90
Views
2
CrossRef citations to date
0
Altmetric
Review

Biomarkers of Parkinson's disease: recent insights, current challenges, and future prospects

, , , &
Pages 1-13 | Published online: 22 Feb 2016

References

  • Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95.
  • Wright-Willis A, Evanoff BA, Lian M, Criswell SR, Racette BA. Geographic and ethnic variation in Parkinson disease: a population-based study of US Medicare beneficiaries. Neuroepidemiology. 2010;34(3):143–151.
  • Pont-Sunyer C, Hotter A, Gaig C, et al. The onset of nonmotor symptoms in Parkinson’s disease (the ONSET PD study). Mov Disord. 2015;30(2):229–237.
  • Noyce AJ, Bestwick JP, Silveira-Moriyama L, et al. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann Neurol. 2012;72(6):893–901.
  • Moccia M, Erro R, Picillo M, et al. Quitting smoking: an early non-motor feature of Parkinson’s disease? Parkinsonism Relat Disord. 2015;21(3):216–220.
  • Schrag A, Horsfall L, Walters K, Noyce A, Petersen I. Prediagnostic presentations of Parkinson’s disease in primary care: a case-control study. Lancet Neurol. 2015;14(1):57–64.
  • Wu YH, Liao YC, Chen YH, Chang MH, Lin CH. Risk of premotor symptoms in patients with newly diagnosed PD: a nationwide, population-based, case-control study in Taiwan. PLoS One. 2015; 10(6):e0130282.
  • Shen CC, Tsai SJ, Perng CL, Kuo BI, Yang AC. Risk of Parkinson disease after depression: a nationwide population-based study. Neurology. 2013;81(17):1538–1544.
  • Adams-Carr KL, Bestwick JP, Shribman S, Lees A, Schrag A, Noyce AJ. Constipation preceding Parkinson’s disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. Epub 2015 Sep 7.
  • Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older males initially diagnosed with idiopathic REM sleep behavior disorder: 16-year update on a previously reported series. Sleep Med. 2013;14(8):744–748.
  • Iranzo A, Tolosa E, Gelpi E, Molinuevo JL, Valldeoriola F, Serradell M, et al. Neurodegenerative disease status and post-mortem pathology in idiopathic rapid-eye-movement sleep behaviour disorder: an observational cohort study. Lancet Neurol. 2013;12(5):443–453.
  • Berg D, Marek K, Ross GW, Poewe W. Defining at-risk populations for Parkinson’s disease: lessons from ongoing studies. Mov Disord. 2012;27(5):656–665.
  • O’Sullivan SS, Williams DR, Gallagher DA, Massey LA, Silveira-Moriyama L, Lees AJ. Nonmotor symptoms as presenting complaints in Parkinson’s disease: a clinicopathological study. Mov Disord. 2008;23(1):101–106.
  • Hu G. Total cholesterol and the risk of Parkinson’s disease: a review for some new findings. Parkinsons Dis. 2010;2010:836962.
  • De Lau LML, Koudstaal PJ, Hofman A, Breteler MMB. Serum cholesterol levels and the risk of Parkinson’s disease. Am J Epidemiol. 2006;164(10):998–1002.
  • Godau J, Herfurth M, Kattner B, Gasser T, Berg D. Increased serum insulin-like growth factor 1 in early idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81(5):536–538.
  • Godau J, Knauel K, Weber K, et al. Serum insulinlike growth factor 1 as possible marker for risk and early diagnosis of Parkinson disease. Arch Neurol. 2011;68(7):925–931.
  • Knekt P, Kilkkinen A, Rissanen H, Marniemi J, Sääksjärvi K, Heliövaara M. Serum vitamin D and the risk of Parkinson disease. Arch Neurol. 2010;67(7):808–811.
  • de Lau L, Koudstaal P, Hofman A, Breteler M. Serum uric acid levels and the risk of Parkinson disease. Ann Neurol. 2005;585:797–800.
  • Weisskopf M, O’Reilly E, Chen H, Schwarzschild M, Ascherio A. Plasma urate and risk of Parkinson’s disease. Am J Epidemiol. 2007;166(5):561–567.
