2,582
Views
0
CrossRef citations to date
0
Altmetric
Commentary and Views

Should evidence of an autolysosomal de-acidification defect in Alzheimer and Parkinson diseases call for caution in prescribing chronic PPI and DMARD?

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 2800-2806 | Received 14 Sep 2022, Accepted 08 May 2023, Published online: 23 Jul 2023

References

  • Vakifahmetoglu-Norberg H, Xia H, Yuan J. Pharmacologic agents targeting autophagy. J Clin Invest. 2015;125:5–13.
  • Mauthe M, Orhon I, Rocchi C, et al. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy. 2018;14:1435–1455.
  • Kim C-H, Han B-S, Moon J, et al. Nuclear receptor Nurr1 agonists enhance its dual functions and improve behavioral deficits in an animal model of Parkinson’s disease. Proc Natl Acad Sci U S A. 2015;112:8756–8761.
  • Chen X, Wang N, Zhu Y, et al. The antimalarial chloroquine suppresses LPS-Induced NLRP3 inflammasome activation and confers protection against murine endotoxic shock. Mediators Inflamm. 2017;2017:6543237.
  • Kong X, Chen L, Jiao L, et al. Astemizole arrests the proliferation of cancer cells by disrupting the EZH2-EED interaction of polycomb repressive complex 2. J Med Chem. 2014;57:9512–9521.
  • Quiros Roldan E, Biasiotto G, Magro P, et al. The possible mechanisms of action of 4-aminoquinolines (chloroquine/hydroxychloroquine) against Sars-Cov-2 infection (COVID-19): a role for iron homeostasis? Pharmacol Res. 2020;158:104904.
  • Bondeson J, Sundler R. Antimalarial drugs inhibit phospholipase A2 activation and induction of interleukin lβ and tumor necrosis factor α in macrophages: implications for their mode of action in rheumatoid arthritis. Gen Pharmacol Vasc Syst. 1998;30:357–366.
  • Chou M-H, Wang J-Y, Lin C-L, et al. DMARD use is associated with a higher risk of dementia in patients with rheumatoid arthritis: a propensity score-matched case–control study. Toxicol Appl Pharmacol. 2017;334:217–222.
  • Van Gool WA, Weinstein HC, Scheltens P, et al. Effect of hydroxychloroquine on progression of dementia in early Alzheimer’s disease: an 18-month randomised, double-blind, placebo-controlled study. Lancet. 2001;358:455–460.
  • Fardet L, Nazareth I, Petersen I. Chronic hydroxychloroquine/chloroquine exposure for connective tissue diseases and risk of Alzheimer’s disease: a population-based cohort study. Ann Rheum Dis. 2019;78:279–282.
  • Savarino V, Dulbecco P, de Bortoli N, et al. The appropriate use of proton pump inhibitors (PPIs): need for a reappraisal. Eur J Intern Med. 2017;37:19–24.
  • Elias E, Targownik LE. The clinician’s guide to proton pump inhibitor related adverse events. Drugs. 2019;79:715–731.
  • Vaezi MF, Yang Y-X, Howden CW. Complications of proton pump inhibitor therapy. Gastroenterology. 2017;153:35–48.
  • Pottegård A, Broe A, Hallas J, et al. Use of proton-pump inhibitors among adults: a Danish nationwide drug utilization study. Therap Adv Gastroenterol. 2016;9:671–678.
  • Daniels B, Pearson S-A, Buckley NA, et al. Long-term use of proton-pump inhibitors: whole-of-population patterns in Australia 2013–2016. Therap Adv Gastroenterol. 2020;13:1756284820913743.
  • Bustillos H, Leer K, Kitten A, et al. A cross-sectional study of national outpatient gastric acid suppressant prescribing in the United States between 2009 and 2015. PLoS ONE. 2018;13:e0208461.
  • Lassalle M, Le Tri T, Bardou M, et al. Use of proton pump inhibitors in adults in France: a nationwide drug utilization study. Eur J Clin Pharmacol. 2020;76(3):449–457. DOI:10.1007/s00228-019-02810-1
  • Kim J-H, Oh J-K, Kim Y-H, et al. Association between proton pump inhibitor use and parkinson’s disease in a Korean Population. Pharmaceuticals (Basel). 2022;15(3):327. DOI:10.3390/ph15030327
  • Torres-Bondia F, Dakterzada F, Galván L, et al. Proton pump inhibitors and the risk of Alzheimer’s disease and non-Alzheimer’s dementias. Sci Rep. 2020;10(1):21046. DOI:10.1038/s41598-020-78199-0
  • Fallahzadeh MK, Borhani Haghighi A, Namazi MR. Proton pump inhibitors: predisposers to Alzheimer disease? J Clin Pharm Ther. 2010;35(2):125–126.
  • Haenisch B, von Holt K, Wiese B, et al. Risk of dementia in elderly patients with the use of proton pump inhibitors. Eur Arch Psychiatry Clin Neurosci. 2015;265(5):419–428. DOI:10.1007/s00406-014-0554-0
  • Gomm W, von Holt K, Thomé F, et al. Association of proton pump inhibitors with risk of dementia: a pharmacoepidemiological claims data analysis. JAMA Neurol. 2016;73(4):410–416. DOI:10.1001/jamaneurol.2015.4791
  • World Health Organization, Risk Reduction of Cognitive Decline and Dementia: WHO Guidelines WHO | Risk Reduction of Cognitive Decline and Dementia [Internet]. Geneva: World Health Organization; 2019 [cited 2022 Jul 29]. Available from 2019: http://www.ncbi.nlm.nih.gov/books/NBK542796/.
  • Norton S , Matthews F.E , Barnes D.E , Yaffe K , Brayne C, et al. Potential for primary prevention of Alzheimer's disease: an analysis of population-based data. Lancet Neurol. 2014;13(8):788–794. DOI:10.1016/S1474-4422(14)70136-X.
  • Ortiz-Guerrero G, Amador-Muñoz D, Calderón-Ospina CA, et al. Proton pump inhibitors and dementia: physiopathological mechanisms and clinical consequences. Neural Plast. 2018;2018:5257285.
  • Rojo LE, Alzate-Morales J, Saavedra IN, et al. Selective interaction of lansoprazole and astemizole with tau polymers: potential new clinical use in diagnosis of Alzheimer’s Disease. J Alzheimers Dis. 2010;19(2):573–589. DOI:10.3233/JAD-2010-1262
  • Cheng FC, Ho YF, Hung LC, et al. Determination and pharmacokinetic profile of omeprazole in rat blood, brain and bile by microdialysis and high-performance liquid chromatography. J Chromatogr A. 2002;949(1–2):35–42. DOI:10.1016/S0021-9673(01)01225-0
  • Nighot M, Nighot P, Ma T. Proton Pump Inhibitors (PPI) induces colonic Tight Junction barrier (TJ) dysfunction via an upregulation of TJ pore forming Caludin-2 protein. Inflamm Bowel Dis. 2021;27:S27–28.
  • Thongon N, Chamniansawat S. Hippocampal synaptic dysfunction and spatial memory impairment in omeprazole-treated rats. Metab Brain Dis. 2022;37:2871–2881.
  • Ali SB, Mahmood K, Saeed R, et al. Elevated anxiety, hypoactivity, memory deficits, decreases of brain serotonin and 5-HT-1A receptors expression in rats treated with omeprazole. Toxicol Res. 2020;37:237–248.
  • Mascolo A, Berrino PM, Gareri P, et al. Neuropsychiatric clinical manifestations in elderly patients treated with hydroxychloroquine: a review article. Inflammopharmacology. 2018;26:1141–1149.
  • Klionsky DJ, Petroni G, Amaravadi RK, et al. Autophagy in major human diseases. EMBO J. 2021 Oct 1;40(19):e108863. https://onlinelibrary.wiley.com/doi/10.15252/embj.2021108863
  • Grosjean I, Roméo B, Domdom M-A, et al. Autophagopathies: from autophagy gene polymorphisms to precision medicine for human diseases. Autophagy. 2022;18:2519–2536.
  • Navarro-Romero A, Montpeyó M, Martinez-Vicente M. The emerging role of the lysosome in Parkinson’s Disease. Cells. 2020;9:2399.
  • Klein AD, Mazzulli JR. Is Parkinson’s disease a lysosomal disorder? Brain. 2018;141:2255–2262.
  • Blauwendraat C, Nalls MA, Singleton AB. The genetic architecture of Parkinson’s disease. Lancet Neurol. 2020;19(2):170–178.
  • Navarro-Romero A, Fernandez-Gonzalez I, Riera J, et al. Lysosomal lipid alterations caused by glucocerebrosidase deficiency promote lysosomal dysfunction, chaperone-mediated-autophagy deficiency, and alpha-synuclein pathology. NPJ Parkinsons Dis. 2022 Oct 6;8(1):126. DOI:10.1038/s41531-022-00397-6
  • Song Q, Meng B, Xu H, et al. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl Neurodegener. 2020;9(1):17. DOI:10.1186/s40035-020-00196-0
  • Dehay B, Bové J, Rodríguez-Muela N, et al. Pathogenic lysosomal depletion in Parkinson’s disease. J Neurosci. 2010;30(37):12535–12544. DOI:10.1523/JNEUROSCI.1920-10.2010
  • Bourdenx M, Daniel J, Genin E, et al. Nanoparticles restore lysosomal acidification defects: implications for Parkinson and other lysosomal-related diseases. Autophagy. 2016;12:472–483.
  • Nixon RA, Wegiel J, Kumar A, et al. Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol. 2005;64(2):113–122. DOI:10.1093/jnen/64.2.113
  • Bai B, Wang X, Li Y, et al. Deep multilayer brain proteomics identifies molecular networks in Alzheimer’s Disease Progression. Neuron. 2020;105:975–991.e7.
  • Lee J-H, Yang D-S, Goulbourne CN, et al. Faulty autolysosome acidification in Alzheimer’s disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat Neurosci. 2022;25(6):688–701. DOI:10.1038/s41593-022-01084-8
  • Lie PPY, Yoo L, Goulbourne CN, et al. Axonal transport of late endosomes and amphisomes is selectively modulated by local Ca2+ efflux and disrupted by PSEN1 loss of function. Sci Adv. 2022;8(17):eabj5716. DOI:10.1126/sciadv.abj5716
  • Lee J-H, McBrayer MK, Wolfe DM, et al. Presenilin 1 maintains lysosomal Ca2+ homeostasis via TRPML1 by regulating vatpase-mediated lysosome acidification. Cell Rep. 2015;12(9):1430–1444. DOI:10.1016/j.celrep.2015.07.050
  • Im E, Jiang Y, Stavrides P, et al. Lysosomal dysfunction in Down Syndrome and Alzheimer mouse models is caused by selective v-ATPase inhibition by Tyr682 phosphorylated APP βCTF. bioRxiv. DOI:10.1101/2022.06.02.494546.
  • Lee J-H, Yu WH, Kumar A, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by alzheimer-related PS1 mutations. Cell. 2010;141:1146–1158.
  • Tong B-K, Wu AJ, Huang AS, et al. Lysosomal TPCN (two pore segment channel) inhibition ameliorates beta-amyloid pathology and mitigates memory impairment in Alzheimer disease. Autophagy. 2022;18(3):624–642. DOI:10.1080/15548627.2021.1945220
  • Wolfe DM, Lee J, Kumar A, et al. Autophagy failure in Alzheimer’s disease and the role of defective lysosomal acidification. Eur J Neurosci. 2013;37(12):1949–1961. DOI:10.1111/ejn.12169
  • Torres M, Jimenez S, Sanchez-Varo R, et al. Defective lysosomal proteolysis and axonal transport are early pathogenic events that worsen with age leading to increased APP metabolism and synaptic Abeta in transgenic APP/PS1 hippocampus. Mol Neurodegener. 2012;7(1):59. DOI:10.1186/1750-1326-7-59
  • Avrahami L, Farfara D, Shaham-Kol M, et al. Inhibition of glycogen synthase kinase-3 ameliorates β-amyloid pathology and restores lysosomal acidification and mammalian target of rapamycin activity in the alzheimer disease mouse model. J Biol Chem. 2013;288(2):1295–1306. DOI:10.1074/jbc.M112.409250
  • Jiang Y, Sato Y, Im E, et al. Lysosomal dysfunction in down syndrome is APP-Dependent and mediated by APP-βCTF (C99). J Neurosci. 2019;39(27):5255–5268. DOI:10.1523/JNEUROSCI.0578-19.2019
  • Lee J-H, Mitchell RR, McNicol JD, et al. Single transcription factor conversion of human blood fate to NPCs with CNS and PNS developmental capacity. Cell Rep. 2015;11(9):1367–1376. DOI:10.1016/j.celrep.2015.04.056
  • Butzlaff M, Hannan SB, Karsten P, et al. Impaired retrograde transport by the Dynein/Dynactin complex contributes to Tau-induced toxicity. Hum Mol Genet. 2015;24:3623–3637.
  • Yu WH, Kumar A, Peterhoff C, et al. Autophagic vacuoles are enriched in amyloid precursor protein-secretase activities: implications for beta-amyloid peptide over-production and localization in Alzheimer’s disease. Int J Biochem Cell Biol. 2004;36:2531–2540.
  • Solvik TA, Nguyen TA, Tony Lin Y-H, et al. Secretory autophagy maintains proteostasis upon lysosome inhibition. J Cell Bio. 2022;221(6):e202110151. DOI:10.1083/jcb.202110151
  • Tai S-Y, Chien C-Y, Wu D-C, et al. Risk of dementia from proton pump inhibitor use in Asian population: a nationwide cohort study in Taiwan. PLoS ONE. 2017;12:e0171006.
  • Kim J-H, Oh J-K, Kim Y-H, et al. Association between proton pump inhibitor use and parkinson’s disease in a Korean population. Pharmaceuticals (Basel). 2022;15:327.
  • Choi HG, Kim J-H, Kim JH, et al. Associations between proton pump inhibitors and Alzheimer’s disease: a nested case–control study using a Korean nationwide health screening cohort. Alzheimers Res Ther. 2022;14(1):91. DOI:10.1186/s13195-022-01032-5
  • Lai S-W, Liao K-F, Lin C-L, et al. Association between Parkinson’s disease and proton pump inhibitors therapy in older people. Biomedicine (Taipei). 2020;10:1–4.
  • Nielsen HH, Qiu J, Friis S, et al. Treatment for Helicobacter pylori infection and risk of Parkinson’s disease in Denmark. Eur J Neurol. 2012;19:864–869.
  • Galatti L, Giustini SE, Sessa A, et al. Neuropsychiatric reactions to drugs: an analysis of spontaneous reports from general practitioners in Italy. Pharmacol Res. 2005;51:211–216.
  • Laudisio A, Incalzi RA, Gemma A, et al. Use of proton-pump inhibitors is associated with depression: a population-based study. Int Psychogeriatr. 2018;30:153–159.
  • Akter S, MdR H, Shahriar M, et al. Cognitive impact after short-term exposure to different proton pump inhibitors: assessment using CANTAB software. Alzheimers Res Ther. 2015;7(1):79. DOI:10.1186/s13195-015-0164-8
  • Polimeni G, Cutroneo P, Gallo A, et al. Rabeprazole and psychiatric symptoms. Ann Pharmacother. 2007;41(7–8):1315–1317. DOI:10.1345/aph.1K134
  • Heckmann JG, Birklein F, Neundörfer B. Omeprazole-induced delirium. J Neurol. 2000;247(1):56–57.
  • Abela M, Aquilina N, Attard A. Omeprazole-induced delirium. The Synapse. 2014;13:18,19.
  • Makunts T, Alpatty S, Lee KC, et al. Proton-pump inhibitor use is associated with a broad spectrum of neurological adverse events including impaired hearing, vision, and memory. Sci Rep. 2019;9:17280.
  • Trevisani S, Cereda J-M. Blurred vision: a rare secondary effect of proton pump inhibitors. Rev Med Suisse. 2012;8(336):811–812, 814.
  • Schönhöfer PS, Werner B, Tröger U. Ocular damage associated with proton pump inhibitors. BMJ. 1997;314(7097):1805.
  • Wang AK, Sharma S, Kim P, et al. Hypomagnesemia in the intensive care unit: choosing your gastrointestinal prophylaxis, a case report and review of the literature. Indian J Crit Care Med. 2014;18:456–460.
  • Rajabally YA, Jacob S. Neuropathy associated with lansoprazole treatment. Muscle Nerve. 2005;31:124–125.
  • Otremba I, Wilczyński K, Szewieczek J. Delirium in the geriatric unit: proton-pump inhibitors and other risk factors. Clin Interv Aging. 2016;11:397–405.
  • Targownik LE, Fisher DA, Saini SD. AGA clinical practice update on de-prescribing of proton pump inhibitors: expert review. Gastroenterology. 2022;162:1334–1342.
  • Booker A, Jacob LE, Rapp M, et al. Risk factors for dementia diagnosis in German primary care practices. Int Psychogeriatr. 2016;28(7):1059–1065. DOI:10.1017/S1041610215002082
  • Cooksey R, Kennedy J, Dennis MS, et al. Proton pump inhibitors and dementia risk: Evidence from a cohort study using linked routinely collected national health data in Wales, UK. PLoS ONE. 2020;15:e0237676.
  • Goldstein FC, Steenland K, Zhao L, et al. Proton Pump Inhibitors and Risk for Mild Cognitive Impairment and Dementia. J Am Geriatr Soc. 2017;65(9):1969–1974. DOI:10.1111/jgs.14956
  • Taipale H, Tolppanen A-M, Tiihonen M, et al. No Association Between Proton Pump Inhibitor Use and Risk of Alzheimer’s Disease. Am J Gastroenterol. 2017;112(12):1802–1808. DOI:10.1038/ajg.2017.196
  • Gray SL, Walker RL, Dublin S, et al. Proton Pump Inhibitor Use and Dementia Risk: Prospective Population Based Study. J Am Geriatr Soc. 2018;66(2):247–253. DOI:10.1111/jgs.15073
  • Lo C-H, Ni P, Yan Y, et al. Association of Proton Pump Inhibitor Use With All-Cause and Cause-Specific Mortality. Gastroenterology. 2022;S0016-5085(22):00729.
  • Song YQ, Li Y, Zhang SL, et al. Proton pump inhibitor use does not increase dementia and Alzheimer’s disease risk: An updated meta-analysis of published studies involving 642305 patients. PLoS ONE. 2019;14(7):e0219213. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6605652/
  • Komatsu M, Waguri S, Chiba T, et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;441:880–884.
  • Komatsu M, Waguri S, Koike M, et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell. 2007;131:1149–1163.
  • Hara T, Nakamura K, Matsui M, et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006;441:885–889.