Estrategias terapéuticas en la enfermedad de Alzheimer

Alfredo Sanabria-Castro, Ileana Alvarado-Echeverría, Cecilia Monge-Bonilla

Resumen


La enfermedad de Alzheimer es el tipo de demencia más común a nivel mundial, sin embargo en la actualidad no existe ningún tratamiento curativo para la misma. La terapéutica aprobada para su manejo se limita a disminuir síntomas específicos y mejorar la calidad de vida de los pacientes. Razón por la cual la búsqueda de nuevas estrategias terapéuticas se ha convertido en una labor necesaria y de gran relevancia. La presente revisión bibliográfica resume brevemente las principales teorías de la patogénesis de la enfermedad de Alzheimer y aborda de lleno los principales fármacos utilizados en el tratamiento presente y futuro de la misma. En ésta se incluyen los tratamientos aprobados, las moléculas en investigación y otras sustancias de uso común.

 

Abstract

Although Alzheimer’s disease is the most common type of dementia currently there is no cure for it. Approved regulatory agencies therapeutic drugs limit to reduce specific symptoms and mildly improve the quality of life of patients. On this matter, the search for new therapeutic strategies has become a relevant necessity. This literature review briefly summarizes the main theories of the pathogenesis of Alzheimer's disease and addresses the main drugs that are used in the present.  Other therapeutic strategies on study and substances commonly used are also discussed.

Palabras clave: Alzheimer’s disease; cholinesterase inhibitors; muscarinic receptors; nicotinic receptors.


Palabras clave


Enfermedad de Alzheimer; inhibidores de la acetilcolinesterasa; receptores muscarínicos; receptores nicotínicos.

Referencias


Mc Khann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. 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. Alzheimers Dement. 2011;7(3):263–9.

Barber RC, Barber RC. The Genetics of Alzheimer’s Disease. Scientifica (Cairo). DOI:10.6064/2012/246210.

Schu MC, Sherva R, Farrer LA, Green RC. The genetics of Alzheimer’s disease. Alzheimer’s Dis Mod Concept, Biol Diagnosis Ther. 2012;28:15–29.

Schaeffer EL, Gattaz WF. Cholinergic and glutamatergic alterations beginning at the early stages of Alzheimer disease: participation of the phospholipase A2 enzyme. Psychopharmacology (Berl). 2008;198(1):1–27.

Albuquerque EX, Pereira EFR, Alkondon M, Rogers SW. Mammalian nicotinic acetylcholine receptors: from structure to function. Physiol Rev. 2009;89(1):73–120.

Watanabe T, Yamagata N, Takasaki K, Sano K, Hayakawa K, Katsurabayashi S, Egashira N, Mishima K, Iwasaki K, Fujiwara M. Decreased acetylcholine release is correlated to memory impairment in the Tg2576 transgenic mouse model of Alzheimer’s disease. Brain Res. 2009;1249:222–8.

Geerts H, Grossberg GT. Pharmacology of acetylcholinesterase inhibitors and N-methyl-D-aspartate receptors for combination therapy in the treatment of Alzheimer’s disease. J Clin Pharmacol. 2006;46(7 Suppl 1):8S–16S.

Lin H, Vicini S, Hsu F-C, Doshi S, Takano H, Coulter DA, Lynch DR. Axonal α7 nicotinic ACh receptors modulate presynaptic NMDA receptor expression and structural plasticity of glutamatergic presynaptic boutons. Proc Natl Acad Sci U S A. 2010;107(38):16661–6.

>Dong X, Wang Y, Qin Z. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol Sin. 2009;30(4):379–87.

Mohandas E, Rajmohan V, Raghunath B. Neurobiology of Alzheimers disease. Indian J Psychiatry. 2009;51(1):55.

Peña F, Gutiérrez-Lerma A, Quiroz-Baez R, Arias C. The role of beta-amyloid protein in synaptic function: implications for Alzheimer’s disease therapy. Curr Neuropharmacol. 2006;4(2):149–63.

Cummings JL, Doody R, Clark C. Disease-modifying therapies for Alzheimer disease: challenges to early intervention. Neurology. 2007;69(16):1622–34.

Wang DS, Dickson DW, Malter JS. Beta-Amyloid degradation and Alzheimer’s disease. J Biomed Biotechnol. 2006;2006(3):58406.

