Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gene Ther ; 21(12): 1029-40, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25231174

RESUMO

Astrocytes are an attractive cell target for gene therapy, but the validation of new therapeutic candidates is needed. We determined whether adeno-associated viral (AAV) vector-mediated overexpression of glutamine synthetase (GS) or excitatory amino-acid transporter 2 (EAAT2), or expression of microRNA targeting adenosine kinase (miR-ADK) in hippocampal astrocytes in the rat brain could modulate susceptibility to kainate-induced seizures and neuronal cell loss. Transgene expression was found predominantly in astrocytes following direct injection of glial-targeting AAV9 vectors by 3 weeks postinjection. ADK expression in miR-ADK vector-injected rats was reduced by 94-96% and was associated with an ~50% reduction in the duration of kainate-induced seizures and greater protection of dentate hilar neurons but not CA3 neurons compared with miR-control vector-injected rats. In contrast, infusion of AAV-GS and EAAT2 vectors did not afford any protection against seizures or neuronal damage as the level of transcriptional activity of the glial fibrillary acidic promoter was too low to drive any significant increase in transgenic GS or EAAT2 relative to the high endogenous levels of these proteins. Our findings support ADK as a prime therapeutic target for gene therapy of temporal lobe epilepsy and suggest that alternative approaches including the use of stronger glial promoters are needed to increase transgenic GS and EAAT2 expression to levels that may be required to affect seizure induction and propagation.


Assuntos
Adenosina Quinase/genética , Epilepsia do Lobo Temporal/terapia , Transportador 2 de Aminoácido Excitatório/genética , Marcação de Genes , Terapia Genética/métodos , Glutamato-Amônia Ligase/genética , Adenosina Quinase/metabolismo , Animais , Astrócitos/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos , Glutamato-Amônia Ligase/metabolismo , Hipocampo/metabolismo , Ácido Caínico/efeitos adversos , Masculino , Neuroglia/metabolismo , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Transgenes/genética
2.
Nat Genet ; 8(2): 148-54, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7842013

RESUMO

Adeno-associated viral (AAV) vectors are non-pathogenic, integrating DNA vectors in which all viral genes are removed and helper virus is completely eliminated. To evaluate this system in the post-mitotic cells of the brain, we found that an AAV vector containing the lacZ gene (AAVlac) resulted in expression of beta-galactosidase up to three months post-injection in vivo. A second vector expressing human tyrosine hydroxylase (AAVth) was injected into the denervated striatum of unilateral 6-hydroxydopamine-lesioned rats. Tyrosine hydroxylase (TH) immunoreactivity was detectable in striatal neurons and glia for up to four months and we also found significant behavioural recovery in lesioned rats treated with AAVth versus AAVlac controls. Safe and stable TH gene transfer into the denervated striatum may have potential for the genetic therapy of Parkinson's disease.


Assuntos
Encéfalo/metabolismo , Dependovirus/genética , Regulação Viral da Expressão Gênica , Terapia Genética , Vetores Genéticos , Doença de Parkinson Secundária/terapia , Proteínas Recombinantes de Fusão/biossíntese , Tirosina 3-Mono-Oxigenase/genética , Adenovírus Humanos/fisiologia , Animais , Apomorfina/toxicidade , Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Corpo Estriado , Citomegalovirus/genética , Genes Reporter , Genes Sintéticos , Vírus Auxiliares/fisiologia , Humanos , Masculino , Microinjeções , Oxidopamina/toxicidade , Doença de Parkinson Secundária/induzido quimicamente , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Vírus 40 dos Símios/genética , Substância Negra/metabolismo , Substância Negra/patologia , Transfecção , Tirosina 3-Mono-Oxigenase/biossíntese , beta-Galactosidase/genética
3.
Nat Med ; 4(10): 1131-5, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9771745

RESUMO

Gene therapy is usually reserved for severe and medically refractory disorders because of the toxicity, potential long-term risks and invasiveness of most gene transfer protocols. Here we show that an orally administered adeno-associated viral vector leads to persistent expression of a beta-galactosidase transgene in both gut epithelial and lamina propria cells, and that this approach results in long-term phenotypic recovery in an animal model of lactose intolerance. A gene 'pill' associated with highly efficient and stable gene expression might be a practical and cost-effective strategy for even relatively mild disorders, such as lactase deficiency.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Intolerância à Lactose/terapia , beta-Galactosidase/genética , Administração Oral , Animais , Glicemia/análise , Peso Corporal , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virologia , Lactase , Lactose/metabolismo , Ratos , Transformação Genética , Transgenes , beta-Galactosidase/deficiência
4.
Nat Med ; 5(4): 448-53, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10202938

