Race modifies the relationship between cognition and Alzheimer's disease cerebrospinal fluid biomarkers

Jennifer C Howell, Kelly D Watts, Monica W Parker, Junjie Wu, Alexander Kollhoff, Thomas S Wingo, Cornelya D Dorbin, Deqiang Qiu, William T Hu, Jennifer C Howell, Kelly D Watts, Monica W Parker, Junjie Wu, Alexander Kollhoff, Thomas S Wingo, Cornelya D Dorbin, Deqiang Qiu, William T Hu

Abstract

Background: African Americans have been reported to have a higher prevalence of Alzheimer's disease (AD) than Caucasians, but etiology-specific AD biomarkers have not been systematically analyzed in older African Americans. Coexisting cerebrovascular disease may also contribute to this increased prevalence. We hypothesized that cerebrospinal fluid (CSF) biomarkers of amyloid, neurodegeneration, and endothelial dysfunction would differ between older African Americans and Caucasians with normal cognition and cognitive impairment associated with AD.

Methods: We prospectively recruited 135 older Americans to undergo detailed clinical, neuropsychological, genetic, magnetic resonance imaging (MRI), and CSF analysis from 2013 to 2015 at Emory University (Atlanta, GA, USA). We compared levels of CSF markers for β-amyloid (Aβ42, Aβ40), total and phosphorylated tau (t-tau and p-tau181, respectively), endothelial dysfunction (soluble vascular cell adhesion molecule 1, soluble intercellular adhesion molecule 1), α-synuclein, and neurodegeneration (neurofilament light chain [NfL]), as well as MRI markers, for hippocampal atrophy and cerebrovascular disease (white matter hyperintensity [WMH] volume).

Results: Sixty-five older African Americans (average age, 69.1 years) and 70 older Caucasians (average age, 70.8 years) were included. After adjusting for demographic variables, AD risk alleles, and cognitive function, older African Americans had lower CSF levels of p-tau181 (difference of 7.4 pg/ml; 95% CI, 3.7-11.2 pg/ml; p < 0.001), t-tau (difference of 23.6 pg/ml; 95% CI, 9.5-37.7; p = 0.001), and Aβ40 (difference of 1.35 ng/ml; 95% CI, 0.29-2.42 ng/ml; p = 0.013) despite similar levels of Aβ42, NfL, WMH volume, and hippocampal volume. Cognitively impaired African Americans also had lower CSF t-tau/Aβ42 (difference of 0.255 per 1-SD change in composite cognition; 95% CI, 0.100-0.409; p = 0.001) and p-tau181/Aβ42 (difference of 0.076 per 1-SD change in composite cognition; 95% CI, 0.031-0.122; p = 0.001). These could not be explained by measured biomarkers of non-AD processes, but African Americans may be more susceptible than Caucasians to the cognitive effects of WMH.

Conclusions: Despite comparable levels of CSF Aβ42 and Aβ42/Aβ40, cognitive impairment in African Americans is associated with smaller changes in CSF tau markers but greater impact from similar WMH burden than Caucasians. Race-associated differences in CSF tau markers and ratios may lead to underdiagnosis of AD in African Americans.

Trial registration: ClinicalTrials.gov, NCT02089555 . Retrospectively registered on 14 March 2014.

Keywords: African American; Amyloid; Dementia; Endothelial dysfunction; Mild cognitive impairment; Tau.

Conflict of interest statement

Ethics approval and consent to participate

This study was approved by the Emory Institutional Review Board (number 66145). Informed consent was obtained from all subjects or their authorized representatives.

Consent for publication

Not applicable.

Competing interests

WTH has a patent (assignee, Emory University) on the use of CSF p/t-tau ratio in the evaluation of frontotemporal lobar degeneration, has received research support from Avid Pharmaceuticals, and has received travel support from Eli Lilly and Hoffman-La Roche. DQ has received research support from Medtronic and Siemens Healthcare.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Cerebrospinal fluid (CSF) levels of tau and amyloid markers in older African Americans and Caucasians according to cognitive function. Composite cognitive Z-scores are shown on the x-axis (lower score corresponds to worse cognitive function). African Americans (closed circles) had lower CSF levels of total tau (t-tau) (a), tau phosphorylated at threonine 181 (p-tau181) (b), and β-amyloid 1–40 (Aβ40) (d) than Caucasians (open circles). Raw values are shown, with dashed lines representing trends among Caucasians and solid lines representing trends among African Americans. The differences persisted after adjusting for age, sex, apolipoprotein E (APOE) and ABCA7 genotypes, and β-amyloid 1–42 (Aβ42) levels (c), which did not differ between the two groups. CSF biomarker t-tau/Aβ42 ratio was lower in African Americans than in Caucasians when there was cognitive impairment (e), but race did not have a significant effect on CSF biomarker Aβ42/Aβ40 (f)
Fig. 2
Fig. 2
Relationship between cerebrospinal fluid (CSF) soluble vascular cell adhesion molecule 1 (sVCAM-1) levels and other vascular markers according to race. In Caucasians (open circles), CSF sVCAM-1 levels strongly correlated with log-transformed white matter hyperintensity (WMH) volumes derived by magnetic resonance imaging (a, b) and the total number of peripheral vascular risk factors (c, d) whether a more (a, c) or less stringent (b, d) threshold was applied to identify subjects with no Alzheimer’s disease pathology. However, there was no such correlation in African Americans (closed circles). Aβ42 β-Amyloid 1–42, t-Tau Total tau

References

    1. Association A’s. 2010 Alzheimer’s disease facts and figures. Alzheimers Dement. 2010;6:158–94. doi: 10.1016/j.jalz.2010.01.009.
    1. Manly JJ, Tang MX, Schupf N, Stern Y, Vonsattel JP, Mayeux R. Frequency and course of mild cognitive impairment in a multiethnic community. Ann Neurol. 2008;63:494–506. doi: 10.1002/ana.21326.
    1. Green RC, Cupples LA, Go R, Benke KS, Edeki T, Griffith PA, Williams M, Hipps Y, Graff-Radford N, Bachman D, Farrer LA. Risk of dementia among white and African American relatives of patients with Alzheimer disease. JAMA. 2002;287:329–36. doi: 10.1001/jama.287.3.329.
    1. Reitz C, Jun G, Naj A, Rajbhandary R, Vardarajan BN, Wang LS, Valladares O, Lin CF, Larson EB, Graff-Radford NR, et al. Variants in the ATP-binding cassette transporter (ABCA7), apolipoprotein E ε4, and the risk of late-onset Alzheimer disease in African Americans. JAMA. 2013;309:1483–92. doi: 10.1001/jama.2013.2973.
    1. Logue MW, Schu M, Vardarajan BN, Buros J, Green RC, Go RC, Griffith P, Obisesan TO, Shatz R, Borenstein A, et al. A comprehensive genetic association study of Alzheimer disease in African Americans. Arch Neurol. 2011;68:1569–79. doi: 10.1001/archneurol.2011.646.
    1. Hollingworth P, Harold D, Sims R, Gerrish A, Lambert JC, Carrasquillo MM, Abraham R, Hamshere ML, Pahwa JS, Moskvina V, et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat Genet. 2011;43:429–35. doi: 10.1038/ng.803.
    1. Mez J, Chung J, Jun G, Kriegel J, Bourlas AP, Sherva R, Logue MW, Barnes LL, Bennett DA, Buxbaum JD, et al. Two novel loci, COBL and SLC10A2, for Alzheimer’s disease in African Americans. Alzheimers Dement. 2017;13:119–29. doi: 10.1016/j.jalz.2016.09.002.
    1. Livney MG, Clark CM, Karlawish JH, Cartmell S, Negron M, Nunez J, Xie SX, Entenza-Cabrera F, Vega IE, Arnold SE. Ethnoracial differences in the clinical characteristics of Alzheimer’s disease at initial presentation at an urban Alzheimer’s disease center. Am J Geriatr Psychiatry. 2011;19:430–9. doi: 10.1097/JGP.0b013e3181f7d881.
    1. Barnes LL, Wilson RS, Li Y, Aggarwal NT, Gilley DW, McCann JJ, Evans DA. Racial differences in the progression of cognitive decline in Alzheimer disease. Am J Geriatr Psychiatry. 2005;13:959–67. doi: 10.1097/00019442-200511000-00006.
    1. Graff-Radford NR, Besser LM, Crook JE, Kukull WA, Dickson DW. Neuropathologic differences by race from the National Alzheimer’s Coordinating Center. Alzheimers Dement. 2016;12:669–77. doi: 10.1016/j.jalz.2016.03.004.
    1. Barnes LL, Leurgans S, Aggarwal NT, Shah RC, Arvanitakis Z, James BD, Buchman AS, Bennett DA, Schneider JA. Mixed pathology is more likely in black than white decedents with Alzheimer dementia. Neurology. 2015;85:528–34. doi: 10.1212/WNL.0000000000001834.
    1. Petersen RC, Aisen PS, Beckett LA, Donohue MC, Gamst AC, Harvey DJ, Jack CR, Jr, Jagust WJ, Shaw LM, Toga AW, et al. Alzheimer’s Disease Neuroimaging Initiative (ADNI): clinical characterization. Neurology. 2010;74:201–9. doi: 10.1212/WNL.0b013e3181cb3e25.
    1. Hu WT, Chen-Plotkin A, Arnold SE, Grossman M, Clark CM, Shaw LM, Pickering E, Kuhn M, Chen Y, McCluskey L, et al. Novel CSF biomarkers for Alzheimer’s disease and mild cognitive impairment. Acta Neuropathol. 2010;119:669–78. doi: 10.1007/s00401-010-0667-0.
    1. Clark CM, Schneider JA, Bedell BJ, Beach TG, Bilker WB, Mintun MA, Pontecorvo MJ, Hefti F, Carpenter AP, Flitter ML, et al. Use of florbetapir-PET for imaging beta-amyloid pathology. JAMA. 2011;305:275–83. doi: 10.1001/jama.2010.2008.
    1. Howell JC, Parker MW, Watts KD, Kollhoff A, Tsvetkova DZ, Hu WT. Research lumbar punctures among African Americans and Caucasians: perception predicts experience. Front Aging Neurosci. 2016;8:296. doi: 10.3389/fnagi.2016.00296.
    1. Hall JR, Wiechmann AR, Johnson LA, Edwards M, Barber RC, Winter AS, Singh M, O’Bryant SE. Biomarkers of vascular risk, systemic inflammation, and microvascular pathology and neuropsychiatric symptoms in Alzheimer’s disease. J Alzheimers Dis. 2013;35:363–71.
    1. Ewers M, Mielke MM, Hampel H. Blood-based biomarkers of microvascular pathology in Alzheimer’s disease. Exp Gerontol. 2010;45:75–9. doi: 10.1016/j.exger.2009.09.005.
    1. Lucas JA, Ivnik RJ, Smith GE, Ferman TJ, Willis FB, Petersen RC, Graff-Radford NR. Mayo’s Older African Americans Normative Studies: norms for Boston Naming Test, Controlled Oral Word Association, Category Fluency, Animal Naming, Token Test, WRAT-3 Reading, Trail Making Test, Stroop Test, and Judgment of Line Orientation. Clin Neuropsychol. 2005;19:243–69. doi: 10.1080/13854040590945337.
    1. Machulda MM, Ivnik RJ, Smith GE, Ferman TJ, Boeve BF, Knopman D, Petersen RC, Tangalos EG. Mayo’s Older Americans Normative Studies: Visual Form Discrimination and copy trial of the Rey-Osterrieth Complex Figure. J Clin Exp Neuropsychol. 2007;29:377–84. doi: 10.1080/13803390600726803.
    1. Hebert LE, Scherr PA, Bennett DA, Bienias JL, Wilson RS, Morris MC, Evans DA. Blood pressure and late-life cognitive function change: a biracial longitudinal population study. Neurology. 2004;62:2021–4. doi: 10.1212/01.WNL.0000129258.93137.4B.
    1. Crane PK, Narasimhalu K, Gibbons LE, Pedraza O, Mehta KM, Tang Y, Manly JJ, Reed BR, Mungas DM. Composite scores for executive function items: demographic heterogeneity and relationships with quantitative magnetic resonance imaging. J Int Neuropsychol Soc. 2008;14:746–59. doi: 10.1017/S1355617708081162.
    1. Crane PK, Carle A, Gibbons LE, Insel P, Mackin RS, Gross A, Jones RN, Mukherjee S, Curtis SM, Harvey D, et al. Development and assessment of a composite score for memory in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) Brain Imaging Behav. 2012;6:502–16. doi: 10.1007/s11682-012-9186-z.
    1. Hu WT, Watts KD, Tailor P, Nguyen TP, Howell JC, Lee RC, Seyfried NT, Gearing M, Hales CM, Levey AI, et al. CSF complement 3 and factor H are staging biomarkers in Alzheimer’s disease. Acta Neuropathol Commun. 2016;4:14. doi: 10.1186/s40478-016-0277-8.
    1. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive impairment: clinical characterization and outcome. Arch Neurol. 1999;56:303–8. doi: 10.1001/archneur.56.3.303.
    1. Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, et al. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging and Alzheimer’s Association Workgroup. Alzheimers Dement. 2011;7:270–9. doi: 10.1016/j.jalz.2011.03.008.
    1. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34:939–44. doi: 10.1212/WNL.34.7.939.
    1. Dubois B, Feldman HH, Jacova C, DeKosky ST, Barberger-Gateau P, Cummings J, Delacourte A, Galasko D, Gauthier S, Jicha G, et al. Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteria. Lancet Neurol. 2007;6:734–46. doi: 10.1016/S1474-4422(07)70178-3.
    1. Lucas JA, Ivnik RJ, Willis FB, Ferman TJ, Smith GE, Parfitt FC, Petersen RC, Graff-Radford NR. Mayo’s Older African Americans Normative Studies: normative data for commonly used clinical neuropsychological measures. Clin Neuropsychol. 2005;19:162–83. doi: 10.1080/13854040590945265.
    1. Shaw LM, Vanderstichele H, Knapik-Czajka M, Clark CM, Aisen PS, Petersen RC, Blennow K, Soares H, Simon A, Lewczuk P, et al. Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol. 2009;65:403–13. doi: 10.1002/ana.21610.
    1. Fischl B, Salat DH, Busa E, Albert M, Dieterich M, Haselgrove C, van der Kouwe A, Killiany R, Kennedy D, Klaveness S, et al. Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron. 2002;33:341–55. doi: 10.1016/S0896-6273(02)00569-X.
    1. Carmichael O, Schwarz C, Drucker D, Fletcher E, Harvey D, Beckett L, Jack CR, Jr, Weiner M, DeCarli C. Longitudinal changes in white matter disease and cognition in the first year of the Alzheimer disease neuroimaging initiative. Arch Neurol. 2010;67:1370–8. doi: 10.1001/archneurol.2010.284.
    1. Bateman RJ, Wen G, Morris JC, Holtzman DM. Fluctuations of CSF amyloid-β levels: implications for a diagnostic and therapeutic biomarker. Neurology. 2007;68:666–9. doi: 10.1212/01.wnl.0000256043.50901.e3.
    1. Hu WT, Watts KD, Shaw LM, Howell JC, Trojanowski JQ, Basra S, Glass JD, Lah JJ, Levey AI. CSF β-amyloid 1–42 – what are we measuring in Alzheimer’s disease? Ann Clin Transl Neurol. 2015;2:131–9. doi: 10.1002/acn3.160.
    1. Jin SC, Carrasquillo MM, Benitez BA, Skorupa T, Carrell D, Patel D, Lincoln S, Krishnan S, Kachadoorian M, Reitz C, et al. TREM2 is associated with increased risk for Alzheimer’s disease in African Americans. Mol Neurodegener. 2015;10:19. doi: 10.1186/s13024-015-0016-9.
    1. Schneider AL, Sharrett AR, Gottesman RF, Coresh J, Coker L, Wruck L, Selnes OA, Deal J, Knopman D, Mosley TH. Normative data for 8 neuropsychological tests in older blacks and whites from the atherosclerosis risk in communities (ARIC) study. Alzheimer Dis Assoc Disord. 2015;29:32–44. doi: 10.1097/WAD.0000000000000042.
    1. Fillenbaum GG, Heyman A, Huber MS, Ganguli M, Unverzagt FW. Performance of elderly African American and white community residents on the CERAD Neuropsychological Battery. J Int Neuropsychol Soc. 2001;7:502–9. doi: 10.1017/S1355617701744062.
    1. Jack CR, Jr, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010;9:119–28. doi: 10.1016/S1474-4422(09)70299-6.
    1. Lee MJ, Seo SW, Na DL, Kim C, Park JH, Kim GH, Kim CH, Noh Y, Cho H, Kim HJ, et al. Synergistic effects of ischemia and β-amyloid burden on cognitive decline in patients with subcortical vascular mild cognitive impairment. JAMA Psychiatry. 2014;71:412–22. doi: 10.1001/jamapsychiatry.2013.4506.
    1. Steenland K, Goldstein FC, Levey A, Wharton W. A Meta-analysis of Alzheimer’s disease incidence and prevalence comparing African-Americans and Caucasians. J Alzheimers Dis. 2016;50:71–6. doi: 10.3233/JAD-150778.
    1. Hu WT, Watts K, Grossman M, Glass J, Lah JJ, Hales C, Shelnutt M, Van Deerlin V, Trojanowski JQ, Levey AI. Reduced CSF p-Tau181 to Tau ratio is a biomarker for FTLD-TDP. Neurology. 2013;81:1945–52. doi: 10.1212/01.wnl.0000436625.63650.27.
    1. Evans W, Fung HC, Steele J, Eerola J, Tienari P, Pittman A, Silva R, Myers A, Vrieze FW, Singleton A, Hardy J. The tau H2 haplotype is almost exclusively Caucasian in origin. Neurosci Lett. 2004;369:183–5. doi: 10.1016/j.neulet.2004.05.119.
    1. Kauwe JS, Cruchaga C, Mayo K, Fenoglio C, Bertelsen S, Nowotny P, Galimberti D, Scarpini E, Morris JC, Fagan AM, et al. Variation in MAPT is associated with cerebrospinal fluid tau levels in the presence of amyloid-β deposition. Proc Natl Acad Sci U S A. 2008;105:8050–4. doi: 10.1073/pnas.0801227105.
    1. Scheible M, Just R, Sturk-Andreaggi K, Saunier J, Parson W, Parsons T, Coble M, Irwin J. The mitochondrial landscape of African Americans: an examination of more than 2500 control region haplotypes from 22 U.S. locations. Forensic Sci Int Genet. 2016;22:139–48. doi: 10.1016/j.fsigen.2016.01.002.

Source: PubMed

3
구독하다