1
|
Terada T, Bunai T, Hashizume T, Matsudaira T, Yokokura M, Takashima H, Konishi T, Obi T, Ouchi Y. Neuroinflammation following anti-parkinsonian drugs in early Parkinson's disease: a longitudinal PET study. Sci Rep 2024; 14:4708. [PMID: 38409373 PMCID: PMC10897150 DOI: 10.1038/s41598-024-55233-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
The progression of neuroinflammation after anti-parkinsonian therapy on the Parkinson's disease (PD) brain and in vivo evidence of the therapy purporting neuroprotection remain unclear. To elucidate this, we examined changes in microglial activation, nigrostriatal degeneration, and clinical symptoms longitudinally after dopamine replacement therapy in early, optimally-controlled PD patients with and without zonisamide treatment using positron emission tomography (PET). We enrolled sixteen PD patients (Hoehn and Yahr stage 1-2), and age-matched normal subjects. PD patients were randomly divided into two groups: one (zonisamide+) that did and one (zonisamide-) that did not undergo zonisamide therapy. Annual changes in neuroinflammation ([11C]DPA713 PET), dopamine transporter availability ([11C]CFT PET) and clinical severity were examined. Voxelwise differentiations in the binding of [11C]DPA713 (BPND) and [11C]CFT (SUVR) were compared with normal data and between the zonisamide+ and zonisamide- PD groups. The cerebral [11C]DPA713 BPND increased with time predominantly over the parieto-occipital region in PD patients. Comparison of the zonisamide+ group with the zonisamide- group showed lower levels in the cerebral [11C]DPA713 BPND in the zonisamide+ group. While the striatal [11C]CFT SUVR decreased longitudinally, the [11C]CFT SUVR in the nucleus accumbens showed a higher binding in the zonisamide+ group. A significant annual increase in attention score were found in the zonisamide+ group. The current results indicate neuroinflammation proceeds to the whole brain even after anti-parkinsonian therapy, but zonisamide coadministration might have the potential to ameliorate proinflammatory responses, exerting a neuroprotective effect in more damaged nigrostriatal regions with enhanced attention in PD.
Collapse
Affiliation(s)
- Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Tomoyasu Bunai
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Takanori Hashizume
- Laboratory of Drug Metabolism and Pharmacokinetics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| | - Takashi Matsudaira
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Masamichi Yokokura
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirotsugu Takashima
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Takashi Konishi
- Department of Neurology, Japanese Red Cross Shizuoka Hospital, Shizuoka, Japan
| | - Tomokazu Obi
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.
- Hamamatsu Medical Imaging Center, Hamamatsu Medical Photonics Foundation, Hamamatsu, Japan.
| |
Collapse
|
2
|
Aboulatta L, Haidar L, Abou-Setta A, Askin N, Rabbani R, Lavu A, Peymani P, Zarychanski R, Eltonsy S. Efficacy and Safety of MAO-B Inhibitors Safinamide and Zonisamide in Parkinson's Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. CNS Drugs 2023; 37:941-956. [PMID: 37973769 DOI: 10.1007/s40263-023-01048-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND OBJECTIVE In Parkinson's disease, safinamide and zonisamide are novel monoamine oxidase-B inhibitors with a dual mechanism of action involving the inhibition of sodium and calcium channels and the subsequent release of glutamate. The aim of this systematic review and meta-analysis was to examine the efficacy and safety of both drugs compared with placebo on motor symptoms, cognitive function, and quality of life in patients with Parkinson's disease. METHODS We searched MEDLINE, EMBASE, Cochrane Central, Scopus, PsycINFO, and trials registries up to March 2023 for randomized controlled trials of adults with Parkinson's disease administered either safinamide or zonisamide and published in English. We excluded single-arm trials or if neither the efficacy nor safety outcomes of interest were reported. Primary outcomes were the change from baseline in Unified Parkinson's Disease Rating Scale section III (UPDRS-III) and serious adverse events. Secondary outcomes included a change from baseline in OFF-time, Parkinson's Disease Questionnaire 39 to evaluate quality of life, and Mini-Mental State Examination for cognitive function assessment. The meta-analysis was conducted using Review Manager 5.4.1. Random-effect models were used to calculate the pooled mean differences (MDs) and risk ratios with 95% confidence intervals (CIs). Subgroup analyses by medication, doses, Parkinson's disease stage, and risk of bias were conducted. We assessed the risk of bias using the Cochrane's risk of bias tool. Sensitivity analysis was conducted, and publication bias were evaluated. This meta-analysis was not externally funded, and the protocol is available on the Open Science Framework Registration ( https://doi.org/10.17605/OSF.IO/AMNP5 ). RESULTS Of 3570 screened citations, 16 trials met inclusion criteria (4314 patients with Parkinson's disease). Ten safinamide trials were conducted in several countries. Six zonisamide trials were included, five of which were conducted in Japan and one in India. UPDRS Part III scores were significantly lower with both monoamine oxidase-B inhibitors than with placebo (MD = - 2.18; 95% CI - 2.88 to - 1.49; I 2 =63%; n = 14 studies). A subgroup analysis showed a significant improvement in UPDRS-III in safinamide (MD = - 2.10; 95% CI - 3.09 to - 1.11; I2 = 71%; n = 8 studies) and zonisamide (MD = - 2.31; 95% CI - 3.35 to - 1.27; I2 = 52%; n = 6 studies) compared with placebo. Monoamine oxidase-B inhibitors significantly decreased OFF-time compared with placebo. No significant differences in cognitive function (Mini-Mental State Examination), whereas an improvement in quality of life (Parkinson's Disease Questionnaire 39 scores) was observed. There was no significant difference in incidence rates of serious adverse events among all examined doses of zonisamide and safinamide compared with placebo. Two trials were reported as a high risk of bias and sensitivity analyses confirmed the primary analysis results. CONCLUSIONS Evidence suggests that novel monoamine oxidase-B inhibitors not only improve motor symptoms but also enhance patients' quality of life. The meta-analysis showed that both medications have a similar safety profile to placebo with regard to serious adverse events. The overall findings emphasize the effectiveness of safinamide and zonisamide in the treatment of Parkinson's disease as adjunct therapy. Further long-term studies examining the impact of these medications on motor and non-motor symptoms are necessary.
Collapse
Affiliation(s)
- Laila Aboulatta
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Lara Haidar
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Ahmed Abou-Setta
- George & Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, MB, Canada
- Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Nicole Askin
- WRHA Virtual Library, University of Manitoba, Winnipeg, MB, Canada
| | - Rasheda Rabbani
- George & Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, MB, Canada
| | - Alekhya Lavu
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Payam Peymani
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Ryan Zarychanski
- Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Cancer Care Manitoba, Winnipeg, MB, Canada
| | - Sherif Eltonsy
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada.
| |
Collapse
|
3
|
Yan R, Cai H, Cui Y, Su D, Cai G, Lin F, Feng T. Comparative efficacy and safety of monoamine oxidase type B inhibitors plus channel blockers and monoamine oxidase type B inhibitors as adjuvant therapy to levodopa in the treatment of Parkinson's disease: a network meta-analysis of randomized controlled trials. Eur J Neurol 2023; 30:1118-1134. [PMID: 36437702 DOI: 10.1111/ene.15651] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 11/01/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE The monoamine oxidase type B inhibitors plus channel blockers (MAO-BIs plus) are a new class of antiparkinsonian drug with additional mechanisms of action for their property as ion channel blockers. The present study aimed to compare the efficacy and safety of MAO-BIs plus and conventional MAO-BIs, as well as their corresponding doses, as adjuvant therapy to levodopa in the treatment of Parkinson's disease (PD). METHOD Randomized controlled trials enrolling PD patients treated with selegiline, rasagiline, safinamide or zonisamide as adjuvant therapy to levodopa were identified. Bayesian network meta-analysis was conducted. RESULTS Thirty-one randomized controlled trials comprising 7142 PD patients were included. Compared with levodopa monotherapy, the combination therapy of MAO-BIs and levodopa was significantly more effective, with a mean difference of 2.74 (1.26-4.18) on the Unified Parkinson's Disease Rating Scale (UPDRS) III score change for selegiline, 2.67 (1.45-3.87) for safinamide, 2.2 (0.98-3.64) for zonisamide and 2.04 (1.24-2.87) for rasagiline. No significant difference was detected amongst MAO-BIs. The surface under the cumulative ranking results showed that safinamide 100 mg and rasagiline 1 mg ranked first in improving UPDRS III and UPDRS II, respectively. Zonisamide 100 mg ranked first in reducing OFF time. For safety outcomes, rasagiline was associated with a higher incidence of adverse events than placebo and safinamide. MAO-BIs plus had a higher probability of being safer agents compared to conventional MAO-BIs. CONCLUSIONS Monoamine oxidase type B inhibitors plus, conventional MAO-BIs and the corresponding doses are similar in efficacy in PD treatment. MAO-BIs plus might be safer than conventional MAO-BIs. Head-to-head comparisons are needed for further investigation.
Collapse
Affiliation(s)
- Rui Yan
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huihui Cai
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yusha Cui
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dongning Su
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Guoen Cai
- Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Fabin Lin
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Tao Feng
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
4
|
Deepthy MS, Karun KM, Harichandrakumar KT, Nair NS. Investigation of the Utility of Multivariate Meta-Analysis Methods in Estimating the Summary Dose Response Curve. J Res Health Sci 2022; 22:e00561. [PMID: 37571932 PMCID: PMC10422157 DOI: 10.34172/jrhs.2022.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Traditional meta-analyses often assess the effectiveness of different doses of the same intervention separately or examine the overall differences between intervention and placebo groups. The present study aimed to model the effect sizes obtained from different doses in multiple studies using a two-stage dose-response meta-analytic approach while taking dose variations into account. METHODS Different dose-response meta-analysis models using linear, quadratic, and restricted cubic spline (RCS) functions were fitted. A two-stage approach utilizing multivariate meta-analysis was performed and the obtained results were compared with those of the univariate meta-analysis. A random effect dose-response meta-analysis was performed using data from an existing systematic review on combination therapy with zonisamide and anti-Parkinson drugs for Parkinson's disease. The effective or optimum dose for producing maximum response was also investigated. Moreover, a sensitivity analysis was performed by changing the knots of the RCS model. RESULTS Dose-response meta-analysis was performed using data from four double-blinded randomized controlled trials with 724 and 309 patients with Parkinson's disease in dose and placebo arms, respectively. The quadratic model yielded the smallest Akaike information criterion (AIC), compared to the linear and RCS models, indicating it to be the best fit for the data. CONCLUSION Compared to the traditional approach, the two-stage approach could model the dose-dependent effect of zonisamide on the Unified Parkinson's Disease Rating Scale (UPRDS) part III score and predict the outcome for different doses through a single analysis.
Collapse
Affiliation(s)
| | - Kalesh Mappilakudy Karun
- Division of Biostatistics, Malankara Orthodox Syrian Church Medical College, Kolenchery, Ernakulam, Kerala, India
| | | | | |
Collapse
|
5
|
Li C, Xue L, Liu Y, Yang Z, Chi S, Xie A. Zonisamide for the Treatment of Parkinson Disease: A Current Update. Front Neurosci 2020; 14:574652. [PMID: 33408605 PMCID: PMC7779619 DOI: 10.3389/fnins.2020.574652] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Zonisamide has been used as an add-on treatment in order to overcome the deficiencies of the general therapies currently used to resolve the motor complications and non-motor symptoms of Parkinson disease. Various trials have been designed to investigate the mechanism of action and treatment effects of zonisamide in this condition. Most clinical trials of zonisamide in Parkinson disease were from Japan. The vast majority of studies used changes in the Unified Parkinson’s Disease Rating Scale (UPDRS) scores and daily “OFF” time as primary endpoints. Based on adequate randomized controlled trials, zonisamide is considered a safe and efficacious add-on treatment in Parkinson disease. The most convincing proof is available for a dosage of 25–50 mg, which was shown to lead to a significant reduction in the UPDRS III score and daily “OFF” time, without increasing disabling dyskinesia. Furthermore, zonisamide may play a beneficial role in improving non-motor symptoms in PD, including impulsive–compulsive disorder, rapid eye movement sleep behavior disorder, and dementia. Among the various mechanisms reported, inhibition of monoamine oxidase-B, blocking of T-type calcium channels, modulation of the levodopa–dopamine metabolism, modulation of receptor expression, and neuroprotection are the most often cited. The mechanisms underlying neuroprotection, including modulation of dopamine turnover, induction of neurotrophic factor expression, inhibition of oxidative stress and apoptosis, inhibition of neuroinflammation, modulation of synaptic transmission, and modulation of gene expression, have been most extensively studied. This review focuses on structure, pharmacokinetics, mechanisms, therapeutic effectiveness, and safety and tolerability of zonisamide in patients with Parkinson disease.
Collapse
Affiliation(s)
- Chengqian Li
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Xue
- Department of Medical Record, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yumei Liu
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhengjie Yang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Rodriguez-López J, Arrojo M, Paz E, Páramo M, Costas J. Identification of relevant hub genes for early intervention at gene coexpression modules with altered predicted expression in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2020; 98:109815. [PMID: 31715283 DOI: 10.1016/j.pnpbp.2019.109815] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/25/2019] [Accepted: 11/08/2019] [Indexed: 01/20/2023]
Abstract
Genetic risk for schizophrenia is due to the joint effect of multiple genes acting mainly at two different processes, prenatal/perinatal neurodevelopment and adolescence/early adulthood synapse maturation. Identification of important genes at the second process is of relevance for early intervention. The aim of this work was to identify gene co-expression modules with altered expression in schizophrenia during adolescence/early adulthood. To this goal, we predicted frontal cortex gene expression in one discovery sample, the largest GWAS of schizophrenia from the Psychiatric Genomics Consortium, using S-prediXcan, and in one target sample, consisting of 625 schizophrenic patients and 819 controls from Spain, using prediXcan. Prediction models were trained on GTEx frontal cortex expression dataset. In parallel, we identified brain co-expression modules from BrainSpan using WGCNA. Then, we estimated polygenic risk scores based on predicted expression (PE-PRS) for each co-expression module in the target sample, based on PE-PRS model from the discovery sample. This analysis led to the identification of a module with mainly adolescence/adulthood expression whose PE-PRS was significantly associated with schizophrenia. The module was significantly enriched in synaptic processes. Several hub genes at this module are drugabble, according to the drug-gene interaction database, and/or involved in synaptic transmission, such as the voltage-gated ion channels SCN2B and KCNAB2, the calcium calmodulin kinases CAMK2A and CAMK1G, or genes involved in synaptic vesicle cycle, such as DNM1, or SYNGR1. Therefore, identification of this module may be the first step in patient stratification based on biology, as well as in drug design and drug repurposing efforts.
Collapse
Affiliation(s)
- Julio Rodriguez-López
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Arrojo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Eduardo Paz
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Mario Páramo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Javier Costas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain.
| |
Collapse
|
7
|
Bentea E, Van Liefferinge J, Verbruggen L, Martens K, Kobayashi S, Deneyer L, Demuyser T, Albertini G, Maes K, Sato H, Smolders I, Lewerenz J, Massie A. Zonisamide attenuates lactacystin-induced parkinsonism in mice without affecting system x c<sup/>. Exp Neurol 2016; 290:15-28. [PMID: 28024798 DOI: 10.1016/j.expneurol.2016.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 11/29/2022]
Abstract
Zonisamide (ZNS), an anticonvulsant drug exhibiting symptomatic effects in Parkinson's disease (PD), was recently reported to exert neuroprotection in rodent models. One of the proposed neuroprotective mechanisms involves increased protein expression of xCT, the specific subunit of the cystine/glutamate antiporter system xc-, inducing glutathione (GSH) synthesis. Here, we investigated the outcome of ZNS treatment in a mouse model of PD based on intranigral proteasome inhibition, and whether the observed effects would be mediated by system xc-. The proteasome inhibitor lactacystin (LAC) was administered intranigrally to male C57BL/6J mice receiving repeated intraperitoneal injections of either ZNS 30mgkg-1 or vehicle. Drug administration was initiated three days prior to stereotaxic LAC injection and was maintained until six days post-surgery. One week after lesion, mice were behaviorally assessed and investigated in terms of nigrostriatal neurodegeneration and molecular changes at the level of the basal ganglia, including expression levels of xCT. ZNS reduced the loss of nigral dopaminergic neurons following LAC injection and the degree of sensorimotor impairment. ZNS failed, however, to modulate xCT expression in basal ganglia of lesioned mice. In a separate set of experiments, the impact of ZNS treatment on system xc- was investigated in control conditions in vivo as well as in vitro. Similarly, ZNS did not influence xCT or glutathione levels in naive male C57BL/6J mice, nor did it alter system xc- activity or glutathione content in vitro. Taken together, these results demonstrate that ZNS treatment provides neuroprotection and behavioral improvement in a PD mouse model based on proteasome inhibition via system xc- independent mechanisms.
Collapse
Affiliation(s)
- Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lise Verbruggen
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katleen Martens
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sho Kobayashi
- Department of Food and Applied Life Sciences, Yamagata University, Yamagata, Japan
| | - Lauren Deneyer
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katrien Maes
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Technology, Niigata University, Niigata, Japan
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan Lewerenz
- Department of Neurology, Ulm University, Ulm, Germany
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
8
|
Devergnas A, Chen E, Ma Y, Hamada I, Pittard D, Kammermeier S, Mullin AP, Faundez V, Lindsley CW, Jones C, Smith Y, Wichmann T. Anatomical localization of Cav3.1 calcium channels and electrophysiological effects of T-type calcium channel blockade in the motor thalamus of MPTP-treated monkeys. J Neurophysiol 2015; 115:470-85. [PMID: 26538609 DOI: 10.1152/jn.00858.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/03/2015] [Indexed: 12/28/2022] Open
Abstract
Conventional anti-Parkinsonian dopamine replacement therapy is often complicated by side effects that limit the use of these medications. There is a continuing need to develop nondopaminergic approaches to treat Parkinsonism. One such approach is to use medications that normalize dopamine depletion-related firing abnormalities in the basal ganglia-thalamocortical circuitry. In this study, we assessed the potential of a specific T-type calcium channel blocker (ML218) to eliminate pathologic burst patterns of firing in the basal ganglia-receiving territory of the motor thalamus in Parkinsonian monkeys. We also carried out an anatomical study, demonstrating that the immunoreactivity for T-type calcium channels is strongly expressed in the motor thalamus in normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys. At the electron microscopic level, dendrites accounted for >90% of all tissue elements that were immunoreactive for voltage-gated calcium channel, type 3.2-containing T-type calcium channels in normal and Parkinsonian monkeys. Subsequent in vivo electrophysiologic studies in awake MPTP-treated Parkinsonian monkeys demonstrated that intrathalamic microinjections of ML218 (0.5 μl of a 2.5-mM solution, injected at 0.1-0.2 μl/min) partially normalized the thalamic activity by reducing the proportion of rebound bursts and increasing the proportion of spikes in non-rebound bursts. The drug also attenuated oscillatory activity in the 3-13-Hz frequency range and increased gamma frequency oscillations. However, ML218 did not normalize Parkinsonism-related changes in firing rates and oscillatory activity in the beta frequency range. Whereas the described changes are promising, a more complete assessment of the cellular and behavioral effects of ML218 (or similar drugs) is needed for a full appraisal of their anti-Parkinsonian potential.
Collapse
Affiliation(s)
- Annaelle Devergnas
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia;
| | - Erdong Chen
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Yuxian Ma
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Ikuma Hamada
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Damien Pittard
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Stefan Kammermeier
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia; Klinikum der Universität München, Neurologische Klinik und Poliklinik, München, Germany
| | - Ariana P Mullin
- Department of Cell Biology, Emory University, Atlanta, Georgia
| | - Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, Georgia; Center for Social Translational Neuroscience, Emory University, Atlanta, Georgia
| | - Craig W Lindsley
- Department of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Carrie Jones
- Department of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia; Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| | - Thomas Wichmann
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia; Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
9
|
Jenner P. Treatment of the later stages of Parkinson's disease - pharmacological approaches now and in the future. Transl Neurodegener 2015; 4:3. [PMID: 25973178 PMCID: PMC4429454 DOI: 10.1186/2047-9158-4-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/01/2015] [Indexed: 11/10/2022] Open
Abstract
The problems associated with the pharmacological treatment of the later stages of Parkinson's disease (PD) remain those seen over many years. These centre on a loss of drug effect ('wearing off') with disease progression, the occurrence of dyskinesia, notably with L-dopa use and the appearance of non-motor symptoms that are largely refractory to dopaminergic medication. Treatment strategies in late PD have been dominated by the use of drug combinations and the subtle manipulation of drug dosage. However, change is occurring as the understanding of the basis of motor complications and fluctuations and non-motor symptoms improves. New pharmacological options are expanding with the advent of longer acting versions of existing dopaminergic drugs, new drug delivery systems and the introduction of non-dopaminergic agents able to manipulate motor function both within the basal ganglia and in other brain regions. Non-dopaminergic agents are also being investigated for the treatment of dyskinesia and for the relief of non-motor symptoms. However, while therapy continues to improve, the treatment of late stage PD remains problematic with non-motor symptoms dominating the unmet need in this patient group.
Collapse
Affiliation(s)
- Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Sciences, Faculty of Health Sciences and Medicine, King's College, London, SE1 1UL UK
| |
Collapse
|
10
|
Yang YC, Tai CH, Pan MK, Kuo CC. The T-type calcium channel as a new therapeutic target for Parkinson's disease. Pflugers Arch 2014; 466:747-55. [PMID: 24531801 DOI: 10.1007/s00424-014-1466-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is one of the most prevalent movement disorder caused by degeneration of the dopaminergic neurons in substantia nigra pars compacta. Deep brain stimulation (DBS) at the subthalamic nucleus (STN) has been a new and effective treatment of PD. It is interesting how a neurological disorder caused by the deficiency of a specific chemical substance (i.e., dopamine) from one site could be so successfully treated by a pure physical maneuver (i.e., DBS) at another site. STN neurons could discharge in the single-spike or the burst modes. A significant increase in STN burst discharges has been unequivocally observed in dopamine-deprived conditions such as PD, and was recently shown to have a direct causal relation with parkinsonian symptoms. The occurrence of burst discharges in STN requires enough available T-type Ca(2+) currents, which could bring the relatively negative membrane potential to the threshold of firing Na(+) spikes. DBS, by injection of negative currents into the extracellular space, most likely would depolarize the STN neuron and then inactivate the T-type Ca(2+) channel. Burst discharges are thus decreased and parkinsonian locomotor deficits ameliorated. Conversely, injection of positive currents into STN itself could induce parkinsonian locomotor deficits in animals without dopaminergic lesions. Local application of T-type Ca(2+) channel blockers into STN would also dramatically decrease the burst discharges and improve parkinsonian locomotor symptoms. Notably, zonisamide, which could inhibit T-type Ca(2+) currents in STN, has been shown to benefit PD patients in a clinical trial. From the pathophysiological perspectives, PD can be viewed as a prototypical disorder of "brain arrhythmias". Modulation of relevant ion channels by physical or chemical maneuvers may be important therapeutic considerations for PD and other diseases related to deranged neural rhythms.
Collapse
Affiliation(s)
- Ya-Chin Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | |
Collapse
|
11
|
Do the basal ganglia inhibit seizure activity in temporal lobe epilepsy? Epilepsy Behav 2012; 25:56-9. [PMID: 22835431 DOI: 10.1016/j.yebeh.2012.04.125] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 04/20/2012] [Accepted: 04/21/2012] [Indexed: 11/22/2022]
Abstract
There is substantial evidence in the literature that the basal ganglia (BG), namely the striatum and pallidum, are involved in temporal lobe epilepsy (TLE). The BG are probably not involved in elaborating clinical seizures, as they do not produce specific epileptiform activity and there is no evident change in the electrical activity in the BG immediately after seizure onset. The data we obtained by direct ictal recording in the BG [1,2], as well as a large body of experimental and clinical evidence reported by other groups, suggest an inhibitory role of the BG during temporal lobe seizures. The BG may have a remote influence on cortical oscillatory processes related to control of epileptic seizures via their feedback pathways to the cortex.
Collapse
|
12
|
Miwa H, Kondo T. T-type calcium channel as a new therapeutic target for tremor. THE CEREBELLUM 2012; 10:563-9. [PMID: 21479969 DOI: 10.1007/s12311-011-0277-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Voltage-gated calcium channels play an important role in many physiological and pathological processes. Accumulating studies suggest that the T-type calcium channel is a potential target for the treatment of various neurological disorders, such as epilepsy, insomnia, and neuropathic pain. Here, we highlight recent advances in our understanding of T-type calcium channel regulation and their implications for tremor disorders. Several T-type calcium channel blockers effectively suppressed experimental tremors that have been suggested to originate from either the cerebellum or basal ganglia. Among T-type calcium channel blockers that have been used clinically, the anti-tremor efficacy of zonisamide garnered our attention. Based on both basic and clinical studies, the possibility is emerging that T-type calcium channel blockers that transit into the central nervous system may have therapeutic potentials for tremor disorders.
Collapse
Affiliation(s)
- Hideto Miwa
- Department of Neurology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan.
| | | |
Collapse
|
13
|
Effects of T-type calcium channel blockers on a parkinsonian tremor model in rats. Pharmacol Biochem Behav 2011; 97:656-9. [DOI: 10.1016/j.pbb.2010.11.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/15/2010] [Accepted: 11/18/2010] [Indexed: 11/19/2022]
|
14
|
Choudhury ME, Moritoyo T, Yabe H, Nishikawa N, Nagai M, Kubo M, Matsuda S, Nomoto M. Zonisamide Attenuates MPTP Neurotoxicity in Marmosets. J Pharmacol Sci 2010; 114:298-303. [DOI: 10.1254/jphs.10120fp] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
15
|
Abstract
Although zonisamide was previously only used to treat epilepsy, recently more applications have been forthcoming. Due to a good side effect profile, a lower frequency of interactions and a more comfortable posology, there are several studies regarding its uses in other pathologies such as migraine, neuropathic pain, essential tremor and various psychiatric diseases. A multicentered, randomized, double-blind, placebo-controlled study conducted in Japan suggested that zonisamide, as an add-on treatment, has efficacy in treating motor symptoms in patients with Parkinson's disease. In addition, other studies support the utility of zonisamide in other symptoms of this disease. The therapeutic doses of zonisamide for the treatment of Parkinson's disease are considerably lower than those for the treatment of epilepsy. This antiepileptic drug has been used in Japan for more than 15 years and so it is expected that it will be safe and well tolerated in patients with Parkinson's disease. However, the pharmacological mechanisms of the antiparkinsonian actions of zonisamide remain unclear and more basic investigation is warranted. The aim of this paper is to review the structure, mechanisms of action, pharmacokinetics and antiparkinsonian action of zonisamide.
Collapse
Affiliation(s)
- Pedro Emilio Bermejo
- Sanatorio Nuestra Señora del Rosario - Hospital Sanitas La Zarzuela, Madrid, Spain
| | | |
Collapse
|
16
|
Miwa H, Kubo T, Suzuki A, Kondo T. Effects of zonisamide on c-Fos expression under conditions of tacrine-induced tremulous jaw movements in rats: a potential mechanism underlying its anti-parkinsonian tremor effect. Parkinsonism Relat Disord 2008; 15:30-5. [PMID: 18693129 DOI: 10.1016/j.parkreldis.2008.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Revised: 02/18/2008] [Accepted: 02/18/2008] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To examine the mechanisms underlying the anti-tremor effect of zonisamide in rats under conditions of tacrine-induced tremulous jaw movements (TJMs). METHODS Male adult rats received systemic administration of either zonisamide (5 or 50mg/kg) or vehicle at 20min prior to the administration of tacrine hydrochloride (5mg/kg). Animals were sacrificed 2h later, and the brains collected and immunostained for quantitative assessment of c-Fos expression. RESULTS There was no effect of zonisamide on tacrine-induced c-Fos expression in the ventrolateral striatum, a primary site of the pharmacological action of tacrine. Zonisamide suppressed the tacrine-induced c-Fos expression in the cortex, the dorsal striatum, and the nucleus accumbens, which are involved in the architecture of the cortico-basal ganglia-thalamocortical circuits. CONCLUSION The anti-TJM effect of zonisamide may not relate to suppression of neural activity specifically in primary tremor-generating sites, but may be due to a more broad inhibitory effect on tremor-related structures such as the cortex or the striatum. This effect of zonisamide may be a contributing mechanism underlying its therapeutic efficacy on parkinsonian tremor.
Collapse
Affiliation(s)
- Hideto Miwa
- Department of Neurology, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 641-8510, Japan.
| | | | | | | |
Collapse
|
17
|
Kothare SV, Kaleyias J. Zonisamide: review of pharmacology, clinical efficacy, tolerability, and safety. Expert Opin Drug Metab Toxicol 2008; 4:493-506. [PMID: 18433351 DOI: 10.1517/17425255.4.4.493] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Zonisamide (ZNS), a sulphonamide derivative, is a new-generation anticonvulsant with multiple potential mechanisms that contribute to its antiepileptic efficacy and may also explain its as yet incompletely assessed utility for non-seizure disorders such as headaches, neuropathic pain, and weight loss. OBJECTIVE A review of the pharmacokinetics, pharmacodynamics, evidence for efficacy in different seizure types and non-seizure conditions, adverse effects, and tolerability of ZNS is presented. METHODS A review of all manuscripts published in the English literature on ZNS was performed in preparing this manuscript. RESULTS/CONCLUSIONS ZNS has a broad label for use in Japan, while the regulatory bodies in the USA and Europe have approved it for use only as an adjunctive therapy for partial seizures in adults. It has favorable pharmacokinetic characteristics, proven efficacy in seizure disorders, and is well tolerated in long-term use.
Collapse
Affiliation(s)
- Sanjeev V Kothare
- Pediatric Sleep Medicine Training Program, Division of Epilepsy & Clinical Neurophysiology, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|