Proton Pump Inhibitors and Dementia: Physiopathological Mechanisms and Clinical Consequences

Gloria Ortiz-Guerrero, Diana Amador-Muñoz, Carlos Alberto Calderón-Ospina, Daniel López-Fuentes, Mauricio Orlando Nava Mesa, Gloria Ortiz-Guerrero, Diana Amador-Muñoz, Carlos Alberto Calderón-Ospina, Daniel López-Fuentes, Mauricio Orlando Nava Mesa

Abstract

Alzheimer's disease (AD) is the most common type of dementia, mainly encompassing cognitive decline in subjects aged ≥65 years. Further, AD is characterized by selective synaptic and neuronal degeneration, vascular dysfunction, and two histopathological features: extracellular amyloid plaques composed of amyloid beta peptide (Aβ) and neurofibrillary tangles formed by hyperphosphorylated tau protein. Dementia and AD are chronic neurodegenerative conditions with a complex physiopathology involving both genetic and environmental factors. Recent clinical studies have shown that proton pump inhibitors (PPIs) are associated with risk of dementia, including AD. However, a recent case-control study reported decreased risk of dementia. PPIs are a widely indicated class of drugs for gastric acid-related disorders, although most older adult users are not treated for the correct indication. Although neurological side effects secondary to PPIs are rare, several preclinical reports indicate that PPIs might increase Aβ levels, interact with tau protein, and affect the neuronal microenvironment through several mechanisms. Considering the controversy between PPI use and dementia risk, as well as both cognitive and neuroprotective effects, the aim of this review is to examine the relationship between PPI use and brain effects from a neurobiological and clinical perspective.

References

    1. Prince M., Wimo A., Guerchet M., Claire Ali G., Wu Y.-T., Prina M. World Alzheimer Report 2015: the Global Impact of Dementia: an Analysis of Prevalence, Incidence, Cost and Trends. London: Alzheimer’s Disease International (ADI); 2015.
    1. World Health Organization. Dementia: a Public Health Priority. World Health Organization. Geneva: World Health Organization; 2012.
    1. Querfurth H. W., LaFerla F. M. Alzheimer’s disease. The New England Journal of Medicine. 2010;362(4):329–344. doi: 10.1056/NEJMra0909142.
    1. Matyas N., Auer S., Gisinger C., et al. Continuing education for the prevention of mild cognitive impairment and Alzheimer’s-type dementia: a systematic review protocol. Systematic Reviews. 2017;6(1):p. 157. doi: 10.1186/s13643-017-0553-0.
    1. Aisen P. S., Cummings J., Jack C. R., et al. On the path to 2025: understanding the Alzheimer’s disease continuum. Alzheimer's Research & Therapy. 2017;9(1):p. 60. doi: 10.1186/s13195-017-0283-5.
    1. Selkoe D. J., Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Molecular Medicine. 2016;8(6):595–608. doi: 10.15252/emmm.201606210.
    1. Goldstein F. C., Steenland K., Zhao L., Wharton W., Levey A. I., Hajjar I. Proton pump inhibitors and risk of mild cognitive impairment and dementia. Journal of the American Geriatrics Society. 2017;65(9):1969–1974. doi: 10.1111/jgs.14956.
    1. Kantor E. D., Rehm C. D., Haas J. S., Chan A. T., Giovannucci E. L. Trends in prescription drug use among adults in the United States from 1999-2012. JAMA. 2015;314(17):1818–1831. doi: 10.1001/jama.2015.13766.
    1. Gomm W., von Holt K., Thomé F., et al. Association of proton pump inhibitors with risk of dementia: a pharmacoepidemiological claims data analysis. JAMA Neurology. 2016;73(4):410–416. doi: 10.1001/jamaneurol.2015.4791.
    1. Heidelbaugh J. J., Inadomi J. M. Magnitude and economic impact of inappropriate use of stress ulcer prophylaxis in non-ICU hospitalized patients. The American Journal of Gastroenterology. 2006;101(10):2200–2205. doi: 10.1111/j.1572-0241.2006.00839.x.
