Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Rev Neurol (Paris) ; 180(5): 378-382, 2024 May.
Article in English | MEDLINE | ID: mdl-38580500

ABSTRACT

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.


Subject(s)
Ataxin-3 , Gene Editing , Machado-Joseph Disease , Humans , Machado-Joseph Disease/therapy , Machado-Joseph Disease/genetics , Ataxin-3/genetics , Gene Editing/methods , Genetic Therapy/methods , Animals , Repressor Proteins/genetics
2.
Gene Ther ; 24(10): 630-639, 2017 10.
Article in English | MEDLINE | ID: mdl-28771234

ABSTRACT

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.


Subject(s)
Huntington Disease/therapy , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNAi Therapeutics/methods , Animals , Dependovirus/genetics , Genetic Vectors/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Male , Microglia/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism , RNAi Therapeutics/adverse effects , Rats , Rats, Sprague-Dawley
3.
Neurobiol Dis ; 102: 105-112, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28286179

ABSTRACT

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.


Subject(s)
Hippocampus/metabolism , Huntingtin Protein/metabolism , Memory Disorders/metabolism , Neurogenesis/physiology , Protein Aggregation, Pathological/metabolism , Spatial Memory/physiology , Animals , Disease Models, Animal , Female , Genetic Vectors , Hippocampus/pathology , Humans , Huntingtin Protein/administration & dosage , Huntingtin Protein/genetics , Huntington Disease , Lentivirus , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/pathology , Mice, Inbred C57BL , Mutation , Neurons/metabolism , Neurons/pathology , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/psychology , Proto-Oncogene Proteins c-fos/metabolism
4.
Gene Ther ; 22(10): 830-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26109254

ABSTRACT

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.


Subject(s)
Astrocytes/physiology , Gene Transfer Techniques , Genetic Vectors , Lentivirus , RNA, Small Interfering , Animals , Central Nervous System/cytology , Central Nervous System/physiology , Gene Expression Regulation , Gene Silencing , Mice , Mice, Transgenic , Tetracycline
5.
Transl Psychiatry ; 3: e253, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23632457

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Neurogenesis/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/physiopathology , Astrocytes/drug effects , Astrocytes/physiology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/physiology , Depression/drug therapy , Depression/metabolism , Depression/physiopathology , Fluoxetine/pharmacology , Gene Expression/physiology , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice , Piperazines/pharmacology , Pyridines/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology
6.
Exp Neurol ; 215(1): 191-200, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19022249

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder resulting from the expansion of a glutamine repeat (polyQ) in the N-terminus of the huntingtin (htt) protein. Expression of polyQ-containing proteins has been previously shown to induce various cellular stress responses. Among these, activation of the c-Jun N-terminal kinase (JNK) cascade has been observed in cellular models of HD. However, the implication of the JNK pathway has not previously been evaluated in the striatum of HD animal models. Here we report that the JNK pathway participates in HD pathology in a rat model of the disease. Increased phosphorylation of the JNK target c-Jun was observed as early as 4 weeks and persisted for 13 weeks after lentiviral-mediated expression of htt171-82Q. In order to assess the importance of this pathway in HD pathology, JNK inhibitors including dominant-negative mutants of upstream kinases (ASK1(K709R), MEKK1(D1369A)), a c-Jun mutant (Delta169c-Jun) and the active domain of the scaffold protein JIP-1/IBI (IBI-JBD) were tested for their ability to mitigate the effect of htt171-82Q. The overexpression of MEKK1(D1369A) and JIP-1/IBI reduced the polyQ-related loss of DARPP-32 expression, while the other inhibitors had no effect. In all cases, the formation of EM48-positive htt inclusions and P-c-Jun immunoreactivity were unaltered. These results suggest that JNK activation is involved in HD and that blockade of this pathway may be of benefit in counteracting HD-related neurotoxicity.


Subject(s)
Huntington Disease/enzymology , Huntington Disease/physiopathology , MAP Kinase Kinase 4/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , CREB-Binding Protein/metabolism , Cell Line, Transformed , Disease Models, Animal , Dopamine/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Female , Gene Expression Regulation/physiology , Humans , Huntingtin Protein , Huntington Disease/genetics , Lentivirus/physiology , Mutation/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Wistar , Serine/metabolism , Time Factors , Transfection/methods
7.
J Gene Med ; 7(5): 530-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15651039

ABSTRACT

The identification of disease-causing genes in familial forms of neurodegenerative disorders and the development of genetic models closely replicating human central nervous system (CNS) pathologies have drastically changed our understanding of the molecular events leading to neuronal cell death. If these achievements open new opportunities of therapeutic interventions, including gene-based therapies, the presence of the blood-brain barrier and the post-mitotic and poor regenerative nature of the target cells constitute important challenges. Efficient delivery systems taking into account the specificity of the CNS are required to administer potential therapeutic candidates. In addition, genetic models in large animals that replicate the late stages of the diseases are in most cases not available for pre-clinical studies. The present review summarizes the potential of viral vectors as tools to create new genetic models of CNS disorders in various species including primates and the recent progress toward viral gene therapy clinical trials for the administration of therapeutic candidates into the brain.


Subject(s)
Genetic Therapy , Genetic Vectors , Models, Biological , Neurodegenerative Diseases/therapy , Viruses/genetics , Blood-Brain Barrier/drug effects , Humans
8.
Hum Gene Ther ; 15(10): 968-75, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15585112

ABSTRACT

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.


Subject(s)
Genetic Therapy/methods , Huntington Disease/genetics , Huntington Disease/therapy , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Cell Line , Cell Survival , Ciliary Neurotrophic Factor/chemistry , Ciliary Neurotrophic Factor/genetics , Codon , Cricetinae , Electrophysiology , Female , Gene Transfer Techniques , Humans , Male , Neurons/metabolism , Polymers/chemistry , Retroviridae/genetics , Time Factors
9.
Neurobiol Dis ; 17(2): 283-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15474365

ABSTRACT

Viral delivery of glial cell line-derived neurotrophic factor (GDNF) currently represents one of the most promising neuroprotective strategies for Parkinson's Disease (PD). However, the effect of this neurotrophic factor has never been tested in the newly available genetic models of PD based on the viral expression of mutated alpha-synuclein. In this study, we evaluated the ability of lentiviral vectors coding for GDNF (lenti-GDNF) to prevent nigral dopaminergic degeneration associated with the lentiviral mediated expression of the A30P mutant human alpha-synuclein (lenti-A30P). This virally based rat model develops a progressive and selective loss of dopamine neurons associated with the appearance of alpha-synuclein containing inclusions, thus recapitulating the major hallmarks of PD. Lenti-GDNF was injected in the substantia nigra 2 weeks before nigral administration of lenti-A30P. Although a robust expression of GDNF was observed in the whole nigrostriatal pathway due to retrograde and/or anterograde transport, lenti-GDNF did not prevent the alpha-synuclein-induced dopaminergic neurodegeneration in the lentiviral-based genetic rat model of PD. These results suggest that sustained GDNF treatment cannot modulate the cellular toxicity related to abnormal folded protein accumulation as mutated human alpha-synuclein.


Subject(s)
Gene Transfer Techniques , Lentivirus/genetics , Nerve Degeneration/etiology , Nerve Degeneration/prevention & control , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease/complications , Animals , Female , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor , Humans , Immunohistochemistry , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Parkinson Disease/etiology , Parkinson Disease/pathology , Rats , Rats, Wistar , Synucleins , alpha-Synuclein
10.
Hum Gene Ther ; 15(7): 669-80, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15242527

ABSTRACT

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.


Subject(s)
Cell Transplantation/methods , Erythropoietin/genetics , Fibroblasts/transplantation , Animals , Cell Survival , Erythropoietin/analysis , Erythropoietin/biosynthesis , Fibroblasts/cytology , Fibroblasts/metabolism , Hematocrit , Humans , Mice , Mice, Inbred DBA , Polymers/chemistry , Sulfones/chemistry , Transplantation, Heterologous
11.
Gene Ther ; 10(9): 818-21, 2003 May.
Article in English | MEDLINE | ID: mdl-12704422

ABSTRACT

For most retinal degeneration disorders, no efficient treatment exists to preserve photoreceptors (PRs) and, consequently, to maintain vision. Gene transfer appears to be a promising approach to prevent PR loss. In order to design adequate vectors to target specific retinal cell types, we have analyzed the expression pattern of three different promoters (mouse phosphoglycerate kinase 1 (PGK), elongation factor-1 (EFS), rhodopsin (Rho)) in newborn and adult DBA/2 mice retinas using self-inactivating lentiviral vectors. At 7 days after intraocular injection and in optimal conditions, cell transduction was observed up to 1.5 mm from the injection site. PGK promoter expression was predominant in the retinal pigment epithelium (RPE), especially in adult mice, whereas the EFS promoter allowed a broad expression in the retina. Finally, as expected, the Rho promoter was specifically expressed in PRs. Differences in the cell types transduced and in transduction efficiency were observed between newborn and adult injected eyes emphasizing the importance of such basic studies for further gene therapy approaches as well as for understanding the transcriptional changes during retinal maturation. Thus, for future attempts to slow or rescue retinal degeneration by lentiviral delivery, PGK and EFS are more suitable to control the expression of a supporting secreted factor, PGK being mainly expressed in RPE and EFS in different cell types throughout the entire retina, whereas Rho should allow to specifically deliver the therapeutic gene to PRs.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Lentivirus/genetics , Promoter Regions, Genetic/genetics , Retinal Degeneration/therapy , Animals , Animals, Newborn , Gene Expression , Genetic Vectors/genetics , Lentivirus/physiology , Mice , Mice, Inbred DBA , Microscopy, Fluorescence , Peptide Elongation Factor 1/genetics , Phosphoglycerate Kinase/genetics , Retina/metabolism , Rhodopsin/genetics , Virus Inactivation
12.
J Gene Med ; 5(3): 246-57, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12666190

ABSTRACT

BACKGROUND: The transplantation of encapsulated cells genetically engineered to secrete human erythropoietin (hEpo) represents an alternative to repeated injections of the recombinant hormone for the treatment of Epo-responsive anemia. In the present study, the ability of primary human foreskin fibroblasts to secrete high levels of hEpo and the importance of cis-acting elements and infection conditions on transgene expression level were assessed. METHODS: The transduction efficiency was first evaluated with beta-galactosidase (LacZ)-encoding retroviral vectors derived from the murine leukemia retrovirus (MLV) pseudotyped either with an amphotropic envelope or with the G glycoprotein of vesicular stomatitis virus (VSV-G). Human fibroblasts were then infected with an amphotropic hEpo-expressing retroviral vector, which was modified by insertion of a post-transcriptional regulatory element from the woodchuck hepatitis virus (WPRE) and a Kozak consensus sequence (KZ). Human Epo production was further optimized by increasing the multiplicity of infection and by selecting high producer cells. The survival and the transgene expression of these fibroblasts were finally evaluated in vivo. The cells were encapsulated into microporous hollow fibers and subcutaneously implanted in nude mice. RESULTS: A secretion level of approximately 5 IU hEpo/10(6) cells/day was obtained with the basal vector. A 7.5-fold increase in transgene expression was observed with the insertion of WPRE and KZ elements. Finally, according to the optimization of infection conditions, we obtained a 40-fold increase in hEpo secretion, reaching approximately 200 IU hEpo/10(6) cells/day. The in vivo experiments showed an increase in the hematocrit during the first 2 weeks and elevated levels exceeding 60% were maintained over a 6-week period. CONCLUSIONS: These results indicate that primary human fibroblasts represent a promising source for encapsulated cell therapy.


Subject(s)
Erythropoietin/genetics , Gene Transfer Techniques , Genetic Vectors , Leukemia Virus, Murine , Animals , Erythropoietin/metabolism , Fibroblasts/metabolism , Fibroblasts/transplantation , Genes, Regulator , Genes, Reporter , Humans , Mice , Mice, Nude , Transduction, Genetic
13.
Hum Gene Ther ; 13(16): 1981-90, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12427308

ABSTRACT

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.


Subject(s)
Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/pharmacology , Genetic Vectors , Huntington Disease/prevention & control , Huntington Disease/therapy , Lentivirus/genetics , Quinolinic Acid/pharmacology , Tetracycline/pharmacology , Animals , Brain/pathology , Choline O-Acetyltransferase/metabolism , DNA, Complementary/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Recombinant Fusion Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 99(16): 10813-8, 2002 Aug 06.
Article in English | MEDLINE | ID: mdl-12122208

ABSTRACT

Parkinson's disease (PD) is characterized by the progressive loss of substantia nigra dopaminergic neurons and the presence of cytoplasmic inclusions named Lewy bodies. Two missense mutations of the alpha-synuclein (alpha-syn; A30P and A53T) have been described in several families with an autosomal dominant form of PD. alpha-Syn also constitutes one of the main components of Lewy bodies in sporadic cases of PD. To develop an animal model of PD, lentiviral vectors expressing different human or rat forms of alpha-syn were injected into the substantia nigra of rats. In contrast to transgenic mice models, a selective loss of nigral dopaminergic neurons associated with a dopaminergic denervation of the striatum was observed in animals expressing either wild-type or mutant forms of human alpha-syn. This neuronal degeneration correlates with the appearance of abundant alpha-syn-positive inclusions and extensive neuritic pathology detected with both alpha-syn and silver staining. Lentiviral-mediated expression of wild-type or mutated forms of human alpha-syn recapitulates the essential neuropathological features of PD. Rat alpha-syn similarly leads to protein aggregation but without cell loss, suggesting that inclusions are not the primary cause of cell degeneration in PD. Viral-mediated genetic models may contribute to elucidate the mechanism of alpha-syn-induced cell death and allow the screening of candidate therapeutic molecules.


Subject(s)
Dopamine , Nerve Degeneration/pathology , Nerve Tissue Proteins/physiology , Neurons/pathology , Parkinson Disease/pathology , Animals , Animals, Genetically Modified , Biomarkers , Brain/metabolism , Brain/pathology , Disease Models, Animal , Gene Expression , Genetic Vectors , HIV-1 , Humans , Lentivirus , Lewy Bodies/metabolism , Lewy Bodies/pathology , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Rats , Substantia Nigra/metabolism , Synucleins , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein
16.
Neurobiol Dis ; 8(3): 433-46, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11442352

ABSTRACT

Neurodegenerative diseases represent promising targets for gene therapy approaches provided effective transfer vectors. In the present study, we evaluated the effectiveness of LacZ-expressing lentiviral vectors with two different internal promoters, the mouse phosphoglycerate kinase 1 (PGK) and cytomegalovirus (CMV), to infect striatal cells. The intrastriatal injection of lenti-beta-Gal vectors lead to 207, 400 +/- 11,500 and 303,100 +/- 4,300 infected cells in adult rats, respectively. Importantly, the beta-galactosidase activity was higher in striatal extracts from PGK-LacZ-injected animals as compared to CMV-LacZ animals. The efficacy of the system was further examined with a potential therapeutic gene for the treatment of Huntington's disease, the human ciliary neurotrophic factor (CNTF). PGK-LacZ- or PGK-CNTF-expressing viruses were stereotaxically injected into the striatum of rats, 3 weeks later the animals were unilaterally lesioned with 180 nmol of quinolinic acid (QA). Control animals displayed 148 +/- 43 apomorphine-induced rotations ipsilateral to the lesion 5 days postlesion as compared to 26 +/- 22 turns/45 min in the CNTF-treated group. The extent of the striatal damage was significantly diminished in the CNTF-treated rats as indicated by the 52 +/- 9.7% decrease of the lesion volume and the sparing of DARPP-32, ChAT and NADPH-d neuronal populations. These results further establish that lentiviruses may represent an efficient gene delivery system for the screening of therapeutic molecules in Huntington's disease.


Subject(s)
Ciliary Neurotrophic Factor/genetics , Genetic Therapy/methods , Genetic Vectors , Huntington Disease/therapy , Lentivirus/genetics , Animals , Cytomegalovirus/genetics , Disease Models, Animal , Female , Gene Expression , Huntington Disease/chemically induced , Neuroprotective Agents , Phosphoglycerate Kinase/genetics , Promoter Regions, Genetic , Quinolinic Acid , Rats , Rats, Wistar , beta-Galactosidase/genetics
17.
Exp Neurol ; 170(1): 48-62, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11421583

ABSTRACT

Multipotent precursors able to generate neurons, astrocytes, and oligodendrocytes have previously been isolated from human brain embryos and recently from neurogenic regions of the adult human brains. The isolation of multipotent neural precursors from adult human should open new perspectives to study adult neurogenesis and for brain repair. The present study describes the in vitro isolation from adult human brains of a progenitor responsive to both epidermal and basic fibroblast growth factors that forms spheres as it proliferates. Single spheres derived from various regions of the brain generate in vitro neurons, astrocytes, and oligodendrocytes. The clonal origin of the spheres was revealed by genomic viral insertion using lentiviral vector. Interestingly, this vector appears to be a potent tool for gene transfer into human neural progeny. Ninety-six percent of the spheres investigated were multipotent. Multipotent precursors were isolated from all brain regions studied, including the temporal and the frontal cortex, the amygdala, the hippocampus, and the ventricular zone. This study is the first evidence that primitive precursors such as multipotent precursors exist in the adult human cortex and can reside far from the ventricles. Neurogenesis derived from adult human progenitors differ to murine neurogenesis by the requirement of laminin for oligodendrocyte generation and by the action of basic-fibroblast growth factor and platelet derived growth factor that prevented the formation of oligodendrocytes and neurons. Moreover, the differentiation of human adult precursors seems to differ from fetal ones: adult precursors do not necessitate the removal of mitogen for differentiation. These results indicate that the study of adult multipotent precursors is a new platform to study adult human neurogenesis, potentially generate neural cells for transplantation, and design protocols for in vivo stimulation.


Subject(s)
Brain/cytology , Neurons/cytology , Stem Cells/cytology , Adolescent , Adult , Amygdala/cytology , Amygdala/drug effects , Antigens, Differentiation/biosynthesis , Brain/drug effects , Cell Differentiation , Cell Division/drug effects , Cell Separation , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Child , Child, Preschool , Clone Cells/cytology , Epidermal Growth Factor/pharmacology , Extrachromosomal Inheritance , Fibroblast Growth Factor 2/pharmacology , Humans , Infant , Middle Aged , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Stem Cells/drug effects
18.
Proc Natl Acad Sci U S A ; 98(13): 7611-6, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11404469

ABSTRACT

Adenosine is an inhibitor of neuronal activity in the brain. The local release of adenosine from grafted cells was evaluated as an ex vivo gene therapy approach to suppress synchronous discharges and epileptic seizures. Fibroblasts were engineered to release adenosine by inactivating the adenosine-metabolizing enzymes adenosine kinase and adenosine deaminase. After encapsulation into semipermeable polymers, the cells were grafted into the brain ventricles of electrically kindled rats, a model of partial epilepsy. Grafted rats provided a nearly complete protection from behavioral seizures and a near-complete suppression of afterdischarges in electroencephalogram recordings, whereas the full tonic-clonic convulsions in control rats remained unaltered. Thus, the local release of adenosine resulting in adenosine concentrations <25 nM at the site of action is sufficient to suppress seizure activity and, therefore, provides a potential therapeutic principle for the treatment of drug-resistant partial epilepsies.


Subject(s)
Adenosine Deaminase/metabolism , Adenosine Kinase/metabolism , Adenosine/physiology , Brain/metabolism , Cell Transplantation , Kindling, Neurologic/physiology , Seizures/therapy , Adenosine/metabolism , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Adenosine Kinase/genetics , Aggression , Animals , Brain/cytology , Cell Line , Cricetinae , Epilepsies, Partial/therapy , Exploratory Behavior , Fibroblasts/cytology , Fibroblasts/physiology , Genetic Therapy , Kindling, Neurologic/drug effects , Male , Mice , Mice, Knockout , Motor Activity , Phenytoin/pharmacology , Rats , Rats, Inbred Strains , Seizures/physiopathology , Social Behavior , Xanthines/pharmacology
19.
Mol Cell Neurosci ; 17(5): 855-71, 2001 May.
Article in English | MEDLINE | ID: mdl-11358483

ABSTRACT

Viral vectors are useful for transferring genes into neurons. Here, we characterized recombinant Semliki Forest virus (SFV), adenovirus type 5 (Ad5), adeno-associated virus type 2 (AAV), lentivirus, and measles virus (MV) by their expression of green fluorescent protein (GFP) in rat hippocampal slice cultures. SFV infected more neurons (>90% of all GFP-positive cells) than AAV, lentivirus, and MV (71, 69, and 62%, respectively), whereas no infected neurons were identified with Ad5. AAV-mediated GFP expression was neuron-specific when the platelet-derived growth factor beta-chain promoter rather than cytomegalovirus promoter was used. Transgene expression occurred rapidly but transiently for SFV, increased slowly but remained stable with AAV and lentivirus, and was fast with MV. Resting membrane potential and conductance, action potentials, firing accommodation, and H-current appeared normal in infected CA1 pyramidal cells. Thus, SFV is useful for short-term and AAV and lentivirus for long-term transduction of hippocampal slices, while MV constitutes a novel vector.


Subject(s)
Gene Expression Regulation, Viral/physiology , Gene Transfer Techniques , Genetic Vectors/physiology , Neurons/virology , Transduction, Genetic/methods , Transgenes/physiology , Viruses/genetics , Adenoviridae/genetics , Adenoviridae/pathogenicity , Animals , Cell Survival/genetics , Dependovirus/genetics , Dependovirus/pathogenicity , Green Fluorescent Proteins , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/virology , Indicators and Reagents/metabolism , Lentivirus/genetics , Lentivirus/pathogenicity , Luminescent Proteins/metabolism , Measles virus/genetics , Measles virus/pathogenicity , Membrane Potentials/genetics , Neurons/cytology , Neurons/metabolism , Organ Culture Techniques , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Pyramidal Cells/virology , Rats , Semliki forest virus/genetics , Semliki forest virus/pathogenicity , Time Factors , Virulence/genetics , Viruses/pathogenicity
20.
Eur J Neurosci ; 14(11): 1753-61, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11860469

ABSTRACT

Ciliary neurotrophic factor prevents behavioural deficits and striatal degeneration in rat and primate models of Huntington's disease. Interleukin-6, another member of the cytokine family, and the chimeric molecule (IL6/IL6R) in which interleukin-6 and its soluble receptor are fused, have been shown to exert trophic action on various neuronal populations in the central nervous system. Therefore, we investigated the neuroprotective effect of these two molecules in the quinolinic acid model of Huntington's disease. LacZ-, interleukin-6- and IL6/IL6R-expressing lentiviral vectors were stereotaxically injected into the striatum of Wistar rats. Three weeks later the animals were lesioned through the intrastriatal injection of 180 nmol of quinolinic acid. The extent of the striatal damage was significantly diminished in the rats that had been treated with interleukin-6 or IL6/IL6R. The neuroprotective effect was, however, more pronounced with the IL6/IL6R chimera than with interleukin-6 as indicated by the volume of the lesions (38.6 +/- 10% in the IL6/IL6R group, 63.3 +/- 3.6% in the IL-6 group and 84.3 +/-2.9% in the control group). Quantitative analysis of striatal interneurons further demonstrated that the IL6/IL6R chimera is more neuroprotective than IL-6 on ChAT- and NADPH-d-immunoreactive neurons. These results suggest that the IL6/IL6R chimera is a potential treatment for Huntington's disease.


Subject(s)
Huntington Disease/drug therapy , Interleukin-6/pharmacology , Neostriatum/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Interleukin-6/genetics , Recombinant Fusion Proteins/pharmacology , Acetylcholine/metabolism , Animals , Disease Models, Animal , Female , Genetic Vectors , Huntington Disease/chemically induced , Huntington Disease/physiopathology , Immunohistochemistry , Interleukin-6/genetics , Interleukin-6/metabolism , Neostriatum/metabolism , Neostriatum/physiopathology , Neurons/metabolism , Quinolinic Acid/pharmacology , Rats , Rats, Wistar , Receptors, Interleukin-6/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...