Skip to main content
Top
Published in: Translational Neurodegeneration 1/2015

Open Access 01-12-2015 | Review

The associations between Parkinson’s disease and cancer: the plot thickens

Authors: Danielle D. Feng, Waijiao Cai, Xiqun Chen

Published in: Translational Neurodegeneration | Issue 1/2015

Login to get access

Abstract

Epidemiological studies support a general inverse association between the risk of cancer development and Parkinson’s disease (PD). In recent years however, increasing amount of eclectic evidence points to a positive association between PD and cancers through different temporal analyses and ethnic groups. This positive association has been supported by several common genetic mutations in SNCA, PARK2, PARK8, ATM, p53, PTEN, and MC1R resulting in cellular changes such as mitochondrial dysfunction, aberrant protein aggregation, and cell cycle dysregulation. Here, we review the epidemiological and biological advances of the past decade in the association between PD and cancers to offer insight on the recent and sometimes contradictory findings.
Literature
1.
go back to reference de Lau LML, Breteler MMB. Epidemiology of Parkinson's disease. The Lancet Neurology 5.6. 2006;525-535. de Lau LML, Breteler MMB. Epidemiology of Parkinson's disease. The Lancet Neurology 5.6. 2006;525-535.
2.
go back to reference Bajaj A, Driver JA, Schernhammer ES. Parkinson’s disease and cancer risk: a systematic review and meta-analysis. Cancer Causes Control. 2010;21:697–707.PubMedCrossRef Bajaj A, Driver JA, Schernhammer ES. Parkinson’s disease and cancer risk: a systematic review and meta-analysis. Cancer Causes Control. 2010;21:697–707.PubMedCrossRef
3.
go back to reference Catalá-López F, Suárez-Pinilla M, Suárez-Pinilla P, Valderas JM, Gómez-Beneyto M, Martinez S, et al. Inverse and direct cancer comorbidity in people with central nervous system disorders: a meta-analysis of cancer incidence in 577,013 participants of 50 observational studies. Psychother Psychosom. 2014;83:89–105.PubMedCrossRef Catalá-López F, Suárez-Pinilla M, Suárez-Pinilla P, Valderas JM, Gómez-Beneyto M, Martinez S, et al. Inverse and direct cancer comorbidity in people with central nervous system disorders: a meta-analysis of cancer incidence in 577,013 participants of 50 observational studies. Psychother Psychosom. 2014;83:89–105.PubMedCrossRef
4.
go back to reference Olsen JH, Friis S, Frederiksen K. Malignant melanoma and other types of cancer preceding Parkinson disease. Epidemiology. 2006;17:582–7.PubMedCrossRef Olsen JH, Friis S, Frederiksen K. Malignant melanoma and other types of cancer preceding Parkinson disease. Epidemiology. 2006;17:582–7.PubMedCrossRef
5.
go back to reference Wirdefeldt K, Weibull CE, Chen H, Kamel F, Lundholm C, Fang F, et al. Parkinson’s disease and cancer: A register-based family study. Am J Epidemiol. 2014;179:85–94.PubMedCentralPubMedCrossRef Wirdefeldt K, Weibull CE, Chen H, Kamel F, Lundholm C, Fang F, et al. Parkinson’s disease and cancer: A register-based family study. Am J Epidemiol. 2014;179:85–94.PubMedCentralPubMedCrossRef
6.
go back to reference Ong ELH, Goldacre R, Goldacre M. Differential risks of cancer types in people with Parkinson’s disease: a national record-linkage study. Eur J Cancer. 2014;50:2456–62.PubMedCrossRef Ong ELH, Goldacre R, Goldacre M. Differential risks of cancer types in people with Parkinson’s disease: a national record-linkage study. Eur J Cancer. 2014;50:2456–62.PubMedCrossRef
7.
go back to reference Lin P-Y, Chang S-N, Hsiao T-H, Huang B-T, Lin C-H, Yang P-C. Association Between Parkinson Disease and Risk of Cancer in Taiwan. JAMA Oncol. 2015;1(5):633–40.PubMedCrossRef Lin P-Y, Chang S-N, Hsiao T-H, Huang B-T, Lin C-H, Yang P-C. Association Between Parkinson Disease and Risk of Cancer in Taiwan. JAMA Oncol. 2015;1(5):633–40.PubMedCrossRef
8.
go back to reference Sun L-M, Liang J-A, Chang S-N, Sung F-C, Muo C-H, Kao C-H. Analysis of Parkinson’s disease and subsequent cancer risk in Taiwan: a nationwide population-based cohort study. Neuroepidemiology. 2011;37:114–9.PubMedCrossRef Sun L-M, Liang J-A, Chang S-N, Sung F-C, Muo C-H, Kao C-H. Analysis of Parkinson’s disease and subsequent cancer risk in Taiwan: a nationwide population-based cohort study. Neuroepidemiology. 2011;37:114–9.PubMedCrossRef
9.
go back to reference Olsen JH, Friis S, Frederiksen K, McLaughlin JK, Mellemkjaer L, Møller H. Atypical cancer pattern in patients with Parkinson’s disease. Br J Cancer. 2005;92:201–5.PubMedCentralPubMedCrossRef Olsen JH, Friis S, Frederiksen K, McLaughlin JK, Mellemkjaer L, Møller H. Atypical cancer pattern in patients with Parkinson’s disease. Br J Cancer. 2005;92:201–5.PubMedCentralPubMedCrossRef
10.
11.
go back to reference Kareus SA, Figueroa KP, Cannon-Albright LA, Pulst SM. Shared predispositions of parkinsonism and cancer: a population-based pedigree-linked study. Arch Neurol. 2012;69:1572–7.PubMedCrossRef Kareus SA, Figueroa KP, Cannon-Albright LA, Pulst SM. Shared predispositions of parkinsonism and cancer: a population-based pedigree-linked study. Arch Neurol. 2012;69:1572–7.PubMedCrossRef
12.
go back to reference Driver JA, Logroscino G, Buring JE, Gaziano JM, Kurth T. A prospective cohort study of cancer incidence following the diagnosis of Parkinson’s disease. Cancer Epidemiol Biomarkers Prev. 2007;16:1260–5.PubMedCrossRef Driver JA, Logroscino G, Buring JE, Gaziano JM, Kurth T. A prospective cohort study of cancer incidence following the diagnosis of Parkinson’s disease. Cancer Epidemiol Biomarkers Prev. 2007;16:1260–5.PubMedCrossRef
13.
go back to reference Fois AF, Wotton CJ, Yeates D, Turner MR, Goldacre MJ. Cancer in patients with motor neuron disease, multiple sclerosis and Parkinson’s disease: record linkage studies. J Neurol Neurosurg Psychiatry. 2010;81:215–21.PubMedCrossRef Fois AF, Wotton CJ, Yeates D, Turner MR, Goldacre MJ. Cancer in patients with motor neuron disease, multiple sclerosis and Parkinson’s disease: record linkage studies. J Neurol Neurosurg Psychiatry. 2010;81:215–21.PubMedCrossRef
15.
go back to reference Rugbjerg K, Kathrine R, Søren F, Lassen CF, Beate R, Olsen JH. Malignant melanoma, breast cancer and other cancers in patients with Parkinson’s disease. Int J Cancer. 2012;131:1904–11.PubMedCentralPubMedCrossRef Rugbjerg K, Kathrine R, Søren F, Lassen CF, Beate R, Olsen JH. Malignant melanoma, breast cancer and other cancers in patients with Parkinson’s disease. Int J Cancer. 2012;131:1904–11.PubMedCentralPubMedCrossRef
17.
go back to reference Skibba JL, Pinckley J, Gilbert EF, Johnson RO. Multiple primary melanoma following administration of levodopa. Arch Pathol. 1972;93:556–61.PubMed Skibba JL, Pinckley J, Gilbert EF, Johnson RO. Multiple primary melanoma following administration of levodopa. Arch Pathol. 1972;93:556–61.PubMed
18.
go back to reference Vermeij J-D, Winogrodzka A, Trip J, Weber WEJ. Parkinson’s disease, levodopa-use and the risk of melanoma. Parkinsonism Relat Disord. 2009;15:551–3.PubMedCrossRef Vermeij J-D, Winogrodzka A, Trip J, Weber WEJ. Parkinson’s disease, levodopa-use and the risk of melanoma. Parkinsonism Relat Disord. 2009;15:551–3.PubMedCrossRef
19.
go back to reference Zanetti R, Loria D, Rosso S. Melanoma, Parkinson’s disease and levodopa: causal or spurious link? A review of the literature. Melanoma Res. 2006;16:201–6.PubMedCrossRef Zanetti R, Loria D, Rosso S. Melanoma, Parkinson’s disease and levodopa: causal or spurious link? A review of the literature. Melanoma Res. 2006;16:201–6.PubMedCrossRef
20.
go back to reference Pan T, Li X, Jankovic J. The association between Parkinson’s disease and melanoma. Int J Cancer. 2011;128:2251–60.PubMedCrossRef Pan T, Li X, Jankovic J. The association between Parkinson’s disease and melanoma. Int J Cancer. 2011;128:2251–60.PubMedCrossRef
21.
go back to reference Paisán-Ruiz C, Houlden H. Common pathogenic pathways in melanoma and Parkinson disease. Neurology. 2010;75:1653–5.PubMedCrossRef Paisán-Ruiz C, Houlden H. Common pathogenic pathways in melanoma and Parkinson disease. Neurology. 2010;75:1653–5.PubMedCrossRef
23.
go back to reference Chorfa A, Bétemps D, Morignat E, Lazizzera C, Hogeveen K, Andrieu T, et al. Specific pesticide-dependent increases in α-synuclein levels in human neuroblastoma (SH-SY5Y) and melanoma (SK-MEL-2) cell lines. Toxicol Sci. 2013;133:289–97.PubMedCrossRef Chorfa A, Bétemps D, Morignat E, Lazizzera C, Hogeveen K, Andrieu T, et al. Specific pesticide-dependent increases in α-synuclein levels in human neuroblastoma (SH-SY5Y) and melanoma (SK-MEL-2) cell lines. Toxicol Sci. 2013;133:289–97.PubMedCrossRef
24.
go back to reference Noyce AJ, Bestwick JP, Silveira-Moriyama L, Hawkes CH, Giovannoni G, Lees AJ, et al. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann Neurol. 2012;72:893–901.PubMedCentralPubMedCrossRef Noyce AJ, Bestwick JP, Silveira-Moriyama L, Hawkes CH, Giovannoni G, Lees AJ, et al. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann Neurol. 2012;72:893–901.PubMedCentralPubMedCrossRef
25.
go back to reference Freedman DM, Michal Freedman D, Alice S, Doody MM, Sowmya Rao R, Linet MS. Risk of melanoma in relation to smoking, alcohol intake, and other factors in a large occupational cohort. Cancer Causes Control. 2003;14:847–57.PubMedCrossRef Freedman DM, Michal Freedman D, Alice S, Doody MM, Sowmya Rao R, Linet MS. Risk of melanoma in relation to smoking, alcohol intake, and other factors in a large occupational cohort. Cancer Causes Control. 2003;14:847–57.PubMedCrossRef
26.
go back to reference DeLancey JO, Hannan LM, Gapstur SM, Thun MJ. Cigarette smoking and the risk of incident and fatal melanoma in a large prospective cohort study. Cancer Causes Control. 2011;22:937–42.PubMedCrossRef DeLancey JO, Hannan LM, Gapstur SM, Thun MJ. Cigarette smoking and the risk of incident and fatal melanoma in a large prospective cohort study. Cancer Causes Control. 2011;22:937–42.PubMedCrossRef
27.
go back to reference Song F, Qureshi AA, Gao X, Li T, Han J. Smoking and risk of skin cancer: a prospective analysis and a meta-analysis. Int J Epidemiol. 2012;41:1694–705.PubMedCentralPubMedCrossRef Song F, Qureshi AA, Gao X, Li T, Han J. Smoking and risk of skin cancer: a prospective analysis and a meta-analysis. Int J Epidemiol. 2012;41:1694–705.PubMedCentralPubMedCrossRef
28.
go back to reference Kenborg L, Lassen CF, Ritz B, Andersen KK, Christensen J, Schernhammer ES, et al. Lifestyle, family history, and risk of idiopathic Parkinson disease: a large Danish case–control study. Am J Epidemiol. 2015;181:808–16.PubMedCrossRef Kenborg L, Lassen CF, Ritz B, Andersen KK, Christensen J, Schernhammer ES, et al. Lifestyle, family history, and risk of idiopathic Parkinson disease: a large Danish case–control study. Am J Epidemiol. 2015;181:808–16.PubMedCrossRef
29.
go back to reference Van der Mark M, Nijssen PCG, Vlaanderen J, Huss A, Mulleners WM, Sas AMG, et al. A case–control study of the protective effect of alcohol, coffee, and cigarette consumption on Parkinson disease risk: time-since-cessation modifies the effect of tobacco smoking. PLoS One. 2014;9:e95297.PubMedCentralPubMedCrossRef Van der Mark M, Nijssen PCG, Vlaanderen J, Huss A, Mulleners WM, Sas AMG, et al. A case–control study of the protective effect of alcohol, coffee, and cigarette consumption on Parkinson disease risk: time-since-cessation modifies the effect of tobacco smoking. PLoS One. 2014;9:e95297.PubMedCentralPubMedCrossRef
30.
go back to reference Fedorow H, Tribl F, Halliday G, Gerlach M, Riederer P, Double KL. Neuromelanin in human dopamine neurons: comparison with peripheral melanins and relevance to Parkinson’s disease. Prog Neurobiol. 2005;75:109–24.PubMedCrossRef Fedorow H, Tribl F, Halliday G, Gerlach M, Riederer P, Double KL. Neuromelanin in human dopamine neurons: comparison with peripheral melanins and relevance to Parkinson’s disease. Prog Neurobiol. 2005;75:109–24.PubMedCrossRef
31.
go back to reference Sprong H, Degroote S, Claessens T, van Drunen J, Oorschot V, Westerink BH, et al. Glycosphingolipids are required for sorting melanosomal proteins in the Golgi complex. J Cell Biol. 2001;155:369–80.PubMedCentralPubMedCrossRef Sprong H, Degroote S, Claessens T, van Drunen J, Oorschot V, Westerink BH, et al. Glycosphingolipids are required for sorting melanosomal proteins in the Golgi complex. J Cell Biol. 2001;155:369–80.PubMedCentralPubMedCrossRef
32.
go back to reference Paisán-Ruiz C, Li A, Schneider SA, Holton JL, Johnson R, Kidd D, et al. Widespread Lewy body and tau accumulation in childhood and adult onset dystonia-parkinsonism cases with PLA2G6 mutations. Neurobiol Aging. 2012;33:814–23.PubMedCentralPubMedCrossRef Paisán-Ruiz C, Li A, Schneider SA, Holton JL, Johnson R, Kidd D, et al. Widespread Lewy body and tau accumulation in childhood and adult onset dystonia-parkinsonism cases with PLA2G6 mutations. Neurobiol Aging. 2012;33:814–23.PubMedCentralPubMedCrossRef
33.
go back to reference Paisan-Ruiz C, Bhatia KP, Li A, Hernandez D, Davis M, Wood NW, et al. Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann Neurol. 2009;65:19–23.PubMedCrossRef Paisan-Ruiz C, Bhatia KP, Li A, Hernandez D, Davis M, Wood NW, et al. Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann Neurol. 2009;65:19–23.PubMedCrossRef
34.
go back to reference Falchi M, Bataille V, Hayward NK, Duffy DL, Bishop JAN, Pastinen T, et al. Genome-wide association study identifies variants at 9p21 and 22q13 associated with development of cutaneous nevi. Nat Genet. 2009;41:915–9.PubMedCrossRef Falchi M, Bataille V, Hayward NK, Duffy DL, Bishop JAN, Pastinen T, et al. Genome-wide association study identifies variants at 9p21 and 22q13 associated with development of cutaneous nevi. Nat Genet. 2009;41:915–9.PubMedCrossRef
35.
36.
go back to reference Piccinini M, Scandroglio F, Prioni S, Buccinnà B, Loberto N, Aureli M, et al. Deregulated sphingolipid metabolism and membrane organization in neurodegenerative disorders. Mol Neurobiol. 2010;41:314–40.PubMedCrossRef Piccinini M, Scandroglio F, Prioni S, Buccinnà B, Loberto N, Aureli M, et al. Deregulated sphingolipid metabolism and membrane organization in neurodegenerative disorders. Mol Neurobiol. 2010;41:314–40.PubMedCrossRef
37.
go back to reference Zecca L, Luigi Z, Zucca FA, Henrik W, David S. Neuromelanin of the substantia nigra: a neuronal black hole with protective and toxic characteristics. Trends Neurosci. 2003;26:578–80.PubMedCrossRef Zecca L, Luigi Z, Zucca FA, Henrik W, David S. Neuromelanin of the substantia nigra: a neuronal black hole with protective and toxic characteristics. Trends Neurosci. 2003;26:578–80.PubMedCrossRef
38.
go back to reference Yerger VB, Malone RE. Melanin and nicotine: A review of the literature. Nicotine Tob Res. 2006;8:487–98.PubMedCrossRef Yerger VB, Malone RE. Melanin and nicotine: A review of the literature. Nicotine Tob Res. 2006;8:487–98.PubMedCrossRef
40.
go back to reference Rockenstein E, Nuber S, Overk CR, Ubhi K, Mante M, Patrick C, et al. Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo. Brain. 2014;137(Pt 5):1496–513.PubMedCentralPubMedCrossRef Rockenstein E, Nuber S, Overk CR, Ubhi K, Mante M, Patrick C, et al. Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo. Brain. 2014;137(Pt 5):1496–513.PubMedCentralPubMedCrossRef
41.
go back to reference Chandra S, Sreeganga C, Gilbert G, Rafael F-C, Schlüter OM, Südhof TC. α-Synuclein Cooperates with CSPα in Preventing Neurodegeneration. Cell. 2005;123:383–96.PubMedCrossRef Chandra S, Sreeganga C, Gilbert G, Rafael F-C, Schlüter OM, Südhof TC. α-Synuclein Cooperates with CSPα in Preventing Neurodegeneration. Cell. 2005;123:383–96.PubMedCrossRef
42.
go back to reference Burre J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Sudhof TC. Alpha-Synuclein Promotes SNARE-Complex Assembly in Vivo and in Vitro. Science. 2010;329:1663–7.PubMedCentralPubMedCrossRef Burre J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Sudhof TC. Alpha-Synuclein Promotes SNARE-Complex Assembly in Vivo and in Vitro. Science. 2010;329:1663–7.PubMedCentralPubMedCrossRef
43.
go back to reference Burré J, Sharma M, Südhof TC. α-Synuclein assembles into higher-order multimers upon membrane binding to promote SNARE complex formation. Proc Natl Acad Sci U S A. 2014;111:E4274–83.PubMedCentralPubMedCrossRef Burré J, Sharma M, Südhof TC. α-Synuclein assembles into higher-order multimers upon membrane binding to promote SNARE complex formation. Proc Natl Acad Sci U S A. 2014;111:E4274–83.PubMedCentralPubMedCrossRef
45.
go back to reference Pirc K, Ulrih NP. α-Synuclein interactions with phospholipid model membranes: Key roles for electrostatic interactions and lipid-bilayer structure. Biochim Biophys Acta. 2015;1848(10 Pt A):2002–12.PubMedCrossRef Pirc K, Ulrih NP. α-Synuclein interactions with phospholipid model membranes: Key roles for electrostatic interactions and lipid-bilayer structure. Biochim Biophys Acta. 2015;1848(10 Pt A):2002–12.PubMedCrossRef
46.
go back to reference Bartels T, Tim B, Ahlstrom LS, Avigdor L, Frits K, Christian H, et al. The N-Terminus of the Intrinsically Disordered Protein α-Synuclein Triggers Membrane Binding and Helix Folding. Biophys J. 2010;99:2116–24.PubMedCentralPubMedCrossRef Bartels T, Tim B, Ahlstrom LS, Avigdor L, Frits K, Christian H, et al. The N-Terminus of the Intrinsically Disordered Protein α-Synuclein Triggers Membrane Binding and Helix Folding. Biophys J. 2010;99:2116–24.PubMedCentralPubMedCrossRef
47.
go back to reference Bachhuber T, Katzmarski N, McCarter JF, Loreth D, Tahirovic S, Kamp F, et al. Inhibition of amyloid-β plaque formation by α-synuclein. Nat Med. 2015;21:802–7.PubMedCrossRef Bachhuber T, Katzmarski N, McCarter JF, Loreth D, Tahirovic S, Kamp F, et al. Inhibition of amyloid-β plaque formation by α-synuclein. Nat Med. 2015;21:802–7.PubMedCrossRef
48.
go back to reference Fung K-M, Rorke LB, Giasson B, Lee VM-Y, Trojanowski JQ. Expression of alpha-, beta-, and gamma-synuclein in glial tumors and medulloblastomas. Acta Neuropathol. 2003;106:167–75.PubMedCrossRef Fung K-M, Rorke LB, Giasson B, Lee VM-Y, Trojanowski JQ. Expression of alpha-, beta-, and gamma-synuclein in glial tumors and medulloblastomas. Acta Neuropathol. 2003;106:167–75.PubMedCrossRef
49.
go back to reference Bruening W, Giasson BI, Klein-Szanto AJ, Lee VM, Trojanowski JQ, Godwin AK. Synucleins are expressed in the majority of breast and ovarian carcinomas and in preneoplastic lesions of the ovary. Cancer. 2000;88:2154–63.PubMedCrossRef Bruening W, Giasson BI, Klein-Szanto AJ, Lee VM, Trojanowski JQ, Godwin AK. Synucleins are expressed in the majority of breast and ovarian carcinomas and in preneoplastic lesions of the ovary. Cancer. 2000;88:2154–63.PubMedCrossRef
50.
go back to reference Welinder C, Jönsson GB, Ingvar C, Lundgren L, Baldetorp B, Olsson H, et al. Analysis of alpha-synuclein in malignant melanoma - development of a SRM quantification assay. PLoS One. 2014;9:e110804.PubMedCentralPubMedCrossRef Welinder C, Jönsson GB, Ingvar C, Lundgren L, Baldetorp B, Olsson H, et al. Analysis of alpha-synuclein in malignant melanoma - development of a SRM quantification assay. PLoS One. 2014;9:e110804.PubMedCentralPubMedCrossRef
51.
go back to reference Bethge N, Lothe RA, Honne H, Andresen K, Trøen G, Eknæs M, et al. Colorectal cancer DNA methylation marker panel validated with high performance in Non-Hodgkin lymphoma. Epigenetics. 2014;9:428–36.PubMedCentralPubMedCrossRef Bethge N, Lothe RA, Honne H, Andresen K, Trøen G, Eknæs M, et al. Colorectal cancer DNA methylation marker panel validated with high performance in Non-Hodgkin lymphoma. Epigenetics. 2014;9:428–36.PubMedCentralPubMedCrossRef
52.
go back to reference El-Agnaf O, Salem SA, Paleologou KE, Curran MD. Detection of oligomeric forms of α-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006;20(3):419–25.PubMedCrossRef El-Agnaf O, Salem SA, Paleologou KE, Curran MD. Detection of oligomeric forms of α-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006;20(3):419–25.PubMedCrossRef
53.
go back to reference Gao L, Tang H, Nie K, Wang L, Zhao J, Gan R, et al. Cerebrospinal fluid alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: a systematic review and meta-analysis. Int J Neurosci. 2014;125(9):645–54.PubMedCrossRef Gao L, Tang H, Nie K, Wang L, Zhao J, Gan R, et al. Cerebrospinal fluid alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: a systematic review and meta-analysis. Int J Neurosci. 2014;125(9):645–54.PubMedCrossRef
54.
go back to reference Hansson O, Hall S, Ohrfelt A, Zetterberg H. Levels of cerebrospinal fluid α-synuclein oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to. Alzheimers Res Ther. 2014;6(3):25.PubMedCentralPubMedCrossRef Hansson O, Hall S, Ohrfelt A, Zetterberg H. Levels of cerebrospinal fluid α-synuclein oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to. Alzheimers Res Ther. 2014;6(3):25.PubMedCentralPubMedCrossRef
55.
56.
go back to reference Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7:42.PubMedCentralPubMedCrossRef Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7:42.PubMedCentralPubMedCrossRef
57.
go back to reference Lachenal G, Pernet-Gallay K, Chivet M, Hemming FJ, Belly A, Bodon G, et al. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol Cell Neurosci. 2011;46:409–18.PubMedCrossRef Lachenal G, Pernet-Gallay K, Chivet M, Hemming FJ, Belly A, Bodon G, et al. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol Cell Neurosci. 2011;46:409–18.PubMedCrossRef
58.
go back to reference Gray MT, Woulfe JM. Striatal blood–brain barrier permeability in Parkinson’s disease. J Cereb Blood Flow Metab. 2015;35:747–50.PubMedCrossRef Gray MT, Woulfe JM. Striatal blood–brain barrier permeability in Parkinson’s disease. J Cereb Blood Flow Metab. 2015;35:747–50.PubMedCrossRef
59.
go back to reference Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, et al. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–4.PubMedCrossRef Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, et al. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–4.PubMedCrossRef
60.
go back to reference Shimura H, Schlossmacher MG, Hattori N, Frosch MP, Trockenbacher A, Schneider R, et al. Ubiquitination of a new form of alpha-synuclein by parkin from human brain: implications for Parkinson’s disease. Science. 2001;293:263–9.PubMedCrossRef Shimura H, Schlossmacher MG, Hattori N, Frosch MP, Trockenbacher A, Schneider R, et al. Ubiquitination of a new form of alpha-synuclein by parkin from human brain: implications for Parkinson’s disease. Science. 2001;293:263–9.PubMedCrossRef
61.
go back to reference Shimura H, Hattori N, Kubo SI, Mizuno Y, Asakawa S, Minoshima S, et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet. 2000;25:302–5.PubMedCrossRef Shimura H, Hattori N, Kubo SI, Mizuno Y, Asakawa S, Minoshima S, et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet. 2000;25:302–5.PubMedCrossRef
62.
go back to reference Hattori N, Kitada T, Matsumine H, Asakawa S, Yamamura Y, Yoshino H, et al. Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann Neurol. 1998;44:935–41.PubMedCrossRef Hattori N, Kitada T, Matsumine H, Asakawa S, Yamamura Y, Yoshino H, et al. Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann Neurol. 1998;44:935–41.PubMedCrossRef
63.
64.
go back to reference Lücking CB, Dürr A, Bonifati V, Vaughan J, De Michele G, Gasser T, et al. Association between Early-Onset Parkinson’s Disease and Mutations in the Parkin Gene. N Engl J Med. 2000;342:1560–7.PubMedCrossRef Lücking CB, Dürr A, Bonifati V, Vaughan J, De Michele G, Gasser T, et al. Association between Early-Onset Parkinson’s Disease and Mutations in the Parkin Gene. N Engl J Med. 2000;342:1560–7.PubMedCrossRef
65.
go back to reference Oliveira SA, Scott WK, Martin ER, Nance MA, Watts RL, Hubble JP, et al. Parkin mutations and susceptibility alleles in late-onset Parkinson’s disease. Ann Neurol. 2003;53:624–9.PubMedCrossRef Oliveira SA, Scott WK, Martin ER, Nance MA, Watts RL, Hubble JP, et al. Parkin mutations and susceptibility alleles in late-onset Parkinson’s disease. Ann Neurol. 2003;53:624–9.PubMedCrossRef
66.
go back to reference Foroud T, Uniacke SK, Liu L, Pankratz N, Rudolph A, Halter C, et al. Heterozygosity for a mutation in the parkin gene leads to later onset Parkinson disease. Neurology. 2003;60:796–801.PubMedCrossRef Foroud T, Uniacke SK, Liu L, Pankratz N, Rudolph A, Halter C, et al. Heterozygosity for a mutation in the parkin gene leads to later onset Parkinson disease. Neurology. 2003;60:796–801.PubMedCrossRef
67.
go back to reference Alcalay RN, Clark LN, Marder KS, Bradley WEC. Lack of association between cancer history and PARKIN genotype: a family based study in PARKIN/Parkinson’s families. Genes Chromosomes Cancer. 2012;51:1109–13.PubMedCentralPubMedCrossRef Alcalay RN, Clark LN, Marder KS, Bradley WEC. Lack of association between cancer history and PARKIN genotype: a family based study in PARKIN/Parkinson’s families. Genes Chromosomes Cancer. 2012;51:1109–13.PubMedCentralPubMedCrossRef
68.
go back to reference Xiong D, Wang Y, You M. PARK2 gene and familial lung cancer: what is the link? Future Oncol. 2015;11:1707–10.PubMedCrossRef Xiong D, Wang Y, You M. PARK2 gene and familial lung cancer: what is the link? Future Oncol. 2015;11:1707–10.PubMedCrossRef
69.
go back to reference Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I, et al. Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet. 2010;42:77–82.PubMedCentralPubMedCrossRef Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I, et al. Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet. 2010;42:77–82.PubMedCentralPubMedCrossRef
70.
go back to reference Xu L, Lin D-C, Yin D, Koeffler HP. An emerging role of PARK2 in cancer. J Mol Med. 2014;92:31–42.PubMedCrossRef Xu L, Lin D-C, Yin D, Koeffler HP. An emerging role of PARK2 in cancer. J Mol Med. 2014;92:31–42.PubMedCrossRef
71.
go back to reference Gong Y, Zack TI, Morris LGT, Lin K, Hukkelhoven E, Raheja R, et al. Pan-cancer genetic analysis identifies PARK2 as a master regulator of G1/S cyclins. Nat Genet. 2014;46:588–94.PubMedCentralPubMedCrossRef Gong Y, Zack TI, Morris LGT, Lin K, Hukkelhoven E, Raheja R, et al. Pan-cancer genetic analysis identifies PARK2 as a master regulator of G1/S cyclins. Nat Genet. 2014;46:588–94.PubMedCentralPubMedCrossRef
72.
go back to reference Ohtsubo M, Theodoras AM, Schumacher J, Roberts JM, Pagano M. Human cyclin E, a nuclear protein essential for the G1-to-S phase transition. Mol Cell Biol. 1995;15:2612–24.PubMedCentralPubMedCrossRef Ohtsubo M, Theodoras AM, Schumacher J, Roberts JM, Pagano M. Human cyclin E, a nuclear protein essential for the G1-to-S phase transition. Mol Cell Biol. 1995;15:2612–24.PubMedCentralPubMedCrossRef
73.
go back to reference Lin D-C, Xu L, Chen Y, Yan H, Hazawa M, Doan N, et al. Genomic and Functional Analysis of the E3 Ligase PARK2 in Glioma. Cancer Res. 2015;75:1815–27.PubMedCrossRef Lin D-C, Xu L, Chen Y, Yan H, Hazawa M, Doan N, et al. Genomic and Functional Analysis of the E3 Ligase PARK2 in Glioma. Cancer Res. 2015;75:1815–27.PubMedCrossRef
74.
go back to reference Staropoli JF, Caroline M, Cécile M, Brenda S, Elena D, Asa A. Parkin Is a Component of an SCF-like Ubiquitin Ligase Complex and Protects Postmitotic Neurons from Kainate Excitotoxicity. Neuron. 2003;37:735–49.PubMedCrossRef Staropoli JF, Caroline M, Cécile M, Brenda S, Elena D, Asa A. Parkin Is a Component of an SCF-like Ubiquitin Ligase Complex and Protects Postmitotic Neurons from Kainate Excitotoxicity. Neuron. 2003;37:735–49.PubMedCrossRef
75.
go back to reference Harbour JW, Luo RX, Dei Santi A, Postigo AA, Dean DC. Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1. Cell. 1999;98:859–69.PubMedCrossRef Harbour JW, Luo RX, Dei Santi A, Postigo AA, Dean DC. Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1. Cell. 1999;98:859–69.PubMedCrossRef
76.
go back to reference L’Episcopo F, Tirolo C, Caniglia S, Testa N, Morale MC, Serapide MF, et al. Targeting Wnt signaling at the neuroimmune interface for dopaminergic neuroprotection/repair in Parkinson’s disease. J Mol Cell Biol. 2014;6:13–26.PubMedCentralPubMedCrossRef L’Episcopo F, Tirolo C, Caniglia S, Testa N, Morale MC, Serapide MF, et al. Targeting Wnt signaling at the neuroimmune interface for dopaminergic neuroprotection/repair in Parkinson’s disease. J Mol Cell Biol. 2014;6:13–26.PubMedCentralPubMedCrossRef
77.
go back to reference Fallon L, Bélanger CML, Corera AT, Kontogiannea M, Regan-Klapisz E, Moreau F, et al. A regulated interaction with the UIM protein Eps15 implicates parkin in EGF receptor trafficking and PI(3)K-Akt signalling. Nat Cell Biol. 2006;8:834–42.PubMedCrossRef Fallon L, Bélanger CML, Corera AT, Kontogiannea M, Regan-Klapisz E, Moreau F, et al. A regulated interaction with the UIM protein Eps15 implicates parkin in EGF receptor trafficking and PI(3)K-Akt signalling. Nat Cell Biol. 2006;8:834–42.PubMedCrossRef
78.
79.
go back to reference Inzelberg R, Hassin-Baer S, Jankovic J. Genetic movement disorders in patients of Jewish ancestry. JAMA Neurol. 2014;71:1567–72.PubMedCrossRef Inzelberg R, Hassin-Baer S, Jankovic J. Genetic movement disorders in patients of Jewish ancestry. JAMA Neurol. 2014;71:1567–72.PubMedCrossRef
80.
go back to reference Di Fonzo A, Yah-Huei W-C, Chin-Song L, van Doeselaar M, Simons EJ, Rohé CF, et al. A common missense variant in the LRRK2 gene, Gly2385Arg, associated with Parkinson’s disease risk in Taiwan. Neurogenetics. 2006;7:133–8.PubMedCrossRef Di Fonzo A, Yah-Huei W-C, Chin-Song L, van Doeselaar M, Simons EJ, Rohé CF, et al. A common missense variant in the LRRK2 gene, Gly2385Arg, associated with Parkinson’s disease risk in Taiwan. Neurogenetics. 2006;7:133–8.PubMedCrossRef
81.
go back to reference Cirnaru MD, Marte A, Belluzzi E, Russo I, Gabrielli M, Longo F, et al. LRRK2 kinase activity regulates synaptic vesicle trafficking and neurotransmitter release through modulation of LRRK2 macro-molecular complex. Front Mol Neurosci. 2014;7:49.PubMedCentralPubMedCrossRef Cirnaru MD, Marte A, Belluzzi E, Russo I, Gabrielli M, Longo F, et al. LRRK2 kinase activity regulates synaptic vesicle trafficking and neurotransmitter release through modulation of LRRK2 macro-molecular complex. Front Mol Neurosci. 2014;7:49.PubMedCentralPubMedCrossRef
82.
go back to reference Piccoli G, Condliffe SB, Bauer M, Giesert F, Boldt K, De Astis S, et al. LRRK2 Controls Synaptic Vesicle Storage and Mobilization within the Recycling Pool. J Neurosci. 2011;31:2225–37.PubMedCrossRef Piccoli G, Condliffe SB, Bauer M, Giesert F, Boldt K, De Astis S, et al. LRRK2 Controls Synaptic Vesicle Storage and Mobilization within the Recycling Pool. J Neurosci. 2011;31:2225–37.PubMedCrossRef
84.
go back to reference Kawashima M, Masatou K, Suzuki SO, Katsumi D-U, Toru I. α-Synuclein is expressed in a variety of brain tumors showing neuronal differentiation. Acta Neuropathol 2000;99:154–160.PubMedCrossRef Kawashima M, Masatou K, Suzuki SO, Katsumi D-U, Toru I. α-Synuclein is expressed in a variety of brain tumors showing neuronal differentiation. Acta Neuropathol 2000;99:154–160.PubMedCrossRef
85.
go back to reference Nichols RJ, Dzamko N, Morrice NA, Campbell DG, Deak M, Ordureau A, et al. 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson’s disease-associated mutations and regulates cytoplasmic localization. Biochem J. 2010;430:393–404.PubMedCentralPubMedCrossRef Nichols RJ, Dzamko N, Morrice NA, Campbell DG, Deak M, Ordureau A, et al. 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson’s disease-associated mutations and regulates cytoplasmic localization. Biochem J. 2010;430:393–404.PubMedCentralPubMedCrossRef
86.
go back to reference Guerreiro PS, Ellen G, da Fonseca TL, Mathias B, Outeiro TF, Katrin E: LRRK2 Promotes Tau Accumulation, Aggregation and Release. Mol Neurobiol 2015. [Epub ahead of print] Guerreiro PS, Ellen G, da Fonseca TL, Mathias B, Outeiro TF, Katrin E: LRRK2 Promotes Tau Accumulation, Aggregation and Release. Mol Neurobiol 2015. [Epub ahead of print]
87.
go back to reference Lesage S, Dürr A, Tazir M, Lohmann E, Leutenegger A-L, Janin S, et al. LRRK2 G2019S as a cause of Parkinson’s disease in North African Arabs. N Engl J Med. 2006;354:422–3.PubMedCrossRef Lesage S, Dürr A, Tazir M, Lohmann E, Leutenegger A-L, Janin S, et al. LRRK2 G2019S as a cause of Parkinson’s disease in North African Arabs. N Engl J Med. 2006;354:422–3.PubMedCrossRef
88.
go back to reference Ozelius LJ, Senthil G, Saunders-Pullman R, Ohmann E, Deligtisch A, Tagliati M, et al. LRRK2 G2019S as a cause of Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2006;354:424–5.PubMedCrossRef Ozelius LJ, Senthil G, Saunders-Pullman R, Ohmann E, Deligtisch A, Tagliati M, et al. LRRK2 G2019S as a cause of Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2006;354:424–5.PubMedCrossRef
90.
go back to reference Beccano-Kelly DA, Kuhlmann N, Tatarnikov I, Volta M, Munsie LN, Chou P, et al. Synaptic function is modulated by LRRK2 and glutamate release is increased in cortical neurons of G2019S LRRK2 knock-in mice. Front Cell Neurosci. 2014;8:301.PubMedCentralPubMedCrossRef Beccano-Kelly DA, Kuhlmann N, Tatarnikov I, Volta M, Munsie LN, Chou P, et al. Synaptic function is modulated by LRRK2 and glutamate release is increased in cortical neurons of G2019S LRRK2 knock-in mice. Front Cell Neurosci. 2014;8:301.PubMedCentralPubMedCrossRef
91.
go back to reference Caesar M, Felk S, Zach S, Brønstad G, Aasly JO, Gasser T, et al. Changes in matrix metalloprotease activity and progranulin levels may contribute to the pathophysiological function of mutant leucine-rich repeat kinase 2. Glia. 2014;62:1075–92.PubMedCrossRef Caesar M, Felk S, Zach S, Brønstad G, Aasly JO, Gasser T, et al. Changes in matrix metalloprotease activity and progranulin levels may contribute to the pathophysiological function of mutant leucine-rich repeat kinase 2. Glia. 2014;62:1075–92.PubMedCrossRef
92.
go back to reference Van Damme P, Van Hoecke A, Lambrechts D, Vanacker P, Bogaert E, van Swieten J, et al. Progranulin functions as a neurotrophic factor to regulate neurite outgrowth and enhance neuronal survival. J Cell Biol. 2008;181:37–41.PubMedCentralPubMedCrossRef Van Damme P, Van Hoecke A, Lambrechts D, Vanacker P, Bogaert E, van Swieten J, et al. Progranulin functions as a neurotrophic factor to regulate neurite outgrowth and enhance neuronal survival. J Cell Biol. 2008;181:37–41.PubMedCentralPubMedCrossRef
93.
go back to reference Hockey LN, Kilpatrick BS, Eden ER, Lin-Moshier Y, Brailoiu GC, Brailoiu E, et al. Dysregulation of lysosomal morphology by pathogenic LRRK2 is corrected by TPC2 inhibition. J Cell Sci. 2015;128:232–8.PubMedCentralPubMedCrossRef Hockey LN, Kilpatrick BS, Eden ER, Lin-Moshier Y, Brailoiu GC, Brailoiu E, et al. Dysregulation of lysosomal morphology by pathogenic LRRK2 is corrected by TPC2 inhibition. J Cell Sci. 2015;128:232–8.PubMedCentralPubMedCrossRef
94.
go back to reference Saha S, Shamol S, Ash PEA, Vivek G, Liqun L, Orian S, et al. Mutations in LRRK2 potentiate age-related impairment of autophagic flux. Mol Neurodegener. 2015;10:26.PubMedPubMedCentralCrossRef Saha S, Shamol S, Ash PEA, Vivek G, Liqun L, Orian S, et al. Mutations in LRRK2 potentiate age-related impairment of autophagic flux. Mol Neurodegener. 2015;10:26.PubMedPubMedCentralCrossRef
95.
go back to reference Ramonet D, Daher JPL, Lin BM, Stafa K, Kim J, Banerjee R, et al. Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2. PLoS One. 2011;6:e18568.PubMedCentralPubMedCrossRef Ramonet D, Daher JPL, Lin BM, Stafa K, Kim J, Banerjee R, et al. Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2. PLoS One. 2011;6:e18568.PubMedCentralPubMedCrossRef
96.
go back to reference Inzelberg R, Cohen OS, Aharon-Peretz J, Schlesinger I, Gershoni-Baruch R, Djaldetti R, et al. The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers. Neurology. 2012;78:781–6.PubMedCrossRef Inzelberg R, Cohen OS, Aharon-Peretz J, Schlesinger I, Gershoni-Baruch R, Djaldetti R, et al. The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers. Neurology. 2012;78:781–6.PubMedCrossRef
97.
go back to reference Agalliu I, San Luciano M, Mirelman A, Giladi N, Waro B, Aasly J, et al. Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis. JAMA Neurol. 2015;72:58–65.PubMedPubMedCentralCrossRef Agalliu I, San Luciano M, Mirelman A, Giladi N, Waro B, Aasly J, et al. Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis. JAMA Neurol. 2015;72:58–65.PubMedPubMedCentralCrossRef
98.
go back to reference Allegra R, Tunesi S, Cilia R, Pezzoli G, Goldwurm S. LRRK2-G2019S mutation is not associated with an increased cancer risk: A kin-cohort study. Mov Disord. 2014;29:1325–6.PubMedCrossRef Allegra R, Tunesi S, Cilia R, Pezzoli G, Goldwurm S. LRRK2-G2019S mutation is not associated with an increased cancer risk: A kin-cohort study. Mov Disord. 2014;29:1325–6.PubMedCrossRef
99.
go back to reference Saunders-Pullman R, Barrett MJ, Stanley KM, Luciano MS, Shanker V, Severt L, et al. LRRK2 G2019S mutations are associated with an increased cancer risk in Parkinson disease. Mov Disord. 2010;25:2536–41.PubMedCentralPubMedCrossRef Saunders-Pullman R, Barrett MJ, Stanley KM, Luciano MS, Shanker V, Severt L, et al. LRRK2 G2019S mutations are associated with an increased cancer risk in Parkinson disease. Mov Disord. 2010;25:2536–41.PubMedCentralPubMedCrossRef
100.
go back to reference Ruiz-Martínez J, de la Riva P, Rodríguez-Oroz MC, Mondragón Rezola E, Bergareche A, Gorostidi A, et al. Prevalence of cancer in Parkinson’s disease related to R1441G and G2019S mutations in LRRK2. Mov Disord. 2014;29:750–5.PubMedCrossRef Ruiz-Martínez J, de la Riva P, Rodríguez-Oroz MC, Mondragón Rezola E, Bergareche A, Gorostidi A, et al. Prevalence of cancer in Parkinson’s disease related to R1441G and G2019S mutations in LRRK2. Mov Disord. 2014;29:750–5.PubMedCrossRef
101.
go back to reference Kondo K, Obitsu S, Teshima R. ALPHA.-Synuclein Aggregation and Transmission Are Enhanced by Leucine-Rich Repeat Kinase 2 in Human Neuroblastoma SH-SY5Y Cells. Biol Pharm Bull. 2011;34:1078–83.PubMedCrossRef Kondo K, Obitsu S, Teshima R. ALPHA.-Synuclein Aggregation and Transmission Are Enhanced by Leucine-Rich Repeat Kinase 2 in Human Neuroblastoma SH-SY5Y Cells. Biol Pharm Bull. 2011;34:1078–83.PubMedCrossRef
102.
go back to reference Looyenga BD, Furge KA, Dykema KJ, Koeman J, Swiatek PJ, Giordano TJ, et al. Chromosomal amplification of leucine-rich repeat kinase-2 (LRRK2) is required for oncogenic MET signaling in papillary renal and thyroid carcinomas. Proc Natl Acad Sci U S A. 2011;108:1439–44.PubMedCentralPubMedCrossRef Looyenga BD, Furge KA, Dykema KJ, Koeman J, Swiatek PJ, Giordano TJ, et al. Chromosomal amplification of leucine-rich repeat kinase-2 (LRRK2) is required for oncogenic MET signaling in papillary renal and thyroid carcinomas. Proc Natl Acad Sci U S A. 2011;108:1439–44.PubMedCentralPubMedCrossRef
103.
go back to reference Bravo-San Pedro JM, Niso-Santano M, Gómez-Sánchez R, Pizarro-Estrella E, Aiastui-Pujana A, Gorostidi A, et al. The LRRK2 G2019S mutant exacerbates basal autophagy through activation of the MEK/ERK pathway. Cell Mol Life Sci. 2013;70:121–36.PubMedCrossRef Bravo-San Pedro JM, Niso-Santano M, Gómez-Sánchez R, Pizarro-Estrella E, Aiastui-Pujana A, Gorostidi A, et al. The LRRK2 G2019S mutant exacerbates basal autophagy through activation of the MEK/ERK pathway. Cell Mol Life Sci. 2013;70:121–36.PubMedCrossRef
104.
105.
go back to reference Lee Y, McKinnon PJ. Responding to DNA double strand breaks in the nervous system. Neuroscience. 2007;145:1365–74.PubMedCrossRef Lee Y, McKinnon PJ. Responding to DNA double strand breaks in the nervous system. Neuroscience. 2007;145:1365–74.PubMedCrossRef
106.
go back to reference Kastan MB, Zhan Q, El-Deiry WS, Carrier F, Jacks T, Walsh WV, et al. A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-telangiectasia. Cell. 1992;71:587–97.PubMedCrossRef Kastan MB, Zhan Q, El-Deiry WS, Carrier F, Jacks T, Walsh WV, et al. A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-telangiectasia. Cell. 1992;71:587–97.PubMedCrossRef
107.
go back to reference Matsuoka S, Rotman G, Ogawa A, Shiloh Y, Tamai K, Elledge SJ. ATM phosphorylates Chk2 in vivo and in vitro. Proc Natl Acad Sci U S A. 2000;97:10389–94.PubMedCentralPubMedCrossRef Matsuoka S, Rotman G, Ogawa A, Shiloh Y, Tamai K, Elledge SJ. ATM phosphorylates Chk2 in vivo and in vitro. Proc Natl Acad Sci U S A. 2000;97:10389–94.PubMedCentralPubMedCrossRef
108.
go back to reference Shiloh Y. ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer. 2003;3:155–68.PubMedCrossRef Shiloh Y. ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer. 2003;3:155–68.PubMedCrossRef
109.
go back to reference Friesner JD, Liu B, Culligan K, Britt AB. Ionizing Radiation–dependent γ-H2AX Focus Formation Requires Ataxia Telangiectasia Mutated and Ataxia Telangiectasia Mutated and Rad3-related. Mol Biol Cell. 2005;16:2566–76.PubMedCentralPubMedCrossRef Friesner JD, Liu B, Culligan K, Britt AB. Ionizing Radiation–dependent γ-H2AX Focus Formation Requires Ataxia Telangiectasia Mutated and Ataxia Telangiectasia Mutated and Rad3-related. Mol Biol Cell. 2005;16:2566–76.PubMedCentralPubMedCrossRef
110.
go back to reference Renwick A, Thompson D, Seal S, Kelly P, Chagtai T, Ahmed M, et al. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet. 2006;38:873–5.PubMedCrossRef Renwick A, Thompson D, Seal S, Kelly P, Chagtai T, Ahmed M, et al. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet. 2006;38:873–5.PubMedCrossRef
111.
go back to reference Gumy-Pause F, Wacker P, Sappino A-P. ATM gene and lymphoid malignancies. Leukemia. 2004;18:238–42.PubMedCrossRef Gumy-Pause F, Wacker P, Sappino A-P. ATM gene and lymphoid malignancies. Leukemia. 2004;18:238–42.PubMedCrossRef
112.
go back to reference Camins A, Pizarro JG, Alvira D, Gutierrez-Cuesta J, de la Torre AV, Folch J, et al. Activation of ataxia telangiectasia muted under experimental models and human Parkinson’s disease. Cell Mol Life Sci. 2010;67:3865–82.PubMedCrossRef Camins A, Pizarro JG, Alvira D, Gutierrez-Cuesta J, de la Torre AV, Folch J, et al. Activation of ataxia telangiectasia muted under experimental models and human Parkinson’s disease. Cell Mol Life Sci. 2010;67:3865–82.PubMedCrossRef
113.
go back to reference Petersen AJ, Rimkus SA, Wassarman DA. ATM kinase inhibition in glial cells activates the innate immune response and causes neurodegeneration in Drosophila. Proc Natl Acad Sci U S A. 2012;109:E656–64.PubMedCentralPubMedCrossRef Petersen AJ, Rimkus SA, Wassarman DA. ATM kinase inhibition in glial cells activates the innate immune response and causes neurodegeneration in Drosophila. Proc Natl Acad Sci U S A. 2012;109:E656–64.PubMedCentralPubMedCrossRef
114.
go back to reference Sunico CR, Nakamura T, Rockenstein E, Mante M, Adame A, Chan SF, et al. S-Nitrosylation of parkin as a novel regulator of p53-mediated neuronal cell death in sporadic Parkinson’s disease. Mol Neurodegener. 2013;8:29.PubMedCentralPubMedCrossRef Sunico CR, Nakamura T, Rockenstein E, Mante M, Adame A, Chan SF, et al. S-Nitrosylation of parkin as a novel regulator of p53-mediated neuronal cell death in sporadic Parkinson’s disease. Mol Neurodegener. 2013;8:29.PubMedCentralPubMedCrossRef
115.
go back to reference Jayadev S, Nesser NK, Hopkins S, Myers SJ, Case A, Lee RJ, et al. Transcription factor p53 influences microglial activation phenotype. Glia. 2011;59:1402–13.PubMedCentralPubMedCrossRef Jayadev S, Nesser NK, Hopkins S, Myers SJ, Case A, Lee RJ, et al. Transcription factor p53 influences microglial activation phenotype. Glia. 2011;59:1402–13.PubMedCentralPubMedCrossRef
116.
go back to reference Davenport CM, Sevastou IG, Hooper C, Pocock JM. Inhibiting p53 pathways in microglia attenuates microglial-evoked neurotoxicity following exposure to Alzheimer peptides. J Neurochem. 2010;112:552–63.PubMedCrossRef Davenport CM, Sevastou IG, Hooper C, Pocock JM. Inhibiting p53 pathways in microglia attenuates microglial-evoked neurotoxicity following exposure to Alzheimer peptides. J Neurochem. 2010;112:552–63.PubMedCrossRef
117.
go back to reference Li J, Ghiani CA, Kim JY, Liu A, Sandoval J, DeVellis J, et al. Inhibition of p53 transcriptional activity: a potential target for future development of therapeutic strategies for primary demyelination. J Neurosci. 2008;28:6118–27.PubMedCentralPubMedCrossRef Li J, Ghiani CA, Kim JY, Liu A, Sandoval J, DeVellis J, et al. Inhibition of p53 transcriptional activity: a potential target for future development of therapeutic strategies for primary demyelination. J Neurosci. 2008;28:6118–27.PubMedCentralPubMedCrossRef
118.
go back to reference Jebelli J, Hooper C, Pocock JM. Microglial p53 activation is detrimental to neuronal synapses during activation-induced inflammation: Implications for neurodegeneration. Neurosci Lett. 2014;583:92–7.PubMedCrossRef Jebelli J, Hooper C, Pocock JM. Microglial p53 activation is detrimental to neuronal synapses during activation-induced inflammation: Implications for neurodegeneration. Neurosci Lett. 2014;583:92–7.PubMedCrossRef
120.
go back to reference Desplats P, Spencer B, Crews L, Pathel P, Morvinski-Friedmann D, Kosberg K, et al. Synuclein Induces Alterations in Adult Neurogenesis in Parkinson Disease Models via p53-mediated Repression of Notch1. J Biol Chem. 2012;287:31691–702.PubMedCentralPubMedCrossRef Desplats P, Spencer B, Crews L, Pathel P, Morvinski-Friedmann D, Kosberg K, et al. Synuclein Induces Alterations in Adult Neurogenesis in Parkinson Disease Models via p53-mediated Repression of Notch1. J Biol Chem. 2012;287:31691–702.PubMedCentralPubMedCrossRef
121.
go back to reference Jacobs WB, Kaplan DR, Miller FD. The p53 family in nervous system development and disease. J Neurochem. 2006;97:1571–84.PubMedCrossRef Jacobs WB, Kaplan DR, Miller FD. The p53 family in nervous system development and disease. J Neurochem. 2006;97:1571–84.PubMedCrossRef
122.
go back to reference Lanni C, Racchi M, Memo M, Govoni S, Uberti D. p53 at the crossroads between cancer and neurodegeneration. Free Radic Biol Med. 2012;52:1727–33.PubMedCrossRef Lanni C, Racchi M, Memo M, Govoni S, Uberti D. p53 at the crossroads between cancer and neurodegeneration. Free Radic Biol Med. 2012;52:1727–33.PubMedCrossRef
123.
go back to reference Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem. 1998;273:13375–8.PubMedCrossRef Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem. 1998;273:13375–8.PubMedCrossRef
124.
go back to reference Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997;275:1943–7.PubMedCrossRef Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997;275:1943–7.PubMedCrossRef
125.
go back to reference Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 1997;15:356–62.PubMedCrossRef Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 1997;15:356–62.PubMedCrossRef
126.
go back to reference Myers MP, Pass I, Batty IH, Van der Kaay J, Stolarov JP, Hemmings BA, et al. The lipid phosphatase activity of PTEN is critical for its tumor supressor function. Proc Natl Acad Sci U S A. 1998;95:13513–8.PubMedCentralPubMedCrossRef Myers MP, Pass I, Batty IH, Van der Kaay J, Stolarov JP, Hemmings BA, et al. The lipid phosphatase activity of PTEN is critical for its tumor supressor function. Proc Natl Acad Sci U S A. 1998;95:13513–8.PubMedCentralPubMedCrossRef
127.
go back to reference Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y, Chandarlapaty S, et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell. 2011;19:575–86.PubMedCentralPubMedCrossRef Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y, Chandarlapaty S, et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell. 2011;19:575–86.PubMedCentralPubMedCrossRef
128.
go back to reference Tan M-H, Mester JL, Ngeow J, Rybicki LA, Orloff MS, Eng C. Lifetime cancer risks in individuals with germline PTEN mutations. Clin Cancer Res. 2012;18:400–7.PubMedCentralPubMedCrossRef Tan M-H, Mester JL, Ngeow J, Rybicki LA, Orloff MS, Eng C. Lifetime cancer risks in individuals with germline PTEN mutations. Clin Cancer Res. 2012;18:400–7.PubMedCentralPubMedCrossRef
129.
go back to reference Song MS, Salmena L, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol. 2012;13:283–96.PubMed Song MS, Salmena L, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol. 2012;13:283–96.PubMed
130.
go back to reference Lachyankar MB, Sultana N, Schonhoff CM, Mitra P, Poluha W, Lambert S, et al. A role for nuclear PTEN in neuronal differentiation. J Neurosci. 2000;20:1404–13.PubMed Lachyankar MB, Sultana N, Schonhoff CM, Mitra P, Poluha W, Lambert S, et al. A role for nuclear PTEN in neuronal differentiation. J Neurosci. 2000;20:1404–13.PubMed
131.
go back to reference Sano T, Lin H, Chen X, Langford LA, Koul D, Bondy ML, et al. Differential expression of MMAC/PTEN in glioblastoma multiforme: relationship to localization and prognosis. Cancer Res. 1999;59:1820–4.PubMed Sano T, Lin H, Chen X, Langford LA, Koul D, Bondy ML, et al. Differential expression of MMAC/PTEN in glioblastoma multiforme: relationship to localization and prognosis. Cancer Res. 1999;59:1820–4.PubMed
132.
go back to reference Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nat Genet. 1998;19:348–55.PubMedCrossRef Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nat Genet. 1998;19:348–55.PubMedCrossRef
133.
go back to reference Groszer M, Erickson R, Scripture-Adams DD, Lesche R, Trumpp A, Zack JA, et al. Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo. Science. 2001;294:2186–9.PubMedCrossRef Groszer M, Erickson R, Scripture-Adams DD, Lesche R, Trumpp A, Zack JA, et al. Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo. Science. 2001;294:2186–9.PubMedCrossRef
134.
go back to reference Backman SA, Stambolic V, Suzuki A, Haight J, Elia A, Pretorius J, et al. Deletion of Pten in mouse brain causes seizures, ataxia and defects in soma size resembling Lhermitte-Duclos disease. Nat Genet. 2001;29:396–403.PubMedCrossRef Backman SA, Stambolic V, Suzuki A, Haight J, Elia A, Pretorius J, et al. Deletion of Pten in mouse brain causes seizures, ataxia and defects in soma size resembling Lhermitte-Duclos disease. Nat Genet. 2001;29:396–403.PubMedCrossRef
135.
go back to reference Kwon CH, Zhu X, Zhang J, Knoop LL, Tharp R, Smeyne RJ, et al. Pten regulates neuronal soma size: a mouse model of Lhermitte-Duclos disease. Nat Genet. 2001;29:404–11.PubMedCrossRef Kwon CH, Zhu X, Zhang J, Knoop LL, Tharp R, Smeyne RJ, et al. Pten regulates neuronal soma size: a mouse model of Lhermitte-Duclos disease. Nat Genet. 2001;29:404–11.PubMedCrossRef
136.
go back to reference Park KK, Liu K, Hu Y, Smith PD, Wang C, Cai B, et al. Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science. 2008;322:963–6.PubMedCentralPubMedCrossRef Park KK, Liu K, Hu Y, Smith PD, Wang C, Cai B, et al. Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science. 2008;322:963–6.PubMedCentralPubMedCrossRef
137.
go back to reference Perandones C, Costanzo RV, Kowaljow V, Pivetta OH, Carminatti H, Radrizzani M. Correlation between synaptogenesis and the PTEN phosphatase expression in dendrites during postnatal brain development. Brain Res Mol Brain Res. 2004;128:8–19.PubMedCrossRef Perandones C, Costanzo RV, Kowaljow V, Pivetta OH, Carminatti H, Radrizzani M. Correlation between synaptogenesis and the PTEN phosphatase expression in dendrites during postnatal brain development. Brain Res Mol Brain Res. 2004;128:8–19.PubMedCrossRef
139.
go back to reference Malagelada C, Jin ZH, Jackson-Lewis V, Przedborski S, Greene LA. Rapamycin Protects against Neuron Death in In Vitro andIn Vivo Models of Parkinson’s Disease. J Neurosci. 2010;30:1166–75.PubMedCentralPubMedCrossRef Malagelada C, Jin ZH, Jackson-Lewis V, Przedborski S, Greene LA. Rapamycin Protects against Neuron Death in In Vitro andIn Vivo Models of Parkinson’s Disease. J Neurosci. 2010;30:1166–75.PubMedCentralPubMedCrossRef
140.
go back to reference Bové J, Martínez-Vicente M, Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci. 2011;12:437–52.PubMedCrossRef Bové J, Martínez-Vicente M, Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci. 2011;12:437–52.PubMedCrossRef
141.
go back to reference Valente EM, Abou-Sleiman PM, Caputo V, Muqit MMK, Harvey K, Gispert S, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 2004;304:1158–60.PubMedCrossRef Valente EM, Abou-Sleiman PM, Caputo V, Muqit MMK, Harvey K, Gispert S, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 2004;304:1158–60.PubMedCrossRef
142.
go back to reference Rogaeva E, Johnson J, Lang AE, Gulick C, Gwinn-Hardy K, Kawarai T, et al. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch Neurol. 2004;61:1898–904.PubMedCrossRef Rogaeva E, Johnson J, Lang AE, Gulick C, Gwinn-Hardy K, Kawarai T, et al. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch Neurol. 2004;61:1898–904.PubMedCrossRef
143.
go back to reference Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, Valent A, et al. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90:809–19.PubMedCrossRef Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, Valent A, et al. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90:809–19.PubMedCrossRef
145.
go back to reference Matsuda S, Nakanishi A, Minami A, Wada Y, Kitagishi Y. Functions and characteristics of PINK1 and Parkin in cancer. Front Biosci. 2015;20:491–501.CrossRef Matsuda S, Nakanishi A, Minami A, Wada Y, Kitagishi Y. Functions and characteristics of PINK1 and Parkin in cancer. Front Biosci. 2015;20:491–501.CrossRef
146.
go back to reference O’Flanagan CH, Morais VA, Wurst W, De Strooper B, O’Neill C. The Parkinson’s gene PINK1 regulates cell cycle progression and promotes cancer-associated phenotypes. Oncogene. 2015;34:1363–74.PubMedCrossRef O’Flanagan CH, Morais VA, Wurst W, De Strooper B, O’Neill C. The Parkinson’s gene PINK1 regulates cell cycle progression and promotes cancer-associated phenotypes. Oncogene. 2015;34:1363–74.PubMedCrossRef
147.
go back to reference O’Flanagan CH, O’Neill C. PINK1 signalling in cancer biology. Biochim Biophys Acta. 1846;2014:590–8. O’Flanagan CH, O’Neill C. PINK1 signalling in cancer biology. Biochim Biophys Acta. 1846;2014:590–8.
