1
|
She R, Zhang Z, Han M, Zhao D, Li X, Zhou J, Chang Y, Zhang X, Li X. Luteolin Exhibits Anxiolytic and Antidepressant Potential in Parkinson's Disease Rat: Antioxidant and Anti-Inflammatory Effects. Rejuvenation Res 2025; 28:67-82. [PMID: 39729251 DOI: 10.1089/rej.2024.0045] [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] [Indexed: 12/28/2024] Open
Abstract
Parkinson's disease (PD) is accompanied by a complex array of nonmotor and motor manifestations. The exploration of anti-inflammatory and antioxidant active ingredient as potential therapeutic interventions in PD-associated mood alterations has gained significant attention. This study aimed to assess the antidepressant and anxiolytic properties of luteolin (LTN), a potent antioxidant and anti-inflammatory component, using a 6-hydroxydopamine (6-OHDA)-induced animal model of PD. Rats were administered LTN (10, 25, and 50 mg/kg, per oral) and fluoxetine (10 mg/kg/per oral) over a 28-day period. Behavioral tests were employed to estimate the depression- and anxiety-like behaviors. Rats treated with LTN exhibited significant improvement in 6-OHDA-induced mood alterations, as per behavioral tests. Additionally, LTN treatment led to increased hippocampal levels of catalase and superoxide dismutase, and a reduction in malondialdehyde. LTN downregulated the gene expression of nuclear factor kappa B (NF-κB)/nod-like receptor (NLR) pyrin domain-containing 3 (NLRP3) axis components, including NF-κB, NLRP3, ASC, and Caspase1 and reduced the protein level of pro-inflammatory cytokines, including interleukin (IL)-6, interleukin (IL)-1β, and tumor necrosis factor alpha (TNF-α), in addition to augmenting the protein levels of TNF-α, IL-1β, and IL-6. Furthermore, LTN exhibited an upregulatory effect on the anti-inflammatory cytokine IL-10 within the hippocampus of 6-OHDA-induced PD rats. Also, molecular docking showed higher affinity between LTN and NF-κB/NLRP3 axis components. These findings highlight the potential anxiolytic and antidepressant impacts of LTN through its antioxidant and anti-inflammatory mechanisms against 6-OHDA-induced alterations in a rat PD model.
Collapse
Affiliation(s)
- Ruifang She
- Department of Neurology, Jinan Central Hospital, Shandong University, Jinan City, People's Republic of China
- Department of Neurology, The Affiliated Taian City Central Hospital of Qingdao University, Taian City, People's Republic of China
| | - Zhaoting Zhang
- Department of Neurology, The Second People's Hospital of Lianyungang City, Lianyungang City, People's Republic of China
| | - Miaomiao Han
- Encephalopathy Department, Zibo Hospital of Integrated Traditional Chinese and Western Medicine, Zibo City, People's Republic of China
| | - Dapeng Zhao
- Department of Neurology, The Affiliated Taian City Central Hospital of Qingdao University, Taian City, People's Republic of China
| | - Xiangting Li
- Department of Pulmonary and Critical Care Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian City, People's Republic of China
| | - Jian Zhou
- Department of Radiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian City, People's Republic of China
| | - Yanyan Chang
- Department of Neurology, The Affiliated Taian City Central Hospital of Qingdao University, Taian City, People's Republic of China
| | - Xinping Zhang
- Department of Neurology, The Affiliated Taian City Central Hospital of Qingdao University, Taian City, People's Republic of China
| | - Xiaohong Li
- Department of Neurology, Jinan Central Hospital, Shandong University, Jinan City, People's Republic of China
| |
Collapse
|
2
|
Jung H, Kim S. E46K α-Synuclein Mutation Fails to Promote Neurite Outgrowth by Not Inducing Cdc42EP2 Expression, Unlike Wild-Type or A53T α-Synuclein in SK-N-SH Cells. Brain Sci 2024; 15:9. [PMID: 39851377 PMCID: PMC11763803 DOI: 10.3390/brainsci15010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES α-Synuclein (α-syn) protein is a major pathological agent of familial Parkinson's disease (PD), and its levels and aggregations determine neurotoxicity in PD pathogenesis. Although the pathophysiological functions of α-syn have been extensively studied, its biological functions remain elusive, and there are reports of wild-type (WT) α-syn and two missense mutations of α-syn (A30P and A53T) inducing protective neuritogenesis through neurite outgrowth. However, the function of another α-syn mutation, E46K, has not been fully elucidated. Thus, we compared the effect of E46K α-syn with other types to identify the mechanisms underlying neurite outgrowth. METHODS We transfected SK-N-SH cells with WT and mutant (A53T and E46K) α-syn to investigate the effects of their overexpression on neurite outgrowth. Then, we compared the differential effects of α-syn on neurite outgrowth using microscopic analysis, including confocal microscopy. We also analyzed the differential regulation of cell division control 42 effector protein 2 (Cdc42EP2) using real-time quantitative polymerase chain reaction and western blot analysis. Finally, to confirm the implication of neurite outgrowth, we knocked down Cdc42EP2 using small interfering RNA. RESULTS Unlike WT and A53T α-syn, E46K α-syn failed to promote neurite outgrowth by not inducing Cdc42EP2 and subsequent βIII-tubulin expression. Cdc42EP2 knockdown impaired neurite outgrowth in WT and A53T α-syn transfectants. CONCLUSIONS Our findings suggest that WT and mutant α-syn are linked to Cdc42EP2 production in neuritogenesis, implying α-syn involvement in the physiological function of axon growth and synapse formation. Thus, α-syn may be a potential therapeutic target for PD.
Collapse
Affiliation(s)
| | - Seonghan Kim
- Department of Anatomy, College of Medicine, Inje University, Busan 47392, Republic of Korea
| |
Collapse
|
3
|
Sun N, Cui WQ, Min XM, Zhang GM, Liu JZ, Wu HY. A new perspective on hippocampal synaptic plasticity and post-stroke depression. Eur J Neurosci 2023; 58:2961-2984. [PMID: 37518943 DOI: 10.1111/ejn.16093] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Post-stroke depression, a common complication after stroke, severely affects the recovery and quality of life of patients with stroke. Owing to its complex mechanisms, post-stroke depression treatment remains highly challenging. Hippocampal synaptic plasticity is one of the key factors leading to post-stroke depression; however, the precise molecular mechanisms remain unclear. Numerous studies have found that neurotrophic factors, protein kinases and neurotransmitters influence depressive behaviour by modulating hippocampal synaptic plasticity. This review further elaborates on the role of hippocampal synaptic plasticity in post-stroke depression by summarizing recent research and analysing possible molecular mechanisms. Evidence for the correlation between hippocampal mechanisms and post-stroke depression helps to better understand the pathological process of post-stroke depression and improve its treatment.
Collapse
Affiliation(s)
- Ning Sun
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Man Min
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guang-Ming Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia-Zheng Liu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Masanetz RK, Baum W, Schett G, Winkler J, Süß P. Cellular plasticity and myeloid inflammation in the adult brain are independent of the transcriptional modulator DREAM. Neurosci Lett 2023; 796:137061. [PMID: 36626960 DOI: 10.1016/j.neulet.2023.137061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
The downstream regulatory element antagonist modulator (DREAM) modulates ion channel function and gene transcription. Functionally, DREAM is implicated in physiological and pathological processes including cell proliferation, inflammation, and nociception. Despite its multiple functions and robust expression in forebrain tissue, neurons and glial cells, the role of DREAM in regard to cellular plasticity and tumor necrosis factor (TNF)-mediated inflammation is largely unexplored. Here, we demonstrate that adult hippocampal neurogenesis as well as the density and plasticity of glial cells in the hippocampus and thalamus are independent of the presence of DREAM. Further, DREAM deletion does not alter the regional myeloid response and inflammatory gene expression induced by chronic peripheral inflammation in mice overexpressing human TNF. Our data suggest that despite their highly dynamic regulation, neural cell plasticity and adult neurogenesis in the hippocampus do not depend on the multifunctional protein DREAM. Furthermore, TNF-mediated myeloid inflammation in the brain persists in the absence of DREAM.
Collapse
Affiliation(s)
- Rebecca Katharina Masanetz
- Department of Molecular Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Wolfgang Baum
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Georg Schett
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Patrick Süß
- Department of Molecular Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Neurology, Friedrich-Alexander-Universität Erlangen Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany.
| |
Collapse
|
5
|
Amanollahi M, Jameie M, Rezaei N. Neuroinflammation as a potential therapeutic target in neuroimmunological diseases. TRANSLATIONAL NEUROIMMUNOLOGY, VOLUME 7 2023:475-504. [DOI: 10.1016/b978-0-323-85841-0.00021-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
6
|
Jellinger KA. The pathobiological basis of depression in Parkinson disease: challenges and outlooks. J Neural Transm (Vienna) 2022; 129:1397-1418. [PMID: 36322206 PMCID: PMC9628588 DOI: 10.1007/s00702-022-02559-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Depression, with an estimated prevalence of about 40% is a most common neuropsychiatric disorder in Parkinson disease (PD), with a negative impact on quality of life, cognitive impairment and functional disability, yet the underlying neurobiology is poorly understood. Depression in PD (DPD), one of its most common non-motor symptoms, can precede the onset of motor symptoms but can occur at any stage of the disease. Although its diagnosis is based on standard criteria, due to overlap with other symptoms related to PD or to side effects of treatment, depression is frequently underdiagnosed and undertreated. DPD has been related to a variety of pathogenic mechanisms associated with the underlying neurodegenerative process, in particular dysfunction of neurotransmitter systems (dopaminergic, serotonergic and noradrenergic), as well as to disturbances of cortico-limbic, striato-thalamic-prefrontal, mediotemporal-limbic networks, with disruption in the topological organization of functional mood-related, motor and other essential brain network connections due to alterations in the blood-oxygen-level-dependent (BOLD) fluctuations in multiple brain areas. Other hypothetic mechanisms involve neuroinflammation, neuroimmune dysregulation, stress hormones, neurotrophic, toxic or metabolic factors. The pathophysiology and pathogenesis of DPD are multifactorial and complex, and its interactions with genetic factors, age-related changes, cognitive disposition and other co-morbidities awaits further elucidation.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
7
|
Nano-MgO composites containing plasmid DNA to silence SNCA gene displays neuroprotective effects in Parkinson's rats induced by 6-hydroxydopamine. Eur J Pharmacol 2022; 922:174904. [DOI: 10.1016/j.ejphar.2022.174904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 11/23/2022]
|
8
|
Zhang X, Deng R, Zhang S, Deng J, Jia JJ, Sun B, Zhou X, Bai J. Thioredoxin-1 regulates calcium homeostasis in MPP + /MPTP-induced Parkinson's disease models. Eur J Neurosci 2021; 54:4827-4837. [PMID: 34132424 DOI: 10.1111/ejn.15355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/04/2021] [Accepted: 06/12/2021] [Indexed: 11/30/2022]
Abstract
Disturbance in calcium (Ca2+ ) homeostasis has been involved in a variety of neuropathological conditions including Parkinson's disease (PD). The Ca2+ channel, transient receptor potential channel 1 (TRPC1), plays a protective role in regulating entry of Ca2+ activated by store depletion of Ca2+ in endoplasmic reticulum (ER). We have showed that thioredoxin-1 (Trx-1) plays a role in suppressing ER stress in PD. However, whether Trx-1 regulates TRPC1 expression in PD is still unknown. In the present study, we demonstrated that treatment of 1-methyl-4-phenylpyridinum ion (MPP+ ) significantly reduced the expression of TRPC1 in PC12 cells, which was restored by Trx-1 overexpression, and further decreased significantly by Trx-1 siRNA. Moreover, we found that Ca2+ entered into the cells was decreased by MPP+ in PC 12 cells, which was restored by Trx-1 overexpression, and further decreased by Trx-1 siRNA. MPP+ significantly increased calcium-dependent cysteine protease calpain1 expression in PC12 cells, which was suppressed by Trx-1 overexpression. Calpain1 expression was increased by Trx-1 siRNA or SKF96365, an inhibitor of TRPC1. Moreover, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) decreased TRPC1 expression in the substantia nigra pars compacta region (SNpc), which was restored in mice overexpressing Trx-1, and further decreased in mice of knockdown Trx-1. Inversely, the expression of calpain1 was increased by MPTP, which was suppressed in mice overexpressing Trx-1, and further increased in mice of knockdown Trx-1. In conclusion, Trx-1 regulates the Ca2+ entry through regulating TRPC1 expression after treatment of MPP+ /MPTP.
Collapse
Affiliation(s)
- Xianwen Zhang
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Ruhua Deng
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Se Zhang
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Juan Deng
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Jing Jing Jia
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Bo Sun
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Xiaoshuang Zhou
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
9
|
Langley MR, Ghaisas S, Palanisamy BN, Ay M, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Characterization of nonmotor behavioral impairments and their neurochemical mechanisms in the MitoPark mouse model of progressive neurodegeneration in Parkinson's disease. Exp Neurol 2021; 341:113716. [PMID: 33839143 PMCID: PMC9797183 DOI: 10.1016/j.expneurol.2021.113716] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/30/2022]
Abstract
Mitochondrial dysfunction has been implicated as a key player in the pathogenesis of Parkinson's disease (PD). The MitoPark mouse, a transgenic mitochondrial impairment model developed by specific inactivation of TFAM in dopaminergic neurons, spontaneously exhibits progressive motor deficits and neurodegeneration, recapitulating several features of PD. Since nonmotor symptoms are now recognized as important features of the prodromal stage of PD, we comprehensively assessed the clinically relevant motor and nonmotor deficiencies from ages 8-24 wk in both male and female MitoPark mice and their littermate controls. As expected, motor deficits in MitoPark mice began around 12-14 wk and became severe by 16-24 wk. Interestingly, MitoPark mice exhibited olfactory deficits in the novel and social scent tests as early as 10-12 wk as compared to age-matched littermate controls. Additionally, male MitoPark mice showed spatial memory deficits before female mice, beginning at 8 wk and becoming most severe at 16 wk, as determined by the Morris water maze. MitoPark mice between 16 and 24 wk spent more time immobile in forced swim and tail suspension tests, and made fewer entries into open arms of the elevated plus maze, indicating a depressive and anxiety-like phenotype, respectively. Importantly, depressive behavior as determined by immobility in forced swim test was reversible by antidepressant treatment with desipramine. Neurochemical and mechanistic studies revealed significant changes in CREB phosphorylation, BDNF, and catecholamine levels as well as neurogenesis in key brain regions. Collectively, our results indicate that MitoPark mice progressively exhibit deficits in olfactory discrimination, cognitive learning and memory, and anxiety- and depression-like behaviors as well as key neurochemical signaling associated with nonmotor deficits in PD. Thus, MitoPark mice can serve as an invaluable model for studying nonmotor deficits in addition to studying the motor deficits related to pathology in PD.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America.
| |
Collapse
|
10
|
Yoo DY, Jung HY, Kim W, Hahn KR, Kwon HJ, Nam SM, Chung JY, Yoon YS, Kim DW, Hwang IK. Entacapone promotes hippocampal neurogenesis in mice. Neural Regen Res 2021; 16:1005-1110. [PMID: 33269743 PMCID: PMC8224137 DOI: 10.4103/1673-5374.300447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Entacapone, a catechol-O-methyltransferase inhibitor, can strengthen the therapeutic effects of levodopa on the treatment of Parkinson’s disease. However, few studies are reported on whether entacapone can affect hippocampal neurogenesis in mice. To investigate the effects of entacapone, a modulator of dopamine, on proliferating cells and immature neurons in the mouse hippocampal dentate gyrus, 60 mice (7 weeks old) were randomly divided into a vehicle-treated group and the groups treated with 10, 50, or 200 mg/kg entacapone. The results showed that 50 and 200 mg/kg entacapone increased the exploration time for novel object recognition. Immunohistochemical staining results revealed that after entacapone treatment, the numbers of Ki67-positive proliferating cells, doublecortin-positive immature neurons, and phosphorylated cAMP response element-binding protein (pCREB)-positive cells were significantly increased. Western blot analysis results revealed that treatment with tyrosine kinase receptor B (TrkB) receptor antagonist significantly decreased the exploration time for novel object recognition and inhibited the expression of phosphorylated TrkB and brain-derived neurotrophic factor (BDNF). Entacapone treatment antagonized the effects of TrkB receptor antagonist. These results suggest that entacapone treatment promoted hippocampal neurogenesis and improved memory function through activating the BDNF-TrkB-pCREB pathway. This study was approved by the Institutional Animal Care and Use Committee of Seoul National University (approval No. SNU-130730-1) on February 24, 2014.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea; Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul; Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, South Korea
| | - Sung Min Nam
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
11
|
Cahill CM, Aleyadeh R, Gao J, Wang C, Rogers JT. Alpha-Synuclein in Alcohol Use Disorder, Connections with Parkinson's Disease and Potential Therapeutic Role of 5' Untranslated Region-Directed Small Molecules. Biomolecules 2020; 10:E1465. [PMID: 33096655 PMCID: PMC7589448 DOI: 10.3390/biom10101465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/28/2020] [Accepted: 10/09/2020] [Indexed: 12/23/2022] Open
Abstract
Alpha-synuclein (α-Syn) is a 140-amino acid (aa) protein encoded by the Synuclein alpha SNCA gene. It is the synaptic protein associated with Parkinson's disease (PD) and is the most highly expressed protein in the Lewy bodies associated with PD and other alpha synucleopathies, including Lewy body dementia (LBD) and multiple system atrophy (MSA). Iron deposits are present in the core of Lewy bodies, and there are reports suggesting that divalent metal ions including Cu2+ and Fe2+ enhance the aggregation of α-Syn. Differential expression of α-Syn is associated with alcohol use disorder (AUD), and specific genetic variants contribute to the risk for alcoholism, including alcohol craving. Spliced variants of α-Syn, leading to the expression of several shorter forms which are more prone to aggregation, are associated with both PD and AUD, and common transcript variants may be able to predict at-risk populations for some movement disorders or subtypes of PD, including secondary Parkinsonism. Both PD and AUD are associated with liver and brain iron dyshomeostasis. Research over the past decade has shown that α-Syn has iron import functions with an ability to oxidize the Fe3+ form of iron to Fe2+ to facilitate its entry into cells. Our prior research has identified an iron-responsive element (IRE) in the 5' untranslated region (5'UTR) of α-Syn mRNA, and we have used the α-Syn 5'UTR to screen for small molecules that modulate its expression in the H4 neuronal cell line. These screens have led us to identify several interesting small molecules capable of both decreasing and increasing α-Syn expression and that may have the potential, together with the recently described mesenchymal stem cell therapies, to normalize α-Syn expression in different regions of the alcoholic and PD brain.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | | | - Jin Gao
- Department of Clinical Psychology, Qilu Hospital of Shandong University, Qingdao 266011, China;
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| |
Collapse
|
12
|
Gruden’ MA, Solov’eva OA, Kudrin VS, Narkevich VB, Sherstnev VV. Neurochemical and Behavioral Features of Action of Pre-Fibrillar Oligomeric Structures of α-Sinuclein in Adult Mice. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Süß P, Hoffmann A, Rothe T, Ouyang Z, Baum W, Staszewski O, Schett G, Prinz M, Krönke G, Glass CK, Winkler J, Schlachetzki JCM. Chronic Peripheral Inflammation Causes a Region-Specific Myeloid Response in the Central Nervous System. Cell Rep 2020; 30:4082-4095.e6. [PMID: 32209470 DOI: 10.1016/j.celrep.2020.02.109] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/15/2020] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Systemic immune dysregulation contributes to the development of neuropsychiatric and neurodegenerative diseases. The precise effect of chronic peripheral immune stimulation on myeloid cells across anatomical brain regions is unclear. Here, we demonstrate brain-region-specific differences in myeloid responses induced by chronic peripheral inflammation. This shift in the myeloid compartment is associated with the appearance of an inflammatory myeloid subpopulation in the cortex, striatum, and thalamus accompanied by regional transcriptomic fingerprints that include induction of chemokines, complement factors, and endothelial adhesion molecules. In contrast, myeloid immune responses within the hippocampus and cerebellum are subtle or absent. Treatment with the anti-tumor necrosis factor α (anti-TNF-α) antibody infliximab ablates the region-specific inflammatory response. A region-specific myeloid cell response to chronic peripheral inflammation is observed in postmortem brains from individuals with rheumatoid arthritis. Our data suggest that chronic peripheral inflammation has heterogeneous effects on the brain, as evidenced by the spectrum of myeloid cell responses observed across brain regions.
Collapse
Affiliation(s)
- Patrick Süß
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Alana Hoffmann
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Tobias Rothe
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie, 91054 Erlangen, Germany
| | - Zhengyu Ouyang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wolfgang Baum
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ori Staszewski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie, 91054 Erlangen, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; Signalling Research Centres for BIOSS and CIBSS, University of Freiburg, 79104 Freiburg im Breisgau, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie, 91054 Erlangen, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Johannes C M Schlachetzki
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
14
|
Srivastava AK, Roy Choudhury S, Karmakar S. Melatonin/polydopamine nanostructures for collective neuroprotection-based Parkinson's disease therapy. Biomater Sci 2020; 8:1345-1363. [PMID: 31912833 DOI: 10.1039/c9bm01602c] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and localized deposition of cytoplasmic fibrillary inclusions as Lewy bodies in the brain. The aberrant phosphorylation of α-synuclein at serine 129 is the key process on its early onset, which alters the cellular conformation to oligomers and insoluble aggregates, underpinning cellular oxidative stress and mitochondrial dysfunction, leading to devastating PD synucleinopathy. The multiple neuroprotective roles of dopamine and melatonin are often demonstrated separately; however, this approach suffers from low and short bioavailability and is associated with side-effects upon overdosing. Herein, highly pleiotropic melatonin-enriched polydopamine nanostructures were fabricated, which showed efficient brain tissue retention, sustainable and prolonged melatonin release, and prevented neuroblastoma cell death elicited by Parkinson's disease-associated and mitochondrial damaging stimuli. The synergistic neuroprotection re-established the mitochondrial membrane potential, reduced the generation of cellular reactive oxygen species (ROS), inhibited the activation of both the caspase-dependent and independent apoptotic pathways, and exhibited an anti-inflammatory effect. At the molecular level, it suppressed α-synuclein phosphorylation at Ser 129 and reduced the cellular deposition of high molecular weight oligomers. The therapeutic assessment on ex vivo organotypic brain slice culture, and in vivo experimental PD model confirmed the superior brain targeting, collective neuroprotection on dopaminergic neurons with reduced alpha-synuclein phosphorylation and deposition in the hippocampal and substantia nigra region of the brain. Thus, nature-inspired melatonin-enriched polydopamine nanostructures conferring collective neuroprotective effects attributes activation of anti-oxidative, anti-inflammatory, and anti-apoptotic pathways may be superior for application in a nanomedicine-based PD therapy.
Collapse
Affiliation(s)
- Anup K Srivastava
- Institute of Nano Science and Technology, Habitat Centre, Sector-64, Mohali, Punjab-160062, India.
| | | | | |
Collapse
|
15
|
Canerina-Amaro A, Pereda D, Diaz M, Rodriguez-Barreto D, Casañas-Sánchez V, Heffer M, Garcia-Esparcia P, Ferrer I, Puertas-Avendaño R, Marin R. Differential Aggregation and Phosphorylation of Alpha Synuclein in Membrane Compartments Associated With Parkinson Disease. Front Neurosci 2019; 13:382. [PMID: 31068782 PMCID: PMC6491821 DOI: 10.3389/fnins.2019.00382] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/02/2019] [Indexed: 12/15/2022] Open
Abstract
The aggregation of α-synuclein (α-syn) is a major factor behind the onset of Parkinson’s disease (PD). Sublocalization of this protein may be relevant for the formation of multimeric α-syn oligomeric configurations, insoluble aggregates that form Lewy bodies in PD brains. Processing of this protein aggregation is regulated by associations with distinct lipid classes. For instance, instability of lipid raft (LR) microdomains, membrane regions with a particular lipid composition, is an early event in the development of PD. However, the relevance of membrane microdomains in the regulation and trafficking of the distinct α-syn configurations associated with PD remains unexplored. In this study, using 6- and 14-month-old healthy and MPTP-treated animals as a model of PD, we have investigated the putative molecular alterations of raft membrane microstructures, and their impact on α-syn dynamics and conformation. A comparison of lipid analyses of LR microstructures and non-raft (NR) fractions showed alterations in gangliosides, cholesterol, polyunsaturated fatty acids (PUFA) and phospholipids in the midbrain and cortex of aged and MPTP-treated mice. In particular, the increase of PUFA and phosphatidylserine (PS) during aging correlated with α-syn multimeric formation in NR. In these aggregates, α-syn was phosphorylated in pSer129, the most abundant post-transductional modification of α-syn promoting toxic aggregation. Interestingly, similar variations in PUFA and PS content correlating with α-syn insoluble accumulation were also detected in membrane microstructures from the human cortex of incidental Parkinson Disease (iPD) and PD, as compared to healthy controls. Furthermore, structural changes in membrane lipid microenvironments may induce rearrangements in raft-interacting proteins involved in other neuropathologies. Therefore, we also investigated the dynamic of other protein markers involved in cognition and memory impairment such as metabotropic glutamate receptor 5 (mGluR5), ionotropic NMDA receptor (NMDAR2B), prion protein (PrPc) and amyloid precursor protein (APP), whose activity depends on membrane lipid organization. We observed a decline of these protein markers in LR fractions with the progression of aging and pathology. Overall, our findings demonstrate that lipid alterations in membranous compartments promoted by brain aging and PD-like injury may have an effect on α-syn aggregation and segregation in abnormal multimeric structures.
Collapse
Affiliation(s)
- Ana Canerina-Amaro
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain.,Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Daniel Pereda
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain.,Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Mario Diaz
- Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, Santa Cruz de Tenerife, Spain.,Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, Faculty of Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Deiene Rodriguez-Barreto
- Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, Faculty of Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Verónica Casañas-Sánchez
- Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, Faculty of Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Marija Heffer
- Department of Biology, University of Osijek School of Medicine, Osijek, Croatia
| | - Paula Garcia-Esparcia
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, Barcelona, Spain.,CIBERNED, Barcelona, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, Barcelona, Spain.,CIBERNED, Barcelona, Spain
| | - Ricardo Puertas-Avendaño
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, Santa Cruz de Tenerife, Spain.,Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
16
|
Ma K, Han C, Zhang G, Guo X, Xia Y, Wan F, Yin S, Kou L, Liu L, Huang J, Xiong N, Wang T. Reduced VMAT2 expression exacerbates the hyposmia in the MPTP model of Parkinson's disease. Biochem Biophys Res Commun 2019; 513:306-312. [PMID: 30954223 DOI: 10.1016/j.bbrc.2019.03.159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/23/2019] [Indexed: 11/17/2022]
Abstract
Hyposmia occurs during the prodromal phase of Parkinson's disease (PD), while the underlying mechanisms remain unclear. Discussed are altered dopamine content and impairment of neurogenesis of olfactory bulbs (OB), which has been suggested to be linked to olfactory dysfunction. Given that mouse with reduced vesicular monoamine transporter 2 (VMAT2) expression is now deemed as a relatively new PD animal model simulating motor and nonmotor symptoms, it may provide a new insight into investigating the mechanisms of hyposmia in the context of PD. In this study, we examined the effect of subacute administration of MPTP on mice with a reduced expression of VMAT2, focusing on the histopathological and biochemical alterations, specifically, TH expression level, dopamine content as well as neurogenesis in OB. Interestingly, mice with a reduced VMAT2 expression displayed more obvious olfactory impairment in response to MPTP administration accompanied by markedly decreased dopaminergic interneurons in OB. Furthermore, neurogenesis in OB was also further impaired after MPTP due to reduced VMAT2 expression. We therefore demonstrated that reduced expression of VMAT2 contributed to the impairment of hyposmia, pathologically, the degeneration of extranigral systems and reduced neurogenesis might be the underlying mechanisms.
Collapse
Affiliation(s)
- Kai Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Han
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| | - Guoxin Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingfang Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
17
|
Hain EG, Sparenberg M, Rasińska J, Klein C, Akyüz L, Steiner B. Indomethacin promotes survival of new neurons in the adult murine hippocampus accompanied by anti-inflammatory effects following MPTP-induced dopamine depletion. J Neuroinflammation 2018; 15:162. [PMID: 29803225 PMCID: PMC5970532 DOI: 10.1186/s12974-018-1179-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 04/25/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by dopaminergic cell loss and inflammation in the substantia nigra (SN) leading to motor deficits but also to hippocampus-associated non-motor symptoms such as spatial learning and memory deficits. The cognitive decline is correlated with impaired adult hippocampal neurogenesis resulting from dopamine deficit and inflammation, represented in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) mouse model of PD. In the inflammatory tissue, cyclooxygenase (COX) is upregulated leading to an ongoing inflammatory process such as prostaglandin-mediated increased cytokine levels. Therefore, inhibition of COX by indomethacin may prevent the inflammatory response and the impairment of adult hippocampal neurogenesis. METHODS Wildtype C57Bl/6 and transgenic Nestin-GFP mice were treated with MPTP followed by short-term or long-term indomethacin treatment. Then, aspects of inflammation and neurogenesis were evaluated by cell counts using immunofluorescence and immunohistochemical stainings in the SN and dentate gyrus (DG). Furthermore, hippocampal mRNA expression of neurogenesis-related genes of the Notch, Wnt, and sonic hedgehog signaling pathways and neurogenic factors were assessed, and protein levels of serum cytokines were measured. RESULTS Indomethacin restored the reduction of the survival rate of new mature neurons and reduced the amount of amoeboid CD68+ cells in the DG after MPTP treatment. Indomethacin downregulated genes of the Wnt and Notch signaling pathways and increased neuroD6 expression. In the SN, indomethacin reduced the pro-inflammatory cellular response without reversing dopaminergic cell loss. CONCLUSION Indomethacin has a pro-neurogenic and thereby restorative effect and an anti-inflammatory effect on the cellular level in the DG following MPTP treatment. Therefore, COX inhibitors such as indomethacin may represent a therapeutic option to restore adult neurogenesis in PD.
Collapse
Affiliation(s)
- Elisabeth G Hain
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany.
| | - Maria Sparenberg
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Justyna Rasińska
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Charlotte Klein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Levent Akyüz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Augustenburger Platz 1, 13353, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Barbara Steiner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
18
|
Leeboonngam T, Pramong R, Sae-Ung K, Govitrapong P, Phansuwan-Pujito P. Neuroprotective effects of melatonin on amphetamine-induced dopaminergic fiber degeneration in the hippocampus of postnatal rats. J Pineal Res 2018; 64. [PMID: 29149481 DOI: 10.1111/jpi.12456] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/10/2017] [Indexed: 12/21/2022]
Abstract
Chronic amphetamine (AMPH) abuse leads to damage of the hippocampus, the brain area associated with learning and memory process. Previous results have shown that AMPH-induced dopamine neurotransmitter release, reactive oxygen species formation, and degenerative protein aggregation lead to neuronal death. Melatonin, a powerful antioxidant, plays a role as a neuroprotective agent. The objective of this study was to investigate whether the protective effect of melatonin on AMPH-induced hippocampal damage in the postnatal rat acts through the dopaminergic pathway. Four-day-old postnatal rats were subcutaneously injected with 5-10 mg/kg AMPH and pretreated with 10 mg/kg melatonin prior to AMPH exposure for seven days. The results showed that melatonin decreased the AMPH-induced hippocampal neuronal degeneration in the dentate gyrus, CA1, and CA3. Melatonin attenuated the reduction in the expression of hippocampal synaptophysin, PSD-95, α-synuclein, and N-methyl-D-aspartate (NMDA) receptor protein and mRNA caused by AMPH. Melatonin attenuated the AMPH-induced reduction in dopamine transporter (DAT) protein expression in the hippocampus and the reduction in mRNA expression in the ventral tegmental area (VTA). Immunofluorescence demonstrated that melatonin not only prevented the AMPH-induced loss of DAT and NMDA receptor but also prevented AMPH-induced α-synuclein overexpression in the dentate gyrus, CA1, and CA3. Melatonin decreased the AMPH-induced reduction in the protein and mRNA of the NMDA receptor downstream signaling molecule, calcium/calmodulin-dependent protein kinase II (CaMKII), and the melatonin receptors (MT1 and MT2). This study showed that melatonin prevented AMPH-induced toxicity in the hippocampus of postnatal rats possibly via its antioxidative effect and mitochondrial protection.
Collapse
Affiliation(s)
- Tanawan Leeboonngam
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Ratchadaporn Pramong
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Kwankanit Sae-Ung
- Innovative Learning Center, Srinakharinwirot University, Bangkok, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakorn Pathom, Thailand
| | | |
Collapse
|
19
|
Chen JF, Wang M, Zhuang YH, Behnisch T. Intracerebroventricularly-administered 1-methyl-4-phenylpyridinium ion and brain-derived neurotrophic factor affect catecholaminergic nerve terminals and neurogenesis in the hippocampus, striatum and substantia nigra. Neural Regen Res 2018; 13:717-726. [PMID: 29722326 PMCID: PMC5950684 DOI: 10.4103/1673-5374.230300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Parkinson's disease is a progressive neurological disease characterized by the degeneration of dopaminergic neurons in the substantia nigra. A highly similar pattern of neurodegeneration can be induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium ion (MPP+), which cause the death of dopaminergic neurons. Administration of MPTP or MPP+ results in Parkinson's disease-like symptoms in rodents. However, it remains unclear whether intracerebroventricular MPP+ administration affects neurogenesis in the substantia nigra and subgranular zone or whether brain-derived neurotrophic factor alters the effects of MPP+. In this study, MPP+ (100 nmol) was intracerebroventricularly injected into mice to model Parkinson's disease. At 7 days after administration, the number of bromodeoxyuridine (BrdU)-positive cells in the subgranular zone of the hippocampal dentate gyrus increased, indicating enhanced neurogenesis. In contrast, a reduction in BrdU-positive cells was detected in the substantia nigra. Administration of brain-derived neurotrophic factor (100 ng) 1 day after MPP+ administration attenuated the effect of MPP+ in the subgranular zone and the substantia nigra. These findings reveal the complex interaction between neurotrophic factors and neurotoxins in the Parkinsonian model that result in distinct effects on the catecholaminergic system and on neurogenesis in different brain regions.
Collapse
Affiliation(s)
- Jun-Fang Chen
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Man Wang
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ying-Han Zhuang
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Synaptic Regulator α-Synuclein in Dopaminergic Fibers Is Essentially Required for the Maintenance of Subependymal Neural Stem Cells. J Neurosci 2017; 38:814-825. [PMID: 29217686 DOI: 10.1523/jneurosci.2276-17.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 11/28/2017] [Indexed: 01/11/2023] Open
Abstract
Synaptic protein α-synuclein (α-SYN) modulates neurotransmission in a complex and poorly understood manner and aggregates in the cytoplasm of degenerating neurons in Parkinson's disease. Here, we report that α-SYN present in dopaminergic nigral afferents is essential for the normal cycling and maintenance of neural stem cells (NSCs) in the brain subependymal zone of adult male and female mice. We also show that premature senescence of adult NSCs into non-neurogenic astrocytes in mice lacking α-SYN resembles the effects of dopaminergic fiber degeneration resulting from chronic exposure to 1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine or intranigral inoculation of aggregated toxic α-SYN. Interestingly, NSC loss in α-SYN-deficient mice can be prevented by viral delivery of human α-SYN into their sustantia nigra or by treatment with l-DOPA, suggesting that α-SYN regulates dopamine availability to NSCs. Our data indicate that α-SYN, present in dopaminergic nerve terminals supplying the subependymal zone, acts as a niche component to sustain the neurogenic potential of adult NSCs and identify α-SYN and DA as potential targets to ameliorate neurogenic defects in the aging and diseased brain.SIGNIFICANCE STATEMENT We report an essential role for the protein α-synuclein present in dopaminergic nigral afferents in the regulation of adult neural stem cell maintenance, identifying the first synaptic regulator with an implication in stem cell niche biology. Although the exact role of α-synuclein in neural transmission is not completely clear, our results indicate that it is required for stemness and the preservation of neurogenic potential in concert with dopamine.
Collapse
|
21
|
Smita SS, Raj Sammi S, Laxman TS, Bhatta RS, Pandey R. Shatavarin IV elicits lifespan extension and alleviates Parkinsonism in Caenorhabditis elegans. Free Radic Res 2017; 51:954-969. [PMID: 29069955 DOI: 10.1080/10715762.2017.1395419] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Shatavarin IV (SIV), a steroidal saponin, is a major bioactive phytomolecule present in roots of Asparagus racemosus (Liliaceae) known for its anticancer activity. Age-associated neurodegenerative Parkinson's disease (PD) is characterised by alpha-synuclein aggregation in dopaminergic neuron resulting in neurodegeneration. The invention of bioactive molecules that delay aging and age-associated disorders endorses development of natural phytomolecule as a therapeutic agent for curing age-related diseases. Therefore, the present study for the first time explores the potential of SIV against aging and Parkinsonism utilising Caenorhabditis elegans model system. SIV significantly attenuated oxidative stress in terms of intracellular reactive oxygen species (ROS) as well as oxidative damage including protein carbonylation and also promotes longevity. SIV also significantly increased the mRNA expression of stress responsive genes namely sod-1, sod-2, sod-3, gst-4, gst-7 and ctl-2 suggesting its anti-oxidant property that might be contributed in the modulation of oxidative stress and promoting lifespan. Additionally, SIV improved PD symptoms by reducing the alpha-synuclein aggregation, lipid accumulation and enhancing dopamine level. Altogether, present findings indicate that SIV possibly utilising ubiquitin-mediated proteasomal system and attenuating oxidative stress by up-regulating PD-associated genes pdr-1, ubc-12 and pink-1. Therefore, this study is a forward step in exploring the anti-aging and anti-Parkinsonism potential of bioactive compound SIV in C. elegans.
Collapse
Affiliation(s)
- Shachi Shuchi Smita
- a Laboratory of Aging Biology, Department of Microbial Technology and Nematology , Central Institute of Medicinal and Aromatic Plants, Council of Scientific and Industrial Research , Lucknow , India
| | - Shreesh Raj Sammi
- a Laboratory of Aging Biology, Department of Microbial Technology and Nematology , Central Institute of Medicinal and Aromatic Plants, Council of Scientific and Industrial Research , Lucknow , India
| | - Tulsankar S Laxman
- b Pharmacokinetics and Metabolism Division , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Rabi S Bhatta
- b Pharmacokinetics and Metabolism Division , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Rakesh Pandey
- a Laboratory of Aging Biology, Department of Microbial Technology and Nematology , Central Institute of Medicinal and Aromatic Plants, Council of Scientific and Industrial Research , Lucknow , India
| |
Collapse
|
22
|
Jung HY, Kim DW, Nam SM, Kim JW, Chung JY, Won MH, Seong JK, Yoon YS, Yoo DY, Hwang IK. Pyridoxine improves hippocampal cognitive function via increases of serotonin turnover and tyrosine hydroxylase, and its association with CB1 cannabinoid receptor-interacting protein and the CB1 cannabinoid receptor pathway. Biochim Biophys Acta Gen Subj 2017; 1861:3142-3153. [PMID: 28935605 DOI: 10.1016/j.bbagen.2017.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 08/28/2017] [Accepted: 09/15/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND In the present study, we investigated the effects of pyridoxine on hippocampal functions and changes in protein profiles based on the proteomic approach. METHODS Eight-week-old mice received intraperitoneal injections of physiological saline (vehicle) or 350mg/kg pyridoxine twice a day for 21days. RESULTS Phosphoglycerate mutase 1 was up-regulated, while CB1 cannabinoid receptor-interacting protein 1 (CRIP1) was down-regulated, in the pyridoxine-treated group. Additionally, the serotonin and tyrosine hydroxylase was increased in the hippocampus of the pyridoxine-treated group than in that of the vehicle-treated group. Furthermore, discrimination indices based on the novel object recognition test were significantly higher in the pyridoxine-treated group than in the vehicle-treated group. Administration of CRIP1a siRNA significantly increases the discrimination index as well as cell proliferation and neuroblast differentiation in the dentate gyrus. In addition, the administration of rimonabant, a CB1 cannabinoid receptor antagonist, for 3weeks significantly decreased the novel object recognition memory, the tyrosine hydroxylase level, the amount of cell proliferation, and neuroblast differentiation in the dentate gyrus. Treatment with pyridoxine significantly increased novel object recognition memory, but slightly ameliorated rimonabant-induced reduction in serotonin, the tyrosine hydroxylase level, the amount of cell proliferation, and neuroblast differentiation in the dentate gyrus. CONCLUSION These results suggest that pyridoxine promotes hippocampal functions by increasing serotonin and tyrosine hydroylase immunoreactivity in the hippocampus. This positive effect may be associated with CRIP1a and CB1 cannabinoid receptor function. GENERAL SIGNIFICANCE Vitamin-B6 enhances hippocampal functions and this is closely associated with CRIP1a and CB1 cannabinoid receptors.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, South Korea
| | - Sung Min Nam
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, South Korea
| | - Je Kyung Seong
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea; KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul 08826, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea; KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul 08826, South Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea.
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea; KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
23
|
Brandt MD, Krüger-Gerlach D, Hermann A, Meyer AK, Kim KS, Storch A. Early Postnatal but Not Late Adult Neurogenesis Is Impaired in the Pitx3-Mutant Animal Model of Parkinson's Disease. Front Neurosci 2017; 11:471. [PMID: 28883785 PMCID: PMC5573808 DOI: 10.3389/fnins.2017.00471] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 08/09/2017] [Indexed: 01/10/2023] Open
Abstract
The generation of new neurons in the adult dentate gyrus has functional implications for hippocampal formation. Reduced hippocampal neurogenesis has been described in various animal models of hippocampal dysfunction such as dementia and depression, which are both common non-motor-symptoms of Parkinson's disease (PD). As dopamine plays an important role in regulating precursor cell proliferation, the loss of dopaminergic neurons in the substantia nigra (SN) in PD may be related to the reduced neurogenesis observed in the neurogenic regions of the adult brain: subventricular zone (SVZ) and dentate gyrus (DG). Here we examined adult hippocampal neurogenesis in the Pitx3-mutant mouse model of PD (aphakia mice), which phenotypically shows a selective embryonic degeneration of dopamine neurons within the SN and to a smaller extent in the ventral tegmental area (VTA). Proliferating cells were labeled with BrdU in aphakia mice and healthy controls from 3 to 42 weeks of age. Three weeks old mutant mice showed an 18% reduction of proliferating cells in the DG and of 26% in the SVZ. Not only proliferation but also the number of new neurons was impaired in young aphakia mice resulting in 33% less newborn cells 4 weeks after BrdU-labeling. Remarkably, however, the decline in the number of proliferating cells in the neurogenic regions vanished in older animals (8–42 weeks) indicating that aging masks the effect of dopamine depletion on adult neurogenesis. Region specific reduction in precursor cells proliferation correlated with the extent of dopaminergic degeneration in mesencephalic subregions (VTA and SN), which supports the theory of age- and region-dependent regulatory effects of dopaminergic projections. Physiological stimulation of adult neurogenesis by physical activity (wheel running) almost doubled the number of proliferating cells in the dentate gyrus of 8 weeks old aphakia mice to a number comparable to that of wild-type mice, abolishing the slight reduction of baseline neurogenesis at this age. The described age-dependent susceptibility of adult neurogenesis to PD-like dopaminergic degeneration and its responsiveness to physical activity might have implications for the understanding of the pathophysiology and treatment of non-motor symptoms in PD.
Collapse
Affiliation(s)
- Moritz D Brandt
- Department of Neurology, Technische Universität DresdenDresden, Germany.,German Center for Neurodegenerative Diseases DresdenDresden, Germany
| | | | - Andreas Hermann
- Department of Neurology, Technische Universität DresdenDresden, Germany.,German Center for Neurodegenerative Diseases DresdenDresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität DresdenDresden, Germany
| | - Anne K Meyer
- Department of Neurology, Technische Universität DresdenDresden, Germany
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, McLean Hospital/Harvard Medical SchoolBelmont, MA, United States
| | - Alexander Storch
- German Center for Neurodegenerative Diseases RostockRostock, Germany.,Department of Neurology, University of RostockRostock, Germany
| |
Collapse
|
24
|
The Cytomegalovirus protein pUL37×1 targets mitochondria to mediate neuroprotection. Sci Rep 2016; 6:31373. [PMID: 27562039 PMCID: PMC4999870 DOI: 10.1038/srep31373] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
There is substantial evidence that mitochondrial dysfunction plays a significant role in the pathogenesis of Parkinson disease (PD). This contribution probably encompasses defects of oxidative phosphorylation, mitochondrial turnover (mitophagy), mitochondrial derived oxidative stress, and apoptotic signalling. Human cytomegalovirus immediate-early protein pUL37 × 1 induces Bax mitochondrial translocation and inactivation to prevent apoptosis. Over-expressing pUL37 × 1 in neuronal cells protects against staurosporin and 6-hydroxydopamine induced apoptosis and cell death. Protection is not enhanced by bax silencing in pUL37 × 1 over-expressing cells, suggesting a bax-dependent mechanism of action. pUL37 × 1 increases glycolysis and induces mitochondrial hyperpolarization, a bax independent anti-apoptotic action. pUL37 × 1 increases glycolysis through activation of phosphofructokinase by a calcium-dependent pathway. The dual anti-apoptotic mechanism of pUL37 × 1 may be considered a novel neuroprotective strategy in diseases where mitochondrial dysfunction and apoptotic pathways are involved.
Collapse
|
25
|
|