1
|
Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement 2024; 20:3034-3053. [PMID: 38305570 PMCID: PMC11032537 DOI: 10.1002/alz.13678] [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: 08/24/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
Astrocytes are abundantly and ubiquitously expressed cell types with diverse functions throughout the central nervous system. Astrocytes show remarkable plasticity and exhibit morphological, molecular, and functional remodeling in response to injury, disease, or infection of the central nervous system, as evident in neurodegenerative diseases. Astroglial mediated inflammation plays a prominent role in the pathogenesis of neurodegenerative diseases. This review focus on the role of astrocytes as essential players in neuroinflammation and discuss their morphological and functional heterogeneity in the normal central nervous system and explore the spatial and temporal variations in astroglial phenotypes observed under different disease conditions. This review discusses the intimate relationship of astrocytes to pathological hallmarks of neurodegenerative diseases. Finally, this review considers the putative therapeutic strategies that can be deployed to modulate the astroglial functions in neurodegenerative diseases. HIGHLIGHTS: Astroglia mediated neuroinflammation plays a key role in the pathogenesis of neurodegenerative diseases. Activated astrocytes exhibit diverse phenotypes in a region-specific manner in brain and interact with β-amyloid, tau, and α-synuclein species as well as with microglia and neuronal circuits. Activated astrocytes are likely to influence the trajectory of disease progression of neurodegenerative diseases, as determined by the stage of disease, individual susceptibility, and state of astroglial priming. Modulation of astroglial activation may be a therapeutic strategy at various stages in the trajectory of neurodegenerative diseases to modify the disease course.
Collapse
Affiliation(s)
- Paul Edison
- Division of NeurologyDepartment of Brain SciencesFaculty of Medicine, Imperial College LondonLondonUK
- Division of Psychological medicine and clinical neurosciencesSchool of Medicine, Cardiff UniversityWalesUK
| |
Collapse
|
2
|
Ismail FS, Faustmann PM, Förster E, Corvace F, Faustmann TJ. Tiagabine and zonisamide differentially regulate the glial properties in an astrocyte-microglia co-culture model of inflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3253-3267. [PMID: 37231170 PMCID: PMC10567966 DOI: 10.1007/s00210-023-02538-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Due to the role of astrocytes and microglia in the pathophysiology of epilepsy and limited studies of antiseizure medication (ASM) effects on glial cells, we studied tiagabine (TGB) and zonisamide (ZNS) in an astrocyte-microglia co-culture model of inflammation. Different concentrations of ZNS (10, 20, 40, 100 µg/ml) or TGB (1, 10, 20, 50 µg/ml) were added to primary rat astrocytes co-cultures with 5-10% (M5, physiological conditions) or 30-40% (M30, pathological inflammatory conditions) microglia for 24 h, aiming to study glial viability, microglial activation, connexin 43 (Cx43) expression and gap-junctional coupling. ZNS led to the reduction of glial viability by only 100 µg/ml under physiological conditions. By contrast, TGB revealed toxic effects with a significant, concentration-dependent reduction of glial viability under physiological and pathological conditions. After the incubation of M30 co-cultures with 20 µg/ml TGB, the microglial activation was significantly decreased and resting microglia slightly increased, suggesting possible anti-inflammatory features of TGB under inflammatory conditions. Otherwise, ZNS caused no significant changes of microglial phenotypes. The gap-junctional coupling was significantly decreased after the incubation of M5 co-cultures with 20 and 50 µg/ml TGB, which can be related to its anti-epileptic activity under noninflammatory conditions. A significant decrease of Cx43 expression and cell-cell coupling was found after the incubation of M30 co-cultures with 10 µg/ml ZNS, suggesting additional anti-seizure effects of ZNS with the disruption of glial gap-junctional communication under inflammatory conditions. TGB and ZNS differentially regulated the glial properties. Developing novel ASMs targeting glial cells may have future potential as an "add-on" therapy to classical ASMs targeting neurons.
Collapse
Affiliation(s)
- Fatme Seval Ismail
- Department of Neurology, University Hospital Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.
| | - Pedro M Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
3
|
Wang C, Liu H, Xu S, Deng Y, Xu B, Yang T, Liu W. Ferroptosis and Neurodegenerative Diseases: Insights into the Regulatory Roles of SLC7A11. Cell Mol Neurobiol 2023; 43:2627-2642. [PMID: 36988772 PMCID: PMC11410137 DOI: 10.1007/s10571-023-01343-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Programed cell death plays a key role in promoting human development and maintaining homeostasis. Ferroptosis is a recently identified pattern of programmed cell death that is closely associated with the onset and progression of neurodegenerative diseases. Ferroptosis is mainly caused by the intracellular accumulation of iron-dependent lipid peroxides. The cysteine/glutamate antibody Solute carrier family 7 member 11 (SLC7A11, also known as xCT) functions to import cysteine for glutathione biosynthesis and antioxidant defense. SLC7A11 has a significant impact on ferroptosis, and inhibition of SLC7A11 expression promotes ferroptosis. Moreover, SLC7A11 is also closely associated with neurodegenerative diseases. In this paper, we summarize the relationship between ferroptosis and neurodegenerative diseases and the role of SLC7A11 during this process. The various regulatory mechanisms of SLC7A11 are also discussed. In conclusion, we are looking forward to a theoretical basis for further understanding the occurrence and development of ferroptosis in SLC7A11 and neurodegenerative diseases, and to seek new clues for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Chen Wang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Haihui Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Si Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
4
|
Sano H, Nambu A. Behavioral effects of zonisamide on L-DOPA-induced dyskinesia in Parkinson's disease model mice. Front Aging Neurosci 2023; 15:1221341. [PMID: 37441679 PMCID: PMC10333504 DOI: 10.3389/fnagi.2023.1221341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
Zonisamide (ZNS; 1,2-benzisoxazole-3-methanesulfonamide) was initially developed and is commonly used as an anticonvulsant drug. However, it has also shown its beneficial effects on Parkinson's disease (PD), a progressive neurodegenerative disorder caused by the loss of dopaminergic neurons in the midbrain. Recent clinical studies have suggested that ZNS can also have beneficial effects on L-DOPA-induced dyskinesia (LID), which is a major side effect of long-term L-DOPA treatments for PD. In the present study, we examined the behavioral effects of ZNS on LID in PD model mice. Acute ZNS treatment did not have any observable behavioral effects on LID. Contrastingly, chronic ZNS treatment with L-DOPA delayed the peak of LID and reduced the severity of LID before the peak but increased the duration of LID in a dose-dependent manner of ZNS compared to PD model mice treated with L-DOPA alone. Thus, ZNS appears to have both beneficial and adverse effects on LID.
Collapse
Affiliation(s)
- Hiromi Sano
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
| |
Collapse
|
5
|
Masood N, Jimenez-Shahed J. Effective Management of "OFF" Episodes in Parkinson's Disease: Emerging Treatment Strategies and Unmet Clinical Needs. Neuropsychiatr Dis Treat 2023; 19:247-266. [PMID: 36721795 PMCID: PMC9884436 DOI: 10.2147/ndt.s273121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Motor complications related to the chronic administration of levodopa and failure to prevent the neurodegenerative disease process counterbalance the pivotal discovery of levodopa as the cornerstone of PD treatment. Excellent motor control is offered early during the course of treatment, but this diminishes as pathological changes in the striatum lead to synaptic dopamine levels becoming completely dependent on exogenous dopamine. This non-physiologic stimulation of dopamine receptors eventually manifests as OFF episodes. As no disease modifying therapy exists for PD that can disrupt these pathological changes, most research and treatment focuses on optimization of dopaminergic stimulation of striatal receptors so that they mimic tonic, physiologic stimulation as closely as possible. Strategies focusing on these challenges have included non-pharmacologic approaches, optimizing levodopa pharmacokinetics, using adjunctive treatments including those with non-dopaminergic mechanisms, and implementing rescue therapies. Device aided therapies, including surgery, are also available. In this review, we will focus on effective management of motor symptoms related to OFF periods, including emerging strategies. Unmet clinical needs will be discussed, including non-motor symptoms, targeted molecular therapies and disease modifying therapy.