  • Chen H, Mosley T, Alonso A, Huang X. Plasma urate and Parkinson’s disease in the Atherosclerosis Risk in Communities (ARIC) study. Am J Epidemiol. 2009;169(9):1064–1069.
  • Alonso A, Rodríguez L, Logroscino G, Hernán M. Gout and risk of Parkinson disease: a prospective study. Neurology. 2007;69(17):1696–1700.
  • De Vera M, Rahman M, Rankin J, Kopec J, Gao X, Choi H. Gout and the risk of Parkinson’s disease: a cohort study. Arthritis Rheum. 2008;59(11):1549–1554.
  • Stern MB, Siderowf A. Parkinson’s at risk syndrome: can Parkinson’s disease be predicted? Mov Disord. 2010;25 Suppl 1:S89–93.
  • Siderowf A, Jennings D, Eberly S, et al. Impaired olfaction and other prodromal features in the Parkinson at-risk syndrome study. Mov Disord. 2012;27(3):406–412.
  • Jennings D, Siderowf A, Stern M, et al. Imaging prodromal Parkinson disease: the Parkinson associated risk syndrome study. Neurology. 2014;83(19):1739–1746.
  • Berg D, Seppi K, Behnke S, et al. Enlarged substantia nigra hyperechogenicity and risk for Parkinson disease: a 37-month 3-center study of 1847 older persons. Arch Neurol. 2011;68(7):932–937.
  • Berg D, Behnke S, Seppi K, et al. Enlarged hyperechogenic substantia nigra as a risk marker for Parkinson’s disease. Mov Disord. 2013;28(2):216–219.
  • Iranzo A, Stockner H, Serradell M, et al. Five-year follow-up of substantia nigra echogenicity in idiopathic REM sleep behavior disorder. Mov Disord. 2014;29(14):1774–1780.
  • Iranzo A, Valldeoriola F, Lomeña F, et al. Serial dopamine transporter imaging of nigrostriatal function in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 2011;10(9):797–805.
  • Vilas D, Ispierto L, Álvarez R, et al. Clinical and imaging markers in premotor LRRK2 G2019S mutation carriers. Parkinsonism Relat Disord. 2015;21(10):1170–1176.
  • Helmich RC, Thaler A, van Nuenen BF, et al. Reorganization of corticostriatal circuits in healthy G2019S LRRK2 carriers. Neurology. 2015;84(4):399–406.
  • Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinic-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55:181–184.
  • Barone P, Antonini A, Colosimo C, et al. The PRIAMO study: a multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson’s disease. Mov Disord. 2009;24(11):1641–1649.
  • Erro R, Santangelo G, Picillo M, et al. Link between non-motor symptoms and cognitive dysfunctions in de novo, drug-naive PD patients. J Neurol. 2012;259(9):1808–1813.
  • Picillo M, Amboni M, Erro R, et al. Gender differences in non-motor symptoms in early, drug naïve Parkinson’s disease. J Neurol. 2013;260(11):2849–2855.
  • Lewis SJG, Foltynie T, Blackwell AD, Robbins TW, Owen AM, Barker RA. Heterogeneity of Parkinson’s disease in the early clinical stages using a data driven approach. J Neurol Neurosurg Psychiatry. 2005;76(3):343–348.
  • Marras C, Lang A. Parkinson’s disease subtypes: lost in translation? J Neurol Neurosurg Psychiatry. 2013;84(4):409–415.
  • Erro R, Vitale C, Amboni M, et al. The heterogeneity of early Parkinson’s disease: a cluster analysis on newly diagnosed untreated patients. PLoS One. 2013;8(8):e70244.
  • Nalls MA, McLean CY, Rick J, et al. Parkinson’s Disease Biomarkers Program and Parkinson’s Progression Marker Initiative investigators. Diagnosis of Parkinson’s disease on the basis of clinical and genetic classification: a population-based modelling study. Lancet Neurol. 2015;14(10):1002–1009.
  • Köllensperger M, Geser F, Seppi K, et al. European MSA Study Group. Red flags for multiple system atrophy. Mov Disord. 2008; 23(8):1093–1099.
  • Todorović Z, Džoljić E, Novaković I, et al. Homocysteine serum levels and MTHFR C677T genotype in patients with Parkinson’s disease, with and without levodopa therapy. J Neurol Sci. 2006;248(1–2):56–61.
  • Levin J, Bötzel K, Giese A, Vogeser M, Lorenzl S. Elevated levels of methylmalonate and homocysteine in Parkinson’s disease, progressive supranuclear palsy and amyotrophic lateral sclerosis. Dement Geriatr Cogn Disord. 2010;29:553–559.
  • Chen W, Cheng X, Zhang X, Zhang Q, Sun H. The expression features of serum Cystatin C and homocysteine of Parkinson’s disease with mild cognitive dysfunction. Eur Rev Med Pharmacol Sci. 2015;19:2957–2963.
  • Scalzo P, Kümmer A, Bretas TL, Cardoso F, Teixeira AL. Serum levels of brain-derived neurotrophic factor correlate with motor impairment in Parkinson’s disease. J Neurol. 2010;257(4):540–545.
  • Ziebell M, Khalid U, Klein AB, et al. Striatal dopamine transporter binding correlates with serum BDNF levels in patients with striatal dopaminergic neurodegeneration. Neurobiol Aging. 2012;33(2):428.e1–5.
  • Pellecchia MT, Pivonello R, Salvatore E, et al. Growth hormone response to arginine test distinguishes multiple system atrophy from Parkinson’s disease and idiopathic late-onset cerebellar ataxia. Clin Endocrinol (Oxf). 2005;62(4):428–433.
  • Picillo M, Erro R, Santangelo G, et al. Insulin-like growth factor-1 and progression of motor symptoms in early, drug-naïve Parkinson’s disease. J Neurol. 2013;260(7):1724–1730.
  • Pellecchia MT, Santangelo G, Picillo M, et al. Insulin-like growth factor-1 predicts cognitive functions at 2-year follow-up in early, drug-naïve Parkinson’s disease. Eur J Neurol. 2014;21(5):802–807.
  • Liu Y, Zhang B-S. Serum 25-hydroxyvitamin D predicts severity in Parkinson’s disease patients. Neurol Sci. 2014;35(1):67–71.
  • Palladino R, Moccia M, De Pascale T, et al. Season of birth and Parkinson’s disease: possible relationship? Neurol Sci. 2015;36(8):1457–1462.
  • Gardener H, Gao X, Chen H, Schwarzschild MA, Spiegelman D, Ascherio A. Prenatal and early life factors and risk of Parkinson’s disease. Mov Disord. 2010;25(11):1560–1567.
  • Mahlknecht P, Stemberger S, Sprenger F, et al. An antibody microarray analysis of serum cytokines in neurodegenerative Parkinsonian syndromes. Proteome Sci. 2012;10(1):71.
  • Moccia M, Picillo M, Erro R, et al. Increased bilirubin levels in de novo Parkinson’s disease. Eur J Neurol. 2015;22(6):954–959.
  • Hatano T, Saiki S, Okuzumi A, Mohney R, Hattori N. Identification of novel biomarkers for Parkinson’s disease by metabolomic technologies. J Neurol Neurosurg Psychiatry. Epub 2015 Mar 20.
  • Bharucha KJ, Friedman JK, Vincent AS, Ross ED. Lower serum ceruloplasmin levels correlate with younger age of onset in Parkinson’s disease. J Neurol. 2008;255(12):1957–1962.
  • Jin L, Wang J, Zhao L, et al. Decreased serum ceruloplasmin levels characteristically aggravate nigral iron deposition in Parkinson’s disease. Brain. 2011;134(1):50–58.
  • Shen C, Guo Y, Luo W, Lin C, Ding M. Serum urate and the risk of Parkinson’s disease: results from a meta-analysis. Can J Neurol Sci. 2012;(88):73–79.
  • Shen L, Ji H-F. Low uric acid levels in patients with Parkinson’s disease: evidence from meta-analysis. BMJ. 2013;3(11):e003620.
  • Schwarzschild MA, Marek K, Eberly S, et al. Serum urate and probability of dopaminergic deficit in early “Parkinson’s disease”. Mov Disord. 2011;26(10):1864–1868.
  • Moccia M, Pappatà S, Erro R, et al. Uric acid relates to dopamine transporter availability in Parkinson’s disease. Acta Neurol Scand. 2015a;123(2):127–131.
  • Mariani S, Ventriglia M, Simonelli I, et al. Association between sex, systemic iron variation and probability of Parkinson’s disease. Int J Neurosci. 2015;126(4):354–360.
  • Fukushima T, Tan X, Luo Y, Kanda H. Relationship between blood levels of heavy metals and Parkinson’s disease in China. Neuroepidemiology. 2010;34(1):18–24.
  • Simonsen A, Kuiperij B, Ali El-Agnaf O, et al. The utility of α-synuclein as biofluid marker in neurodegenerative diseases: a systematic review of the literature. Biomark Med. Epub 2015 Aug 28.
  • Williams SM, Schulz P, Sierks MR. Oligomeric α-synuclein and β-amyloid variants as potential biomarkers for Parkinson’s and Alzheimer’s diseases. Eur J Neurosci. Epub 2015 Aug 31.
  • Gupta V, Garg RK, Khattri S. Serological Analysis of Alpha-synuclein and NF-κB in Parkinson’s Disease Patients. J Clin Diagn Res. 2015;9(5):BC01–04.
  • Shi M, Liu C, Cook TJ et al. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014;128(5):639–650.
  • Alves G, Pedersen KF, Bloem BR, et al. Cerebrospinal fluid amyloid-β and phenotypic heterogeneity in de novo Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2013;84:537–543.
  • Stav AL, Aarsland D, Johansen KK, Hessen E, Auning E, Fladby T. Amyloid-β and α-synuclein cerebrospinal fluid biomarkers and cognition in early Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:758–764.
  • Bibl M, Mollenhauer B, Esselmann H, et al. CSF amyloid-β-peptides in Alzheimer’s disease, dementia with Lewy bodies and Parkinson’s disease dementia. Brain. 2006;129:1177–1187.
  • van Dijk KD, Teunissen CE, Drukarch B, et al. Diagnostic cerebrospinal fluid biomarkers for Parkinson’s disease: a pathogenetically based approach. Neurobiol Dis. 2010;39:229–241.
  • Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Agúndez JAG. Cerebrospinal fluid biochemical studies in patients with Parkinson’s disease: toward a potential search for biomarkers for this disease. Front Cell Neurosci. 2014;11:369.
  • Wennström M, Surova Y, Hall S, et al. Low CSF levels of both α-synuclein and the α-synuclein cleaving enzyme neurosin in patients with synucleinopathy. PLoS One. 2013;8:e53250.
  • Přikrylová Vranová H, Mareš J, Nevrlý M, et al. CSF markers of neurodegeneration in Parkinson’s disease. J Neural Transm. 2010;117:1177–1181.
  • Waragai M, Wei J, Fujita M, et al. Increased level of DJ-1 in the cerebrospinal fluids of sporadic Parkinson’s disease. Biochem Biophys Res Commun. 2006;345:967–972.
  • Herbert MK, Eeftens JM, Aerts MB, et al. CSF levels of DJ-1 and tau distinguish MSA patients from PD patients and controls. Parkinsonism Relat Disord. 2014;20:112–115.
  • Kikuchi A, Takeda A, Onodera H, et al. Systemic increase of oxidative nucleic acid damage in Parkinson’s disease and multiple system atrophy. Neurobiol Dis. 2002;9:244–248.
  • Buhmann C, Arlt S, Kontush A, et al. Plasma and CSF markers of oxidative stress are increased in Parkinson’s disease and influenced by antiparkinsonian medication. Neurobiol Dis. 2004;15:160–170.
  • Öhman A, Forsgren L. NMR metabonomics of cerebrospinal fluid distinguishes between Parkinson’s disease and controls. Neurosci Lett. 2015;594:36–39.
  • Trupp M, Jonsson P, Ohrfelt A, et al. Metabolite and peptide levels in plasma and CSF differentiating healthy controls from patients with newly diagnosed Parkinson’s disease. J Parkinsons Dis. 2014;4(3):549–560.
  • Bajaj N, Hauser RA, Grachev ID. Clinical utility of dopamine transporter single photon emission CT (DaT-SPECT) with (123I) ioflupane in diagnosis of parkinsonian syndromes. J Neurol Neurosurg Psychiatry. 2013;84(11):1288–1295.
  • Niethammer M, Eidelberg D. Metabolic brain networks in translational neurology: concepts and applications. Ann Neurol. 2012;72(5):635–647.
  • Chung EJ, Lee WY, Yoon WT, Kim BJ, Lee GH. Scintigraphy for differentiating Parkinson’s disease with autonomic dysfunction from Parkinsonism-predominant multiple system atrophy. Mov Disord. 2009;24(11):1650–1655.
  • Reimão S, Pita Lobo P, Neutel D, et al. Substantia nigra neuromelanin magnetic resonance imaging in de novo Parkinson’s disease patients. Eur J Neurol. 2015;22(3):540–546.
  • Zhang D, Liu X, Chen J, Liu B. Distinguishing patients with Parkinson’s disease subtypes from normal controls based on functional network regional efficiencies. PLoS One. 2014;9(12):e115131.
  • Bunzeck N, Singh-Curry V, Eckart C, et al. Motor phenotype and magnetic resonance measures of basal ganglia iron levels in Parkinson’s disease. Parkinsonism Relat Disord. 2013;19(12):1136–1142.
  • Moccia M, Pappatà S, Picillo M, et al. Dopamine transporter availability in motor subtypes of de novo drug-naïve Parkinson’s disease. J Neurol. 2014;261(11):2112–2118.
  • Pellecchia MT, Picillo M, Santangelo G, et al. Cognitive performances and DAT imaging in early Parkinson’s disease with mild cognitive impairment: a preliminary study. Acta Neurol Scand. 2015;131(5):275–281.
  • Erro R, Pappatà S, Amboni M, et al. Anxiety is associated with striatal dopamine transporter availability in newly diagnosed untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2012;18(9):1034–1038.
  • Rajput AH, Sitte HH, Rajput A, Fenton ME, Pifl C, Hornykiewicz O. Globus pallidus dopamine and Parkinson motor subtypes: clinical and brain biochemical correlation. Neurology. 2008;70(16 Pt 2):1403–1410.
  • Burn DJ, Rowan EN, Allan LM, Molloy S, O’Brien JT, McKeith IG. Motor subtype and cognitive decline in Parkinson’s disease, Parkinson’s disease with dementia, and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2006;77(5):585–589.
  • Picillo M, Amboni M, Erro R, et al. Segmental progression of cardinal motor symptoms in Parkinson’s disease: a pilot study suggesting a practical approach to rate disease course in the early stages. Parkinsonism Relat Disord. 2013;19(12):1143–1148.
  • Amboni M, Barone P, Picillo M, et al. A two-year follow-up study of executive dysfunctions in parkinsonian patients with freezing of gait at on-state. Mov Disord. 2010;25(6):800–802.
  • Amboni M, Cozzolino A, Longo K, Picillo M, Barone P. Freezing of gait and executive functions in patients with Parkinson’s disease. Mov Disord. 2008;23(3):395–400.
  • Amboni M, Barone P, Iuppariello L, et al. Gait patterns in Parkinsonian patients with or without mild cognitive impairment. Mov Disord. 2012;27(12):1536–1543.
  • Picillo M, Dubbioso R, Iodice R, et al. Short-latency afferent inhibition in patients with Parkinson’s disease and freezing of gait. J Neural Transm. 2015;122(11):1533–1540.
  • Erro R, Picillo M, Vitale C, et al. Non-motor symptoms in early Parkinson’s disease: a 2-year follow-up study on previously untreated patients. J Neurol Neurosurg Psychiatry. 2013;84(1):14–17.
  • Picillo M, Erro R, Amboni M et al. Gender differences in non-motor symptoms in early Parkinson’s disease: a 2-years follow-up study on previously untreated patients. Parkinsonism Relat Disord. 2014;20(8):850–854.
  • Santangelo G, Vitale C, Picillo M, et al. Mild Cognitive Impairment in newly diagnosed Parkinson’s disease: A longitudinal prospective study. Parkinsonism Relat Disord. 2015;21(10):1219–1226.
  • Picillo M, Barone P, Pellecchia MT, et al. Evolution of mild cognitive impairment in Parkinson disease. Neurology. 2014;82(15):1384.
  • Santangelo G, Vitale C, Trojano L, et al. Relationship between apathy and cognitive dysfunctions in de novo untreated Parkinson’s disease: a prospective longitudinal study. Eur J Neurol. 2015;22(2):253–260.
  • Erro R, Santangelo G, Barone P, et al. Do subjective memory complaints herald the onset of mild cognitive impairment in Parkinson disease? J Geriatr Psychiatry Neurol. 2014;27(4):276–281.
  • Manganelli F, Vitale C, Santangelo G, et al. Functional involvement of central cholinergic circuits and visual hallucinations in Parkinson’s disease. Brain. 2009;132(Pt 9):2350–2355.
  • Moccia M, Mollenhauer B, Erro R, Picillo M, Palladino R, Barone P. Non-Motor Correlates of Smoking Habits in de Novo Parkinson’s Disease. J Parkinsons Dis. 2015;5(4):913–924.
  • Kotagal V, Albin RL, Müller ML, Koeppe RA, Frey KA, Bohnen NI. Modifiable cardiovascular risk factors and axial motor impairments in Parkinson disease. Neurology. 2014;82:1514–1520.
  • Costa A, Peppe A, Carlesimo GA et al. Brain-derived neurotrophic factor serum levels correlate with cognitive performance in Parkinson’s disease patients with mild cognitive impairment. Front Behav Neurosci. 2015;9:253.
  • Chen-Plotkin AS, Hu WT, Siderowf A et al. Plasma EGF levels predict cognitive decline in Parkinson’s disease. Ann Neurol. 2011;69(4):655–663.
  • Pellecchia MT, Santangelo G, Picillo M, et al. Serum epidermal growth factor predicts cognitive functions in early, drug-naive Parkinson’s disease patients. J Neurol. 2013;260(2):438–444.
  • Meamar R, Shaabani P, Tabibian SR, Aghaye Ghazvini MR, Feizi A. The effects of uric acid, serum vitamin D3, and their interaction on Parkinson’s disease severity. Parkinsons Dis. Epub 2015 Feb 24.
  • Dufek M, Rektorova I, Thon V, Lokaj J, Rektor I. Interleukin-6 May Contribute to Mortality in Parkinson’s Disease Patients: A 4-Year Prospective Study. Parkinsons Dis. Epub 2015 Aug 16.
  • Hofmann KW, Schuh AFS, Saute J, et al. Interleukin-6 serum levels in patients with Parkinson’s disease. Neurochem Res. 2009;34(8):1401–1404.
  • Tang P, Chong L, Li X, et al. Correlation between serum RANTES levels and the severity of Parkinson’s disease. Oxid Med Cell Longev. 2014;2014:208408.
  • Jiménez-Jiménez F, Molina J, de Bustos F, et al. Serum levels of coenzyme Q10 in patients with Parkinson’s disease. J Neural Transm. 2000;107:177–181.
  • Ascherio A, LeWitt P, Xu K, et al. Urate predicts rate of clinical decline in Parkinsons disease. Arch Neurol. 2009;66(12):1460–1468.
  • Zhang H, Guo J, He D, et al. Lower serum UA levels in Parkinson’s disease patients in the Chinese population. Neurosci Lett. 2012;514(2):152–155.
  • Jesús S, Pérez I, Cáceres-Redondo MT, et al. Low serum uric acid concentration in Parkinson’s disease in southern Spain. Eur J Neurol. 2013;20(1):208–210.
  • Pan M, Gao H, Long L et al. Serum uric acid in patients with Parkinson’s disease and vascular parkinsonism: a cross-sectional study. Neuroimmunomodulation. 2013;20(1):19–28.
  • Lolekha P, Wongwan P, Kulkantrakorn K. Association between serum uric acid and motor subtypes of Parkinson’s disease. J Clin Neurosci. 2015;22(8):1264–1267.
  • Fukae J, Ishikawa K, Hatano T, et al. Serum uric acid concentration is linked to wearing-off fluctuation in Japanese Parkinson’s disease patients. J Parkinsons Dis. 2014;4(3):499–505.
  • Moccia M, Picillo M, Erro R, et al. Presence and progression of non-motor symptoms in relation to uric acid in de novo Parkinson’s disease. Eur J Neurol. 2015;22(1):93–98.
  • Moccia M, Picillo M, Erro R, et al. Is serum uric acid related to non-motor symptoms in de-novo Parkinson’s disease patients? Parkinsonism Relat Disord. 2014;20(7):772–775.
  • Annanmaki T, Pessala-Driver A, Hokkanen L, Murros K. Uric acid associates with cognition in Parkinson’s disease. Parkinsonism Relat Disord. 2008;14(7):576–578.
  • Annanmaki T, Pohja M, Parviainen T, Hakkinen P, Murros K. Uric acid and cognition in Parkinson’s disease: a follow-up study. Parkinsonism Relat Disord. 2011;17(5):333–337.
  • Schwarzschild MA, Ascherio A, Beal MF, et al. Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol. 2014;71(2):141–150.
  • Yu SY, Sun L, Liu Z, et al. Sleep disorders in Parkinson’s disease: clinical features, iron metabolism and related mechanism. PLoS One. 2013;8(12):e82924.
  • Hu Y, Yu SY, Zuo L-J, et al. Investigation on Abnormal Iron Metabolism and Related Inflammation in Parkinson Disease Patients with Probable RBD. PLoS One. 2015;10(10):e0138997.
  • Fukushima T, Tan X, Luo Y, et al. Heavy metals in blood and urine and its relation to depressive symptoms in Parkinson’s disease patients. Fukushima J Med Sci. 2013;59(2):76–80.
  • van Dijk KD, Bidinosti M, Weiss A, Raijmakers P, Berendse HW, van de Berg WDJ. Reduced α-synuclein levels in cerebrospinal fluid in Parkinson’s disease are unrelated to clinical and imaging measures of disease severity. Eur J Neurol. 2014;21(3):388–394.
  • Hall S, Surova Y, Öhrfelt A, Zetterberg H, Lindqvist D, Hansson O. CSF biomarkers and clinical progression of Parkinson disease. Neurology. 2015;84:57–63.
  • Liu C, Cholerton B, Shi M, et al. CSF tau and tau/Aβ42 predict cognitive decline in Parkinson’s disease. Parkinsonism Relat. Disord. 2015;21:271–276.
  • Kang JH, Chen-Plotkin AS., Siderowf A. Association of cerebrospinal fluid β-amyoid 1-42, T-tau, P-tau181, and α-synuclein levels with clinical features of drug-naive patients with early Parkinson disease. JAMA Neurol. 2013;70(10):1277–1287.
  • OldeDubbelink KT, Schoonheim MM, Deijen JB, Twisk JW, Barkhof F, Berendse HW. Functional connectivity and cognitive decline over 3 years in Parkinson disease. Neurology. 2014;83(22):2046–2053.
  • Amboni M, Tessitore A, Esposito F, et al. Resting-state functional connectivity associated with mild cognitive impairment in Parkinson’s disease. J Neurol. 2015;262(2):425–434.
  • Tessitore A, Amboni M, Esposito F, et al. Resting-state brain connectivity in patients with Parkinson’s disease and freezing of gait. Parkinsonism Relat Disord. 2012;18(6):781–787.
  • Jia X, Liang P, Li Y, Shi L, Wang D, Li K. Longitudinal Study of Gray Matter Changes in Parkinson Disease. AJNR Am J Neuroradiol. 2015;36(12):2219–2226.
  • Ibarretxe-Bilbao N, Junque C, Segura B, et al. Progression of cortical thinning in early Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2013;84(8):875–881.
  • Zarei M, Ibarretxe-Bilbao N, Compta Y et al. Cortical thinning is associated with disease stages and dementia in Parkinson’s disease. Mov Disord. 2012;27(14):1746–1753.
  • Ulla M, Bonny JM, Ouchchane L, Rieu I, Claise B, Durif F. Is R2* a new MRI biomarker for the progression of Parkinson’s disease? A longitudinal follow-up. PLoS One. 2013;8(3):e57904.
  • McKeown MJ, Peavy GM. Biomarkers in Parkinson disease: it’s time to combine. Neurology. 2015;84(24):2392–2393.
  • Campbell MC, Koller JM, Snyder AZ, Buddhala C, Kotzbauer PT, Perlmutter JS. CSF proteins and resting-state functional connectivity in Parkinson disease. Neurology. 2015;84(24):2413–2421.
  • Rosenthal LS, Drake D, Alcalay RN, et al. PDBP consortium. The NINDS Parkinson’s disease biomarkers program. Mov Disord. Epub 2015 Oct 7.