You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Association of Peripheral Lymphocyte Subsets with Cognitive Decline and Dementia: The Cardiovascular Health Study

INTRODUCTION

Neuroinflammation is implicated in rapid progression of Alzheimer’s disease (AD) and in altered learning and memory [1, 2]. Large scale genomic studies support the role of the immune system in dementia [3–10]. Immune cells have been identified in brain tissue and cerebrospinal fluid of people with AD, and the levels of immune cells in blood may be associated with dementias, perhaps reflecting premature immunosenescence and chronic inflammation [11–18]. For example, higher circulating proportions of natural killer (NK) cells were apparent before AD onset and the depletion of NK cells improved cognitive function in mice [19]. In humans, high NK cell activity was associated with worse cognitive performance of AD patients [14, 20, 21]. Increased numbers of CD8 + T effector memory CD45RA+(CD8 TEMRA) cells and of CD4 + T effector memory CD45RA+(CD4 TEMRA) cells were identified in the blood of patients with AD [12, 22, 23]. Prior studies report both decreased and increased proportions of regulatory T cells (Treg) in blood of patients with AD [22–27]. Immunosenescence of T cells may promote AD by decreasing anti-amyloid antibodies, which could result in less clearance of amyloid plaques [28], or by producing high levels of pro-inflammatory cytokines and oxidative stress, which could damage neurons [29, 30]. However, many of the associations between peripheral immune cells and cognitive outcomes are based on small, cross-sectional studies. Clarifying the association of the peripheral adaptive and innate immune system with risk for cognitive decline and dementia in a large, population-based setting may illuminate underlying biological processes and lead to the development of better therapeutics.

The Cardiovascular Health Study (CHS) is a population-based, prospective cohort study that included serial cognitive evaluations, dementia adjudication, and assessment of 18 types of peripheral innate and adaptive immune cell subsets. Importantly, CHS allows evaluation of the association of circulating immune cells with both cross-sectional and longitudinal cognitive and dementia outcomes. We hypothesized a priori that high proportions of NK, and both CD4 + and CD8 + TEMRA cells would be associated with worse prospective cross-sectional global cognition, worse cognitive decline, prevalent dementia, and shorter time to death from dementia. We hypothesized that higher proportions of Treg protect against these adverse cognitive outcomes. We investigated all other available immune cell subsets as exploratory hypotheses to broadly characterize the relationship of the peripheral immune system with cognitive decline and dementia.

METHODS AND MATERIALS

Study design and approval

The CHS is a population-based, longitudinal cohort of 5,888 men and women aged 65 years or older at enrollment in 1989–93 [31]. Analytic baseline for this analysis was defined as the 1998–1999 CHS exam because this study leverages immune cell phenotype data obtained [32] from the 1998–1999 exams. Institutional review boards at the University of Washington and at each study site approved the study. All CHS participants provided written informed consent.

Immune cell measurement

Detailed methods for immune cell phenotyping and flow cytometry gating strategies have been published [32, 33]. Briefly, as part of the 1998–1999 exam, peripheral blood mononuclear cells (PBMCs) were cryopreserved. Flow cytometry was used to differentiate cell types based on surface marker expression. Cell phenotypes were expressed as proportions of larger “parent” populations, as indicated in Table 2. Poor sample quality and technical assay errors resulted in missing data, which appear to be missing at random. IgG antibodies to cytomegalovirus (CMV) were measured in serum by enzyme immunoassay (Diamedix Corp., Miami Lakes, FL); the inter-assay coefficients of variation of CMV titer were 5.1% –6.8%.

Cognition and dementia adjudication

Global cognitive performance was assessed with the 100-point Modified Mini Mental State Exam (3MSE) for participants in 1998–1999 and was repeated among participants remaining in the study in 2005–2006. Participants who did not attend an in-person exam were contacted via telephone to complete a Telephone Interview for Cognitive Status (TICS) exam. The TICS score explains 67% of variability in 3MSE in CHS and can be used to estimate 3MSE score with a correlation of 0.82 between actual and TICS-estimated 3MSE [34, 35]. We used TICS score to estimate the 3MSE score when 3MSE was missing. TICS was used to estimate one score at the 1998–1999 exam and 194 scores at the 2005–2006 exam.

A committee of neurologists and psychiatrists used Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) criteria to adjudicate dementia prior to 1998–1999 [36–38]. Death from dementia was ascertained on all participants through 2015 with evidence of advanced dementia prior to death without evidence of another cause [39].

Statistical analysis

For main analyses we used multiple imputation with chained equations (40 imputations) to impute all missing data, including any missing covariates, cell phenotypes, and cognitive outcomes. We imputed 20 cross-sectional cognitive scores, 996 changes in cognitive score, 402 statuses of prevalent dementia, and 19 times to death with dementia. Data were imputed in blocks of immune cells, covariates, and cognitive outcomes. Additional covariates associated with aging-related outcomes in CHS were included in the imputation process to improve estimates [40–44]. In the main imputed analyses, all participants were included in all analyses.

Due to co-linearity, we analyzed each cell phenotype separately, per standard deviation (SD) higher value. Associations between immune cell proportion per 1-SD and both cross-sectional 3MSE score and change in 3MSE between 1998–1999 and 2005–2006 were assessed using linear regression. We used Poisson regression to determine relative risk of prevalent dementia per 1-SD higher immune cell proportion. We used Cox proportional hazards regression to determine hazard ratio of death from dementia per 1-SD higher immune cell proportion. Censoring occurred at death without noted dementia or in 2015. All analyses were adjusted for age, sex, Black race, systolic blood pressure, smoking status, statin use, education, prevalent diabetes, APOE4 allele carrier status, body mass index (BMI), CMV antibody titer, assay batch, and study site.

In exploratory analyses, we repeated all analyses stratified by sex. In sensitivity analyses, we excluded 135 participants who had experienced a stroke prior to the 1998–1999 exam. To identify potential influence of imputed missing data, we performed complete case analysis for associations between immune cell proportion and three outcomes: cross-sectional cognitive function, prevalent dementia, and time-to-death from dementia. We weighted individuals with 2005–2006 data by the inverse probability of the likelihood of having cognitive scores in 2005–2006 to perform a sensitivity analysis of the association of immune cell proportions and seven-year change in cognitive function [45].

For main endpoints, a p-value less than 0.0125 was considered significant to account for the four primary cell types assessed. All other analyses were exploratory and a p-value of 0.05 was considered significant. We conducted all analyses in RStudio (R version 3.6.3). The data that support the findings of this study are available upon reasonable request through the CHS Coordinating Center (CHS-NHLBI.org).

RESULTS

Our analysis included 1928 CHS participants with at least one peripheral immune subset measured. Table 1 presents their characteristics at analytic baseline. Cognitive evaluations were performed seven years later in a subset of 932 (48%) participants and scores declined a mean of 6.5 points (standard deviation = 9.7). Over 16 years of follow up, 272 (14%) participants died from dementia. Table 2 presents immune cells as proportions of their parent population.

Table 1

Characteristics of CHS subjects with immune cell data (n = 1928) at analytic baseline

VariableMean or numberStandard deviation (SD) or %
Age, y (mean, SD)79.74.4
Male (n, %)73238.0%
Black (n, %)33917.6%
Study site (n, %)
  North Carolina53627.8%
  California52127.0%
  Maryland41021.3%
  Pittsburgh46123.9%
Education past grade 12 (n, %)94048.8%
Systolic blood pressure, mmHg (mean, SD)135.220.5
Smoking status (n, %)
  Current1516.6%
  Never93844.1%
  Former83949.3%
At least one APOE4 allele (n, %)53429.0%
BMI, kg/m2 (mean, SD)26.64.4
Prior Stroke (n, %)1357.0%
Diabetes (n, %)37219.4%
Statin user (n, %)30715.9%
CMV antibody titer, EU/mL (mean, SD)190.3183.6
Prevalent dementia (n, %)56336.9%
3MSE (mean, SD)90.411.6

* percentages based on number of participants with data for that variable. No data were missing for age, sex, race, study site, presence of prior stroke, or statin use. The number of missing observations for the other variables are as follows: 3 for level of education, 1 for blood pressure, 25 for smoking status, 84 for APOE4 genotype, 143 for BMI, 10 for prevalent diabetes, 189 for CMV antibody titer, 402 for prevalent dementia, and 1 for 3MSE cognitive score.

Table 2

Cellular phenotypes with their molecular description, parent population, number of samples with data (N), means and standard deviations (SD)

Cellular phenotypeMolecular descriptionParent populationNMeanSD
Primary hypotheses
Natural killerCD3-CD56 + CD16+% Lymphocytes1,5565.14.8
TregCD4 + CD25 + CD127–CD4+1,5456.64.6
CD4 + TEMRACD4 + CD45RA + CD28-CD57+CD4+1,6706.95.9
CD8 + TEMRACD8 + CD45RA + CD28-CD57+CD8+1,67523.614.1
Exploratory hypotheses
γδ T cellsCD3+γδ+CD3+1,5395.54.9
B cellsCD19+% Lymphocytes1,55619.715.9
T helper cellsCD4+% Lymphocytes1,67350.114.5
Cytotoxic T cellsCD8+% Lymphocytes1,69117.39.7
Th1CD4 + CD194-CXCR3 + CD196–CD4+1,32620.28.0
Th2CD4 + CD194 + CXCR3-CD196–CD4+1,3264.73.8
Th17CD4 + CD194 + CXCR3-CD196+CD4+1,3263.22.6
Naïve CD4 + cellsCD4 + CD45RA+CD4+1,69025.812.6
Memory CD4 + cellsCD4 + CD45RO+CD4+1,69049.715.5
Activated/mature CD4 + cellsCD4 + CD38+CD4+1,68833.415.8
Naïve CD8 + cellsCD8 + CD45RA+CD8+1,70942.616.6
Memory CD8 + cellsCD8 + CD45RO+CD8+1,70230.214.4
Activated/mature CD8 + cellsCD8 + CD38+CD8+1,70734.318.5
Memory B cellsCD19 + CD27+CD19+1,55726.419.8

Table 3 presents associations between each immune cell proportion and both cross-sectional cognitive function and longitudinal change in cognitive function after seven years using imputed data where missing. No immune cell proportions were associated with any cognitive outcomes after accounting for multiple comparisons of the primary cell types. However, higher proportions of Treg were associated with greater decline in cognitive function over seven years if not accounting for multiple comparisons. This association was supported by suggestive associations of higher proportions of Treg with both worse cross-sectional cognitive function and higher risk of prevalent dementia. Supplementary Table 1 presents sensitivity analyses using complete case analysis for cross-sectional cognitive function (no missing data were imputed) and inverse probability weighting for change in cognitive function. These analyses were similarly null.

Table 3

Associations of lymphocyte subsets (per 1 SD) with cognitive outcomes. Analyses for cross-sectional 3MSE score and change in 3MSE score over seven years are based on multiple linear regression. Analysis of prevalent dementia is based on Poisson regression. Analysis of time to death from dementia is based on Cox proportional hazards regression. All analyses adjust for age, sex, Black race, systolic blood pressure, smoking, statin use, education, prevalent diabetes, APOE4 carrier status, BMI, CMV antibody titer, assay batch, and study site. Participants were censored at death without dementia or in 2015. All 1928 individuals are included in each analysis. Missing data were imputed, including covariates (see Table 1), immune cells (see Table 2), and outcomes. We imputed 20 cross-sectional cognitive scores, 996 changes in cognitive score, 402 statuses of prevalent dementia, and 19 times to death with dementia. Beta values that are negative indicate lower cognitive score and a greater decline in cognition over 7 years

