Log in

Enhancing the Activity of Glucocerebrosidase as a Treatment for Parkinson Disease

  • Leading Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Mutations in the glucocerebrosidase (GBA1) gene are the most common genetic risk factor for Parkinson disease (PD). Homozygous or compound heterozygous GBA1 mutations cause the lysosomal storage disorder Gaucher disease (GD), characterized by deficient activity of the glucocerebrosidase enzyme (GCase). Both individuals with GD type I and heterozygous carriers of pathogenic variants of GBA1 have an increased risk of develo** PD, by approximately ten- to 20-fold compared to non-carriers. GCase activity is also reduced in PD patients without GBA1 mutations, suggesting that the GCase lysosomal pathway might be involved in PD pathogenesis. Available evidence indicates that GCase can affect α-synuclein pathology in different ways. Misfolded GCase proteins are retained in the endoplasmic reticulum, altering the lysosomal trafficking of the enzyme and disrupting protein trafficking. Also, deficient GCase leads to accumulation of substrates that in turn may bind α-synuclein and promote pathological formation of aggregates. Furthermore, α-synuclein itself can lower the enzymatic activity of GCase, indicating that a bidirectional interaction exists between GCase and α-synuclein. Targeted therapies aimed at enhancing GCase activity, augmenting the trafficking of misfolded GCase proteins by small molecule chaperones, or reducing substrate accumulation, have been tested in preclinical and clinical trials. This article reviews the molecular mechanisms linking GCase to α-synuclein and discusses the therapeutic drugs that by targeting the GCase pathway can influence PD progression.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price includes VAT (Germany)

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Grabowski GA. Phenotype, diagnosis, and treatment of Gaucher's disease. Lancet. 2008;372(9645):1263–71.

    Article  CAS  PubMed  Google Scholar 

  2. Stirnemann J, Vigan M, Hamroun D, Heraoui D, Rossi-Semerano L, Berger MG, et al. The French Gaucher's disease registry: clinical characteristics, complications and treatment of 562 patients. Orphanet J Rare Dis. 2012;7:77.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Hruska KS, LaMarca ME, Scott CR, Sidransky E. Gaucher disease: mutation and polymorphism spectrum in the glucocerebrosidase gene (GBA). Hum Mutat. 2008;29(5):567–83.

    Article  CAS  PubMed  Google Scholar 

  4. Sidransky E, Lopez G. The link between the GBA gene and parkinsonism. Lancet Neurol. 2012;11(11):986–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Alfonso P, Rodriguez-Rey JC, Ganan A, Perez-Calvo JI, Giralt M, Giraldo P, et al. Expression and functional characterization of mutated glucocerebrosidase alleles causing Gaucher disease in Spanish patients. Blood Cells Mol Dis. 2004;32(1):218–25.

    Article  CAS  PubMed  Google Scholar 

  6. Malini E, Grossi S, Deganuto M, Rosano C, Parini R, Dominisini S, et al. Functional analysis of 11 novel GBA alleles. Eur J Human Genet. 2014;22(4):511–6.

    Article  CAS  Google Scholar 

  7. Neudorfer O, Giladi N, Elstein D, Abrahamov A, Turezkite T, Aghai E, et al. Occurrence of Parkinson's syndrome in type I Gaucher disease. QJM. 1996;89(9):691–4.

    Article  CAS  PubMed  Google Scholar 

  8. Goker-Alpan O, Schiffmann R, LaMarca ME, Nussbaum RL, McInerney-Leo A, Sidransky E. Parkinsonism among Gaucher disease carriers. J Med Genet. 2004;41(12):937–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–40.

    Article  CAS  PubMed  Google Scholar 

  10. Tayebi N, Walker J, Stubblefield B, Orvisky E, LaMarca ME, Wong K, et al. Gaucher disease with parkinsonian manifestations: does glucocerebrosidase deficiency contribute to a vulnerability to parkinsonism? Mol Genet Metab. 2003;79(2):104–9.

    CAS  PubMed  Google Scholar 

  11. Wong K, Sidransky E, Verma A, Mixon T, Sandberg GD, Wakefield LK, et al. Neuropathology provides clues to the pathophysiology of Gaucher disease. Mol Genet Metab. 2004;82(3):192–207.

    Article  CAS  PubMed  Google Scholar 

  12. Zhang Y, Shu L, Sun Q, Zhou X, Pan H, Guo J, et al. Integrated genetic analysis of racial differences of common GBA variants in Parkinson's disease: a meta-analysis. Front Mol Neurosci. 2018;11:43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Riboldi GM, Di Fonzo AB. GBA, Gaucher disease, and Parkinson's disease: from genetic to clinic to new therapeutic approaches. Cells. 2019;8(4):364.

    Article  CAS  PubMed Central  Google Scholar 

  14. Anheim M, Elbaz A, Lesage S, Durr A, Condroyer C, Viallet F, et al. Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers. Neurology. 2012;78(6):417–20.

    Article  CAS  PubMed  Google Scholar 

  15. Rana HQ, Balwani M, Bier L, Alcalay RN. Age-specific Parkinson disease risk in GBA mutation carriers: information for genetic counseling. Genet Med. 2013;15(2):146–9.

    Article  CAS  PubMed  Google Scholar 

  16. Gan-Or Z, Amshalom I, Kilarski LL, Bar-Shira A, Gana-Weisz M, Mirelman A, et al. Differential effects of severe vs mild GBA mutations on Parkinson disease. Neurology. 2015;84(9):880–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nalls MA, Duran R, Lopez G, Kurzawa-Akanbi M, McKeith IG, Chinnery PF, et al. A multicenter study of glucocerebrosidase mutations in dementia with Lewy bodies. JAMA Neurol. 2013;70(6):727–35.

    Article  PubMed  Google Scholar 

  18. Guerreiro R, Ross OA, Kun-Rodrigues C, Hernandez DG, Orme T, Eicher JD, et al. Investigating the genetic architecture of dementia with Lewy bodies: a two-stage genome-wide association study. Lancet Neurol. 2018;17(1):64–74.

    Article  PubMed  Google Scholar 

  19. Alcalay RN, Dinur T, Quinn T, Sakanaka K, Levy O, Waters C, et al. Comparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes. JAMA Neurol. 2014;71(6):752–7.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Gegg ME, Burke D, Heales SJ, Cooper JM, Hardy J, Wood NW, et al. Glucocerebrosidase deficiency in substantia nigra of parkinson disease brains. Ann Neurol. 2012;72(3):455–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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. 2015;138(Pt 9):2648–58.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Parnetti L, Paciotti S, Eusebi P, Dardis A, Zampieri S, Chiasserini D, et al. Cerebrospinal fluid beta-glucocerebrosidase activity is reduced in Parkinson's disease patients. Mov Disord. 2017;32(10):1423–31.

    Article  CAS  PubMed  Google Scholar 

  23. Hipp MS, Kasturi P, Hartl FU. The proteostasis network and its decline in ageing. Nat Rev Mol Cell Biol. 2019;20(7):421–35.

    Article  CAS  PubMed  Google Scholar 

  24. Fernandes HJ, Hartfield EM, Christian HC, Emmanoulidou E, Zheng Y, Booth H, et al. ER stress and autophagic perturbations lead to elevated extracellular alpha-synuclein in GBA-N370S Parkinson's iPSC-derived dopamine neurons. Stem Cell Rep. 2016;6(3):342–56.

    Article  CAS  Google Scholar 

  25. Garcia-Sanz P, Orgaz L, Bueno-Gil G, Espadas I, Rodriguez-Traver E, Kulisevsky J, et al. N370S-GBA1 mutation causes lysosomal cholesterol accumulation in Parkinson's disease. Mov Disord. 2017;32(10):1409–22.

