Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Mol Genet Metab ; 133(4): 378-385, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34154922

RESUMO

Mucopolysaccharidosis (MPS) VII is a lysosomal storage disorder characterized by deficient ß-glucuronidase activity, leading to accumulation of incompletely degraded heparan, dermatan and chondroitin sulfate glycosaminoglycans. Patients with MPS VII exhibit progressive spinal deformity, which decreases quality of life. Previously, we demonstrated that MPS VII dogs exhibit impaired initiation of secondary ossification in the vertebrae and long bones. The objective of this study was to build on these findings and comprehensively characterize how vertebral bone disease manifests progressively in MPS VII dogs throughout postnatal growth. Vertebrae were collected postmortem from MPS VII and healthy control dogs at seven ages ranging from 9 to 365 days. Microcomputed tomography and histology were used to characterize bone properties in primary and secondary ossification centers. Serum was analyzed for bone turnover biomarkers. Results demonstrated that not only was secondary ossification delayed in MPS VII vertebrae, but that it progressed aberrantly and was markedly diminished even at 365 days-of-age. Within primary ossification centers, bone volume fraction and bone mineral density were significantly lower in MPS VII at 180 and 365 days-of-age. MPS VII growth plates exhibited significantly lower proliferative and hypertrophic zone cellularity at 90 days-of-age, while serum bone-specific alkaline phosphatase (BAP) was significantly lower in MPS VII dogs at 180 days-of-age. Overall, these findings establish that vertebral bone formation is significantly diminished in MPS VII dogs in both primary and secondary ossification centers during postnatal growth.


Assuntos
Doenças Ósseas/fisiopatologia , Progressão da Doença , Mucopolissacaridose VII/complicações , Coluna Vertebral/patologia , Animais , Animais Recém-Nascidos , Doenças Ósseas/genética , Osso e Ossos/patologia , Cães , Feminino , Crescimento e Desenvolvimento , Masculino , Mucopolissacaridose VII/genética , Osteogênese
2.
Mol Genet Metab ; 122S: 25-34, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29153844

RESUMO

The mucopolysaccharidosis (MPS) disorders are a group of lysosomal storage diseases caused by lysosomal enzyme deficits that lead to glycosaminoglycan accumulation, affecting various tissues throughout the body based on the specific enzyme deficiency. These disorders are characterized by their progressive nature and a variety of somatic manifestations and neurological symptoms. There are established treatments for some MPS disorders, but these mostly alleviate somatic and non-neurological symptoms and do not cure the disease. Patients with MPS I, II, III, and VII can present with neurological manifestations such as neurocognitive decline and behavioral problems. Treatment of these neurological manifestations remains challenging due to the blood-brain barrier (BBB) that limits delivery of therapeutic agents to the central nervous system (CNS). New therapies that circumvent this barrier and target brain disease in MPS are currently under development. They primarily focus on facilitating penetration of drugs through the BBB, delivery of recombinant enzyme to the brain by gene therapy, or direct CNS administration. This review summarizes existing and potential future treatment approaches that target brain disease in MPS. The information in this review is based on current literature and presentations and discussions during a closed meeting by an international group of experts with extensive experience in managing and treating MPS.


Assuntos
Encéfalo/efeitos dos fármacos , Disfunção Cognitiva/terapia , Terapia de Reposição de Enzimas/métodos , Mucopolissacaridoses/terapia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/metabolismo , Criança , Comportamento Infantil/efeitos dos fármacos , Desenvolvimento Infantil/efeitos dos fármacos , Pré-Escolar , Ensaios Clínicos como Assunto , Disfunção Cognitiva/diagnóstico , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Congressos como Assunto , Portadores de Fármacos/química , Terapia Genética/métodos , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/toxicidade , Transplante de Células-Tronco Hematopoéticas , Humanos , Injeções Intraventriculares , Injeções Espinhais , Chaperonas Moleculares/uso terapêutico , Mucopolissacaridoses/diagnóstico , Mucopolissacaridoses/genética , Mucopolissacaridoses/patologia , Nanopartículas/química , Proteínas Recombinantes/uso terapêutico
3.
Mol Ther ; 24(2): 206-216, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26447927

RESUMO

Mucopolysaccharidosis VII (MPS VII) is a lysosomal storage disease arising from mutations in ß-d-glucuronidase (GUSB), which results in glycosaminoglycan (GAG) accumulation and a variety of clinical manifestations including neurological disease. Herein, MPS VII dogs were injected intravenously (i.v.) and/or intrathecally (i.t.) via the cisterna magna with AAV9 or AAVrh10 vectors carrying the canine GUSB cDNA. Although i.v. injection alone at 3 days of age resulted in normal cerebrospinal fluid (CSF) GUSB activity, brain tissue homogenates had only ~1 to 6% normal GUSB activity and continued to have elevated GAG storage. In contrast, i.t. injection at 3 weeks of age resulted in CSF GUSB activity 44-fold normal while brain tissue homogenates had >100% normal GUSB activity and reduced GAGs compared with untreated dogs. Markers for secondary storage and inflammation were eliminated in i.t.-treated dogs and reduced in i.v.-treated dogs compared with untreated dogs. Given that i.t.-treated dogs expressed higher levels of GUSB in the CNS tissues compared to those treated i.v., we conclude that i.t. injection of AAV9 or AAVrh10 vectors is more effective than i.v. injection alone in the large animal model of MPS VII.


Assuntos
Doenças do Sistema Nervoso Central/terapia , Terapia Genética/métodos , Glucuronidase/genética , Mucopolissacaridose VII/terapia , Animais , Animais Recém-Nascidos , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Cães , Vetores Genéticos/administração & dosagem , Glucuronidase/líquido cefalorraquidiano , Glicosaminoglicanos/metabolismo , Injeções Intravenosas , Injeções Espinhais , Masculino , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(41): 14894-9, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25267637

RESUMO

Patients with mucopolysaccharidosis type I (MPS I), a genetic deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), exhibit accumulation of glycosaminoglycans in tissues, with resulting diverse clinical manifestations including neurological, ocular, skeletal, and cardiac disease. MPS I is currently treated with hematopoietic stem cell transplantation or weekly enzyme infusions, but these therapies have significant drawbacks for patient safety and quality of life and do not effectively address some of the most critical clinical sequelae, such as life-threatening cardiac valve involvement. Using the naturally occurring feline model of MPS I, we tested liver-directed gene therapy as a means of achieving long-term systemic IDUA reconstitution. We treated four MPS I cats at 3-5 mo of age with an adeno-associated virus serotype 8 vector expressing feline IDUA from a liver-specific promoter. We observed sustained serum enzyme activity for 6 mo at ∼ 30% of normal levels in one animal, and in excess of normal levels in three animals. Remarkably, treated animals not only demonstrated reductions in glycosaminoglycan storage in most tissues, but most also exhibited complete resolution of aortic valve lesions, an effect that has not been previously observed in this animal model or in MPS I patients treated with current therapies. These data point to clinically meaningful benefits of the robust enzyme expression achieved with hepatic gene transfer that extend beyond the economic and quality of life advantages over lifelong enzyme infusions.


Assuntos
Doenças Cardiovasculares/terapia , Terapia Genética , Fígado/metabolismo , Mucopolissacaridose I/terapia , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Doenças Cardiovasculares/patologia , Gatos , Dependovirus/genética , Feminino , Vetores Genéticos/metabolismo , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/metabolismo , Iduronidase/sangue , Iduronidase/genética , Iduronidase/uso terapêutico , Fígado/patologia , Masculino , Dados de Sequência Molecular , Mucopolissacaridose I/sangue , Mucopolissacaridose I/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Trombina/metabolismo , Distribuição Tecidual , Transdução Genética
5.
Mol Genet Metab ; 119(1-2): 124-30, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27386755

RESUMO

High fidelity animal models of human disease are essential for preclinical evaluation of novel gene and protein therapeutics. However, these studies can be complicated by exaggerated immune responses against the human transgene. Here we demonstrate that dogs with a genetic deficiency of the enzyme α-l-iduronidase (IDUA), a model of the lysosomal storage disease mucopolysaccharidosis type I (MPS I), can be rendered immunologically tolerant to human IDUA through neonatal exposure to the enzyme. Using MPS I dogs tolerized to human IDUA as neonates, we evaluated intrathecal delivery of an adeno-associated virus serotype 9 vector expressing human IDUA as a therapy for the central nervous system manifestations of MPS I. These studies established the efficacy of the human vector in the canine model, and allowed for estimation of the minimum effective dose, providing key information for the design of first-in-human trials. This approach can facilitate evaluation of human therapeutics in relevant animal models, and may also have clinical applications for the prevention of immune responses to gene and protein replacement therapies.


Assuntos
Terapia de Reposição de Enzimas , Iduronidase/genética , Doenças por Armazenamento dos Lisossomos/terapia , Mucopolissacaridose I/terapia , Animais , Modelos Animais de Doenças , Cães , Terapia Genética , Vetores Genéticos , Glicosaminoglicanos/metabolismo , Humanos , Iduronidase/deficiência , Iduronidase/uso terapêutico , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/patologia , Mucopolissacaridose I/genética , Mucopolissacaridose I/patologia , Transgenes
6.
J Exp Biol ; 219(Pt 12): 1866-74, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27045093

RESUMO

The surface area of the maxilloturbinals and fronto-ethmoturbinals is commonly used as an osteological proxy for the respiratory and the olfactory epithelium, respectively. However, this assumption does not fully account for animals with short snouts in which these two turbinal structures significantly overlap, potentially placing fronto-ethmoturbinals in the path of respiratory airflow. In these species, it is possible that anterior fronto-ethmoturbinals are covered with non-sensory (respiratory) epithelium instead of olfactory epithelium. In this study, we analyzed the distribution of olfactory and non-sensory, respiratory epithelia on the turbinals of two domestic cats (Felis catus) and a bobcat (Lynx rufus). We also conducted a computational fluid dynamics simulation of nasal airflow in the bobcat to explore the relationship between epithelial distribution and airflow patterns. The results showed that a substantial amount of respiratory airflow passes over the anterior fronto-ethmoturbinals, and that contrary to what has been observed in caniform carnivorans, much of the anterior ethmoturbinals are covered by non-sensory epithelium. This confirms that in short-snouted felids, portions of the fronto-ethmoturbinals have been recruited for respiration, and that estimates of olfactory epithelial coverage based purely on fronto-ethmoturbinal surface area will be exaggerated. The correlation between the shape of the anterior fronto-ethmoturbinals and the direction of respiratory airflow suggests that in short-snouted species, CT data alone are useful in assessing airflow patterns and epithelium distribution on the turbinals.


Assuntos
Gatos/fisiologia , Lynx/fisiologia , Cavidade Nasal/fisiologia , Ventilação Pulmonar , Mucosa Respiratória/fisiologia , Animais , Masculino , Mucosa Olfatória/fisiologia
7.
Mol Ther ; 23(8): 1298-1307, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26022732

RESUMO

The potential host immune response to a nonself protein poses a fundamental challenge for gene therapies targeting recessive diseases. We demonstrate in both dogs and nonhuman primates that liver-directed gene transfer using an adeno-associated virus (AAV) vector in neonates induces a persistent state of immunological tolerance to the transgene product, substantially improving the efficacy of subsequent vector administration targeting the central nervous system (CNS). We applied this approach to a canine model of mucopolysaccharidosis type I (MPS I), a progressive neuropathic lysosomal storage disease caused by deficient activity of the enzyme α-l-iduronidase (IDUA). MPS I dogs treated systemically in the first week of life with a vector expressing canine IDUA did not develop antibodies against the enzyme and exhibited robust expression in the CNS upon intrathecal AAV delivery at 1 month of age, resulting in complete correction of brain storage lesions. Newborn rhesus monkeys treated systemically with AAV vector expressing human IDUA developed tolerance to the transgene, resulting in high cerebrospinal fluid (CSF) IDUA expression and no antibody induction after subsequent CNS gene therapy. These findings suggest that inducing tolerance to the transgene product during a critical period in immunological development can improve the efficacy and safety of gene therapy.


Assuntos
Sistema Nervoso Central/metabolismo , Dependovirus/genética , Terapia Genética/métodos , Iduronidase/genética , Mucopolissacaridose I/genética , Mucopolissacaridose I/terapia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Cães , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Células HEK293 , Humanos , Iduronidase/deficiência , Macaca mulatta , Transgenes
8.
Mol Genet Metab ; 116(3): 195-203, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26422116

RESUMO

Mucopolysaccharidosis (MPS) VII is a lysosomal storage disorder characterized by deficient ß-glucuronidase activity, which leads to the accumulation of incompletely degraded glycosaminoglycans (GAGs). MPS VII patients present with severe skeletal abnormalities, which are particularly prevalent in the spine. Incomplete cartilage-to-bone conversion in MPS VII vertebrae during postnatal development is associated with progressive spinal deformity and spinal cord compression. The objectives of this study were to determine the earliest postnatal developmental stage at which vertebral bone disease manifests in MPS VII and to identify the underlying cellular basis of impaired cartilage-to-bone conversion, using the naturally-occurring canine model. Control and MPS VII dogs were euthanized at 9 and 14 days-of-age, and vertebral secondary ossification centers analyzed using micro-computed tomography, histology, qPCR, and protein immunoblotting. Imaging studies and mRNA analysis of bone formation markers established that secondary ossification commences between 9 and 14 days in control animals, but not in MPS VII animals. mRNA analysis of differentiation markers revealed that MPS VII epiphyseal chondrocytes are unable to successfully transition from proliferation to hypertrophy during this critical developmental window. Immunoblotting demonstrated abnormal persistence of Sox9 protein in MPS VII cells between 9 and 14 days-of-age, and biochemical assays revealed abnormally high intra and extracellular GAG content in MPS VII epiphyseal cartilage at as early as 9 days-of-age. In contrast, assessment of vertebral growth plates and primary ossification centers revealed no significant abnormalities at either age. The results of this study establish that failed vertebral bone formation in MPS VII can be traced to the failure of epiphyseal chondrocytes to undergo hypertrophic differentiation at the appropriate developmental stage, and suggest that aberrant processing of Sox9 protein may contribute to this cellular dysfunction. These results also highlight the importance of early diagnosis and therapeutic intervention to prevent the progression of debilitating skeletal disease in MPS patients.


Assuntos
Condrócitos/citologia , Epífises/citologia , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/fisiopatologia , Osteogênese , Animais , Doenças Ósseas/etiologia , Doenças Ósseas/fisiopatologia , Diferenciação Celular , Cães , Glicosaminoglicanos/metabolismo , Humanos , Hipertrofia , Coluna Vertebral/fisiologia , Microtomografia por Raio-X
9.
Mol Ther ; 22(4): 762-73, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24343103

RESUMO

Severe deficiency in lysosomal ß-glucuronidase (ß-glu) enzymatic activity results in mucopolysaccharidosis (MPS) VII, an orphan disease with symptoms often appearing in early childhood. Symptoms are variable, but many patients have multiple organ disorders including neurological defects. At the cellular level, deficiency in ß-glu activity leads to abnormal accumulation of glycosaminoglycans (GAGs), and secondary accumulation of GM2 and GM3 gangliosides, which have been linked to neuroinflammation. There have been encouraging gene transfer studies in the MPS VII mouse brain, but this is the first study attempting the correction of the >200-fold larger and challenging canine MPS VII brain. Here, the efficacy of a helper-dependent (HD) canine adenovirus (CAV-2) vector harboring a human GUSB expression cassette (HD-RIGIE) in the MPS VII dog brain was tested. Vector genomes, ß-glu activity, GAG content, lysosome morphology and neuropathology were analyzed and quantified. Our data demonstrated that CAV-2 vectors preferentially transduced neurons and axonal retrograde transport from the injection site to efferent regions was efficient. HD-RIGIE injections, associated with mild and transient immunosuppression, corrected neuropathology in injected and noninjected structures throughout the cerebrum. These data support the clinical evaluation of HD CAV-2 vectors to treat the neurological defects associated with MPS VII and possibly other neuropathic lysosomal storage diseases.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Mucopolissacaridose VII/genética , beta-Glucosidase/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Cães , Regulação Enzimológica da Expressão Gênica , Glicosaminoglicanos/metabolismo , Humanos , Camundongos , Mucopolissacaridose VII/terapia , Mucopolissacaridose VII/veterinária , beta-Glucosidase/administração & dosagem , beta-Glucosidase/biossíntese
10.
Mol Ther ; 22(12): 2018-2027, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25027660

RESUMO

Enzyme replacement therapy has revolutionized the treatment of the somatic manifestations of lysosomal storage diseases (LSD), although it has been ineffective in treating central nervous system (CNS) manifestations of these disorders. The development of neurotrophic vectors based on novel serotypes of adeno-associated viruses (AAV) such as AAV9 provides a potential platform for stable and efficient delivery of enzymes to the CNS. We evaluated the safety and efficacy of intrathecal delivery of AAV9 expressing α-l-iduronidase (IDUA) in a previously described feline model of mucopolysaccharidosis I (MPS I). A neurological phenotype has not been defined in these animals, so our analysis focused on the biochemical and histological CNS abnormalities characteristic of MPS I. Five MPS I cats were dosed with AAV9 vector at 4-7 months of age and followed for 6 months. Treated animals demonstrated virtually complete correction of biochemical and histological manifestations of the disease throughout the CNS. There was a range of antibody responses against IDUA in this cohort which reduced detectable enzyme without substantially reducing efficacy; there was no evidence of toxicity. This first demonstration of the efficacy of intrathecal gene therapy in a large animal model of a LSD should pave the way for translation into the clinic.


Assuntos
Gatos , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Terapia Genética/métodos , Iduronidase/sangue , Iduronidase/líquido cefalorraquidiano , Mucopolissacaridose I/terapia , Animais , Dependovirus/enzimologia , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Injeções Espinhais , Mucopolissacaridose I/enzimologia , Mucopolissacaridose I/genética , Mucopolissacaridose I/patologia , Especificidade de Órgãos
11.
Mol Genet Metab ; 110(3): 311-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23860311

RESUMO

Mucopolysaccharidosis VII (MPS VII) is due to deficient activity of the lysosomal enzyme ß-glucuronidase (GUSB) and results in the accumulation of glycosaminoglycans (GAGs). This study determined the long-term effect of neonatal intravenous injection of a gamma retroviral vector (RV) on cardiac valve disease in MPS VII dogs. Transduced hepatocytes secreted GUSB into the blood for up to 11 years at levels similar to or greater than those achieved with enzyme replacement therapy (ERT). Valve regurgitation and thickening were scored from 0 (normal) to +4 (severely abnormal). At 1 year, untreated MPS VII dogs had mitral regurgitation, mitral valve thickening, aortic regurgitation, and aortic valve thickening scores of 2.3 ± 0.7, 2.3 ± 0.6, 1.8 ± 0.5, and 1.6 ± 0.7, respectively, which were higher than the values of 0.6 ± 0.1, 0.1 ± 0.4, 0.3 ± 0.8, and 0.1 ± 0.4, respectively, in treated MPS VII dogs. Treated MPS VII dogs maintained low aortic regurgitation and aortic valve thickening scores in their lifetime. Although mitral regurgitation and mitral valve thickening scores increased to 2.0 at ≥ 8 years of age in the treated MPS VII dogs, older normal dogs from the colony had similar scores, making it difficult to assess mitral valve disease. Older treated dogs had calcification within the mitral and the aortic valve annulus, while GUSB staining demonstrated enzyme activity within the mitral valve. We conclude that neonatal RV-mediated gene therapy reduced cardiac valve disease in MPS VII dogs for up to 11 years, and propose that neonatal initiation of ERT should have a similar effect.


Assuntos
Gammaretrovirus/genética , Terapia Genética , Vetores Genéticos/genética , Doenças das Valvas Cardíacas/etiologia , Doenças das Valvas Cardíacas/terapia , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/genética , Animais , Animais Recém-Nascidos , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/patologia , Modelos Animais de Doenças , Cães , Ecocardiografia , Feminino , Glucuronidase/genética , Glucuronidase/metabolismo , Doenças das Valvas Cardíacas/diagnóstico por imagem , Masculino , Valva Mitral/diagnóstico por imagem , Valva Mitral/patologia
12.
Mol Genet Metab ; 109(2): 183-93, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23628461

RESUMO

Mucopolysaccharidosis (MPS) VII is a lysosomal storage disease due to deficient activity of ß-glucuronidase (GUSB), and results in glycosaminoglycan accumulation. Skeletal manifestations include bone dysplasia, degenerative joint disease, and growth retardation. One gene therapy approach for MPS VII involves neonatal intravenous injection of a gamma retroviral vector expressing GUSB, which results in stable expression in liver and secretion of enzyme into blood at levels predicted to be similar or higher to enzyme replacement therapy. The goal of this study was to evaluate the long-term effect of neonatal gene therapy on skeletal manifestations in MPS VII dogs. Treated MPS VII dogs could walk throughout their lives, while untreated MPS VII dogs could not stand beyond 6 months and were dead by 2 years. Luxation of the coxofemoral joint and the patella, dysplasia of the acetabulum and supracondylar ridge, deep erosions of the distal femur, and synovial hyperplasia were reduced, and the quality of articular bone was improved in treated dogs at 6 to 11 years of age compared with untreated MPS VII dogs at 2 years or less. However, treated dogs continued to have osteophyte formation, cartilage abnormalities, and an abnormal gait. Enzyme activity was found near synovial blood vessels, and there was 2% as much GUSB activity in synovial fluid as in serum. We conclude that neonatal gene therapy reduces skeletal abnormalities in MPS VII dogs, but clinically-relevant abnormalities remain. Enzyme replacement therapy will probably have similar limitations long-term.


Assuntos
Glucuronidase/genética , Mucopolissacaridose VII/terapia , Animais , Animais Recém-Nascidos , Cães , Feminino , Cabeça do Fêmur/patologia , Terapia Genética , Glucuronidase/metabolismo , Membro Posterior/patologia , Cápsula Articular/irrigação sanguínea , Cápsula Articular/enzimologia , Articulações/patologia , Masculino , Mucopolissacaridose VII/diagnóstico por imagem , Mucopolissacaridose VII/patologia , Radiografia , Resultado do Tratamento
13.
Mol Genet Metab ; 110(3): 319-28, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23856419

RESUMO

Mucopolysaccharidosis VII (MPS VII) is due to the deficient activity of ß-glucuronidase (GUSB) and results in the accumulation of glycosaminoglycans (GAGs) in lysosomes and multisystemic disease with cardiovascular manifestations. The goal here was to determine the pathogenesis of mitral valve (MV) disease in MPS VII dogs. Untreated MPS VII dogs had a marked reduction in the histochemical signal for structurally-intact collagen in the MV at 6 months of age, when mitral regurgitation had developed. Electron microscopy demonstrated that collagen fibrils were of normal diameter, but failed to align into large parallel arrays. mRNA analysis demonstrated a modest reduction in the expression of genes that encode collagen or collagen-associated proteins such as the proteoglycan decorin which helps collagen fibrils assemble, and a marked increase for genes that encode proteases such as cathepsins. Indeed, enzyme activity for cathepsin B (CtsB) was 19-fold normal. MPS VII dogs that received neonatal intravenous injection of a gamma retroviral vector had an improved signal for structurally-intact collagen, and reduced CtsB activity relative to that seen in untreated MPS VII dogs. We conclude that MR in untreated MPS VII dogs was likely due to abnormalities in MV collagen structure. This could be due to upregulation of enzymes that degrade collagen or collagen-associated proteins, to the accumulation of GAGs that compete with proteoglycans such as decorin for binding to collagen, or to other causes. Further delineation of the etiology of abnormal collagen structure may lead to treatments that improve biomechanical properties of the MV and other tissues.


