Imaging beta-amyloid (Aβ) burden in the brains of middle-aged individuals with alcohol-use disorders: a [11C]PIB PET study

Margaret R Flanigan, Sarah K Royse, David P Cenkner, Katelyn M Kozinski, Clara J Stoughton, Michael L Himes, Davneet S Minhas, Brian Lopresti, Meryl A Butters, Rajesh Narendran, Margaret R Flanigan, Sarah K Royse, David P Cenkner, Katelyn M Kozinski, Clara J Stoughton, Michael L Himes, Davneet S Minhas, Brian Lopresti, Meryl A Butters, Rajesh Narendran

Abstract

No in vivo human studies have examined the prevalence of Alzheimer's disease (AD) neuropathology in individuals with alcohol-use disorder (AUD), although recent research suggests that a relationship between the two exists. Therefore, this study used Pittsburgh Compound-B ([11C]PiB) PET imaging to test the hypothesis that AUD is associated with greater brain amyloid (Aβ) burden in middle-aged adults compared to healthy controls. Twenty healthy participants (14M and 6F) and 19 individuals with AUD (15M and 4F), all aged 40-65 years, underwent clinical assessment, MRI, neurocognitive testing, and positron emission tomography (PET) imaging. Global [11C]PiB standard uptake value ratios (SUVRs), cortical thickness, gray matter volumes (GMVs), and neurocognitive function in subjects with AUD were compared to healthy controls. These measures were selected because they are considered markers of risk for future AD and other types of neurocognitive dysfunction. The results of this study showed no significant differences in % global Aβ positivity or subthreshold Aβ loads between AUD and controls. However, relative to controls, we observed a significant 6.1% lower cortical thickness in both AD-signature regions and in regions not typically associated with AD, lower GMV in the hippocampus, and lower performance on tests of attention as well as immediate and delayed memory in individuals with AUD. This suggest that Aβ accumulation is not greater in middle-aged individuals with AUD. However, other markers of neurodegeneration, such as impaired memory, cortical thinning, and reduced hippocampal GMV, are present. Further studies are needed to elucidate the patterns and temporal staging of AUD-related pathophysiology and cognitive impairment. Imaging β-amyloid in middle age alcoholics as a mechanism that increases their risk for Alzheimer's disease; Registration Number: NCT03746366 .

Conflict of interest statement

The authors declare no competing interest.

References

    1. Piumatti G, Moore S, Berridge D, Sarkar C, Gallacher J. The relationship between alcohol use and long-term cognitive decline in middle and late life: a longitudinal analysis using UK Biobank. J. Public Health. 2018;40:313–314.
    1. Stavro K, Pelletier J, Potvin S. Widespread and sustained cognitive deficits in alcoholism: a meta-analysis. Addict. Biol. 2013;18:203–213.
    1. Schwarzinger M, et al. Contribution of alcohol use disorders to the burden of dementia in France 2008-13: a nationwide retrospective cohort study. Lancet Public Health. 2018;3:e124–e132.
    1. Kim SR, et al. Effects of chronic alcohol consumption on expression levels of APP and Aβ-producing enzymes. BMB Rep. 2011;44:135–139.
    1. Huang D, et al. Ethanol alters APP processing and aggravates Alzheimer-associated phenotypes. Mol. Neurobiol. 2018;55:5006–5018.
    1. Aho L, Karkola K, Juusela J, Alafuzoff I. Heavy alcohol consumption and neuropathological lesions: a post-mortem human study. J. Neurosci. Res. 2009;87:2786–2792.
    1. Huang WJ, Zhang X, Chen WW. Association between alcohol and Alzheimer’s disease. Exp. Ther. Med. 2016;12:1247–1250.
    1. Kim JW, et al. Association of moderate alcohol intake with in vivo amyloid-beta deposition in human brain: a cross-sectional study. PLoS Med. 2020;17:e1003022.
    1. Crowe SF, Cammisuli DM, Stranks EK. Widespread cognitive deficits in alcoholism persistent following prolonged abstinence: an updated meta-analysis of studies that used standardised neuropsychological assessment tools. Arch. Clin. Neuropsychol. 2019;35:31–45.
    1. Dickson DW, et al. Identification of normal and pathological aging in prospectively studied nondemented elderly humans. Neurobiol. Aging. 1992;13:179–189.
    1. Jansen WJ, et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA. 2015;313:1924–1938.
    1. Klunk WE, et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann. Neurol. 2004;55:306–319.
    1. Schwarz CG, et al. A large-scale comparison of cortical thickness and volume methods for measuring Alzheimer’s disease severity. Neuroimage Clin. 2016;11:802–812.
    1. Cohen AD, et al. Classification of amyloid-positivity in controls: comparison of visual read and quantitative approaches. Neuroimage. 2013;71:207–215.
    1. Silverman W, Wisniewski HM, Bobinski M, Wegiel J. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol. Aging. 1997;18:377–379.
    1. McLellan AT, et al. The Fifth Edition of the Addiction Severity Index. J. Subst. Abus. Treat. 1992;9:199–213.
    1. Maisto SA, Sobell LC, Cooper AM, Sobell MB. Comparison of two techniques to obtain retrospective reports of drinking behavior from alcohol abusers. Addict. Behav. 1982;7:33–38.
    1. Skinner H. & Horn J. Alcohol Dependence Scale (ADS) User’s Guide (Addiction Research Foundation, 1984).
    1. Selzer ML. Michigan Alcoholism Screening Test (MAST): preliminary report. Univ. Mich. Med. Cent. J. 1968;34:143–145.
    1. Flannery BA, Volpicelli JR, Pettinati HM. Psychometric properties of the Penn Alcohol Craving Scale. Alcohol Clin. Exp. Res. 1999;23:1289–1295.
    1. Fagerstrom K, Heatherton TF, Koslowski T. Nicotine addiction and its assessment. Ear Nose Throat J. 1991;69:763–765.
    1. Berger L, et al. Ethyl glucuronide in hair and fingernails as a long-term alcohol biomarker. Addiction. 2014;109:425–431.
    1. Corder EH, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–923.
    1. Reiman EM, et al. Fibrillar amyloid-beta burden in cognitively normal people at 3 levels of genetic risk for Alzheimer’s disease. Proc. Natl Acad. Sci. USA. 2009;106:6820–6825.
    1. Rowe CC, et al. Amyloid imaging results from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol. Aging. 2010;31:1275–1283.
    1. Fleisher AS, et al. Apolipoprotein E epsilon4 and age effects on florbetapir positron emission tomography in healthy aging and Alzheimer disease. Neurobiol. Aging. 2013;34:1–12.
    1. Lopresti BJ, et al. Influence of apolipoprotein-E genotype on brain amyloid load and longitudinal trajectories. Neurobiol. Aging. 2020;94:111–120.
    1. Randolph C, Tierney MC, Mohr E, Chase TN. The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS): preliminary clinical validity. J. Clin. Exp. Neuropsychol. 1998;20:310–319.
    1. Sullivan JT, Sykora K, Schneiderman J, Naranjo CA, Sellers EM. Assessment of alcohol withdrawal: the revised clinical institute withdrawal assessment for alcohol scale (CIWA-Ar) Br. J. Addict. 1989;84:1353–1357.
    1. Mathis CA, et al. Synthesis and evaluation of 11C-labeled 6-substituted 2-arylbenzothiazoles as amyloid imaging agents. J. Med. Chem. 2003;46:2740–2754.
    1. Cohen AD, et al. Basal cerebral metabolism may modulate the cognitive effects of Abeta in mild cognitive impairment: an example of brain reserve. J. Neurosci. 2009;29:14770–14778.
    1. McNamee RL, et al. Consideration of optimal time window for Pittsburgh compound B PET summed uptake measurements. J. Nucl. Med. 2009;50:348–355.
    1. Sullivan, K. J. et al. Alzheimer’s disease pathology in a community-based sample of older adults without dementia: The MYHAT neuroimaging study. Brain Imaging Behav. 10.1007/s11682-020-00334-2 (2020).
    1. Fischl B. FreeSurfer. Neuroimage. 2012;62:774–781.
    1. Tziortzi AC, et al. Imaging dopamine receptors in humans with [11C]-(+)-PHNO: dissection of D3 signal and anatomy. Neuroimage. 2011;54:264–277.
    1. Rousset OG, Ma Y, Evans AC. Correction for partial volume effects in PET: principle and validation. J. Nucl. Med. 1998;39:904–911.
    1. Fischl B, Dale AM. Measuring the thickness of the human cerebral cortex from magnetic resonance images. Proc. Natl Acad. Sci. USA. 2000;97:11050–11055.
    1. Jack CR, et al. Defining imaging biomarker cut points for brain aging and Alzheimer’s disease. Alzheimers Dement. 2017;13:205–216.
    1. Anticevic A, et al. Global prefrontal and fronto-amygdala dysconnectivity in bipolar I disorder with psychosis history. Biol. Psychiatry. 2013;73:565–573.
    1. Fortier CB, et al. Reduced cortical thickness in abstinent alcoholics and association with alcoholic behavior. Alcohol Clin. Exp. Res. 2011;35:2193–2201.
    1. Bae S, et al. Prefrontal cortical thickness deficit in detoxified alcohol-dependent patients. Exp. Neurobiol. 2016;25:333–341.
    1. Wang GY, et al. Longitudinal mapping of gyral and sulcal patterns of cortical thickness and brain volume regain during early alcohol abstinence. Eur. Addict. Res. 2016;22:80–89.
    1. Uhlmann A, et al. Grey matter structural differences in alcohol-dependent individuals with and without comorbid depression/anxiety-an MRI study. Eur. Arch. Psychiatry Clin. Neurosci. 2019;269:285–294.
    1. Wilson S, Bair JL, Thomas KM, Iacono WG. Problematic alcohol use and reduced hippocampal volume: a meta-analytic review. Psychol. Med. 2017;47:2288–2301.
    1. Dickerson BC, et al. The cortical signature of Alzheimer’s disease: regionally specific cortical thinning relates to symptom severity in very mild to mild AD dementia and is detectable in asymptomatic amyloid-positive individuals. Cereb. Cortex. 2009;19:497–510.
    1. Pettigrew C, et al. Cortical thickness in relation to clinical symptom onset in preclinical AD. Neuroimage Clin. 2016;12:116–122.
    1. Devanand DP, et al. Hippocampal and entorhinal atrophy in mild cognitive impairment: prediction of Alzheimer disease. Neurology. 2007;68:828–836.
    1. Mueller SG, et al. Hippocampal atrophy patterns in mild cognitive impairment and Alzheimer’s disease. Hum. Brain Mapp. 2010;31:1339–1347.
    1. Schottenbauer MA, Hommer D, Weingartner H. Memory deficits among alcoholics: performance on a selective reminding task. Neuropsychol. Dev. Cogn. B Aging Neuropsychol. Cogn. 2007;14:505–516.
    1. Tedstone D, Coyle K. Cognitive impairments in sober alcoholics: performance on selective and divided attention tasks. Drug Alcohol Depend. 2004;75:277–286.
    1. Cordovil De Sousa Uva M, et al. Distinct effects of protracted withdrawal on affect, craving, selective attention and executive functions among alcohol-dependent patients. Alcohol Alcohol. 2010;45:241–246.
    1. Kühn S, et al. Plasticity of hippocampal subfield volume cornu ammonis 2+3 over the course of withdrawal in patients with alcohol dependence. JAMA Psychiatry. 2014;71:806–811.
    1. Fan LY, et al. The relation between brain amyloid deposition, cortical atrophy, and plasma biomarkers in amnesic mcgnitive impairment and Alzheimer’s disease. Front Aging Neurosci. 2018;10:175.
    1. Jack CR, et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013;12:207–216.
    1. Mattsson N, et al. Association of brain amyloid-β with cerebral perfusion and structure in Alzheimer’s disease and mild cognitive impairment. Brain. 2014;137:1550–1561.
    1. Durazzo TC, et al. Cortical thickness, surface area, and volume of the brain reward system in alcohol dependence: relationships to relapse and extended abstinence. Alcohol Clin. Exp. Res. 2011;35:1187–1200.
    1. Im S, et al. Surface-based parameters of brain imaging in male patients with alcohol use disorder. Psychiatry Investig. 2016;13:511–517.
    1. Burzynska AZ, et al. Cortical thickness is linked to executive functioning in adulthood and aging. Hum. Brain Mapp. 2012;33:1607–1620.
    1. Lee MD, Wagenmakers EJ. Bayesian Cognitive Modeling: A Practical Course. Cambridge Univ. Press; 2013.

Source: PubMed

3
Předplatit