101
|
Lanznaster D, Dal-Cim T, Piermartiri TCB, Tasca CI. Guanosine: a Neuromodulator with Therapeutic Potential in Brain Disorders. Aging Dis 2016; 7:657-679. [PMID: 27699087 PMCID: PMC5036959 DOI: 10.14336/ad.2016.0208] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/08/2016] [Indexed: 12/20/2022] Open
Abstract
Guanosine is a purine nucleoside with important functions in cell metabolism and a protective role in response to degenerative diseases or injury. The past decade has seen major advances in identifying the modulatory role of extracellular action of guanosine in the central nervous system (CNS). Evidence from rodent and cell models show a number of neurotrophic and neuroprotective effects of guanosine preventing deleterious consequences of seizures, spinal cord injury, pain, mood disorders and aging-related diseases, such as ischemia, Parkinson’s and Alzheimer’s diseases. The present review describes the findings of in vivo and in vitro studies and offers an update of guanosine effects in the CNS. We address the protein targets for guanosine action and its interaction with glutamatergic and adenosinergic systems and with calcium-activated potassium channels. We also discuss the intracellular mechanisms modulated by guanosine preventing oxidative damage, mitochondrial dysfunction, inflammatory burden and modulation of glutamate transport. New and exciting avenues for future investigation into the protective effects of guanosine include characterization of a selective guanosine receptor. A better understanding of the neuromodulatory action of guanosine will allow the development of therapeutic approach to brain diseases.
Collapse
Affiliation(s)
- Débora Lanznaster
- 2Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil; 3CAPES Foundation, Ministry of Education of Brazil, Brasília - DF 70040-020, Brazil
| | - Tharine Dal-Cim
- 2Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil; 3CAPES Foundation, Ministry of Education of Brazil, Brasília - DF 70040-020, Brazil
| | - Tetsadê C B Piermartiri
- 2Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil; 3CAPES Foundation, Ministry of Education of Brazil, Brasília - DF 70040-020, Brazil
| | - Carla I Tasca
- 1Departamento de Bioquímica,; 2Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil
| |
Collapse
|
102
|
Bega D, Kim S, Zhang Y, Elm J, Schneider J, Hauser R, Fraser A, Simuni T. Predictors of Functional Decline in Early Parkinson's Disease: NET-PD LS1 Cohort. JOURNAL OF PARKINSONS DISEASE 2016; 5:773-82. [PMID: 26639661 DOI: 10.3233/jpd-150668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Data on predictors of decline in PD are largely based on de-novo populations and limited to the use of motor outcomes that fail to capture the full scope of disease. OBJECTIVE Determine the clinical predictors of decline in early treated PD using a novel multi-domain measure. METHODS Data from NINDS Exploratory Trials in PD Long-Term Study 1 (NET-PD LS1), a multicenter Phase 3 study of creatine in early treated PD, were analyzed. Functional decline was defined by a global outcome metric (GO) that consisted of: Schwab and England ADL scale, PD 39-item Questionnaire, Unified PD Rating Scale, Ambulatory Capacity Score, Symbol Digit Modalities Test, and Modified Rankin Scale. Univariate and multivariate models were used to test the association of predictors of interest with a standardized rank-sum of the GO. RESULTS 765 of 1741 participants completed five-year assessments and were included. Older age at disease onset (p < 0.0001), higher baseline levodopa equivalent dose (p = 0.01), and worse Scales for Outcomes of Parkinson's Disease Cognition score (p = 0.001) at baseline were the strongest predictors of functional decline in multivariate analysis. PD symptom subtype was not a significant predictor of outcome (p = 0.42). The full model was only a modest predictor of change in GO (R2 = 0.186). CONCLUSIONS This is the largest study to systematically assess predictors of functional decline in early treated PD over several years, and the first to use a multi-domain outcome measure of decline. Older age at disease onset and worse cognition, and not PD subtype, were predictors of decline.
Collapse
Affiliation(s)
- Danny Bega
- Northwestern University, Chicago, IL, USA
| | - Soeun Kim
- University of Texas Health Science Center, Houston, TX, USA
| | - Yunxi Zhang
- University of Texas Health Science Center, Houston, TX, USA
| | - Jordan Elm
- Medical University of South Carolina, Charleston, SC, USA
| | | | | | - Andy Fraser
- Oregon Health Sciences University, Portland, OR, USA
| | | |
Collapse
|
103
|
O'Reilly ÉIJ, Liu D, Johns DR, Cudkowicz ME, Paganoni S, Schwarzschild MA, Leitner M, Ascherio A. Serum urate at trial entry and ALS progression in EMPOWER. Amyotroph Lateral Scler Frontotemporal Degener 2016; 18:120-125. [PMID: 27677562 DOI: 10.1080/21678421.2016.1214733] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our objective was to determine whether serum urate predicts ALS progression. A study population comprised adult participants of EMPOWER (n = 942), a phase III clinical trial to evaluate the efficacy of dexpramipexole to treat ALS. Urate was measured in blood samples collected during enrollment as part of the routine block chemistry. We measured outcomes by combined assessment of function and survival rank (CAFs), and time to death, by 12 months. Results showed that in females there was not a significant relation between urate and outcomes. In males, outcomes improved with increasing urate (comparing highest to lowest urate quartile: CAFS was 53 points better with p for trend = 0.04; and hazard ratio for death was 0.60 with p for trend = 0.07), but with adjustment for body mass index (BMI) at baseline, a predictor of both urate levels and prognosis, associations were attenuated and no longer statistically significant. Overall, participants with urate levels equal to or above the median (5.1 mg/dl) appeared to have a survival advantage compared to those below (hazard ratio adjusted for BMI: 0.67; 95% confidence interval 0.47-0.95). In conclusion, these findings suggest that while the association between urate at baseline and ALS progression is partially explained by BMI, there may be an independent beneficial effect of urate.
Collapse
Affiliation(s)
- ÉIlis J O'Reilly
- a Department of Nutrition, Harvard TH Chan School of Public Health, Boston. Channing Division of Network Medicine , Harvard Medical School and Brigham and Women's Hospital , Boston , Massachusetts
| | - Dawei Liu
- b ALS Neuroscience Discovery and Development , Biogen , Cambridge , Massachusetts
| | - Donald R Johns
- b ALS Neuroscience Discovery and Development , Biogen , Cambridge , Massachusetts
| | - Merit E Cudkowicz
- c Department of Neurology , Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease , Boston , Massachusetts
| | - Sabrina Paganoni
- c Department of Neurology , Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease , Boston , Massachusetts.,d Department of Physical Medicine and Rehabilitation , Harvard Medical School, Spaulding Rehabilitation Hospital , Boston, Massachusetts; VA Healthcare System , Boston , Massachusetts , and
| | - Michael A Schwarzschild
- c Department of Neurology , Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease , Boston , Massachusetts
| | - Melanie Leitner
- b ALS Neuroscience Discovery and Development , Biogen , Cambridge , Massachusetts
| | - Alberto Ascherio
- e Department of Epidemiology and Nutrition , Harvard TH Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Harvard Medical School and Brigham and Women's Hospital , Boston , Massachusetts , USA
| |
Collapse
|
104
|
Pellecchia MT, Savastano R, Moccia M, Picillo M, Siano P, Erro R, Vallelunga A, Amboni M, Vitale C, Santangelo G, Barone P. Lower serum uric acid is associated with mild cognitive impairment in early Parkinson's disease: a 4-year follow-up study. J Neural Transm (Vienna) 2016; 123:1399-1402. [PMID: 27682634 DOI: 10.1007/s00702-016-1622-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/06/2016] [Indexed: 01/29/2023]
Abstract
Cognitive deficits are common in Parkinson's disease (PD) and many patients eventually develop dementia; however, its occurrence is unpredictable. Serum uric acid (UA) has been proposed as a biomarker of PD, both in the preclinical and clinical phase of the disease. The aim of this pilot study was to evaluate relationships between baseline serum UA levels and occurrence of mild cognitive impairment (MCI) at 4-year follow-up in a cohort of early PD patients. Early PD patients, not presenting concomitant diseases, cognitive impairment or treatment possibly interfering with UA levels, underwent neuropsychological testing at baseline and 4-year follow-up. UA levels were determined in serum at baseline. MCI was found in 23 out of 42 PD patients completing 4-year follow-up. Patients presenting MCI had significantly higher age at onset and lower Frontal Assessment Battery scores at baseline as compared with patients cognitively intact. Logistic regression analysis showed that both serum UA levels (OR = 0.54, p = 0.044) and age (OR = 1.16, p = 0.009) contribute to the occurrence of MCI at 4-year follow-up. Our pilot study suggests that lower levels of serum UA in the early disease stages are associated to the later occurrence of MCI. These results need to be confirmed by further studies on larger samples.
Collapse
Affiliation(s)
- Maria Teresa Pellecchia
- Neuroscience Section, Department of Medicine and Surgery, Center for Neurodegenerative Diseases (CEMAND), University of Salerno, Salerno, Italy.
| | | | - Marcello Moccia
- Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University, Naples, Italy
| | - Marina Picillo
- Neuroscience Section, Department of Medicine and Surgery, Center for Neurodegenerative Diseases (CEMAND), University of Salerno, Salerno, Italy
| | - Pietro Siano
- AOU San Giovanni di Dio e Ruggi d'Aragona, Salerno, Italy
| | - Roberto Erro
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Annamaria Vallelunga
- Neuroscience Section, Department of Medicine and Surgery, Center for Neurodegenerative Diseases (CEMAND), University of Salerno, Salerno, Italy
| | | | - Carmine Vitale
- IDC Hermitage-Capodimonte, Naples, Italy.,University of Naples Parthenope, Naples, Italy
| | - Gabriella Santangelo
- IDC Hermitage-Capodimonte, Naples, Italy.,Neuropsychology Laboratory, Department of Psychology, Second University of Naples, Caserta, Italy
| | - Paolo Barone
- Neuroscience Section, Department of Medicine and Surgery, Center for Neurodegenerative Diseases (CEMAND), University of Salerno, Salerno, Italy
| |
Collapse
|
105
|
Abstract
Despite an increased understanding of the pathogenesis of Parkinson's disease (PD), and a number of drugs designed to ameliorate symptoms, finding an effective neuroprotective therapy remains elusive. For decades now, several promising agents targeting different pathways have been explored as potential treatments that could help slow disease progression, but these have met with limited success. There are hurdles to overcome, particularly given that there is no exact animal model of PD and also no reliable biomarkers for PD. Without biomarkers, it is not possible to demonstrate, in the context of a clinical trial, that an intervention prevents neuronal degeneration. However, given the compelling scientific rationale of several compounds, an unrelenting pursuit continues. There have been hundreds of human studies looking at neuroprotection in PD. This article will briefly summarize several of the neuroprotective treatments that have been evaluated in large clinical trials, and will also outline some of the newer therapies that are currently being explored.
