Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 31(3): 616-630, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36635967

RESUMO

Assessment of anti-adeno-associated virus (AAV) antibodies in patients prior to systemic gene therapy administration is an important consideration regarding efficacy and safety of the therapy. Approximately 30%-60% of individuals have pre-existing anti-AAV antibodies. Seroprevalence is impacted by multiple factors, including geography, age, capsid serotype, and assay type. Anti-AAV antibody assays typically measure (1) transduction inhibition by detecting the neutralizing capacity of antibodies and non-antibody neutralizing factors, or (2) total anti-capsid binding antibodies, regardless of neutralizing activity. Presently, there is a paucity of head-to-head data and standardized approaches associating assay results with clinical outcomes. In addition, establishing clinically relevant screening titer cutoffs is complex. Thus, meaningful comparisons across assays are nearly impossible. Although complex, establishing screening assays in routine clinical practice to identify patients with antibody levels that may impact favorable treatment outcomes is achievable for both transduction inhibition and total antibody assays. Formal regulatory approval of such assays as companion diagnostic tests will confirm their suitability for specific recombinant AAV gene therapies. This review covers current approaches to measure anti-AAV antibodies in patient plasma or serum, their potential impact on therapeutic safety and efficacy, and investigative strategies to mitigate the effects of pre-existing anti-AAV antibodies in patients.


Assuntos
Anticorpos Neutralizantes , Dependovirus , Humanos , Dependovirus/genética , Estudos Soroepidemiológicos , Vetores Genéticos/genética , Terapia Genética/métodos , Anticorpos Antivirais , Proteínas do Capsídeo/genética
2.
J Inherit Metab Dis ; 45(3): 481-492, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34918784

RESUMO

Classic galactosemia (CG) is a rare disorder of autosomal recessive inheritance. It is caused predominantly by point mutations as well as deletions in the gene encoding the enzyme galactose-1-phosphate uridyltransferase (GALT). The majority of the more than 350 mutations identified in the GALT gene cause a significant reduction in GALT enzyme activity resulting in the toxic buildup of galactose metabolites that in turn is associated with cellular stress and injury. Consequently, developing a therapeutic strategy that reverses both the oxidative and ER stress in CG cells may be helpful in combating this disease. Recombinant adeno-associated virus (AAV)-mediated gene therapy to restore GALT activity offers the potential to address the unmet medical needs of galactosemia patients. Here, utilizing fibroblasts derived from CG patients we demonstrated that AAV-mediated augmentation of GALT protein and activity resulted in the prevention of ER and oxidative stress. We also demonstrate that these CG patient fibroblasts exhibit reduced CD109 and TGFßRII protein levels and that these effectors of cellular homeostasis could be restored following AAV-mediated expression of GALT. Finally, we show initial in vivo proof-of-concept restoration of galactose metabolism in a GALT knockout mouse model following treatment with AAV-GALT.


Assuntos
Galactosemias , UTP-Hexose-1-Fosfato Uridililtransferase , Animais , Fibroblastos/metabolismo , Galactose/metabolismo , Galactosemias/genética , Galactosemias/terapia , Humanos , Camundongos , Camundongos Knockout , UTP-Hexose-1-Fosfato Uridililtransferase/genética , UTP-Hexose-1-Fosfato Uridililtransferase/metabolismo
3.
J Neurochem ; 156(5): 692-701, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32743826

RESUMO

Most lysosomal storage diseases (LSDs) have a significant neurological component, including types 2 and 3 Gaucher disease (neuronal forms of Gaucher disease; nGD). No therapies are currently available for nGD since the recombinant enzymes used in the systemic form of Gaucher disease do not cross the blood-brain barrier (BBB). However, a number of promising approaches are currently being tested, including substrate reduction therapy (SRT), in which partial inhibition of the synthesis of the glycosphingolipids (GSLs) that accumulate in nGD lowers their accumulation. We now induce nGD in mice by injection with conduritol B-epoxide (CBE), an irreversible inhibitor of acid beta-glucosidase (GCase), the enzyme defective in nGD, with or without co-injection with Genz-667161, a prototype for SRT which crosses the BBB. Significant neuropathology, and a reduction in lifespan, was observed upon CBE injection, and this was largely reversed by co-injection with Genz-667161, along with a reduction in glucosylceramide and glucosylsphingosine levels. Analysis of gene expression by RNAseq revealed that Genz-667161 largely reversed the changes in genes and pathways that were differentially expressed upon CBE injection, specifically pathways of GSL metabolism, lipoproteins and other lipid metabolic pathways, lipid droplets, astrocyte activation, neuronal function, and to some extent, neuroinflammation. Together, this demonstrates the efficacy of SRT to reverse the effects of substrate accumulation on pathological components and pathways in nGD brain.


