51
|
Abstract
Parkinson's disease (PD) is a chronic, debilitating neurodegenerative disorder characterized clinically by a variety of progressive motor and nonmotor symptoms. Currently, there is a dearth of diagnostic tools available to predict, diagnose or mitigate disease risk or progression, leading to a challenging dilemma within the healthcare management system. The search for a reliable biomarker for PD that reflects underlying pathology is a high priority in PD research. Currently, there is no reliable single biomarker predictive of risk for motor and cognitive decline, and there have been few longitudinal studies of temporal progression. A combination of multiple biomarkers might facilitate earlier diagnosis and more accurate prognosis in PD. In this review, we focus on the recent developments of serial biomarkers for PD from a variety of clinical, biochemical, genetic and neuroimaging perspectives.
Collapse
Affiliation(s)
- Anastasia Bougea
- Neurochemistry Laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Neuroscience Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
52
|
Ghanta MK, Elango P, L V K S B. Current Therapeutic Strategies and Perspectives for Neuroprotection in Parkinson's Disease. Curr Pharm Des 2020; 26:4738-4746. [PMID: 32065086 DOI: 10.2174/1381612826666200217114658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/10/2020] [Indexed: 02/04/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder of dopaminergic striatal neurons in basal ganglia. Treatment of Parkinson's disease (PD) through dopamine replacement strategies may provide improvement in early stages and this treatment response is related to dopaminergic neuronal mass which decreases in advanced stages. This treatment failure was revealed by many studies and levodopa treatment became ineffective or toxic in chronic stages of PD. Early diagnosis and neuroprotective agents may be a suitable approach for the treatment of PD. The essentials required for early diagnosis are biomarkers. Characterising the striatal neurons, understanding the status of dopaminergic pathways in different PD stages may reveal the effects of the drugs used in the treatment. This review updates on characterisation of striatal neurons, electrophysiology of dopaminergic pathways in PD, biomarkers of PD, approaches for success of neuroprotective agents in clinical trials. The literature was collected from the articles in database of PubMed, MedLine and other available literature resources.
Collapse
Affiliation(s)
- Mohan K Ghanta
- Department of Pharmacology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research (DU), Porur, Chennai-600116, Tamil Nadu, India
| | - P Elango
- Department of Pharmacology, Panimalar Medical College Hospital & Research Institute, Poonamallee, Chennai-600123, Tamil Nadu, India
| | - Bhaskar L V K S
- Department of Zoology, Guru Ghasidas University, Bilaspur, 495009 (CG), India
| |
Collapse
|
53
|
Lee DY, Oh M, Kim SJ, Oh JS, Chung SJ, Kim JS. Bilirubin-Related Differential Striatal [18F]FP-CIT Uptake in Parkinson Disease. Clin Nucl Med 2019; 44:855-859. [PMID: 31490312 DOI: 10.1097/rlu.0000000000002749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF THE REPORT Oxidative stress is a leading factor in the pathogenesis of idiopathic Parkinson disease (IPD). Two intrinsic antioxidative molecules, bilirubin and uric acid, are known to protect dopaminergic neurons from oxidative stress in IPD. The objective of this study was to determine the relationship between basal serum levels of 2 molecules and dopaminergic deficit assessed by dopamine transporter imaging with F-fluorinated-N-3-fluoropropyl-2-β-carboxymethoxy-3-β-(4-iodophenyl)nortropane ([F]FP-CIT) PET/CT in patients with early-stage drug-naive IPD. METHODS Cases of IPD patients who possess the levels of uric acid and bilirubin within a month from [F]FP-CIT PET/CT from January 2011 to December 2016 were retrospectively reviewed. As a control, the same criteria applied to patients with essential tremor (ET). PET images were analyzed using volume-of-interest templates for 12 striatal subregions and 1 occipital area, and the specific-to-nonspecific binding ratio (SNBR) was calculated. RESULTS One hundred five patients with drug-naive, early-stage IPD and 62 patients with ET were finally included. Levels of bilirubin were significantly higher in the IPD group than in controls (P = 0.026), and bilirubin level was the factor showing the most correlations with SNBR in IPD (P < 0.001), whereas uric acid showed no such difference or relationship. Furthermore, levels of bilirubin showed a positive correlation with SNBR in more affected posterior putamen in the IPD group (Pearson correlation coefficient, ρ = 0.456; P < 0.001), but a negative one in the ET group (ρ = -0.440, P < 0.001). CONCLUSIONS Bilirubin, not uric acid, was the most significant antioxidant marker for dopaminergic deficit in early-stage drug-naive IPD assessed by [F]FP-CIT PET/CT.
Collapse
Affiliation(s)
| | | | | | | | - Sun Ju Chung
- Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | |
Collapse
|
54
|
Shen Y, Li J, Schwarzschild M, Pavlova M, He S, Ascherio A, Wu S, Cui L, Gao X. Plasma urate concentrations and possible REM sleep behavior disorder. Ann Clin Transl Neurol 2019; 6:2368-2376. [PMID: 31714690 PMCID: PMC6917330 DOI: 10.1002/acn3.50929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/02/2022] Open
Abstract
Objective To examine how urate concentrations are related to the risk of having possible REM sleep behavior disorder (pRBD) in a community‐based cohort. Methods The study included 12,923 Chinese adults of the Kailuan Study, free of Parkinson disease (PD) and dementia. Plasma urate concentrations were measured in 2006, 2008, and 2010. Cumulative average urate concentration was used as primary exposure. In 2012, we determined pRBD status using a validated RBD questionnaire‐Hong Kong (RBDQ‐HK). Logistic regression analysis was performed to estimate the association between urate concentrations during 2006–2010 and odds of having pRBD in 2012 or pRBD case with symptom onset within 1 year. Results Higher average urate concentrations were associated with a lower odds of pRBD (P‐trend <0.001). The adjusted odds ratio (OR), for the highest versus lowest urate quintiles, was 0.43 (95% confidence intervals (CIs) 0.32–0.57). Significant association was consistently observed when we examined the association of a single urate assessment (2006 or 2010) or the rate of change in urate concentrations during 2006–2010 with pRBD (P‐trend <0.001 for all). However, restricting to pRBD onset during 2011–2012, we observed a nonsignificant trend between high urate concentration and high odds of pRBD (P‐trend = 0.09). Interpretation Higher average urate concentrations were associated with a lower likelihood of having pRBD, but not new‐onset pRBD. Because of its observational study design, the result should be interpreted with caution due to the possibility of residual confounding.
Collapse
Affiliation(s)
- Yun Shen
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junjuan Li
- Department of Nephrology, Kailuan General Hospital, Tangshan, China
| | - Michael Schwarzschild
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Milena Pavlova
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Songbin He
- Department of Neurology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Alberto Ascherio
- Department of Nutrition and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Liufu Cui
- Department of Rheumatology, Kailuan General Hospital, Tangshan, China
| | - Xiang Gao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
55
|
Lawton M, Baig F, Toulson G, Morovat A, Evetts SG, Ben-Shlomo Y, Hu MT. Blood biomarkers with Parkinson's disease clusters and prognosis: The oxford discovery cohort. Mov Disord 2019; 35:279-287. [PMID: 31693246 PMCID: PMC7028059 DOI: 10.1002/mds.27888] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 09/03/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022] Open
Abstract
Background Predicting prognosis in Parkinson's disease (PD) has important implications for individual prognostication and clinical trials design and targeting novel treatments. Blood biomarkers could help in this endeavor. Methods We identified 4 blood biomarkers that might predict prognosis: apolipoprotein A1, C‐reactive protein, uric acid and vitamin D. These biomarkers were measured in baseline serum from 624 Parkinson's disease subjects (median disease duration, 1.0 years; interquartile range, 0.5–2.0) from the Oxford Discovery prospective cohort. We compared these biomarkers against PD subtypes derived from clinical features in the baseline cohort using data‐driven approaches. We used multilevel models with MDS‐UPDRS parts I, II, and III and Montreal Cognitive Assessment as outcomes to test whether the biomarkers predicted subsequent progression in motor and nonmotor domains. We compared the biomarkers against age of PD onset and age at diagnosis. The q value, a false‐discovery rate alternative to P values, was calculated as an adjustment for multiple comparisons. Results Apolipoprotein A1 and C‐reactive protein levels differed across our PD subtypes, with severe motor disease phenotype, poor psychological well‐being, and poor sleep subtype having reduced apolipoprotein A1 and higher C‐reactive protein levels. Reduced apolipoprotein A1, higher C‐reactive protein, and reduced vitamin D were associated with worse baseline activities of daily living (MDS‐UPDRS II). Conclusion Baseline clinical subtyping identified a pro‐inflammatory biomarker profile significantly associated with a severe motor/nonmotor disease phenotype, lending biological validity to subtyping approaches. No blood biomarker predicted motor or nonmotor prognosis. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Michael Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Fahd Baig
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Greg Toulson
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alireza Morovat
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Samuel G Evetts
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michele T Hu
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK.,Department of Clinical Neurology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
56
|
Mandal AK, Mount DB. Interaction Between ITM2B and GLUT9 Links Urate Transport to Neurodegenerative Disorders. Front Physiol 2019; 10:1323. [PMID: 31695625 PMCID: PMC6818471 DOI: 10.3389/fphys.2019.01323] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/03/2019] [Indexed: 11/13/2022] Open
Abstract
Hyperuricemia plays a critical causative role in gout. In contrast, hyperuricemia has a protective effect in neurodegenerative disorders, including Alzheimer's Disease. Genetic variation in the SLC2A9 gene, encoding the urate transporter GLUT9, exerts the largest single-gene effect on serum uric acid (SUA). We report here the identification of two GLUT9-interacting proteins, integral membrane protein 2B (ITM2B) and transmembrane protein 85 (TMEM85), isolated from a human kidney cDNA library using the dual-membrane yeast two-hybrid system. ITM2B is a ubiquitously expressed, N-glycosylated transmembrane regulatory protein, involved in familial dementias and retinal dystrophy; the function of TMEM85 is less defined. Using coimmunoprecipitation, we confirmed the physical interaction between ITM2B or TMEM85 and N-terminal GLUT9 isoforms (GLUT9a and GLUT9b) in transfected HEK 293T cells and Xenopus oocytes, wherein ITM2B but not TMEM85 inhibited GLUT9-mediated urate uptake. Additionally, co-expression of ITM2B with GLUT9 in oocytes inhibited N-glycosylation of GLUT9a more than GLUT9b and stimulated urate efflux by both isoforms. However, urate uptake by N-glycosylation and N-terminal deletion GLUT9 mutants was efficiently inhibited by ITM2B, indicating that neither N-glycosylation nor the N terminus is necessary for functional interaction of GLUT9 with ITM2B. Notably, ITM2B variants linked to familial Danish dementia and retinal dystrophy significantly attenuated the inhibition of GLUT9-mediated urate influx. We propose ITM2B as a potential regulatory link between urate homeostasis and neurodegenerative disorders.
Collapse
Affiliation(s)
- Asim K. Mandal
- Renal Divisions, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - David B. Mount
- Renal Divisions, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| |
Collapse
|
57
|
Oh Y‐S, Kim J‐S, Yoo S‐W, Hwang E‐J, Lyoo CH, Lee K‐S. Gender difference in the effect of uric acid on striatal dopamine in early Parkinson's disease. Eur J Neurol 2019; 27:258-264. [DOI: 10.1111/ene.14070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Y. ‐S. Oh
- Department of Neurology College of Medicine The Catholic University of Korea Seoul Korea
| | - J. ‐S. Kim
- Department of Neurology College of Medicine The Catholic University of Korea Seoul Korea
| | - S. ‐W. Yoo
- Department of Neurology College of Medicine The Catholic University of Korea Seoul Korea
| | - E. ‐J. Hwang
- Department of Radiology College of Medicine The Catholic University of Korea Seoul Korea
| | - C. H. Lyoo
- Department of Neurology Gangnam Severance Hospital Yonsei University College of Medicine Seoul Korea
| | - K. ‐S. Lee
- Department of Neurology College of Medicine The Catholic University of Korea Seoul Korea
| |
Collapse
|
58
|
Nag N, Jelinek GA. A Narrative Review of Lifestyle Factors Associated with Parkinson's Disease Risk and Progression. NEURODEGENER DIS 2019; 19:51-59. [PMID: 31487721 DOI: 10.1159/000502292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/23/2019] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease is a complex slowly progressive neurodegenerative disorder with motor and non-motor symptoms affecting daily living. Despite effective symptomatic treatments, with various degrees of side effects, no disease-modifying therapeutic options presently exist. Symptoms progress, with an accumulating burden, reducing the quality of life and forming the impression that medications are no longer effective. Adopting positive lifestyle behaviours can empower patients, improve the quality of life, alleviate symptoms, and potentially slow disease progression. Lifestyle behaviours including nutrition, cognitive enrichment, physical activity, and stress management have beneficial effects on brain health and quality of life. While some evidence of an association of lifestyle with Parkinson's disease risk and progression exists, the sparse and often conflicting data make it difficult to provide clinical recommendations. Herein, we highlight studies showing promising associations between lifestyle and Parkinson's disease. Given the increasing aging of populations worldwide and the prevalence of neurological disorders, further research into self-management through adoption of positive lifestyle behaviours is clearly warranted to better enable individualized care.
Collapse
Affiliation(s)
- Nupur Nag
- Neuroepidemiology Unit, Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia,
| | - George A Jelinek
- Neuroepidemiology Unit, Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
59
|
Schwarzschild MA, Macklin EA, Bakshi R, Battacharyya S, Logan R, Espay AJ, Hung AY, Bwala G, Goetz CG, Russell DS, Goudreau JL, Parashos SA, Saint-Hilaire MH, Rudolph A, Hare JM, Curhan GC, Ascherio A. Sex differences by design and outcome in the Safety of Urate Elevation in PD (SURE-PD) trial. Neurology 2019; 93:e1328-e1338. [PMID: 31484712 DOI: 10.1212/wnl.0000000000008194] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To investigate whether women and men with Parkinson disease (PD) differ in their biochemical and clinical responses to long-term treatment with inosine. METHODS The Safety of Urate Elevation in Parkinson's Disease (SURE-PD) trial enrolled 75 people with early PD and baseline serum urate below 6 mg/dL and randomized them to 3 double-blinded treatment arms: oral placebo or inosine titrated to produce mild (6.1-7.0 mg/dL) or moderate (7.1-8.0 mg/dL) serum urate elevation for up to 2 years. Parkinsonism, serum urate, and plasma antioxidant capacity were measured at baseline and repeatedly on treatment; CSF urate was assessed once, at 3 months. Here in secondary analyses results are stratified by sex. RESULTS Inosine produced an absolute increase in average serum urate from baseline that was 50% greater in women (3.0 mg/dL) than in men (2.0 mg/dL), consistent with expected lower baseline levels in women. Similarly, only among women was CSF urate significantly greater on mild or moderate inosine (+87% [p < 0.001] and +98% [p < 0.001], respectively) than on placebo (in contrast to men: +10% [p = 0.6] and +14% [p = 0.4], respectively). Women in the higher inosine dosing group showed a 7.0 Unified Parkinson's Disease Rating Scale (UPDRS) points/year lower rate of decline vs placebo (p = 0.01). In women, slower rates of UPDRS change were associated with greater increases in serum urate (r = -0.52; p = 0.001), and with greater increases in plasma antioxidant capacity (r = -0.44; p = 0.006). No significant associations were observed in men. CONCLUSIONS Inosine produced greater increases in serum and CSF urate in women compared to men in the SURE-PD trial, consistent with the study's design and with preliminary evidence for slower clinical decline in early PD among women treated with urate-elevating doses of inosine. CLINICALTRIALSGOV IDENTIFIER NCT00833690. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that inosine produced greater urate elevation in women than men and may slow PD progression in women.
Collapse
Affiliation(s)
- Michael A Schwarzschild
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA.
| | - Eric A Macklin
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Rachit Bakshi
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Shamik Battacharyya
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Robert Logan
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Alberto J Espay
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Albert Y Hung
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Grace Bwala
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Christopher G Goetz
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - David S Russell
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - John L Goudreau
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Sotirios A Parashos
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Marie H Saint-Hilaire
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Alice Rudolph
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Joshua M Hare
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Gary C Curhan
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Alberto Ascherio
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | | |
Collapse
|
60
|
Feeding-produced subchronic high plasma levels of uric acid improve behavioral dysfunction in 6-hydroxydopamine-induced mouse model of Parkinson's disease. Behav Pharmacol 2019; 30:89-94. [PMID: 29847340 DOI: 10.1097/fbp.0000000000000413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The development of Parkinson's disease (PD) involves the degeneration of dopaminergic neurons caused by oxidative stress. Accumulating clinical evidence indicates that high blood levels of uric acid (UA), an intrinsic antioxidative substance, are associated with reduced risk of PD. However, this hypothesis has not been confirmed by in-vivo experiments. The present study investigated the effects of UA on behavioral abnormalities in the development of PD. We used unilateral 6-hydroxydopamine-lesioned mice, which were fed on a diet containing 1% UA and 2.5% potassium oxonate (an uricase inhibitor) to induce hyperuricemia. A significant elevation in UA levels was found in groups that were fed a UA diet. The 6-hydroxydopamine-lesioned mice showed impaired rotarod performance and increased apomorphine-induced contralateral rotations. These behavioral abnormalities were significantly reversed by feeding a UA diet for 1 week before and 5 weeks after surgery (subchronic hyperuricemia). These behavioral improvements occurred in parallel with recovery of tyrosine hydroxylase protein levels in the lesioned striatal side. The present study with a dietary hyperuricemia mice model confirms that UA exerts a neuroprotective effect on dopaminergic neuronal loss, improving motor dysfunction and ameliorating PD development.
Collapse
|
61
|
Mantri S, Morley JF, Siderowf AD. The importance of preclinical diagnostics in Parkinson disease. Parkinsonism Relat Disord 2019; 64:20-28. [DOI: 10.1016/j.parkreldis.2018.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/02/2018] [Accepted: 09/08/2018] [Indexed: 01/21/2023]
|
62
|
Pohl H, Vallelian F, Herfs G. [Light and Shadow of Hyperuricemia - Neuroprotection, Comorbidities and Therapeutic Strategies]. PRAXIS 2019; 108:527-533. [PMID: 31185840 DOI: 10.1024/1661-8157/a003253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Light and Shadow of Hyperuricemia - Neuroprotection, Comorbidities and Therapeutic Strategies Abstract. Hyperuricemia can lead to gout but also favor the appearance of comorbidities like hypertension, kidney insufficiency, type 2 diabetes, myocardial infarction and strokes. Uric acid does not only have, however, negative impact on the body, but seems to influence positively certain inflammatory and degenerative neurological diseases. The inflammatory reaction at the center of a gout attack is mediated by interleukin 1. Therefore, antagonists against interleukin 1 or IL-1 receptors can be used for treatment if colchicine, steroids or nonsteroidal anti-inflammatory drugs are ineffective or contraindicated. First-line drug for urate-lowering therapy is allopurinol that positively influences comorbidities as well.
Collapse
Affiliation(s)
- Heiko Pohl
- 1 Klinik und Poliklinik für Innere Medizin, Universitätsspital Zürich
| | | | - Gregor Herfs
- 1 Klinik und Poliklinik für Innere Medizin, Universitätsspital Zürich
| |
Collapse
|
63
|
Guttuso T, Andrzejewski KL, Lichter DG, Andersen JK. Targeting kinases in Parkinson's disease: A mechanism shared by LRRK2, neurotrophins, exenatide, urate, nilotinib and lithium. J Neurol Sci 2019; 402:121-130. [PMID: 31129265 DOI: 10.1016/j.jns.2019.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022]
Abstract
Several kinases have been implicated in the pathogenesis of Parkinson's disease (PD), most notably leucine-rich repeat kinase 2 (LRRK2), as LRRK2 mutations are the most common genetic cause of a late-onset parkinsonism that is clinically indistinguishable from sporadic PD. More recently, several other kinases have emerged as promising disease-modifying targets in PD based on both preclinical studies and clinical reports on exenatide, the urate precursor inosine, nilotinib and lithium use in PD patients. These kinases include protein kinase B (Akt), glycogen synthase kinases-3β and -3α (GSK-3β and GSK-3α), c-Abelson kinase (c-Abl) and cyclin-dependent kinase 5 (cdk5). Activities of each of these kinases are involved either directly or indirectly in phosphorylating tau or increasing α-synuclein levels, intracellular proteins whose toxic oligomeric forms are strongly implicated in the pathogenesis of PD. GSK-3β, GSK-3α and cdk5 are the principle kinases involved in phosphorylating tau at sites critical for the formation of tau oligomers. Exenatide analogues, urate, nilotinib and lithium have been shown to affect one or more of the above kinases, actions that can decrease the formation and increase the clearance of intraneuronal phosphorylated tau and α-synuclein. Here we review the current preclinical and clinical evidence supporting kinase-targeting agents as potential disease-modifying therapies for PD patients enriched with these therapeutic targets and incorporate LRRK2 physiology into this novel model.
Collapse
Affiliation(s)
- Thomas Guttuso
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - Kelly L Andrzejewski
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - David G Lichter
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - Julie K Andersen
- The Buck Institute for Research on Aging, Novato, CA, United States of America.
| |
Collapse
|
64
|
Nybo SE, Lamberts JT. Integrated use of LC/MS/MS and LC/Q-TOF/MS targeted metabolomics with automated label-free microscopy for quantification of purine metabolites in cultured mammalian cells. Purinergic Signal 2019; 15:17-25. [PMID: 30604179 PMCID: PMC6439090 DOI: 10.1007/s11302-018-9643-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/11/2018] [Indexed: 01/27/2023] Open
Abstract
Purine metabolites have been implicated as clinically relevant biomarkers of worsening or improving Parkinson's disease (PD) progression. However, the identification of purine molecules as biomarkers in PD has largely been determined using non-targeted metabolomics analysis. The primary goal of this study was to develop an economical targeted metabolomics approach for the routine detection of purine molecules in biological samples. Specifically, this project utilized LC/MS/MS and LC/QTOF/MS to accurately quantify levels of six purine molecules in samples from cultured N2a murine neuroblastoma cells. The targeted metabolomics workflow was integrated with automated label-free digital microscopy, which enabled normalization of purine concentration per unit cell in the absence of fluorescent dyes. The established method offered significantly enhanced selectivity compared to previously published procedures. In addition, this study demonstrates that a simple, quantitative targeted metabolomics approach can be developed to identify and quantify purine metabolites in biological samples. We envision that this method could be broadly applicable to quantification of purine metabolites from other complex biological samples, such as cerebrospinal fluid or blood.
Collapse
Affiliation(s)
- S Eric Nybo
- College of Pharmacy, Department of Pharmaceutical Sciences, Ferris State University, 220 Ferris Drive, Big Rapids, MI, 49307, USA
| | - Jennifer T Lamberts
- College of Pharmacy, Department of Pharmaceutical Sciences, Ferris State University, 220 Ferris Drive, Big Rapids, MI, 49307, USA.
| |
Collapse
|
65
|
Chen X, Wei QQ, Chen Y, Cao B, Ou R, Hou Y, Yuan X, Zhang L, Liu H, Shang H. Clinical disease stage related changes of serological factors in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2019; 20:53-60. [PMID: 30784318 DOI: 10.1080/21678421.2018.1550516] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Little is known whether disease clinical stage would influence the serological values in Amyotrophic lateral sclerosis (ALS). We aimed to explore the association between the levels of serological factors with clinical progression determined by the King's College staging system. METHODS ALS Patients were registered from May 2008 to December 2016. The differences of serological values between patients and healthy controls, and the correlation of these serological values with disease stage were examined. RESULTS A total of 571 patients and 571 age-/gender-/BMI-matched healthy controls were included. The levels of creatinine, uric acid (UA), albumin, total protein, total cholesterol, and high-density lipoprotein (HDL) were significantly lower, and the low-density lipoprotein/HDL ratio was higher in ALS patients than those in healthy controls. The levels of UA, albumin, and total protein were significantly reversely correlated with diseases stages. The longitudinal observation of 81 ALS patients also showed that the levels of UA, creatinine, albumin, total protein, and HDL were significantly decreased in the second hematological examinations. CONCLUSIONS In the present study, ALS patients and control subjects were evenly matched with regard to sex, age, and BMI value, this finding could be considered as a metabolite signature in ALS. The changes of metabolite-based serological factors with progression of disease stage might be related to the pathophysiology of disease, and might have clinical utility in clinical practice.
Collapse
Affiliation(s)
- Xueping Chen
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Qian-Qian Wei
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Yongping Chen
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Bei Cao
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - RuWei Ou
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Yanbing Hou
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Xiaoqin Yuan
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Lingyu Zhang
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Hui Liu
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Huifang Shang
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| |
Collapse
|
66
|
Johnson TA, Jinnah HA, Kamatani N. Shortage of Cellular ATP as a Cause of Diseases and Strategies to Enhance ATP. Front Pharmacol 2019; 10:98. [PMID: 30837873 PMCID: PMC6390775 DOI: 10.3389/fphar.2019.00098] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/24/2019] [Indexed: 12/14/2022] Open
Abstract
Germline mutations in cellular-energy associated genes have been shown to lead to various monogenic disorders. Notably, mitochondrial disorders often impact skeletal muscle, brain, liver, heart, and kidneys, which are the body’s top energy-consuming organs. However, energy-related dysfunctions have not been widely seen as causes of common diseases, although evidence points to such a link for certain disorders. During acute energy consumption, like extreme exercise, cells increase the favorability of the adenylate kinase reaction 2-ADP -> ATP+AMP by AMP deaminase degrading AMP to IMP, which further degrades to inosine and then to purines hypoxanthine -> xanthine -> urate. Thus, increased blood urate levels may act as a barometer of extreme energy consumption. AMP deaminase deficient subjects experience some negative effects like decreased muscle power output, but also positive effects such as decreased diabetes and improved prognosis for chronic heart failure patients. That may reflect decreased energy consumption from maintaining the pool of IMP for salvage to AMP and then ATP, since de novo IMP synthesis requires burning seven ATPs. Similarly, beneficial effects have been seen in heart, skeletal muscle, or brain after treatment with allopurinol or febuxostat to inhibit xanthine oxidoreductase, which catalyzes hypoxanthine -> xanthine and xanthine -> urate reactions. Some disorders of those organs may reflect dysfunction in energy-consumption/production, and the observed beneficial effects related to reinforcement of ATP re-synthesis due to increased hypoxanthine levels in the blood and tissues. Recent clinical studies indicated that treatment with xanthine oxidoreductase inhibitors plus inosine had the strongest impact for increasing the pool of salvageable purines and leading to increased ATP levels in humans, thereby suggesting that this combination is more beneficial than a xanthine oxidoreductase inhibitor alone to treat disorders with ATP deficiency.
Collapse
Affiliation(s)
| | - H A Jinnah
- Departments of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | | |
Collapse
|
67
|
Zhang C, Yang Y, Liang W, Wang T, Wang S, Wang X, Wang Y, Jiang H, Feng H. Neuroprotection by urate on the mutant hSOD1-related cellular and Drosophila models of amyotrophic lateral sclerosis: Implication for GSH synthesis via activating Akt/GSK3β/Nrf2/GCLC pathways. Brain Res Bull 2019; 146:287-301. [PMID: 30690059 DOI: 10.1016/j.brainresbull.2019.01.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 12/13/2022]
Abstract
Oxidative stress has been considered as a principal mechanism of motor neuron death in amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease which could be caused by dominant mutations in an antioxidant enzyme superoxide dismutase-1 (SOD1). The aim of the present study was to investigate the potential neuroprotective effects and mechanisms of urate, an important endogenous antioxidant and a biomarker of favorable ALS progression rates, in the mutant human SOD1-related cellular and Drosophila models of ALS. Our results showed that urate treatment provided neuroprotective effects as confirmed by enhanced survival, attenuated motor impairments, reduced oxidative damage and increased antioxidant defense in hSOD1-G85R-expressing Drosophila models of ALS. In vitro studies, we demonstrated that urate protected motor neurons (NSC-34 cells) against hSOD1-G93A-induced cell damage and apoptosis by decreasing reactive oxygen specials (ROS) production and oxidative damage. Moreover, urate markedly increased the expression and activation of nuclear factor erythroid 2-related factor 2 (Nrf2), stimulated Nrf2-targeted antioxidant gene glutathione cysteine ligase catalytic subunit (GCLC) expression and glutathione (GSH) synthesis by upregulating Akt/GSK3β pathway. Furthermore, the inhibition of Akt pathway with LY294002 abolished urate-mediated elevation of GSH synthesis and neuroprotective effects both in vivo and in vitro. Overall, these results suggested that, in addition to its direct scavenging of ROS, urate markedly enhanced GSH expression by activating Akt/GSK3β/Nrf2/GCLC pathway, and thus offering neuroprotective effects on motor neurons against oxidative stress.
Collapse
Affiliation(s)
- Chunting Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Yueqing Yang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Weiwei Liang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Tianhang Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Shuyu Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Xudong Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Ying Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Hongquan Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China
| | - Honglin Feng
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, PR China.
| |
Collapse
|
68
|
Shao Y, Le W. Recent advances and perspectives of metabolomics-based investigations in Parkinson's disease. Mol Neurodegener 2019; 14:3. [PMID: 30634989 PMCID: PMC6330496 DOI: 10.1186/s13024-018-0304-2] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/06/2018] [Indexed: 12/24/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease of the central nervous system (CNS), which affects mostly older adults. In recent years, the incidence of PD has been dramatically increasing with the aging population expanding. Due to the lack of effective biomarkers, the accurate diagnosis and precise treatment of PD are currently compromised. Notably, metabolites have been considered as the most direct reflection of the physiological and pathological conditions in individuals and represent attractive candidates to provide deep insights into disease phenotypes. By profiling the metabolites in biofluids (cerebrospinal fluid, blood, urine), feces and brain tissues, metabolomics has become a powerful and promising tool to identify novel biomarkers and provide valuable insights into the etiopathogenesis of neurological diseases. In this review, we will summarize the recent advancements of major analytical platforms implemented in metabolomics studies, dedicated to the improvement and extension of metabolome coverage for in-depth biological research. Based on the current metabolomics studies in both clinical populations and experimental PD models, this review will present new findings in metabolomics biomarkers research and abnormal metabolic pathways in PD, and will discuss the correlation between metabolomic changes and clinical conditions of PD. A better understanding of the biological underpinning of PD pathogenesis might offer novel diagnostic, prognostic, and therapeutic approaches to this devastating disease.
Collapse
Affiliation(s)
- Yaping Shao
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
69
|
Nicholson K, Chan J, Macklin EA, Levine‐Weinberg M, Breen C, Bakshi R, Grasso DL, Wills A, Jahandideh S, Taylor AA, Beaulieu D, Ennist DL, Andronesi O, Ratai E, Schwarzschild MA, Cudkowicz M, Paganoni S. Pilot trial of inosine to elevate urate levels in amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2018; 5:1522-1533. [PMID: 30564619 PMCID: PMC6292193 DOI: 10.1002/acn3.671] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/13/2018] [Accepted: 09/10/2018] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To test the safety, tolerability, and urate-elevating capability of the urate precursor inosine taken orally or by feeding tube in people with amyotrophic lateral sclerosis (ALS). METHODS This was a pilot, open-label trial in 25 participants with ALS. Treatment duration was 12 weeks. The dose of inosine was titrated at pre-specified time points to elevate serum urate levels to 7-8 mg/dL. Primary outcomes were safety (as assessed by the occurrence of adverse events [AEs]) and tolerability (defined as the ability to complete the 12-week study on study drug). Secondary outcomes included biomarkers of oxidative stress and damage. As an exploratory analysis, observed outcomes were compared with a virtual control arm built using prediction algorithms to estimate ALSFRS-R scores. RESULTS Twenty-four out of 25 participants (96%) completed 12 weeks of study drug treatment. One participant was unable to comply with study visits and was lost to follow-up. Serum urate rose to target levels in 6 weeks. No serious AEs attributed to study drug and no AEs of special concern, such as urolithiasis and gout, occurred. Selected biomarkers of oxidative stress and damage had significant changes during the study period. Observed changes in ALSFRS-R did not differ from baseline predictions. INTERPRETATION Inosine appeared safe, well tolerated, and effective in raising serum urate levels in people with ALS. These findings, together with epidemiological observations and preclinical data supporting a neuroprotective role of urate in ALS models, provide the rationale for larger clinical trials testing inosine as a potential disease-modifying therapy for ALS.
Collapse
Affiliation(s)
- Katharine Nicholson
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
| | - James Chan
- MGH Biostatistics CenterBostonMassachusetts
| | | | - Mark Levine‐Weinberg
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
| | - Christopher Breen
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
| | - Rachit Bakshi
- MassGeneral Institute for Neurodegenerative DiseaseBostonMassachusetts
| | - Daniela L. Grasso
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
| | - Anne‐Marie Wills
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
| | | | | | | | | | - Ovidiu Andronesi
- MGH Department of RadiologyA. A. Martinos Center for Biomedical ImagingBostonMassachusetts
| | - Eva‐Maria Ratai
- MGH Department of RadiologyA. A. Martinos Center for Biomedical ImagingBostonMassachusetts
| | | | - Merit Cudkowicz
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
| | - Sabrina Paganoni
- Neurological Clinical Research Institute (NCRI)Massachusetts General Hospital (MGH)BostonMassachusetts
- Spaulding Rehabilitation HospitalBostonMassachusetts
| |
Collapse
|
70
|
Abstract
The past decade has seen tremendous efforts in biomarker discovery and validation for neurodegenerative diseases. The source and type of biomarkers has continued to grow for central nervous system diseases, from biofluid-based biomarkers (blood or cerebrospinal fluid (CSF)), to nucleic acids, tissue, and imaging. While DNA remains a predominant biomarker used to identify familial forms of neurodegenerative diseases, various types of RNA have more recently been linked to familial and sporadic forms of neurodegenerative diseases during the past few years. Imaging approaches continue to evolve and are making major contributions to target engagement and early diagnostic biomarkers. Incorporation of biomarkers into drug development and clinical trials for neurodegenerative diseases promises to aid in the development and demonstration of target engagement and drug efficacy for neurologic disorders. This review will focus on recent advancements in developing biomarkers for clinical utility in Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
| | - Robert Bowser
- Iron Horse Diagnostics, Inc., Scottsdale, AZ, 85255, USA.
- Divisions of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
71
|
Tasca CI, Lanznaster D, Oliveira KA, Fernández-Dueñas V, Ciruela F. Neuromodulatory Effects of Guanine-Based Purines in Health and Disease. Front Cell Neurosci 2018; 12:376. [PMID: 30459558 PMCID: PMC6232889 DOI: 10.3389/fncel.2018.00376] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 10/02/2018] [Indexed: 12/18/2022] Open
Abstract
The function of guanine-based purines (GBPs) is mostly attributed to the intracellular modulation of heteromeric and monomeric G proteins. However, extracellular effects of guanine derivatives have also been recognized. Thus, in the central nervous system (CNS), a guanine-based purinergic system that exerts neuromodulator effects, has been postulated. The thesis that GBPs are neuromodulators emerged from in vivo and in vitro studies, in which neurotrophic and neuroprotective effects of these kinds of molecules (i.e., guanosine) were demonstrated. GBPs induce several important biological effects in rodent models and have been shown to reduce seizures and pain, stabilize mood disorder behavior and protect against gliomas and diseases related with aging, such as ischemia or Parkinson and Alzheimer diseases. In vitro studies to evaluate the protective and trophic effects of guanosine, and of the nitrogenous base guanine, have been fundamental for understanding the mechanisms of action of GBPs, as well as the signaling pathways involved in their biological roles. Conversely, although selective binding sites for guanosine have been identified in the rat brain, GBP receptors have not been still described. In addition, GBP neuromodulation may depend on the capacity of GBPs to interact with well-known membrane proteins in glutamatergic and adenosinergic systems. Overall, in this review article, we present up-to-date GBP biology, focusing mainly on the mechanisms of action that may lead to the neuromodulator role of GBPs observed in neurological disorders.
Collapse
Affiliation(s)
- Carla I Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Débora Lanznaster
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,UMR 1253, Team 2, INSERM/University of Tours, Tours, France
| | - Karen A Oliveira
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Victor Fernández-Dueñas
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
72
|
He R, Yan X, Guo J, Xu Q, Tang B, Sun Q. Recent Advances in Biomarkers for Parkinson's Disease. Front Aging Neurosci 2018; 10:305. [PMID: 30364199 PMCID: PMC6193101 DOI: 10.3389/fnagi.2018.00305] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 09/14/2018] [Indexed: 02/04/2023] Open
Abstract
Parkinson's disease (PD) is one of the common progressive neurodegenerative disorders with several motor and non-motor symptoms. Most of the motor symptoms may appear at a late stage where most of the dopaminergic neurons have been already damaged. In order to provide better clinical intervention and treatment at the onset of disease, it is imperative to find accurate biomarkers for early diagnosis, including prodromal diagnosis and preclinical diagnosis. At the same time, these reliable biomarkers can also be utilized to monitor the progress of the disease. In this review article, we will discuss recent advances in the development of PD biomarkers from different aspects, including clinical, biochemical, neuroimaging and genetic aspects. Although various biomarkers for PD have been developed so far, their specificity and sensitivity are not ideal when applied individually. So, the combination of multimodal biomarkers will greatly improve the diagnostic accuracy and facilitate the implementation of personalized medicine.
Collapse
Affiliation(s)
- Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Parkinson’s Disease Center of Beijing Institute for Brain Disorders, Beijing, China
- Collaborative Innovation Center for Brain Science, Shanghai, China
- Collaborative Innovation Center for Genetics and Development, Shanghai, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Parkinson’s Disease Center of Beijing Institute for Brain Disorders, Beijing, China
- Collaborative Innovation Center for Brain Science, Shanghai, China
- Collaborative Innovation Center for Genetics and Development, Shanghai, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Qiying Sun
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
73
|
Cova I, Priori A. Diagnostic biomarkers for Parkinson's disease at a glance: where are we? J Neural Transm (Vienna) 2018; 125:1417-1432. [PMID: 30145631 PMCID: PMC6132920 DOI: 10.1007/s00702-018-1910-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder whose aetiology remains unclear: degeneration involves several neurotransmission systems, resulting in a heterogeneous disease characterized by motor and non-motor symptoms. PD causes progressive disability that responds only to symptomatic therapies. Future advances include neuroprotective strategies for use in at-risk populations before the clinical onset of disease, hence the continuing need to identify reliable biomarkers that can facilitate the clinical diagnosis of PD. In this evaluative review, we summarize information on potential diagnostic biomarkers for use in the clinical and preclinical stages of PD.
Collapse
Affiliation(s)
- Ilaria Cova
- Neurology Unit, L. Sacco University Hospital, Milan, Italy
| | - Alberto Priori
- Department of Health Sciences, "Aldo Ravelli" Research Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan and ASST Santi Paolo e Carlo, Milan, Italy.
| |
Collapse
|
74
|
Doyle C, Cristofaro V, Sullivan MP, Adam RM. Inosine - a Multifunctional Treatment for Complications of Neurologic Injury. Cell Physiol Biochem 2018; 49:2293-2303. [PMID: 30261493 DOI: 10.1159/000493831] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/18/2018] [Indexed: 01/31/2023] Open
Abstract
Spinal cord injury (SCI) caused by trauma or disease leads to motor and sensory abnormalities that depend on the level, severity and duration of the lesion. The most obvious consequence of SCI is paralysis affecting lower and upper limbs. SCI also leads to loss of bladder and bowel control, both of which have a deleterious, life-long impact on the social, psychological, functional, medical and economic well being of affected individuals. Currently, there is neither a cure for SCI nor is there adequate management of its consequences. Although medications provide symptomatic relief for the complications of SCI including muscle spasms, lower urinary tract dysfunction and hyperreflexic bowel, strategies for repair of spinal injuries and recovery of normal limb and organ function are still to be realized. In this review, we discuss experimental evidence supporting the use of the naturally occurring purine nucleoside inosine to improve the devastating sequelae of SCI. Evidence suggests inosine is a safe, novel agent with multifunctional properties that is effective in treating complications of SCI and other neuropathies.
Collapse
Affiliation(s)
- Claire Doyle
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivian Cristofaro
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Division of Urology, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Maryrose P Sullivan
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Division of Urology, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
75
|
Kobylecki CJ, Nordestgaard BG, Afzal S. Plasma urate and risk of Parkinson's disease: A mendelian randomization study. Ann Neurol 2018; 84:178-190. [PMID: 30014508 DOI: 10.1002/ana.25292] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/26/2018] [Accepted: 07/01/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Urate is a potent antioxidant, and high plasma urate has been associated with lower incidence of Parkinson's disease (PD) in epidemiological studies. We tested the hypothesis that high concentrations of plasma urate are associated with low incidence of PD. METHODS We performed observational and genetic analyses using plasma urate and the urate SLC2A9 rs7442295 and ABCG2 rs2231142 genotype in >102,000 individuals from the CGPS (Copenhagen General Population Study). Information on PD and mortality was from national patient and death registries. Incidences of PD were calculated using Cox regression, Fine and Gray competing-risks regression, and instrumental variable analyses. RESULTS In total, 398 individuals were diagnosed with PD, of which 285 were incident cases. The multivariable adjusted hazard ratio for PD was 0.56 (95% confidence interval [CI], 0.41-0.77) for the highest versus the lowest tertile of plasma urate (p for trend across 3 groups, 8 × 10-5 ). Each one-allele increase in the combined allele score was associated with 19μmol/l (95% CI, 18.5-19.9) higher plasma urate. In observational analyses, a 50μmol/l higher plasma urate was associated with a hazard ratio of 0.85 (0.77-0.92) for PD; in instrumental variable analyses, 50μmol/l higher plasma urate was associated with an odds ratio of 1.20 (0.85-1.71) for PD. INTERPRETATION High plasma urate was associated with lower risk of PD in observational analyses; however, in instrumental variable analysis, high plasma urate was not associated with low risk of PD. Thus, our data do not support a causal relationship between high plasma urate and low risk of PD. Ann Neurol 2018;84:178-190.
Collapse
Affiliation(s)
- Camilla J Kobylecki
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
76
|
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
|
77
|
Lee Y, Park YH, Lee JJ, Sohn YH, Lee JM, Lee PH. Gender-specific effect of uric acid on resting-state functional networks in de novo Parkinson's disease. Parkinsonism Relat Disord 2018; 52:49-54. [DOI: 10.1016/j.parkreldis.2018.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 02/20/2018] [Accepted: 03/25/2018] [Indexed: 11/16/2022]
|
78
|
Bao LH, Zhang YN, Zhang JN, Gu L, Yang HM, Huang YY, Xia N, Zhang H. Urate inhibits microglia activation to protect neurons in an LPS-induced model of Parkinson's disease. J Neuroinflammation 2018; 15:131. [PMID: 29720230 PMCID: PMC5932803 DOI: 10.1186/s12974-018-1175-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/23/2018] [Indexed: 01/30/2023] Open
Abstract
Background Multiple risk factors contribute to the progression of Parkinson’s disease, including oxidative stress and neuroinflammation. Epidemiological studies have revealed a link between higher urate level and a lower risk of developing PD. However, the mechanistic basis for this association remains unclear. Urate protects dopaminergic neurons from cell death induced by oxidative stress. Here, we investigated a novel role of urate in microglia activation in a lipopolysaccharide (LPS)-induced PD model. Methods We utilized Griess, ELISA, real-time PCR, Western blot, immunohistochemistry, and immunofluorescence to detect the neuroinflammation. For Griess, ELISA, Western blot, and immunofluorescence assay, cells were seeded in 6-well plates pre-coated with poly-l-lysine (PLL) and incubated for 24 h with the indicated drugs. For real-time PCR assay, cells were seeded in 6-well plates pre-coated with PLL and incubated for 6 h with the indicated drugs. For animal experiments, rats were injected with urate or its vehicle twice daily for five consecutive days before and after stereotaxic surgery. Rats were killed and brain tissues were harvested after 4 weeks of LPS injection. Results In cultured BV2 cells and rat primary microglia, urate suppressed proinflammatory cytokine production and inducible cyclooxygenase 2 and nitric oxide synthase expression to protect dopaminergic neurons from the toxic effects of activated microglia. The neuroprotective effects of urate may also be associated with the stimulation of anti-inflammatory factors interleukin 10 and transforming growth factor β1. Intracellular urate level was increased in a dose-dependent manner upon co-treatment with urate and LPS as compared with LPS alone, an effect that was abrogated by pretreatment with probenecid (PBN), an inhibitor of both glucose transporter 9 and urate transporter 1 (URAT1). PBN also abolished the anti-inflammatory effect of urate. Consistent with these in vitro observations, the number of tyrosine hydroxylase-positive neurons was decreased and the loss of motor coordination was reversed by urate administration in an LPS-induced rat model of PD. Additionally, increased plasma urate level abolished the reduction of URAT1 expression, the increase in the expression of interleukin-1β, and the number of ionized calcium-binding adaptor molecule 1-positive microglia along with changes in their morphology. Conclusions Urate protects neurons against cytotoxicity induced by microglia activation via modulating urate transporter-mediated intracellular urate level. Electronic supplementary material The online version of this article (10.1186/s12974-018-1175-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Hui Bao
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ya-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Yi-Ying Huang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ning Xia
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
79
|
Hughes KC, Gao X, O'Reilly EJ, Kim IY, Wang M, Weisskopf MG, Schwarzschild MA, Ascherio A. Genetic variants related to urate and risk of Parkinson's disease. Parkinsonism Relat Disord 2018; 53:4-9. [PMID: 29789205 DOI: 10.1016/j.parkreldis.2018.04.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/13/2018] [Accepted: 04/28/2018] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Higher urate concentrations have been associated with a lower risk of developing Parkinson's disease (PD) and with slower rates of clinical decline in PD patients. Whether these associations reflect a neuroprotective effect of urate is unclear. Our objective was to assess whether genetic variants that modify circulating urate levels are also associated with altered PD risk. METHODS Participants were from three large ongoing cohort studies: the Nurses' Health Study (NHS), the Health Professionals Follow-up Study (HPFS), and the Cancer Prevention Study II Nutrition Cohort (CPS-IIN). We examined associations between single nucleotide polymorphisms (SNPs) in SLC2A9 and other genes involved in urate transport and PD risk using conditional logistic regression among 1451 cases and 3135 matched controls. We assessed associations between SNPs and plasma urate levels in a subset of 1174 control participants with linear regression models. RESULTS We found the expected associations between SNPs in SLC2A9 and plasma urate levels among men and women; however, SNPs in other genes tended not to be associated with urate. Each SNP in SLC2A9 explained less than 7% of the variance in plasma urate. We did not find significant associations between the SNPs in SLC2A9 and PD risk among men or women. CONCLUSION Our results do not support an association between genetic variants associated with circulating urate levels and risk of PD, but larger investigations are needed to determine whether the modest genetic effects on blood urate contribute to predict PD risk.
Collapse
Affiliation(s)
- Katherine C Hughes
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Xiang Gao
- Department of Nutritional Health, The Pennsylvania State University, University Park, PA, USA
| | - Eilis J O'Reilly
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; School of Public Health, College of Medicine, University College Cork, Ireland
| | - Iris Y Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marc G Weisskopf
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael A Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Alberto Ascherio
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
80
|
Wang JN, Zhang HJ, Li JQ, Ding WJ, Ma ZJ. Bioactive Indolocarbazoles from the Marine-Derived Streptomyces sp. DT-A61. JOURNAL OF NATURAL PRODUCTS 2018; 81:949-956. [PMID: 29558119 DOI: 10.1021/acs.jnatprod.7b01058] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nine new indolocarbazoles (1-9) were isolated from the marine-derived Streptomyces sp. DT-A61. Among them compounds 1-8 featured a hydroxy group at the C-3 or C-9 position. All purified compounds were identified by 1D and 2D NMR and HRESIMS data. The absolute configurations of 4-6, 8, and 9 were determined by electronic circular dichroism spectroscopic data. Compound 7 exhibited significant activity against human prostate PC-3 cancer cells with an IC50 value of 0.16 μM. Compounds 1, 5, 6, and 9 showed moderate inhibition against the same cell line with IC50 values of 8.0, 3.6, 3.1, and 5.6 μM. Compound 2 displayed a notable inhibitory effect against Rho-associated protein kinase (ROCK2) with an IC50 value of 5.7 nM, which was similar to the positive control staurosporine (IC50 7.8 nM).
Collapse
Affiliation(s)
- Jia-Nan Wang
- Institute of Marine Biology, Ocean College , Zhejiang University , No. 1 Zheda Road , Zhoushan 316021 , People's Republic of China
| | - Hao-Jian Zhang
- Institute of Marine Biology, Ocean College , Zhejiang University , No. 1 Zheda Road , Zhoushan 316021 , People's Republic of China
| | - Jia-Qi Li
- Institute of Marine Biology, Ocean College , Zhejiang University , No. 1 Zheda Road , Zhoushan 316021 , People's Republic of China
| | - Wan-Jing Ding
- Institute of Marine Biology, Ocean College , Zhejiang University , No. 1 Zheda Road , Zhoushan 316021 , People's Republic of China
| | - Zhong-Jun Ma
- Institute of Marine Biology, Ocean College , Zhejiang University , No. 1 Zheda Road , Zhoushan 316021 , People's Republic of China
| |
Collapse
|
81
|
Bi M, Jiao Q, Du X, Jiang H. Glut9-mediated Urate Uptake Is Responsible for Its Protective Effects on Dopaminergic Neurons in Parkinson's Disease Models. Front Mol Neurosci 2018; 11:21. [PMID: 29434538 PMCID: PMC5790811 DOI: 10.3389/fnmol.2018.00021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/15/2018] [Indexed: 01/10/2023] Open
Abstract
Considerable evidence has shown that elevated plasma or cerebrospinal fluid (CSF) urate levels correlated with a reduced risk of Parkinson’s disease (PD). Based on its anti-oxidative properties, urate might serve as one of promising neuroprotective candidates for PD. However, how urate is transported through cell membranes to exert its effects inside the cells in PD is largely unknown. To elucidate this, we showed that increased intracellular urate exerted its neuroprotective effects against 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity in MES23.5 cells and elevated urate could antagonize 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigral dopaminergic neuronal death in urate oxidase (UOx) knockout (KO) mice. Its transporter, glucose transporter type 9 (Glut9), was observed up-regulated, which was caused by the activation of p53. These protective effects could be abolished by Glut9 blocker and p53 inhibitor. These results suggested that Glut9 was a functional urate transporter, whose up-regulation by activation of p53 resulted in the increased intracellular urate levels in PD models. Our findings suggest that Glut9 could be modified to modulate urate levels in dopaminergic neurons and urate-elevating strategies without increasing systemic levels to avoid side effects might serve as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College, Qingdao University, Qingdao, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
82
|
Parkinson’s Disease: Contemporary Concepts and Clinical Management. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
83
|
O'Reilly ÉJ, Bjornevik K, Schwarzschild MA, McCullough ML, Kolonel LN, Le Marchand L, Manson JE, Ascherio A. Pre-diagnostic plasma urate and the risk of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2017; 19:194-200. [PMID: 29277115 DOI: 10.1080/21678421.2017.1418005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To prospectively examine for the first time the association between plasma urate levels measured in healthy participants and future amyotrophic lateral sclerosis (ALS) risk. METHODS A pooled case-control study nested in five US prospective cohorts comprising 319,617 participants who provided blood, of which 275 had ALS during follow-up. Pre-diagnostic plasma urate was determined for all participants using a clinical colorimetric enzyme assay. Gender-specific multivariable-adjusted rate ratios (RR) of ALS incidence or death estimated by conditional logistic regression and pooled using inverse-variance weighting. RESULTS In age- and matching factor-adjusted analyses, a 1 mg/dL increase in urate concentration was associated with RR = 0.88 (95% CI: [0.78, 0.997] p = 0.044). After adjustment for BMI, a strong predictor of ALS and urate levels, and other potential covariates, the RR = 0.89 (95% CI: [0.78, 1.02]; p = 0.08 for 1mg/dL increase in urate). CONCLUSION Elevation of plasma urate was modestly inversely associated with the risk of ALS and warrants further study for a potential role in this disease.
Collapse
Affiliation(s)
- Éilis J O'Reilly
- a School of Public Health , College of Medicine, University College Cork , Cork , Ireland.,b Department of Nutrition , Harvard TH Chan School of Public Health , Boston , MA , USA
| | - Kjetil Bjornevik
- c Department of Global Public Health and Primary Care , University of Bergen , Bergen , Norway.,d The Norwegian Multiple Sclerosis Competence Center, Department of Neurology , Haukeland University Hospital , Bergen , Norway
| | | | | | - Laurence N Kolonel
- g Epidemiology Program , University of Hawaii Cancer Center , Honolulu , HI , USA
| | - Loic Le Marchand
- g Epidemiology Program , University of Hawaii Cancer Center , Honolulu , HI , USA
| | - Joann E Manson
- h Department of Medicine Brigham and Women's Hospital , Harvard Medical School , Boston , MA , USA.,i Department of Epidemiology , Harvard TH Chan School of Public Health , Boston , MA , USA , and
| | - Alberto Ascherio
- b Department of Nutrition , Harvard TH Chan School of Public Health , Boston , MA , USA.,i Department of Epidemiology , Harvard TH Chan School of Public Health , Boston , MA , USA , and.,j Channing Division of Network Medicine , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| |
Collapse
|
84
|
Palacios N. Air pollution and Parkinson's disease - evidence and future directions. REVIEWS ON ENVIRONMENTAL HEALTH 2017; 32:303-313. [PMID: 28731859 DOI: 10.1515/reveh-2017-0009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/12/2017] [Indexed: 05/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease of unknown etiology that is thought to be caused by a complex combination of environmental and/or genetic factors. Air pollution exposure is linked to numerous adverse effects on human health, including brain inflammation and oxidative stress, processes that are believed to contribute to the development and progression of PD. This review provides an overview of recent advances in the epidemiology of air pollution and PD, including evidence of the effects of various pollutants (ozone, PM10, PM2.5, PM2.5-10, NOx, NO2, CO, traffic air pollution, second-hand smoking) on PD risk. Based on this evidence, promising opportunities for future research are outlined, including: (1) studies of smaller particle sizes that cross the blood-brain barrier, (2) studies of the effects of air pollution on PD mortality and/or progression; (3) studies of interactions of air pollution with gene environment and other environmental factors.
Collapse
|
85
|
Iwaki H, Ando R, Miyaue N, Tada S, Tsujii T, Yabe H, Nishikawa N, Nagai M, Nomoto M. One year safety and efficacy of inosine to increase the serum urate level for patients with Parkinson's disease in Japan. J Neurol Sci 2017; 383:75-78. [DOI: 10.1016/j.jns.2017.10.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 09/27/2017] [Accepted: 10/23/2017] [Indexed: 02/04/2023]
|
86
|
The Single Nucleotide Polymorphism rs1014290 of the SLC2A9 Gene Is Associated with Uric Acid Metabolism in Parkinson's Disease. PARKINSONS DISEASE 2017; 2017:7184927. [PMID: 29158942 PMCID: PMC5660784 DOI: 10.1155/2017/7184927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/14/2017] [Accepted: 08/22/2017] [Indexed: 11/18/2022]
Abstract
Individuals with Parkinson's disease (PD) have lower uric acid levels than those without PD, and the CC genotype and C minor allele of a single nucleotide polymorphism (SNP), rs1014290 of SLC2A9, are associated with lower uric acid levels. We investigated the association of rs1014290 with uric acid metabolism in a cohort of PD cases (220) and controls (110) in a Han Chinese population. Uric acid levels were determined and rs1014290 was assayed using a mutation-sensitive on/off switch technology. PD uric acid levels (291.65 ± 76.29 μmol/L) were significantly lower than the controls (325.73 ± 74.23 μmol/L, P < 0.001, t-test). Individuals with rs1014290 TT and CT genotypes had higher uric acid levels, and those with the CC genotype had the lowest uric acid levels among both control and PD cases. The CC genotype and the C minor allele were statistically more frequent in the PD group compared to the control group. Those with the CC genotype had a statistically significant higher risk of PD than those with the TT or TC genotype (odds ratio [OR] = 2.249, 95% confidence interval [CI]: 1.129–4.480, and P = 0.021). Thus, SLC2A9 rs1014290 is related to lower uric acid levels in PD patients and can be a risk factor for PD in the Han population.
Collapse
|
87
|
Yu Z, Zhang S, Wang D, Fan M, Gao F, Sun W, Li Z, Li S. The significance of uric acid in the diagnosis and treatment of Parkinson disease: An updated systemic review. Medicine (Baltimore) 2017; 96:e8502. [PMID: 29137045 PMCID: PMC5690738 DOI: 10.1097/md.0000000000008502] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/04/2017] [Accepted: 10/08/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Parkinson disease (PD) is a neurodegenerative disease characterized by chronic and progressive loss of dopaminergic neurons in substansia nigra pars compacta. Oxidative stress is proposed to play a critical role in the pathogenesis of PD. Uric acid (UA), as an important physiological antioxidant, is identified a molecular predictor associated with a decreased risk and a slower disease progression for PD and potential neuroprotectant of PD by increasing epidemiological and clinical evidences. Within this review, we will present a comprehensive overview of the data linking UA to PD in recent years. METHODS We searched PubMed, EMBASE, Web of Science databases for relevant studies. Any observational or experimental studies that evaluated UA and PD were our goal of searching the electric databases. RESULTS Twelve studies that evaluated UA and PD were identified in this review. We reviewed the roles of UA in the pathogenesis of PD, the association of UA with morbidity, severity/progression, nonmotor symptoms, motor complications of PD, with an attempt to provide new ideas for diagnosis and treatment in PD. CONCLUSION Our findings supported that lots of clinical and epidemiological data observed lower UA levels in PD patients. Manipulation of UA or its precursors' concentration could be effective to treat or prevent PD. However, it is still suspectable that higher UA levels are better enough to PD patients. Furthermore, for the complex nature of PD and its heterogeneous genetic and environmental influences, it is inadequate for just manipulating UA in treating the disease.
Collapse
Affiliation(s)
- Zhange Yu
- Department of Acupuncture, China-Japan Friendship Hospital, Beijing
| | - Shuai Zhang
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province
| | - Dongdong Wang
- Department of Orthopedics, Tumd Right Banner Hospital, Baotou City
| | - Meng Fan
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Fuqiang Gao
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Wei Sun
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Zirong Li
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Shiliang Li
- Department of Acupuncture, China-Japan Friendship Hospital, Beijing
| |
Collapse
|
88
|
Chahine LM, Stern MB. Parkinson's Disease Biomarkers: Where Are We and Where Do We Go Next? Mov Disord Clin Pract 2017; 4:796-805. [PMID: 30363472 DOI: 10.1002/mdc3.12545] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/09/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Background Objective measures of Parkinson's disease (PD) are needed for purposes of diagnosis and prognostication, as well as identification of those at risk of PD. In this qualitative review, we provide an overview of the current state of the field of PD biomarker development, delineate challenges, and discuss how the field is evolving. Methods A search of PubMed was conducted for articles pertaining to objective biomarkers for PD. Articles were selected based on relevance and methodology; where available, meta-analyses, systematic reviews, and comprehensive qualitative review articles were preferentially referenced. Results There are several potential sources of objective PD biomarkers including biofluids, peripheral tissue, imaging, genetics, and technology based objective motor testing. Approaches to biomarker identification include the candidate biomarker approach and unbiased discovery methods, each of which has advantages and disadvantages. Several emerging techniques hold promise in each of these areas. Advances in technology and bioinformatics, and the increasing availability of biobanks, are expected to facilitate future PD biomarker development. Conclusions The field of objective biomarkers for PD has made great progress but much remains to be done in translating putative biomarkers into tools useful in the clinic and for research. Multimodal biomarker platforms have the potential to capitalize on the utility and strengths of individual biomarkers. Rigorous methodology and standards for replication of findings will be key to meaningful progress in the field.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology Parkinson's Disease and Movement Disorders Center Perelman School of Medicine University of Pennsylvania Philadelphia PA
| | - Matthew B Stern
- Department of Neurology Parkinson's Disease and Movement Disorders Center Perelman School of Medicine University of Pennsylvania Philadelphia PA
| |
Collapse
|
89
|
Mikami T, Sorimachi M. Uric acid contributes greatly to hepatic antioxidant capacity besides protein. Physiol Res 2017; 66:1001-1007. [PMID: 28937257 DOI: 10.33549/physiolres.933555] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Uric acid is the end-product of purine nucleotide metabolism and an increase in uric acid concentration in the body results in hyperuricemia, ultimately leading to gout. However, uric acid is a potent antioxidant and interacts with reactive oxygen species (ROS) to be non-enzymatically converted to allantoin. Uric acid accounts for approximately 60 % of antioxidant capacity in the plasma; however, its contribution to tissue antioxidant capacity is unknown. In this study, the contribution of uric acid to tissue antioxidant capacity and its conversion to allantoin by scavenging ROS in tissue were examined. The results showed that a decrease in hepatic uric acid content via allopurinol administration significantly reduced hepatic total-radical trapping antioxidant parameter (TRAP) content in protein-free cytosol. Additionally, treating protein-free cytosol with uricase led to a further reduction of hepatic TRAP content. Allantoin was also detected in the solution containing protein-free cytosol that reacted with ROS. These findings suggest that in the absence of protein, uric acid contributes greatly to antioxidant capacity in the liver, where uric acid is converted to allantoin by scavenging ROS.
Collapse
Affiliation(s)
- T Mikami
- Department of Health and Sports Science, Nippon Medical School, Musashino, Tokyo, Japan.
| | | |
Collapse
|
90
|
Paganoni S, Nicholson K, Chan J, Shui A, Schoenfeld D, Sherman A, Berry J, Cudkowicz M, Atassi N. Urate levels predict survival in amyotrophic lateral sclerosis: Analysis of the expanded Pooled Resource Open-Access ALS clinical trials database. Muscle Nerve 2017; 57:430-434. [PMID: 28857199 DOI: 10.1002/mus.25950] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Urate has been identified as a predictor of amyotrophic lateral sclerosis (ALS) survival in some but not all studies. Here we leverage the recent expansion of the Pooled Resource Open-Access ALS Clinical Trials (PRO-ACT) database to study the association between urate levels and ALS survival. METHODS Pooled data of 1,736 ALS participants from the PRO-ACT database were analyzed. Cox proportional hazards regression models were used to evaluate associations between urate levels at trial entry and survival. RESULTS After adjustment for potential confounders (i.e., creatinine and body mass index), there was an 11% reduction in risk of reaching a survival endpoint during the study with each 1-mg/dL increase in uric acid levels (adjusted hazard ratio 0.89, 95% confidence interval 0.82-0.97, P < 0.01). DISCUSSION Our pooled analysis provides further support for urate as a prognostic factor for survival in ALS and confirms the utility of the PRO-ACT database as a powerful resource for ALS epidemiological research. Muscle Nerve 57: 430-434, 2018.
Collapse
Affiliation(s)
- Sabrina Paganoni
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge St, Suite 600 Boston, Massachusetts, 02114.,Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, Massachusetts.,VA Boston Healthcare System, Boston, Massachusetts
| | - Katharine Nicholson
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge St, Suite 600 Boston, Massachusetts, 02114
| | - James Chan
- Massachusetts General Hospital Biostatistics Center, Boston, Massachusetts
| | - Amy Shui
- Massachusetts General Hospital Biostatistics Center, Boston, Massachusetts
| | - David Schoenfeld
- Massachusetts General Hospital Biostatistics Center, Boston, Massachusetts
| | - Alexander Sherman
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge St, Suite 600 Boston, Massachusetts, 02114
| | - James Berry
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge St, Suite 600 Boston, Massachusetts, 02114
| | - Merit Cudkowicz
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge St, Suite 600 Boston, Massachusetts, 02114
| | - Nazem Atassi
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge St, Suite 600 Boston, Massachusetts, 02114
| | | |
Collapse
|
91
|
Tropea TF, Chen-Plotkin AS. Unlocking the mystery of biomarkers: A brief introduction, challenges and opportunities in Parkinson Disease. Parkinsonism Relat Disord 2017; 46 Suppl 1:S15-S18. [PMID: 28793971 DOI: 10.1016/j.parkreldis.2017.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 10/19/2022]
Abstract
First described 200 years ago, Parkinson Disease (PD) exhibits considerable heterogeneity in clinical presentation, as well as trajectory of motor and non-motor decline. This heterogeneity, in turn, complicates the planning of clinical research, particularly trials of disease-modifying therapies, as well as the care of PD patients. While clinical features have been used to delineate subgroups of PD patients, clinical subtyping is hampered by change in features over time, and clinical subtyping may fail to capture the biological processes underlying heterogeneity. In contrast, biomarkers - objective measures that serve as indicators of normal biological processes, pathogenic processes, or pharmacologic responses to therapeutic interventions - have promise to delineate molecularly-defined subgroups of PD patients who may be most likely to benefit from specific therapeutic interventions. Here we review the present role of genetic and biochemical biomarkers in PD. Moreover, we highlight areas where the use of biomarkers may benefit clinical trial planning, as well as clinical care through the application of a "precision medicine" approach, in the near term.
Collapse
Affiliation(s)
- Thomas F Tropea
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, United States
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, United States.
| |
Collapse
|
92
|
Abstract
Oxidative stress has been implicated as a core contributor to the initiation and progression of multiple neurological diseases. Genetic and environmental factors can produce oxidative stress through mitochondrial dysfunction leading to the degeneration of dopaminergic and other neurons underlying Parkinson disease (PD). Although clinical trials of antioxidants have thus far failed to demonstrate slowed progression of PD, oxidative stress remains a compelling target. Rather than prompting abandonment of antioxidant strategies, these failures have raised the bar for justifying drug and dosing selections and for improving study designs to test for disease modification by antioxidants. Urate, the main antioxidant found in plasma as well as the end product of purine metabolism in humans, has emerged as a promising potential neuroprotectant with advantages that distinguish it from previously tested antioxidant agents. Uniquely, higher urate levels in plasma or cerebrospinal fluid (CSF) have been linked to both a lower risk of developing PD and to a slower rate of its subsequent progression in numerous large prospective epidemiological and clinical cohorts. Laboratory evidence that urate confers neuroprotection in cellular and animal models of PD, possibly via the Nrf2 antioxidant response pathway, further strengthened its candidacy for rapid clinical translation. An early phase trial of the urate precursor inosine demonstrated its capacity to safely produce well tolerated, long-term elevation of plasma and CSF urate in early PD, supporting a phase 3 trial now underway to determine whether oral inosine dosed to elevate urate to concentrations predictive of favorable prognosis in PD slows clinical decline in people with recently diagnosed, dopamine transporter-deficient PD.
Collapse
Affiliation(s)
- Grace F Crotty
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Alberto Ascherio
- Departments of Epidemiology and Nutrition, Harvard School of Public Health, Boston, MA, USA
| | | |
Collapse
|
93
|
Gwinn K, David KK, Swanson-Fischer C, Albin R, Hillaire-Clarke CS, Sieber BA, Lungu C, Bowman FD, Alcalay RN, Babcock D, Dawson TM, Dewey RB, Foroud T, German D, Huang X, Petyuk V, Potashkin JA, Saunders-Pullman R, Sutherland M, Walt DR, West AB, Zhang J, Chen-Plotkin A, Scherzer CR, Vaillancourt DE, Rosenthal LS. Parkinson's disease biomarkers: perspective from the NINDS Parkinson's Disease Biomarkers Program. Biomark Med 2017; 11:451-473. [PMID: 28644039 PMCID: PMC5619098 DOI: 10.2217/bmm-2016-0370] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/11/2017] [Indexed: 11/21/2022] Open
Abstract
Biomarkers for Parkinson's disease (PD) diagnosis, prognostication and clinical trial cohort selection are an urgent need. While many promising markers have been discovered through the National Institute of Neurological Disorders and Stroke Parkinson's Disease Biomarker Program (PDBP) and other mechanisms, no single PD marker or set of markers are ready for clinical use. Here we discuss the current state of biomarker discovery for platforms relevant to PDBP. We discuss the role of the PDBP in PD biomarker identification and present guidelines to facilitate their development. These guidelines include: harmonizing procedures for biofluid acquisition and clinical assessments, replication of the most promising biomarkers, support and encouragement of publications that report negative findings, longitudinal follow-up of current cohorts including the PDBP, testing of wearable technologies to capture readouts between study visits and development of recently diagnosed (de novo) cohorts to foster identification of the earliest markers of disease onset.
Collapse
Affiliation(s)
- Katrina Gwinn
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karen K David
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Christine Swanson-Fischer
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Roger Albin
- Neurology Service & GRECC, VAAAHS, UM Udall Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Beth-Anne Sieber
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Codrin Lungu
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - F DuBois Bowman
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University, New York, NY, USA
| | - Debra Babcock
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ted M Dawson
- Neuroregeneration & Stem Cell Programs, Institute for Cell Engineering, Solomon H Snyder Department of Neuroscience, Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard B Dewey
- Department of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tatiana Foroud
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dwight German
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuemei Huang
- Department of Neurology, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Vlad Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Judith A Potashkin
- Department of Cellular & Molecular Pharmacology, Rosalind Franklin University of Medicine & Science, North Chicago, IL, USA
| | - Rachel Saunders-Pullman
- Department of Neurology, Mount Sinai Beth Israel & Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret Sutherland
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, MD, USA
| | - David R Walt
- Department of Chemistry, Tufts University, Medford, MA, USA
| | - Andrew B West
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jing Zhang
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Alice Chen-Plotkin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Clemens R Scherzer
- Department of Neurology, Harvard Medical School, Brigham & Women's Hospital, Cambridge, MA, USA
| | - David E Vaillancourt
- Departments of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL, USA
| | - Liana S Rosenthal
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
94
|
Huertas I, Jesús S, Lojo JA, García-Gómez FJ, Cáceres-Redondo MT, Oropesa-Ruiz JM, Carrillo F, Vargas-Gonzalez L, Martín Rodríguez JF, Gómez-Garre P, García-Solís D, Mir P. Lower levels of uric acid and striatal dopamine in non-tremor dominant Parkinson's disease subtype. PLoS One 2017; 12:e0174644. [PMID: 28358829 PMCID: PMC5373593 DOI: 10.1371/journal.pone.0174644] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/13/2017] [Indexed: 11/26/2022] Open
Abstract
Parkinson’s disease (PD) patients who present with tremor and maintain a predominance of tremor have a better prognosis. Similarly, PD patients with high levels of uric acid (UA), a natural neuroprotectant, have also a better disease course. Our aim was to investigate whether PD motor subtypes differ in their levels of UA, and if these differences correlate with the degree of dopamine transporter (DAT) availability. We included 75 PD patients from whom we collected information about their motor symptoms, DAT imaging and UA concentration levels. Based on the predominance of their motor symptoms, patients were classified into postural instability and gait disorder (PIGD, n = 36), intermediate (I, n = 22), and tremor-dominant (TD, n = 17) subtypes. The levels of UA and striatal DAT were compared across subtypes and the correlation between these two measures was also explored. We found that PIGD patients had lower levels of UA (3.7 vs 4.5 vs 5.3 mg/dL; P<0.001) and striatal DAT than patients with an intermediate or TD phenotype. Furthermore, UA levels significantly correlated with the levels of striatal DAT. We also observed that some PIGD (25%) and I (45%) patients had a predominance of tremor at disease onset. We speculate that UA might be involved in the maintenance of the less damaging TD phenotype and thus also in the conversion from TD to PIGD. Low levels of this natural antioxidant could lead to a major neuronal damage and therefore influence the conversion to a more severe motor phenotype.
Collapse
Affiliation(s)
- Ismael Huertas
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Antonio Lojo
- Servicio de Medicina Nuclear. UDIM. Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - María Teresa Cáceres-Redondo
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Juan Manuel Oropesa-Ruiz
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Fátima Carrillo
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Laura Vargas-Gonzalez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Juan Francisco Martín Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David García-Solís
- Servicio de Medicina Nuclear. UDIM. Hospital Universitario Virgen del Rocío, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- * E-mail:
| |
Collapse
|
95
|
Song JU, Hwang J, Ahn JK. Serum uric acid is positively associated with pulmonary function in Korean health screening examinees. Mod Rheumatol 2017; 27:1057-1065. [PMID: 28693364 DOI: 10.1080/14397595.2017.1285981] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To determine whether level of serum uric acid (SUA) is linked to pulmonary function in health screening examinees without overt medical conditions. METHODS We performed a cross-sectional study on 69,928 Koreans that participated in an annual health check. RESULTS Percent predicted forced vital capacity (FVC%) and forced expiratory volume in 1 s (FEV1%) were positively correlated with SUA in both genders (FVC%: r = .361; FEV1%: r = .314 in males and FVC%: r = .413; FEV1%: r = .382 in females, all p < .001). When the 2nd, 3rd and lowest FEV1% quartile were compared to the highest quartile group (the reference) by regression analysis, the adjusted ORs for hyperuricemia in males were 0.876 (95% CI, 0.809-0.949), 0.631 (0.574-0.695), and 0.311 (0.278-0.349), respectively. The adjusted ORs for hyperuricemia when the 2nd, 3rd and lowest FEV1% quartile were compared to the highest quartile in males were 0.791 (95% CI, 0.729-0.859), 0.565 (0.513-0.623), and 0.302 (0.270-0.337), respectively (p for trend <.001). Similarly, the adjusted OR of having hyperuricemia in females decreased significantly across FEV1% and FVC% quartile groups compared to the reference. CONCLUSIONS Hyperuricemia may have a positive effect on pulmonary function in middle aged healthy population.
Collapse
Affiliation(s)
- Jae-Uk Song
- a Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kangbuk Samsung Hospital , Sungkyunkwan University School of Medicine , Seoul , South Korea
| | - Jiwon Hwang
- b Division of Rheumatology, Department of Internal Medicine , National Police Hospital , Seoul , South Korea
| | - Joong Kyong Ahn
- c Division of Rheumatology, Department of Internal Medicine, Kangbuk Samsung Hospital , Sungkyunkwan University School of Medicine , Seoul , South Korea
| |
Collapse
|
96
|
Picillo M, Nicoletti A, Fetoni V, Garavaglia B, Barone P, Pellecchia MT. The relevance of gender in Parkinson’s disease: a review. J Neurol 2017; 264:1583-1607. [DOI: 10.1007/s00415-016-8384-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
|
97
|
Ouma S, Fukae J, Fujioka S, Yamamoto S, Hatano T, Yoritaka A, Okuma Y, Kashihara KI, Hattori N, Tsuboi Y. The Risk Factors for the Wearing-off Phenomenon in Parkinson's Disease in Japan: A Cross-sectional, Multicenter Study. Intern Med 2017; 56:1961-1966. [PMID: 28768964 PMCID: PMC5577070 DOI: 10.2169/internalmedicine.56.7667] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective Parkinson's disease (PD) is a common, progressive, neurodegenerative disorder. With progression of PD, the wearing-off phenomenon occurs more frequently as a motor complication, decreasing the patient's quality of life. The aim of this study was to investigate the risk factors for the wearing-off phenomenon in Japanese PD patients. Methods All of the study participants were clinically diagnosed as having PD. Each patient was assessed for the wearing-off phenomenon based on the findings of clinical assessments and interviews that were conducted during a single visit. The risk factors for wearing-off were analyzed by univariate and multivariate logistic regression analyses. Results Wearing-off was observed in 101 of the 180 (56.1%) patients who were enrolled in this study. The multivariate logistic regression analysis revealed that the onset of PD at ≥69 years of age (odds ratio [OR], 0.22; 95% confidence interval [CI], 0.05-0.88; p=0.032), female sex (OR, 6.49; 95% CI, 2.34-17.99; p<0.001), catechol-O-methyltransferase (COMT) inhibitor treatment (OR, 19.59; 95% CI, 3.55-108.11; p<0.001) and a high daily levodopa dosage (≥600 mg/day) (OR, 7.69; 95% CI, 1.41-41.84; p=0.018) were independent predictive factors for wearing-off in Japanese PD patients. Conclusion Age at the symptomatic disease onset, female sex, COMT inhibitor treatment, and a high daily levodopa dose were associated with the occurrence of wearing-off in Japanese PD patients. Physicians need to consider the risk factors and carefully choose medications for PD patients to postpone the occurrence of this phenomenon for as long as possible.
Collapse
Affiliation(s)
- Shinji Ouma
- Department of Neurology, Fukuoka University School of Medicine, Japan
| | - Jiro Fukae
- Department of Neurology, Fukuoka University School of Medicine, Japan
| | - Shinsuke Fujioka
- Department of Neurology, Fukuoka University School of Medicine, Japan
| | | | - Taku Hatano
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Asako Yoritaka
- Department of Neurology, Juntendo Koshigaya Hospital, Japan
| | - Yasuyuki Okuma
- Department of Neurology, Juntendo Shizuoka Hospital, Japan
| | | | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University School of Medicine, Japan
| |
Collapse
|
98
|
Paganoni S, Schwarzschild MA. Urate as a Marker of Risk and Progression of Neurodegenerative Disease. Neurotherapeutics 2017; 14:148-153. [PMID: 27995438 PMCID: PMC5233635 DOI: 10.1007/s13311-016-0497-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Urate is a naturally occurring antioxidant whose levels are associated with reduced risk of developing Parkinson's disease (PD) and Alzheimer's disease. Urate levels are also associated with favorable progression in PD, amyotrophic lateral sclerosis, Huntington's disease, and multisystem atrophy. These epidemiological data are consistent with laboratory studies showing that urate exhibits neuroprotective effects by virtue of its antioxidant properties in several preclinical models. This body of evidence supports the hypothesis that urate may represent a shared pathophysiologic mechanism across neurodegenerative diseases. Most importantly, beyond its role as a molecular predictor of disease risk and progression, urate may constitute a novel therapeutic target. Indeed, clinical trials of urate elevation in PD and amyotrophic lateral sclerosis are testing the impact of raising peripheral urate levels on disease outcomes. These studies will contribute to unraveling the neuroprotective potential of urate in human pathology. In parallel, preclinical experiments are deepening our understanding of the molecular pathways that underpin urate's activities. Altogether, these efforts will bring about new insights into the translational potential of urate, its determinants, and its targets and their relevance to neurodegeneration.
Collapse
Affiliation(s)
- Sabrina Paganoni
- Harvard Medical School, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA.
- VA Boston Healthcare System, Boston, MA, USA.
| | - Michael A Schwarzschild
- Harvard Medical School, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- MassGeneral Institute for Neurodegenerative Disease (MIND), Boston, MA, USA
| |
Collapse
|
99
|
Lower Serum Bilirubin and Uric Acid Concentrations in Patients with Parkinson's Disease in China. Cell Biochem Biophys 2016; 72:49-56. [PMID: 25449297 DOI: 10.1007/s12013-014-0402-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The objective of the study is to investigate the correlation between bilirubin and uric acid (UA) concentrations and symptoms of Parkinson's disease (PD) in Chinese population. A total of 425 PD patients and 460 controls were included in the current study. Patients were diagnosed by a neurologist and assessed using the Hoehn & Yahr (H&Y) scale. Venous blood samples were collected, and bilirubin and UA concentrations were analyzed. Compared to controls, indirect bilirubin (IBIL) and UA concentrations were lower in PD patients (P IBIL = 0.015, P UA = 0.000). Serum IBIL in different age subgroups and H&Y stage subgroups were also lower compared to the control group (P IBIL = 0.000, P UA = 0.000) but were not significantly different among these subgroups. Females in the control group had significantly lower serum IBIL and UA concentrations than males (P IBIL = 0.000, P UA = 0.000) and the PD group (P IBIL = 0.027, P UA = 0.000). In early PD (patients with <2-year medical history and no treatment), serum IBIL and UA concentrations were also lower than the controls (P IBIL = 0.013, P UA = 0.000). Although IBIL concentration was positively correlated with UA concentration in controls (R IBIL = 0.229, P IBIL = 0.004), this positive association was not observed in the PD group (R IBIL = -0.032, P IBIL = 0.724). Decreased levels of serum IBIL and UA were observed in PD patients. It is possible that individuals with decreased serum bilirubin and UA concentrations lack the endogenous defense system to prevent peroxynitrite and other free radicals from damaging and destroying dopaminergic cells in the substantia nigra. Our results provide a basis for further investigation into the role of bilirubin in PD.
Collapse
|
100
|
Hatefi M, Dastjerdi MM, Ghiasi B, Rahmani A. Association of Serum Uric Acid Level with the Severity of Brain Injury and Patient's Outcome in Severe Traumatic Brain Injury. J Clin Diagn Res 2016; 10:OC20-OC24. [PMID: 28208906 DOI: 10.7860/jcdr/2016/21918.8993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/25/2016] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The prognostic value of serum Uric Acid (UA) levels in Traumatic Brain Injury (TBI) is unclear. AIM To investigate the relationship between serum UA levels and prognosis of patients with TBI when in hospital and at six months after discharge. MATERIALS AND METHODS All patients attended our emergency department during July 2014 and December 2015 and were consecutively entered into the study and among 890 evaluated candidates based on inclusion criteria we finally investigated the serum UA levels of 725 TBI patients. Computed Tomography (CT) images of the brain were obtained within the first 24 hours of hospitalization. Outcome was assessed using the Glasgow Outcome Scale (GOS) score at discharge and at six months after discharge. RESULTS Data of 725 patients (42.89% men; mean age: 54.69±12.37 years) were analyzed. Mean±Standard Deviation (SD) of Glasgow Coma Scale (GCS) scores was 4.65±1.76. Serum levels of UA, when in hospital and at six months after discharge, among those who died were lower than those who survived (in hospital: 0.126±0.026 vs. 0.243±0.942 mmol/l, p = 0.000; 6 months post-discharge: 0.130±0.044 vs. 0.286±0.069 mmol/l, p<0.001). The mean UA plasma was significantly different between deceased and alive patients according to GOS scores (p<0.001 and p=0.030, respectively). The UA levels showed a significant relationship with GCS scores and severity of brain injury assessed using the Marshall Classification Score (p=0.005). CONCLUSION Our results showed a strong relationship between UA levels and patients' outcomes either in hospital or at six months after discharge. Serum UA level could be considered as a valuable marker for evaluating the severity of brain injury and outcomes of TBI.
Collapse
Affiliation(s)
- Masoud Hatefi
- Assistant Professor, Department of Neurosurgery, Faculty of Medicine, Ilam University of Medical Science , Ilam, Iran
| | - Masoud Moghadas Dastjerdi
- Assistant Professor, Department of Emergency Medicine, School of Medicine, Isfahan University of Medical Science , Isfahan, Iran
| | - Bahareh Ghiasi
- Assistant Professor, Department of Nephrology, Faculty of Medicine, Ilam University of Medical Science , Ilam, Iran
| | - Asghar Rahmani
- Student's Researches Committee, Ilam University of Medical Sciences , Ilam, Iran
| |
Collapse
|