    1. Eid S. M., Boueiz A., Paranji S., Mativo C., Landis R., Abougergi M. S. Patterns and predictors of proton pump inhibitor overuse among academic and non-academic hospitalists. Internal Medicine. 2010;49(23):2561–2568. doi: 10.2169/internalmedicine.49.4064.
    1. Akter S., Hassan M. R., Shahriar M., Akter N., Abbas M. G., Bhuiyan M. A. Cognitive impact after short-term exposure to different proton pump inhibitors: assessment using CANTAB software. Alzheimer's Research & Therapy. 2015;7(1):p. 79. doi: 10.1186/s13195-015-0164-8.
    1. Yu L. Y., Sun L. N., Zhang X. H., et al. A review of the novel application and potential adverse effects of proton pump inhibitors. Advances in Therapy. 2017;34(5):1070–1086. doi: 10.1007/s12325-017-0532-9.
    1. Haenisch B., von Holt K., Wiese B., et al. Risk of dementia in elderly patients with the use of proton pump inhibitors. European Archives of Psychiatry and Clinical Neuroscience. 2015;265(5):419–428. doi: 10.1007/s00406-014-0554-0.
    1. 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(2, article e0171006) doi: 10.1371/journal.pone.0171006.
    1. Booker A., Jacob L. E., Rapp M., Bohlken J., Kostev K. Risk factors for dementia diagnosis in German primary care practices. International Psychogeriatrics. 2016;28(7):1059–1065. doi: 10.1017/S1041610215002082.
    1. Hashioka S., Klegeris A., McGeer P. L. Proton pump inhibitors reduce interferon-γ-induced neurotoxicity and STAT3 phosphorylation of human astrocytes. Glia. 2011;59(5):833–840. doi: 10.1002/glia.21157.
    1. Hashioka S., Klegeris A., McGeer P. L. Proton pump inhibitors exert anti-inflammatory effects and decrease human microglial and monocytic THP-1 cell neurotoxicity. Experimental Neurology. 2009;217(1):177–183. doi: 10.1016/j.expneurol.2009.02.002.
    1. Driel I. R., Callaghan J. M. Proton and potassium transport by H+/K+-ATPases. Clinical and Experimental Pharmacology and Physiology. 1995;22(12):952–960. doi: 10.1111/j.1440-1681.1995.tb02332.x.
    1. Modyanov N. N., Petrukhin K. E., Sverdlov V. E., et al. The family of human Na, K-ATPase genes. ATP1AL1 gene is transcriptionally competent and probably encodes the related ion transport ATPase. FEBS Letters. 1991;278(1):91–94. doi: 10.1016/0014-5793(91)80091-g.
    1. Wang D., Hiesinger P. R. The vesicular ATPase: a missing link between acidification and exocytosis. The Journal of Cell Biology. 2013;203(2):171–173. doi: 10.1083/jcb.201309130.
    1. Tabares L., Betz B. Multiple functions of the vesicular proton pump in nerve terminals. Neuron. 2010;68(6):1020–1022. doi: 10.1016/j.neuron.2010.12.012.
    1. Bublitz M., Poulsen H., Morth J. P., Nissen P. In and out of the cation pumps: P-type ATPase structure revisited. Current Opinion in Structural Biology. 2010;20(4):431–439. doi: 10.1016/j.sbi.2010.06.007.
    1. Sweadner K. J., Donnet C. Structural similarities of Na, K-ATPase and SERCA, the Ca2+-ATPase of the sarcoplasmic reticulum. The Biochemical Journal. 2001;356(3):685–704. doi: 10.1042/bj3560685.
    1. Jaisser F., Horisberger J. D., Geering K., Rossier B. C. Mechanisms of urinary K+ and H+ excretion: primary structure and functional expression of a novel H, K-ATPase. The Journal of Cell Biology. 1993;123(6):1421–1429. doi: 10.1083/jcb.123.6.1421.
