Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
Add more filters

Publication year range
1.
J Neurosci ; 40(35): 6812-6819, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32690616

ABSTRACT

Parkinson's disease (PD) is characterized by severe locomotor deficits and is commonly treated with the dopamine precursor L-DOPA, but its prolonged usage causes dyskinesias referred to as L-DOPA-induced dyskinesia (LID). Several studies in animal models of PD have suggested that dyskinesias are associated with a heightened opioid cotransmitter tone, observations that have led to the notion of a LID-related hyperactive opioid transmission that should be corrected by µ opioid receptor antagonists. Reports that both antagonists and agonists of the µ opioid receptor may alleviate LID severity in primate models of PD and LID, together with the failure of nonspecific antagonist to improve LID in pilot clinical trials in patients, raises doubt about the reliability of the available data on the opioid system in PD and LID. After in vitro characterization of the functional activity at the µ opioid receptor, we selected prototypical agonists, antagonists, and partial agonists at the µ opioid receptor. We then showed that both oral and discrete intracerebral administration of a µ receptor agonist, but not of an antagonist as long thought, ameliorated LIDs in the gold-standard bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned female macaque model of PD and LID. The results call for a reappraisal of opioid pharmacology in the basal ganglia as well as for the development of brain nucleus-targeted µ opioid receptor agonists.SIGNIFICANCE STATEMENT µ opioid receptors have long been considered as a viable target for alleviating the severity of L-DOPA-induced hyperkinetic side effects, induced by the chronic treatment of Parkinson's disease motor symptoms with L-DOPA. Conflicting results between experimental parkinsonism and Parkinson's disease patients, however, dampened the enthusiasm for the target. Here we reappraise the pharmacology and then demonstrate that both oral and discrete intracerebral administration of a µ receptor agonist, but not of an antagonist as long thought, ameliorates LIDs in the gold-standard bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson's disease, calling for a reappraisal of the opioid pharmacology as well as for the development of brain nucleus-targeted µ receptor agonists.


Subject(s)
Dyskinesias/drug therapy , MPTP Poisoning/physiopathology , Receptors, Opioid, mu/agonists , Animals , CHO Cells , Cricetinae , Cricetulus , Dyskinesias/etiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use , Female , Humans , Levodopa/adverse effects , Levodopa/therapeutic use , MPTP Poisoning/drug therapy , Macaca fascicularis , Neurotransmitter Agents/administration & dosage , Neurotransmitter Agents/pharmacology , Neurotransmitter Agents/therapeutic use , Receptors, Opioid, mu/antagonists & inhibitors
2.
Biochem Biophys Res Commun ; 556: 16-22, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33836343

ABSTRACT

Evidence suggests constipation precedes motor dysfunction and is the most common gastrointestinal symptom in Parkinson's disease (PD). 5-HT4 receptor (5-HT4R) agonist prucalopride has been approved to treat chronic constipation. Here, we reported intraperitoneal injection of prucalopride for 7 days increased dopamine and decreased dopamine turnover. Prucalopride administration improved motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrathydropyridine (MPTP)-induced PD mouse models. Prucalopride treatment also ameliorated intestinal barrier impairment and increased IL-6 release in PD model mice. However, prucalopride treatment exerted no impact on JAK2/STAT3 pathway, suggesting that prucalopride may stimulate IL-6 via JAK2/STAT3-independent pathway. In conclusion, prucalopride exerted beneficial effects in MPTP-induced Parkinson's disease mice by attenuating the loss of dopamine, improving motor dysfunction and intestinal barrier.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Benzofurans/pharmacology , Benzofurans/therapeutic use , Intestinal Mucosa/drug effects , Motor Skills/drug effects , Parkinson Disease/prevention & control , Parkinson Disease/physiopathology , Animals , Body Weight/drug effects , Disease Models, Animal , Dopamine/metabolism , Eating/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Janus Kinase 2/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/physiopathology , MPTP Poisoning/prevention & control , Male , Mice , Mice, Inbred C57BL , Neostriatum/metabolism , Parkinson Disease/drug therapy , Parkinson Disease, Secondary/physiopathology , Parkinson Disease, Secondary/prevention & control , STAT3 Transcription Factor/metabolism
3.
Learn Mem ; 27(9): 372-379, 2020 09.
Article in English | MEDLINE | ID: mdl-32817303

ABSTRACT

Parkinson's disease (PD) is characterized clinically by progressive motor dysfunction; overt parkinsonism is often preceded by prodromal symptoms including disturbances in the sleep-wake cycle. Up to 80% of patients with PD also develop dementia. In humans, there are three major apolipoprotein E isoforms: E2, E3, and E4. Increased rate of dementia in PD may be associated with E4 isoform. To better understand prodromal changes associated with E4, we exposed young (3-5 mo) male and female mice expressing E3 or E4 via targeted replacement to a subchronic dosage of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We hypothesized that E4 mice would be more susceptible to MPTP-related behavioral and cognitive changes. MPTP-treated E4 mice explored novel objects longer than genotype-matched saline-treated mice. In contrast, saline-treated E3 mice preferentially explored the novel object whereas MPTP-treated E3 mice did not and showed impaired object recognition. MPTP treatment altered swim speed of E4, but not E3, mice in the water maze compared to controls. Thus, E4 carriage may influence the preclinical symptoms associated with PD. Increased efforts are warranted to study early time points in this disease model.


Subject(s)
Apolipoprotein E3 , Apolipoprotein E4 , Behavior, Animal , MPTP Poisoning/genetics , MPTP Poisoning/physiopathology , Motor Activity , Recognition, Psychology , Spatial Learning , Animals , Behavior, Animal/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Recognition, Psychology/physiology , Spatial Learning/physiology
4.
Cereb Cortex ; 29(1): 167-177, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29190329

ABSTRACT

Parkinson's disease is associated with abnormal oscillatory electrical activities of neurons and neuronal ensembles throughout the basal ganglia-thalamocortical network. It has recently been documented in patients with advanced parkinsonism that the amplitude of gamma-band oscillations (50-200 Hz) in electrocorticogram recordings from the primary motor cortex is abnormally coupled to the phase of beta band oscillations within the same signals. It is not known when in the course of the disease the abnormal phase-amplitude coupling (PAC) arises, and whether it is influenced by arousal or prior exposure to dopaminergic medications. To address these issues, we analyzed the relationship between the severity of parkinsonian motor signs and the extent of PAC in a progressive model of parkinsonism, using primates that were not exposed to levodopa prior to testing. PAC was measured in electrocorticogram signals from the primary motor cortex and the supplementary motor area in 3 monkeys that underwent weekly injections of small doses of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rendering them progressively parkinsonian. We found that parkinsonism was associated with increased coupling between the phase of low-frequency (4-10 Hz) oscillations and the amplitude of oscillations in the high gamma band (50-150 Hz). These changes only reached significance when the animals became fully parkinsonian. The increased PAC was normalized after levodopa treatment. We also found a similar increase in PAC during sleep, even in normal animals. The identified PAC was independent of concomitant changes in spectral power in the 2.9-9.8Hz or 49.8-150.4 Hz ranges. We conclude that PAC is predominately a sign of advanced parkinsonism, and is, thus, not essential for the development of parkinsonism. However, increased PAC appears to correlate with the severity of fully developed parkinsonism.


Subject(s)
Disease Models, Animal , Disease Progression , Gamma Rhythm/physiology , MPTP Poisoning/physiopathology , Motor Cortex/physiopathology , Animals , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Female , MPTP Poisoning/drug therapy , Macaca mulatta , Male , Motor Cortex/drug effects , Primates
5.
J Integr Neurosci ; 19(2): 209-215, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32706185

ABSTRACT

Assessing the finger fine motor ability is extremely important. However, conventional behavioral tests in monkeys are complicated and costly. We attempted to develop a new task to assess the precise finger grip in Parkinson's disease monkeys based on the principles of objectification, multipurpose, and simplification. This study involved seven adult male cynomolgus monkeys. A gripping test based on the previous food reaching test was developed. Parallel experiments of food reaching test and gripping test affected by the treatments of levodopa and deep brain stimulation of the subthalamic nucleus were performed to verify the utility of the gripping test. We found that gross motor ability (measured by food reaching test) could be significantly improved by both the subthalamic nucleus and levodopa administration, which reproduced the results of our previous study. The finger fine motor ability (measured by the gripping test) could be significantly improved by levodopa administration, but not by the subthalamic nucleus. Our results verified the utility and reliability of the gripping test, which is a simple, convenient, and objective task for evaluating the finger fine motor skill in Parkinson's disease monkeys. Mechanisms of the efficacy of deep brain stimulation on fine motor ability require further investigation.


Subject(s)
Antiparkinson Agents/pharmacology , Deep Brain Stimulation , Fingers , Levodopa/pharmacology , Motor Activity , Motor Skills , Neuropsychological Tests/standards , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Subthalamic Nucleus , Animals , Antiparkinson Agents/administration & dosage , Behavior, Animal/drug effects , Behavior, Animal/physiology , Disease Models, Animal , Fingers/physiopathology , Levodopa/administration & dosage , MPTP Poisoning/physiopathology , MPTP Poisoning/therapy , Macaca fascicularis , Male , Motor Activity/drug effects , Motor Activity/physiology , Motor Skills/drug effects , Motor Skills/physiology , Parkinson Disease/drug therapy , Reproducibility of Results
6.
Neurobiol Dis ; 128: 40-48, 2019 08.
Article in English | MEDLINE | ID: mdl-30086388

ABSTRACT

The pedunculopontine nucleus (PPN) included in the caudal mesencephalic reticular formation (cMRF) plays a key role in the control of locomotion and wake state. Regarding its involvement in the neurodegenerative process observed in Parkinson disease (PD), deep brain stimulation of the PPN was proposed to treat levodopa-resistant gait disorders. However, the precise role of the cMRF in the pathophysiology of PD, particularly in freezing of gait and other non-motor symptoms is still not clear. Here, using micro electrode recording (MER) in 2 primates, we show that dopamine depletion did not alter the mean firing rate of the overall cMRF neurons, particularly the putative non-cholinergic ones, but only a decreased activity of the regular neurons sub-group (though to be the cholinergic PPN neurons). Interestingly, a significant increase in the relative proportion of cMRF neurons with a burst pattern discharge was observed after MPTP intoxication. The present results question the hypothesis of an over-inhibition of the CMRF by the basal ganglia output structures in PD. The decreased activity observed in the regular neurons could explain some non-motor symptoms in PD regarding the strong involvement of the cholinergic neurons on the modulation of the thalamo-cortical system. The increased burst activity under dopamine depletion confirms that this specific spike discharge pattern activity also observed in other basal ganglia nuclei and in different pathologies could play a mojor role in the pathophysiology of the disease and could explain several symptoms of PD including the freezing of gait. The present data will have to be replicated in a larger number of animals and will have to investigate more in details how the modification of the spike discharge of the cMRF neurons in the parkinsonian state could alter functions such as locomotion and attentional state. This will ultimely allow a better comprehension of the pathophysiology of freezing of gait.


Subject(s)
Action Potentials/physiology , MPTP Poisoning/physiopathology , Neurons/physiology , Pedunculopontine Tegmental Nucleus/physiopathology , Reticular Formation/physiopathology , Animals , Macaca fascicularis , Male
7.
Behav Pharmacol ; 30(1): 36-44, 2019 02.
Article in English | MEDLINE | ID: mdl-29847337

ABSTRACT

Parkinson's disease (PD) causes major changes in dopaminergic neurons of the brain, resulting in motor symptoms in older adults. A previous study showed that Danshensu alleviates the cognitive decline by attenuating neuroinflammation. In the present study, we investigated the neuroprotective effect of Danshensu in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. C57BL/6 mice were randomly divided into the following four groups: control, MPTP, Danshensu at 15 mg/kg, and Danshensu at 60 mg/kg. The mice were administered Danshensu intragastrically for 14 days. In the behavioral tests, Danshensu treatment alleviated motor dysfunction induced by MPTP. The number of tyrosine hydroxylase-positive neurons in the substantia nigra was significantly reduced in the MPTP group, relative to the control group; Danshensu partially blocked this reduction in tyrosine hydroxylase-positive neurons. In addition, Danshensu attenuated the reductions in striatal dopamine and 5-HT levels induced by MPTP. Danshensu also diminished the increase in Iba1-positive cells in the substantia nigra and reduced the levels of interleukin-1ß and tumor necrosis factor-α in the striatum. These findings suggest that Danshensu exerts neuroprotective effects and improves motor function in PD mice, at least in part, by reducing neuroinflammation.


Subject(s)
Lactates/therapeutic use , MPTP Poisoning/drug therapy , MPTP Poisoning/physiopathology , Neuroprotective Agents/therapeutic use , Animals , Disease Models, Animal , Dopamine/metabolism , Gene Expression Regulation/drug effects , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Psychomotor Performance/drug effects , Rotarod Performance Test , Serotonin/metabolism , Tumor Necrosis Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism
8.
J Immunol ; 198(11): 4312-4326, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28446566

ABSTRACT

Although the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model is the most widely used animal model for Parkinson's disease (PD), it is known that nigrostriatal pathologies do not persist in the acute MPTP mouse model. This study highlights the importance of adaptive immunity in driving persistent and progressive disease in acute MPTP-intoxicated mice. Although marked infiltration of T cells into the nigra was found on 1 d of MPTP insult, T cell infiltration decreased afterward, becoming normal on 30 d of insult. Interestingly, twice-weekly supplementation of RANTES and eotaxin, chemokines that are involved in T cell trafficking, drove continuous T cell infiltration to the nigra and incessant glial inflammation. Supplementation of RANTES and eotaxin was also associated with the induction of nigral α-synuclein pathology, persistent loss of dopaminergic neurons and striatal neurotransmitters, and continuous impairment of motor functions in MPTP-intoxicated mice. In contrast, supplementation of TNF-α and IL-1ß, widely studied proinflammatory cytokines, did not induce persistent disease in MPTP-insulted mice. Our results suggest that induction of adaptive immunity by RANTES and eotaxin could hold the key for driving persistent nigrostriatal pathologies in the MPTP mouse model, and that targeting these factors may halt disease progression in PD patients.


Subject(s)
Adaptive Immunity , MPTP Poisoning/immunology , MPTP Poisoning/physiopathology , Substantia Nigra/immunology , Acute Disease , Animals , Chemokine CCL11/administration & dosage , Chemokine CCL5/administration & dosage , Disease Progression , Inflammation , Interleukin-1beta/administration & dosage , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Substantia Nigra/pathology , Tumor Necrosis Factor-alpha/administration & dosage
9.
Dig Dis Sci ; 64(12): 3502-3517, 2019 12.
Article in English | MEDLINE | ID: mdl-31187328

ABSTRACT

BACKGROUND: Gastrointestinal (GI) motility dysfunction is the most common non-motor symptom of Parkinson's disease (PD). Studies have indicated that GI motility functions are impaired before the onset of PD. AIMS: To investigate the underlying mechanism of PD-induced GI dysmotility in MPTP (1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine)-induced animal model. METHODS: C57BL/6 mice were administered with or without a selective dopamine neurotoxin, MPTP, to induce parkinsonian symptoms. In addition to in vivo studies, in vitro experiments were also conducted in colon specimens using l-methyl-4-phenylpyridinium (MPP+), a metabolic product of MPTP. Gastric emptying, colon motility, nitrergic relaxation, and western blot experiments were performed as reported. RESULTS: MPTP-induced PD mice showed decreased expression of nuclear factor erythroid 2-related factor (Nrf2) and its target phase II genes in gastric and colon neuromuscular tissues. Decreased levels of tetrahydrobiopterin (BH4, a critical cofactor for nNOS dimerization) associated with uncoupling of nNOS in gastric and colon tissues exposed to MPTP. Impaired enteric nitrergic system led to delayed gastric emptying and slower colonic motility compared to the control mice. In vitro results in colon specimens confirm that activation of Nrf2 restored MPP+-induced suppression of alpha-synuclein, tyrosine hydroxylase (TH), Nrf2, and heme oxygenase-1. In vitro exposure to L-NAME [N(w)-nitro-L-arginine methyl ester], a NOS synthase inhibitor, reduced protein expression of TH in colon tissue homogenates. CONCLUSIONS: Loss of Nrf2/BH4/nNOS expression in PD impairs antioxidant gene expression, which deregulates NO synthesis, thereby contributing to the development of GI dysmotility and constipation. Nitric oxide appears to be important to maintain dopamine synthesis in the colon.


Subject(s)
Gastrointestinal Motility/physiology , MPTP Poisoning/genetics , NF-E2-Related Factor 2/genetics , Nitric Oxide Synthase Type I/genetics , Nitric Oxide/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Biopterins/analogs & derivatives , Biopterins/metabolism , Blotting, Western , Colon/drug effects , Colon/metabolism , Colon/physiopathology , Constipation , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gastric Emptying/physiology , Gene Expression Regulation , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/metabolism , Parkinson Disease/physiopathology , Parkinsonian Disorders , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/drug effects , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
10.
Clin Auton Res ; 29(4): 415-425, 2019 08.
Article in English | MEDLINE | ID: mdl-31338635

ABSTRACT

PURPOSE: Systemic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces degeneration of dopaminergic neurons and reproduces the motor features of Parkinson disease (PD); however, the effect of MPTP on extranigral structures has been poorly studied. The aim of this research was to study the cardiac sympathetic innervation of control and MPTP-treated monkeys in order to describe the influence of MPTP toxicity on cardiac tissue. METHODS: Eight monkeys were included in the study and divided into two groups, four monkeys serving as controls and four forming the MPTP group. Sections from the anterior left ventricle were immunohistochemically examined to characterize the sympathetic fibers of cardiac tissue. The intensity of immunoreactivity in the nerve fibers was quantitatively analyzed using ImageJ software. RESULTS: As occurs in PD, the sympathetic peripheral nervous system is affected in MPTP-treated monkeys. The percentage of tyrosine hydroxylase immunoreactive fibers in the entire fascicle area was markedly lower in the MPTP group (24.23%) than the control group (35.27%) (p < 0.05), with preservation of neurofilament immunoreactive fibers in the epicardium of MPTP-treated monkeys. Alpha-synuclein deposits were observed in sections of the anterior left ventricle of MPTP-treated monkeys but not in control animals, whereas phosphorylated synuclein aggregates were not observed in either controls or MPTP-treated monkeys. CONCLUSION: The peripheral autonomic system can also be affected by neurotoxins that specifically inhibit mitochondrial complex I.


Subject(s)
Disease Models, Animal , Heart/innervation , Heart/physiopathology , MPTP Poisoning/physiopathology , Animals , MPTP Poisoning/metabolism , Macaca fascicularis , Male , Primates , Random Allocation , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , alpha-Synuclein/metabolism
11.
J Neurosci ; 37(17): 4584-4592, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28363982

ABSTRACT

Transforming growth factor-ß (TGF-ß) plays an important role in the development and maintenance of embryonic dopaminergic (DA) neurons in the midbrain. To study the function of TGF-ß signaling in the adult nigrostriatal system, we generated transgenic mice with reduced TGF-ß signaling in mature neurons. These mice display age-related motor deficits and degeneration of the nigrostriatal system. Increasing TGF-ß signaling in the substantia nigra through adeno-associated virus expressing a constitutively active type I receptor significantly reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration and motor deficits. These results suggest that TGF-ß signaling is critical for adult DA neuron survival and that modulating this signaling pathway has therapeutic potential in Parkinson disease.SIGNIFICANCE STATEMENT We show that reducing Transforming growth factor-ß (TGF-ß) signaling promotes Parkinson disease-related pathologies and motor deficits, and increasing TGF-ß signaling reduces neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a parkinsonism-inducing agent. Our results provide a rationale to pursue a means of increasing TGF-ß signaling as a potential therapy for Parkinson's disease.


Subject(s)
MPTP Poisoning/physiopathology , Neostriatum/physiopathology , Neurodegenerative Diseases/physiopathology , Signal Transduction , Substantia Nigra/physiopathology , Transforming Growth Factor beta/deficiency , Animals , Cell Survival/genetics , Gait Disorders, Neurologic/chemically induced , Gait Disorders, Neurologic/physiopathology , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/chemically induced , Postural Balance , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics
12.
Neuroimage ; 183: 132-141, 2018 12.
Article in English | MEDLINE | ID: mdl-30102999

ABSTRACT

BACKGROUND: Dopaminergic and serotonergic degenerations alter pharmacological neurotransmission and structural markers in Parkinson's disease (PD). Alteration of diffusion measures in key brain regions depict MPTP/MDMA lesions in the monkey model of PD. Whether dopatherapy impacts such diffusion measures remains an open question. OBJECTIVES: The aim of this study was to investigate the consequences of l-DOPA treatment on diffusion alterations, PET imaging and immunohistochemical markers in MPTP/MDMA-intoxicated monkeys. METHODS: We acquired PET imaging and measures of mean diffusivity and fractional anisotropy longitudinally and correlated them with behavior and post-mortem fiber quantification. RESULTS: Severity of l-DOPA-induced dyskinesia was correlated to serotonin transporter radioligand binding increases in the ventral striatum and the anterior cingulate cortex and decreases of mean diffusivity in the ventral striatum. After lesion of serotonergic fibers by MDMA and the second l-DOPA period, diffusion measures were no more altered while the serotonergic binding still increased in all regions of interest, despite abolition of dyskinesia. Interestingly, in the anterior cingulate cortex, the SERT radioligand binding was negatively correlated to the number of SERT fibers. CONCLUSION: These results show that the increase of SERT radioligand binding is not systematically paralleled by an increase of SERT fibers and does not always reflect the presence of LID. More specifically, our study suggest that SERT increase may be underpinned by an increased density of serotonergic fibers after MPTP and the first l-DOPA period, and by an elevation of SERT itself after MDMA and the second l-DOPA period. This highlights that DTI is complementary to PET imaging to decipher pathophysiological mechanisms underlying l-DOPA-induced dyskinesia in a non-human primate model of PD.


Subject(s)
Brain , Dopamine Agents/pharmacology , Dyskinesia, Drug-Induced , Levodopa/pharmacology , Nerve Fibers , Parkinson Disease, Secondary , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Diffusion Tensor Imaging , Disease Models, Animal , Dyskinesia, Drug-Induced/diagnostic imaging , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Immunohistochemistry , MPTP Poisoning/diagnostic imaging , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Macaca fascicularis , Multimodal Imaging , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Nerve Fibers/drug effects , Nerve Fibers/pathology , Parkinson Disease, Secondary/diagnostic imaging , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Parkinson Disease, Secondary/physiopathology , Positron-Emission Tomography
13.
J Neural Transm (Vienna) ; 125(9): 1355-1360, 2018 09.
Article in English | MEDLINE | ID: mdl-29247391

ABSTRACT

Trazodone is a clinically available anti-depressant that exhibits affinity for serotonin 1A and 2A receptors, as well as for alpha-adrenoceptors, suggesting that it may be useful to treat L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia and psychosis that are encountered in advanced Parkinson's disease (PD). Here, we investigated the anti-dyskinetic and anti-psychotic effects of trazodone in the parkinsonian non-human primate. 6 common marmosets were rendered parkinsonian by administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Following repeated administration of L-DOPA to induce stable dyskinesia and psychosis-like behaviours (PLBs), trazodone (0.1, 1 and 10 mg/kg) or vehicle was administered in combination with L-DOPA and its effects on dyskinesia, PLBs and parkinsonism were determined. The addition of trazodone 10 mg/kg to L-DOPA reduced peak dose dyskinesia by ≈ 39% (P < 0.01) and peak dose PLBs by ≈ 17% (P < 0.01). However, parkinsonian disability was significantly worsened by trazodone 10 mg/kg (P < 0.05) and duration of anti-parkinsonian action was diminished by ≈ 21% (P < 0.05). Our results suggest that trazodone may be effective in alleviating L-DOPA-induced dyskinesia and psychosis in PD, but its deleterious effect on motor function is a concern and may limit its tolerability and usefulness in clinical settings.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/drug therapy , Levodopa/toxicity , MPTP Poisoning/physiopathology , Parkinsonian Disorders/physiopathology , Serotonin Receptor Agonists/therapeutic use , Trazodone/therapeutic use , Animals , Antiparkinson Agents/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Behavior, Animal/drug effects , Callithrix , Disorders of Excessive Somnolence/chemically induced , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/prevention & control , Female , Levodopa/therapeutic use , MPTP Poisoning/drug therapy , MPTP Poisoning/psychology , Male , Motor Activity/drug effects , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/psychology , Serotonin Receptor Agonists/pharmacology , Trazodone/pharmacology , Trazodone/toxicity
14.
J Neurophysiol ; 117(6): 2242-2249, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28228579

ABSTRACT

A number of studies suggest that Parkinson's disease (PD) is associated with alterations of neuronal activity patterns in the basal-ganglia-thalamocortical circuit. There are limited electrophysiological data, however, describing how the premotor cortex, which is involved in movement and decision-making, is likely impacted in PD. In this study, spontaneous local field potential (LFP) and single unit neuronal activity were recorded in the dorsal premotor area of nonhuman primates in both the naïve and parkinsonian state using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of parkinsonism. In both animals, we observed a shift of power in LFP power spectral densities (1-350 Hz) from higher to lower frequency bands; parkinsonism resulted in increased power in frequencies <8 Hz and decreased power at frequencies >30 Hz. A comparable but not identical trend was observed in the power spectral analysis of single unit spike trains: alpha power increased in both animals and gamma power decreased in one; power in other frequency bands remaining unchanged. Although not consistent across animals, we also observed changes in discharge rates and bursting activity. Overall, the LFP and single unit analysis suggest that abnormalities in premotor neural activity are a feature of parkinsonism, although specific details of those abnormalities may differ between subjects. This study further supports the concept that PD is a network disorder that induces abnormal spontaneous neural activities across the basal-ganglia-thalamocortical circuit including the premotor cortex and provides foundational knowledge for future studies regarding the relationship between changes in neuronal activity in this region and the development of motor deficits in PD.NEW & NOTEWORTHY This study begins to fill a gap in knowledge regarding how Parkinson's disease (PD) may cause abnormal functioning of the premotor cortex. It is novel as the premotor activity is examined in both the naïve and parkinsonian states, in the same subjects, at the single unit and LFP level. It provides foundational knowledge on which to build future studies to explore the relationships between premotor activities and specific parkinsonian motor and cognitive deficits.


Subject(s)
Evoked Potentials , MPTP Poisoning/physiopathology , Motor Cortex/physiopathology , Neurons/physiology , Alpha Rhythm , Animals , Female , Gamma Rhythm , Macaca mulatta , Male
15.
J Neurophysiol ; 118(5): 2654-2669, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28835526

ABSTRACT

Oscillatory neural activity in different frequency bands and phase-amplitude coupling (PAC) are hypothesized to be biomarkers of Parkinson's disease (PD) that could explain dysfunction in the motor circuit and be used for closed-loop deep brain stimulation (DBS). How these putative biomarkers change from the normal to the parkinsonian state across nodes in the motor circuit and within the same subject, however, remains unknown. In this study, we characterized how parkinsonism and vigilance altered oscillatory activity and PAC within the primary motor cortex (M1), subthalamic nucleus (STN), and globus pallidus (GP) in two nonhuman primates. Static and dynamic analyses of local field potential (LFP) recordings indicate that 1) after induction of parkinsonism using the neurotoxin MPTP, low-frequency power (8-30 Hz) increased in the STN and GP in both subjects, but increased in M1 in only one subject; 2) high-frequency power (~330 Hz) was present in the STN in both normal subjects but absent in the parkinsonian condition; 3) elevated PAC measurements emerged in the parkinsonian condition in both animals, but in different sites in each animal (M1 in one subject and GPe in the other); and 4) the state of vigilance significantly impacted how oscillatory activity and PAC were expressed in the motor circuit. These results support the hypothesis that changes in low- and high-frequency oscillatory activity and PAC are features of parkinsonian pathophysiology and provide evidence that closed-loop DBS systems based on these biomarkers may require subject-specific configurations as well as adaptation to changes in vigilance.NEW & NOTEWORTHY Chronically implanted electrodes were used to record neural activity across multiple nodes in the basal ganglia-thalamocortical circuit simultaneously in a nonhuman primate model of Parkinson's disease, enabling within-subject comparisons of electrophysiological biomarkers between normal and parkinsonian conditions and different vigilance states. This study improves our understanding of the role of oscillatory activity and phase-amplitude coupling in the pathophysiology of Parkinson's disease and supports the development of more effective DBS therapies based on pathophysiological biomarkers.


Subject(s)
Arousal , Globus Pallidus/physiopathology , MPTP Poisoning/physiopathology , Motor Cortex/physiopathology , Animals , Deep Brain Stimulation , Evoked Potentials , Female , Macaca mulatta , Subthalamic Nucleus/physiopathology
16.
Biochem Biophys Res Commun ; 493(4): 1443-1449, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28986252

ABSTRACT

Uric acid has neuroprotective effect on Parkinson's disease (PD) by inhibiting oxidative damage and neuronal cell death. Our previous study has shown that uric acid protected dopaminergic cell line damage through inhibiting accumulation of NF-E2-related factor 2 (Nrf2). This study aimed to investigate its in vivo neuroprotective effect. PD was induced by MPTP intraperitoneally injection for 7 d in male C57BL/6 mice. Mice were treated with either uric acid (intraperitoneally injection 250 mg/kg) or saline for a total of 13 d. We showed that uric acid improved behavioral performances and cognition of PD mice, increased TH-positive dopaminergic neurons and decreased GFAP-positive astrocytes in substantia nigra (SN). Uric acid increased mRNA and protein expressions of Nrf2 and three Nrf2-responsive genes, including γ-glutamate-cysteine ligase catalytic subunit (γ-GCLC), heme oxygenase-1 (HO-1) and NQO1. Uric acid significantly increased superoxide dismutase (SOD), CAT, glutathione (GSH) levels and decreased malondialdehyde (MDA) level in SN regions of MPTP-treated mice. Uric acid inhibited the hippocampal expression of IL-1ß and decreased serum and hippocampus levels of interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α). In conclusion, uric acid demonstrates neuroprotective properties for dopaminergic neurons in PD mice through modulation of neuroinflammation and oxidative stress.


Subject(s)
Antioxidant Response Elements/drug effects , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Uric Acid/pharmacology , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Cytokines/blood , Cytokines/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Inflammation/drug therapy , Inflammation/pathology , MPTP Poisoning/drug therapy , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Parkinson Disease/psychology , Signal Transduction/drug effects
17.
Ann Neurol ; 79(1): 59-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26456231

ABSTRACT

OBJECTIVE: To examine whether near-infrared light (NIr) treatment reduces clinical signs and/or offers neuroprotection in a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkey model of Parkinson disease. METHODS: We implanted an optical fiber device that delivered NIr (670 nm) to the midbrain of macaque monkeys, close to the substantia nigra of both sides. MPTP injections (1.5-2.1mg/kg) were made over a 5- to 7-day period, during which time the NIr device was turned on. This was then followed by a 3-week survival period. Monkeys were evaluated clinically (eg, posture, bradykinesia) and behaviorally (open field test), and their brains were processed for immunohistochemistry and stereology. RESULTS: All monkeys in the MPTP group developed severe clinical and behavioral impairment (mean clinical scores = 21-34; n = 11). By contrast, the MPTP-NIr group developed much less clinical and behavioral impairment (n = 9); some monkeys developed moderate clinical signs (mean scores = 11-15; n = 3), whereas the majority--quite remarkably--developed few clinical signs (mean scores = 1-6; n = 6). The monkeys that developed moderate clinical signs had hematic fluid in their optical fibers at postmortem, presumably limiting NIr exposure and overall clinical improvement. NIr was not toxic to brain tissue and offered neuroprotection to dopaminergic cells and their terminations against MPTP insult, particularly in animals that developed few clinical signs. INTERPRETATION: Our findings indicate NIr to be an effective therapeutic agent in a primate model of the disease and create the template for translation into clinical trials.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Behavior, Animal/radiation effects , Infrared Rays/therapeutic use , MPTP Poisoning/prevention & control , Mesencephalon/radiation effects , Neurotoxins/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Low-Level Light Therapy , MPTP Poisoning/physiopathology , Macaca fascicularis , Male , Mesencephalon/drug effects , Neurotoxins/administration & dosage , Optical Fibers
18.
Toxicol Appl Pharmacol ; 326: 34-42, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28400118

ABSTRACT

Reactive oxygen species are a well-defined therapeutic target for Parkinson's disease (PD) and pharmacological agents that catalytically scavenge reactive species are promising neuroprotective strategies for treatment. Metalloporphyrins are synthetic catalytic antioxidants that mimic the body's own antioxidant enzymes i.e. superoxide dismutases and catalase. The goal of this study was to determine if newly designed metalloporphyrins have enhanced pharmacodynamics including oral bioavailability, longer plasma elimination half-lives, penetrate the blood brain barrier, and show promise for PD treatment. Three metalloporphyrins (AEOL 11216, AEOL 11203 and AEOL 11114) were identified in this study as potential candidates for further pre-clinical development. Each of these compounds demonstrated blood brain barrier permeability by the i.p. route and two of three compounds (AEOL 11203 and AEOL 11114) were orally bioavailable. All of these compounds protected against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity, including dopamine depletion in the striatum, dopaminergic neuronal loss in the substantial nigra, and increased oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain of the mice without inhibiting MPTP metabolism. Daily therapeutic dosing of these metalloporphyrins were well tolerated without accumulation of brain manganese levels or behavioral alterations assessed by open field and rotarod tests. The study identified two orally active metalloporphyrins and one injectable metalloporphyrin as clinical candidates for further development in PD.


Subject(s)
Antioxidants/pharmacology , Antiparkinson Agents/pharmacology , Brain/drug effects , MPTP Poisoning/prevention & control , Metalloporphyrins/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Administration, Oral , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacokinetics , Behavior, Animal/drug effects , Biological Availability , Biomarkers/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/physiopathology , Capillary Permeability , Disease Models, Animal , Dopamine/metabolism , Drug Design , Drug Evaluation, Preclinical , Half-Life , Injections, Intraperitoneal , MPTP Poisoning/etiology , MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/pharmacokinetics , Mice, Inbred C57BL , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Rotarod Performance Test
19.
J Neurosci ; 35(15): 6231-40, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878293

ABSTRACT

While beta oscillations often occur within the parkinsonian basal ganglia, how these oscillations emerge from a naive state and change with disease severity is not clear. To address this question, a progressive, nonhuman primate model of Parkinson's disease was developed using staged injections of MPTP. Within each parkinsonian state (naive, mild, moderate, and severe), spontaneous local field potentials were recorded throughout the sensorimotor globus pallidus. In the naive state, beta oscillations (11-32 Hz) occurred in half of the recordings, indicating spontaneous beta oscillations in globus pallidus are not pathognomonic. Mild and moderate states were characterized by a narrower distribution of beta frequencies that shifted toward the 8-15 Hz range. Additionally, coupling between the phase of beta and the amplitude of high-frequency oscillations (256-362 Hz) emerged in the mild state and increased with severity. These findings provide a novel mechanistic framework to understand how progressive loss of dopamine translates into abnormal information processing in the pallidum through alterations in oscillatory activity. The results suggest that rather than the emergence of oscillatory activity in one frequency spectrum or the other, parkinsonian motor signs may relate more to the development of altered coupling across multiple frequency spectrums.


Subject(s)
Beta Rhythm/physiology , Globus Pallidus/physiopathology , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Animals , Biological Clocks/physiology , Brain Mapping , Disease Models, Animal , Female , Macaca mulatta , Magnetic Resonance Imaging , Male , Movement/physiology , Severity of Illness Index , Spectrum Analysis , Statistics as Topic
20.
J Neurosci ; 35(50): 16463-78, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26674871

ABSTRACT

Vasoactive intestinal peptide (VIP) mediates a broad range of biological responses by activating two related receptors, VIP receptor 1 and 2 (VIPR1 and VIPR2). Although the use of native VIP facilitates neuroprotection, clinical application of the hormone is limited due to VIP's rapid metabolism and inability to distinguish between VIPR1 and VIPR2 receptors. In addition, activation of both receptors by therapeutics may increase adverse secondary toxicities. Therefore, we developed metabolically stable and receptor-selective agonists for VIPR1 and VIPR2 to improve pharmacokinetic and pharmacodynamic therapeutic end points. Selective agonists were investigated for their abilities to protect mice against MPTP-induced neurodegeneration used to model Parkinson's disease (PD). Survival of tyrosine hydroxylase neurons in the substantia nigra was determined by stereological tests after MPTP intoxication in mice pretreated with either VIPR1 or VIPR2 agonist or after adoptive transfer of splenic cell populations from agonist-treated mice administered to MPTP-intoxicated animals. Treatment with VIPR2 agonist or splenocytes from agonist-treated mice resulted in increased neuronal sparing. Immunohistochemical tests showed that agonist-treated mice displayed reductions in microglial responses, with the most pronounced effects in VIPR2 agonist-treated, MPTP-intoxicated mice. In parallel studies, we observed reductions in proinflammatory cytokine release that included IL-17A, IL-6, and IFN-γ and increases in GM-CSF transcripts in CD4(+) T cells recovered from VIPR2 agonist-treated animals. Moreover, a phenotypic shift of effector to regulatory T cells was observed. These results support the use of VIPR2-selective agonists as neuroprotective agents for PD treatment. SIGNIFICANCE STATEMENT: Vasoactive intestinal peptide receptor 2 can elicit immune transformation in a model of Parkinson's disease (PD). Such immunomodulatory capabilities can lead to neuroprotection by attenuating microglial activation and by slowing degradation of neuronal cell bodies and termini in MPTP-intoxicated mice. The protective mechanism arises from altering a Th1/Th2 immune cytokine response into an anti-inflammatory and neuronal sparing profile. These results are directly applicable for the development of novel PD therapies.


Subject(s)
Dopaminergic Neurons/drug effects , Dopaminergic Neurons/immunology , MPTP Poisoning/drug therapy , MPTP Poisoning/immunology , Neuroprotective Agents/therapeutic use , Oligopeptides/pharmacology , Receptors, Vasoactive Intestinal Peptide/agonists , Animals , CD4-Positive T-Lymphocytes/metabolism , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cytokines/metabolism , Humans , Immunohistochemistry , MPTP Poisoning/physiopathology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Oligopeptides/pharmacokinetics , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Spleen/cytology , Spleen/drug effects , Substantia Nigra/enzymology , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL