Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Rev Neurol (Paris) ; 180(5): 378-382, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38580500

RESUMEN

Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease, is a neurodegenerative disease caused by expanded polyglutamine repeats in exon 10 of the ataxin-3 gene, ATXN3. The accumulation of mutant ATXN3 protein leads to severe clinical manifestations and premature death. Clinically, SCA3 pathology is characterized by progressive ataxia leading to motor incoordination that may affect balance, gait and speech, and neuropathologically by a progressive degeneration of the spinal cord and cerebellum, as well as the cerebral cortex and basal ganglia. Although SCA3 is a rare disease, it is the most common autosomal dominant spinocerebellar ataxia worldwide. Its geographical distribution varies worldwide, with peak prevalence in certain regions of Brazil, Portugal and China. In 1994, the identification of the polyglutamine expansion in the ATXN3 gene made it possible not only to diagnose this pathology but also to dissect the mechanisms leading to cellular degeneration. As a monogenic disease for which only symptomatic treatment is available, the ATXN3 gene represents an attractive therapeutic target for gene editing strategies.


Asunto(s)
Ataxina-3 , Edición Génica , Enfermedad de Machado-Joseph , Humanos , Enfermedad de Machado-Joseph/terapia , Enfermedad de Machado-Joseph/genética , Ataxina-3/genética , Edición Génica/métodos , Terapia Genética/métodos , Animales , Proteínas Represoras/genética
2.
Gene Ther ; 24(10): 630-639, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28771234

RESUMEN

Huntington's disease (HD) is a fatal progressive neurodegenerative disorder caused by a mutation in the huntingtin (HTT) gene. To date, there is no treatment to halt or reverse the course of HD. Lowering of either total or only the mutant HTT expression is expected to have therapeutic benefit. This can be achieved by engineered micro (mi)RNAs targeting HTT transcripts and delivered by an adeno-associated viral (AAV) vector. We have previously showed a miHTT construct to induce total HTT knock-down in Hu128/21 HD mice, while miSNP50T and miSNP67T constructs induced allele-selective HTT knock-down in vitro. In the current preclinical study, the mechanistic efficacy and gene specificity of these selected constructs delivered by an AAV serotype 5 (AAV5) vector was addressed using an acute HD rat model. Our data demonstrated suppression of mutant HTT messenger RNA, which almost completely prevented mutant HTT aggregate formation, and ultimately resulted in suppression of DARPP-32-associated neuronal dysfunction. The AAV5-miHTT construct was found to be the most efficient, although AAV5-miSNP50T demonstrated the anticipated mutant HTT allele selectivity and no passenger strand expression. Ultimately, AAV5-delivered-miRNA-mediated HTT lowering did not cause activation of microglia or astrocytes suggesting no immune response to the AAV5 vector or therapeutic precursor sequences. These preclinical results suggest that using gene therapy to knock-down HTT may provide important therapeutic benefit for HD patients and raised no safety concerns, which supports our ongoing efforts for the development of an RNA interference-based gene therapy product for HD.


Asunto(s)
Enfermedad de Huntington/terapia , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Tratamiento con ARN de Interferencia/métodos , Animales , Dependovirus/genética , Vectores Genéticos/genética , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Masculino , Microglía/metabolismo , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/metabolismo , Tratamiento con ARN de Interferencia/efectos adversos , Ratas , Ratas Sprague-Dawley
3.
Neurobiol Dis ; 102: 105-112, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28286179

RESUMEN

Huntington's disease (HD) is a genetic neurodegenerative disorder characterized by a triad of motor, psychiatric and cognitive deficits with the latter classically attributed to disruption of fronto-striatal circuits. However, emerging evidence suggests that some of the cognitive deficits in HD may have their origin in other structures including the hippocampus. Hippocampal abnormalities have been reported in HD mouse models particularly in terms of performance on the Morris Water Maze. However, in these animals, it is difficult to be certain whether the spatial memory deficits are due to local pathology within this structure or their poor mobility and motivation. Thus, a better model of hippocampal dysfunction in HD is needed especially given that we have previously shown that patients with HD have hippocampal-related problems from the very earliest stages of disease. In this study, our aim was therefore to understand the cellular and behavioural consequences of local overexpression of mutant huntingtin (mHTT) in the hippocampus of adult mice. We found that a targeted injection of a lentivirus, encoding an N-terminal of mHTT with 82 CAG repeats, into the murine hippocampus led to the focal formation of mHTT aggregates, long-term spatial memory impairments with decreased neurogenesis and expression of the immediate early gene c-fos. This study has therefore shown for the first time that local expression of mHTT in the dentate gyrus has deleterious effects, including its neurogenic capacity, with functional behavioural consequences, which fits well with recent data on hippocampal deficits seen in patients with HD.


Asunto(s)
Hipocampo/metabolismo , Proteína Huntingtina/metabolismo , Trastornos de la Memoria/metabolismo , Neurogénesis/fisiología , Agregación Patológica de Proteínas/metabolismo , Memoria Espacial/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Hipocampo/patología , Humanos , Proteína Huntingtina/administración & dosificación , Proteína Huntingtina/genética , Enfermedad de Huntington , Lentivirus , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/patología , Ratones Endogámicos C57BL , Mutación , Neuronas/metabolismo , Neuronas/patología , Agregación Patológica de Proteínas/complicaciones , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/psicología , Proteínas Proto-Oncogénicas c-fos/metabolismo
4.
Gene Ther ; 22(10): 830-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26109254

RESUMEN

Cell-type-specific gene silencing is critical to understand cell functions in normal and pathological conditions, in particular in the brain where strong cellular heterogeneity exists. Molecular engineering of lentiviral vectors has been widely used to express genes of interest specifically in neurons or astrocytes. However, we show that these strategies are not suitable for astrocyte-specific gene silencing due to the processing of small hairpin RNA (shRNA) in a cell. Here we develop an indirect method based on a tetracycline-regulated system to fully restrict shRNA expression to astrocytes. The combination of Mokola-G envelope pseudotyping, glutamine synthetase promoter and two distinct microRNA target sequences provides a powerful tool for efficient and cell-type-specific gene silencing in the central nervous system. We anticipate our vector will be a potent and versatile system to improve the targeting of cell populations for fundamental as well as therapeutic applications.


Asunto(s)
Astrocitos/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus , ARN Interferente Pequeño , Animales , Sistema Nervioso Central/citología , Sistema Nervioso Central/fisiología , Regulación de la Expresión Génica , Silenciador del Gen , Ratones , Ratones Transgénicos , Tetraciclina
5.
Nat Med ; 2(6): 696-9, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8640564

RESUMEN

Neuronal growth factors hold promise for providing therapeutic benefits in various neurological disorders. As a means of ensuring adequate central nervous system delivery of growth factors and minimizing significant adverse side effects associated with systemic delivery methods, we have developed an ex vivo gene therapy approach for protein delivery using encapsulated genetically modified xenogeneic cells. Ciliary neurotrophic factor (CNTF) has been shown in various rodent models to reduce the motor neuron cell death similar to that seen in amyotrophic lateral sclerosis (ALS). The initial trials focusing on the systemic administration of CNTF for ALS have been discontinued as a result of major side effects, thus preventing determination of the potential efficacy of the molecule. In order to deliver CNTF directly to the nervous system, we conducted a phase I study in which six ALS patients were implanted with polymer capsules containing genetically engineered baby hamster kidney cells releasing approximately 0.5 microgram of human CNTF per day in vitro. The CNTF-releasing implants were surgically placed within the lumbar intrathecal space. Nanogram levels of CNTF were measured within the patients' cerebrospinal fluid (CSF) for at least 17 weeks post-transplantation, whereas it was undetectable before implantation. Intrathecal delivery of CNTF was not associated with the limiting side effects observed with systemic delivery. These results demonstrate that neurotrophic factors can be continuously delivered within the CSF of humans by an ex vivo gene therapy approach, opening new avenues for the treatment of neurological diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Implantes de Medicamentos/química , Inyecciones Espinales , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/uso terapéutico , Esclerosis Amiotrófica Lateral/terapia , Animales , Trasplante de Células/métodos , Factor Neurotrófico Ciliar , Cricetinae , Implantes de Medicamentos/administración & dosificación , Terapia Genética/métodos , Vectores Genéticos/química , Vectores Genéticos/genética , Humanos , Riñón/citología , Riñón/fisiología , Vértebras Lumbares/cirugía , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/uso terapéutico
6.
Science ; 290(5492): 767-73, 2000 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-11052933

RESUMEN

Lentiviral delivery of glial cell line-derived neurotrophic factor (lenti-GDNF) was tested for its trophic effects upon degenerating nigrostriatal neurons in nonhuman primate models of Parkinson's disease (PD). We injected lenti-GDNF into the striatum and substantia nigra of nonlesioned aged rhesus monkeys or young adult rhesus monkeys treated 1 week prior with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Extensive GDNF expression with anterograde and retrograde transport was seen in all animals. In aged monkeys, lenti-GDNF augmented dopaminergic function. In MPTP-treated monkeys, lenti-GDNF reversed functional deficits and completely prevented nigrostriatal degeneration. Additionally, lenti-GDNF injections to intact rhesus monkeys revealed long-term gene expression (8 months). In MPTP-treated monkeys, lenti-GDNF treatment reversed motor deficits in a hand-reach task. These data indicate that GDNF delivery using a lentiviral vector system can prevent nigrostriatal degeneration and induce regeneration in primate models of PD and might be a viable therapeutic strategy for PD patients.


Asunto(s)
Dihidroxifenilalanina/análogos & derivados , Dopamina/metabolismo , Terapia Genética , Degeneración Nerviosa/prevención & control , Factores de Crecimiento Nervioso , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/terapia , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Envejecimiento , Animales , Antígenos CD/análisis , Dihidroxifenilalanina/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial , Lentivirus/genética , Macaca mulatta , Neostriado/metabolismo , Neostriado/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/uso terapéutico , Neuronas/enzimología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/fisiopatología , Trastornos Parkinsonianos/terapia , Desempeño Psicomotor , Sustancia Negra/metabolismo , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo
8.
Hum Gene Ther ; 13(16): 1981-90, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12427308

RESUMEN

The ability to regulate gene expression constitutes a prerequisite for the development of gene therapy strategies aimed at the treatment of neurologic disorders. In the present work, we used tetracycline (Tet)-regulated lentiviral vectors to investigate the dose-dependent neuroprotective effect of human ciliary neurotrophic factor (CNTF) in the quinolinic acid (QA) model of Huntington's disease (HD). The Tet system was split in two lentiviruses, the first one containing the CNTF or green fluorescent protein (GFP) cDNAs under the control of the Tet-response element (TRE) and a second vector encoding the transactivator (tTA). Preliminary coinfection study demonstrated that 63.8% +/- 2.0% of infected cells contain at least two viral copies. Adult rats were then injected with CNTF- and GFP-expressing viral vectors followed 3 weeks later by an intrastriatal administration of QA. A significant reduction of apomorphine-induced rotations was observed in the CNTF-on group. In contrast, GFP-treated animals or CNTF-off rats displayed an ipsilateral turning behavior in response to apomorphine. A selective sparing of DARPP-32-, choline acetyltransferase (ChAT)-, and NADPH-d-positive neurons was observed in the striatum of CNTF-on rats compared to GFP animals and CNTF-off group. Enzyme-linked immunosorbent assay (ELISA) performed on striatal samples of rats sacrificed at the same time point indicated that this neuroprotective effect was associated with the production of 15.5 +/- 4.7 ng CNTF per milligram of protein whereas the residual CNTF expression in the off state (0.54 +/- 0.02 ng/mg of protein) was not sufficient to protect against QA toxicity. These results establish the proof of principle of neurotrophic factor dosing for neurodegenerative diseases and demonstrate the feasibility of lentiviral-mediated tetracycline-regulated gene transfer in the brain.


Asunto(s)
Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/farmacología , Vectores Genéticos , Enfermedad de Huntington/prevención & control , Enfermedad de Huntington/terapia , Lentivirus/genética , Ácido Quinolínico/farmacología , Tetraciclina/farmacología , Animales , Encéfalo/patología , Colina O-Acetiltransferasa/metabolismo , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Neuronas/citología , Neuronas/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/metabolismo
9.
Hum Gene Ther ; 8(16): 1881-9, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9382954

RESUMEN

Current therapy for several forms of anemia involves a weekly regime of multiple subcutaneous injections of recombinant human erythropoietin (hEpo). In an effort to provide a physiologically regulated administration of erythropoietin, we are developing cell lines genetically engineered to release hEpo as a function of oxygen tension. C2C12 cells were transfected using a vector containing the hEpo cDNA driven by the hypoxia-responsive promoter to the murine phosphoglycerate kinase gene. In vitro, these cells showed a threefold increase in hEpo secretion as oxygen levels were shifted from 21% to 1.3% oxygen. To test in vivo response, C2C12-hEpo cells were encapsulated in a microporous membrane and implanted subcutaneously on the dorsal flank of DBA/2J mice. On average, serum hEpo levels in animals exposed to 7% oxygen were two-fold higher than values seen in their control counterparts kept at 21% oxygen. Similar studies employing rats confirmed that hEpo delivery is regulated as a function of oxygen tension. These results suggest the feasibility of developing an oxygen-regulated, encapsulated cell-based system for hEpo delivery.


Asunto(s)
Eritropoyetina/genética , Regulación de la Expresión Génica , Terapia Genética/métodos , Oxígeno/fisiología , Factores de Transcripción , Transgenes , Anemia/terapia , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Composición de Medicamentos , Eritropoyetina/sangre , Eritropoyetina/metabolismo , Histocitoquímica , Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Endogámicos DBA , Proteínas Nucleares/genética , Oxígeno/sangre , Presión Parcial , Fosfoglicerato Quinasa/genética , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes
10.
Hum Gene Ther ; 7(17): 2135-46, 1996 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-8934227

RESUMEN

Neurotrophic factors hold promise for the treatment of neurodegenerative diseases. Intrathecal transplantation of polymer encapsulated cell lines genetically engineered to release neurotrophic factors provides a means to deliver them continuously behind the blood-brain barrier. Long-term delivery, however, may benefit from the use of conditionally mitotic cells to avoid the overgrowth observed with continuously dividing cell lines. Myoblast lines have all the advantages of dividing cell lines, i.e., unlimited availability, possibility for in vitro screening for the presence of pathogens, suitability for stable gene transfer and clonal selection. Furthermore they can be differentiated into a nonmitotic stage upon exposure to low-serum-containing medium. In this study, mouse C2C12 myoblasts were transfected with a pNUT expression vector containing the human ciliary neurotrophic factor (CNTF) gene. hCNTF expression and bioactivity were demonstrated by Northern blot, ELISA assay, and measurement of choline acetyltransferase (ChAT) activity in embryonic spinal cord motor neuron cultures. One C2C12 clone was found to secrete 200 ng of CNTF/10(6) cells per day. The rate of secretion of hCNTF was not altered upon differentiation of C2C12 myoblasts. A bromodeoxyuridine (BrdU) proliferation assay indicated that approximately 12% of the myoblasts continue to divide after 4 days in low-serum-containing medium. The presence of the herpes simplex thymidine kinase gene (HSV-tk) in the expression vector, however, provides a way to eliminate these dividing myoblasts upon exposure to ganciclovir, therefore increasing the safety of the encapsulation technology using established cell lines. Encapsulated hCNTF-C2C12 cells can partially rescue motor neurons from axotomy-induced cell death. In adult rats, intrathecal implantation of encapsulated hCNTF-C2C12 cells or control C2C12 confirmed the long-term survival of these cells and their potential use as a source of neurotophic factors for the treatment of neurodegenerative diseases.


Asunto(s)
Enfermedades del Sistema Nervioso Central/terapia , Regulación de la Expresión Génica , Terapia Genética , Vectores Genéticos/farmacocinética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacocinética , Animales , Antivirales/farmacología , Northern Blotting , Cápsulas , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/patología , Colina O-Acetiltransferasa/metabolismo , Factor Neurotrófico Ciliar , Sistemas de Liberación de Medicamentos/métodos , Ensayo de Inmunoadsorción Enzimática , Ganciclovir/farmacología , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos C3H , Fibras Musculares Esqueléticas/citología , Neuronas/metabolismo , Ratas , Recombinación Genética , Simplexvirus/efectos de los fármacos , Simplexvirus/genética , Timidina Quinasa/genética , Transfección
11.
Hum Gene Ther ; 15(7): 669-80, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15242527

RESUMEN

Allogeneic cells are the most attractive source for cell transplantation, as the use of xenogeneic cells is hampered by safety concerns and the use of autologous cells involves practical difficulties. The immune rejection of allogeneic cells can be overcome by physical immunoprotection provided by polymer encapsulation. To study the variability of cell and donor sources, we compared different primary human cells as candidates for gene therapy-mediated delivery of human erythropoietin (hEpo). DARC-3.1 fibroblasts, MDX-01 fibroblasts, and ARPE-19 retinal pigment epithelial cells were encapsulated into polyethersulfone hollow fibers and implanted for 1 month in nude mice as well as in immunocompetent and FK506-immunosuppressed mice to test their in vivo resistance, with the assumption that xenogeneic conditions constitute a stringent model for human application. DARC-3.1 fibroblasts showed the best survival, prompting us to evaluate cell lineages from the same donor (DARC-3.2) or another donor (DARC-4.3 and DARC-4.4). With the exception of DARC-4.3, the remaining three lineages showed comparable survival in immunocompetent C3H and DBA/2J mice. DARC-3.1 fibroblasts were retrovirally engineered with hEpo cDNA, reaching a secretion level of 170 IU of hEpo per 10(6) cells per day. Encapsulated DARC-3.1-hEpo cells led to significantly increased hematocrits in the various hosts and under various transplantation conditions. The present study shows that encapsulated primary human DARC-3.1 fibroblasts are able to survive under xenogeneic conditions and, once engineered with hEpo cDNA, to increase the hematocrit of transplanted mice.


Asunto(s)
Trasplante de Células/métodos , Eritropoyetina/genética , Fibroblastos/trasplante , Animales , Supervivencia Celular , Eritropoyetina/análisis , Eritropoyetina/biosíntesis , Fibroblastos/citología , Fibroblastos/metabolismo , Hematócrito , Humanos , Ratones , Ratones Endogámicos DBA , Polímeros/química , Sulfonas/química , Trasplante Heterólogo
12.
Hum Gene Ther ; 11(1): 179-90, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10646649

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is able to protect dopaminergic neurons against various insults and constitutes therefore a promising candidate for the treatment of Parkinson's disease. Lentiviral vectors that infect quiescent neuronal cells may allow the localized delivery of GDNF, thus avoiding potential side effects related to the activation of other brain structures. To test this hypothesis in a setting ensuring both maximal biosafety and optimal transgene expression, a self-inactivating (SIN) lentiviral vector was modified by insertion of the posttranscriptional regulatory element of the woodchuck hepatitis virus, and particles were produced with a multiply attenuated packaging system. After a single injection of 2 microl of a lacZ-expressing vector (SIN-W-LacZ) in the substantia nigra of adult rats, an average of 40.1 +/- 6.0% of the tyrosine hydroxylase (TH)-positive neurons were transduced as compared with 5.0 +/- 2.1% with the first-generation lentiviral vector. Moreover, the SIN-W vector expressing GDNF under the control of the mouse phosphoglycerate kinase 1 (PGK) promoter was able to protect nigral dopaminergic neurons after medial forebrain bundle axotomy. Expression of hGDNF in the nanogram range was detected in extracts of mesencephalon of animals injected with an SIN-W-PGK-GDNF vector, whereas it was undetectable in animals injected with a control vector. Lentiviral vectors with enhanced expression and safety features further establish the potential use of these vectors for the local delivery of bioactive molecules into defined structures of the central nervous system.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus/genética , Factores de Crecimiento Nervioso , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/genética , Transgenes , Animales , Línea Celular , ADN Complementario , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos , Prosencéfalo/metabolismo , Ratas , Ratas Wistar , Sustancia Negra/metabolismo , Transducción Genética
13.
Hum Gene Ther ; 11(12): 1723-9, 2000 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-10954906

RESUMEN

Huntington's disease (HD) is an autosomal dominant genetic disease with devastating clinical effects on cognitive, psychological, and motor functions. These clinical symptoms primarily relate to the progressive loss of medium-spiny GABA-ergic neurons of the striatum. There is no known treatment to date. Several neurotrophic factors have, however, demonstrated the capacity to protect striatal neurons in various experimental models of HD. This includes the ciliary neurotrophic factor (CNTF), the substance examined in this protocol. An ex vivo gene therapy approach based on encapsulated genetically modified BHK cells will be used for the continuous and long-term intracerebral delivery of CNTF. A device, containing up to 106 human CNTF-producing BHK cells surrounded by a semipermeable membrane, will be implanted into the right lateral ventricle of 6 patients. Capsules releasing 0.15-0.5 microg CNTF/day will be used. In this phase I study, the principal goal will be the evaluation of the safety and tolerability of the procedure. As a secondary goal, HD symptoms will be analyzed using a large battery of neuropsychological, motor, neurological, and neurophysiological tests and the striatal pathology monitored using MRI and PET-scan imaging. It is expected that the gene therapy approach described in this protocol will mitigate the side effects associated with the peripheral administration of recombinant hCNTF and allow a well-tolerated, continuous intracerebroventricular delivery of the neuroprotective factor.


Asunto(s)
Factor Neurotrófico Ciliar/genética , Terapia Genética , Enfermedad de Huntington/terapia , Animales , Línea Celular , Ventrículos Cerebrales/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Protocolos Clínicos , Ensayos Clínicos Fase I como Asunto , Cricetinae , Técnicas de Transferencia de Gen , Humanos , Selección de Paciente
14.
Hum Gene Ther ; 11(8): 1177-87, 2000 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-10834619

RESUMEN

Huntington's disease (HD) is an inherited disorder characterized by cognitive impairments, motor deficits, and progressive dementia. These symptoms result from progressive neurodegenerative changes mainly affecting the neostriatum. This pathology is fatal in 10 to 20 years and there is currently no treatment for HD. Early in the course of the disease, initial clinical manifestations are due to striatal neuronal dysfunction, which is later followed by massive neuronal death. A major therapeutic objective is therefore to reverse striatal dysfunction prior to cell death. Using a primate model reproducing the clinical features and the progressive neuronal degeneration typical of HD, we tested the therapeutic effects of direct intrastriatal infusion of ciliary neurotrophic factor (CNTF). To achieve a continuous delivery of CNTF over the full period of evaluation, we took advantage of the macroencapsulation technique. Baby hamster kidney (BHK) cells previously engineered to produce human CNTF were encapsulated into semipermeable membranes and implanted bilaterally into striata. We show here that intracerebral delivery of low doses of CNTF at the onset of symptoms not only protects neurons from degeneration but also restores neostriatal functions. CNTF-treated primates recovered, in particular, cognitive and motor functions dependent on the anatomofunctional integrity of frontostriatal pathways that were distinctively altered in this HD model. These results support the hypothesis that CNTF infusion into the striatum of HD patients not only could block the degeneration of neurons but also alleviated motor and cognitive symptoms associated with persistent neuronal dysfunction.


Asunto(s)
Encéfalo/patología , Factor Neurotrófico Ciliar/genética , Terapia Genética/métodos , Enfermedad de Huntington/terapia , Animales , Encéfalo/metabolismo , Calbindinas , Línea Celular , Factor Neurotrófico Ciliar/administración & dosificación , Convulsivantes/farmacología , Cricetinae , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Humanos , Inmunohistoquímica , Macaca fascicularis , Imagen por Resonancia Magnética , Destreza Motora , Manifestaciones Neuroconductuales , Nitrocompuestos , Propionatos/farmacología , Putamen/metabolismo , Ratas , Proteína G de Unión al Calcio S100/metabolismo , Succinato Deshidrogenasa/metabolismo , Factores de Tiempo , Transfección , Transgenes
15.
Hum Gene Ther ; 15(10): 968-75, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15585112

RESUMEN

Huntington's disease (HD) is a monogenic neurodegenerative disease that affects the efferent neurons of the striatum. The protracted evolution of the pathology over 15 to 20 years, after clinical onset in adulthood, underscores the potential of therapeutic tools that would aim at protecting striatal neurons. Proteins with neuroprotective effects in the adult brain have been identified, among them ciliary neurotrophic factor (CNTF), which protected striatal neurons in animal models of HD. Accordingly, we have carried out a phase I study evaluating the safety of intracerebral administration of this protein in subjects with HD, using a device formed by a semipermeable membrane encapsulating a BHK cell line engineered to synthesize CNTF. Six subjects with stage 1 or 2 HD had one capsule implanted into the right lateral ventricle; the capsule was retrieved and exchanged for a new one every 6 months, over a total period of 2 years. No sign of CNTF-induced toxicity was observed; however, depression occurred in three subjects after removal of the last capsule, which may have correlated with the lack of any future therapeutic option. All retrieved capsules were intact but contained variable numbers of surviving cells, and CNTF release was low in 13 of 24 cases. Improvements in electrophysiological results were observed, and were correlated with capsules releasing the largest amount of CNTF. This phase I study shows the safety, feasibility, and tolerability of this gene therapy procedure. Heterogeneous cell survival, however, stresses the need for improving the technique.


Asunto(s)
Terapia Genética/métodos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Fármacos Neuroprotectores/farmacología , Animales , Encéfalo/metabolismo , Línea Celular , Supervivencia Celular , Factor Neurotrófico Ciliar/química , Factor Neurotrófico Ciliar/genética , Codón , Cricetinae , Electrofisiología , Femenino , Técnicas de Transferencia de Gen , Humanos , Masculino , Neuronas/metabolismo , Polímeros/química , Retroviridae/genética , Factores de Tiempo
16.
Hum Gene Ther ; 7(7): 851-60, 1996 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-8860837

RESUMEN

The gene therapy approach presented in this protocol employs a polymer encapsulated, xenogenic, transfected cell line to release human ciliary neurotrophic factor (hCNTF) for the treatment of Amyotrophic Lateral Sclerosis (ALS). A tethered device, containing around 10(6) genetically modified cells surrounded by a semipermeable membrane, is implanted intrathecally; it provides for slow continuous release of hCNTF at a rate of 0.25 to 1.0 micrograms/24 hours. The semipermeable membrane prevents immunologic rejection of the cells and interposes a physical, virally impermeable barrier between cells and host. Moreover, the device and the cells it contains may be retrieved in the event of side effects. A vector containing the human CNTF gene was transfected into a line of baby hamster kidney cells (BHK) with calcium phosphate using a dihydrofolate reductase-based selection vector with a SV40 promoter and contains a HSV-tk killer gene. hCNTF is a potent neurotrophic factor which may have utility for the treatment of ALS. Systemic delivery of hCNTF in humans has been frustrated by peripheral side effects, the molecule's short half life, and its inability to cross the blood-brain barrier. The gene therapy approach described in this protocol is expected to mitigate such difficulties by local intrathecal delivery of a known quantity of continuously-synthesized hCNTF from a retrievable implant.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Terapia Genética/métodos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/uso terapéutico , Prótesis e Implantes , Animales , Cápsulas/química , Cápsulas/uso terapéutico , Línea Celular , Trasplante de Células/métodos , Células Cultivadas , Factor Neurotrófico Ciliar , Protocolos Clínicos , Cricetinae , Ganciclovir/farmacología , Vectores Genéticos/genética , Vectores Genéticos/farmacología , Vectores Genéticos/toxicidad , Humanos , Riñón/citología , Proteínas del Tejido Nervioso/efectos adversos , Polímeros/química , Polímeros/uso terapéutico , Primates , Ratas , Ovinos , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Transfección
17.
Gene ; 122(2): 297-304, 1992 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-1487144

RESUMEN

Developmentally regulated mechanisms involving alternative RNA splicing and/or polyadenylation, as well as transcription termination, are implicated in controlling the levels of secreted mu (mu s), membrane mu (mu m) and delta immunoglobulin (Ig) heavy chain mRNAs during B cell differentiation (mu gene encodes the mu heavy chain). Using expression vectors constructed with genomic DNA segments composed of the mu m polyadenylation signal region, we analyzed poly(A) site utilization and termination of transcription in stably transfected myeloma cells and in murine fibroblast L cells. We found that the gene segment containing the mu m poly(A) signals, along with 536 bp of downstream flanking sequence, acted as a transcription terminator in both myeloma cells and L cell fibroblasts. Neither a 141-bp DNA fragment (which directed efficient polyadenylation at the mu m site), nor the 536-bp flanking nucleotide sequence alone, were sufficient to obtain a similar regulation. This shows that the mu m poly(A) region plays a central role in controlling developmentally regulated transcription termination by blocking downstream delta gene expression. Because this gene segment exhibited the same RNA processing and termination activities in fibroblasts, it appears that these processes are not tissue-specific.


Asunto(s)
Genes de Inmunoglobulinas , Cadenas mu de Inmunoglobulina/genética , Poli A , Regiones Terminadoras Genéticas , Empalme Alternativo , Animales , Secuencia de Bases , Northern Blotting , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Células L , Ratones , Datos de Secuencia Molecular , Células Tumorales Cultivadas
18.
Novartis Found Symp ; 231: 202-15; discussion 215-9, 302-6, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11131539

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor disturbances caused by an alteration of the dopaminergic nigrostriatal system. Current symptomatic treatments for PD include dopaminergic drug administration, deep brain stimulation, ablative surgery and fetal cell transplantation. Though these approaches have significant beneficial effects, they are hampered by limiting side-effects, but more importantly they do not change the disease progression. Alternative restorative and neuroprotective strategies have therefore to be considered. Neuroprotective effects of neurotrophic factors, anti-apoptotic and antioxidant molecules are currently being investigated for this purpose. Among neurotrophic molecules, the potential of the glial cell line-derived neurotrophic factor (GDNF) to protect the nigral dopaminergic neurons and/or rescue striatal dopamine levels has been extensively documented. For GDNF to become a clinical reality, appropriate delivery techniques will have to be developed. This chapter focuses on the potential of encapsulated cells and viral vectors to locally release neurotrophic factors in experimental models of PD.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Factores de Crecimiento Nervioso , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores , Enfermedad de Parkinson/terapia , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos
19.
Cell Transplant ; 5(5): 577-87, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8889216

RESUMEN

The neurodegenerative disease amyotrophic lateral sclerosis (ALS) results from the progressive loss of motoneurons, leading to death in a few years. Ciliary neurotrophic factor (CNTF), which decreases naturally occurring and axotomy-induced cell death, may result in slowing of motoneuron loss and has been evaluated as a treatment for ALS. Effective administration of this protein to motoneurons may be hampered by the exceedingly short half-life of CNTF, and the inability to deliver effective concentration into the central nervous system after systemic administration in vivo. The constitutive release of CNTF from genetically engineered cells may represent a solution to this delivery problem. In this work, baby hamster kidney (BHK) cells stably tranfected with a chimeric plasmid construct containing the gene for human or mouse CNTF were encapsulated in polymer fibers, which prevents immune rejection and allow long-term survival of the transplanted cells. In vitro bioassays show that the encapsulated transfected cells release bioactive CNTF. In vivo, systemic delivery of human and mouse CNTF from encapsulated cells was observed to rescue 26 and 27% more facial motoneurons, respectively, as compared to capsules containing parent BHK cells 1 wk postaxotomy in neonatal rats. With local application of CNTF on the nerve stump and by systemic delivery through repeated subcutaneous injections, 15 and 13% more rescue effects were observed. These data illustrate the potential of using encapsulated genetically engineered cells to continuously release CNTF to slow down motoneuron degeneration following axotomy and suggest that encapsulated cell delivery of neurotrophic factors may provide a general method for effective administration of therapeutic proteins for the treatment of neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Axones/ultraestructura , Sistemas de Liberación de Medicamentos/métodos , Neuronas Motoras/citología , Proteínas del Tejido Nervioso/metabolismo , Animales , Northern Blotting , Muerte Celular , Células Cultivadas , Embrión de Pollo , Factor Neurotrófico Ciliar , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Ingeniería Genética/métodos , Humanos , Ratones , Neuronas Motoras/trasplante , Polímeros , Ratas , Ratas Sprague-Dawley
20.
Transl Psychiatry ; 3: e253, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23632457

RESUMEN

The therapeutic activity of selective serotonin (5-HT) reuptake inhibitors (SSRIs) relies on long-term adaptation at pre- and post-synaptic levels. The sustained administration of SSRIs increases the serotonergic neurotransmission in response to a functional desensitization of the inhibitory 5-HT1A autoreceptor in the dorsal raphe. At nerve terminal such as the hippocampus, the enhancement of 5-HT availability increases brain-derived neurotrophic factor (BDNF) synthesis and signaling, a major event in the stimulation of adult neurogenesis. In physiological conditions, BDNF would be expressed at functionally relevant levels in neurons. However, the recent observation that SSRIs upregulate BDNF mRNA in primary cultures of astrocytes strongly suggest that the therapeutic activity of antidepressant drugs might result from an increase in BDNF synthesis in this cell type. In this study, by overexpressing BDNF in astrocytes, we balanced the ratio between astrocytic and neuronal BDNF raising the possibility that such manipulation could positively reverberate on anxiolytic-/antidepressant-like activities in transfected mice. Our results indicate that BDNF overexpression in hippocampal astrocytes produced anxiolytic-/antidepressant-like activity in the novelty suppressed feeding in relation with the stimulation of hippocampal neurogenesis whereas it did not potentiate the effects of the SSRI fluoxetine on these parameters. Moreover, overexpressing BDNF revealed the anxiolytic-like activity of fluoxetine in the elevated plus maze while attenuating 5-HT neurotransmission in response to a blunted downregulation of the 5-HT1A autoreceptor. These results emphasize an original role of hippocampal astrocytes in the synthesis of BDNF, which can act through neurogenesis-dependent and -independent mechanisms to regulate different facets of anxiolytic-like responses.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Neurogénesis/fisiología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Antidepresivos de Segunda Generación/farmacología , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Ansiedad/fisiopatología , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/análisis , Factor Neurotrófico Derivado del Encéfalo/fisiología , Depresión/tratamiento farmacológico , Depresión/metabolismo , Depresión/fisiopatología , Fluoxetina/farmacología , Expresión Génica/fisiología , Hipocampo/química , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Masculino , Ratones , Piperazinas/farmacología , Piridinas/farmacología , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA