51
|
Sun HL, Wu YW, Bian HG, Yang H, Wang H, Meng XM, Jin J. Function of Uric Acid Transporters and Their Inhibitors in Hyperuricaemia. Front Pharmacol 2021; 12:667753. [PMID: 34335246 PMCID: PMC8317579 DOI: 10.3389/fphar.2021.667753] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Disorders of uric acid metabolism may be associated with pathological processes in many diseases, including diabetes mellitus, cardiovascular disease, and kidney disease. These diseases can further promote uric acid accumulation in the body, leading to a vicious cycle. Preliminary studies have proven many mechanisms such as oxidative stress, lipid metabolism disorders, and rennin angiotensin axis involving in the progression of hyperuricaemia-related diseases. However, there is still lack of effective clinical treatment for hyperuricaemia. According to previous research results, NPT1, NPT4, OAT1, OAT2, OAT3, OAT4, URAT1, GLUT9, ABCG2, PDZK1, these urate transports are closely related to serum uric acid level. Targeting at urate transporters and urate-lowering drugs can enhance our understanding of hyperuricaemia and hyperuricaemia-related diseases. This review may put forward essential references or cross references to be contributed to further elucidate traditional and novel urate-lowering drugs benefits as well as provides theoretical support for the scientific research on hyperuricemia and related diseases.
Collapse
Affiliation(s)
- Hao-Lu Sun
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Yi-Wan Wu
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - He-Ge Bian
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Hui Yang
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Heng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan Jin
- Department of Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
52
|
Yoo HS, Chung SJ, Lee YH, Ye BS, Sohn YH, Kwon H, Lee PH. Urate is closely linked to white matter integrity in multiple system atrophy. Ann Clin Transl Neurol 2021; 7:1029-1039. [PMID: 32588990 PMCID: PMC7318089 DOI: 10.1002/acn3.51073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Objective We aimed to investigate the association of the serum urate level with cortical thickness and white matter integrity in multiple system atrophy (MSA). Methods We recruited 75 MSA patients and 42 controls who underwent brain MRI and measured serum urate level at baseline. Using cortical thickness and tract‐based spatial statistics analyses, we investigated the correlation between serum urate levels and cortical thickness or diffusion tensor imaging (DTI) measures in controls and MSA patients. Interaction effects were analyzed to find different patterns of correlation according to sex and clinical subtype. We evaluated the relationship between serum urate levels, DTI measures, and total UMSARS score, using path analysis. Results Serum urate levels showed a positive correlation with FA values in the corpus callosum and a negative correlation with MD values in widespread regions including cerebellar, brainstem, and cerebral white matter in patients with MSA. Both sexes showed a negative correlation between serum urate levels and MD values without significant interaction effect. In subgroup analysis according to subtype, patients with cerebellar subtype showed a negative correlation. Serum urate levels did not correlated with cortical thickness. Path analysis showed that MD values in middle and inferior cerebellar peduncle mediated the association between serum urate level and total UMSAR score. Interpretation The present study demonstrated that serum urate levels played a pivotal role in white matter disintegrity and clinical disability in MSA. It would provide an evidence of the role of urate as a potential neuroprotective factor against white matter neurodegeneration in MSA.
Collapse
Affiliation(s)
- Han Soo Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yang Hyun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hunki Kwon
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
53
|
Guo SM, Liu YT, He SR, Wu MS, Tseng WT, Wu RC, Wu IC. Differential relationship of uric acid to mortality and clinical biomarkers of aging according to grip strength in older adults: a cohort study. Aging (Albany NY) 2021; 13:10555-10583. [PMID: 33820873 PMCID: PMC8064196 DOI: 10.18632/aging.202820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/04/2021] [Indexed: 12/25/2022]
Abstract
Uric acid is both a pro-oxidant and antioxidant. We investigated serum uric acid's association with mortality and aging biomarkers in older adults with varying levels of grip strength. A total of 5329 community-dwelling adults aged ≥55 years underwent assessments of serum uric acid levels, grip strength, and biomarkers of diverse physiological systems. The primary outcome was all-cause mortality. We observed a significant (P < .001) interaction between uric acid levels and grip strength on all-cause mortality risk. Among participants with low grip strength, a nonlinear association (P for nonlinearity = .006) was observed between serum uric acid levels and mortality risk after multivariate adjustment. Compared with participants with neither extreme uric acid levels nor low grip strength, those with a combination of high serum uric acid and low grip strength exhibited greater risks of mortality (adjusted hazard ratio [aHR], 1.52; 95% confidence interval [CI], 1.15–2.02) and deviations in biomarkers of specific systems, so did those with a combination of low serum uric acid and low grip strength (aHR, 1.52; 95% CI, 1.13–2.05). In conclusion, there was a J-shaped association between serum uric acid and the risk of all-cause mortality in older adults. This was primarily true for those with low grip strength.
Collapse
Affiliation(s)
- Sin-Mei Guo
- The Department of Family Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Yen-Tze Liu
- The Department of Family Medicine, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Holistic Wellness, MingDao University, Changhua, Taiwan
| | - Sin-Ru He
- The Department of Family Medicine, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Shiang Wu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Wei-Ting Tseng
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Ray-Chin Wu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - I-Chien Wu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
54
|
Abstract
The links between diet and Parkinson's disease (PD) are unclear and incomprehensible. However, numerous studies have demonstrated the correlation between diet, nutrients and health condition in PD patients. They indicate the possibility of management of the disease, which might be possible through nutrition. Pharmaceutical treatment as well as a complementary holistic approach to the patients should be considered. It is of critical importance to understand how the diet and nutrients might influence PD. A better understanding of the relationship between diet and PD could help to better manage the disease explain promising therapeutic approaches, minimize motor and nonmotor symptoms and disease progression based on a personalized diet. In this review, the recent literature on the observed nutrition disorders and the possible role of diet and nutrients in the prevention and potential regression of PD, as well as dietary interventions and supplementation used to manage the disease is revised.
Collapse
Affiliation(s)
- Paulina Gątarek
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
| | - Joanna Kałużna-Czaplińska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
55
|
Antioxidant Therapeutics in Parkinson's Disease: Current Challenges and Opportunities. Antioxidants (Basel) 2021; 10:antiox10030453. [PMID: 33803945 PMCID: PMC7998929 DOI: 10.3390/antiox10030453] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is considered one of the pathological mechanisms that cause Parkinson’s disease (PD), which has led to the investigation of several antioxidants molecules as a potential therapeutic treatment against the disease. Although preclinical studies have demonstrated the efficacy of these compounds to maintain neuronal survival and activity in PD models, these results have not been reflected in clinical trials, antioxidants have not been able to act as disease modifiers in terms of clinical symptoms. Translational medicine currently faces the challenge of redesigning clinical trials to standardize criteria when testing molecules to reduce responses’ variability. Herein, we discuss current challenges and opportunities regarding several non-enzymatic antioxidants’ therapeutic molecules for PD patients’ potential treatment.
Collapse
|
56
|
Medically Important Alterations in Transport Function and Trafficking of ABCG2. Int J Mol Sci 2021; 22:ijms22062786. [PMID: 33801813 PMCID: PMC8001156 DOI: 10.3390/ijms22062786] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Several polymorphisms and mutations in the human ABCG2 multidrug transporter result in reduced plasma membrane expression and/or diminished transport function. Since ABCG2 plays a pivotal role in uric acid clearance, its malfunction may lead to hyperuricemia and gout. On the other hand, ABCG2 residing in various barrier tissues is involved in the innate defense mechanisms of the body; thus, genetic alterations in ABCG2 may modify the absorption, distribution, excretion of potentially toxic endo- and exogenous substances. In turn, this can lead either to altered therapy responses or to drug-related toxic reactions. This paper reviews the various types of mutations and polymorphisms in ABCG2, as well as the ways how altered cellular processing, trafficking, and transport activity of the protein can contribute to phenotypic manifestations. In addition, the various methods used for the identification of the impairments in ABCG2 variants and the different approaches to correct these defects are overviewed.
Collapse
|
57
|
Bougea A, Koros C, Papagiannakis N, Simitsi AM, Prentakis A, Papadimitriou D, Pachi I, Antonelou R, Angelopoulou E, Beratis I, Bozi M, Papageorgiou SG, Trapali XG, Stamelou M, Stefanis L. Serum Uric Acid in LRRK2 Related Parkinson's Disease: Longitudinal Data from the PPMI Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:633-640. [PMID: 33682725 DOI: 10.3233/jpd-202337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Previous studies have highlighted serum uric acid as a putative idiopathic Parkinson's disease (iPD) biomarker. Only one study, so far, showed higher levels of serum uric acid in leucine-rich repeat kinase 2 (LRRK + 2) carriers compared to those who developed PD, however a longitudinal comparison between LRRK2 + PD and healthy controls (HC) has not been performed. OBJECTIVE The aim of this study was to determine whether there are longitudinal differences in serum uric acid between iPD, LRRK2 + PD and HC and their association with motor and non-motor features. METHODS Longitudinal data of uric acid of 282 de novo iPD, 144 LRRK2 + PD patients, and 195 age-matched HC were obtained from the Parkinson's Progression Markers Initiative (PPMI) database. We also used longitudinal Montreal Cognitive Assessment (MoCA), Movement Disorder Society-Unified Parkinson's Disease Rating Scale part III (MDS-UPDRS-III), Geriatric Depression Scale (GDS) scores, and DaTSCAN striatal binding ratios (SBRs). RESULTS Longitudinal uric acid measurements were significantly lower in LRRK2 + PD patients compared to HC up to 5 years follow-up. There was no significant impact or correlation of adjusted or unadjusted uric acid levels with MoCA, MDS-UPDRS III, or GDS scores, the presence of RBD or DAT-SCAN SBRs. CONCLUSION LRRK2 + PD group had significantly lower uric acid concentrations compared to HC after adjusting for age, sex and baseline BMI up to 5 years follow-up. There were no significant associations between uric acid levels and indices of disease severity. These findings identify serum uric acid as a marker linked to LRRK2 + PD.
Collapse
Affiliation(s)
- Anastasia Bougea
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Koros
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Papagiannakis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina-Maria Simitsi
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Prentakis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Nuclear Medicine Unit, Attikon Hospital, Athens, Greece
| | | | - Ioanna Pachi
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Roubina Antonelou
- 2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthalia Angelopoulou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ion Beratis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Bozi
- 2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sokratis G Papageorgiou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Stamelou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Neurology Clinic, Philipps University, Marburg, Germany.,Parkinson's disease and Movement Disorders Department, HYGEIA Hospital, Athens, Greece
| | - Leonidas Stefanis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
58
|
Mencke P, Boussaad I, Romano CD, Kitami T, Linster CL, Krüger R. The Role of DJ-1 in Cellular Metabolism and Pathophysiological Implications for Parkinson's Disease. Cells 2021; 10:347. [PMID: 33562311 PMCID: PMC7915027 DOI: 10.3390/cells10020347] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 11/16/2022] Open
Abstract
DJ-1 is a multifunctional protein associated with pathomechanisms implicated in different chronic diseases including neurodegeneration, cancer and diabetes. Several of the physiological functions of DJ-1 are not yet fully understood; however, in the last years, there has been increasing evidence for a potential role of DJ-1 in the regulation of cellular metabolism. Here, we summarize the current knowledge on specific functions of DJ-1 relevant to cellular metabolism and their role in modulating metabolic pathways. Further, we illustrate pathophysiological implications of the metabolic effects of DJ-1 in the context of neurodegeneration in Parkinson´s disease.
Collapse
Affiliation(s)
- Pauline Mencke
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg;
| | - Ibrahim Boussaad
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg;
| | - Chiara D. Romano
- Biospecimen Research Group, Integrated Biobank of Luxembourg, Luxembourg Institute of Health (LIH), 3531 Dudelange, Luxembourg;
- Enzymology & Metabolism, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg;
| | - Toshimori Kitami
- RIKEN Outpost Laboratory, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg;
| | - Carole L. Linster
- Enzymology & Metabolism, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg;
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg;
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), 1210 Luxembourg (Belair), Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), 1445 Strassen, Luxembourg
| |
Collapse
|
59
|
Zhang Y, Li J, Zhang X, Song D, Tian T. Advances of Mechanisms-Related Metabolomics in Parkinson's Disease. Front Neurosci 2021; 15:614251. [PMID: 33613180 PMCID: PMC7887307 DOI: 10.3389/fnins.2021.614251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial disorder characterized by progressively debilitating dopaminergic neurodegeneration in the substantia nigra and the striatum, along with various metabolic dysfunctions and molecular abnormalities. Metabolomics is an emerging study and has been demonstrated to play important roles in describing complex human diseases by integrating endogenous and exogenous sources of alterations. Recently, an increasing amount of research has shown that metabolomics profiling holds great promise in providing unique insights into molecular pathogenesis and could be helpful in identifying candidate biomarkers for clinical detection and therapies of PD. In this review, we briefly summarize recent findings and analyze the application of molecular metabolomics in familial and sporadic PD from genetic mutations, mitochondrial dysfunction, and dysbacteriosis. We also review metabolic biomarkers to assess the functional stage and improve therapeutic strategies to postpone or hinder the disease progression.
Collapse
Affiliation(s)
| | | | | | | | - Tian Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
60
|
Gentile F, Doneddu PE, Riva N, Nobile-Orazio E, Quattrini A. Diet, Microbiota and Brain Health: Unraveling the Network Intersecting Metabolism and Neurodegeneration. Int J Mol Sci 2020; 21:E7471. [PMID: 33050475 PMCID: PMC7590163 DOI: 10.3390/ijms21207471] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence gives support for the idea that extra-neuronal factors may affect brain physiology and its predisposition to neurodegenerative diseases. Epidemiological and experimental studies show that nutrition and metabolic disorders such as obesity and type 2 diabetes increase the risk of Alzheimer's and Parkinson's diseases after midlife, while the relationship with amyotrophic lateral sclerosis is uncertain, but suggests a protective effect of features of metabolic syndrome. The microbiota has recently emerged as a novel factor engaging strong interactions with neurons and glia, deeply affecting their function and behavior in these diseases. In particular, recent evidence suggested that gut microbes are involved in the seeding of prion-like proteins and their spreading to the central nervous system. Here, we present a comprehensive review of the impact of metabolism, diet and microbiota in neurodegeneration, by affecting simultaneously several aspects of health regarding energy metabolism, immune system and neuronal function. Advancing technologies may allow researchers in the future to improve investigations in these fields, allowing the buildup of population-based preventive interventions and development of targeted therapeutics to halt progressive neurologic disability.
Collapse
Affiliation(s)
- Francesco Gentile
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.G.); (N.R.)
- Neuromuscular and Neuroimmunology Service, Humanitas Clinical and Research Institute IRCCS, 20089 Milan, Italy; (P.E.D.); (E.N.-O.)
| | - Pietro Emiliano Doneddu
- Neuromuscular and Neuroimmunology Service, Humanitas Clinical and Research Institute IRCCS, 20089 Milan, Italy; (P.E.D.); (E.N.-O.)
| | - Nilo Riva
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.G.); (N.R.)
- Department of Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Eduardo Nobile-Orazio
- Neuromuscular and Neuroimmunology Service, Humanitas Clinical and Research Institute IRCCS, 20089 Milan, Italy; (P.E.D.); (E.N.-O.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.G.); (N.R.)
| |
Collapse
|
61
|
Hung AY, Schwarzschild MA. Approaches to Disease Modification for Parkinson's Disease: Clinical Trials and Lessons Learned. Neurotherapeutics 2020; 17:1393-1405. [PMID: 33205384 PMCID: PMC7851299 DOI: 10.1007/s13311-020-00964-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Despite many clinical trials over the last three decades, the goal of demonstrating that a treatment slows the progression of Parkinson's disease (PD) remains elusive. Research advances have shed new insight into cellular pathways contributing to PD pathogenesis and offer increasingly compelling therapeutic targets. Here we review recent and ongoing clinical trials employing novel strategies toward disease modification, including those targeting alpha-synuclein and those repurposing drugs approved for other indications. Active and passive immunotherapy approaches are being studied with the goal to modify the spread of alpha-synuclein pathology in the brain. Classes of currently available drugs that have been proposed to have potential disease-modifying effects for PD include calcium channel blockers, antioxidants, anti-inflammatory agents, iron-chelating agents, glucagon-like peptide 1 agonists, and cAbl tyrosine kinase inhibitors. The mechanistic diversity of these treatments offers hope, but to date, results from these trials have been disappointing. Nevertheless, they provide useful lessons in guiding future therapeutic development.
Collapse
Affiliation(s)
- Albert Y Hung
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.
| | - Michael A Schwarzschild
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
| |
Collapse
|
62
|
Yakhine-Diop SM, Morales-García JA, Niso-Santano M, González-Polo RA, Uribe-Carretero E, Martinez-Chacon G, Durand S, Maiuri MC, Aiastui A, Zulaica M, Ruíz-Martínez J, López de Munain A, Pérez-Tur J, Pérez-Castillo A, Kroemer G, Bravo-San Pedro JM, Fuentes JM. Metabolic alterations in plasma from patients with familial and idiopathic Parkinson's disease. Aging (Albany NY) 2020; 12:16690-16708. [PMID: 32903216 PMCID: PMC7521510 DOI: 10.18632/aging.103992] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/15/2020] [Indexed: 01/24/2023]
Abstract
The research of new biomarkers for Parkinson's disease is essential for accurate and precocious diagnosis, as well as for the discovery of new potential disease mechanisms and drug targets. The main objective of this work was to identify metabolic changes that might serve as biomarkers for the diagnosis of this neurodegenerative disorder. For this, we profiled the plasma metabolome from mice with neurotoxin-induced Parkinson's disease as well as from patients with familial or sporadic Parkinson's disease. By using mass spectrometry technology, we analyzed the complete metabolome from healthy volunteers compared to patients with idiopathic or familial (carrying the G2019S or R1441G mutations in the LRRK2 gene) Parkinson's disease, as well as, from mice treated with 6-hydroxydopamine to induce Parkinson disease. Both human and murine Parkinson was accompanied by an increase in plasma levels of unconjugated bile acids (cholic acid, deoxycholic acid and lithocholic acid) and purine base intermediary metabolites, in particular hypoxanthine. The comprehensive metabolomic analysis of plasma from Parkinsonian patients underscores the importance of bile acids and purine metabolism in the pathophysiology of this disease. Therefore, plasma measurements of certain metabolites related to these pathways might contribute to the diagnosis of Parkinson's Disease.
Collapse
Affiliation(s)
- Sokhna M.S. Yakhine-Diop
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - José A. Morales-García
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto de Investigaciones Biomédicas (CSIC-UAM) “Alberto Sols” (CSIC-UAM), Madrid, Spain,Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Mireia Niso-Santano
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Rosa A. González-Polo
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Elisabet Uribe-Carretero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Guadalupe Martinez-Chacon
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Sylvere Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université de Paris, Sorbonne Université, Paris, France
| | - Ana Aiastui
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Cell Culture Platform, Biodonostia Health Research Institute, San Sebastián, Spain,Neuroscience Area of Biodonostia Health Research Institute, Donostia University Hospital, San Sebastián, Spain
| | - Miren Zulaica
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Neuroscience Area of Biodonostia Health Research Institute, Donostia University Hospital, San Sebastián, Spain
| | - Javier Ruíz-Martínez
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Neuroscience Area of Biodonostia Health Research Institute, Donostia University Hospital, San Sebastián, Spain,Donostia University Hospital, Department of Neurology, OSAKIDETZA, Spain,Ilundain Foundation, San Sebastian, Spain
| | - Adolfo López de Munain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Neuroscience Area of Biodonostia Health Research Institute, Donostia University Hospital, San Sebastián, Spain,Donostia University Hospital, Department of Neurology, OSAKIDETZA, Spain,Ilundain Foundation, San Sebastian, Spain,Department of Neurosciences, University of the Basque Country UPV-EHU, San Sebastián, Spain
| | - Jordi Pérez-Tur
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto de Biomedicina de Valencia-CSIC, Unidad de Genética Molecular, Valencia, Spain,Unidad Mixta de Genética y Neurología, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Ana Pérez-Castillo
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto de Investigaciones Biomédicas (CSIC-UAM) “Alberto Sols” (CSIC-UAM), Madrid, Spain
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université de Paris, Sorbonne Université, Paris, France,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, France,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - José M. Bravo-San Pedro
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - José M. Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| |
Collapse
|
63
|
Watanabe H, Hattori T, Kume A, Misu K, Ito T, Koike Y, Johnson TA, Kamitsuji S, Kamatani N, Sobue G. Improved Parkinsons disease motor score in a single-arm open-label trial of febuxostat and inosine. Medicine (Baltimore) 2020; 99:e21576. [PMID: 32871874 PMCID: PMC7458241 DOI: 10.1097/md.0000000000021576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cellular energetics play an important role in Parkinsons disease etiology, but no treatments directly address this deficiency. Our past research showed that treatment with febuxostat and inosine increased blood hypoxanthine and ATP in healthy adults, and a preliminary trial in 3 Parkinson's disease patients suggested some symptomatic improvements with no adverse effects. METHODS To examine the efficacy on symptoms and safety in a larger group of Parkinsons disease patients, we conducted a single-arm, open-label trial at 5 Japanese neurology clinics and enrolled thirty patients (nmales = 11; nfemales = 19); 26 patients completed the study (nmales = 10; nfemales = 16). Each patient was administered febuxostat 20 mg and inosine 500 mg twice-per-day (after breakfast and dinner) for 8 weeks. The primary endpoint was the difference of MDS-UPDRS Part III score immediately before and after 57 days of treatment. RESULTS Serum hypoxanthine concentrations were raised significantly after treatment (Pre = 11.4 μM; Post = 38.1 μM; P < .0001). MDS-UPDRS Part III score was significantly lower after treatment (Pre = 28.1 ± 9.3; Post = 24.7 ± 10.8; mean ± SD; P = .0146). Sixteen adverse events occurred in 13/29 (44.8%) patients, including 1 serious adverse event (fracture of the second lumbar vertebra) that was considered not related to the treatment. CONCLUSIONS The results of this study suggest that co-administration of febuxostat and inosine is relatively safe and effective for improving symptoms of Parkinsons disease patients. Further controlled trials need to be performed to confirm the symptomatic improvement and to examine the disease-modifying effect in long-term trials.
Collapse
Affiliation(s)
- Hirohisa Watanabe
- Nagoya University Graduate School of Medicine, Brain and Mind Research Center, Nagoya
- Fujita Health University School of Medicine, Department of Neurology, Toyoake
| | | | | | | | | | | | | | | | | | - Gen Sobue
- Nagoya University Graduate School of Medicine, Brain and Mind Research Center, Nagoya
| |
Collapse
|
64
|
Ellmore TM, Suescun J, Castriotta RJ, Schiess MC. A Study of the Relationship Between Uric Acid and Substantia Nigra Brain Connectivity in Patients With REM Sleep Behavior Disorder and Parkinson's Disease. Front Neurol 2020; 11:815. [PMID: 32849245 PMCID: PMC7419698 DOI: 10.3389/fneur.2020.00815] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 06/29/2020] [Indexed: 01/28/2023] Open
Abstract
Low levels of the natural antioxidant uric acid (UA) and the presence of REM sleep behavior disorder (RBD) are both associated with an increased likelihood of developing Parkinson's disease (PD). RBD and PD are also accompanied by basal ganglia dysfunction including decreased nigrostriatal and nigrocortical resting state functional connectivity. Despite these independent findings, the relationship between UA and substantia nigra (SN) functional connectivity remains unknown. In the present study, voxelwise analysis of covariance was used in a cross-sectional design to explore the relationship between UA and whole-brain SN functional connectivity using the eyes-open resting state fMRI method in controls without RBD, patients with idiopathic RBD, and PD patients with and without RBD. The results showed that controls exhibited a positive relationship between UA and SN functional connectivity with left lingual gyrus. The positive relationship was reduced in patients with RBD and PD with RBD, and the relationship was found to be negative in PD patients. These results are the first to show differential relationships between UA and SN functional connectivity among controls, prodromal, and diagnosed PD patients in a ventral occipital region previously documented to be metabolically and structurally altered in RBD and PD. More investigation, including replication in longitudinal designs with larger samples, is needed to understand the pathophysiological significance of these changes.
Collapse
Affiliation(s)
- Timothy M Ellmore
- Department of Psychology, The City College of New York, New York, NY, United States
| | - Jessika Suescun
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
| | - Richard J Castriotta
- Department of Clinical Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States
| | - Mya C Schiess
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
| |
Collapse
|
65
|
Von Seggern M, Szarowicz C, Swanson M, Cavotta S, Pike ST, Lamberts JT. Purine molecules in Parkinson's disease: Analytical techniques and clinical implications. Neurochem Int 2020; 139:104793. [PMID: 32650026 DOI: 10.1016/j.neuint.2020.104793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 10/23/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that primarily affects patients over the age of 65. PD is characterized by loss of neurons in the substantia nigra and dopamine deficiency in the striatum. Once PD is clinically diagnosed by the observation of motor dysfunction, the disease is already in its advance stages. Consequently, there is a major push to identify clinical biomarkers that are useful for the earlier detection of PD. Using untargeted metabolomics, several research groups have identified purine molecules, and specifically urate, as important biomarkers related to PD. This review will summarize recent findings in the field of purine metabolomics and biomarker identification for PD, including in the areas of PD pathophysiology, diagnosis, prognosis and treatment. In addition, this article will summarize and examine the primary research techniques that are employed to quantify purine molecules in both experimental systems and human subjects.
Collapse
Affiliation(s)
| | - Carlye Szarowicz
- College of Arts & Sciences, Ferris State University, Big Rapids, MI, USA; Shimadzu Core Laboratory for Academic and Research Excellence, Ferris State University, Big Rapids, MI, USA
| | - Matthew Swanson
- College of Arts & Sciences, Ferris State University, Big Rapids, MI, USA; Shimadzu Core Laboratory for Academic and Research Excellence, Ferris State University, Big Rapids, MI, USA
| | - Samantha Cavotta
- College of Pharmacy, Ferris State University, Big Rapids, MI, USA
| | - Schuyler T Pike
- College of Arts & Sciences, Ferris State University, Big Rapids, MI, USA; Shimadzu Core Laboratory for Academic and Research Excellence, Ferris State University, Big Rapids, MI, USA
| | | |
Collapse
|
66
|
Chang KH, Chen CM. The Role of Oxidative Stress in Parkinson's Disease. Antioxidants (Basel) 2020; 9:antiox9070597. [PMID: 32650609 PMCID: PMC7402083 DOI: 10.3390/antiox9070597] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Parkinson’s disease (PD) is caused by progressive neurodegeneration of dopaminergic (DAergic) neurons with abnormal accumulation of α-synuclein in substantia nigra (SN). Studies have suggested the potential involvement of dopamine, iron, calcium, mitochondria and neuroinflammation in contributing to overwhelmed oxidative stress and neurodegeneration in PD. Function studies on PD-causative mutations of SNCA, PRKN, PINK1, DJ-1, LRRK2, FBXO7 and ATP13A2 further indicate the role of oxidative stress in the pathogenesis of PD. Therefore, it is reasonable that molecules involved in oxidative stress, such as DJ-1, coenzyme Q10, uric acid, 8-hydroxy-2’-deoxyguanosin, homocysteine, retinoic acid/carotenes, vitamin E, glutathione peroxidase, superoxide dismutase, xanthine oxidase and products of lipid peroxidation, could be candidate biomarkers for PD. Applications of antioxidants to modulate oxidative stress could be a strategy in treating PD. Although a number of antioxidants, such as creatine, vitamin E, coenzyme Q10, pioglitazone, melatonin and desferrioxamine, have been tested in clinical trials, none of them have demonstrated conclusive evidence to ameliorate the neurodegeneration in PD patients. Difficulties in clinical studies may be caused by the long-standing progression of neurodegeneration, lack of biomarkers for premotor stage of PD and inadequate drug delivery across blood–brain barrier. Solutions for these challenges will be warranted for future studies with novel antioxidative treatment in PD patients.
Collapse
Affiliation(s)
| | - Chiung-Mei Chen
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8347); Fax: +886-3-3288849
| |
Collapse
|
67
|
Ugrumov M. Development of early diagnosis of Parkinson's disease: Illusion or reality? CNS Neurosci Ther 2020; 26:997-1009. [PMID: 32597012 PMCID: PMC7539842 DOI: 10.1111/cns.13429] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
The fight against neurodegenerative diseases, Alzheimer disease and Parkinson's disease (PD), is a challenge of the 21st century. The low efficacy of treating patients is due to the late diagnosis and start of therapy, after the degeneration of most specific neurons and depletion of neuroplasticity. It is believed that the development of early diagnosis (ED) and preventive treatment will delay the onset of specific symptoms. This review evaluates methodologies for developing ED of PD. Since PD is a systemic disease, and the degeneration of certain neurons precedes that of nigrostriatal dopaminergic neurons that control motor function, the current methodology is based on searching biomarkers, such as premotor symptoms and changes in body fluids (BF) in patients. However, all attempts to develop ED were unsuccessful. Therefore, it is proposed to enhance the current methodology by (i) selecting among biomarkers found in BF in patients at the clinical stage those that are characteristics of animal models of the preclinical stage, (ii) searching biomarkers in BF in subjects at the prodromal stage, selected by detecting premotor symptoms and failure of the nigrostriatal dopaminergic system. Moreover, a new methodology was proposed for the development of ED of PD using a provocative test, which is successfully used in internal medicine.
Collapse
Affiliation(s)
- Michael Ugrumov
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology RAS, Moscow, Russia
| |
Collapse
|
68
|
Jung JH, Chung SJ, Yoo HS, Lee YH, Baik K, Ye BS, Sohn YH, Lee PH. Sex-specific association of urate and levodopa-induced dyskinesia in Parkinson's disease. Eur J Neurol 2020; 27:1948-1956. [PMID: 32441832 DOI: 10.1111/ene.14337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/14/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE As a major antioxidant, uric acid (UA) is known to be associated with the clinical progression of Parkinson's disease (PD). This study investigated whether baseline UA levels are associated with the risk for levodopa-induced dyskinesia (LID) in PD in a sex-dependent manner. METHODS In all, 152 patients with de novo PD (78 males and 74 females) who were followed up for >2 years were enrolled. The effect of baseline serum UA levels on LID-free survival was assessed by Cox regression, separately for sex, whilst being adjusted for potential confounding factors. The optimal UA level cut-off value to determine the high-risk group for LID was set using Contal and O'Quigley's method. RESULTS Levodopa-induced dyskinesia developed in 23 (29.5%) male patients and 30 (40.5%) female patients. Cox regression showed a significant interaction between UA level and sex. Higher UA levels were associated with a higher risk for LID in male PD patients (hazard ratio 1.380; 95% confidence interval 1.038-1.835; P = 0.027), although this relationship was not observed in female PD patients. The optimal UA level cut-off for LID in male PD was 7.2 mg/dl, and the high UA group had a 5.7-fold higher risk of developing LID than the low UA group. CONCLUSIONS Contrary to a presumptive beneficial role of UA, the present study demonstrated that higher UA levels are associated with increased risk of LID occurrence in male patients with PD, suggesting a sex-dependent role of UA in LID.
Collapse
Affiliation(s)
- J H Jung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - S J Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea.,Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - H S Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Y H Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - K Baik
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - B S Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Y H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - P H Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
69
|
Cerri S, Mus L, Blandini F. Parkinson's Disease in Women and Men: What's the Difference? JOURNAL OF PARKINSONS DISEASE 2020; 9:501-515. [PMID: 31282427 PMCID: PMC6700650 DOI: 10.3233/jpd-191683] [Citation(s) in RCA: 392] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increasing evidence points to biological sex as an important factor in the development and phenotypical expression of Parkinson’s disease (PD). Risk of developing PD is twice as high in men than women, but women have a higher mortality rate and faster progression of the disease. Moreover, motor and nonmotor symptoms, response to treatments and disease risk factors differ between women and men. Altogether, sex-related differences in PD support the idea that disease development might involve distinct pathogenic mechanisms (or the same mechanism but in a different way) in male and female patients. This review summarizes the most recent knowledge concerning differences between women and men in PD clinical features, risk factors, response to treatments and mechanisms underlying the disease pathophysiology. Unraveling how the pathology differently affect the two sexes might allow the development of tailored interventions and the design of innovative programs that meet the distinct needs of men and women, improving patient care.
Collapse
Affiliation(s)
- Silvia Cerri
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Liudmila Mus
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Blandini
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
70
|
Barp A, Gerardi F, Lizio A, Sansone VA, Lunetta C. Emerging Drugs for the Treatment of Amyotrophic Lateral Sclerosis: A Focus on Recent Phase 2 Trials. Expert Opin Emerg Drugs 2020; 25:145-164. [PMID: 32456491 DOI: 10.1080/14728214.2020.1769067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease involving both upper and lower motor neurons and resulting in increasing disability and death 3-5 years after onset of symptoms. Over 40 large clinical trials for ALS have been negative, except for Riluzole that offers a modest survival benefit, and Edaravone that modestly reduces disease progression in patients with specific characteristics. Thus, the discovery of efficient disease modifying therapy is an urgent need. AREAS COVERED Although the cause of ALS remains unclear, many studies have demonstrated that neuroinflammation, proteinopathies, glutamate-induced excitotoxicity, microglial activation, oxidative stress, and mitochondrial dysfunction may play a key role in the pathogenesis. This review highlights recent discoveries relating to these diverse mechanisms and their implications for the development of therapy. Ongoing phase 2 clinical trials aimed to interfere with these pathophysiological mechanisms are discussed. EXPERT OPINION This review describes the challenges that the discovery of an efficient drug therapy faces and how these issues may be addressed. With the continuous advances coming from basic research, we provided possible suggestions that may be considered to improve performance of clinical trials and turn ALS research into a 'fertile ground' for drug development for this devastating disease.
Collapse
Affiliation(s)
- Andrea Barp
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy.,Dept. Biomedical Sciences of Health, University of Milan , Milan, Italy
| | | | - Andrea Lizio
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy
| | - Valeria Ada Sansone
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy.,Dept. Biomedical Sciences of Health, University of Milan , Milan, Italy
| | | |
Collapse
|
71
|
Trifonova OP, Maslov DL, Balashova EE, Urazgildeeva GR, Abaimov DA, Fedotova EY, Poleschuk VV, Illarioshkin SN, Lokhov PG. Parkinson's Disease: Available Clinical and Promising Omics Tests for Diagnostics, Disease Risk Assessment, and Pharmacotherapy Personalization. Diagnostics (Basel) 2020; 10:E339. [PMID: 32466249 PMCID: PMC7277996 DOI: 10.3390/diagnostics10050339] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease is the second most frequent neurodegenerative disease, representing a significant medical and socio-economic problem. Modern medicine still has no answer to the question of why Parkinson's disease develops and whether it is possible to develop an effective system of prevention. Therefore, active work is currently underway to find ways to assess the risks of the disease, as well as a means to extend the life of patients and improve its quality. Modern studies aim to create a method of assessing the risk of occurrence of Parkinson's disease (PD), to search for the specific ways of correction of biochemical disorders occurring in the prodromal stage of Parkinson's disease, and to personalize approaches to antiparkinsonian pharmacotherapy. In this review, we summarized all available clinically approved tests and techniques for PD diagnostics. Then, we reviewed major improvements and recent advancements in genomics, transcriptomics, and proteomics studies and application of metabolomics in PD research, and discussed the major metabolomics findings for diagnostics and therapy of the disease.
Collapse
Affiliation(s)
- Oxana P. Trifonova
- Laboratory of mass spectrometry-based metabolomics diagnostics, Institute of Biomedical Chemistry, 10 building 8, Pogodinskaya street, 119121 Moscow, Russia; (D.L.M.); (E.E.B.); (P.G.L.)
| | - Dmitri L. Maslov
- Laboratory of mass spectrometry-based metabolomics diagnostics, Institute of Biomedical Chemistry, 10 building 8, Pogodinskaya street, 119121 Moscow, Russia; (D.L.M.); (E.E.B.); (P.G.L.)
| | - Elena E. Balashova
- Laboratory of mass spectrometry-based metabolomics diagnostics, Institute of Biomedical Chemistry, 10 building 8, Pogodinskaya street, 119121 Moscow, Russia; (D.L.M.); (E.E.B.); (P.G.L.)
| | - Guzel R. Urazgildeeva
- 5th Neurological Department (Department of Neurogenetics), Research Centre of Neurology, Volokolamskoe shosse, 80, 125367 Moscow, Russia; (G.R.U.); (D.A.A.); (E.Y.F.); (V.V.P.); (S.N.I.)
| | - Denis A. Abaimov
- 5th Neurological Department (Department of Neurogenetics), Research Centre of Neurology, Volokolamskoe shosse, 80, 125367 Moscow, Russia; (G.R.U.); (D.A.A.); (E.Y.F.); (V.V.P.); (S.N.I.)
| | - Ekaterina Yu. Fedotova
- 5th Neurological Department (Department of Neurogenetics), Research Centre of Neurology, Volokolamskoe shosse, 80, 125367 Moscow, Russia; (G.R.U.); (D.A.A.); (E.Y.F.); (V.V.P.); (S.N.I.)
| | - Vsevolod V. Poleschuk
- 5th Neurological Department (Department of Neurogenetics), Research Centre of Neurology, Volokolamskoe shosse, 80, 125367 Moscow, Russia; (G.R.U.); (D.A.A.); (E.Y.F.); (V.V.P.); (S.N.I.)
| | - Sergey N. Illarioshkin
- 5th Neurological Department (Department of Neurogenetics), Research Centre of Neurology, Volokolamskoe shosse, 80, 125367 Moscow, Russia; (G.R.U.); (D.A.A.); (E.Y.F.); (V.V.P.); (S.N.I.)
| | - Petr G. Lokhov
- Laboratory of mass spectrometry-based metabolomics diagnostics, Institute of Biomedical Chemistry, 10 building 8, Pogodinskaya street, 119121 Moscow, Russia; (D.L.M.); (E.E.B.); (P.G.L.)
| |
Collapse
|
72
|
Christine CW, Auinger P, Saleh N, Tian M, Bottiglieri T, Arning E, Tran NK, Ueland PM, Green R, the Parkinson Study Group—DATATOP Investigators. Relationship of Cerebrospinal Fluid Vitamin B12 Status Markers With Parkinson's Disease Progression. Mov Disord 2020; 35:1466-1471. [PMID: 32407590 PMCID: PMC7496300 DOI: 10.1002/mds.28073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Using blood specimens from untreated early Parkinson's disease (PD) patients from the DATATOP trial, we found that subjects in the low serum vitamin B12 tertile experienced greater annualized change in ambulatory capacity score, whereas those with moderately elevated (>15 μmol/L) total homocysteine had greater annualized declines in the Mini‐Mental State Exam. Methods In this this study we sought to determine whether levels of cerebrospinal fluid (CSF) B12 markers were also associated with progression of PD. Results The annualized change in the UPDRS “walking” item, a component of the ambulatory capacity score, was worse in the low B12 tertile. No association with change in the Mini‐Mental State Exam was seen for those 7% with the highest baseline CSF total homocysteine. Conclusions In these untreated early‐PD subjects, low CSF B12 predicted greater worsening of the UPDRS “walking” item, whereas CSF total homocysteine was not associated with progression of cognitive impairment. These findings extend and partially support our findings in serum. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Peggy Auinger
- Center for Health and TechnologyUniversity of RochesterRochesterNew YorkUSA
| | - Nasrin Saleh
- Department of Pathology and Laboratory MedicineUC DavisSacramentoCaliforniaUSA
| | - Miao Tian
- Department of Pathology and Laboratory MedicineUC DavisSacramentoCaliforniaUSA
| | - Teodoro Bottiglieri
- Institute of Metabolic Disease, Baylor Scott & White Research InstituteDallasTexasUSA
| | - Erland Arning
- Institute of Metabolic Disease, Baylor Scott & White Research InstituteDallasTexasUSA
| | - Nam K. Tran
- Department of Pathology and Laboratory MedicineUC DavisSacramentoCaliforniaUSA
| | | | - Ralph Green
- Department of Pathology and Laboratory MedicineUC DavisSacramentoCaliforniaUSA
| | | |
Collapse
|
73
|
Sleeman I, Lawson RA, Yarnall AJ, Duncan GW, Johnston F, Khoo TK, Burn DJ. Urate and Homocysteine: Predicting Motor and Cognitive Changes in Newly Diagnosed Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2020; 9:351-359. [PMID: 30909247 PMCID: PMC6597987 DOI: 10.3233/jpd-181535] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Urate and homocysteine are potential biomarkers for disease progression in Parkinson's disease (PD). Baseline serum urate concentration has been shown to predict motor but not cognitive decline. The relationship between serum homocysteine concentration and cognitive and motor impairment is unknown. OBJECTIVES The aim of this study was to examine the association between baseline serum urate and homocysteine, and prospective measures of disease progression and cognition over 54 months in early PD. METHODS 154 newly diagnosed PD participants and 99 age-matched controls completed a schedule of assessments at baseline, 18, 36 and 54 months. The Movement Disorders Society Unified Parkinson's Disease Scale Part III (MDS-UPDRS III) was used to assess motor severity. The Montreal Cognitive Assessment (MoCA) was used to assess global cognition. Serum samples drawn at baseline were analysed for urate, homocysteine, red cell folate and vitamin B12 concentrations. RESULTS Baseline urate was 331.4±83.8 and 302.7±78.0μmol/L for control and PD participants, respectively (p = 0.015). Baseline homocysteine was 9.6±3.3 and 11.1±3.8μmol/L for controls and PD participants, respectively (p < 0.01). Linear mixed effects modelling showed that lower baseline urate (β= 0.02, p < 0.001) and higher homocysteine (β= 0.29, p < 0.05) predicted decline in motor function. Only higher homocysteine concentrations at baseline, however, predicted declining MoCA scores over 54 months (β= 0.11, p < 0.01). CONCLUSIONS Lower serum urate concentration is associated with worsening motor function; while higher homocysteine concentration is associated with change in motor function and cognitive decline. Therefore, urate and homocysteine may be suitable biomarkers for predicting motor and cognitive decline in early PD.
Collapse
Affiliation(s)
- Isobel Sleeman
- Institute of Applied Health Sciences, University of Aberdeen, UK.,Institute of Neuroscience, Newcastle University, UK
| | | | | | - Gordon W Duncan
- Institute of Neuroscience, Newcastle University, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | - Tien K Khoo
- School of Medicine and Menzies Health Institute Queensland, Griffith University, Australia.,School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | - David J Burn
- Faculty of Medical Sciences, Newcastle University, UK
| |
Collapse
|
74
|
Franco R, Rivas-Santisteban R, Reyes-Resina I, Navarro G, Martínez-Pinilla E. Microbiota and Other Preventive Strategies and Non-genetic Risk Factors in Parkinson's Disease. Front Aging Neurosci 2020; 12:12. [PMID: 32226375 PMCID: PMC7080700 DOI: 10.3389/fnagi.2020.00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
The exact cause of Parkinson’s disease (PD), the second most prevalent neurodegenerative disease in modern societies, is still unknown. Many scientists point out that PD is caused by a complex interaction between different factors. Although the main risk factor is age, there are other influences, genetic and environmental, that individually or in combination may trigger neurodegenerative changes leading to PD. Nowadays, research remains focused on better understanding which environmental factors are related to the risk of developing PD and why. In line with the knowledge on evidence on exposures that prevent/delay PD onset or that impact on disease progression, the aims of this review were: (i) to comment on the non-genetic risk factors that mainly affect idiopathic PD; and (ii) to comment on seemingly reliable preventive interventions. We discuss both environmental factors that may affect the central nervous system (CNS) or the intestinal tract, and the likely mechanisms underlying noxious or protective actions. Knowledge on risk, protective factors, and mechanisms may help to envisage why nigral dopaminergic neurons are so vulnerable in PD and, eventually, to design new strategies for PD prevention and/or anti-PD therapy. This article reviews the variety of the known and suspected environmental factors, such as lifestyle, gut microbiota or pesticide exposition, and distinguishes between those that are harmful or beneficial for the PD acquisition or progression. In fact, the review covers one of the most novel players in the whole picture, and we address the role of microbiota on keeping a healthy CNS and/or on preventing the “side-effects” related to aging.
Collapse
Affiliation(s)
- Rafael Franco
- Chemistry School, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Rivas-Santisteban
- Chemistry School, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Eva Martínez-Pinilla
- Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
75
|
Songsomboon C, Tanprawate S, Soontornpun A, Wantaneeyawong C, Louthrenoo W. Serum Uric Acid, Serum Uric Acid to Serum Creatinine Ratio and Serum Bilirubin in Patients With Parkinson's Disease: A Case-Control Study. J Clin Med Res 2020; 12:172-179. [PMID: 32231753 PMCID: PMC7092755 DOI: 10.14740/jocmr4079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/15/2020] [Indexed: 01/20/2023] Open
Abstract
Background Studies have shown that a low serum uric acid (SUA) level associates with Parkinson’s disease (PD), but many of them did not exclude patients with impaired renal function. Studies on the association between serum bilirubin level and PD also are limited. This study determined the association between SUA level, SUA/serum creatinine (SCr) ratio and serum bilirubin levels in PD patients with normal renal and liver functions. Methods The PD patients from a neurological clinic, and the controls from the club for the elderly, were recruited into this study. The PD stage and motor and non-motor function were determined by the Hoehn-Yahr (H&Y) scale and unified Parkinson’s disease rating scale (UPDRS), respectively. Results Sixty-one PD patients and 135 controls participated. The SUA/SCr ratio, but not SUA, was significantly lower in the PD patients than in the controls (4.12 ± 0.90 vs. 4.59 ± 1.04, P = 0.003). Serum total bilirubin (TB) and indirect bilirubin (IDB) were significantly higher in the PD patients (7.92 ± 3.67 µmol/L vs. 6.59 ± 2.78 µmol/L, P = 0.003 and 4.52 ± 2.48 µmol/L vs. 3.26 ± 1.82 µmol/L, P < 0.001), respectively. Serum TB and IDB, but not SUA or SUA/SCr ratio, were associated negatively with PD stages (P = 0.010 and P = 0.014, respectively). There was no association between TB, IDB, SUA or SUA/SCr ratio and PD disease duration or motor subtypes. No significant correlation was found between SUA or SUA/SCr ratio, serum TB and IDB. Conclusion The SUA/SCr ratio is more sensitive than SUA in determining their association with PD. The high serum TB and IDB levels in PD patients compared with the controls suggest that serum bilirubin might play a role in the pathogenesis of PD. However, the lack of association between SUA or the SUA/SCr ratio and serum TB or IDB suggests that these two biomarkers play a different role in the etiopathogenesis of PD.
Collapse
Affiliation(s)
- Chayanon Songsomboon
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Current address: Lampang Hospital, Lampang, Thailand
| | - Surat Tanprawate
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Atiwat Soontornpun
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chayasak Wantaneeyawong
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Current address: The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Worawit Louthrenoo
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
76
|
Abstract
BACKGROUND Formate is a one-carbon molecule at the crossroad between cellular and whole body metabolism, between host and microbiome metabolism, and between nutrition and toxicology. This centrality confers formate with a key role in human physiology and disease that is currently unappreciated. SCOPE OF REVIEW Here we review the scientific literature on formate metabolism, highlighting cellular pathways, whole body metabolism, and interactions with the diet and the gut microbiome. We will discuss the relevance of formate metabolism in the context of embryonic development, cancer, obesity, immunometabolism, and neurodegeneration. MAJOR CONCLUSIONS We will conclude with an outlook of some open questions bringing formate metabolism into the spotlight.
Collapse
Affiliation(s)
| | - Johannes Meiser
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Alexei Vazquez
- Cancer Research UK Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
77
|
Baik K, Chung SJ, Yoo HS, Lee YH, Jung JH, Sohn YH, Lee PH. Sex‐dependent association of urate on the patterns of striatal dopamine depletion in Parkinson’s disease. Eur J Neurol 2020; 27:773-778. [DOI: 10.1111/ene.14152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023]
Affiliation(s)
- K. Baik
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
| | - S. J. Chung
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
| | - H. S. Yoo
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
| | - Y. H. Lee
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
| | - J. H. Jung
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
| | - Y. H. Sohn
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
| | - P. H. Lee
- Department of Neurology Yonsei University College of Medicine Seoul South Korea
- Severance Biomedical Science Institute Yonsei University College of Medicine Seoul South Korea
| |
Collapse
|
78
|
Rasagiline and selegiline modulate mitochondrial homeostasis, intervene apoptosis system and mitigate α-synuclein cytotoxicity in disease-modifying therapy for Parkinson's disease. J Neural Transm (Vienna) 2020; 127:131-147. [PMID: 31993732 DOI: 10.1007/s00702-020-02150-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/21/2020] [Indexed: 12/16/2022]
Abstract
Parkinson's disease has been considered as a motor neuron disease with dopamine (DA) deficit caused by neuronal loss in the substantia nigra, but now proposed as a multi-system disorder associated with α-synuclein accumulation in neuronal and non-neuronal systems. Neuroprotection in Parkinson's disease has intended to halt or reverse cell death of nigro-striatal DA neurons and prevent the disease progression, but clinical studies have not presented enough beneficial results, except the trial of rasagiline by delayed start design at low dose of 1 mg/day only. Now strategy of disease-modifying therapy should be reconsidered taking consideration of accumulation and toxicity of α-synuclein preceding the manifest of motor symptoms. Hitherto neuroprotective therapy has been aimed to mitigate non-specific risk factors; oxidative stress, mitochondrial dysfunction, apoptosis, deficits of neurotrophic factors (NTFs), inflammation and accumulation of pathogenic protein. Future disease-modify therapy should target more specified pathogenic factors, including deregulated mitochondrial homeostasis, deficit of NTFs and α-synuclein toxicity. Selegiline and rasagiline, inhibitors of type B monoamine oxidase, have been proved to exhibit potent neuroprotective function: regulation of mitochondrial apoptosis system, maintenance of mitochondrial function, increased expression of genes coding antioxidant enzymes, anti-apoptotic Bcl-2 and pro-survival NTFs, and suppression of oligomerization and aggregation of α-synuclein and the toxicity in cellular and animal experiments. However, the present available pharmacological therapy starts too late to reverse disease progression, and future disease-modifying therapy should include also non-pharmacological complementary therapy during the prodromal stage.
Collapse
|
79
|
Abstract
In a range of neurological conditions, including movement disorders, sex-related differences are emerging not only in brain anatomy and function, but also in pathogenesis, clinical features and response to treatment. In Parkinson disease (PD), for example, oestrogens can influence the severity of motor symptoms, whereas elevation of androgens can exacerbate tic disorders. Nevertheless, the real impact of sex differences in movement disorders remains under-recognized. In this article, we provide an up-to-date review of sex-related differences in PD and the most common hyperkinetic movement disorders, namely, essential tremor, dystonia, Huntington disease and other chorea syndromes, and Tourette syndrome and other chronic tic disorders. We highlight the most relevant clinical aspects of movement disorders that differ between men and women. Increased recognition of these differences and their impact on patient care could aid the development of tailored approaches to the management of movement disorders and enable the optimization of preclinical research and clinical studies.
Collapse
|
80
|
Abstract
Parkinson's disease (PD) is a chronic, debilitating neurodegenerative disorder characterized clinically by a variety of progressive motor and nonmotor symptoms. Currently, there is a dearth of diagnostic tools available to predict, diagnose or mitigate disease risk or progression, leading to a challenging dilemma within the healthcare management system. The search for a reliable biomarker for PD that reflects underlying pathology is a high priority in PD research. Currently, there is no reliable single biomarker predictive of risk for motor and cognitive decline, and there have been few longitudinal studies of temporal progression. A combination of multiple biomarkers might facilitate earlier diagnosis and more accurate prognosis in PD. In this review, we focus on the recent developments of serial biomarkers for PD from a variety of clinical, biochemical, genetic and neuroimaging perspectives.
Collapse
Affiliation(s)
- Anastasia Bougea
- Neurochemistry Laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Neuroscience Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
81
|
Ghanta MK, Elango P, L V K S B. Current Therapeutic Strategies and Perspectives for Neuroprotection in Parkinson's Disease. Curr Pharm Des 2020; 26:4738-4746. [PMID: 32065086 DOI: 10.2174/1381612826666200217114658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/10/2020] [Indexed: 02/04/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder of dopaminergic striatal neurons in basal ganglia. Treatment of Parkinson's disease (PD) through dopamine replacement strategies may provide improvement in early stages and this treatment response is related to dopaminergic neuronal mass which decreases in advanced stages. This treatment failure was revealed by many studies and levodopa treatment became ineffective or toxic in chronic stages of PD. Early diagnosis and neuroprotective agents may be a suitable approach for the treatment of PD. The essentials required for early diagnosis are biomarkers. Characterising the striatal neurons, understanding the status of dopaminergic pathways in different PD stages may reveal the effects of the drugs used in the treatment. This review updates on characterisation of striatal neurons, electrophysiology of dopaminergic pathways in PD, biomarkers of PD, approaches for success of neuroprotective agents in clinical trials. The literature was collected from the articles in database of PubMed, MedLine and other available literature resources.
Collapse
Affiliation(s)
- Mohan K Ghanta
- Department of Pharmacology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research (DU), Porur, Chennai-600116, Tamil Nadu, India
| | - P Elango
- Department of Pharmacology, Panimalar Medical College Hospital & Research Institute, Poonamallee, Chennai-600123, Tamil Nadu, India
| | - Bhaskar L V K S
- Department of Zoology, Guru Ghasidas University, Bilaspur, 495009 (CG), India
| |
Collapse
|
82
|
Salamon A, Zádori D, Szpisjak L, Klivényi P, Vécsei L. Neuroprotection in Parkinson's disease: facts and hopes. J Neural Transm (Vienna) 2019; 127:821-829. [PMID: 31828513 PMCID: PMC7242234 DOI: 10.1007/s00702-019-02115-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022]
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide. Behind the symptoms there is a complex pathological mechanism which leads to a dopaminergic cell loss in the substantia nigra pars compacta. Despite the strong efforts, curative treatment has not been found yet. To prevent a further cell death, numerous molecules were tested in terms of neuroprotection in preclinical (in vitro, in vivo) and in clinical studies as well. The aim of this review article is to summarize our knowledge about the extensively tested neuroprotective agents (Search period: 1991–2019). We detail the underlying pathological mechanism and summarize the most important results of the completed animal and clinical trials. Although many positive results have been reported in the literature, there is still no evidence that any of them should be used in clinical practice (Cochrane analysis was performed). Therefore, further studies are needed to better understand the pathomechanism of PD and to find the optimal neuroprotective agent(s).
Collapse
Affiliation(s)
- András Salamon
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - Dénes Zádori
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - László Szpisjak
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - Péter Klivényi
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary. .,MTA-SZTE Neuroscience Research Group, Szeged, Hungary.
| |
Collapse
|
83
|
Chittoor G, Haack K, Balakrishnan P, Bizon C, Laston S, Best LG, MacCluer JW, North KE, Umans JG, Franceschini N, Prasad G, Macias-Kauffer L, Villarreal-Molina T, Bharadwaj D, Canizales-Quinteros S, Navas-Acien A, Cole SA, Voruganti VS. Fine mapping and identification of serum urate loci in American Indians: The Strong Heart Family Study. Sci Rep 2019; 9:17899. [PMID: 31784582 PMCID: PMC6884539 DOI: 10.1038/s41598-019-52924-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 10/12/2019] [Indexed: 02/07/2023] Open
Abstract
While studies have reported genetic loci affecting serum urate (SU) concentrations, few studies have been conducted in minority populations. Our objective for this study was to identify genetic loci regulating SU in a multigenerational family-based cohort of American Indians, the Strong Heart Family Study (SHFS). We genotyped 162,718 single nucleotide polymorphisms (SNPs) in 2000 SHFS participants using an Illumina MetaboChip array. A genome-wide association analysis of SU was conducted using measured genotype analysis approach accounting for kinships in SOLAR, and meta-analysis in METAL. Our results showed strong association of SU with rs4481233, rs9998811, rs7696092 and rs13145758 (minor allele frequency (MAF) = 25-44%; P < 3 × 10-14) of solute carrier family 2, member 9 (SLC2A9) and rs41481455, rs2231142 and rs1481012 (MAF = 29%; p < 3 × 10-9) of ATP-binding cassette protein, subfamily G, member 2 (ABCG2). Carriers of G alleles of rs9998811, rs4148155 and rs1481012 and A alleles of rs4481233, rs7696092 and rs13145758 and rs2231142 had lower SU concentrations as compared to non-carriers. Genetic analysis of SU conditional on significant SLC2A9 and ABCG2 SNPs revealed new loci, nucleobindin 1 (NUCB1) and neuronal PAS domain protein 4 (NPAS4) (p <6× 10-6). To identify American Indian-specific SNPs, we conducted targeted sequencing of key regions of SLC2A9. A total of 233 SNPs were identified of which 89 were strongly associated with SU (p < 7.1 × 10-10) and 117 were American Indian specific. Analysis of key SNPs in cohorts of Mexican-mestizos, European, Indian and East Asian ancestries showed replication of common SNPs, including our lead SNPs. Our results demonstrate the association of SU with uric acid transporters in a minority population of American Indians and potential novel associations of SU with neuronal-related genes which warrant further investigation.
Collapse
Affiliation(s)
- Geetha Chittoor
- Department of Nutrition, and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
- Biomedical and Translational Informatics, Geisinger, Danville, PA, USA
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Poojitha Balakrishnan
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Christopher Bizon
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Sandra Laston
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Lyle G Best
- Missouri Breaks Industries Research Inc., Eagle Butte, SD, USA
| | - Jean W MacCluer
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jason G Umans
- Medstar Health Research Institute, Hyattsville, MD, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gauri Prasad
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110 020, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi, 110 020, India
| | - Luis Macias-Kauffer
- Laboratorio de Enfermedades Cardiovasculares, INMEGEN, Mexico City, 14610, Mexico
| | | | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi, 110 020, India
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110 067, India
| | - Samuel Canizales-Quinteros
- Unidad de Genomica de Poblaciones Aplicada a la Salud Facultad de Quimica, UNAM-Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - V S Voruganti
- Department of Nutrition, and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA.
| |
Collapse
|
84
|
Shen Y, Li J, Schwarzschild M, Pavlova M, He S, Ascherio A, Wu S, Cui L, Gao X. Plasma urate concentrations and possible REM sleep behavior disorder. Ann Clin Transl Neurol 2019; 6:2368-2376. [PMID: 31714690 PMCID: PMC6917330 DOI: 10.1002/acn3.50929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/02/2022] Open
Abstract
Objective To examine how urate concentrations are related to the risk of having possible REM sleep behavior disorder (pRBD) in a community‐based cohort. Methods The study included 12,923 Chinese adults of the Kailuan Study, free of Parkinson disease (PD) and dementia. Plasma urate concentrations were measured in 2006, 2008, and 2010. Cumulative average urate concentration was used as primary exposure. In 2012, we determined pRBD status using a validated RBD questionnaire‐Hong Kong (RBDQ‐HK). Logistic regression analysis was performed to estimate the association between urate concentrations during 2006–2010 and odds of having pRBD in 2012 or pRBD case with symptom onset within 1 year. Results Higher average urate concentrations were associated with a lower odds of pRBD (P‐trend <0.001). The adjusted odds ratio (OR), for the highest versus lowest urate quintiles, was 0.43 (95% confidence intervals (CIs) 0.32–0.57). Significant association was consistently observed when we examined the association of a single urate assessment (2006 or 2010) or the rate of change in urate concentrations during 2006–2010 with pRBD (P‐trend <0.001 for all). However, restricting to pRBD onset during 2011–2012, we observed a nonsignificant trend between high urate concentration and high odds of pRBD (P‐trend = 0.09). Interpretation Higher average urate concentrations were associated with a lower likelihood of having pRBD, but not new‐onset pRBD. Because of its observational study design, the result should be interpreted with caution due to the possibility of residual confounding.
Collapse
Affiliation(s)
- Yun Shen
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junjuan Li
- Department of Nephrology, Kailuan General Hospital, Tangshan, China
| | - Michael Schwarzschild
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Milena Pavlova
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Songbin He
- Department of Neurology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Alberto Ascherio
- Department of Nutrition and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Liufu Cui
- Department of Rheumatology, Kailuan General Hospital, Tangshan, China
| | - Xiang Gao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
85
|
Modifiable risk and protective factors in disease development, progression and clinical subtypes of Parkinson's disease: What do prospective studies suggest? Neurobiol Dis 2019; 134:104671. [PMID: 31706021 DOI: 10.1016/j.nbd.2019.104671] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/23/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder whose pathogenesis depends on a combination of genetic and environmental factors. The aim of the present review was to provide an updated description of the findings emerging from prospective longitudinal cohort studies on the possible risk/protective factors underlying the development, progression and clinical subtypes of PD. We reviewed all the environmental, lifestyle, dietary, comorbid and pharmacological factors that have been investigated as possible modifiable protective/risk factors for PD by longitudinal studies. Only a few factors have the epidemiological evidence and the biological plausibility to be considered risk (pesticides, dairy products, β2-adrenoreceptor antagonists) or protective (smoking, caffeine and tea intake, physical activity, gout, vitamin E intake, non-steroidal anti-inflammatory drugs and β2-adrenoreceptor agonists) factors for PD. Caffeine intake and physical activity also seem to slow down the progression of the disease, thus representing good candidates for primary prevention and disease modifying strategies in PD. Possible modifiable risk factors of PD subtypes is almost unknown and this might depend on the uncertain biological and neuropathological reliability of clinical subtypes. The results of the present review suggest that only eleven risk/protective factors may be associated with the risk of PD. It may be possible to target some of these factors for preventive interventions aimed at reducing the risk of developing and the rate of progression of PD.
Collapse
|
86
|
Lawton M, Baig F, Toulson G, Morovat A, Evetts SG, Ben-Shlomo Y, Hu MT. Blood biomarkers with Parkinson's disease clusters and prognosis: The oxford discovery cohort. Mov Disord 2019; 35:279-287. [PMID: 31693246 PMCID: PMC7028059 DOI: 10.1002/mds.27888] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 09/03/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022] Open
Abstract
Background Predicting prognosis in Parkinson's disease (PD) has important implications for individual prognostication and clinical trials design and targeting novel treatments. Blood biomarkers could help in this endeavor. Methods We identified 4 blood biomarkers that might predict prognosis: apolipoprotein A1, C‐reactive protein, uric acid and vitamin D. These biomarkers were measured in baseline serum from 624 Parkinson's disease subjects (median disease duration, 1.0 years; interquartile range, 0.5–2.0) from the Oxford Discovery prospective cohort. We compared these biomarkers against PD subtypes derived from clinical features in the baseline cohort using data‐driven approaches. We used multilevel models with MDS‐UPDRS parts I, II, and III and Montreal Cognitive Assessment as outcomes to test whether the biomarkers predicted subsequent progression in motor and nonmotor domains. We compared the biomarkers against age of PD onset and age at diagnosis. The q value, a false‐discovery rate alternative to P values, was calculated as an adjustment for multiple comparisons. Results Apolipoprotein A1 and C‐reactive protein levels differed across our PD subtypes, with severe motor disease phenotype, poor psychological well‐being, and poor sleep subtype having reduced apolipoprotein A1 and higher C‐reactive protein levels. Reduced apolipoprotein A1, higher C‐reactive protein, and reduced vitamin D were associated with worse baseline activities of daily living (MDS‐UPDRS II). Conclusion Baseline clinical subtyping identified a pro‐inflammatory biomarker profile significantly associated with a severe motor/nonmotor disease phenotype, lending biological validity to subtyping approaches. No blood biomarker predicted motor or nonmotor prognosis. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Michael Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Fahd Baig
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Greg Toulson
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alireza Morovat
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Samuel G Evetts
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michele T Hu
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK.,Department of Clinical Neurology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
87
|
Mandal AK, Mount DB. Interaction Between ITM2B and GLUT9 Links Urate Transport to Neurodegenerative Disorders. Front Physiol 2019; 10:1323. [PMID: 31695625 PMCID: PMC6818471 DOI: 10.3389/fphys.2019.01323] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/03/2019] [Indexed: 11/13/2022] Open
Abstract
Hyperuricemia plays a critical causative role in gout. In contrast, hyperuricemia has a protective effect in neurodegenerative disorders, including Alzheimer's Disease. Genetic variation in the SLC2A9 gene, encoding the urate transporter GLUT9, exerts the largest single-gene effect on serum uric acid (SUA). We report here the identification of two GLUT9-interacting proteins, integral membrane protein 2B (ITM2B) and transmembrane protein 85 (TMEM85), isolated from a human kidney cDNA library using the dual-membrane yeast two-hybrid system. ITM2B is a ubiquitously expressed, N-glycosylated transmembrane regulatory protein, involved in familial dementias and retinal dystrophy; the function of TMEM85 is less defined. Using coimmunoprecipitation, we confirmed the physical interaction between ITM2B or TMEM85 and N-terminal GLUT9 isoforms (GLUT9a and GLUT9b) in transfected HEK 293T cells and Xenopus oocytes, wherein ITM2B but not TMEM85 inhibited GLUT9-mediated urate uptake. Additionally, co-expression of ITM2B with GLUT9 in oocytes inhibited N-glycosylation of GLUT9a more than GLUT9b and stimulated urate efflux by both isoforms. However, urate uptake by N-glycosylation and N-terminal deletion GLUT9 mutants was efficiently inhibited by ITM2B, indicating that neither N-glycosylation nor the N terminus is necessary for functional interaction of GLUT9 with ITM2B. Notably, ITM2B variants linked to familial Danish dementia and retinal dystrophy significantly attenuated the inhibition of GLUT9-mediated urate influx. We propose ITM2B as a potential regulatory link between urate homeostasis and neurodegenerative disorders.
Collapse
Affiliation(s)
- Asim K. Mandal
- Renal Divisions, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - David B. Mount
- Renal Divisions, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| |
Collapse
|
88
|
Nag N, Jelinek GA. A Narrative Review of Lifestyle Factors Associated with Parkinson's Disease Risk and Progression. NEURODEGENER DIS 2019; 19:51-59. [PMID: 31487721 DOI: 10.1159/000502292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/23/2019] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease is a complex slowly progressive neurodegenerative disorder with motor and non-motor symptoms affecting daily living. Despite effective symptomatic treatments, with various degrees of side effects, no disease-modifying therapeutic options presently exist. Symptoms progress, with an accumulating burden, reducing the quality of life and forming the impression that medications are no longer effective. Adopting positive lifestyle behaviours can empower patients, improve the quality of life, alleviate symptoms, and potentially slow disease progression. Lifestyle behaviours including nutrition, cognitive enrichment, physical activity, and stress management have beneficial effects on brain health and quality of life. While some evidence of an association of lifestyle with Parkinson's disease risk and progression exists, the sparse and often conflicting data make it difficult to provide clinical recommendations. Herein, we highlight studies showing promising associations between lifestyle and Parkinson's disease. Given the increasing aging of populations worldwide and the prevalence of neurological disorders, further research into self-management through adoption of positive lifestyle behaviours is clearly warranted to better enable individualized care.
Collapse
Affiliation(s)
- Nupur Nag
- Neuroepidemiology Unit, Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia,
| | - George A Jelinek
- Neuroepidemiology Unit, Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
89
|
Schwarzschild MA, Macklin EA, Bakshi R, Battacharyya S, Logan R, Espay AJ, Hung AY, Bwala G, Goetz CG, Russell DS, Goudreau JL, Parashos SA, Saint-Hilaire MH, Rudolph A, Hare JM, Curhan GC, Ascherio A. Sex differences by design and outcome in the Safety of Urate Elevation in PD (SURE-PD) trial. Neurology 2019; 93:e1328-e1338. [PMID: 31484712 DOI: 10.1212/wnl.0000000000008194] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To investigate whether women and men with Parkinson disease (PD) differ in their biochemical and clinical responses to long-term treatment with inosine. METHODS The Safety of Urate Elevation in Parkinson's Disease (SURE-PD) trial enrolled 75 people with early PD and baseline serum urate below 6 mg/dL and randomized them to 3 double-blinded treatment arms: oral placebo or inosine titrated to produce mild (6.1-7.0 mg/dL) or moderate (7.1-8.0 mg/dL) serum urate elevation for up to 2 years. Parkinsonism, serum urate, and plasma antioxidant capacity were measured at baseline and repeatedly on treatment; CSF urate was assessed once, at 3 months. Here in secondary analyses results are stratified by sex. RESULTS Inosine produced an absolute increase in average serum urate from baseline that was 50% greater in women (3.0 mg/dL) than in men (2.0 mg/dL), consistent with expected lower baseline levels in women. Similarly, only among women was CSF urate significantly greater on mild or moderate inosine (+87% [p < 0.001] and +98% [p < 0.001], respectively) than on placebo (in contrast to men: +10% [p = 0.6] and +14% [p = 0.4], respectively). Women in the higher inosine dosing group showed a 7.0 Unified Parkinson's Disease Rating Scale (UPDRS) points/year lower rate of decline vs placebo (p = 0.01). In women, slower rates of UPDRS change were associated with greater increases in serum urate (r = -0.52; p = 0.001), and with greater increases in plasma antioxidant capacity (r = -0.44; p = 0.006). No significant associations were observed in men. CONCLUSIONS Inosine produced greater increases in serum and CSF urate in women compared to men in the SURE-PD trial, consistent with the study's design and with preliminary evidence for slower clinical decline in early PD among women treated with urate-elevating doses of inosine. CLINICALTRIALSGOV IDENTIFIER NCT00833690. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that inosine produced greater urate elevation in women than men and may slow PD progression in women.
Collapse
Affiliation(s)
- Michael A Schwarzschild
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA.
| | - Eric A Macklin
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Rachit Bakshi
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Shamik Battacharyya
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Robert Logan
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Alberto J Espay
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Albert Y Hung
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Grace Bwala
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Christopher G Goetz
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - David S Russell
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - John L Goudreau
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Sotirios A Parashos
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Marie H Saint-Hilaire
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Alice Rudolph
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Joshua M Hare
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Gary C Curhan
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | - Alberto Ascherio
- From the Departments of Neurology (M.A.S., R.B., S.B., R.L., A.Y.H., G.B.) and Medicine (E.A.M.), Massachusetts General Hospital, Boston; University of Cincinnati (A.J.E.), OH; Rush University (C.G.G.), Chicago, IL; Michigan State University (J.L.G.), East Lansing; Struthers Parkinson's Center (S.A.P.), Minneapolis, MN; Boston University (M.H.S.-H.), MA; University of Rochester (A.R.), NY; University of Miami (J.M.H.), FL; Brigham and Women's Hospital (G.C.C.), Boston, MA; Yale University School of Medicine (D.S.R.), New Haven, CT; and Department of Nutrition (A.A.), Harvard School of Public Health, Boston, MA
| | | |
Collapse
|
90
|
Nutritional Risk Factors, Microbiota and Parkinson's Disease: What Is the Current Evidence? Nutrients 2019; 11:nu11081896. [PMID: 31416163 PMCID: PMC6722832 DOI: 10.3390/nu11081896] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/31/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a frequent neurodegenerative disease among elderly people. Genetic and underlying environmental factors seem to be involved in the pathogenesis of PD related to degeneration of dopaminergic neurons in the striatum. In previous experimental researches oxidative stress, mitochondrial dysfunction, homocysteine, and neuroinflammation have been reported as potential mechanisms. Among environmental factors, nutrition is one of the most investigated areas as it is a potentially modifiable factor. The purpose of this review is to provide current knowledge regarding the relation between diet and PD risk. We performed a comprehensive review including the most relevant studies from the year 2000 onwards including prospective studies, nested case-control studies, and meta-analysis. Among dietary factors we focused on specific nutrients and food groups, alcoholic beverages, uric acid, and dietary patterns. Furthermore, we included studies on microbiota as recent findings have shown a possible impact on neurodegeneration. As a conclusion, there are still many controversies regarding the relationship between PD and diet which, beside methodological differences among studies, may be due to underlying genetic and gender-specific factors. However, some evidence exists regarding a potential protective effect of uric acid, poly-unsaturated fatty acids, coffee, and tea but mainly in men, whereas dairy products, particularly milk, might increase PD risk through contaminant mediated effect.
Collapse
|
91
|
Feeding-produced subchronic high plasma levels of uric acid improve behavioral dysfunction in 6-hydroxydopamine-induced mouse model of Parkinson's disease. Behav Pharmacol 2019; 30:89-94. [PMID: 29847340 DOI: 10.1097/fbp.0000000000000413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The development of Parkinson's disease (PD) involves the degeneration of dopaminergic neurons caused by oxidative stress. Accumulating clinical evidence indicates that high blood levels of uric acid (UA), an intrinsic antioxidative substance, are associated with reduced risk of PD. However, this hypothesis has not been confirmed by in-vivo experiments. The present study investigated the effects of UA on behavioral abnormalities in the development of PD. We used unilateral 6-hydroxydopamine-lesioned mice, which were fed on a diet containing 1% UA and 2.5% potassium oxonate (an uricase inhibitor) to induce hyperuricemia. A significant elevation in UA levels was found in groups that were fed a UA diet. The 6-hydroxydopamine-lesioned mice showed impaired rotarod performance and increased apomorphine-induced contralateral rotations. These behavioral abnormalities were significantly reversed by feeding a UA diet for 1 week before and 5 weeks after surgery (subchronic hyperuricemia). These behavioral improvements occurred in parallel with recovery of tyrosine hydroxylase protein levels in the lesioned striatal side. The present study with a dietary hyperuricemia mice model confirms that UA exerts a neuroprotective effect on dopaminergic neuronal loss, improving motor dysfunction and ameliorating PD development.
Collapse
|
92
|
|
93
|
Dietiker C, Kim S, Zhang Y, Christine CW. Characterization of Vitamin B12 Supplementation and Correlation with Clinical Outcomes in a Large Longitudinal Study of Early Parkinson's Disease. J Mov Disord 2019; 12:91-96. [PMID: 31158942 PMCID: PMC6547038 DOI: 10.14802/jmd.18049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/21/2019] [Indexed: 01/01/2023] Open
Abstract
Objective In Parkinson’s disease (PD), vitamin B12 levels are lower, and comorbid B12 deficiency has been associated with the development of neuropathy and early gait instability. Because little is known about B12 supplement use in PD, we sought to evaluate its use in a large PD cohort and, as an exploratory analysis, to determine whether baseline characteristics or disease progression differed according to B12 supplementation. Methods We utilized data collected as part of the National Institutes of Health Exploratory Trials in PD (NET-PD) Long-term Study (LS-1), a longitudinal study of 1,741 participants. We stratified subjects into 4 groups according to daily supplement use: no B12, multivitamin (MVI) containing < 100 μg B12, B12 ≥ 100 μg, and MVI + B12 ≥ 100 μg. Clinical outcomes were assessed at 3 years for each group using the Unified Parkinson’s Disease Rating Scale (UPDRS), its subscores, and selected individual questions. Results Of the 1,147 participants who completed the 3-year visit, 41% took an MVI, 2% took B12, 3% took MVI + B12, and 54% reported taking no supplements. At 3 years, no significant differences in clinical outcomes were observed. However, there was a trend toward lower hazard ratios for developing sensory symptoms (UPDRS Item 17) in the MVI (p = 0.08) and B12 + MVI (p = 0.08) groups compared to that in the no supplement group. Conclusion These results show that supplementation with vitamin B12 ≥ 100 μg is uncommon in early PD. The finding of a trend toward a lower hazard ratio for the development of sensory symptoms in those taking an MVI or B12 + MVI warrants further study.
Collapse
Affiliation(s)
- Cameron Dietiker
- Department of Neurology, Movement Disorder and Neuromodulation Center, University of California San Francisco, San Francisco, CA, USA
| | - Soeun Kim
- Department of Biostatistics and Data Science, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Yunxi Zhang
- Department of Biostatistics and Data Science, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Chadwick W Christine
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
94
|
Guttuso T, Andrzejewski KL, Lichter DG, Andersen JK. Targeting kinases in Parkinson's disease: A mechanism shared by LRRK2, neurotrophins, exenatide, urate, nilotinib and lithium. J Neurol Sci 2019; 402:121-130. [PMID: 31129265 DOI: 10.1016/j.jns.2019.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022]
Abstract
Several kinases have been implicated in the pathogenesis of Parkinson's disease (PD), most notably leucine-rich repeat kinase 2 (LRRK2), as LRRK2 mutations are the most common genetic cause of a late-onset parkinsonism that is clinically indistinguishable from sporadic PD. More recently, several other kinases have emerged as promising disease-modifying targets in PD based on both preclinical studies and clinical reports on exenatide, the urate precursor inosine, nilotinib and lithium use in PD patients. These kinases include protein kinase B (Akt), glycogen synthase kinases-3β and -3α (GSK-3β and GSK-3α), c-Abelson kinase (c-Abl) and cyclin-dependent kinase 5 (cdk5). Activities of each of these kinases are involved either directly or indirectly in phosphorylating tau or increasing α-synuclein levels, intracellular proteins whose toxic oligomeric forms are strongly implicated in the pathogenesis of PD. GSK-3β, GSK-3α and cdk5 are the principle kinases involved in phosphorylating tau at sites critical for the formation of tau oligomers. Exenatide analogues, urate, nilotinib and lithium have been shown to affect one or more of the above kinases, actions that can decrease the formation and increase the clearance of intraneuronal phosphorylated tau and α-synuclein. Here we review the current preclinical and clinical evidence supporting kinase-targeting agents as potential disease-modifying therapies for PD patients enriched with these therapeutic targets and incorporate LRRK2 physiology into this novel model.
Collapse
Affiliation(s)
- Thomas Guttuso
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - Kelly L Andrzejewski
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - David G Lichter
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - Julie K Andersen
- The Buck Institute for Research on Aging, Novato, CA, United States of America.
| |
Collapse
|
95
|
Perez-Gomez MV, Bartsch LA, Castillo-Rodriguez E, Fernandez-Prado R, Kanbay M, Ortiz A. Potential Dangers of Serum Urate-Lowering Therapy. Am J Med 2019; 132:457-467. [PMID: 30611833 DOI: 10.1016/j.amjmed.2018.12.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022]
Abstract
In observational studies, high serum urate levels are associated with adverse outcomes, including mortality. However, the hypothesis that urate-lowering may improve nongout outcomes has not been confirmed by placebo-controlled clinical trials. On the contrary, 7 recent placebo-controlled trials of urate-lowering drugs with different mechanisms of action (uricosuric: lesinurad; xanthine oxidase inhibition: febuxostat; uricase: pegloticase) have observed higher mortality or trends to higher mortality in gout patients, with the largest decreases in serum urate. Because all urate-lowering mechanisms were implicated, this raises safety concerns about urate-lowering itself. Far from unexpected, the higher mortality associated with more intense urate-lowering is in line with the U-shaped association of urate with mortality in some observational studies. Urate accounts for most of the antioxidant capacity of plasma, and strategies to increase urate are undergoing clinical trials in neurological disease. Post hoc analysis of recent trials should explore whether the magnitude of urate-lowering is associated with adverse outcomes, and safety trials are needed before guidelines recommend lowering serum urate below certain thresholds.
Collapse
Affiliation(s)
- Maria Vanessa Perez-Gomez
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz Universidad Autónoma Madrid (UAM), Spain; Red de Investigación Renal (REDinREN), Madrid, Spain; Fundacion Renal Iñigo Alvarez de Toledo (FRIAT), Madrid, Spain
| | | | - Esmeralda Castillo-Rodriguez
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz Universidad Autónoma Madrid (UAM), Spain; Red de Investigación Renal (REDinREN), Madrid, Spain; Fundacion Renal Iñigo Alvarez de Toledo (FRIAT), Madrid, Spain
| | - Raul Fernandez-Prado
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz Universidad Autónoma Madrid (UAM), Spain; Red de Investigación Renal (REDinREN), Madrid, Spain; Fundacion Renal Iñigo Alvarez de Toledo (FRIAT), Madrid, Spain
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz Universidad Autónoma Madrid (UAM), Spain; Red de Investigación Renal (REDinREN), Madrid, Spain; Fundacion Renal Iñigo Alvarez de Toledo (FRIAT), Madrid, Spain.
| |
Collapse
|
96
|
Nybo SE, Lamberts JT. Integrated use of LC/MS/MS and LC/Q-TOF/MS targeted metabolomics with automated label-free microscopy for quantification of purine metabolites in cultured mammalian cells. Purinergic Signal 2019; 15:17-25. [PMID: 30604179 PMCID: PMC6439090 DOI: 10.1007/s11302-018-9643-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/11/2018] [Indexed: 01/27/2023] Open
Abstract
Purine metabolites have been implicated as clinically relevant biomarkers of worsening or improving Parkinson's disease (PD) progression. However, the identification of purine molecules as biomarkers in PD has largely been determined using non-targeted metabolomics analysis. The primary goal of this study was to develop an economical targeted metabolomics approach for the routine detection of purine molecules in biological samples. Specifically, this project utilized LC/MS/MS and LC/QTOF/MS to accurately quantify levels of six purine molecules in samples from cultured N2a murine neuroblastoma cells. The targeted metabolomics workflow was integrated with automated label-free digital microscopy, which enabled normalization of purine concentration per unit cell in the absence of fluorescent dyes. The established method offered significantly enhanced selectivity compared to previously published procedures. In addition, this study demonstrates that a simple, quantitative targeted metabolomics approach can be developed to identify and quantify purine metabolites in biological samples. We envision that this method could be broadly applicable to quantification of purine metabolites from other complex biological samples, such as cerebrospinal fluid or blood.
Collapse
Affiliation(s)
- S Eric Nybo
- College of Pharmacy, Department of Pharmaceutical Sciences, Ferris State University, 220 Ferris Drive, Big Rapids, MI, 49307, USA
| | - Jennifer T Lamberts
- College of Pharmacy, Department of Pharmaceutical Sciences, Ferris State University, 220 Ferris Drive, Big Rapids, MI, 49307, USA.
| |
Collapse
|
97
|
Yan LY, He QF, Lu MY, Wang SL, Qi ZQ, Dong HR. Association between carotid plaque and Parkinson's disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:94. [PMID: 31019944 DOI: 10.21037/atm.2019.01.68] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Epidemiological studies show that patients with Parkinson's disease (PD) are prone to have a reduced incidence of ischemic cerebrovascular disease. Previous studies show the correlation between PD and the lipids serum levels. The PD,s patients are found with a reduced serum level of triglyceride and low-density lipoprotein cholesterol (LDL-C); thus, the level of serum uric acid (UA) is closely related to the occurrence and development of PD. Patients with low serum UA levels have a higher chance of developing PD than the ones who do not. However, the relationship between carotid plaques and PD is still unknown. Methods Our study was based on 68 patients with PD (known as the PD group) and 81 people without PD (known as the control group). Patients in the PD group were of the same age and gender. Both groups were recorded and analyzed for UA, LDL-C, and carotid plaques or intima-media thickness (IMT). The PD group was then divided into three subgroups: the stable plaque group, the unstable plaque group, and the non-plaque group. Results In the present study, the PD group showed a significantly lower level of UA and LDL-C than the control group (P<0.01); somehow there were no statistically significant differences in the IMT and plaque incidence between the two groups (P>0.05). There were also no significant differences (P>0.05) in both the LDL-C and UA levels in all subgroups, but there was a close relation in both age and duration of disease to IMT. According to the Hoehn and Yahr staging scale, serum levels of LDL-C were inversely correlated in PD patients, while UA was related to the duration of the disease. Conclusions Our study suggested that there were no differences in carotid artery arteriosclerosis plaque and IMT, but the PD progress was indeed correlated with IMT. Meanwhile, LDL-C and UA had different priorities in H&Y and disease progression.
Collapse
Affiliation(s)
- Lan-Yun Yan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qing-Fang He
- Department of Neurology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| | - Min-Yan Lu
- Department of Neurology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| | - Sheng-Long Wang
- Department of Neurology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| | - Zhi-Qiang Qi
- Department of Neurology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| | - Hai-Rong Dong
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Department of Neurology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| |
Collapse
|
98
|
Chen X, Wei QQ, Chen Y, Cao B, Ou R, Hou Y, Yuan X, Zhang L, Liu H, Shang H. Clinical disease stage related changes of serological factors in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2019; 20:53-60. [PMID: 30784318 DOI: 10.1080/21678421.2018.1550516] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Little is known whether disease clinical stage would influence the serological values in Amyotrophic lateral sclerosis (ALS). We aimed to explore the association between the levels of serological factors with clinical progression determined by the King's College staging system. METHODS ALS Patients were registered from May 2008 to December 2016. The differences of serological values between patients and healthy controls, and the correlation of these serological values with disease stage were examined. RESULTS A total of 571 patients and 571 age-/gender-/BMI-matched healthy controls were included. The levels of creatinine, uric acid (UA), albumin, total protein, total cholesterol, and high-density lipoprotein (HDL) were significantly lower, and the low-density lipoprotein/HDL ratio was higher in ALS patients than those in healthy controls. The levels of UA, albumin, and total protein were significantly reversely correlated with diseases stages. The longitudinal observation of 81 ALS patients also showed that the levels of UA, creatinine, albumin, total protein, and HDL were significantly decreased in the second hematological examinations. CONCLUSIONS In the present study, ALS patients and control subjects were evenly matched with regard to sex, age, and BMI value, this finding could be considered as a metabolite signature in ALS. The changes of metabolite-based serological factors with progression of disease stage might be related to the pathophysiology of disease, and might have clinical utility in clinical practice.
Collapse
Affiliation(s)
- Xueping Chen
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Qian-Qian Wei
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Yongping Chen
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Bei Cao
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - RuWei Ou
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Yanbing Hou
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Xiaoqin Yuan
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Lingyu Zhang
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Hui Liu
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Huifang Shang
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| |
Collapse
|
99
|
Johnson TA, Jinnah HA, Kamatani N. Shortage of Cellular ATP as a Cause of Diseases and Strategies to Enhance ATP. Front Pharmacol 2019; 10:98. [PMID: 30837873 PMCID: PMC6390775 DOI: 10.3389/fphar.2019.00098] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/24/2019] [Indexed: 12/14/2022] Open
Abstract
Germline mutations in cellular-energy associated genes have been shown to lead to various monogenic disorders. Notably, mitochondrial disorders often impact skeletal muscle, brain, liver, heart, and kidneys, which are the body’s top energy-consuming organs. However, energy-related dysfunctions have not been widely seen as causes of common diseases, although evidence points to such a link for certain disorders. During acute energy consumption, like extreme exercise, cells increase the favorability of the adenylate kinase reaction 2-ADP -> ATP+AMP by AMP deaminase degrading AMP to IMP, which further degrades to inosine and then to purines hypoxanthine -> xanthine -> urate. Thus, increased blood urate levels may act as a barometer of extreme energy consumption. AMP deaminase deficient subjects experience some negative effects like decreased muscle power output, but also positive effects such as decreased diabetes and improved prognosis for chronic heart failure patients. That may reflect decreased energy consumption from maintaining the pool of IMP for salvage to AMP and then ATP, since de novo IMP synthesis requires burning seven ATPs. Similarly, beneficial effects have been seen in heart, skeletal muscle, or brain after treatment with allopurinol or febuxostat to inhibit xanthine oxidoreductase, which catalyzes hypoxanthine -> xanthine and xanthine -> urate reactions. Some disorders of those organs may reflect dysfunction in energy-consumption/production, and the observed beneficial effects related to reinforcement of ATP re-synthesis due to increased hypoxanthine levels in the blood and tissues. Recent clinical studies indicated that treatment with xanthine oxidoreductase inhibitors plus inosine had the strongest impact for increasing the pool of salvageable purines and leading to increased ATP levels in humans, thereby suggesting that this combination is more beneficial than a xanthine oxidoreductase inhibitor alone to treat disorders with ATP deficiency.
Collapse
Affiliation(s)
| | - H A Jinnah
- Departments of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | | |
Collapse
|
100
|
Shao Y, Le W. Recent advances and perspectives of metabolomics-based investigations in Parkinson's disease. Mol Neurodegener 2019; 14:3. [PMID: 30634989 PMCID: PMC6330496 DOI: 10.1186/s13024-018-0304-2] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/06/2018] [Indexed: 12/24/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease of the central nervous system (CNS), which affects mostly older adults. In recent years, the incidence of PD has been dramatically increasing with the aging population expanding. Due to the lack of effective biomarkers, the accurate diagnosis and precise treatment of PD are currently compromised. Notably, metabolites have been considered as the most direct reflection of the physiological and pathological conditions in individuals and represent attractive candidates to provide deep insights into disease phenotypes. By profiling the metabolites in biofluids (cerebrospinal fluid, blood, urine), feces and brain tissues, metabolomics has become a powerful and promising tool to identify novel biomarkers and provide valuable insights into the etiopathogenesis of neurological diseases. In this review, we will summarize the recent advancements of major analytical platforms implemented in metabolomics studies, dedicated to the improvement and extension of metabolome coverage for in-depth biological research. Based on the current metabolomics studies in both clinical populations and experimental PD models, this review will present new findings in metabolomics biomarkers research and abnormal metabolic pathways in PD, and will discuss the correlation between metabolomic changes and clinical conditions of PD. A better understanding of the biological underpinning of PD pathogenesis might offer novel diagnostic, prognostic, and therapeutic approaches to this devastating disease.
Collapse
Affiliation(s)
- Yaping Shao
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|