Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38858083

ABSTRACT

Decades of research have identified the pathological and pathophysiological hallmarks of Parkinson's disease (PD): profound deficit in brain dopamine and other monoamines, pathological α-synuclein aggregation, synaptic and neuronal network dysfunction, aberrant proteostasis, altered energy homeostasis, inflammation, and neuronal cell death. The purpose of this contribution is to present the phenocopy aspect, pathogenic, and etiologic nonhuman primate (NHP) models of PD to readers with limited prior knowledge of PD so that they are ready to start working on PD. How NHPs, the closest species to man on which we can model diseases, contribute to the knowledge progress and how these models represent an invaluable translational step in therapeutic development are highlighted.

2.
NPJ Parkinsons Dis ; 9(1): 135, 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37726343

ABSTRACT

Aggregation of α-synuclein (α-syn) is the cornerstone of neurodegenerative diseases termed synucleinopathies, which include Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB), and Multiple System Atrophy (MSA). These synucleinopathies are characterized by the deposit of aggregated α-syn in intracellular inclusions observable in neurons and glial cells. In PD and DLB, these aggregates, predominantly located in neurons, are called Lewy Bodies (LBs). These LBs are one of the pathological hallmarks of PD and DLB, alongside dopaminergic neuron loss in the substantia nigra. Previous studies have demonstrated the ability of PD patient-derived LB fractions to induce nigrostriatal neurodegeneration and α-syn pathology when injected into the striatum or the enteric nervous system of non-human primates. Here, we report the pathological consequences of injecting these LB fractions into the cortex of non-human primates. To this end, we inoculated mesencephalic PD patient-derived LB fractions into the prefrontal cortex of baboon monkeys terminated one year later. Extensive analyses were performed to evaluate pathological markers known to be affected in LB pathologies. We first assessed the hypothesized presence of phosphorylated α-syn at S129 (pSyn) in the prefrontal cortices. Second, we quantified the neuronal, microglial, and astrocytic cell survival in the same cortices. Third, we characterized these cortical LB injections' putative impact on the integrity of the nigrostriatal system. Overall, we observed pSyn accumulation around the injection site in the dorsal prefrontal cortex, in connected cortical regions, and further towards the striatum, suggesting α-syn pathological propagation. The pathology was also accompanied by neuronal loss in these prefrontal cortical regions and the caudate nucleus, without, however, loss of nigral dopamine neurons. In conclusion, this pilot study provides novel data demonstrating the toxicity of patient-derived extracts, their potential to propagate from the cortex to the striatum in non-human primates, and a possible primate model of DLB.

3.
Brain ; 145(3): 1001-1017, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35285474

ABSTRACT

Synucleinopathies encompass several neurodegenerative diseases, which include Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. These diseases are characterized by the deposit of α-synuclein aggregates in intracellular inclusions in neurons and glial cells. Unlike Parkinson's disease and dementia with Lewy bodies, where aggregates are predominantly neuronal, multiple system atrophy is associated with α-synuclein cytoplasmic inclusions in oligodendrocytes. Glial cytoplasmic inclusions are the pathological hallmark of multiple system atrophy and are associated with neuroinflammation, modest demyelination and, ultimately, neurodegeneration. To evaluate the possible pathogenic role of glial cytoplasmic inclusions, we inoculated glial cytoplasmic inclusion-containing brain fractions obtained from multiple system atrophy patients into the striatum of non-human primates. After a 2-year in vivo phase, extensive histochemical and biochemical analyses were performed on the whole brain. We found loss of both nigral dopamine neurons and striatal medium spiny neurons, as well as loss of oligodendrocytes in the same regions, which are characteristics of multiple system atrophy. Furthermore, demyelination, neuroinflammation and α-synuclein pathology were also observed. These results show that the α-synuclein species in multiple system atrophy-derived glial cytoplasmic inclusions can induce a pathological process in non-human primates, including nigrostriatal and striatofugal neurodegeneration, oligodendroglial cell loss, synucleinopathy and gliosis. The present data pave the way for using this experimental model for MSA research and therapeutic development.


Subject(s)
Demyelinating Diseases , Lewy Body Disease , Multiple System Atrophy , Parkinson Disease , Synucleinopathies , Animals , Brain/pathology , Demyelinating Diseases/pathology , Humans , Inclusion Bodies/metabolism , Lewy Body Disease/pathology , Multiple System Atrophy/pathology , Parkinson Disease/pathology , alpha-Synuclein/metabolism
4.
J Med Chem ; 64(12): 8246-8262, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34107215

ABSTRACT

Adenosine A1/A2A receptors (A1R/A2AR) represent targets in nondopaminergic treatment of motor disorders such as Parkinson's disease (PD). As an innovative strategy, multitargeting ligands (MTLs) were developed to achieve comprehensive PD therapies simultaneously addressing comorbid symptoms such as sleep disruption. Recognizing the wake-promoting capacity of histamine H3 receptor (H3R) antagonists in combination with the "caffeine-like effects" of A1R/A2AR antagonists, we designed A1R/A2AR/H3R MTLs, where a piperidino-/pyrrolidino(propyloxy)phenyl H3R pharmacophore was introduced with overlap into an adenosine antagonist arylindenopyrimidine core. These MTLs showed distinct receptor binding profiles with overall nanomolar H3R affinities (Ki < 55 nM). Compound 4 (ST-2001, Ki (A1R) = 11.5 nM, Ki (A2AR) = 7.25 nM) and 12 (ST-1992, Ki (A1R) = 11.2 nM, Ki (A2AR) = 4.01 nM) were evaluated in vivo. l-DOPA-induced dyskinesia was improved after administration of compound 4 (1 mg kg-1, i.p. rats). Compound 12 (2 mg kg-1, p.o. mice) increased wakefulness representing novel pharmacological tools for PD therapy.


Subject(s)
Adenosine A1 Receptor Antagonists/therapeutic use , Adenosine A2 Receptor Antagonists/therapeutic use , Histamine H3 Antagonists/therapeutic use , Parkinson Disease, Secondary/drug therapy , Adenosine A1 Receptor Antagonists/chemical synthesis , Adenosine A1 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/metabolism , Animals , Dyskinesias/drug therapy , Histamine H3 Antagonists/chemical synthesis , Histamine H3 Antagonists/metabolism , Humans , Levodopa/pharmacology , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Piperidines/chemical synthesis , Piperidines/metabolism , Piperidines/therapeutic use , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Pyrrolidines/chemical synthesis , Pyrrolidines/metabolism , Pyrrolidines/therapeutic use , Rats, Sprague-Dawley , Receptor, Adenosine A2A/metabolism , Receptors, Histamine H3/metabolism , Wakefulness/drug effects
5.
Heliyon ; 7(2): e06302, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33665452

ABSTRACT

Woodchuck Hepatitis Virus Post-transcriptional Regulatory Element (WPRE) is thought to enhance transgene expression of target genes delivered by adeno-associated viral (AAV) vectors. This study assessed the protein expression of α-synuclein, phosphorylated α-synuclein at Serine 129, extent of nigrostriatal degeneration as well as subsequent behavioral deficits induced by unilateral intranigral stereotactic injection in male adult C57BL/6J mice of an AAV2/9 expressing A53T human α-synuclein under the control of the synapsin promoter in presence or absence of the WPRE. The presence of WPRE enabled to achieve greater nigrostriatal degeneration and synucleinopathy which was concomitant with worsened forelimb use asymmetry. This work refines a mouse Parkinson's disease model in which anatomo-pathology is related to behavioral deficits.

6.
Neurobiol Dis ; 150: 105255, 2021 03.
Article in English | MEDLINE | ID: mdl-33421564

ABSTRACT

In Parkinson's disease, synucleinopathy is hypothesized to spread from the enteric nervous system, via the vagus nerve, to the central nervous system. Recent evidences collected in non-human primates challenge however the hypothesis of a transmission of α-synuclein (α-syn) pathology through the vagus nerve. Would the hypothesis whereby the bloodstream acts as a route for long-distance transmission of pathological α-syn hold true, an inter-individual transmission of synucleinopathy could occur via blood contact. Here, we used a parabiosis approach to join the circulatory systems of wild type and GFP transgenic C57BL/6 J mice, for which one of the partners parabiont received a stereotaxic intranigral injection of patient-derived α-syn aggregates. While the Lewy Body-receiving mice exhibited a loss of dopamine neurons and an increase in nigral S129 phosphorylated α-syn immunoreactivity, their parabiotic bloodstream-sharing partners did not show any trend for a lesion or change in S129 phosphorylated-α-syn levels. Altogether, our study suggests that, in the patient-derived α-synuclein aggregates-injected mouse model and within the selected time frame, the disease is not "transmitted" through the bloodstream.


Subject(s)
Lewy Bodies/transplantation , Neostriatum/pathology , Neurons/pathology , Parabiosis , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Substantia Nigra/pathology , alpha-Synuclein/metabolism , Animals , Mice , Mice, Transgenic , Stereotaxic Techniques , alpha-Synuclein/blood
7.
Cereb Cortex ; 30(12): 6469-6480, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32776091

ABSTRACT

OBJECTIVE: Whether the basal ganglia are involved in the cortical synchronization during focal seizures is still an open question. In the present study, we proposed to synchronize cortico-striatal activities acutely inducing striatal disinhibition, performing GABA-antagonist injections within the putamen in primates. METHOD: Experiments were performed on three fascicularis monkeys. During each experimental session, low volumes of bicuculline (0.5-4 µL) were injected at a slow rate of 1 µL/min. Spontaneous behavioral changes were classified according to Racine's scale modified for primates. These induced motor behaviors were correlated with electromyographic, electroencephalographic, and putaminal and pallidal local field potentials changes in activity. RESULTS: acute striatal desinhibition induced focal motor seizures. Seizures were closely linked to cortical epileptic activity synchronized with a striatal paroxysmal activity. These changes in striatal activity preceded the cortical epileptic activity and the induced myoclonia, and both cortical and subcortical activities were coherently synchronized during generalized seizures. INTERPRETATION: Our results strongly suggest the role of the sensorimotor striatum in the regulation and synchronization of cortical excitability. These dramatic changes in the activity of this "gating" pathway might influence seizure susceptibility by modulating the threshold for the initiation of focal motor seizures.


Subject(s)
Cerebral Cortex/physiopathology , Cortical Synchronization , Putamen/physiopathology , Seizures/physiopathology , Animals , Bicuculline/administration & dosage , Female , GABA-A Receptor Antagonists/administration & dosage , Macaca fascicularis , Male , Neural Pathways/physiopathology , Putamen/drug effects , Rats, Sprague-Dawley , Seizures/etiology
8.
Brain ; 143(5): 1462-1475, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32380543

ABSTRACT

In Parkinson's disease, synucleinopathy is hypothesized to spread from the enteric nervous system, via the vagus nerve, to the CNS. Here, we compare, in baboon monkeys, the pathological consequences of either intrastriatal or enteric injection of α-synuclein-containing Lewy body extracts from patients with Parkinson's disease. This study shows that patient-derived α-synuclein aggregates are able to induce nigrostriatal lesions and enteric nervous system pathology after either enteric or striatal injection in a non-human primate model. This finding suggests that the progression of α-synuclein pathology might be either caudo-rostral or rostro-caudal, varying between patients and disease subtypes. In addition, we report that α-synuclein pathological lesions were not found in the vagal nerve in our experimental setting. This study does not support the hypothesis of a transmission of α-synuclein pathology through the vagus nerve and the dorsal motor nucleus of the vagus. Instead, our results suggest a possible systemic mechanism in which the general circulation would act as a route for long-distance bidirectional transmission of endogenous α-synuclein between the enteric and the central nervous systems. Taken together, our study provides invaluable primate data exploring the role of the gut-brain axis in the initiation and propagation of Parkinson's disease pathology and should open the door to the development and testing of new therapeutic approaches aimed at interfering with the development of sporadic Parkinson's disease.


Subject(s)
Brain/pathology , Neuroimmunomodulation/physiology , Parkinson Disease/physiopathology , Vagus Nerve/pathology , alpha-Synuclein/toxicity , Aged , Animals , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Female , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Male , Papio , alpha-Synuclein/administration & dosage
9.
Mov Disord ; 33(10): 1632-1642, 2018 10.
Article in English | MEDLINE | ID: mdl-29756234

ABSTRACT

Parkinson's disease motor symptoms are treated with levodopa, but long-term treatment leads to disabling dyskinesia. Altered synaptic transmission and maladaptive plasticity of corticostriatal glutamatergic projections play a critical role in the pathophysiology of dyskinesia. Because the noble gas xenon inhibits excitatory glutamatergic signaling, primarily through allosteric antagonism of the N-methyl-d-aspartate receptors, we aimed to test its putative antidyskinetic capabilities. We first studied the direct effect of xenon gas exposure on corticostriatal plasticity in a murine model of levodopa-induced dyskinesia We then studied the impact of xenon inhalation on behavioral dyskinetic manifestations in the gold-standard rat and primate models of PD and levodopa-induced dyskinesia. Last, we studied the effect of xenon inhalation on axial gait and posture deficits in a primate model of PD with levodopa-induced dyskinesia. This study shows that xenon gas exposure (1) normalized synaptic transmission and reversed maladaptive plasticity of corticostriatal glutamatergic projections associated with levodopa-induced dyskinesia, (2) ameliorated dyskinesia in rat and nonhuman primate models of PD and dyskinesia, and (3) improved gait performance in a nonhuman primate model of PD. These results pave the way for clinical testing of this unconventional but safe approach. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Parkinsonian Disorders/drug therapy , Xenon/therapeutic use , Administration, Inhalation , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/etiology , Gait Disorders, Neurologic/drug therapy , Gait Disorders, Neurologic/etiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , MPTP Poisoning/drug therapy , Mice , Mice, Transgenic , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/complications , Rats , Sensation Disorders/drug therapy , Sensation Disorders/etiology , Sympatholytics/toxicity , Time Factors
10.
Animal Model Exp Med ; 1(4): 314-321, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30891581

ABSTRACT

BACKGROUND: The brain bioavailability of novel small molecules developed to address central nervous system disease is classically documented through ex vivo or in vivo analyses conducted in rodent models. Data acquired in rodent models are, however, not easily transferrable to human as the pharmacokinetic and pharmacodynamics profiles of the species are quite different. METHODS: Using drugs selected for their differential transport across the blood-brain barrier, we here demonstrate the feasibility of brain microdialysis in normal vigil macaque monkey by measuring brain extracellular fluid bioavailability of carbamazepine, digoxin, oxycodone, and quinidine. RESULTS: All drugs, but digoxin, were found in dialysate samples. Drugs that are substrate of P-glycoprotein show a difference of bioavailability or brain pharmacokinetic parameters between rodents and primates. CONCLUSION: Data suggest that brain microdialysis in vigil macaque monkey, the species of choice for classic pharmacokinetic/pharmacodynamics studies could help predicting human brain bioavailability of a small molecule depending on the protein involved in the efflux transport from the brain.

11.
Acta Neuropathol Commun ; 3: 46, 2015 Jul 25.
Article in English | MEDLINE | ID: mdl-26205255

ABSTRACT

INTRODUCTION: Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons as well as the presence of proteinaceous inclusions named Lewy bodies. α-synuclein (α-syn) is a major constituent of Lewy bodies, and the first disease-causing protein characterized in PD. Several α-syn-based animal models of PD have been developed to investigate the pathophysiology of PD, but none of them recapitulate the full picture of the disease. Ageing is the most compelling and major risk factor for developing PD but its impact on α-syn toxicity remains however unexplored. In this study, we developed and exploited a recombinant adeno-associated viral (AAV) vector of serotype 9 overexpressing mutated α-syn to elucidate the influence of ageing on the dynamics of PD-related neurodegeneration associated with α-syn pathology in different mammalian species. RESULTS: Identical AAV pseudotype 2/9 vectors carrying the DNA for human mutant p.A53T α-syn were injected into the substantia nigra to induce neurodegeneration and synucleinopathy in mice, rats and monkeys. Rats were used first to validate the ability of this serotype to replicate α-syn pathology and second to investigate the relationship between the kinetics of α-syn-induced nigrostriatal degeneration and the progressive onset of motor dysfunctions, strikingly reminiscent of the impairments observed in PD patients. In mice, AAV2/9-hα-syn injection into the substantia nigra was associated with accumulation of α-syn and phosphorylated hα-syn, regardless of mouse strain. However, phenotypic mutants with either accelerated senescence or resistance to senescence did not display differential susceptibility to hα-syn overexpression. Of note, p-α-syn levels correlated with nigrostriatal degeneration in mice. In monkeys, hα-syn-induced degeneration of the nigrostriatal pathway was not affected by the age of the animals. Unlike mice, monkeys did not exhibit correlations between levels of phosphorylated α-syn and neurodegeneration. CONCLUSIONS: In conclusion, AAV2/9-mediated hα-syn induces robust nigrostriatal neurodegeneration in mice, rats and monkeys, allowing translational comparisons among species. Ageing, however, neither exacerbated nigrostriatal neurodegeneration nor α-syn pathology per se. Our unprecedented multi-species investigation thus favours the multiple-hit hypothesis for PD wherein ageing would merely be an aggravating, additive, factor superimposed upon an independent disease process.


Subject(s)
Aging , MPTP Poisoning/pathology , Striatonigral Degeneration/pathology , Substantia Nigra/metabolism , alpha-Synuclein/metabolism , Animals , Biomechanical Phenomena , Callithrix , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , MPTP Poisoning/chemically induced , Mice , Motor Activity , Principal Component Analysis , Psychomotor Performance/physiology , Rats , Striatonigral Degeneration/etiology , Time Factors , Transduction, Genetic , Tyrosine 3-Monooxygenase/metabolism
12.
Sci Rep ; 4: 3730, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24429495

ABSTRACT

L-dopa remains the mainstay treatment for Parkinson's disease (PD), although in later stages, treatment is complicated by L-dopa-induced dyskinesias (LID). Current evidence links LID to excessive striatal L-dopa-derived dopamine (DA) release, while the possibility of a direct involvement of L-dopa itself in LID has been largely ignored. Here we show that L-dopa can alter basal ganglia activity and produce LID without enhancing striatal DA release in parkinsonian non-human primates. These data may have therapeutic implications for the management of advanced PD since they suggest that LID could result from diverse mechanisms of action of L-dopa.


Subject(s)
Dopamine Agents/adverse effects , Dyskinesias/etiology , Levodopa/adverse effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Benserazide/adverse effects , Benserazide/pharmacokinetics , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Agents/pharmacokinetics , Drug Combinations , Dyskinesias/metabolism , Female , Levodopa/pharmacokinetics , Macaca mulatta , Male , Parkinson Disease/metabolism
13.
Neurobiol Dis ; 62: 179-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24103779

ABSTRACT

Long-term l-3,4-dihydroxyphenylalanine (l-DOPA) treatment in Parkinson's disease (PD) leads to l-DOPA-induced dyskinesia (LID), a condition thought to primarily involve the dopamine D1 receptor-expressing striatal medium spiny neurons. Activation of the D1 receptor results in increased expression of several molecular markers, in particular the members of the immediate-early gene (IEG) family, a class of genes rapidly transcribed in response to an external stimulus. However, several dopaminoceptive structures in the brain that are likely to be affected by the exogenously produced DA have received little attention although they might play a key role in mediating those l-DOPA-induced abnormal behaviours. ΔFosB, ARC, FRA2 and Zif268 IEGs expression patterns were thus characterised, using unbiased stereological methods, in the whole brain of dyskinetic and non-dyskinetic rats to identify brain nuclei displaying a transcriptional response specifically related to LID. Within the basal ganglia, the striatum and the substantia nigra pars reticulata showed an increased expression of all four IEGs in dyskinetic compared to non-dyskinetic rats. Outside the basal ganglia, there was a striking increased expression of the four IEGs in the motor cortex, the bed nucleus of the stria terminalis, the dorsal hippocampus, the pontine nuclei, the cuneiform nucleus and the pedunculopontine nuclei. Moreover, the zona incerta and the lateral habenula displayed an overexpression of ΔFosB, ARC and Zif268. Among these structures, the IEG expression in the striatum, the bed nucleus of the stria terminalis, the lateral habenula, the pontine nuclei and the cuneiform nucleus correlate with LID severity. These results illustrate a global transcriptional response to a dyskinetic state in the whole brain suggesting the possible involvement of these structures in LID.


Subject(s)
Antiparkinson Agents/toxicity , Basal Ganglia/metabolism , Brain/metabolism , Dyskinesia, Drug-Induced/metabolism , Immediate-Early Proteins/metabolism , Levodopa/toxicity , Animals , Cytoskeletal Proteins/metabolism , Early Growth Response Protein 1/metabolism , Fos-Related Antigen-2/metabolism , Male , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
14.
Neurosci Res ; 77(4): 242-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24135129

ABSTRACT

The serotonin system has emerged as a potential target for anti-dyskinetic therapy in Parkinson's disease. In fact, serotonin neurons can convert L-DOPA into dopamine, and mediate its synaptic release. However, they lack a feedback control mechanism able to regulate synaptic dopamine levels, which leads to un-physiological stimulation of post-synaptic striatal dopamine receptors. Accordingly, drugs able to dampen the activity of serotonin neurons can suppress L-DOPA-induced dyskinesia in animal models of Parkinson's disease. Here, we investigated the ability of the 5-HT1A/1B receptor agonist anpirtoline to counteract L-DOPA-induced dyskinesia in L-DOPA-primed 6-OHDA-lesioned rats and MPTP-treated macaques. Results suggest that anpirtoline dose-dependently reduced dyskinesia both in rats and monkeys; however, the effect in MPTP-treated macaques was accompanied by a worsening of the Parkinson's disease score at significantly effective doses (1.5 and 2.0mg/kg). At a lower dose (0.75mg/kg), anpirtoline markedly reduced dyskinesia in 4 out of 5 subjects, but statistical significance was prevented by the presence of a non-responsive subject. These results provide further evidence that the serotonin neurons contribute both to the pro-dyskinetic effect of L-DOPA and to its therapeutic efficacy in the rat and monkey models of Parkinson's disease.


Subject(s)
Dyskinesia, Drug-Induced/drug therapy , Piperidines/therapeutic use , Pyridines/therapeutic use , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Animals , Antiparkinson Agents/adverse effects , Female , Levodopa/adverse effects , Macaca fascicularis , Parkinsonian Disorders/drug therapy , Rats , Rats, Sprague-Dawley
15.
Mov Disord ; 28(8): 1088-96, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23389842

ABSTRACT

The serotonin (5-hydroxytryptamine [5HT]) system has recently emerged as an important player in the appearance of l-3,4-dihydroxyphenylalanine (levodopa [l-dopa])-induced dyskinesia in animal models of Parkinson's disease. In fact, dopamine released as a false transmitter from serotonin neurons appears to contribute to the pulsatile stimulation of dopamine receptors, leading to the appearance of the abnormal involuntary movements. Thus, drugs able to dampen the activity of serotonin neurons hold promise for the treatment of dyskinesia. The authors investigated the ability of the mixed 5-HT 1A/1B receptor agonist eltoprazine to counteract l-dopa-induced dyskinesia in 6-hydroxydopamine-lesioned rats and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaques. The data demonstrated that eltoprazine is extremely effective in suppressing dyskinesia in experimental models, although this effect was accompanied by a partial worsening of the therapeutic effect of l-dopa. Interestingly, eltoprazine was found to (synergistically) potentiate the antidyskinetic effect of amantadine. The current data indicated that eltoprazine is highly effective in counteracting dyskinesia in preclinical models. However, the partial worsening of the l-dopa effect observed after eltoprazine administration represents a concern; whether this side effect is due to a limitation of the animal models or to an intrinsic property of eltoprazine needs to be addressed in ongoing clinical trials. The data also suggest that the combination of low doses of eltoprazine with amantadine may represent a valid strategy to increase the antidyskinetic effect and reduce the eltoprazine-induced worsening of l-dopa therapeutic effects.


Subject(s)
Dyskinesia, Drug-Induced/drug therapy , Piperazines/therapeutic use , Serotonin Receptor Agonists/therapeutic use , Amantadine/therapeutic use , Amphetamine , Animals , Apomorphine/pharmacology , Disease Models, Animal , Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/etiology , Female , Levodopa/adverse effects , Macaca fascicularis , Medial Forebrain Bundle/physiology , Motor Activity/drug effects , Oxidopamine/toxicity , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinsonian Disorders/drug therapy , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Tyrosine 3-Monooxygenase/metabolism
16.
J Clin Invest ; 122(11): 3977-89, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23041629

ABSTRACT

L-DOPA-induced dyskinesia (LID), a detrimental consequence of dopamine replacement therapy for Parkinson's disease, is associated with an alteration in dopamine D1 receptor (D1R) and glutamate receptor interactions. We hypothesized that the synaptic scaffolding protein PSD-95 plays a pivotal role in this process, as it interacts with D1R, regulates its trafficking and function, and is overexpressed in LID. Here, we demonstrate in rat and macaque models that disrupting the interaction between D1R and PSD-95 in the striatum reduces LID development and severity. Single quantum dot imaging revealed that this benefit was achieved primarily by destabilizing D1R localization, via increased lateral diffusion followed by increased internalization and diminished surface expression. These findings indicate that altering D1R trafficking via synapse-associated scaffolding proteins may be useful in the treatment of dyskinesia in Parkinson's patients.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Levodopa/adverse effects , Membrane Proteins/metabolism , Receptors, Dopamine D1/metabolism , Synapses/metabolism , Animals , Corpus Striatum/pathology , Disks Large Homolog 4 Protein , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/pathology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Levodopa/pharmacology , Macaca , Male , Membrane Proteins/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/genetics , Synapses/genetics
17.
Cold Spring Harb Perspect Med ; 2(3): a009308, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22393538

ABSTRACT

The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) primate models of Parkinson's disease (PD) reproduce most, although not all, of the clinical and pathological hallmarks of PD. The present contribution presents the possibilities offered by the MPTP monkey models of PD to readers with minimal knowledge of PD, emphasizing the diversity of species, route and regimen of administration, symptoms and pathological features. Readers would eventually find out that there is not a single MPTP monkey model of PD but instead MPTP monkey models of PD, each addressing a specific experimental need.


Subject(s)
Disease Models, Animal , MPTP Poisoning , Parkinson Disease , Animals , Cercopithecinae , Chronobiology Disorders/physiopathology , Cognition Disorders , Disease Progression , Haplorhini , Lewy Bodies/pathology , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Movement/physiology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Sleep Wake Disorders/physiopathology , Translational Research, Biomedical
18.
J Neurosci ; 32(2): 681-91, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22238104

ABSTRACT

Aberrant membrane localization of dopamine D(1) receptor (D1R) is associated with L-DOPA-induced dyskinesia (LID), a major complication of L-DOPA treatment in Parkinson's disease (PD). Since the proteasome plays a central role in modulating neuronal response through regulation of neurotransmitter receptor intraneuronal fate, we hypothesized that the ubiquitine-proteasome proteolytic pathway could be impaired in LID. Those LIDs are actually associated with a striatum-specific decrease in proteasome catalytic activity and accumulation of polyubiquitinated proteins in experimental rodent and monkey parkinsonism. We then demonstrated that such decreased proteasome catalytic activity (1) results from D1R activation and (2) feed-back the D1R abnormal trafficking, i.e., its exaggerated cell surface abundance. We further showed that the genetic invalidation of the E3 ubiquitin-protein ligase parkin PD gene leads to exaggerated abnormal involuntary movements compared with wild-type mice. We thus established in an unprecedented series of experimental models that impairment of the ubiquitine-proteasome system at specific nodes (E3 ligase parkin, polyubiquitination, proteasome catalytic activity) leads to the same phenomenon, i.e., aberrant behavioral response to dopamine replacement therapy in PD, highlighting the intimate interplay between dopamine receptor and proteasome activity in a nondegenerative context.


Subject(s)
Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Receptors, Dopamine D1/agonists , Animals , Disease Models, Animal , Dopamine Agonists/toxicity , Dyskinesia, Drug-Induced/physiopathology , Female , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Parkinsonian Disorders/enzymology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/physiology
19.
Brain ; 134(Pt 8): 2321-38, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21742735

ABSTRACT

Morphine is endogenously synthesized in the central nervous system and endogenous dopamine is thought to be necessary for endogenous morphine formation. As Parkinson's disease results from the loss of dopamine and is associated with central pain, we considered how endogenous morphine is regulated in the untreated and l-DOPA-treated parkinsonian brain. However, as the cellular origin and overall distribution of endogenous morphine remains obscure in the pathological adult brain, we first characterized the distribution of endogenous morphine-like compound immunoreactive cells in the rat striatum. We then studied changes in the endogenous morphine-like compound immunoreactivity of medium spiny neurons in normal, Parkinson's disease-like and l-DOPA-treated Parkinson's disease-like conditions in experimental (rat and monkey) and human Parkinson's disease. Our results reveal an unexpected dramatic upregulation of neuronal endogenous morphine-like compound immunoreactivity and levels in experimental and human Parkinson's disease, only partially normalized by l-DOPA treatment. Our data suggest that endogenous morphine formation is more complex than originally proposed and that the parkinsonian brain experiences a dramatic upregulation of endogenous morphine immunoreactivity. The functional consequences of such endogenous morphine upregulation are as yet unknown, but based upon the current knowledge of morphine signalling, we hypothesize that it is involved in fatigue, depression and pain symptoms experienced by patients with Parkinson's disease.


Subject(s)
Brain/metabolism , Parkinsonian Disorders/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Aged , Analysis of Variance , Animals , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Choline O-Acetyltransferase/metabolism , Chromatography, High Pressure Liquid/methods , Dendrites/metabolism , Dendrites/ultrastructure , Disease Models, Animal , Dopamine/metabolism , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/deficiency , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Female , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/metabolism , Humans , Levodopa/pharmacology , Macaca fascicularis , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Immunoelectron/methods , Middle Aged , Nerve Growth Factors/metabolism , Organic Chemicals/metabolism , Oxidopamine/adverse effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Postmortem Changes , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Tandem Mass Spectrometry , alpha-Methyltyrosine/pharmacology
20.
PLoS One ; 5(10): e13306, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20967255

ABSTRACT

BACKGROUND: The A11 diencephalospinal pathway is crucial for sensorimotor integration and pain control at the spinal cord level. When disrupted, it is thought to be involved in numerous painful conditions such as restless legs syndrome and migraine. Its anatomical organization, however, remains largely unknown in the non-human primate (NHP). We therefore characterized the anatomy of this pathway in the NHP. METHODS AND FINDINGS: In situ hybridization of spinal dopamine receptors showed that D1 receptor mRNA is absent while D2 and D5 receptor mRNAs are mainly expressed in the dorsal horn and D3 receptor mRNA in both the dorsal and ventral horns. Unilateral injections of the retrograde tracer Fluoro-Gold (FG) into the cervical spinal enlargement labeled A11 hypothalamic neurons quasi-exclusively among dopamine areas. Detailed immunohistochemical analysis suggested that these FG-labeled A11 neurons are tyrosine hydroxylase-positive but dopa-decarboxylase and dopamine transporter-negative, suggestive of a L-DOPAergic nucleus. Stereological cell count of A11 neurons revealed that this group is composed by 4002±501 neurons per side. A 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) intoxication with subsequent development of a parkinsonian syndrome produced a 50% neuronal cell loss in the A11 group. CONCLUSION: The diencephalic A11 area could be the major source of L-DOPA in the NHP spinal cord, where it may play a role in the modulation of sensorimotor integration through D2 and D3 receptors either directly or indirectly via dopamine formation in spinal dopa-decarboxylase-positives cells.


Subject(s)
Diencephalon/anatomy & histology , Primates/anatomy & histology , Spinal Cord/anatomy & histology , Animals , Diencephalon/metabolism , Dopamine/metabolism , Immunohistochemistry , In Situ Hybridization , RNA, Messenger/genetics , Receptors, Dopamine/genetics , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...