Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 181: 106116, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37054900

ABSTRACT

Tauopathy is a typical feature of Alzheimer's disease of major importance because it strongly correlates with the severity of cognitive deficits experienced by patients. During the pathology, it follows a characteristic spatiotemporal course which takes its origin in the transentorhinal cortex, and then gradually invades the entire forebrain. To study the mechanisms of tauopathy, and test new therapeutic strategies, it is necessary to set-up relevant and versatile in vivo models allowing to recapitulate tauopathy. With this in mind, we have developed a model of tauopathy by overexpression of the human wild-type Tau protein in retinal ganglion cells in mice (RGCs). This overexpression led to the presence of hyperphosphorylated forms of the protein in the transduced cells as well as to their progressive degeneration. The application of this model to mice deficient in TREM2 (Triggering Receptor Expressed on Myeloid cells-2, an important genetic risk factor for AD) as well as to 15-month-old mice showed that microglia actively participate in the degeneration of RGCs. Surprisingly, although we were able to detect the transgenic Tau protein up to the terminal arborization of RGCs at the level of the superior colliculi, spreading of the transgenic Tau protein to post-synaptic neurons was detected only in aged animals. This suggests that there may be neuron-intrinsic- or microenvironment mediators facilitating this spreading that appear with aging.


Subject(s)
Alzheimer Disease , Tauopathies , Animals , Humans , Mice , Alzheimer Disease/metabolism , Disease Models, Animal , Membrane Glycoproteins/metabolism , Mice, Transgenic , Microglia/metabolism , Receptors, Immunologic/metabolism , Retinal Ganglion Cells/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Tauopathies/pathology , Visual Pathways/metabolism
2.
Brain ; 146(1): 149-166, 2023 01 05.
Article in English | MEDLINE | ID: mdl-35298632

ABSTRACT

Huntington's disease is a fatal neurodegenerative disease characterized by striatal neurodegeneration, aggregation of mutant Huntingtin and the presence of reactive astrocytes. Astrocytes are important partners for neurons and engage in a specific reactive response in Huntington's disease that involves morphological, molecular and functional changes. How reactive astrocytes contribute to Huntington's disease is still an open question, especially because their reactive state is poorly reproduced in experimental mouse models. Here, we show that the JAK2-STAT3 pathway, a central cascade controlling astrocyte reactive response, is activated in the putamen of Huntington's disease patients. Selective activation of this cascade in astrocytes through viral gene transfer reduces the number and size of mutant Huntingtin aggregates in neurons and improves neuronal defects in two complementary mouse models of Huntington's disease. It also reduces striatal atrophy and increases glutamate levels, two central clinical outcomes measured by non-invasive magnetic resonance imaging. Moreover, astrocyte-specific transcriptomic analysis shows that activation of the JAK2-STAT3 pathway in astrocytes coordinates a transcriptional program that increases their intrinsic proteolytic capacity, through the lysosomal and ubiquitin-proteasome degradation systems. This pathway also enhances their production and exosomal release of the co-chaperone DNAJB1, which contributes to mutant Huntingtin clearance in neurons. Together, our results show that the JAK2-STAT3 pathway controls a beneficial proteostasis response in reactive astrocytes in Huntington's disease, which involves bi-directional signalling with neurons to reduce mutant Huntingtin aggregation, eventually improving disease outcomes.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Animals , Mice , Huntington Disease/genetics , Astrocytes/metabolism , Proteostasis , Neurodegenerative Diseases/pathology , Neurons/metabolism , Huntingtin Protein/genetics , Huntingtin Protein/metabolism
3.
Neurobiol Dis ; 155: 105398, 2021 07.
Article in English | MEDLINE | ID: mdl-34019997

ABSTRACT

The role played by microglia has taken the center of the stage in the etiology of Alzheimer's disease (AD). Several genome-wide association studies carried out on large cohorts of patients have indeed revealed a large number of genetic susceptibility factors corresponding to genes involved in neuroinflammation and expressed specifically by microglia in the brain. Among these genes TREM2, a cell surface receptor expressed by microglia, arouses strong interest because its R47H variant confers a risk of developing AD comparable to the ε4 allele of the APOE gene. Since this discovery, a growing number of studies have therefore examined the role played by TREM2 in the evolution of amyloid plaques and neurofibrillary tangles, the two brain lesions characteristic of AD. Many studies report conflicting results, reflecting the complex nature of microglial activation in AD. Here, we investigated the impact of TREM2 deficiency in the THY-Tau22 transgenic line, a well-characterized model of tauopathy. Our study reports an increase in the severity of tauopathy lesions in mice deficient in TREM2 occurring at an advanced stage of the pathology. This exacerbation of pathology was associated with a reduction in microglial activation indicated by typical morphological features and altered expression of specific markers. However, it was not accompanied by any further changes in memory performance. Our longitudinal study confirms that a defect in microglial TREM2 signaling leads to an increase in neuronal tauopathy occurring only at late stages of the disease.


Subject(s)
Disease Models, Animal , Membrane Glycoproteins/deficiency , Microglia/metabolism , Receptors, Immunologic/deficiency , Tauopathies/metabolism , Thy-1 Antigens/genetics , tau Proteins/genetics , Animals , Female , Humans , Longitudinal Studies , Male , Maze Learning/physiology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Receptors, Immunologic/genetics , Tauopathies/genetics , Tauopathies/pathology
4.
J Cereb Blood Flow Metab ; 35(6): 917-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25833344

ABSTRACT

Ciliary neurotrophic factor (CNTF) is neuroprotective against multiple pathologic conditions including metabolic impairment, but the mechanisms are still unclear. To delineate CNTF effects on brain energy homeostasis, we performed a multimodal imaging study, combining in vivo proton magnetic resonance spectroscopy, high-performance liquid chromatography analysis, and in situ glutamate imaging by chemical exchange saturation transfer. Unexpectedly, we found that CNTF expression through lentiviral gene transfer in the rat striatum significantly decreased the levels of neuronal metabolites (N-acetyl-aspartate, N-acetyl-aspartyl-glutamate, and glutamate). This preclinical study shows that CNTF remodels brain metabolism, and suggests that decreased levels of neuronal metabolites may occur in the absence of neuronal dysfunction.


Subject(s)
Astrocytes/metabolism , Ciliary Neurotrophic Factor/metabolism , Corpus Striatum/metabolism , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Glutamic Acid/metabolism , Humans , Magnetic Resonance Imaging , Male , Neuroprotective Agents/metabolism , Rats, Sprague-Dawley
5.
J Neurosci ; 35(6): 2817-29, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673868

ABSTRACT

Astrocyte reactivity is a hallmark of neurodegenerative diseases (ND), but its effects on disease outcomes remain highly debated. Elucidation of the signaling cascades inducing reactivity in astrocytes during ND would help characterize the function of these cells and identify novel molecular targets to modulate disease progression. The Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway is associated with reactive astrocytes in models of acute injury, but it is unknown whether this pathway is directly responsible for astrocyte reactivity in progressive pathological conditions such as ND. In this study, we examined whether the JAK/STAT3 pathway promotes astrocyte reactivity in several animal models of ND. The JAK/STAT3 pathway was activated in reactive astrocytes in two transgenic mouse models of Alzheimer's disease and in a mouse and a nonhuman primate lentiviral vector-based model of Huntington's disease (HD). To determine whether this cascade was instrumental for astrocyte reactivity, we used a lentiviral vector that specifically targets astrocytes in vivo to overexpress the endogenous inhibitor of the JAK/STAT3 pathway [suppressor of cytokine signaling 3 (SOCS3)]. SOCS3 significantly inhibited this pathway in astrocytes, prevented astrocyte reactivity, and decreased microglial activation in models of both diseases. Inhibition of the JAK/STAT3 pathway within reactive astrocytes also increased the number of huntingtin aggregates, a neuropathological hallmark of HD, but did not influence neuronal death. Our data demonstrate that the JAK/STAT3 pathway is a common mediator of astrocyte reactivity that is highly conserved between disease states, species, and brain regions. This universal signaling cascade represents a potent target to study the role of reactive astrocytes in ND.


Subject(s)
Alzheimer Disease/physiopathology , Astrocytes , Huntington Disease/physiopathology , Janus Kinases , STAT3 Transcription Factor , Signal Transduction , Alzheimer Disease/pathology , Animals , Electron Transport Complex IV/metabolism , Humans , Huntington Disease/pathology , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
6.
PLoS One ; 9(6): e100760, 2014.
Article in English | MEDLINE | ID: mdl-24971751

ABSTRACT

Tau is a microtubule-associated protein that aggregates in neurodegenerative disorders known as tauopathies. Recently, studies have suggested that Tau may be secreted and play a role in neural network signalling. However, once deregulated, secreted Tau may also participate in the spreading of Tau pathology in hierarchical pathways of neurodegeneration. The mechanisms underlying neuron-to-neuron Tau transfer are still unknown; given the known role of extra-cellular vesicles in cell-to-cell communication, we wondered whether these vesicles could carry secreted Tau. We found, among vesicles, that Tau is predominately secreted in ectosomes, which are plasma membrane-originating vesicles, and when it accumulates, the exosomal pathway is activated.


Subject(s)
Cell-Derived Microparticles/metabolism , tau Proteins/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Exosomes/metabolism , Extracellular Fluid/metabolism , Humans , Microscopy, Electron , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar
7.
PLoS One ; 9(6): e99341, 2014.
Article in English | MEDLINE | ID: mdl-24926995

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder resulting from polyglutamine expansion in the huntingtin (HTT) protein and for which there is no cure. Although suppression of both wild type and mutant HTT expression by RNA interference is a promising therapeutic strategy, a selective silencing of mutant HTT represents the safest approach preserving WT HTT expression and functions. We developed small hairpin RNAs (shRNAs) targeting single nucleotide polymorphisms (SNP) present in the HTT gene to selectively target the disease HTT isoform. Most of these shRNAs silenced, efficiently and selectively, mutant HTT in vitro. Lentiviral-mediated infection with the shRNAs led to selective degradation of mutant HTT mRNA and prevented the apparition of neuropathology in HD rat's striatum expressing mutant HTT containing the various SNPs. In transgenic BACHD mice, the mutant HTT allele was also silenced by this approach, further demonstrating the potential for allele-specific silencing. Finally, the allele-specific silencing of mutant HTT in human embryonic stem cells was accompanied by functional recovery of the vesicular transport of BDNF along microtubules. These findings provide evidence of the therapeutic potential of allele-specific RNA interference for HD.


Subject(s)
Brain/cytology , Genetic Therapy/methods , Huntington Disease/therapy , Mutant Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , RNA, Small Interfering/genetics , Animals , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/cytology , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/genetics , In Vitro Techniques , Male , Mice , Mutant Proteins/genetics , Polymorphism, Single Nucleotide , RNA Isoforms/metabolism , RNA Stability , Rats , Rats, Wistar
8.
Acta Neuropathol Commun ; 2: 14, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24479894

ABSTRACT

BACKGROUND: In sporadic Tauopathies, neurofibrillary degeneration (NFD) is characterised by the intraneuronal aggregation of wild-type Tau proteins. In the human brain, the hierarchical pathways of this neurodegeneration have been well established in Alzheimer's disease (AD) and other sporadic tauopathies such as argyrophilic grain disorder and progressive supranuclear palsy but the molecular and cellular mechanisms supporting this progression are yet not known. These pathways appear to be associated with the intercellular transmission of pathology, as recently suggested in Tau transgenic mice. However, these conclusions remain ill-defined due to a lack of toxicity data and difficulties associated with the use of mutant Tau. RESULTS: Using a lentiviral-mediated rat model of hippocampal NFD, we demonstrated that wild-type human Tau protein is axonally transferred from ventral hippocampus neurons to connected secondary neurons even at distant brain areas such as olfactory and limbic systems indicating a trans-synaptic protein transfer. Using different immunological tools to follow phospho-Tau species, it was clear that Tau pathology generated using mutated Tau remains near the IS whereas it spreads much further using the wild-type one. CONCLUSION: Taken together, these results support a novel mechanism for Tau protein transfer compared to previous reports based on transgenic models with mutant cDNA. It also demonstrates that mutant Tau proteins are not suitable for the development of experimental models helpful to validate therapeutic intervention interfering with Tau spreading.


Subject(s)
Neurons/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Animals , Brain/pathology , Cell Differentiation/genetics , Cells, Cultured , Disease Models, Animal , Disease Progression , Embryo, Mammalian , Gene Transfer Techniques , Humans , Microfluidic Analytical Techniques , Microscopy, Confocal , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Protein Transport/physiology , RNA, Messenger/metabolism , Rats , Rats, Wistar
9.
Mol Ther ; 21(7): 1358-68, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23609018

ABSTRACT

Most models for tauopathy use a mutated form of the Tau gene, MAPT, that is found in frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17) and that leads to rapid neurofibrillary degeneration (NFD). Use of a wild-type (WT) form of human Tau protein to model the aggregation and associated neurodegenerative processes of Tau in the mouse brain has thus far been unsuccessful. In the present study, we generated an original "sporadic tauopathy-like" model in the rat hippocampus, encoding six Tau isoforms as found in humans, using lentiviral vectors (LVs) for the delivery of a human WT Tau. The overexpression of human WT Tau in pyramidal neurons resulted in NFD, the morphological characteristics and kinetics of which reflected the slow and sporadic neurodegenerative processes observed in sporadic tauopathies, unlike the rapid neurodegenerative processes leading to cell death and ghost tangles triggered by the FTDP-17 mutant Tau P301L. This new model highlights differences in the molecular and cellular mechanisms underlying the pathological processes induced by WT and mutant Tau and suggests that preference should be given to animal models using WT Tau in the quest to understand sporadic tauopathies.


Subject(s)
Brain/metabolism , Brain/pathology , Lentivirus/genetics , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Rats , Rats, Wistar , Tauopathies/genetics , tau Proteins/genetics
10.
Hum Gene Ther Methods ; 23(4): 242-54, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22934828

ABSTRACT

Tissue-targeted expression is of major interest for studying the contribution of cellular subpopulations to neurodegenerative diseases. However, in vivo methods to investigate this issue are limited. Here, we report an analysis of the cell specificity of expression of fluorescent reporter genes driven by six neuronal promoters, with the ubiquitous phosphoglycerate kinase 1 (PGK) promoter used as a reference. Quantitative analysis of AcGFPnuc expression in the striatum and hippocampus of rodents showed that all lentiviral vectors (LV) exhibited a neuronal tropism; however, there was substantial diversity of transcriptional activity and cell-type specificity of expression. The promoters with the highest activity were those of the 67 kDa glutamic acid decarboxylase (GAD67), homeobox Dlx5/6, glutamate receptor 1 (GluR1), and preprotachykinin 1 (Tac1) genes. Neuron-specific enolase (NSE) and dopaminergic receptor 1 (Drd1a) promoters showed weak activity, but the integration of an amplification system into the LV overcame this limitation. In the striatum, the expression profiles of Tac1 and Drd1a were not limited to the striatonigral pathway, whereas in the hippocampus, Drd1a and Dlx5/6 showed the expected restricted pattern of expression. Regulation of the Dlx5/6 promoter was observed in a disease condition, whereas Tac1 activity was unaffected. These vectors provide safe tools that are more selective than others available, for the administration of therapeutic molecules in the central nervous system (CNS). Nevertheless, additional characterization of regulatory elements in neuronal promoters is still required.


Subject(s)
Brain/metabolism , Promoter Regions, Genetic/genetics , Animals , Brain/pathology , Genes, Reporter , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lentivirus/genetics , Male , Mice , Neurons/metabolism , Neurons/pathology , Phosphoglycerate Kinase/genetics , Phosphoglycerate Kinase/metabolism , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Rats , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Tachykinins/genetics , Tachykinins/metabolism , Transcription, Genetic , Transduction, Genetic
11.
J Neurosci ; 31(20): 7392-401, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21593323

ABSTRACT

Astrocytes support neuronal antioxidant capacity by releasing glutathione, which is cleaved to cysteine in brain extracellular space. Free cysteine is then taken up by neurons through excitatory amino acid transporter 3 [EAAT3; also termed Slc1a1 (solute carrier family 1 member 1)] to support de novo glutathione synthesis. Activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) pathway by oxidative stress promotes astrocyte release of glutathione, but it remains unknown how this release is coupled to neuronal glutathione synthesis. Here we evaluated transcriptional regulation of the neuronal cysteine transporter EAAT3 by the Nrf2-ARE pathway. Nrf2 activators and Nrf2 overexpression both produced EAAT3 transcriptional activation in C6 cells. A conserved ARE-related sequence was found in the EAAT3 promoter of several mammalian species. This ARE-related sequence was bound by Nrf2 in mouse neurons in vivo as observed by chromatin immunoprecipitation. Chemical activation of the Nrf2-ARE pathway in mouse brain increased both neuronal EAAT3 levels and neuronal glutathione content, and these effects were abrogated in mice genetically deficient in either Nrf2 or EAAT3. Selective overexpression of Nrf2 in brain neurons by lentiviral gene transfer was sufficient to upregulate both neuronal EAAT3 protein and glutathione content. These findings identify a mechanism whereby Nrf2 activation can coordinate astrocyte glutathione release with neuronal glutathione synthesis through transcriptional upregulation of neuronal EAAT3 expression.


Subject(s)
Excitatory Amino Acid Transporter 3/biosynthesis , Glutathione/biosynthesis , NF-E2-Related Factor 2/physiology , Neurons/metabolism , Up-Regulation/physiology , Animals , Cell Line, Tumor , Excitatory Amino Acid Transporter 3/deficiency , Excitatory Amino Acid Transporter 3/genetics , Male , Marmota , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Rats , Up-Regulation/genetics
12.
Brain ; 134(Pt 5): 1400-15, 2011 May.
Article in English | MEDLINE | ID: mdl-21478185

ABSTRACT

Machado-Joseph disease, also known as spinocerebellar ataxia type 3, is the most common of the dominantly inherited ataxias worldwide and is characterized by mutant ataxin-3 misfolding, intracellular accumulation of aggregates and neuronal degeneration. Here we investigated the implication of autophagy, the major pathway for organelle and protein turnover, in the accumulation of mutant ataxin-3 aggregates and neurodegeneration found in Machado-Joseph disease and we assessed whether specific stimulation of this pathway could mitigate the disease. Using tissue from patients with Machado-Joseph disease, transgenic mice and a lentiviral-based rat model, we found an abnormal expression of endogenous autophagic markers, accumulation of autophagosomes and decreased levels of beclin-1, a crucial protein in the early nucleation step of autophagy. Lentiviral vector-mediated overexpression of beclin-1 led to stimulation of autophagic flux, mutant ataxin-3 clearance and overall neuroprotective effects in neuronal cultures and in a lentiviral-based rat model of Machado-Joseph disease. These data demonstrate that autophagy is a key degradation pathway, with beclin-1 playing a significant role in alleviating Machado-Joseph disease pathogenesis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Machado-Joseph Disease/genetics , Membrane Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Aged , Animals , Apoptosis Regulatory Proteins/genetics , Ataxin-3 , Autophagy-Related Proteins , Beclin-1 , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Machado-Joseph Disease/pathology , Machado-Joseph Disease/physiopathology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Rats , Rats, Wistar , Sequestosome-1 Protein , Transfection/methods , Trinucleotide Repeat Expansion/genetics
13.
Hum Mol Genet ; 19(15): 3053-67, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20494921