    Article  CAS  PubMed  Google Scholar 

  26. McNeill A, Magalhaes J, Shen C, Chau KY, Hughes D, Mehta A, et al. Ambroxol improves lysosomal biochemistry in glucocerebrosidase mutation-linked Parkinson disease cells. Brain. 2014;137(Pt 5):1481–95.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Bendikov-Bar I, Ron I, Filocamo M, Horowitz M. Characterization of the ERAD process of the L444P mutant glucocerebrosidase variant. Blood Cells Mol Dis. 2011;46(1):4–10.

    Article  CAS  PubMed  Google Scholar 

  28. Maor G, Rencus-Lazar S, Filocamo M, Steller H, Segal D, Horowitz M. Unfolded protein response in Gaucher disease: from human to Drosophila. Orphanet J Rare Dis. 2013;8:140.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, et al. Acid beta-glucosidase mutants linked to Gaucher disease, Parkinson disease, and Lewy body dementia alter alpha-synuclein processing. Ann Neurol. 2011;69(6):940–53.

    Article  CAS  PubMed  Google Scholar 

  30. Sardi SP, Clarke J, Viel C, Chan M, Tamsett TJ, Treleaven CM, et al. Augmenting CNS glucocerebrosidase activity as a therapeutic strategy for parkinsonism and other Gaucher-related synucleinopathies. Proc Natl Acad Sci USA. 2013;110(9):3537–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xu YH, Xu K, Sun Y, Liou B, Quinn B, Li RH, et al. Multiple pathogenic proteins implicated in neuronopathic Gaucher disease mice. Hum Mol Genet. 2014;23(15):3943–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Papadopoulos VE, Nikolopoulou G, Antoniadou I, Karachaliou A, Arianoglou G, Emmanouilidou E, et al. Modulation of beta-glucocerebrosidase increases alpha-synuclein secretion and exosome release in mouse models of Parkinson's disease. Hum Mol Genet. 2018;27(10):1696–710.

    CAS  PubMed  Google Scholar 

  33. Maor G, Rapaport D, Horowitz M. The effect of mutant GBA1 on accumulation and aggregation of alpha-synuclein. Hum Mol Genet. 2019;28(11):1768–81.

    Article  CAS  PubMed  Google Scholar 

  34. Fishbein I, Kuo YM, Giasson BI, Nussbaum RL. Augmentation of phenotype in a transgenic Parkinson mouse heterozygous for a Gaucher mutation. Brain. 2014;137(Pt 12):3235–47.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Goker-Alpan O, Stubblefield BK, Giasson BI, Sidransky E. Glucocerebrosidase is present in alpha-synuclein inclusions in Lewy body disorders. Acta Neuropathol. 2010;120(5):641–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yap TL, Gruschus JM, Velayati A, Westbroek W, Goldin E, Moaven N, et al. Alpha-synuclein interacts with glucocerebrosidase providing a molecular link between Parkinson and Gaucher diseases. J Biol Chem. 2011;286(32):28080–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cleeter MW, Chau KY, Gluck C, Mehta A, Hughes DA, Duchen M, et al. Glucocerebrosidase inhibition causes mitochondrial dysfunction and free radical damage. Neurochem Int. 2013;62(1):1–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Manning-Bog AB, Schule B, Langston JW. Alpha-synuclein-glucocerebrosidase interactions in pharmacological Gaucher models: a biological link between Gaucher disease and Parkinsonism. Neurotoxicology. 2009;30(6):1127–32.

    Article  CAS  PubMed  Google Scholar 

  39. Rocha EM, Smith GA, Park E, Cao H, Graham AR, Brown E, et al. Sustained systemic glucocerebrosidase inhibition induces brain alpha-synuclein aggregation, microglia and complement C1q activation in mice. Antioxid Redox Signal. 2015;23(6):550–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mus L, Siani F, Giuliano C, Ghezzi C, Cerri S, Blandini F. Development and biochemical characterization of a mouse model of Parkinson's disease bearing defective glucocerebrosidase activity. Neurobiol Dis. 2019;124:289–96.

    Article  CAS  PubMed  Google Scholar 

  41. Henderson MX, Sedor S, McGeary I, Cornblath EJ, Peng C, Riddle DM, et al. Glucocerebrosidase activity modulates neuronal susceptibility to pathological alpha-synuclein insult. Neuron. 2020;105(5):822–36 e7.

    Article  CAS  PubMed  Google Scholar 

  42. Bae EJ, Yang NY, Song M, Lee CS, Lee JS, Jung BC, et al. Glucocerebrosidase depletion enhances cell-to-cell transmission of alpha-synuclein. Nat Commun. 2014;5:4755.

    Article  CAS  PubMed  Google Scholar 

  43. Zunke F, Moise AC, Belur NR, Gelyana E, Stojkovska I, Dzaferbegovic H, et al. Reversible conformational conversion of alpha-synuclein into toxic assemblies by glucosylceramide. Neuron. 2018;97(1):92–107 e10.

    Article  CAS  PubMed  Google Scholar 

  44. Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, Caldwell GA, et al. Gaucher disease glucocerebrosidase and alpha-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell. 2011;146(1):37–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Taguchi YV, Liu J, Ruan J, Pacheco J, Zhang X, Abbasi J, et al. Glucosylsphingosine promotes alpha-synuclein pathology in mutant GBA-associated Parkinson's disease. J Neurosci. 2017;37(40):9617–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ikuno M, Yamakado H, Akiyama H, Parajuli LK, Taguchi K, Hara J, et al. GBA haploinsufficiency accelerates alpha-synuclein pathology with altered lipid metabolism in a prodromal model of Parkinson's disease. Hum Mol Genet. 2019;28(11):1894–904.

    Article  CAS  PubMed  Google Scholar 

  47. Rocha EM, Smith GA, Park E, Cao H, Brown E, Hallett P, et al. Progressive decline of glucocerebrosidase in aging and Parkinson's disease. Ann Clin Transl Neurol. 2015;2(4):433–8.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Gegg ME, Sweet L, Wang BH, Shihabuddin LS, Sardi SP, Schapira AH. No evidence for substrate accumulation in Parkinson brains with GBA mutations. Mov Disord. 2015;30(8):1085–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Abbott SK, Li H, Munoz SS, Knoch B, Batterham M, Murphy KE, et al. Altered ceramide acyl chain length and ceramide synthase gene expression in Parkinson's disease. Mov Disord. 2014;29(4):518–26.

    Article  CAS  PubMed  Google Scholar 

  50. Kim MJ, Jeon S, Burbulla LF, Krainc D. Acid ceramidase inhibition ameliorates alpha-synuclein accumulation upon loss of GBA1 function. Hum Mol Genet. 2018;27(11):1972–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Heinrich M, Wickel M, Winoto-Morbach S, Schneider-Brachert W, Weber T, Brunner J, et al. Ceramide as an activator lipid of cathepsin D. Adv Exp Med Biol. 2000;477:305–15.

    Article  CAS  PubMed  Google Scholar 

  52. Sevlever D, Jiang P, Yen SH. Cathepsin D is the main lysosomal enzyme involved in the degradation of alpha-synuclein and generation of its carboxy-terminally truncated species. Biochemistry. 2008;47(36):9678–87.

    Article  CAS  PubMed  Google Scholar 

  53. Yang SY, Gegg M, Chau D, Schapira A. Glucocerebrosidase activity, cathepsin D and monomeric alpha-synuclein interactions in a stem cell derived neuronal model of a PD associated GBA1 mutation. Neurobiol Dis. 2020;134:104620.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dehay B, Martinez-Vicente M, Caldwell GA, Caldwell KA, Yue Z, Cookson MR, et al. Lysosomal impairment in Parkinson's disease. Mov Disord. 2013;28(6):725–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schondorf 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 

  56. Magalhaes J, Gegg ME, Migdalska-Richards A, Doherty MK, Whitfield PD, Schapira AH. Autophagic lysosome reformation dysfunction in glucocerebrosidase deficient cells: relevance to Parkinson disease. Hum Mol Genet. 2016;25(16):3432–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pan T, Rawal P, Wu Y, **e W, Jankovic J, Le W. Rapamycin protects against rotenone-induced apoptosis through autophagy induction. Neuroscience. 2009;164(2):541–51.

    Article  CAS  PubMed  Google Scholar 

  58. Barton NW, Brady RO, Dambrosia JM, Di Bisceglie AM, Doppelt SH, Hill SC, et al. Replacement therapy for inherited enzyme deficiency–macrophage-targeted glucocerebrosidase for Gaucher's disease. N Engl J Med. 1991;324(21):1464–70.

    Article  CAS  PubMed  Google Scholar 

  59. Stojkovska I, Krainc D, Mazzulli JR. Molecular mechanisms of alpha-synuclein and GBA1 in Parkinson's disease. Cell Tissue Res. 2018;373(1):51–60.

    Article  CAS  PubMed  Google Scholar 

  60. Sardi SP, Clarke J, Kinnecom C, Tamsett TJ, Li L, Stanek LM, et al. CNS expression of glucocerebrosidase corrects alpha-synuclein pathology and memory in a mouse model of Gaucher-related synucleinopathy. Proc Natl Acad Sci USA. 2011;108(29):12101–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Rocha EM, Smith GA, Park E, Cao H, Brown E, Hayes MA, et al. Glucocerebrosidase gene therapy prevents alpha-synucleinopathy of midbrain dopamine neurons. Neurobiol Dis. 2015;82:495–503.

    Article  CAS  PubMed  Google Scholar 

  62. Rockenstein E, Clarke J, Viel C, Panarello N, Treleaven CM, Kim C, et al. Glucocerebrosidase modulates cognitive and motor activities in murine models of Parkinson's disease. Hum Mol Genet. 2016;25(13):2645–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Morabito G, Giannelli SG, Ordazzo G, Bido S, Castoldi V, Indrigo M, et al. AAV-PHP.B-mediated global-scale expression in the mouse nervous system enables GBA1 gene therapy for wide protection from synucleinopathy. Mol Therapy. 2017;25(12):2727–42.

    Article  CAS  Google Scholar 

  64. Chang TC, Mendell JT. microRNAs in vertebrate physiology and human disease. Annu Rev Genom Hum Genet. 2007;8:215–39.

    Article  CAS  Google Scholar 

  65. Siebert M, Westbroek W, Chen YC, Moaven N, Li Y, Velayati A, et al. Identification of miRNAs that modulate glucocerebrosidase activity in Gaucher disease cells. RNA Biol. 2014;11(10):1291–300.

    Article  PubMed  Google Scholar 

  66. Hoss AG, Labadorf A, Beach TG, Latourelle JC, Myers RH. microRNA profiles in Parkinson's disease prefrontal cortex. Front Aging Neurosci. 2016;8:36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Straniero L, Rimoldi V, Samarani M, Goldwurm S, Di Fonzo A, Kruger 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 

  68. Burbulla LF, Jeon S, Zheng J, Song P, Silverman RB, Krainc D. A modulator of wild-type glucocerebrosidase improves pathogenic phenotypes in dopaminergic neuronal models of Parkinson's disease. Sci Transl Med. 2019;11(514):eaau6870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Burbulla LF, Song P, Mazzulli JR, Zampese E, Wong YC, Jeon S, et al. Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson's disease. Science. 2017;357(6357):1255–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nguyen M, Krainc D. LRRK2 phosphorylation of auxilin mediates synaptic defects in dopaminergic neurons from patients with Parkinson's disease. Proc Natl Acad Sci USA. 2018;115(21):5576–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Magalhaes J, Gegg ME, Migdalska-Richards A, Schapira AH. Effects of ambroxol on the autophagy-lysosome pathway and mitochondria in primary cortical neurons. Sci Rep. 2018;8(1):1385.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Migdalska-Richards A, Daly L, Bezard E, Schapira AH. Ambroxol effects in glucocerebrosidase and alpha-synuclein transgenic mice. Ann Neurol. 2016;80(5):766–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Migdalska-Richards A, Ko WKD, Li Q, Bezard E, Schapira AHV. Oral ambroxol increases brain glucocerebrosidase activity in a nonhuman primate. Synapse. 2017;71(7):e21967.

    Article  PubMed Central  CAS  Google Scholar 

  74. Kim S, Yun SP, Lee S, Umanah GE, Bandaru VVR, Yin X, et al. GBA1 deficiency negatively affects physiological alpha-synuclein tetramers and related multimers. Proc Natl Acad Sci USA. 2018;115(4):798–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Sardi SP, Viel C, Clarke J, Treleaven CM, Richards AM, Park H, et al. Glucosylceramide synthase inhibition alleviates aberrations in synucleinopathy models. Proc Natl Acad Sci USA. 2017;114(10):2699–704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. 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. 2019;34(1):9–21.

    Article  PubMed  Google Scholar 

  77. Do J, McKinney C, Sharma P, Sidransky E. Glucocerebrosidase and its relevance to Parkinson disease. Mol Neurodegener. 2019;14(1):36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Khanna R, Benjamin ER, Pellegrino L, Schilling A, Rigat BA, Soska R, et al. The pharmacological chaperone isofagomine increases the activity of the Gaucher disease L444P mutant form of beta-glucosidase. FEBS J. 2010;277(7):1618–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Richter F, Fleming SM, Watson M, Lemesre V, Pellegrino L, Ranes B, et al. A GCase chaperone improves motor function in a mouse model of synucleinopathy. Neurotherapeutics. 2014;11(4):840–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sun Y, Liou B, Xu YH, Quinn B, Zhang W, Hamler R, et al. Ex vivo and in vivo effects of isofagomine on acid beta-glucosidase variants and substrate levels in Gaucher disease. J Biol Chem. 2012;287(6):4275–87.

    Article  CAS  PubMed  Google Scholar 

  81. Jung O, Patnaik S, Marugan J, Sidransky E, Westbroek W. Progress and potential of non-inhibitory small molecule chaperones for the treatment of Gaucher disease and its implications for Parkinson disease. Expert Rev Proteom. 2016;13(5):471–9.

    Article  CAS  Google Scholar 

  82. Goldin E, Zheng W, Motabar O, Southall N, Choi JH, Marugan J, et al. High throughput screening for small molecule therapy for Gaucher disease using patient tissue as the source of mutant glucocerebrosidase. PLoS ONE. 2012;7(1):e29861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Patnaik S, Zheng W, Choi JH, Motabar O, Southall N, Westbroek W, et al. Discovery, structure-activity relationship, and biological evaluation of noninhibitory small molecule chaperones of glucocerebrosidase. J Med Chem. 2012;55(12):5734–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Aflaki E, Stubblefield BK, Maniwang E, Lopez G, Moaven N, Goldin E, et al. Macrophage models of Gaucher disease for evaluating disease pathogenesis and candidate drugs. Sci Transl Med. 2014;6(240):240–73.

    Article  CAS  Google Scholar 

  85. Mazzulli JR, Zunke F, Tsunemi T, Toker NJ, Jeon S, Burbulla LF, et al. Activation of beta-glucocerebrosidase reduces pathological alpha-synuclein and restores lysosomal function in Parkinson's patient midbrain neurons. J Neurosci. 2016;36(29):7693–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Aflaki E, Borger DK, Moaven N, Stubblefield BK, Rogers SA, Patnaik S, et al. A new glucocerebrosidase chaperone reduces alpha-synuclein and glycolipid levels in iPSC-derived dopaminergic neurons from patients with Gaucher disease and parkinsonism. J Neurosci. 2016;36(28):7441–522.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Silveira CRA, MacKinley J, Coleman K, Li Z, Finger E, Bartha R, et al. Ambroxol as a novel disease-modifying treatment for Parkinson's disease dementia: protocol for a single-centre, randomized, double-blind, placebo-controlled trial. BMC Neurol. 2019;19(1):20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Maegawa GH, Tropak MB, Buttner JD, Rigat BA, Fuller M, Pandit D, et al. Identification and characterization of ambroxol as an enzyme enhancement agent for Gaucher disease. J Biol Chem. 2009;284(35):23502–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Mullin S, Smith L, Lee K, D'Souza G, Woodgate P, Elflein J, et al. Ambroxol for the treatment of patients with Parkinson disease with and without glucocerebrosidase gene mutations: a nonrandomized, noncontrolled trial. JAMA Neurol. 2020;77(4):427–34.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Bendikov-Bar I, Maor G, Filocamo M, Horowitz M. Ambroxol as a pharmacological chaperone for mutant glucocerebrosidase. Blood Cells Mol Dis. 2013;50(2):141–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Yang SY, Beavan M, Chau KY, Taanman JW, Schapira AHV. A human neural crest stem cell-derived dopaminergic neuronal model recapitulates biochemical abnormalities in GBA1 mutation carriers. Stem Cell Rep. 2017;8(3):728–42.

    Article  CAS  Google Scholar 

  92. Fois G, Hobi N, Felder E, Ziegler A, Miklavc P, Walther P, et al. A new role for an old drug: Ambroxol triggers lysosomal exocytosis via pH-dependent Ca(2)(+) release from acidic Ca(2)(+) stores. Cell Calcium. 2015;58(6):628–37.

    Article  CAS  PubMed  Google Scholar 

  93. Fog CK, Zago P, Malini E, Solanko LM, Peruzzo P, Bornaes C, et al. The heat shock protein amplifier arimoclomol improves refolding, maturation and lysosomal activity of glucocerebrosidase. EBioMedicine. 2018;38:142–53.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Reczek D, Schwake M, Schroder J, Hughes H, Blanz J, ** X, et al. LIMP-2 is a receptor for lysosomal mannose-6-phosphate-independent targeting of beta-glucocerebrosidase. Cell. 2007;131(4):770–83.

    Article  CAS  PubMed  Google Scholar 

  95. Do CB, Tung JY, Dorfman E, Kiefer AK, Drabant EM, Francke U, et al. Web-based genome-wide association study identifies two novel loci and a substantial genetic component for Parkinson's disease. PLoS Genet. 2011;7(6):e1002141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Rothaug M, Zunke F, Mazzulli JR, Schweizer M, Altmeppen H, Lullmann-Rauch R, et al. LIMP-2 expression is critical for beta-glucocerebrosidase activity and alpha-synuclein clearance. Proc Natl Acad Sci USA. 2014;111(43):15573–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Zunke F, Andresen L, Wesseler S, Groth J, Arnold P, Rothaug M, et al. Characterization of the complex formed by beta-glucocerebrosidase and the lysosomal integral membrane protein type-2. Proc Natl Acad Sci USA. 2016;113(14):3791–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ysselstein D, Nguyen M, Young TJ, Severino A, Schwake M, Merchant K, et al. LRRK2 kinase activity regulates lysosomal glucocerebrosidase in neurons derived from Parkinson's disease patients. Nat Commun. 2019;10(1):5570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lu J, Yang C, Chen M, Ye DY, Lonser RR, Brady RO, et al. Histone deacetylase inhibitors prevent the degradation and restore the activity of glucocerebrosidase in Gaucher disease. Proc Natl Acad Sci USA. 2011;108(52):21200–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Yang C, Rahimpour S, Lu J, Pacak K, Ikejiri B, Brady RO, et al. Histone deacetylase inhibitors increase glucocerebrosidase activity in Gaucher disease by modulation of molecular chaperones. Proc Natl Acad Sci USA. 2013;110(3):966–71.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anthony H. V. Schapira.

Ethics declarations

Funding

None.

Conflict of Interest

AS is a principle investigator of the AIM-PD and MOVES-PD studies. EM has no conflicts to report.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Menozzi, E., Schapira, A.H.V. Enhancing the Activity of Glucocerebrosidase as a Treatment for Parkinson Disease. CNS Drugs 34, 915–923 (2020). https://doi.org/10.1007/s40263-020-00746-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-020-00746-0

Navigation