1
|
Dai Z, Liu S, Liu C. Detection of Parkinson's disease using nocturnal breathing signals based on multifractal detrended fluctuation analysis. CHAOS (WOODBURY, N.Y.) 2024; 34:123151. [PMID: 39671704 DOI: 10.1063/5.0237878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/25/2024] [Indexed: 12/15/2024]
Abstract
Parkinson's disease (PD) is a highly prevalent neurodegenerative disorder that poses a significant challenge in terms of accurate and cost-effective diagnosis. This study focuses on the use of fractal features derived from nocturnal breathing signals to diagnose PD. Our study includes 49 individuals with Parkinson's disease (PD group), 49 relatively healthy individuals without PD (HC group), 49 individuals without PD but with other diseases (NoPD group), as well as 12 additional PD patients and 200 healthy individuals for testing. Using multifractal detrended fluctuation analysis, we extracted fractal features from nocturnal breathing signals, with logistic regression models applied to diagnose PD, as demonstrated in receiver operating characteristic curves. Eight fractal features show significant diagnostic potential for PD, including generalized Hurst exponents for the Airflow, Thorax, and Abdomen signals and the multifractal spectrum width of the SaO2 signal. Finally, the area under the receiver operating characteristic curve (AUC) of the training data set of the PD and HC groups for all four signals is 0.911, and the AUC of the testing data set is 0.929. These results demonstrate the potential of this work to enhance the accuracy of PD diagnosis in clinical settings.
Collapse
Affiliation(s)
- Zhong Dai
- School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Shutang Liu
- School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Changan Liu
- Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
2
|
Li Y, Shi C, Liu R, Yang J, Wang J. Alpha-synuclein affects certain iron transporters of BV2 microglia cell through its ferric reductase activity. J Neurophysiol 2024; 132:446-453. [PMID: 38919150 DOI: 10.1152/jn.00106.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Alpha-synuclein (α-syn) is a major component of Lewy bodies, which is a biomarker of Parkinson's disease (PD). It accumulates in substantia nigra pars compacta (SNpc) to form insoluble aggregates and cause neurotoxicity, which is often accompanied by iron deposition. We compared the iron reductase activity between monomeric α-syn (M-α-syn) and oligomeric α-syn (O-α-syn) and investigated the effect of α-syn on iron metabolism of BV2 microglia cells as well. α-syn had ferric reductase activity, and O-α-syn had stronger enzyme activity than M-α-syn. M-α-syn upregulated iron uptake protein, divalent metal transporter1 (DMT1) expression, and iron influx but did not regulate iron release protein ferroportin1 (FPN1) expression and iron efflux. O-α-syn elevated the expression of both DMT1 and FPN1 and thus increased the iron influx and efflux in BV2 microglial cells, but the expressions of iron regulatory protein1 (IRP1) and hypoxia-inducible factor 2α (HIF-2α) had no significant change. Moreover, both M-α-syn and O-α-syn could increase the mRNA expressions of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in BV2 microglia cells. Both types of α-syn can activate microglia, which leads to increased expressions of proinflammatory factors. α-syn can affect DMT1 and FPN1 expressions in BV2 microglia cells, which might be through its ferric reductase activity.NEW & NOTEWORTHY The effects of monomeric α-syn (M-α-syn) and oligomeric α-syn (O-α-syn) on the iron metabolism of BV2 microglia cells were detected by exogenous α-syn treatment. This study provides a strong experimental basis for α-syn involvement in iron metabolism in microglia.
Collapse
Affiliation(s)
- Yinghui Li
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Chengkui Shi
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Rong Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Jiahua Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Jun Wang
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Zhang X, Wang Y, Lv J. STAT4 targets KISS1 to inhibit the oxidative damage, inflammation and neuronal apoptosis in experimental PD models by inactivating the MAPK pathway. Neurochem Int 2024; 175:105683. [PMID: 38341034 DOI: 10.1016/j.neuint.2024.105683] [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: 08/23/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Oxidative stress and neuroinflammation are proven to play critical roles in the pathogenesis of Parkinson's disease (PD). As reported, patients with PD have lower level of STAT4 compared with healthy subjects. However, the biological functions and mechanisms of STAT4 in PD pathogenesis remain uncertain. This study aimed to investigate the roles and related mechanisms of STAT4 in PD development. METHODS The intraperitoneal injection of MPTP (20 mg/kg) dissolved in physiological saline was performed to mimic PD-like conditions in vivo. MPP + solution was prepared for cell model of PD. Cell viability was measured by CCK-8. Griess reaction was conducted to measure NO concentrations. The mRNA and protein levels were evaluated by RT-qPCR and western blotting. ROS generation was assessed by DCFH-DA. The levels of inflammatory cytokines were measured by ELISA. Cell apoptosis was examined by flow cytometry and western blotting. Moreover, the SH-SY5Y cells were treated with conditioned medium from LPS-stimulated microglia and subjected to CCK-8 assays and ELISA. Mechanistically, CHIP assays and luciferase reporter assays were performed to verify the binding relationship between KISS1 and STAT4. For in vivo analysis, the histological changes of midbrain tissues of mice were determined by hematoxylin and eosin staining. The expression of tyrosine hydroxylase (TH) was detected by immunohistochemistry staining. Iba-1 positive microglial cells in the striatum were assessed by immunofluorescence staining. RESULTS For in vitro analysis, STAT4 level was downregulated after MPP+ treatment, and STAT4 upregulation inhibited the oxidative damage, inflammation and apoptosis in SH-SY5Y cells. STAT4 bound at +215-228 region of KISS1, and KISS1 upregulation counteracted the protection of STAT4 upregulation against cell damage. Moreover, STAT4 upregulation inhibited cell viability loss and inflammation induced by conditioned medium from LPS-treated microglia, whereas KISS1 upregulation had the opposite effect. For in vivo analysis, the protective effects of STAT4 upregulation against inflammatory response, oxidative stress, dopaminergic neuronal loss and microglia activation were attenuated by KISS1 upregulation. Moreover, the inactivation of MAPK pathway caused by STAT4 upregulation was reversed by KISS1 upregulation, and MAPK inhibition attenuated the MPP+-induced inflammation, oxidative stress and apoptosis in SH-SY5Y cells. CONCLUSION STAT4 inhibits KISS1 to attenuate the oxidative damage, inflammation and neuronal apoptosis in PD by inactivating the MAPK pathway.
Collapse
Affiliation(s)
- XiaoLei Zhang
- Department of Neurology, Shanxi People's Hospital, Taiyuan 030012, China.
| | - Yu Wang
- Department of Neurology, Fifth Hospital of Shanxi Medical University, Taiyuan 030012, China
| | - Jia Lv
- Department of Neurology, Fifth Hospital of Shanxi Medical University, Taiyuan 030012, China
| |
Collapse
|
4
|
Pathomechanisms of cognitive impairment in progressive supranuclear palsy. J Neural Transm (Vienna) 2023; 130:481-493. [PMID: 36862189 DOI: 10.1007/s00702-023-02613-w] [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: 01/16/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disorder characterized by early postural instability and falls, oculomotor dysfunction (vertical supranuclear gaze palsy), parkinsonism with poor response to levodopa, pseudobulbar palsy, and cognitive impairment. This four-repeat tauopathy is morphologically featured by accumulation of tau protein in neurons and glia causing neuronal loss and gliosis in the extrapyramidal system associated with cortical atrophy and white matter lesions. Cognitive impairment being frequent in PSP and more severe than in multiple system atrophy and Parkinson disease, is dominated by executive dysfunction, with milder difficulties in memory, and visuo-spatial and naming dysfunctions. Showing longitudinal decline, it has been related to a variety of pathogenic mechanisms associated with the underlying neurodegenerative process, such as involvement of cholinergic and muscarinergic dysfunctions, and striking tau pathology in frontal and temporal cortical regions associated with reduced synaptic density. Altered striatofrontal, fronto-cerebellar, parahippocampal, and multiple subcortical structures, as well as widespread white matter lesions causing extensive connectivity disruptions in cortico-subcortical and cortico-brainstem connections, support the concept that PSP is a brain network disruption disorder. The pathophysiology and pathogenesis of cognitive impairment in PSP, as in other degenerative movement disorders, are complex and deserve further elucidation as a basis for adequate treatment to improve the quality of life of patients with this fatal disease.
Collapse
|
5
|
Shreevatsa B, Dharmashekara C, Jain AS, Amachawadi R, Achar RR, Syed A, Shivamallu C, Kollur SP, Frau J, Flores-Holguín N, Glossman-Mitnik D. An insight into reactivity and bioactivity properties of quorum sensing peptides against PDE10A: a computational peptidology approach. J Mol Model 2022; 28:209. [PMID: 35789297 DOI: 10.1007/s00894-022-05176-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022]
Abstract
Peptides are currently the most promising lead molecules. Quorum sensing peptides have a variety of structural features and are regularly exposed to post-translational modifications. Antiparkinsonian drugs lose their efficacy after a long period of use, and patients develop motor problems such as drug-induced dyskinesia (DIDs). The interaction between PDE10A and cAMP is necessary for dopamine neurotransmission and may play a role in Parkinson's disease pathogenesis. cAMP and cGMP are cyclic nucleotides that act as secondary messengers in the signal transduction pathway, influencing a range of CNS activities. PDE enzymes hydrolyze phosphodiester bonds to break down cAMP and cGMP, allowing them to control intracellular levels of these second messengers effectively. PDE expression, and hence cyclic nucleotide levels and their downstream targets, may change with age and in numerous age-related illnesses, including Parkinson's disease, according to mounting evidence. At the peak of dyskinesias, cyclic nucleotide levels were lower, and using phosphodiesterase inhibitors before antiparkinsonian medicines reduced the severity of dyskinesias. In a recent study, PapRIV was found to have the ability to activate BV-2 microglia cells, indicating that this quorum sensing peptide may play a role in gut-brain contact. As a result of the current in silico work, mainly focused on QSPs as a lead molecule for inhibiting PDE10A, the SRNAT QSP sequence has been a potent molecule in molecular docking and molecular dynamics simulations. Furthermore, we can test the efficiency of therapeutic components in vitro and in vivo utilizing this computational approach against PDE10A.
Collapse
Affiliation(s)
- Bhargav Shreevatsa
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India
| | - Chandan Dharmashekara
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India
| | - Anisha S Jain
- Department of Microbiology, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India
| | - Raghavendra Amachawadi
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, 66506-5800, Kansas, USA
| | - Raghu Ram Achar
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India.
| | - Shiva Prasad Kollur
- Department of Sciences, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Mysuru, 570026, Karnataka, India.
| | - Juan Frau
- Departament de Química, Universitat de les Illes Balears, Palma de Mallorca, E-07122, Illes Balears, Spain
| | - Norma Flores-Holguín
- Laboratorio Virtual NANOCOSMOS, Departamento de Medio Ambiente y Energía, Centro de Investigación en Materiales Avanzados, Chihuahua, 31136, Chihuahua, Mexico
| | - Daniel Glossman-Mitnik
- Laboratorio Virtual NANOCOSMOS, Departamento de Medio Ambiente y Energía, Centro de Investigación en Materiales Avanzados, Chihuahua, 31136, Chihuahua, Mexico.
| |
Collapse
|
6
|
Lu J, Wang X, Wu A, Cao Y, Dai X, Liang Y, Li X. Ginsenosides in central nervous system diseases: Pharmacological actions, mechanisms, and therapeutics. Phytother Res 2022; 36:1523-1544. [PMID: 35084783 DOI: 10.1002/ptr.7395] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/11/2022]
Abstract
The nervous system is one of the most complex physiological systems, and central nervous system diseases (CNSDs) are serious diseases that affect human health. Ginseng (Panax L.), the root of Panax species, are famous Chinese herbs that have been used for various diseases in China, Japan, and Korea since ancient times, and remain a popular natural medicine used worldwide in modern times. Ginsenosides are the main active components of ginseng, and increasing evidence has demonstrated that ginsenosides can prevent CNSDs, including neurodegenerative diseases, memory and cognitive impairment, cerebral ischemia injury, depression, brain glioma, multiple sclerosis, which has been confirmed in numerous studies. Therefore, this review summarizes the potential pathways by which ginsenosides affect the pathogenesis of CNSDs mainly including antioxidant effects, anti-inflammatory effects, anti-apoptotic effects, and nerve protection, which provides novel ideas for the treatment of CNSDs.
Collapse
Affiliation(s)
- Jing Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anxin Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Youdan Liang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Daneshvar Kakhaki R, Kouchaki E, Dadgostar E, Behnam M, Tamtaji OR, Nikoueinejad H, Akbari H. The correlation of helios and neuropilin-1 frequencies with parkinson disease severity. Clin Neurol Neurosurg 2020; 192:105833. [PMID: 32305590 DOI: 10.1016/j.clineuro.2020.105833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 02/11/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Parkinson disease (PD), a neurodegenerative disease, has also some immunologic basis in which several regulatory factors, like Helios and Neuropilin-1 (NRP-1) may show some roles in its pathogenesis. We aimed to evaluate the circulatory frequency of T regulatory cells (Tregs) expressing Helios and NRP-1 in PD. PATIENTS AND METHODS In this case-control study, 83 patients with PD and 83 healthy controls were enrolled. The diagnosis of PD was accomplished in accordance with clinical diagnostic criteria of the UK Parkinson Disease Society Brain Bank. The modified Hoehn and Yahr (H and Y) were used to measure the severity of PD. Flow cytometry was used to evaluate the circulatory frequency of CD4+CD25+Foxp3+Tregs expressing and Helios and NRP-1 in all participants. Also, correlation of H and Y with such frequencies was evaluated. RESULTS Our findings showed that frequency of CD4+CD25+Foxp3+Tregs expressing NRP-1 (P = 0.04) and Helios (P = 0.01) in patients with PD was significantly higher than those in healthy subjects. The frequency of Tregs expressing Helios and NRP-1 showed a negative correlation with H and Y criteria and disease duration. Multiple linear regression analysis indicated that the severity of PD is the only effective factor on the frequency of CD4+CD25+Foxp3+NRP-1+Tregs (P = 0.012) and CD4+CD25+FoxP3+ Helios + Tregs (P = 0.038). CONCLUSION Our study showed that the increased frequency of Tregs expressing Helios and NRP-1 is associated with the severity of PD.
Collapse
Affiliation(s)
- Reza Daneshvar Kakhaki
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Ebrahim Kouchaki
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Mohammad Behnam
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hassan Nikoueinejad
- Department of Immunology, Baqiyatallah University of Medical Sciences, Tehran, Iran; Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Baqiyatallah Hospital, Mollasadra Ave., Vanak Sq., P.O. Box: 19395-5487, Tehran, Iran.
| | - Hossein Akbari
- Department of Biostatistics and Public Health, Faculty of Health, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Hwang SR, Hwang SW, Chen JC, Hwang JH. Association of periodic limb movements during sleep and Parkinson disease: A retrospective clinical study. Medicine (Baltimore) 2019; 98:e18444. [PMID: 31861016 PMCID: PMC6940135 DOI: 10.1097/md.0000000000018444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Both of periodic limb movements during sleep (PLMS) and Parkinson disease (PD) were related with dopaminergic system dysfunction. We aimed to investigate the detailed association of PLMS severity and PD.Clinical and overnight polysomnographic data of 2230 adults older than 40 from a community hospital between November 2011 and June 2017 in Taiwan were collected retrospectively. The association of PLMS severity and PD was analyzed by Fisher exact test, univariate, and multivariate logistic regression.The mean age was 55.6 years old (standard deviation = 9.8, range = 40-91) for all subjects. There were 2205 subjects without PD and 25 subjects with PD in this study. The distribution of PLMS severity was not significantly different between subjects without PD and with PD (Fischer exact test, P = .215). Also, PLMS was not significantly associated with PD using univariate and multivariate logistic regression.The PLMS severity was not associated with PD.
Collapse
Affiliation(s)
| | | | - Jin-Cherng Chen
- Departments of Neurosurgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi
- School of Medicine, Tzu Chi University, Hualien
| | - Juen-Haur Hwang
- School of Medicine, Tzu Chi University, Hualien
- Department of Otolaryngology-Head and Neck Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
9
|
Masi A, Narducci R, Mannaioni G. Harnessing ionic mechanisms to achieve disease modification in neurodegenerative disorders. Pharmacol Res 2019; 147:104343. [PMID: 31279830 DOI: 10.1016/j.phrs.2019.104343] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/19/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022]
Abstract
Progressive neuronal death is the key pathogenic event leading to clinical symptoms in neurodegenerative disorders (NDDs). Neuroprotective treatments are virtually unavailable, partly because of the marked internal heterogeneity of the mechanisms underlying pathology. Targeted neuroprotection would require deep mechanistic knowledge across the entire aetiological spectrum of each NDD and the development of tailored treatments. Although ideal, this strategy appears challenging, as it would require a degree of characterization of both the disease and the patient that is currently unavailable. The alternate strategy is to search for commonalities across molecularly distinct NDD forms and exploit these for the development of drugs with broad-spectrum efficacy. In this view, mounting evidence points to ionic mechanisms (IMs) as targets with potential therapeutic efficacy across distinct NDD subtypes. The scope of this review is to present clinical and preclinical evidence supporting the link between disruption of IMs and neuronal death in specific NDDs and to critically revise past and ongoing attempts of harnessing IMs for the development of neuroprotective treatments.
Collapse
Affiliation(s)
- A Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy; School of Pharmacy, University of Camerino, Camerino, Italy.
| | - R Narducci
- Italian Institute of Technology (IIT), Department of Neuroscience and Brain Technologies, Genova, Italy
| | - G Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy; Toxicology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| |
Collapse
|
10
|
Increased serum levels of TNF-α and decreased serum levels of IL-27 in patients with Parkinson disease and their correlation with disease severity. Clin Neurol Neurosurg 2019; 166:76-79. [PMID: 29408778 DOI: 10.1016/j.clineuro.2018.01.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/16/2018] [Accepted: 01/20/2018] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Immunological basis of neurodegenerative diseases including Alzheimer and Parkinson disease (PD) has some important roles in their pathogenesis. There are conflicting studies to serum level of TNF-α in PD. Also, according to our finding there is no report evaluating serum level of IL-27 in PD. This study correlates the serum level of those factors with severity of PD. PATIENTS AND METHODS In this case-control study, 83 patients with PD and 83 healthy volunteers were enrolled. The diagnosis was fulfilled in accordance with clinical diagnostic criteria of the UK Parkinson's Disease Society Brain Bank by two neurologists. The modified Hoehn and Yahr (H and Y) scale was used to evaluate the severity of PD. Serum levels of TNF-α and IL-27 were measured by Elisa. Correlation of H and Y scale with serum levels of these cytokines was evaluated. RESULTS The serum levels of TNF-α were increased and serum levels of IL-27 were decreased in patients with PD compared to those in healthy subjects (P < 0.0001). There was a significant correlation between serum levels of TNF-α and IL-27 with H and Y scale. CONCLUSION Our study showed that the serum levels of TNF-α and IL-27 may be important prognostic biomarkers of PD.
Collapse
|
11
|
Carbone C, Costa A, Provensi G, Mannaioni G, Masi A. The Hyperpolarization-Activated Current Determines Synaptic Excitability, Calcium Activity and Specific Viability of Substantia Nigra Dopaminergic Neurons. Front Cell Neurosci 2017; 11:187. [PMID: 28701928 PMCID: PMC5487410 DOI: 10.3389/fncel.2017.00187] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/16/2017] [Indexed: 12/28/2022] Open
Abstract
Differential vulnerability between Substantia Nigra pars compacta (SNpc) and Ventral Tegmental Area (VTA) dopaminergic (DAergic) neurons is a hallmark of Parkinson’s disease (PD). Understanding the molecular bases of this key histopathological aspect would foster the development of much-needed disease-modifying therapies. Non-heterogeneous DAergic degeneration is present in both toxin-based and genetic animal models, suggesting that cellular specificity, rather than causing factors, constitutes the background for differential vulnerability. In this regard, we previously demonstrated that MPP+, a neurotoxin able to cause selective nigrostriatal degeneration in animal rodents and primates, inhibits the Hyperpolarization-activated current (Ih) in SNpc DAergic neurons and that pharmacological Ih antagonism causes potentiation of evoked Excitatory post-synaptic potentials (EPSPs). Of note, the magnitude of such potentiation is greater in the SNpc subfield, consistent with higher Ih density. In the present work, we show that Ih block-induced synaptic potentiation leads to the amplification of somatic calcium responses (SCRs) in vitro. This effect is specific for the SNpc subfield and largely mediated by L-Type calcium channels, as indicated by sensitivity to the CaV 1 blocker isradipine. Furthermore, Ih is downregulated by low intracellular ATP and determines the efficacy of GABAergic inhibition in SNpc DAergic neurons. Finally, we show that stereotaxic administration of Ih blockers causes SNpc-specific neurodegeneration and hemiparkinsonian motor phenotype in rats. During PD progression, Ih downregulation may result from mitochondrial dysfunction and, in concert with PD-related disinhibition of excitatory inputs, determine a SNpc-specific disease pathway.
Collapse
Affiliation(s)
- Carmen Carbone
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy
| | - Alessia Costa
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy
| | - Gustavo Provensi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy
| | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy.,Toxicology Unit, Azienda Ospedaliero-Universitaria CareggiFlorence, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy.,Toxicology Unit, Azienda Ospedaliero-Universitaria CareggiFlorence, Italy
| |
Collapse
|
12
|
Zhang J, Zhang Z, Bao J, Yu Z, Cai M, Li X, Wu T, Xiang J, Cai D. Jia-Jian-Di-Huang-Yin-Zi decoction reduces apoptosis induced by both mitochondrial and endoplasmic reticulum caspase12 pathways in the mouse model of Parkinson's disease. JOURNAL OF ETHNOPHARMACOLOGY 2017; 203:69-79. [PMID: 28163115 DOI: 10.1016/j.jep.2016.12.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/16/2016] [Accepted: 12/18/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a classical prescription of traditional Chinese medicine (TCM), Jia-Jian-Di-Huang-Yin-Zi decoction (JJDHYZ) has been used to treat the symptoms of neurological disorders with a long history. AIM OF THE STUDY To evaluate the effects and possible mechanisms of JJDHYZ on a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced subacute mouse model of Parkinson's disease. MATERIALS AND METHODS Adult male C57BL/6 mice were randomly divided into five groups: control, MPTP, JJDHYZ low dosage (JJDHYZ-L, 8.5g/kg/day), medium dosage (JJDHYZ-M, 17g/kg/day) and high dosage (JJDHYZ-H, 34g/kg/day). Behavioral tests, immunohistochemistry, immunofluorescence, and high-performance liquid chromatography (HPLC) were conducted to evaluate the neuroprotective effects of JJDHYZ. The mechanism was further explored using TdT-mediated dUTP nick end labeling staining and transmission electron microscopy. The protein expression of Bax, Bcl-2, cytochrome c, full-length caspase9, cleaved caspase9, cleaved caspase3, caspase12 and C/EBP homologous protein was assessed. The toxicity on hepatocytes and renal cells was detected using the enzyme-linked immunosorbent assay kits. RESULTS JJDHYZ-H restored the behavior performance impaired by MPTP, and reduced the loss of tyrosine hydroxylase. Additionally, it blocked the apoptosis, activated cleaved caspase3 and protected the ultrastructural integrity of mitochondria by regulating the expression of proteins in both mitochondrial and endoplasmic reticulum (ER) caspase12 pathways. CONCLUSIONS JJDHYZ-H showed behavior recovery and dopamine neuron protection by inhibiting the apoptotic activities associated with mitochondrial and ER caspase12 pathways.
Collapse
Affiliation(s)
- Jingsi Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhennian Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jie Bao
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhonghai Yu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ting Wu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
13
|
Koo JH, Cho JY, Lee UB. Treadmill exercise alleviates motor deficits and improves mitochondrial import machinery in an MPTP-induced mouse model of Parkinson's disease. Exp Gerontol 2017; 89:20-29. [PMID: 28062370 DOI: 10.1016/j.exger.2017.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/26/2016] [Accepted: 01/02/2017] [Indexed: 11/16/2022]
Abstract
Alpha-synuclein (α-Syn) accumulation is significantly correlated with motor deficits and mitochondrial dysfunction in Parkinson's disease (PD), but the molecular mechanism underlying its pathogenesis is unclear. In this study, we investigated the effects of treadmill exercise on motor deficits and mitochondrial dysfunction in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Treadmill exercise inhibited dopaminergic neuron loss by promoting the expression of tyrosine hydroxylase (TH) and dopamine transporter (DAT) and seemed to improve cell survival by reducing α-Syn expression. Most importantly, treadmill exercise increased expression of the mitochondrial import machinery proteins TOM-40, TOM-20, and TIM-23. This was associated with decreased α-Syn expression and subsequent upregulation of the mitochondrial proteins COX-I, COX-IV, and mtHSP70. Taken together, these results indicate that treadmill exercise may ameliorate motor deficits and improve mitochondrial dysfunction by reducing α-Syn expression in the MPTP-induced mouse model of PD.
Collapse
Affiliation(s)
- Jung-Hoon Koo
- Department of Exercise Biochemistry, Korea National Sport University, Seoul 138-763, Republic of Korea; Institute of Sport Science, Korea National Sport University, Seoul, 138-763, Republic of Korea
| | - Joon-Yong Cho
- Department of Exercise Biochemistry, Korea National Sport University, Seoul 138-763, Republic of Korea
| | - Ung-Bae Lee
- Department of Beauty Health Science, Shinhan University, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
14
|
Annesley SJ, Lay ST, De Piazza SW, Sanislav O, Hammersley E, Allan CY, Francione LM, Bui MQ, Chen ZP, Ngoei KRW, Tassone F, Kemp BE, Storey E, Evans A, Loesch DZ, Fisher PR. Immortalized Parkinson's disease lymphocytes have enhanced mitochondrial respiratory activity. Dis Model Mech 2016; 9:1295-1305. [PMID: 27638668 PMCID: PMC5117226 DOI: 10.1242/dmm.025684] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022] Open
Abstract
In combination with studies of post-mortem Parkinson's disease (PD) brains, pharmacological and genetic models of PD have suggested that two fundamental interacting cellular processes are impaired - proteostasis and mitochondrial respiration. We have re-examined the role of mitochondrial dysfunction in lymphoblasts isolated from individuals with idiopathic PD and an age-matched control group. As previously reported for various PD cell types, the production of reactive oxygen species (ROS) by PD lymphoblasts was significantly elevated. However, this was not due to an impairment of mitochondrial respiration, as is often assumed. Instead, basal mitochondrial respiration and ATP synthesis are dramatically elevated in PD lymphoblasts. The mitochondrial mass, genome copy number and membrane potential were unaltered, but the expression of indicative respiratory complex proteins was also elevated. This explains the increased oxygen consumption rates by each of the respiratory complexes in experimentally uncoupled mitochondria of iPD cells. However, it was not attributable to increased activity of the stress- and energy-sensing protein kinase AMPK, a regulator of mitochondrial biogenesis and activity. The respiratory differences between iPD and control cells were sufficiently dramatic as to provide a potentially sensitive and reliable biomarker of the disease state, unaffected by disease duration (time since diagnosis) or clinical severity. Lymphoblasts from control and PD individuals thus occupy two distinct, quasi-stable steady states; a 'normal' and a 'hyperactive' state characterized by two different metabolic rates. The apparent stability of the 'hyperactive' state in patient-derived lymphoblasts in the face of patient ageing, ongoing disease and mounting disease severity suggests an early, permanent switch to an alternative metabolic steady state. With its associated, elevated ROS production, the 'hyperactive' state might not cause pathology to cells that are rapidly turned over, but brain cells might accumulate long-term damage leading ultimately to neurodegeneration and the loss of mitochondrial function observed post-mortem. Whether the 'hyperactive' state in lymphoblasts is a biomarker specifically of PD or more generally of neurodegenerative disease remains to be determined.
Collapse
Affiliation(s)
- Sarah J Annesley
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Sui T Lay
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Shawn W De Piazza
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Oana Sanislav
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Eleanor Hammersley
- Department of Psychology and Counselling, School of Psychology and Public Health, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3986, Australia
| | - Claire Y Allan
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Lisa M Francione
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Minh Q Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Zhi-Ping Chen
- Department of Medicine, University of Melbourne St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Kevin R W Ngoei
- Department of Medicine, University of Melbourne St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | | | - Bruce E Kemp
- Department of Medicine, University of Melbourne St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Elsdon Storey
- Department of Medicine (Neuroscience), Monash University, (Alfred Hospital Campus), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Andrew Evans
- Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria 3052, Australia
| | - Danuta Z Loesch
- Department of Psychology and Counselling, School of Psychology and Public Health, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3986, Australia
| | - Paul R Fisher
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, College of Science Health and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| |
Collapse
|
15
|
Surmeier DJ, Schumacker PT, Guzman JD, Ilijic E, Yang B, Zampese E. Calcium and Parkinson's disease. Biochem Biophys Res Commun 2016; 483:1013-1019. [PMID: 27590583 DOI: 10.1016/j.bbrc.2016.08.168] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 01/07/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world. Its causes are poorly understood and there is no proven therapeutic strategy for slowing disease progression. The core motor symptoms of PD are caused by the death of dopaminergic neurons in the substantia nigra pars compacta (SNc). In these neurons, Ca2+entry through plasma membrane Cav1 channels drives a sustained feed-forward stimulation of mitochondrial oxidative phosphorylation. Although this design helps prevent bioenergetic failure when activity needs to be sustained, it leads to basal mitochondrial oxidant stress. Over decades, this basal oxidant stress could compromise mitochondrial function and increase mitophagy, resulting in increased vulnerability to other proteostatic stressors, like elevated alpha synuclein expression. Because this feedforward mechanism is no longer demanded by our lifestyle, it could be dispensed with. Indeed, use of dihydropyridines - negative allosteric modulators of Cav1 Ca2+ channels - comes with little or no effect on brain function but is associated with decreased risk and progression of PD. An ongoing, NIH sponsored, Phase 3 clinical trial in North America is testing the ability of one member of the dihydropyridine class (isradipine) to slow PD progression in early stage patients. The review summarizes the rationale for the trial and outlines some unanswered questions.
Collapse
Affiliation(s)
- D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA.
| | - Paul T Schumacker
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Jaime D Guzman
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Ema Ilijic
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Ben Yang
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Enrico Zampese
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| |
Collapse
|
16
|
Neuroprotective Transcription Factors in Animal Models of Parkinson Disease. Neural Plast 2015; 2016:6097107. [PMID: 26881122 PMCID: PMC4736191 DOI: 10.1155/2016/6097107] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/10/2015] [Accepted: 07/14/2015] [Indexed: 11/28/2022] Open
Abstract
A number of transcription factors, including En1/2, Foxa1/2, Lmx1a/b, Nurr1, Otx2, and Pitx3, with key roles in midbrain dopaminergic (mDA) neuron development, also regulate adult mDA neuron survival and physiology. Mouse models with targeted disruption of some of these genes display several features reminiscent of Parkinson disease (PD), in particular the selective and progressive loss of mDA neurons in the substantia nigra pars compacta (SNpc). The characterization of these animal models has provided valuable insights into various mechanisms of PD pathogenesis. Therefore, the dissection of the mechanisms and survival signalling pathways engaged by these transcription factors to protect mDA neuron from degeneration can suggest novel therapeutic strategies. The work on En1/2-mediated neuroprotection also highlights the potential of protein transduction technology for neuroprotective approaches in PD.
Collapse
|
17
|
Heng Y, Zhang QS, Mu Z, Hu JF, Yuan YH, Chen NH. Ginsenoside Rg1 attenuates motor impairment and neuroinflammation in the MPTP-probenecid-induced parkinsonism mouse model by targeting α-synuclein abnormalities in the substantia nigra. Toxicol Lett 2015; 243:7-21. [PMID: 26723869 DOI: 10.1016/j.toxlet.2015.12.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/14/2015] [Accepted: 12/21/2015] [Indexed: 11/27/2022]
Abstract
Parkinson's disease (PD) is pathologically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of aggregated α-synuclein in specific central nervous system (CNS) regions. Disease development is attributed to α-synuclein abnormalities, particularly aggregation and phosphorylation. The ginsenoside Rg1, an active component of ginseng, possesses neuroprotective and anti-inflammatory effects. The purpose of the present study was to evaluate these activities of Rg1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid (MPTP/p)-induced PD mouse model for the first time and to elucidate the underlying mechanisms. Oral treatment with Rg1 significantly attenuated the high MPTP-induced mortality, behavior defects, loss of dopamine neurons and abnormal ultrastructure changes in the SNpc. Other assays indicated that the protective effect of Rg1 may be mediated by its anti-neuroinflammatory properties. Rg1 regulated MPTP-induced reactive astrocytes and microglia and decreased the release of cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the SNpc. Rg1 also alleviated the unusual MPTP-induced increase in oligomeric, phosphorylated and disease-related α-synuclein in the SNpc. In conclusion, Rg1 protects dopaminergic neurons, most likely by reducing aberrant α-synuclein-mediated neuroinflammation, and holds promise for PD therapeutics.
Collapse
Affiliation(s)
- Yang Heng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qiu-Shuang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zheng Mu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jin-Feng Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
18
|
Dissecting the role of Engrailed in adult dopaminergic neurons--Insights into Parkinson disease pathogenesis. FEBS Lett 2015; 589:3786-94. [PMID: 26459030 DOI: 10.1016/j.febslet.2015.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/18/2015] [Accepted: 10/06/2015] [Indexed: 11/23/2022]
Abstract
The homeoprotein Engrailed (Engrailed-1/Engrailed-2, collectively En1/2) is not only a survival factor for mesencephalic dopaminergic (mDA) neurons during development, but continues to exert neuroprotective and physiological functions in adult mDA neurons. Loss of one En1 allele in the mouse leads to progressive demise of mDA neurons in the ventral midbrain starting from 6 weeks of age. These mice also develop Parkinson disease-like motor and non-motor symptoms. The characterization of En1 heterozygous mice have revealed striking parallels to central mechanisms of Parkinson disease pathogenesis, mainly related to mitochondrial dysfunction and retrograde degeneration. Thanks to the ability of homeoproteins to transduce cells, En1/2 proteins have also been used to protect mDA neurons in various experimental models of Parkinson disease. This neuroprotection is partly linked to the ability of En1/2 to regulate the translation of certain nuclear-encoded mitochondrial mRNAs for complex I subunits. Other transcription factors that govern mDA neuron development (e.g. Foxa1/2, Lmx1a/b, Nurr1, Otx2, Pitx3) also continue to function for the survival and maintenance of mDA neurons in the adult and act through partially overlapping but also diverse mechanisms.
Collapse
|
19
|
Chen JC, Tsai TY, Li CY, Hwang JH. Obstructive sleep apnea and risk of Parkinson's disease: a population-based cohort study. J Sleep Res 2015; 24:432-7. [PMID: 25810019 DOI: 10.1111/jsr.12289] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/01/2015] [Indexed: 01/10/2023]
Abstract
Sleep disorders could be associated with neurodegenerative diseases. This study aimed to determine the risk of Parkinson's disease in patients with obstructive sleep apnea. The incident cases of newly diagnosed obstructive sleep apnea were identified between 2000 and 2009 from the medical claims database of National Health Institute of Taiwan. The risk of Parkinson's disease onset at least 1 year after the diagnosis of obstructive sleep apnea was measured during and up to 11 years of period, compared to that of age- and gender-matched controls estimated in the same period. A total of 5864 patients with newly diagnosed obstructive sleep apnea and 23,269 subjects without obstructive sleep apnea were identified for data analysis. The study reported that the incidence of Parkinson's disease in the obstructive sleep apnea cohort was approximately two times higher than that in the control cohort (2.57 versus 1.32 per 1000 person-years), with an adjusted hazard ratio of 1.84. Furthermore, the risk of Parkinson's disease was particularly greater for the obstructive sleep apnea with insomnia subgroup (adjusted hazard ratio = 1.97, 95% confidence interval = 1.44-2.69) than for the control cohort. The sex-age-specific analysis further discovered that the most elevated risk of Parkinson's disease onset was noted in female obstructive sleep apnea patients aged 50-69 years (adjusted hazard ratio = 2.82). This population-based study indicated that patients with obstructive sleep apnea, especially those who suffered from insomnia, are at an increased risk of Parkinson's disease onset.
Collapse
Affiliation(s)
- Jin-Cherng Chen
- Department of Neurosurgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tzung-Yi Tsai
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan.,Department of Environmental and Occupational Health, National Cheng Kung University, Tainan, Taiwan.,Department of Nursing, Tzu Chi College of Technology, Hualien, Taiwan
| | - Chung-Yi Li
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Juen-Haur Hwang
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Otolaryngology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| |
Collapse
|
20
|
Weinert M, Selvakumar T, Tierney TS, Alavian KN. Isolation, culture and long-term maintenance of primary mesencephalic dopaminergic neurons from embryonic rodent brains. J Vis Exp 2015. [PMID: 25741798 DOI: 10.3791/52475] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Degeneration of mesencephalic dopaminergic (mesDA) neurons is the pathological hallmark of Parkinson's diseae. Study of the biological processes involved in physiological functions and vulnerability and death of these neurons is imparative to understanding the underlying causes and unraveling the cure for this common neurodegenerative disorder. Primary cultures of mesDA neurons provide a tool for investigation of the molecular, biochemical and electrophysiological properties, in order to understand the development, long-term survival and degeneration of these neurons during the course of disease. Here we present a detailed method for the isolation, culturing and maintenance of midbrain dopaminergic neurons from E12.5 mouse (or E14.5 rat) embryos. Optimized cell culture conditions in this protocol result in presence of axonal and dendritic projections, synaptic connections and other neuronal morphological properties, which make the cultures suitable for study of the physiological, cell biological and molecular characteristics of this neuronal population.
Collapse
Affiliation(s)
- Maria Weinert
- Division of Brain Sciences, Department of Medicine, Imperial College London
| | | | - Travis S Tierney
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School
| | - Kambiz N Alavian
- Division of Brain Sciences, Department of Medicine, Imperial College London; Department of Internal Medicine, Endocrinology, Yale University School of Medicine;
| |
Collapse
|
21
|
Graham J, Hobson D, Ponnampalam A. High affinity hemoglobin and Parkinson’s disease. Med Hypotheses 2014; 83:819-21. [DOI: 10.1016/j.mehy.2014.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/03/2014] [Accepted: 10/20/2014] [Indexed: 10/24/2022]
|
22
|
Synthesis, pharmacological study and docking calculations of new benzo[f]coumarin derivatives as dual inhibitors of enzymatic systems involved in neurodegenerative diseases. Future Med Chem 2014; 6:371-83. [DOI: 10.4155/fmc.14.9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Due to the complex etiology of neurodegenerative diseases, there is growing interest in multitarget drugs. In this study we synthesized and evaluated a new series of compounds, with benzo[f]coumarin structure, as potential inhibitors of MAO-A, MAO-B, AChE and BuChE. Results: In vitro studies show that most of the studied compounds inhibited the activity of MAO-B in the nano- to micro-molar range. 3-(3´-methoxyphenyl)benzo[f]coumarin is the most active compound with an IC50 value against MAO-B of 2.44 nM. Most of the derivatives exhibited an important selectivity profile against the MAO-B isoform. Some of them also acted as in vitro inhibitors of BuChE, with 3-(2´-hydroxyphenyl)benzo[f]coumarin being the most active with an IC50 value of 1.13 µM. In addition, a theoretical study of the physicochemical properties of the new compounds, as well as a docking study in both MAO isoforms, were carried out. Important structure–activity relationships were obtained. Conclusion: Important preliminary structure–activity relationships were concluded from the experimental results. These results encourage us to further explore the potential of this chemical family as potential drug candidates for the treatment of Alzheimer‘s disease.
Collapse
|
23
|
Fujita KA, Ostaszewski M, Matsuoka Y, Ghosh S, Glaab E, Trefois C, Crespo I, Perumal TM, Jurkowski W, Antony PMA, Diederich N, Buttini M, Kodama A, Satagopam VP, Eifes S, del Sol A, Schneider R, Kitano H, Balling R. Integrating pathways of Parkinson's disease in a molecular interaction map. Mol Neurobiol 2014; 49:88-102. [PMID: 23832570 PMCID: PMC4153395 DOI: 10.1007/s12035-013-8489-4] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/13/2013] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a major neurodegenerative chronic disease, most likely caused by a complex interplay of genetic and environmental factors. Information on various aspects of PD pathogenesis is rapidly increasing and needs to be efficiently organized, so that the resulting data is available for exploration and analysis. Here we introduce a computationally tractable, comprehensive molecular interaction map of PD. This map integrates pathways implicated in PD pathogenesis such as synaptic and mitochondrial dysfunction, impaired protein degradation, alpha-synuclein pathobiology and neuroinflammation. We also present bioinformatics tools for the analysis, enrichment and annotation of the map, allowing the research community to open new avenues in PD research. The PD map is accessible at http://minerva.uni.lu/pd_map .
Collapse
Affiliation(s)
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Integrated Biobank of Luxembourg, Luxembourg City, Luxembourg
| | | | - Samik Ghosh
- The Systems Biology Institute, Minato-ku, Tokyo, Japan
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Christophe Trefois
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Isaac Crespo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Thanneer M. Perumal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Wiktor Jurkowski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Paul M. A. Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Nico Diederich
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Department of Neuroscience, Centre Hospitalier Luxembourg, Luxembourg City, Luxembourg
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Akihiko Kodama
- Faculty of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Venkata P. Satagopam
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Serge Eifes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Hiroaki Kitano
- The Systems Biology Institute, Minato-ku, Tokyo, Japan
- Sony Computer Science Laboratories, Shinagawa-ku, Tokyo, Japan
- Division of Systems Biology, Cancer Institute, Tokyo, Japan
- Open Biology Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa Japan
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
24
|
Truini A, Frontoni M, Cruccu G. Parkinson's disease related pain: a review of recent findings. J Neurol 2013; 260:330-4. [PMID: 23180185 PMCID: PMC3535357 DOI: 10.1007/s00415-012-6754-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 12/31/2022]
Abstract
In this review we summarize progress in research on Parkinson's disease-related pain as reported in articles published in the Journal of Neurology in the years 2011 and 2012.
Collapse
Affiliation(s)
- A. Truini
- Department of Neurology and Psychiatry, University Sapienza, Viale Università 30, 00185 Rome, Italy
| | - M. Frontoni
- Department of Neurology and Psychiatry, University Sapienza, Viale Università 30, 00185 Rome, Italy
| | - G. Cruccu
- Department of Neurology and Psychiatry, University Sapienza, Viale Università 30, 00185 Rome, Italy
| |
Collapse
|
25
|
Aboud AA, Tidball AM, Kumar KK, Neely MD, Ess KC, Erikson KM, Bowman AB. Genetic risk for Parkinson's disease correlates with alterations in neuronal manganese sensitivity between two human subjects. Neurotoxicology 2012; 33:1443-1449. [PMID: 23099318 DOI: 10.1016/j.neuro.2012.10.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 10/01/2012] [Accepted: 10/15/2012] [Indexed: 01/16/2023]
Abstract
Manganese (Mn) is an environmental risk factor for Parkinson's disease (PD). Recessive inheritance of PARK2 mutations is strongly associated with early onset PD (EOPD). It is widely assumed that the influence of PD environmental risk factors may be enhanced by the presence of PD genetic risk factors in the genetic background of individuals. However, such interactions may be difficult to predict owing to the complexities of genetic and environmental interactions. Here we examine the potential of human induced pluripotent stem (iPS) cell-derived early neural progenitor cells (NPCs) to model differences in Mn neurotoxicity between a control subject (CA) with no known PD genetic risk factors and a subject (SM) with biallelic loss-of-function mutations in PARK2 and family history of PD but no evidence of PD by neurological exam. Human iPS cells were generated from primary dermal fibroblasts of both subjects. We assessed several outcome measures associated with Mn toxicity and PD. No difference in sensitivity to Mn cytotoxicity or mitochondrial fragmentation was observed between SM and CA NPCs. However, we found that Mn exposure was associated with significantly higher reactive oxygen species (ROS) generation in SM compared to CA NPCs despite significantly less intracellular Mn accumulation. Thus, this report offers the first example of human subject-specific differences in PD-relevant environmental health related phenotypes that are consistent with pathogenic interactions between known genetic and environmental risk factors for PD.
Collapse
Affiliation(s)
- Asad A Aboud
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA
| | - Andrew M Tidball
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA
| | - Kevin K Kumar
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA; Vanderbilt Medical Scientist Training Program, Nashville, TN 37232-8552, USA
| | - M Diana Neely
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA
| | - Kevin C Ess
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA; Vanderbilt Center for Stem Cell Biology and The Department of Pediatrics, Nashville, TN 37232-8552, USA
| | - Keith M Erikson
- University of North Carolina-Greensboro, Nutrition Department, Greensboro, NC 27402-6107, USA
| | - Aaron B Bowman
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA; Vanderbilt Center for Stem Cell Biology and The Department of Pediatrics, Nashville, TN 37232-8552, USA; Vanderbilt Center in Molecular Toxicology, Vanderbilt University, Nashville, TN 37232-8552, USA.
| |
Collapse
|