Collapse
Affiliation(s)
- Ariane Park
- Department of Neurology, The Ohio State University, Columbus, OH, USA.
| | - Mark Stacy
- Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
106
|
Tuite P. Magnetic resonance imaging as a potential biomarker for Parkinson's disease. Transl Res 2016; 175:4-16. [PMID: 26763585 DOI: 10.1016/j.trsl.2015.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 01/01/2023]
Abstract
Although a magnetic resonance imaging (MRI) biomarker for Parkinson's disease (PD) remains an unfulfilled objective, there have been numerous developments in MRI methodology and some of these have shown promise for PD. With funding from the National Institutes of Health and the Michael J Fox Foundation there will be further validation of structural, diffusion-based, and iron-focused MRI methods as possible biomarkers for PD. In this review, these methods and other strategies such as neurochemical and metabolic MRI have been covered. One of the challenges in establishing a biomarker is in the selection of individuals as PD is a heterogeneous disease with varying clinical features, different etiologies, and a range of pathologic changes. Additionally, longitudinal studies are needed of individuals with clinically diagnosed PD and cohorts of individuals who are at great risk for developing PD to validate methods. Ultimately an MRI biomarker will be useful in the diagnosis of PD, predicting the course of PD, providing a means to track its course, and provide an approach to select and monitor treatments.
Collapse
Affiliation(s)
- Paul Tuite
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
107
|
|
108
|
Polivka J, Polivka J, Krakorova K, Peterka M, Topolcan O. Current status of biomarker research in neurology. EPMA J 2016; 7:14. [PMID: 27379174 PMCID: PMC4931703 DOI: 10.1186/s13167-016-0063-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 06/02/2016] [Indexed: 01/18/2023]
Abstract
Neurology is one of the typical disciplines where personalized medicine has been recently becoming an important part of clinical practice. In this article, the brief overview and a number of examples of the use of biomarkers and personalized medicine in neurology are described. The various issues in neurology are described in relation to the personalized medicine and diagnostic, prognostic as well as predictive blood and cerebrospinal fluid biomarkers. Such neurological domains discussed in this work are neuro-oncology and primary brain tumors glioblastoma and oligodendroglioma, cerebrovascular diseases focusing on stroke, neurodegenerative disorders especially Alzheimer's and Parkinson's diseases and demyelinating diseases such as multiple sclerosis. Actual state of the art and future perspectives in diagnostics and personalized treatment in diverse domains of neurology are given.
Collapse
Affiliation(s)
- Jiri Polivka
- Department of Neurology, Faculty of Medicine in Plzen, Charles University Prague, Husova 3, 301 66 Plzen, Czech Republic ; Department of Neurology, Faculty Hospital Plzen, E. Benese 13, 305 99 Plzen, Czech Republic
| | - Jiri Polivka
- Department of Histology and Embryology, Charles University Prague, Husova 3, 301 66 Plzen, Czech Republic ; Biomedical Centre, Faculty of Medicine in Plzen, Charles University Prague, Husova 3, 301 66 Plzen, Czech Republic
| | - Kristyna Krakorova
- Department of Neurology, Faculty of Medicine in Plzen, Charles University Prague, Husova 3, 301 66 Plzen, Czech Republic ; Department of Neurology, Faculty Hospital Plzen, E. Benese 13, 305 99 Plzen, Czech Republic
| | - Marek Peterka
- Department of Neurology, Faculty of Medicine in Plzen, Charles University Prague, Husova 3, 301 66 Plzen, Czech Republic ; Department of Neurology, Faculty Hospital Plzen, E. Benese 13, 305 99 Plzen, Czech Republic
| | - Ondrej Topolcan
- Central Imunoanalytical Laboratory, Faculty Hospital Plzen, E. Benese 13, 305 99 Plzen, Czech Republic
| |
Collapse
|
109
|
Neuroprotective and Therapeutic Strategies against Parkinson's Disease: Recent Perspectives. Int J Mol Sci 2016; 17:ijms17060904. [PMID: 27338353 PMCID: PMC4926438 DOI: 10.3390/ijms17060904] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/18/2022] Open
Abstract
Parkinsonism is a progressive motor disease that affects 1.5 million Americans and is the second most common neurodegenerative disease after Alzheimer’s. Typical neuropathological features of Parkinson’s disease (PD) include degeneration of dopaminergic neurons located in the pars compacta of the substantia nigra that project to the striatum (nigro-striatal pathway) and depositions of cytoplasmic fibrillary inclusions (Lewy bodies) which contain ubiquitin and α-synuclein. The cardinal motor signs of PD are tremors, rigidity, slow movement (bradykinesia), poor balance, and difficulty in walking (Parkinsonian gait). In addition to motor symptoms, non-motor symptoms that include autonomic and psychiatric as well as cognitive impairments are pressing issues that need to be addressed. Several different mechanisms play an important role in generation of Lewy bodies; endoplasmic reticulum (ER) stress induced unfolded proteins, neuroinflammation and eventual loss of dopaminergic neurons in the substantia nigra of mid brain in PD. Moreover, these diverse processes that result in PD make modeling of the disease and evaluation of therapeutics against this devastating disease difficult. Here, we will discuss diverse mechanisms that are involved in PD, neuroprotective and therapeutic strategies currently in clinical trial or in preclinical stages, and impart views about strategies that are promising to mitigate PD pathology.
Collapse
|
110
|
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and the numbers are projected to double in the next two decades with the increase in the aging population. An important focus of current research is to develop interventions to slow the progression of the disease. However, prerequisites to it include the development of reliable biomarkers for early diagnosis which would identify at-risk groups and disease progression. In this review, we present updated evidence of already known clinical biomarkers (such as hyposmia and rapid eye movement (REM) sleep behavior disorder (RBD)) and neuroimaging biomarkers, as well as newer possible markers in the blood, CSF, and other tissues. While several promising candidates and methods to assess these biomarkers are on the horizon, it is becoming increasingly clear that no one candidate will clearly fulfill all the roles as a single biomarker. A multimodal and combinatorial approach to develop a battery of biomarkers will likely be necessary in the future.
Collapse
Affiliation(s)
- Shyamal H Mehta
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic College of Medicine, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| | - Charles H Adler
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic College of Medicine, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
| |
Collapse
|
111
|
Sherzai AZ, Tagliati M, Park K, Gatto NM, Pezeshkian S, Sherzai D. Micronutrients and Risk of Parkinson's Disease: A Systematic Review. Gerontol Geriatr Med 2016; 2:2333721416644286. [PMID: 28138496 PMCID: PMC5119866 DOI: 10.1177/2333721416644286] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder. Although the precise pathogenetic mechanisms of PD remain undetermined, there appears to be both genetic and environmental factors that contribute to the risk of developing PD. With regard to environmental risk factors, there has been significant interest related to the role of diet, nutrition, and nutrients on the onset and progression of PD. As the current treatments are predominantly focused on symptomatic management, efforts must be directed toward prevention of the PD and identification of potentially modifiable risk and preventive factors. This comprehensive review gives an overview of studies examining the role of micronutrients in PD, and provides guidance on the value of the reported outcomes.
Collapse
Affiliation(s)
- Ayesha Z Sherzai
- Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michele Tagliati
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Katherine Park
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | | | - Shant Pezeshkian
- Loma Linda University School of Public Health, Loma Linda, CA, USA
| | - Dean Sherzai
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
112
|
Martino R, Candundo H, Lieshout PV, Shin S, Crispo JAG, Barakat-Haddad C. Onset and progression factors in Parkinson's disease: A systematic review. Neurotoxicology 2016; 61:132-141. [PMID: 27058967 DOI: 10.1016/j.neuro.2016.04.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 12/18/2022]
Abstract
Current research has identified several factors thought to be associated with the onset and progression of Parkinson's Disease (PD); however, whether certain factors contribute to or are protective against PD remains unclear. As such, a systematic search of the literature was performed using variations of MeSH and keyword search terms to identify and summarize systematic reviews and primary studies pertaining to factors associated with the onset and progression of PD. Factors referred to both traditional risk factors and prodromal markers. The following databases were searched: MEDLINE, MEDLINE In-Process, EMBASE, PsycINFO, Scopus, Web of Science, Cochrane Database of Systematic Reviews, Cumulative Index to Nursing and Allied Health Literature (CINAHL), ProQuest Dissertations & Theses, AARP AgeLine, and PDGene. A quality assessment of included systematic reviews was completed using the validated Assessment of the Methodological Quality of Systematic Reviews (AMSTAR) tool. Data extraction targeted reported factors, risk estimates, and 95% confidence intervals (CI). Findings identified 11 systematic reviews of sufficient quality reporting factors for PD onset, and no systematic reviews reporting factors for PD progression. In addition, 93 primary articles were identified, of which, 89 articles addressed factors related to PD onset and 4 articles addressed factors related to the PD progression. Pesticide exposure, rural living, well-water drinking, and farming occupation were consistently found to be positively associated with the onset of PD. Moreover, family history and polymorphisms to key genes were also found to be positively associated with the onset of PD. Conversely, coffee consumption, cigarette smoking, and some polymorphisms were consistently found to be negatively associated with the onset of PD. Urate was the only identified factor linked to the progression of PD; it was mostly found to be negatively associated with PD. In sum, the evidence was systematically found and summarized in the literature pertaining to factors related to the onset and progression of PD.
Collapse
Affiliation(s)
- Rosemary Martino
- Department of Speech-Language Pathology, University of Toronto, Toronto, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Canada; Health Care and Outcomes Research, Krembil Research Institute, University Health Network, Toronto, Canada.
| | - Hamilton Candundo
- Faculty of Health Sciences, University of Ontario Institute of Technology, Toronto, Canada
| | - Pascal van Lieshout
- Department of Speech-Language Pathology, University of Toronto, Toronto, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Canada; Department of Psychology, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
| | - Sabina Shin
- Faculty of Health Sciences, University of Ontario Institute of Technology, Toronto, Canada
| | - James A G Crispo
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| | | |
Collapse
|
113
|
Sampat R, Young S, Rosen A, Bernhard D, Millington D, Factor S, Jinnah HA. Potential mechanisms for low uric acid in Parkinson disease. J Neural Transm (Vienna) 2016; 123:365-70. [PMID: 26747026 PMCID: PMC5912672 DOI: 10.1007/s00702-015-1503-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/27/2015] [Indexed: 12/25/2022]
Abstract
Several epidemiologic studies have described an association between low serum uric acid (UA) and Parkinson disease (PD). Uric acid is a known antioxidant, and one proposed mechanism of neurodegeneration in PD is oxidative damage of dopamine neurons. However, other complex metabolic pathways may contribute. The purpose of this study is to elucidate potential mechanisms of low serum UA in PD. Subjects who met diagnostic criteria for definite or probable PD (n = 20) and controls (n = 20) aged 55-80 years were recruited. Twenty-four hour urine samples were collected from all participants, and both uric acid and allantoin were measured and corrected for body mass index (BMI). Urinary metabolites were compared using a twoway ANOVA with diagnosis and sex as the explanatory variables. There were no significant differences between PD and controls for total UA (p = 0.60), UA corrected for BMI (p = 0.37), or in the interaction of diagnosis and sex on UA (p = 0.24). Similarly, there were no significant differences between PD and controls for allantoin (p = 0.47), allantoin corrected for BMI (p = 0.57), or in the interaction of diagnosis and sex on allantoin (p = 0.78). Allantoin/UA ratios also did not significantly differ by diagnosis (p = 0.99). Our results imply that low serum UA in PD may be due to an intrinsic mechanism that alters the homeostatic set point for serum UA in PD, and may contribute to relatively lower protection against oxidative damage. These findings provide indirect support for neuroprotection trials aimed at raising serum UA.
Collapse
Affiliation(s)
- Radhika Sampat
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA.
| | - Sarah Young
- Department of Biochemistry, Duke University, Durham, NC, 27713, USA
| | - Ami Rosen
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Douglas Bernhard
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - David Millington
- Department of Biochemistry, Duke University, Durham, NC, 27713, USA
| | - Stewart Factor
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - H A Jinnah
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA.
- Department of Human Genetics and Pediatrics, Emory University, Suite 6300 Woodruff Memorial Building, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
114
|
Wills AMA, Pérez A, Wang J, Su X, Morgan J, Rajan SS, Leehey MA, Pontone GM, Chou KL, Umeh C, Mari Z, Boyd J. Association Between Change in Body Mass Index, Unified Parkinson's Disease Rating Scale Scores, and Survival Among Persons With Parkinson Disease: Secondary Analysis of Longitudinal Data From NINDS Exploratory Trials in Parkinson Disease Long-term Study 1. JAMA Neurol 2016; 73:321-8. [PMID: 26751506 PMCID: PMC5469290 DOI: 10.1001/jamaneurol.2015.4265] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Greater body mass index (BMI, calculated as weight in kilograms divided by height in meters squared) is associated with improved survival among persons with Huntington disease or amyotrophic lateral sclerosis. Weight loss is common among persons with Parkinson disease (PD) and is associated with worse quality of life. OBJECTIVE To explore the association between change in BMI, Unified Parkinson's Disease Rating Scale (UPDRS) motor and total scores, and survival among persons with PD and to test whether there is a positive association between BMI at randomization and survival. DESIGN, SETTING, AND PARTICIPANTS Secondary analysis (from May 27, 2014, to October 13, 2015) of longitudinal data (3-6 years) from 1673 participants who started the National Institute of Neurological Disorders and Stroke Exploratory Trials in PD Long-term Study-1 (NET-PD LS-1). This was a double-blind randomized placebo-controlled clinical trial of creatine monohydrate (10 g/d) that was performed at 45 sites throughout the United States and Canada. Participants with early (within 5 years of diagnosis) and treated (receiving dopaminergic therapy) PD were enrolled from March 2007 to May 2010 and followed up until September 2013. MAIN OUTCOMES AND MEASURES Change across time in motor UPDRS score, change across time in total UPDRS score, and time to death. Generalized linear mixed models were used to estimate the effect of BMI on the change in motor and total UPDRS scores after controlling for covariates. Survival was analyzed using Cox proportional hazards models of time to death. A participant's BMI was measured at randomization, and BMI trajectory groups were classified according to whether participants experienced weight loss ("decreasing BMI"), weight stability ("stable BMI"), or weight gain ("increasing BMI") during the study. RESULTS Of the 1673 participants (mean [SD] age, 61.7 [9.6] years; 1074 [64.2%] were male), 158 (9.4%) experienced weight loss (decreasing BMI), whereas 233 (13.9%) experienced weight gain (increasing BMI). After adjusting for covariates, we found that the weight-loss group's mean (SE) motor UPDRS score increased by 1.48 (0.28) (P < .001) more points per visit than the weight-stable group's mean (SE) motor UPDRS score. The weight-gain group's mean (SE) motor UPDRS score decreased by -0.51 (0.24) (P = .03) points per visit, relative to the weight-stable group. While there was an unadjusted difference in survival between the 3 BMI trajectory groups (log-rank P < .001), this was not significant after adjusting for covariates. CONCLUSIONS AND RELEVANCE Change in BMI was inversely associated with change in motor and total UPDRS scores in the NET-PD LS-1. Change in BMI was not associated with survival; however, these results were limited by the low number of deaths in the NET-PD LS-1. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00449865.
Collapse
Affiliation(s)
- Anne-Marie A Wills
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Adriana Pérez
- Department of Biostatistics, The University of Texas Health Science Center at Houston UTHealth, School of Public Health, Austin
| | - Jue Wang
- UTHealth, The University of Texas School of Public Health, Houston
| | - Xiao Su
- UTHealth, The University of Texas School of Public Health, Houston
| | - John Morgan
- Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta
| | - Suja S Rajan
- Department of Management, Policy and Community Health, The University of Texas Health Science Center at Houston UTHealth, School of Public Health, Houston
| | - Maureen A Leehey
- Department of Neurology, University of Colorado Hospital and University of Colorado School of Medicine, Aurora
| | - Gregory M Pontone
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Kelvin L Chou
- Departments of Neurology and Neurosurgery, University of Michigan, Ann Arbor
| | - Chizoba Umeh
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zoltan Mari
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - James Boyd
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington
| |
Collapse
|
115
|
Santos-García D, Mir P, Cubo E, Vela L, Rodríguez-Oroz MC, Martí MJ, Arbelo JM, Infante J, Kulisevsky J, Martínez-Martín P. COPPADIS-2015 (COhort of Patients with PArkinson's DIsease in Spain, 2015), a global--clinical evaluations, serum biomarkers, genetic studies and neuroimaging--prospective, multicenter, non-interventional, long-term study on Parkinson's disease progression. BMC Neurol 2016; 16:26. [PMID: 26911448 PMCID: PMC4766717 DOI: 10.1186/s12883-016-0548-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/19/2016] [Indexed: 12/19/2022] Open
Abstract
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder causing motor and non-motor symptoms that can affect independence, social adjustment and the quality of life (QoL) of both patients and caregivers. Studies designed to find diagnostic and/or progression biomarkers of PD are needed. We describe here the study protocol of COPPADIS-2015 (COhort of Patients with PArkinson’s DIsease in Spain, 2015), an integral PD project based on four aspects/concepts: 1) PD as a global disease (motor and non-motor symptoms); 2) QoL and caregiver issues; 3) Biomarkers; 4) Disease progression. Methods/design Observational, descriptive, non-interventional, 5-year follow-up, national (Spain), multicenter (45 centers from 15 autonomous communities), evaluation study. Specific goals: (1) detailed study (clinical evaluations, serum biomarkers, genetic studies and neuroimaging) of a population of PD patients from different areas of Spain, (2) comparison with a control group and (3) follow-up for 5 years. COPPADIS-2015 has been specifically designed to assess 17 proposed objectives. Study population: approximately 800 non-dementia PD patients, 600 principal caregivers and 400 control subjects. Study evaluations: (1) baseline includes motor assessment (e.g., Unified Parkinson’s Disease Rating Scale part III), non-motor symptoms (e.g., Non-Motor Symptoms Scale), cognition (e.g., Parkinson’s Disease Cognitive Rating Scale), mood and neuropsychiatric symptoms (e.g., Neuropsychiatric Inventory), disability, QoL (e.g., 39-item Parkinson’s disease Quality of Life Questionnaire Summary-Index) and caregiver status (e.g., Zarit Caregiver Burden Inventory); (2) follow-up includes annual (patients) or biannual (caregivers and controls) evaluations. Serum biomarkers (S-100b protein, TNF-α, IL-1, IL-2, IL-6, vitamin B12, methylmalonic acid, homocysteine, uric acid, C-reactive protein, ferritin, iron) and brain MRI (volumetry, tractography and MTAi [Medial Temporal Atrophy Index]), at baseline and at the end of follow-up, and genetic studies (DNA and RNA) at baseline will be performed in a subgroup of subjects (300 PD patients and 100 control subjects). Study periods: (1) recruitment period, from November, 2015 to February, 2017 (basal assessment); (2) follow-up period, 5 years; (3) closing date of clinical follow-up, May, 2022. Funding: Public/Private. Discussion COPPADIS-2015 is a challenging initiative. This project will provide important information on the natural history of PD and the value of various biomarkers.
Collapse
Affiliation(s)
- Diego Santos-García
- Sección de Neurología, Complejo Hospitalario Universitario de Ferrol (CHUF), Hospital Arquitecto Marcide, c/Avenida La Residencia, s/n, 15405, Ferrol, Spain.
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, CSIC y Universidad de Sevilla, Sevilla, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sevilla, Spain.
| | - Esther Cubo
- Servicio de Neurología, Hospital Universitario de Burgos, Burgos, Spain.
| | - Lydia Vela
- Unidad de Neurología, Fundación Hospital de Alcorcón, Madrid, Spain.
| | | | - Maria José Martí
- Unidad de Parkinson y Trastornos del Movimiento, Servicio de Neurología, Instituto Clínico de Neurociencias, Hospital Clínic, Barcelona, Spain.
| | - José Matías Arbelo
- Unidad de Trastornos del Movimiento y enfermedad de Parkinson, Servicio de Neurología, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Jon Infante
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital Universitario Marqués de Valdecilla, Santander, Spain.
| | - Jaime Kulisevsky
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital de Sant Pau, Barcelona, Spain.
| | - Pablo Martínez-Martín
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sevilla, Spain. .,Centro Nacional de Epidemiología, Instituto de Salud Carlos III, Madrid, Spain.
| | | |
Collapse
|
116
|
Jackson EK, Boison D, Schwarzschild MA, Kochanek PM. Purines: forgotten mediators in traumatic brain injury. J Neurochem 2016; 137:142-53. [PMID: 26809224 DOI: 10.1111/jnc.13551] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/12/2022]
Abstract
Recently, the topic of traumatic brain injury has gained attention in both the scientific community and lay press. Similarly, there have been exciting developments on multiple fronts in the area of neurochemistry specifically related to purine biology that are relevant to both neuroprotection and neurodegeneration. At the 2105 meeting of the National Neurotrauma Society, a session sponsored by the International Society for Neurochemistry featured three experts in the field of purine biology who discussed new developments that are germane to both the pathomechanisms of secondary injury and development of therapies for traumatic brain injury. This included presentations by Drs. Edwin Jackson on the novel 2',3'-cAMP pathway in neuroprotection, Detlev Boison on adenosine in post-traumatic seizures and epilepsy, and Michael Schwarzschild on the potential of urate to treat central nervous system injury. This mini review summarizes the important findings in these three areas and outlines future directions for the development of new purine-related therapies for traumatic brain injury and other forms of central nervous system injury. In this review, novel therapies based on three emerging areas of adenosine-related pathobiology in traumatic brain injury (TBI) were proposed, namely, therapies targeting 1) the 2',3'-cyclic adenosine monophosphate (cAMP) pathway, 2) adenosine deficiency after TBI, and 3) augmentation of urate after TBI.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, Oregon, USA
| | - Michael A Schwarzschild
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
117
|
Brody DM, Litvan I, Warner S, Riley DE, Hall DA, Kluger BM, Shprecher DR, Cunningham CR. Relationship between uric acid levels and progressive supranuclear palsy. Mov Disord 2016; 31:663-7. [PMID: 26890571 DOI: 10.1002/mds.26535] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 10/29/2015] [Accepted: 11/29/2015] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION The pathophysiology of both PD and PSP is characterized by a pro-oxidant state. Uric acid is an oxidative stress marker. High uric acid blood levels have been associated with a reduced risk of PD and a decreased rate of disease progression. We investigated whether a low serum concentration of uric acid is also associated with PSP. METHODS We measured serum uric acid concentrations in a subsample of the ENGENE PSP Cohort that included 75 cases and 75 frequency-matched-by-sex healthy controls (69 spouses, 6 in-laws) from four centers willing to participate (Case Western, Rush University, University of Utah, and University of Louisville). Case severity was characterized using the total PSP-Rating Scale, UPDRS, and Mattis Dementia Rating Scale. Unconditional logistic regression, Pearson's chi-squared test, and analysis of variance were used, as appropriate. RESULTS The mean uric acid level among cases (4.0 mg/dL) was not significantly lower than that of controls (4.1 mg/dL). When controlling for sex, there were no between-group statistical differences in uric acid levels. Uric acid levels were not correlated with disease severity. CONCLUSIONS The results of this study do not provide evidence of uric acid having a protective role in PSP, even if oxidative injury is important in the pathophysiology of this disorder. The lack of statistical significance suggests that there is no direct association between uric acid levels and PSP. However, a small inverse association cannot be excluded. © 2016 Movement Disorder Society.
Collapse
Affiliation(s)
- David M Brody
- Movement Disorder Center, Department of Neurosciences, University of California San Diego, San Diego, California, USA
| | - Irene Litvan
- Movement Disorder Center, Department of Neurosciences, University of California San Diego, San Diego, California, USA.,Division of Movement Disorders, Department of Neurology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | | | | | - Deborah A Hall
- Department of Neurology, Rush University Medical Center, Chicago, Illinois, USA
| | - Benzi M Kluger
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Christopher R Cunningham
- Division of Movement Disorders, Department of Neurology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
118
|
Algarni MA, Stoessl AJ. The role of biomarkers and imaging in Parkinson’s disease. Expert Rev Neurother 2016; 16:187-203. [DOI: 10.1586/14737175.2016.1135056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
119
|
Bhattacharyya S, Bakshi R, Logan R, Ascherio A, Macklin EA, Schwarzschild MA. Oral Inosine Persistently Elevates Plasma antioxidant capacity in Parkinson's disease. Mov Disord 2016; 31:417-21. [DOI: 10.1002/mds.26483] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/06/2015] [Accepted: 10/18/2015] [Indexed: 11/08/2022] Open
Affiliation(s)
- Shamik Bhattacharyya
- Department of Neurology, Molecular Neurobiology Lab; Massachusetts General Hospital; Boston Massachusetts USA
- Departments of Neurology; Brigham and Women's Hospital; Boston Massachusetts USA
- Harvard Medical School; Boston Massachusetts USA
| | - Rachit Bakshi
- Department of Neurology, Molecular Neurobiology Lab; Massachusetts General Hospital; Boston Massachusetts USA
- Harvard Medical School; Boston Massachusetts USA
| | - Robert Logan
- Department of Neurology, Molecular Neurobiology Lab; Massachusetts General Hospital; Boston Massachusetts USA
| | - Alberto Ascherio
- Departments of Epidemiology and Nutrition; Harvard School of Public Health; Boston Massachusetts USA
- Harvard Medical School; Boston Massachusetts USA
| | - Eric A. Macklin
- Department of Medicine, Biostatistics Center; Massachusetts General Hospital; Boston Massachusetts USA
- Harvard Medical School; Boston Massachusetts USA
| | - Michael A. Schwarzschild
- Department of Neurology, Molecular Neurobiology Lab; Massachusetts General Hospital; Boston Massachusetts USA
- Harvard Medical School; Boston Massachusetts USA
| |
Collapse
|
120
|
Gao X, O'Reilly ÉJ, Schwarzschild MA, Ascherio A. Prospective study of plasma urate and risk of Parkinson disease in men and women. Neurology 2016; 86:520-6. [PMID: 26764029 DOI: 10.1212/wnl.0000000000002351] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/15/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To examine whether higher plasma urate concentrations are associated with a lower risk of developing Parkinson disease (PD) and whether there is a sex difference in the potential urate-PD relationship. METHODS We conducted a nested case-control study based on 90,214 participants of 3 ongoing US cohorts. We identified 388 new PD cases (202 men and 186 women) since blood collection, which were then matched to 1,267 controls. PD cases were confirmed by medical record review. Conditional logistic regression estimated relative risks (RRs) and 95% confidence intervals (95% CIs), after adjustment for age, smoking, caffeine intake, plasma concentrations of cholesterol and ferritin, and other covariates. We also conducted a meta-analysis to combine our study with 3 previously published prospective studies on urate and PD risk. RESULTS In the present nested case-control study, the multivariate-adjusted RRs of PD comparing extreme quartiles of urate were 0.63 (95% CI 0.35, 1.10; ptrend = 0.049) in men and 1.04 (95% CI 0.61, 1.78; ptrend = 0.44) in women (pheterogeneity = 0.001). In the meta-analysis, the pooled RRs comparing 2 extreme quartiles of urate were 0.63 (95% CI 0.42, 0.95) in men and 0.89 (95% CI 0.57, 1.40) in women. CONCLUSION We observed that men, but not women, with higher urate concentrations had a lower future risk of developing PD, suggesting that urate could be protective against PD risk or could slow disease progression during the preclinical stage of disease.
Collapse
Affiliation(s)
- Xiang Gao
- From the Department of Nutritional Health (X.G.), The Pennsylvania State University, University Park; Department of Nutrition (E.J.O., A.A.), Harvard School of Public Health; Channing Division of Network Medicine (E.J.O., A.A.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; and Department of Neurology (M.A.S.), Massachusetts General Hospital and Harvard Medical School, Boston.
| | - Éilis J O'Reilly
- From the Department of Nutritional Health (X.G.), The Pennsylvania State University, University Park; Department of Nutrition (E.J.O., A.A.), Harvard School of Public Health; Channing Division of Network Medicine (E.J.O., A.A.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; and Department of Neurology (M.A.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Michael A Schwarzschild
- From the Department of Nutritional Health (X.G.), The Pennsylvania State University, University Park; Department of Nutrition (E.J.O., A.A.), Harvard School of Public Health; Channing Division of Network Medicine (E.J.O., A.A.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; and Department of Neurology (M.A.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Alberto Ascherio
- From the Department of Nutritional Health (X.G.), The Pennsylvania State University, University Park; Department of Nutrition (E.J.O., A.A.), Harvard School of Public Health; Channing Division of Network Medicine (E.J.O., A.A.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; and Department of Neurology (M.A.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
121
|
Nazeri A, Roostaei T, Sadaghiani S, Chakravarty MM, Eberly S, Lang AE, Voineskos AN. Genome-wide variant by serum urate interaction in Parkinson's disease. Ann Neurol 2015; 78:731-41. [PMID: 26284320 DOI: 10.1002/ana.24504] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 08/04/2015] [Accepted: 08/04/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Serum urate levels have been associated with risk for and progression of Parkinson's disease (PD). Urate-related compounds are therapeutic candidates in neuroprotective efforts to slow PD progression. A urate-elevating agent is currently under investigation as a potential disease-modifying strategy in people with PD. However, PD is a heterogeneous disorder, and genetic variation may explain divergence in disease severity and progression. METHODS We conducted a genome-wide association study to identify gene variant × serum urate interaction effects on the striatal (123) I-ioflupane (DaTscan) binding ratio measured using single photon emission computed tomography in patients with possible PD from the Parkinson's Progression Markers Initiative (PPMI, n = 360). Follow-up analyses were conducted to assess gene variant × serum urate interaction effects on magnetic resonance imaging-derived regional brain volumes and clinical status. We then attempted to replicate our primary analysis in patients who entered the Parkinson Research Examination of CEP-1347 Trial (PRECEPT) with a clinical diagnosis of PD (n = 349). RESULTS Rs1109303 (T>G) variant within the INPP5K gene on chromosome 17p13.3 demonstrated a genome-wide significant interaction with serum urate level to predict striatal dopamine transporter density among all PPMI participants (n = 359) with possible PD (p = 2.01 × 10(-8) ; after excluding participants with SWEDD [scan without evidence of dopaminergic deficit]: p = 1.12 × 10(-9) ; n = 316). Independent of striatal dopamine transporter density, similar effects on brain atrophy, bradykinesia, anxiety, and depression were observed. No effect was present in the PRECEPT sample at baseline; however, in non-SWEDD PD participants in PRECEPT (n = 309), we observed a significant longitudinal genotype × serum urate interaction effect, consistent in direction with the PPMI sample, on progression of striatal dopamine transporter density over the 22-month follow-up. INTERPRETATION Genetic profile combined with serum urate level can be used to predict disease severity and potential disease progression in patients with PD. These results may be relevant to therapeutic efforts targeting the urate pathway.
Collapse
Affiliation(s)
- Arash Nazeri
- Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Tina Roostaei
- Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Shokufeh Sadaghiani
- Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Institute, Montreal, Quebec, Canada.,Departments of Psychiatry and Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Shirley Eberly
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY
| | - Anthony E Lang
- Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, University Health Network, Division of Neurology, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Aristotle N Voineskos
- Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Underserved Populations Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
122
|
Chen X, Xie C, Sun L, Ding J, Cai H. Longitudinal Metabolomics Profiling of Parkinson's Disease-Related α-Synuclein A53T Transgenic Mice. PLoS One 2015; 10:e0136612. [PMID: 26317866 PMCID: PMC4552665 DOI: 10.1371/journal.pone.0136612] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022] Open
Abstract
Metabolic homeostasis is critical for all biological processes in the brain. The metabolites are considered the best indicators of cell states and their rapid fluxes are extremely sensitive to cellular changes. While there are a few studies on the metabolomics of Parkinson's disease, it lacks longitudinal studies of the brain metabolic pathways affected by aging and the disease. Using ultra-high performance liquid chromatography and tandem mass spectroscopy (UPLC/MS), we generated the metabolomics profiling data from the brains of young and aged male PD-related α-synuclein A53T transgenic mice as well as the age- and gender-matched non-transgenic (nTg) controls. Principal component and unsupervised hierarchical clustering analyses identified distinctive metabolites influenced by aging and the A53T mutation. The following metabolite set enrichment classification revealed the alanine metabolism, redox and acetyl-CoA biosynthesis pathways were substantially disturbed in the aged mouse brains regardless of the genotypes, suggesting that aging plays a more prominent role in the alterations of brain metabolism. Further examination showed that the interaction effect of aging and genotype only disturbed the guanosine levels. The young A53T mice exhibited lower levels of guanosine compared to the age-matched nTg controls. The guanosine levels remained constant between the young and aged nTg mice, whereas the aged A53T mice showed substantially increased guanosine levels compared to the young mutant ones. In light of the neuroprotective function of guanosine, our findings suggest that the increase of guanosine metabolism in aged A53T mice likely represents a protective mechanism against neurodegeneration, while monitoring guanosine levels could be applicable to the early diagnosis of the disease.
Collapse
Affiliation(s)
- Xi Chen
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Chengsong Xie
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Lixin Sun
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Jinhui Ding
- Bioinformatics Core, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Huaibin Cai
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| |
Collapse
|
123
|
Swanson CR, Berlyand Y, Xie SX, Alcalay RN, Chahine LM, Chen-Plotkin AS. Plasma apolipoprotein A1 associates with age at onset and motor severity in early Parkinson's disease patients. Mov Disord 2015. [PMID: 26207725 DOI: 10.1002/mds.26290] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Development of robust plasma-based biomarkers in Parkinson's disease (PD) could lead to new approaches for identifying those at risk for PD and developing novel therapies. Here, we validate plasma apolipoprotein A1 (ApoA1) as a correlate of age at onset and motor severity in PD. METHODS Plasma ApoA1 and high-density lipoprotein at baseline, 6 months, and 12 months were measured in 254 research volunteers (154 patients with PD and 100 normal controls) enrolled in the Parkinson's Progression Markers Initiative (PPMI) study. RESULTS Lower baseline plasma ApoA1 levels associate with an earlier age at PD onset in early-stage, drug-naïve PPMI PD patients (P = 0.023). Moreover, lower baseline ApoA1 levels trend toward association with worse motor severity in PPMI PD patients (p = 0.080). Over 12 months of follow-up, plasma ApoA1 levels do not predict motor decline in the PPMI PD cohort. Finally, a meta-analysis of five PD cohorts encompassing >1,000 patients confirms significant association of lower plasma ApoA1 with earlier age at PD onset (P < 0.001) and greater motor severity (P < 0.001). CONCLUSIONS Our results confirm the previously reported association of lower plasma ApoA1 levels with two clinical features suggesting poorer dopaminergic system integrity-earlier age at PD onset and greater motor severity-in early-stage, drug-naïve PD patients. This is the first report of a plasma-based biomarker evaluated in the PPMI study. Future investigations are warranted evaluating plasma ApoA1 as a longitudinal correlate of disease progression as well as investigating the potential of ApoA1 as a therapeutic target in PD.
Collapse
Affiliation(s)
- Christine R Swanson
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yosef Berlyand
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sharon X Xie
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Lama M Chahine
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
124
|
Rosas HD, Doros G, Bhasin S, Thomas B, Gevorkian S, Malarick K, Matson W, Hersch SM. A systems-level "misunderstanding": the plasma metabolome in Huntington's disease. Ann Clin Transl Neurol 2015; 2:756-68. [PMID: 26273688 PMCID: PMC4531058 DOI: 10.1002/acn3.214] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/10/2015] [Accepted: 04/11/2015] [Indexed: 12/17/2022] Open
Abstract
Objective Huntington’s disease (HD) is a rare neurodegenerative disease caused by the expansion of an N-terminal repeat in the huntingtin protein. The protein is expressed in all cells in the body; hence, peripheral tissues, such as blood, may recapitulate processes in the brain. The plasma metabolome may provide a window into active processes that influence brain health and a unique opportunity to noninvasively identify processes that may contribute to neurodegeneration. Alterations in metabolic pathways in brain have been shown to profoundly impact HD. Therefore, identification and quantification of critical metabolomic perturbations could provide novel biomarkers for disease onset and disease progression. Methods We analyzed the plasma metabolomic profiles from 52 premanifest (PHD), 102 early symptomatic HD, and 140 healthy controls (NC) using liquid chromatography coupled with a highly sensitive electrochemical detection platform. Results Alterations in tryptophan, tyrosine, purine, and antioxidant pathways were identified, including many related to energetic and oxidative stress and derived from the gut microbiome. Multivariate statistical modeling demonstrated mutually distinct metabolomic profiles, suggesting that the processes that determine onset were likely distinct from those that determine progression. Gut microbiome-derived metabolites particularly differentiated the PHD metabolome, while the symptomatic HD metabolome was increasingly influenced by metabolites that may reflect mutant huntingtin toxicity and neurodegeneration. Interpretation Understanding the complex changes in the delicate balance of the metabolome and the gut microbiome in HD, and how they relate to disease onset, progression, and phenotypic variability in HD are critical questions for future research.
Collapse
Affiliation(s)
- Herminia D Rosas
- Department of Neurology Boston, Massachusetts ; Center for Neuro-imaging of Aging and Neurodegenerative Diseases Boston, Massachusetts ; Athinoula A. Martinos Center for Biomedical Imaging Charlestown, Massachusetts ; Radiology, Massachusetts General Hospital and Harvard Medical School Boston, Massachusetts
| | - Gheorghe Doros
- Department of Biostatistics, School of Public Health, Boston University Boston, Massachusetts
| | - Swati Bhasin
- Edith Nourse Rogers Memorial Veterans Hospital Bedford, Massachusetts
| | - Beena Thomas
- Edith Nourse Rogers Memorial Veterans Hospital Bedford, Massachusetts
| | - Sona Gevorkian
- Department of Neurology Boston, Massachusetts ; Center for Neuro-imaging of Aging and Neurodegenerative Diseases Boston, Massachusetts ; Athinoula A. Martinos Center for Biomedical Imaging Charlestown, Massachusetts
| | - Keith Malarick
- Department of Neurology Boston, Massachusetts ; Center for Neuro-imaging of Aging and Neurodegenerative Diseases Boston, Massachusetts ; Athinoula A. Martinos Center for Biomedical Imaging Charlestown, Massachusetts
| | - Wayne Matson
- Edith Nourse Rogers Memorial Veterans Hospital Bedford, Massachusetts
| | - Steven M Hersch
- Department of Neurology Boston, Massachusetts ; MassGeneral Institutes for Neurodegenerative Disease, Laboratory of Neurodegeneration and Neurotherapeutics, Boston University Boston, Massachusetts
| |
Collapse
|
125
|
Clinically meaningful parameters of progression and long-term outcome of Parkinson disease: An international consensus statement. Parkinsonism Relat Disord 2015; 21:675-82. [PMID: 25952959 DOI: 10.1016/j.parkreldis.2015.04.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/09/2015] [Accepted: 04/27/2015] [Indexed: 02/03/2023]
Abstract
Parkinson disease (PD) is associated with a clinical course of variable duration, severity, and a combination of motor and non-motor features. Recent PD research has focused primarily on etiology rather than clinical progression and long-term outcomes. For the PD patient, caregivers, and clinicians, information on expected clinical progression and long-term outcomes is of great importance. Today, it remains largely unknown what factors influence long-term clinical progression and outcomes in PD; recent data indicate that the factors that increase the risk to develop PD differ, at least partly, from those that accelerate clinical progression and lead to worse outcomes. Prospective studies will be required to identify factors that influence progression and outcome. We suggest that data for such studies is collected during routine office visits in order to guarantee high external validity of such research. We report here the results of a consensus meeting of international movement disorder experts from the Genetic Epidemiology of Parkinson's Disease (GEO-PD) consortium, who convened to define which long-term outcomes are of interest to patients, caregivers and clinicians, and what is presently known about environmental or genetic factors influencing clinical progression or long-term outcomes in PD. We propose a panel of rating scales that collects a significant amount of phenotypic information, can be performed in the routine office visit and allows international standardization. Research into the progression and long-term outcomes of PD aims at providing individual prognostic information early, adapting treatment choices, and taking specific measures to provide care optimized to the individual patient's needs.
Collapse
|
126
|
Lu N, Dubreuil M, Zhang Y, Neogi T, Rai SK, Ascherio A, Hernán MA, Choi HK. Gout and the risk of Alzheimer's disease: a population-based, BMI-matched cohort study. Ann Rheum Dis 2015; 75:547-51. [PMID: 25739830 DOI: 10.1136/annrheumdis-2014-206917] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/13/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE While gout is associated with cardiovascular (CV)-metabolic comorbidities and their sequelae, the antioxidant effects of uric acid may have neuroprotective benefits. We evaluated the potential impact of incident gout on the risk of developing Alzheimer's disease (AD) in a general population context. METHODS We conducted an age-matched, sex-matched, entry-time-matched and body mass index (BMI)-matched cohort study using data from The Health Improvement Network, an electronic medical record database representative of the UK general population, from 1 January 1995 to 31 December 2013. Up to five non-gout individuals were matched to each case of incident gout by age, sex, year of enrolment and BMI. We compared incidence rates of AD between the gout and comparison cohorts, excluding individuals with prevalent gout or dementia at baseline. Multivariate hazard ratios (HRs) were calculated, while adjusting for smoking, alcohol use, physician visits, social deprivation index, comorbidities and medication use. We repeated the same analysis among patients with incident osteoarthritis (OA) as a negative control exposure. RESULTS We identified 309 new cases of AD among 59 224 patients with gout (29% female, mean age 65 years) and 1942 cases among 238 805 in the comparison cohort over a 5-year median follow up (1.0 vs 1.5 per 1000 person-years, respectively). Univariate (age-matched, sex-matched, entry-time-matched and BMI-matched) and multivariate HRs for AD among patients with gout were 0.71 (95% CI 0.62 to 0.80) and 0.76 (95% CI 0.66 to 0.87), respectively. The inverse association persisted among subgroups stratified by sex, age group (<75 and ≥75 years), social deprivation index and history of CV disease. The association between incident OA and the risk of incident AD was null. CONCLUSIONS These findings provide the first general population-based evidence that gout is inversely associated with the risk of developing AD, supporting the purported potential neuroprotective role of uric acid.
Collapse
Affiliation(s)
- Na Lu
- Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Massachusetts, USA Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maureen Dubreuil
- Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Massachusetts, USA Section of Rheumatology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yuqing Zhang
- Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Tuhina Neogi
- Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sharan K Rai
- Arthritis Research Centre of Canada, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alberto Ascherio
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Miguel A Hernán
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Hyon K Choi
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
127
|
Moccia M, Pappatà S, Erro R, Picillo M, Vitale C, Amboni M, Longo K, Palladino R, Barone P, Pellecchia MT. Uric acid relates to dopamine transporter availability in Parkinson's disease. Acta Neurol Scand 2015; 131:127-31. [PMID: 25288358 DOI: 10.1111/ane.12295] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND Diagnosing Parkinson's disease (PD) and tracking its progression may require the combination of reliable biomarkers. Among them, both serum uric acid (UA) and dopamine transporter (DaT) binding deserve more investigations. AIMS OF THE STUDY We aimed to investigate the relationship between serum UA levels and DaT availability in newly diagnosed, drug-naïve PD patients, by means of semiquantitative [(123) I]FP-CIT-SPECT. METHODS We recruited 52 newly diagnosed, drug-naïve PD patients, and performed serum UA dosage and [(123) I]FP-CIT-SPECT. RESULTS Pearson's correlation analysis showed that UA levels were significantly higher in patients with higher averaged, ipsilateral and contralateral DaT binding in caudate, putamen, and striatum. CONCLUSIONS We showed, for the first time, by regional semiquantitative analysis of DaT binding in PD patients that UA levels significantly correlates with the severity of dopaminergic impairment in caudate, putamen, and striatum. This study broadens our knowledge on the importance of UA as a biomarker of PD.
Collapse
Affiliation(s)
- M. Moccia
- Department of Neuroscience, Reproductive Science and Odontostomatology; Federico II University; Naples Italy
| | - S. Pappatà
- Institute of Biostructure and Bioimaging; CNR; Naples Italy
| | - R. Erro
- Sobell Department of Motor Neuroscience and Movement Disorders; University College London (UCL), Institute of Neurology; London UK
- Dipartimento di Scienze Neurologiche e del Movimento; Università di Verona, Policlinico Borgo Roma; Verona Italy
| | - M. Picillo
- Department of Neuroscience, Reproductive Science and Odontostomatology; Federico II University; Naples Italy
| | - C. Vitale
- University of Naples Parthenope; Naples Italy
- IDC Hermitage-Capodimonte; Naples Italy
| | - M. Amboni
- IDC Hermitage-Capodimonte; Naples Italy
| | - K. Longo
- IDC Hermitage-Capodimonte; Naples Italy
| | - R. Palladino
- Department of Primary Care and Public Health; Imperial College; London UK
- Department of Public Health; Federico II University; Naples Italy
| | - P. Barone
- Center for Neurodegenerative Diseases (CEMAND); Neuroscience Section; Department of Medicine; University of Salerno; Salerno Italy
| | - Maria Teresa Pellecchia
- Center for Neurodegenerative Diseases (CEMAND); Neuroscience Section; Department of Medicine; University of Salerno; Salerno Italy
| |
Collapse
|
128
|
Matsuo H, Tomiyama H, Satake W, Chiba T, Onoue H, Kawamura Y, Nakayama A, Shimizu S, Sakiyama M, Funayama M, Nishioka K, Shimizu T, Kaida K, Kamakura K, Toda T, Hattori N, Shinomiya N. ABCG2 variant has opposing effects on onset ages of Parkinson's disease and gout. Ann Clin Transl Neurol 2015; 2:302-6. [PMID: 25815357 PMCID: PMC4369280 DOI: 10.1002/acn3.167] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/30/2014] [Indexed: 11/29/2022] Open
Abstract
Uric acid (urate) has been suggested to play a protective role in Parkinson's disease onset through its antioxidant activity. Dysfunction of ABCG2, a high-capacity urate exporter, is a major cause for early-onset gout based on hyperuricemia. In this study, the effects of a dysfunctional ABCG2 variant (Q141K, rs2231142) were analyzed on the ages at onset of gout patients (N = 507) and Parkinson's disease patients (N = 1015). The Q141K variant hastened the gout onset (P = 0.0027), but significantly associated with later Parkinson's disease onset (P = 0.025). Our findings will be helpful for development of more effective prevention of Parkinson's disease.
Collapse
Affiliation(s)
- Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| | - Hiroyuki Tomiyama
- Department of Neurology, Juntendo University School of Medicine Tokyo, Japan
| | - Wataru Satake
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine Kobe, Japan
| | - Toshinori Chiba
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| | - Hiroyuki Onoue
- Department of Internal Medicine, National Defense Medical College Tokorozawa, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| | - Masayuki Sakiyama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| | - Manabu Funayama
- Department of Neurology, Juntendo University School of Medicine Tokyo, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine Tokyo, Japan
| | | | - Kenichi Kaida
- Department of Internal Medicine, National Defense Medical College Tokorozawa, Japan
| | - Keiko Kamakura
- Department of Internal Medicine, National Defense Medical College Tokorozawa, Japan ; Department of Physical Therapy, School of Health Sciences, Tokyo University of Technology Tokyo, Japan
| | - Tatsushi Toda
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine Kobe, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine Tokyo, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College Tokorozawa, Japan
| |
Collapse
|
129
|
Quik M, Bordia T, Zhang D, Perez XA. Nicotine and Nicotinic Receptor Drugs. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 124:247-71. [DOI: 10.1016/bs.irn.2015.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
130
|
Athauda D, Foltynie T. The ongoing pursuit of neuroprotective therapies in Parkinson disease. Nat Rev Neurol 2014; 11:25-40. [PMID: 25447485 DOI: 10.1038/nrneurol.2014.226] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many agents developed for neuroprotective treatment of Parkinson disease (PD) have shown great promise in the laboratory, but none have translated to positive results in patients with PD. Potential neuroprotective drugs, such as ubiquinone, creatine and PYM50028, have failed to show any clinical benefits in recent high-profile clinical trials. This 'failure to translate' is likely to be related primarily to our incomplete understanding of the pathogenic mechanisms underlying PD, and excessive reliance on data from toxin-based animal models to judge which agents should be selected for clinical trials. Restricted resources inevitably mean that difficult compromises must be made in terms of trial design, and reliable estimation of efficacy is further hampered by the absence of validated biomarkers of disease progression. Drug development in PD dementia has been mostly unsuccessful; however, emerging biochemical, genetic and pathological evidence suggests a link between tau and amyloid-β deposition and cognitive decline in PD, potentially opening up new possibilities for therapeutic intervention. This Review discusses the most important 'druggable' disease mechanisms in PD, as well as the most-promising drugs that are being evaluated for their potential efficiency in treatment of motor and cognitive impairments in PD.
Collapse
Affiliation(s)
- Dilan Athauda
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
131
|
Yang F, Trolle Lagerros Y, Bellocco R, Adami HO, Fang F, Pedersen NL, Wirdefeldt K. Physical activity and risk of Parkinson's disease in the Swedish National March Cohort. ACTA ACUST UNITED AC 2014; 138:269-75. [PMID: 25410713 DOI: 10.1093/brain/awu323] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Physical exercise has been associated with neuroprotective effects in the nigrostriatal dopaminergic system. To examine the impact of physical activity on Parkinson's disease risk prospectively, we followed 43 368 individuals who provided extensive information on physical activity at baseline. We estimated hazard ratios with 95% confidence intervals using Cox proportional hazards regression. During an average of 12.6 years of follow-up, 286 incident Parkinson's disease cases were identified. In males, there was an inverse association with Parkinson's disease for total physical activity (hazard ratio 0.55, 95% confidence interval 0.35-0.87 for medium versus low level), for sum of household, commuting and leisure time exercise (hazard ratio 0.53, 95% confidence interval 0.33-0.85 for high versus low level), and for household and commuting physical activity specifically (hazard ratio 0.50, 95% confidence interval 0.31-0.81 for >6 versus <2 h per week). No association was observed for leisure time exercise or occupational physical activity with Parkinson's disease, among either males or females. Meta-analysis of the present study and five previous prospective studies showed a pooled hazard ratio of 0.66 (95% confidence interval 0.57-0.78) for highest versus lowest physical activity level. Our results indicate that a medium level of physical activity lowers Parkinson's disease risk.
Collapse
Affiliation(s)
- Fei Yang
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ylva Trolle Lagerros
- 2 Department of Medicine, Unit of Clinical Epidemiology, Karolinska Institutet, T2, 17176 Stockholm, Sweden
| | - Rino Bellocco
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 3 Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Hans-Olov Adami
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 4 Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
| | - Fang Fang
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Nancy L Pedersen
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 5 Department of Psychology, University of Southern California, Los Angeles, CA 90089-1061, USA
| | - Karin Wirdefeldt
- 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden 6 Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
132
|
Kim TH, Lee JH. Serum uric acid and nigral iron deposition in Parkinson's disease: a pilot study. PLoS One 2014; 9:e112512. [PMID: 25386854 PMCID: PMC4227693 DOI: 10.1371/journal.pone.0112512] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/06/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Uric acid (UA) is an endogenous antioxidant which is known to reduce oxidative stress and also chelate iron ion. Recent studies have provided evidence that UA may play a neuroprotective role in Parkinson's disease (PD). However, it is unknown whether UA relates to nigral iron deposition, which is a characteristic pathophysiological alteration in PD. The aim of this study was to determine the potential relationship of these two markers in patients with PD. METHODS A total of 30 patients of PD and 25 age- and gender- matched healthy controls underwent 3-Tesla MRI and laboratory tests including serum UA levels. We assessed iron levels by measuring phase shift values using susceptibility-weighted image. Mean phase shift values of the substantia nigra (SN), red nucleus, head of the caudate nucleus, globus pallidus, putamen, thalamus, and frontal white matter were calculated and correlated with serum UA levels. RESULTS Serum UA levels were significantly decreased in the PD patients than in the controls. Phase shift values in bilateral SN were significantly increased in the PD patients than in the controls. There was no significant correlation between serum UA levels and nigral phase shift values. CONCLUSIONS As previous studies, low serum UA level and increased nigral iron content in the PD was reconfirmed in this study. However, we failed to find the relationship between these two markers. Our data suggest that serum UA may not be important determinant of nigral iron deposition in PD.
Collapse
Affiliation(s)
- Tae-Hyoung Kim
- Department of Neurology, Pusan National University Yangsan Hospital, Research Institute for Convergence of Biomedical Science and Technology, Yangsan, Korea
| | - Jae-Hyeok Lee
- Department of Neurology, Pusan National University Yangsan Hospital, Research Institute for Convergence of Biomedical Science and Technology, Yangsan, Korea
- * E-mail:
| |
Collapse
|
133
|
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia have several important features in common. They are progressive, they affect a relatively inaccessible organ, and we have no disease-modifying therapies for them. For these brain-based diseases, current diagnosis and evaluation of disease severity rely almost entirely on clinical examination, which may be only a rough approximation of disease state. Thus, the development of biomarkers-objective, relatively easily measured, and precise indicators of pathogenic processes-could improve patient care and accelerate therapeutic discovery. Yet existing, rigorously tested neurodegenerative disease biomarkers are few, and even fewer biomarkers have translated into clinical use. To find new biomarkers for these diseases, an unbiased, high-throughput screening approach may be needed. In this review, I will describe the potential utility of such an approach to biomarker discovery, using Parkinson's disease as a case example.
Collapse
Affiliation(s)
- Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3 West Gates, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
134
|
Atassi N, Berry J, Shui A, Zach N, Sherman A, Sinani E, Walker J, Katsovskiy I, Schoenfeld D, Cudkowicz M, Leitner M. The PRO-ACT database: design, initial analyses, and predictive features. Neurology 2014; 83:1719-25. [PMID: 25298304 DOI: 10.1212/wnl.0000000000000951] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To pool data from completed amyotrophic lateral sclerosis (ALS) clinical trials and create an open-access resource that enables greater understanding of the phenotype and biology of ALS. METHODS Clinical trials data were pooled from 16 completed phase II/III ALS clinical trials and one observational study. Over 8 million de-identified longitudinally collected data points from over 8,600 individuals with ALS were standardized across trials and merged to create the Pooled Resource Open-Access ALS Clinical Trials (PRO-ACT) database. This database includes demographics, family histories, and longitudinal clinical and laboratory data. Mixed effects models were used to describe the rate of disease progression measured by the Revised ALS Functional Rating Scale (ALSFRS-R) and vital capacity (VC). Cox regression models were used to describe survival data. Implementing Bonferroni correction, the critical p value for 15 different tests was p = 0.003. RESULTS The ALSFRS-R rate of decline was 1.02 (±2.3) points per month and the VC rate of decline was 2.24% of predicted (±6.9) per month. Higher levels of uric acid at trial entry were predictive of a slower drop in ALSFRS-R (p = 0.01) and VC (p < 0.0001), and longer survival (p = 0.02). Higher levels of creatinine at baseline were predictive of a slower drop in ALSFRS-R (p = 0.01) and VC (p < 0.0001), and longer survival (p = 0.01). Finally, higher body mass index (BMI) at baseline was associated with longer survival (p < 0.0001). CONCLUSION The PRO-ACT database is the largest publicly available repository of merged ALS clinical trials data. We report that baseline levels of creatinine and uric acid, as well as baseline BMI, are strong predictors of disease progression and survival.
