donors. Pathological thresholds were set at: <72µg/mL native HK, >14.4% HK cleavage for men; <38µg/mL; native HK, >33.0% HK cleavage for women, with >98% specificity achieved for all parameters. In plasma from patients undergoing recovery two months after oestrogen/progestin combination withdrawal (n = 13) or two weeks after AE attack (n = 2), HK cleavage was not fully restored, suggesting its use as a post-attack assay. CONCLUSION: As a diagnostic tool, HK cleavage can offer physicians supportive arguments for kinin production in suspected AE cases and improve patient follow-up in clinical trials or prophylactic management.

7.
Mol Immunol ; 71: 161-165, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26897339

RESUMEN

C1 Inhibitor (C1Inh), a member of the Serine proteinase inhibitor family, is the most heavily glycosylated plasma protein. This work investigated the impact of C1Inh glycosylation on its function regarding protease targets and autoantibody binding. C1Inh deglycosylation was found to affect its function with O-linked polysaccharides, but not with N-linked polysaccharides, in controlling the contact phase but not C1s target, thus indicating the N-terminal domain's involvement in C1Inh function. Instructive samples demonstrated that O-deglycosylation strongly suppressed autoantibody binding, suggesting the polysaccharide motif is an antibody target. The autoantibodies did not directly affect C1Inh function.


Asunto(s)
Proteínas Inactivadoras del Complemento 1/metabolismo , Angioedema Hereditario Tipos I y II/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Western Blotting , Proteína Inhibidora del Complemento C1 , Ensayo de Inmunoadsorción Enzimática , Glicoproteínas/metabolismo , Glicosilación , Humanos , Polisacáridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...