201
|
De Luca MA, Cauli O, Morelli M, Simola N. Elevation of striatal urate in experimental models of Parkinson's disease: a compensatory mechanism triggered by dopaminergic nigrostriatal degeneration? J Neurochem 2014; 131:284-9. [PMID: 24989117 DOI: 10.1111/jnc.12809] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/26/2014] [Accepted: 07/01/2014] [Indexed: 02/04/2023]
Abstract
Epidemiological studies have indicated an inverse association between high uricemia and incidence of Parkinson's disease (PD). To investigate the link between endogenous urate and neurotoxic changes involving the dopaminergic nigrostriatal system, this study evaluated the modifications in the striatal urate levels in two models of PD. To this end, a partial dopaminergic degeneration was induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mice, while a severe dopaminergic degeneration was elicited by unilateral medial forebrain bundle infusion of 6-hydroxydopamine (6-OHDA) in rats. Urate levels were measured by in vivo microdialysis at 7 or 14 days from toxin exposure. The results obtained demonstrated higher urate levels in the dopamine-denervated striatum of 6-OHDA-lesioned rats compared with the intact striatum. Moreover, an inverse correlation between urate and dopamine levels was observed in the same area. In contrast, only a trend to significant increase in striatal urate was observed in MPTP-treated mice. These results demonstrate that a damage to the dopaminergic nigrostriatal system elevates the striatal levels of urate, and suggest that this could be an endogenous compensatory mechanism to attenuate dopaminergic neurodegeneration. This finding may be important in light of the epidemiological and preclinical evidences that indicate a link between urate and development of PD.
Collapse
Affiliation(s)
- Maria Antonietta De Luca
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| | | | | | | |
Collapse
|
202
|
Moccia M, Picillo M, Erro R, Vitale C, Longo K, Amboni M, Santangelo G, Spina E, De Rosa A, De Michele G, Santoro L, Barone P, Pellecchia MT. Is serum uric acid related to non-motor symptoms in de-novo Parkinson's disease patients? Parkinsonism Relat Disord 2014; 20:772-5. [DOI: 10.1016/j.parkreldis.2014.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 02/26/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
|
203
|
Metabolomics of Human Brain Aging and Age-Related Neurodegenerative Diseases. J Neuropathol Exp Neurol 2014; 73:640-57. [DOI: 10.1097/nen.0000000000000091] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
204
|
González-Aramburu I, Sánchez-Juan P, Sierra M, Fernández-Juan E, Sánchez-Quintana C, Berciano J, Combarros O, Infante J. Serum uric acid and risk of dementia in Parkinson's disease. Parkinsonism Relat Disord 2014; 20:637-9. [DOI: 10.1016/j.parkreldis.2014.02.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/09/2014] [Accepted: 02/24/2014] [Indexed: 10/25/2022]
|
205
|
Cipriani S, Bakshi R, Schwarzschild MA. Protection by inosine in a cellular model of Parkinson's disease. Neuroscience 2014; 274:242-9. [PMID: 24880154 DOI: 10.1016/j.neuroscience.2014.05.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 05/14/2014] [Accepted: 05/16/2014] [Indexed: 02/06/2023]
Abstract
Inosine (hypoxanthine 9-beta-D-ribofuranoside), a purine nucleoside with multiple intracellular roles, also serves as an extracellular modulatory signal. On neurons, it can produce anti-inflammatory and trophic effects that confer protection against toxic influences in vivo and in vitro. The protective effects of inosine treatment might also be mediated by its metabolite urate. Urate in fact possesses potent antioxidant properties and has been reported to be protective in preclinical Parkinson's disease (PD) studies and to be an inverse risk factor for both the development and progression of PD. In this study we assessed whether inosine might protect rodent MES 23.5 dopaminergic cell line from oxidative stress in a cellular model of PD, and whether its effects could be attributed to urate. MES 23.5 cells cultured alone or in presence of enriched murine astroglial cultures MES 23.5-astrocytes co-cultures were pretreated with inosine (0.1-100 μM) for 24 h before addition of the oxidative stress inducer H₂O₂ (200 μM). Twenty-four hours later, cell viability was quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay or immunocytochemistry in pure and MES 23.5-astrocytes co-cultures, respectively. H₂O₂-toxic effect on dopaminergic cells was reduced when they were cultured with astrocytes, but not when they were cultured alone. Moreover, in MES 23.5-astrocytes co-cultures, indicators of free radical generation and oxidative damage, evaluated by nitrite (NO₂(-)) release and protein carbonyl content, respectively, were attenuated. Conditioned medium experiments indicated that the protective effect of inosine relies on the release of a protective factor from inosine-stimulated astrocytes. Purine levels were measured in the cellular extract and conditioned medium using high-performance liquid chromatography (HPLC) method. Urate concentration was not significantly increased by inosine treatment however there was a significant increase in levels of other purine metabolites, such as adenosine, hypoxanthine and xanthine. In particular, in MES 23.5-astrocytes co-cultures, inosine medium content was reduced by 99% and hypoxanthine increased by 127-fold. Taken together these data raise the possibility that inosine might have a protective effect in PD that is independent of any effects mediated through its metabolite urate.
Collapse
Affiliation(s)
- S Cipriani
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, 114 16th street, Boston, MA 02129, USA.
| | - R Bakshi
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, 114 16th street, Boston, MA 02129, USA
| | - M A Schwarzschild
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, 114 16th street, Boston, MA 02129, USA
| |
Collapse
|
206
|
Schwarzschild MA, Ascherio A, Beal MF, Cudkowicz ME, Curhan GC, Hare JM, Hooper DC, Kieburtz KD, Macklin EA, Oakes D, Rudolph A, Shoulson I, Tennis MK, Espay AJ, Gartner M, Hung A, Bwala G, Lenehan R, Encarnacion E, Ainslie M, Castillo R, Togasaki D, Barles G, Friedman JH, Niles L, Carter JH, Murray M, Goetz CG, Jaglin J, Ahmed A, Russell DS, Cotto C, Goudreau JL, Russell D, Parashos SA, Ede P, Saint-Hilaire MH, Thomas CA, James R, Stacy MA, Johnson J, Gauger L, Antonelle de Marcaida J, Thurlow S, Isaacson SH, Carvajal L, Rao J, Cook M, Hope-Porche C, McClurg L, Grasso DL, Logan R, Orme C, Ross T, Brocht AFD, Constantinescu R, Sharma S, Venuto C, Weber J, Eaton K. Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol 2014; 71:141-50. [PMID: 24366103 DOI: 10.1001/jamaneurol.2013.5528] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IMPORTANCE Convergent biological, epidemiological, and clinical data identified urate elevation as a candidate strategy for slowing disability progression in Parkinson disease (PD). OBJECTIVE To determine the safety, tolerability, and urate-elevating capability of the urate precursor inosine in early PD and to assess its suitability and potential design features for a disease-modification trial. DESIGN, SETTING, AND PARTICIPANTS The Safety of Urate Elevation in PD (SURE-PD) study, a randomized, double-blind, placebo-controlled, dose-ranging trial of inosine, enrolled participants from 2009 to 2011 and followed them for up to 25 months at outpatient visits to 17 credentialed clinical study sites of the Parkinson Study Group across the United States. Seventy-five consenting adults (mean age, 62 years; 55% women) with early PD not yet requiring symptomatic treatment and a serum urate concentration less than 6 mg/dL (the approximate population median) were enrolled. INTERVENTIONS Participants were randomized to 1 of 3 treatment arms: placebo or inosine titrated to produce mild (6.1-7.0 mg/dL) or moderate (7.1-8.0 mg/dL) serum urate elevation using 500-mg capsules taken orally up to 2 capsules 3 times per day. They were followed for up to 24 months (median, 18 months) while receiving the study drug plus 1 washout month. MAIN OUTCOMES AND MEASURES The prespecified primary outcomes were absence of unacceptable serious adverse events (safety), continued treatment without adverse event requiring dose reduction (tolerability), and elevation of urate assessed serially in serum and once (at 3 months) in cerebrospinal fluid. RESULTS Serious adverse events (17), including infrequent cardiovascular events, occurred at the same or lower rates in the inosine groups relative to placebo. No participant developed gout and 3 receiving inosine developed symptomatic urolithiasis. Treatment was tolerated by 95% of participants at 6 months, and no participant withdrew because of an adverse event. Serum urate rose by 2.3 and 3.0 mg/dL in the 2 inosine groups (P < .001 for each) vs placebo, and cerebrospinal fluid urate level was greater in both inosine groups (P = .006 and <.001, respectively). Secondary analyses demonstrated nonfutility of inosine treatment for slowing disability. CONCLUSIONS AND RELEVANCE Inosine was generally safe, tolerable, and effective in raising serum and cerebrospinal fluid urate levels in early PD. The findings support advancing to more definitive development of inosine as a potential disease-modifying therapy for PD. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00833690.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - David Oakes
- University of Rochester, Rochester, New York
| | | | | | | | | | | | - Albert Hung
- Massachusetts General Hospital, Boston, Massachusetts
| | - Grace Bwala
- Massachusetts General Hospital, Boston, Massachusetts
| | - Richard Lenehan
- Scott & White Memorial Hospital/Texas A&M University, Temple
| | | | - Melissa Ainslie
- Scott & White Memorial Hospital/Texas A&M University, Temple
| | | | | | - Gina Barles
- University of Southern California, Los Angeles
| | | | - Lisa Niles
- Butler Hospital, Providence, Rhode Island
| | | | | | | | - Jeana Jaglin
- Rush University Medical Center, Chicago, Illinois
| | | | - David S Russell
- Institute of Neurodegenerative Disorders, New Haven, Connecticut
| | - Candace Cotto
- Institute of Neurodegenerative Disorders, New Haven, Connecticut
| | | | | | | | - Patricia Ede
- Struthers Parkinson's Center, Golden Valley, Minnesota
| | | | | | | | | | | | | | | | | | - Stuart H Isaacson
- Parkinson's Disease & Movement Disorder Center of Boca Raton, Boca Raton, Florida
| | - Lisbeth Carvajal
- Parkinson's Disease & Movement Disorder Center of Boca Raton, Boca Raton, Florida
| | | | - Maureen Cook
- Ochsner Clinic Foundation, New Orleans, Louisiana
| | | | - Lauren McClurg
- Administrative Coordination Center, Massachusetts General Hospital, Boston
| | - Daniela L Grasso
- Administrative Coordination Center, Massachusetts General Hospital, Boston
| | - Robert Logan
- Administrative Coordination Center, Massachusetts General Hospital, Boston
| | - Constance Orme
- Clinical Coordination Center, University of Rochester, Rochester, New York
| | - Tori Ross
- Clinical Coordination Center, University of Rochester, Rochester, New York
| | - Alicia F D Brocht
- Clinical Coordination Center, University of Rochester, Rochester, New York
| | | | - Saloni Sharma
- Clinical Coordination Center, University of Rochester, Rochester, New York
| | - Charles Venuto
- Clinical Coordination Center, University of Rochester, Rochester, New York
| | - Joseph Weber
- Clinical Coordination Center, University of Rochester, Rochester, New York
| | - Ken Eaton
- Clinical Coordination Center, University of Rochester, Rochester, New York
| |
Collapse
|
207
|
Camilleri A, Vassallo N. The centrality of mitochondria in the pathogenesis and treatment of Parkinson's disease. CNS Neurosci Ther 2014; 20:591-602. [PMID: 24703487 DOI: 10.1111/cns.12264] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/07/2014] [Accepted: 03/08/2014] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is an incurable neurodegenerative disorder leading to progressive motor impairment and for which there is no cure. From the first postmortem account describing a lack of mitochondrial complex I in the substantia nigra of PD sufferers, the direct association between mitochondrial dysfunction and death of dopaminergic neurons has ever since been consistently corroborated. In this review, we outline common pathways shared by both sporadic and familial PD that remarkably and consistently converge at the level of mitochondrial integrity. Furthermore, such knowledge has incontrovertibly established mitochondria as a valid therapeutic target in neurodegeneration. We discuss several mitochondria-directed therapies that promote the preservation, rescue, or restoration of dopaminergic neurons and which have been identified in the laboratory and in preclinical studies. Some of these have progressed to clinical trials, albeit the identification of an unequivocal disease-modifying neurotherapeutic is still elusive. The challenge is therefore to improve further, not least by more research on the molecular mechanisms and pathophysiological consequences of mitochondrial dysfunction in PD.
Collapse
Affiliation(s)
- Angelique Camilleri
- Department of Physiology and Biochemistry, University of Malta, Msida 2080, Malta
| | | |
Collapse
|
208
|
Kachroo A, Schwarzschild MA. Allopurinol reduces levels of urate and dopamine but not dopaminergic neurons in a dual pesticide model of Parkinson's disease. Brain Res 2014; 1563:103-9. [PMID: 24680743 DOI: 10.1016/j.brainres.2014.03.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 03/03/2014] [Accepted: 03/19/2014] [Indexed: 01/21/2023]
Abstract
Robust epidemiological data link higher levels of the antioxidant urate to a reduced risk of developing Parkinson׳s disease (PD) and to a slower rate of its progression. Allopurinol, an inhibitor of xanthine oxidoreductase (XOR), blocks the oxidation of xanthine to urate. The present study sought to determine whether lowering levels of urate using allopurinol results in exacerbated neurotoxicity in a dual pesticide mouse model of PD. Although oral allopurinol reduced serum and striatal urate levels 4-fold and 1.3-fold, respectively, it did not alter the multiple motor deficits induced by chronic (7 week) intermittent (biweekly) exposure to intraperitoneal Paraquat (PQ) plus Maneb (MB). However, striatal dopamine content, which was unaffected after either allopurinol or chronic pesticide exposure alone, was significantly reduced by 22% in mice exposed to the combination. Stereological assessment showed that the numbers of dopaminergic nigral neurons were significantly reduced by 29% and the tyrosine hydroxylase (TH) negative neurons unaffected after PQ+MB treatments. This reduction in TH-positive neurons was not affected by allopurinol treatment. Of note, despite the expectation of exacerbated oxidative damage due to the reduction in urate, protein carbonyl levels, a marker of oxidative damage, were actually reduced in the presence of allopurinol. Overall, allopurinol lowered urate levels but did not exacerbate dopaminergic neuron degeneration, findings suggesting that basal levels of urate in mice do not appreciably protect against oxidative damage and neurotoxicity in the PQ+MB model of PD, and/or that allopurinol produces an antioxidant benefit offsetting its detrimental urate-lowering effect.
Collapse
Affiliation(s)
- Anil Kachroo
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA.
| | - Michael A Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
209
|
Mellick GD, Silburn PA, Sutherland GT, Siebert GA. Exploiting the potential of molecular profiling in Parkinson’s disease: current practice and future probabilities. Expert Rev Mol Diagn 2014; 10:1035-50. [PMID: 21080820 DOI: 10.1586/erm.10.86] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- George D Mellick
- Eskitis Institute for Cell & Molecular Therapies, School of Biomolecular & Physical Sciences, Griffith University, Brisbane, QLD 4111, Australia.
| | | | | | | |
Collapse
|
210
|
Yasuda D, Takahashi K, Ohe T, Nakamura S, Mashino T. Antioxidant activities of 5-hydroxyoxindole and its 3-hydroxy-3-phenacyl derivatives: The suppression of lipid peroxidation and intracellular oxidative stress. Bioorg Med Chem 2013; 21:7709-14. [DOI: 10.1016/j.bmc.2013.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
|
211
|
Saracchi E, Fermi S, Brighina L. Emerging candidate biomarkers for Parkinson's disease: a review. Aging Dis 2013; 5:27-34. [PMID: 24490114 DOI: 10.14366/ad.2014.050027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/06/2013] [Accepted: 10/07/2013] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease is a chronic neurodegenerative disorder leading to progressive motor impairment affecting more than 1% of the over-65 population. In spite of considerable progress in identifying the genetic and biochemical basis of PD, to date the diagnosis remains clinical and disease-modifying therapies continue to be elusive. A cornerstone in recent PD research is the investigation of biological markers that could help in identifying at-risk population or to track disease progression and response to therapies. Although none of these parameters has been validated for routine clinical practice yet, however some biochemical candidates hold great promise for application in PD patients, especially in the early stages of disease, and it is likely that in the future the diagnosis of PD will require a combination of genetic, imaging and laboratory data. In this review we discuss the most interesting biochemical markers for PD (including the "-omics" techniques), focusing on the methodological challenges in using ex vivo blood/CSF/tissue-based biomarkers and suggesting alternative strategies to overcome the difficulties that still prevent their actual use.
Collapse
Affiliation(s)
- Enrico Saracchi
- Department of Neurology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Silvia Fermi
- Neurology Department, Azienda Ospedaliera di Lodi, 26900 Lodi, Italy
| | - Laura Brighina
- Department of Neurology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
212
|
Advanced Parkinson's disease: Clinical characteristics and treatment (part 1). NEUROLOGÍA (ENGLISH EDITION) 2013. [DOI: 10.1016/j.nrleng.2013.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
213
|
Dexter DT, Jenner P. Parkinson disease: from pathology to molecular disease mechanisms. Free Radic Biol Med 2013; 62:132-144. [PMID: 23380027 DOI: 10.1016/j.freeradbiomed.2013.01.018] [Citation(s) in RCA: 485] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
Parkinson disease (PD) is a complex neurodegenerative disorder with both motor and nonmotor symptoms owing to a spreading process of neuronal loss in the brain. At present, only symptomatic treatment exists and nothing can be done to halt the degenerative process, as its cause remains unclear. Risk factors such as aging, genetic susceptibility, and environmental factors all play a role in the onset of the pathogenic process but how these interlink to cause neuronal loss is not known. There have been major advances in the understanding of mechanisms that contribute to nigral dopaminergic cell death, including mitochondrial dysfunction, oxidative stress, altered protein handling, and inflammation. However, it is not known if the same processes are responsible for neuronal loss in nondopaminergic brain regions. Many of the known mechanisms of cell death are mirrored in toxin-based models of PD, but neuronal loss is rapid and not progressive and limited to dopaminergic cells, and drugs that protect against toxin-induced cell death have not translated into neuroprotective therapies in humans. Gene mutations identified in rare familial forms of PD encode proteins whose functions overlap widely with the known molecular pathways in sporadic disease and these have again expanded our knowledge of the neurodegenerative process but again have so far failed to yield effective models of sporadic disease when translated into animals. We seem to be missing some key parts of the jigsaw, the trigger event starting many years earlier in the disease process, and what we are looking at now is merely part of a downstream process that is the end stage of neuronal death.
Collapse
Affiliation(s)
- David T Dexter
- Parkinson's Disease Research Group, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London SE1 9NH, UK.
| |
Collapse
|
214
|
Tanner CM. Alpha-synuclein: one key opens many locks. Mov Disord 2013; 28:1176-8. [PMID: 23925937 DOI: 10.1002/mds.25596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 11/08/2022] Open
|
215
|
Abstract
PURPOSE OF REVIEW Gout is a painful inflammatory arthritis associated with hyperuricemia, with a prevalence of almost 10 million in the USA. Reduced renal excretion of urate is the underlying hyperuricemic mechanism in the vast majority of gout patients; most of the genes that affect serum urate level (SUA) encode urate transporters or associated regulatory proteins. Acquired influences can also modulate SUA and renal urate excretion, sometimes precipitating acute gout. Coincidentally, the prevalence of renal comorbidities in gout - hypertension, chronic kidney disease (CKD), and nephrolithiasis - is very high. RECENT FINDINGS Recent advances in genetics and molecular physiology have greatly enhanced the understanding of renal reabsorption and secretion of filtered urate. Moreover, baseline SUA appears to be set by the net balance of absorption and secretion across epithelial cells in the kidney and intestine. There have also been substantial advances in the management of gout in patients with CKD. SUMMARY The stage is set for an increasingly molecular understanding of baseline and regulated urate transport by the kidney and intestine. The increasing prevalence of gout with CKD will be balanced by an expanding spectrum of therapeutic options for this important disease.
Collapse
|
216
|
Kulisevsky J, Luquin MR, Arbelo JM, Burguera JA, Carrillo F, Castro A, Chacón J, García-Ruiz PJ, Lezcano E, Mir P, Martinez-Castrillo JC, Martínez-Torres I, Puente V, Sesar A, Valldeoriola-Serra F, Yañez R. [Advanced Parkinson's disease: clinical characteristics and treatment (part 1)]. Neurologia 2013; 28:503-21. [PMID: 23856182 DOI: 10.1016/j.nrl.2013.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/16/2013] [Accepted: 05/02/2013] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION A large percentage of patients with Parkinson's disease (PD) develop motor fluctuations, dyskinesias, and severe non-motor symptoms within 3 to 5 years of starting dopaminergic therapy, and these motor complications are refractory to treatment. Several authors refer to this stage of the disease as advanced Parkinson's disease. OBJECTIVE To define the clinical manifestations of advanced PD and the risk factors for reaching this stage of the disease. DEVELOPMENT This consensus document has been prepared by using an exhaustive literature search and by discussion of the contents by an expert group on movement disorders of the Sociedad Española de Neurología (Spanish Neurology Society), coordinated by two of the authors (JK and MRL). CONCLUSIONS Severe motor fluctuations and dyskinesias, axial motor symptoms resistant to levodopa, and cognitive decline are the main signs in the clinical phenotype of advanced PD.
Collapse
Affiliation(s)
- J Kulisevsky
- Servicio de Neurología, Hospital Sant Pau, IIB Sant Pau, CIBERNED, Universitat Autònoma de Barcelona, Barcelona, España.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Cao B, Guo X, Chen K, Song W, Huang R, Wei QQ, Zhao B, Shang HF. Uric acid is associated with the prevalence but not disease progression of multiple system atrophy in Chinese population. J Neurol 2013; 260:2511-5. [PMID: 23801150 DOI: 10.1007/s00415-013-7006-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 06/10/2013] [Accepted: 06/10/2013] [Indexed: 02/05/2023]
Abstract
Oxidative stress is involved in the pathogenesis of multiple system atrophy (MSA). Uric acid has an antioxidative effect. Our aim is to clarify the correlations between serum uric acid and MSA in Chinese population. A total of 234 patients with probable MSA and 240 age- and gender- matched healthy controls were included in the study. The serum uric acid levels of all the patients and controls were evaluated. The Unified MSA Rating Scale (UMSARS) was used to assess the severity and the mean rate of annualized changes of UMSARS to assess the progression of MSA. The mean age of MSA patients was 58.90 ± 9.00 years and the mean disease duration was 2.60 ± 1.75 years. The serum uric acid levels of MSA patients were significantly lower than that of controls in males (p = 0.0001). The occurrence of MSA was increased in the lowest uric acid quartiles compared with the highest uric acid quartiles (p = 0.005). In a gender-specific analysis, increased occurrence was found in the lowest quartiles and second quartiles compared with the highest quartiles in males (p = 0.001 and p = 0.0001 respectively), but not in females. No correlation was found between the mean rate of annualized changes and serum levels of uric acid, as well as other independent factors, such as age, BMI, gender, subtype (C-type or P-type) and disease duration at the initial visit in 107 followed-up patients. MSA patients have lower levels of serum uric acid than controls. High levels of serum uric acid may be associated with a lower prevalence of MSA in the Chinese population, especially in males. However, serum uric acid does not deteriorate or ameliorate the progression of MSA.
Collapse
Affiliation(s)
- Bei Cao
- Department of Neurology, West China Hospital, SiChuan University, Chengdu, 610041, Sichuan, China
| | | | | | | | | | | | | | | |
Collapse
|
218
|
Liu R, Guo X, Park Y, Wang J, Huang X, Hollenbeck A, Blair A, Chen H. Alcohol Consumption, Types of Alcohol, and Parkinson's Disease. PLoS One 2013; 8:e66452. [PMID: 23840473 PMCID: PMC3686735 DOI: 10.1371/journal.pone.0066452] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/04/2013] [Indexed: 02/04/2023] Open
Abstract
Background The epidemiologic evidence on alcohol consumption and Parkinson’s disease (PD) is equivocal. We prospectively examined total alcohol consumption and consumption of specific types of alcoholic beverage in relation to future risk of PD. Methods The study comprised 306,895 participants (180,235 male and 126,660 female) ages 50–71 years in 1995–1996 from the NIH-AARP Diet and Health Study. Consumption of alcoholic beverages in the past 12 months was assessed in 1995–1996. Multivariate odds ratios (OR) and 95% confidence intervals (CI) were obtained from logistic regression models. Results A total of 1,113 PD cases diagnosed between 2000 and 2006 were included in the analysis. Total alcohol consumption was not associated with PD. However, the association differed by types of alcoholic beverages. Compared with non-beer drinkers, the multivariate ORs for beer drinkers were 0.79 (95% CI: 0.68, 0.92) for <1 drink/day, 0.73 (95% CI: 0.50, 1.07) for 1–1.99 drinks/day, and 0.86 (95% CI: 0.60, 1.21) for ≥2 drinks/day. For liquor consumption, a monotonic increase in PD risk was suggested: ORs (95% CI) were 1.06 (0.91, 1.23), 1.22 (0.94, 1.58), and 1.35 (1.02, 1.80) for <1, 1–1.99, and ≥2 drinks/day, respectively (P for trend <0.03). Additional analyses among exclusive drinkers of one specific type of alcoholic beverage supported the robustness of these findings. The results for wine consumption were less clear, although a borderline lower PD risk was observed when comparing wine drinkers of 1–1.99 drinks/day with none drinkers (OR = 0.74, 95% CI: 0.53, 1.02). Conclusions Our results suggest that beer and liquor consumption may have opposite associations with PD: low to moderate beer consumption with lower PD risk and greater liquor consumption with higher risk. These findings and potential underlying mechanisms warrant further investigations.
Collapse
Affiliation(s)
- Rui Liu
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| | - Xuguang Guo
- Westat Inc., Research Triangle Park, North Carolina, United States of America
| | - Yikyung Park
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Jian Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Xuemei Huang
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | | | - Aaron Blair
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Honglei Chen
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
219
|
Yao JK, Dougherty GG, Reddy RD, Matson WR, Kaddurah-Daouk R, Keshavan MS. Associations between purine metabolites and monoamine neurotransmitters in first-episode psychosis. Front Cell Neurosci 2013; 7:90. [PMID: 23781173 PMCID: PMC3678099 DOI: 10.3389/fncel.2013.00090] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/22/2013] [Indexed: 12/31/2022] Open
Abstract
Schizophrenia (SZ) is a biochemically complex disorder characterized by widespread defects in multiple metabolic pathways whose dynamic interactions, until recently, have been difficult to examine. Rather, evidence for these alterations has been collected piecemeal, limiting the potential to inform our understanding of the interactions amongst relevant biochemical pathways. We herein review perturbations in purine and neurotransmitter metabolism observed in early SZ using a metabolomic approach. Purine catabolism is an underappreciated, but important component of the homeostatic response of mitochondria to oxidant stress. We have observed a homeostatic imbalance of purine catabolism in first-episode neuroleptic-naïve patients with SZ (FENNS). Precursor and product relationships within purine pathways are tightly correlated. Although some of these correlations persist across disease or medication status, others appear to be lost among FENNS suggesting that steady formation of the antioxidant uric acid (UA) via purine catabolism is altered early in the course of illness. As is the case for within-pathway correlations, there are also significant cross-pathway correlations between respective purine and tryptophan (TRP) pathway metabolites. By contrast, purine metabolites show significant cross-pathway correlation only with tyrosine, and not with its metabolites. Furthermore, several purine metabolites (UA, guanosine, or xanthine) are each significantly correlated with 5-hydroxyindoleacetic acid (5-HIAA) in healthy controls, but not in FENNS at baseline or 4-week after antipsychotic treatment. Taken together, the above findings suggest that purine catabolism strongly associates with the TRP pathways leading to serotonin (5-hydroxytryptamine, 5-HT) and kynurenine metabolites. The lack of a significant correlation between purine metabolites and 5-HIAA, suggests alterations in key 5-HT pathways that may both be modified by and contribute to oxidative stress via purine catabolism in FENNS.
Collapse
Affiliation(s)
- Jeffrey K Yao
- Medical Research Service, VA Pittsburgh Healthcare System Pittsburgh, PA, USA ; Department of Psychiatry, University of Pittsburgh School of Medicine Pittsburgh, PA, USA ; Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
220
|
González-Aramburu I, Sánchez-Juan P, Jesús S, Gorostidi A, Fernández-Juan E, Carrillo F, Sierra M, Gómez-Garre P, Cáceres-Redondo MT, Berciano J, Ruiz-Martínez J, Combarros O, Mir P, Infante J. Genetic variability related to serum uric acid concentration and risk of Parkinson's disease. Mov Disord 2013; 28:1737-40. [PMID: 23712608 DOI: 10.1002/mds.25507] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/28/2013] [Accepted: 04/09/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Low serum uric acid (UA) levels have been associated with increased Parkinson's disease (PD) risk and accelerated disease progression. We analyzed the effect of polymorphisms in 9 genes influencing serum UA concentration on the risk of PD. METHODS We genotyped SLC2A9 rs734553, ABCG2 rs2231142, SLC17A1 rs1183201, SLC22A11 rs17300741, SLC22A12 rs505802, GCKR rs780094, PDZK1 rs12129861, LRRC16A+SCGN rs742132, and SLC16A9 rs12356193 in 1061 PD patients and 754 controls. For each subject we calculated a cumulative genetic risk score (GRS), defined as the total number of PD-risk alleles (range, 2-15) associated to lower serum UA levels. Serum UA levels were measured in a subgroup of 365 PD cases and 132 controls. RESULTS Serum UA levels were significantly lower in men with PD than in controls. Subjects (both men and women) carrying more than 9 risk alleles (third GRS tertile) had a 1.5 higher risk of developing PD than subjects with less than 8 risk alleles (first GRS tertile). An inverse correlation was observed between higher GRS and lower serum UA concentration in both men and women. CONCLUSIONS Genetic variability influencing serum UA levels might modify susceptibility to PD.
Collapse
Affiliation(s)
- Isabel González-Aramburu
- Service of Neurology, Universitary Hospital Marqués de Valdecilla (IFIMAV), University of Cantabria (UC), Santander, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Nath AK, Roberts LD, Liu Y, Mahon SB, Kim S, Ryu JH, Werdich A, Januzzi JL, Boss GR, Rockwood GA, MacRae CA, Brenner M, Gerszten RE, Peterson RT. Chemical and metabolomic screens identify novel biomarkers and antidotes for cyanide exposure. FASEB J 2013; 27:1928-38. [PMID: 23345455 PMCID: PMC3633825 DOI: 10.1096/fj.12-225037] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 01/14/2013] [Indexed: 11/11/2022]
Abstract
Exposure to cyanide causes a spectrum of cardiac, neurological, and metabolic dysfunctions that can be fatal. Improved cyanide antidotes are needed, but the ideal biological pathways to target are not known. To understand better the metabolic effects of cyanide and to discover novel cyanide antidotes, we developed a zebrafish model of cyanide exposure and scaled it for high-throughput chemical screening. In a screen of 3120 small molecules, we discovered 4 novel antidotes that block cyanide toxicity. The most potent antidote was riboflavin. Metabolomic profiling of cyanide-treated zebrafish revealed changes in bile acid and purine metabolism, most notably by an increase in inosine levels. Riboflavin normalizes many of the cyanide-induced neurological and metabolic perturbations in zebrafish. The metabolic effects of cyanide observed in zebrafish were conserved in a rabbit model of cyanide toxicity. Further, humans treated with nitroprusside, a drug that releases nitric oxide and cyanide ions, display increased circulating bile acids and inosine. In summary, riboflavin may be a novel treatment for cyanide toxicity and prophylactic measure during nitroprusside treatment, inosine may serve as a biomarker of cyanide exposure, and metabolites in the bile acid and purine metabolism pathways may shed light on the pathways critical to reversing cyanide toxicity.
Collapse
Affiliation(s)
- Anjali K. Nath
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, MA, USA
| | - Lee D. Roberts
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, MA, USA
| | - Yan Liu
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, MA, USA
| | - Sari B. Mahon
- Pulmonary and Critical Care Medicine, University of California–Irvine Medical Center, Orange, California, USA
| | - Sonia Kim
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Justine H. Ryu
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Andreas Werdich
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James L. Januzzi
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Gerry R. Boss
- Department of Medicine, University of California–San Diego, La Jolla, California, USA; and
| | - Gary A. Rockwood
- Analytical Toxicology Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Aberdeen, Maryland, USA
| | - Calum A. MacRae
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Brenner
- Pulmonary and Critical Care Medicine, University of California–Irvine Medical Center, Orange, California, USA
| | - Robert E. Gerszten
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, MA, USA
| | - Randall T. Peterson
- Cardiovascular Research Center and
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, MA, USA
| |
Collapse
|
222
|
Martín RUS, Francke PV, Illanes FL, Gazmuri AJ, Rivera JS, Ferńndez JG, Martínez JS, Juri C. Plasma urate in REM sleep behavior disorder. Mov Disord 2013; 28:1150-1. [DOI: 10.1002/mds.25441] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 11/10/2022] Open
Affiliation(s)
- Reinaldo Uribe-San Martín
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
| | | | - Felipe López Illanes
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
| | - Alex Jones Gazmuri
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
| | - Julio Salazar Rivera
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
| | - Jaime Godoy Ferńndez
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
| | - Julia Santín Martínez
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
| | - Carlos Juri
- Department of Neurology; School of Medicine. Pontificia Universidad Católica de Chile; Santiago Chile
- Department of Neurology; Hospital Sotero del Río; Santiago Chile
| |
Collapse
|
223
|
Abstract
OBJECTIVE Serum urate may exert protective effects against Parkinson's disease (PD) through its antioxidant capacities. In this article, we examine the hypothesis that high serum urate levels are associated with lower risk of PD. METHODS We searched NCBI (PubMed), ISI Web of Science and EMBASE for studies that reported the risk of PD associated with serum urate. Fixed or random effects meta-analysis was used to pool results across studies, and further analysis was used to assess the effects by gender. RESULTS Six studies met the inclusion criteria involving a total of 33 185 participants. Overall, we found a 33% reduction in PD incidence among persons with high serum urate level (relative risk [RR]=0.67; 95% confidence interval [CI], 0.50-0.91). Subgroup analysis was performed with 20 641 men and 12 544 women included, indicating statistically significant protective effects of serum urate in men (RR=0.60; 95% CI, 0.40-0.90) but not in women. A dose-response trend of serum urate to reduce PD risk was also observed involving 11 795 participants (RR=0.77; 95% CI, 0.68-0.88). Additionally, high serum urate levels seemed to slow the clinical decline of PD patients (RR=0.56; 95% CI, 0.43-0.72). CONCLUSIONS In light of these findings, our study confirms previous findings of a robust association between high serum urate level and PD risk, especially in men. It also suggests that long-term exposure to high serum urate may be linked to the delay of PD progression, however more well-designed investigations are needed.
Collapse
|
224
|
McFarland NR, Burdett T, Desjardins CA, Frosch MP, Schwarzschild MA. Postmortem brain levels of urate and precursors in Parkinson's disease and related disorders. NEURODEGENER DIS 2013; 12:189-98. [PMID: 23467193 PMCID: PMC3809155 DOI: 10.1159/000346370] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 12/09/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that urate may play an important role in neurodegenerative disease. In Parkinson's disease (PD) higher, but still normal, levels of blood and cerebrospinal fluid urate have been associated with a lower rate of disease progression. OBJECTIVE We explored the hypothesis that lower levels of urate and its purine precursors in brain may be associated with PD and related neurodegenerative disorders, including Alzheimer's disease (AD) and Lewy body dementia (DLB). METHODS Human postmortem brain tissues were obtained from PD, AD, and DLB patients and non-neurodegenerative disease controls. We measured urate and other purine pathway analytes in the frontal and temporal cortex, striatum, and cerebellum, using high-performance liquid chromatography with electrochemical and ultraviolet detection. RESULTS Age was well-matched among groups. Mean postmortem interval for samples was 16.3 ± 9.9 h. Urate levels in cortical and striatal tissue trended lower in PD and AD compared to controls in males only. These findings correlated with increased urate in male versus female control tissues. By contrast, in DLB urate levels were significantly elevated relative to PD and AD. Measurement of urate precursors suggested a decrease in xanthine in PD compared to AD in females only, and relative increases in inosine and adenosine in DLB and AD samples among males. Xanthine and hypoxanthine were more concentrated in striatal tissue than in other brain regions. CONCLUSIONS Though limited in sample size, these findings lend support to the inverse association between urate levels and PD, as well as possibly AD. The finding of increased urate in DLB brain tissue is novel and warrants further study.
Collapse
Affiliation(s)
- Nikolaus R McFarland
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida College of Medicine, Gainesville, Fla., USA
| | | | | | | | | |
Collapse
|
225
|
Gao J, Xu H, Huang X, Chen H. Short communication: genetic variations of SLC2A9 in relation to Parkinson's disease. Transl Neurodegener 2013; 2:5. [PMID: 23422251 PMCID: PMC3598344 DOI: 10.1186/2047-9158-2-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/17/2013] [Indexed: 11/19/2022] Open
Abstract
Background Epidemiological studies showed that higher plasma urate was associated with lower risk for Parkinson’s disease (PD) and slower disease progression. Recent genome-wide association studies (GWAS) consistently showed that several single nucleotide polymorphisms (SNPs) in the solute carrier family 2 member 9 gene (SLC2A9 ) were associated with plasma urate concentration and the risk of gout. Methods We conducted a case–control study to examine twelve tag SNPs of the SLC2A9 gene in relation to PD among 788 cases and 911 controls of European ancestry. Odds ratios (OR) and 95% confidence intervals (CI) were derived from logistic regression models, adjusting for age, sex, smoking and caffeine consumption. Results These SNPs were all in linkage disequilibrium (R2 > 0.7). None of them were associated with PD risk. Among women, however, there was a suggestion that the presence of the minor allele of one SNP (rs7442295) was related to a small increase in PD risk [OR (95% CI) = 1.48 (1.01-2.16)]. Conclusion This study provides little support for genetic variations of SLC2A9 and PD risk.
Collapse
Affiliation(s)
- Jianjun Gao
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| | | | | | | |
Collapse
|
226
|
Constantinescu R, Andreasson U, Holmberg B, Zetterberg H. Serum and cerebrospinal fluid urate levels in synucleinopathies versus tauopathies. Acta Neurol Scand 2013; 127:e8-12. [PMID: 22998191 DOI: 10.1111/ane.12012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND Low levels of serum urate are associated with a higher risk of Parkinson's disease (PD). Higher serum and cerebrospinal fluid (CSF) urate levels are associated with slower rates of clinical decline in PD and in multiple system atrophy (MSA). AIMS To compare CSF and blood urate levels in healthy controls, patients with synucleinopathies and with tauopathies. METHODS We investigated urate levels in serum and CSF from 18 healthy controls, 19 patients with synucleinopathies (six patients with PD and 13 with MSA), and 24 patients with tauopathies (18 with progressive supranuclear palsy and six with corticobasal degeneration). None of the patients were treated with dopaminergic medications. RESULTS No significant differences were seen when comparing serum and CSF urate levels from controls across the parkinsonian diagnostic groups. However, in men, serum urate levels were significantly lower in the synucleinopathy group compared with the tauopathy group (P = 0.046), although with a broad overlap. CONCLUSION Our study suggests that urate levels might provide new insights into the potential pathophysiological mechanisms underlying Parkinsonism and thereby contribute to the future management of these disorders.
Collapse
Affiliation(s)
- R. Constantinescu
- Department of Neurology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - U. Andreasson
- Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - B. Holmberg
- Department of Neurology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - H. Zetterberg
- Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| |
Collapse
|
227
|
Constantinescu R, Mondello S. Cerebrospinal fluid biomarker candidates for parkinsonian disorders. Front Neurol 2013; 3:187. [PMID: 23346074 PMCID: PMC3549487 DOI: 10.3389/fneur.2012.00187] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 12/21/2012] [Indexed: 01/21/2023] Open
Abstract
The Parkinsonian disorders are a large group of neurodegenerative diseases including idiopathic Parkinson’s disease (PD) and atypical Parkinsonian disorders (APD), such as multiple system atrophy, progressive supranuclear palsy, corticobasal degeneration, and dementia with Lewy bodies. The etiology of these disorders is not known although it is considered to be a combination of genetic and environmental factors. One of the greatest obstacles for developing efficacious disease-modifying treatment strategies is the lack of biomarkers. Reliable biomarkers are needed for early and accurate diagnosis, to measure disease progression, and response to therapy. In this review several of the most promising cerebrospinal biomarker candidates are discussed. Alpha-synuclein seems to be intimately involved in the pathogenesis of synucleinopathies and its levels can be measured in the cerebrospinal fluid and in plasma. In a similar way, tau protein accumulation seems to be involved in the pathogenesis of tauopathies. Urate, a potent antioxidant, seems to be associated to the risk of developing PD and with its progression. Neurofilament light chain levels are increased in APD compared with PD and healthy controls. The new “omics” techniques are potent tools offering new insights in the patho-etiology of these disorders. Some of the difficulties encountered in developing biomarkers are discussed together with future perspectives.
Collapse
Affiliation(s)
- Radu Constantinescu
- Department of Neurology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg Gothenburg, Sweden
| | | |
Collapse
|
228
|
Burdett TC, Desjardins CA, Logan R, McFarland NR, Chen X, Schwarzschild MA. Efficient determination of purine metabolites in brain tissue and serum by high-performance liquid chromatography with electrochemical and UV detection. Biomed Chromatogr 2013; 27:122-9. [PMID: 22674671 PMCID: PMC9979337 DOI: 10.1002/bmc.2760] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 04/26/2012] [Indexed: 12/27/2022]
Abstract
The purine metabolic pathway has been implicated in neurodegeneration and neuroprotection. High-performance liquid chromatography (HPLC) is widely used to determine purines and metabolites. However, methods for analysis of multiple purines in a single analysis have not been standardized, especially in brain tissue. We report the development and validation of a reversed-phase HPLC method combining electrochemical and UV detection after a short gradient run to measure seven purine metabolites (adenosine, guanosine, inosine, guanine, hypoxanthine, xanthine and urate) from the entire purine metabolic pathway. The limit of detection (LoD) for each analyte was determined. The LoD using UV absorption was 0.001 mg/dL for hypoxanthine (Hyp), inosine (Ino), guanosine (Guo) and adenosine (Ado), and those using coulometric electrodes were 0.001 mg/dL for guanine (Gua), 0.0001 mg/dL for urate (UA) and 0.0005 mg/dL for xanthine (Xan). The intra- and inter-day coefficient of variance was generally <8%. Using this method, we determined basal levels of these metabolites in mouse brain and serum, as well as in post-mortem human brain. Peak identities were confirmed by enzyme degradation. Spike recovery was performed to assess accuracy. All recoveries fell within 80-120%. Our HPLC method provides a sensitive, rapid, reproducible and low-cost method for determining multiple purine metabolites in a single analysis in serum and brain specimens.
Collapse
Affiliation(s)
- Thomas C. Burdett
- The MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th, Charlestown, MA 02129, USA
| | - Cody A. Desjardins
- The MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th, Charlestown, MA 02129, USA
| | - Robert Logan
- The MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th, Charlestown, MA 02129, USA
| | - Nikolaus R. McFarland
- University of Florida, Department of Neurology Center for Translational Research in Neurodegenerative Disease, PO Box 100159, Gainesville, FL 32610, USA
| | - Xiqun Chen
- The MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th, Charlestown, MA 02129, USA,Correspondence to: Xiqun Chen, The MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School 114 16th, Charlestown, MA 02129, USA.
| | - Michael A. Schwarzschild
- The MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th, Charlestown, MA 02129, USA
| |
Collapse
|
229
|
Pienaar IS, Chinnery PF. Existing and emerging mitochondrial-targeting therapies for altering Parkinson's disease severity and progression. Pharmacol Ther 2013; 137:1-21. [DOI: 10.1016/j.pharmthera.2012.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 08/07/2012] [Indexed: 02/07/2023]
|
230
|
Pan M, Gao H, Long L, Xu Y, Liu M, Zou J, Wu A, Wei X, Chen X, Tang B, Wang Q. Serum uric acid in patients with Parkinson's disease and vascular parkinsonism: a cross-sectional study. Neuroimmunomodulation 2013; 20:19-28. [PMID: 23154271 DOI: 10.1159/000342483] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 08/03/2012] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Elevation of serum uric acid (UA) is correlated with a decreased risk of Parkinson's disease (PD); however, the association and clinical relevance of serum UA levels in patients with PD and vascular parkinsonism (VP) are unknown. OBJECTIVE We performed a cross-sectional study of 160 Chinese patients with PD and VP to determine whether UA levels in patients could predict the outcomes. METHODS Serum UA levels were divided into quartiles and the association between UA and the severity of PD or VP was investigated in each quartile. RESULTS The serum levels of UA in PD were significantly lower than those in normal subjects and VP. The serum UA levels in PD patients were significantly correlated with some clinical parameters. Strong correlations were observed in male PD patients, but significant correlations were observed only between UA and the non-motor symptoms (NMS) of burden of sleep/fatigue and mood in female PD patients. PD patients in the lowest quartile of serum UA levels had significant correlations between UA and the unified Parkinson's disease rating scale, the modified Hoehn and Yahr staging scale and NMS burden for attention/memory. CONCLUSION Our findings support the hypothesis that subjects with low serum UA levels may be more prone to developing PD and indicate that the inverse relationship between UA and severity of PD was robust for men but weak for women. Our results strongly imply that either low serum UA level is a deteriorative predictor or that serum UA level serves as an indirect biomarker of prediction in PD but not in VP patients.
Collapse
Affiliation(s)
- Mengqiu Pan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Chen X, Wu G, Schwarzschild MA. Urate in Parkinson's disease: more than a biomarker? Curr Neurol Neurosci Rep 2012; 12:367-75. [PMID: 22580741 DOI: 10.1007/s11910-012-0282-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with characteristic motor manifestations. Although appreciation of PD as a multisystem disorder has grown, loss of dopaminergic neurons in the substantia nigra remains a pathological and neurochemical hallmark, accounting for the substantial symptomatic benefits of dopamine replacement therapies. However, currently no treatment has been shown to prevent or forestall the progression of the disease in spite of tremendous efforts. Among multiple environmental and genetic factors that have been implicated in the pathogenesis of PD, oxidative stress is proposed to play a critical role. A recent confluence of clinical, epidemiological, and laboratory evidence identified urate, an antioxidant and end product of purine metabolism, as not only a molecular predictor for both reduced risk and favorable progression of PD but also a potential neuroprotectant for the treatment of PD. This review summarizes recent findings on urate in PD and their clinical implications.
Collapse
Affiliation(s)
- Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA.
| | | | | |
Collapse
|
232
|
Bird SS, Sheldon DP, Gathungu RM, Vouros P, Kautz R, Matson WR, Kristal BS. Structural characterization of plasma metabolites detected via LC-electrochemical coulometric array using LC-UV fractionation, MS, and NMR. Anal Chem 2012; 84:9889-98. [PMID: 23106399 DOI: 10.1021/ac302278u] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liquid chromatography (LC) separation combined with electrochemical coulometric array detection (EC) is a sensitive, reproducible, and robust technique that can detect hundreds of redox-active metabolites down to the level of femtograms on column, making it ideal for metabolomics profiling. EC detection cannot, however, structurally characterize unknown metabolites that comprise these profiles. Several aspects of LC-EC methods prevent a direct transfer to other structurally informative analytical methods, such as LC-MS and NMR. These include system limits of detection, buffer requirements, and detection mechanisms. To address these limitations, we developed a workflow based on the concentration of plasma, metabolite extraction, and offline LC-UV fractionation. Pooled human plasma was used to provide sufficient material necessary for multiple sample concentrations and platform analyses. Offline parallel LC-EC and LC-MS methods were established that correlated standard metabolites between the LC-EC profiling method and the mass spectrometer. Peak retention times (RT) from the LC-MS and LC-EC system were linearly related (r(2) = 0.99); thus, LC-MS RTs could be directly predicted from the LC-EC signals. Subsequent offline microcoil-NMR analysis of these collected fractions was used to confirm LC-MS characterizations by providing complementary, structural data. This work provides a validated workflow that is transferrable across multiple platforms and provides the unambiguous structural identifications necessary to move primary mathematically driven LC-EC biomarker discovery into biological and clinical utility.
Collapse
Affiliation(s)
- Susan S Bird
- Department of Neurosurgery, Brigham and Women's Hospital, and Harvard Medical School, 221 Longwood Avenue, LMRC-322, Boston, Massachusetts 02115, United States
| | | | | | | | | | | | | |
Collapse
|
233
|
Shoemaker LD, Achrol AS, Sethu P, Steinberg GK, Chang SD. Clinical neuroproteomics and biomarkers: from basic research to clinical decision making. Neurosurgery 2012; 70:518-25. [PMID: 21866062 DOI: 10.1227/neu.0b013e3182333a26] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Clinical neuroproteomics aims to advance our understanding of disease and injury affecting the central and peripheral nervous systems through the study of protein expression and the discovery of protein biomarkers to facilitate diagnosis and treatment. The general premise of the biomarker field is that in vivo factors present in either tissue or circulating biofluids, reflect pathological changes, and can be identified and analyzed. This approach offers an opportunity to illuminate changes occurring at both the population and patient levels toward the realization of personalized medicine. This review is intended to provide research-driven clinicians with an overview of protein biomarkers of disease and injury for clinical use and to highlight methodology and potential pitfalls. We examine the neuroproteomic biomarker field and discuss the hallmarks and the challenges of clinically relevant biomarker discovery relating to central nervous system pathology. We discuss the issues in the maturation of potential biomarkers from discovery to Food and Drug Administration approval and review several platforms for protein biomarker discovery, including protein microarray and mass spectrometry-based proteomics. We describe the application of microfluidic technologies to the evolution of a robust clinical test. Finally, we highlight several biomarkers currently in use for cancer, ischemia, and injury in the central nervous system. Future efforts using these technologies will result in the maturation of existing and the identification of de novo biomarkers that could guide clinical decision making and advance diagnostic and therapeutic options for the treatment of neurological disease and injury.
Collapse
Affiliation(s)
- Lorelei D Shoemaker
- Department of Neurosurgery, Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University, Stanford, California 94305, USA
| | | | | | | | | |
Collapse
|
234
|
Wang J, Hoekstra JG, Zuo C, Cook TJ, Zhang J. Biomarkers of Parkinson's disease: current status and future perspectives. Drug Discov Today 2012; 18:155-62. [PMID: 22982303 DOI: 10.1016/j.drudis.2012.09.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 08/14/2012] [Accepted: 09/05/2012] [Indexed: 01/09/2023]
Abstract
This review summarizes major advances in biomarker discovery for diagnosis, differential diagnosis and progression of Parkinson's disease (PD), with emphasis on neuroimaging and biochemical markers. Potential strategies to develop biomarkers capable of predicting PD in the prodromal stage before the appearance of motor symptoms or correlating with nonmotor symptoms, an active area of research, are also discussed.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
235
|
Abstract
Though the search for cerebrospinal fluid (CSF) biomarkers of Parkinson's disease (PD) began more than 40 years ago, the most promising results are relatively recent. Disease-specific indicators have been sought among the hundreds of proteins and other biochemicals found in CSF (which is contiguous with the extracellular fluid compartment of the brain). Initially, research focused on the selective neurotransmitter disturbance in PD. While investigations of dopamine metabolism (as reflected by its major metabolite, homovanillic acid [HVA]) have been relatively uninformative, we found that indexing HVA concentration to that of the purine metabolite xanthine permits differentiation of PD specimens from healthy controls (p < 0.0016) [Brain Research 2011;1408:88-97]. In another recent biomarker study, we utilized ultrahigh-performance liquid chromatography linked to gas chromatography-mass spectrometry for metabolomic analysis [Movement Disorders 2011;26(Suppl 2):S193]. Using t-tests to differentiate PD and control groups at p < 0.02, we found changes in compounds not known to be related to the neurodegenerative process (4 increased in CSF concentration and 8 decreased). Other recent investigations have reported distinctive biomarker findings in proteins and other biochemicals. The ultimate goal is for CSF biomarkers also found in peripheral biospecimens, aiding in diagnostic screening applications and providing further clues as to the etiology of PD.
Collapse
Affiliation(s)
- Peter LeWitt
- Department of Neurology, Henry Ford Hospital, Michigan 48322, USA.
| |
Collapse
|
236
|
Agarwal PA, Stoessl AJ. Biomarkers for trials of neuroprotection in Parkinson's disease. Mov Disord 2012; 28:71-85. [DOI: 10.1002/mds.25065] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 04/19/2012] [Accepted: 04/23/2012] [Indexed: 02/06/2023] Open
Affiliation(s)
- Pankaj A. Agarwal
- Pacific Parkinson's Research Centre; University of British Columbia; Vancouver; British Columbia; Canada
| | | |
Collapse
|
237
|
Mollenhauer B, Zhang J. Biochemical premotor biomarkers for Parkinson's disease. Mov Disord 2012; 27:644-50. [PMID: 22508282 DOI: 10.1002/mds.24956] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A biomarker is a biological characteristic that is objectively measured and evaluated as an indicator of normal biological or pathologic processes or of pharmacologic responses to a therapeutic intervention. We reviewed the current status of target protein biomarkers (eg, total/oligomeric α-synuclein and DJ-1) in cerebrospinal fluid, as well as on unbiased processes that can be used to discover novel biomarkers. We have also provide details about strategies toward potential populations/models and technologies, including the need for standardized sampling techniques, to pursue the identification of new biochemical markers in the premotor stage of Parkinson's disease in the future.
Collapse
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik and Georg August University Goettingen, Kassel, Germany.
| | | |
Collapse
|
238
|
Cipriani S, Desjardins CA, Burdett TC, Xu Y, Xu K, Schwarzschild MA. Protection of dopaminergic cells by urate requires its accumulation in astrocytes. J Neurochem 2012; 123:172-81. [PMID: 22671773 DOI: 10.1111/j.1471-4159.2012.07820.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Urate is the end product of purine metabolism and a major antioxidant circulating in humans. Recent data link higher levels of urate with a reduced risk of developing Parkinson's disease and with a slower rate of its progression. In this study, we investigated the role of astrocytes in urate-induced protection of dopaminergic cells in a cellular model of Parkinson's disease. In mixed cultures of dopaminergic cells and astrocytes oxidative stress-induced cell death and protein damage were reduced by urate. By contrast, urate was not protective in pure dopaminergic cell cultures. Physical contact between dopaminergic cells and astrocytes was not required for astrocyte-dependent rescue as shown by conditioned medium experiments. Urate accumulation in dopaminergic cells and astrocytes was blocked by pharmacological inhibitors of urate transporters expressed differentially in these cells. The ability of a urate transport blocker to prevent urate accumulation into astroglial (but not dopaminergic) cells predicted its ability to prevent dopaminergic cell death. Transgenic expression of uricase reduced urate accumulation in astrocytes and attenuated the protective influence of urate on dopaminergic cells. These data indicate that urate might act within astrocytes to trigger release of molecule(s) that are protective for dopaminergic cells.
Collapse
Affiliation(s)
- Sara Cipriani
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA 02129, USA.
| | | | | | | | | | | |
Collapse
|
239
|
Parkinson's disease. Transl Neurosci 2012. [DOI: 10.1017/cbo9780511980053.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
240
|
Urate and its transgenic depletion modulate neuronal vulnerability in a cellular model of Parkinson's disease. PLoS One 2012; 7:e37331. [PMID: 22606360 PMCID: PMC3351394 DOI: 10.1371/journal.pone.0037331] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/19/2012] [Indexed: 02/03/2023] Open
Abstract
Urate is a major antioxidant as well as the enzymatic end product of purine metabolism in humans. Higher levels correlate with a reduced risk of developing Parkinson's disease (PD) and with a slower rate of PD progression. In this study we investigated the effects of modulating intracellular urate concentration on 1-methyl-4-phenyl-pyridinium (MPP+)-induced degeneration of dopaminergic neurons in cultures of mouse ventral mesencephalon prepared to contain low (neuron-enriched cultures) or high (neuron-glial cultures) percentage of astrocytes. Urate, added to the cultures 24 hours before and during treatment with MPP+, attenuated the loss of dopaminergic neurons in neuron-enriched cultures and fully prevented their loss and atrophy in neuron-astrocyte cultures. Exogenous urate was found to increase intracellular urate content in cortical neuronal cultures. To assess the effect of reducing cellular urate content on MPP+-induced toxicity, mesencephalic neurons were prepared from mice over-expressing urate oxidase (UOx). Transgenic UOx expression decreased endogenous urate content both in neurons and astrocytes. Dopaminergic neurons expressing UOx were more susceptible to MPP+ in mesencephalic neuron-enriched cultures and to a greater extent in mesencephalic neuron-astrocyte cultures. Our findings correlate intracellular urate content in dopaminergic neurons with their toxin resistance in a cellular model of PD and suggest a facilitative role for astrocytes in the neuroprotective effect of urate.
Collapse
|
241
|
Abstract
Incidence and prevalence of gout have markedly increased over the last few decades in keeping with the rise in prevalence of obesity and metabolic syndrome. Until recently, management of gout in patients with associated metabolic syndrome and comorbid illnesses such as renal impairment was difficult because of limited treatment options. However, significant progress has been made in the last few years, with introduction of new treatments such as interleukin-1 antagonists for management of acute gout, and febuxostat and pegloticase for chronic gout. The association of gout with alcohol, dietary purines and fructose ingestion has been confirmed in large prospective studies, thus enabling the clinician to now provide evidence-based advice to patients. Recent efficacy and safety data favour lower over higher doses of colchicine, and oral corticosteroids over non-steroidal anti-inflammatory drugs for patients with acute gout. Local ice therapy might help to differentiate gout from other forms of inflammatory arthritis, and supplementation with vitamin C help to reduce risk of gout. Several other drugs with rational mechanisms of action are in the pipeline, and likely to be introduced over the next few years. A new era has thus begun in the field of gout.
Collapse
Affiliation(s)
- E Suresh
- Rheumatology Department, Kettering General Hospital, Rothwell Road, Kettering, NN16 8UZ, UK.
| | | |
Collapse
|
242
|
Gao X, Simon KC, Schwarzschild MA, Ascherio A. Prospective study of statin use and risk of Parkinson disease. ACTA ACUST UNITED AC 2012; 69:380-4. [PMID: 22410446 DOI: 10.1001/archneurol.2011.1060] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
OBJECTIVE To prospectively examine whether use of statins is associated with altered risk of Parkinson disease (PD). DESIGN, SETTING, AND PARTICIPANTS A prospective study including 38 192 men and 90 874 women participating in 2 ongoing US cohorts, the Health Professional Follow-up Study and the Nurses' Health Study, was conducted. Information on regular cholesterol-lowering drug use (≥2 times/wk) was collected in 1994 in both cohorts via questionnaire. Relative risks (RRs) and 95% CIs were computed using Cox proportional hazards models adjusting for age, smoking, caffeine intake, duration of hypercholesterolemia, and other covariates. MAIN OUTCOME MEASURE Incident PD. RESULTS During 12 years of follow-up (1994-2006), we documented 644 incident PD cases (338 women and 306 men). The risk of PD was lower among current statin users (adjusted pooled RR = 0.74; 95% CI, 0.54-1.00; P = .049) relative to nonusers. A significant association was observed in participants younger than 60 years at baseline (adjusted pooled RR = 0.31; 95% CI, 0.11-0.86; P = .02) but not among those who were older (adjusted pooled RR = 0.83; 95% CI, 0.60-1.14; P = .25) (P for interaction = .03). CONCLUSIONS We found that regular use of statins was associated with a modest reduction in PD risk. The possibility that some statins may reduce PD risk deserves further consideration.
Collapse
Affiliation(s)
- Xiang Gao
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
243
|
Arts IC, Coolen EJ, Bours MJ, Huyghebaert N, Stuart MAC, Bast A, Dagnelie PC. Adenosine 5'-triphosphate (ATP) supplements are not orally bioavailable: a randomized, placebo-controlled cross-over trial in healthy humans. J Int Soc Sports Nutr 2012; 9:16. [PMID: 22510240 PMCID: PMC3441280 DOI: 10.1186/1550-2783-9-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 04/06/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nutritional supplements designed to increase adenosine 5'-triphosphate (ATP) concentrations are commonly used by athletes as ergogenic aids. ATP is the primary source of energy for the cells, and supplementation may enhance the ability to maintain high ATP turnover during high-intensity exercise. Oral ATP supplements have beneficial effects in some but not all studies examining physical performance. One of the remaining questions is whether orally administered ATP is bioavailable. We investigated whether acute supplementation with oral ATP administered as enteric-coated pellets led to increased concentrations of ATP or its metabolites in the circulation. METHODS Eight healthy volunteers participated in a cross-over study. Participants were given in random order single doses of 5000 mg ATP or placebo. To prevent degradation of ATP in the acidic environment of the stomach, the supplement was administered via two types of pH-sensitive, enteric-coated pellets (targeted at release in the proximal or distal small intestine), or via a naso-duodenal tube. Blood ATP and metabolite concentrations were monitored by HPLC for 4.5 h (naso-duodenal tube) or 7 h (pellets) post-administration. Areas under the concentration vs. time curve were calculated and compared by paired-samples t-tests. RESULTS ATP concentrations in blood did not increase after ATP supplementation via enteric-coated pellets or naso-duodenal tube. In contrast, concentrations of the final catabolic product of ATP, uric acid, were significantly increased compared to placebo by ~50% after administration via proximal-release pellets (P = 0.003) and naso-duodenal tube (P = 0.001), but not after administration via distal-release pellets. CONCLUSIONS A single dose of orally administered ATP is not bioavailable, and this may explain why several studies did not find ergogenic effects of oral ATP supplementation. On the other hand, increases in uric acid after release of ATP in the proximal part of the small intestine suggest that ATP or one of its metabolites is absorbed and metabolized. Uric acid itself may have ergogenic effects, but this needs further study. Also, more studies are needed to determine whether chronic administration of ATP will enhance its oral bioavailability.
Collapse
Affiliation(s)
- Ilja Cw Arts
- Department of Epidemiology, Maastricht University, P,O, Box 616, Maastricht, MD, 6200, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
244
|
Zhang HN, Guo JF, He D, Lei LF, Wang YQ, Wang CY, Tan LM, Yan XX, Shen L, Tang BS. Lower serum UA levels in Parkinson's disease patients in the Chinese population. Neurosci Lett 2012; 514:152-5. [DOI: 10.1016/j.neulet.2012.02.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 01/31/2012] [Accepted: 02/23/2012] [Indexed: 10/28/2022]
|
245
|
Martinez-Fong D, Bannon MJ, Trudeau LE, Gonzalez-Barrios JA, Arango-Rodriguez ML, Hernandez-Chan NG, Reyes-Corona D, Armendáriz-Borunda J, Navarro-Quiroga I. NTS-Polyplex: a potential nanocarrier for neurotrophic therapy of Parkinson's disease. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 8:1052-69. [PMID: 22406187 DOI: 10.1016/j.nano.2012.02.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 02/14/2012] [Accepted: 02/20/2012] [Indexed: 10/28/2022]
Abstract
UNLABELLED Nanomedicine has focused on targeted neurotrophic gene delivery to the brain as a strategy to stop and reverse neurodegeneration in Parkinson's disease. Because of improved transfection ability, synthetic nanocarriers have become candidates for neurotrophic therapy. Neurotensin (NTS)-polyplex is a "Trojan horse" synthetic nanocarrier system that enters dopaminergic neurons through NTS receptor internalization to deliver a genetic cargo. The success of preclinical studies with different neurotrophic genes supports the possibility of using NTS-polyplex in nanomedicine. In this review, we describe the mechanism of NTS-polyplex transfection. We discuss the concept that an effective neurotrophic therapy requires a simultaneous effect on the axon terminals and soma of the remaining dopaminergic neurons. We also discuss the future of this strategy for the treatment of Parkinson's disease. FROM THE CLINICAL EDITOR This review paper focuses on nanomedicine-based treatment of Parkinson's disease, a neurodegenerative condition with existing symptomatic but no curative treatment. Neurotensin-polyplex is a synthetic nanocarrier system that enables delivery of genetic cargo to dopaminergic neurons via NTS receptor internalization.
Collapse
|
246
|
Benetti F, Gustincich S, Legname G. Gene expression profiling and therapeutic interventions in neurodegenerative diseases: a comprehensive study on potentiality and limits. Expert Opin Drug Discov 2012; 7:245-59. [PMID: 22468955 DOI: 10.1517/17460441.2012.659661] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Neurodegenerative diseases are incurable debilitating disorders of the nervous system that affect approximately 30 million people worldwide. Despite profuse efforts attempting to define the molecular mechanisms underlying neurodegeneration, many aspects of these pathologies remain elusive. The novelty of their mechanisms represents a challenge to biology, to their related biomarkers identification and drug discovery. Because of their multifactorial aspects and complexity, gene expression analysis platforms have been extensively used to investigate altered pathways during degeneration and to identify potential biomarkers and drug targets. AREAS COVERED This work offers an overview of the gene expression profiling studies carried out on Alzheimer's disease, Huntington's disease, Parkinson's disease and prion disease specimens. Therapeutic approaches are also discussed. EXPERT OPINION Although many therapeutic approaches have been tested, some of them acting on several altered cellular pathways, no effective cures for these neurodegenerative diseases have been identified. Microarray technology must be associated with functional proteomics and physiology in an effort to identify specific and selective biomarkers and druggable targets, thus allowing the successful discovery of disease-modifying therapeutic treatments.
Collapse
Affiliation(s)
- Federico Benetti
- Laboratory of Prion Biology, Neurobiology Sector, Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy
| | | | | |
Collapse
|
247
|
Uric acid levels predict survival in men with amyotrophic lateral sclerosis. J Neurol 2012; 259:1923-8. [PMID: 22323210 DOI: 10.1007/s00415-012-6440-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/21/2012] [Accepted: 01/24/2012] [Indexed: 10/14/2022]
Abstract
Elevated uric acid levels have recently been found to be associated with slower disease progression in Parkinson's disease, Huntington's disease, multiple system atrophy, and mild cognitive impairment. The aim of this study is to determine whether serum uric acid levels predict survival in amyotrophic lateral sclerosis (ALS). A total of 251 people with ALS enrolled in two multicenter clinical trials were included in our analysis. The main outcome measure was survival time, which was calculated as time to death, tracheostomy, or permanent assistive ventilation, with any event considered a survival endpoint. Cox proportional hazards models were used to estimate the hazard ratio (HR) of reaching a survival endpoint according to baseline uric acid levels after adjusting for markers of disease severity (FVC, total ALSFRS-R score, time since symptom onset, diagnostic delay, BMI, bulbar vs. spinal onset, age, and riluzole use). There was a dose-dependent survival advantage in men, but not women, with higher baseline uric acid levels (logrank test: p = 0.018 for men, p = 0.81 for women). There was a 39% reduction in risk of death during the study for men with each 1 mg/dl increase in uric acid levels (adjusted HR: 0.61, 95% CI 0.39-0.96, p = 0.03). This is the first study to demonstrate that serum uric acid is associated with prolonged survival in ALS, after adjusting for markers of disease severity. Similar to previous reports in Parkinson's disease, this association was seen in male subjects only.
Collapse
|
248
|
Abstract
Parkinson's disease (PD) is the most common age-related motoric neurodegenerative disease initially described in the 1800's by James Parkinson as the 'Shaking Palsy'. Loss of the neurotransmitter dopamine was recognized as underlying the pathophysiology of the motor dysfunction; subsequently discovery of dopamine replacement therapies brought substantial symptomatic benefit to PD patients. However, these therapies do not fully treat the clinical syndrome nor do they alter the natural history of this disorder motivating clinicians and researchers to further investigate the clinical phenotype, pathophysiology/pathobiology and etiology of this devastating disease. Although the exact cause of sporadic PD remains enigmatic studies of familial and rare toxicant forms of this disorder have laid the foundation for genome wide explorations and environmental studies. The combination of methodical clinical evaluation, systematic pathological studies and detailed genetic analyses have revealed that PD is a multifaceted disorder with a wide-range of clinical symptoms and pathology that include regions outside the dopamine system. One common thread in PD is the presence of intracytoplasmic inclusions that contain the protein, α-synuclein. The presence of toxic aggregated forms of α-synuclein (e.g., amyloid structures) are purported to be a harbinger of subsequent pathology. In fact, PD is both a cerebral amyloid disease and the most common synucleinopathy, that is, diseases that display accumulations of α-synuclein. Here we present our current understanding of PD etiology, pathology, clinical symptoms and therapeutic approaches with an emphasis on misfolded α-synuclein.
Collapse
Affiliation(s)
- Timothy R. Mhyre
- Department of Neuroscience, Georgetown University Medical Center, NRB EP08, 3970 Reservoir Road NW, 20057, Washington, DC, USA,
| | - James T. Boyd
- University of Vermont College of Medicine, 1 South Prospect Street, DU-Arnold 4416-UHC, 05401, Burlington, VT, USA,
| | - Robert W. Hamill
- Department of Neurology, University of Vermont College of Medicine, 89 Beaumont Avenue, Given Hall Room C225, 05405, Burlington, VT, USA,
| | - Kathleen A. Maguire-Zeiss
- Department of Neuroscience, Center for Neural Injury and RecoveryGeorgetown University Medical Center, 3970 Reservoir Road, NW NRB EP08, 20057, Washington, DC, USA,
| |
Collapse
|
249
|
Abstract
High uric acid (UA) levels can cause gout, urolithiasis and acute and chronic nephropathy, all of which are due to the deposit of urate crystals. There is also increasing evidence of relationships of hyperuricemia with other important disorders, including hypertension, chronic renal disease, metabolic syndrome and cardiovascular disease, as well as an increased mortality, although a causal relationship between these conditions has not been clearly established. On the other hand, low UA levels are not known to cause any disorder or disease. However, in the last few years a higher prevalence and progression of some neurological diseases have been associated with a low UA, and it is possible that they may predispose to some other disorders, mainly due to the decrease in its antioxidant activity. In this article, the known negative effects of UA are reviewed, as well as the much less-known possible positive actions, and their therapeutic implications.
Collapse
Affiliation(s)
- B Alvarez-Lario
- Sección de Reumatología, Complejo Asistencial Universitario de Burgos, Avda. del Cid 96, 09005 Burgos, Spain.
| | | |
Collapse
|
250
|
Seidl SE, Potashkin JA. The promise of neuroprotective agents in Parkinson's disease. Front Neurol 2011; 2:68. [PMID: 22125548 PMCID: PMC3221408 DOI: 10.3389/fneur.2011.00068] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 10/21/2011] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by loss of dopamine neurons in the substantia nigra of the brain. Since there are limited treatment options for PD, neuroprotective agents are currently being tested as a means to slow disease progression. Agents targeting oxidative stress, mitochondrial dysfunction, and inflammation are prime candidates for neuroprotection. This review identifies Rasagiline, Minocycline, and creatine, as the most promising neuroprotective agents for PD, and they are all currently in phase III trials. Other agents possessing protective characteristics in delaying PD include stimulants, vitamins, supplements, and other drugs. Additionally, combination therapies also show benefits in slowing PD progression. The identification of neuroprotective agents for PD provides us with therapeutic opportunities for modifying the course of disease progression and, perhaps, reducing the risk of onset when preclinical biomarkers become available.
Collapse
Affiliation(s)
- Stacey E Seidl
- Department of Biological Sciences, DePaul University Chicago, IL, USA
| | | |
Collapse
|