Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
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.
Brain ; 143(7): 2058-2072, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32671406

RESUMO

Intravascular injection of certain adeno-associated virus vector serotypes can cross the blood-brain barrier to deliver a gene into the CNS. However, gene distribution has been much more limited within the brains of large animals compared to rodents, rendering this approach suboptimal for treatment of the global brain lesions present in most human neurogenetic diseases. The most commonly used serotype in animal and human studies is 9, which also has the property of being transported via axonal pathways to distal neurons. A small number of other serotypes share this property, three of which were tested intravenously in mice compared to 9. Serotype hu.11 transduced fewer cells in the brain than 9, rh8 was similar to 9, but hu.32 mediated substantially greater transduction than the others throughout the mouse brain. To evaluate the potential for therapeutic application of the hu.32 serotype in a gyrencephalic brain of larger mammals, a hu.32 vector expressing the green fluorescent protein reporter gene was evaluated in the cat. Transduction was widely distributed in the cat brain, including in the cerebral cortex, an important target since mental retardation is an important component of many of the human neurogenetic diseases. The therapeutic potential of a hu.32 serotype vector was evaluated in the cat homologue of the human lysosomal storage disease alpha-mannosidosis, which has globally distributed lysosomal storage lesions in the brain. Treated alpha-mannosidosis cats had reduced severity of neurological signs and extended life spans compared to untreated cats. The extent of therapy was dose dependent and intra-arterial injection was more effective than intravenous delivery. Pre-mortem, non-invasive magnetic resonance spectroscopy and diffusion tensor imaging detected differences between the low and high doses, and showed normalization of grey and white matter imaging parameters at the higher dose. The imaging analysis was corroborated by post-mortem histological analysis, which showed reversal of histopathology throughout the brain with the high dose, intra-arterial treatment. The hu.32 serotype would appear to provide a significant advantage for effective treatment of the gyrencephalic brain by systemic adeno-associated virus delivery in human neurological diseases with widespread brain lesions.


Assuntos
Encéfalo/virologia , Dependovirus , Modelos Animais de Doenças , Terapia Genética/métodos , Vetores Genéticos , alfa-Manosidose/genética , Animais , Encéfalo/patologia , Gatos , Técnicas de Transferência de Genes , Transdução Genética
3.
Mol Ther ; 28(6): 1455-1463, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32330426

RESUMO

Mucopolysaccharidosis type I (MPS I) is an autosomal recessive lysosomal storage disease characterized by severe phenotypes, including corneal clouding. MPS I is caused by mutations in alpha-l-iduronidase (IDUA), a ubiquitous enzyme that catalyzes the hydrolysis of glycosaminoglycans. Currently, no treatment exists to address MPS I corneal clouding other than corneal transplantation, which is complicated by a high risk for rejection. Investigation of an adeno-associated virus (AAV) IDUA gene addition strategy targeting the corneal stroma addresses this deficiency. In MPS I canines with early or advanced corneal disease, a single intrastromal AAV8G9-IDUA injection was well tolerated at all administered doses. The eyes with advanced disease demonstrated resolution of corneal clouding as early as 1 week post-injection, followed by sustained corneal transparency until the experimental endpoint of 25 weeks. AAV8G9-IDUA injection in the MPS I canine eye with early corneal disease prevented the development of advanced corneal changes while restoring clarity. Biodistribution studies demonstrated vector genomes in ocular compartments other than the cornea and in some systemic organs; however, a capsid antibody response was detected in only the highest dosed subject. Collectively, the results suggest that intrastromal AAV8G9-IDUA therapy prevents and reverses visual impairment associated with MPS I corneal clouding.


Assuntos
Doenças da Córnea/etiologia , Doenças da Córnea/terapia , Técnicas de Transferência de Genes , Terapia Genética , Mucopolissacaridose I/complicações , Mucopolissacaridose I/genética , Animais , Animais Geneticamente Modificados , Doenças da Córnea/diagnóstico , Dependovirus/genética , Modelos Animais de Doenças , Cães , Feminino , Imunofluorescência , Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Reporter , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Iduronidase/genética , Masculino , Transgenes , Resultado do Tratamento
4.
Mol Ther ; 24(1): 26-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26354342

RESUMO

Lysosomal storage diseases (LSDs) are debilitating neurometabolic disorders for most of which long-term effective therapies have not been developed. Gene therapy is a potential treatment but a critical barrier to treating the brain is the need for global correction. We tested the efficacy of cisterna magna infusion of adeno-associated virus type 1 (AAV1) expressing feline alpha-mannosidase gene in the postsymptomatic alpha-mannosidosis (AMD) cat, a homologue of the human disease. Lysosomal alpha-mannosidase (MANB) activity in the cerebrospinal fluid (CSF) and serum were increased above the control values in untreated AMD cats. Clinical neurological signs were delayed in onset and reduced in severity. The lifespan of the treated cats was significantly extended. Postmortem histopathology showed resolution of lysosomal storage lesions throughout the brain. MANB activity in brain tissue was significantly above the levels of untreated tissues. The results demonstrate that a single cisterna magna injection of AAV1 into the CSF can mediate widespread neuronal transduction of the brain and meaningful clinical improvement. Thus, cisterna magna gene delivery by AAV1 appears to be a viable strategy for treatment of the whole brain in AMD and should be applicable to many of the neurotropic LSDs as well as other neurogenetic disorders.


Assuntos
Doenças do Gato/terapia , Cisterna Magna/metabolismo , Dependovirus/genética , alfa-Manosidase/genética , alfa-Manosidose/veterinária , Idade de Início , Animais , Encéfalo/enzimologia , Doenças do Gato/patologia , Gatos , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Injeções , Lisossomos/metabolismo , alfa-Manosidase/sangue , alfa-Manosidase/líquido cefalorraquidiano , alfa-Manosidase/metabolismo , alfa-Manosidose/patologia , alfa-Manosidose/terapia
5.
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
6.
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
7.
J Neurosci Res ; 94(11): 1007-17, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27638585

RESUMO

Globoid cell leukodystrophy (GLD), or Krabbe's disease, is a debilitating and always fatal pediatric neurodegenerative disease caused by a mutation in the gene encoding the hydrolytic enzyme galactosylceramidase (GALC). In the absence of GALC, progressive loss of myelin and accumulation of a neurotoxic substrate lead to incapacitating loss of motor and cognitive function and death, typically by 2 years of age. Currently, there is no cure. Recent convincing evidence of the therapeutic potential of combining gene and cell therapies in the murine model of GLD has accelerated the requirement for validated markers of disease to evaluate therapeutic efficacy. Here we demonstrate clinically relevant and quantifiable measures of central (CNS) and peripheral (PNS) nervous system disease progression in the naturally occurring canine model of GLD. As measured by brainstem auditory-evoked response testing, GLD dogs demonstrated a significant increase in I-V interpeak latency and hearing threshold at all time points. Motor nerve conduction velocities (NCVs) in GLD dogs were significantly lower than normal by 12-16 weeks of age, and sensory NCV was significantly lower than normal by 8-12 weeks of age, serving as a sensitive indicator of peripheral nerve dysfunction. Post-mortem histological evaluations confirmed neuroimaging and electrodiagnostic assessments and detailed loss of myelin and accumulation of storage product in the CNS and the PNS. Additionally, cerebrospinal fluid psychosine concentrations were significantly elevated in GLD dogs, demonstrating potential as a biochemical marker of disease. These data demonstrate that CNS and PNS disease progression can be quantified over time in the canine model of GLD with tools identical to those used to assess human patients. © 2016 Wiley Periodicals, Inc.


Assuntos
Potenciais Evocados Auditivos do Tronco Encefálico/genética , Leucodistrofia de Células Globoides/complicações , Leucodistrofia de Células Globoides/genética , Doenças do Sistema Nervoso , Animais , Modelos Animais de Doenças , Cães , Estimulação Elétrica , Feminino , Galactosilceramidase/genética , Humanos , Leucodistrofia de Células Globoides/diagnóstico por imagem , Leucodistrofia de Células Globoides/veterinária , Imageamento por Ressonância Magnética , Masculino , Mutação de Sentido Incorreto/genética , Sistema Nervoso/diagnóstico por imagem , Sistema Nervoso/patologia , Sistema Nervoso/fisiopatologia , Doenças do Sistema Nervoso/diagnóstico por imagem , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/terapia , Condução Nervosa/genética , Psicosina/líquido cefalorraquidiano
8.
J Pharmacol Exp Ther ; 358(2): 254-61, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27307499

RESUMO

Niemann-Pick type C (NPC) 1 disease is a rare, inherited, neurodegenerative disease. Clear evidence of the therapeutic efficacy of 2-hydroxypropyl-ß-cyclodextrin (HPßCD) in animal models resulted in the initiation of a phase I/IIa clinical trial in 2013 and a phase IIb/III trial in 2015. With clinical trials ongoing, validation of a biomarker to track disease progression and serve as a supporting outcome measure of therapeutic efficacy has become compulsory. In this study, we evaluated calcium-binding protein calbindin D-28K (calbindin) concentrations in the cerebrospinal fluid (CSF) as a biomarker of NPC1 disease. In the naturally occurring feline model, CSF calbindin was significantly elevated at 3 weeks of age, prior to the onset of cerebellar dysfunction, and steadily increased to >10-fold over normal at end-stage disease. Biweekly intrathecal administration of HPßCD initiated prior to the onset of neurologic dysfunction completely normalized CSF calbindin in NPC1 cats at all time points analyzed when followed up to 78 weeks of age. Initiation of HPßCD after the onset of clinical signs (16 weeks of age) resulted in a delayed reduction of calbindin levels in the CSF. Evaluation of CSF from patients with NPC1 revealed that calbindin concentrations were significantly elevated compared with CSF samples collected from unaffected patients. Off-label treatment of patients with NPC1 with miglustat, an inhibitor of glycosphingolipid biosynthesis, significantly decreased CSF calbindin compared with pretreatment concentrations. These data suggest that the CSF calbindin concentration is a sensitive biomarker of NPC1 disease that could be instrumental as an outcome measure of therapeutic efficacy in ongoing clinical trials.


Assuntos
Biomarcadores/líquido cefalorraquidiano , Calbindina 1/líquido cefalorraquidiano , Progressão da Doença , Doença de Niemann-Pick Tipo C/líquido cefalorraquidiano , 2-Hidroxipropil-beta-Ciclodextrina , Adolescente , Adulto , Animais , Gatos , Criança , Pré-Escolar , Feminino , Glicoesfingolipídeos/biossíntese , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Doença de Niemann-Pick Tipo C/metabolismo , Fatores de Tempo , Adulto Jovem , beta-Ciclodextrinas/farmacologia
9.
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
10.
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
11.
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
12.
Mol Ther Methods Clin Dev ; 32(2): 101272, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38946937

RESUMO

Alpha-mannosidosis is caused by a genetic deficiency of lysosomal alpha-mannosidase, leading to the widespread presence of storage lesions in the brain and other tissues. Enzyme replacement therapy is available but is not approved for treating the CNS, since the enzyme does not penetrate the blood-brain barrier. However, intellectual disability is a major manifestation of the disease; thus, a complimentary treatment is needed. While enzyme replacement therapy into the brain is technically feasible, it requires ports and frequent administration over time that are difficult to manage medically. Infusion of adeno-associated viral vectors into the cerebrospinal fluid is an attractive route for broadly targeting brain cells. We demonstrate here the widespread post-symptomatic correction of the globally distributed storage lesions by infusion of a high dose of AAV1-feline alpha-mannosidase (fMANB) into the CSF via the cisterna magna in the gyrencephalic alpha-mannosidosis cat brain. Significant improvements in clinical parameters occurred, and widespread global correction was documented pre-mortem by non-invasive magnetic resonance imaging. Postmortem analysis demonstrated high levels of MANB activity and reversal of lysosomal storage lesions throughout the brain. Thus, CSF treatment by adeno-associated viral vector gene therapy appears to be a suitable complement to systemic enzyme replacement therapy to potentially treat the whole patient.

13.
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
14.
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
15.
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
16.
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
17.
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
18.
Mol Ther ; 19(3): 461-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21119624

RESUMO

Mucopolysaccharidosis VI (MPS VI) is caused by deficient arylsulfatase B (ARSB) activity resulting in lysosomal storage of glycosaminoglycans (GAGs). MPS VI is characterized by dysostosis multiplex, organomegaly, corneal clouding, and heart valve thickening. Gene transfer to a factory organ like liver may provide a lifetime source of secreted ARSB. We show that intravascular administration of adeno-associated viral vectors (AAV) 2/8-TBG-felineARSB in MPS VI cats resulted in ARSB expression up to 1 year, the last time point of the study. In newborn cats, normal circulating ARSB activity was achieved following delivery of high vector doses (6 × 10(13) genome copies (gc)/kg) whereas delivery of AAV2/8 vector doses as low as 2 × 10(12) gc/kg resulted in higher than normal serum ARSB levels in juvenile MPS VI cats. In MPS VI cats showing high serum ARSB levels, independent of the age at treatment, we observed: (i) clearance of GAG storage, (ii) improvement of long bone length, (iii) reduction of heart valve thickness, and (iv) improvement in spontaneous mobility. Thus, AAV2/ 8-mediated liver gene transfer represents a promising therapeutic strategy for MPS VI patients.


Assuntos
Dependovirus , Técnicas de Transferência de Genes , Fígado , Mucopolissacaridose VI/terapia , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Gatos , Dependovirus/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glicosaminoglicanos/metabolismo , Células HEK293 , Humanos , Fígado/metabolismo , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Mucopolissacaridose VI/enzimologia , Mucopolissacaridose VI/patologia , N-Acetilgalactosamina-4-Sulfatase/genética , N-Acetilgalactosamina-4-Sulfatase/metabolismo , Fenótipo , Resultado do Tratamento
19.
Mol Ther Methods Clin Dev ; 26: 384-393, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36034772

RESUMO

Multiple studies have examined the transduction characteristics of different AAV serotypes in the mouse brain, where they can exhibit significantly different patterns of transduction. The pattern of transduction also varies with the route of administration. Much less information exists for the transduction characteristics in large-brained animals. Large animal models have brains that are closer in size and organization to the human brain, such as being gyrencephalic compared to the lissencephalic rodent brains, pathway organization, and certain electrophysiologic properties. Large animal models are used as translational intermediates to develop gene therapies to treat human diseases. Various AAV serotypes and routes of delivery have been used to study the correction of pathology in the brain in lysosomal storage diseases. In this study, we evaluated the ability of selected AAV serotypes to transduce cells in the cat brain when delivered into the cerebrospinal fluid via the cisterna magna. We previously showed that AAV1 transduced significantly greater numbers of cells than AAV9 in the cat brain by this route. In the present study, we evaluated serotypes closely related to AAVs 1 and 9 (AAVs 6, AS, hu32) that may mediate more extensive transduction, as well as AAVs 4 and 5, which primarily transduce choroid plexus epithelial (CPE) and ependymal lining cells in the rodent brain. The related serotypes tended to have similar patterns of transduction but were divergent in some specific brain structures.

20.
PLoS One ; 16(12): e0260805, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34855883

RESUMO

BACKGROUND: Hereditary angioedema (HAE) is a rare disease that manifests as recurrent and debilitating angioedema attacks, significantly impacting patients' quality of life. OBJECTIVE: To assess communication dynamics between patients with HAE and treating physicians and the impact this has on the treatment of HAE in the United States. METHODS: This observational study used an institutional review board-approved protocol to collect four sources of patient-physician communication data from the period between January 2015 and May 2017: in-office conversations between patients aged ≥18 years with HAE and physicians, follow-up dictations with physicians, telephone interviews with patients and physicians, and publicly available social media posts from patients. Participant language was qualitatively assessed and key communication elements and communication gaps identified. RESULTS: Twenty-five in-office conversations, 14 follow-up physician dictations, and 17 telephone interviews were conducted with a total of 29 unique patients, 4 caregivers, and 14 physicians. In-office conversations were generally physician-driven and focused primarily on symptom frequency, location, and severity; lexicon from both parties centered on "episodes" and "swelling." During visits, impact on quality of life was not routinely assessed by physicians nor discussed proactively by patients; however, during telephone interviews and online, patients frequently described the multifaceted burden of HAE. Patients highlighted the difficulties they experience by using repetition, emphasis, and metaphors; they also varied the descriptors used for attacks depending on the communication goal. Physicians used intensifiers to emphasize the necessity of rescue medication access, whereas prophylactic treatments were positioned as an option for frequent or laryngeal attacks. CONCLUSION: Vocabulary differences suggest that the full impact of HAE is not consistently communicated by patients to physicians during clinical visits, indicating the potential for misaligned understanding of disease burden. A patient-driven, rather than physician-driven approach to the discussions may elicit valuable information that could help to optimize treatment approaches.


Assuntos
Angioedemas Hereditários/epidemiologia , Comunicação , Medidas de Resultados Relatados pelo Paciente , Médicos/psicologia , Qualidade de Vida , Índice de Gravidade de Doença , Adolescente , Adulto , Idoso , Angioedemas Hereditários/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inquéritos e Questionários , Estados Unidos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA