Generic placeholder image

Current Alzheimer Research

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Research Article

APOE Genotypes and Brain Imaging Classes in Normal Cognition, Mild Cognitive Impairment, and Alzheimer’s Disease: A Longitudinal Study

Author(s): Cecilia Camarda*, Paola Torelli , Carmela Pipia, Gianluca Sottile*, Giovanna Cilluffo and Rosolino Camarda

Volume 17, Issue 8, 2020

Page: [766 - 780] Pages: 15

DOI: 10.2174/1567205017666201109093314

Price: $65

Abstract

Objective: To evaluate in 419 stroke-free cognitively normal subjects (CN) aged 45-82 years covering during a long prospective study (11.54 ± 1.47 years) the preclinical to dementia spectrum: 1) the distribution of small vessel disease (V) and brain atrophy (A) aggregated as following: V−/A−, V−/A+, V+/A−, V+/A+; 2) the relationship of these imaging classes with individual apolipoprotein E (APOE) genotypes; 3) the risk of progression to Alzheimer Disease (AD) of the individual APOE genotypes.

Methods: Participants underwent one baseline (t0), and 4 clinical and neuropsychological assessments (t1,t2,t3, and t4). Brain MRI was performed in all subjects at t0, t2, t3 and t4.. White matter hyperintensities were assessed through two visual rating scales. Lacunes were also rated. Subcortical and global brain atrophy were determined through the bicaudate ratio and the lateral ventricle to brain ratio, respectively. APOE genotypes were determined at t0 in all subjects. Cox proportional hazard model was used to evaluate the risk of progression to AD.

Results: The imaging class of mixed type was very common in AD, and in non amnestic mild cognitive impaired APOE ε4 non carriers. In these subjects, frontal and parieto-occipital regions were most affected by small vessel disease.

Conclusion: Our findings suggest that the APOE ε3 allele is probably linked to the brain vascular pathology.

Keywords: APOE genotypes, white matter hyperintensities, lacunes, caudate atrophy, global cerebral atrophy, brain imaging classes.

« Previous
[1]
Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclinical stages of 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): 280-92.
[http://dx.doi.org/10.1016/j.jalz.2011.03.003 ] [PMID: 21514248]
[2]
Petersen RC. Mild cognitive impairment as a diagnostic entity. J Intern Med 2004; 256(3): 183-94.
[http://dx.doi.org/10.1111/j.1365-2796.2004.01388.x ] [PMID: 15324362]
[3]
Jack CR Jr. Brain atrophy on magnetic resonance imaging as a biomarker of neurodegeneration. JAMA Neurol 2016; 73(10): 1179-82.
[http://dx.doi.org/10.1001/jamaneurol.2016.2843 ] [PMID: 27548303]
[4]
McKhann GM, Knopman DS, 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. Alzheimers Dement 2011; 7(3): 263-9.
[http://dx.doi.org/10.1016/j.jalz.2011.03.005 ] [PMID: 21514250]
[5]
Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cognitive impairment 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): 270-9.
[http://dx.doi.org/10.1016/j.jalz.2011.03.008 ] [PMID: 21514249]
[6]
Jack CR Jr, Knopman DS, Jagust WJ, et al. Tracking pathophysiological processes in Alzheimer’s disease: An updated hypothetical model of dynamic biomarkers. Lancet Neurol 2013; 12(2): 207-16.
[http://dx.doi.org/10.1016/S1474-4422(12)70291-0 ] [PMID: 23332364]
[7]
Villemagne VL, Burnham S, Bourgeat P, et al. Australian Imaging Biomarkers and Lifestyle (AIBL) Research Group. Amyloid β deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: A prospective cohort study. Lancet Neurol 2013; 12(4): 357-67.
[http://dx.doi.org/10.1016/S1474-4422(13)70044-9 ] [PMID: 23477989]
[8]
Dubois B, Hampel H, Feldman HH, et al. Proceedings of the Meeting of the International Working Group (IWG) and the American Alzheimer’s Association on “The Preclinical State of AD”; July 23, 2015; Washington DC, USA. Preclinical Alzheimer’s disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 2016; 12(3): 292-323.
[http://dx.doi.org/10.1016/j.jalz.2016.02.002 ] [PMID: 27012484]
[9]
Schilling S, DeStefano AL, Sachdev PS, et al. APOE genotype and MRI markers of cerebrovascular disease: Systematic review and meta-analysis. Neurology 2013; 81(3): 292-300.
[http://dx.doi.org/10.1212/WNL.0b013e31829bfda4 ] [PMID: 23858411]
[10]
Santos CY, Snyder PJ, Wu WC, Zhang M, Echeverria A, Alber J. Pathophysiologic relationship between Alzheimer’s disease, cerebrovascular disease, and cardiovascular risk: A review and synthesis. Alzheimers Dement (Amst) 2017; 7: 69-87.
[http://dx.doi.org/10.1016/j.dadm.2017.01.005 ] [PMID: 28275702]
[11]
Vemuri P, Lesnick TG, Przybelski SA, et al. Vascular and amyloid pathologies are independent predictors of cognitive decline in normal elderly. Brain 2015; 138(Pt 3): 761-71.
[http://dx.doi.org/10.1093/brain/awu393 ] [PMID: 25595145]
[12]
Bennett DA, Schneider JA, Buchman AS, Barnes LL, Boyle PA, Wilson RS. Overview and findings from the rush Memory and Aging Project. Curr Alzheimer Res 2012; 9(6): 646-63.
[http://dx.doi.org/10.2174/156720512801322663 ] [PMID: 22471867]
[13]
Wardlaw JM, Smith EE, Biessels GJ, et al. STandards for ReportIng Vascular changes on nEuroimaging (STRIVE v1). Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol 2013; 12(8): 822-38.
[http://dx.doi.org/10.1016/S1474-4422(13)70124-8 ] [PMID: 23867200]
[14]
Gorelick PB, Scuteri A, Black SE, et al. American Heart Association Stroke Council, Council on Epidemiology and Prevention, Council on Cardiovascular Nursing, Council on Cardiovascular Radiology and Intervention, and Council on Cardiovascular Surgery and Anesthesia. Vascular contributions to cognitive impairment and dementia: A statement for healthcare professionals from the american heart association/american stroke association. Stroke 2011; 42(9): 2672-713.
[http://dx.doi.org/10.1161/STR.0b013e3182299496 ] [PMID: 21778438]
[15]
Pantoni L. Cerebral small vessel disease: From pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol 2010; 9(7): 689-701.
[http://dx.doi.org/10.1016/S1474-4422(10)70104-6 ] [PMID: 20610345]
[16]
Reijmer YD, Freeze WM, Leemans A, Biessels GJ. Utrecht Vascular Cognitive Impairment Study Group. The effect of lacunar infarcts on white matter tract integrity. Stroke 2013; 44(7): 2019-21.
[http://dx.doi.org/10.1161/STROKEAHA.113.001321 ] [PMID: 23686971]
[17]
Lambert C, Sam Narean J, Benjamin P, Zeestraten E, Barrick TR, Markus HS. Characterising the grey matter correlates of leukoaraiosis in cerebral small vessel disease. Neuroimage Clin 2015; 9: 194-205.
[http://dx.doi.org/10.1016/j.nicl.2015.07.002 ] [PMID: 26448913]
[18]
Raz N, Lindenberger U, Rodrigue KM, et al. Regional brain changes in aging healthy adults: general trends, individual differences and modifiers. Cereb Cortex 2005; 15(11): 1676-89.
[http://dx.doi.org/10.1093/cercor/bhi044 ] [PMID: 15703252]
[19]
Habes M, Erus G, Toledo JB, et al. White matter hyperintensities and imaging patterns of brain ageing in the general population. Brain 2016; 139(Pt 4): 1164-79.
[http://dx.doi.org/10.1093/brain/aww008 ] [PMID: 26912649]
[20]
Rizvi B, Narkhede A, Last BS, Budge M, Tosto G, Manly JJ, et al. The effect of white matter hyperintensities on cognition is mediated by cortical atrophy. Neurobiol Aging 2018; 64: 25-32.
[http://dx.doi.org/10.1016/j.neurobiolaging.2017.12.006]
[21]
Gunning-Dixon FM, Raz N. The cognitive correlates of white matter abnormalities in normal aging: a quantitative review. Neuropsychology 2000; 14(2): 224-32.
[http://dx.doi.org/10.1037/0894-4105.14.2.224 ] [PMID: 10791862]
[22]
Prins ND, van Dijk EJ, den Heijer T, et al. Cerebral small-vessel disease and decline in information processing speed, executive function and memory. Brain 2005; 128(Pt 9): 2034-41.
[http://dx.doi.org/10.1093/brain/awh553 ] [PMID: 15947059]
[23]
Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 1993; 261(5123): 921-3.
[http://dx.doi.org/10.1126/science.8346443 ] [PMID: 8346443]
[24]
Corder EH, Saunders AM, Risch NJ, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet 1994; 7(2): 180-4.
[http://dx.doi.org/10.1038/ng0694-180 ] [PMID: 7920638]
[25]
Sudlow C, Martínez González NA, Kim J, Clark C. Does apolipoprotein E genotype influence the risk of ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage? Systematic review and meta-analyses of 31 studies among 5961 cases and 17,965 controls. Stroke 2006; 37(2): 364-70.
[http://dx.doi.org/10.1161/01.STR.0000199065.12908.62 ] [PMID: 16385096]
[26]
Montagne A, Nation DA, Sagare AP, et al. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 2020; 581(7806): 71-6.
[http://dx.doi.org/10.1038/s41586-020-2247-3 ] [PMID: 32376954]
[27]
Morgen K, Schneider M, Frölich L, et al. Apolipoprotein E-dependent load of white matter hyperintensities in Alzheimer’s disease: A voxel-based lesion mapping study. Alzheimers Res Ther 2015; 7(1): 27.
[http://dx.doi.org/10.1186/s13195-015-0111-8 ] [PMID: 25984242]
[28]
Morgen K, Frölich L, Tost H, et al. APOE-dependent phenotypes in subjects with mild cognitive impairment converting to Alzheimer’s disease. J Alzheimers Dis 2013; 37(2): 389-401.
[http://dx.doi.org/10.3233/JAD-130326 ] [PMID: 23948881]
[29]
Groot C, Sudre CH, Barkhof F, et al. Clinical phenotype, atrophy, and small vessel disease in APOEε2 carriers with Alzheimer disease. Neurology 2018; 91(20): e1851-9.
[http://dx.doi.org/10.1212/WNL.0000000000006503 ] [PMID: 30341156]
[30]
Camarda C, Torelli P, Camarda R, Battaglini I, Gagliardo C, Monastero R. Isolated, subtle, neurological abnormalities in neurologically and cognitively healthy aging subjects. J Neurol 2015; 262(5): 1328-39.
[http://dx.doi.org/10.1007/s00415-015-7716-5 ] [PMID: 25825125]
[31]
Camarda C, Pipia C, Azzarello D, et al. Vascular risk factors, vascular diseases, and imaging findings in a hospital-based cohort of mild cognitive impairment types. Curr Alzheimer Res 2018; 15(7): 679-90.
[http://dx.doi.org/10.2174/1567205015666180119110712 ] [PMID: 29357798]
[32]
Hixson JE, Vernier DT. Restriction isotyping of human apolipoprotein E by gene amplification and cleavage with HhaI. J Lipid Res 1990; 31(3): 545-8.
[PMID: 2341813]
[33]
Katz S, Ford AB, Moskowitz RW, Jackson BA, Jaffe MW. The index of ADL: A standardized measure of biological and psychosocial function. JAMA 1963; 185(12): 914-9.
[http://dx.doi.org/10.1001/jama.1963.03060120024016 ] [PMID: 14044222]
[34]
Lawton MP, Brody EM. Assessment of older people: Self-maintaining and instrumental activities of daily living. Gerontologist 1969; 9(3): 179-86.
[http://dx.doi.org/10.1093/geront/9.3_Part_1.179 ] [PMID: 5349366]
[35]
Hachinski VC, Iliff LD, Zilhka E, et al. Cerebral blood flow in dementia. Arch Neurol 1975; 32(9): 632-7.
[http://dx.doi.org/10.1001/archneur.1975.00490510088009 ] [PMID: 1164215]
[36]
Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975; 12(3): 189-98.
[http://dx.doi.org/10.1016/0022-3956(75)90026-6 ] [PMID: 1202204]
[37]
Morris JC. The Clinical Dementia Rating (CDR): Current version and scoring rules. Neurology 1993; 43(11): 2412-4.
[http://dx.doi.org/10.1212/WNL.43.11.2412-a ] [PMID: 8232972]
[38]
Wahlund LO, Barkhof F, Fazekas F, et al. European task force on age-related white matter changes. A new rating scale for age-related white matter changes applicable to MRI and CT. Stroke 2001; 32(6): 1318-22.
[http://dx.doi.org/10.1161/01.STR.32.6.1318 ] [PMID: 11387493]
[39]
Fazekas F, Chawluk JB, Alavi A, Hurtig HI, Zimmerman RA. MR signal abnormalities at 1.5 T in Alzheimer’s dementia and normal aging. AJR Am J Roentgenol 1987; 149(2): 351-6.
[http://dx.doi.org/10.2214/ajr.149.2.351 ] [PMID: 3496763]
[40]
Jessen F, Amariglio RE, van Boxtel M, et al. Subjective Cognitive Decline Initiative (SCD-I) Working Group. A conceptual framework for research on subjective cognitive decline in preclinical Alzheimer’s disease. Alzheimers Dement 2014; 10(6): 844-52.
[http://dx.doi.org/10.1016/j.jalz.2014.01.001 ] [PMID: 24798886]
[41]
Erkinjuntti T, Inzitari D, Pantoni L, Wallin A, Scheltens P, Rockwood K, et al. Research criteria for subcortical vascular dementia in clinical trials. J Neural Transm Suppl 2000; 59: 23-30.
[42]
Wallin A, Román GC, Esiri M, et al. Update on vascular cognitive impairment associated with subcortical small-vessel disease. J Alzheimers Dis 2018; 62(3): 1417-41.
[http://dx.doi.org/10.3233/JAD-170803 ] [PMID: 29562536]
[43]
McKeith IG, Galasko D, Kosaka K, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): Report of the consortium on DLB international workshop. Neurology 1996; 47(5): 1113-24.
[http://dx.doi.org/10.1212/WNL.47.5.1113 ] [PMID: 8909416]
[44]
Brun A, Englund B, Gustafson L, et al. The Lund and Manchester Groups. Clinical and neuropathological criteria for frontotemporal dementia. J Neurol Neurosurg Psychiatry 1994; 57(4): 416-8.
[http://dx.doi.org/10.1136/jnnp.57.4.416 ] [PMID: 8163988]
[45]
Therneau TM, Grambsch PM. Modeling survival data extending the cox model. New York: Springer-Verlag 2000.
[http://dx.doi.org/10.1007/978-1-4757-3294-8]
[46]
Grambsch P, Therneau T. Proportional hazards tests and diagnostics based on weighted residuals. Biometrika 1994; 81(3): 515-26.
[http://dx.doi.org/10.1093/biomet/81.3.515]
[47]
Schipper HM, Apolipoprotein E. Implications for AD neurobiology, epidemiology and risk assessment. Neurobiol Aging 2011; 32(5): 778-90.
[http://dx.doi.org/10.1016/j.neurobiolaging.2009.04.021 ] [PMID: 19482376]
[48]
Liu L, Caselli RJ. Age stratification corrects bias in estimated hazard of APOE genotype for Alzheimer’s disease. Alzheimers Dement (N Y) 2018; 4: 602-8.
[http://dx.doi.org/10.1016/j.trci.2018.09.006 ] [PMID: 30450407]
[49]
Bilgel M, An Y, Zhou Y, et al. Individual estimates of age at detectable amyloid onset for risk factor assessment. Alzheimers Dement 2016; 12(4): 373-9.
[http://dx.doi.org/10.1016/j.jalz.2015.08.166 ] [PMID: 26588863]
[50]
Liu CC, Liu CC, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat Rev Neurol 2013; 9(2): 106-18.
[http://dx.doi.org/10.1038/nrneurol.2012.263 ] [PMID: 23296339]
[51]
Ba M, Kong M, Li X, Ng KP, Rosa-Neto P, Gauthier S. Is ApoE ɛ 4 a good biomarker for amyloid pathology in late onset Alzheimer’s disease? Transl Neurodegener 2016; 5: 20-4.
[http://dx.doi.org/10.1186/s40035-016-0067-z ] [PMID: 27891223]
[52]
Ossenkoppele R, Jansen WJ, Rabinovici GD, et al. Amyloid PET Study Group. Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. JAMA 2015; 313(19): 1939-49.
[http://dx.doi.org/10.1001/jama.2015.4669 ] [PMID: 25988463]
[53]
Grothe MJ, Villeneuve S, Dyrba M, Bartrés-Faz D, Wirth M. Alzheimer’s Disease Neuroimaging Initiative. Multimodal characterization of older APOE2 carriers reveals selective reduction of amyloid load. Neurology 2017; 88(6): 569-76.
[http://dx.doi.org/10.1212/WNL.0000000000003585 ] [PMID: 28062720]
[54]
Nelson PT, Pious NM, Jicha GA, et al. APOE- and APOE- correlate with increased amyloid accumulation in cerebral vasculature. J Neuropathol Exp Neurol 2013; 72(7): 708-15.
[http://dx.doi.org/10.1097/NEN.0b013e31829a25b9 ] [PMID: 23771217]
[55]
Greenberg SM, Bacskai BJ, Hernandez-Guillamon M, Pruzin J, Sperling R, van Veluw SJ. Cerebral amyloid angiopathy and Alzheimer disease - one peptide, two pathways. Nat Rev Neurol 2020; 16(1): 30-42.
[http://dx.doi.org/10.1038/s41582-019-0281-2 ] [PMID: 31827267]
[56]
Nicoll JA, Burnett C, Love S, et al. High frequency of apolipoprotein E epsilon 2 allele in hemorrhage due to cerebral amyloid angiopathy. Ann Neurol 1997; 41(6): 716-21.
[http://dx.doi.org/10.1002/ana.410410607 ] [PMID: 9189032]
[57]
Oveisgharan S, Buchman AS, Yu L, Farfel J, Hachinski V, Gaiteri C, et al. APOE genotype, incident AD and MCI, cognitive decline, and AD pathology in older adults. Neurology 2018; 90(24): 2127-34.
[http://dx.doi.org/10.1212/WNL.0000000000005677 ] [PMID: 29752306]
[58]
Wolk DA, Dickerson BC. Alzheimer’s Disease Neuroimaging Initiative. Apolipoprotein E (APOE) genotype has dissociable effects on memory and attentional-executive network function in Alzheimer’s disease. Proc Natl Acad Sci USA 2010; 107(22): 10256-61.
[http://dx.doi.org/10.1073/pnas.1001412107 ] [PMID: 20479234]
[59]
Gouw AA, Seewann A, van der Flier WM, et al. Heterogeneity of small vessel disease: a systematic review of MRI and histopathology correlations. J Neurol Neurosurg Psychiatry 2011; 82(2): 126-35.
[http://dx.doi.org/10.1136/jnnp.2009.204685 ] [PMID: 20935330]
[60]
Prins ND, Scheltens P. White matter hyperintensities, cognitive impairment and dementia: An update. Nat Rev Neurol 2015; 11(3): 157-65.
[http://dx.doi.org/10.1038/nrneurol.2015.10 ] [PMID: 25686760]
[61]
Wardlaw JM, Valdés Hernández MC, Muñoz-Maniega S. What are white matter hyperintensities made of? Relevance to vascular cognitive impairment. J Am Heart Assoc 2015; 4(6)
[http://dx.doi.org/10.1161/JAHA.114.001140 ] [PMID: 26104658]
[62]
Kapeller P, Barber R, Vermeulen RJ, et al. European Task Force of Age Related White Matter Changes. Visual rating of age-related white matter changes on magnetic resonance imaging: scale comparison, interrater agreement, and correlations with quantitative measurements. Stroke 2003; 34(2): 441-5.
[http://dx.doi.org/10.1161/01.STR.0000049766.26453.E9 ] [PMID: 12574557]
[63]
Gao FQ, Swartz RH, Scheltens P, et al. Complexity of MRI white matter hyperintensity assessments in relation to cognition in aging and dementia from the Sunnybrook Dementia Study. J Alzheimers Dis 2011; 26(Suppl. 3): 379-88.
[http://dx.doi.org/10.3233/JAD-2011-0058 ] [PMID: 21971477]
[64]
Yates PA, Desmond PM, Phal PM, et al. AIBL Research Group. Incidence of cerebral microbleeds in preclinical Alzheimer disease. Neurology 2014; 82(14): 1266-73.
[http://dx.doi.org/10.1212/WNL.0000000000000285 ] [PMID: 24623839]
[65]
Zhu YC, Dufouil C, Soumaré A, Mazoyer B, Chabriat H, Tzourio C. High degree of dilated Virchow-Robin spaces on MRI is associated with increased risk of dementia. J Alzheimers Dis 2010; 22(2): 663-72.
[http://dx.doi.org/10.3233/JAD-2010-100378 ] [PMID: 20847444]
[66]
Passiak BS, Liu D, Kresge HA, et al. Perivascular spaces contribute to cognition beyond other small vessel disease markers. Neurology 2019; 92(12): e1309-21.
[http://dx.doi.org/10.1212/WNL.0000000000007124 ] [PMID: 30814324]
[67]
Doraiswamy PM, Patterson L, Na C, et al. Bicaudate index on magnetic resonance imaging: effects of normal aging. J Geriatr Psychiatry Neurol 1994; 7(1): 13-7.
[http://dx.doi.org/10.1177/089198879400700103 ] [PMID: 8192824]
[68]
Nestor SM, Rupsingh R, Borrie M, et al. Alzheimer’s Disease Neuroimaging Initiative. Ventricular enlargement as a possible measure of Alzheimer’s disease progression validated using the Alzheimer’s disease neuroimaging initiative database. Brain 2008; 131(Pt 9): 2443-54.
[http://dx.doi.org/10.1093/brain/awn146 ] [PMID: 18669512]
[69]
Jack CR Jr, Bennett DA, Blennow K, et al. Contributors. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14(4): 535-62.
[http://dx.doi.org/10.1016/j.jalz.2018.02.018 ] [PMID: 29653606]
[70]
Sweeney MD, Montagne A, Sagare AP, et al. Vascular dysfunction-The disregarded partner of Alzheimer’s disease. Alzheimers Dement 2019; 15(1): 158-67.
[http://dx.doi.org/10.1016/j.jalz.2018.07.222 ] [PMID: 30642436]
[71]
Jagust W, Jack CR Jr, Bennett DA, et al. “Alzheimer’s disease” is neither “Alzheimer’s clinical syndrome” nor “dementia”. Alzheimers Dement 2019; 15(1): 153-7.
[http://dx.doi.org/10.1016/j.jalz.2018.11.002] [PMID: 30642435]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy