Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
2.
J Psychiatr Res ; 141: 66-73, 2021 09.
Article in English | MEDLINE | ID: mdl-34175744

ABSTRACT

Little is known regarding the brain substrates of Gambling Disorder, including surface brain morphometry, and whether these are linked to the clinical profile. A better understanding of the brain substrates will likely help determine targets to treat patients. Hence, the aim of this study was two-fold, that is to examine surface-based morphometry in 17 patients with gambling disorder as compared to norms of healthy individuals (2713 and 2790 subjects for cortical and subcortical anatomical scans, respectively) and to assess the clinical relevance of morphometry in patients with Gambling Disorder. This study measured brain volume, surface and thickness in Gambling Disorder. We compared these measures to those of a normative database that controlled for factors such as age and sex. We also tested for correlations with gambling-related behaviors, such as gambling severity and duration, impulsivity, and depressive symptoms (assessed using the South Oaks Gambling Screen, years of gambling, Barratt Impulsiveness Scale, and Beck Depression Inventory, respectively). Patients displayed thinner prefrontal and parietal cortices, greater volume and thickness of the occipital and the entorhinal cortices, and greater volume of subcortical regions as compared to the norms of healthy individuals. There were positive correlations between surface area of occipital regions and depressive symptoms. This work contributes to better characterize the brain substrates of Gambling Disorder, which appear to resemble those of substance use disorders and Internet Gaming Disorder.


Subject(s)
Gambling , Adult , Brain/diagnostic imaging , Gambling/diagnostic imaging , Humans , Impulsive Behavior , Internet Addiction Disorder , Magnetic Resonance Imaging , Psychiatric Status Rating Scales
3.
Brain Connect ; 11(10): 815-821, 2021 12.
Article in English | MEDLINE | ID: mdl-34128386

ABSTRACT

Background/Introduction: Transcranial direct current stimulation (tDCS) delivered over the dorsolateral prefrontal cortex (DLPFC) while patients are at rest can decrease craving in patients with substance-related and addictive disorders. Yet, the effects of tDCS on resting-state brain activity remain unknown in this population. This study examined the effects of tDCS on resting-state functional connectivity (rsFC) with concurrent stimulation and functional magnetic resonance imaging in patients with gambling disorder. Methods: This was a randomized, sham-controlled, double-blind, crossover study. The anodal and cathodal electrodes were applied over the right and left DLPFC, respectively. Patients received 30 min of active and sham stimulation on separate days. rsFC was assessed before and during stimulation with seed-based analyses. Results: There was a significant increase of rsFC between the right DLPFC seed and the right superior parietal lobule during active stimulation as compared to during sham stimulation (p = 0.0059, corrected for multiple comparisons). There was also a positive correlation between rsFC change of this frontoparietal network and brain volume of the right DLPFC (p = 0.0042, corrected for multiple comparisons). Discussion: A single session of tDCS targeting the DLPFC strengthened functional connectivity in a frontoparietal circuit, known to be implicated in cognitive control, especially in patients with a greater volume of the region under the anode electrode. Impact statement Transcranial direct current stimulation increased the functional connectivity of a frontoparietal circuit in patients with gambling disorder. These changes were larger in patients with greater volume of the dorsolateral prefrontal cortex. Transcranial direct current stimulation strengthened the connectivity of a brain network known to be associated with cognitive control.


Subject(s)
Gambling , Transcranial Direct Current Stimulation , Brain/diagnostic imaging , Cross-Over Studies , Dorsolateral Prefrontal Cortex , Humans , Magnetic Resonance Imaging , Prefrontal Cortex/diagnostic imaging , Transcranial Magnetic Stimulation
4.
Eur J Neurosci ; 51(12): 2412-2422, 2020 06.
Article in English | MEDLINE | ID: mdl-31944461

ABSTRACT

l-3,4-Dihydroxyphenylalanine (l-DOPA) is the most effective treatment for Parkinson's disease (PD), but its use over a long period is marred by motors complications such as dyskinesia. We previously demonstrated that selective metabotropic glutamate 2/3 (mGlu2/3 ) receptor activation with LY-354,740 alleviates dyskinesia in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned marmoset and the 6-hydroxydopamine (6-OHDA)-lesioned rat. Here, we sought to determine the role played by selective mGlu2 activation in the anti-dyskinetic effect of mGlu2/3 stimulation and have investigated the effect of the highly selective mGlu2 positive allosteric modulator LY-487,379 at alleviating established, and preventing the development of, l-DOPA-induced dyskinesia in the 6-OHDA-lesioned rat. First, dyskinetic 6-OHDA-lesioned rats were administered l-DOPA in combination with LY-487,379 (0.1, 1 and 10 mg/kg) or vehicle, and the severity of dyskinesia was determined. Second, 6-OHDA-lesioned rats were administered LY-487,379 (0.1 or 1 mg/kg), started concurrently with l-DOPA, once daily for 22 days, and dyskinesia severity was evaluated weekly for four consecutive weeks. We also assessed the effect of LY-487,379 on l-DOPA anti-parkinsonian effect. We found that acute challenges of LY-487,379 0.1 mg/kg in combination with l-DOPA, significantly diminished dyskinesia severity, by ≈54% (p < .01), when compared to vehicle. Moreover, animals treated with l-DOPA/LY-487,379 0.1 and 1 mg/kg during the dyskinesia induction phase exhibited milder dyskinesia, by ≈74% and ≈61%, respectively (both p < .01), when compared to l-DOPA/vehicle. LY-487,379 did not impair l-DOPA anti-parkinsonian activity. These results suggest that mGlu2 activation may be an effective and promising therapeutic strategy to alleviate the severity and prevent the development of dyskinesia.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Animals , Antiparkinson Agents , Callithrix , Disease Models, Animal , Dyskinesia, Drug-Induced/drug therapy , Levodopa , Oxidopamine/toxicity , Rats
6.
J Biol Chem ; 294(13): 4889-4897, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30696767

ABSTRACT

The Nr4a subfamily of nuclear receptor comprises three members in mammalian cells: Nur77/Nr4a1, Nurr1/Nr4a2, and Nor1/Nr4a3. Nr4a proteins play key roles in the regulation of glucose homeostasis in peripheral metabolic tissues. However, their biological functions in ß-cells remain relatively uncharacterized. Here we sought to investigate the potential role of Nor1 in the regulation of ß-cell mass and, in particular, ß-cell survival/apoptosis. We used histological analysis to examine the consequences of genetic deletion of either Nur77 and Nor1 on ß-cell mass, investigated the expression patterns of Nr4as in human islets and INS cells and performed gain- and loss-of-function experiments to further characterize the role of Nor1 in ß-cell apoptosis. Surprisingly, Nor1 knockout mice displayed increased ß-cell mass, whereas mice with genetic deletion of Nur77 did not exhibit any significant differences compared with their WT littermates. The increase in ß-cell mass in Nor1 knockout mice was accompanied by improved glucose tolerance. A gene expression study performed in both human islets and INS cells revealed that Nor1 expression is significantly increased by pro-inflammatory cytokines and, to a lesser extent, by elevated concentrations of glucose. Nor1 overexpression in both INS and human islet cells caused apoptosis, whereas siRNA-mediated Nor1 knockdown prevented cytokine-induced ß-cell death. Finally, Nor1 expression was up-regulated in islets of individuals with type 2 diabetes. Altogether, our results uncover that Nor1 negatively regulates ß-cell mass. Nor1 represents a promising molecular target in diabetes treatment to prevent ß-cell destruction.


Subject(s)
Apoptosis , DNA-Binding Proteins/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Up-Regulation , Animals , Cytokines , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Humans , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics
7.
Exp Neurol ; 304: 143-153, 2018 06.
Article in English | MEDLINE | ID: mdl-29530712

ABSTRACT

Parkinson's disease (PD) is an idiopathic progressive neurodegenerative disorder characterized by the loss of midbrain dopamine neurons. Levodopa (l-dopa) is the main pharmacological approach to relieve PD motor symptoms. However, chronic treatment with l-Dopa is inevitably associated with the generation of abnormal involuntary movements (l-Dopa-induced dyskinesia). We have previously shown that Nr4a1 (Nur77), a transcription factor of the nuclear receptor family, is closely associated with dopamine neurotransmission in the mature brain. However, the role of Nr4a1 in the etiology of PD and its treatment remain elusive. We report here that the neurotoxin 6-hydroxydopamine in rat lead to a rapid up-regulation of Nr4a1 in the substantia nigra. Genetic disruption of Nr4a1 in rat reduced neurotoxin-induced dopamine cell loss and l-Dopa-induced dyskinesia, whereas virally-driven striatal overexpression of Nr4a1 enhanced or partially restored involuntary movements induced by chronic l-Dopa in wild type and Nr4a1-deficient rats, respectively. Collectively, these results suggest that Nr4a1 is involved in dopamine cell loss and l-Dopa-induced dyskinesia in experimental PD.


Subject(s)
Dopaminergic Neurons/pathology , Dyskinesia, Drug-Induced/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Parkinsonian Disorders/metabolism , Animals , Antiparkinson Agents/toxicity , Gene Knockout Techniques , Levodopa/toxicity , Mice , Parkinsonian Disorders/pathology , Rats
8.
Neuropharmacology ; 133: 289-306, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29407218

ABSTRACT

The dopamine transporter (DAT) is abundantly expressed in the striatum where it removes extracellular dopamine into the cytosol of presynaptic nerve terminals. It is the target of drugs of abuse and antidepressants. There is a loss of the DAT in Parkinson's disease affecting release of levodopa implicated in levodopa-induced dyskinesias. This study investigated the effect of cholesterol on DAT, serotonin transporter (SERT) and vesicular monoamine transporter 2 (VMAT2) in monkey and rat brains in vitro. DAT protein levels measured by Western blot remained unchanged with in vitro methyl-ß-cyclodextrin (MCD) incubations to remove membrane cholesterol or with incubations to increase membrane cholesterol content. By contrast, striatal DAT specific binding labelled with [125I]RTI-121 or with [125I]RTI-55 decreased with increasing concentrations of MCD and increased with cholesterol loading. Moreover, [125I]RTI-121 specific binding of striatal membranes depleted of cholesterol with MCD was restored to initial DAT content with addition of cholesterol showing its rapid and reversible effect. By contrast, striatal VMAT2 and SERT specific binding showed no or limited changes by cholesterol manipulations. Similar results were obtained for monkey caudate nucleus, putamen and nucleus accumbens. Membrane microviscosity was assessed by fluorescence polarization spectroscopy, using the probe 1,6-diphenyl-1,3,5-hexatriene. DAT changes positively correlated with changes of membrane microviscosity in rat and monkey brain regions investigated and with membrane cholesterol contents. Similar findings were observed with desmosterol but to a lower extent than with cholesterol. These results show an important effect of cholesterol on the DAT associated with microviscosity changes that should be considered in drug therapies.


Subject(s)
Brain/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Membrane Transport Proteins/metabolism , Animals , Brain/drug effects , Cell Membrane/drug effects , Cholesterol/pharmacology , Cocaine/analogs & derivatives , Cocaine/pharmacokinetics , Cyclodextrins/pharmacology , Dopamine/pharmacology , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Iodine Radioisotopes/pharmacokinetics , Macaca fascicularis , Male , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley
9.
J Neurosci ; 38(6): 1335-1350, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29295823

ABSTRACT

The energetic costs of behavioral chronic stress are unlikely to be sustainable without neuronal plasticity. Mitochondria have the capacity to handle synaptic activity up to a limit before energetic depletion occurs. Protective mechanisms driven by the induction of neuronal genes likely evolved to buffer the consequences of chronic stress on excitatory neurons in prefrontal cortex (PFC), as this circuitry is vulnerable to excitotoxic insults. Little is known about the genes involved in mitochondrial adaptation to the buildup of chronic stress. Using combinations of genetic manipulations and stress for analyzing structural, transcriptional, mitochondrial, and behavioral outcomes, we characterized NR4A1 as a stress-inducible modifier of mitochondrial energetic competence and dendritic spine number in PFC. NR4A1 acted as a transcription factor for changing the expression of target genes previously involved in mitochondrial uncoupling, AMP-activated protein kinase activation, and synaptic growth. Maintenance of NR4A1 activity by chronic stress played a critical role in the regressive synaptic organization in PFC of mouse models of stress (male only). Knockdown, dominant-negative approach, and knockout of Nr4a1 in mice and rats (male only) protected pyramidal neurons against the adverse effects of chronic stress. In human PFC tissues of men and women, high levels of the transcriptionally active NR4A1 correlated with measures of synaptic loss and cognitive impairment. In the context of chronic stress, prolonged expression and activity of NR4A1 may lead to responses of mitochondria and synaptic connectivity that do not match environmental demand, resulting in circuit malfunction between PFC and other brain regions, constituting a pathological feature across disorders.SIGNIFICANCE STATEMENT The bioenergetic cost of chronic stress is too high to be sustainable by pyramidal prefrontal neurons. Cellular checkpoints have evolved to adjust the responses of mitochondria and synapses to the buildup of chronic stress. NR4A1 plays such a role by controlling the energetic competence of mitochondria with respect to synapse number. As an immediate-early gene, Nr4a1 promotes neuronal plasticity, but sustained expression or activity can be detrimental. NR4A1 expression and activity is sustained by chronic stress in animal models and in human studies of neuropathologies sensitive to the buildup of chronic stress. Therefore, antagonism of NR4A1 is a promising avenue for preventing the regressive synaptic reorganization in cortical systems in the context of chronic stress.


Subject(s)
Mitochondria/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Prefrontal Cortex/physiopathology , Stress, Psychological/physiopathology , Synapses/metabolism , Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Count , Chronic Disease , Cognition Disorders/etiology , Cognition Disorders/psychology , Dendritic Spines , Female , Gene Expression Regulation/genetics , Hindlimb Suspension , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics , Prefrontal Cortex/cytology , Pyramidal Cells/physiology , Rats , Stress, Psychological/psychology
10.
Neurotox Res ; 25(1): 124-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24277157

ABSTRACT

Nuclear receptors (Nurs) represent a large family of gene expression regulating proteins. Gathering evidence indicates an important role for Nurs as transcription factors in dopamine neurotransmission. Nur77, a member of the Nur superfamily, plays a role in mediating the effects of antiparkinsonian and neuroleptic drugs. Besides, Nur77 survival and apoptotic roles depend largely on its subcellular localization. Estrogens are known for their neuroprotective properties, as demonstrated in animal and clinical studies. However, their action on Nur77 translocation pertaining to neuroprotection has not been investigated yet. The aim of our study was to perform a kinetic study on the effect of neurotoxic 6-hydroxydopamine (6-OHDA) and 17ß-estradiol (E2) on the subcellular localization of Nur77 with reference to the modulation of apoptosis in PC12 cells. Our results demonstrate that E2 administration alone does not affect Nur77 cytoplasmic/nuclear ratio, mRNA levels, or apoptosis in PC12 cells. The neurotoxin 6-OHDA significantly enhances cytoplasmic localization of Nur77 after merely 3 h, while precipitating apoptosis. 6-OHDA also increases Nur77 transcription, which could partly explain the rise in cytoplasmic localization of the protein. Finally, treatment with both E2 and 6-OHDA delays Nur77 accumulation in the cytoplasm and delays cell death for a few hours in our cellular paradigm. Pre-treatment with E2 does not alter the increase in levels of Nur77 mRNA produced by 6-OHDA, suggesting that a raise in nuclear translocation is likely responsible for the stabilization of the cytoplasmic/nuclear ratio until 6 h. These results suggest an intriguing cooperation between E2 and Nur77 toward cellular fate guidance.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/metabolism , Cytoplasm/metabolism , Estradiol/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Nucleus/drug effects , Cells, Cultured , Cytoplasm/drug effects , Oxidopamine/toxicity , PC12 Cells , Rats
11.
Neurochem Int ; 61(7): 1160-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22940695

ABSTRACT

After chronic use of l-3,4-dihydroxyphenylalanine (l-DOPA), most Parkinson's disease (PD) patients suffer from its side effects, especially motor complications called l-DOPA-induced dyskinesia (LID). 5-HT(1A) agonists were tested to treat LID but many were reported to worsen parkinsonism. In this study, we evaluated changes in concentration of serotonin and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) and of 5-HT(1A) receptors in control monkeys, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkeys, dyskinetic MPTP monkeys treated chronically with l-DOPA, low dyskinetic MPTP monkeys treated with l-DOPA and drugs of various pharmacological activities: Ro 61-8048 (an inhibitor of kynurenine hydroxylase) or docosahexaenoic acid (DHA) and dyskinetic MPTP monkeys treated with l-DOPA+naltrexone (an opioid receptor antagonist). Striatal serotonin concentrations were reduced in MPTP monkeys compared to controls. Higher striatal 5-HIAA/serotonin concentration ratios in l-DOPA-treated monkeys compared to untreated monkeys suggest an intense activity of serotonin axon terminals but this value was similar in dyskinetic and nondyskinetic animals treated with or without adjunct treatment with l-DOPA. As measured by autoradiography with [(3)H]8-hydroxy-2-(di-n-propyl) aminotetralin (8-OH-DPAT), a decrease of 5-HT(1A) receptor specific binding was observed in the posterior/dorsal region of the anterior cingulate gyrus and posterior/ventral area of the superior frontal gyrus of MPTP monkeys compared to controls. An increase of 5-HT(1A) receptor specific binding was observed in the hippocampus of MPTP monkeys treated with l-DOPA regardless to their adjunct treatment. Cortical 5-HT(1A) receptor specific binding was increased in the l-DOPA-treated MPTP monkeys alone or with DHA or naltrexone and this increase was prevented in low dyskinetic MPTP monkeys treated with l-DOPA and Ro 61-8048. These results highlight the importance of 5-HT(1A) receptor alterations in treatment of PD with l-DOPA.


Subject(s)
Brain/drug effects , Levodopa/pharmacology , Parkinsonian Disorders/drug therapy , Receptors, Serotonin, 5-HT1/drug effects , Animals , Behavior, Animal , Brain/metabolism , Female , Macaca fascicularis , Parkinsonian Disorders/metabolism
12.
Front Pharmacol ; 3: 153, 2012.
Article in English | MEDLINE | ID: mdl-22912617

ABSTRACT

Dopamine D(2) receptor antagonists modulate gene transcription in the striatum. However, the molecular mechanism underlying this effect remains elusive. Here we used the expression of Nur77, a transcription factor of the orphan nuclear receptor family, as readout to explore the role of dopamine, glutamate, and adenosine receptors in the effect of a dopamine D(2) antagonist in the striatum. First, we investigated D(2) antagonist-induced Nur77 mRNA in D(2L) receptor knockout mice. Surprisingly, deletion of the D(2L) receptor isoform did not reduce eticlopride-induced upregulation of Nur77 mRNA levels in the striatum. Next, we tested if an ibotenic acid-induced cortical lesion could block the effect of eticlopride on Nur77 expression. Cortical lesions strongly reduced eticlopride-induced striatal upregulation of Nur77 mRNA. Then, we investigated if glutamatergic neurotransmission could modulate eticlopride-induced Nur77 expression. A combination of a metabotropic glutamate type 5 (mGlu5) and adenosine A(2A) receptor antagonists abolished eticlopride-induced upregulation of Nur77 mRNA levels in the striatum. Direct modulation of Nur77 expression by striatal glutamate and adenosine receptors was confirmed using corticostriatal organotypic cultures. Taken together, these results indicate that blockade of postsynaptic D(2) receptors is not sufficient to trigger striatal transcriptional activity and that interaction with corticostriatal presynaptic D(2) receptors and subsequent activation of postsynaptic glutamate and adenosine receptors in the striatum is required. Thus, these results uncover an unappreciated role of presynaptic D(2) heteroreceptors and support a prominent role of glutamate in the effect of D(2) antagonists.

13.
Int J Neuropsychopharmacol ; 15(4): 509-21, 2012 May.
Article in English | MEDLINE | ID: mdl-21524335

ABSTRACT

Different patterns of expression of the transcription factors of Nur77 and Nor-1 are induced following acute administration of typical and atypical antipsychotic drugs. The pharmacological profile of atypical antipsychotics suggests that serotonergic and/or adrenergic receptors might contribute to these reported differences. In order to test this possibility, we examined the abilities of serotonin 5-HT(1A) and 5-HT(2A/2C), and α1- and α2-adrenergic receptor drugs to modify the pattern of Nur77 (NR4A1) and Nor-1 (NR4A3) mRNA expression induced by haloperidol. Various groups of mice were treated with either saline, DOI, a 5-HT(2A/2C) agonist, MDL11939, a 5-HT(2A) antagonist, 8-OH-DPAT, a 5-HT(1A) agonist, prazosin, an α1-adrenergic antagonist and idazoxan, an α2-adrenergic antagonist, alone or in combination with haloperidol. The 5-HT(2A/2C) agonist DOI alone significantly increased Nur77 expression in the medial striatum and nucleus accumbens. DOI reduced Nor-1 expression, while MDL11939 increased the expression of this transcript in the cortex. Prazosin reduced Nur77 expression in the dorsal striatum and nucleus accumbens. Interestingly, 8-OH-DPAT and MDL11939 partially prevented haloperidol-induced Nur77 up-regulation, while MDL11939 completely abolished Nor-1 expression in the striatum. In addition, MDL11939 decreased haloperidol-induced Nur77 and Nor-1 mRNA levels in the ventral tegmental area. On the contrary, idazoxan (α2 antagonist) consistently potentiated haloperidol-induced Nur77, but not Nor-1 mRNA levels in the striatum, whereas prazosin (α1 antagonist) remained without effect. Taken together, these results show the ability of a 5-HT(1A) agonist or a 5-HT(2A) antagonist to reduce haloperidol-induced Nur77 and Nor-1 striatal expression, suggesting that these serotonin receptor subtypes participate in the differential pattern of gene expression induced by typical and atypical antipsychotic drugs.


Subject(s)
Adrenergic Agents/pharmacology , Brain/drug effects , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Serotonin Agents/pharmacology , Animals , Brain/anatomy & histology , DNA-Binding Proteins/genetics , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Haloperidol/pharmacology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , RNA, Messenger/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics
14.
J Neurochem ; 114(6): 1651-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20569301

ABSTRACT

Cystamine has shown significant neuroprotective properties in preclinical studies of Parkinson's disease (PD) and Huntington's disease (HD). Cysteamine, its FDA-approved reduced form, is scheduled to be tested for clinical efficacy in HD patients. Here, we studied the key cystamine metabolites, namely cysteamine, hypotaurine and taurine, as well as cysteine, in order to identify which one is more distinctively responsible for the neuroprotective action of cystamine. After a single administration of cystamine (10, 50 or 200 mg/kg), naïve mice were perfused with phosphate-buffered saline (PBS) at 1, 3, 12, 24 or 48 h post-injection and brain and plasma samples were analyzed by two distinct HPLC methods. Although plasma levels remained under the detection threshold, significant increases in cysteamine brain levels were detected with the 50 and 200 mg/kg doses in mice perfused 1 and 3 h following cystamine injection. To further assess cysteamine as the candidate molecule for pre-clinical and clinical trials in PD, we evaluated its capacity to cross the blood brain barrier. Using an in situ cerebral perfusion technique, we determined that the brain transport coefficient (Clup) of cysteamine (259 µM) was 0.15 ± 0.02 µL/g/s and was increased up to 0.34 ± 0.07 µL/g/s when co-perfused in the presence of cysteine. Taken together, these results strongly suggest that cysteamine is the neuroactive metabolite of cystamine and may further support its therapeutic use in neurodegenerative diseases, particularly in HD and PD.


Subject(s)
Brain/metabolism , Cystamine/metabolism , Neuroprotective Agents/metabolism , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Cystamine/pharmacology , Cysteamine/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Taurine/analogs & derivatives , Taurine/metabolism
15.
Neuropharmacology ; 58(1): 286-96, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19576910

ABSTRACT

Dopamine denervation in Parkinson's disease and repeated Levodopa (L-DOPA) administration that induces dyskinesias are associated with an enhancement of basal ganglia neuropeptide transmission. Various adjunct non-dopaminergic treatments to Levodopa were shown to reduce and/or prevent dyskinesias. The aim of this study was to seek if non-dopaminergic drug treatments to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) lesioned monkeys combined with L-DOPA to prevent dyskinesia were associated with changes of striatal neuropeptides. Chronic treatment with Ro 61-8048 a kynurenine hydroxylase inhibitor, docosahexaenoic acid (DHA) a polyunsaturated fatty acid (omega-3), naltrexone an opioidergic antagonist and CI-1041 an N-methyl-D-aspartate (NMDA) glutamate receptor antagonist with L-DOPA prevented dyskinesias to various extents except naltrexone whereas all MPTP monkeys treated with L-DOPA alone developed dyskinesias. Striatal preproenkephalin (PPE), preprodynorphin (PPD) and preprotachykinin A (PPT-A) mRNA levels were measured by in situ hybridization. An increase of PPE and PPD mRNA levels was observed in anterior caudate nucleus of L-DOPA treated MPTP monkeys compared to controls and to Saline-treated MPTP monkeys whereas PPT-A mRNA levels were unchanged. Striatal PPE and PPD mRNA levels remained elevated in L-DOPA plus naltrexone-treated MPTP monkeys, while co-treatment with DHA, CI-1041 or Ro 61-8048 prevented their increase to various extents. Maximal dyskinesias scores of MPTP monkeys correlated significantly with striatal PPE and PPD mRNA levels but not with PPT-A mRNA levels. These results show that drugs displaying a wide range of pharmacological activities can modulate L-DOPA induced dyskinesias and this activity is correlated with striatal PPD and PPE mRNA levels suggesting a convergent mechanism.


Subject(s)
Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Corpus Striatum/metabolism , Dyskinesia, Drug-Induced , Levodopa/adverse effects , Neuropeptides/metabolism , Animals , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Cocaine/analogs & derivatives , Cocaine/metabolism , Corpus Striatum/drug effects , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Dopamine/metabolism , Dopamine Uptake Inhibitors/metabolism , Dynorphins/genetics , Dynorphins/metabolism , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/pathology , Enkephalins/genetics , Enkephalins/metabolism , Female , Iodine Isotopes/metabolism , Macaca fascicularis , Naltrexone/pharmacology , Naltrexone/therapeutic use , Ovariectomy , Parkinsonian Disorders/drug therapy , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tachykinins/genetics , Tachykinins/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use , Time Factors
16.
J Neurosci ; 29(50): 15923-32, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016108

ABSTRACT

Transcription factors involved in the specification and differentiation of neurons often continue to be expressed in the adult brain, but remarkably little is known about their late functions. Nurr1, one such transcription factor, is essential for early differentiation of midbrain dopamine (mDA) neurons but continues to be expressed into adulthood. In Parkinson's disease, Nurr1 expression is diminished and mutations in the Nurr1 gene have been identified in rare cases of disease; however, the significance of these observations remains unclear. Here, a mouse strain for conditional targeting of the Nurr1 gene was generated, and Nurr1 was ablated either at late stages of mDA neuron development by crossing with mice carrying Cre under control of the dopamine transporter locus or in the adult brain by transduction of adeno-associated virus Cre-encoding vectors. Nurr1 deficiency in maturing mDA neurons resulted in rapid loss of striatal DA, loss of mDA neuron markers, and neuron degeneration. In contrast, a more slowly progressing loss of striatal DA and mDA neuron markers was observed after ablation in the adult brain. As in Parkinson's disease, neurons of the substantia nigra compacta were more vulnerable than cells in the ventral tegmental area when Nurr1 was ablated at late embryogenesis. The results show that developmental pathways play key roles for the maintenance of terminally differentiated neurons and suggest that disrupted function of Nurr1 and other developmental transcription factors may contribute to neurodegenerative disease.


Subject(s)
Mesencephalon/cytology , Mesencephalon/growth & development , Neurons/cytology , Neurons/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Age Factors , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Gene Targeting , Integrases/genetics , Mesencephalon/physiology , Mice , Mice, Transgenic , Neurogenesis/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Pregnancy
17.
Neurobiol Dis ; 36(1): 213-22, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19635563

ABSTRACT

We have previously shown that docosahexaenoic acid (DHA) significantly reduced L-Dopa-induced dyskinesia (LID) in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkeys (Samadi et al., Ann. Neurol. 59:282-288, 2006). In the present study, we measured for the first time mRNA levels of Nur77, an orphan nuclear receptor that participates to adaptive and/or aberrant dopamine-related behaviors, and retinoid X receptor gamma1 (RXRgamma1), a putative brain receptor for DHA and transcriptional partner of Nur77, in MPTP monkeys treated with L-Dopa and DHA. The RXRgamma1 mRNA is strongly expressed in monkey caudate nucleus and putamen, but no change in levels of RXRgamma1 was observed following MPTP and L-Dopa treatments. On the other hand, denervation reduced Nur77 mRNA levels, whereas chronic L-Dopa treatment strongly induced Nur77 transcripts. These modulations are taking place in substance P positive cells and are associated with both caudate-putamen matrix and striosome compartments. Interestingly, combination of L-Dopa with DHA further increases Nur77 mRNA levels in the anterior caudate-putamen, and mainly in striosomes. This is accompanied by a significant inverse correlation between Nur77 mRNA levels and dyskinetic scores. Taken together, our results show that Nur77 expression is modulated following dopamine denervation and chronic L-Dopa therapy in a non-human primate model of Parkinson's disease, and suggest that strong modulation of Nur77 expression might be linked to a reduced risk to develop LIDs.


Subject(s)
Antiparkinson Agents/adverse effects , Docosahexaenoic Acids/pharmacology , Dyskinesia, Drug-Induced , Levodopa/adverse effects , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , RNA, Messenger/metabolism , Acetylcholinesterase/metabolism , Analysis of Variance , Animals , Autoradiography/methods , Brain/drug effects , Brain/metabolism , Cocaine/analogs & derivatives , Cocaine/metabolism , Disease Models, Animal , Drug Interactions , Dyskinesia, Drug-Induced/drug therapy , Female , Iodine Isotopes/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Macaca fascicularis , Protein Binding/drug effects , Statistics as Topic
18.
J Neurochem ; 109(5): 1469-82, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19457163

ABSTRACT

Animal models are invaluable tools to study neurodegenerative disorders but a general consensus on the most accurate rodent model of Parkinson's disease has not been reached. Here, we examined how different methods of MPTP administration influence the degeneration of the dopaminergic (DA) system. Adult male C57BL/6 mice were treated with the same cumulative dose of MPTP following four distinct procedures: (i) subacute i.p. injections; (ii) 28-day chronic s.c. infusion; (iii) 28-day chronic i.p. infusion; and (iv) 14-day chronic i.p. infusion. Subacute MPTP treatment significantly affected all aspects of the DA system within the nigral and striatal territories. In contrast, the 28-day chronic s.c. infusion did not significantly alter any components of the DA system. The 28- and 14-day chronic i.p. infusions induced loss of tyrosine hydroxylase (TH)-positive cells correlated with a decrease in Nurr1 mRNA levels, but no significant decrease in the density of TH striatal fibers. Importantly, however, only the 14-day chronic MPTP i.p. infusion protocol promoted the formation of neuronal inclusions as noted by the expression of alpha-synuclein protein within the cytoplasm of TH nigral neurons. Overall, we found that the 14-day chronic MPTP i.p. infusion reproduces more accurately the pathological characteristics of early stage Parkinson's disease.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Dopamine/metabolism , Intranuclear Inclusion Bodies/metabolism , MPTP Poisoning/metabolism , Nerve Degeneration/metabolism , alpha-Synuclein/metabolism , Animals , Corpus Striatum/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drug Administration Routes , Drug Administration Schedule , Exploratory Behavior/drug effects , MPTP Poisoning/complications , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Nerve Degeneration/etiology , Nuclear Receptor Subfamily 4, Group A, Member 2 , Substantia Nigra/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Psychopharmacology (Berl) ; 202(4): 635-48, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18843482

ABSTRACT

INTRODUCTION: The major substrate underlying amphetamine (AMPH)-induced locomotor activity is associated with dopamine forebrain circuits. Brain regions associated with AMPH-induced locomotor activity express high levels of retinoid receptors. However, the role of these transcription factors in dopamine-mediated effects remains poorly understood. Two nuclear receptor families, the retinoic acid receptors (RAR) and the retinoid X receptors (RXR), transduce retinoic acid signal. RARs are specifically involved in retinoid signaling, whereas RXRs also participate in other signaling pathways as partners for other nuclear receptors such as Nur77, an orphan member of the nuclear receptor family expresses in dopamine system. MATERIALS AND METHODS: To explore the role of retinoid receptors and Nur77 in AMPH-induced locomotor activity, we administered selective retinoid receptor drugs in combination with AMPH in adult wild-type and Nur77-deficient mice. At a low dose, AMPH similarly increased ambulatory activity in wild-type and Nur77-deficient mice, while it did not alter non-ambulatory activity. RESULTS AND DISCUSSION: At a high dose, AMPH did not alter ambulatory activity anymore, while non-ambulatory activity strongly increased in wild-type mice. Nur77-deficient mice still displayed a higher ambulatory activity with no change in non-ambulatory activity. HX531, a synthetic RXR antagonist, blocks AMPH-induced ambulatory activity, whereas RAR drugs tested remained without effect. Interestingly, the effect of HX531 was abolished in Nur77-deficient mice, suggesting that this orphan nuclear receptor is essential for the action of the RXR drug. CONCLUSION: This study shows that RXR and Nur77 participate in AMPH-induced locomotor activity and prompts for further investigations on the role of Nur77 and RXR in addiction and reward-related behaviors.


Subject(s)
Amphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , DNA-Binding Proteins/genetics , Motor Activity/drug effects , Motor Activity/genetics , Receptors, Steroid/genetics , Retinoid X Receptors/genetics , Animals , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , DNA-Binding Proteins/drug effects , Dibenzazepines/pharmacology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neostriatum/drug effects , Neostriatum/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Receptors, Dopamine D2/drug effects , Receptors, Steroid/drug effects , Retinoid X Receptors/antagonists & inhibitors
20.
Eur J Neurosci ; 28(11): 2231-42, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19019202

ABSTRACT

A role of serotonin receptors (5-HTRs) in spinal rhythmogenesis has been proposed several years ago based mainly upon data showing that bath-applied 5-HT could elicit locomotor-like rhythms in in vitro isolated spinal cord preparations. Such a role was partially confirmed in vivo after revealing that systemically administered 5-HTR(2) agonists, such as quipazine, could induce some locomotor-like movements (LM) in completely spinal cord-transected (Tx) rodents. However, given the limited binding selectivity of currently available 5-HTR(2) agonists, it has remained difficult to determine clearly if one receptor subtype is specifically associated with LM induction. In situ hybridization, data using tissues from L1-L2 spinal cord segments, where critical locomotor network elements have been identified in mice, revealed greater 5-HTR(2A) mRNA levels in low-thoracic Tx than non-Tx animals. This expression level remained elevated for several days, specifically in the lateral intermediate zone, where peak values were detected at 1 week post-Tx and returned to normal at 3 weeks post-Tx. Behavioral and kinematic analyses revealed quipazine-induced LM in 1-week Tx mice either non-pretreated or pretreated with selective 5-HTR(2B) and/or 5-HTR(2C) antagonists. In contrast, LM completely failed to be induced by quipazine in animals pretreated with selective 5-HTR(2A) antagonists. Altogether, these results provide strong evidence suggesting that 5-HTR(2A) are specifically associated with spinal locomotor network activation and LM generation induced by quipazine in Tx animals. These findings may contribute to design drug treatments aimed at promoting locomotor function recovery in chronic spinal cord-injured patients.


Subject(s)
Paralysis/metabolism , Quipazine/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Animals , Disease Models, Animal , Hindlimb/innervation , Hindlimb/physiopathology , Male , Mice , Motor Activity/drug effects , Motor Activity/physiology , Movement/drug effects , Movement/physiology , Nerve Net/drug effects , Nerve Net/metabolism , Paralysis/drug therapy , Paralysis/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2B/drug effects , Receptor, Serotonin, 5-HT2B/genetics , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Serotonin, 5-HT2C/drug effects , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Recovery of Function/drug effects , Recovery of Function/physiology , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Spinal Cord/drug effects , Spinal Cord/physiopathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Thoracic Vertebrae , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...