    1. Shin J. M., Kim N. Pharmacokinetics and pharmacodynamics of the proton pump inhibitors. Journal of Neurogastroenterology and Motility. 2013;19(1):25–35. doi: 10.5056/jnm.2013.19.1.25.
    1. Shin J. M., Munson K., Vagin O., Sachs G. The gastric HK-ATPase: structure, function, and inhibition. Pflugers Archiv: European Journal of Physiology. 2009;457(3):609–622. doi: 10.1007/s00424-008-0495-4.
    1. Cheng F. C., Ho Y. F., Hung L. C., Chen C. F., Tsai T. H. Determination and pharmacokinetic profile of omeprazole in rat blood, brain and bile by microdialysis and high-performance liquid chromatography. Journal of Chromatography A. 2002;949(1-2):35–42. doi: 10.1016/S0021-9673(01)01225-0.
    1. Rojo L. E., Alzate-Morales J., Saavedra I. N., Davies P., Maccioni R. B. Selective interaction of lansoprazole and astemizole with tau polymers: potential new clinical use in diagnosis of Alzheimer’s disease. Journal of Alzheimer's Disease. 2010;19(2):573–589. doi: 10.3233/JAD-2010-1262.
    1. Liang J.-F., Chen Y.-T., Fuh J.-L., et al. Proton pump inhibitor-related headaches: a nationwide population-based case-crossover study in Taiwan. Cephalalgia. 2014;35(3):203–210. doi: 10.1177/0333102414535114.
    1. Martin R. M., Dunn N. R., Freemantle S., Shakir S. The rates of common adverse events reported during treatment with proton pump inhibitors used in general practice in England: cohort studies. British Journal of Clinical Pharmacology. 2000;50(4):366–372. doi: 10.1046/j.1365-2125.2000.00262.x.
    1. Chimirri S., Aiello R., Mazzitello C., et al. Vertigo/dizziness as a drugs’ adverse reaction. Journal of Pharmacology & Pharmacotherapeutics. 2013;4(5):104–109. doi: 10.4103/0976-500X.120969.
    1. Sebastian Domingo J. J. Omeprazole-induced hallucinations. Not as rare as you might think. Gastroenterología y Hepatología. 2017;41(3) doi: 10.1016/j.gastrohep.2017.05.002.
    1. Hanneken A. M., Babai N., Thoreson W. B. Oral proton pump inhibitors disrupt horizontal cell-cone feedback and enhance visual hallucinations in macular degeneration patients. Investigative Ophthalmology & Visual Science. 2013;54(2):1485–1489. doi: 10.1167/iovs.12-11091.
    1. Heckmann J. G., Birklein F., Neundörfer B. Omeprazole-induced delirium. Journal of Neurology. 2000;247(1):56–57. doi: 10.1007/s004150050011.
    1. de la Coba Ortiz C., Argüelles Arias F., Martín de Argila de Prados C., et al. Proton-pump inhibitors adverse effects: a review of the evidence and position statement by the Sociedad Española de Patología Digestiva. Revista Española de Enfermedades Digestivas. 2016;108(4):207–224. doi: 10.17235/reed.2016.4232/2016.
    1. Badiola N., Alcalde V., Pujol A., et al. The proton-pump inhibitor lansoprazole enhances amyloid beta production. PLoS One. 2013;8(3, article e58837) doi: 10.1371/journal.pone.0058837.
    1. Younkin S. G. The role of Aβ42 in Alzheimer’s disease. Journal of Physiology-Paris. 1998;92(3-4):289–292. doi: 10.1016/S0928-4257(98)80035-1.
    1. DaRocha-Souto B., Scotton T. C., Coma M., et al. Brain oligomeric β-amyloid but not total amyloid plaque burden correlates with neuronal loss and astrocyte inflammatory response in amyloid precursor protein/tau transgenic mice. Journal of Neuropathology & Experimental Neurology. 2011;70(5):360–376. doi: 10.1097/NEN.0b013e318217a118.
    1. Fallahzadeh M. K., Borhani Haghighi A., Namazi M. R. Proton pump inhibitors: predisposers to Alzheimer disease? Journal of Clinical Pharmacy and Therapeutics. 2010;35(2):125–126. doi: 10.1111/j.1365-2710.2009.01100.x.
    1. Namazi M. R., Jowkar F. A succinct review of the general and immunological pharmacologic effects of proton pump inhibitors. Journal of Clinical Pharmacy and Therapeutics. 2008;33(3):215–217. doi: 10.1111/j.1365-2710.2008.00907.x.
    1. Visser P. J., Vos S., van Rossum I., Scheltens P. Comparison of International Working Group criteria and National Institute on Aging–Alzheimer’s Association criteria for Alzheimer’s disease. Alzheimer’s & Dementia. 2012;8(6):560–563. doi: 10.1016/j.jalz.2011.10.008.
    1. McKhann G. M., Knopman D. S., Chertkow H., et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia. 2011;7(3):263–269. doi: 10.1016/j.jalz.2011.03.005.
    1. Carrillo M. C., Dean R. A., Nicolas F., et al. Revisiting the framework of the National Institute on Aging-Alzheimer’s Association diagnostic criteria. Alzheimer’s & Dementia. 2013;9(5):594–601. doi: 10.1016/j.jalz.2013.05.1762.
    1. Dawbarn D., Allen S. J. Neurobiology of Alzheimer’s Disease. Oxford: Bios Scientific; 1995.
    1. Farías G. A., Vial C., Maccioni R. B. Specific macromolecular interactions between tau and the microtubule system. Molecular and Cellular Biochemistry. 1992;112(1):81–88. doi: 10.1007/bf00229646.
    1. Kosik K. S., Joachim C. L., Selkoe D. J. Microtubule-associated protein tau (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America. 1986;83(11):4044–4048. doi: 10.1073/pnas.83.11.4044.
    1. Maccioni R. B., Cambiazo V. Role of microtubule-associated proteins in the control of microtubule assembly. Physiological Reviews. 1995;75(4):835–864. doi: 10.1152/physrev.1995.75.4.835.
    1. Billingsley M. L., Kincaid R. L. Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. The Biochemical Journal. 1997;323(3) Part 3:577–591. doi: 10.1042/bj3230577.
    1. Rojo L. E., Fernández J. A., Maccioni A. A., Jimenez J. M., Maccioni R. B. Neuroinflammation: implications for the pathogenesis and molecular diagnosis of Alzheimer’s disease. Archives of Medical Research. 2008;39(1):1–16. doi: 10.1016/j.arcmed.2007.10.001.
    1. Okamura N., Suemoto T., Furumoto S., et al. Quinoline and benzimidazole derivatives: candidate probes for in vivo imaging of tau pathology in Alzheimer’s disease. The Journal of Neuroscience. 2005;25(47):10857–10862. doi: 10.1523/JNEUROSCI.1738-05.2005.
    1. Braak H., Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathologica. 1991;82(4):239–259. doi: 10.1007/BF00308809.
    1. Fawaz M. V., Brooks A. F., Rodnick M. E., et al. High affinity radiopharmaceuticals based upon lansoprazole for PET imaging of aggregated tau in Alzheimer’s disease and progressive supranuclear palsy: synthesis, preclinical evaluation, and lead selection. ACS Chemical Neuroscience. 2014;5(8):718–730. doi: 10.1021/cn500103u.
    1. Villemagne V. L., Okamura N. Tau imaging in the study of ageing, Alzheimer’s disease, and other neurodegenerative conditions. Current Opinion in Neurobiology. 2016;36:43–51. doi: 10.1016/j.conb.2015.09.002.
    1. Spillantini M. G., Goedert M. Tau pathology and neurodegeneration. The Lancet Neurology. 2013;12(6):609–622. doi: 10.1016/S1474-4422(13)70090-5.
    1. Oh R., Brown D. L. Vitamin B12 deficiency. American Family Physician. 2003;67(5):979–986.
    1. Hunt A., Harrington D., Robinson S. Vitamin B12 deficiency. BMJ. 2014;349, article g5226 doi: 10.1136/bmj.g5226.
    1. Savarino V., Dulbecco P., Savarino E. Are proton pump inhibitors really so dangerous? Digestive and Liver Disease. 2016;48(8):851–859. doi: 10.1016/j.dld.2016.05.018.
    1. Lam J. R., Schneider J. L., Zhao W., Corley D. A. Proton pump inhibitor and histamine 2 receptor antagonist use and vitamin B12 deficiency. JAMA. 2013;310(22):2435–2442. doi: 10.1001/jama.2013.280490.
    1. Lombardo L., Foti M., Ruggia O., Chiecchio A. Increased incidence of small intestinal bacterial overgrowth during proton pump inhibitor therapy. Clinical Gastroenterology and Hepatology. 2010;8(6):504–508. doi: 10.1016/j.cgh.2009.12.022.
    1. Linder L., Tamboue C., Clements J. N. Drug-induced vitamin B12 deficiency: a focus on proton pump inhibitors and histamine-2 antagonists. Journal of Pharmacy Practice. 2017;30(6):639–642. doi: 10.1177/0897190016663092.
    1. Rafiee S., Asadollahi K., Riazi G., Ahmadian S., Saboury A. A. Vitamin B12 inhibits tau fibrillization via binding to cysteine residues of tau. ACS Chemical Neuroscience. 2017;8(12):2676–2682. doi: 10.1021/acschemneuro.7b00230.
    1. Penninx B. W., Guralnik J. M., Ferrucci L., Fried L. P., Allen R. H., Stabler S. P. Vitamin B12 deficiency and depression in physically disabled older women: epidemiologic evidence from the Women’s Health and Aging Study. The American Journal of Psychiatry. 2000;157(5):715–721. doi: 10.1176/appi.ajp.157.5.715.
    1. Ma F., Wu T., Zhao J., et al. Plasma homocysteine and serum folate and vitamin B12 levels in mild cognitive impairment and Alzheimer’s disease: a case-control study. Nutrients. 2017;9(12) doi: 10.3390/nu9070725.
    1. Mandelkow E. M., Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harbor Perspectives in Medicine. 2012;2(7, article a006247) doi: 10.1101/cshperspect.a006247.
    1. Bottiglieri T., Arning E., Wasek B., Nunbhakdi-Craig V., Sontag J. M., Sontag E. Acute administration of L-DOPA induces changes in methylation metabolites, reduced protein phosphatase 2A methylation, and hyperphosphorylation of tau protein in mouse brain. The Journal of Neuroscience. 2012;32(27):9173–9181. doi: 10.1523/JNEUROSCI.0125-12.2012.
    1. Wei W., Liu Y. H., Zhang C. E., et al. Folate/vitamin-B12 prevents chronic hyperhomocysteinemia-induced tau hyperphosphorylation and memory deficits in aged rats. Journal of Alzheimer's Disease. 2011;27(3):639–650. doi: 10.3233/JAD-2011-110770.
    1. Zhang C. E., Tian Q., Wei W., et al. Homocysteine induces tau phosphorylation by inactivating protein phosphatase 2A in rat hippocampus. Neurobiology of Aging. 2008;29(11):1654–1665. doi: 10.1016/j.neurobiolaging.2007.04.015.
    1. Zhang C. E., Wei W., Liu Y. H., et al. Hyperhomocysteinemia increases β-amyloid by enhancing expression of γ-secretase and phosphorylation of amyloid precursor protein in rat brain. The American Journal of Pathology. 2009;174(4):1481–1491. doi: 10.2353/ajpath.2009.081036.
    1. Ravaglia G., Forti P., Maioli F., et al. Homocysteine and folate as risk factors for dementia and Alzheimer disease. The American Journal of Clinical Nutrition. 2005;82(3):636–643. doi: 10.1093/ajcn/82.3.636.
    1. Heneka M. T., Rodríguez J. J., Verkhratsky A. Neuroglia in neurodegeneration. Brain Research Reviews. 2010;63(1-2):189–211. doi: 10.1016/j.brainresrev.2009.11.004.
    1. Mizuno T., Kuno R., Nitta A., et al. Protective effects of nicergoline against neuronal cell death induced by activated microglia and astrocytes. Brain Research. 2005;1066(1-2):78–85. doi: 10.1016/j.brainres.2005.10.050.
    1. Gonzalez-Reyes R. E., Nava-Mesa M. O., Vargas-Sanchez K., Ariza-Salamanca D., Mora-Muñoz L. Involvement of astrocytes in Alzheimer’s disease from a neuroinflammatory and oxidative stress perspective. Frontiers in Molecular Neuroscience. 2017;10:p. 427. doi: 10.3389/fnmol.2017.00427.
    1. Sofroniew M. V., Vinters H. V. Astrocytes: biology and pathology. Acta Neuropathologica. 2010;119(1):7–35. doi: 10.1007/s00401-009-0619-8.
    1. Cronican A. A., Fitz N. F., Pham T., et al. Proton pump inhibitor lansoprazole is a nuclear liver X receptor agonist. Biochemical Pharmacology. 2010;79(9):1310–1316. doi: 10.1016/j.bcp.2009.12.018.
    1. Batchelor R., Gilmartin J. F., Kemp W., Hopper I., Liew D. Dementia, cognitive impairment and proton pump inhibitor therapy: a systematic review. Journal of Gastroenterology and Hepatology. 2017;32(8):1426–1435. doi: 10.1111/jgh.13750.
    1. Herghelegiu A. M., Prada G. I., Nacu R. Prolonged use of proton pomp inhibitors and cognitive function in older adults. Farmácia. 2016;64(2):262–267.
    1. Delgado M. G., Calleja S., Suarez L., Pascual J. Recurrent confusional episodes associated with hypomagnesaemia due to esomeprazol. BMJ Case Reports. 2013;2013, article bcr2013200501 doi: 10.1136/bcr-2013-200501.
    1. Pasina L., Zanotta D., Puricelli S., Bonoldi G. Acute neurological symptoms secondary to hypomagnesemia induced by proton pump inhibitors: a case series. European Journal of Clinical Pharmacology. 2016;72(5):641–643. doi: 10.1007/s00228-016-2024-2.
    1. Bebarta V. S., King J. A., McDonough M. Proton pump inhibitor-induced rhabdomyolysis and hyponatremic delirium. The American Journal of Emergency Medicine. 2008;26(4):519.e1–519.e2. doi: 10.1016/j.ajem.2007.08.026.
    1. Fujii S., Tanimukai H., Kashiwagi Y. Comparison and analysis of delirium induced by histamine H2 receptor antagonists and proton pump inhibitors in cancer patients. Case Reports in Oncology. 2012;5(2):409–412. doi: 10.1159/000341873.
    1. Otremba I., Wilczyński K., Szewieczek J. Delirium in the geriatric unit: proton-pump inhibitors and other risk factors. Clinical Interventions in Aging. 2016;11:397–405. doi: 10.2147/CIA.S103349.
    1. Soares F. C., de Oliveira T. C., de Macedo L. D., et al. CANTAB object recognition and language tests to detect aging cognitive decline: an exploratory comparative study. Clinical Interventions in Aging. 2015;10:37–48. doi: 10.2147/CIA.S68186.
    1. Lochhead P., Hagan K., Joshi A. D., et al. Association between proton pump inhibitor use and cognitive function in women. Gastroenterology. 2017;153(4):971–979.e4. doi: 10.1053/j.gastro.2017.06.061.
    1. Li W., Zeng S., Yu L.-S., Zhou Q. Pharmacokinetic drug interaction profile of omeprazole with adverse consequences and clinical risk management. Therapeutics and Clinical Risk Management. 2013;9:259–271. doi: 10.2147/TCRM.S43151.
    1. Parsons C., Johnston S., Mathie E., et al. Potentially inappropriate prescribing in older people with dementia in care homes. Drugs & Aging. 2012;29(2):143–155. doi: 10.2165/11598560-000000000-00000.
    1. Labenz J., Petersen K. U., Rösch W., Koelz H. R. A summary of Food and Drug Administration-reported adverse events and drug interactions occurring during therapy with omeprazole, lansoprazole and pantoprazole. Alimentary Pharmacology and Therapeutics. 2003;17(8):1015–1019. doi: 10.1046/j.1365-2036.2003.01550.x.
    1. Gallacher J., Elwood P., Pickering J., Bayer A., Fish M., Ben-Shlomo Y. Benzodiazepine use and risk of dementia: evidence from the Caerphilly Prospective Study (CaPS) Journal of Epidemiology & Community Health. 2012;66(10):869–873. doi: 10.1136/jech-2011-200314.
    1. Billioti de Gage S., Pariente A., Begaud B. Is there really a link between benzodiazepine use and the risk of dementia? Expert Opinion on Drug Safety. 2015;14(5):733–747. doi: 10.1517/14740338.2015.1014796.
    1. Nava-Mesa M. O., Jimenez-Diaz L., Yajeya J., Navarro-Lopez J. D. GABAergic neurotransmission and new strategies of neuromodulation to compensate synaptic dysfunction in early stages of Alzheimer’s disease. Frontiers in Cellular Neuroscience. 2014;8:p. 167. doi: 10.3389/fncel.2014.00167.
    1. Stonnington C. M., Snyder P. J., Hentz J. G., Reiman E. M., Caselli R. J. Double-blind crossover study of the cognitive effects of lorazepam in healthy apolipoprotein E (APOE)-ε4 carriers. The Journal of Clinical Psychiatry. 2009;70(10):1379–1384. doi: 10.4088/JCP.08m04593.
    1. Gugler R., Hartmann M., Rudi J., et al. Lack of pharmacokinetic interaction of pantoprazole with diazepam in man. British Journal of Clinical Pharmacology. 1996;42(2):249–252. doi: 10.1046/j.1365-2125.1996.40619.x.
    1. Lefebvre R. A., Flouvat B., Karolac-Tamisier S., Moerman E., Ganse E. V. Influence of lansoprazole treatment on diazepam plasma concentrations. Clinical Pharmacology & Therapeutics. 1992;52(5):458–463. doi: 10.1038/clpt.1992.172.
    1. Ishizaki T., Chiba K., Manabe K., et al. Comparison of the interaction potential of a new proton pump inhibitor, E3810, versus omeprazole with diazepam in extensive and poor metabolizers of S-mephenytoin 4′-hydroxylation. Clinical Pharmacology & Therapeutics. 1995;58(2):155–164. doi: 10.1016/0009-9236(95)90193-0.
    1. Pasinetti G. M. From epidemiology to therapeutic trials with anti-inflammatory drugs in Alzheimer’s disease: the role of NSAIDs and cyclooxygenase in β-amyloidosis and clinical dementia. Journal of Alzheimer’s Disease. 2002;4(5):435–445. doi: 10.3233/JAD-2002-4510.
    1. McGeer P. L., McGeer E., Rogers J., Sibley J. Anti-inflammatory drugs and Alzheimer disease. The Lancet. 1990;335(8696):p. 1037. doi: 10.1016/0140-6736(90)91101-F.
    1. Ghallab A. In vitro test systems and their limitations. EXCLI Journal. 2013;12:1024–1026.
    1. Bell R. D., Zlokovic B. V. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathologica. 2009;118(1):103–113. doi: 10.1007/s00401-009-0522-3.
    1. Shah G. N., Mooradian A. D. Age-related changes in the blood-brain barrier. Experimental Gerontology. 1997;32(4-5):501–519. doi: 10.1016/S0531-5565(96)00158-1.
    1. Kleine T. O., Hackler R., Zofel P. Age-related alterations of the blood-brain-barrier (bbb) permeability to protein molecules of different size. Zeitschrift für Gerontologie. 1993;26(4):256–259.

Source: PubMed

3
S'abonner