51
|
Paul R, Dutta A, Phukan BC, Mazumder MK, Justin-Thenmozhi A, Manivasagam T, Bhattacharya P, Borah A. Accumulation of Cholesterol and Homocysteine in the Nigrostriatal Pathway of Brain Contributes to the Dopaminergic Neurodegeneration in Mice. Neuroscience 2018; 388:347-356. [DOI: 10.1016/j.neuroscience.2018.07.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 07/15/2018] [Accepted: 07/23/2018] [Indexed: 11/26/2022]
|
52
|
Brown EG, Goldman SM, Tanner CM. Mendel and urate: Acid test or random noise? Parkinsonism Relat Disord 2018; 53:1-3. [PMID: 30100365 DOI: 10.1016/j.parkreldis.2018.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 08/06/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Ethan G Brown
- Department of Neurology, University of California - San Francisco, San Francisco, CA, USA; Department of Neurology, Weil Institute for Neurosciences, University of California - San Francisco, San Francisco, CA, USA
| | - Samuel M Goldman
- Department of Neurology, University of California - San Francisco, San Francisco, CA, USA; Division of Occupational and Environmental Medicine, University of California - San Francisco, San Francisco, CA, USA; Medical Service, San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
| | - Caroline M Tanner
- Department of Neurology, University of California - San Francisco, San Francisco, CA, USA; Department of Neurology, Weil Institute for Neurosciences, University of California - San Francisco, San Francisco, CA, USA; Parkinson's Disease Research, Education and Clinical Center, San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA.
| |
Collapse
|
53
|
Ng YW, Say YH. Palmitic acid induces neurotoxicity and gliatoxicity in SH-SY5Y human neuroblastoma and T98G human glioblastoma cells. PeerJ 2018; 6:e4696. [PMID: 29713567 PMCID: PMC5924683 DOI: 10.7717/peerj.4696] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022] Open
Abstract
Background Obesity-related central nervous system (CNS) pathologies like neuroinflammation and reactive gliosis are associated with high-fat diet (HFD) related elevation of saturated fatty acids like palmitic acid (PA) in neurons and astrocytes of the brain. Methods Human neuroblastoma cells SH-SY5Y (as a neuronal model) and human glioblastoma cells T98G (as an astrocytic model), were treated with 100–500 µM PA, oleic acid (OA) or lauric acid (LA) for 24 h or 48 h, and their cell viability was assessed by 3-(4,5-dimetylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effects of stable overexpression of γ-synuclein (γ-syn), a neuronal protein recently recognized as a novel regulator of lipid handling in adipocytes, and transient overexpression of Parkinson’s disease (PD) α-synuclein [α-syn; wild-type (wt) and its pathogenic mutants A53T, A30P and E46K] in SH-SY5Y and T98G cells, were also evaluated. The effects of co-treatment of PA with paraquat (PQ), a Parkinsonian pesticide, and leptin, a hormone involved in the brain-adipose axis, were also assessed. Cell death mode and cell cycle were analyzed by Annexin V/PI flow cytometry. Reactive oxygen species (ROS) level was determined using 2′,7′-dichlorofluorescien diacetate (DCFH-DA) assay and lipid peroxidation level was determined using thiobarbituric acid reactive substances (TBARS) assay. Results MTT assay revealed dose- and time-dependent PA cytotoxicity on SH-SY5Y and T98G cells, but not OA and LA. The cytotoxicity was significantly lower in SH-SY5Y-γ-syn cells, while transient overexpression of wt α-syn or its PD mutants (A30P and E46K, but not A53T) modestly (but still significantly) rescued the cytotoxicity of PA in SH-SY5Y and T98G cells. Co-treatment of increasing concentrations of PQ exacerbated PA’s neurotoxicity. Pre-treatment of leptin, an anti-apoptotic adipokine, did not successfully rescue SH-SY5Y cells from PA-induced cytotoxicity—suggesting a mechanism of PA-induced leptin resistance. Annexin V/PI flow cytometry analysis revealed PA-induced increase in percentages of cells in annexin V-positive/PI-negative quadrant (early apoptosis) and subG0-G1 fraction, accompanied by a decrease in G2-M phase cells. The PA-induced ROS production and lipid peroxidation was at greater extent in T98G as compared to that in SH-SY5Y. Discussion In conclusion, PA induces apoptosis by increasing oxidative stress in neurons and astrocytes. Taken together, the results suggest that HFD may cause neuronal and astrocytic damage, which indirectly proposes that CNS pathologies involving neuroinflammation and reactive gliosis could be prevented via the diet regimen.
Collapse
Affiliation(s)
- Yee-Wen Ng
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Kampar Campus, Kampar, Perak, Malaysia
| | - Yee-How Say
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Kampar Campus, Kampar, Perak, Malaysia
| |
Collapse
|
54
|
LaHue SC, Comella CL, Tanner CM. The best medicine? The influence of physical activity and inactivity on Parkinson's disease. Mov Disord 2017; 31:1444-1454. [PMID: 27477046 DOI: 10.1002/mds.26728] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/01/2016] [Accepted: 06/12/2016] [Indexed: 12/14/2022] Open
Abstract
The incidence of Parkinson's disease (PD) is expected to increase as our population ages and will likely strain the projected capacity of our health care system. Despite being the most common movement disorder, there have been few noninvasive therapeutic advances for people with PD since the first levodopa clinical trial in 1961. The study of PD pathogenesis, combined with an appreciation for the biochemical mechanisms by which physical activity and exercise may impact physiology, has resulted in emerging hypotheses for new modifiable risk factors for PD. Physical activity and exercise as a means of preventing PD, or maintaining the functionality of people with PD, are a promising area of investigation. Conversely, physical inactivity is implicated in many disease states, some of which are also correlated with the development of PD, such as metabolic syndrome. The primary relationship between these diseases is likely rooted in heightened inflammation and oxidative stress at the cellular level. Physical activity and exercise as a means of attenuating inflammation have led to increased interest in related potential therapeutic targets for PD. Ultimately, these findings may translate into low-cost, universally available therapies for PD disease modification or prevention. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sara C LaHue
- Kaiser Permanente San Francisco Medical Center, San Francisco, California, USA
| | | | - Caroline M Tanner
- San Francisco Veterans Affairs Medical Center and Department of Neurology, University of California, San Francisco, California, USA.
| |
Collapse
|
55
|
Morgan AH, Rees DJ, Andrews ZB, Davies JS. Ghrelin mediated neuroprotection - A possible therapy for Parkinson's disease? Neuropharmacology 2017; 136:317-326. [PMID: 29277488 DOI: 10.1016/j.neuropharm.2017.12.027] [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] [Received: 10/06/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 12/31/2022]
Abstract
Parkinson's disease is a common age-related neurodegenerative disorder affecting 10 million people worldwide, but the mechanisms underlying its pathogenesis are still unclear. The disease is characterised by dopamine nerve cell loss in the mid-brain and intra-cellular accumulation of α-synuclein that results in motor and non-motor dysfunction. In this review, we discuss the neuroprotective effects of the stomach hormone, ghrelin, in models of Parkinson's disease. Recent findings suggest that it may modulate mitochondrial function and autophagic clearance of impaired organelle in response to changes in cellular energy balance. We consider the putative cellular mechanisms underlying ghrelin-action and the possible role of ghrelin mimetics in slowing or preventing Parkinson's disease progression. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Alwena H Morgan
- Molecular Neurobiology, Institute of Life Science, Medical School, Swansea University, UK
| | - Daniel J Rees
- Molecular Neurobiology, Institute of Life Science, Medical School, Swansea University, UK
| | - Zane B Andrews
- Biomedicine Discovery Institute & Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Jeffrey S Davies
- Molecular Neurobiology, Institute of Life Science, Medical School, Swansea University, UK.
| |
Collapse
|
56
|
On phagocytes and macular degeneration. Prog Retin Eye Res 2017; 61:98-128. [DOI: 10.1016/j.preteyeres.2017.06.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/29/2017] [Accepted: 06/05/2017] [Indexed: 12/17/2022]
|
57
|
Leehey M, Luo S, Sharma S, Wills AMA, Bainbridge JL, Wong PS, Simon DK, Schneider J, Zhang Y, Pérez A, Dhall R, Christine CW, Singer C, Cambi F, Boyd JT. Association of metabolic syndrome and change in Unified Parkinson's Disease Rating Scale scores. Neurology 2017; 89:1789-1794. [PMID: 28972194 DOI: 10.1212/wnl.0000000000004572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/28/2017] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To explore the association between metabolic syndrome and the Unified Parkinson's Disease Rating Scale (UPDRS) scores and, secondarily, the Symbol Digit Modalities Test (SDMT). METHODS This is a secondary analysis of data from 1,022 of 1,741 participants of the National Institute of Neurological Disorders and Stroke Exploratory Clinical Trials in Parkinson Disease Long-Term Study 1, a randomized, placebo-controlled trial of creatine. Participants were categorized as having or not having metabolic syndrome on the basis of modified criteria from the National Cholesterol Education Program Adult Treatment Panel III. Those who had the same metabolic syndrome status at consecutive annual visits were included. The change in UPDRS and SDMT scores from randomization to 3 years was compared in participants with and without metabolic syndrome. RESULTS Participants with metabolic syndrome (n = 396) compared to those without (n = 626) were older (mean [SD] 63.9 [8.1] vs 59.9 [9.4] years; p < 0.0001), were more likely to be male (75.3% vs 57.0%; p < 0.0001), and had a higher mean uric acid level (men 5.7 [1.3] vs 5.3 [1.1] mg/dL, women 4.9 [1.3] vs 3.9 [0.9] mg/dL, p < 0.0001). Participants with metabolic syndrome experienced an additional 0.6- (0.2) unit annual increase in total UPDRS (p = 0.02) and 0.5- (0.2) unit increase in motor UPDRS (p = 0.01) scores compared with participants without metabolic syndrome. There was no difference in the change in SDMT scores. CONCLUSIONS Persons with Parkinson disease meeting modified criteria for metabolic syndrome experienced a greater increase in total UPDRS scores over time, mainly as a result of increases in motor scores, compared to those who did not. Further studies are needed to confirm this finding. CLINICALTRIALSGOV IDENTIFIER NCT00449865.