RESUMO

The mammalian brain has a high degree of plasticity, with dentate granule cell neurogenesis and glial proliferation stimulated by an enriched environment combining both complex inanimate and social stimulation. Moreover, rodents exposed to an enriched environment both before and after a cerebral insult show improved cognitive performance. One of the most robust associations of environmental enrichment is improved learning and memory in the Morris water maze, a spatial task that mainly involves the hippocampus. Furthermore, clinical evidence showing an association between higher educational attainment and reduced risk of Alzheimer and Parkinson-related dementia indicates that a stimulating environment has positive effects on cerebral health that may provide some resilience to cerebral insults. Here we show that in addition to its effects on neurogenesis, an enriched environment reduces spontaneous apoptotic cell death in the rat hippocampus by 45%. Moreover, these environmental conditions protect against kainate-induced seizures and excitotoxic injury. The enriched environment induces expression of glial-derived neurotrophic factor and brain-derived neurotrophic factor and increases phosphorylation of the transcription factor cyclic-AMP response element binding protein, indicating that the influence of the environment on spontaneous apoptosis and cerebral resistance to insults may be mediated through transcription factor activation and induction of growth factor expression.


Assuntos
Apoptose , Meio Ambiente , Hipocampo/patologia , Ácido Caínico/efeitos adversos , Fatores de Crescimento Neural , Convulsões/prevenção & controle , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hipocampo/crescimento & desenvolvimento , Hibridização In Situ , Masculino , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fosforilação , RNA Mensageiro/isolamento & purificação , Ratos , Ratos Wistar , Convulsões/induzido quimicamente
5.
Gene Ther ; 17(5): 643-52, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20220782

RESUMO

Neuropeptide Y (NPY) is an endogenous peptide with powerful anticonvulsant properties. Its overexpression in the rat hippocampus, mediated by the local application of recombinant adeno-associated viral (rAAV) vectors carrying the human NPY gene, results in significant reduction of seizures in acute and chronic seizure models. In this study, we characterized a more efficient rAAV-NPY vector to improve cell transfection in the injected area. The changes included pseudotyping with the AAV vector serotype 1 (rAAV1), and using the strong constitutive hybrid CBA promoter, which contains a cytomegalovirus enhancer and chicken beta-actin promoter sequences. We compared NPY expression and the associated anticonvulsant effects of this new vector, with those mediated by the former rAAV vector with chimeric serotype 1/2 (rAAV1/2). In addition, we investigated whether rAAV serotype 1 vector-mediated chronic NPY overexpression causes behavioural deficits that may detract from the clinical utility of this therapeutic approach. We report that rAAV-NPY serotype 1 vector has significantly improved anticonvulsant activity when compared with serotype 1/2 vector, as assessed by measuring EEG seizure activity in kainic acid treated rats. rAAV1-mediated NPY overexpression in naive rats did not result in alterations of physiological functions such as learning and memory, anxiety and locomotor activity. In addition, we did not observe glia activation, or humoral immune responses against serotype 1 vector, which could inactivate gene expression. Our findings show that rAAV1-NPY vector with the CBA promoter mediates powerful anticonvulsant effects and seems to be safe in rodents, thus it may be considered a vector of choice for possible clinical applications.


Assuntos
Epilepsia do Lobo Temporal/terapia , Terapia Genética/métodos , Hipocampo/metabolismo , Neuropeptídeo Y/genética , Convulsões/terapia , Transdução Genética/métodos , Actinas/genética , Animais , Dependovirus , Epilepsia do Lobo Temporal/fisiopatologia , Vetores Genéticos , Imunidade Humoral , Ácido Caínico/efeitos adversos , Aprendizagem , Masculino , Memória , Atividade Motora , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Convulsões/fisiopatologia
6.
Science ; 266(5189): 1399-403, 1994 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-7669103

RESUMO

One therapeutic approach to treating Parkinson's disease is to convert endogenous striatal cells into levo-3,4-dihydroxyphenylalanine (L-dopa)-producing cells. A defective herpes simplex virus type 1 vector expressing human tyrosine hydroxylase was delivered into the partially denervated striatum of 6-hydroxydopamine-lesioned rats, used as a model of Parkinson's disease. Efficient behavioral and biochemical recovery was maintained for 1 year after gene transfer. Biochemical recovery included increases in both striatal tyrosine hydroxylase enzyme activity and in extracellular dopamine concentrations. Persistence of human tyrosine hydroxylase was revealed by expression of RNA and immunoreactivity.