148.
go back to reference Raimondi S, Sera F, Gandini S, Iodice S, Caini S, Maisonneuve P, et al. MC1R variants, melanoma and red hair color phenotype: a meta-analysis. Int J Cancer. 2008;122:2753–60.PubMedCrossRef Raimondi S, Sera F, Gandini S, Iodice S, Caini S, Maisonneuve P, et al. MC1R variants, melanoma and red hair color phenotype: a meta-analysis. Int J Cancer. 2008;122:2753–60.PubMedCrossRef
149.
go back to reference D’Orazio JA, Nobuhisa T, Cui R, Arya M, Spry M, Wakamatsu K, et al. Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature. 2006;443:340–4.PubMedCrossRef D’Orazio JA, Nobuhisa T, Cui R, Arya M, Spry M, Wakamatsu K, et al. Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature. 2006;443:340–4.PubMedCrossRef
150.
go back to reference Wong KY, Rajora N, Boccoli G, Catania A, Lipton JM. A potential mechanism of local anti-inflammatory action of alpha-melanocyte-stimulating hormone within the brain: modulation of tumor necrosis factor-alpha production by human astrocytic cells. Neuroimmunomodulation. 1997;4:37–41.PubMed Wong KY, Rajora N, Boccoli G, Catania A, Lipton JM. A potential mechanism of local anti-inflammatory action of alpha-melanocyte-stimulating hormone within the brain: modulation of tumor necrosis factor-alpha production by human astrocytic cells. Neuroimmunomodulation. 1997;4:37–41.PubMed
151.
go back to reference Teare KA, Pearson RG, Shakesheff KM, Haycock JW. Alpha-MSH inhibits inflammatory signalling in Schwann cells. Neuroreport. 2004;15:493–8.PubMedCrossRef Teare KA, Pearson RG, Shakesheff KM, Haycock JW. Alpha-MSH inhibits inflammatory signalling in Schwann cells. Neuroreport. 2004;15:493–8.PubMedCrossRef
152.
go back to reference Xia Y, Wikberg JE, Chhajlani V. Expression of melanocortin 1 receptor in periaqueductal gray matter. Neuroreport. 1995;6:2193–6.PubMedCrossRef Xia Y, Wikberg JE, Chhajlani V. Expression of melanocortin 1 receptor in periaqueductal gray matter. Neuroreport. 1995;6:2193–6.PubMedCrossRef
153.
go back to reference Tell-Marti G, Puig-Butille JA, Potrony M, Badenas C, Mila M, Malvehy J, et al. The MC1R melanoma risk variant p. R160W is associated with Parkinson disease. Ann Neurol. 2015;77:889–94.PubMedCrossRef Tell-Marti G, Puig-Butille JA, Potrony M, Badenas C, Mila M, Malvehy J, et al. The MC1R melanoma risk variant p. R160W is associated with Parkinson disease. Ann Neurol. 2015;77:889–94.PubMedCrossRef
154.
go back to reference Dong J, Gao J, Nalls M, Gao X, Huang X, Han J, et al. Susceptibility loci for pigmentation and melanoma in relation to Parkinson’s disease. Neurobiol Aging. 2014;35:1512. e5–10.PubMedCentralPubMedCrossRef Dong J, Gao J, Nalls M, Gao X, Huang X, Han J, et al. Susceptibility loci for pigmentation and melanoma in relation to Parkinson’s disease. Neurobiol Aging. 2014;35:1512. e5–10.PubMedCentralPubMedCrossRef
155.
go back to reference Roe CM, Fitzpatrick AL, Xiong C, Sieh W, Kuller L, Miller JP, et al. Cancer linked to Alzheimer disease but not vascular dementia. Neurology. 2010;74:106–12.PubMedCentralPubMedCrossRef Roe CM, Fitzpatrick AL, Xiong C, Sieh W, Kuller L, Miller JP, et al. Cancer linked to Alzheimer disease but not vascular dementia. Neurology. 2010;74:106–12.PubMedCentralPubMedCrossRef
156.
go back to reference Romero JP, Benito-León J, Louis ED, Bermejo-Pareja F. Alzheimer’s disease is associated with decreased risk of cancer-specific mortality: a prospective study (NEDICES). J Alzheimers Dis. 2014;40:465–73.PubMed Romero JP, Benito-León J, Louis ED, Bermejo-Pareja F. Alzheimer’s disease is associated with decreased risk of cancer-specific mortality: a prospective study (NEDICES). J Alzheimers Dis. 2014;40:465–73.PubMed
157.
go back to reference Musicco M, Adorni F, Di Santo S, Prinelli F, Pettenati C, Caltagirone C, et al. Inverse occurrence of cancer and Alzheimer disease: a population-based incidence study. Neurology. 2013;81:322–8.PubMedCrossRef Musicco M, Adorni F, Di Santo S, Prinelli F, Pettenati C, Caltagirone C, et al. Inverse occurrence of cancer and Alzheimer disease: a population-based incidence study. Neurology. 2013;81:322–8.PubMedCrossRef
158.
go back to reference Realmuto S, Cinturino A, Arnao V, Mazzola MA, Cupidi C, Aridon P, et al. Tumor diagnosis preceding Alzheimer’s disease onset: is there a link between cancer and Alzheimer's disease? J Alzheimers Dis. 2012;31:177–82.PubMed Realmuto S, Cinturino A, Arnao V, Mazzola MA, Cupidi C, Aridon P, et al. Tumor diagnosis preceding Alzheimer’s disease onset: is there a link between cancer and Alzheimer's disease? J Alzheimers Dis. 2012;31:177–82.PubMed
159.
go back to reference Driver JA, Beiser A, Au R, Kreger BE, Splansky GL, Kurth T, et al. Inverse association between cancer and Alzheimer’s disease: results from the Framingham Heart Study. BMJ. 2012;344:e1442.PubMedCentralPubMedCrossRef Driver JA, Beiser A, Au R, Kreger BE, Splansky GL, Kurth T, et al. Inverse association between cancer and Alzheimer’s disease: results from the Framingham Heart Study. BMJ. 2012;344:e1442.PubMedCentralPubMedCrossRef
160.
161.
go back to reference Nudelman KNH, Risacher SL, West JD, McDonald BC, Gao S, Saykin AJ, et al. Association of cancer history with Alzheimer’s disease onset and structural brain changes. Front Physiol. 2014;5:423.PubMedCentralPubMedCrossRef Nudelman KNH, Risacher SL, West JD, McDonald BC, Gao S, Saykin AJ, et al. Association of cancer history with Alzheimer’s disease onset and structural brain changes. Front Physiol. 2014;5:423.PubMedCentralPubMedCrossRef
162.
go back to reference Griffin RJ, Moloney A, Kelliher M, Johnston JA, Ravid R, Dockery P, et al. Activation of Akt/PKB, increased phosphorylation of Akt substrates and loss and altered distribution of Akt and PTEN are features of Alzheimer’s disease pathology. J Neurochem. 2005;93:105–17.PubMedCrossRef Griffin RJ, Moloney A, Kelliher M, Johnston JA, Ravid R, Dockery P, et al. Activation of Akt/PKB, increased phosphorylation of Akt substrates and loss and altered distribution of Akt and PTEN are features of Alzheimer’s disease pathology. J Neurochem. 2005;93:105–17.PubMedCrossRef
164.
go back to reference Stein TD, Johnson JA. Lack of neurodegeneration in transgenic mice overexpressing mutant amyloid precursor protein is associated with increased levels of transthyretin and the activation of cell survival pathways. J Neurosci. 2002;22:7380–8.PubMed Stein TD, Johnson JA. Lack of neurodegeneration in transgenic mice overexpressing mutant amyloid precursor protein is associated with increased levels of transthyretin and the activation of cell survival pathways. J Neurosci. 2002;22:7380–8.PubMed
165.
go back to reference Wei W, Wang X, Kusiak JW. Signaling Events in Amyloid β-Peptide-induced Neuronal Death and Insulin-like Growth Factor I Protection. J Biol Chem. 2002;277:17649–56.PubMedCrossRef Wei W, Wang X, Kusiak JW. Signaling Events in Amyloid β-Peptide-induced Neuronal Death and Insulin-like Growth Factor I Protection. J Biol Chem. 2002;277:17649–56.PubMedCrossRef
166.
go back to reference Lo RY, Tanner CM, Van Den Eeden SK, Albers KB, Leimpeter AD, Nelson LM. Comorbid cancer in Parkinson’s disease. Mov Disord. 2010;25:1809–17.PubMedCrossRef Lo RY, Tanner CM, Van Den Eeden SK, Albers KB, Leimpeter AD, Nelson LM. Comorbid cancer in Parkinson’s disease. Mov Disord. 2010;25:1809–17.PubMedCrossRef
167.
go back to reference Becker C, Brobert GP, Johansson S, Jick SS, Meier CR. Cancer risk in association with Parkinson disease: a population-based study. Parkinsonism Relat Disord. 2010;16:186–90.PubMedCrossRef Becker C, Brobert GP, Johansson S, Jick SS, Meier CR. Cancer risk in association with Parkinson disease: a population-based study. Parkinsonism Relat Disord. 2010;16:186–90.PubMedCrossRef
168.
go back to reference Elbaz A, Peterson BJ, Bower JH, Yang P, Maraganore DM, McDonnell SK, et al. Risk of cancer after the diagnosis of Parkinson’s disease: a historical cohort study. Mov Disord. 2005;20:719–25.PubMedCrossRef Elbaz A, Peterson BJ, Bower JH, Yang P, Maraganore DM, McDonnell SK, et al. Risk of cancer after the diagnosis of Parkinson’s disease: a historical cohort study. Mov Disord. 2005;20:719–25.PubMedCrossRef
169.
go back to reference Minami Y, Yamamoto R, Nishikouri M, Fukao A, Hisamichi S. Mortality and cancer incidence in patients with Parkinson’s disease. J Neurol. 2000;247:429–34.PubMedCrossRef Minami Y, Yamamoto R, Nishikouri M, Fukao A, Hisamichi S. Mortality and cancer incidence in patients with Parkinson’s disease. J Neurol. 2000;247:429–34.PubMedCrossRef
170.
go back to reference Constantinescu R, Elm J, Auinger P, Sharma S, Augustine EF, Khadim L, et al. Malignant melanoma in early-treated Parkinson’s disease: the NET-PD trial. Mov Disord. 2014;29:263–5.PubMedCentralPubMedCrossRef Constantinescu R, Elm J, Auinger P, Sharma S, Augustine EF, Khadim L, et al. Malignant melanoma in early-treated Parkinson’s disease: the NET-PD trial. Mov Disord. 2014;29:263–5.PubMedCentralPubMedCrossRef
171.
go back to reference Bertoni JM, Arlette JP, Fernandez HH, Fitzer-Attas C, Frei K, Hassan MN, et al. Increased melanoma risk in Parkinson disease: a prospective clinicopathological study. Arch Neurol. 2010;67:347–52.PubMedCrossRef Bertoni JM, Arlette JP, Fernandez HH, Fitzer-Attas C, Frei K, Hassan MN, et al. Increased melanoma risk in Parkinson disease: a prospective clinicopathological study. Arch Neurol. 2010;67:347–52.PubMedCrossRef
172.
go back to reference Schwid SR, Janice B, David O, Lynn S, Caroline T, Misser F, et al. Cancer incidence in a trial of an antiapoptotic agent for Parkinson’s disease. Mov Disord. 2010;25:1801–8.PubMedCrossRef Schwid SR, Janice B, David O, Lynn S, Caroline T, Misser F, et al. Cancer incidence in a trial of an antiapoptotic agent for Parkinson’s disease. Mov Disord. 2010;25:1801–8.PubMedCrossRef
173.
go back to reference Baade PD, Fritschi L, Freedman DM. Mortality due to amyotrophic lateral sclerosis and Parkinson’s disease among melanoma patients. Neuroepidemiology. 2007;28:16–20.PubMedCrossRef Baade PD, Fritschi L, Freedman DM. Mortality due to amyotrophic lateral sclerosis and Parkinson’s disease among melanoma patients. Neuroepidemiology. 2007;28:16–20.PubMedCrossRef
174.
go back to reference Bellani S, Sousa VL, Ronzitti G, Valtorta F, Meldolesi J, Chieregatti E. The regulation of synaptic function by α-synuclein. Commun Integr Biol. 2010;3:106–9.PubMedCentralPubMedCrossRef Bellani S, Sousa VL, Ronzitti G, Valtorta F, Meldolesi J, Chieregatti E. The regulation of synaptic function by α-synuclein. Commun Integr Biol. 2010;3:106–9.PubMedCentralPubMedCrossRef
175.
go back to reference Janezic S, Threlfell S, Dodson PD, Dowie MJ, Taylor TN, Potgieter D, et al. Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc Natl Acad Sci U S A. 2013;110:E4016–25.PubMedCentralPubMedCrossRef Janezic S, Threlfell S, Dodson PD, Dowie MJ, Taylor TN, Potgieter D, et al. Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc Natl Acad Sci U S A. 2013;110:E4016–25.PubMedCentralPubMedCrossRef
176.
go back to reference Hüls S, Högen T, Vassallo N, Danzer KM, Hengerer B, Giese A, et al. AMPA-receptor-mediated excitatory synaptic transmission is enhanced by iron-induced α-synuclein oligomers. J Neurochem. 2011;117:868–78.PubMedCrossRef Hüls S, Högen T, Vassallo N, Danzer KM, Hengerer B, Giese A, et al. AMPA-receptor-mediated excitatory synaptic transmission is enhanced by iron-induced α-synuclein oligomers. J Neurochem. 2011;117:868–78.PubMedCrossRef
178.
go back to reference Diógenes MJ, Dias RB, Rombo DM, Vicente Miranda H, Maiolino F, Guerreiro P, et al. Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J Neurosci. 2012;32:11750–62.PubMedCrossRef Diógenes MJ, Dias RB, Rombo DM, Vicente Miranda H, Maiolino F, Guerreiro P, et al. Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J Neurosci. 2012;32:11750–62.PubMedCrossRef
179.
go back to reference Brück D, Wenning GK, Stefanova N, Fellner L: Glia and alpha-synuclein in neurodegeneration: A complex interaction. Neurobiol Dis 2015. [Epub ahead of print] Brück D, Wenning GK, Stefanova N, Fellner L: Glia and alpha-synuclein in neurodegeneration: A complex interaction. Neurobiol Dis 2015. [Epub ahead of print]
180.
go back to reference Sampaio-Marques B, Felgueiras C, Silva A, Rodrigues M, Tenreiro S, Franssens V, et al. SNCA (α-synuclein)-induced toxicity in yeast cells is dependent on sirtuin 2 (Sir2)-mediated mitophagy. Autophagy. 2012;8:1494–509.PubMedCrossRef Sampaio-Marques B, Felgueiras C, Silva A, Rodrigues M, Tenreiro S, Franssens V, et al. SNCA (α-synuclein)-induced toxicity in yeast cells is dependent on sirtuin 2 (Sir2)-mediated mitophagy. Autophagy. 2012;8:1494–509.PubMedCrossRef
181.
go back to reference Zhou C, Chen H, Han L, Wang A, Chen L-A. Identification of featured biomarkers in different types of lung cancer with DNA microarray. Mol Biol Rep. 2014;41:6357–63.PubMedCrossRef Zhou C, Chen H, Han L, Wang A, Chen L-A. Identification of featured biomarkers in different types of lung cancer with DNA microarray. Mol Biol Rep. 2014;41:6357–63.PubMedCrossRef
182.
go back to reference Li W-H, Zhang H, Guo Q, Wu X-D, Xu Z-S, Dang C-X, et al. Detection of SNCA and FBN1 methylation in the stool as a biomarker for colorectal cancer. Dis Markers. 2015;2015:657570.PubMedCentralPubMed Li W-H, Zhang H, Guo Q, Wu X-D, Xu Z-S, Dang C-X, et al. Detection of SNCA and FBN1 methylation in the stool as a biomarker for colorectal cancer. Dis Markers. 2015;2015:657570.PubMedCentralPubMed
183.
go back to reference Fung K-M, Kar-Ming F, Rorke LB, Benoit G, Lee VM-Y, Trojanowski JQ. Expression of alpha-, beta-, and gamma-synuclein in glial tumors and medulloblastomas. Acta Neuropathol. 2003;106:167–75.PubMedCrossRef Fung K-M, Kar-Ming F, Rorke LB, Benoit G, Lee VM-Y, Trojanowski JQ. Expression of alpha-, beta-, and gamma-synuclein in glial tumors and medulloblastomas. Acta Neuropathol. 2003;106:167–75.PubMedCrossRef
184.
go back to reference Matsuo Y, Kamitani T. Parkinson’s Disease-Related Protein, alpha-Synuclein. Malignant Melanoma PLoS ONE. 2010;5:e10481.PubMedCrossRef Matsuo Y, Kamitani T. Parkinson’s Disease-Related Protein, alpha-Synuclein. Malignant Melanoma PLoS ONE. 2010;5:e10481.PubMedCrossRef
185.
go back to reference Corcoran C, Rani S, O’Driscoll L. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate. 2014;74:1320–34.PubMedCrossRef Corcoran C, Rani S, O’Driscoll L. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate. 2014;74:1320–34.PubMedCrossRef
186.
go back to reference Cirnaru MD, Marte A, Belluzzi E, Russo I, Gabrielli M, Longo F, et al. LRRK2 kinase activity regulates synaptic vesicle trafficking and neurotransmitter release through modulation of LRRK2 macro-molecular complex. Front Mol Neurosci. 2014;7:49.PubMedCentralPubMedCrossRef Cirnaru MD, Marte A, Belluzzi E, Russo I, Gabrielli M, Longo F, et al. LRRK2 kinase activity regulates synaptic vesicle trafficking and neurotransmitter release through modulation of LRRK2 macro-molecular complex. Front Mol Neurosci. 2014;7:49.PubMedCentralPubMedCrossRef
187.
go back to reference Orenstein SJ, Kuo S-H, Tasset I, Arias E, Koga H, Fernandez-Carasa I, et al. Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci. 2013;16:394–406.PubMedCentralPubMedCrossRef Orenstein SJ, Kuo S-H, Tasset I, Arias E, Koga H, Fernandez-Carasa I, et al. Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci. 2013;16:394–406.PubMedCentralPubMedCrossRef
188.
go back to reference Bahnassawy L, Nicklas S, Palm T, Menzl I, Birzele F, Gillardon F, et al. The parkinson’s disease-associated LRRK2 mutation R1441G inhibits neuronal differentiation of neural stem cells. Stem Cells Dev. 2013;22:2487–96.PubMedCrossRef Bahnassawy L, Nicklas S, Palm T, Menzl I, Birzele F, Gillardon F, et al. The parkinson’s disease-associated LRRK2 mutation R1441G inhibits neuronal differentiation of neural stem cells. Stem Cells Dev. 2013;22:2487–96.PubMedCrossRef
189.
go back to reference Ho CC-Y, Rideout HJ, Ribe E, Troy CM, Dauer WT. The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration. J Neurosci. 2009;29:1011–6.PubMedCentralPubMedCrossRef Ho CC-Y, Rideout HJ, Ribe E, Troy CM, Dauer WT. The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration. J Neurosci. 2009;29:1011–6.PubMedCentralPubMedCrossRef
190.
go back to reference Wang X, Yan MH, Fujioka H, Liu J, Wilson-Delfosse A, Chen SG, et al. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet. 2012;21:1931–44.PubMedCentralPubMedCrossRef Wang X, Yan MH, Fujioka H, Liu J, Wilson-Delfosse A, Chen SG, et al. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet. 2012;21:1931–44.PubMedCentralPubMedCrossRef
192.
go back to reference Parisiadou L, Xie C, Cho HJ, Lin X, Gu X-L, Long C-X, et al. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci. 2009;29:13971–80.PubMedCentralPubMedCrossRef Parisiadou L, Xie C, Cho HJ, Lin X, Gu X-L, Long C-X, et al. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci. 2009;29:13971–80.PubMedCentralPubMedCrossRef
193.
194.
go back to reference Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher RJ, et al. Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of α-synuclein, and apoptotic cell death in aged mice. Proc Natl Acad Sci. 2010;107:9879–84.PubMedCentralPubMedCrossRef Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher RJ, et al. Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of α-synuclein, and apoptotic cell death in aged mice. Proc Natl Acad Sci. 2010;107:9879–84.PubMedCentralPubMedCrossRef
195.
go back to reference Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, et al. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis. 2011;41:706–16.PubMedCentralPubMedCrossRef Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, et al. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis. 2011;41:706–16.PubMedCentralPubMedCrossRef
196.
go back to reference Helton TD, Otsuka T, Lee M-C, Mu Y, Ehlers MD. Pruning and loss of excitatory synapses by the parkin ubiquitin ligase. Proc Natl Acad Sci U S A. 2008;105:19492–7.PubMedCentralPubMedCrossRef Helton TD, Otsuka T, Lee M-C, Mu Y, Ehlers MD. Pruning and loss of excitatory synapses by the parkin ubiquitin ligase. Proc Natl Acad Sci U S A. 2008;105:19492–7.PubMedCentralPubMedCrossRef
197.
go back to reference Zhang Y, Gao J, Chung KK, Huang H, Dawson VL, Dawson TM. Parkin functions as an E2-dependent ubiquitin- protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc Natl Acad Sci U S A. 2000;97:13354–9.PubMedCentralPubMedCrossRef Zhang Y, Gao J, Chung KK, Huang H, Dawson VL, Dawson TM. Parkin functions as an E2-dependent ubiquitin- protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc Natl Acad Sci U S A. 2000;97:13354–9.PubMedCentralPubMedCrossRef
198.
go back to reference Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron. 2015;85:257–73.PubMedCrossRef Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron. 2015;85:257–73.PubMedCrossRef
199.
go back to reference Cesari R, Martin ES, Calin GA, Pentimalli F, Bichi R, McAdams H, et al. Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25-q27. Proc Natl Acad Sci U S A. 2003;100:5956–61.PubMedCentralPubMedCrossRef Cesari R, Martin ES, Calin GA, Pentimalli F, Bichi R, McAdams H, et al. Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25-q27. Proc Natl Acad Sci U S A. 2003;100:5956–61.PubMedCentralPubMedCrossRef
200.
go back to reference Devireddy S, Liu A, Lampe T, Hollenbeck PJ. The Organization of Mitochondrial Quality Control and Life Cycle in the Nervous System In Vivo in the Absence of PINK1. J Neurosci. 2015;35:9391–401.PubMedPubMedCentralCrossRef Devireddy S, Liu A, Lampe T, Hollenbeck PJ. The Organization of Mitochondrial Quality Control and Life Cycle in the Nervous System In Vivo in the Absence of PINK1. J Neurosci. 2015;35:9391–401.PubMedPubMedCentralCrossRef
201.
go back to reference Yang Y, Ouyang Y, Yang L, Beal MF, McQuibban A, Vogel H, et al. Pink1 regulates mitochondrial dynamics through interaction with the fission/fusion machinery. Proc Natl Acad Sci U S A. 2008;105:7070–5.PubMedCentralPubMedCrossRef Yang Y, Ouyang Y, Yang L, Beal MF, McQuibban A, Vogel H, et al. Pink1 regulates mitochondrial dynamics through interaction with the fission/fusion machinery. Proc Natl Acad Sci U S A. 2008;105:7070–5.PubMedCentralPubMedCrossRef
202.
go back to reference Kitada T, Pisani A, Porter DR, Yamaguchi H, Tscherter A, Martella G, et al. Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proc Natl Acad Sci U S A. 2007;104:11441–6.PubMedCentralPubMedCrossRef Kitada T, Pisani A, Porter DR, Yamaguchi H, Tscherter A, Martella G, et al. Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proc Natl Acad Sci U S A. 2007;104:11441–6.PubMedCentralPubMedCrossRef
203.
go back to reference Ye M, Zhou D, Zhou Y, Sun C. Parkinson’s disease-associated PINK1 G309D mutation increases abnormal phosphorylation of Tau. IUBMB Life. 2015;67:286–90.PubMedCrossRef Ye M, Zhou D, Zhou Y, Sun C. Parkinson’s disease-associated PINK1 G309D mutation increases abnormal phosphorylation of Tau. IUBMB Life. 2015;67:286–90.PubMedCrossRef
204.
go back to reference Park J, Lee SB, Lee S, Kim Y, Song S, Kim S, et al. Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin. Nature. 2006;441:1157–61.PubMedCrossRef Park J, Lee SB, Lee S, Kim Y, Song S, Kim S, et al. Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin. Nature. 2006;441:1157–61.PubMedCrossRef
206.
go back to reference Berthier A, Arnaud B, Samuel N, Judit J-S, Isabel R, Francisca R, et al. PINK1 displays tissue-specific subcellular location and regulates apoptosis and cell growth in breast cancer cells☆. Hum Pathol. 2011;42:75–87.PubMedCrossRef Berthier A, Arnaud B, Samuel N, Judit J-S, Isabel R, Francisca R, et al. PINK1 displays tissue-specific subcellular location and regulates apoptosis and cell growth in breast cancer cells☆. Hum Pathol. 2011;42:75–87.PubMedCrossRef
207.
go back to reference Devine MJ, Plun-Favreau H, Wood NW. Parkinson’s disease and cancer: two wars, one front. Nat Rev Cancer. 2011;11:812–23.PubMedCrossRef Devine MJ, Plun-Favreau H, Wood NW. Parkinson’s disease and cancer: two wars, one front. Nat Rev Cancer. 2011;11:812–23.PubMedCrossRef
208.
go back to reference Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, et al. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S A. 2005;102:5215–20.PubMedCentralPubMedCrossRef Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, et al. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S A. 2005;102:5215–20.PubMedCentralPubMedCrossRef
209.
go back to reference Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich A. DJ-1 is a redox-dependent molecular chaperone that inhibits alpha-synuclein aggregate formation. PLoS Biol. 2004;2:e362.PubMedCentralPubMedCrossRef Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich A. DJ-1 is a redox-dependent molecular chaperone that inhibits alpha-synuclein aggregate formation. PLoS Biol. 2004;2:e362.PubMedCentralPubMedCrossRef
210.
go back to reference Ariga H, Takahashi-Niki K, Kato I, Maita H, Niki T, Iguchi-Ariga SMM. Neuroprotective Function of DJ-1 in Parkinson’s Disease. Oxid Med Cell Longev. 2013;2013:683920.PubMedCentralPubMedCrossRef Ariga H, Takahashi-Niki K, Kato I, Maita H, Niki T, Iguchi-Ariga SMM. Neuroprotective Function of DJ-1 in Parkinson’s Disease. Oxid Med Cell Longev. 2013;2013:683920.PubMedCentralPubMedCrossRef
211.
go back to reference Vasseur S, Afzal S, Tardivel-Lacombe J, Park DS, Iovanna JL, Mak TW. DJ-1/PARK7 is an important mediator of hypoxia-induced cellular responses. Proc Natl Acad Sci U S A. 2009;106:1111–6.PubMedCentralPubMedCrossRef Vasseur S, Afzal S, Tardivel-Lacombe J, Park DS, Iovanna JL, Mak TW. DJ-1/PARK7 is an important mediator of hypoxia-induced cellular responses. Proc Natl Acad Sci U S A. 2009;106:1111–6.PubMedCentralPubMedCrossRef
213.
go back to reference Martinat C, Shendelman S, Jonason A, Leete T, Beal MF, Yang L, et al. Sensitivity to oxidative stress in DJ-1-deficient dopamine neurons: an ES- derived cell model of primary Parkinsonism. PLoS Biol. 2004;2, e327.PubMedCentralPubMedCrossRef Martinat C, Shendelman S, Jonason A, Leete T, Beal MF, Yang L, et al. Sensitivity to oxidative stress in DJ-1-deficient dopamine neurons: an ES- derived cell model of primary Parkinsonism. PLoS Biol. 2004;2, e327.PubMedCentralPubMedCrossRef
214.
go back to reference Hayashi T, Ishimori C, Takahashi-Niki K, Taira T, Kim Y-C, Maita H, et al. DJ-1 binds to mitochondrial complex I and maintains its activity. Biochem Biophys Res Commun. 2009;390:667–72.PubMedCrossRef Hayashi T, Ishimori C, Takahashi-Niki K, Taira T, Kim Y-C, Maita H, et al. DJ-1 binds to mitochondrial complex I and maintains its activity. Biochem Biophys Res Commun. 2009;390:667–72.PubMedCrossRef
215.
go back to reference Wang Y, Liu W, He X, Zhou F. Parkinson’s disease-associated DJ-1 mutations increase abnormal phosphorylation of tau protein through Akt/GSK-3β pathways. J Mol Neurosci. 2013;51:911–8.PubMedCrossRef Wang Y, Liu W, He X, Zhou F. Parkinson’s disease-associated DJ-1 mutations increase abnormal phosphorylation of tau protein through Akt/GSK-3β pathways. J Mol Neurosci. 2013;51:911–8.PubMedCrossRef
216.
go back to reference Kawate T, Iwaya K, Kikuchi R, Kaise H, Oda M, Sato E, et al. DJ-1 protein expression as a predictor of pathological complete remission after neoadjuvant chemotherapy in breast cancer patients. Breast Cancer Res Treat. 2013;139:51–9.PubMedCrossRef Kawate T, Iwaya K, Kikuchi R, Kaise H, Oda M, Sato E, et al. DJ-1 protein expression as a predictor of pathological complete remission after neoadjuvant chemotherapy in breast cancer patients. Breast Cancer Res Treat. 2013;139:51–9.PubMedCrossRef
217.
go back to reference Zeng H-Z, Qu Y-Q, Zhang W-J, Xiu B, Deng A-M, Liang A-B. Proteomic analysis identified DJ-1 as a cisplatin resistant marker in non-small cell lung cancer. Int J Mol Sci. 2011;12:3489–99.PubMedCentralPubMedCrossRef Zeng H-Z, Qu Y-Q, Zhang W-J, Xiu B, Deng A-M, Liang A-B. Proteomic analysis identified DJ-1 as a cisplatin resistant marker in non-small cell lung cancer. Int J Mol Sci. 2011;12:3489–99.PubMedCentralPubMedCrossRef
218.
go back to reference He XY, Liu BY, Yao WY, Zhao XJ, Zheng Z, Li JF, et al. Serum DJ-1 as a diagnostic marker and prognostic factor for pancreatic cancer. J Dig Dis. 2011;12:131–7.PubMedCrossRef He XY, Liu BY, Yao WY, Zhao XJ, Zheng Z, Li JF, et al. Serum DJ-1 as a diagnostic marker and prognostic factor for pancreatic cancer. J Dig Dis. 2011;12:131–7.PubMedCrossRef
219.
go back to reference Li Y, Cui J, Zhang C-H, Yang D-J, Chen J-H, Zan W-H, et al. High-expression of DJ-1 and loss of PTEN associated with tumor metastasis and correlated with poor prognosis of gastric carcinoma. Int J Med Sci. 2013;10:1689–97.PubMedCentralPubMedCrossRef Li Y, Cui J, Zhang C-H, Yang D-J, Chen J-H, Zan W-H, et al. High-expression of DJ-1 and loss of PTEN associated with tumor metastasis and correlated with poor prognosis of gastric carcinoma. Int J Med Sci. 2013;10:1689–97.PubMedCentralPubMedCrossRef
220.
go back to reference Hod Y. Differential control of apoptosis by DJ-1 in prostate benign and cancer cells. J Cell Biochem. 2004;92:1221–33.PubMedCrossRef Hod Y. Differential control of apoptosis by DJ-1 in prostate benign and cancer cells. J Cell Biochem. 2004;92:1221–33.PubMedCrossRef
221.
go back to reference Drechsel DN, Hyman AA, Cobb MH, Kirschner MW. Modulation of the dynamic instability of tubulin assembly by the microtubule-associated protein tau. Mol Biol Cell. 1992;3:1141–54.PubMedCentralPubMedCrossRef Drechsel DN, Hyman AA, Cobb MH, Kirschner MW. Modulation of the dynamic instability of tubulin assembly by the microtubule-associated protein tau. Mol Biol Cell. 1992;3:1141–54.PubMedCentralPubMedCrossRef
222.
go back to reference Mietelska-Porowska A, Wasik U, Goras M, Filipek A, Niewiadomska G. Tau protein modifications and interactions: their role in function and dysfunction. Int J Mol Sci. 2014;15:4671–713.PubMedCentralPubMedCrossRef Mietelska-Porowska A, Wasik U, Goras M, Filipek A, Niewiadomska G. Tau protein modifications and interactions: their role in function and dysfunction. Int J Mol Sci. 2014;15:4671–713.PubMedCentralPubMedCrossRef
223.
go back to reference Pooler AM, Noble W, Hanger DP. A role for tau at the synapse in Alzheimer’s disease pathogenesis. Neuropharmacology. 2014;76:Pt A:1–8.PubMedCrossRef Pooler AM, Noble W, Hanger DP. A role for tau at the synapse in Alzheimer’s disease pathogenesis. Neuropharmacology. 2014;76:Pt A:1–8.PubMedCrossRef
224.
go back to reference Souter S, Lee G. Tubulin-independent tau in Alzheimer’s disease and cancer: implications for disease pathogenesis and treatment. Curr Alzheimer Res. 2010;7:697–707.PubMedCrossRef Souter S, Lee G. Tubulin-independent tau in Alzheimer’s disease and cancer: implications for disease pathogenesis and treatment. Curr Alzheimer Res. 2010;7:697–707.PubMedCrossRef
225.
go back to reference Rouzier R, Rajan R, Wagner P, Hess KR, Gold DL, Stec J, et al. Microtubule-associated protein tau: a marker of paclitaxel sensitivity in breast cancer. Proc Natl Acad Sci U S A. 2005;102:8315–20.PubMedCentralPubMedCrossRef Rouzier R, Rajan R, Wagner P, Hess KR, Gold DL, Stec J, et al. Microtubule-associated protein tau: a marker of paclitaxel sensitivity in breast cancer. Proc Natl Acad Sci U S A. 2005;102:8315–20.PubMedCentralPubMedCrossRef
226.
go back to reference Smoter M, Bodnar L, Grala B, Stec R, Zieniuk K, Kozlowski W, et al. Tau protein as a potential predictive marker in epithelial ovarian cancer patients treated with paclitaxel/platinum first-line chemotherapy. J Exp Clin Cancer Res. 2013;32:25.PubMedCentralPubMedCrossRef Smoter M, Bodnar L, Grala B, Stec R, Zieniuk K, Kozlowski W, et al. Tau protein as a potential predictive marker in epithelial ovarian cancer patients treated with paclitaxel/platinum first-line chemotherapy. J Exp Clin Cancer Res. 2013;32:25.PubMedCentralPubMedCrossRef
227.
go back to reference Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J. Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci. 2006;26:7212–21.PubMedCrossRef Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J. Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci. 2006;26:7212–21.PubMedCrossRef
230.
go back to reference Baldus CD, Liyanarachchi S, Mrózek K, Auer H, Tanner SM, Guimond M, et al. Acute myeloid leukemia with complex karyotypes and abnormal chromosome 21: Amplification discloses overexpression of APP, ETS2, and ERG genes. Proc Natl Acad Sci U S A. 2004;101:3915–20.PubMedCentralPubMedCrossRef Baldus CD, Liyanarachchi S, Mrózek K, Auer H, Tanner SM, Guimond M, et al. Acute myeloid leukemia with complex karyotypes and abnormal chromosome 21: Amplification discloses overexpression of APP, ETS2, and ERG genes. Proc Natl Acad Sci U S A. 2004;101:3915–20.PubMedCentralPubMedCrossRef
231.
go back to reference Yamada Y, Fujimura T, Takahashi S, Takayama K, Urano T, Murata T, et al. Clinical significance of amyloid precursor protein in patients with testicular germ cell tumor. Adv Urol. 2013;2013:348438.PubMedCentralPubMedCrossRef Yamada Y, Fujimura T, Takahashi S, Takayama K, Urano T, Murata T, et al. Clinical significance of amyloid precursor protein in patients with testicular germ cell tumor. Adv Urol. 2013;2013:348438.PubMedCentralPubMedCrossRef
232.
233.
go back to reference Lee J-H, Paull TT. ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex. Science. 2005;308:551–4.PubMedCrossRef Lee J-H, Paull TT. ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex. Science. 2005;308:551–4.PubMedCrossRef
234.
go back to reference Thompson D, Duedal S, Kirner J, McGuffog L, Last J, Reiman A, et al. Cancer risks and mortality in heterozygous ATM mutation carriers. J Natl Cancer Inst. 2005;97:813–22.PubMedCrossRef Thompson D, Duedal S, Kirner J, McGuffog L, Last J, Reiman A, et al. Cancer risks and mortality in heterozygous ATM mutation carriers. J Natl Cancer Inst. 2005;97:813–22.PubMedCrossRef
235.
go back to reference Prokopcova J, Jana P, Zdenek K, Banwell CM, Petr P. The role of ATM in breast cancer development. Breast Cancer Res Treat. 2006;104:121–8.PubMedCrossRef Prokopcova J, Jana P, Zdenek K, Banwell CM, Petr P. The role of ATM in breast cancer development. Breast Cancer Res Treat. 2006;104:121–8.PubMedCrossRef
236.
go back to reference Bishop DT, Demenais F, Iles MM, Harland M, Taylor JC, Corda E, et al. Genome-wide association study identifies three loci associated with melanoma risk. Nat Genet. 2009;41:920–5.PubMedCentralPubMedCrossRef Bishop DT, Demenais F, Iles MM, Harland M, Taylor JC, Corda E, et al. Genome-wide association study identifies three loci associated with melanoma risk. Nat Genet. 2009;41:920–5.PubMedCentralPubMedCrossRef
Metadata
Title
The associations between Parkinson’s disease and cancer: the plot thickens
Authors
Danielle D. Feng
Waijiao Cai
Xiqun Chen
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Translational Neurodegeneration / Issue 1/2015
Electronic ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-015-0043-z

Other articles of this Issue 1/2015

Translational Neurodegeneration 1/2015 Go to the issue