Gan-Or Z, Giladi N, Rozovski U, Shifrin C, Rosner S, Gurevich T, et al. Genotype-phenotype correlations between GBA mutations and Parkinson disease risk and onset. Neurology. 2008;70(24):2277–83.
Article
CAS
PubMed
Google Scholar
Lesage S, Anheim M, Condroyer C, Pollak P, Durif F, Dupuits C, et al. Large-scale screening of the Gaucher’s disease-related glucocerebrosidase gene in Europeans with Parkinson’s disease. Hum Mol Genet. 2011;20(1):202–10.
Article
CAS
PubMed
Google Scholar
Li Y, Sekine T, Funayama M, Li L, Yoshino H, Nishioka K, et al. Clinicogenetic study of GBA mutations in patients with familial Parkinson’s disease. Neurobiol Aging. 2014;35(4):935.e3–8.
Article
CAS
Google Scholar
Sidransky E, Samaddar T, Tayebi N. Mutations in GBA are associated with familial Parkinson disease susceptibility and age at onset. Neurology. 2009;73(17):1424–5 author reply 1425-1426.
Article
PubMed
Google Scholar
Neumann J, Bras J, Deas E, O’Sullivan SS, Parkkinen L, Lachmann RH, et al. Glucocerebrosidase mutations in clinical and pathologically proven Parkinson’s disease. Brain J Neurol. 2009;132(Pt 7):1783–94.
Article
Google Scholar
Riboldi GM, Di Fonzo AB. GBA, Gaucher disease, and Parkinson’s disease: from genetic to clinic to new therapeutic approaches. Cells. 2019;8(4):8(4):364.
Velez-Pardo C, Lorenzo-Betancor O, Jimenez-Del-Rio M, Moreno S, Lopera F, Cornejo-Olivas M, et al. The distribution and risk effect of GBA variants in a large cohort of PD patients from Colombia and Peru. Parkinsonism Relat Disord. 2019;63:204–8.
Article
PubMed
PubMed Central
Google Scholar
Zhang Y, Shu L, Zhou X, Pan H, Xu Q, Guo J, et al. A meta-analysis of GBA-related clinical symptoms in Parkinson’s disease. Park Dis. 2018;2018:3136415.
Google Scholar
Bandres-Ciga S, Saez-Atienzar S, Bonet-Ponce L, Billingsley K, Vitale D, Blauwendraat C, et al. The endocytic membrane trafficking pathway plays a major role in the risk of Parkinson’s disease. Mov Disord Off J Mov Disord Soc. 2019;34(4):460–8.
Article
CAS
Google Scholar
Henderson MX, Sedor S, McGeary I, Cornblath EJ, Peng C, Riddle DM, et al. Glucocerebrosidase activity modulates neuronal susceptibility to pathological α-Synuclein insult. Neuron. 2020;105(5):822–836.e7.
Article
CAS
PubMed
Google Scholar
Kilpatrick BS, Magalhaes J, Beavan MS, McNeill A, Gegg ME, Cleeter MWJ, et al. Endoplasmic reticulum and lysosomal Ca2+ stores are remodelled in GBA1-linked Parkinson disease patient fibroblasts. Cell Calcium. 2016 Jan;59(1):12–20.
Article
CAS
PubMed
PubMed Central
Google Scholar
Maor G, Cabasso O, Krivoruk O, Rodriguez J, Steller H, Segal D, et al. The contribution of mutant GBA to the development of Parkinson disease in Drosophila. Hum Mol Genet. 2016;25(13):2712–27.
CAS
PubMed
PubMed Central
Google Scholar
Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, Caldwell GA, et al. Gaucher disease glucocerebrosidase and α-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell. 2011;146(1):37–52.
Article
CAS
PubMed
PubMed Central
Google Scholar
Papadopoulos VE, Nikolopoulou G, Antoniadou I, Karachaliou A, Arianoglou G, Emmanouilidou E, et al. Modulation of β-glucocerebrosidase increases α-synuclein secretion and exosome release in mouse models of Parkinson’s disease. Hum Mol Genet. 2018;27(10):1696–710.
CAS
PubMed
Google Scholar
Ron I, Horowitz M. ER retention and degradation as the molecular basis underlying Gaucher disease heterogeneity. Hum Mol Genet. 2005;14(16):2387–98.
Article
CAS
PubMed
Google Scholar
Sanchez-Martinez A, Beavan M, Gegg ME, Chau KY, Whitworth AJ, Schapira AHV. Parkinson disease-linked GBA mutation effects reversed by molecular chaperones in human cell and fly models. Sci Rep. 2016 Aug;19(6):31380.
Article
CAS
Google Scholar
Suzuki T, Shimoda M, Ito K, Hanai S, Aizawa H, Kato T, et al. Expression of human Gaucher disease gene GBA generates neurodevelopmental defects and ER stress in Drosophila eye. PLoS One. 2013;8(8):e69147.
Article
CAS
PubMed
PubMed Central
Google Scholar
Blauwendraat C, Reed X, Krohn L, Heilbron K, Bandres-Ciga S, Tan M, et al. Genetic modifiers of risk and age at onset in GBA associated Parkinson’s disease and Lewy body dementia. Brain J Neurol. 2020;143(1):234–48.
Article
Google Scholar
Alcalay RN, Levy OA, Waters CC, Fahn S, Ford B, Kuo SH, et al. Glucocerebrosidase activity in Parkinson’s disease with and without GBA mutations. Brain J Neurol. 2015;138(Pt 9):2648–58.
Article
Google Scholar
Gan-Or Z, Bar-Shira A, Gurevich T, Giladi N, Orr-Urtreger A. Homozygosity for the MTX1 c.184T>a (p.S63T) alteration modifies the age of onset in GBA-associated Parkinson’s disease. Neurogenetics. 2011;12(4):325–32.
Article
CAS
PubMed
Google Scholar
Omer N, Giladi N, Gurevich T, Bar-Shira A, Gana-Weisz M, Goldstein O, et al. A possible modifying effect of the G2019S mutation in the LRRK2 gene on GBA Parkinson’s disease. Mov Disord Off J Mov Disord Soc. 2020;35(7):1249–53.
Article
CAS
Google Scholar
Sanyal A, DeAndrade MP, Novis HS, Lin S, Chang J, Lengacher N, et al. Lysosome and inflammatory defects in GBA1-mutant astrocytes are normalized by LRRK2 inhibition. Mov Disord Off J Mov Disord Soc. 2020;35(5):760–73.
Article
CAS
Google Scholar
Schierding W, Farrow S, Fadason T, Graham OEE, Pitcher TL, Qubisi S, et al. Common variants Coregulate expression of GBA and modifier genes to delay Parkinson’s disease onset. Mov Disord Off J Mov Disord Soc. 2020;35(8):1346–56.
Article
CAS
Google Scholar
Straniero L, Rimoldi V, Samarani M, Goldwurm S, Di Fonzo A, Krüger R, et al. The GBAP1 pseudogene acts as a ceRNA for the glucocerebrosidase gene GBA by sponging miR-22-3p. Sci Rep. 2017;7(1):12702.
Article
PubMed
PubMed Central
CAS
Google Scholar
Chahine LM, Qiang J, Ashbridge E, Minger J, Yearout D, Horn S, et al. Clinical and biochemical differences in patients having Parkinson disease with vs without GBA mutations. JAMA Neurol. 2013;70(7):852–8.
Article
PubMed
PubMed Central
Google Scholar
Dzamko N, Gysbers A, Perera G, Bahar A, Shankar A, Gao J, et al. Toll-like receptor 2 is increased in neurons in Parkinson’s disease brain and may contribute to alpha-synuclein pathology. Acta Neuropathol (Berl). 2017;133(2):303–19.
Article
CAS
Google Scholar
Keatinge M, Bui H, Menke A, Chen YC, Sokol AM, Bai Q, et al. Glucocerebrosidase 1 deficient Danio rerio mirror key pathological aspects of human Gaucher disease and provide evidence of early microglial activation preceding alpha-synuclein-independent neuronal cell death. Hum Mol Genet. 2015;24(23):6640–52.
Article
CAS
PubMed
PubMed Central
Google Scholar
Mullin S, Stokholm MG, Hughes D, Mehta A, Parbo P, Hinz R, et al. Brain microglial activation increased in Glucocerebrosidase (GBA) mutation carriers without Parkinson’s disease. Mov Disord Off J Mov Disord Soc. 2021;36(3):774–9.
Article
CAS
Google Scholar
Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis. 2006;21(2):404–12.
Article
CAS
PubMed
Google Scholar
Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, et al. Inflammatory dysregulation of blood monocytes in Parkinson’s disease patients. Acta Neuropathol (Berl). 2014;128(5):651–63.
Article
CAS
Google Scholar
Harms AS, Thome AD, Yan Z, Schonhoff AM, Williams GP, Li X, et al. Peripheral monocyte entry is required for alpha-Synuclein induced inflammation and neurodegeneration in a model of Parkinson disease. Exp Neurol. 2018;300:179–87.
Article
CAS
PubMed
Google Scholar
McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38(8):1285–91.
Article
CAS
PubMed
Google Scholar
Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T. Interleukin (IL)-1 beta, IL-2, IL-4, IL-6 and transforming growth factor-alpha levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson’s disease. Neurosci Lett. 1996;211(1):13–6.
Article
CAS
PubMed
Google Scholar
Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di Nicola M, et al. Peripheral cytokines profile in Parkinson’s disease. Brain Behav Immun. 2009;23(1):55–63.
Article
CAS
PubMed
Google Scholar
Terada T, Yokokura M, Yoshikawa E, Futatsubashi M, Kono S, Konishi T, et al. Extrastriatal spreading of microglial activation in Parkinson’s disease: a positron emission tomography study. Ann Nucl Med. 2016;30(8):579–87.
Article
CAS
PubMed
Google Scholar
Funk N, Wieghofer P, Grimm S, Schaefer R, Bühring HJ, Gasser T, et al. Characterization of peripheral hematopoietic stem cells and monocytes in Parkinson’s disease. Mov Disord Off J Mov Disord Soc. 2013;28(3):392–5.
Article
Google Scholar
Holmans P, Moskvina V, Jones L, Sharma M, International Parkinson’s Disease Genomics Consortium, Vedernikov A, et al. A pathway-based analysis provides additional support for an immune-related genetic susceptibility to Parkinson’s disease. Hum Mol Genet. 2013;22(5):1039–49.
Article
CAS
PubMed
Google Scholar
Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet. 2014;46(9):989–93.
Article
CAS
PubMed
PubMed Central
Google Scholar
Raj T, Rothamel K, Mostafavi S, Ye C, Lee MN, Replogle JM, et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science. 2014;344(6183):519–23.
Article
CAS
PubMed
PubMed Central
Google Scholar
Wissemann WT, Hill-Burns EM, Zabetian CP, Factor SA, Patsopoulos N, Hoglund B, et al. Association of Parkinson disease with structural and regulatory variants in the HLA region. Am J Hum Genet. 2013;93(5):984–93.
Article
CAS
PubMed
PubMed Central
Google Scholar
Navarro E, Udine E, de Paiva Lopes K, Parks M, Riboldi G, Schilder BM, et al. Dysregulation of mitochondrial and proteolysosomal genes in Parkinson’s disease myeloid cells. Nat Aging. 2021;1(9):850–63. https://doi.org/10.1038/s43587-021-00110-x.
Article
PubMed
PubMed Central
Google Scholar
Atashrazm F, Hammond D, Perera G, Dobson-Stone C, Mueller N, Pickford R, et al. Reduced glucocerebrosidase activity in monocytes from patients with Parkinson’s disease. Sci Rep. 2018;8(1):15446.
Article
PubMed
PubMed Central
CAS
Google Scholar
Hughes LP, Pereira MMM, Hammond DA, Kwok JB, Halliday GM, Lewis SJG, et al. Glucocerebrosidase activity is reduced in cryopreserved Parkinson’s disease patient monocytes and inversely correlates with motor severity. J Parkinsons Dis. 2021;11(3):1157–65.
Article
CAS
PubMed
PubMed Central
Google Scholar
Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55(3):181–4.
Article
CAS
PubMed
PubMed Central
Google Scholar
Craig DW, Hutchins E, Violich I, Alsop E, Gibbs JR, Levy S, et al. RNA sequencing of whole blood reveals early alterations in immune cells and gene expression in Parkinson’s disease. Nat Aging. 2021;1(1):734–47.
Article
Google Scholar
Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81(3):559–75.
Article
CAS
PubMed
PubMed Central
Google Scholar
Carmi S, Hui KY, Kochav E, Liu X, Xue J, Grady F, et al. Sequencing an Ashkenazi reference panel supports population-targeted personal genomics and illuminates Jewish and European origins. Nat Commun. 2014;9(5):4835.
Article
CAS
Google Scholar
Shah, H. PgmNr 1856: RAPiD—An Agile and Dependable RNA-Seq Framework (American Society of Human Genetics, 2015).
Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The sequence alignment/map format and SAMtools. Bioinforma Oxf Engl. 2009;25(16):2078–9.
Article
CAS
Google Scholar
Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinforma Oxf Engl. 2014;30(15):2114–20.
Article
CAS
Google Scholar
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinforma Oxf Engl. 2013;29(1):15–21.
Article
CAS
Google Scholar
Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323.
Article
CAS
PubMed
PubMed Central
Google Scholar
Law CW, Alhamdoosh M, Su S, Dong X, Tian L, Smyth GK, et al. RNA-seq analysis is easy as 1–2-3 with limma, Glimma and edgeR. F1000Research. 2018;5:ISCB Comm J-1408.
Article
PubMed Central
Google Scholar
Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinforma Oxf Engl. 2012;28(6):882–3.
Article
CAS
Google Scholar
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50.