Assuntos
Corpo Estriado/enzimologia , Terapia Genética , Atividade Motora , Doença de Parkinson/terapia , Simplexvirus/genética , Tirosina 3-Mono-Oxigenase/genética , Animais , Sequência de Bases , Corpo Estriado/metabolismo , Denervação , Modelos Animais de Doenças , Dopamina/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Levodopa/metabolismo , Masculino , Dados de Sequência Molecular , Neurônios/enzimologia , Doença de Parkinson/metabolismo , Ratos , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/metabolismo
7.
Science ; 287(5457): 1453-60, 2000 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-10688787

RESUMO

The brain is generally considered immunoprivileged, although increasing examples of immunological responses to brain antigens, neuronal expression of major histocompatibility class I genes, and neurological autoimmunity have been recognized. An adeno-associated virus (AAV) vaccine generated autoantibodies that targeted a specific brain protein, the NR1 subunit of the N-methyl-D-aspartate (NMDA) receptor. After peroral administration of the AAV vaccine, transgene expression persisted for at least 5 months and was associated with a robust humoral response in the absence of a significant cell-mediated response. This single-dose vaccine was associated with strong anti-epileptic and neuroprotective activity in rats for both a kainate-induced seizure model and also a middle cerebral artery occlusion stroke model at 1 to 5 months following vaccination. Thus, a vaccination strategy targeting brain proteins is feasible and may have therapeutic potential for neurological disorders.


Assuntos
Autoanticorpos/imunologia , Epilepsia do Lobo Temporal/terapia , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/imunologia , Acidente Vascular Cerebral/terapia , Vacinas de DNA/uso terapêutico , Administração Oral , Animais , Afinidade de Anticorpos , Autoanticorpos/análise , Autoanticorpos/sangue , Autoanticorpos/líquido cefalorraquidiano , Barreira Hematoencefálica , Dependovirus/genética , Epilepsia do Lobo Temporal/patologia , Mapeamento de Epitopos , Epitopos , Vetores Genéticos , Hipocampo/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Ativação Linfocitária , Atividade Motora , Ratos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/biossíntese , Estado Epiléptico/prevenção & controle , Transgenes , Vacinação
8.
Neuroscience ; 151(2): 352-60, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18082971

RESUMO

Neuronal activity regulated pentraxin (Narp) is a secreted, synaptic protein that has been implicated in modulating synaptic transmission. However, it is unclear how Narp secretion is regulated. Since we noted prominent Narp immunostaining in vasopressin neurons of the hypothalamus and in the posterior pituitary, we assessed whether it, like vasopressin, is released into the systemic circulation in an activity-dependent fashion. Consistent with this hypothesis, electron microscopic studies of the posterior pituitary demonstrated that Narp is located in secretory vesicles containing vasopressin. Using affinity chromatography, we detected Narp in plasma and found that these levels are markedly decreased by hypophysectomy. In addition, we confirmed that injection of a viral Narp construct into the hypothalamus restores plasma Narp levels in Narp knockout mice. In checking for activity-dependent secretion of Narp from the posterior pituitary, we found that several stimuli known to trigger vasopressin release, i.e. hypovolemia, dehydration and endotoxin, elevate plasma Narp levels. Taken together, these findings provide compelling evidence that Narp is secreted from vasopressin neurons in an activity-dependent fashion.


Assuntos
Proteína C-Reativa/metabolismo , Atividade Motora/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Vasopressinas/fisiologia , Adenoviridae/genética , Animais , Cromatografia de Afinidade , DNA Complementar/biossíntese , DNA Complementar/genética , Desidratação/fisiopatologia , Vetores Genéticos , Humanos , Hipovolemia/fisiopatologia , Imuno-Histoquímica , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Microscopia Imunoeletrônica , Proteínas do Tecido Nervoso/sangue , Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
9.
J Clin Invest ; 95(5): 2403-8, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7738204

RESUMO

Glucose modulates beta cell insulin secretion via effects on ATP-sensitive potassium (KATP) channels. To test the hypothesis that glucose exerts a similar effect on neuronal function, local glucose availability was varied in awake rats using microdialysis in the substantia nigra, the brain region with the highest density of KATP channels. 10 mM glucose perfusion increased GABA release by 111 +/- 42%, whereas the sulfonylurea, glipizide, increased GABA release by 84 +/- 20%. In contrast, perfusion of the KATP channel activator, lemakalim, or depletion of ATP by perfusion of 2-deoxyglucose with oligomycin inhibited GABA release by 44 +/- 8 and 45 +/- 11%, respectively. Moreover, the inhibition of GABA release by 2-deoxyglucose and oligomycin was blocked by glipizide. During systemic insulin-induced hypoglycemia (1.8 +/- 0.3 mM), nigral dialysate GABA concentrations decreased by 49 +/- 4% whereas levels of dopamine in striatal dialysates increased by 119 +/- 18%. We conclude that both local and systemic glucose availability influences nigral GABA release via an effect on KATP channels and that inhibition of GABA release may in part mediate the hyperexcitability associated with hypoglycemia. These data support the hypothesis that glucose acts as a signaling molecule, and not simply as an energy-yielding fuel, for neurons.


Assuntos
Desoxiglucose/farmacologia , Glucose/fisiologia , Neurônios/fisiologia , Canais de Potássio/fisiologia , Substância Negra/fisiologia , Ácido gama-Aminobutírico/metabolismo , Trifosfato de Adenosina/farmacologia , Análise de Variância , Animais , Glipizida/farmacologia , Hipoglicemia/fisiopatologia , Insulina/farmacologia , Masculino , Microdiálise/métodos , Neurônios/efeitos dos fármacos , Oligomicinas/farmacologia , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos
10.
J Clin Invest ; 93(4): 1677-82, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8163668

RESUMO

The central nervous system has been implicated in the activation of counterregulatory hormone release during hypoglycemia. However, the precise loci involved are not established. To determine the role of the ventromedial hypoglycemia, we performed hypoglycemic clamp studies in conscious Sprague-Dawley rats with bilateral VMH lesions produced by local ibotenic acid injection 2 wk earlier. Rats with lesions in the lateral hypothalamic area, frontal lobe, sham operated (stereotaxic needle placement into hypothalamus without injection), and naive animals served as control groups. The clamp study had two phases. For the first hour plasma glucose was fixed by a variable glucose infusion at euglycemia (approximately 5.9 mM). Thereafter, for an additional 90 min, glucose was either allowed to fall to (a) mild hypoglycemia (approximately 3.0 mM) or (b) more severe hypoglycemia (approximately 2.5 mM). Glucagon and catecholamine responses of lateral hypothalamic area-, frontal lobe-lesioned, sham operated, and naive animals were virtually identical at each hypoglycemic plateau. In contrast, glucagon, epinephrine, and norepinephrine responses in the VMH-lesioned rats were markedly inhibited; hormones were diminished by 50-60% during mild and by 75-80% during severe hypoglycemia as compared with the other groups. We conclude that the VMH plays a crucial role in triggering the release of glucagon and catecholamines during hypoglycemia.


Assuntos
Hormônios/sangue , Hipoglicemia/sangue , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Epinefrina/sangue , Glucagon/sangue , Insulina/sangue , Masculino , Norepinefrina/sangue , Ratos , Ratos Sprague-Dawley
11.
J Clin Invest ; 96(1): 370-7, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7615807

RESUMO

To determine the relationship between circulating metabolic fuels and their local concentrations in peripheral tissues we measured glycerol, glucose, and amino acids by microdialysis in muscle and adipose interstitium of 10 fasted, nonobese human subjects during (a) baseline, (b) euglycemic hyperinsulinemia (3 mU/kg per min for 3 h) and, (c) local norepinephrine reuptake blockade (NOR). At baseline, interstitial glycerol was strikingly higher (P < 0.0001) in muscle (3710 microM) and adipose tissue (2760 microM) compared with plasma (87 microM), whereas interstitial glucose (muscle 3.3, fat 3.6 mM) was lower (P < 0.01) than plasma levels (4.8 mM). Taurine, glutamine, and alanine levels were higher in muscle than in adipose or plasma (P < 0.05). Euglycemic hyperinsulinemia did not affect interstitial glucose, but induced a fall in plasma glycerol and amino acids paralleled by similar changes in the interstitium of both tissues. Local NOR provoked a fivefold increase in glycerol (P < 0.001) and twofold increase in norepinephrine (P < 0.01) in both muscle and adipose tissues. To conclude, interstitial substrate levels in human skeletal muscle and adipose tissue differ substantially from those in the circulation and this disparity is most pronounced for glycerol which is raised in muscle as well as adipose tissue. In muscle, insulin suppressed and NOR increased interstitial glycerol concentrations. Our data suggest unexpectedly high rates of intramuscular lipolysis in humans that may play an important role in fuel metabolism.


Assuntos
Tecido Adiposo/química , Aminoácidos/análise , Espaço Extracelular/química , Glucose/análise , Glicerol/análise , Lipólise , Músculo Esquelético/química , Adolescente , Adulto , Calibragem , Desipramina/farmacologia , Feminino , Humanos , Insulina/farmacologia , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/metabolismo
12.
Trends Neurosci ; 14(10): 428-32, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1722360

RESUMO

A genetic analysis of mammalian neuronal physiology might now be possible due to the development of defective herpes simplex virus vectors, which allow gene transfer directly into mature neurons, in culture or in the adult brain. Genetically altered proteins that play critical roles in neuronal physiology, including those responsible for the generation of action potentials, synthesis and release of neurotransmitters, and signal transduction enzymes, can now be stably expressed in neurons. The effect of such altered proteins on neuronal physiology can therefore be examined, using the tools of modern neuroscience. Genetic manipulation is biochemically specific and stable, and can be targeted both to a particular cell type and to a particular subregion of the cell to yield insights into the molecular basis for specific brain functions.


Assuntos
Neurônios/fisiologia , Simplexvirus/genética , Transfecção , Animais , Humanos , Biologia Molecular
13.
Trends Neurosci ; 24(12): 706-12, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11718875

RESUMO

A fundamental problem in neuroscience has been the creation of suitable in vivo model systems to study basic neurological phenomena and pathology of the central nervous system (CNS). Somatic cell genetic engineering with viral vectors provides a versatile tool to model normal brain physiology and a variety of neurological diseases.


Assuntos
Encéfalo/fisiologia , Técnicas de Transferência de Genes , Vírus/genética , Animais , Encefalopatias/fisiopatologia , Vetores Genéticos , Mamíferos
14.
Cancer Res ; 61(12): 4827-36, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11406559

RESUMO

The fragile histidine triad (FHIT) gene is a tumor suppressor gene that is altered by deletion in a large fraction of human tumors, including pancreatic cancer. To evaluate the potential of FHIT gene therapy, we developed recombinant adenoviral and adenoassociated viral (AAV) FHIT vectors and tested these vectors in vitro and in vivo for activity against human pancreatic cancer cells. Our data show that viral FHIT gene delivery results in apoptosis by activation of the caspase pathway. Furthermore, Fhit overexpression enhances the susceptibility of pancreatic cancer cells to exogenous inducers of apoptosis. In vivo results show that FHIT gene transfer delays tumor growth and prolongs survival in a murine model mimicking human disease.


Assuntos
Hidrolases Anidrido Ácido , Apoptose/genética , Proteínas de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas/genética , Adenoviridae/genética , Animais , Caspases/metabolismo , Ciclo Celular/fisiologia , Divisão Celular/genética , Fragmentação do DNA , Feminino , Técnicas de Transferência de Genes , Genes Supressores de Tumor , Terapia Genética , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Nus , Mitocôndrias/fisiologia , Neoplasias Pancreáticas/metabolismo , Biossíntese de Proteínas , Transdução de Sinais/fisiologia , Transdução Genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Diabetes ; 44(2): 180-4, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7859938

RESUMO

To test the hypothesis that nuclei of the ventromedial hypothalamus (VMH) play a key role in the detection of counterregulatory responses to hypoglycemia, we delivered the glucopenic agent 2-deoxyglucose via bilaterally placed microdialysis probes into the VMH of conscious, chronically catheterized rats. The goal was to produce cellular glucopenia localized to the VMH. The volume of brain tissue exposed to 2-deoxyglucose was determined by adding [3H]2-deoxyglucose to the dialysate; its distribution in cerebral tissue was almost exclusively limited to the VMH. Rats with microdialysis probes placed into the frontal lobes served as a control group. Local perfusion of 2-deoxyglucose (but not glucose) into the VMH caused a prompt twofold increase in plasma glucose in association with a striking elevation of plasma glucagon (3.5-fold), epinephrine (30-fold), and norepinephrine (3.5-fold). No effect was seen when 2-deoxyglucose was delivered into the frontal lobes. We conclude that glucopenia localized to the VMH triggers the release of counterregulatory hormones that defend against hypoglycemia. Thus, the neurons that sense glucopenia may be situated in the VMH.


Assuntos
Epinefrina/metabolismo , Glucagon/metabolismo , Hipoglicemia/fisiopatologia , Hipotálamo Médio/fisiopatologia , Insulina/metabolismo , Norepinefrina/metabolismo , Animais , Glicemia/metabolismo , Desoxiglucose/administração & dosagem , Desoxiglucose/metabolismo , Desoxiglucose/farmacologia , Homeostase , Hipotálamo Médio/efeitos dos fármacos , Secreção de Insulina , Masculino , Ratos , Ratos Sprague-Dawley
16.
Hum Gene Ther ; 12(12): 1589-91, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11529246

RESUMO

This gene transfer experiment is the first Parkinson's Disease (PD) protocol to be submitted to the Recombinant DNA Advisory Committee. The principal investigators have uniquely focused their careers on both pre-clinical work on gene transfer in the brain and clinical expertise in management and surgical treatment of patients with PD. They have extensively used rodent models of PD for proof-of-principle experiments on the utility of different vector systems. PD is an excellent target for gene therapy, because it is a complex acquired disease of unknown etiology (apart from some rare familial cases) yet it is characterized by a specific neuroanatomical pathology, the degeneration of dopamine neurons of the substantia nigra (SN) with loss of dopamine input to the striatum. This pathology results in focal changes in the function of several deep brain nuclei, which have been well-characterized in humans and animal models and which account for many of the motor symptoms of PD. Our original approaches, largely to validate in vivo gene transfer in the brain, were designed to facilitate dopamine transmission in the striatum using an AAV vector expressing dopamine-synthetic enzymes. Although these confirmed the safety and potential efficacy of AAV, complex patient responses to dopamine augmenting medication as well as poor results and complications of human transplant studies suggested that this would be a difficult and potentially dangerous clinical strategy using current approaches. Subsequently, we and others investigated the use of growth factors, including GDNF. These showed some encouraging effects on dopamine neuron survival and regeneration in both rodent and primate models; however, uncertain consequences of long-term growth factor expression and question regarding timing of therapy in the disease course must be resolved before any clinical study can be contemplated. We now propose to infuse into the subthalamic nucleus (STN) recombinant AAV vectors expressing the two isoforms of the enzyme glutamic acid decarboxylase (GAD-65 and GAD-67), which synthesizes the major inhibitory neurotransmitter in the brain, GABA. The STN is a very small nucleus (140 cubic mm or 0.02% of the total brain volume, consisting of approximately 300,000 neurons) which is disinhibited in PD, leading to pathological excitation of its targets, the internal segment of the globus pallidus (GPi) and substantia nigra pars reticulata (SNpr). Increased GPi/SNpr outflow is believed responsible for many of the cardinal symptoms of PD, i.e., tremor, rigidity, bradykinesia, and gait disturbance. A large amount of data based on lesioning, electrical stimulation, and local drug infusion studies with GABA-agonists in human PD patients have reinforced this circuit model of PD and the central role of the STN. Moreover, the closest conventional surgical intervention to our proposal, deep brain stimulation (DBS) of the STN, has shown remarkable efficacy in even late stage PD, unlike the early failures associated with recombinant GDNF infusion or cell transplantation approaches in PD. We believe that our gene transfer strategy will not only palliate symptoms by inhibiting STN activity, as with DBS, but we also have evidence that the vector converts excitatory STN projections to inhibitory projections. This additional dampening of outflow GPi/SNpr outflow may provide an additional advantage over DBS. Moreover, of perhaps the greatest interest, our preclinical data suggests that this strategy may also be neuroprotective, so this therapy may slow the degeneration of dopaminergic neurons. We will use both GAD isoforms since both are typically expressed in inhibitory neurons in the brain, and our data suggest that the combination of both isoforms is likely to be most beneficial. Our preclinical data includes three model systems: (1) old, chronically lesioned parkinsonian rats in which intraSTN GAD gene transfer results not only in improvement in both drug-induced asymmetrical behavior (apomorphine symmetrical rotations), but also in spontaneous behaviors. In our second model, GAD gene transfer precedes the generation of a dopamine lesion. Here GAD gene transfer showed remarkable neuroprotection. Finally, we carried out a study where GAD-65 and GAD-67 were used separately in monkeys that were resistant to MPTP lesioning and hence showed minimal symptomatology. Nevertheless GAD gene transfer showed no adverse effects and small improvements in both Parkinson rating scales and activity measures were obtained. In the proposed clinical trial, all patients will have met criteria for and will have given consent for STN DBS elective surgery. Twenty patients will all receive DBS electrodes, but in addition they will be randomized into two groups, to receive either a solution containing rAAV-GAD, or a solution which consists just of the vector vehicle, physiological saline. Patients, care providers, and physicians will be blind as to which solution any one patient receives. All patients, regardless of group, will agree to not have the DBS activated until the completion and unblinding of the study. Patients will be assessed with a core clinical assessment program modeled on the CAPSIT, and in addition will also undergo a preop and several postop PET scans. At the conclusion of the study, if any patient with sufficient symptomatic improvement will be offered DBS removal if they so desire. Any patients with no benefit will simply have their stimulators activated, which would normally be appropriate therapy for them and which requires no additional operations. If any unforeseen symptoms occur from STN production of GABA, this might be controlled by blocking STN GABA release with DBS, or STN lesioning could be performed using the DBS electrode. Again, this treatment would not subject the patient to additional invasive brain surgery. The trial described here reflects an evolution in our thinking about the best strategy to make a positive impact in Parkinson Disease by minimizing risk and maximizing potential benefit. To our knowledge, this proposal represents the first truly blinded, completely controlled gene or cell therapy study in the brain, which still provides the patient with the same surgical procedure which they would normally receive and should not subject the patient to additional surgical procedures regardless of the success or failure of the study. This study first and foremost aims to maximally serve the safety interests of the individual patient while simultaneously serving the public interest in rigorously determining in a scientific fashion if gene therapy can be effective to any degree in treating Parkinson's disease.


Assuntos
Protocolos Clínicos , Terapia por Estimulação Elétrica/métodos , Técnicas de Transferência de Genes , Terapia Genética/legislação & jurisprudência , Terapia Genética/métodos , Glutamato Descarboxilase/genética , Doença de Parkinson/terapia , Núcleo Celular/metabolismo , Terapia Combinada , Dependovirus/genética , Vetores Genéticos , Glutamato Descarboxilase/química , Humanos , Isoformas de Proteínas
17.
J Clin Endocrinol Metab ; 81(6): 2401-4, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8964885

RESUMO

Pituitary adenomas are common intracranial neoplasms, for which surgery and radiation are usually not curative. In attempting to develop gene therapy as a better approach to treating pituitary adenomas, we chose lactotroph adenomas as a model. The rationale for the use of this model is based on the observation that dopamine agonists decrease prolactin secretion by lactotroph adenomas, and also decrease their size. We transfected primary cultures of human lactotroph adenoma cells with an adenovirus vector containing a cDNA which encodes a human tyrosine hydroxylase, the rate-limiting enzyme in the biosynthesis of dopamine. Transfection induced expression of tyrosine hydroxylase and increased production of dopamine, resulting in the predicted biologic effect of decreased prolactin secretion. These results demonstrate the potential for gene therapy of lactotroph adenomas and perhaps other pituitary adenomas, which are less amenable to pharmacologic treatment than lactotroph adenomas.


Assuntos
Adenoma/metabolismo , Expressão Gênica , Neoplasias Hipofisárias/metabolismo , Prolactina/metabolismo , Transfecção , Tirosina 3-Mono-Oxigenase/genética , Adenoma/patologia , Adenoviridae/genética , Adulto , Dopamina/metabolismo , Feminino , Vetores Genéticos , Humanos , Levodopa/metabolismo , Masculino , Adeno-Hipófise/patologia , Neoplasias Hipofisárias/patologia , Prolactina/antagonistas & inibidores , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Mol Neurobiol ; 5(2-4): 131-41, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1668383

RESUMO

GAP-43 is a presynaptic membrane phosphoprotein that has been implicated in both the development and the modulation of neural connections. The availability of cDNA clones for GAP-43 makes it possible to examine with greater precision its role in neuronal outgrowth and physiology. We used Northern blots and in situ hybridization with GAP-43 antisense RNA probes to show that GAP-43 is expressed selectively in associative regions of the adult brain. Immunocytochemical analyses showed alterations in the pattern of GAP-43 expression in the hippocampus during reactive synaptogenesis following lesions of the perforant pathway. Genetic intervention methodology was used to analyze the molecular nature of GAP-43 involvement in synaptic plasticity. GAP-43-transfected PC12 cells displayed an enhanced response to nerve growth factor, suggesting that GAP-43 may be directly involved in neurite extension and in the modulation of the neuronal response to extrinsic trophic factors. Studies of PC12 cell transfectants, in which the synthesis of GAP-43 was blocked by expression of GAP-43 antisense RNA, showed that evoked dopamine release was significantly attenuated in these cells. The use of gene transfer into neurons with the HSV-1 vector is presented as a method of analyzing the interaction of GAP-43 with signal transduction systems during neurotransmitter release.


Assuntos
Química Encefálica , Glicoproteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Plasticidade Neuronal/fisiologia , Animais , DNA/genética , DNA Antissenso/genética , Proteína GAP-43 , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Humanos , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Antissenso/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais , Simplexvirus/genética , Transfecção
19.
FEBS Lett ; 247(2): 438-44, 1989 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-2523822

RESUMO

Extracellular fluid levels of the neurotoxin quinolinic acid in the corpus striatum of rats, measured by in vivo microdialysis, were increased in a dose-dependent manner following the intraperitoneal administration of tryptophan. The lowest dose of tryptophan (12.5 mg/kg), equivalent to about 5% of the normal daily intake, increased peak quinolinic acid levels nearly 3-fold. At higher doses of tryptophan (up to 250 mg/kg), concentrations of quinolinic acid increased over 200-fold and exceeded potentially neurotoxic levels (10 microM). In contrast, the increase in extracellular serotonin following even the highest tryptophan dose was small (less than 2-fold). These data indicate that quinolinic acid is present in the extracellular fluid where it may function as a neuromodulator and that it is very responsive to physiological changes in precursor availability.


Assuntos
Corpo Estriado/metabolismo , Espaço Extracelular/metabolismo , Piridinas/metabolismo , Ácidos Quinolínicos/metabolismo , Serotonina/metabolismo , Triptofano/metabolismo , Animais , Corpo Estriado/efeitos dos fármacos , Diálise , Relação Dose-Resposta a Droga , Cinética , Masculino , Ácido Quinolínico , Ratos , Ratos Endogâmicos , Triptofano/administração & dosagem , Triptofano/farmacologia
20.
Neurology ; 40(4): 691-5, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2181342

RESUMO

Over the past 2 decades, a number of studies have demonstrated that amino acids act as precursors for the biosynthesis of a variety of neuroactive compounds, including catecholamines and indoleamines. For example, the aromatic amino acid L-tryptophan is a precursor for serotonin biosynthesis. Based on this observed precursor relationship, dietary tryptophan supplementation is used to treat a number of neurologic disorders attributed to alterations in serotoninergic neurotransmission. Recent studies have revealed that, in addition to serotonin, a number of neuroactive compounds, the kynurenines, are metabolities of tryptophan. Of these, perhaps the most important is quinolinic acid, a neurotoxin that acts at the N-methyl-D-aspartate (NMDA) receptor and whose precursor responsiveness to tryptophan far exceeds that of serotonin. In the central nervous system, kynurenines, and in particular quinolinic acid, may modulate excitatory amino acid transmission, and may act as neurotoxic agents implicated in the pathogenesis of several neurologic diseases.


Assuntos
Cinurenina/metabolismo , Doenças do Sistema Nervoso/metabolismo , Triptofano/metabolismo , Animais , Encéfalo/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA