Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
Cell Rep Med ; 4(10): 101208, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37774703

ABSTRACT

Dyskinesia is involuntary movement caused by long-term medication with dopamine-related agents: the dopamine agonist 3,4-dihydroxy-L-phenylalanine (L-DOPA) to treat Parkinson's disease (L-DOPA-induced dyskinesia [LID]) or dopamine antagonists to treat schizophrenia (tardive dyskinesia [TD]). However, it remains unknown why distinct types of medications for distinct neuropsychiatric disorders induce similar involuntary movements. Here, we search for a shared structural footprint using magnetic resonance imaging-based macroscopic screening and super-resolution microscopy-based microscopic identification. We identify the enlarged axon terminals of striatal medium spiny neurons in LID and TD model mice. Striatal overexpression of the vesicular gamma-aminobutyric acid transporter (VGAT) is necessary and sufficient for modeling these structural changes; VGAT levels gate the functional and behavioral alterations in dyskinesia models. Our findings indicate that lowered type 2 dopamine receptor signaling with repetitive dopamine fluctuations is a common cause of VGAT overexpression and late-onset dyskinesia formation and that reducing dopamine fluctuation rescues dyskinesia pathology via VGAT downregulation.


Subject(s)
Dyskinesia, Drug-Induced , Parkinsonian Disorders , Mice , Animals , Dopamine Agonists/adverse effects , Levodopa/adverse effects , Dopamine , Antiparkinson Agents/adverse effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Oxidopamine/adverse effects , gamma-Aminobutyric Acid/adverse effects
2.
Mol Biol Rep ; 50(5): 4535-4549, 2023 May.
Article in English | MEDLINE | ID: mdl-36853472

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder caused by the degeneration of dopaminergic neurons. This leads to the pathogenesis of multiple basal ganglia-thalamomotor loops and diverse neurotransmission alterations. Dopamine replacement therapy, and on top of that, levodopa and l-3,4-dihydroxyphenylalanine (L-DOPA), is the gold standard treatment, while it develops numerous complications. Levodopa-induced dyskinesia (LID) is well-known as the most prominent side effect. Several studies have been devoted to tackling this problem. Studies showed that metabotropic glutamate receptor 5 (mGluR5) antagonists and 5-hydroxytryptamine receptor 1B (5HT1B) agonists significantly reduced LID when considering the glutamatergic overactivity and compensatory mechanisms of serotonergic neurons after L-DOPA therapy. Moreover, it is documented that these receptors act through an adaptor protein called P11 (S100A10). This protein has been thought to play a crucial role in LID due to its interactions with numerous ion channels and receptors. Lately, experiments have shown successful evidence of the effects of P11 blockade on alleviating LID greater than 5HT1B and mGluR5 manipulations. In contrast, there is a trace of ambiguity in the exact mechanism of action. P11 has shown the potential to be a promising target to diminish LID and prolong L-DOPA therapy in parkinsonian patients owing to further studies and experiments.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Humans , Levodopa/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Parkinson Disease/drug therapy , Basal Ganglia/metabolism , Basal Ganglia/pathology , Dopamine/metabolism , Dopamine/pharmacology , Dopamine/therapeutic use
3.
Mov Disord ; 37(8): 1693-1706, 2022 08.
Article in English | MEDLINE | ID: mdl-35535012

ABSTRACT

BACKGROUND: In advanced stages of Parkinson's disease (PD), dyskinesia and motor fluctuations become seriously debilitating and therapeutic options become scarce. Aberrant activity of striatal cholinergic interneurons (SCIN) has been shown to be critical to PD and dyskinesia, but the systemic administration of cholinergic medications can exacerbate extrastriatal-related symptoms. Thus, targeting the mechanisms causing pathological SCIN activity in severe PD with motor fluctuations and dyskinesia is a promising therapeutic alternative. METHODS: We used ex vivo electrophysiological recordings combined with pharmacology to study the alterations in intracellular signaling that contribute to the altered SCIN physiology observed in the 6-hydroxydopamine mouse model of PD treated with levodopa. RESULTS: The altered phenotypes of SCIN of parkinsonian mice during the "off levodopa" state resulting from aberrant Kir/leak and Kv1.3 currents can be rapidly reverted by acute inhibition of cAMP-ERK1/2 signaling. Inverse agonists that inhibit the ligand-independent activity of D5 receptors, like clozapine, restore Kv1.3 and Kir/leak currents and SCIN normal physiology in dyskinetic mice. CONCLUSION: Our work unravels a signaling pathway involved in the dysregulation of membrane currents causing SCIN hyperexcitability and burst-pause activity in parkinsonian mice treated with levodopa (l-dopa). These changes persist during off-medication periods due to tonic mechanisms that can be acutely reversed by pharmacological interventions. Thus, targeting the D5-cAMP-ERK1/2 signaling pathway selectively in SCIN may have therapeutic effects in PD and dyskinesia by restoring the normal SCIN function. © 2022 International Parkinson and Movement Disorder Society.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Animals , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Cholinergic Agents/therapeutic use , Corpus Striatum/metabolism , Disease Models, Animal , Dyskinesia, Drug-Induced/pathology , Interneurons/metabolism , Levodopa/pharmacology , Levodopa/therapeutic use , Mice , Oxidopamine/toxicity
4.
Neuroscience ; 492: 92-107, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35367290

ABSTRACT

Amantadine and clozapine have proved to reduce abnormal involuntary movements (AIMs) in preclinical and clinical studies of L-DOPA-Induced Dyskinesias (LID). Even though both drugs decrease AIMs, they may have different action mechanisms by using different receptors and signaling profiles. Here we asked whether there are differences in how they modulate neuronal activity of multiple striatal neurons within the striatal microcircuit at histological level during the dose-peak of L-DOPA in ex-vivo brain slices obtained from dyskinetic mice. To answer this question, we used calcium imaging to record the activity of dozens of neurons of the dorsolateral striatum before and after drugs administration in vitro. We also developed an analysis framework to extract encoding insights from calcium imaging data by quantifying neuronal activity, identifying neuronal ensembles by linking neurons that coactivate using hierarchical cluster analysis and extracting network parameters using Graph Theory. The results show that while both drugs reduce LIDs scores behaviorally in a similar way, they have several different and specific actions on modulating the dyskinetic striatal microcircuit. The extracted features were highly accurate in separating amantadine and clozapine effects by means of principal components analysis (PCA) and support vector machine (SVM) algorithms. These results predict possible synergistic actions of amantadine and clozapine on the dyskinetic striatal microcircuit establishing a framework for a bioassay to test novel antidyskinetic drugs or treatments in vitro.


Subject(s)
Clozapine , Dyskinesia, Drug-Induced , Amantadine/pharmacology , Animals , Antiparkinson Agents/pharmacology , Calcium , Clozapine/pharmacology , Corpus Striatum , Disease Models, Animal , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Levodopa/toxicity , Mice , Neurons , Oxidopamine/pharmacology
5.
Neurosci Lett ; 765: 136251, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34536508

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease caused by the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc), characterized by motor dysfunction. While PD symptoms are well treated with L-DOPA, continuous use can cause L-DOPA-induced dyskinesia (LID). We have previously demonstrated that sub-anesthetic ketamine attenuated LID development in rodents, measured by abnormal involuntary movements (AIMs), and reduced the density of maladaptive striatal dendritic mushroom spines. Microglia may play a role by phagocytosing maladaptive neuronal spines. In this exploratory study, we hypothesized that ketamine would prevent AIMs and change microglia ramified morphology - an indicator of a microglia response. Unilaterally 6-hydroxydopamine (6-OHDA)-lesioned rats were primed with daily injections of L-DOPA for 14 days, treated on days 0 and 7 for 10-hours with sub-anesthetic ketamine (i.p.), and we replicated that this attenuated LID development. We further extended our prior work by showing that while ketamine treatment did lead to an increase of striatal interleukin-6 in dyskinetic rats, indicating a modulation of an inflammatory response, it did not change microglia number or morphology in the dyskinetic striatum. Yet an increase of CD68 in the SNpc of 6-OHDA-lesioned hemispheres post-ketamine indicates increased microglia phagocytosis suggestive of a lingering microglial response to 6-OHDA injury in the SNpc pointing to possible anti-inflammatory action in the PD model in addition to anti-dyskinetic action. In conclusion, we provide further support for sub-anesthetic ketamine treatment of LID. The mechanisms of action for ketamine, specifically related to inflammation and microglia phagocytic functions, are emerging, and require further examination.


Subject(s)
Dyskinesia, Drug-Induced/prevention & control , Excitatory Amino Acid Antagonists/administration & dosage , Ketamine/administration & dosage , Levodopa/administration & dosage , Parkinson Disease/drug therapy , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/pathology , Humans , Levodopa/adverse effects , Male , Microglia/drug effects , Microglia/pathology , Phagocytosis/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Substantia Nigra/drug effects , Substantia Nigra/pathology
6.
Neurotoxicology ; 86: 104-113, 2021 09.
Article in English | MEDLINE | ID: mdl-34331976

ABSTRACT

Parkinson's disease (PD) is a progressive and self-propelling neurodegenerative disorder, which is characterized by motor symptoms, such as rigidity, tremor, slowness of movement and problems with gait. These symptoms become worse over time. To date, Dopamine (DA) replacement therapy with 3, 4-dihydroxy-l-phenylalanine (L-DOPA) is still the most effective pharmacotherapy for motor symptoms of PD. Unfortunately, motor fluctuations consisting of wearing-off effect actions and dyskinesia tend to occur in a few years of starting l-DOPA. Currently, l-DOPA-induced dyskinesia (LID) is troublesome and the pathogenesis of LID requires further investigation. Importantly, a new intervention for LID is imminent. Thus, this review mainly summarized the clinical features, risk factors and pathogenesis of LID to provide updatefor the development of therapeutic targets and new approaches for the treatment of LID.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Animals , Dopamine/metabolism , Dyskinesia, Drug-Induced/pathology , Humans , Parkinson Disease/pathology
7.
Parkinsonism Relat Disord ; 90: 8-14, 2021 09.
Article in English | MEDLINE | ID: mdl-34325387

ABSTRACT

INTRODUCTION: Levodopa-induced dyskinesia is a complication of levodopa therapy and negatively impacts the quality of life of patients. We aimed to elucidate white matter alterations in Parkinson's disease with levodopa-induced dyskinesia using advanced diffusion magnetic resonance imaging techniques. METHODS: The enrolled subjects included 26 clinically confirmed Parkinson's disease patients without levodopa-induced dyskinesia, 25 Parkinson's disease patients with levodopa-induced dyskinesia, and 23 healthy controls. Subjects were imaged using a 3-T magnetic resonance scanner. Diffusion tensor imaging, diffusion kurtosis imaging, and neurite orientation dispersion and density imaging findings were compared between groups with a group-wise whole brain approach and a region-of-interest analysis for each white matter tract. Additionally, logistic regression analysis was used to calculate odds ratios for levodopa-induced dyskinesia. RESULTS: Group-wise tract-based spatial statistical analysis revealed significant white matter differences in isotropic diffusion, complexity, or heterogeneity, and neurite density between healthy controls and Parkinson's disease patients without levodopa-induced dyskinesia and between patients with and without levodopa-induced dyskinesia. Region-of-interest analysis revealed similar alterations using a group-wise whole-brain approach in the external capsule, inferior fronto-occipital fasciculus, inferior longitudinal fasciculus, and uncinate fasciculus. These tracts had an odds ratio of approximately 2.3 for the presence of levodopa-induced dyskinesia. CONCLUSIONS: Our findings suggest that Parkinson's disease with levodopa-induced dyskinesia produces less white matter microstructural disruption, especially in temporal lobe fibers, than Parkinson's disease without levodopa-induced dyskinesia. These fibers has a more than 2-fold odds ratio for the presence of levodopa-induced dyskinesia and might be associated with the pathogenesis of the sequela.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Parkinson Disease/drug therapy , White Matter/pathology , Aged , Biomarkers/analysis , Case-Control Studies , Diffusion Magnetic Resonance Imaging , Dyskinesia, Drug-Induced/etiology , Female , Humans , Male , Nerve Fibers/pathology , Odds Ratio , Temporal Lobe/pathology , White Matter/ultrastructure
8.
Neurotox Res ; 39(3): 705-719, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33687725

ABSTRACT

Interferon-γ (IFN-γ) is a proinflammatory cytokine that activates glial cells. IFN-γ is increased in the plasma and brain of Parkinson's disease patients, suggesting its potential role in the disease. We investigated whether the IFN-γ deficiency could interfere with nigrostriatal degeneration induced by the neurotoxin 6-hydroxydopamine, L-DOPA-induced dyskinesia, and the neuroinflammatory features as astrogliosis, microgliosis, and induced nitric oxide synthase (iNOS) immunoreactivity induced by L-DOPA treatment. Wild type (WT) and IFN-γ knockout (IFN-γ/KO) mice received unilateral striatal microinjections of 6-hydroxydopamine. Animals were sacrificed 1, 3, 7, and 21 days after lesions. Additional group of WT and IFN-γ/KO parkinsonian mice, after 3 weeks of neurotoxin injection, received L-DOPA (intraperitoneally, for 21 days) resulting in dyskinetic-like behavior. Tyrosine hydroxylase immunostaining indicated the starting of dopaminergic lesion since the first day past toxin administration, progressively increased until the third day when it stabilized. There was no difference in the lesion and L-DOPA-induced dyskinesia intensity between WT and IFN-γ/KO mice. Remarkably, IFN-γ/KO mice treated with L-DOPA presented in the lesioned striatum an increase of iNOS and glial fibrilary acid protein (GFAP) density, compared with the WT group. Morphological analysis revealed the rise of astrocytes and microglia reactivity in IFN-γ/KO mice exibiting dyskinesia. In conclusion, IFN-γ/KO mice presented an intensification of the inflammatory reaction accompanying L-DOPA treatment and suggest that iNOS and GFAP increase, and the activation of astrocytes and microglia induced afterward L-DOPA treatment was IFN-γ independent events. Intriguingly, IFN-γ absence did not affect the degeneration of dopaminergic neurons or LID development.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/deficiency , Levodopa/toxicity , Parkinsonian Disorders/metabolism , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/pathology , Interferon-gamma/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidopamine/toxicity , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology
9.
J Neurosci ; 40(18): 3675-3691, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32238479

ABSTRACT

The transcription factor Nurr1 has been identified to be ectopically induced in the striatum of rodents expressing l-DOPA-induced dyskinesia (LID). In the present study, we sought to characterize Nurr1 as a causative factor in LID expression. We used rAAV2/5 to overexpress Nurr1 or GFP in the parkinsonian striatum of LID-resistant Lewis or LID-prone Fischer-344 (F344) male rats. In a second cohort, rats received the Nurr1 agonist amodiaquine (AQ) together with l-DOPA or ropinirole. All rats received a chronic DA agonist and were evaluated for LID severity. Finally, we performed single-unit recordings and dendritic spine analyses on striatal medium spiny neurons (MSNs) in drug-naïve rAAV-injected male parkinsonian rats. rAAV-GFP injected LID-resistant hemi-parkinsonian Lewis rats displayed mild LID and no induction of striatal Nurr1 despite receiving a high dose of l-DOPA. However, Lewis rats overexpressing Nurr1 developed severe LID. Nurr1 agonism with AQ exacerbated LID in F344 rats. We additionally determined that in l-DOPA-naïve rats striatal rAAV-Nurr1 overexpression (1) increased cortically-evoked firing in a subpopulation of identified striatonigral MSNs, and (2) altered spine density and thin-spine morphology on striatal MSNs; both phenomena mimicking changes seen in dyskinetic rats. Finally, we provide postmortem evidence of Nurr1 expression in striatal neurons of l-DOPA-treated PD patients. Our data demonstrate that ectopic induction of striatal Nurr1 is capable of inducing LID behavior and associated neuropathology, even in resistant subjects. These data support a direct role of Nurr1 in aberrant neuronal plasticity and LID induction, providing a potential novel target for therapeutic development.SIGNIFICANCE STATEMENT The transcription factor Nurr1 is ectopically induced in striatal neurons of rats exhibiting levodopa-induced dyskinesia [LID; a side-effect to dopamine replacement strategies in Parkinson's disease (PD)]. Here we asked whether Nurr1 is causing LID. Indeed, rAAV-mediated expression of Nurr1 in striatal neurons was sufficient to overcome LID-resistance, and Nurr1 agonism exacerbated LID severity in dyskinetic rats. Moreover, we found that expression of Nurr1 in l-DOPA naïve hemi-parkinsonian rats resulted in the formation of morphologic and electrophysiological signatures of maladaptive neuronal plasticity; a phenomenon associated with LID. Finally, we determined that ectopic Nurr1 expression can be found in the putamen of l-DOPA-treated PD patients. These data suggest that striatal Nurr1 is an important mediator of the formation of LID.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Parkinsonian Disorders/metabolism , Aged , Animals , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/pathology , Female , Humans , Male , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Sprague-Dawley
10.
Sci Rep ; 10(1): 2542, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054879

ABSTRACT

The dopamine D5 receptor (D5R) is a Gαs-coupled dopamine receptor belonging to the dopamine D1-like receptor family. Together with the dopamine D2 receptor it is highly expressed in striatal cholinergic interneurons and therefore is poised to be a positive regulator of cholinergic activity in response to L-DOPA in the dopamine-depleted parkinsonian brain. Tonically active cholinergic interneurons become dysregulated during chronic L-DOPA administration and participate in the expression of L-DOPA induced dyskinesia. The molecular mechanisms involved in this process have not been elucidated, however a correlation between dyskinesia severity and pERK expression in cholinergic cells has been described. To better understand the function of the D5 receptor and how it affects cholinergic interneurons in L-DOPA induced dyskinesia, we used D5R knockout mice that were rendered parkinsonian by unilateral 6-OHDA injection. In the KO mice, expression of pERK was strongly reduced indicating that activation of these cells is at least in part driven by the D5 receptor. Similarly, pS6, another marker for the activity status of cholinergic interneurons was also reduced. However, mice lacking D5R exhibited slightly worsened locomotor performance in response to L-DOPA and enhanced LID scores. Our findings suggest that D5R can modulate L-DOPA induced dyskinesia and is a critical activator of CINs via pERK and pS6.


Subject(s)
Dopamine/metabolism , Dyskinesia, Drug-Induced/genetics , Levodopa/adverse effects , Receptors, Dopamine D5/genetics , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dyskinesia, Drug-Induced/pathology , Humans , Interneurons/drug effects , Interneurons/metabolism , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Oxidopamine/pharmacology , Parkinson Disease/genetics , Parkinson Disease/pathology
11.
Mol Imaging Biol ; 22(3): 634-642, 2020 06.
Article in English | MEDLINE | ID: mdl-31392531

ABSTRACT

PURPOSE: The development of L-DOPA-induced dyskinesia (LID) is one of the most severe side effects of chronic L-DOPA treatment in Parkinson's disease patients. [11C]DASB positron emission tomography (PET) provides a prominent tool to visualize and quantify serotonin transporter (SERT) pathology in vivo in patients and in animal models. To evaluate the effect of chronic L-DOPA treatment on SERT availability in an animal model of LID, we performed a longitudinal PET study. PROCEDURES: Rats received a unilateral 6-hydroxydopamine (6-OHDA) lesion, and striatal and extrastriatal SERT expression levels were studied with [11C]DASB, a marker of SERT availability, before and after daily treatment with L-DOPA. Dyskinesias were evaluated at different time points over a period of 21 days. RESULTS: [11C]DASB binding was found to be decreased after 6-OHDA lesions in the striatum, cortex, and hippocampus 5 weeks after 6-OHDA injection in the lesioned hemisphere of the rat brain. Chronic L-DOPA priming resulted in a relative preservation of SERT availability in the lesioned and healthy hemisphere compared to baseline measurements. CONCLUSIONS: Our longitudinal PET data support a preservation of SERT availability after the induction of L-DOPA-induced dyskinesia, which is in line with previous reports in dyskinetic PD patients.


Subject(s)
Brain/pathology , Carbon Radioisotopes/pharmacokinetics , Dyskinesia, Drug-Induced/diagnostic imaging , Motor Activity/physiology , Positron-Emission Tomography/methods , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/chemistry , Disease Models, Animal , Dopamine Agents/toxicity , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/pathology , Levodopa/toxicity , Male , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley
12.
Mol Neurobiol ; 57(2): 736-751, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31468338

ABSTRACT

Dopamine and serotonin in the basal ganglia interact in a bidirectional manner. On the one hand, serotonin (5-HT) receptors regulate the effects of dopamine agonists on several levels, ranging from molecular signaling to behavior. These interactions include 5-HT receptor-mediated facilitation of dopamine receptor-induced gene regulation in striatal output pathways, which involves the 5-HT1B receptor and others. Conversely, there is evidence that dopamine action by psychostimulants regulates 5-HT1B receptor expression in the striatum. To further investigate the effects of dopamine and agonists on 5-HT receptors, we assessed the expression of 5-HT1B and other serotonin receptor subtypes in the striatum after unilateral dopamine depletion by 6-OHDA and subsequent treatment with L-DOPA (5 mg/kg; 4 weeks). Neither dopamine depletion nor L-DOPA treatment produced significant changes in 5-HT2C, 5-HT4, or 5-HT6 receptor expression in the striatum. In contrast, the 6-OHDA lesion caused a (modest) increase in 5-HT1B mRNA levels throughout the striatum. Moreover, repeated L-DOPA treatment markedly further elevated 5-HT1B expression in the dopamine-depleted striatum, an effect that was most robust in the sensorimotor striatum. A minor L-DOPA-induced increase in 5-HT1B expression was also seen in the intact striatum. These changes in 5-HT1B expression mimicked changes in the expression of neuropeptide markers (dynorphin, enkephalin mRNA) in striatal projection neurons. After repeated L-DOPA treatment, the severity of L-DOPA-induced dyskinesias and turning behavior was positively correlated with the increase in 5-HT1B expression in the associative, but not sensorimotor, striatum ipsilateral to the lesion, suggesting that associative striatal 5-HT1B receptors may play a role in L-DOPA-induced behavioral abnormalities.


Subject(s)
Corpus Striatum/metabolism , Dopamine/deficiency , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Receptor, Serotonin, 5-HT1B/metabolism , Animals , Behavior, Animal , Dynorphins/metabolism , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/pathology , Enkephalins/metabolism , Gene Expression Regulation , Male , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
13.
Pharmacogenomics ; 20(17): 1199-1223, 2019 11.
Article in English | MEDLINE | ID: mdl-31686592

ABSTRACT

Identifying biomarkers which can be used as a diagnostic tool is a major objective of pharmacogenetic studies. Most mental and many neurological disorders have a compiled multifaceted nature, which may be the reason why this endeavor has hitherto not been very successful. This is also true for tardive dyskinesia (TD), an involuntary movement complication of long-term treatment with antipsychotic drugs. The observed associations of specific gene variants with the prevalence and severity of a disorder can also be applied to try to elucidate the pathogenesis of the condition. In this paper, this strategy is used by combining pharmacogenetic knowledge with theories on the possible role of a dysfunction of specific cellular elements of neostriatal parts of the (dorsal) extrapyramidal circuits: various glutamatergic terminals, medium spiny neurons, striatal interneurons and ascending monoaminergic fibers. A peculiar finding is that genetic variants which would be expected to increase the neostriatal dopamine concentration are not associated with the prevalence and severity of TD. Moreover, modifying the sensitivity to glutamatergic long-term potentiation (and excitotoxicity) shows a relationship with levodopa-induced dyskinesia, but not with TD. Contrasting this, TD is associated with genetic variants that modify vulnerability to oxidative stress. Reducing the oxidative stress burden of medium spiny neurons may also be the mechanism behind the protective influence of 5-HT2 receptor antagonists. It is probably worthwhile to discriminate between neostriatal matrix and striosomal compartments when studying the mechanism of TD and between orofacial and limb-truncal components in epidemiological studies.


Subject(s)
Dyskinesia, Drug-Induced/genetics , Oxidative Stress/drug effects , Schizophrenia/drug therapy , Tardive Dyskinesia/genetics , Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Dopamine/genetics , Dopamine/metabolism , Dyskinesia, Drug-Induced/pathology , Excitatory Amino Acid Agents/adverse effects , Excitatory Amino Acid Agents/therapeutic use , Humans , Neostriatum/drug effects , Neostriatum/pathology , Pharmacogenetics , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Receptors, Serotonin, 5-HT2/genetics , Schizophrenia/complications , Schizophrenia/genetics , Schizophrenia/pathology , Serotonin 5-HT2 Receptor Antagonists/adverse effects , Serotonin 5-HT2 Receptor Antagonists/therapeutic use , Spinal Cord/drug effects , Spinal Cord/pathology , Tardive Dyskinesia/chemically induced , Tardive Dyskinesia/pathology
14.
Aging (Albany NY) ; 11(21): 9661-9671, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31699957

ABSTRACT

Abnormal dopaminergic modulation of the cortico-basal ganglia motor loops results in the emergence of levodopa-induced dyskinesia (LID). We focused on alterations in the gray matter (GM) volume and the cortical thickness of the brain, especially in cortico-basal ganglia motor loops, in Parkinson's disease (PD) with diphasic dyskinesia. 48 PD patients with diphasic dyskinesia, 60 PD patients without dyskinesia and 48 healthy controls (HC) were included. Voxel-based morphometry (VBM) was applied to get GM images from MRI brain images. FreeSurfer was used to get cortical thickness. Distinct analyses of covariance (ANCOVA) and linear contrasts were performed for early- and late-onset PD groups. The severity of diphasic dyskinesia was evaluated by the Unified Dyskinesia Rating Scale (UDysRS). Finally, the correlations between mean volumes of clusters showing differences and the UDysRS scores were performed by Pearson's correlation. The GM volumes of precentral gyri were increased in PD patients with diphasic dyskinesia when compared with those without dyskinesia, which were positively correlated with UDysRS scores in PD patients with diphasic dyskinesia. However, there was no significant difference in cortical thickness among groups. The increased precentral gyri GM volumes might be associated with the pathogenesis and the severity of diphasic dyskinesia.


Subject(s)
Dyskinesia, Drug-Induced/pathology , Frontal Lobe/pathology , Gray Matter/pathology , Parkinson Disease/pathology , Adult , Age of Onset , Case-Control Studies , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Female , Frontal Lobe/diagnostic imaging , Gray Matter/diagnostic imaging , Humans , Levodopa/adverse effects , Magnetic Resonance Imaging , Male , Middle Aged , Parkinson Disease/complications , Parkinson Disease/drug therapy
15.
Ann Clin Transl Neurol ; 6(11): 2251-2260, 2019 11.
Article in English | MEDLINE | ID: mdl-31643140

ABSTRACT

OBJECTIVE: The precise pathogenesis or neural correlates underlying levodopa-induced dyskinesia (LID) remains poorly understood. There is growing evidence of the involvement of the cerebellum in Parkinson's disease (PD). The present study evaluated the role of motor cerebellar connectivity in determining vulnerability to LID. METHODS: We enrolled 25 de novo patients with PD who developed LID within 5 years of levodopa treatment, 26 propensity score-matched PD patients who had not developed LID, and 24 age- and sex-matched healthy controls. We performed a comparative analysis of resting-state functional connectivity (FC) between the motor cerebellum and whole brain between the groups. RESULTS: The patients with PD had increased FC bewteen the motor cerebellum and posterior cortical and cerebellar regions, while no gray matter regions had decreased FC with the motor cerebellum compared to the control participant. The patients with PD who were vulnerable to the development of LID had a significantly higher FC between the motor cerebellum lobule VIIIb and the left inferior frontal gyrus than those who were resistant to LID development. The connectivity of the motor cerebellum and left inferior frontal gyrus was negatively correlated with the latency from PD onset to the occurrence of LID. INTERPRETATION: Increased FC between the motor cerebellum and left inferior frontal gyrus in de novo patients with PD could be an important determinant of vulnerability to LID.


Subject(s)
Antiparkinson Agents/adverse effects , Cerebellum/pathology , Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Neural Pathways/pathology , Parkinson Disease/drug therapy , Aged , Dyskinesia, Drug-Induced/etiology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Retrospective Studies
16.
Proc Natl Acad Sci U S A ; 116(37): 18664-18672, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31455727

ABSTRACT

Long-term dopamine (DA) replacement therapy in Parkinson's disease (PD) leads to the development of abnormal involuntary movements known as l-Dopa-induced dyskinesia (LID). The transcription factor ΔFosB that is highly up-regulated in the striatum following chronic l-Dopa exposure may participate in the mechanisms of altered neuronal responses to DA generating LID. To identify intrinsic effects of elevated ΔFosB on l-Dopa responses, we induced transgenic ΔFosB overexpression in the striatum of parkinsonian nonhuman primates kept naïve of l-Dopa treatment. Elevated ΔFosB levels led to consistent appearance of LID since the initial acute l-Dopa tests. In line with this motor response, striatal projection neurons (SPNs) responded to DA with changes in firing frequency that reversed at the peak of the motor response, and these unstable SPN activity changes in response to DA are typically associated with the emergence of LID. Transgenic ΔFosB overexpression also induced up-regulation of other molecular markers of LID. These results support an autonomous role of striatal ΔFosB in the adaptive mechanisms altering motor responses to chronic DA replacement in PD.


Subject(s)
Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Neostriatum/pathology , Parkinson Disease/drug therapy , Proto-Oncogene Proteins c-fos/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Humans , Macaca fascicularis , Male , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fos/genetics , Up-Regulation/drug effects
17.
Sci Rep ; 9(1): 9248, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31239456

ABSTRACT

The objective of this study was to investigate the impact of levodopa therapy-induced complications on the quality of life (QoL) of Parkinson's disease (PD) patients in Singapore over a 1-year follow-up period. 274 PD patients were prospectively recruited, of which 78 patients completed the follow-up. Patients were evaluated on: (1) motor symptoms, (2) non-motor symptoms, (3) levodopa therapy-induced complications and (4) QoL. Levodopa-induced complications including dyskinesia and OFF symptoms occurred in 13.5% and 55.9% of the study population, respectively. In patients who completed the 1-year follow-up, there was a trend suggestive of increasing dyskinesia duration, more disabling dyskinesia as well as longer, more sudden and unpredictable OFF periods. There was a significant decline in the overall QoL at follow-up, in particular, activities of daily living, emotional well-being, cognition and communication domains were the most affected. The multivariable analysis demonstrated that worsening of UPDRS IV total score over 1-year interval was associated with worsening in PDQ-Summary Index score (d = 0.671, p = 0.014). In conclusion, levodopa-induced complications had significant adverse impacts on QoL. This study substantiates the importance for clinicians to closely monitor and promptly manage levodopa therapy-induced complications that may arise in patients.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/epidemiology , Levodopa/adverse effects , Parkinson Disease/drug therapy , Quality of Life , Severity of Illness Index , Activities of Daily Living , Aged , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/pathology , Female , Humans , Incidence , Longitudinal Studies , Male , Middle Aged , Parkinson Disease/pathology , Singapore/epidemiology , Treatment Outcome
18.
BMC Neurosci ; 20(1): 5, 2019 Feb 13.
Article in English | MEDLINE | ID: mdl-30760214

ABSTRACT

BACKGROUND: Accurately assessing promising therapeutic interventions for human diseases depends, in part, on the reproducibility of preclinical disease models. With the development of transgenic mice, the rapid adaptation of a 6-OHDA mouse model of Parkinson's disease that was originally described for the use in rats has come with a lack of a comprehensive characterization of lesion progression. In this study we therefore first characterised the time course of neurodegeneration in the substantia nigra pars compacta and striatum over a 4 week period following 6-OHDA injection into the medial forebrain bundle of mice. We then utilised the model to assess the anti-dyskinetic efficacy of recombinant activin A, a putative neuroprotectant and anti-inflammatory that is endogenously upregulated during the course of Parkinson's disease. RESULTS: We found that degeneration of fibers in the striatum was fully established within 1 week following 6-OHDA administration, but that the loss of neurons continued to progress over time, becoming fully established 3 weeks after the 6-OHDA injection. In assessing the anti-dyskinetic efficacy of activin A using this model we found that treatment with activin A did not significantly reduce the severity, or delay the time-of-onset, of dyskinesia. CONCLUSION: First, the current study concludes that a 3 week duration is required to establish a complete lesion of the nigrostriatal tract following 6-OHDA injection into the medial forebrain bundle of mice. Second, we found that activin A was not anti-dyskinetic in this model.


Subject(s)
Activins/pharmacology , Dyskinesia, Drug-Induced/drug therapy , Medial Forebrain Bundle/physiopathology , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Disease Progression , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Levodopa/pharmacology , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/pathology , Mice, Inbred C57BL , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Random Allocation , Treatment Failure
19.
Neurotox Res ; 35(3): 575-583, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30729450

ABSTRACT

Organophosphates (OPs) inhibit cholinesterase and hyperactivate the acetylcholinergic nervous system in the brain, causing motor disorders (e.g., tremor and seizures). Here, we performed behavioral and immunohistochemical studies in mice and rats to investigate the tremorgenic mechanism of paraoxon, an active metabolite of parathion. Treating animals with paraoxon (0.15-0.6 mg/kg, i.p.) elicited kinetic tremor in a dose-dependent manner. Expressional analysis of Fos protein, a biomarker of neural excitation, revealed that a tremorgenic dose of paraoxon (0.6 mg/kg) significantly and region-specifically elevated Fos expression in the cerebral cortex (e.g., sensory cortex), hippocampal CA1, globus pallidus, medial habenula, and inferior olive (IO) among 48 brain regions examined. A moderate increase in Fos expression was also observed in the dorsolateral striatum while the change was not statistically significant. Paraoxon-induced tremor was inhibited by the nicotinic acetylcholine (nACh) receptor antagonist mecamylamine (MEC), but not affected by the muscarinic acetylcholine receptor antagonist trihexyphenidyl (THP). In addition, paraoxon-induced Fos expression in the IO was also antagonized by MEC, but not by THP, and lesioning of the IO markedly suppressed tremorgenic action of paraoxon. The present results suggest that OPs elicit kinetic tremor at least partly by activating IO neurons via nACh receptors.


Subject(s)
Brain/drug effects , Brain/metabolism , Dyskinesia, Drug-Induced/metabolism , Paraoxon/adverse effects , Tremor/chemically induced , Tremor/metabolism , Animals , Brain/pathology , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Gene Expression/drug effects , Male , Mecamylamine/pharmacology , Mice , Muscarinic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nicotinic Antagonists/pharmacology , Oncogene Proteins v-fos/metabolism , Rats , Receptors, Nicotinic/metabolism , Tremor/drug therapy , Tremor/pathology , Trihexyphenidyl/pharmacology
20.
Neurotox Res ; 35(3): 635-653, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30673988

ABSTRACT

L-dopa is still considered as the gold standard therapy for Parkinson's disease (PD); however, L-dopa-induced dyskinesia (LID) is a serious complication of long-term L-dopa treatment. The present study investigated the therapeutic potential of sitagliptin and liraglutide in comparisons with L-dopa against PD. In addition, their capacity to modulate L-dopa dose and/or side effects was investigated, too. Rats were injected with rotenone (3 mg/kg/day, s.c.) for 10 consecutive days to induce the experimental PD. The rotenone-treated rats were administered sitagliptin (30 mg/kg/day, p.o.) and liraglutide (50 µg/kg, s.c.) for 16 days either alone or together with L-dopa/carbidopa (50/25 mg/kg/day, i.p.). Scoring of LID was done on days 2, 4, 8, 12, and 16 in all L-dopa-treated groups. Twenty-four hours after the last administered dose of tested drugs, the behavior of rats in each group was screened by using the open-field test. Sitagliptin and liraglutide revealed marked attenuation of LID scores; in addition, they markedly increased animals' motor performance. Moreover, they preserved substantia nigra pars compacta (SNpc) tyrosine hydroxylase (TH) and vesicular monoamine transporter 2-positive (VMAT2) cells with prominent increase of the striatal dopamine (DA) content. On the other hand, they significantly decreased nigral neuromelanin (NM)-positive cells, activated microglia, gliosis, and other pathological changes. In conclusion, sitagliptin and liraglutide could be a promising therapeutic challenger in PD, modifying L-dopa effect and/or allowing the use of L-dopa with fewer side effects.


Subject(s)
Antiparkinson Agents/pharmacology , Carbidopa/pharmacology , Dyskinesia, Drug-Induced/prevention & control , Levodopa/pharmacology , Liraglutide/pharmacology , Parkinsonian Disorders/drug therapy , Sitagliptin Phosphate/pharmacology , Animals , Antiparkinson Agents/adverse effects , Carbidopa/adverse effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Drug Combinations , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Liraglutide/adverse effects , Male , Motor Activity/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , Random Allocation , Rats, Wistar , Rotenone , Sitagliptin Phosphate/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...