Assuntos
Doenças das Valvas Cardíacas/etiologia , Valva Mitral/patologia , Mucopolissacaridose VII/complicações , Animais , Cordas Tendinosas/metabolismo , Colágeno/metabolismo , Cães , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Glicosaminoglicanos/metabolismo , Doenças das Valvas Cardíacas/metabolismo , Masculino , Valva Mitral/metabolismo , Mucopolissacaridose VII/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Transdução de Sinais
14.
Mol Ther ; 20(5): 898-907, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22395531

RESUMO

Mucopolysaccharidosis (MPS) VI is due to a deficiency in the activity of N-acetylgalactosamine 4-sulfatase (4S), also known as arylsulfatase B. Previously, retroviral vector (RV)-mediated neonatal gene therapy reduced the clinical manifestations of MPS I and MPS VII in mice and dogs. However, sulfatases require post-translational modification by sulfatase-modifying factors. MPS VI cats were injected intravenously (i.v.) with a gamma RV-expressing feline 4S, resulting in 5 ± 3 copies of RV per 100 cells in liver. Liver and serum 4S activity were 1,450 ± 1,720 U/mg (26-fold normal) and 107 ± 60 U/ml (13-fold normal), respectively, and were directly proportional to the liver 4S protein levels for individual cats. This study suggests that sulfatase-modifying factor (SUMF) activity in liver was sufficient to result in active enzyme despite overexpression of 4S. RV-treated MPS VI cats achieved higher body weights and longer appendicular skeleton lengths, had reduced articular cartilage erosion, and reduced aortic valve thickening and aortic dilatation compared with untreated MPS VI cats, although cervical vertebral bone lengths were not improved. This demonstrates that therapeutic expression of a functional sulfatase protein can be achieved with neonatal gene therapy using a gamma RV, but some aspects of bone disease remain difficult to treat.


Assuntos
Doenças do Gato/terapia , Vírus da Leucemia Murina de Moloney/genética , Mucopolissacaridose VI/veterinária , N-Acetilgalactosamina-4-Sulfatase/genética , Animais , Animais Recém-Nascidos , Peso Corporal , Doenças do Gato/enzimologia , Doenças do Gato/genética , Gatos , Feminino , Terapia Genética , Vetores Genéticos , Injeções Intravenosas , Masculino , Mucopolissacaridose VI/enzimologia , Mucopolissacaridose VI/genética , Mucopolissacaridose VI/terapia , N-Acetilgalactosamina-4-Sulfatase/metabolismo , Processamento de Proteína Pós-Traducional
15.
Hum Mol Genet ; 19(4): 584-96, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19934113

RESUMO

Human acute intermittent porphyria (AIP), the most common acute hepatic porphyria, is an autosomal dominant inborn error of heme biosynthesis due to the half-normal activity of hydroxymethylbilane synthase (HMB-synthase). Here, we describe the first naturally occurring animal model of AIP in four unrelated cat lines who presented phenotypically as congenital erythropoietic porphyria (CEP). Affected cats had erythrodontia, brownish urine, fluorescent bones, and markedly elevated urinary uroporphyrin (URO) and coproporphyrin (COPRO) consistent with CEP. However, their uroporphyrinogen-III-synthase (URO-synthase) activities (deficient in CEP) were normal. Notably, affected cats had half-normal HMB-synthase activities and elevated urinary 5-aminolevulinic acid (ALA) and porphobilinogen (PBG), the deficient enzyme and accumulated metabolites in human AIP. Sequencing the feline HMB-synthase gene revealed different mutations in each line: a duplication (c.189dupT), an in-frame 3 bp deletion (c.842_844delGAG) identical to that causing human AIP and two missense mutations, c.250G>A (p.A84T) and c.445C>T (p.R149W). Prokaryotic expression of mutations c.842_844delGAG and c.445C>T resulted in mutant enzymes with <1% wild-type activity, whereas c.250G>A expressed a stable enzyme with approximately 35% of wild-type activity. The discolored teeth from the affected cats contained markedly elevated URO I and III, accounting for the CEP-like phenocopy. In three lines, the phenotype was an autosomal dominant trait, while affected cats with the c.250G>A (p.A84T) mutation were homozygous, a unique recessive form of AIP. These animal models may permit further investigation of the pathogenesis of the acute, life-threatening neurological attacks in human AIP and the evaluation of therapeutic strategies. GenBank Accession Numbers: GQ850461-GQ850464.


Assuntos
Doenças do Gato/enzimologia , Gatos/genética , Modelos Animais de Doenças , Hidroximetilbilano Sintase/genética , Mutação , Porfiria Aguda Intermitente/enzimologia , Porfiria Eritropoética/enzimologia , Animais , Osso e Ossos/metabolismo , Doenças do Gato/genética , Doenças do Gato/metabolismo , Gatos/metabolismo , Coproporfirinas/urina , Feminino , Humanos , Hidroximetilbilano Sintase/química , Hidroximetilbilano Sintase/metabolismo , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/metabolismo , Porfiria Eritropoética/genética , Porfiria Eritropoética/metabolismo , Porfirinas/metabolismo , Dente/metabolismo , Uroporfirinas/urina
16.
Mol Genet Metab ; 107(1-2): 145-52, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22510705

RESUMO

Mucopolysaccharidosis VII (MPS VII) is due to deficient ß-glucuronidase (GUSB) activity, which leads to accumulation of chondroitin, heparan, and dermatan sulfate glycosaminoglycans in various tissues including those of the spine. Associated spine disease can be due to abnormalities in the vertebrae, the intervertebral disks, or other spine tissues. The goal of this study was to determine if neonatal gene therapy could prevent lumbar spine disease in MPS VII dogs. MPS VII dogs were injected intravenously with a retroviral vector (RV) expressing canine GUSB at 2 to 3 days after birth, which resulted in transduction of hepatocytes that secreted GUSB into blood. Expression was stable for up to 11 years, and mean survival was increased from 0.4 years in untreated dogs to 6.1 years in treated dogs. Despite a profound positive clinical effect, 6-month-old RV-treated MPS VII dogs still had hypoplastic ventral epiphyses with reduced calcification in the lumbar spine, which resulted in a reduced stiffness and increased range of motion that were not improved relative to untreated MPS VII dogs. At six to 11 years of age, ventral vertebrae remained hypoplastic in RV-treated MPS VII dogs, and there was desiccation of the nucleus pulposus in some disks. Histochemical staining demonstrated that disks did not have detectable GUSB activity despite high serum GUSB activity, which is likely due to poor diffusion into this relatively avascular structure. Thus, neonatal gene therapy cannot prevent lumbar spine disease in MPS VII dogs, which predicts that enzyme replacement therapy (ERT) will similarly be relatively ineffective even if started at birth.


Assuntos
Terapia Genética , Vértebras Lombares , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/terapia , Doenças da Coluna Vertebral/etiologia , Doenças da Coluna Vertebral/terapia , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Fenômenos Biomecânicos , Cálcio/metabolismo , Cães , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glucuronidase/sangue , Glicosaminoglicanos/urina , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/metabolismo , Vértebras Lombares/patologia , Masculino , Mucopolissacaridose VII/mortalidade , Radiografia , Retroviridae/genética , Doenças da Coluna Vertebral/diagnóstico
17.
Mol Genet Metab ; 107(1-2): 153-60, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22513347

RESUMO

Mucopolysaccharidosis type VII (MPS VII) is characterized by deficient ß-glucuronidase (GUSB) activity, which leads to accumulation of chondroitin, heparan and dermatan sulfate glycosaminoglycans (GAGs), and multisystemic disease. MPS VII patients can develop kypho-scoliotic deformity and spinal cord compression due to disease of intervertebral disks, vertebral bodies, and associated tissues. We have previously demonstrated in MPS VII dogs that intervertebral disks degenerate, vertebral bodies have irregular surfaces, and vertebral body epiphyses have reduced calcification, but the pathophysiological mechanisms underlying these changes are unclear. We hypothesized that some of these manifestations could be due to upregulation of destructive proteases, possibly via the binding of GAGs to Toll-like receptor 4 (TLR4), as has been proposed for other tissues in MPS models. In this study, the annulus fibrosus of the intervertebral disk of 6-month-old MPS VII dogs had cathepsin B and K activities that were 117- and 2-fold normal, respectively, which were associated with elevations in mRNA levels for these cathepsins as well as TLR4. The epiphyses of MPS VII dogs had a marked elevation in mRNA for the cartilage-associated gene collagen II, consistent with a developmental delay in the conversion of the cartilage to bone in this region. The spine obtained at autopsy from a young man with MPS VII exhibited similar increased cartilage in the vertebral bodies adjacent to the end plates, disorganization of the intervertebral disks, and irregular vertebral end plate morphology. These data suggest that the pathogenesis of destructive changes in the spine in MPS VII may involve upregulation of cathepsins. Inhibition of destructive proteases, such as cathepsins, might reduce spine disease in patients with MPS VII or related disorders.


Assuntos
Vértebras Lombares , Mucopolissacaridose VII/complicações , Doenças da Coluna Vertebral/etiologia , Animais , Animais Geneticamente Modificados , Catepsinas/genética , Catepsinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Cães , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Humanos , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/patologia , Masculino , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Radiografia , Doenças da Coluna Vertebral/diagnóstico por imagem , Doenças da Coluna Vertebral/patologia
18.
Mol Genet Metab ; 104(3): 395-403, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21778099

RESUMO

Gene transfer vectors based on adeno-associated virus 8 (AAV8) are highly efficient in liver transduction and can be easily administered by intravenous injection. In mice, AAV8 transduces predominantly hepatocytes near central veins and yields lower transduction levels in hepatocytes in periportal regions. This transduction bias has important implications for gene therapy that aims to correct metabolic liver enzymes because metabolic zonation along the porto-central axis requires the expression of therapeutic proteins within the zone where they are normally localized. In the present study we compared the expression pattern of AAV8 expressing green fluorescent protein (GFP) in liver between mice, dogs, and non-human primates. We confirmed the pericentral dominance in transgene expression in mice with AAV8 when the liver-specific thyroid hormone-binding globulin (TBG) promoter was used but also observed the same expression pattern with the ubiquitous chicken ß-actin (CB) and cytomegalovirus (CMV) promoters, suggesting that transduction zonation is not caused by promoter specificity. Predominantly pericentral expression was also found in dogs injected with AAV8. In contrast, in cynomolgus and rhesus macaques the expression pattern from AAV vectors was reversed, i.e. transgene expression was most intense around portal areas and less intense or absent around central veins. Infant rhesus macaques as well as newborn mice injected with AAV8 however showed a random distribution of transgene expression with neither portal nor central transduction bias. Based on the data in monkeys, adult humans treated with AAV vectors are predicted to also express transgenes predominantly in periportal regions whereas infants are likely to show a uniform transduction pattern in liver.


Assuntos
Dependovirus , Terapia Genética/métodos , Vetores Genéticos , Hepatócitos/citologia , Fígado/metabolismo , Transdução Genética/métodos , Animais , Cães , Proteínas de Fluorescência Verde/metabolismo , Processamento de Imagem Assistida por Computador , Modelos Lineares , Macaca , Camundongos , Microscopia de Fluorescência , Transgenes/genética
19.
Neurochem Res ; 36(9): 1612-22, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21136152

RESUMO

To facilitate the study of the chemical pathology of galactosylsphingosine (psychosine, GalSph) in Krabbe disease and glucosylsphingosine (GlcSph) in Gaucher disease, we have devised a facile method for the effective separation of these two glycosylsphingosines from other glycosphingolipids (GSLs) in Krabbe brain and Gaucher spleen samples. The procedure involves the use of acetone to selectively extract GalSph and GlcSph, respectively, from Krabbe brain and Gaucher spleen samples. Since acetone does not extract other GSLs except modest amounts of galactosylceramide, sulfatide, and glucosylceramide, the positively charged GalSph or GlcSph in the acetone extract can be readily separated from other GSLs by batchwise cation-exchange chromatography using a Waters Accell Plus CM Cartridge. GalSph or GlcSph enriched by this simple procedure can be readily analyzed by thin-layer chromatography or high-performance liquid chromatography.


Assuntos
Doença de Gaucher/patologia , Glicoesfingolipídeos/química , Leucodistrofia de Células Globoides/patologia , Psicosina/análogos & derivados , Psicosina/isolamento & purificação , Adolescente , Adulto , Animais , Química Encefálica , Criança , Pré-Escolar , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia em Camada Fina/métodos , Cães , Humanos , Lactente , Macaca mulatta , Camundongos , Camundongos Mutantes , Psicosina/análise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Baço/química
20.
Mol Ther Methods Clin Dev ; 20: 247-257, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33473358

RESUMO

Adeno-associated viral (AAV) vectors have emerged as the preferred platform for in vivo gene transfer because of their combined efficacy and safety. However, insertional mutagenesis with the subsequent development of hepatocellular carcinomas (HCCs) has been recurrently noted in newborn mice treated with high doses of AAV, and more recently, the association of wild-type AAV integrations in a subset of human HCCs has been documented. Here, we address, in a comprehensive, prospective study, the long-term risk of tumorigenicity in young adult mice following delivery of single-stranded AAVs targeting liver. HCC incidence in mice treated with therapeutic and reporter AAVs was low, in contrast to what has been previously documented in mice treated as newborns with higher doses of AAV. Specifically, HCCs developed in 6 out 76 of AAV-treated mice, and a pathogenic integration of AAV was found in only one tumor. Also, no evidence of liver tumorigenesis was found in juvenile AAV-treated mucopolysaccharidosis type VI (MPS VI) cats followed as long as 8 years after vector administration. Together, our results support the low risk of tumorigenesis associated with AAV-mediated gene transfer targeting juvenile/young adult livers, although constant monitoring of subjects enrolled in AAV clinical trial is advisable.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA