Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
2.
Mol Ther ; 29(9): 2806-2820, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34298128

RESUMEN

Non-human primates (NHPs) are a preferred animal model for optimizing adeno-associated virus (AAV)-mediated CNS gene delivery protocols before clinical trials. In spite of its inherent appeal, it is challenging to compare different serotypes, delivery routes, and disease indications in a well-powered, comprehensive, multigroup NHP experiment. Here, a multiplex barcode recombinant AAV (rAAV) vector-tracing strategy has been applied to a systemic analysis of 29 distinct, wild-type (WT), AAV natural isolates and engineered capsids in the CNS of eight macaques. The report describes distribution of each capsid in 15 areas of the macaques' CNS after intraparenchymal (putamen) injection, or cerebrospinal fluid (CSF)-mediated administration routes (intracisternal, intrathecal, or intracerebroventricular). To trace the vector biodistribution (viral DNA) and targeted tissues transduction (viral mRNA) of each capsid in each of the analyzed CNS areas, quantitative next-generation sequencing analysis, assisted by the digital-droplet PCR technology, was used. The report describes the most efficient AAV capsid variants targeting specific CNS areas after each route of administration using the direct side-by-side comparison of WT AAV isolates and a new generation of rationally designed capsids. The newly developed bioinformatics and visualization algorithms, applicable to the comparative analysis of several mammalian brain models, have been developed and made available in the public domain.


Asunto(s)
Proteínas de la Cápside/genética , Sistema Nervioso Central/química , Dependovirus/fisiología , Vectores Genéticos/administración & dosificación , Algoritmos , Animales , Sistema Nervioso Central/virología , ADN Viral/genética , Bases de Datos Genéticas , Dependovirus/genética , Vías de Administración de Medicamentos , Secuenciación de Nucleótidos de Alto Rendimiento , Primates , ARN Mensajero/genética , ARN Viral/genética , Distribución Tisular , Transducción Genética
3.
Gene Ther ; 28(7-8): 447-455, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33244179

RESUMEN

Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal recessive lysosomal disease caused by defective production of the enzyme α-N-acetylglucosaminidase. It is characterized by severe and complex central nervous system degeneration. Effective therapies will likely target early onset disease and overcome the blood-brain barrier. Modifications of adeno-associated viral (AAV) vector capsids that enhance transduction efficiency have been described in the retina. Herein, we describe for the first time, a transduction assessment of two intracranially administered adeno-associated virus serotype 8 variants, in which specific surface-exposed tyrosine (Y) and threonine (T) residues were substituted with phenylalanine (F) and valine (V) residues, respectively. A double-mutant (Y444 + 733F) and a triple-mutant (Y444 + 733F + T494V) AAV8 were evaluated for their efficacy for the potential treatment of MPS IIIB in a neonatal setting. We evaluated biodistribution and transduction profiles of both variants compared to the unmodified parental AAV8, and assessed whether the method of vector administration would modulate their utility. Vectors were administered through four intracranial routes: six sites (IC6), thalamic (T), intracerebroventricular, and ventral tegmental area into neonatal mice. Overall, we conclude that the IC6 method resulted in the widest biodistribution within the brain. Noteworthy, we demonstrate that GFP intensity was significantly more robust with AAV8 (double Y-F + T-V) compared to AAV8 (double Y-F). This provides proof of concept for the enhanced utility of IC6 administration of the capsid modified AAV8 (double Y-F + T-V) as a valid therapeutic approach for the treatment of MPS IIIB, with further implications for other monogenic diseases.


Asunto(s)
Cápside , Mucopolisacaridosis III , Animales , Encéfalo , Dependovirus/genética , Vectores Genéticos/genética , Ratones , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/terapia , Distribución Tisular , Transducción Genética
5.
Exp Neurol ; 325: 113159, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31843492

RESUMEN

Parkinson's disease (PD) is a debilitating condition resulting in motor and non-motor symptoms affecting approximately 10 million people worldwide. Currently, there are no pharmacological treatments that can cure the condition or effectively halt its progression. The focus of PD research has been primarily on the neurobiological basis and consequences of dopamine (DA) neuron degeneration given that the loss of DA neurons projecting from the substantia nigra to the dorsal striatum results in the development of cardinal PD motor symptoms. Alternatively, gastrointestinal dysfunction is well recognized in PD patients, and often occurs prior to the development of motor symptoms. The gut microbiota, which contains thousands of bacterial species, play important roles in intestinal barrier integrity and function, metabolism, immunity and brain function. Pre-clinical and clinical studies suggest an important link between alterations in the composition of the gut microbiota and psychiatric and neurological conditions, including PD. Several reports have documented gut dysbiosis and alterations in the composition of the gut microbiota in PD patients. Therefore, the goal of this study was to explore the contribution of the gut microbiota to the behavioral and neurochemical alterations in a rodent toxin model of DA depletion that reproduces the motor symptoms associated with PD. We observed that chronic treatment of adult rats with non-absorbable antibiotics ameliorates the neurotoxicity of 6-hydroxydopamine (6-OHDA) in a unilateral lesion model. Specifically, immunohistochemistry against the dopaminergic neuron marker tyrosine hydroxylase (TH) showed an attenuation of the degree of 6-OHDA-induced dopaminergic neuron loss in antibiotic treated animals compared to control animals. In addition, we observed a reduction in the expression of pro-inflammatory markers in the striatum of antibiotic-treated animals. The degree of motor dysfunction after 6-OHDA was also attenuated in antibiotic-treated animals as measured by paw-rearing measurements in the cylinder test, forepaw stepping test, and ipsilateral rotations observed in the amphetamine-induced rotation test. These results implicate the gut microbiota as a potential contributor to pathology in the development of PD. Further studies are necessary to understand the specific mechanisms involved in transducing alterations in the gut microbiota to changes in dopaminergic neuron loss and motor dysfunction.


Asunto(s)
Antibacterianos/farmacología , Neuronas Dopaminérgicas/patología , Microbioma Gastrointestinal/efectos de los fármacos , Degeneración Nerviosa/patología , Trastornos Parkinsonianos/patología , Animales , Bacitracina/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Natamicina/farmacología , Neomicina/farmacología , Degeneración Nerviosa/etiología , Trastornos Parkinsonianos/complicaciones , Trastornos Parkinsonianos/microbiología , Ratas , Ratas Sprague-Dawley , Vancomicina/farmacología
6.
Acta Neuropathol Commun ; 5(1): 47, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28619074

RESUMEN

Multiple system atrophy (MSA) is a horrible and unrelenting neurodegenerative disorder with an uncertain etiology and pathophysiology. MSA is a unique proteinopathy in which alpha-synuclein (α-syn) accumulates preferentially in oligodendroglia rather than neurons. Glial cytoplasmic inclusions (GCIs) of α-syn are thought to elicit changes in oligodendrocyte function, such as reduced neurotrophic support and demyelination, leading to neurodegeneration. To date, only a murine model using one of three promoters exist to study this disease. We sought to develop novel rat and nonhuman primate (NHP) models of MSA by overexpressing α-syn in oligodendroglia using a novel oligotrophic adeno-associated virus (AAV) vector, Olig001. To establish tropism, rats received intrastriatal injections of Olig001 expressing GFP. Histological analysis showed widespread expression of GFP throughout the striatum and corpus callosum with >95% of GFP+ cells co-localizing with oligodendroglia and little to no expression in neurons or astrocytes. We next tested the efficacy of this vector in rhesus macaques with intrastriatal injections of Olig001 expressing GFP. As in rats, we observed a large number of GFP+ cells in gray matter and white matter tracts of the striatum and the corpus callosum, with 90-94% of GFP+ cells co-localizing with an oligodendroglial marker. To evaluate the potential of our vector to elicit MSA-like pathology in NHPs, we injected rhesus macaques intrastriatally with Olig001 expressing the α-syn transgene. Histological analysis 3-months after injection demonstrated widespread α-syn expression throughout the striatum as determined by LB509 and phosphorylated serine-129 α-syn immunoreactivity, all of which displayed as tropism similar to that seen with GFP. As in MSA, Olig001-α-syn GCIs in our model were resistant to proteinase K digestion and caused microglial activation. Critically, demyelination was observed in the white matter tracts of the corpus callosum and striatum of Olig001-α-syn but not Olig001-GFP injected animals, similar to the human disease. These data support the concept that this vector can provide novel rodent and nonhuman primate models of MSA.


Asunto(s)
Encéfalo/metabolismo , Modelos Animales de Enfermedad , Atrofia de Múltiples Sistemas/metabolismo , Oligodendroglía/metabolismo , alfa-Sinucleína/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/patología , Dependovirus/genética , Endopeptidasa K/metabolismo , Femenino , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Macaca mulatta , Masculino , Microglía/metabolismo , Microglía/patología , Atrofia de Múltiples Sistemas/patología , Neuronas/metabolismo , Neuronas/patología , Oligodendroglía/patología , Ratas Sprague-Dawley , alfa-Sinucleína/genética
7.
Front Aging Neurosci ; 8: 127, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27303293

RESUMEN

The primary cilia of forebrain neurons assemble around birth and become enriched with neuromodulatory receptors. Our understanding of the permanence of these structures and their associated signaling pathways in the aging brain is poor, but they are worthy of investigation because disruptions in neuronal cilia signaling have been implicated in changes in learning and memory, depression-like symptoms, and sleep anomalies. Here, we asked whether neurons in aged forebrain retain primary cilia and whether the staining characteristics of aged cilia for type 3 adenylyl cyclase (ACIII), somatostatin receptor 3 (SSTR3), and pericentrin resemble those of cilia in younger forebrain. To test this, we analyzed immunostained sections of forebrain tissues taken from young and aged male Fischer 344 (F344) and F344 × Brown Norway (F344 × BN) rats. Analyses of ACIII and SSTR3 in young and aged cortices of both strains of rats revealed that the staining patterns in the neocortex and hippocampus were comparable. Virtually every NeuN positive cell examined possessed an ACIII positive cilium. The lengths of ACIII positive cilia in neocortex were similar between young and aged for both strains, whereas in F344 × BN hippocampus, the cilia lengths increased with age in CA1 and CA3, but not in dentate gyrus (DG). Additionally, the percentages of ACIII positive cilia that were also SSTR3 positive did not differ between young and aged tissues in either strain. We also found that pericentrin, a protein that localizes to the basal bodies of neuronal cilia and functions in primary cilia assembly, persisted in aged cortical neurons of both rat strains. Collectively, our data show that neurons in aged rat forebrain possess primary cilia and that these cilia, like those present in younger brain, continue to localize ACIII, SSTR3, and pericentrin. Further studies will be required to determine if the function and signaling pathways regulated by cilia are similar in aged compared to young brain.

8.
Neurobiol Dis ; 48(2): 212-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22426391

RESUMEN

The field of in vivo gene therapy has matured to the point where there are numerous clinical trials underway including late-stage clinical trials. Several viral vectors are especially efficient and support lifetime protein expression in the brain and a number of clinical trials are underway for various progressive or chronic neurological disorders including Parkinson's disease, Alzheimer's disease, and Batten's disease. To date, however, none of the vectors in clinical use have any direct way to reverse or control their transgene product in the event continued protein expression should become problematic. Several schemes that use elements within the vector design have been developed that allow an external drug or pro-drug to alter ongoing protein expression after in vivo gene transfer. The most promising and most studied regulated protein expression methods for in vivo gene transfer are reviewed. In addition, potential scientific and clinical advantages of transgene regulation for gene therapy are discussed.


Asunto(s)
Regulación de la Expresión Génica/genética , Terapia Genética/métodos , Enfermedades del Sistema Nervioso/terapia , Animales , Terapia Genética/tendencias , Vectores Genéticos , Humanos , Regiones Promotoras Genéticas , Bibliotecas de Moléculas Pequeñas , Transcripción Genética/genética , Transcripción Genética/fisiología , Transgenes , Virus/genética
9.
Mol Ther ; 20(3): 544-54, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22008908

RESUMEN

Neurotrophic factors are integrally involved in the development of the nigrostriatal system and in combination with gene therapy, possess great therapeutic potential for Parkinson's disease (PD). Pleiotrophin (PTN) is involved in the development, maintenance, and repair of the nigrostriatal dopamine (DA) system. The present study examined the ability of striatal PTN overexpression, delivered via psueudotyped recombinant adeno-associated virus type 2/1 (rAAV2/1), to provide neuroprotection and functional restoration from 6-hydroxydopamine (6-OHDA). Striatal PTN overexpression led to significant neuroprotection of tyrosine hydroxylase immunoreactive (THir) neurons in the substantia nigra pars compacta (SNpc) and THir neurite density in the striatum, with long-term PTN overexpression producing recovery from 6-OHDA-induced deficits in contralateral forelimb use. Transduced striatal PTN levels were increased threefold compared to adult striatal PTN expression and approximated peak endogenous developmental levels (P1). rAAV2/1 vector exclusively transduced neurons within the striatum and SNpc with approximately half the total striatal volume routinely transduced using our injection parameters. Our results indicate that striatal PTN overexpression can provide neuroprotection for the 6-OHDA lesioned nigrostriatal system based upon morphological and functional measures and that striatal PTN levels similar in magnitude to those expressed in the striatum during development are sufficient to provide neuroprotection from Parkinsonian insult.


Asunto(s)
Proteínas Portadoras/genética , Cuerpo Estriado/metabolismo , Citocinas/genética , Trastornos Parkinsonianos/terapia , Animales , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Orden Génico , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Masculino , Oxidopamina , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/genética , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Transducción Genética
10.
Exp Neurol ; 229(2): 201-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21459087

RESUMEN

The use of recombinant lentiviral vectors (rLV) is emerging as a viable candidate for clinical gene therapy of the central nervous system. New generation vectors are being produced while addressing viral safety concerns as well as production capabilities. Furthermore, the ability to combine envelope proteins targeting specific cell types with specific promoters guiding the expression of the genetic payload will allow researchers and clinicians to precisely guide transgene expression to anatomically and phenotypically distinct populations of cells. In a recent issue of Experimental Neurology, Cannon and colleagues demonstrate the ability to transduce specific populations of cells in the rat midbrain by using differently pseudotyped lentiviral vectors which results in significant differences in transgene spread throughout the nigrostriatal tract. These results highlight the potential utility of rLV in clinical applications as well as in research involving neurodegenerative disease.


Asunto(s)
Vectores Genéticos/genética , Lentivirus/genética , Transducción Genética/métodos , Animales , Ratas
11.
Hum Mol Genet ; 20(14): 2770-82, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21515588

RESUMEN

Recent studies have implicated an N-terminal caspase-6 cleavage product of mutant huntingtin (htt) as an important mediator of toxicity in Huntington's disease (HD). To directly assess the consequences of such fragments on neurologic function, we produced transgenic mice that express a caspase-6 length N-terminal fragment of mutant htt (N586) with both normal (23Q) and disease (82Q) length glutamine repeats. In contrast to mice expressing N586-23Q, mice expressing N586-82Q accumulate large cytoplasmic inclusion bodies that can be visualized with antibodies to epitopes throughout the N586 protein. However, biochemical analyses of aggregated mutant huntingtin in these mice demonstrated that the inclusion bodies are composed largely of a much smaller htt fragment (terminating before residue 115), with lesser amounts of full-length N586-82Q fragments. Mice expressing the N586-82Q fragment show symptoms typical of previously generated mice expressing mutant huntingtin fragments, including failure to maintain weight, small brain weight and reductions in specific mRNAs in the striatum. Uniquely, these N586-82Q mice develop a progressive movement disorder that includes dramatic deficits in motor performance on the rotarod and ataxia. Our findings suggest that caspase-6-derived fragments of mutant htt are capable of inducing novel HD-related phenotypes, but these fragments are not terminal cleavage products as they are subject to further proteolysis. In this scenario, mutant htt fragments derived from caspase 6, or possibly other proteases, could mediate HD pathogenesis via a 'hit and run' type of mechanism in which caspase-6, or other larger N-terminal fragments, mediate a neurotoxic process before being cleaved to a smaller fragment that accumulates pathologically.


Asunto(s)
Cuerpo Estriado/metabolismo , Expresión Génica , Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/metabolismo , Mutación Missense , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Sustitución de Aminoácidos , Animales , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , Caspasa 6 , Cuerpo Estriado/patología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Cuerpos de Inclusión/patología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Prueba de Desempeño de Rotación con Aceleración Constante
13.
Exp Neurol ; 228(2): 173-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21192926

RESUMEN

The discovery of the gene mutation responsible for Huntington's disease (HD), huntingtin, in 1993 allowed for a better understanding of the pathology of and enabled the development of animal models. HD is caused by the expansion of a polyglutamine repeat region in the N-terminal of the huntingtin protein. Here we examine the behavioral, transcriptional, histopathological and anatomical characteristics of a knock-in HD mouse model with a 140 polyglutamine expansion in the huntingtin protein. This CAG 140 model contains a portion of the human exon 1 with 140 CAG repeats knocked into the mouse huntingtin gene. We have longitudinally examined the rearing behavior, accelerating rotarod, constant speed rotarod and gait for age-matched heterozygote, homozygote and non-transgenic mice and have found a significant difference in the afflicted mice. However, while there were significant differences between the non-transgenic and the knock-in mice, these behaviors were not progressive. As in HD, we show that the CAG 140 mice also have a significant decrease in striatally enriched mRNA transcripts. In addition, striatal neuronal intranuclear inclusion density increases with age. Lastly these CAG 140 mice show slight cortical thinning compared to non-transgenic mice, similarly to the cortical thinning recently reported in HD.


Asunto(s)
Conducta Animal/fisiología , Técnicas de Sustitución del Gen/métodos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Estudios Transversales , Modelos Animales de Enfermedad , Femenino , Marcha/genética , Tamización de Portadores Genéticos/métodos , Homocigoto , Humanos , Proteína Huntingtina , Enfermedad de Huntington/psicología , Estudios Longitudinales , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/genética , Prueba de Desempeño de Rotación con Aceleración Constante
14.
Mol Ther ; 18(10): 1758-68, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20664530

RESUMEN

We present genetic evidence that an in vivo role of α-synuclein (α-syn) is to inhibit phospholipase D2 (PLD2), an enzyme that is believed to participate in vesicle trafficking, membrane signaling, and both endo- and exocytosis. Overexpression of PLD2 in rat substantia nigra pars compacta (SNc) caused severe neurodegeneration of dopamine (DA) neurons, loss of striatal DA, and an associated ipsilateral amphetamine-induced rotational asymmetry. Coexpression of human wild type α-syn suppressed PLD2 neurodegeneration, DA loss, and amphetamine-induced rotational asymmetry. However, an α-syn mutant defective for inhibition of PLD2 in vitro also failed to inhibit PLD toxicity in vivo. Further, reduction of PLD2 activity in SNc, either by siRNA knockdown of PLD2 or overexpression of α-syn, both produced an unusual contralateral amphetamine-induced rotational asymmetry, opposite to that seen with overexpression of PLD2, suggesting that PLD2 and α-syn were both involved in DA release or reuptake. Finally, α-syn coimmunoprecipitated with PLD2 from extracts prepared from striatal tissues. Taken together, our data demonstrate that α-syn is an inhibitor of PLD2 in vivo, and confirm earlier reports that α-syn inhibits PLD2 in vitro. Our data also demonstrate that it is possible to use viral-mediated gene transfer to study gene interactions in vivo.


Asunto(s)
Degeneración Nerviosa/metabolismo , Fosfolipasa D/metabolismo , Sustancia Negra/metabolismo , Sustancia Negra/patología , alfa-Sinucleína/metabolismo , Animales , Dependovirus/genética , Dopamina/metabolismo , Vectores Genéticos/genética , Immunoblotting , Inmunohistoquímica , Microscopía Confocal , Degeneración Nerviosa/genética , Fosfolipasa D/genética , Plásmidos/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , alfa-Sinucleína/genética
15.
Mol Ther ; 18(8): 1450-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20551914

RESUMEN

Two small-interfering RNAs (siRNAs) targeting alpha-synuclein (alpha-syn) and three control siRNAs were cloned in an adeno-associated virus (AAV) vector and unilaterally injected into rat substantia nigra pars compacta (SNc). Reduction of alpha-syn resulted in a rapid (4 week) reduction in the number of tyrosine hydroxylase (TH) positive cells and striatal dopamine (DA) on the injected side. The level of neurodegeneration induced by the different siRNAs correlated with their ability to downregulate alpha-syn protein and mRNA in tissue culture and in vivo. Examination of various SNc neuronal markers indicated that neurodegeneration was due to cell loss and not just downregulation of DA synthesis. Reduction of alpha-syn also resulted in a pronounced amphetamine induced behavioral asymmetry consistent with the level of neurodegeneration. In contrast, none of the three control siRNAs, which targeted genes not normally expressed in SNc, showed evidence of neurodegeneration or behavioral asymmetry, even at longer survival times. Moreover, co-expression of both rat alpha-syn and alpha-syn siRNA partially reversed the neurodegenerative and behavioral effects of alpha-syn siRNA alone. Our data show that alpha-syn plays an important role in the rat SNc and suggest that both up- and downregulation of wild-type alpha-syn expression increase the risk of nigrostriatal pathology.


Asunto(s)
Sustancia Negra/metabolismo , Sustancia Negra/patología , alfa-Sinucleína/metabolismo , Animales , Encéfalo , Silenciador del Gen/fisiología , Humanos , Immunoblotting , Inmunohistoquímica , Microscopía Confocal , Interferencia de ARN , Ratas , alfa-Sinucleína/genética
16.
Curr Opin Mol Ther ; 12(2): 240-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20373268

RESUMEN

To date, only five drugs have been approved for the treatment of Alzheimer's disease (AD); however, these agents impact the symptoms rather than the progression of the disease. It is well established that nerve growth factor (NGF) enhances the function and survival of basal forebrain cholinergic neurons that are vulnerable in AD. However, NGF does not cross the blood-brain barrier, and intraventricular NGF injections in animals and humans were associated with significant side effects. Adeno-associated virus (AAV)-based gene delivery is a novel technology being developed for administration of NGF to the brain to treat AD symptoms and progression. Indeed, the efficacy of ex vivo gene delivery was demonstrated in patients with AD who experienced improvements in cerebral metabolism and cognition compared with pre-operative function without adverse events. CERE-110 (AAV2-NGF), under development by Ceregene Inc, is an AAV serotype 2-based vector expressing human NGF delivered to the nucleus basalis of Meynert by stereotactic injection for the treatment of AD. Animal studies have established the preclinical efficacy of CERE-110, revealing an excellent safety profile. CERE-110 has passed phase I clinical testing and a multicenter phase II clinical trial has commenced. CERE-110 is a promising candidate for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Factores de Crecimiento Nervioso/genética , Anciano , Enfermedad de Alzheimer/genética , Animales , Ensayos Clínicos como Asunto , Dependovirus/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos
17.
Discov Med ; 9(46): 204-11, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20350486

RESUMEN

Given improvements in viral vector design, production and efficiency of transduction in the central nervous system (CNS), as well as increased knowledge of neuropathological mechanisms in neurological disorders, success in treating a CNS disorder with gene transfer seems inevitable. Several different vector systems have been studied extensively and the adeno-associated viral vector system has been utilized in most early stage clinical trials in neurological disorders. Other vector systems, such as lentivirus, adenovirus, and herpes simplex virus are also viable vector platforms that should fill significant clinical niches based on their specific characteristics. In addition to the choice of the appropriate vector, the proper choice of transgene for the appropriate strategy to treat a neurological disorder is also critical. The example of glial cell line-derived neurotrophic factor ligands to treat Parkinson's disease is used to illustrate the importance of the interface between interpretation of pre-clinical data and consideration of the natural history of the disorder. This interface dictates the proper design of clinical trials that are capable of testing whether the treatment is actually successful.


Asunto(s)
Terapia Genética/métodos , Enfermedad de Parkinson/terapia , Ensayos Clínicos como Asunto , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factores Neurotróficos Derivados de la Línea Celular Glial/uso terapéutico , Humanos
18.
Mol Ther ; 18(3): 579-87, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19773746

RESUMEN

We examined the transduction efficiency of different adeno-associated virus (AAV) capsid serotypes encoding for green fluorescent protein (GFP) flanked by AAV2 inverted terminal repeats in the nonhuman primate basal ganglia as a prelude to translational studies, as well as clinical trials in patients with Parkinson's disease (PD). Six intact young adult cynomolgus monkeys received a single 10 microl injection of AAV2/1-GFP, AAV2/5-GFP, or AAV2/8-GFP pseudotyped vectors into the caudate nucleus and putamen bilaterally in a pattern that resulted in each capsid serotype being injected into at least four striatal sites. GFP immunohistochemistry revealed excellent transduction rates for each AAV pseudotype. Stereological estimates of GFP+ cells within the striatum revealed that AAV2/5-GFP transduces significantly higher number of cells than AAV2/8-GFP (P < 0.05) and there was no significant difference between AAV2/5-GFP and AAV2/1-GFP (P = 0.348). Consistent with this result, Cavalieri estimates revealed that AAV2/5-GFP resulted in a significantly larger transduction volume than AAV2/8-GFP (P < 0.05). Each pseudotype transduced striatal neurons effectively [>95% GFP+ cells colocalized neuron-specific nuclear protein (NeuN)]. The current data suggest that AAV2/5 and AAV2/1 are superior to AAV2/8 for gene delivery to the nonhuman primate striatum and therefore better candidates for therapeutic applications targeting this structure.


Asunto(s)
Ganglios Basales/metabolismo , Cápside/metabolismo , Dependovirus/metabolismo , Técnicas Genéticas , Terapia Genética/métodos , Animales , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Macaca fascicularis , Masculino , Microscopía Confocal/métodos , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Plásmidos/metabolismo , Primates
19.
Curr Gene Ther ; 9(5): 375-88, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19860652

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) family of ligands (GDFLs) as well as other trophic factors have, in animal models of Parkinson's disease (PD), demonstrated the potential for excellent ameliorative properties. Clinical trials that have mechanically injected GDNF intracerebrally, while demonstrating relative safety, have been clinically disappointing to date. Likewise, recombinant adeno-associated virus (rAAV) delivered neurturin (cere-120) has also been demonstrated to be safe in humans, however clinical results have been negative. The failure of the major clinical trials has cast some doubt in the field about trophic factor delivery for the treatment of PD. In this review, we make the case that GDFLs are likely to function only when there are remaining dopamine neurons in the nigrostriatal pathway as opposed to other candidate modes of action. Thus, it is our view that utilizing earlier stage PD patients who have significant nigrostriatal dopamine innervation remaining would be more ideal to demonstrate the efficacy of GDFLs. This is particularly true when considering a novel delivery method such as gene transfer. However, if earlier stage patients are to be enrolled in GDFL gene transfer trials, then a much better safety profile must be demonstrated by preclinical experiments. One important safety advance might be the use of an external regulation system to control the expression level of the transgene. However, gene regulation systems pose unique safety issues and we will discuss these in detail. It is our view that GDFLs still remain as a promising therapeutic approach for PD.


Asunto(s)
Terapia Genética , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Enfermedad de Parkinson/terapia , Adenoviridae/genética , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos
20.
J Neurochem ; 111(2): 355-67, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19686242

RESUMEN

Viral vector-mediated gene transfer is emerging as a novel therapeutic approach with clinical utility in treatment of Parkinson's disease. Recombinant adeno-associated viral (rAAV) vector in particular has been utilized for continuous l-3,4 dihydroxyphenylalanine (DOPA) delivery by expressing the tyrosine hydroxylase (TH) and GTP cyclohydrolase 1 (GCH1) genes which are necessary and sufficient for efficient synthesis of DOPA from dietary tyrosine. The present study was designed to determine the optimal stoichiometric relationship between TH and GCH1 genes for ectopic DOPA production and the cellular machinery involved in its synthesis, storage, and metabolism. For this purpose, we injected a fixed amount of rAAV5-TH vector and increasing amounts of rAAV5-GCH1 into the striatum of rats with complete unilateral dopamine lesion. After 7 weeks the animals were killed for either biochemical or histological analysis. We show that increasing the availability of 5,6,7,8-tetrahydro-l-biopterin (BH4) in the same cellular compartment as the TH enzyme resulted in better efficiency in DOPA synthesis, most likely by hindering inactivation of the enzyme and increasing its stability. Importantly, the BH4 synthesis from ectopic GCH1 expression was saturable, yielding optimal TH enzyme functionality between GCH1 : TH ratios of 1 : 3 and 1 : 7.


Asunto(s)
Dopamina/biosíntesis , GTP Ciclohidrolasa/genética , Terapia Genética/métodos , Trastornos Parkinsonianos/terapia , Tirosina 3-Monooxigenasa/genética , Animales , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Dependovirus/genética , Dihidroxifenilalanina/metabolismo , Dopamina/metabolismo , Femenino , GTP Ciclohidrolasa/metabolismo , Inmunohistoquímica , Oxidopamina , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Ratas , Ratas Sprague-Dawley , Simpaticolíticos , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...