Collapse
Affiliation(s)
- Maureen Leehey
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC.
| | - Sheng Luo
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Saloni Sharma
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Anne-Marie A Wills
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Jacquelyn L Bainbridge
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Pei Shieen Wong
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - David K Simon
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Jay Schneider
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Yunxi Zhang
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Adriana Pérez
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Rohit Dhall
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Chadwick W Christine
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Carlos Singer
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Franca Cambi
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - James T Boyd
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| |
Collapse
|
58
|
Hypercholesterolemia causes psychomotor abnormalities in mice and alterations in cortico-striatal biogenic amine neurotransmitters: Relevance to Parkinson's disease. Neurochem Int 2017; 108:15-26. [DOI: 10.1016/j.neuint.2017.01.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 12/20/2022]
|
59
|
Sharma JC, Lewis A. Weight in Parkinson's Disease: Phenotypical Significance. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 134:891-919. [PMID: 28805588 DOI: 10.1016/bs.irn.2017.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Body weight in Parkinson's disease (PD) is a significant nonmotor feature. Weight homeostasis is a complex physiological process and gets deranged in PD patients leading to changes in weight. While both the low and high body weight have been reported as risk factors for PD, the majority of PD patients have a lower weight and a subset of patients lose weight during the course of the disease, while a small proportion gain weight. A number of clinical parameters such as older age, impaired cognition, severity of disease, and an imbalance of food intake determined by satiety and hunger hormones have been reported to be associated with but not the cause of weight change. Low body weight and weight loss have a negative impact on disease severity, dyskinesia quality of life, and mortality indicative of disease progression. An early assessment of olfactory impairment seems to identify patients at risk of weight loss, the patients with more severe olfactory loss-anosmic group, lose weight as compared to the patients with some preservation of olfaction, the hyposmic group. Higher levodopa dose per kilogram body weight increases the risk of dyskinesia, higher body weight seems to be protective against this complication. The identification of PD patients according to the nonmotor phenotype of "Park-olfaction-weight-phenotype" and the "olfaction-weight-dyskinesia" triad should help to develop strategies to prevent weight reduction and improve general health and complications of PD patients. The phenotype seems to reflect a differential prodromal pathology and influence clinical disease. Higher body weight patients would benefit from life style changes to achieve a healthy profile. Weight monitoring and weight orientated approach to management of PD patients should help to improve their outcome. Body weight change might be a surrogate to disease progression and may be used to investigate neuroprotection strategies.
Collapse
Affiliation(s)
- Jagdish C Sharma
- Geriatric Medicine (Movement Disorders), Lincoln County Hospital, Lincoln, United Kingdom; University of Lincoln, Lincoln, United Kingdom.
| | - Anna Lewis
- Geriatric Medicine (Movement Disorders), Lincoln County Hospital, Lincoln, United Kingdom; University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
60
|
Noyce AJ, Kia DA, Hemani G, Nicolas A, Price TR, De Pablo-Fernandez E, Haycock PC, Lewis PA, Foltynie T, Davey Smith G, Schrag A, Lees AJ, Hardy J, Singleton A, Nalls MA, Pearce N, Lawlor DA, Wood NW. Estimating the causal influence of body mass index on risk of Parkinson disease: A Mendelian randomisation study. PLoS Med 2017; 14:e1002314. [PMID: 28609445 PMCID: PMC5469450 DOI: 10.1371/journal.pmed.1002314] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 05/04/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Both positive and negative associations between higher body mass index (BMI) and Parkinson disease (PD) have been reported in observational studies, but it has been difficult to establish causality because of the possibility of residual confounding or reverse causation. To our knowledge, Mendelian randomisation (MR)-the use of genetic instrumental variables (IVs) to explore causal effects-has not previously been used to test the effect of BMI on PD. METHODS AND FINDINGS Two-sample MR was undertaken using genome-wide association (GWA) study data. The associations between the genetic instruments and BMI were obtained from the GIANT consortium and consisted of the per-allele difference in mean BMI for 77 independent variants that reached genome-wide significance. The per-allele difference in log-odds of PD for each of these variants was estimated from a recent meta-analysis, which included 13,708 cases of PD and 95,282 controls. The inverse-variance weighted method was used to estimate a pooled odds ratio (OR) for the effect of a 5-kg/m2 higher BMI on PD. Evidence of directional pleiotropy averaged across all variants was sought using MR-Egger regression. Frailty simulations were used to assess whether causal associations were affected by mortality selection. A combined genetic IV expected to confer a lifetime exposure of 5-kg/m2 higher BMI was associated with a lower risk of PD (OR 0.82, 95% CI 0.69-0.98). MR-Egger regression gave similar results, suggesting that directional pleiotropy was unlikely to be biasing the result (intercept 0.002; p = 0.654). However, the apparent protective influence of higher BMI could be at least partially induced by survival bias in the PD GWA study, as demonstrated by frailty simulations. Other important limitations of this application of MR include the inability to analyse non-linear associations, to undertake subgroup analyses, and to gain mechanistic insights. CONCLUSIONS In this large study using two-sample MR, we found that variants known to influence BMI had effects on PD in a manner consistent with higher BMI leading to lower risk of PD. The mechanism underlying this apparent protective effect warrants further study.
Collapse
Affiliation(s)
- Alastair J. Noyce
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
- Centre for Neuroscience and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Demis A. Kia
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Aude Nicolas
- Laboratory for Neurogenetics, National Institute on Aging National Institutes of Health, Bethesda, Maryland, United States of America
| | - T. Ryan Price
- Laboratory for Neurogenetics, National Institute on Aging National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eduardo De Pablo-Fernandez
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Philip C. Haycock
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Patrick A. Lewis
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
- School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, University College London, London, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | | | - Anette Schrag
- Department of Clinical Neurosciences, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Andrew J. Lees
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Andrew Singleton
- Laboratory for Neurogenetics, National Institute on Aging National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mike A. Nalls
- Laboratory for Neurogenetics, National Institute on Aging National Institutes of Health, Bethesda, Maryland, United States of America
- Data Tecnica International, Glen Echo, Maryland, United States of America
| | - Neil Pearce
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Debbie A. Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Nicholas W. Wood
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
61
|
Jang Y, Lee MJ, Han J, Kim SJ, Ryu I, Ju X, Ryu MJ, Chung W, Oh E, Kweon GR, Heo JY. A High-fat Diet Induces a Loss of Midbrain Dopaminergic Neuronal Function That Underlies Motor Abnormalities. Exp Neurobiol 2017; 26:104-112. [PMID: 28442947 PMCID: PMC5403908 DOI: 10.5607/en.2017.26.2.104] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Movement defects in obesity are associated with peripheral muscle defects, arthritis, and dysfunction of motor control by the brain. Although movement functionality is negatively correlated with obesity, the brain regions and downstream signaling pathways associated with movement defects in obesity are unclear. A dopaminergic neuronal pathway from the substantia nigra (SN) to the striatum is responsible for regulating grip strength and motor initiation through tyrosine hydroxylase (TH) activity-dependent dopamine release. We found that mice fed a high-fat diet exhibited decreased movement in open-field tests and an increase in missteps in a vertical grid test compared with normally fed mice. This motor abnormality was associated with a significant reduction of TH in the SN and striatum. We further found that phosphorylation of c-Jun N-terminal kinase (JNK), which modulates TH expression in the SN and striatum, was decreased under excess-energy conditions. Our findings suggest that high calorie intake impairs motor function through JNK-dependent dysregulation of TH in the SN and striatum.
Collapse
Affiliation(s)
- Yunseon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Jeongsu Han
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Ilhwan Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Xianshu Ju
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Woosuk Chung
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Korea
| |
Collapse
|
62
|
Frago LM, Chowen JA. Involvement of Astrocytes in Mediating the Central Effects of Ghrelin. Int J Mol Sci 2017; 18:ijms18030536. [PMID: 28257088 PMCID: PMC5372552 DOI: 10.3390/ijms18030536] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/16/2017] [Accepted: 02/25/2017] [Indexed: 12/31/2022] Open
Abstract
Although astrocytes are the most abundant cells in the mammalian brain, much remains to be learned about their molecular and functional features. Astrocytes express receptors for numerous hormones and metabolic factors, including the appetite-promoting hormone ghrelin. The metabolic effects of ghrelin are largely opposite to those of leptin, as it stimulates food intake and decreases energy expenditure. Ghrelin is also involved in glucose-sensing and glucose homeostasis. The widespread expression of the ghrelin receptor in the central nervous system suggests that this hormone is not only involved in metabolism, but also in other essential functions in the brain. In fact, ghrelin has been shown to promote cell survival and neuroprotection, with some studies exploring the use of ghrelin as a therapeutic agent against metabolic and neurodegenerative diseases. In this review, we highlight the possible role of glial cells as mediators of ghrelin's actions within the brain.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28009 Madrid, Spain.
- Department of Pediatrics, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28009 Madrid, Spain.
| |
Collapse
|
63
|
Diagnosis and body mass index effects on hippocampal volumes and neurochemistry in bipolar disorder. Transl Psychiatry 2017; 7:e1071. [PMID: 28350397 PMCID: PMC5404613 DOI: 10.1038/tp.2017.42] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 11/14/2016] [Accepted: 01/15/2017] [Indexed: 12/14/2022] Open
Abstract
We previously reported that higher body mass index (BMI) was associated with greater hippocampal glutamate+glutamine in people with bipolar disorder (BD), but not in non-BD healthy comparator subjects (HSs). In the current report, we extend these findings by examining the impact of BD diagnosis and BMI on hippocampal volumes and the concentrations of several additional neurochemicals in 57 early-stage BD patients and 31 HSs. Using 3-T magnetic resonance imaging and magnetic resonance spectroscopy, we measured bilateral hippocampal volumes and the hippocampal concentrations of four neurochemicals relevant to BD: N-acetylaspartate+N-acteylaspartylglutamate (tNAA), creatine+phosphocreatine (Cre), myoinositol (Ins) and glycerophosphocholine+phosphatidylcholine (Cho). We used multivariate factorial analysis of covariance to investigate the impact of diagnosis (patient vs HS) and BMI category (normal weight vs overweight/obese) on these variables. We found a main effect of diagnosis on hippocampal volumes, with patients having smaller hippocampi than HSs. There was no association between BMI and hippocampal volumes. We found diagnosis and BMI effects on hippocampal neurochemistry, with patients having lower Cre, Ins and Cho, and overweight/obese subjects having higher levels of these chemicals. In patient-only models that controlled for clinical and treatment variables, we detected an additional association between higher BMI and lower tNAA that was absent in HSs. To our knowledge, this was the first study to investigate the relative contributions of BD diagnosis and BMI to hippocampal volumes, and only the second to investigate their contributions to hippocampal chemistry. It provides further evidence that diagnosis and elevated BMI both impact limbic brain areas relevant to BD.
Collapse
|
64
|
Cholesterol contributes to dopamine-neuronal loss in MPTP mouse model of Parkinson's disease: Involvement of mitochondrial dysfunctions and oxidative stress. PLoS One 2017; 12:e0171285. [PMID: 28170429 PMCID: PMC5295696 DOI: 10.1371/journal.pone.0171285] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/19/2017] [Indexed: 01/24/2023] Open
Abstract
Hypercholesterolemia is a known contributor to the pathogenesis of Alzheimer’s disease while its role in the occurrence of Parkinson’s disease (PD) is only conjecture and far from conclusive. Altered antioxidant homeostasis and mitochondrial functions are the key mechanisms in loss of dopaminergic neurons in the substantia nigra (SN) region of the midbrain in PD. Hypercholesterolemia is reported to cause oxidative stress and mitochondrial dysfunctions in the cortex and hippocampus regions of the brain in rodents. However, the impact of hypercholesterolemia on the midbrain dopaminergic neurons in animal models of PD remains elusive. We tested the hypothesis that hypercholesterolemia in MPTP model of PD would potentiate dopaminergic neuron loss in SN by disrupting mitochondrial functions and antioxidant homeostasis. It is evident from the present study that hypercholesterolemia in naïve animals caused dopamine neuronal loss in SN with subsequent reduction in striatal dopamine levels producing motor impairment. Moreover, in the MPTP model of PD, hypercholesterolemia exacerbated MPTP-induced reduction of striatal dopamine as well as dopaminergic neurons in SN with motor behavioral depreciation. Activity of mitochondrial complexes, mainly complex-I and III, was impaired severely in the nigrostriatal pathway of hypercholesterolemic animals treated with MPTP. Hypercholesterolemia caused oxidative stress in the nigrostriatal pathway with increased generation of hydroxyl radicals and enhanced activity of antioxidant enzymes, which were further aggravated in the hypercholesterolemic mice with Parkinsonism. In conclusion, our findings provide evidence of increased vulnerability of the midbrain dopaminergic neurons in PD with hypercholesterolemia.
Collapse
|
65
|
De Pablo-Fernández E, Breen DP, Bouloux PM, Barker RA, Foltynie T, Warner TT. Neuroendocrine abnormalities in Parkinson's disease. J Neurol Neurosurg Psychiatry 2017; 88:176-185. [PMID: 27799297 DOI: 10.1136/jnnp-2016-314601] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/06/2016] [Accepted: 10/13/2016] [Indexed: 12/20/2022]
Abstract
Neuroendocrine abnormalities are common in Parkinson's disease (PD) and include disruption of melatonin secretion, disturbances of glucose, insulin resistance and bone metabolism, and body weight changes. They have been associated with multiple non-motor symptoms in PD and have important clinical consequences, including therapeutics. Some of the underlying mechanisms have been implicated in the pathogenesis of PD and represent promising targets for the development of disease biomarkers and neuroprotective therapies. In this systems-based review, we describe clinically relevant neuroendocrine abnormalities in Parkinson's disease to highlight their role in overall phenotype. We discuss pathophysiological mechanisms, clinical implications, and pharmacological and non-pharmacological interventions based on the current evidence. We also review recent advances in the field, focusing on the potential targets for development of neuroprotective drugs in Parkinson's disease and suggest future areas for research.
Collapse
Affiliation(s)
- Eduardo De Pablo-Fernández
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, London, UK
| | - David P Breen
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Pierre M Bouloux
- Centre for Neuroendocrinology, Royal Free Campus, UCL Institute of Neurology, London, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology, London, UK
| | - Thomas T Warner
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, London, UK
| |
Collapse
|
66
|
Ascherio A, Schwarzschild MA. The epidemiology of Parkinson's disease: risk factors and prevention. Lancet Neurol 2016; 15:1257-1272. [PMID: 27751556 DOI: 10.1016/s1474-4422(16)30230-7] [Citation(s) in RCA: 1202] [Impact Index Per Article: 133.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022]
Abstract
Since 2006, several longitudinal studies have assessed environmental or behavioural factors that seem to modify the risk of developing Parkinson's disease. Increased risk of Parkinson's disease has been associated with exposure to pesticides, consumption of dairy products, history of melanoma, and traumatic brain injury, whereas a reduced risk has been reported in association with smoking, caffeine consumption, higher serum urate concentrations, physical activity, and use of ibuprofen and other common medications. Randomised trials are investigating the possibility that some of the negative risk factors might be neuroprotective and thus beneficial in individuals with early Parkinson's disease, particularly with respect to smoking (nicotine), caffeine, and urate. In the future, it might be possible to identify Parkinson's disease in its prodromal phase and to promote neuroprotective interventions before the onset of motor symptoms. At this time, however, the only intervention that seems justifiable for the primary prevention of Parkinson's disease is the promotion of physical activity, which is likely to be beneficial for the prevention of several chronic diseases.
Collapse
Affiliation(s)
- Alberto Ascherio
- Departments of Epidemiology and Nutrition, Harvard T H Chan School of Public Health, Boston, MA, USA; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
67
|
Raider K, Ma D, Harris JL, Fuentes I, Rogers RS, Wheatley JL, Geiger PC, Yeh HW, Choi IY, Brooks WM, Stanford JA. A high fat diet alters metabolic and bioenergetic function in the brain: A magnetic resonance spectroscopy study. Neurochem Int 2016; 97:172-80. [PMID: 27125544 PMCID: PMC4900919 DOI: 10.1016/j.neuint.2016.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 11/18/2022]
Abstract
Diet-induced obesity and associated metabolic effects can lead to neurological dysfunction and increase the risk of developing Alzheimer's disease (AD) and Parkinson's disease (PD). Despite these risks, the effects of a high-fat diet on the central nervous system are not well understood. To better understand the mechanisms underlying the effects of high fat consumption on brain regions affected by AD and PD, we used proton magnetic resonance spectroscopy ((1)H-MRS) to measure neurochemicals in the hippocampus and striatum of rats fed a high fat diet vs. normal low fat chow. We detected lower concentrations of total creatine (tCr) and a lower glutamate-to-glutamine ratio in the hippocampus of high fat rats. Additional effects observed in the hippocampus of high fat rats included higher N-acetylaspartylglutamic acid (NAAG), and lower myo-inositol (mIns) and serine (Ser) concentrations. Post-mortem tissue analyses revealed lower phosphorylated AMP-activated protein kinase (pAMPK) in the striatum but not in the hippocampus of high fat rats. Hippocampal pAMPK levels correlated significantly with tCr, aspartate (Asp), phosphoethanolamine (PE), and taurine (Tau), indicating beneficial effects of AMPK activation on brain metabolic and energetic function, membrane turnover, and edema. A negative correlation between pAMPK and glucose (Glc) indicates a detrimental effect of brain Glc on cellular energy response. Overall, these changes indicate alterations in neurotransmission and in metabolic and bioenergetic function in the hippocampus and in the striatum of rats fed a high fat diet.
Collapse
Affiliation(s)
- Kayla Raider
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Delin Ma
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Janna L Harris
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Isabella Fuentes
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Robert S Rogers
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joshua L Wheatley
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paige C Geiger
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hung-Wen Yeh
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - In-Young Choi
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - William M Brooks
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS 66160, USA; Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John A Stanford
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
68
|
Wei ZB, Yuan YF, Jaouen F, Ma MS, Hao CJ, Zhang Z, Chen Q, Yuan Z, Yu L, Beurrier C, Li W. SLC35D3 increases autophagic activity in midbrain dopaminergic neurons by enhancing BECN1-ATG14-PIK3C3 complex formation. Autophagy 2016; 12:1168-79. [PMID: 27171858 DOI: 10.1080/15548627.2016.1179402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Searching for new regulators of autophagy involved in selective dopaminergic (DA) neuron loss is a hallmark in the pathogenesis of Parkinson disease (PD). We here report that an endoplasmic reticulum (ER)-associated transmembrane protein SLC35D3 is selectively expressed in subsets of midbrain DA neurons in about 10% TH (tyrosine hydroxylase)-positive neurons in the substantia nigra pars compacta (SNc) and in about 22% TH-positive neurons in the ventral tegmental area (VTA). Loss of SLC35D3 in ros (roswell mutant) mice showed a reduction of 11.9% DA neurons in the SNc and 15.5% DA neuron loss in the VTA with impaired autophagy. We determined that SLC35D3 enhanced the formation of the BECN1-ATG14-PIK3C3 complex to induce autophagy. These results suggest that SLC35D3 is a new regulator of tissue-specific autophagy and plays an important role in the increased autophagic activity required for the survival of subsets of DA neurons.
Collapse
Affiliation(s)
- Zong-Bo Wei
- a State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Ye-Feng Yuan
- a State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Florence Jaouen
- c Aix-Marseille University, Center National de la Recherche Scientifique , UMR 7288 , Institut de Biologie du Développement de Marseille, Marseille , France
| | - Mei-Sheng Ma
- d School of Life Sciences, Tsinghua University , Beijing , China
| | - Chan-Juan Hao
- a State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China.,e Center for Medical Genetics, Beijing Children's Hospital, Capital Medical University, Beijing Pediatric Research Institute , Beijing , China
| | - Zhe Zhang
- a State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| | - Quan Chen
- f Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Zengqiang Yuan
- g Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Li Yu
- d School of Life Sciences, Tsinghua University , Beijing , China
| | - Corinne Beurrier
- c Aix-Marseille University, Center National de la Recherche Scientifique , UMR 7288 , Institut de Biologie du Développement de Marseille, Marseille , France
| | - Wei Li
- a State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China.,e Center for Medical Genetics, Beijing Children's Hospital, Capital Medical University, Beijing Pediatric Research Institute , Beijing , China.,h Center of Alzheimer Disease, Beijing Institute for Brain Disorders , Beijing , China
| |
Collapse
|
69
|
Ungprasert P, Srivali N, Kittanamongkolchai W. Risk of Parkinson's Disease Among Patients with Psoriasis: A Systematic Review and Meta-analysis. Indian J Dermatol 2016; 61:152-6. [PMID: 27057013 PMCID: PMC4817438 DOI: 10.4103/0019-5154.177771] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background: Patients with psoriasis might be at a higher risk of developing Parkinson's disease (PD) as a result of the detrimental effect of chronic inflammation on the neuronal tissue. This meta-analysis aimed to investigate this risk by comprehensively reviewing all available data. Methods: We conducted a systematic review and meta-analysis of cohort and case–control studies that reported relative risk, hazard ratio, odds ratio, or standardized incidence ratio comparing the risk of PD in patients with psoriasis versus subjects without psoriasis. Pooled risk ratio and 95% confidence interval (CI) were calculated using random-effect, generic inverse variance methods of DerSimonian and Laird. Results: Three retrospective studies and one case–control study met our eligibility criteria and were included in this meta-analysis. The pooled risk ratio of PD in patients with psoriasis versus participants without psoriasis was 1.38 (95% CI, 1.15–1.66). The statistical heterogeneity was low with an I2 of 35%. Conclusions: Our meta-analysis demonstrated a statistically significant increased risk of PD among patients with psoriasis.
Collapse
Affiliation(s)
- Patompong Ungprasert
- Department of Internal Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Narat Srivali
- Department of Internal Medicine, Division of Pulmonology and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
70
|
Libertini G, Ferrara N. Aging of perennial cells and organ parts according to the programmed aging paradigm. AGE (DORDRECHT, NETHERLANDS) 2016; 38:35. [PMID: 26957493 PMCID: PMC5005898 DOI: 10.1007/s11357-016-9895-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 02/22/2016] [Indexed: 06/05/2023]
Abstract
If aging is a physiological phenomenon-as maintained by the programmed aging paradigm-it must be caused by specific genetically determined and regulated mechanisms, which must be confirmed by evidence. Within the programmed aging paradigm, a complete proposal starts from the observation that cells, tissues, and organs show continuous turnover: As telomere shortening determines both limits to cell replication and a progressive impairment of cellular functions, a progressive decline in age-related fitness decline (i.e., aging) is a clear consequence. Against this hypothesis, a critic might argue that there are cells (most types of neurons) and organ parts (crystalline core and tooth enamel) that have no turnover and are subject to wear or manifest alterations similar to those of cells with turnover. In this review, it is shown how cell types without turnover appear to be strictly dependent on cells subjected to turnover. The loss or weakening of the functions fulfilled by these cells with turnover, due to telomere shortening and turnover slowing, compromises the vitality of the served cells without turnover. This determines well-known clinical manifestations, which in their early forms are described as distinct diseases (e.g., Alzheimer's disease, Parkinson's disease, age-related macular degeneration, etc.). Moreover, for the two organ parts (crystalline core and tooth enamel) without viable cells or any cell turnover, it is discussed how this is entirely compatible with the programmed aging paradigm.
Collapse
Affiliation(s)
- Giacinto Libertini
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| | - Nicola Ferrara
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
71
|
Wills AMA, Pérez A, Wang J, Su X, Morgan J, Rajan SS, Leehey MA, Pontone GM, Chou KL, Umeh C, Mari Z, Boyd J. Association Between Change in Body Mass Index, Unified Parkinson's Disease Rating Scale Scores, and Survival Among Persons With Parkinson Disease: Secondary Analysis of Longitudinal Data From NINDS Exploratory Trials in Parkinson Disease Long-term Study 1. JAMA Neurol 2016; 73:321-8. [PMID: 26751506 PMCID: PMC5469290 DOI: 10.1001/jamaneurol.2015.4265] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Greater body mass index (BMI, calculated as weight in kilograms divided by height in meters squared) is associated with improved survival among persons with Huntington disease or amyotrophic lateral sclerosis. Weight loss is common among persons with Parkinson disease (PD) and is associated with worse quality of life. OBJECTIVE To explore the association between change in BMI, Unified Parkinson's Disease Rating Scale (UPDRS) motor and total scores, and survival among persons with PD and to test whether there is a positive association between BMI at randomization and survival. DESIGN, SETTING, AND PARTICIPANTS Secondary analysis (from May 27, 2014, to October 13, 2015) of longitudinal data (3-6 years) from 1673 participants who started the National Institute of Neurological Disorders and Stroke Exploratory Trials in PD Long-term Study-1 (NET-PD LS-1). This was a double-blind randomized placebo-controlled clinical trial of creatine monohydrate (10 g/d) that was performed at 45 sites throughout the United States and Canada. Participants with early (within 5 years of diagnosis) and treated (receiving dopaminergic therapy) PD were enrolled from March 2007 to May 2010 and followed up until September 2013. MAIN OUTCOMES AND MEASURES Change across time in motor UPDRS score, change across time in total UPDRS score, and time to death. Generalized linear mixed models were used to estimate the effect of BMI on the change in motor and total UPDRS scores after controlling for covariates. Survival was analyzed using Cox proportional hazards models of time to death. A participant's BMI was measured at randomization, and BMI trajectory groups were classified according to whether participants experienced weight loss ("decreasing BMI"), weight stability ("stable BMI"), or weight gain ("increasing BMI") during the study. RESULTS Of the 1673 participants (mean [SD] age, 61.7 [9.6] years; 1074 [64.2%] were male), 158 (9.4%) experienced weight loss (decreasing BMI), whereas 233 (13.9%) experienced weight gain (increasing BMI). After adjusting for covariates, we found that the weight-loss group's mean (SE) motor UPDRS score increased by 1.48 (0.28) (P < .001) more points per visit than the weight-stable group's mean (SE) motor UPDRS score. The weight-gain group's mean (SE) motor UPDRS score decreased by -0.51 (0.24) (P = .03) points per visit, relative to the weight-stable group. While there was an unadjusted difference in survival between the 3 BMI trajectory groups (log-rank P < .001), this was not significant after adjusting for covariates. CONCLUSIONS AND RELEVANCE Change in BMI was inversely associated with change in motor and total UPDRS scores in the NET-PD LS-1. Change in BMI was not associated with survival; however, these results were limited by the low number of deaths in the NET-PD LS-1. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00449865.
Collapse
Affiliation(s)
- Anne-Marie A Wills
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Adriana Pérez
- Department of Biostatistics, The University of Texas Health Science Center at Houston UTHealth, School of Public Health, Austin
| | - Jue Wang
- UTHealth, The University of Texas School of Public Health, Houston
| | - Xiao Su
- UTHealth, The University of Texas School of Public Health, Houston
| | - John Morgan
- Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta
| | - Suja S Rajan
- Department of Management, Policy and Community Health, The University of Texas Health Science Center at Houston UTHealth, School of Public Health, Houston
| | - Maureen A Leehey
- Department of Neurology, University of Colorado Hospital and University of Colorado School of Medicine, Aurora
| | - Gregory M Pontone
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Kelvin L Chou
- Departments of Neurology and Neurosurgery, University of Michigan, Ann Arbor
| | - Chizoba Umeh
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zoltan Mari
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - James Boyd
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington
| |
Collapse
|
72
|
Lee JJ, Oh JS, Ham JH, Lee DH, Lee I, Sohn YH, Kim JS, Lee PH. Association of body mass index and the depletion of nigrostriatal dopamine in Parkinson's disease. Neurobiol Aging 2016; 38:197-204. [DOI: 10.1016/j.neurobiolaging.2015.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/17/2015] [Accepted: 11/17/2015] [Indexed: 11/16/2022]
|
73
|
Csoti I, Jost WH, Reichmann H. Parkinson's disease between internal medicine and neurology. J Neural Transm (Vienna) 2016; 123:3-17. [PMID: 26298728 PMCID: PMC4713462 DOI: 10.1007/s00702-015-1443-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 08/10/2015] [Indexed: 02/07/2023]
Abstract
General medical problems and complications have a major impact on the quality of life in all stages of Parkinson's disease. To introduce an effective treatment, a comprehensive analysis of the various clinical symptoms must be undertaken. One must distinguish between (1) diseases which arise independently of Parkinson's disease, and (2) diseases which are a direct or indirect consequence of Parkinson's disease. Medical comorbidity may induce additional limitations to physical strength and coping strategies, and may thus restrict the efficacy of the physical therapy which is essential for treating hypokinetic-rigid symptoms. In selecting the appropriate medication for the treatment of any additional medical symptoms, which may arise, its limitations, contraindications and interactions with dopaminergic substances have to be taken into consideration. General medical symptoms and organ manifestations may also arise as a direct consequence of the autonomic dysfunction associated with Parkinson's disease. As the disease progresses, additional non-parkinsonian symptoms can be of concern. Furthermore, the side effects of Parkinson medications may necessitate the involvement of other medical specialists. In this review, we will discuss the various general medical aspects of Parkinson's disease.
Collapse
Affiliation(s)
- Ilona Csoti
- Gertrudis-Clinic Parkinson-Center, Karl-Ferdinand-Broll-Str. 2-4, 35638, Leun, Germany.
| | - Wolfgang H Jost
- Parkinson-Klinik Wolfach, Kreuzbergstr.12-24, 77709, Wolfach, Germany.
| | - Heinz Reichmann
- Department of Neurology, University of Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| |
Collapse
|
74
|
Hargrave SL, Davidson TL, Zheng W, Kinzig KP. Western diets induce blood-brain barrier leakage and alter spatial strategies in rats. Behav Neurosci 2015; 130:123-35. [PMID: 26595878 DOI: 10.1037/bne0000110] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Western diet (WD) intake induces obesity and metabolic dysfunction. The present study examined the effects of WD on hippocampal-dependent cognitive functioning and blood-brain barrier (BBB) permeability as a function of exposure duration, obesity phenotype, and peripheral markers of energy regulation. The use of hippocampal-dependent "place" or hippocampal-independent "response" strategies in a Y maze was assessed in male rats following 10, 40, and 90 days of WD exposure in diet-induced obese (DIO) rats, in diet resistant (DR) rats that are relatively insensitive to the obesogenic properties of WD, and in chow-fed controls. Insulin, glucose, and BBB permeability throughout several loci in the hippocampus, striatum, and cerebellum were evaluated in relation to duration of WD exposure, obesity phenotype, and type of strategy used. DIO rats had increased body weight and adiposity throughout the study, and elevated 10-day glucose and 90-day insulin levels. Throughout the study, chow-fed and DR rats reliably relied on a place strategy. DIO rats, in contrast, favored a response strategy at the 10- and 90-day time points. BBB leakage was observed in the dorsal striatum and multiple subregions of the hippocampus of DIO, but not DR or chow-fed rats. Increased ventral hippocampal BBB permeability and blood glucose levels were associated with reduced place strategy use. These data indicate that WD-induced BBB leakage is dependent on duration of diet exposure as well as obesity phenotype, and implicates BBB leakage and impaired glucoregulation in behavioral strategy and cognitive performance.
Collapse
Affiliation(s)
| | | | - Wei Zheng
- School of Health Sciences, Purdue University
| | | |
Collapse
|
75
|
Sääksjärvi K, Knekt P, Männistö S, Lyytinen J, Heliövaara M. Prospective study on the components of metabolic syndrome and the incidence of Parkinson's disease. Parkinsonism Relat Disord 2015; 21:1148-55. [DOI: 10.1016/j.parkreldis.2015.07.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/26/2015] [Accepted: 07/20/2015] [Indexed: 10/23/2022]
|
76
|
Paul R, Choudhury A, Borah A. Cholesterol - A putative endogenous contributor towards Parkinson's disease. Neurochem Int 2015; 90:125-33. [PMID: 26232622 DOI: 10.1016/j.neuint.2015.07.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 07/25/2015] [Accepted: 07/27/2015] [Indexed: 01/20/2023]
Abstract
Elevated levels of cholesterol and its metabolites (oxysterols) have been reported to be associated not only with several metabolic syndromes, but also become a prognostic risk factor of neurodegenerative diseases particularly Alzheimer's disease. The incidence and the prospect of Alzheimer's disease with respect to elevated levels of cholesterol have been studied extensively and reviewed earlier. Recently, several interesting findings have shown the occurrence of equivalent Parkinsonian pathologies in cellular neuronal models, mediated by oxysterols or excess exposure to cholesterol. In this regard, oxysterols are particular in causing alpha-synuclein aggregation and destruction of dopamine containing neurons in in vitro models, which is linked to their direct influence on oxidative stress provoking potency. Inspite of the significant in vitro reports, which suggest the relativeness of cholesterol or oxysterol towards Parkinsonism, several prospective clinical reports provided a negative or no correlation. However, few prospective clinical studies showed a positive correlation between plasma cholesterol and incidence of Parkinson's disease (PD). Also, few significant studies have convincingly demonstrated that high fat diet exacerbates parkinsonian pathologies, including loss of dopaminergic neurons and oxidative stress parameters in animal models of PD. The present review brings together all the neuropathological proceedings mediated by excess cholesterol or its metabolites in brain in the light of their contribution towards the onset of PD. Also we have reviewed the possibilities of cholesterol lowering efficacy of statin therapy, in reducing the occurrence of PD.
Collapse
Affiliation(s)
- Rajib Paul
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Amarendranath Choudhury
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India.
| |
Collapse
|
77
|
Håglin L. High serum phosphate concentration as the result of smoking might underlie the lower risk of Parkinson's disease. Med Hypotheses 2015. [PMID: 26206759 DOI: 10.1016/j.mehy.2015.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Epidemiological studies have found a negative association between cigarette smoking and Parkinson's disease (PD): PD patients are approximately 50% less likely to have smoked cigarettes than age- and sex-matched controls. In both women and men, the PD protection effect of smoking may be explained by higher levels of phosphate in serum (S-P) and triglycerides (S-TG) in smokers compared to non-smokers. That is, the protecting effect from smoking could be mediated by either a high S-P or high S-TG levels. I suggest that higher S-P as the result of smoking may be associated with intracellular depletion of Pi in skeletal muscle and that this depletion of Pi is associated with increased availability of phosphate for the brain. This increased phosphate availability would protect against PD, as oxidative phosphorylation in the mitochondria is a central and persistent phenomenon in the pathogenesis cascade of PD. Phosphate is necessary for energy production in the form of creatine phosphate (CP) and adenosine-tri-phosphate (ATP) in the brain and skeletal muscle. As such, hypophosphatemia increases risk of cell death. In some clinical instances, this energy depletion may pre-dispose to dopamine neuron death. Mitochondrial dysfunction is associated with the generation of oxidative stress and mediates the induction of apoptosis. Studies with NMR spectroscopy reveal that an energy deficit in brain cells is a strong mark for PD although this differed for men and women. Compared to women, men had lower serum phosphate and ATP levels in the brain (about 15% lower). In addition to sex differences, age, stress, and malnutrition may cause low serum phosphate levels, a situation that could contribute to the lack of energy available to the brain and the risk of PD. As hypophosphatemia is present in overnutrition and has an inverse relation with a high BMI, both obesity and malnutrition are considered to be presumptive risk factor for PD.
Collapse
Affiliation(s)
- L Håglin
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Sweden.
| |
Collapse
|
78
|
Brauer R, Bhaskaran K, Chaturvedi N, Dexter DT, Smeeth L, Douglas I. Glitazone Treatment and Incidence of Parkinson's Disease among People with Diabetes: A Retrospective Cohort Study. PLoS Med 2015; 12:e1001854. [PMID: 26196151 PMCID: PMC4511413 DOI: 10.1371/journal.pmed.1001854] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/12/2015] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Recent in vitro and animal experiments suggest that peroxisome proliferation-activated receptor gamma (PPARɣ) agonist medications, such as antidiabetic glitazone (GTZ) drugs, are neuroprotective in models of Parkinson's disease (PD). These findings have not been tested in humans. We hypothesized that individuals prescribed GTZ drugs would have a lower incidence of PD compared to individuals prescribed other treatments for diabetes. METHODS AND FINDINGS Using primary care data from the United Kingdom Clinical Practice Research Datalink (CPRD), we conducted a retrospective cohort study in which individuals with diabetes who were newly prescribed GTZ (GTZ-exposed group) were matched by age, sex, practice, and diabetes treatment stage with up to five individuals prescribed other diabetes treatments (other antidiabetic drug-exposed group). Patients were followed up from 1999 until the first recording of a PD diagnosis, end of observation in the database, or end of the study (1 August 2013). An incidence rate ratio (IRR) was calculated using conditional Poisson regression, adjusted for possible confounders. 44,597 GTZ exposed individuals were matched to 120,373 other antidiabetic users. 175 GTZ-exposed individuals were diagnosed with PD compared to 517 individuals in the other antidiabetic drug-exposed group. The incidence rate (IR) of PD in the GTZ-exposed group was 6.4 per 10,000 patient years compared with 8.8 per 10,000 patient years in those prescribed other antidiabetic treatments (IRR 0.72, 95% confidence interval [CI] 0.60-0.87). Adjustments for potential confounding variables, including smoking, other medications, head injury, and disease severity, had no material impact (fully adjusted IRR 0.75, 0.59-0.94). The risk was reduced in those with current GTZ prescriptions (current GTZ-exposed IRR 0.59, 0.46-0.77) but not reduced among those with past prescriptions (past GTZ-exposed IRR 0.85, 0.65-1.10). Our study only included patients with diabetes who did not have a PD diagnosis when they were first prescribed GTZ, and thus, it cannot establish whether GTZ use prevents or slows the progression of PD. CONCLUSIONS In patients with diabetes, a current prescription for GTZ is associated with a reduction in incidence of PD. This suggests PPAR gamma pathways may be a fruitful drug target in PD.
Collapse
Affiliation(s)
- Ruth Brauer
- Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Krishnan Bhaskaran
- Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Nishi Chaturvedi
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
| | - David T. Dexter
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Liam Smeeth
- Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ian Douglas
- Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
79
|
Wang YL, Wang YT, Li JF, Zhang YZ, Yin HL, Han B. Body Mass Index and Risk of Parkinson's Disease: A Dose-Response Meta-Analysis of Prospective Studies. PLoS One 2015; 10:e0131778. [PMID: 26121579 PMCID: PMC4488297 DOI: 10.1371/journal.pone.0131778] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/06/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND A number of epidemiologic studies examining the relationship between body mass index (BMI) and the future occurrence of Parkinson's disease (PD) reported largely inconsistent findings. We conducted a dose-response meta-analysis of prospective studies to clarify this association. METHODS Eligible prospective studies were identified by a search of PubMed and by checking the references of related publications. The generalized least squares trend estimation was employed to compute study-specific relative risks (RR) and 95% confidence intervals (CI) for an increase in BMI of 5 kg/m2, and the random-effects model was used to compute summary RR and 95% CI. RESULTS A total of 10 prospective studies were included in the final analysis. An increase in BMI of 5 kg/m2 was not associated with PD risk, with a summary RR of 1.00 (95% CI = 0.89-1.12). Results of subgroup analysis found similar results except for a week positive association in studies that adjusted for alcohol consumption (RR = 1.13, 95% CI = 0.99-1.29), and a week inverse association in studies that did not (RR = 0.90, 95% CI = 0.78-1.04). In a separate meta-analysis, no significant association between overweight (25 kg/m2 ≤ BMI ≤29.9 kg/m2), obesity (BMI≥30 kg/m2) or excess weight (BMI≥25 kg/m2) and PD risk was observed. CONCLUSION This meta-analysis does not support the notion that higher BMI materially increases PD risk. However, a week positive BMI-PD association that may be masked by confounders still cannot be excluded, and future prospective studies with a good control for potential confounding factors are needed.
Collapse
Affiliation(s)
- Yun-Liang Wang
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, 255000, China
- * E-mail:
| | - Yu-Tong Wang
- School of Medicine, Henan University, Kaifeng, 475000, China
| | - Jin-Feng Li
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, 255000, China
| | - Yu-Zheng Zhang
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, 255000, China
| | - Hong-Lei Yin
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, 255000, China
| | - Bing Han
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, 255000, China
| |
Collapse
|
80
|
Deleidi M, Jäggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Front Neurosci 2015; 9:172. [PMID: 26089771 PMCID: PMC4453474 DOI: 10.3389/fnins.2015.00172] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
As we age, the immune system undergoes a process of senescence accompanied by the increased production of proinflammatory cytokines, a chronic subclinical condition named as “inflammaging”. Emerging evidence from human and experimental models suggest that immune senescence also affects the central nervous system and promotes neuronal dysfunction, especially within susceptible neuronal populations. In this review we discuss the potential role of immune aging, inflammation and metabolic derangement in neurological diseases. The discovery of novel therapeutic strategies targeting age-linked inflammation may promote healthy brain aging and the treatment of neurodegenerative as well as neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michela Deleidi
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Madeline Jäggle
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Graziella Rubino
- Department of Internal Medicine II, Center for Medical Research, University of Tübingen Tübingen, Germany
| |
Collapse
|
81
|
Lai CY, Chou MC, Lin CL, Kao CH. Increased risk of Parkinson disease in patients with carbon monoxide intoxication: a population-based cohort study. Medicine (Baltimore) 2015; 94:e869. [PMID: 25984676 PMCID: PMC4602584 DOI: 10.1097/md.0000000000000869] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The present study evaluated the association of carbon monoxide intoxication (COI) with Parkinson disease (PD).A total of 9012 adults newly diagnosed with COI were enrolled in this study as the COI cohort. The control (non-COI) cohort, comprising 36,048 participants, was matched for each COI patient according to age, sex, and the year of hospitalization. We calculated the hazard ratios (HR) and 95% confidence intervals by using a Cox proportional hazards regression model.The overall incidence of PD (per 10,000 person-year) in the COI and non-COI cohorts was 27.4 and 2.53, respectively. After adjustment for age, sex, and comorbidities, the COI patients exhibited a 9.08-fold increased risk for PD. The COI patients without comorbidity exhibited a significantly higher risk of PD (adjusted HR = 15.8) than did the COI patients without comorbidity (adjusted HR = 4.15). Patients with COI and receiving hyperbaric oxygen therapy exhibited a 14.3-fold increased risk of PD; the adjusted HR of patients who did not receive hyperbaric oxygen treatment was increased 7.97-fold.The risk of PD increased in the COI patients and the significance increased in young people. COI is a crucial factor leading to PD.
Collapse
Affiliation(s)
- Ching-Yuan Lai
- From the Department of Emergency Medicine, China Medical University Hospital, Taichung (C-YL); Department of Radiology, E-Da Hospital/I-Shou University, Kaohsiung (M-CC); Management Office for Health Data, China Medical University Hospital (C-LL); College of Medicine, China Medical University (C-LL); Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine, China Medical University (C-HK); Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan (C-HK)
| | | | | | | |
Collapse
|
82
|
Cardiovascular risk factors and the risk of Parkinson's disease. Eur J Clin Nutr 2014; 69:729-33. [PMID: 25514902 DOI: 10.1038/ejcn.2014.259] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 10/15/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND/OBJECTIVES The objective of this study was to investigate whether serum triglycerides (S-TG), cholesterol, blood pressure and waist/height ratio are risk factors for Parkinson's disease (PD). SUBJECTS AND METHODS A population-based sample within the Northern Sweden Health and Disease Study (NSHDS) was used in this study (n=101 790 subjects). Cases with PD were identified prospectively in a community-based study of idiopathic Parkinsonism in the period 2004-2009 in the county of Västerbotten in northern Sweden. The case database obtained was crosslinked to the NSHDS. Eighty-four of 147 patients with PD had visited the primary health care 2-8 years before diagnosis for participation in the NSHDS. For each case, four referents from the NSHDS population were selected, matched for sex, age, year of health survey, subcohort and geographic area. RESULTS Cases had lower mean S-TG levels (P=0.007). After stratification for sex, the lower S-TG remained significant for men (P=0.006) but not for women (P=0.450), and these were confirmed by the conditional logistic regression for all cases, none adjusted (hazard ratio (HR): 0.65; 95% confidence interval (CI): 0.42, 0.99) and after adjusting for age, body mass index (BMI) and physical activity (HR: 0.61; 95% CI: 0.39, 0.96). Systolic blood pressure (SBP) was negatively associated with PD risk after adjustments for age, BMI and physical activity (HR: 0.98; 95% CI: 0.97-0.99). Smoking and former smoking were associated with a reduced risk for PD. CONCLUSIONS We found lower S-TG and SBP 2-8 years before a diagnosis of PD. Smoking was confirmed to be negatively associated with PD, whereas recreational activity indicates a risk for women.
Collapse
|
83
|
Yang F, Trolle Lagerros Y, Bellocco R, Adami HO, Fang F, Pedersen NL, Wirdefeldt K. Physical activity and risk of Parkinson's disease in the Swedish National March Cohort. ACTA ACUST UNITED AC 2014; 138:269-75. [PMID: 25410713 DOI: 10.1093/brain/awu323] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Physical exercise has been associated with neuroprotective effects in the nigrostriatal dopaminergic system. To examine the impact of physical activity on Parkinson's disease risk prospectively, we followed 43 368 individuals who provided extensive information on physical activity at baseline. We estimated hazard ratios with 95% confidence intervals using Cox proportional hazards regression. During an average of 12.6 years of follow-up, 286 incident Parkinson's disease cases were identified. In males, there was an inverse association with Parkinson's disease for total physical activity (hazard ratio 0.55, 95% confidence interval 0.35-0.87 for medium versus low level), for sum of household, commuting and leisure time exercise (hazard ratio 0.53, 95% confidence interval 0.33-0.85 for high versus low level), and for household and commuting physical activity specifically (hazard ratio 0.50, 95% confidence interval 0.31-0.81 for >6 versus <2 h per week). No association was observed for leisure time exercise or occupational physical activity with Parkinson's disease, among either males or females. Meta-analysis of the present study and five previous prospective studies showed a pooled hazard ratio of 0.66 (95% confidence interval 0.57-0.78) for highest versus lowest physical activity level. Our results indicate that a medium level of physical activity lowers Parkinson's disease risk.
Collapse
Affiliation(s)
- Fei Yang
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ylva Trolle Lagerros
- 2 Department of Medicine, Unit of Clinical Epidemiology, Karolinska Institutet, T2, 17176 Stockholm, Sweden
| | - Rino Bellocco
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 3 Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Hans-Olov Adami
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 4 Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
| | - Fang Fang
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Nancy L Pedersen
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 5 Department of Psychology, University of Southern California, Los Angeles, CA 90089-1061, USA
| | - Karin Wirdefeldt
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 6 Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
84
|
Xie B, Lin F, Peng L, Ullah K, Wu H, Qing H, Deng Y. Methylglyoxal increases dopamine level and leads to oxidative stress in SH-SY5Y cells. Acta Biochim Biophys Sin (Shanghai) 2014; 46:950-6. [PMID: 25274329 DOI: 10.1093/abbs/gmu094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
More and more studies have suggested that methylglyoxal (MGO) induced by type-2 diabetes is related to Parkinson's disease (PD). However, little is known about the molecular mechanism. In this study, we explored the MGO toxicity in neuroblastoma SH-SY5Y cells. Neurotoxicity of MGO was measured by mitochondrial membrane potential, malondialdehyde, and methylthiazoletetrazolium assays. The levels of dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC), and 1-methyl-4-phenyl-1,2,3,4-tetrahydroisoquinoline (salsolinol) were detected by liquid chromatography-mass spectrometry/mass spectrometry. The expressions of tyrosine hydroxylase (TH) and dopamine transporter (DAT) were detected by reverse transcriptase polymerase chain reaction and western blot analysis. The results showed that MGO induced an increase in TH and DAT expressions in SH-SY5Y neuroblastoma cells, while the levels of dopamine, DOPAC, and endogenous neurotoxin salsolinol also increased. Aminoguanidine (AG) is an inhibitor of MGO. It was found that AG could decrease the reactive oxygen species (ROS) level induced by MGO, but could not inhibit an increase of TH, DAT and dopamine. The increase of dopamine, DOPAC and salsolinol levels could lead to high ROS and mitochondrial damage. This study suggests that ROS caused by dopamine could contribute to the damage of dopaminergic neurons when MGO is increased during the course of diabetes.
Collapse
Affiliation(s)
- Bingjie Xie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Fankai Lin
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Lei Peng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Kaleem Ullah
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hanyan Wu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
85
|
Spielman LJ, Little JP, Klegeris A. Inflammation and insulin/IGF-1 resistance as the possible link between obesity and neurodegeneration. J Neuroimmunol 2014; 273:8-21. [PMID: 24969117 DOI: 10.1016/j.jneuroim.2014.06.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 06/03/2014] [Accepted: 06/04/2014] [Indexed: 12/17/2022]
Abstract
Obesity is a growing epidemic that contributes to several brain disorders including Alzheimer's, Parkinson's, and Huntington's diseases. Obesity could promote these diseases through several different mechanisms. Here we review evidence supporting the involvement of two recently recognized factors linking obesity with neurodegeneration: the induction of pro-inflammatory cytokines and onset of insulin and insulin-like growth factor 1 (IGF-1) resistance. Excess peripheral pro-inflammatory mediators, some of which can cross the blood brain barrier, may trigger neuroinflammation, which subsequently exacerbates neurodegeneration. Insulin and IGF-1 resistance leads to weakening of neuroprotective signaling by these molecules and can contribute to onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lindsay J Spielman
- Department of Biology, University of British Columbia Okanagan Campus, 3333 University Way, Kelowna, BC, V1V 1V7 Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan Campus, 3333 University Way, Kelowna, BC, V1V 1V7 Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, 3333 University Way, Kelowna, BC, V1V 1V7 Canada.
| |
Collapse
|
86
|
Skow A, Douglas I, Smeeth L. The association between Parkinson's disease and anti-epilepsy drug carbamazepine: a case-control study using the UK General Practice Research Database. Br J Clin Pharmacol 2014; 76:816-22. [PMID: 23432592 DOI: 10.1111/bcp.12100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 02/12/2013] [Indexed: 01/14/2023] Open
Abstract
AIMS To investigate whether the use of carbamazepine is associated with reduced risk of Parkinson's disease. METHODS We conducted a population-based, matched case-control study of patients randomly selected from the UK General Research Practice Database. We identified 8549 patients with Parkinson's disease using diagnosis criteria with a positive predictive value of 90%. These patients were compared with 42, 160 control subjects matched for age, sex and general practice. RESULTS Overall, 3.0% of cases (257 of 8549) had at least one recorded prescription for carbamazepine compared with 2.5% (1050 of 42, 160) of controls. The crude odds ratio for the association between Parkinson's disease and carbamazepine was 1.22 (95% confidence interval 1.06-1.40), but this reduced to 0.93 (95% confidence interval 0.81-1.08, P = 0.34) after adjusting for annual consultation rate. Further adjustment for body mass index, smoking status, alcohol consumption or use of calcium channel blockers did not affect results. There was no evidence that risk decreased with higher doses or longer duration of carbamazepine use. CONCLUSIONS There was little to no evidence that use of carbamazepine is associated with reduced risk of Parkinson's disease. Although the study was underpowered, it does indicate that any effect of carbamazepine is likely to be small.
Collapse
Affiliation(s)
- Aine Skow
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | | | | |
Collapse
|
87
|
Kistner A, Lhommée E, Krack P. Mechanisms of body weight fluctuations in Parkinson's disease. Front Neurol 2014; 5:84. [PMID: 24917848 PMCID: PMC4040467 DOI: 10.3389/fneur.2014.00084] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 05/16/2014] [Indexed: 11/13/2022] Open
Abstract
Typical body weight changes are known to occur in Parkinson’s disease (PD). Weight loss has been reported in early stages as well as in advanced disease and malnutrition may worsen the clinical state of the patient. On the other hand, an increasing number of patients show weight gain under dopamine replacement therapy or after surgery. These weight changes are multifactorial and involve changes in energy expenditure, perturbation of homeostatic control, and eating behavior modulated by dopaminergic treatment. Comprehension of the different mechanisms contributing to body weight is a prerequisite for the management of body weight and nutritional state of an individual PD patient. This review summarizes the present knowledge and highlights the necessity of evaluation of body weight and related factors, as eating behavior, energy intake, and expenditure in PD.
Collapse
Affiliation(s)
- Andrea Kistner
- Movement Disorder Unit, Department of Psychiatry and Neurology, University Hospital Grenoble , Grenoble , France ; Unité 836, Équipe 11, INSERM, Grenoble Institut des Neurosciences , Grenoble , France
| | - Eugénie Lhommée
- Movement Disorder Unit, Department of Psychiatry and Neurology, University Hospital Grenoble , Grenoble , France ; Unité 836, Équipe 11, INSERM, Grenoble Institut des Neurosciences , Grenoble , France
| | - Paul Krack
- Movement Disorder Unit, Department of Psychiatry and Neurology, University Hospital Grenoble , Grenoble , France ; Unité 836, Équipe 11, INSERM, Grenoble Institut des Neurosciences , Grenoble , France ; Joseph Fourier University , Grenoble , France
| |
Collapse
|
88
|
Metabolic syndrome: an important risk factor for Parkinson's disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:729194. [PMID: 24955210 PMCID: PMC4052080 DOI: 10.1155/2014/729194] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/21/2014] [Indexed: 12/26/2022]
Abstract
Metabolic syndrome is becoming commoner due to a rise in obesity rates among adults. Generally speaking, a person with metabolic syndrome is twice as likely to develop cardiovascular disease and five times as likely to develop diabetes as someone without metabolic syndrome. Increasing oxidative stress in metabolic syndrome and Parkinson's disease is mentioned in the comprehensive articles; however, the system review about clear relation between metabolic syndrome and Parkinson's disease is deficient. In this review, we will focus on the analysis that the metabolic syndrome may be a risk factor for Parkinson's disease and the preventions that reduce the incident of Parkinson's disease by regulating the oxidative stress.
Collapse
|
89
|
Is elevated norepinephrine an etiological factor in some cases of Parkinson’s disease? Med Hypotheses 2014; 82:462-9. [DOI: 10.1016/j.mehy.2014.01.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/12/2014] [Accepted: 01/22/2014] [Indexed: 11/19/2022]
|
90
|
Reduced risk of Parkinson's disease associated with lower body mass index and heavy leisure-time physical activity. Eur J Epidemiol 2014; 29:285-92. [PMID: 24633681 DOI: 10.1007/s10654-014-9887-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
The risk factors for Parkinson's disease (PD) are not well established. We therefore examined the prediction of various lifestyle factors on the incidence of PD in a cohort drawn from the Finnish Mobile Clinic Health Examination Survey, conducted in 1973-1976. The study population comprised 6,715 men and women aged 50-79 years and free of PD at the baseline. All of the subjects completed a baseline health examination (including height and weight measurements) and a questionnaire providing information on leisure-time physical activity, smoking, and alcohol consumption. During a 22-year follow-up, 101 incident cases of PD occurred. The statistical analyses were based on Cox's model including age, sex, education, community density, occupation, coffee consumption, body mass index (BMI), leisure-time physical activity, smoking and alcohol consumption as independent variables. At first, BMI was not associated with PD risk, but after exclusion of the first 15 years of follow-up, an elevated risk appeared at higher BMI levels (P for trend 0.02). Furthermore, subjects with heavy leisure-time physical activity had a lower PD risk than those with no activity [relative risk (RR) 0.27, 95 % confidence interval (CI) 0.08-0.90]. In variance with findings for other chronic diseases, current smokers had a lower PD risk than those who had never smoked (RR 0.23, 95 % CI 0.08-0.67), and individuals with moderate alcohol intake (at the level of <5 g/day) had an elevated PD risk compared to non-drinkers. The results support the hypothesis that lifestyle factors predict the occurrence of Parkinson's disease, but more research is needed.
Collapse
|
91
|
Hosseini Tabatabaei N, Babakhani B, Hosseini Tabatabaei A, Vahabi Z, Soltanzadeh A. Non-genetic factors associated with the risk of Parkinson's disease in Iranian patients. FUNCTIONAL NEUROLOGY 2014; 28:107-13. [PMID: 24125560 DOI: 10.11138/fneur/2013.28.2.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of this study was to investigate some of nongenetic factors associated with Parkinson's disease (PD) in a sample of Iranian patients. This case-control study included 75 patients with idiopathic PD and 75 control patients. The patients in the control group were found to have drunk more glasses of tea per day than the case group before the onset of their problem (p=0.019). Every extra glass of tea per day decreased the risk of PD by 0.8 times (OR=0.8, 95%CI=0.73-0.97, p=0.02). Each cup of coffee per week decreased the chance of developing PD by 0.5 times (OR=0.5, 95%CI=0.28-0.9, p=0.021). A previous history of evening work increased PD risk by 4.6 times (OR=4.6, 95%CI=1.29-16.86, p=0.019) while major stressful events increased it by 13.5 times (OR=13.5, 95%CI=4.7-38.1, p=0.0001). In conclusion, coffee and tea consumption may exert a protective effect against PD, while evening work and stress may be risk factors for the development of the disease.
Collapse
|
92
|
Weise CM, Hohenadel MG, Krakoff J, Votruba SB. Body composition and energy expenditure predict ad-libitum food and macronutrient intake in humans. Int J Obes (Lond) 2014; 38:243-51. [PMID: 23736368 PMCID: PMC3909024 DOI: 10.1038/ijo.2013.85] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 04/15/2013] [Accepted: 05/08/2013] [Indexed: 11/09/2022]
Abstract
BACKGROUND Obesity is the result of chronic positive energy balance. The mechanisms underlying the regulation of energy homeostasis and food intake are not understood. Despite large increases in fat mass (FM), recent evidence indicates that fat-free mass (FFM) rather than FM is positively associated with intake in humans. METHODS In 184 humans (73 females/111 males; age 34.5±8.8 years; percentage body fat: 31.6±8.1%), we investigated the relationship of FFM index (FFMI, kg m(-2)), FM index (FMI, kg m(-2)); and 24-h energy expenditure (EE, n=127) with ad-libitum food intake using a 3-day vending machine paradigm. Mean daily calories (CAL) and macronutrient intake (PRO, CHO, FAT) were determined and used to calculate the relative caloric contribution of each (%PRO, %CHO, %FAT) and percent of caloric intake over weight maintaining energy needs (%WMENs). RESULTS FFMI was positively associated with CAL (P<0.0001), PRO (P=0.0001), CHO (P=0.0075) and FAT (P<0.0001). This remained significant after adjusting for FMI. Total EE predicted CAL and macronutrient intake (all P<0.0001). FMI was positively associated with CAL (P=0.019), PRO (P=0.025) and FAT (P=0.0008). In models with both FFMI and FMI, FMI was negatively associated with CAL (P=0.019) and PRO (P=0.033). Both FFMI and FMI were negatively associated with %CHO and positively associated with %FAT (all P<0.001). EE and FFMI (adjusted for FMI) were positively (EE P=0.0085; FFMI P=0.0018) and FMI negatively (P=0.0018; adjusted for FFMI) associated with %WMEN. CONCLUSION Food and macronutrient intake are predicted by FFMI and to a lesser degree by FMI. FFM and FM may have opposing effects on energy homeostasis.
Collapse
Affiliation(s)
- Christopher M Weise
- Obesity and Diabetes Clinical Research Section, NIDDK-NIH, DHHS, Phoenix, AZ, USA
| | | | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, NIDDK-NIH, DHHS, Phoenix, AZ, USA
| | - Susanne B Votruba
- Obesity and Diabetes Clinical Research Section, NIDDK-NIH, DHHS, Phoenix, AZ, USA
| |
Collapse
|
93
|
Truong DD, Wolters EC. Recognition and management of Parkinson’s disease during the premotor (prodromal) phase. Expert Rev Neurother 2014; 9:847-57. [DOI: 10.1586/ern.09.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
94
|
Chen J, Guan Z, Wang L, Song G, Ma B, Wang Y. Meta-analysis: overweight, obesity, and Parkinson's disease. Int J Endocrinol 2014; 2014:203930. [PMID: 24672544 PMCID: PMC3941583 DOI: 10.1155/2014/203930] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/07/2013] [Accepted: 12/09/2013] [Indexed: 11/17/2022] Open
Abstract
Objective. Parkinson's disease (PD) is a severe neurological disease and its risk factors remain largely unknown. A meta-analysis was carried out to investigate the relationship of overweight and obesity with PD. Methods. We used PubMed, EMBASE, and the Chinese National Knowledge Infrastructure (CNKI) databases to identify studies of associations between overweight/obesity and PD. Overweight, obesity, and PD were used as keywords, and published works were retrieved until September 30, 2013. The extracted data were classified (BMI ≥ 30, 25 ≤ BMI < 30, and BMI < 25) according to BMI values and analyzed using RevMan5.2 and Stata11.0. Results. Four cohort studies and three case-control studies were used to evaluate the association between overweight/obesity and PD, including 2857 PD patients and 5, 683, 939 cases of non-PD controls. There was a statistically significant difference between 25 ≤ BMI < 30 and BMI < 25 in the cohort study (RR = 1.17, 95% CI, 1.03-1.32, P = 0.03), but there was no difference between BMI ≥ 30 and BMI < 25 or BMI ≥ 30 and 25 ≤ BMI < 30, where the respective RR was 1.16 and 0.84; the respective 95% CI was 0.67-2.01 and 0.61-1.15, respectively, and the P values were 0.60 and 0.28, respectively. Case-control studies showed that there was no statistical difference between any two groups. Conclusion. Meta-analysis showed that overweight might be a potential risk factor of PD. Demonstration of a causal role of overweight/obesity in PD development could have important therapeutic implications.
Collapse
Affiliation(s)
- Jinhu Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang 050017, China
| | - Zhenlong Guan
- Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Liqin Wang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang 050000, China
| | - Guangyao Song
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang 050017, China
| | - Boqing Ma
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang 050017, China
| | - Yanqin Wang
- Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
- *Yanqin Wang:
| |
Collapse
|
95
|
Nutrition and Gastrointestinal Health as Modulators of Parkinson’s Disease. PHARMA-NUTRITION 2014. [DOI: 10.1007/978-3-319-06151-1_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
96
|
Abstract
Parkinson disease (PD) is a progressive, neurodegenerative movement disorder. PD was originally attributed to neuronal loss within the substantia nigra pars compacta, and a concomitant loss of dopamine. PD is now thought to be a multisystem disorder that involves not only the dopaminergic system, but other neurotransmitter systems whose role may become more prominent as the disease progresses (189). PD is characterized by four cardinal symptoms, resting tremor, rigidity, bradykinesia, and postural instability, all of which are motor. However, PD also may include any combination of a myriad of nonmotor symptoms (195). Both motor and nonmotor symptoms may impact the ability of those with PD to participate in exercise and/or impact the effects of that exercise on those with PD. This article provides a comprehensive overview of PD, its symptoms and progression, and current treatments for PD. Among these treatments, exercise is currently at the forefront. People with PD retain the ability to participate in many forms of exercise and generally respond to exercise interventions similarly to age-matched subjects without PD. As such, exercise is currently an area receiving substantial research attention as investigators seek interventions that may modify the progression of the disease, perhaps through neuroprotective mechanisms.
Collapse
Affiliation(s)
- Gammon M Earhart
- Program in Physical Therapy, Washington University School of Medicine, St. Louis, Missouri, USA.
| | | |
Collapse
|
97
|
Palacios N, Ascherio A. Reply to: Diabetes and risk of Parkinson's disease. Mov Disord 2013; 28:258. [PMID: 23436636 DOI: 10.1002/mds.25208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 08/23/2012] [Indexed: 11/09/2022] Open
|
98
|
Ådén E, Carlsson M, Poortvliet E, Stenlund H, Linder J, Edström M, Forsgren L, Håglin L. Dietary intake and olfactory function in patients with newly diagnosed Parkinson's disease: a case-control study. Nutr Neurosci 2013; 14:25-31. [DOI: 10.1179/174313211x12966635733312] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
99
|
Lu M, Hu G. Targeting metabolic inflammation in Parkinson's disease: implications for prospective therapeutic strategies. Clin Exp Pharmacol Physiol 2013; 39:577-85. [PMID: 22126374 DOI: 10.1111/j.1440-1681.2011.05650.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. Parkinson's disease (PD) is one of the most common neurodegenerative disorders and is characterized by a progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Although the aetiology of PD has not been clarified as yet, it is believed that ageing, diet, diabetes and adiposity are associated with PD. 2. Type 2 diabetes and lipid abnormalities share multiple common pathophysiological mechanisms with PD. In particular, inflammation plays a critical role in the destruction of both pancreatic islet β-cells and dopaminergic neurons in the substantia nigra. Emerging evidence indicates that dysfunctions of energy metabolism evoke metabolic inflammation, which differs to the narrow concept of inflammation, participating in systemic pathological processes such as neurodegenerative disease and diabetes. 3. The brain is considered an immunologically privileged organ, free from immune reactions, because it is protected by the blood-brain barrier (BBB). However, studies have shown that there is gradual impairment of neurovascular function with ageing and in neurodegenerative disorders, resulting in abnormal states, including increased BBB permeability. Consequently, harmful elements that would not normally be able to cross the BBB, such as pro-inflammatory factors, reactive oxygen species and neurotoxins, infiltrate into the brain, triggering neural injury. 4. Currently, the drugs available for the treatment of PD only ameliorate the symptoms of the disease. Therapeutic strategies aimed at stopping or modifying disease progression are still being sought. Most recent studies suggest that both central and peripheral inflammation may be dysregulated in PD. Therefore, therapeutic strategies aimed at modulating systemic inflammatory reactions or energy metabolism may represent a goal in neuroprotection in PD.
Collapse
Affiliation(s)
- Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | | |
Collapse
|
100
|
Shared dysregulated pathways lead to Parkinson's disease and diabetes. Trends Mol Med 2013; 19:176-86. [PMID: 23375873 DOI: 10.1016/j.molmed.2013.01.002] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/12/2012] [Accepted: 01/05/2013] [Indexed: 12/11/2022]
|