101
|
Gureev AP, Popov VN. Nrf2/ARE Pathway as a Therapeutic Target for the Treatment of Parkinson Diseases. Neurochem Res 2019; 44:2273-2279. [PMID: 30617864 DOI: 10.1007/s11064-018-02711-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 02/07/2023]
Abstract
Instead of the progress in the understanding of etiology of Parkinson's disease (PD), effective methods to prevent the progression of the disease have not been developed and only symptomatic treatment is currently possible. One of possible pathways to slow the progression of the disease is protection of dopaminergic neurons by maintaining mitochondrial quality control in neuron cells. Recent studies showed that the most promising target for pharmacological effects on mitochondria is the Nrf2/ARE signaling cascade. It participates in the maintenance of mitochondrial homeostasis, which is provided by an optimal ratio in the processes of mitochondrial biogenesis and mitophagy, as well as the optimal ratio of ROS production and ROS scavenging. Nrf2 activators are capable of modulating these processes, maintaining mitochondrial homeostasis in neurons. In addition, Nrf2 can synergistically interact with other transcription factors, for example, PGC-1a in the regulation of mitochondrial biogenesis and YY1 with the increase of antioxidant defense. All this makes Nrf2 an optimal target for drugs that could support the mitochondrial quality control, which, in combination with antioxidant protection, can significantly slow down the pathogenesis of PD. Some of these compounds have undergone laboratory studies and are at the stage of clinical trials now.
Collapse
Affiliation(s)
- Artem P Gureev
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, Voronezh, Russia.
| | - Vasily N Popov
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, Voronezh, Russia
| |
Collapse
|
102
|
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
|
103
|
Heyes N, Kapoor P, Kerr ID. Polymorphisms of the Multidrug Pump ABCG2: A Systematic Review of Their Effect on Protein Expression, Function, and Drug Pharmacokinetics. Drug Metab Dispos 2018; 46:1886-1899. [PMID: 30266733 DOI: 10.1124/dmd.118.083030] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022] Open
Abstract
The widespread expression and polyspecificity of the multidrug ABCG2 efflux transporter make it an important determinant of the pharmacokinetics of a variety of substrate drugs. Null ABCG2 expression has been linked to the Junior blood group. Polymorphisms affecting the expression or function of ABCG2 may have clinically important roles in drug disposition and efficacy. The most well-studied single nucleotide polymorphism (SNP), Q141K (421C>A), is shown to decrease ABCG2 expression and activity, resulting in increased total drug exposure and decreased resistance to various substrates. The effect of Q141K can be rationalized by inspection of the ABCG2 structure, and the effects of this SNP on protein processing may make it a target for pharmacological intervention. The V12M SNP (34G>A) appears to improve outcomes in cancer patients treated with tyrosine kinase inhibitors, but the reasons for this are yet to be established, and this residue's role in the mechanism of the protein is unexplored by current biochemical and structural approaches. Research into the less-common polymorphisms is confined to in vitro studies, with several polymorphisms shown to decrease resistance to anticancer agents such as SN-38 and mitoxantrone. In this review, we present a systematic analysis of the effects of ABCG2 polymorphisms on ABCG2 function and drug pharmacokinetics. Where possible, we use recent structural advances to present a molecular interpretation of the effects of SNPs and indicate where we need further in vitro experiments to fully resolve how SNPs impact ABCG2 function.
Collapse
Affiliation(s)
- Niall Heyes
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Parth Kapoor
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Ian D Kerr
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
104
|
Mollenhauer B, Zimmermann J, Sixel‐Döring F, Focke NK, Wicke T, Ebentheuer J, Schaumburg M, Lang E, Friede T, Trenkwalder C. Baseline predictors for progression 4 years after Parkinson's disease diagnosis in the De Novo Parkinson Cohort (DeNoPa). Mov Disord 2018; 34:67-77. [DOI: 10.1002/mds.27492] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/26/2022] Open
Affiliation(s)
- Brit Mollenhauer
- Paracelsus‐Elena‐Klinik Kassel Germany
- Department of Neurology University Medical Centre Goettingen Goettingen Germany
| | | | | | - Niels K. Focke
- Department of Clinical Neurophysiology University Medical Centre Goettingen Goettingen Germany
| | | | | | | | | | - Tim Friede
- Department of Medical Statistics University Medical Centre Goettingen Goettingen Germany
| | - Claudia Trenkwalder
- Paracelsus‐Elena‐Klinik Kassel Germany
- Department of Neurosurgery University Medical Centre GoettingenGoettingen Germany
| | | |
Collapse
|
105
|
Dissociation between urate and blood pressure in mice and in people with early Parkinson's disease. EBioMedicine 2018; 37:259-268. [PMID: 30415890 PMCID: PMC6284456 DOI: 10.1016/j.ebiom.2018.10.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/05/2018] [Accepted: 10/12/2018] [Indexed: 02/02/2023] Open
Abstract
Background Epidemiological, laboratory and clinical studies have established an association between elevated urate and high blood pressure (BP). However, the inference of causality remains controversial. A naturally occurring antioxidant, urate may also be neuroprotective, and urate-elevating treatment with its precursor inosine is currently under clinical development as a potential disease-modifying strategy for Parkinson's disease (PD). Methods Our study takes advantage of a recently completed phase II trial evaluating oral inosine in de novo non-disabling early PD with no major cardiovascular and nephrological conditions, and of three lines of genetically engineered mice: urate oxidase (UOx) global knockout (gKO), conditional KO (cKO), and transgenic (Tg) mice with markedly elevated, mildly elevated, and substantially reduced serum urate, respectively, to systematically investigate effects of urate-modifying manipulation on BP. Findings Among clinical trial participants, change in serum urate but not changes in systolic, diastolic and orthostatic BP differed by treatment group. There was no positive correlation between urate elevations and changes in systolic, diastolic and orthostatic BP ((p = .05 (in inverse direction), 0.30 and 0.63, respectively)). Between UOx gKO, cKO, or Tg mice and their respective wildtype littermates there were no significant differences in systolic or diastolic BP or in their responses to BP-regulating interventions. Interpretation Our complementary preclinical and human studies of urate modulation in animal models and in generally healthy early PD do not support a hypertensive effect of urate elevation or an association between urate and BP. Fund U.S. Department of Defense, RJG Foundation, Michael J. Fox Foundation LEAPS program, National Institutes of Health, American Federation for Aging Research, Parkinson's Disease Foundation Advancing Parkinson's Therapies initiative.
Collapse
|
106
|
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
|
107
|
Chinese Multidisciplinary Expert Consensus on the Diagnosis and Treatment of Hyperuricemia and Related Diseases. Chin Med J (Engl) 2018; 130:2473-2488. [PMID: 29052570 PMCID: PMC5684625 DOI: 10.4103/0366-6999.216416] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
108
|
Taga A, Maragakis NJ. Current and emerging ALS biomarkers: utility and potential in clinical trials. Expert Rev Neurother 2018; 18:871-886. [DOI: 10.1080/14737175.2018.1530987] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Arens Taga
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
109
|
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
|
110
|
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
|
111
|
Phillips MCL, Murtagh DKJ, Gilbertson LJ, Asztely FJS, Lynch CDP. Low-fat versus ketogenic diet in Parkinson's disease: A pilot randomized controlled trial. Mov Disord 2018; 33:1306-1314. [PMID: 30098269 PMCID: PMC6175383 DOI: 10.1002/mds.27390] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/15/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Preliminary evidence suggests that diet manipulation may influence motor and nonmotor symptoms in PD, but conflict exists regarding the ideal fat to carbohydrate ratio. Objectives: We designed a pilot randomized, controlled trial to compare the plausibility, safety, and efficacy of a low‐fat, high‐carbohydrate diet versus a ketogenic diet in a hospital clinic of PD patients. Methods: We developed a protocol to support PD patients in a diet study and randomly assigned patients to a low‐fat or ketogenic diet. Primary outcomes were within‐ and between‐group changes in MDS‐UPDRS Parts 1 to 4 over 8 weeks. Results: We randomized 47 patients, of which 44 commenced the diets and 38 completed the study (86% completion rate for patients commencing the diets). The ketogenic diet group maintained physiological ketosis. Both groups significantly decreased their MDS‐UPDRS scores, but the ketogenic group decreased more in Part 1 (−4.58 ± 2.17 points, representing a 41% improvement in baseline Part 1 scores) compared to the low‐fat group (−0.99 ± 3.63 points, representing an 11% improvement) (P < 0.001), with the largest between‐group decreases observed for urinary problems, pain and other sensations, fatigue, daytime sleepiness, and cognitive impairment. There were no between‐group differences in the magnitude of decrease for Parts 2 to 4. The most common adverse effects were excessive hunger in the low‐fat group and intermittent exacerbation of the PD tremor and/or rigidity in the ketogenic group. Conclusions: It is plausible and safe for PD patients to maintain a low‐fat or ketogenic diet for 8 weeks. Both diet groups significantly improved in motor and nonmotor symptoms; however, the ketogenic group showed greater improvements in nonmotor symptoms. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | | | | | - Fredrik J S Asztely
- Department of Neurology, Waikato Hospital, Hamilton, New Zealand.,Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
112
|
Abstract
The development of an intervention to slow or halt disease progression remains the greatest unmet therapeutic need in Parkinson's disease. Given the number of failures of various novel interventions in disease-modifying clinical trials in combination with the ever-increasing costs and lengthy processes for drug development, attention is being turned to utilizing existing compounds approved for other indications as novel treatments in Parkinson's disease. Advances in rational and systemic drug repurposing have identified a number of drugs with potential benefits for Parkinson's disease pathology and offer a potentially quicker route to drug discovery. Here, we review the safety and potential efficacy of the most promising candidates repurposed as potential disease-modifying treatments for Parkinson's disease in the advanced stages of clinical testing.
Collapse
Affiliation(s)
- Dilan Athauda
- Department of Clinical and Movement Neurosciences, UCL Institute of Neurology and National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Institute of Neurology and National Hospital for Neurology & Neurosurgery, Queen Square, London, UK.
| |
Collapse
|
113
|
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]
|
114
|
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
|
115
|
Kakkar AK, Singh H, Medhi B. Old wines in new bottles: Repurposing opportunities for Parkinson's disease. Eur J Pharmacol 2018; 830:115-127. [PMID: 29689247 DOI: 10.1016/j.ejphar.2018.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is a chronic progressive neurological disorder characterized by accumulation of Lewy bodies and profound loss of substantia nigra dopaminergic neurons. PD symptomatology is now recognized to include both cardinal motor as well as clinically significant non-motor symptoms. Despite intensive research, the current understanding of molecular mechanisms underlying neurodegeneration in PD is limited and has hampered the development of novel symptomatic and disease modifying therapies. The currently available treatment options are only partially or transiently effective and fail to restore the lost dopaminergic neurons or retard disease progression. Given the escalating drug development costs, lengthening timelines and declining R&D efficiency, industry and academia are increasingly focusing on ways to repurpose existing molecules as an accelerated route for drug discovery. The field of PD therapeutics is witnessing vigorous repurposing activity supported by big data analytics, computational models, and high-throughput drug screening systems. Here we review the mechanisms, efficacy, and safety of several emerging drugs currently aspiring to be repositioned for PD pharmacotherapy.
Collapse
Affiliation(s)
- Ashish Kumar Kakkar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
116
|
Cardoso F. Vitamin B12 and Parkinson's Disease: What is the Relationship? Mov Disord 2018; 33:702-703. [PMID: 29508925 DOI: 10.1002/mds.27366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Francisco Cardoso
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, The Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
117
|
Christine CW, Auinger P, Joslin A, Yelpaala Y, Green R. Vitamin B12 and Homocysteine Levels Predict Different Outcomes in Early Parkinson's Disease. Mov Disord 2018; 33:762-770. [DOI: 10.1002/mds.27301] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/12/2017] [Accepted: 12/31/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Peggy Auinger
- Center for Human Experimental Therapeutics; University of Rochester; Rochester New York USA
| | - Amelia Joslin
- Department of Pathology and Laboratory Medicine; UC Davis; Davis California USA
| | - Yuora Yelpaala
- Department of Pathology and Laboratory Medicine; UC Davis; Davis California USA
| | - Ralph Green
- Department of Pathology and Laboratory Medicine; UC Davis; Davis California USA
| | | |
Collapse
|
118
|
Stoessel D, Schulte C, Teixeira dos Santos MC, Scheller D, Rebollo-Mesa I, Deuschle C, Walther D, Schauer N, Berg D, Nogueira da Costa A, Maetzler W. Promising Metabolite Profiles in the Plasma and CSF of Early Clinical Parkinson's Disease. Front Aging Neurosci 2018; 10:51. [PMID: 29556190 PMCID: PMC5844983 DOI: 10.3389/fnagi.2018.00051] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) shows high heterogeneity with regard to the underlying molecular pathogenesis involving multiple pathways and mechanisms. Diagnosis is still challenging and rests entirely on clinical features. Thus, there is an urgent need for robust diagnostic biofluid markers. Untargeted metabolomics allows establishing low-molecular compound biomarkers in a wide range of complex diseases by the measurement of various molecular classes in biofluids such as blood plasma, serum, and cerebrospinal fluid (CSF). Here, we applied untargeted high-resolution mass spectrometry to determine plasma and CSF metabolite profiles. We semiquantitatively determined small-molecule levels (≤1.5 kDa) in the plasma and CSF from early PD patients (disease duration 0-4 years; n = 80 and 40, respectively), and sex- and age-matched controls (n = 76 and 38, respectively). We performed statistical analyses utilizing partial least square and random forest analysis with a 70/30 training and testing split approach, leading to the identification of 20 promising plasma and 14 CSF metabolites. These metabolites differentiated the test set with an AUC of 0.8 (plasma) and 0.9 (CSF). Characteristics of the metabolites indicate perturbations in the glycerophospholipid, sphingolipid, and amino acid metabolism in PD, which underscores the high power of metabolomic approaches. Further studies will enable to develop a potential metabolite-based biomarker panel specific for PD.
Collapse
Affiliation(s)
- Daniel Stoessel
- Metabolomic Discoveries GmbH, Potsdam, Germany
- Department of Biochemistry and Biology, Universität Potsdam, Potsdam, Germany
- Max Planck Institute für Molekulare Pflanzenphysiologie, Potsdam, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, German Center for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | | | | | - Irene Rebollo-Mesa
- Exploratory Statistics, Global Exploratory Development, UCB Pharma SA, Slough, United Kingdom
| | - Christian Deuschle
- Department of Neurodegeneration, German Center for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Dirk Walther
- Department of Biochemistry and Biology, Universität Potsdam, Potsdam, Germany
- Max Planck Institute für Molekulare Pflanzenphysiologie, Potsdam, Germany
| | | | - Daniela Berg
- Department of Neurodegeneration, German Center for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Andre Nogueira da Costa
- Experimental Medicine and Diagnostics, Global Exploratory Development, UCB Biopharma SPRL, Brussels, Belgium
| | - Walter Maetzler
- Department of Neurodegeneration, German Center for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
119
|
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
|
120
|
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
|
121
|
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]
|
122
|
Yasutake Y, Tomita K, Higashiyama M, Furuhashi H, Shirakabe K, Takajo T, Maruta K, Sato H, Narimatsu K, Yoshikawa K, Okada Y, Kurihara C, Watanabe C, Komoto S, Nagao S, Matsuo H, Miura S, Hokari R. Uric acid ameliorates indomethacin-induced enteropathy in mice through its antioxidant activity. J Gastroenterol Hepatol 2017; 32:1839-1845. [PMID: 28295549 DOI: 10.1111/jgh.13785] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIM Uric acid is excreted from blood into the intestinal lumen, yet the roles of uric acid in intestinal diseases remain to be elucidated. The study aimed to determine whether uric acid could reduce end points associated with nonsteroidal anti-inflammatory drug (NSAID)-induced enteropathy. METHODS A mouse model of NSAID-induced enteropathy was generated by administering indomethacin intraperitoneally to 8-week-old male C57BL/6 mice, and then vehicle or uric acid was administered orally. A group of mice treated with indomethacin was also concurrently administered inosinic acid, a uric acid precursor, and potassium oxonate, an inhibitor of uric acid metabolism, intraperitoneally. For in vitro analysis, Caco-2 cells treated with indomethacin were incubated in the presence or absence of uric acid. RESULTS Oral administration of uric acid ameliorated NSAID-induced enteropathy in mice even though serum uric acid levels did not increase. Intraperitoneal administration of inosinic acid and potassium oxonate significantly elevated serum uric acid levels and ameliorated NSAID-induced enteropathy in mice. Both oral uric acid treatment and intraperitoneal treatment with inosinic acid and potassium oxonate significantly decreased lipid peroxidation in the ileum of mice with NSAID-induced enteropathy. Treatment with uric acid protected Caco-2 cells from indomethacin-induced oxidative stress, lipid peroxidation, and cytotoxicity. CONCLUSIONS Uric acid within the intestinal lumen and in serum had a protective effect against NSAID-induced enteropathy in mice, through its antioxidant activity. Uric acid could be a promising therapeutic target for NSAID-induced enteropathy.
Collapse
Affiliation(s)
- Yuichi Yasutake
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Kengo Tomita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Masaaki Higashiyama
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Hirotaka Furuhashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Kazuhiko Shirakabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Takeshi Takajo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Koji Maruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Hirokazu Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Kazuyuki Narimatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Kenichi Yoshikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Yoshikiyo Okada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Chie Kurihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Chikako Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Shunsuke Komoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Shigeaki Nagao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Soichiro Miura
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| | - Ryota Hokari
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, Tokorozawa-shi, Saitama, Japan
| |
Collapse
|
123
|
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
|
124
|
Leehey M, Luo S, Sharma S, Wills AMA, Bainbridge JL, Wong PS, Simon DK, Schneider J, Zhang Y, Pérez A, Dhall R, Christine CW, Singer C, Cambi F, Boyd JT. Association of metabolic syndrome and change in Unified Parkinson's Disease Rating Scale scores. Neurology 2017; 89:1789-1794. [PMID: 28972194 DOI: 10.1212/wnl.0000000000004572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/28/2017] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To explore the association between metabolic syndrome and the Unified Parkinson's Disease Rating Scale (UPDRS) scores and, secondarily, the Symbol Digit Modalities Test (SDMT). METHODS This is a secondary analysis of data from 1,022 of 1,741 participants of the National Institute of Neurological Disorders and Stroke Exploratory Clinical Trials in Parkinson Disease Long-Term Study 1, a randomized, placebo-controlled trial of creatine. Participants were categorized as having or not having metabolic syndrome on the basis of modified criteria from the National Cholesterol Education Program Adult Treatment Panel III. Those who had the same metabolic syndrome status at consecutive annual visits were included. The change in UPDRS and SDMT scores from randomization to 3 years was compared in participants with and without metabolic syndrome. RESULTS Participants with metabolic syndrome (n = 396) compared to those without (n = 626) were older (mean [SD] 63.9 [8.1] vs 59.9 [9.4] years; p < 0.0001), were more likely to be male (75.3% vs 57.0%; p < 0.0001), and had a higher mean uric acid level (men 5.7 [1.3] vs 5.3 [1.1] mg/dL, women 4.9 [1.3] vs 3.9 [0.9] mg/dL, p < 0.0001). Participants with metabolic syndrome experienced an additional 0.6- (0.2) unit annual increase in total UPDRS (p = 0.02) and 0.5- (0.2) unit increase in motor UPDRS (p = 0.01) scores compared with participants without metabolic syndrome. There was no difference in the change in SDMT scores. CONCLUSIONS Persons with Parkinson disease meeting modified criteria for metabolic syndrome experienced a greater increase in total UPDRS scores over time, mainly as a result of increases in motor scores, compared to those who did not. Further studies are needed to confirm this finding. CLINICALTRIALSGOV IDENTIFIER NCT00449865.
Collapse
Affiliation(s)
- Maureen Leehey
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC.
| | - Sheng Luo
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Saloni Sharma
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Anne-Marie A Wills
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Jacquelyn L Bainbridge
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Pei Shieen Wong
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - David K Simon
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Jay Schneider
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Yunxi Zhang
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Adriana Pérez
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Rohit Dhall
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Chadwick W Christine
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Carlos Singer
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Franca Cambi
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - James T Boyd
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| |
Collapse
|
125
|
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
|
126
|
Havelund JF, Heegaard NHH, Færgeman NJK, Gramsbergen JB. Biomarker Research in Parkinson's Disease Using Metabolite Profiling. Metabolites 2017; 7:E42. [PMID: 28800113 PMCID: PMC5618327 DOI: 10.3390/metabo7030042] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 01/08/2023] Open
Abstract
Biomarker research in Parkinson's disease (PD) has long been dominated by measuring dopamine metabolites or alpha-synuclein in cerebrospinal fluid. However, these markers do not allow early detection, precise prognosis or monitoring of disease progression. Moreover, PD is now considered a multifactorial disease, which requires a more precise diagnosis and personalized medication to obtain optimal outcome. In recent years, advanced metabolite profiling of body fluids like serum/plasma, CSF or urine, known as "metabolomics", has become a powerful and promising tool to identify novel biomarkers or "metabolic fingerprints" characteristic for PD at various stages of disease. In this review, we discuss metabolite profiling in clinical and experimental PD. We briefly review the use of different analytical platforms and methodologies and discuss the obtained results, the involved metabolic pathways, the potential as a biomarker and the significance of understanding the pathophysiology of PD. Many of the studies report alterations in alanine, branched-chain amino acids and fatty acid metabolism, all pointing to mitochondrial dysfunction in PD. Aromatic amino acids (phenylalanine, tyrosine, tryptophan) and purine metabolism (uric acid) are also altered in most metabolite profiling studies in PD.
Collapse
Affiliation(s)
- Jesper F Havelund
- Villum Centre for Bioanalytical Sciences, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark.
| | - Niels H H Heegaard
- Department of Autoimmunology and Biomarkers, Statens Serum Institute, DK-2300 Copenhagen, Denmark.
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark.
| | - Nils J K Færgeman
- Villum Centre for Bioanalytical Sciences, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark.
| | - Jan Bert Gramsbergen
- Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark.
| |
Collapse
|
127
|
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
|
128
|
Havelund JF, Andersen AD, Binzer M, Blaabjerg M, Heegaard NHH, Stenager E, Faergeman NJ, Gramsbergen JB. Changes in kynurenine pathway metabolism in Parkinson patients with L-DOPA-induced dyskinesia. J Neurochem 2017. [PMID: 28628213 DOI: 10.1111/jnc.14104] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
L-3,4-Dihydroxyphenylalanine (L-DOPA) is the most effective drug in the symptomatic treatment of Parkinson's disease, but chronic use is associated with L-DOPA-induced dyskinesia in more than half the patients after 10 years of treatment. L-DOPA treatment may affect tryptophan metabolism via the kynurenine pathway. Altered levels of kynurenine metabolites can affect glutamatergic transmission and may play a role in the development of L-DOPA-induced dyskinesia. In this study, we assessed kynurenine metabolites in plasma and cerebrospinal fluid of Parkinson's disease patients and controls. Parkinson patients (n = 26) were clinically assessed for severity of motor symptoms (UPDRS) and L-DOPA-induced dyskinesia (UDysRS). Plasma and cerebrospinal fluid samples were collected after overnight fasting and 1-2 h after intake of L-DOPA or other anti-Parkinson medication. Metabolites were analyzed in plasma and cerebrospinal fluid of controls (n = 14), Parkinson patients receiving no L-DOPA (n = 8), patients treated with L-DOPA without dyskinesia (n = 8), and patients with L-DOPA-induced dyskinesia (n = 10) using liquid chromatography-mass spectrometry. We observed approximately fourfold increase in the 3-hydroxykynurenine/kynurenic acid ratio in plasma of Parkinson's patients with L-DOPA-induced dyskinesia. Anthranilic acid levels were decreased in plasma and cerebrospinal fluid of this patient group. 5-Hydroxytryptophan levels were twofold increased in all L-DOPA-treated Parkinson's patients. We conclude that a higher 3-hydroxykynurenine/kynurenic acid ratio in plasma may serve as a biomarker for L-DOPA-induced dyskinesia. Longitudinal studies including larger patients cohorts are needed to verify whether the changes observed here may serve as a prognostic marker for L-DOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Jesper F Havelund
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Andreas D Andersen
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.,Focused Research Unit in Neurology, Hospital of Southern Jutland, Aabenraa, Denmark.,Department of Neurology, Hospital of Southern Jutland, Sønderborg, Denmark.,Odense Patient data Exploratory Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Michael Binzer
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.,Focused Research Unit in Neurology, Hospital of Southern Jutland, Aabenraa, Denmark
| | - Morten Blaabjerg
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Niels H H Heegaard
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Autoimmunology & Biomarkers, Statens Serum Institut, Copenhagen, Denmark
| | - Egon Stenager
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.,Focused Research Unit in Neurology, Hospital of Southern Jutland, Aabenraa, Denmark
| | - Nils J Faergeman
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Jan Bert Gramsbergen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
129
|
Synthesis and characterisation of non-bonded 1.7 μm thin-shell (TS1.7-100 nm) silica particles for the rapid separation and analysis of uric acid and creatinine in human urine by hydrophilic interaction chromatography. J Chromatogr A 2017; 1506:37-44. [DOI: 10.1016/j.chroma.2017.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/04/2017] [Accepted: 05/02/2017] [Indexed: 02/04/2023]
|
130
|
Hui M, Carr A, Cameron S, Davenport G, Doherty M, Forrester H, Jenkins W, Jordan KM, Mallen CD, McDonald TM, Nuki G, Pywell A, Zhang W, Roddy E. The British Society for Rheumatology Guideline for the Management of Gout. Rheumatology (Oxford) 2017; 56:e1-e20. [DOI: 10.1093/rheumatology/kex156] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Indexed: 12/13/2022] Open
|
131
|
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
|
132
|
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
|
133
|
Fukae J, Fujioka S, Yanamoto S, Mori A, Nomi T, Hatano T, Fukuhara K, Ouma S, Hattori N, Tsuboi Y. Serum uric acid level is linked to the disease progression rate in male patients with multiple system atrophy. Clin Neurol Neurosurg 2017; 158:15-19. [PMID: 28432997 DOI: 10.1016/j.clineuro.2017.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/26/2017] [Accepted: 04/02/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Multiple system atrophy (MSA) is a progressive neurodegenerative disorder that may be caused in part by oxidative stress. Uric acid (UA) protects neurons in neurodegenerative disorders via antioxidative effects. The aim of this study was to investigate the relationship between the serum UA concentration and disease progression in MSA patients. PATIENTS AND METHODS A total of 53 Japanese MSA patients were enrolled in this study. The disease progression rate was estimated by the rate of global disability scale change per year. The relationship between the serum UA concentration and disease progression was assessed by Spearman's correlation analysis. Disease progression depending on the UA concentration was also estimated by multivariate logistic regression analysis. RESULTS MSA patients with the highest serum UA concentration had lower disease progression rates than those with the lowest concentration. Spearman's correlation analysis showed an inverse correlation between the serum UA concentration and disease progression in male patients. Multivariate logistic regression analysis confirmed that the UA concentration was independently related to disease progression only in male patients. CONCLUSION These results suggest that serum UA may be associated with disease progression in male patients with MSA.
Collapse
Affiliation(s)
- Jiro Fukae
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan; Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Shinsuke Fujioka
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Shosaburo Yanamoto
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Akio Mori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takahiro Nomi
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kousuke Fukuhara
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Shinji Ouma
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan.
| |
Collapse
|
134
|
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
|
135
|
Abstract
This article summarizes (1) the recent achievements to further improve symptomatic therapy of motor Parkinson’s disease (PD) symptoms, (2) the still-few attempts to systematically search for symptomatic therapy of non-motor symptoms in PD, and (3) the advances in the development and clinical testing of compounds which promise to offer disease modification in already-manifest PD. However, prevention (that is, slowing or stopping PD in a prodromal stage) is still a dream and one reason for this is that we have no consensus on primary endpoints for clinical trials which reflect the progression in prodromal stages of PD, such as in rapid eye movement sleep behavior disorder (RBD) —a methodological challenge to be met in the future.
Collapse
Affiliation(s)
- Wolfgang H Oertel
- Department of Neurology, University Clinic, Philipps Universität Marburg, Marburg, Germany; Institute for Neurogenomics, Helmholtz Center for Health and Environment, Munich, Germany
| |
Collapse
|
136
|
Baena CP, Suemoto CK, Barreto SM, Lotufo PA, Benseñor I. Serum uric acid is associated with better executive function in men but not in women: Baseline assessment of the ELSA-Brasil study. Exp Gerontol 2017; 92:82-86. [PMID: 28300627 DOI: 10.1016/j.exger.2017.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Serum uric acid (SUA) may protect against free radical stress damage and was previously linked to cognitive impairment in older adults, but evidence in middle-aged adults is scarce. PURPOSE We sought to analyze whether SUA is associated with cognitive performance in apparently healthy middle-aged participants in the ELSA-Brasil cohort study. METHODS We excluded participants older than age 65, those taking allopurinol, benzbromarone, or medications that could impair cognitive performance, those with previous stroke, and those with incomplete data on cognitive tests or SUA. The Consortium to Establish a Registry for Alzheimer's Disease Word List Memory Test (CERAD-WLMT), the Semantic Fluency Test (SFT), and the Trail Making Test version B (TMT) were used as dependent variables. Sex-specific linear regression models were used to assess the association between SUA and cognitive tests, adjusted by age, education, hypertension, dyslipidemia, diabetes, smoking, alcohol consumption, body mass index, coronary heart disease, renal function, depression, aspirin use, thyroid function, and menopausal status (in women). We used the Bonferroni procedure to control for the false discovery rate associated with multiple comparisons. RESULTS We analyzed cross-sectional data from 6751 women and 5464 men. Mean age and standard deviation (SD) of the sample was 49.6 (SD 7.4) years for men and 49.9 (SD 7.3) years for women. The majority of men (52%) and women (51%) were white. Mean SUA value was 4.75 (SD 1.16) mg/dL in women and 6.44 (SD 1.39) mg/dL in men. Multivariate linear models showed no association in women and a significant inverse association between SUA levels and TMT (β=-3.106, 95% CI=-4.594; -1.618, p=0.0004) in men. CONCLUSION In a middle-aged subset population, SUA is associated with better performance on an executive function test in men, but not in women in the ELSA-Brasil cohort study.
Collapse
Affiliation(s)
- Cristina Pellegrino Baena
- Center for Clinical and Epidemiologic Research of the University of São Paulo, São Paulo, Brazil; School of Medicine, Pontifícia Universidade Católica Paraná, Curitiba, Brazil.
| | - Claudia Kimie Suemoto
- Center for Clinical and Epidemiologic Research of the University of São Paulo, São Paulo, Brazil; Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Sandhi Maria Barreto
- Department of Preventive Medicine, Faculty of Medicine, Unversidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paulo Andrade Lotufo
- Center for Clinical and Epidemiologic Research of the University of São Paulo, São Paulo, Brazil
| | - Isabela Benseñor
- Center for Clinical and Epidemiologic Research of the University of São Paulo, São Paulo, Brazil
| |
Collapse
|
137
|
Serum uric acid levels and freezing of gait in Parkinson's disease. Neurol Sci 2017; 38:955-960. [PMID: 28251464 DOI: 10.1007/s10072-017-2871-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/23/2017] [Indexed: 02/05/2023]
Abstract
Uric acid (UA) is a natural antioxidant and iron scavenger in the human body, which has been hypothesized to exert an anti-oxidative effect in Parkinson's disease (PD). This study aimed to investigate the relationship between serum UA levels and freezing of gait (FOG) in PD. A total of 321 Chinese PD patients with fasting serum UA evaluated were included in the cross-sectional study. Demographics, clinical features, and therapeutic regimen were collected. The Unified PD Rating Scale (UPDRS) III and Hoehn and Yahr (H and Y) stage were used to evaluate the severity of disease, and the Frontal Assessment Battery (FAB) and Montreal Cognitive Assessment (MoCA) scales were used to assess the cognitive function. Patients with FOG showed lower proportion of male, longer disease duration, lower body mass index, lower concentrations of serum UA, higher total levodopa equivalent daily dosage, higher UPDRS III score, greater median H and Y stage, lower scores of FAB and MoCA, and higher frequencies of motor fluctuation, dyskinesia, falls, and festination compared to patients without FOG (P < 0.05). The binary logistic regression model indicated that high UPDRS III score (OR = 1.049, P < 0.001), fluctuation (OR = 2.677, P = 0.035), dyskinesia (OR = 6.294, P = 0.003), festination (OR = 3.948, P < 0.001), falls (OR = 7.528, P < 0.001), and low serum UA levels (OR = 0.990, P < 0.001) were associated with FOG. Our study suggests that low serum UA concentration is associated with the occurrence of FOG in PD.
Collapse
|
138
|
LeWitt PA, Li J, Lu M, Guo L, Auinger P. Metabolomic biomarkers as strong correlates of Parkinson disease progression. Neurology 2017; 88:862-869. [PMID: 28179471 DOI: 10.1212/wnl.0000000000003663] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/29/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether a Parkinson disease (PD)-specific biochemical signature might be found in the total body metabolic milieu or in the CSF compartment, especially since this disorder has systemic manifestations beyond the progressive loss of dopaminergic nigrostriatal neurons. METHODS Our goal was to discover biomarkers of PD progression. Using ultra-high-performance liquid chromatography linked to gas chromatography and tandem mass spectrometry, we measured concentrations of small-molecule (≤1.5 kDa) constituents of plasma and CSF from 49 unmedicated, mildly affected patients with PD (mean age 61.4 years; mean duration of PD 11.4 months). Specimens were collected twice (baseline and final) at intervals up to 24 months. During this time, mean Unified Parkinson's Disease Rating Scale (UPDRS) parts 2 + 3 scores increased 47% (from 28.8 to 42.2). Measured compounds underwent unbiased univariate and multivariate analyses, including fitting data into multiple linear regression with variable selection using least absolute shrinkage and selection operator (LASSO). RESULTS Of 575 identified plasma and 383 CSF biochemicals, LASSO led to selection of 15 baseline plasma constituents with high positive correlation (0.87, p = 2.2e-16) to baseline-to-final change in UPDRS parts 2 + 3 scores. Three of the compounds had xanthine structures, and 4 were either medium- or long-chain fatty acids. For the 15 LASSO-selected biomarkers, pathway enrichment software found no overrepresentation among metabolic pathways. CSF concentrations of the dopamine metabolite homovanillate showed little change between baseline and final collections and minimal correlation with worsening UPDRS parts 2 + 3 scores (0.29, p = 0.041). CONCLUSIONS Metabolomic profiling of plasma yielded strong prediction of PD progression and offered biomarkers that may provide new insights into PD pathogenesis.
Collapse
Affiliation(s)
- Peter A LeWitt
- From the Departments of Neurology (P.A.L.) and Public Health Science (J.L., M.L.), Henry Ford Health System; Wayne State University School of Medicine (P.A.L.), Detroit MI; Metabolon, Inc (L.G.), Durham, NC; and Center for Human Experimental Therapeutics (P.A.), University of Rochester, NY.
| | - Jia Li
- From the Departments of Neurology (P.A.L.) and Public Health Science (J.L., M.L.), Henry Ford Health System; Wayne State University School of Medicine (P.A.L.), Detroit MI; Metabolon, Inc (L.G.), Durham, NC; and Center for Human Experimental Therapeutics (P.A.), University of Rochester, NY
| | - Mei Lu
- From the Departments of Neurology (P.A.L.) and Public Health Science (J.L., M.L.), Henry Ford Health System; Wayne State University School of Medicine (P.A.L.), Detroit MI; Metabolon, Inc (L.G.), Durham, NC; and Center for Human Experimental Therapeutics (P.A.), University of Rochester, NY
| | - Lining Guo
- From the Departments of Neurology (P.A.L.) and Public Health Science (J.L., M.L.), Henry Ford Health System; Wayne State University School of Medicine (P.A.L.), Detroit MI; Metabolon, Inc (L.G.), Durham, NC; and Center for Human Experimental Therapeutics (P.A.), University of Rochester, NY
| | - Peggy Auinger
- From the Departments of Neurology (P.A.L.) and Public Health Science (J.L., M.L.), Henry Ford Health System; Wayne State University School of Medicine (P.A.L.), Detroit MI; Metabolon, Inc (L.G.), Durham, NC; and Center for Human Experimental Therapeutics (P.A.), University of Rochester, NY
| | | |
Collapse
|
139
|
Mandal AK, Mercado A, Foster A, Zandi-Nejad K, Mount DB. Uricosuric targets of tranilast. Pharmacol Res Perspect 2017; 5:e00291. [PMID: 28357121 PMCID: PMC5368959 DOI: 10.1002/prp2.291] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/24/2016] [Accepted: 11/10/2016] [Indexed: 11/08/2022] Open
Abstract
Uric acid, generated from the metabolism of purines, has both proven and emerging roles in human disease. Serum uric acid in humans is determined by production and by the net balance of reabsorption and secretion in kidney and intestine. In the human kidney, epithelial reabsorption dominates over secretion, such that in normal subjects there is at least 90% net reabsorption of filtered urate resulting in a fractional excretion of <10%. Tranilast, an anti-inflammatory drug with pleiotropic effects, has a marked hypouricemic, uricosuric effect in humans. We report here that tranilast is a potent inhibitor of [14C]-urate transport mediated by the major reabsorptive urate transporters (URAT1, GLUT9, OAT4, and OAT10) in Xenopus oocytes; this provides an unequivocal molecular mechanism for the drug's uricosuric effect. Tranilast was found to inhibit urate transport mediated by URAT1 and GLUT9 in a fully reversible and noncompetitive (mixed) manner. In addition, tranilast inhibits the secretory urate transporters NPT1, OAT1, and OAT3 without affecting the secretory efflux pump ABCG2. Notably, while benzbromarone and probenecid inhibited urate as well as nicotinate transport, tranilast inhibited the urate transport function of URAT1, GLUT9, OAT4, OAT10, and NPT1, without significantly affecting nicotinate transport mediated by SMCT1 (IC 50 ~1.1 mmol/L), SMCT2 (IC 50 ~1.0 mmol/L), and URAT1 (IC 50 ~178 μmol/L). In summary, tranilast causes uricosuria by inhibiting all the major reabsorptive urate transporters, selectively affecting urate over nicotinate transport. These data have implications for the treatment of hyperuricemia and gout, the pharmacology of tranilast, and the structure-function analysis of urate transport.
Collapse
Affiliation(s)
- Asim K Mandal
- Renal Divisions VA Boston Healthcare System and Brigham and Women's Hospital Boston Massachusetts
| | - Adriana Mercado
- Renal Divisions Departamento de Nefrología Instituto Nacional de Cardiología Ignacio Chávez Mexico City Mexico
| | - Andria Foster
- Renal Divisions VA Boston Healthcare System and Brigham and Women's Hospital Boston Massachusetts
| | - Kambiz Zandi-Nejad
- Renal Division Beth Israel Deaconess Medical Center Boston Massachusetts
| | - David B Mount
- Renal Divisions VA Boston Healthcare System and Brigham and Women's Hospital Boston Massachusetts
| |
Collapse
|
140
|
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]
|
141
|
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
|
142
|
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
|
143
|
Andersen AD, Binzer M, Stenager E, Gramsbergen JB. Cerebrospinal fluid biomarkers for Parkinson's disease - a systematic review. Acta Neurol Scand 2017; 135:34-56. [PMID: 26991855 DOI: 10.1111/ane.12590] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2016] [Indexed: 12/14/2022]
Abstract
Diagnosis of Parkinson's disease (PD) relies on clinical history and physical examination, but misdiagnosis is common in early stages. Identification of biomarkers for PD may allow early and more precise diagnosis and monitoring of dopamine replacement strategies and disease modifying treatments. Developments in analytical chemistry allow the detection of large numbers of molecules in plasma or cerebrospinal fluid, associated with the pathophysiology or pathogenesis of PD. This systematic review includes cerebrospinal fluid biomarker studies focusing on different disease pathways: oxidative stress, neuroinflammation, lysosomal dysfunction and proteins involved in PD and other neurodegenerative disorders, focusing on four clinical domains: their ability to (1) distinguish PD from healthy subjects and other neurodegenerative disorders as well as their relation to (2) disease duration after initial diagnosis, (3) severity of disease (motor symptoms) and (4) cognitive dysfunction. Oligomeric alpha-synuclein might be helpful in the separation of PD from controls. Through metabolomics, changes in purine and tryptophan metabolism have been discovered in patients with PD. Neurofilament light chain (NfL) has a significant role in distinguishing PD from other neurodegenerative diseases. Several oxidative stress markers are related to disease severity, with the antioxidant urate also having a prognostic value in terms of disease severity. Increased levels of amyloid and tau-proteins correlate with cognitive decline and may have prognostic value for cognitive deficits in PD. In the future, larger longitudinal studies, corroborating previous research on viable biomarker candidates or using metabolomics identifying a vast amount of potential biomarkers, could be a good approach.
Collapse
Affiliation(s)
- A. D. Andersen
- Department of Neurology; Hospital of Southern Jutland; Sønderborg Denmark
- Institute of Regional Health Research; University of Southern Denmark; Odense Denmark
- Focused Research Group in Neurology; Hospital of Southern Jutland; Sønderborg Denmark
| | - M. Binzer
- Institute of Regional Health Research; University of Southern Denmark; Odense Denmark
- Focused Research Group in Neurology; Hospital of Southern Jutland; Sønderborg Denmark
| | - E. Stenager
- Institute of Regional Health Research; University of Southern Denmark; Odense Denmark
- Focused Research Group in Neurology; Hospital of Southern Jutland; Sønderborg Denmark
- The Multiple Sclerosis Clinic of Southern Jutland; (Vejle, Sonderborg, Esbjerg) Denmark
| | - J. B. Gramsbergen
- Institute of Molecular Medicine, Neurobiological Research; University of Southern Denmark; Odense Denmark
| |
Collapse
|
144
|
Tomioka NH, Tamura Y, Takada T, Shibata S, Suzuki H, Uchida S, Hosoyamada M. Immunohistochemical and in situ hybridization study of urate transporters GLUT9/URATv1, ABCG2, and URAT1 in the murine brain. Fluids Barriers CNS 2016; 13:22. [PMID: 27955673 PMCID: PMC5154092 DOI: 10.1186/s12987-016-0046-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/26/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Uric acid (UA) is known to exert neuroprotective effects in the brain. However, the mechanism of UA regulation in the brain is not well characterized. In our previous study, we described that the mouse urate transporter URAT1 is localized to the cilia and apical surface of ventricular ependymal cells. To further strengthen the hypothesis that UA is transported transcellularly at the ependymal cells, we aimed to assess the distribution of other UA transporters in the murine brain. METHODS Immunostaining and highly-sensitive in situ hybridization was used to assess the distribution of UA transporters: GLUT9/URATv1, ABCG2, and URAT1. RESULTS Immunostaining for GLUT9 was observed in ependymal cells, neurons, and brain capillaries. Immunostaining for ABCG2 was observed in the choroid plexus epithelium and brain capillaries, but not in ependymal cells. These results were validated by in situ hybridization. CONCLUSIONS We propose that given their specific expression patterns in ependymal, choroid plexus epithelial, and brain capillary endothelial cells in this study, UA may be transported by these UA transporters in the murine brain. This may provide a novel strategy for targeted neuroprotection.
Collapse
Affiliation(s)
- Naoko H. Tomioka
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605 Japan
| | - Yoshifuru Tamura
- Department of Internal Medicine, Teikyo University School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605 Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Shigeru Shibata
- Department of Internal Medicine, Teikyo University School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605 Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Shunya Uchida
- Department of Internal Medicine, Teikyo University School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605 Japan
| | - Makoto Hosoyamada
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605 Japan
| |
Collapse
|
145
|
Hughes KC, Gao X, Kim IY, Rimm E, Wang M, Weisskopf MG, Schwarzschild MA, Ascherio A. Intake of antioxidant vitamins and risk of Parkinson's disease. Mov Disord 2016; 31:1909-1914. [PMID: 27787934 PMCID: PMC5154924 DOI: 10.1002/mds.26819] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Oxidative stress is proposed to be one of the potential mechanisms leading to neurodegeneration in Parkinson's disease. However, previous epidemiologic studies investigating associations between antioxidant vitamins, such as vitamins E and C and carotenoids, and PD risk have produced inconsistent results. OBJECTIVE The objective of this work was to prospectively examine associations between intakes of antioxidant vitamins, including vitamins E and C and carotenoids, and PD risk. METHODS Cases were identified in two large cohorts: the Nurses' Health Study and the Health Professionals Follow-up Study. Cohort members completed semiquantitative food frequency questionnaires every 4 years. RESULTS A total of 1036 PD cases were identified. Dietary intakes of vitamin E and carotenoids were not associated with PD risk; the multivariable-adjusted relative risk comparing extreme intake quintiles were 0.93 (95% confidence interval: 0.75-1.14) and 0.97 (95% confidence interval: 0.69-1.37), respectively. Dietary vitamin C intake was significantly associated with reduced PD risk (relative risk: 0.81; 95% confidence interval: 0.65-1.01; ptrend , 0.01); however, this result was not significant in a 4-year lag analysis. For vitamins E and C, intake from foods and supplements combined were also unrelated to PD risk. CONCLUSIONS Our results do not support the hypothesis that intake of antioxidant vitamins reduces the risk of PD. © 2016 International Parkinson and Movement Disorder Society.
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
| | - Iris Y. Kim
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Eric Rimm
- 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 Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 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 Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
146
|
Arnaldi D, Antelmi E, St Louis EK, Postuma RB, Arnulf I. Idiopathic REM sleep behavior disorder and neurodegenerative risk: To tell or not to tell to the patient? How to minimize the risk? Sleep Med Rev 2016; 36:82-95. [PMID: 28082168 DOI: 10.1016/j.smrv.2016.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 10/23/2016] [Accepted: 11/02/2016] [Indexed: 11/17/2022]
Abstract
Most people with idiopathic REM sleep behavior disorder (iRBD) have an underlying synucleinopathy, mainly Parkinson's disease (PD) or dementia with Lewy bodies, with median conversion time of 4-9 y from iRBD diagnosis and of 11-16 y from symptom onset. Subtle signs and imaging tests indicate concomitant neurodegeneration in widespread brain areas. Risk factor studies suggest that iRBD patients may have prior head injury, occupational farming, pesticide exposure, low education level and possibly more frequent family history of dream-enactment behavior (but not of PD), plus unexpected risk factors (smoking, ischemic heart disease and inhaled corticosteroid use). Unlike PD, caffeine and smoking appear not to have a protective role. Prior depression and antidepressant use may be early neurodegenerative signs rather than exclusively causative factors. Age, hyposmia, impaired color vision, abnormal dopaminergic imaging, mild cognitive impairment and possibly sleepiness, may identify patients at greater risk of more rapid conversion. The consensus is to generally disclose the neurodegenerative risk to patients (with the caveat that phenoconversion and its temporal course remain uncertain in individuals without "soft neurodegenerative signs" and those under 50 y of age), to suggest a healthy lifestyle and to take part in prospective cohort studies in anticipation of eventual neuroprotective trials.
Collapse
Affiliation(s)
- Dario Arnaldi
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Italy
| | - Elena Antelmi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Italy
| | - Erik K St Louis
- Mayo Center for Sleep Medicine, Departments of Neurology and Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Ronald B Postuma
- Department of Neurology, Montreal General Hospital, Montreal, Quebec, Canada
| | - Isabelle Arnulf
- Sleep Disorder Unit, Pitie-Salpetriere Hospital, APHP, Pierre and Marie Curie University, Paris, France.
| |
Collapse
|
147
|
Abstract
BACKGROUND Over the past several years, the concept of prodromal Parkinson disease (PD) has been increasingly recognized. This term refers to individuals who do not fulfill motor diagnostic criteria for PD, but who have clinical, genetic, or biomarker characteristics suggesting risk of developing PD in the future. Clinical diagnosis of prodromal PD has low specificity, prompting the need for objective biomarkers with higher specificity. In this qualitative review, we discuss objectively defined putative biomarkers for PD and prodromal PD. METHODS We searched Pubmed and Embase for articles pertaining to objective biomarkers for PD and their application in prodromal cohorts. Articles were selected based on relevance and methodology. KEY FINDINGS Objective biomarkers of demonstrated utility in prodromal PD include ligand-based imaging and transcranial sonography. Development of serum, cerebrospinal fluid, and tissue-based biomarkers is underway, but their application in prodromal PD has yet to meaningfully occur. Combining objective biomarkers with clinical or genetic prodromal features increases the sensitivity and specificity for identifying prodromal PD. CONCLUSIONS Several objective biomarkers for prodromal PD show promise but require further study, including their application to and validation in prodromal cohorts followed longitudinally. Accurate identification of prodromal PD will likely require a multimodal approach. (JINS, 2016, 22, 956-967).
Collapse
|
148
|
Ascherio A, Schwarzschild MA. The epidemiology of Parkinson's disease: risk factors and prevention. Lancet Neurol 2016; 15:1257-1272. [PMID: 27751556 DOI: 10.1016/s1474-4422(16)30230-7] [Citation(s) in RCA: 1220] [Impact Index Per Article: 135.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022]
Abstract
Since 2006, several longitudinal studies have assessed environmental or behavioural factors that seem to modify the risk of developing Parkinson's disease. Increased risk of Parkinson's disease has been associated with exposure to pesticides, consumption of dairy products, history of melanoma, and traumatic brain injury, whereas a reduced risk has been reported in association with smoking, caffeine consumption, higher serum urate concentrations, physical activity, and use of ibuprofen and other common medications. Randomised trials are investigating the possibility that some of the negative risk factors might be neuroprotective and thus beneficial in individuals with early Parkinson's disease, particularly with respect to smoking (nicotine), caffeine, and urate. In the future, it might be possible to identify Parkinson's disease in its prodromal phase and to promote neuroprotective interventions before the onset of motor symptoms. At this time, however, the only intervention that seems justifiable for the primary prevention of Parkinson's disease is the promotion of physical activity, which is likely to be beneficial for the prevention of several chronic diseases.
Collapse
Affiliation(s)
- Alberto Ascherio
- Departments of Epidemiology and Nutrition, Harvard T H Chan School of Public Health, Boston, MA, USA; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
149
|
O'Reilly ÉIJ, Liu D, Johns DR, Cudkowicz ME, Paganoni S, Schwarzschild MA, Leitner M, Ascherio A. Serum urate at trial entry and ALS progression in EMPOWER. Amyotroph Lateral Scler Frontotemporal Degener 2016; 18:120-125. [PMID: 27677562 DOI: 10.1080/21678421.2016.1214733] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our objective was to determine whether serum urate predicts ALS progression. A study population comprised adult participants of EMPOWER (n = 942), a phase III clinical trial to evaluate the efficacy of dexpramipexole to treat ALS. Urate was measured in blood samples collected during enrollment as part of the routine block chemistry. We measured outcomes by combined assessment of function and survival rank (CAFs), and time to death, by 12 months. Results showed that in females there was not a significant relation between urate and outcomes. In males, outcomes improved with increasing urate (comparing highest to lowest urate quartile: CAFS was 53 points better with p for trend = 0.04; and hazard ratio for death was 0.60 with p for trend = 0.07), but with adjustment for body mass index (BMI) at baseline, a predictor of both urate levels and prognosis, associations were attenuated and no longer statistically significant. Overall, participants with urate levels equal to or above the median (5.1 mg/dl) appeared to have a survival advantage compared to those below (hazard ratio adjusted for BMI: 0.67; 95% confidence interval 0.47-0.95). In conclusion, these findings suggest that while the association between urate at baseline and ALS progression is partially explained by BMI, there may be an independent beneficial effect of urate.
Collapse
Affiliation(s)
- ÉIlis J O'Reilly
- a Department of Nutrition, Harvard TH Chan School of Public Health, Boston. Channing Division of Network Medicine , Harvard Medical School and Brigham and Women's Hospital , Boston , Massachusetts
| | - Dawei Liu
- b ALS Neuroscience Discovery and Development , Biogen , Cambridge , Massachusetts
| | - Donald R Johns
- b ALS Neuroscience Discovery and Development , Biogen , Cambridge , Massachusetts
| | - Merit E Cudkowicz
- c Department of Neurology , Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease , Boston , Massachusetts
| | - Sabrina Paganoni
- c Department of Neurology , Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease , Boston , Massachusetts.,d Department of Physical Medicine and Rehabilitation , Harvard Medical School, Spaulding Rehabilitation Hospital , Boston, Massachusetts; VA Healthcare System , Boston , Massachusetts , and
| | - Michael A Schwarzschild
- c Department of Neurology , Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease , Boston , Massachusetts
| | - Melanie Leitner
- b ALS Neuroscience Discovery and Development , Biogen , Cambridge , Massachusetts
| | - Alberto Ascherio
- e Department of Epidemiology and Nutrition , Harvard TH Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Harvard Medical School and Brigham and Women's Hospital , Boston , Massachusetts , USA
| |
Collapse
|
150
|
Abstract
Despite an increased understanding of the pathogenesis of Parkinson's disease (PD), and a number of drugs designed to ameliorate symptoms, finding an effective neuroprotective therapy remains elusive. For decades now, several promising agents targeting different pathways have been explored as potential treatments that could help slow disease progression, but these have met with limited success. There are hurdles to overcome, particularly given that there is no exact animal model of PD and also no reliable biomarkers for PD. Without biomarkers, it is not possible to demonstrate, in the context of a clinical trial, that an intervention prevents neuronal degeneration. However, given the compelling scientific rationale of several compounds, an unrelenting pursuit continues. There have been hundreds of human studies looking at neuroprotection in PD. This article will briefly summarize several of the neuroprotective treatments that have been evaluated in large clinical trials, and will also outline some of the newer therapies that are currently being explored.
Collapse
Affiliation(s)
- Ariane Park
- Department of Neurology, The Ohio State University, Columbus, OH, USA.
| | - Mark Stacy
- Duke University Medical Center, Durham, NC, USA
| |
Collapse
|