Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(13)2023 Jul 02.
Article in English | MEDLINE | ID: mdl-37446179

ABSTRACT

Cholesterol metabolism dysregulation is associated with several neurological disorders. In Huntington's disease (HD), several enzymes involved in cholesterol metabolism are downregulated, among which the neuronal cholesterol 24-hydroxylase, CYP46A1, is of particular interest. The restoration of CYP46A1 expression in striatal neurons of HD mouse models is beneficial for motor behavior, cholesterol metabolism, transcriptomic activity, and alleviates neuropathological hallmarks induced by mHTT. Among the genes regulated after CYP46A1 restoration, those involved in cholesterol synthesis and efflux may explain the positive effect of CYP46A1 on cholesterol precursor metabolites. Since cholesterol homeostasis results from a fine-tuning between neurons and astrocytes, we quantified the distribution of key genes regulating cholesterol metabolism and efflux in astrocytes and neurons using in situ hybridization coupled with S100ß and NeuN immunostaining, respectively. Neuronal expression of CYP46A1 in the striatum of HD zQ175 mice increased key cholesterol synthesis driver genes (Hmgcr, Dhcr24), specifically in neurons. This effect was associated with an increase of the srebp2 transcription factor gene that regulates most of the genes encoding for cholesterol enzymes. However, the cholesterol efflux gene, ApoE, was specifically upregulated in astrocytes by CYP46A1, probably though a paracrine effect. In summary, the neuronal expression of CYP46A1 has a dual and specific effect on neurons and astrocytes, regulating cholesterol metabolism. The neuronal restoration of CYP46A1 in HD paves the way for future strategies to compensate for mHTT toxicity.


Subject(s)
Huntington Disease , Mice , Animals , Cholesterol 24-Hydroxylase/genetics , Huntington Disease/metabolism , Neurons/metabolism , Cholesterol/metabolism , Homeostasis , Disease Models, Animal , Corpus Striatum/metabolism
3.
Front Aging Neurosci ; 14: 797220, 2022.
Article in English | MEDLINE | ID: mdl-35517051

ABSTRACT

Huntington's disease (HD) is an autosomal dominant genetic disorder caused by an expansion of the CAG repeat in the first exon of Huntingtin's gene. The associated neurodegeneration mainly affects the striatum and the cortex at early stages and progressively spreads to other brain structures. Targeting HD at its earlier stages is under intense investigation. Numerous drugs were tested, with a rate of success of only 3.5% approved molecules used as symptomatic treatment. The restoration of cholesterol metabolism, which is central to the brain homeostasis and strongly altered in HD, could be an interesting disease-modifying strategy. Cholesterol is an essential membrane component in the central nervous system (CNS); alterations of its homeostasis have deleterious consequences on neuronal functions. The levels of several sterols, upstream of cholesterol, are markedly decreased within the striatum of HD mouse model. Transcription of cholesterol biosynthetic genes is reduced in HD cell and mouse models as well as post-mortem striatal and cortical tissues from HD patients. Since the dynamic of brain cholesterol metabolism is complex, it is essential to establish the best method to target it in HD. Cholesterol, which does not cross the blood-brain-barrier, is locally synthesized and renewed within the brain. All cell types in the CNS synthesize cholesterol during development but as they progress through adulthood, neurons down-regulate their cholesterol synthesis and turn to astrocytes for their full supply. Cellular levels of cholesterol reflect the dynamic balance between synthesis, uptake and export, all integrated into the context of the cross talk between neurons and glial cells. In this review, we describe the latest advances regarding the role of cholesterol deregulation in neuronal functions and how this could be a determinant factor in neuronal degeneration and HD progression. The pathways and major mechanisms by which cholesterol and sterols are regulated in the CNS will be described. From this overview, we discuss the main clinical strategies for manipulating cholesterol metabolism in the CNS, and how to reinstate a proper balance in HD.

4.
Sci Adv ; 7(43): eabg5970, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34669474

ABSTRACT

Addictive drugs increase dopamine in the nucleus accumbens (NAc), where it persistently shapes excitatory glutamate transmission and hijacks natural reward processing. Here, we provide evidence, from mice to humans, that an underlying mechanism relies on drug-evoked heteromerization of glutamate N-methyl-d-aspartate receptors (NMDAR) with dopamine receptor 1 (D1R) or 2 (D2R). Using temporally controlled inhibition of D1R-NMDAR heteromerization, we unraveled their selective implication in early phases of cocaine-mediated synaptic, morphological, and behavioral responses. In contrast, preventing D2R-NMDAR heteromerization blocked the persistence of these adaptations. Interfering with these heteromers spared natural reward processing. Notably, we established that D2R-NMDAR complexes exist in human samples and showed that, despite a decreased D2R protein expression in the NAc, individuals with psychostimulant use disorder display a higher proportion of D2R forming heteromers with NMDAR. These findings contribute to a better understanding of molecular mechanisms underlying addiction and uncover D2R-NMDAR heteromers as targets with potential therapeutic value.

5.
J Med Chem ; 64(9): 5667-5688, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33949859

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS) that causes severe motor, sensory, and cognitive impairments. Kallikrein-related peptidase (KLK)6 is the most abundant serine protease secreted in the CNS, mainly by oligodendrocytes, the myelin-producing cells of the CNS, and KLK6 is assumed to be a robust biomarker of MS, since it is highly increased in the cerebrospinal fluid (CSF) of MS patients. Here, we report the design and biological evaluation of KLK6's low-molecular-weight inhibitors, para-aminobenzyl derivatives. Interestingly, selected hit compounds were selective of the KLK6 proteolytic network encompassing KLK1 and plasmin that also participate in the development of MS physiopathology. Moreover, hits were found noncytotoxic on primary cultures of murine neurons and oligodendrocyte precursor cells (OPCs). Among them, two compounds (32 and 42) were shown to promote the differentiation of OPCs into mature oligodendrocytes in vitro constituting thus emerging leads for the development of regenerative therapies.


Subject(s)
Cell Differentiation/drug effects , Kallikreins/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Animals , Benzene Derivatives/chemistry , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacology , Binding Sites , Catalytic Domain , Cells, Cultured , Drug Design , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Humans , Kallikreins/metabolism , Kinetics , Mice , Molecular Docking Simulation , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Structure-Activity Relationship
6.
BMC Res Notes ; 13(1): 210, 2020 Apr 10.
Article in English | MEDLINE | ID: mdl-32276655

ABSTRACT

OBJECTIVE: Compromised brain cholesterol turnover and altered regulation of brain cholesterol metabolism have been allied with some neurodegenerative diseases, including Huntington's disease (HD). Following our previous studies in HD, in this study we aim to investigate in vitro in a neuroblastoma cellular model of HD, the effect of CYP46A1 overexpression, an essential enzyme in cholesterol metabolism, on huntingtin aggregation and levels. RESULTS: We found that CYP46A1 reduces the quantity and size of mutant huntingtin aggregates in cells, as well as the levels of mutant huntingtin protein. Additionally, our results suggest that the observed beneficial effects of CYP46A1 in HD cells are linked to the activation of autophagy. Taken together, our results further demonstrate that CYP46A1 is a pertinent target to counteract HD progression.


Subject(s)
Autophagy , Cholesterol 24-Hydroxylase/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Neuroblastoma , Animals , Cell Line, Tumor , Cells, Cultured , Huntington Disease/enzymology , Mice , Mutant Proteins
8.
Brain ; 142(8): 2432-2450, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31286142

ABSTRACT

Dysfunctions in brain cholesterol homeostasis have been extensively related to brain disorders. The main pathway for brain cholesterol elimination is its hydroxylation into 24S-hydroxycholesterol by the cholesterol 24-hydrolase, CYP46A1. Increasing evidence suggests that CYP46A1 has a role in the pathogenesis and progression of neurodegenerative disorders, and that increasing its levels in the brain is neuroprotective. However, the mechanisms underlying this neuroprotection remain to be fully understood. Huntington's disease is a fatal autosomal dominant neurodegenerative disease caused by an abnormal CAG expansion in huntingtin's gene. Among the multiple cellular and molecular dysfunctions caused by this mutation, altered brain cholesterol homeostasis has been described in patients and animal models as a critical event in Huntington's disease. Here, we demonstrate that a gene therapy approach based on the delivery of CYP46A1, the rate-limiting enzyme for cholesterol degradation in the brain, has a long-lasting neuroprotective effect in Huntington's disease and counteracts multiple detrimental effects of the mutated huntingtin. In zQ175 Huntington's disease knock-in mice, CYP46A1 prevented neuronal dysfunctions and restored cholesterol homeostasis. These events were associated to a specific striatal transcriptomic signature that compensates for multiple mHTT-induced dysfunctions. We thus explored the mechanisms for these compensations and showed an improvement of synaptic activity and connectivity along with the stimulation of the proteasome and autophagy machineries, which participate to the clearance of mutant huntingtin (mHTT) aggregates. Furthermore, BDNF vesicle axonal transport and TrkB endosome trafficking were restored in a cellular model of Huntington's disease. These results highlight the large-scale beneficial effect of restoring cholesterol homeostasis in neurodegenerative diseases and give new opportunities for developing innovative disease-modifying strategies in Huntington's disease.


Subject(s)
Brain/metabolism , Cholesterol 24-Hydroxylase/therapeutic use , Cholesterol/metabolism , Genetic Therapy , Genetic Vectors/therapeutic use , Huntington Disease/therapy , Neuroprotective Agents/therapeutic use , Animals , Autophagy , Axonal Transport , Brain-Derived Neurotrophic Factor/physiology , Cells, Cultured , Cerebral Cortex/physiopathology , Cholesterol 24-Hydroxylase/genetics , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Dependovirus/genetics , Endosomes/metabolism , Gene Knock-In Techniques , Genetic Vectors/genetics , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/physiopathology , Neuroprotective Agents/administration & dosage , Oxysterols/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Aggregation, Pathological , Protein-Tyrosine Kinases/physiology , Rotarod Performance Test , Synaptic Transmission , Transcriptome
9.
Biochimie ; 153: 70-79, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30107216

ABSTRACT

Huntington's Disease (HD) is an autosomal dominant neurodegenerative disease caused by abnormal polyglutamine expansion in huntingtin (mHtt) protein leading to degeneration of striatal neurons. Excitotoxicity, consecutive to overstimulation of N-methyl d-aspartate receptors (NMDARs) has a pivotal role in many neurological disorders including HD. Mutant Htt causes enhanced NMDA sensitivity, alteration of NMDAR expression and localization in neurons. Excitotoxic events initiate neuronal death in numerous ways, including activation of apoptotic cascades. Among the NMDAR subunits involved in glutamatergic-mediated excitotoxicity, GluN2B has been extensively reported. In addition to excitotoxicity, alteration of cholesterol metabolism has been observed in HD, with a decrease of cholesterol precursor synthesis along with an increase of cholesterol accumulation, which is deleterious for neurons. Expression of Cholesterol Hydroxylase enzyme, CYP46A1, which converts cholesterol into 24 S-hydroxycholesterol is down-regulated in HD. We found that CYP46A1 overexpression is beneficial in HD neurons and mouse model, but the mechanisms involved still remain unclear. In this study we addressed the effect of CYP46A1 on NMDAR-mediated excitotoxicity in HD primary neurons and its role in modulating cholesterol and localization of GLUN2B in lipid rafts. We showed that CYP46A1 is protective against NMDAR-mediated excitotoxicity in two different HD neuronal cell models. Cholesterol as well as GluN2B level in lipid raft, are significantly increased by mHtt. Despite a clear effect of CYP46A1 in reducing cholesterol content in lipid raft extracts from wild type neurons, CYP46A1 overexpression in HD neurons could not normalize the increased cholesterol levels in lipid rafts. This study highlights the beneficial role of CYP46A1 against NMDAR-mediated excitotoxicity and gives further insights into the cellular mechanisms underlying CYP46A1-mediated neuroprotection.


Subject(s)
Cholesterol 24-Hydroxylase/metabolism , Huntington Disease/prevention & control , Membrane Microdomains/metabolism , N-Methylaspartate/toxicity , Animals , Cholesterol/metabolism , Corpus Striatum/cytology , Corpus Striatum/enzymology , Female , Homeostasis , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Male , Mice , Mutation , Neurons/enzymology , Receptors, N-Methyl-D-Aspartate/metabolism
10.
Biol Psychiatry ; 82(11): 806-818, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28545678

ABSTRACT

BACKGROUND: Repeated cocaine exposure produces new spine formation in striatal projection neurons (SPNs) of the nucleus accumbens. However, an acute exposure to cocaine can trigger long-lasting synaptic plasticity in SPNs leading to behavioral alterations. This raises the intriguing question as to whether a single administration of cocaine could enduringly modify striatal connectivity. METHODS: A three-dimensional morphometric analysis of presynaptic glutamatergic boutons and dendritic spines was performed on SPNs 1 hour and 1 week after a single cocaine administration. Time-lapse two-photon microscopy in adult slices was used to determine the precise molecular-events sequence responsible for the rapid spine formation. RESULTS: A single injection triggered a rapid synaptogenesis and persistent increase in glutamatergic connectivity in SPNs from the shell part of the nucleus accumbens, specifically. Synapse formation occurred through clustered growth of active spines contacting pre-existing axonal boutons. Spine growth required extracellular signal-regulated kinase activation, while spine stabilization involved transcription-independent protein synthesis driven by mitogen-activated protein kinase interacting kinase-1, downstream from extracellular signal-regulated kinase. The maintenance of new spines driven by mitogen-activated protein kinase interacting kinase-1 was essential for long-term connectivity changes induced by cocaine in vivo. CONCLUSIONS: Our study originally demonstrates that a single administration of cocaine is able to induce stable synaptic rewiring in the nucleus accumbens, which will likely influence responses to subsequent drug exposure. It also unravels a new functional role for cocaine-induced extracellular signal-regulated kinase pathway independently of nuclear targets. Finally, it reveals that mitogen-activated protein kinase interacting kinase-1 has a pivotal role in cocaine-induced connectivity.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Kinase 1/metabolism , Neurogenesis/drug effects , Nucleus Accumbens/drug effects , Synapses/physiology , Animals , Dendritic Spines/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neostriatum/metabolism , Nucleus Accumbens/cytology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Dopamine D1/metabolism , Sirolimus/pharmacology , Synapses/drug effects , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism
11.
Eur J Neurosci ; 45(1): 198-206, 2017 01.
Article in English | MEDLINE | ID: mdl-27717053

ABSTRACT

Huntington's disease, an inherited neurodegenerative disorder, results from abnormal polyglutamine extension in the N-terminal region of the huntingtin protein. This mutation causes preferential degeneration of striatal projection neurons. We previously demonstrated, in vitro, that dopaminergic D2 receptor stimulation acted in synergy with expanded huntingtin to increase aggregates formation and striatal death through activation of the Rho/ROCK signaling pathway. In vivo, in a lentiviral-mediated model of expanded huntingtin expression in the rat striatum, we found that the D2 antagonist haloperidol protects striatal neurons against expanded huntingtin-mediated toxicity. Two variant transcripts are generated by alternative splicing of the of D2 receptor gene, the D2R-Long and the D2R-Short, which are thought to play different functional roles. We show herein that overexpression of D2R-Short, but not D2R-Long in cell lines is associated with activation of the RhoA/ROCK signaling pathway. In striatal neurons in culture, the selective D2 agonist Quinpirole triggers phosphorylation of cofilin, a downstream effector of ROCK, which is abrogated by siRNAs that knockdown both D2R-Long and D2R-Short, but not by siRNAs targeting D2R-Long alone. Aggregate formation and neuronal death induced by expanded huntingtin, were potentiated by Quinpirole. This D2 agonist-mediated effect was selectively inhibited by the siRNA targeting both D2R-Long and D2R-Short but not D2R-Long alone. Our data provide evidence for a specific coupling of D2R-Short to the RhoA/ROCK/cofilin pathway, and its involvement in striatal vulnerability to expanded huntingtin. A new route for targeting Rho-ROCK signaling in Huntington's disease is unraveled with our findings.


Subject(s)
Corpus Striatum/metabolism , Huntington Disease/metabolism , Neostriatum/metabolism , Neurons/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction , Dopamine/metabolism , Humans , Huntingtin Protein/metabolism , Huntington Disease/genetics , Protein Isoforms/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
12.
Brain ; 139(Pt 3): 953-70, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26912634

ABSTRACT

Huntington's disease is an autosomal dominant neurodegenerative disease caused by abnormal polyglutamine expansion in huntingtin (Exp-HTT) leading to degeneration of striatal neurons. Altered brain cholesterol homeostasis has been implicated in Huntington's disease, with increased accumulation of cholesterol in striatal neurons yet reduced levels of cholesterol metabolic precursors. To elucidate these two seemingly opposing dysregulations, we investigated the expression of cholesterol 24-hydroxylase (CYP46A1), the neuronal-specific and rate-limiting enzyme for cholesterol conversion to 24S-hydroxycholesterol (24S-OHC). CYP46A1 protein levels were decreased in the putamen, but not cerebral cortex samples, of post-mortem Huntington's disease patients when compared to controls. Cyp46A1 mRNA and CYP46A1 protein levels were also decreased in the striatum of the R6/2 Huntington's disease mouse model and in SThdhQ111 cell lines. In vivo, in a wild-type context, knocking down CYP46A1 expression in the striatum, via an adeno-associated virus-mediated delivery of selective shCYP46A1, reproduced the Huntington's disease phenotype, with spontaneous striatal neuron degeneration and motor deficits, as assessed by rotarod. In vitro, CYP46A1 restoration protected SThdhQ111 and Exp-HTT-expressing striatal neurons in culture from cell death. In the R6/2 Huntington's disease mouse model, adeno-associated virus-mediated delivery of CYP46A1 into the striatum decreased neuronal atrophy, decreased the number, intensity level and size of Exp-HTT aggregates and improved motor deficits, as assessed by rotarod and clasping behavioural tests. Adeno-associated virus-CYP46A1 infection in R6/2 mice also restored levels of cholesterol and lanosterol and increased levels of desmosterol. In vitro, lanosterol and desmosterol were found to protect striatal neurons expressing Exp-HTT from death. We conclude that restoring CYP46A1 activity in the striatum promises a new therapeutic approach in Huntington's disease.


Subject(s)
Cholesterol/metabolism , Huntington Disease/enzymology , Huntington Disease/prevention & control , Steroid Hydroxylases/biosynthesis , Aged , Aged, 80 and over , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cholesterol 24-Hydroxylase , Female , Humans , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Middle Aged
13.
Front Neurol ; 4: 127, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24167500

ABSTRACT

Huntington's Disease (HD) is a genetic neurodegenerative disease caused by a CAG expansion in the gene encoding Huntingtin (Htt). It is characterized by chorea, cognitive, and psychiatric disorders. The most affected brain region is the striatum, and the clinical symptoms are directly correlated to the rate of striatal degeneration. The wild-type Htt is a ubiquitous protein and its deletion is lethal. Mutated (expanded) Htt produces excitotoxicity, mitochondrial dysfunctions, axonal transport deficit, altered proteasome activity, and gene dysregulation. Transcriptional dysregulation occurs at early neuropathological stages in HD patients. Multiple genes are dysregulated, with overlaps of altered transcripts between mouse models of HD and patient brains. Nuclear localization of Exp-Htt interferes with transcription factors, co-activators, and proteins of the transcriptional machinery. Another key mechanism described so far, is an alteration of cytoplasmic retention of the transcriptional repressor REST, which is normally associated with wild-type Htt. As such, Exp-Htt causes alteration of transcription of multiple genes involved in neuronal survival, plasticity, signaling, and mitochondrial biogenesis and respiration. Besides these transcriptional dysregulations, Exp-Htt affects the chromatin structure through altered post-translational modifications (PTM) of histones and methylation of DNA. Multiple alterations of histone PTM are described, including acetylation, methylation, ubiquitylation, polyamination, and phosphorylation. Exp-Htt also affects the expression and regulation of non-coding microRNAs (miRNAs). First multiple neural miRNAs are controlled by REST, and dysregulated in HD, with concomitant de-repression of downstream mRNA targets. Second, Exp-Htt protein or RNA may also play a major role in the processing of miRNAs and hence pathogenesis. These pleiotropic effects of Exp-Htt on gene expression may represent seminal deleterious effects in the pathogenesis of HD.

14.
Brain Struct Funct ; 217(2): 421-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21822732

ABSTRACT

Dendritic spines are postsynaptic structures the morphology of which correlates with the strength of synaptic efficacy. Measurements of spine density and spine morphology are achievable using recent imaging and bioinformatics tools. The three-dimensional automated analysis requires optimization of image acquisition and treatment. Here, we studied the critical steps for optimal confocal microscopy imaging of dendritic spines. We characterize the deconvolution process and show that it improves spine morphology analysis. With this method, images of dendritic spines from medium spiny neurons are automatically detected by the software Neuronstudio, which retrieves spine density as well as spine diameter and volume. This approach is illustrated with three-dimensional analysis of dendritic spines in a mouse model of Huntington's disease: the transgenic R6/2 mice. In symptomatic mutant mice, we confirm the decrease in spine density, and the method brings further information and show a decrease in spine volume and dendrite diameter. Moreover, we show a significant decrease in spine density at presymptomatic age which so far has gone unnoticed.


Subject(s)
Dendritic Spines/pathology , Huntington Disease/pathology , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Animals , Disease Models, Animal , Humans , Huntingtin Protein , Huntington Disease/genetics , Image Processing, Computer-Assisted , Mice , Mice, Mutant Strains , Mice, Transgenic , Nerve Tissue Proteins/genetics , Software
15.
Front Neuroanat ; 5: 55, 2011.
Article in English | MEDLINE | ID: mdl-22007160

ABSTRACT

Huntington's Disease (HD) is the most frequent neurodegenerative disease caused by an expansion of polyglutamines (CAG). The main clinical manifestations of HD are chorea, cognitive impairment, and psychiatric disorders. The transmission of HD is autosomal dominant with a complete penetrance. HD has a single genetic cause, a well-defined neuropathology, and informative pre-manifest genetic testing of the disease is available. Striatal atrophy begins as early as 15 years before disease onset and continues throughout the period of manifest illness. Therefore, patients could theoretically benefit from therapy at early stages of the disease. One important characteristic of HD is the striatal vulnerability to neurodegeneration, despite similar expression of the protein in other brain areas. Aggregation of the mutated Huntingtin (HTT), impaired axonal transport, excitotoxicity, transcriptional dysregulation as well as mitochondrial dysfunction, and energy deficits, are all part of the cellular events that underlie neuronal dysfunction and striatal death. Among these non-exclusive mechanisms, an alteration of striatal signaling is thought to orchestrate the downstream events involved in the cascade of striatal dysfunction.

16.
Lab Chip ; 11(21): 3663-73, 2011 Nov 07.
Article in English | MEDLINE | ID: mdl-21922081

ABSTRACT

Various experimental models are used to study brain development and degeneration. They range from whole animal models, which preserve anatomical structures but strongly limit investigations at the cellular level, to dissociated cell culture systems that allow detailed observation of cell phenotypes but lack the highly ordered physiological neuron connection architecture. We describe here a platform comprising independent cell culture chambers separated by an array of "axonal diodes". This array involves asymmetric micro-channels, imposing unidirectional axon connectivity with 97% selectivity. It allows the construction of complex, oriented neuronal networks not feasible with earlier platforms. Different neuronal subtypes could be co-cultivated for weeks, and sequential seeding of different cell populations reproduced physiological network development. To illustrate possible applications, we created and characterized a cortico-striatal oriented network. Functional synaptic connections were established. The activation of striatal differentiation by cortical axons, and the synchronization of neural activity were demonstrated. Each neuronal population and subcompartment could be chemically addressed individually. The directionality of neural pathways being a key feature of the nervous system organization, the axon diode concept brings in a paradigmatic change in neuronal culture platforms, with potential applications for studying neuronal development, synaptic transmission and neurodegenerative disorder such as Alzheimer and Parkinson diseases at the sub-cellular, cellular and network levels.


Subject(s)
Axons/physiology , Microfluidic Analytical Techniques , Nerve Net/cytology , Neurons/cytology , Aniline Compounds/chemistry , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Transgenic , Nerve Net/metabolism , Nerve Net/physiology , Neurons/metabolism , Xanthenes/chemistry
17.
Hum Mol Genet ; 20(12): 2422-34, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21493629

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder due to abnormal polyglutamine expansion in huntingtin protein (Exp-Htt). This expansion causes protein aggregation, leading to neuronal dysfunction and death. We have previously shown that mitogen- and stress-activated kinase (MSK-1), a nuclear protein kinase involved in chromatin remodeling through histone H3 phosphorylation, is deficient in the striatum of HD patients and model mice. Restoring MSK-1 expression in cultured striatal cells prevented neuronal dysfunction and death induced by Exp-Htt. Here we extend these observations in a rat model of HD based on striatal lentiviral expression of Exp-Htt (LV-Exp-HTT). MSK-1 overexpression attenuated Exp-Htt-induced down-regulation of DARPP-32 expression 4 and 10 weeks after infection and enhanced NeuN staining after 10 weeks. LV-MSK-1 induced constitutive hyperphosphorylation of H3 and cAMP-responsive element binding protein (CREB), indicating that MSK-1 has spontaneous catalytic activity. MSK-1 overexpression also upregulated peroxisome proliferator-activated receptor γ coactivator alpha (PGC-1α), a transcriptional co-activator involved in mitochondrial biogenesis. Chromatin immunoprecipitation indicated that transcriptional regulation of PGC-1α is directly linked to increased binding of MSK-1, along with H3 and CREB phosphorylation of the PGC-1α promoter. MSK-1 knock-out mice showed spontaneous striatal atrophy as they aged, as well as higher susceptibility to systemic administration of the mitochondrial neurotoxin 3-NP. These results indicate that MSK-1 activation is an important and key event in the signaling cascade that regulates PGC-1α expression. Strategies aimed at restoring MSK-1 expression in the striatum might offer a new therapeutic approach to HD.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Corpus Striatum/metabolism , Gene Expression Regulation/drug effects , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Nuclear Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/pharmacology , Analysis of Variance , Animals , Chromatin Assembly and Disassembly/physiology , Chromatin Immunoprecipitation , DNA Repeat Expansion/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation/physiology , Genetic Vectors/genetics , Huntingtin Protein , Immunohistochemistry , Lentivirus , Mice , Mice, Knockout , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Neuroprotective Agents/metabolism , Nuclear Proteins/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Adv Exp Med Biol ; 685: 45-63, 2010.
Article in English | MEDLINE | ID: mdl-20687494

ABSTRACT

Huntington's disease (HD) is one of the most frequently found neurodegenerative disorders. Its main clinical manifestations are chorea, cognitive impairment and psychiatric disorders. It is an autosomal-dominant disorder with almost complete penetrance. The mutation responsible for HD, unstable expansion of a CAG repeat, is located in the 5' terminal section of the gene (IT15) that encodes huntingtin protein (Htt). The pathophysiology of HD is not entirely clear. One intriguing characteristic of HD is the special vulnerability of the striatum tomutated Htt, despite similar expression of the mutated protein in other brain regions. Aggregation of mutated Htt, transcriptional dysregulation, altered energy metabolism, excitotoxicity, impaired axonal transport and altered synaptic transmission culminate in neuronal dysfunction and death. There is currently no way of preventing or slowing down the disease progression and death usually occurs at about 20 years after diagnosis.


Subject(s)
Chromosome Disorders/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion , Animals , Axons/metabolism , Axons/pathology , Basal Ganglia/metabolism , Basal Ganglia/pathology , Basal Ganglia/physiopathology , Biological Transport/genetics , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosome Disorders/physiopathology , Energy Metabolism/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Huntington Disease/physiopathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics
19.
PLoS One ; 4(12): e8287, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20016831

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a polyglutamine-expanded related neurodegenerative disease. Despite the ubiquitous expression of expanded, polyQ-Huntingtin (ExpHtt) in the brain, striatal neurons present a higher susceptibility to the mutation. A commonly admitted hypothesis is that Dopaminergic inputs participate to this vulnerability. We previously showed that D2 receptor stimulation increased aggregate formation and neuronal death induced by ExpHtt in primary striatal neurons in culture, and chronic D2 antagonist treatment protects striatal dysfunctions induced by ExpHtt in a lentiviral-induced model system in vivo. The present work was designed to elucidate the signalling pathways involved, downstream D2 receptor (D2R) stimulation, in striatal vulnerability to ExpHtt. METHODOLOGY/PRINCIPAL FINDINGS: Using primary striatal neurons in culture, transfected with a tagged-GFP version of human exon 1 ExpHtt, and siRNAs against D2R or D1R, we confirm that DA potentiates neuronal dysfunctions via D2R but not D1R stimulation. We demonstrate that D2 agonist treatment induces neuritic retraction and growth cone collapse in Htt- and ExpHtt expressing neurons. We then tested a possible involvement of the Rho/ROCK signalling pathway, which plays a key role in the dynamic of the cytoskeleton, in these processes. The pharmacological inhibitors of ROCK (Y27632 and Hydroxyfasudil), as well as siRNAs against ROCK-II, reversed D2-related effects on neuritic retraction and growth cone collapse. We show a coupling between D2 receptor stimulation and Rho activation, as well as hyperphosphorylation of Cofilin, a downstream effector of ROCK-II pathway. Importantly, D2 agonist-mediated potentiation of aggregate formation and neuronal death induced by ExpHtt, was totally reversed by Y27632 and Hydroxyfasudil and ROCK-II siRNAs. CONCLUSIONS/SIGNIFICANCE: Our data provide the first demonstration that D2R-induced vulnerability in HD is critically linked to the activation of the Rho/ROCK signalling pathway. The inclusion of Rho/ROCK inhibitors could be an interesting therapeutic option aimed at forestalling the onset of the disease.


Subject(s)
Neostriatum/physiopathology , Neurons/enzymology , Peptides/toxicity , Receptors, Dopamine D2/metabolism , Serotonin Plasma Membrane Transport Proteins/toxicity , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Cell Death/drug effects , Dopamine/pharmacology , Enzyme Activation/drug effects , Growth Cones/drug effects , Growth Cones/pathology , Humans , Mice , Neostriatum/drug effects , Neostriatum/enzymology , Neurites/drug effects , Neurites/metabolism , Neurons/drug effects , Protein Structure, Quaternary , Quinpirole/pharmacology , RNA, Small Interfering , Receptors, Dopamine D1/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Trinucleotide Repeat Expansion/genetics
20.
J Neurochem ; 108(6): 1323-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19183268

ABSTRACT

Drugs of abuse induce neuroadaptations through regulation of gene expression. Although much attention has focused on transcription factor activities, new concepts have recently emerged on the role of chromatin remodelling as a prerequisite for regulation of gene expression in neurons. Thus, for transcription to occur, chromatin must be decondensed, a dynamic process that depends on post-translational modifications of histones. We review here these modifications with a particular emphasis on the role of histone H3 phosphorylation at the promoter of specific genes, including c-fos and c-jun. We trace the signalling pathways involved in H3 phosphorylation and provide evidence for a role of mitogen and stress-activated protein kinase-1 (MSK1) downstream from the MAPK/extracellular-signal regulated kinase (ERK) cascade. In response to cocaine, MSK1 controls an early phase of histone H3 phosphorylation at the c-fos promoter in striatal neurons. MSK1 action may be potentiated by the concomitant inhibition of protein phosphatase 1 by nuclear translocation of dopamine- and cAMP-regulated phosphoprotein Mr = 32 000. H3 phosphorylation by MSK1 is critically involved in c-fos transcription, and cocaine-induced locomotor sensitization. Thus, ERK plays a dual role in gene regulation and drug addiction by direct activation of transcription factors and by chromatin remodelling.


Subject(s)
Brain/physiopathology , Chromatin Assembly and Disassembly/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Illicit Drugs/pharmacology , Signal Transduction/physiology , Animals , Brain/drug effects , Brain/enzymology , Histones/metabolism , Humans , Phosphorylation , Protein Processing, Post-Translational/drug effects , Substance-Related Disorders/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...