Assuntos
Modelos Animais de Doenças , Doença de Gaucher/metabolismo , Doença de Gaucher/patologia , Glucosilceramidase/antagonistas & inibidores , Glicoesfingolipídeos/antagonistas & inibidores , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Doença de Gaucher/tratamento farmacológico , Glucosilceramidase/metabolismo , Glicoesfingolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia
4.
Mol Ther ; 28(2): 664-676, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31843448

RESUMO

Patients with α-dystroglycanopathies, a subgroup of rare congenital muscular dystrophies, present with a spectrum of clinical manifestations that includes muscular dystrophy as well as CNS and ocular abnormalities. Although patients with α-dystroglycanopathies are genetically heterogeneous, they share a common defect of aberrant post-translational glycosylation modification of the dystroglycan alpha-subunit, which renders it defective in binding to several extracellular ligands such as laminin-211 in skeletal muscles, agrin in neuromuscular junctions, neurexin in the CNS, and pikachurin in the eye, leading to various symptoms. The genetic heterogeneity associated with the development of α-dystroglycanopathies poses significant challenges to developing a generalized treatment to address the spectrum of genetic defects. Here, we propose the development of a bispecific antibody (biAb) that functions as a surrogate molecular linker to reconnect laminin-211 and the dystroglycan beta-subunit to ameliorate sarcolemmal fragility, a primary pathology in patients with α-dystroglycan-related muscular dystrophies. We show that the treatment of LARGEmyd-3J mice, an α-dystroglycanopathy model, with the biAb improved muscle function and protected muscles from exercise-induced damage. These results demonstrate the viability of a biAb that binds to laminin-211 and dystroglycan simultaneously as a potential treatment for patients with α-dystroglycanopathy.


Assuntos
Anticorpos Biespecíficos/farmacologia , Distroglicanas/metabolismo , Laminina/metabolismo , Síndrome de Walker-Warburg/metabolismo , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/metabolismo , Modelos Animais de Doenças , Distroglicanas/imunologia , Expressão Gênica , Humanos , Imuno-Histoquímica , Injeções Intramusculares , Laminina/genética , Laminina/imunologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Ligação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas/genética , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Síndrome de Walker-Warburg/tratamento farmacológico , Síndrome de Walker-Warburg/etiologia
5.
Mol Ther ; 27(8): 1495-1506, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31208914

RESUMO

Neuronopathic glycosphingolipidoses are a sub-group of lysosomal storage disorders for which there are presently no effective therapies. Here, we evaluated the potential of substrate reduction therapy (SRT) using an inhibitor of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide (GL1) and related glycosphingolipids. The substrates that accumulate in Sandhoff disease (e.g., ganglioside GM2 and its nonacylated derivative, lyso-GM2) are distal to the drug target, GCS. Treatment of Sandhoff mice with a GCS inhibitor that has demonstrated CNS access (Genz-682452) reduced the accumulation of GL1 and GM2, as well as a variety of disease-associated substrates in the liver and brain. Concomitant with these effects was a significant decrease in the expression of CD68 and glycoprotein non-metastatic melanoma B protein (Gpnmb) in the brain, indicating a reduction in microgliosis in the treated mice. Moreover, using in vivo imaging, we showed that the monocytic biomarker translocator protein (TSPO), which was elevated in Sandhoff mice, was normalized following Genz-682452 treatment. These positive effects translated in turn into a delay (∼28 days) in loss of motor function and coordination, as measured by rotarod latency, and a significant increase in longevity (∼17.5%). Together, these results support the development of SRT for the treatment of gangliosidoses, particularly in patients with residual enzyme activity.


Assuntos
Carbamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Glucosiltransferases/antagonistas & inibidores , Quinuclidinas/farmacologia , Doença de Sandhoff/enzimologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Ligantes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Knockout , Imagem Molecular , Receptores de GABA/metabolismo , Doença de Sandhoff/diagnóstico , Doença de Sandhoff/genética , Doença de Sandhoff/terapia , Esfingolipídeos/metabolismo , Cadeia beta da beta-Hexosaminidase/genética , Cadeia beta da beta-Hexosaminidase/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(10): 2699-2704, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28223512

RESUMO

Mutations in the glucocerebrosidase gene (GBA) confer a heightened risk of developing Parkinson's disease (PD) and other synucleinopathies, resulting in a lower age of onset and exacerbating disease progression. However, the precise mechanisms by which mutations in GBA increase PD risk and accelerate its progression remain unclear. Here, we investigated the merits of glucosylceramide synthase (GCS) inhibition as a potential treatment for synucleinopathies. Two murine models of synucleinopathy (a Gaucher-related synucleinopathy model, GbaD409V/D409V and a A53T-α-synuclein overexpressing model harboring wild-type alleles of GBA, A53T-SNCA mouse model) were exposed to a brain-penetrant GCS inhibitor, GZ667161. Treatment of GbaD409V/D409V mice with the GCS inhibitor reduced levels of glucosylceramide and glucosylsphingosine in the central nervous system (CNS), demonstrating target engagement. Remarkably, treatment with GZ667161 slowed the accumulation of hippocampal aggregates of α-synuclein, ubiquitin, and tau, and improved the associated memory deficits. Similarly, prolonged treatment of A53T-SNCA mice with GZ667161 reduced membrane-associated α-synuclein in the CNS and ameliorated cognitive deficits. The data support the contention that prolonged antagonism of GCS in the CNS can affect α-synuclein processing and improve behavioral outcomes. Hence, inhibition of GCS represents a disease-modifying therapeutic strategy for GBA-related synucleinopathies and conceivably for certain forms of sporadic disease.


Assuntos
Carbamatos/farmacologia , Inibidores Enzimáticos/administração & dosagem , Glucosiltransferases/antagonistas & inibidores , Doença de Parkinson/tratamento farmacológico , Quinuclidinas/farmacologia , alfa-Sinucleína/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Glucosiltransferases/genética , Humanos , Camundongos , Mutação , Doença de Parkinson/enzimologia , Doença de Parkinson/patologia , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Ubiquitina/metabolismo , Proteínas tau/metabolismo
7.
Hum Mol Genet ; 26(6): 1182-1192, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28158561

RESUMO

Fabry disease is caused by deficient activity of α-galactosidase A and subsequent accumulation of glycosphingolipids (mainly globotriaosylceramide, Gb3), leading to multisystem organ dysfunction. Oxidative stress and nitric oxide synthase (NOS) uncoupling are thought to contribute to Fabry cardiovascular diseases. We hypothesized that decreased tetrahydrobiopterin (BH4) plays a role in the pathogenesis of Fabry disease. We found that BH4 was decreased in the heart and kidney but not in the liver and aorta of Fabry mice. BH4 was also decreased in the plasma of female Fabry patients, which was not corrected by enzyme replacement therapy (ERT). Gb3 levels were inversely correlated with BH4 levels in animal tissues and cultured patient cells. To investigate the role of BH4 deficiency in disease phenotypes, 12-month-old Fabry mice were treated with gene transfer-mediated ERT or substrate reduction therapy (SRT) for 6 months. In the Fabry mice receiving SRT but not ERT, BH4 deficiency was restored, concomitant with ameliorated cardiac and renal hypertrophy. Additionally, glutathione levels were decreased in Fabry mouse tissues in a sex-dependent manner. Renal BH4 levels were closely correlated with glutathione levels and inversely correlated with cardiac and kidney weight. In conclusion, this study showed that BH4 deficiency occurs in Fabry disease and may contribute to the pathogenesis of the disease through oxidative stress associated with a reduced antioxidant capacity of cells and NOS uncoupling. This study also suggested dissimilar efficacy of ERT and SRT in correcting pre-existing pathologies in Fabry disease.


Assuntos
Biopterinas/análogos & derivados , Terapia de Reposição de Enzimas , Doença de Fabry/genética , alfa-Galactosidase/genética , Animais , Biopterinas/deficiência , Biopterinas/genética , Biopterinas/metabolismo , Modelos Animais de Doenças , Doença de Fabry/mortalidade , Doença de Fabry/fisiopatologia , Feminino , Glutationa/metabolismo , Glicoesfingolipídeos/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo/genética , alfa-Galactosidase/biossíntese , alfa-Galactosidase/metabolismo
8.
Gene Ther ; 25(3): 205-219, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29785047

RESUMO

The successful application of adeno-associated virus (AAV) gene delivery vectors as a therapeutic paradigm will require efficient gene delivery to the appropriate cells in affected organs. In this study, we utilized a rational design approach to introduce modifications to the AAV2 and AAVrh8R capsids and the resulting variants were evaluated for transduction activity in the retina and brain. The modifications disrupted either capsid/receptor binding or altered capsid surface charge. Specifically, we mutated AAV2 amino acids R585A and R588A, which are required for binding to its receptor, heparan sulfate proteoglycans, to generate a variant referred to as AAV2-HBKO. In contrast to parental AAV2, the AAV2-HBKO vector displayed low-transduction activity following intravitreal delivery to the mouse eye; however, following its subretinal delivery, AAV2-HBKO resulted in significantly greater photoreceptor transduction. Intrastriatal delivery of AAV2-HBKO to mice facilitated widespread striatal and cortical expression, in contrast to the restricted transduction pattern of the parental AAV2 vector. Furthermore, we found that altering the surface charge on the AAVrh8R capsid by modifying the number of arginine residues on the capsid surface had a profound impact on subretinal transduction. The data further validate the potential of capsid engineering to improve AAV gene therapy vectors for clinical applications.


Assuntos
Terapia Genética/métodos , Parvovirinae/crescimento & desenvolvimento , Parvovirinae/imunologia , Animais , Encéfalo/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Dependovirus/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos , Células HeLa , Heparitina Sulfato , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Transdução Genética/métodos
9.
Hum Mol Genet ; 25(13): 2645-2660, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27126635

RESUMO

Mutations in GBA1, the gene encoding glucocerebrosidase, are associated with an enhanced risk of developing synucleinopathies such as Parkinson's disease (PD) and dementia with Lewy bodies. A higher prevalence and increased severity of motor and non-motor symptoms is observed in PD patients harboring mutant GBA1 alleles, suggesting a link between the gene or gene product and disease development. Interestingly, PD patients without mutations in GBA1 also exhibit lower levels of glucocerebrosidase activity in the central nervous system (CNS), implicating this lysosomal enzyme in disease pathogenesis. Here, we investigated whether modulation of glucocerebrosidase activity in murine models of synucleinopathy (expressing wild type Gba1) affected α-synuclein accumulation and behavioral phenotypes. Partial inhibition of glucocerebrosidase activity in PrP-A53T-SNCA mice using the covalent inhibitor conduritol-B-epoxide induced a profound increase in soluble α-synuclein in the CNS and exacerbated cognitive and motor deficits. Conversely, augmenting glucocerebrosidase activity in the Thy1-SNCA mouse model of PD delayed the progression of synucleinopathy. Adeno-associated virus-mediated expression of glucocerebrosidase in the Thy1-SNCA mouse striatum led to decrease in the levels of the proteinase K-resistant fraction of α-synuclein, amelioration of behavioral aberrations and protection from loss of striatal dopaminergic markers. These data indicate that increasing glucocerebrosidase activity can influence α-synuclein homeostasis, thereby reducing the progression of synucleinopathies. This study provides robust in vivo evidence that augmentation of CNS glucocerebrosidase activity is a potential therapeutic strategy for PD, regardless of the mutation status of GBA1.


Assuntos
Glucosilceramidase/metabolismo , Glucosilceramidase/fisiologia , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Dopamina , Doença de Gaucher/genética , Expressão Gênica , Glucosilceramidase/genética , Glucosilceramidase/uso terapêutico , Humanos , Camundongos , Atividade Motora/efeitos dos fármacos , Mutação , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , alfa-Sinucleína/líquido cefalorraquidiano , alfa-Sinucleína/metabolismo
10.
Lancet ; 390(10089): 50-61, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526489

RESUMO

BACKGROUND: Long-term intraocular injections of vascular endothelial growth factor (VEGF)-neutralising proteins can preserve central vision in many patients with neovascular age-related macular degeneration. We tested the safety and tolerability of a single intravitreous injection of an AAV2 vector expressing the VEGF-neutralising protein sFLT01 in patients with advanced neovascular age-related macular degeneration. METHODS: This was a phase 1, open-label, dose-escalating study done at four outpatient retina clinics in the USA. Patients were assigned to each cohort in order of enrolment, with the first three patients being assigned to and completing the first cohort before filling positions in the following treatment groups. Patients aged 50 years or older with neovascular age-related macular degeneration and a baseline best-corrected visual acuity score of 20/100 or less in the study eye were enrolled in four dose-ranging cohorts (cohort 1, 2 × 108 vector genomes (vg); cohort 2, 2 × 109 vg; cohort 3, 6 × 109 vg; and cohort 4, 2 × 1010 vg, n=3 per cohort) and one maximum tolerated dose cohort (cohort 5, 2 × 1010 vg, n=7) and followed up for 52 weeks. The primary objective of the study was to assess the safety and tolerability of a single intravitreous injection of AAV2-sFLT01, through the measurement of eye-related adverse events. This trial is registered with ClinicalTrials.gov, number NCT01024998. FINDINGS: 19 patients with advanced neovascular age-related macular degeneration were enrolled in the study between May 18, 2010, and July 14, 2014. All patients completed the 52-week trial period. Two patients in cohort 4 (2 × 1010 vg) experienced adverse events that were possibly study-drug related: pyrexia and intraocular inflammation that resolved with a topical steroid. Five of ten patients who received 2 × 1010 vg had aqueous humour concentrations of sFLT01 that peaked at 32·7-112·0 ng/mL (mean 73·7 ng/mL, SD 30·5) by week 26 with a slight decrease to a mean of 53·2 ng/mL at week 52 (SD 17·1). At baseline, four of these five patients were negative for anti-AAV2 serum antibodies and the fifth had a very low titre (1:100) of anti-AAV2 antibodies, whereas four of the five non-expressers of sFLT01 had titres of 1:400 or greater. In 11 of 19 patients with intraretinal or subretinal fluid at baseline judged to be reversible, six showed substantial fluid reduction and improvement in vision, whereas five showed no fluid reduction. One patient in cohort 5 showed a large decrease in vision between weeks 26 and 52 that was not thought to be vector-related. INTERPRETATION: Intravitreous injection of AAV2-sFLT01 seemed to be safe and well tolerated at all doses. Additional studies are needed to identify sources of variability in expression and anti-permeability activity, including the potential effect of baseline anti-AAV2 serum antibodies. FUNDING: Sanofi Genzyme, Framingham, MA, USA.


Assuntos
Terapia Genética/métodos , Degeneração Macular/terapia , Parvovirinae/genética , Proteínas Recombinantes de Fusão/genética , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/biossíntese , Inibidores da Angiogênese/genética , Neovascularização de Coroide/diagnóstico por imagem , Neovascularização de Coroide/fisiopatologia , Neovascularização de Coroide/terapia , Dependovirus , Feminino , Terapia Genética/efeitos adversos , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intravítreas , Degeneração Macular/diagnóstico por imagem , Degeneração Macular/fisiopatologia , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Tomografia de Coerência Óptica , Acuidade Visual
11.
J Inherit Metab Dis ; 41(2): 231-238, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29110178

RESUMO

Fabry disease is a glycosphingolipidosis caused by deficient activity of α-galactosidase A; it is one of a few diseases that are associated with priapism, an abnormal prolonged erection of the penis. The goal of this study was to investigate the pathogenesis of Fabry disease-associated priapism in a mouse model of the disease. We found that Fabry mice develop late-onset priapism. Neuronal nitric oxide synthase (nNOS), which was predominantly present as the 120-kDa N-terminus-truncated form, was significantly upregulated in the penis of 18-month-old Fabry mice compared to wild type controls (~fivefold). Endothelial NOS (eNOS) was also upregulated (~twofold). NO level in penile tissues of Fabry mice was significantly higher than wild type controls at 18 months. Gene transfer-mediated enzyme replacement therapy reversed abnormal nNOS expression in the Fabry mouse penis. The penile nNOS level was restored by antiandrogen treatment, suggesting that hyperactive androgen receptor signaling in Fabry mice may contribute to nNOS upregulation. However, the phosphodiesterase-5A expression level and the adenosine content in the penis, which are known to play roles in the development of priapism in other etiologies, were unchanged in Fabry mice. In conclusion, these data suggested that increased nNOS (and probably eNOS) content and the consequential elevated NO production and high arterial blood flow in the penis may be the underlying mechanism of priapism in Fabry mice. Furthermore, in combination with previous findings, this study suggested that regulation of NOS expression is susceptible to α-galactosidase A deficiency, and this may represent a general pathogenic mechanism of Fabry vasculopathy.


Assuntos
Doença de Fabry/complicações , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ereção Peniana , Pênis/enzimologia , Priapismo/etiologia , Animais , Modelos Animais de Doenças , Terapia de Reposição de Enzimas/métodos , Doença de Fabry/enzimologia , Doença de Fabry/fisiopatologia , Doença de Fabry/terapia , Terapia Genética/métodos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Pênis/fisiopatologia , Priapismo/enzimologia , Priapismo/fisiopatologia , Priapismo/terapia , Fluxo Sanguíneo Regional , Transdução de Sinais , Regulação para Cima , alfa-Galactosidase/biossíntese , alfa-Galactosidase/genética
12.
Nature ; 488(7409): 111-5, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22859208

RESUMO

Antisense oligonucleotides (ASOs) hold promise for gene-specific knockdown in diseases that involve RNA or protein gain-of-function effects. In the hereditary degenerative disease myotonic dystrophy type 1 (DM1), transcripts from the mutant allele contain an expanded CUG repeat and are retained in the nucleus. The mutant RNA exerts a toxic gain-of-function effect, making it an appropriate target for therapeutic ASOs. However, despite improvements in ASO chemistry and design, systemic use of ASOs is limited because uptake in many tissues, including skeletal and cardiac muscle, is not sufficient to silence target messenger RNAs. Here we show that nuclear-retained transcripts containing expanded CUG (CUG(exp)) repeats are unusually sensitive to antisense silencing. In a transgenic mouse model of DM1, systemic administration of ASOs caused a rapid knockdown of CUG(exp) RNA in skeletal muscle, correcting the physiological, histopathologic and transcriptomic features of the disease. The effect was sustained for up to 1 year after treatment was discontinued. Systemically administered ASOs were also effective for muscle knockdown of Malat1, a long non-coding RNA (lncRNA) that is retained in the nucleus. These results provide a general strategy to correct RNA gain-of-function effects and to modulate the expression of expanded repeats, lncRNAs and other transcripts with prolonged nuclear residence.


Assuntos
Núcleo Celular/genética , Inativação Gênica , Distrofia Miotônica/genética , Distrofia Miotônica/terapia , RNA/antagonistas & inibidores , RNA/genética , Alelos , Animais , Sequência de Bases , Núcleo Celular/efeitos dos fármacos , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofia Miotônica/patologia , Distrofia Miotônica/fisiopatologia , Miotonina Proteína Quinase , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas Serina-Treonina Quinases/genética , RNA/metabolismo , RNA Longo não Codificante , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA não Traduzido/genética , Ribonuclease H/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Expansão das Repetições de Trinucleotídeos/genética
13.
Mol Ther ; 25(2): 331-341, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28109959

RESUMO

As the most common subtype of Leber congenital amaurosis (LCA), LCA10 is a severe retinal dystrophy caused by mutations in the CEP290 gene. The most frequent mutation found in patients with LCA10 is a deep intronic mutation in CEP290 that generates a cryptic splice donor site. The large size of the CEP290 gene prevents its use in adeno-associated virus (AAV)-mediated gene augmentation therapy. Here, we show that targeted genomic deletion using the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system represents a promising therapeutic approach for the treatment of patients with LCA10 bearing the CEP290 splice mutation. We generated a cellular model of LCA10 by introducing the CEP290 splice mutation into 293FT cells and we showed that guide RNA pairs coupled with SpCas9 were highly efficient at removing the intronic splice mutation and restoring the expression of wild-type CEP290. In addition, we demonstrated that a dual AAV system could effectively delete an intronic fragment of the Cep290 gene in the mouse retina. To minimize the immune response to prolonged expression of SpCas9, we developed a self-limiting CRISPR/Cas9 system that minimizes the duration of SpCas9 expression. These results support further studies to determine the therapeutic potential of CRISPR/Cas9-based strategies for the treatment of patients with LCA10.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Amaurose Congênita de Leber/genética , Processamento Alternativo , Animais , Antígenos de Neoplasias/genética , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Feminino , Expressão Gênica , Ordem dos Genes , Marcação de Genes , Loci Gênicos , Íntrons , Amaurose Congênita de Leber/terapia , Camundongos , Mutação , Proteínas de Neoplasias/genética , RNA Guia de Cinetoplastídeos , RNA Mensageiro/genética , Retina/metabolismo , Deleção de Sequência , Reparo Gênico Alvo-Dirigido
14.
Proc Natl Acad Sci U S A ; 112(26): 8100-5, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26056266

RESUMO

Recent genetic evidence suggests that aberrant glycosphingolipid metabolism plays an important role in several neuromuscular diseases including hereditary spastic paraplegia, hereditary sensory neuropathy type 1, and non-5q spinal muscular atrophy. Here, we investigated whether altered glycosphingolipid metabolism is a modulator of disease course in amyotrophic lateral sclerosis (ALS). Levels of ceramide, glucosylceramide, galactocerebroside, lactosylceramide, globotriaosylceramide, and the gangliosides GM3 and GM1 were significantly elevated in spinal cords of ALS patients. Moreover, enzyme activities (glucocerebrosidase-1, glucocerebrosidase-2, hexosaminidase, galactosylceramidase, α-galactosidase, and ß-galactosidase) mediating glycosphingolipid hydrolysis were also elevated up to threefold. Increased ceramide, glucosylceramide, GM3, and hexosaminidase activity were also found in SOD1(G93A) mice, a familial model of ALS. Inhibition of glucosylceramide synthesis accelerated disease course in SOD1(G93A) mice, whereas infusion of exogenous GM3 significantly slowed the onset of paralysis and increased survival. Our results suggest that glycosphingolipids are likely important participants in pathogenesis of ALS and merit further analysis as potential drug targets.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Glicoesfingolipídeos/fisiologia , Esclerose Lateral Amiotrófica/enzimologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Gangliosídeo G(M3)/administração & dosagem , Glucosiltransferases/antagonistas & inibidores , Humanos , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Transgênicos , Medula Espinal/fisiopatologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
15.
Mol Ther ; 24(6): 1019-1029, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26948439

RESUMO

Gaucher disease (GD) is caused by a deficiency of glucocerebrosidase and the consequent lysosomal accumulation of unmetabolized glycolipid substrates. Enzyme-replacement therapy adequately manages the visceral manifestations of nonneuronopathic type-1 Gaucher patients, but not the brain disease in neuronopathic types 2 and 3 GD. Substrate reduction therapy through inhibition of glucosylceramide synthase (GCS) has also been shown to effectively treat the visceral disease. Here, we evaluated the efficacy of a novel small molecule inhibitor of GCS with central nervous system (CNS) access (Genz-682452) to treat the brain disease. Treatment of the conduritol ß epoxide-induced mouse model of neuronopathic GD with Genz-682452 reduced the accumulation of liver and brain glycolipids (>70% and >20% respectively), extent of gliosis, and severity of ataxia. In the genetic 4L;C* mouse model, Genz-682452 reduced the levels of substrate in the brain by >40%, the extent of gliosis, and paresis. Importantly, Genz-682452-treated 4L;C* mice also exhibited an ~30% increase in lifespan. Together, these data indicate that an orally available antagonist of GCS that has CNS access is effective at attenuating several of the neuropathologic and behavioral manifestations associated with mouse models of neuronopathic GD. Therefore, Genz-682452 holds promise as a potential therapeutic approach for patients with type-3 GD.


Assuntos
Carbamatos/administração & dosagem , Sistema Nervoso Central/metabolismo , Inibidores Enzimáticos/administração & dosagem , Doença de Gaucher/tratamento farmacológico , Glucosiltransferases/antagonistas & inibidores , Glicolipídeos/metabolismo , Quinuclidinas/administração & dosagem , Administração Oral , Animais , Carbamatos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/induzido quimicamente , Doença de Gaucher/metabolismo , Humanos , Inositol/análogos & derivados , Fígado/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Camundongos , Quinuclidinas/farmacologia , Distribuição Tecidual , Resultado do Tratamento
16.
Hum Mol Genet ; 23(3): 730-48, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24057669

RESUMO

The GM2 gangliosidoses are progressive neurodegenerative disorders due to defects in the lysosomal ß-N-acetylhexosaminidase system. Accumulation of ß-hexosaminidases A and B substrates is presumed to cause this fatal condition. An authentic mouse model of Sandhoff disease (SD) with pathological characteristics resembling those noted in infantile GM2 gangliosidosis has been described. We have shown that expression of ß-hexosaminidase by intracranial delivery of recombinant adeno-associated viral vectors to young adult SD mice can prevent many features of the disease and extends lifespan. To investigate the nature of the neurological injury in GM2 gangliosidosis and the extent of its reversibility, we have examined the evolution of disease in the SD mouse; we have moreover explored the effects of gene transfer delivered at key times during the course of the illness. Here we report greatly increased survival only when the therapeutic genes are expressed either before the disease is apparent or during its early manifestations. However, irrespective of when treatment was administered, widespread and abundant expression of ß-hexosaminidase with consequent clearance of glycoconjugates, α-synuclein and ubiquitinated proteins, and abrogation of inflammatory responses and neuronal loss was observed. We also show that defects in myelination occur in early life and cannot be easily resolved when treatment is given to the adult brain. These results indicate that there is a limited temporal opportunity in which function and survival can be improved-but regardless of resolution of the cardinal pathological features of GM2 gangliosidosis, a point is reached when functional deterioration and death cannot be prevented.


Assuntos
Encéfalo/enzimologia , Vetores Genéticos/farmacologia , Doença de Sandhoff/genética , Doença de Sandhoff/patologia , Doença de Sandhoff/terapia , Doença de Tay-Sachs/patologia , beta-N-Acetil-Hexosaminidases/genética , Animais , Encéfalo/efeitos dos fármacos , Dependovirus/genética , Modelos Animais de Doenças , Gangliosídeo G(M2)/genética , Gangliosídeo G(M2)/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intralesionais , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Doença de Sandhoff/mortalidade , Ubiquitina/metabolismo , alfa-Sinucleína/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
17.
Am J Pathol ; 185(3): 651-65, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25553976

RESUMO

Fabry disease is an X-linked lysosomal storage disease caused by deficient activity of α-galactosidase A and the resultant systemic accumulation of globotrioasylceramide (GL-3) and related glycolipids. α-Galactosidase A gene knockout (Gla KO) mice have no α-galactosidase A activity and progressively accumulate GL-3 in tissues and fluids, similarly to FD patients. The nature and temporal effects of the progressive substrate accumulation on tissue histology in these mice have not previously been characterized. Here, we report the pathology of young to old (3 to 17 months old) Gla KO mice and compare these changes with those in strain-matched control animals. Gla KO mice accumulated GL-3 in various tissues and fluids with age. Lysosomal GL-3 inclusions increased with age in multiple cell types, including renal epithelial, intestinal, and vascular smooth muscle cells, and neurons in trigeminal and dorsal root ganglia, as detected by light and electron microscopy. However, unlike the case for male FD patients with the type 1 classic phenotype, GL-3 inclusions were not detected in vascular endothelial cells or cardiomyocytes. The histological changes in Gla KO mice better resemble the type 2 later-onset phenotype observed in patients with residual α-galactosidase A activity. GL-3 accumulation in the small intestine and sensory ganglia of Gla KO mice provides a model for study of enteropathy and neuropathy in Fabry disease.


Assuntos
Doença de Fabry/patologia , Intestinos/patologia , Rim/patologia , Músculo Liso Vascular/patologia , alfa-Galactosidase/genética , Animais , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , Modelos Animais de Doenças , Progressão da Doença , Doença de Fabry/genética , Doença de Fabry/metabolismo , Feminino , Humanos , Mucosa Intestinal/metabolismo , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , alfa-Galactosidase/metabolismo
18.
Mol Genet Metab ; 117(4): 447-55, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26857995

RESUMO

Mucolipidoses II and III (ML II and ML III) are lysosomal disorders in which the mannose 6-phosphate recognition marker is absent from lysosomal hydrolases and other glycoproteins due to mutations in GNPTAB, which encodes two of three subunits of the heterohexameric enzyme, N-acetylglucosamine-1-phosphotransferase. Both disorders are caused by the same gene, but ML II represents the more severe phenotype. Bone manifestations of ML II include hip dysplasia, scoliosis, rickets and osteogenesis imperfecta. In this study, we sought to determine whether a recombinant adeno-associated viral vector (AAV2/8-GNPTAB) could confer high and prolonged gene expression of GNPTAB and thereby influence the pathology in the cartilage and bone tissue of a GNPTAB knock out (KO) mouse model. The results demonstrated significant increases in bone mineral density and content in AAV2/8-GNPTAB-treated as compared to non-treated KO mice. We also showed that IL-6 (interleukin-6) expression in articular cartilage was reduced in AAV2/8-GNPTAB treated ML II mice. Together, these data suggest that AAV-mediated expression of GNPTAB in ML II mice can attenuate bone loss via inhibition of IL-6 production. This study emphasizes the value of the MLII KO mouse to recapitulate the clinical manifestations of the disease and highlights its amenability to therapy.


Assuntos
Desmineralização Patológica Óssea/etiologia , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/genética , Mucolipidoses/genética , Mucolipidoses/patologia , Transdução Genética , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Animais , Desmineralização Patológica Óssea/diagnóstico , Desmineralização Patológica Óssea/terapia , Densidade Óssea , Modelos Animais de Doenças , Ordem dos Genes , Marcação de Genes , Loci Gênicos , Terapia Genética , Vetores Genéticos/administração & dosagem , Genótipo , Humanos , Camundongos , Camundongos Knockout , Mucolipidoses/terapia , Fenótipo
19.
J Inherit Metab Dis ; 39(3): 447-455, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26960552

RESUMO

Fabry disease is caused by deficient activity of α-galactosidase A and subsequent intracellular accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3). Vascular endothelial cells may play important roles in disease pathogenesis, and are one of the main target cell types in therapeutic interventions. In this study, we generated immortalized aortic endothelial cell lines from a mouse model of Fabry disease. These cells retained endothelial cell-specific markers and functions. Gb3 expression level in one of these clones (referred to as FMEC2) was highly susceptible to culture media, and appeared to be regulated by glucosylceramide synthase. Results also showed that Gb3 could be upregulated by hydrocortisone. FMEC2 express the mannose 6-phosphate receptor and sortilin but not the mannose receptor. Uptake studies suggested that sortilin plays a role in the binding and internalization of mammalian cell-produced α-galactosidase A. Moss-aGal (a plant-made enzyme) was endocytosed by FMEC2 via a receptor other than the aforementioned receptors. In conclusion, this study suggests that glucosylceramide synthase and hydrocortisone may play important roles in modulating Gb3 levels in Fabry mouse aortic endothelial cells, and that endocytosis of recombinant α-galactosidase A involves a combination of multiple receptors depending on the properties of the enzyme.


Assuntos
Aorta/metabolismo , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Doença de Fabry/enzimologia , Doença de Fabry/metabolismo , Triexosilceramidas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Endocitose/fisiologia , Endotélio Vascular/enzimologia , Glucosiltransferases/metabolismo , Glicoesfingolipídeos/metabolismo , Lectinas Tipo C/metabolismo , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor IGF Tipo 2/metabolismo , Receptores de Superfície Celular/metabolismo , alfa-Galactosidase/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 35(4): 911-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25614286

RESUMO

OBJECTIVE: Studies of mice with mild Marfan syndrome (MFS) have correlated the development of thoracic aortic aneurysm (TAA) with improper stimulation of noncanonical (Erk-mediated) TGFß signaling by the angiotensin type I receptor (AT1r). This correlation was largely based on comparable TAA modifications by either systemic TGFß neutralization or AT1r antagonism. However, subsequent investigations have called into question some key aspects of this mechanism of arterial disease in MFS. To resolve these controversial points, here we made a head-to-head comparison of the therapeutic benefits of TGFß neutralization and AT1r antagonism in mice with progressively severe MFS (Fbn1(mgR/mgR) mice). APPROACH AND RESULTS: Aneurysm growth, media degeneration, aortic levels of phosphorylated Erk and Smad proteins and the average survival of Fbn1(mgR/mgR) mice were compared after a ≈3-month-long treatment with placebo and either the AT1r antagonist losartan or the TGFß-neutralizing antibody 1D11. In contrast to the beneficial effect of losartan, TGFß neutralization either exacerbated or mitigated TAA formation depending on whether treatment was initiated before (postnatal day 16; P16) or after (P45) aneurysm formation, respectively. Biochemical evidence-related aneurysm growth with Erk-mediated AT1r signaling, and medial degeneration with TGFß hyperactivity that was in part AT1r dependent. Importantly, P16-initiated treatment with losartan combined with P45-initiated administration of 1D11 prevented death of Fbn1(mgR/mgR) mice from ruptured TAA. CONCLUSIONS: By demonstrating that promiscuous AT1r and TGFß drive partially overlapping processes of arterial disease in MFS mice, our study argues for a therapeutic strategy against TAA that targets both signaling pathways although sparing the early protective role of TGFß.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Anticorpos Neutralizantes/farmacologia , Aorta Torácica/efeitos dos fármacos , Aneurisma da Aorta Torácica/prevenção & controle , Losartan/farmacologia , Síndrome de Marfan/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/patologia , Ruptura Aórtica/genética , Ruptura Aórtica/metabolismo , Ruptura Aórtica/patologia , Ruptura Aórtica/prevenção & controle , Modelos Animais de Doenças , Progressão da Doença , Fibrilina-1 , Fibrilinas , Humanos , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Síndrome de Marfan/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas dos Microfilamentos/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutação , Fosforilação , Receptor Tipo 1 de Angiotensina/metabolismo , Proteína Smad2/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA