Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
1.
Neurosci Lett ; 825: 137706, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38431040

ABSTRACT

INTRODUCTION: Levodopa-induced dyskinesia (LID) is a debilitating motor feature in a subset of patients with Parkinson's disease (PD) after prolonged therapeutic administration of levodopa. Preliminary animal and human studies are suggestive of a key role of dopamine type 3 (D3) receptor polymorphism (Ser9Gly; rs6280) in LID. Its contribution to development of LID among Indian PD patients has remained relatively unexplored and merits further investigation. METHODS AND MATERIALS: 200 well-characterised PD patients (100 without LID and 100 with LID) and 100 age-matched healthy controls were recruited from the outpatient department of Institute of Neurosciences Kolkata. MDS-UPDRS (Unified Parkinson's Disease Rating Scale from International Movement Disorder Society) Part III and AIMS (abnormal involuntary movement scale) were performed for estimation of severity of motor features and LID respectively in the ON state of the disease. Participants were analysed for the presence of Ser9Gly single nucleotide variant (SNV) (rs6280) by polymerase chain reaction followed by restriction fragment length polymorphism techniques. RESULTS: The frequency of AA genotype (serine type) was more frequently present in PD patients with LID compared to PD patients without LID (50 % vs 28 %; P = 0.002; OR = 2.57, 95 % CI: 1.43 - 4.62). The abnormal involuntary movement scale score was significantly higher in PD patients with AA genotype compared to carriers of glycine allele (AG + GG) (4.08 ± 3.35; P = 0.002). CONCLUSION: We observed a significant association of serine type SNV (rs6280) in D3 receptor gene in a cohort of PD patients with LID from India. More severe motor severity was found in patients with glycine substitution of the same SNV. The current study emphasised the role of D3 receptor in the pathogenesis of LID.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Animals , Humans , Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/drug therapy , Glycine , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Receptors, Dopamine D3/genetics , Serine/genetics
2.
CNS Neurosci Ther ; 29(10): 2925-2939, 2023 10.
Article in English | MEDLINE | ID: mdl-37101388

ABSTRACT

BACKGROUND: Levodopa (L-DOPA) is considered the most reliable drug for treating Parkinson's disease (PD) clinical symptoms. Regrettably, long-term L-DOPA therapy results in the emergence of drug-induced abnormal involuntary movements (AIMs) in most PD patients. The mechanisms underlying motor fluctuations and dyskinesia induced by L-DOPA (LID) are still perplexing. METHODS: Here, we first performed the analysis on the microarray data set (GSE55096) from the gene expression omnibus (GEO) repository and identified the differentially expressed genes (DEGs) using linear models for microarray analysis (Limma) R packages from the Bioconductor project. 12 genes (Nr4a2, Areg, Tinf2, Ptgs2, Pdlim1, Tes, Irf6, Tgfb1, Serpinb2, Lipg, Creb3l1, Lypd1) were found to be upregulated. Six genes were validated on quantitative polymerase chain reaction and subsequently, Amphiregulin (Areg) was selected (based on log2 fold change) for further experiments to unravel its involvement in LID. Areg LV_shRNA was used to knock down Areg to explore its therapeutic role in the LID model. RESULTS: Western blotting and immunofluorescence results show that AREG is significantly expressed in the LID group relative to the control. Dyskinetic movements in LID mice were alleviated by Areg knockdown, and the protein expression of delta FOSB, the commonly attributable protein in LID, was decreased. Moreover, Areg knockdown reduced the protein expression of P-ERK. In order to ascertain whether the inhibition of the ERK pathway (a common pathway known to mediate levodopa-induced dyskinesia) could also impede Areg, the animals were injected with an ERK inhibitor (PD98059). Afterward, the AIMs, AREG, and ERK protein expression were measured relative to the control group. A group treated with ERK inhibitor had a significant decrease of AREG and phosphorylated ERK protein expression relative to the control group. CONCLUSION: Taken together, our results indicate unequivocal involvement of Areg in levodopa-induced dyskinesia, thus a target for therapy development.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Mice , Animals , Levodopa/adverse effects , Parkinson Disease/drug therapy , Oxidopamine/toxicity , Antiparkinson Agents/therapeutic use , Amphiregulin/genetics , Amphiregulin/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/metabolism , Disease Models, Animal
3.
Neurosci Lett ; 801: 137140, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36813078

ABSTRACT

INTRODUCTION: Levodopa-induced dyskinesia (LID) is a common motor complication in Parkinson's disease (PD). Several genes in the levodopa metabolic pathway, such as COMT, DRDx and MAO-B, were reported associated with LID. However, there has been no systematic analyses between common variants in levodopa metabolic pathway genes and LID in a large sample of the Chinese population. METHODS: Through the whole exome sequencing (WES) and target region sequencing, we aimed to explore the potential associations between common single nucleotide polymorphisms (SNPs) in the levodopa metabolic pathway and LID in Chinese PD individuals. Five hundred and two PD individuals were enrolled in our study, among them, 348 individuals underwent WES, and 154 individuals underwent target region sequencing. We acquired the genetic profile of 11 genes including COMT, DDC, DRD1-5, SLC6A3, TH and MAO-A/B. We established a stepwise strategy to filter SNPs, which finally included 34 SNPs in our analyses. And we used a two-stage study, with discovery (348 individuals with WES) and the replication (all 502 individuals) to confirm our findings. RESULTS: Among the 502 PD individuals, 104 (20.7%) were diagnosed with LID. In the discovery stage, we found that COMT rs6269, DRD2 rs6275 and DRD2 rs1076560 were associated with LID. In the replication stage, associations between the three above-mentioned SNPs and LID were still present in all 502 individuals. CONCLUSION: We demonstrated that in the Chinese population, COMT rs6269, DRD2 rs6275 and rs1076560 were significantly associated with LID. And rs6275 was reported associated with LID for the first time.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Humans , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/genetics , Polymorphism, Single Nucleotide , Monoamine Oxidase/genetics
4.
Commun Biol ; 5(1): 872, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008531

ABSTRACT

Although levodopa is the most effective medication for Parkinson's disease, long-term levodopa treatment is largely compromised due to late motor complications, including levodopa-induced dyskinesia (LID). However, the genetic basis of LID pathogenesis has not been fully understood. Here, we discover genes pathogenic for LID using Drosophila genetics and behavioral analyses combined with genome-wide association studies on 578 patients clinically diagnosed with LID. Similar to the therapeutic effect of levodopa in patients, acute levodopa treatments restore the motor defect of Parkinson's disease model flies, while prolonged treatments cause LID-related symptoms, such as increased yawing, freezing and abrupt acceleration of locomotion. These symptoms require dopamine 1-like receptor 1 and are induced by neuronal overexpression of the receptor. Among genes selected from our analyses in the patient genome, neuronal knockdown of adenylyl cyclase 2 suppresses the levodopa-induced phenotypes and the receptor overexpression-induced symptoms in Drosophila. Together, our study provides genetic insights for LID pathogenesis through the D1-like receptor-adenylyl cyclase 2 signaling axis.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Animals , Drosophila/genetics , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/genetics , Genome-Wide Association Study , Genomics , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/genetics
5.
Psychiatry Res ; 317: 114791, 2022 11.
Article in English | MEDLINE | ID: mdl-36030699

ABSTRACT

Genome-wide association studies (GWAS) have identified some variants for movement-related adverse antipsychotic effects (MAAE), while how these variants confer MAAE remains unclear. We used the probabilistic Mendelian randomization (PMR) method to identify candidate proteins for MAAE by integrating MAAE GWASs and protein quantitative trait loci (pQTL) data. An independent pQTL data from the Banner project and brain-derived eQTL data were used to perform confirmatory PMR. A total of 56 proteins were identified as candidate targets for MAAE after false discovery rates (FDR) correction, such as GRIN2B, ADRA1A, and PED4B. 12 genes were replicated in the confirmatory PMR, and 18 genes had consistent evidence at the transcript level. Furthermore, we investigated the associations between candidate proteins and the motor symptoms of Parkinson's disease (PD). There were 24, 38, and 10 candidate proteins that were significantly associated with PD, PD motor subtypes, and PD motor progression, respectively. Enrichment analysis identified 34 GO terms and 17 pathways that may be involved in MAAE, such as glutamatergic synapse, glutamate receptor complex, and GABAergic synapse. Our study identified multiple candidate genes and pathways that were associated with MAAE, providing new insights into the biological mechanism of MAAE and targets for further mechanistic and therapeutic studies.


Subject(s)
Antipsychotic Agents , Dyskinesia, Drug-Induced , Parkinson Disease , Proteome , Humans , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Genetic Predisposition to Disease , Genome-Wide Association Study , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Polymorphism, Single Nucleotide/genetics , Proteome/genetics , Proteome/metabolism , Mendelian Randomization Analysis , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/metabolism , Quantitative Trait Loci
8.
Gene Ther ; 28(12): 760-770, 2021 12.
Article in English | MEDLINE | ID: mdl-33707771

ABSTRACT

L-Dopa-induced dyskinesia (LID) is associated with the upregulation of striatal ∆FosB in animal models and patients with Parkinson's disease (PD). A mechanistic role of ∆FosB is suspected because its transgenic overexpression leads to the early appearance of LID in rodents and primates. This study in rodents is aimed at exploring the therapeutic potential of striatal ∆FosB gene suppression to control LID in patients with PD. To determine the effect of reducing striatal ∆FosB expression, we used RNAi gene knockdown in a rat model of PD and assessed abnormal involuntary movements (AIMs) in response to L-Dopa. Rats with dopamine depletion received striatal injections of rAAV-∆FosB shRNA or a control virus before exposure to chronic L-Dopa treatment. The development of AIMs during the entire L-Dopa treatment period was markedly inhibited by ∆FosB gene knockdown and its associated molecular changes. The antiparkinsonian action of L-Dopa was unchanged by ∆FosB gene knockdown. These results suggest a major role for ∆FosB in the development of LID and support exploring strategies to reduce striatal ∆FosB levels in patients with PD.


Subject(s)
Dyskinesia, Drug-Induced , Levodopa , Animals , Antiparkinson Agents/adverse effects , Antiparkinson Agents/metabolism , Corpus Striatum/metabolism , Disease Models, Animal , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/genetics , Humans , Levodopa/adverse effects , Levodopa/metabolism , Oxidopamine , Rats
9.
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
10.
Parkinsonism Relat Disord ; 84: 52-60, 2021 03.
Article in English | MEDLINE | ID: mdl-33561612

ABSTRACT

INTRODUCTION: Levodopa-induced dyskinesia frequently complicates long-term Parkinson's disease. More in-depth knowledge regarding the role of genetic factors in dyskinesia development may be important to identify parkinsonian patients who are more prone to developing dyskinesia and clarify the molecular mechanisms underlying this condition. For this reason, we systematically reviewed studies investigating genetic factors involved in dyskinesia. METHODS: A systematic search of genetic factors in Parkinson's disease dyskinesia was performed using the MEDLINE (through PubMed up to June 2019) and EMBASE databases according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. A meta-analysis was conducted using a random effect model. RESULTS: The literature search retrieved 33 studies assessing genes and variants possibly associated with dyskinesia in Parkinson's disease. The studies were published between 1984 and 2019 and included a total of 27,092 subjects of different ethnicities. Overall, 37 genes were analyzed in the studies reviewed, of which 22 were possibly associated with dyskinesia. The studies reported a total of 158 variants, of which 94 were possibly related to dyskinesia. CONCLUSION: The studies reviewed demonstrated inconsistent results, possibly due to differences in screening methods and in the comparison of clinical data in a large variety of genetically- and ethnically-diverse populations. The meta-analysis failed to demonstrate any association between the rs6280 in the DRD3 gene, rs1799836 in the MAO-B, rs4680 in the COMT gene, rs34637584 in the LRRK2 gene and LID susceptibility. The role of genetic factors in LID susceptibility is still unclear and further studies are required.


Subject(s)
Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/genetics , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Humans
11.
Exp Neurol ; 336: 113534, 2021 02.
Article in English | MEDLINE | ID: mdl-33249031

ABSTRACT

Parkinson's Disease (PD) is symptomatically managed with L-DOPA but chronic use results in L-DOPA-induced dyskinesia (LID) characterized by abnormal involuntary movements (AIMs). In LID, dopamine D3 receptors (D3R) are upregulated on D1 receptor (D1R)-bearing medium spiny neurons where the can synergistically drive downstream signaling and motor behaviors. Despite evidence implying D1R-D3R cooperativity in LID, the dyskinesiogenic role of D3R has never been directly tested. To this end, we developed a specific cre-dependent microRNA (miRNA) to irreversibly prevent D3R upregulation in D1R striatal cells. D1-Cre rats received unilateral 6-hydroxydopamine lesions. Three weeks later, rats received an adeno-associated virus expressing either D3R miRNA or a scrambled (SCR) miRNA delivered into the striatum. After 4 weeks, rats received chronic L-DOPA (6 mg/kg) or vehicle. AIMs development and motor behaviors were assayed throughout treatment. At the conclusion of the experiment, efficacy and fidelity of the miRNA strategy was analyzed using in situ hybridization (ISH). ISH analyses demonstrated that D1R+/D3R+ cells were upregulated in LID and that the selective D3R miRNA reduced D1R+/D3R+ co-expression. Importantly, silencing of D3R also significantly attenuated LID development without impacting L-DOPA efficacy or other locomotion. These data highlight a dyskinesiogenic role of D3R within D1R cells in LID and highlight aberrant D1R-D3R interactions as targets of LID management.


Subject(s)
Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/prevention & control , Levodopa/adverse effects , Neostriatum/pathology , Receptors, Dopamine D1/genetics , Receptors, Dopamine D3/genetics , Animals , Behavior, Animal , Dyskinesia, Drug-Induced/psychology , Female , Genetic Therapy , Hydroxydopamines , Male , MicroRNAs/genetics , Neostriatum/metabolism , Psychomotor Performance , Rats
12.
J Neuroinflammation ; 17(1): 243, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32807174

ABSTRACT

BACKGROUND/AIMS: The renin-angiotensin system (RAS) is altered in Parkinson's disease (PD), a disease due to substantia nigra neurodegeneration and whose dopamine-replacement therapy, using the precursor levodopa, leads to dyskinesias as the main side effect. Angiotensin AT1 and AT2 receptors, mainly known for their role in regulating water homeostasis and blood pressure and able to form heterodimers (AT1/2Hets), are present in the central nervous system. We assessed the functionality and expression of AT1/2Hets in Parkinson disease (PD). METHODS: Immunocytochemistry was used to analyze the colocalization between angiotensin receptors; bioluminescence resonance energy transfer was used to detect AT1/2Hets. Calcium and cAMP determination, MAPK activation, and label-free assays were performed to characterize signaling in homologous and heterologous systems. Proximity ligation assays were used to quantify receptor expression in mouse primary cultures and in rat striatal sections. RESULTS: We confirmed that AT1 and AT2 receptors form AT1/2Hets that are expressed in cells of the central nervous system. AT1/2Hets are novel functional units with particular signaling properties. Importantly, the coactivation of the two receptors in the heteromer reduces the signaling output of angiotensin. Remarkably, AT1/2Hets that are expressed in both striatal neurons and microglia make possible that candesartan, the antagonist of AT1, increases the effect of AT2 receptor agonists. In addition, the level of striatal expression increased in the unilateral 6-OH-dopamine lesioned rat PD model and was markedly higher in parkinsonian-like animals that did not become dyskinetic upon levodopa chronic administration if compared with expression in those that became dyskinetic. CONCLUSION: The results indicate that boosting the action of neuroprotective AT2 receptors using an AT1 receptor antagonist constitutes a promising therapeutic strategy in PD.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Renin-Angiotensin System/physiology , Substantia Nigra/metabolism , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Dyskinesia, Drug-Induced/genetics , HEK293 Cells , Humans , Levodopa , Mice , Phosphorylation , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Signal Transduction/physiology
13.
Genes Brain Behav ; 19(8): e12690, 2020 11.
Article in English | MEDLINE | ID: mdl-32741046

ABSTRACT

Dyskinesia induced by long-term L-Dopa (LID) therapy in Parkinson disease is associated with altered striatal function whose molecular bases remain unclear. Here, a transcriptomic approach was applied for comprehensive analysis of distinctively regulated genes in striatal tissue, their specific pathways, and functional- and disease-associated networks in a rodent model of LID. This approach has identified transforming growth factor beta type 1 (TGFß1) as a highly upregulated gene in dyskinetic animals. TGFß1 pathway is a top aberrantly regulated pathway in the striatum following LID development based on differentially expressed genes (> 1.5 fold change and P < 0.05). The induction of TGFß1 pathway specific genes, TGFß1, INHBA, AMHR2 and PMEPA1 was also associated with regulation of NPTX2, PDP1, SCG2, SYNPR, TAC1, TH, TNNT1 genes. Transcriptional network and upstream regulator analyses have identified AKT-centered functional and ERK-centered disease networks revealing the association of TGFß1, IL-1ß and TNFα with LID development. Therefore, results support that TGFß1 pathway is a major contributor to the pathogenic mechanisms of LID.


Subject(s)
Dyskinesia, Drug-Induced/metabolism , Signal Transduction , Transcriptome , Transforming Growth Factor beta1/metabolism , Animals , Antiparkinson Agents/toxicity , Brain/metabolism , Dyskinesia, Drug-Induced/genetics , Gene Regulatory Networks , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Levodopa/toxicity , Male , Rats , Rats, Sprague-Dawley , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Tachykinins/genetics , Tachykinins/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/genetics , Up-Regulation
14.
Exp Neurol ; 330: 113327, 2020 08.
Article in English | MEDLINE | ID: mdl-32387398

ABSTRACT

Numerous genes, and alterations in their expression, have been identified as risk factors for developing levodopa-induced dyskinesia (LID). However, our understanding of the complexities of molecular changes remains insufficient for development of clinical treatment. In the current study we used gene array, in situ hybridization, immunohistochemistry, and microdialysis to provide a unique compare and contrast assessment of the relationship of four candidate genes to LID, employing three genetically distinct rat strains (Sprague-Dawley (SD), Fischer-344 (F344) and Lewis-RT.1) showing differences in dyskinesia susceptibility and 'first-ever LID' versus 'chronic LID' expression in subjects displaying equal dyskinesia severity. In these studies, rat strains were easily distinguishable for their LID propensity with: 1) a majority of SD rats expressing LID (LID+) and a subset being resistant (LID-); 2) all F344 rats readily developing (LID+); and 3) all Lewis rats being LID-resistant (LID-). Following chronic levodopa, LID+ SD rats showed significant increases in candidate gene expression: Nr4a2/(Nurr1) > > Trh > Inhba = Fosb. However, SD rats with long-standing striatal dopamine (DA) depletion treated with first-ever versus chronic high-dose levodopa revealed that despite identical levels of LID severity: 1) Fosb and Nurr1 transcripts but not protein were elevated with acute LID expression; 2) FOSB/ΔFOSB and NURR1 proteins were elevated only with chronic LID; and 3) Trh transcript and protein were elevated only with chronic LID. Strikingly, despite similar levodopa-induced striatal DA release in both LID-expressing F344 and LID-resistant Lewis rats, Fosb, Trh, Inhba transcripts were significantly elevated in both strains; however, Nurr1 mRNA was significantly increased only in LID+ F344 rats. These findings suggest a need to reevaluate currently accepted genotype-to-phenotype relationships in the expression of LID, specifically that of Fosb, a transcription factor generally assumed to play a causal role, and Nurr1, a transcription factor that has received significant attention in PD research linked to its critical role in the survival and function of midbrain DA neurons but who's striatal expression, generally below levels of detection, has remained largely unexplored as a regulator of LID. Finally these studies introduce a novel 'model' (inbred F344 vs inbred Lewis) that may provide a powerful tool for investigating the role for 'dyskinesia-resistance' genes downstream of 'dyskinesia-susceptibility' genes in modulating LID expression, a concept that has received considerably less attention and offers a new ways of thinking about antidyskinetic therapies.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Genotype , Male , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Phenotype , Rats , Rats, Inbred F344 , Rats, Inbred Lew
15.
Neurosci Lett ; 729: 134932, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32224226

ABSTRACT

The brain histaminergic and dopaminergic systems closely interact, and some evidence also suggests significant involvement of histamine in Parkinson's disease (PD), where dopaminergic neurons degenerate. To further investigate histamine-dopamine interactions, particularly in the context of PD, a genetic lack of histamine and a mouse model of PD and levodopa-induced dyskinesia were here combined. Dopaminergic lesions were induced in histidine decarboxylase knockout and wildtype mice by 6-hydroxydopamine injections into the medial forebrain bundle. Post-lesion motor dysfunction was studied by measuring drug-induced rotational behavior and dyskinesia. Striatal tissue from both lesioned and naïve animals was used to investigate dopaminergic, serotonergic and histaminergic biomarkers. Histamine deficiency increased amphetamine-induced rotation but did not affect levodopa-induced dyskinesia. qPCR measurements revealed increased striatal expression of D1 and D2 receptor, DARPP-32, and H3 receptor mRNA, and synaptosomal release experiments in naïve mice indicated increased dopamine release. A lack of histamine thus causes pre- and postsynaptic upregulation of striatal dopaminergic neurotransmission which may be reflected in post-lesion motor behavior. Disturbances or manipulations of the histaminergic system may thus have significant consequences for dopaminergic neurotransmission and motor behavior in both healthy and disease conditions. The findings also represent new evidence for the complex interplay between dopamine and histamine within the nigrostriatal pathway.


Subject(s)
Dopaminergic Neurons/metabolism , Dyskinesia, Drug-Induced/metabolism , Levodopa/pharmacology , Synaptic Transmission/drug effects , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dyskinesia, Drug-Induced/genetics , Mice , Oxidopamine/metabolism , Parkinson Disease/metabolism , Up-Regulation/drug effects
16.
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
17.
Brain Behav ; 10(3): e01537, 2020 03.
Article in English | MEDLINE | ID: mdl-32022467

ABSTRACT

OBJECTIVE: The aim of the study was to evaluate the association of individual and combined single-nucleotide polymorphisms in brain-derived neurotrophic factor (BDNF), dopamine transporter (DAT), and catechol-O-methyltransferase (COMT) genes with the occurrence of motor levodopa-induced complications (MLIC) in Parkinson's disease (PD). MATERIALS AND METHODS: We studied 76 patients with PD (MLIC occurred in 56.6%) and 60 controls. Allelic discrimination of rs6265 BDNF (Val66Met), rs397595 DAT (SLC6A3), and rs4680 COMT (Val158Met) genes were genotyped. Odds ratios (OR) and 95% confidence intervals (95% CI) were calculated using multinominal logistic regression. Orthogonal partial least squares (OPLS) analysis and OPLS discriminant analysis (OPLS-DA) were used to analyze qualitative genetic data. RESULTS: The risk of PD in subjects with the AG BDNF genotype was increased sixfold (OR = 6.12, 95% CI = 2.88-13.02, p < .0001), and AG BDNF and AG DAT genotypes were correlated with PD in OPLS-DA (VIP > 1). There were no differences in distributions of BDNF, DAT and COMT genotypes between PD groups with and without MLIC, while OPLS model showed that genotype combination of AG BDNF, AG DAT, and GG COMT was correlated with MLIC and genotypes combination of GG BDNF, AA DAT, and AA COMT with lack of MLIC in PD patients (VIP > 1). CONCLUSIONS: Our results confirmed the association of rs6265 BDNF (Val66Met) with the risk of PD and suggest a synergic effect of rs6265 BDNF (Val66Met), rs397595 DAT (SLC6A3), and rs4680 COMT (Val158Met) polymorphisms on the occurrence of MLIC.


Subject(s)
Antiparkinson Agents/adverse effects , Brain-Derived Neurotrophic Factor/genetics , Catechol O-Methyltransferase/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Dyskinesia, Drug-Induced/genetics , Levodopa/adverse effects , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Alleles , Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/etiology , Female , Genotype , Humans , Levodopa/therapeutic use , Male , Middle Aged , Parkinson Disease/drug therapy , Pharmacogenetics
18.
World J Biol Psychiatry ; 21(1): 72-77, 2020 01.
Article in English | MEDLINE | ID: mdl-30623717

ABSTRACT

Objectives: Acetylcholine M (muscarinic) receptors are possibly involved in tardive dyskinesia (TD). The authors tried to verify this hypothesis by testing for possible associations between two muscarinic receptor genes (CHRM1 and CHRM2) polymorphisms and TD in patients with schizophrenia.Methods: A total of 472 patients with schizophrenia were recruited. TD was assessed cross-sectionally using the Abnormal Involuntary Movement Scale. Fourteen allelic variants of CHRM1 and CHRM2 were genotyped using Applied Biosystems amplifiers (USA) and the MassARRAY System by Agena Bioscience.Results: The prevalence of the rs1824024*GG genotype of the CHRM2 gene was lower in TD patients compared to the group without it (χ2 = 6.035, p = 0.049). This suggested that this genotype has a protective effect for the development of TD (OR = 0.4, 95% CI: 0.19-0.88). When age, gender, duration of schizophrenia and dosage of antipsychotic treatment were added as covariates in regression analysis, the results did not reach statistical significance.Conclusions: This study did identify associations between CHRM2 variations and TD; the results of logistic regression analysis with covariates suggest that the association is, however, likely to be secondary to other concomitant factors.


Subject(s)
Antipsychotic Agents/adverse effects , Dyskinesia, Drug-Induced/genetics , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M2/genetics , Schizophrenia/genetics , Adult , Alleles , Dyskinesia, Drug-Induced/complications , Female , Genetic Predisposition to Disease , Hospitals, Psychiatric , Humans , Logistic Models , Male , Middle Aged , Pharmacogenetics , Polymorphism, Single Nucleotide , Schizophrenia/complications , Schizophrenia/drug therapy , Severity of Illness Index
19.
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
20.
Neuropharmacology ; 162: 107829, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31666199

ABSTRACT

Parkinson's disease (PD) is characterized by progressive loss of midbrain dopaminergic neurons and treated with the dopamine precursor, 3,4-dihydroxy-l-phenylalanine (L-DOPA). Prolonged L-DOPA treatment is however associated with waning efficacy and the induction of L-DOPA induced dyskinesia (LID). GPR88 is an orphan G-protein Coupled Receptor (GPCR) expressed in dopaminoceptive striatal medium spiny neurons (MSNs) and their afferent corticostriatal glutamatergic neurons. Here, we studied the role of GPR88 in experimental parkinsonism and LID. Chronic L-DOPA administration to male GPR88 KO mice, subjected to unilateral 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle, resulted in more rotations than in their WT counterparts. Conversely, GPR88 KO mice had a lower abnormal involuntary movements (AIMs) score. These behavioral responses were accompanied by altered transcription of L-DOPA upregulated genes in lesioned GPR88 KO compared to WT striata. In accordance with a role for serotonin neurons in LID development, WT but not GPR88 KO striata exhibited 5-hydroxytryptamine displacement upon repeated L-DOPA treatment. Intact male GPR88 KO mice showed diminished tacrine-induced PD-like tremor and spontaneous hyperlocomotion. Dopamine and its metabolites were not increased in male GPR88 KO mice, but biosensor recordings revealed increased spontaneous/basal and evoked glutamate release in striata of male GPR88 KO mice. In conclusion, genetic deletion of GPR88 promotes l-DOPA-induced rotation and spontaneous locomotion yet suppresses the induction of LIDs and also reduces tremor. These data provide behavioral, neurochemical and molecular support that GPR88 antagonism may favour motor relief in PD patients without aggravating the induction of motor side effects.


Subject(s)
Antiparkinson Agents/pharmacology , Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/genetics , Levodopa/pharmacology , Locomotion/drug effects , Movement/drug effects , Parkinsonian Disorders/genetics , Receptors, G-Protein-Coupled/genetics , Adrenergic Agents/toxicity , Animals , Cholinesterase Inhibitors/toxicity , Corpus Striatum/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/physiopathology , GABAergic Neurons , Glutamic Acid/metabolism , Locomotion/genetics , Male , Medial Forebrain Bundle , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Oxidopamine/toxicity , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/physiology , Serotonin/metabolism , Tacrine/toxicity , Tremor
SELECTION OF CITATIONS
SEARCH DETAIL
...