Deane R, Bell RD, Sagare A, Zlokovic B V. Clearance of amyloid-beta peptide across the blood-brain barrier: implication for therapies in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2009;8(1):16–30.

Siegel, G.S., Chauhan, N., Karczmar AG. Links between Amyloid and Tau Biology in Alzheimer’s Disease and their Cholinergic Aspects; in: Karczmar AG (ed): Exploring the Vertebrate Central Cholinergic Nervous System. New York, Springer, 2007, pp. 597–603.

Fuentes G P, Slachevsky Ch A. Enfermedad de Alzheimer: Actualización en terapia farmacológica. Rev Med Chil. 2005;133(2):224–30.

Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, Gerstein H, Yu G-Q, Mucke L. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science. 2007;316(5825):750–4.

Kuret J, Congdon EE, Li G, Yin H, Yu X, Zhong Q. Evaluating triggers and enhancers of tau fibrillization. Microsc Res Tech. 2005;67(3-4):141–55.

Rafii MS, Aisen PS. Recent developments in Alzheimer’s disease therapeutics. BMC Med. DOI: 10.1186/1741-7015-7-7.

Stoothoff WH, Johnson GVW. Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta. 2005;1739(2-3):280–97.

De Strooper B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol Rev. 2010;90(2):465–94.

Parnetti L, Mignini F, Tomassoni D, Traini E, Amenta F. Cholinergic precursors in the treatment of cognitive impairment of vascular origin: ineffective approaches or need for re-evaluation? J Neurol Sci. 2007;257(1-2):264–9.

Wilson D, Peters R, Ritchie K, Ritchie CW. Latest Advances on Interventions that May Prevent, Delay or Ameliorate Dementia. Ther Adv Chronic Dis. 2011;2(3):161–73.

Roberson ED, Mucke L. 100 years and counting: prospects for defeating Alzheimer’s disease. Science. 2006;314(5800):781–4.

Sabbagh MN. Drug development for Alzheimer’s disease: where are we now and where are we headed? Am J Geriatr Pharmacother. 2009;7(3):167–85.

Schneider LS, Dagerman KS, Higgins JPT, McShane R. Lack of evidence for the efficacy of memantine in mild Alzheimer disease. Arch Neurol. 2011;68(8):991–8.

Álvarez-Sabín J, Román GC. Citicoline in vascular cognitive impairment and vascular dementia after stroke. Stroke. 2011;42(SUPPL. 1) S40-3.

Bishara D, Sauer J, Taylor D. The pharmacological management of Alzheimer’s disease. Prog Neurol Psychiatry. 2015;19(4):9–16.

Fisher A. M1 muscarinic agonists target major hallmarks of Alzheimer’s disease-an update. Curr Alzheimer Res. 2007;4(5):577–80.

Wess J, Eglen RM, Gautam D. Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nat Rev Drug Discov. 2007;6(9):721–33.

Fisher A. Cholinergic treatments with emphasis on M1 muscarinic agonists as potential disease-modifying agents for Alzheimer’s disease. Neurotherapeutics. 2008;5(3):433–42.

Knudsen GM. Assessment of neuroreceptor changes in healthy ageing and in Alzheimer’s disease with emission tomography. Int Congr Ser. 2003;1252:299–308.

Jones CK, Brady AE, Davis AA, Xiang Z, Bubser M, Tantawy MN, Kane AS, Bridges TM, Kennedy JP, Bradley SR, Peterson TE, Ansari MS, Baldwin RM, Kessler RM, Deutch AY, Lah JJ, Levey AI, Lindsley CW, Conn PJ. Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J Neurosci. 2008;28(41):10422–33.

Potter PE. Investigational medications for treatment of patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 Suppl 8):S27–36.

Fisher A. M1 muscarinic agonists target major hallmarks of Alzheimer’s disease - The pivotal role of brain M1 receptors. Neurodegener Dis. 2008; 5(3-4):237-40.

Fisher A, Medeiros R, Barner N, Natan N, Brandeis R, Elkon H, Nahum V, Grigoryan G, Segal M, LaFerla F. M1 Muscarinic Agonists and a Multipotent Activator of Sigma1/M1 Muscarinic Receptors: Future Therapeutics of Alzheimer’s Disease. Alzheimer’s Dement. DOI: 10.1016/j.alz.2014.04.039

Thathiah A, De Strooper B. The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nat Rev Neurosci. 2011;12(2):73–87.

Digby GJ, Shirey JK, Conn PJ. Allosteric activators of muscarinic receptors as novel approaches for treatment of CNS disorders. Mol Biosyst. 2010;6(8):1345–54.

Shen J, Wu J. Nicotinic Cholinergic Mechanisms in Alzheimer’s Disease. Int Rev Neurobiol. 2015;124:275–92.

Shimohama S. Nicotinic receptor-mediated neuroprotection in neurodegenerative disease models. Biol Pharm Bull. 2009;32(3):332–6.

Serra G, Koukopoulos A, De Chiara L, Koukopoulos AE, Tondo L, Girardi P, Baldessarini RJ, Serra G. Three-year, naturalistic, mirror-image assessment of adding memantine to the treatment of 30 treatment-resistant patients with bipolar disorder. J Clin Psychiatry. 2015;76(1):e91–7.

Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595–608.

Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513–33.

Caccamo A, Oddo S, Billings LM, Green KN, Martinez-Coria H, Fisher A, LaFerla FM. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron. 2006;49(5):671–82.

Standridge JB. Pharmacotherapeutic approaches to the treatment of Alzheimer’s disease. Clin Ther. 2004;26(5):615–30.

Jacobsen JS, Reinhart P, Pangalos MN. Current concepts in therapeutic strategies targeting cognitive decline and disease modification in Alzheimer’s disease. NeuroRx. 2005;2(4):612–26.

Burke A, Hall GR, Yaari R, Fleisher A, Dougherty J, Young J, Brand H, Tariot P. Pharmacological treatment of cognitive decline in Alzheimer’s Disease; in: Hajba, L (eds): Pocket Reference to Alzheimer’s Disease Management. London, Springer Healthcare Ltd., 2015, pp. 33–7.

Kim Y-H, Lee Y, Kim D, Jung MW, Lee C-J. Scopolamine-induced learning impairment reversed by physostigmine in zebrafish. Neurosci Res. 2010;67(2):156–61.

Pepeu G, Giovannini MG. Cholinesterase inhibitors and beyond. Curr Alzheimer Res. 2009;6(2):86–96.

Zouridakis M, Zisimopoulou P, Poulas K, Tzartos SJ. Recent advances in understanding the structure of nicotinic acetylcholine receptors. IUBMB Life. 2009;61(4):407–23.

Holtzman DM, Morris JC, Goate AM. Alzheimer’s disease: the challenge of the second century. Sci Transl Med. DOI:10.1126/scitranslmed.3002369

Soares H, Raha N, Sikpi M, Liston D, Brodney M, Coffman K, Tate B, Qiu R, Wang EQ, Li X, Hidi R, Banerjee S, Jhee S, Ereshefsky L, Fullerton T. Aβ> variability and effect of gamma secretase inhibition on cerebrospinal fluid levels of Aβ in healthy volunteers. Alzheimer’s Dement. 2009;5(4):252–3.

Pollack SJ, Lewis H. Secretase inhibitors for Alzheimer’s disease: challenges of a promiscuous protease. Curr Opin Investig Drugs. 2005;6(1):35–47.

Tomita T. Secretase inhibitors and modulators for Alzheimer’s disease treatment. Expert Rev Neurother. 2009;9(5):661–79.

Lundkvist J, Näslund J. Gamma-secretase: a complex target for Alzheimer’s disease. Curr Opin Pharmacol. 2007;7(1):112–8.

Churcher I, Beher D. Gamma-secretase as a therapeutic target for the treatment of Alzheimer’s disease. Curr Pharm Des. 2005;11(26):3363–82.

Martone RL, Zhou H, Atchison K, Comery T, Xu JZ, Huang X, Gong X, Jin M, Kreft A, Harrison B, Mayer SC, Aschmies S, Gonzales C, Zaleska MM, Riddell DR, Wagner E, Lu P, Sun S-C, Sonnenberg-Reines J, Oganesian A, Adkins K, Leach MW, Clarke DW, Huryn D, Abou-Gharbia M, Magolda R, Bard J, Frick G, Raje S, Forlow SB, Balliet C, Burczynski ME, Reinhart PH, Wan HI, Pangalos MN, Jacobsen JS. Begacestat (GSI-953): a novel, selective thiophene sulfonamide inhibitor of amyloid precursor protein gamma-secretase for the treatment of Alzheimer’s disease. J Pharmacol Exp Ther. 2009;331(2):598–608.

Wolfe MS. Inhibition and modulation of gamma-secretase for Alzheimer’s disease. Neurotherapeutics. 2008;5(3):391–8.

He G, Luo W, Li P, Remmers C, Netzer WJ, Hendrick J, Bettayeb K, Flajolet M, Gorelick F, Wennogle LP, Greengard P. Gamma-secretase activating protein is a therapeutic target for Alzheimer’s disease. Nature. 2010;467(7311):95–8.

Serneels L, Van Biervliet J, Craessaerts K, Dejaegere T, Horré K, Van Houtvin T, Esselmann H, Paul S, Schäfer MK, Berezovska O, Hyman BT, Sprangers B, Sciot R, Moons L, Jucker M, Yang Z, May PC, Karran E, Wiltfang J, D’Hooge R, De Strooper B. gamma-Secretase heterogeneity in the Aph1 subunit: relevance for Alzheimer’s disease. Science. 2009;324(5927):639–42.

Baumann S, Höttecke N, Schubenel R, Baumann K, Schmidt B. NSAID-derived gamma-secretase modulators. Part III: Membrane anchoring. Bioorg Med Chem Lett. 2009;19(24):6986–90.

Sastre M, Gentleman SM. NSAIDs: How they Work and their Prospects as Therapeutics in Alzheimer’s Disease. Front Aging Neurosci. 2010;2(20):1–6.

Zotova E, Nicoll JA, Kalaria R, Holmes C, Boche D. Inflammation in Alzheimer’s disease: relevance to pathogenesis and therapy. Alzheimers Res Ther. 2010;2(1):1–9.

Citron M. Beta-secretase inhibition for the treatment of Alzheimer’s disease--promise and challenge. Trends Pharmacol Sci. 2004;25(2):92–7.

Lindsley SR, Moore KP, Rajapakse HA, Selnick HG, Young MB, Zhu H, Munshi S, Kuo L, McGaughey GB, Colussi D, Crouthamel M-C, Lai M-T, Pietrak B, Price EA, Sankaranarayanan S, Simon AJ, Seabrook GR, Hazuda DJ, Pudvah NT, Hochman JH, Graham SL, Vacca JP, Nantermet PG. Design, synthesis, and SAR of macrocyclic tertiary carbinamine BACE-1 inhibitors. Bioorg Med Chem Lett. 2007;17(14):4057–61.

Ghosh AK, Gemma S, Tang J. beta-Secretase as a therapeutic target for Alzheimer’s disease. Neurotherapeutics. 2008;5(3):399–408.

Dominguez D, Tournoy J, Hartmann D, Huth T, Cryns K, Deforce S, Serneels L, Camacho IE, Marjaux E, Craessaerts K, Roebroek AJM, Schwake M, D’Hooge R, Bach P, Kalinke U, Moechars D, Alzheimer C, Reiss K, Saftig P, De Strooper B. Phenotypic and biochemical analyses of BACE1- and BACE2-deficient mice. J Biol Chem. 2005;280(35):30797–806.

Laird FM, Cai H, Savonenko A V, Farah MH, He K, Melnikova T, Wen H, Chiang H-C, Xu G, Koliatsos VE, Borchelt DR, Price DL, Lee H-K, Wong PC. BACE1, a major determinant of selective vulnerability of the brain to amyloid-beta amyloidogenesis, is essential for cognitive, emotional, and synaptic functions. J Neurosci. 2005;25(50):11693–709.

Savonenko A V, Melnikova T, Laird FM, Stewart K-A, Price DL, Wong PC. Alteration of BACE1-dependent NRG1/ErbB4 signaling and schizophrenia-like phenotypes in BACE1-null mice. Proc Natl Acad Sci U S A. 2008;105(14):5585–90.

Hu X, Hicks CW, He W, Wong P, Macklin WB, Trapp BD, Yan R. Bace1 modulates myelination in the central and peripheral nervous system. Nat Neurosci. 2006;9(12):1520–5.

Silvestri R. Boom in the development of non-peptidic beta-secretase (BACE1) inhibitors for the treatment of Alzheimer’s disease. Med Res Rev. 2009;29(2):295–338.

Seabrook GR, Ray WJ, Shearman M, Hutton M. Beyond amyloid: the next generation of Alzheimer’s disease therapeutics. Mol Interv. 2007;7(5):261–70.

Evin G. Future Therapeutics in Alzheimer’s Disease: Development Status of BACE Inhibitors. BioDrugs. 2016;30(3):173–94.

Eketjäll S, Janson J, Kaspersson K, Bogstedt A, Jeppsson F, Fälting J, Haeberlein SB, Kugler AR, Alexander RC, Cebers G. AZD3293: A Novel, Orally Active BACE1 Inhibitor with High Potency and Permeability and Markedly Slow Off-Rate Kinetics. J Alzheimers Dis. 2016;50(4):1109–23.

Blazer LL, Neubig RR. Small molecule protein-protein interaction inhibitors as CNS therapeutic agents: current progress and future hurdles. Neuropsychopharmacology. 2009;34(1):126–41.

Gervais F, Paquette J, Morissette C, Krzywkowski P, Yu M, Azzi M, Lacombe D, Kong X, Aman A, Laurin J, Szarek WA, Tremblay P. Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol Aging. 2007;28(4):537–47.

Kooistra J, Milojevic J, Melacini G, Ortega J. A new function of human HtrA2 as an amyloid-beta oligomerization inhibitor. J Alzheimers Dis. 2009;17(2):281–94.

Zhao W, Wang J, Ho L, Ono K, Teplow DB, Pasinetti GM. Identification of antihypertensive drugs which inhibit amyloid-beta protein oligomerization. J Alzheimers Dis. 2009;16(1):49–57.

Citron M. Alzheimer’s disease: strategies for disease modification. Nat Rev Drug Discov. 2010;9(5):387–98.

Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, Masters CL, Targum S, Bush AI, Murdoch R, Wilson J, Ritchie CW. Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol. 2008;7(9):779–86.

McGowan E, Eriksen J, Hutton M. A decade of modeling Alzheimer’s disease in transgenic mice. Trends Genet. 2006 May;22(5):281–9.

Dickey CA, Kamal A, Lundgren K, Klosak N, Bailey RM, Dunmore J, Ash P, Shoraka S, Zlatkovic J, Eckman CB, Patterson C, Dickson DW, Nahman NS, Hutton M, Burrows F, Petrucelli L. The high-affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins. J Clin Invest. 2007;117(3):648–58.

Pickhardt M, Larbig G, Khlistunova I, Coksezen A, Meyer B, Mandelkow E-M, Schmidt B, Mandelkow E. Phenylthiazolyl-hydrazide and its derivatives are potent inhibitors of tau aggregation and toxicity in vitro and in cells. Biochemistry. 2007;46(35):10016–23.

Le Corre S, Klafki HW, Plesnila N, Hübinger G, Obermeier A, Sahagún H, Monse B, Seneci P, Lewis J, Eriksen J, Zehr C, Yue M, McGowan E, Dickson DW, Hutton M, Roder HM. An inhibitor of tau hyperphosphorylation prevents severe motor impairments in tau transgenic mice. Proc Natl Acad Sci U S A. 2006;103(25):9673–8.

Gong C-X, Liu F, Grundke-Iqbal I, Iqbal K. Dysregulation of protein phosphorylation/dephosphorylation in Alzheimer’s disease: a therapeutic target. J Biomed Biotechnol. DOI: 10.1155/JBB/2006/31825.

Funderburk SF, Marcellino BK, Yue Z. Cell “self-eating” (autophagy) mechanism in Alzheimer’s disease. Mt Sinai J Med. 77(1):59–68.

Mendes CT, Mury FB, de Sá Moreira E, Alberto FL, Forlenza OV, Dias-Neto E, Gattaz WF. Lithium reduces Gsk3b mRNA levels: implications for Alzheimer Disease. Eur Arch Psychiatry Clin Neurosci. 2009;259(1):16–22.

Rinne JO, Brooks DJ, Rossor MN, Fox NC, Bullock R, Klunk WE, Mathis CA, Blennow K, Barakos J, Okello AA, Rodriguez Martinez de Liano S, Liu E, Koller M, Gregg KM, Schenk D, Black R, Grundman M. 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol. 2010;9(4):363–72.

Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, Eisner L, Kirby L, Rovira MB, Forette F, Orgogozo J-M. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64(9):1553–62.

Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A, Jones RW, Bullock R, Love S, Neal JW, Zotova E, Nicoll JAR. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet (London, England). 2008;372(9634):216–23.

Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, Raskind M, Sabbagh M, Honig LS, Doody R, van Dyck CH, Mulnard R, Barakos J, Gregg KM, Liu E, Lieberburg I, Schenk D, Black R, Grundman M. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2009;73(24):2061–70.

Morgan D. Immunotherapy for Alzheimer’s disease. J Intern Med. 2011;269(1):54–63.

Falkentoft AC, Hasselbalch SG. Immunotherapy for Alzheimer’s disease. Ugeskr Laeger. 2016;178(3):2–6.

Schroeder SK, Joly-Amado A, Gordon MN, Morgan D. Tau-Directed Immunotherapy: A Promising Strategy for Treating Alzheimer’s Disease and Other Tauopathies. J Neuroimmune Pharmacol. 2016;11(1):9–25.

Sparks DL, Kryscio RJ, Connor DJ, Sabbagh MN, Sparks LM, Lin Y, Liebsack C. Cholesterol and cognitive performance in normal controls and the influence of elective statin use after conversion to mild cognitive impairment: results in a clinical trial cohort. Neurodegener Dis. 2010;7(1-3):183–6.

Feldman HH, Doody RS, Kivipelto M, Sparks DL, Waters DD, Jones RW, Schwam E, Schindler R, Hey-Hadavi J, DeMicco DA, Breazna A. Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease: LEADe. Neurology. 2010;74(12):956–64.

McGuinness B, O’Hare J, Craig D, Bullock R, Malouf R, Passmore P. Statins for the treatment of dementia. Cochrane database Syst Rev. 2010;(8):CD007514.

Kosicek M, Malnar M, Goate A, Hecimovic S. Cholesterol accumulation in Niemann Pick type C (NPC) model cells causes a shift in APP localization to lipid rafts. Biochem Biophys Res Commun. 2010;393(3):404–9.

Morris G, Walder K, Puri BK, Berk M, Maes M. The Deleterious Effects of Oxidative and Nitrosative Stress on Palmitoylation, Membrane Lipid Rafts and Lipid-Based Cellular Signalling: New Drug Targets in Neuroimmune Disorders. Mol Neurobiol. DOI: 10.1007/s12035-015-9392-y

Zhou X, Yang C, Liu Y, Li P, Yang H, Dai J, Qu R, Yuan L. Lipid rafts participate in aberrant degradative autophagic-lysosomal pathway of amyloid-beta peptide in Alzheimer’s disease. Neural Regen Res. 2014;9(1):92–100.

Roychaudhuri R, Yang M, Hoshi MM, Teplow DB. Amyloid beta-protein assembly and Alzheimer disease. J Biol Chem. 2009;284(8):4749–53.

Guardia-Laguarta C, Coma M, Pera M, Clarimón J, Sereno L, Agulló JM, Molina-Porcel L, Gallardo E, Deng A, Berezovska O, Hyman BT, Blesa R, Gómez-Isla T, Lleó A. Mild cholesterol depletion reduces amyloid-beta production by impairing APP trafficking to the cell surface. J Neurochem. 2009;110(1):220–30.

Rushworth J V, Hooper NM. Lipid Rafts: Linking Alzheimer’s Amyloid-β Production, Aggregation, and Toxicity at Neuronal Membranes. Int J Alzheimers Dis. DOI: 10.4061/2011/603052.




DOI: https://doi.org/10.25009/eb.v7i15.2577

Enlaces refback

  • No hay ningún enlace refback.


 

eNeurobiología es una publicación cuatrimestral editada por el Instituto de Investigaciones Cerebrales de la Universidad Veracruzana. Estamos ubicados en Av. Dr. Luis Castelazo Ayala, s/n, colonia Industrial Ánimas, C.P. 91190, Xalapa-Enríquez, Veracruz, México. Teléfono: 8418900 ext. 13062, www.iice.uv.mx; eneurobiologia@uv.mx. Reserva de Derechos al Uso Exclusivo 04-2023-061314100600-102, otorgada por el Instituto Nacional de Derechos de Autor. ISSN: 2007-3054. Esta obra está bajo una Licencia Creative Commons Attribution 4.0 International.