Collapse
Affiliation(s)
- Nazem Atassi
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA.
| | - James Berry
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Amy Shui
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Neta Zach
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Alexander Sherman
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Ervin Sinani
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Jason Walker
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Igor Katsovskiy
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - David Schoenfeld
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Merit Cudkowicz
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| | - Melanie Leitner
- From the Neurological Clinical Research Institute (NCRI), Department of Neurology (N.A., J.B., A. Sherman, E.S., J.W., I.K., M.C.), and the Biostatistics Center (A. Shui, D.S.), Massachusetts General Hospital, Boston; and Prize4Life (N.Z., M.L.), Cambridge, MA
| |
Collapse
|
135
|
Simon KC, Eberly S, Gao X, Oakes D, Tanner CM, Shoulson I, Fahn S, Schwarzschild MA, Ascherio A. Mendelian randomization of serum urate and parkinson disease progression. Ann Neurol 2014; 76:862-8. [PMID: 25257975 DOI: 10.1002/ana.24281] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Higher serum urate concentrations predict more favorable prognosis in individuals with Parkinson disease (PD). The purpose of this study was to test the causality of this association using a Mendelian randomization approach. METHODS The study was conducted among participants in DATATOP and PRECEPT, 2 randomized trials among patients with early PD. The 808 patients with available DNA were genotyped for 3 SLC2A9 single nucleotide polymorphisms (SNPs) that identify an allele associated with lower urate concentrations, and for selected SNPs in other genes encoding urate transporters that have modest or no effect on serum urate levels. An SLC2A9 score was created based on the total number of minor alleles at the 3 SLC2A9 loci. Primary outcome was disability requiring dopaminergic treatment. RESULTS Serum urate concentrations were 0.69mg/dl lower among individuals with ≥4 SLC2A9 minor alleles as compared to those with ≤2 (p = 0.0002). The hazard ratio (HR) for progression to disability requiring dopaminergic treatment increased with increasing SLC2A9 score (HR = 1.16, 95% confidence interval [CI] = 1.00-1.35, p = 0.056). In a comparative analysis, the HR was 1.27 (95% CI = 1.00-1.61, p = 0.0497) for a 0.5mg/dl genetically conferred decrease in serum urate, and 1.05 (95% CI = 1.01-1.10, p = 0.0133) for a 0.5mg/dl decrease in measured serum urate. No associations were found between polymorphisms in other genes associated with urate that do not affect serum urate and PD progression. INTERPRETATION This Mendelian randomization analysis adds to the evidence of a causal protective effect of high urate levels.
Collapse
Affiliation(s)
- Kelly Claire Simon
- Department of Nutrition, Harvard School of Public Health, Boston, MA; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Swanson CR, Li K, Unger TL, Gallagher MD, Van Deerlin VM, Agarwal P, Leverenz J, Roberts J, Samii A, Gross RG, Hurtig H, Rick J, Weintraub D, Trojanowski JQ, Zabetian C, Chen-Plotkin AS. Lower plasma apolipoprotein A1 levels are found in Parkinson's disease and associate with apolipoprotein A1 genotype. Mov Disord 2014; 30:805-12. [PMID: 25227208 DOI: 10.1002/mds.26022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/25/2014] [Accepted: 07/16/2014] [Indexed: 01/17/2023] Open
Abstract
The discovery of novel plasma-based biomarkers could lead to new approaches in the treatment of Parkinson's disease (PD). Here, we explore the role of plasma apolipoprotein A1 (ApoA1) as a risk marker for PD and evaluate the influence of APOA1 promoter variation on plasma ApoA1 levels. Plasma ApoA1 and the single-nucleotide polymorphism, rs670, were assayed in a discovery cohort (cohort 1) of 301 PD patients, 80 normal controls (NCs), and 165 subjects with other neurodegenerative diseases, as well as a cohort (cohort 2) of 158 PD patients from a second clinical site. Additionally, rs670 was genotyped in a third cohort of 1,494 PD and 925 NC subjects from both clinical sites. Compared to both normal and disease controls, PD patients have lower plasma ApoA1 (P < 0.001 for both comparisons). Moreover, in PD patients, plasma ApoA1 levels are correlated with genotype at the APOA1 promoter polymorphism, rs670. Specifically, lower plasma ApoA1 levels were found in rs670 major allele (G) homozygotes in both cohort 1 (P = 0.009) and in a replication cohort (cohort 2; n = 158 PD patients; P = 0.024). Finally, evaluating rs670 genotype frequencies in 1,930 PD cases versus 997 NCs, the rs670 GG genotype shows a trend toward association (odds ratio: 1.1; P = 0.10) with PD. Our results are compatible with a model whereby circulating ApoA1 levels may be useful in risk-stratifying subjects for the development of PD, with higher ApoA1 levels suggesting relative protection. Future studies evaluating modulation of ApoA1 as a novel therapeutic strategy in PD are warranted. © 2014 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Christine R Swanson
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine Li
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Travis L Unger
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael D Gallagher
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Cellular and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pinky Agarwal
- Booth Gardner Parkinson's Care Center, Evergreen Hospital Medical Center, Kirkland, Washington, USA
| | - James Leverenz
- Cleveland Clinic Ruvo Center for Brain Health, Cleveland, Ohio, USA
| | - John Roberts
- Virginia Mason Medical Center, Seattle, Washington, USA
| | - Ali Samii
- VA Puget Sound Health Care System and Department of Neurology, University of Washington Seattle, Seattle, Washington, USA
| | - Rachel Goldmann Gross
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Howard Hurtig
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jacqueline Rick
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cyrus Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington Seattle, Seattle, Washington, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
137
|
Zheng Z, Guo X, Wei Q, Song W, Cao B, Huang R, Ou R, Chen X, Shang H. Serum uric acid level is associated with the prevalence but not with survival of amyotrophic lateral sclerosis in a Chinese population. Metab Brain Dis 2014; 29:771-5. [PMID: 24577631 DOI: 10.1007/s11011-014-9510-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 02/13/2014] [Indexed: 02/05/2023]
Abstract
Oxidative stress is involved in the pathogenesis of amyotrophic lateral sclerosis (ALS). Serum uric acid (UA) has anti-oxidative effect. To clarify the associations between UA and sporadic ALS (SALS) in Chinese population. A total of 512 SALS patients and 501 age- and gender- matched healthy controls were included, with fasting serum UA evaluated. The ALS functional rating scale-Revised was used to assess disease severity, and the mean change of ALSFRS-R per month (△ALS-FRS-R/m) to assess its progression. Four hundred forty-five patients were followed up. Survival and progression were estimated according to quartiles and gender-specific quartiles of UA. The mean age of SALS patients was 53.3 ± 11.7 years. UA level of SALS was significantly lower than controls (p = 0.001). Male SALS had higher UA than female (p = 0.001). The occurrence of SALS was increased in the lowest, second and third quartiles compared with the highest quartile (p = 0.001). In the gender-specific analysis, increased occurrence was found in the lowest and second quartiles compared with third and fourth quartiles in both males and females SALS (p(f) = 0.001 and p(m) < 0.05). No correlation was found between △ALS-FRS-R/m and UA (p > 0.05), nor between SALS survival and UA. SALS patients have low level of serum UA than controls. Low level of uric acid may be associated with increased occurrence of SALS in Chinese population. Uric acid level may not contribute to the survival or progression of SALS.
Collapse
Affiliation(s)
- Zhenzhen Zheng
- Department of Neurology, West China Hospital, SiChuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Moccia M, Picillo M, Erro R, Vitale C, Longo K, Amboni M, Santangelo G, Palladino R, Capo G, Orefice G, Barone P, Pellecchia MT. Presence and progression of non-motor symptoms in relation to uric acid inde novoParkinson's disease. Eur J Neurol 2014; 22:93-8. [DOI: 10.1111/ene.12533] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 06/09/2014] [Indexed: 12/25/2022]
Affiliation(s)
- M. Moccia
- Department of Neuroscience, Reproductive Science and Odontostomatology; Federico II University; Naples Italy
| | - M. Picillo
- Neuroscience Section; Department of Medicine; Center for Neurodegenerative Diseases (CEMAND); University of Salerno; Salerno Italy
| | - R. Erro
- Sobell Department of Motor Neuroscience and Movement Disorders; University College London (UCL) Institute of Neurology; London UK
- Department of Neurological and Movement Sciences; University of Verona; Policlinico Borgo Roma; Verona Italy
| | - C. Vitale
- IDC Hermitage Capodimonte; Naples Italy
- Department of Motor Sciences; University Parthenope; Naples Italy
| | - K. Longo
- IDC Hermitage Capodimonte; Naples Italy
| | - M. Amboni
- IDC Hermitage Capodimonte; Naples Italy
| | - G. Santangelo
- Neuropsychology Laboratory; Department of Psychology; Second University of Naples; Caserta Italy
| | - R. Palladino
- Department of Primary Care and Public Health; Imperial College; London UK
- Department of Public Health; Federico II University; Naples Italy
| | - G. Capo
- AOU San Giovanni di Dio e Ruggi d'Aragona; Salerno Italy
| | - G. Orefice
- Department of Neuroscience, Reproductive Science and Odontostomatology; Federico II University; Naples Italy
| | - P. Barone
- Neuroscience Section; Department of Medicine; Center for Neurodegenerative Diseases (CEMAND); University of Salerno; Salerno Italy
| | - M. T. Pellecchia
- Neuroscience Section; Department of Medicine; Center for Neurodegenerative Diseases (CEMAND); University of Salerno; Salerno Italy
| |
Collapse
|
139
|
Nakamura T, Sato A, Kitsukawa T, Momiyama T, Yamamori T, Sasaoka T. Distinct motor impairments of dopamine D1 and D2 receptor knockout mice revealed by three types of motor behavior. Front Integr Neurosci 2014; 8:56. [PMID: 25076876 PMCID: PMC4097398 DOI: 10.3389/fnint.2014.00056] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 06/20/2014] [Indexed: 11/13/2022] Open
Abstract
Both D1R and D2R knock out (KO) mice of the major dopamine receptors show significant motor impairments. However, there are some discrepant reports, which may be due to the differences in genetic background and experimental procedures. In addition, only few studies directly compared the motor performance of D1R and D2R KO mice. In this paper, we examined the behavioral difference among N10 congenic D1R and D2R KO, and wild type (WT) mice. First, we examined spontaneous motor activity in the home cage environment for consecutive 5 days. Second, we examined motor performance using the rota-rod task, a standard motor task in rodents. Third, we examined motor ability with the Step-Wheel task in which mice were trained to run in a motor-driven turning wheel adjusting their steps on foothold pegs to drink water. The results showed clear differences among the mice of three genotypes in three different types of behavior. In monitoring spontaneous motor activities, D1R and D2R KO mice showed higher and lower 24 h activities, respectively, than WT mice. In the rota-rod tasks, at a low speed, D1R KO mice showed poor performance but later improved, whereas D2R KO mice showed a good performance at early days without further improvement. When first subjected to a high speed task, the D2R KO mice showed poorer rota-rod performance at a low speed than the D1R KO mice. In the Step-Wheel task, across daily sessions, D2R KO mice increased the duration that mice run sufficiently close to the spout to drink water, and decreased time to touch the floor due to missing the peg steps and number of times the wheel was stopped, which performance was much better than that of D1R KO mice. These incongruent results between the two tasks for D1R and D2R KO mice may be due to the differences in the motivation for the rota-rod and Step-Wheel tasks, aversion- and reward-driven, respectively. The Step-Wheel system may become a useful tool for assessing the motor ability of WT and mutant mice.
Collapse
Affiliation(s)
- Toru Nakamura
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan ; Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI) Okazaki, Japan
| | - Asako Sato
- Laboratory of Neurochemistry, National Institute for Basic Biology Okazaki, Japan ; Department of Laboratory Animal Science, Kitasato University School of Medicine Sagamihara, Japan
| | - Takashi Kitsukawa
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan ; KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University Osaka, Japan
| | - Toshihiko Momiyama
- Department of Pharmacology, Jikei University School of Medicine Tokyo, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan ; Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI) Okazaki, Japan
| | - Toshikuni Sasaoka
- Laboratory of Neurochemistry, National Institute for Basic Biology Okazaki, Japan ; Department of Laboratory Animal Science, Kitasato University School of Medicine Sagamihara, Japan ; Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University Niigata, Japan
| |
Collapse
|
140
|
Abstract
There is a pressing need for biomarkers to diagnose Parkinson's disease (PD), assess disease severity, and prognosticate course. Various types of biologic specimens are potential candidates for identifying biomarkers--defined here as surrogate indicators of physiological or pathophysiological states--but blood has the advantage of being minimally invasive to obtain. There are, however, several challenges to identifying biomarkers in blood. Several candidate biomarkers identified in other diseases or in other types of biological fluids are being pursued as blood-based biomarkers in PD. In addition, unbiased discovery is underway using techniques including metabolomics, proteomics, and gene expression profiling. In this review, we summarize these techniques and discuss the challenges and successes of blood-based biomarker discovery in PD. Blood-based biomarkers that are discussed include α-synuclein, DJ-1, uric acid, epidermal growth factor, apolipoprotein-A1, and peripheral inflammatory markers.
Collapse
Affiliation(s)
- Lama M Chahine
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | | | | |
Collapse
|
141
|
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: 2.1] [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]
|
142
|
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: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
143
|
Zhang N, Shu HY, Huang T, Zhang QL, Li D, Zhang GQ, Peng XY, Liu CF, Luo WF, Hu LF. Nrf2 signaling contributes to the neuroprotective effects of urate against 6-OHDA toxicity. PLoS One 2014; 9:e100286. [PMID: 24959672 PMCID: PMC4069024 DOI: 10.1371/journal.pone.0100286] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/26/2014] [Indexed: 01/07/2023] Open
Abstract
Background Mounting evidence shows that urate may become a biomarker of Parkinson's disease (PD) diagnosis and prognosis and a neuroprotectant candidate for PD therapy. However, the cellular and molecular mechanisms underlying its neuroprotective actions remain poorly understood. Results In this study, we showed that urate pretreatment protected dopaminergic cell line (SH-SY5Y and MES23.5) against 6-hydroxydopamine (6-OHDA)- and hydrogen peroxide- induced cell damage. Urate was found to be accumulated into SH-SY5Y cells after 30 min treatment. Moreover, urate induced NF-E2-related factor 2 (Nrf2) accumulation by inhibiting its ubiquitinationa and degradation, and also promoted its nuclear translocation; however, it did not modulate Nrf2 mRNA level or Kelch-like ECH-associated protein 1 (Keap1) expression. In addition, urate markedly up-regulated the transcription and protein expression of γ-glutamate-cysteine ligase catalytic subunit (γ-GCLC) and heme oxygenase-1 (HO-1), both of which are controlled by Nrf2 activity. Furthermore, Nrf2 knockdown by siRNA abolished the intracellular glutathione augmentation and the protection exerted by urate pretreatment. Conclusion Our findings demonstrated that urate treatment may result in Nrf2-targeted anti-oxidant genes transcription and expression by reducing Nrf2 ubiquitination and degradation and promoting its nuclear translocation, and thus offer neuroprotection on dopaminergic cells against oxidative stresses.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Hai-Yang Shu
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Tingting Huang
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Qi-Lin Zhang
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Da Li
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Guan-Qun Zhang
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Yan Peng
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Chun-Feng Liu
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Wei-Feng Luo
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- * E-mail: (LFH); (WFL)
| | - Li-Fang Hu
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmacology, Soochow University, Suzhou, Jiangsu, China
- * E-mail: (LFH); (WFL)
| |
Collapse
|
144
|
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: 2.7] [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
|
145
|
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: 174] [Impact Index Per Article: 17.4] [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
|
146
|
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: 12] [Impact Index Per Article: 1.2] [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
|
147
|
Yue HY, Huang S, Chang J, Heo C, Yao F, Adhikari S, Gunes F, Liu LC, Lee TH, Oh ES, Li B, Zhang JJ, Huy TQ, Luan NV, Lee YH. ZnO nanowire arrays on 3D hierachical graphene foam: biomarker detection of Parkinson's disease. ACS NANO 2014; 8:1639-46. [PMID: 24405012 DOI: 10.1021/nn405961p] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
We report that vertically aligned ZnO nanowire arrays (ZnO NWAs) were fabricated on 3D graphene foam (GF) and used to selectively detect uric acid (UA), dopamine (DA), and ascorbic acid (AA) by a differential pulse voltammetry method. The optimized ZnO NWA/GF electrode provided a high surface area and high selectivity with a detection limit of 1 nM for UA and DA. The high selectivity in the oxidation potential was explained by the gap difference between the lowest unoccupied and highest occupied molecular orbitals of a biomolecule for a set of given electrodes. This method was further used to detect UA levels in the serum of patients with Parkinson's disease (PD). The UA level was 25% lower in PD patients than in healthy individuals. This finding strongly implies that UA can be used as a biomarker for PD.
Collapse
Affiliation(s)
- Hong Yan Yue
- Centre for Integrated Nanostructure Physics, Institute for Basic Science, Sungkyunkwan University , Suwon 440-746, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
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: 1.3] [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
|
149
|
AlDakheel A, Kalia LV, Lang AE. Pathogenesis-targeted, disease-modifying therapies in Parkinson disease. Neurotherapeutics 2014; 11:6-23. [PMID: 24085420 PMCID: PMC3899477 DOI: 10.1007/s13311-013-0218-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Parkinson disease is an inexorably progressive neurodegenerative disorder. Multiple attempts have been made to establish therapies for Parkinson disease which provide neuroprotection or disease modification-two related, but not identical, concepts. However, to date, none of these attempts have succeeded. Many challenges exist in this field of research, including a complex multisystem disorder that includes dopaminergic and non-dopaminergic features; poorly understood and clearly multifaceted disease pathogenic mechanisms; a lack of reliable animal models; an absence of effective biomarkers of disease state, progression, and target engagement; and the confounding effects of potent symptomatic therapy. In this article, we will review previous, ongoing, and potential future trials designed to alter the progressive course of the disease from the perspective of the targeted underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Amaal AlDakheel
- />Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson’s Disease, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Lorraine V. Kalia
- />Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson’s Disease, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Anthony E. Lang
- />Movement Disorders Unit, Toronto Western Hospital, 399 Bathurst Street, 7 McLaughlin Wing, Toronto, M5T 2S8 ON Canada
| |
Collapse
|
150
|
Xia X, He F, Wu X, Peng F, Huang F, Yu X. Relationship between serum uric acid and all-cause and cardiovascular mortality in patients treated with peritoneal dialysis. Am J Kidney Dis 2013; 64:257-64. [PMID: 24176223 DOI: 10.1053/j.ajkd.2013.08.027] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 08/30/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Although serum uric acid level appears to be associated with mortality in individuals treated with hemodialysis, the relationship between serum uric acid level and death is uncertain in patients treated with peritoneal dialysis (PD). STUDY DESIGN Cohort study. SETTING & PARTICIPANTS 985 patients from a single PD center in South China followed up for a median of 25.3 months. PREDICTOR Serum uric acid level. OUTCOMES & MEASUREMENTS The association of baseline sex-specific uric acid level with all-cause and cardiovascular mortality was evaluated. Models were adjusted for age, body mass index, comorbidity score, residual kidney function, total Kt/V, allopurinol and angiotensin-converting enzyme inhibitor/angiotensin receptor blocker use, and laboratory test results, including hemoglobin, serum albumin, creatinine, calcium, phosphorus, triglycerides, low-density lipoprotein cholesterol, and high-sensitivity C-reactive protein. RESULTS Mean age was 48.3±15.4 (SD) years, and 23% had diabetes. Mean uric acid level was 7.0±1.3 (range, 3.8-19.8) mg/dL. During follow-up, 144 deaths were recorded, of which 64 were due to cardiovascular events. In multivariable models, the highest sex-specific tertile of uric acid level was associated with increased risk of all-cause mortality (HR, 1.93; 95% CI, 1.27-2.93; P=0.004) and cardiovascular mortality (HR, 3.31; 95% CI, 1.70-6.41; P<0.001) compared to the lowest tertile. Adjusted Cox regression models showed that the HRs per 1-mg/dL higher uric acid level for all-cause and cardiovascular mortality were 1.33 (95% CI, 1.14-1.56; P<0.001) and 1.44 (95% CI, 1.17-1.77; P=0.001) for men and 1.03 (95% CI, 0.86-1.24; P=0.8) and 1.16 (95% CI, 0.97-1.38; P=0.1) for women, respectively. A formal test for interaction indicated that the association of uric acid level with all-cause and cardiovascular mortality differed by sex (β = -0.06 [P=0.02] and β = -0.10 [P=0.02], respectively). LIMITATIONS Single measurement of uric acid at baseline. Cause of death determined by death certificates and expert consensus. CONCLUSIONS Elevated serum uric acid level is an independent risk factor for all-cause and cardiovascular mortality in men treated with PD.
Collapse
Affiliation(s)
- Xi Xia
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Feng He
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xianfeng Wu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fenfen Peng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fengxian Huang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|