Collapse
Affiliation(s)
- Nbaa Masood
- Department of Neurology, Icahn School of Medicine at Mount Sinai, Mount Sinai West, New York, NY, USA
| | - Joohi Jimenez-Shahed
- Department of Neurology, Icahn School of Medicine at Mount Sinai, Mount Sinai West, New York, NY, USA
| |
Collapse
|
6
|
Hastings N, Kuan WL, Osborne A, Kotter MRN. Therapeutic Potential of Astrocyte Transplantation. Cell Transplant 2022; 31:9636897221105499. [PMID: 35770772 PMCID: PMC9251977 DOI: 10.1177/09636897221105499] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell transplantation is an attractive treatment strategy for a variety of brain disorders, as it promises to replenish lost functions and rejuvenate the brain. In particular, transplantation of astrocytes has come into light recently as a therapy for amyotrophic lateral sclerosis (ALS); moreover, grafting of astrocytes also showed positive results in models of other conditions ranging from neurodegenerative diseases of older age to traumatic injury and stroke. Despite clear differences in etiology, disorders such as ALS, Parkinson's, Alzheimer's, and Huntington's diseases, as well as traumatic injury and stroke, converge on a number of underlying astrocytic abnormalities, which include inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. In this review, we examine these convergent pathways leading to astrocyte dysfunction, and explore the existing evidence for a therapeutic potential of transplantation of healthy astrocytes in various models. Existing literature presents a wide variety of methods to generate astrocytes, or relevant precursor cells, for subsequent transplantation, while described outcomes of this type of treatment also differ between studies. We take technical differences between methodologies into account to understand the variability of therapeutic benefits, or lack thereof, at a deeper level. We conclude by discussing some key requirements of an astrocyte graft that would be most suitable for clinical applications.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andrew Osborne
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R N Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Ion-Channel Antiepileptic Drugs: An Analytical Perspective on the Therapeutic Drug Monitoring (TDM) of Ezogabine, Lacosamide, and Zonisamide. ANALYTICA 2021. [DOI: 10.3390/analytica2040016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The term seizures includes a wide array of different disorders with variable etiology, which currently represent one of the most important classes of neurological illnesses. As a consequence, many different antiepileptic drugs (AEDs) are currently available, exploiting different activity mechanisms and providing different levels of performance in terms of selectivity, safety, and efficacy. AEDs are currently among the psychoactive drugs most frequently involved in therapeutic drug monitoring (TDM) practices. Thus, the plasma levels of AEDs and their metabolites are monitored and correlated to administered doses, therapeutic efficacy, side effects, and toxic effects. As for any analytical endeavour, the quality of plasma concentration data is only as good as the analytical method allows. In this review, the main techniques and methods are described, suitable for the TDM of three AEDs belonging to the class of ion channel agents: ezogabine (or retigabine), lacosamide, and zonisamide. In addition to this analytical overview, data are provided, pertaining to two of the most important use cases for the TDM of antiepileptics: drug–drug interactions and neuroprotection activity studies. This review contains 146 references.
Collapse
|
8
|
Asanuma M, Miyazaki I. Glutathione and Related Molecules in Parkinsonism. Int J Mol Sci 2021; 22:ijms22168689. [PMID: 34445395 PMCID: PMC8395390 DOI: 10.3390/ijms22168689] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Glutathione (GSH) is the most abundant intrinsic antioxidant in the central nervous system, and its substrate cysteine readily becomes the oxidized dimeric cystine. Since neurons lack a cystine transport system, neuronal GSH synthesis depends on cystine uptake via the cystine/glutamate exchange transporter (xCT), GSH synthesis, and release in/from surrounding astrocytes. Transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), a detoxifying master transcription factor, is expressed mainly in astrocytes and activates the gene expression of various phase II drug-metabolizing enzymes or antioxidants including GSH-related molecules and metallothionein by binding to the antioxidant response element (ARE) of these genes. Accumulating evidence has shown the involvement of dysfunction of antioxidative molecules including GSH and its related molecules in the pathogenesis of Parkinson’s disease (PD) or parkinsonian models. Furthermore, we found several agents targeting GSH synthesis in the astrocytes that protect nigrostriatal dopaminergic neuronal loss in PD models. In this article, the neuroprotective effects of supplementation and enhancement of GSH and its related molecules in PD pathology are reviewed, along with introducing new experimental findings, especially targeting of the xCT-GSH synthetic system and Nrf2–ARE pathway in astrocytes.
Collapse
|
9
|
Cerebrospinal fluid levels of oxidative stress measured using diacron-reactive oxygen metabolites and biological antioxidant potential in patients with Parkinson's disease and progressive supranuclear palsy. Neurosci Lett 2021; 757:135975. [PMID: 34023404 DOI: 10.1016/j.neulet.2021.135975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. Because no curative therapy is available for PD, elucidation of its pathophysiology is important to establish more effective treatments. Oxidative stress (OS) has gained attention and been investigated as one of the candidates involved in the pathogenesis of PD. This study aimed to evaluate OS in the cerebrospinal fluid (CSF) of patients with PD and progressive supranuclear palsy (PSP) using diacron-reactive oxygen metabolites (d-ROMs) and biological antioxidant potential (BAP) tests, which can easily assess OS in liquid samples. Results were compared to the clinical background of patients and with those of the normal control (NC) group. CSF samples were obtained from 69 patients with PD, 14 patients with PSP, and 22 individuals in the NC group. OS levels and antioxidant capacity were measured using d-ROMs and BAP tests, respectively. CSF d-ROM levels were extremely low (<10 U.CARR) in all 3 groups than the plasma d-ROM levels. Antioxidant capacity was significantly higher in patients with PSP (1074 ± 79 μM) than in patients with PD (918 ± 350 μM) (p = 0.019). In the PD group, antioxidant capacity was significantly lower in patients with tremor (858 ± 269 μM) than in those without tremor (1132 ± 505 μM) (p = 0.004). Our study suggests that the CSF level of OS is under homeostatic control of antioxidative mechanisms in healthy individuals as well as those with neurodegenerative diseases, and increased antioxidant capacity can indicate the CSF level of OS. The lower CSF level of OS in the tremor dominant subtype of PD may be the reason for the benign clinical course.
Collapse
|
10
|
Tousi B, Leverenz JB. The Application of Zonisamide to Patients Suffering from Dementia with Lewy Bodies: Emerging Clinical Data. Drug Des Devel Ther 2021; 15:1811-1817. [PMID: 33976533 PMCID: PMC8106402 DOI: 10.2147/dddt.s240865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/08/2021] [Indexed: 11/23/2022] Open
Abstract
Zonisamide is an anti-epileptic medication with multiple mechanisms of action and a favorable safety profile. Zonisamide may interact with Lewy body dementia pathophysiology through a mechanism unrelated to its original indication. Zonisamide has shown efficacy as adjunct therapy for the management of motor symptoms in patients with Parkinson's disease (PD). Given that dementia with Lewy bodies (DLB) and PD are considered subtypes of a Lewy body disease spectrum, zonisamide was investigated for the treatment of parkinsonism in DLB. Phase II and phase III clinical trials were conducted in patients with DLB in Japan. In both studies, participants were randomized to receive 12 weeks of zonisamide 25 or 50 mg/day or placebo. Zonisamide significantly improved the Unified Parkinson's Disease Rating Scale Part III (UPDRS-III) without affecting the Mini-Mental State Examination (MMSE) or Neuropsychiatry Inventory-10 (NPI-10) scores at week 12. In 2018, zonisamide received Japanese regulatory approval for the additional indication of parkinsonism in DLB. This review discusses the emerging clinical data on zonisamide in the field of DLB.
Collapse
Affiliation(s)
- Babak Tousi
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
11
|
Zhao YS, Zhang M, Li Q. Protective effects of 1,2-benzisoxazole-3-methanesulfonamide (zonisamide)- loaded polymeric micelles against neurotoxicity in spinal cord: In vitro. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
He J, Zhang X, He W, Xie Y, Chen Y, Yang Y, Chen R. Neuroprotective effects of zonisamide on cerebral ischemia injury via inhibition of neuronal apoptosis. ACTA ACUST UNITED AC 2021; 54:e10498. [PMID: 33656055 PMCID: PMC7917778 DOI: 10.1590/1414-431x202010498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
It is known that neuronal apoptosis contributes to pathology of cerebral ischemia injury. Zonisamide (ZNS) has shown anti-apoptosis effects in recent studies. The present study investigated whether the anti-apoptotic effect can account for the neuroprotective action of ZNS on cerebral ischemia. Neuronal cells were maintained under oxygen-glucose deprivation conditions to simulate cerebral ischemia and treated with ZNS simultaneously. The apoptosis of the cells and expression of apoptosis-related proteins were investigated by flow cytometry and western blot analysis, respectively. A cerebral ischemia mouse model was created via middle cerebral artery occlusion, and the mice were treated with ZNS. Neurological deficit scores and infarct volumes of the cerebral ischemia mice were measured. The apoptosis status of the neuronal cells was evaluated by TUNEL staining. In vitro, the ZNS treatment inhibited both the apoptosis of the neuronal cells and apoptosis-related protein expression (caspase-3, caspase-8, and calpain-1) induced by the oxygen-glucose deprivation. The anti-apoptosis effect of ZNS could occur through the blocking of reactive oxygen species. Moreover, ZNS treatment significantly ameliorated neurological deficits and reduced infarct volumes in the cerebral ischemia mice model. In this study, ZNS exerted neuroprotective effects by inhibition of apoptosis in neuronal cells in cerebral ischemia. Therefore, ZNS might be a promising therapy for cerebral ischemia.
Collapse
Affiliation(s)
- Junna He
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weiliang He
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yanzhao Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yanxia Chen
- Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yang Yang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rong Chen
- Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, China
| |
Collapse
|
13
|
Angelopoulou E, Paudel YN, Piperi C. Emerging role of S100B protein implication in Parkinson's disease pathogenesis. Cell Mol Life Sci 2021; 78:1445-1453. [PMID: 33052436 PMCID: PMC11073186 DOI: 10.1007/s00018-020-03673-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/10/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
The exact etiology of Parkinson's disease (PD) remains obscure, lacking effective diagnostic and prognostic biomarkers. In search of novel molecular factors that may contribute to PD pathogenesis, emerging evidence highlights the multifunctional role of the calcium-binding protein S100B that is widely expressed in the brain and predominantly in astrocytes. Preclinical evidence points towards the possible time-specific contributing role of S100B in the pathogenesis of neurodegenerative disorders including PD, mainly by regulating neuroinflammation and dopamine metabolism. Although existing clinical evidence presents some contradictions, estimation of S100B in the serum and cerebrospinal fluid seems to hold a great promise as a potential PD biomarker, particularly regarding the severity of motor and non-motor PD symptoms. Furthermore, given the recent development of S100B inhibitors that are able to cross the blood brain barrier, novel opportunities are arising in the research field of PD therapeutics. In this review, we provide an update on recent advances in the implication of S100B protein in the pathogenesis of PD and discuss relevant studies investigating the biomarker potential of S100B in PD, aiming to shed more light on clinical targeting approaches related to this incurable disorder.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527, Athens, Greece.
| |
Collapse
|
14
|
Ishiguro S, Shinada T, Wu Z, Karimazawa M, Uchidate M, Nishimura E, Yasuno Y, Ebata M, Sillapakong P, Ishiguro H, Ebata N, Ni J, Jiang M, Goryo M, Otsu K, Harada H, Suzuki K. A novel cyclic peptide (Naturido) modulates glia-neuron interactions in vitro and reverses ageing-related deficits in senescence-accelerated mice. PLoS One 2021; 16:e0245235. [PMID: 33503058 PMCID: PMC7840003 DOI: 10.1371/journal.pone.0245235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/26/2020] [Indexed: 12/27/2022] Open
Abstract
The use of agents that target both glia and neurons may represent a new strategy for the treatment of ageing disorders. Here, we confirmed the presence of the novel cyclic peptide Naturido that originates from a medicinal fungus (Isaria japonica) grown on domestic silkworm (Bombyx mori). We found that Naturido significantly enhanced astrocyte proliferation and activated the single copy gene encoding the neuropeptide VGF and the neuron-derived NGF gene. The addition of the peptide to the culture medium of primary hippocampal neurons increased dendrite length, dendrite number and axon length. Furthermore, the addition of the peptide to primary microglial cultures shifted CGA-activated microglia towards anti-inflammatory and neuroprotective phenotypes. These findings of in vitro glia–neuron interactions led us to evaluate the effects of oral administration of the peptide on brain function and hair ageing in senescence-accelerated mice (SAMP8). In vivo analyses revealed that spatial learning ability and hair quality were improved in Naturido-treated mice compared with untreated mice, to the same level observed in the normal ageing control (SAMR1). These data suggest that Naturido may be a promising glia–neuron modulator for the treatment of not only senescence, but also Alzheimer’s disease and other neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Tetsuro Shinada
- Graduate School of Science, Osaka City University, Sumiyoshi-ku, Osaka, Japan
| | - Zhou Wu
- Faculty of Dental Science, Department of Aging Science and Pharmacology, Kyushu University, Fukuoka, Japan
- Faculty of Dental Science, OBT Research Center, Kyushu University, Fukuoka, Japan
| | | | - Michimasa Uchidate
- Faculty of Science and Engineering, Iwate University, Ueda, Morioka, Japan
| | - Eiji Nishimura
- Graduate School of Science, Osaka City University, Sumiyoshi-ku, Osaka, Japan
| | - Yoko Yasuno
- Graduate School of Science, Osaka City University, Sumiyoshi-ku, Osaka, Japan
| | - Makiko Ebata
- Biococoon Laboratories, Inc., Ueda, Morioka, Japan
| | | | | | | | - Junjun Ni
- Faculty of Dental Science, Department of Aging Science and Pharmacology, Kyushu University, Fukuoka, Japan
| | - Muzhou Jiang
- Faculty of Dental Science, Department of Aging Science and Pharmacology, Kyushu University, Fukuoka, Japan
| | | | - Keishi Otsu
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, Yahaba, Japan
| | - Hidemitsu Harada
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, Yahaba, Japan
| | - Koichi Suzuki
- Biococoon Laboratories, Inc., Ueda, Morioka, Japan
- Iwate University, Ueda, Morioka, Japan
- * E-mail:
| |
Collapse
|
15
|
Asanuma M, Miyazaki I. [Anti-oxidants in astrocytes as target of neuroprotection for Parkinson's disease]. Nihon Yakurigaku Zasshi 2021; 156:14-20. [PMID: 33390474 DOI: 10.1254/fpj.20071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Recently, it has been reported that dysfunction of astrocytes is involved vulnerability of neuronal cells in several neurological disorders. Glutathione (GSH) is the most abundant intrinsic antioxidant in the central nervous system, and its substrate cysteine is readily becomes the oxidized dimeric cystine. Since neurons lack a cystine transport system, neuronal GSH synthesis depends on cystine uptake via the cystine/glutamate exchange transporter (xCT), GSH synthesis and release in/from surrounding astrocytes. The expression and release of the zinc-binding protein metallothionein (MT) in astrocytes, which is a strong antioxidant, is induced and exerts neuroprotective in the case of dopaminergic neuronal damage. In addition, the transcription factor Nrf2 induces expression of MT-1 and GSH related molecules. We previously revealed that several antiepileptic drugs, serotonin 5-HT1A receptor agonists, plant-derived chemicals (phytochemicals) increased xCT expression, Nrf2 activation, GSH or MT expression and release in/from astrocytes, and exerted a neuroprotective effect against dopaminergic neurodegeneration in Parkinson's disease model. Our serial studies on neuroprotection via antioxidant defense mechanism of astrocytes have found three target molecular systems of astrocytes for neuroprotection: (1) xCT-GSH synthetic system, (2) Nrf2 system and (3) 5-HT1A receptor-Nrf2-MT system, 5-HT1A-S100β system. In this article, possible neuroprotective strategy for Parkinson's disease has been reviewed targeting antioxidative molecules in astrocytes.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentist and Pharmaceutical Sciences
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentist and Pharmaceutical Sciences
| |
Collapse
|
16
|
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: 5.2] [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
|
17
|
Miyazaki I, Asanuma M. Neuron-Astrocyte Interactions in Parkinson's Disease. Cells 2020; 9:cells9122623. [PMID: 33297340 PMCID: PMC7762285 DOI: 10.3390/cells9122623] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/20/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PD patients exhibit motor symptoms such as akinesia/bradykinesia, tremor, rigidity, and postural instability due to a loss of nigrostriatal dopaminergic neurons. Although the pathogenesis in sporadic PD remains unknown, there is a consensus on the involvement of non-neuronal cells in the progression of PD pathology. Astrocytes are the most numerous glial cells in the central nervous system. Normally, astrocytes protect neurons by releasing neurotrophic factors, producing antioxidants, and disposing of neuronal waste products. However, in pathological situations, astrocytes are known to produce inflammatory cytokines. In addition, various studies have reported that astrocyte dysfunction also leads to neurodegeneration in PD. In this article, we summarize the interaction of astrocytes and dopaminergic neurons, review the pathogenic role of astrocytes in PD, and discuss therapeutic strategies for the prevention of dopaminergic neurodegeneration. This review highlights neuron-astrocyte interaction as a target for the development of disease-modifying drugs for PD in the future.
Collapse
|
18
|
Hara N, Morino H, Matsuda Y, Satoh K, Hashimoto K, Maruyama H, Kawakami H. Zonisamide can ameliorate the voltage-dependence alteration of the T-type calcium channel Ca V3.1 caused by a mutation responsible for spinocerebellar ataxia. Mol Brain 2020; 13:163. [PMID: 33243296 PMCID: PMC7690142 DOI: 10.1186/s13041-020-00700-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/16/2020] [Indexed: 11/25/2022] Open
Abstract
Spinocerebellar ataxia (SCA) 42 is caused by a mutation in CACNA1G, which encodes the low voltage-gated calcium channel CaV3.1 (T-type). Patients with SCA42 exhibit a pure form of cerebellar ataxia. We encountered a patient with the p.Arg1715His mutation, suffering from intractable resting tremor, particularly head tremor. This symptom improved with the administration of low-dose of zonisamide (ZNS), a T-type calcium channel blocker effective for treating Parkinson’s disease and epilepsy. Previous electrophysiological studies showed that the voltage dependence of this mutant CaV3.1 was shifted toward the positive potential. This abnormal shift was considered a factor related to disease onset and symptoms. In this study, we performed whole-cell recordings of GFP-expressing HEK293T cells that expressed wild-type or mutant CaV3.1 and investigated the changes in the abnormal shift of voltage dependence of the mutant CaV3.1. The results showed that ZNS in an amount equivalent to the patient’s internal dose significantly ameliorated the abnormal shift in the mutant CaV3.1, giving values close to those in the wild-type. On the other hand, ZNS did not affect the voltage dependence of wild-type CaV3.1. Because CaV3.1 is known to be involved in tremogenesis, modulation of the voltage dependence of mutant CaV3.1 by ZNS might have contributed to improvement in the intractable tremor of our patient with SCA42. Moreover, efonidipine, another T-type calcium channel blocker, had no effect on tremors in our patient with SCA42 and did not improve the abnormal shift in the voltage dependence of the mutant CaV3.1. This indicates that ZNS is distinct from other T-type calcium channel blockers in terms of modulation of the voltage dependence of the mutant CaV3.1.
Collapse
Affiliation(s)
- Naoyuki Hara
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Hiroyuki Morino
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Yukiko Matsuda
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kenichi Satoh
- The Center for Data Science Education and Research, Shiga University, 1-1-1 Banba, Hikone, Shiga, 522-8522, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Hideshi Kawakami
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| |
Collapse
|
19
|
Zonisamide ameliorates progression of cervical spondylotic myelopathy in a rat model. Sci Rep 2020; 10:13138. [PMID: 32753675 PMCID: PMC7403578 DOI: 10.1038/s41598-020-70068-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 07/08/2020] [Indexed: 12/15/2022] Open
Abstract
Cervical spondylotic myelopathy (CSM) is caused by chronic compression of the spinal cord and is the most common cause of myelopathy in adults. No drug is currently available to mitigate CSM. Herein, we made a rat model of CSM by epidurally implanting an expanding water-absorbent polymer underneath the laminae compress the spinal cord. The CSM rats exhibited progressive motor impairments recapitulating human CSM. CSM rats had loss of spinal motor neurons, and increased lipid peroxidation in the spinal cord. Zonisamide (ZNS) is clinically used for epilepsy and Parkinson's disease. We previously reported that ZNS protected primary spinal motor neurons against oxidative stress. We thus examined the effects of ZNS on our rat CSM model. CSM rats with daily intragastric administration of 0.5% methylcellulose (n = 11) and ZNS (30 mg/kg/day) in 0.5% methylcellulose (n = 11). Oral administration of ZNS ameliorated the progression of motor impairments, spared the number of spinal motor neurons, and preserved myelination of the pyramidal tracts. In addition, ZNS increased gene expressions of cystine/glutamate exchange transporter (xCT) and metallothionein 2A in the spinal cord in CSM rats, and also in the primary astrocytes. ZNS increased the glutathione (GSH) level in the spinal motor neurons of CSM rats. ZNS potentially ameliorates loss of the spinal motor neurons and demyelination of the pyramidal tracts in patients with CSM.
Collapse
|
20
|
Zeng Z, Roussakis AA, Lao-Kaim NP, Piccini P. Astrocytes in Parkinson's disease: from preclinical assays to in vivo imaging and therapeutic probes. Neurobiol Aging 2020; 95:264-270. [PMID: 32905922 DOI: 10.1016/j.neurobiolaging.2020.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/30/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is increasingly thought to be associated with glial pathology. Recently, research in neurodegenerative disorders has applied a greater focus to better understanding the role of astrocytes in the disease pathophysiology. In this article, we review results from the latest preclinical and clinical work, including functional imaging studies on astrocytes in PD and highlight key molecules that may prove valuable as biomarkers. We discuss how astrocytes may contribute to the initiation and progression of PD. We additionally present trials of investigational medicinal products and the current background for the design of future clinical trials.
Collapse
Affiliation(s)
- Zhou Zeng
- Department of Brain Sciences, Imperial College London, Neurology Imaging Unit, London, UK; Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | - Nicholas P Lao-Kaim
- Department of Brain Sciences, Imperial College London, Neurology Imaging Unit, London, UK
| | - Paola Piccini
- Department of Brain Sciences, Imperial College London, Neurology Imaging Unit, London, UK.
| |
Collapse
|
21
|
Miyawaki Y, Samata B, Kikuchi T, Nishimura K, Takahashi J. Zonisamide promotes survival of human-induced pluripotent stem cell-derived dopaminergic neurons in the striatum of female rats. J Neurosci Res 2020; 98:1575-1587. [PMID: 32506530 PMCID: PMC7497107 DOI: 10.1002/jnr.24668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 11/12/2022]
Abstract
The transplantation of dopaminergic (DA) progenitors derived from pluripotent stem cells improves the behavior of Parkinson's disease model animals. However, the survival of DA progenitors is low, and the final yield of DA neurons is only approximately 0.3%–2% the number of transplanted cells. Zonisamide (ZNS) increases the number of survived DA neurons upon the transplantation of mouse‐induced pluripotent stem (iPS) cell‐derived DA progenitors in the rat striatum. In this study, we induced DA progenitors from human iPS cells and transplanted them into the striatum of female rats with daily administration of ZNS. The number of survived DA neurons was evaluated 1 and 4 months after transplantation by immunohistochemistry, which revealed that the number of survived DA neurons was significantly increased with the administration of ZNS. To assess the mechanism of action of ZNS, we performed a gene expression analysis to compare the gene expression profiles in striatum treated with or without ZNS. The analysis revealed that the expression of SLIT‐and NTRK‐like protein 6 (SLITRK6) was upregulated in rat striatum treated with ZNS. In conclusion, ZNS promotes the survival of DA neurons after the transplantation of human‐iPS cell‐derived DA progenitors in the rat striatum. SLITRK6 is suggested to be involved in this supportive effect of ZNS by modulating the environment of the host brain.
Collapse
Affiliation(s)
- Yoshifumi Miyawaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Bumpei Samata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Tetsuhiro Kikuchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Kaneyasu Nishimura
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| |
Collapse
|
22
|
Genome-wide association study identifies zonisamide responsive gene in Parkinson's disease patients. J Hum Genet 2020; 65:693-704. [PMID: 32355309 PMCID: PMC8075945 DOI: 10.1038/s10038-020-0760-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
Long-term treatment of Parkinson's disease (PD) by levodopa leads to motor complication "wearing-off". Zonisamide is a nondopaminergic antiparkinsonian drug that can improve "wearing-off" although response to the treatment varies between individuals. To clarify the genetic basis of zonisamide responsiveness, we conducted a genome-wide association study (GWAS) on 200 PD patients from a placebo-controlled clinical trial, including 67 responders whose "off" time decreased ≥1.5 h after 12 weeks of zonisamide treatment and 133 poor responders. We genotyped and evaluated the association between 611,492 single nucleotide polymorphisms (SNPs) and "off" time reduction. We also performed whole-genome imputation, gene- and pathway-based analyses of GWAS data. For promising SNPs, we examined single-tissue expression quantitative trait loci (eQTL) data in the GTEx database. SNP rs16854023 (Mouse double minute 4, MDM4) showed genome-wide significant association with reduced "off" time (PAdjusted = 4.85 × 10-9). Carriers of responsive genotype showed >7-fold decrease in mean "off" time compared to noncarriers (1.42 h vs 0.19 h; P = 2.71 × 10-7). In silico eQTL data indicated that zonisamide sensitivity is associated with higher MDM4 expression. Among the 37 pathways significantly influencing "off" time, calcium and glutamate signaling have also been associated with anti-epileptic effect of zonisamide. MDM4 encodes a negative regulator of p53. The association between improved motor fluctuation and MDM4 upregulation implies that p53 inhibition may prevent dopaminergic neuron loss and consequent motor symptoms. This is the first genome-wide pharmacogenetics study on antiparkinsonian drug. The findings provide a basis for improved management of "wearing-off" in PD by genotype-guided zonisamide treatment.
Collapse
|
23
|
Takaku S, Sango K. Zonisamide enhances neurite outgrowth from adult rat dorsal root ganglion neurons, but not proliferation or migration of Schwann cells. Histochem Cell Biol 2019; 153:177-184. [PMID: 31879799 PMCID: PMC7060162 DOI: 10.1007/s00418-019-01839-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2019] [Indexed: 01/06/2023]
Abstract
Zonisamide, an anti-epileptic and anti-Parkinson’s disease drug, displays neurotrophic activity on cultured motor neurons and facilitates axonal regeneration after peripheral nerve injury in mice, but its underlying mechanisms remain unclear. In this study, zonisamide enhanced neurite outgrowth from cultured adult rat dorsal root ganglion (DRG) neurons in a concentration-dependent manner (1 μM < 10 μM < 100 μM), and its activity was significantly attenuated by co-treatment with a phosphatidyl inositol-3′-phosphate-kinase (PI3K) inhibitor LY294002 or a mitogen-activated protein kinase (MAPK) inhibitor U0126. In agreement with these findings, 100 μM zonisamide for 1 h induced phosphorylation of AKT and ERK1/2, key molecules of PI3K and MAPK signaling pathways, respectively in mouse neuroblastoma × rat DRG neuron hybrid cells ND7/23. In contrast, zonisamide failed to promote proliferation or migration of immortalized Fischer rat Schwann cells 1 (IFRS1). These findings suggest that the beneficial effects of zonisamide on peripheral nerve regeneration may be attributable to its direct actions on neurons through PI3K and MAPK pathways, rather than the stimulation of Schwann cells.
Collapse
Affiliation(s)
- Shizuka Takaku
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| |
Collapse
|
24
|
Mashima K, Takahashi S, Minami K, Izawa Y, Abe T, Tsukada N, Hishiki T, Suematsu M, Kajimura M, Suzuki N. Neuroprotective Role of Astroglia in Parkinson Disease by Reducing Oxidative Stress Through Dopamine-Induced Activation of Pentose-Phosphate Pathway. ASN Neuro 2019; 10:1759091418775562. [PMID: 29768946 PMCID: PMC5960859 DOI: 10.1177/1759091418775562] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress plays an important role in the onset and progression of Parkinson disease. Although released dopamine at the synaptic terminal is mostly reabsorbed by dopaminergic neurons, some dopamine is presumably taken up by astroglia. This study examined the dopamine-induced astroglial protective function through the activation of the pentose-phosphate pathway (PPP) to reduce reactive oxygen species (ROS). In vitro experiments were performed using striatal neurons and cortical or striatal astroglia prepared from Sprague-Dawley rats or C57BL/6 mice. The rates of glucose phosphorylation in astroglia were evaluated using the [14C]deoxyglucose method. PPP activity was measured using [1-14C]glucose and [6-14C]glucose after acute (60 min) or chronic (15 hr) exposure to dopamine. ROS production was measured using 2',7'-dichlorodihydrofluorescein diacetate. The involvement of the Kelch-like ECH-associated protein 1 (Keap1) or nuclear factor-erythroid-2-related factor 2 (Nrf2) system was evaluated using Nrf2 gene knockout mice, immunohistochemistry, and quantitative reverse transcription polymerase chain reaction analysis for heme oxygenase-1. Acute exposure to dopamine elicited increases in astroglial glucose consumption with lactate release. PPP activity in astroglia was robustly enhanced independently of Na+-dependent monoamine transporters. In contrast, chronic exposure to dopamine induced moderate increases in PPP activity via the Keap1/Nrf2 system. ROS production from dopamine increased gradually over 12 hr. Dopamine induced neuronal cell damage that was prevented by coculturing with astroglia but not with Nrf2-deficient astroglia. Dopamine-enhanced astroglial PPP activity in both acute and chronic manners may possibly reduce neuronal oxidative stress.
Collapse
Affiliation(s)
- Kyoko Mashima
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichi Takahashi
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kazushi Minami
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshikane Izawa
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Takato Abe
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan.,2 Department of Neurology, Graduate School of Medicine, Osaka City University, Japan
| | - Naoki Tsukada
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- 3 Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan.,4 Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- 4 Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Kajimura
- 5 Department of Biology, Keio University School of Medicine, Yokohama, Japan
| | - Norihiro Suzuki
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
25
|
Iwaki H, Tagawa M, Iwasaki K, Kawakami K, Nomoto M. Comparison of zonisamide with non-levodopa, anti-Parkinson's disease drugs in the incidence of Parkinson's disease-relevant symptoms. J Neurol Sci 2019; 402:145-152. [DOI: 10.1016/j.jns.2019.05.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/22/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
|
26
|
Asanuma M, Okumura-Torigoe N, Miyazaki I, Murakami S, Kitamura Y, Sendo T. Region-Specific Neuroprotective Features of Astrocytes against Oxidative Stress Induced by 6-Hydroxydopamine. Int J Mol Sci 2019; 20:ijms20030598. [PMID: 30704073 PMCID: PMC6387089 DOI: 10.3390/ijms20030598] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 01/09/2023] Open
Abstract
In previous studies, we found regional differences in the induction of antioxidative molecules in astrocytes against oxidative stress, postulating that region-specific features of astrocytes lead region-specific vulnerability of neurons. We examined region-specific astrocytic features against dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) as an oxidative stress using co-culture of mesencephalic neurons and mesencephalic or striatal astrocytes in the present study. The 6-OHDA-induced reduction of mesencephalic dopamine neurons was inhibited by co-culturing with astrocytes. The co-culture of midbrain neurons with striatal astrocytes was more resistant to 6-OHDA than that with mesencephalic astrocytes. Furthermore, glia conditioned medium from 6-OHDA-treated striatal astrocytes showed a greater protective effect on the 6-OHDA-induced neurotoxicity and oxidative stress than that from mesencephalic astrocytes. The cDNA microarray analysis showed that the number of altered genes in both mesencephalic and striatal astrocytes was fewer than that changed in either astrocyte. The 6-OHDA treatment, apparently up-regulated expressions of Nrf2 and some anti-oxidative or Nrf2-regulating phase II, III detoxifying molecules related to glutathione synthesis and export in the striatal astrocytes but not mesencephalic astrocytes. There is a profound regional difference of gene expression in astrocytes induced by 6-OHDA. These results suggest that protective features of astrocytes against oxidative stress are more prominent in striatal astrocytes, possibly by secreting humoral factors in striatal astrocytes.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medical, Dental and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Nao Okumura-Torigoe
- Department of Clinical Pharmacy, Okayama University Graduate School of Medical, Dental and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medical, Dental and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Shinki Murakami
- Department of Medical Neurobiology, Okayama University Graduate School of Medical, Dental and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Yoshihisa Kitamura
- Department of Clinical Pharmacy, Okayama University Graduate School of Medical, Dental and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Toshiaki Sendo
- Department of Clinical Pharmacy, Okayama University Graduate School of Medical, Dental and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| |
Collapse
|
27
|
The effects of zonisamide on L-DOPA-induced dyskinesia in Parkinson's disease model mice. Neurochem Int 2019; 124:171-180. [PMID: 30639196 DOI: 10.1016/j.neuint.2019.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 01/24/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by the loss of dopaminergic neurons in the midbrain and shows motor dysfunctions. Zonisamide (ZNS, 1,2-benzisoxazole-3-methanesulfonamide), which was originally developed as an antiepileptic drug, was also found to have beneficial effects on motor symptoms in PD. In the current study, we have investigated the behavioral and physiological effects of ZNS on L-DOPA-induced dyskinesia (LID) in PD model mice. Chronic administration of L-DOPA plus ZNS in PD model mice was shown to increase the duration and severity of LID compared with PD model mice that were treated with L-DOPA alone. To elucidate the neural mechanism of the effects of ZNS on LID, we examined neuronal activity in the output nuclei of the basal ganglia, i.e., the substantia nigra pars reticulata (SNr). Chronic administration of L-DOPA plus ZNS in PD mice decreased the firing rate in the SNr while they showed apparent LID. In addition, chronic treatment of L-DOPA plus ZNS in PD mice changed cortically evoked responses in the SNr during LID. In the control state, motor cortical stimulation induces the triphasic response composed of early excitation, inhibition, and late excitation. In contrast, L-DOPA plus ZNS-treated PD mice showed longer inhibition and reduced late excitation. Previous studies proposed that inhibition in the SNr is derived from the direct pathway and releases movements, and that late excitation is derived from the indirect pathway and stops movements. These changes of the direct and indirect pathways possibly underlie the effects of ZNS on LID.
Collapse
|
28
|
Ueno SI, Saiki S, Fujimaki M, Takeshige-Amano H, Hatano T, Oyama G, Ishikawa KI, Yamaguchi A, Nojiri S, Akamatsu W, Hattori N. Zonisamide Administration Improves Fatty Acid β-Oxidation in Parkinson's Disease. Cells 2018; 8:cells8010014. [PMID: 30597973 PMCID: PMC6356654 DOI: 10.3390/cells8010014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/20/2018] [Accepted: 12/25/2018] [Indexed: 12/03/2022] Open
Abstract
Although many experimental studies have shown the favorable effects of zonisamide on mitochondria using models of Parkinson’s disease (PD), the influence of zonisamide on metabolism in PD patients remains unclear. To assess metabolic status under zonisamide treatment in PD, we performed a pilot study using a comprehensive metabolome analysis. Plasma samples were collected for at least one year from 30 patients with PD: 10 without zonisamide medication and 20 with zonisamide medication. We performed comprehensive metabolome analyses of plasma with capillary electrophoresis time-of-flight mass spectrometry and liquid chromatography time-of-flight mass spectrometry. We also measured disease severity using Hoehn and Yahr (H&Y) staging and the Unified Parkinson’s Disease Rating Scale (UPDRS) motor section, and analyzed blood chemistry. In PD with zonisamide treatment, 15 long-chain acylcarnitines (LCACs) tended to be increased, of which four (AC(12:0), AC(12:1)-1, AC(16:1), and AC(16:2)) showed statistical significance. Of these, two LCACs (AC(16:1) and AC(16:2)) were also identified by partial least squares analysis. There was no association of any LCAC with age, disease severity, levodopa daily dose, or levodopa equivalent dose. Because an upregulation of LCACs implies improvement of mitochondrial β-oxidation, zonisamide might be beneficial for mitochondrial β-oxidation, which is suppressed in PD.
Collapse
Affiliation(s)
- Shin-Ichi Ueno
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Shinji Saiki
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Motoki Fujimaki
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Haruka Takeshige-Amano
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Taku Hatano
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Genko Oyama
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Kei-Ichi Ishikawa
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Akihiro Yamaguchi
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
29
|
A mechanistic approach to explore the neuroprotective potential of zonisamide in seizures. Inflammopharmacology 2018; 26:1125-1131. [PMID: 29644555 DOI: 10.1007/s10787-018-0478-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/30/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Epilepsy, a disease of the brain, is one of the most common serious neurological conditions. It is associated with a group of processes which alter energy metabolism, interrupt cellular ionic homeostasis, cause receptor dysfunction, activate inflammatory cascade, alter neurotransmitter uptake and result in neuronal damage. The increasing knowledge and understanding about the basis of neuronal changes in epilepsy lead to investigate the mechanistic pathway of neuroprotective agents in epilepsy. With this background, the present study is designed to reveal the molecular and biochemical mechanisms involved in the neuroprotective potential of zonisamide in epilepsy. METHODS Seizure-induced neuronal damage was produced by maximal electroshock seizures in animals. The oxidative stress and neuroinflammatory and apoptotic markers were assessed in the brain tissue of animals. RESULTS AND DISCUSSION The present findings revealed that zonisamide treatment prevented the development of seizures in animals. Seizures-induced free radicals production and neuroinflammation were markedly ameliorated by zonisamide administration. In conclusion, the present study demonstrated the mechanisms behind the strong neuroprotective potential of zonisamide against seizures by attenuating the oxidative stress, inflammatory cascade and neuronal death associated with progression of seizures. It can be further developed as a neuroprotective agent for epilepsy and other neurodegenerative disorders.
Collapse
|
30
|
Simmons AD. Parkinson’s Disease. Integr Med (Encinitas) 2018. [DOI: 10.1016/b978-0-323-35868-2.00015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
Uemura MT, Asano T, Hikawa R, Yamakado H, Takahashi R. Zonisamide inhibits monoamine oxidase and enhances motor performance and social activity. Neurosci Res 2017. [DOI: 10.1016/j.neures.2017.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Liu B, Teschemacher AG, Kasparov S. Astroglia as a cellular target for neuroprotection and treatment of neuro-psychiatric disorders. Glia 2017; 65:1205-1226. [PMID: 28300322 PMCID: PMC5669250 DOI: 10.1002/glia.23136] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/12/2022]
Abstract
Astrocytes are key homeostatic cells of the central nervous system. They cooperate with neurons at several levels, including ion and water homeostasis, chemical signal transmission, blood flow regulation, immune and oxidative stress defense, supply of metabolites and neurogenesis. Astroglia is also important for viability and maturation of stem-cell derived neurons. Neurons critically depend on intrinsic protective and supportive properties of astrocytes. Conversely, all forms of pathogenic stimuli which disturb astrocytic functions compromise neuronal functionality and viability. Support of neuroprotective functions of astrocytes is thus an important strategy for enhancing neuronal survival and improving outcomes in disease states. In this review, we first briefly examine how astrocytic dysfunction contributes to major neurological disorders, which are traditionally associated with malfunctioning of processes residing in neurons. Possible molecular entities within astrocytes that could underpin the cause, initiation and/or progression of various disorders are outlined. In the second section, we explore opportunities enhancing neuroprotective function of astroglia. We consider targeting astrocyte-specific molecular pathways which are involved in neuroprotection or could be expected to have a therapeutic value. Examples of those are oxidative stress defense mechanisms, glutamate uptake, purinergic signaling, water and ion homeostasis, connexin gap junctions, neurotrophic factors and the Nrf2-ARE pathway. We propose that enhancing the neuroprotective capacity of astrocytes is a viable strategy for improving brain resilience and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Beihui Liu
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
| | - Anja G. Teschemacher
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
| | - Sergey Kasparov
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
- Institute for Chemistry and BiologyBaltic Federal UniversityKaliningradRussian Federation
| |
Collapse
|
33
|
Finsterer J, Scorza FA. Effects of antiepileptic drugs on mitochondrial functions, morphology, kinetics, biogenesis, and survival. Epilepsy Res 2017; 136:5-11. [PMID: 28732239 DOI: 10.1016/j.eplepsyres.2017.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/22/2017] [Accepted: 07/04/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Antiepileptic drugs (AEDs) exhibit adverse and beneficial effects on mitochondria, which have a strong impact on the treatment of patients with a mitochondrial disorder (MID) with epilepsy (mitochondrial epilepsy). This review aims at summarizing and discussing recent findings concerning the effect of AEDs on mitochondrial functions and the clinical consequences with regard to therapy of mitochondrial epilepsy and of MIDs in general. METHODS Literature review. RESULTS AEDs may interfere with the respiratory chain, with non-respiratory chain enzymes, carrier proteins, or mitochondrial biogenesis, with carrier proteins, membrane-bound channels or receptors and the membrane potential, with anti-oxidative defense mechanisms, with morphology, dynamics and survival of mitochondria, and with the mtDNA. There are AEDs of which adverse effects outweigh beneficial effects, such as valproic acid, carbamazepine, phenytoin, or phenobarbital and there are AEDs in which beneficial effects dominate over mitochondrial toxic effects, such as lamotrigine, levetiracetam, gabapentin, or zonisamide. However, from most AEDs only little is known about their interference with mitochondria. CONCLUSIONS Mitochondrial epilepsy might be initially treated with AEDs with low mitochondrial toxic potential. Only in case mitochondrial epilepsy is refractory to these AEDs, AEDs with higher mitochondrial toxic potential might be tried. In patients carrying POLG1 mutations AEDs with high mitochondrial toxic potential are contraindicated.
Collapse
Affiliation(s)
| | - Fulvio A Scorza
- Disciplina de Neurociência, Escola Paulista de Medicina/Universidade Federal de São Paulo, (EPM/UNIFESP), São Paulo, Brazil.
| |
Collapse
|
34
|
McBean GJ, López MG, Wallner FK. Redox-based therapeutics in neurodegenerative disease. Br J Pharmacol 2017; 174:1750-1770. [PMID: 27477685 PMCID: PMC5446580 DOI: 10.1111/bph.13551] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 06/02/2016] [Accepted: 07/01/2016] [Indexed: 12/13/2022] Open
Abstract
This review describes recent developments in the search for effective therapeutic agents that target redox homeostasis in neurodegenerative disease. The disruption to thiol redox homeostasis in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis is discussed, together with the experimental strategies that are aimed at preventing, or at least minimizing, oxidative damage in these diseases. Particular attention is given to the potential of increasing antioxidant capacity by targeting the Nrf2 pathway, the development of inhibitors of NADPH oxidases that are likely candidates for clinical use, together with strategies to reduce nitrosative stress and mitochondrial dysfunction. We describe the shortcomings of compounds that hinder their progression to the clinic and evaluate likely avenues for future research. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- G J McBean
- School of Biomolecular and Biomedical Science, Conway InstituteUniversity College DublinDublinIreland
| | - M G López
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of MedicineUniversidad Autónoma de MadridMadridSpain
| | - F K Wallner
- Redoxis ABSweden and University of SkövdeSkövdeSweden
| |
Collapse
|
35
|
Grewal GK, Kukal S, Kanojia N, Saso L, Kukreti S, Kukreti R. Effect of Oxidative Stress on ABC Transporters: Contribution to Epilepsy Pharmacoresistance. Molecules 2017; 22:molecules22030365. [PMID: 28264441 PMCID: PMC6155434 DOI: 10.3390/molecules22030365] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 01/16/2023] Open
Abstract
Epilepsy is a neurological disorder affecting around 1%–2% of population worldwide and its treatment includes use of antiepileptic drugs to control seizures. Failure to respond to antiepileptic drug therapy is a major clinical problem and over expression of ATP-binding cassette transporters is considered one of the major reasons for pharmacoresistance. In this review, we have summarized the regulation of ABC transporters in response to oxidative stress due to disease and antiepileptic drugs. Further, ketogenic diet and antioxidants were examined for their role in pharmacoresistance. The understanding of signalling pathways and mechanism involved may help in identifying potential therapeutic targets and improving drug response.
Collapse
Affiliation(s)
- Gurpreet Kaur Grewal
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India.
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi 110007, India.
| | - Samiksha Kukal
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India.
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi 110007, India.
| | - Neha Kanojia
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India.
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi 110007, India.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy.
| | - Shrikant Kukreti
- Nucleic Acids Research Lab, Department of Chemistry, University of Delhi (North Campus), Delhi 110007, India.
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India.
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi 110007, India.
| |
Collapse
|
36
|
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: 10] [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
|
37
|
Murakami S, Miyazaki I, Asanuma M. Neuroprotective effect of fermented papaya preparation by activation of Nrf2 pathway in astrocytes. Nutr Neurosci 2016; 21:176-184. [PMID: 27841081 DOI: 10.1080/1028415x.2016.1253171] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Nuclear factor erythroid 2-related factor (Nrf2) in astrocyte plays important roles in brain homeostasis. Fermented papaya preparation (FPP) has anti-oxidative, anti-inflammatory, immunoregulatory properties. The present study investigated the effects of FPP on activation of Nrf2 and release of Nrf2-regulated neuroprotective antioxidants and detoxifying molecules. METHODS Primary cultured astrocytes from rat embryos were treated with FPP for 6 or 24 hours. The expression levels of nuclear Nrf2 and cytoplasmic Nrf2-regulated molecules were determined by western blot analysis and immunohistochemistry. Glutathione levels were measured in cells and medium. Dopaminergic neurons were exposed 6-hydroxydopamine (6-OHDA) with/without pre-treatment with FPP astrocytes. Mice were treated orally with FPP for 2 weeks. RESULTS FPP increased nuclear translocation of Nrf2 in striatal astrocytes, induced up-regulation of NAD(P)H quinine oxidoreductase-1, glutathione-S transferase and hemeoxygenase-1, and increased glutathione level and the percentage of metallothionein-expressing astrocytes. Moreover, FPP suppressed 6-OHDA-induced dopaminergic neuronal loss in not only neuron-astrocyte mixed culture, but also neuron-rich cultures pre-treated with glial conditioned medium. Two-week oral treatment of mice with FPP resulted in Nrf2 activation and increase in glutathione level in striatum. DISCUSSION The results indicated that FPP enhances the anti-oxidative capacity through activation of Nrf2 in astrocytes, suggesting it may provide neuroprotection in oxidative stress-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Shinki Murakami
- a Department of Medical Neurobiology , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan.,b SAIDO Corporation , Fukuoka , Japan
| | - Ikuko Miyazaki
- a Department of Medical Neurobiology , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Masato Asanuma
- a Department of Medical Neurobiology , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| |
Collapse
|
38
|
Miyazaki I, Asanuma M. Serotonin 1A Receptors on Astrocytes as a Potential Target for the Treatment of Parkinson's Disease. Curr Med Chem 2016; 23:686-700. [PMID: 26795196 PMCID: PMC4997990 DOI: 10.2174/0929867323666160122115057] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/12/2015] [Accepted: 01/22/2016] [Indexed: 12/30/2022]
Abstract
Astrocytes are the most abundant neuron-supporting glial cells in the central nervous system. The neuroprotective role of astrocytes has been demonstrated in various neurological disorders such as amyotrophic lateral sclerosis, spinal cord injury, stroke and Parkinson’s disease (PD). Astrocyte dysfunction or loss-of-astrocytes increases the susceptibility of neurons to cell death, while astrocyte transplantation in animal studies has therapeutic advantage. We reported recently that stimulation of serotonin 1A (5-HT1A) receptors on astrocytes promoted astrocyte proliferation and upregulated antioxidative molecules to act as a neuroprotectant in parkinsonian mice. PD is a progressive neurodegenerative disease with motor symptoms such as tremor, bradykinesia, rigidity and postural instability, that are based on selective loss of nigrostriatal dopaminergic neurons, and with non-motor symptoms such as orthostatic hypotension and constipation based on peripheral neurodegeneration. Although dopaminergic therapy for managing the motor disability associated with PD is being assessed at present, the main challenge remains the development of neuroprotective or disease-modifying treatments. Therefore, it is desirable to find treatments that can reduce the progression of dopaminergic cell death. In this article, we summarize first the neuroprotective properties of astrocytes targeting certain molecules related to PD. Next, we review neuroprotective effects induced by stimulation of 5-HT1A receptors on astrocytes. The review discusses new promising therapeutic strategies based on neuroprotection against oxidative stress and prevention of dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | | |
Collapse
|
39
|
Nagatsu T, Nagatsu I. Tyrosine hydroxylase (TH), its cofactor tetrahydrobiopterin (BH4), other catecholamine-related enzymes, and their human genes in relation to the drug and gene therapies of Parkinson's disease (PD): historical overview and future prospects. J Neural Transm (Vienna) 2016; 123:1255-1278. [PMID: 27491309 DOI: 10.1007/s00702-016-1596-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/14/2016] [Indexed: 12/21/2022]
Abstract
Tyrosine hydroxylase (TH), which was discovered at the National Institutes of Health (NIH) in 1964, is a tetrahydrobiopterin (BH4)-requiring monooxygenase that catalyzes the first and rate-limiting step in the biosynthesis of catecholamines (CAs), such as dopamine, noradrenaline, and adrenaline. Since deficiencies of dopamine and noradrenaline in the brain stem, caused by neurodegeneration of dopamine and noradrenaline neurons, are mainly related to non-motor and motor symptoms of Parkinson's disease (PD), we have studied human CA-synthesizing enzymes [TH; BH4-related enzymes, especially GTP-cyclohydrolase I (GCH1); aromatic L-amino acid decarboxylase (AADC); dopamine β-hydroxylase (DBH); and phenylethanolamine N-methyltransferase (PNMT)] and their genes in relation to PD in postmortem brains from PD patients, patients with CA-related genetic diseases, mice with genetically engineered CA neurons, and animal models of PD. We purified all human CA-synthesizing enzymes, produced their antibodies for immunohistochemistry and immunoassay, and cloned all human genes, especially the human TH gene and the human gene for GCH1, which synthesizes BH4 as a cofactor of TH. This review discusses the historical overview of TH, BH4-, and other CA-related enzymes and their genes in relation to the pathophysiology of PD, the development of drugs, such as L-DOPA, and future prospects for drug and gene therapy for PD, especially the potential of induced pluripotent stem (iPS) cells.
Collapse
Affiliation(s)
- Toshiharu Nagatsu
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
- Department of Brain Functions, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan.
| | - Ikuko Nagatsu
- Department of Anatomy, School of Medicine, Fujita Health University, Toyoake, 470-1192, Japan
| |
Collapse
|
40
|
Takeshima M, Miyazaki I, Murakami S, Kita T, Asanuma M. l-Theanine protects against excess dopamine-induced neurotoxicity in the presence of astrocytes. J Clin Biochem Nutr 2016. [PMID: 27698535 DOI: 10.3164/jcbn.16.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
l-Theanine (γ-glutamylethylamide), a component of green tea, is considered to have regulatory and neuroprotective roles in the brain. The present study was designed to determine the effect of l-theanine on excess dopamine-induced neurotoxicity in both cell culture and animal experiments. The primary cultured mesencephalic neurons or co-cultures of mesencephalic neurons and striatal astrocytes were pretreated with l-theanine for 72 h, and then treated with excess dopamine for further 24 h. The cell viability of dopamine neurons and levels of glutathione were evaluated. Excess dopamine-induced neurotoxicity was significantly attenuated by 72 h preincubation with l-theanine in neuron-astrocyte co-cultures but not in neuron-rich cultures. Exposure to l-theanine increased the levels of glutathione in both astrocytes and glial conditioned medium. The glial conditioned medium from l-theanine-pretreated striatal astrocytes attenuated dopamine-induced neurotoxicity and quinoprotein formation in mesencephalic neurons. In addition, replacement of l-glutamate with l-theanine in an in vitro cell-free glutathione-synthesis system produced glutathione-like thiol compounds. Furthermore, l-theanine administration (4 mg/kg, p.o.) for 14 days significantly increased glutathione levels in the striatum of mice. The results suggest that l-theanine provides neuroprotection against oxidative stress-induced neuronal damage by humoral molecules released from astrocytes, probably including glutathione.
Collapse
Affiliation(s)
- Mika Takeshima
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shinki Murakami
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; SAIDO Co., Fukuoka 810-0021, Japan
| | - Taizo Kita
- Laboratory of Pharmacology, Kyushu Nutrition Welfare University School of Health Science, Fukuoka 803-8511, Japan
| | - Masato Asanuma
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
41
|
Asanuma M, Miyazaki I. 3-O-Methyldopa inhibits astrocyte-mediated dopaminergic neuroprotective effects of L-DOPA. BMC Neurosci 2016; 17:52. [PMID: 27456338 PMCID: PMC4960704 DOI: 10.1186/s12868-016-0289-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We evaluated the effects of 3-O-methyldopa (3-OMD), a metabolite of L-DOPA which is formed by catechol-O-methyltransferase (COMT), on the uptake, metabolism, and neuroprotective effects of L-DOPA in striatal astrocytes. We examined changes in the numbers of dopaminergic neurons after treatment with L-DOPA and 3-OMD or entacapone, a peripheral COMT inhibitor, using primary cultured mesencephalic neurons and striatal astrocytes. RESULTS The number of tyrosine hydroxylase-positive dopaminergic neurons was not affected by L-DOPA treatment in mesencephalic neurons alone. However, the increase in viability of dopaminergic neurons in the presence of astrocytes was further enhanced after methyl-L-DOPA treatment (25 µM) in mixed cultured mesencephalic neurons and striatal astrocytes. The neuroprotective effect of 25 µM L-DOPA was almost completely inhibited by simultaneous treatment with 3-OMD (10 or 100 µM), and was enhanced by concomitant treatment with entacapone (0.3 µM). The uptake of L-DOPA into and the release of glutathione from striatal astrocytes after L-DOPA treatment (100 µM) were inhibited by simultaneous exposure to 3-OMD (100 µM). CONCLUSIONS These data suggest that L-DOPA exerts its neuroprotective effect on dopaminergic neurons via astrocytes and that 3-OMD competes with L-DOPA by acting on target molecule(s) (possibly including glutathione) released from astrocytes. Since some amount of entacapone can cross the blood-brain barrier, this reagent may enhance L-DOPA transportation by inhibiting COMT and increase the astrocyte-mediated neuroprotective effects of L-DOPA on dopaminergic neurons.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan. .,Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.,Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| |
Collapse
|
42
|
Takeshima M, Miyazaki I, Murakami S, Kita T, Asanuma M. l-Theanine protects against excess dopamine-induced neurotoxicity in the presence of astrocytes. J Clin Biochem Nutr 2016; 59:93-99. [PMID: 27698535 PMCID: PMC5018574 DOI: 10.3164/jcbn.16-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 04/12/2016] [Indexed: 01/05/2023] Open
Abstract
l-Theanine (γ-glutamylethylamide), a component of green tea, is considered to have regulatory and neuroprotective roles in the brain. The present study was designed to determine the effect of l-theanine on excess dopamine-induced neurotoxicity in both cell culture and animal experiments. The primary cultured mesencephalic neurons or co-cultures of mesencephalic neurons and striatal astrocytes were pretreated with l-theanine for 72 h, and then treated with excess dopamine for further 24 h. The cell viability of dopamine neurons and levels of glutathione were evaluated. Excess dopamine-induced neurotoxicity was significantly attenuated by 72 h preincubation with l-theanine in neuron-astrocyte co-cultures but not in neuron-rich cultures. Exposure to l-theanine increased the levels of glutathione in both astrocytes and glial conditioned medium. The glial conditioned medium from l-theanine-pretreated striatal astrocytes attenuated dopamine-induced neurotoxicity and quinoprotein formation in mesencephalic neurons. In addition, replacement of l-glutamate with l-theanine in an in vitro cell-free glutathione-synthesis system produced glutathione-like thiol compounds. Furthermore, l-theanine administration (4 mg/kg, p.o.) for 14 days significantly increased glutathione levels in the striatum of mice. The results suggest that l-theanine provides neuroprotection against oxidative stress-induced neuronal damage by humoral molecules released from astrocytes, probably including glutathione.
Collapse
Affiliation(s)
- Mika Takeshima
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shinki Murakami
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; SAIDO Co., Fukuoka 810-0021, Japan
| | - Taizo Kita
- Laboratory of Pharmacology, Kyushu Nutrition Welfare University School of Health Science, Fukuoka 803-8511, Japan
| | - Masato Asanuma
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
43
|
Extrastriatal spreading of microglial activation in Parkinson's disease: a positron emission tomography study. Ann Nucl Med 2016; 30:579-87. [PMID: 27299437 DOI: 10.1007/s12149-016-1099-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/06/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND The neuroinflammatory glial response contributes to the degenerative process in Parkinson's disease (PD). However, the pattern of microglial progression remains unclear. METHODS We evaluated microglial activation in early stage PD patients by quantifying changes in neuroinflammation using PET with [(11)C]DPA713, a selective PET tracer for microglial activation. Eleven PD patients (Hoehn and Yahr stages 1-2) without dementia underwent the [(11)C]DPA713 PET scan two times with 1 year apart. The binding potential (BPND) was estimated with the simplified reference tissue model. Voxelwise and regions of interest analyses were used to compare the regional BPND among groups. RESULTS Significant increase in [(11)C]DPA713 BPND was found extrastriatally in the occipital, temporal and parietal cortex in PD patients, and the degree of BPND became much higher over the brain regions predominantly in the temporal and occipital cortex 1 year later. CONCLUSION The current results indicated that an extrastriatal spreading of microglial activation reflects one of PD pathophysiology occurring at an early stage.
Collapse
|
44
|
Affiliation(s)
- Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Yagi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
45
|
Kuhn DM, Angoa-Pérez M, Thomas DM. Nucleus accumbens invulnerability to methamphetamine neurotoxicity. ILAR J 2016; 52:352-65. [PMID: 23382149 DOI: 10.1093/ilar.52.3.352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Methamphetamine (Meth) is a neurotoxic drug of abuse that damages neurons and nerve endings throughout the central nervous system. Emerging studies of human Meth addicts using both postmortem analyses of brain tissue and noninvasive imaging studies of intact brains have confirmed that Meth causes persistent structural abnormalities. Animal and human studies have also defined a number of significant functional problems and comorbid psychiatric disorders associated with long-term Meth abuse. This review summarizes the salient features of Meth-induced neurotoxicity with a focus on the dopamine (DA) neuronal system. DA nerve endings in the caudate-putamen (CPu) are damaged by Meth in a highly delimited manner. Even within the CPu, damage is remarkably heterogeneous, with ventral and lateral aspects showing the greatest deficits. The nucleus accumbens (NAc) is largely spared the damage that accompanies binge Meth intoxication, but relatively subtle changes in the disposition of DA in its nerve endings can lead to dramatic increases in Meth-induced toxicity in the CPu and overcome the normal resistance of the NAc to damage. In contrast to the CPu, where DA neuronal deficiencies are persistent, alterations in the NAc show a partial recovery. Animal models have been indispensable in studies of the causes and consequences of Meth neurotoxicity and in the development of new therapies. This research has shown that increases in cytoplasmic DA dramatically broaden the neurotoxic profile of Meth to include brain structures not normally targeted for damage. The resistance of the NAc to Meth-induced neurotoxicity and its ability to recover reveal a fundamentally different neuroplasticity by comparison to the CPu. Recruitment of the NAc as a target of Meth neurotoxicity by alterations in DA homeostasis is significant in light of the numerous important roles played by this brain structure.
Collapse
|
46
|
Tohge R, Sakamoto S, Takahashi M. A case of cystinuria presenting with cerebellar ataxia and dementia. Pract Neurol 2016; 16:296-9. [PMID: 26929440 DOI: 10.1136/practneurol-2016-001374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2016] [Indexed: 01/22/2023]
Abstract
Cystinuria normally manifests as recurrent urinary stones and renal dysfunction, but can present to neurologists with ataxia, posterior column impairment, intellectual deficiency and pyramidal and extrapyramidal signs; the neuroradiological features include cerebellar, brainstem and cerebral atrophy. It is an autosomal recessive disease caused by a transport disorder of cystine and dibasic amino acids in renal proximal tubules. Most cases have an SLC3A1 and/or SLC7A9 gene mutation but some recent Japanese patients have had distinct heterozygous gene mutations. We report a patient with cystinuria with a heterozygous P482L mutation in the SLC7A9 gene, presenting with atrophy in the cerebellum, brainstem and cerebrum and with no urinary stones. Cystine, an amino acid comprising two cysteine molecules, is transported into cells via a cystine transporter. It is essential for producing hydrogen sulfate and the cellular antioxidant glutathione: these exert neuroprotection in astrocytes and cerebellar Purkinje cells. Although cystinuria is a metabolic disorder associated with renal dysfunction, we suspect that a trafficking defect of transporter rBAT-BAT1 in brain might cause neuronal degeneration, leading to cerebellar and cerebral atrophy.
Collapse
Affiliation(s)
- Rie Tohge
- Department of Neurology, Osaka Red Cross Hospital, Osaka, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Makio Takahashi
- Department of Neurology, Osaka Red Cross Hospital, Osaka, Japan
| |
Collapse
|
47
|
Abstract
Some of the side and beneficial effects of antiepileptic drugs (AEDs) are mediated via the influence on mitochondria. This is of particular importance in patients requiring AED treatment for mitochondrial epilepsy. AED treatment in patients with mitochondrial disorders should rely on the known influences of AEDs on these organelles. AEDs may influence various mitochondrial functions or structures in a beneficial or detrimental way. There are AEDs in which the toxic effect outweighs the beneficial effect, such as valproic acid (VPA), carbamazepine (CBZ), phenytoin (PHT), or phenobarbital (PB). There are, however, also AEDs in which the beneficial effect on mitochondria outweighs the mitochondrion-toxic effect, such as gabapentin (GBT), lamotrigine (LTG), levetiracetam (LEV), or zonisamide (ZNS). In the majority of the AEDs, however, information about their influence of mitochondria is lacking. In clinical practice mitochondrial epilepsy should be initially treated with AEDs with low mitochondrion-toxic potential. Only in cases of ineffectivity or severe mitochondrial epilepsy, mitochondrion-toxic AEDs should be given. This applies for AEDs given orally or intravenously.
Collapse
|
48
|
Yagi H, Ohkawara B, Nakashima H, Ito K, Tsushima M, Ishii H, Noto K, Ohta K, Masuda A, Imagama S, Ishiguro N, Ohno K. Zonisamide Enhances Neurite Elongation of Primary Motor Neurons and Facilitates Peripheral Nerve Regeneration In Vitro and in a Mouse Model. PLoS One 2015; 10:e0142786. [PMID: 26571146 PMCID: PMC4646494 DOI: 10.1371/journal.pone.0142786] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 10/27/2015] [Indexed: 12/25/2022] Open
Abstract
No clinically applicable drug is currently available to enhance neurite elongation after nerve injury. To identify a clinically applicable drug, we screened pre-approved drugs for neurite elongation in the motor neuron-like NSC34 cells. We found that zonisamide, an anti-epileptic and anti-Parkinson’s disease drug, promoted neurite elongation in cultured primary motor neurons and NSC34 cells in a concentration-dependent manner. The neurite-scratch assay revealed that zonisamide enhanced neurite regeneration. Zonisamide was also protective against oxidative stress-induced cell death of primary motor neurons. Zonisamide induced mRNA expression of nerve growth factors (BDNF, NGF, and neurotrophin-4/5), and their receptors (tropomyosin receptor kinase A and B). In a mouse model of sciatic nerve autograft, intragastric administration of zonisamide for 1 week increased the size of axons distal to the transected site 3.9-fold. Zonisamide also improved the sciatic function index, a marker for motor function of hindlimbs after sciatic nerve autograft, from 6 weeks after surgery. At 8 weeks after surgery, zonisamide was protective against denervation-induced muscle degeneration in tibialis anterior, and increased gene expression of Chrne, Colq, and Rapsn, which are specifically expressed at the neuromuscular junction. We propose that zonisamide is a potential therapeutic agent for peripheral nerve injuries as well as for neuropathies due to other etiologies.
Collapse
Affiliation(s)
- Hideki Yagi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Nakashima
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenyu Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikito Tsushima
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hisao Ishii
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Hand Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimitoshi Noto
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Hand Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kyotaro Ohta
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Ishiguro
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail:
| |
Collapse
|
49
|
Miyazaki I, Murakami S, Torigoe N, Kitamura Y, Asanuma M. Neuroprotective effects of levetiracetam target xCT in astrocytes in parkinsonian mice. J Neurochem 2015; 136:194-204. [DOI: 10.1111/jnc.13405] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Ikuko Miyazaki
- Department of Brain Science; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- Department of Medical Neurobiology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | - Shinki Murakami
- Department of Brain Science; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- Department of Medical Neurobiology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- SAIDO Co.; Fukuoka Japan
| | - Nao Torigoe
- Department of Clinical Pharmacy; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | - Yoshihisa Kitamura
- Department of Clinical Pharmacy; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | - Masato Asanuma
- Department of Brain Science; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- Department of Medical Neurobiology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| |
Collapse
|
50
|
Van Liefferinge J, Bentea E, Demuyser T, Albertini G, Follin-Arbelet V, Holmseth S, Merckx E, Sato H, Aerts JL, Smolders I, Arckens L, Danbolt NC, Massie A. Comparative analysis of antibodies to xCT (Slc7a11): Forewarned is forearmed. J Comp Neurol 2015; 524:1015-32. [DOI: 10.1002/cne.23889] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Virginie Follin-Arbelet
- Department of Molecular Medicine, Institute of Basic Medical Sciences; University of Oslo; Oslo 0317 Norway
| | - Silvia Holmseth
- Department of Molecular Medicine, Institute of Basic Medical Sciences; University of Oslo; Oslo 0317 Norway
| | - Ellen Merckx
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Hideyo Sato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Technology; Niigata University; Niigata Niigata Prefecture 950-2181 Japan
| | - Joeri L. Aerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics; KU Leuven; Leuven 3000 Belgium
| | - Niels C. Danbolt
- Department of Molecular Medicine, Institute of Basic Medical Sciences; University of Oslo; Oslo 0317 Norway
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| |
Collapse
|