Article
CAS
PubMed
PubMed Central
Google Scholar
Brechtmann F, Mertes C, Matusevičiūtė A, Yépez VA, Avsec Ž, Herzog M, et al. OUTRIDER: a statistical method for detecting aberrantly expressed genes in RNA sequencing data. Am J Hum Genet. 2018;103(6):907–17.
Article
CAS
PubMed
PubMed Central
Google Scholar
Cuperfain AB, Zhang ZL, Kennedy JL, Gonçalves VF. The complex interaction of mitochondrial genetics and mitochondrial pathways in psychiatric disease. Mol Neuropsychiatry. 2018;4(1):52–69.
CAS
PubMed
PubMed Central
Google Scholar
Cummings BB, Marshall JL, Tukiainen T, Lek M, Donkervoort S, Foley AR, et al. Improving genetic diagnosis in Mendelian disease with transcriptome sequencing. Sci Transl Med. 2017;9(386):eaal5209.
Article
PubMed
PubMed Central
Google Scholar
Kremer LS, Bader DM, Mertes C, Kopajtich R, Pichler G, Iuso A, et al. Genetic diagnosis of Mendelian disorders via RNA sequencing. Nat Commun. 2017;8:15824.
Article
CAS
PubMed
PubMed Central
Google Scholar
Luo J, Sun L, Lin X, Liu G, Yu J, Parisiadou L, et al. A calcineurin- and NFAT-dependent pathway is involved in α-synuclein-induced degeneration of midbrain dopaminergic neurons. Hum Mol Genet. 2014;23(24):6567–74.
Article
CAS
PubMed
PubMed Central
Google Scholar
Gonzalo S, Kreienkamp R, Askjaer P. Hutchinson-Gilford progeria syndrome: a premature aging disease caused by LMNA gene mutations. Ageing Res Rev. 2017;33:18–29.
Article
CAS
PubMed
Google Scholar
Pan Z, Li GF, Sun ML, Xie L, Liu D, Zhang Q, et al. MicroRNA-1224 splicing CircularRNA-Filip1l in an Ago2-dependent manner regulates chronic inflammatory pain via targeting Ubr5. J Neurosci. 2019;39(11):2125–43.
Article
CAS
PubMed
PubMed Central
Google Scholar
Bhambhani V, Introne WJ, Lungu C, Cullinane A, Toro C. Chediak-Higashi syndrome presenting as young-onset levodopa-responsive parkinsonism. Mov Disord Off J Mov Disord Soc. 2013;28(2):127–9.
Article
Google Scholar
Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, et al. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Park Dis. 2017;3:11.
Article
CAS
Google Scholar
Li X, Sundquist J, Sundquist K. Subsequent risks of Parkinson disease in patients with autoimmune and related disorders: a nationwide epidemiological study from Sweden. Neurodegener Dis. 2012;10(1–4):277–84.
Article
CAS
PubMed
Google Scholar
Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, et al. T cells from patients with Parkinson’s disease recognize α-synuclein peptides. Nature. 2017;546(7660):656–61.
Article
CAS
PubMed
PubMed Central
Google Scholar
Chang D, Nalls MA, Hallgrímsdóttir IB, Hunkapiller J, van der Brug M, Cai F, et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat Genet. 2017;49(10):1511–6.
Article
CAS
PubMed
PubMed Central
Google Scholar
Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 2019;18(12):1091–102.
Article
CAS
PubMed
PubMed Central
Google Scholar
Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, et al. Parkinson disease. Nat Rev Dis Primer. 2017;3:17013.
Article
Google Scholar
Blandini F, Cilia R, Cerri S, Pezzoli G, Schapira AHV, Mullin S, et al. Glucocerebrosidase mutations and synucleinopathies: toward a model of precision medicine. Mov Disord Off J Mov Disord Soc. 2019;34(1):9–21.
Article
Google Scholar
Fishbein I, Kuo YM, Giasson BI, Nussbaum RL. Augmentation of phenotype in a transgenic Parkinson mouse heterozygous for a Gaucher mutation. Brain J Neurol. 2014;137(Pt 12):3235–47.
Article
Google Scholar
Schöndorf DC, Aureli M, McAllister FE, Hindley CJ, Mayer F, Schmid B, et al. iPSC-derived neurons from GBA1-associated Parkinson’s disease patients show autophagic defects and impaired calcium homeostasis. Nat Commun. 2014;5:4028.
Article
PubMed
CAS
Google Scholar
Kim S, Wong YC, Gao F, Krainc D. Dysregulation of mitochondria-lysosome contacts by GBA1 dysfunction in dopaminergic neuronal models of Parkinson’s disease. Nat Commun. 2021;12(1):1807.
Article
CAS
PubMed
PubMed Central
Google Scholar