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder previously thought to be of primary neuronal origin, despite ubiquitous expression of mutant huntingtin (mHtt). We tested the hypothesis that mHtt expressed in astrocytes may contribute to the pathogenesis of HD. To better understand the contribution of astrocytes in HD in vivo, we developed a novel mouse model using lentiviral vectors that results in selective expression of mHtt into striatal astrocytes. Astrocytes expressing mHtt developed a progressive phenotype of reactive astrocytes that was characterized by a marked decreased expression of both glutamate transporters, GLAST and GLT-1, and of glutamate uptake. These effects were associated with neuronal dysfunction, as observed by a reduction in DARPP-32 and NR2B expression. Parallel studies in brain samples from HD subjects revealed early glial fibrillary acidic protein expression in striatal astrocytes from Grade 0 HD cases. Astrogliosis was associated with morphological changes that increased with severity of disease, from Grades 0 through 4 and was more prominent in the putamen. Combined immunofluorescence showed co-localization of mHtt in astrocytes in all striatal HD specimens, inclusive of Grade 0 HD. Consistent with the findings from experimental mice, there was a significant grade-dependent decrease in striatal GLT-1 expression from HD subjects. These findings suggest that the presence of mHtt in astrocytes alters glial glutamate transport capacity early in the disease process and may contribute to HD pathogenesis.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Huntington Disease/metabolism , Neostriatum/pathology , Peptides/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Aged , Amino Acid Transport System X-AG/metabolism , Animals , Astrocytes/pathology , Biological Transport , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Down-Regulation , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Humans , Huntington Disease/pathology , Lentivirus/genetics , Mice , Middle Aged , Mutant Proteins/metabolism , Neostriatum/metabolism , Neurons/metabolism , Neurons/pathology , Phenotype , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
14.
Hum Mol Genet ; 19(12): 2380-94, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20308049

ABSTRACT

Machado-Joseph disease or spinocerebellar ataxia type 3 (MJD/SCA3) is a fatal, autosomal dominant disorder caused by a cytosine-adenine-guanine expansion in the coding region of the MJD1 gene. RNA interference has potential as a therapeutic approach but raises the issue of the role of wild-type ataxin-3 (WT ATX3) in MJD and of whether the expression of the wild-type protein must be maintained. To address this issue, we both overexpressed and silenced WT ATX3 in a rat model of MJD. We showed that (i) overexpression of WT ATX3 did not protect against MJD pathology, (ii) knockdown of WT ATX3 did not aggravate MJD pathology and that (iii) non-allele-specific silencing of ataxin-3 strongly reduced neuropathology in a rat model of MJD. Our findings indicate that therapeutic strategies involving non-allele-specific silencing to treat MJD patients may be safe and effective.


Subject(s)
Machado-Joseph Disease/therapy , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA Interference , Repressor Proteins/genetics , Animals , Ataxin-3 , Cell Line , Disease Models, Animal , Humans , Machado-Joseph Disease/genetics , Machado-Joseph Disease/pathology , Male , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Repressor Proteins/physiology
15.
Ann Neurol ; 65(3): 276-85, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19334076

ABSTRACT

OBJECTIVE: Huntington's disease (HD) is a fatal autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion in the huntingtin (htt) protein. No cure is available to date to alleviate neurodegeneration. Recent studies have demonstrated that RNA interference represents a promising approach for the treatment of autosomal dominant disorders. But whether an allele-specific silencing of mutant htt or a nonallele-specific silencing should be considered has not been addressed. METHODS: We developed small hairpin RNA targeting mutant or wild-type htt transcripts, or both. RESULTS: We confirmed the therapeutic potential of sihtt administered with lentiviral vectors in rodent models of HD and showed that initiation of small interfering RNA treatment after the onset of HD symptoms is still efficacious and reduces the HD-like pathology. We then addressed the question of the impact of nonallele-specific silencing and demonstrated that silencing of endogenous htt to 25 to 35% in vivo is altering several pathways associated with known htt functions but is not inducing overt toxicity or increasing striatal vulnerability up to 9 months after treatment. INTERPRETATION: These data indicate that the coincident silencing of the wild-type and mutant htt may be considered as a therapeutic tool for HD.


Subject(s)
Huntington Disease/therapy , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Animals , Cell Line, Transformed , Corpus Striatum/metabolism , Deoxyglucose , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Doxycycline/metabolism , Exons/genetics , Female , Gene Expression Regulation/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Rats , Rats, Wistar , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Succinate Dehydrogenase/metabolism
16.
Glia ; 57(6): 667-79, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-18942755

ABSTRACT

Astrocytes are involved in key physiological brain processes, such as glutamatergic transmission and energy metabolism, often altered in neurodegenerative diseases. Targeted gene expression in astrocytes is needed to assess the contribution of these cells to physiological processes and for the development of new therapeutic strategies. However, most of the viral vectors currently used for gene transfer in the central nervous system (CNS) are highly neurotropic. We used mokola pseudotyping to shift the tropism of lentiviral vectors toward astrocytes and a detargeting strategy with miRNA to eliminate residual expression in neuronal cells. In primary cultures, we showed that incorporating target sequences for the neuron-specific miR124 effectively abolished transgene expression in neurons post-transcriptionally. Targeted expression of the LacZ reporter gene in astrocytes was achieved in the hippocampus, striatum, and cerebellum of the adult mouse in vivo. As a proof-of-principle, this new lentiviral vector was used to either overexpress or downregulate (RNA interference) the glial glutamate transporter GLAST into striatal astrocytes in vivo. These vectors provide new opportunities for cell type-specific gene transfer in the CNS.


Subject(s)
Astrocytes/physiology , Genetic Vectors , Lentivirus/genetics , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cells, Cultured , Cerebellum/physiology , Corpus Striatum/physiology , Gene Expression , Gene Targeting , Gene Transfer Techniques , Hippocampus/physiology , Lac Operon , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Neurons/physiology , RNA Interference
17.
PLoS One ; 3(10): e3341, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18841197

ABSTRACT

Recent studies have demonstrated that RNAi is a promising approach for treating autosomal dominant disorders. However, discrimination between wild-type and mutant transcripts is essential, to preserve wild-type expression and function. A single nucleotide polymorphism (SNP) is present in more than 70% of patients with Machado-Joseph disease (MJD). We investigated whether this SNP could be used to inactivate mutant ataxin-3 selectively. Lentiviral-mediated silencing of mutant human ataxin-3 was demonstrated in vitro and in a rat model of MJD in vivo. The allele-specific silencing of ataxin-3 significantly decreased the severity of the neuropathological abnormalities associated with MJD. These data demonstrate that RNAi has potential for use in MJD treatment and constitute the first proof-of-principle for allele-specific silencing in the central nervous system.


Subject(s)
Alleles , Disease Models, Animal , Gene Silencing , Machado-Joseph Disease/genetics , Mutation , Nerve Tissue Proteins/genetics , RNA/genetics , Animals , Ataxin-3 , Base Sequence , Brain/metabolism , Cell Line , DNA Primers , Humans , Machado-Joseph Disease/physiopathology , Male , Polymorphism, Single Nucleotide , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...