Cross-sectional 3 MSE scoreChange over seven yearsCross-sectional dementiaTime to dementia death
Cellular phenotypeBeta95% CIpBeta95% CIpRelative Risk95% CIpHazard Ratio95% CIp
Natural killer0.06–1.71, 1.840.94–0.09–3.90, 3.730.961.150.77, 1.700.501.160.80, 1.690.44
Treg–0.52–1.08, 0.030.065–1.30–2.36, –0.240.0181.100.99, 1.220.0861.130.78, 1.640.52
CD4 + TEMRA–0.12–0.80, 0.550.720.47–0.80, 1.730.470.980.83, 1.160.821.070.76, 1.520.68
CD8 + TEMRA0.05–0.55, 0.650.860.24–0.91, 1.390.681.030.91, 1.180.631.050.74, 1.490.79
γδ T cells0.41–1.81, 2.620.72–0.93–4.85, 2.990.641.000.66, 1.500.981.060.75, 1.500.76
B cells0.26–0.22, 0.730.290.08–0.85, 1.010.860.970.87, 1.080.541.100.77, 1.570.60
T helper cells–0.15–0.33, 0.020.09–0.03–0.37, 0.310.861.010.97, 1.050.741.080.76, 1.530.67
Cytotoxic T cells0.11–0.02, 0.250.09–0.07–0.31, 0.170.561.010.98, 1.040.701.080.76, 1.540.67
Th10.14–0.17, 0.460.38–0.25–0.87, 0.370.440.990.92, 1.060.791.080.76, 1.530.67
Th2–0.12–2.51, 2.280.921.59–4.50, 7.680.610.990.57, 1.710.971.080.75, 1.550.68
Th170.34–2.88, 3.560.841.51–2.14, 5.160.420.990.53, 1.840.971.080.76, 1.530.68
Naïve CD4 + cells0.12–0.42, 0.660.670.14–0.86, 1.150.780.900.79, 1.030.141.000.70, 1.430.99
Memory CD4 + cells–0.04–0.39, 0.300.81–0.13–0.83, 0.560.711.060.97, 1.150.191.010.71, 1.440.97
Activated/mature CD4 + cells0.10–0.45, 0.640.730.24–0.73, 1.200.630.950.84, 1.080.451.090.77, 1.550.62
Naïve CD8 + cells0.06–0.06, 0.170.33–0.13–0.36, 0.090.240.990.96, 1.020.471.080.76, 1.530.66
Memory CD8 + cells–0.25–0.59, 0.100.160.21–0.41, 0.820.511.040.96, 1.130.291.090.77, 1.540.64
Activated/mature CD8 + cells0.10–0.45, 0.640.730.34–0.80, 1.480.560.970.85, 1.100.601.060.71, 1.570.79
Memory B cells–0.15–0.74, 0.450.63–1.00–2.29, 0.300.131.090.96, 1.230.211.160.81, 1.670.42

p-value threshold for the primary endpoints is 0.0125. Bolded cells are primary hypotheses. CI, Confidence Interval.

Additionally, no immune cell proportions were associated with prevalent dementia or time to death from dementia (Table 3). Supplementary Table 1 presents sensitivity analyses, using complete case analysis for both prevalent dementia and time to death from dementia.

We did not observe associations between immune cell subsets and any of the outcomes in analyses stratified by sex, when excluding participants with stroke prior to blood collection, or when evaluating prevalent AD specifically rather than all-cause dementia.

Exploratory analysis using principal components of the immune cell distributions did not identify significant associations.

4DISCUSSION

In a large, population-based, longitudinal cohort of older adults with well-defined outcomes, we did not identify associations of peripheral immune cells with either cross-sectional or longitudinal cognitive outcomes after accounting for multiple comparisons. However, there were marginally significant associations of Treg with worse cognitive decline in both imputed and weighted probability models when not accounting for multiple comparisons, and this association was supported by suggestive associations of Treg with both worse cross-sectional cognitive function and prevalent dementia. Higher proportions of Treg may reflect ongoing mobilization in response to inflammation.

The immune cell subsets that we measured at a single timepoint in blood may not reflect features of the immune system most important for cognitive decline, or dynamic temporal intrapersonal variability in cell levels. Overall numbers, activity, or location of immune cells may better reflect pathology than immune cell proportions in peripheral blood. For example, all B and CD4 + T cell count may be diminished in dementia, which may not be captured when evaluating proportions [13, 18]. Treg and NK cells from patients with AD are reported to have altered function [14, 20, 21, 46]. Neurodegeneration may be driven by proinflammatory cytokines and chemokines produced by the immune cells [47]. For example, IL-1β, IL-6, and TNFα are thought to induce neuronal death [47]. Additionally, we may not have evaluated all relevant immune cell subphenotypes. For example, specific Treg subtypes might be more associated with pathologies than proportions of Treg overall [26]. Furthermore, cells may act in concert to affect cognitive decline and AD [48–50], and we have evaluated each cell type independently.

Peripheral immune cells may not reflect immune cells in the brain and cerebrospinal fluid, which may be more important for cognitive decline and dementia. The role of the immune system may vary by type of dementia [13]. NK cells may be diminished in vascular dementia, but not in AD, and the distribution of naïve and memory T cells may be altered only in AD [13]. The majority of dementia in CHS was AD, but ∼25% had vascular dementia and ∼10% had mixed dementia based on adjudicated diagnoses. Our dementia endpoint included all dementia subtypes and limiting our analysis to adjudicated AD did not affect our findings.

Other limitations of the study include large amounts of missing data and that participants with cognitive data are known to be healthier than those without cognitive data or who did not survive to the 1998–1999 exam. Survival and participation bias is especially likely to affect longitudinal analysis of cognitive decline, where participants experiencing greater cognitive decline were less likely to be re-examined in follow-up. We attempted to account for missing data through multiple imputation with chained equations and with probability weighting based on likelihood of participation in the follow up exam, but selection bias remains a concern. Nonetheless, sensitivity analyses were also null. Other sources of bias include that death from dementia is specific, but not sensitive, and we likely underestimated the number of participants who died with dementia. Immune cells were measured at only one time point, several years after cohort development. Participants must have survived and been healthy enough to participate in a blood draw during the 1998–1999 exam. These older participants may already have experienced changes in their immune system that could affect cognitive function and decline.

The role of the immune system in dementia is likely complex. While neuroinflammation is well established with respect to AD, anti-inflammatory therapy for AD has had poor results [51]. Our findings that peripheral immune cells, measured as proportions, are not associated with cross-sectional global cognition, cognitive decline, prevalent dementia, or time to death with dementia may reflect the complexity of both the immune system and its role in AD and related dementias. Further studies are needed to clarify associations between Treg and subsequent dementia and cognitive decline.

ACKNOWLEDGMENTS

This project was supported by grants HL120854 and HL135625 from the National Heart Lung and Blood Institute (NHLBI). The CHS cohort was supported by contracts HHSN268201200036C, HHSN268200800007C, HHSN268201800001C, N01HC55222, N01HC85079, N01HC85080, N01HC85081, N01HC85082, N01HC85083, N01HC85086, 75N92021D00006, and grants U01HL080295 and U01HL130114 from the National Heart, Lung, and Blood Institute (NHLBI), with additional contribution from the National Institute of Neurological Disorders and Stroke (NINDS). Additional support was provided by R01AG023629 from the National Institute on Aging (NIA). Alison Fohner is supported by K01AG071689. A full list of principal CHS investigators and institutions can be found at CHS-NHLBI.org. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Authors’ disclosures available online (https://www.j-alz.com/manuscript-disclosures/22-0091r1).

SUPPLEMENTARY MATERIAL

[1] The supplementary material is available in the electronic version of this article: https://dx.doi.org/10.3233/JAD-220091.

REFERENCES

[1] 

Kipnis J , Cohen H , Cardon M , Ziv Y , Schwartz M ((2004) ) T cell deficiency leads to cognitive dysfunction: Implications for therapeutic vaccination for schizophrenia and other psychiatric conditions.Proc Natl Acad Sci U S A 101: , 8180–8185.

[2] 

Lee S , Cho HJ , Ryu JH ((2021) ) Innate immunity and cell death in Alzheimer’s disease.ASN Neuro 13: , 17590914211051908.

[3] 

Jun G , Naj AC , Beecham GW , Wang L-S , Buros J , Gallins PJ , Buxbaum JD , Ertekin-Taner N , Fallin MD , Friedland R , Inzelberg R , Kramer P , Rogaeva E , St George-Hyslop P , Alzheimer’s Disease Genetics Consortium, Cantwell LB , Dombroski BA , Saykin AJ , Reiman EM , Bennett DA , Morris JC , Lunetta KL , Martin ER , Montine TJ , Goate AM , Blacker D , Tsuang DW , Beekly D , Cupples LA , Hakonarson H , Kukull W , Foroud TM , Haines J , Mayeux R , Farrer LA , Pericak-Vance MA , Schellenberg GD ((2010) ) Meta-analysis confirms CR1, CLU, and PICALM as alzheimer disease risk loci and reveals interactions with APOE genotypes.Arch Neurol 67: , 1473–1484.

[4] 

Lambert JC , Heath S , Even G , Campion D , Sleegers K , Hiltunen M , Combarros O , Zelenika D , Bullido MJ , Tavernier B , Letenneur L , Bettens K , Berr C , Pasquier F , Fiévet N , Barberger-Gateau P , Engelborghs S , De Deyn P , Mateo I , Franck A , Helisalmi S , Porcellini E , Hanon O , de Pancorbo MM , Lendon C , Dufouil C , Jaillard C , Leveillard T , Alvarez V , Bosco P , Mancuso M , Panza F , Nacmias B , Bossù P , Piccardi P , Annoni G , Seripa D , Galimberti D , Hannequin D , Licastro F , Soininen H , Ritchie K , Blanché H , Dartigues JF , Tzourio C , Gut I , VanBroeckhoven C , Alpévitch A , Lathrop M , Amouyel P ((2009) ) Genome-wide association study identifies variants at CLU andCR1 associated with Alzheimer’s disease.Nat Genet 41: , 1094–1099.

[5] 

Lambert JC , Ibrahim-Verbaas CA , Harold D , Naj AC , Sims R , Bellenguez C , DeStafano AL , Bis JC , Beecham GW , Grenier-Boley B , Russo G , Thorton-Wells TA , Jones N , Smith AV , Chouraki V , Thomas C , Ikram MA , Zelenika D , Vardarajan BN , Kamatani Y , Lin CF , Gerrish A , Schmidt H , Kunkle B , Dunstan ML , Ruiz A , Bihoreau MT , Choi SH , Reitz C , Pasquier F , Cruchaga C , Craig D , Amin N , Berr C , Lopez OL , De Jager PL , Deramecourt V , Johnston JA , Evans D , Lovestone S , Letenneur L , Morón FJ , Rubinsztein DC , Eiriksdottir G , Sleegers K , Goate AM , Fiévet N , Huentelman MW , Gill M , Brown K , Kamboh MI , Keller L , Barberger-Gateau P , McGuiness B , Larson EB , Green R , Myers AJ , Dufouil C , Todd S , Wallon D , Love S , Rogaeva E , Gallacher J , StGeorge-Hyslop P , Clarimon J , Lleo A , Bayer A , Tsuang DW , Yu L , Tsolaki M , Bossù P , Spalletta G , Proitsi P , Collinge J , Sorbi S , Sanchez-Garcia F , Fox NC , Hardy J , Deniz Naranjo MC , Bosco P , Clarke R , Brayne C , Galimberti D , Mancuso M , Matthews F , European Alzheimer’s Disease Initiative (EADI); Genetic and Environmental Risk in Alzheimer’s Disease; Alzheimer’s Disease Genetic Consortium; Cohorts for Heart and Aging Research in Genomic Epidemiology; Moebus S , Mecocci P , Del Zompo M , Maier W , Hampel H , Pilotto A , Bullido M , Panza F , Caffarra P , Nacmias B , Gilbert JR , Mayhaus M , Lannefelt L , Hakonarson H , Pichler S , Carrasquillo MM , Ingelsson M , Beekly D , Alvarez V , Zou F , Valladares O , Younkin SG , Coto E , Hamilton-Nelson KL , Gu W , Razquin C , Pastor P , Mateo I , OwenMJ , Faber KM , Jonsson PV , Combarros O , O’Donovan MC , Cantwell LB , Soininen H , Blacker D , Mead S , Mosley TH Jr. , Bennett DA , Harris TB , Fratiglioni L , Holmes C , de Bruijn RF , Passmore P , Montine TJ , Bettens K , Rotter JI , Brice A , Morgan K , Foroud TM , Kukull WA , Hannequin D , Powell JF , Nalls MA , Ritchie K , Lunetta KL , Kauwe JS , Boerwinkle E , Riemenschneider M , Boada M , Hiltuenen M , Martin ER , Schmidt R , Rujescu D , Wang LS , Dartigues JF , Mayeux R , Tzourio C , Hofman A , Nöthen MM , Graff C , Psaty BM , Jones L , Haines JL , Holmans PA , Lathrop M , Pericak-Vance MA , Launer LJ , Farrer LA , vanDuijn CM , Van Broeckhoven C , Moskvina V , Seshadri S , Williams J , Schellenberg GD , Amouyel P ((2013) ) Meta-analysis of 74,046individuals identifies 11 new susceptibility loci for Alzheimer’sdisease.Nat Genet 45: , 1452–1458.

[6] 

Malik M , Parikh I , Vasquez JB , Smith C , Tai L , Bu G , LaDu MJ , Fardo DW , Rebeck GW , Estus S ((2015) ) Genetics ignite focus on microglial inflammation in Alzheimer’s disease.Mol Neurodegener 10: , 52.

[7] 

Naj AC , Jun G , Beecham GW , Wang L-S , Vardarajan BN , Buros J , Gallins PJ , Buxbaum JD , Jarvik GP , Crane PK , Larson EB , Bird TD , Boeve BF , Graff-Radford NR , De Jager PL , Evans D , Schneider JA , Carrasquillo MM , Ertekin-Taner N , Younkin SG , Cruchaga C , Kauwe JSK , Nowotny P , Kramer P , Hardy J , Huentelman MJ , Myers AJ , Barmada MM , Demirci FY , Baldwin CT , Green RC , Rogaeva E , St George-Hyslop P , Arnold SE , Barber R , Beach T , Bigio EH , Bowen JD , Boxer A , Burke JR , Cairns NJ , Carlson CS , Carney RM , Carroll SL , Chui HC , Clark DG , Corneveaux J , Cotman CW , Cummings JL , DeCarli C , DeKosky ST , Diaz-Arrastia R , Dick M , Dickson DW , Ellis WG , Faber KM , Fallon KB , Farlow MR , Ferris S , Frosch MP , Galasko DR , Ganguli M , Gearing M , Geschwind DH , Ghetti B , Gilbert JR , Gilman S , Giordani B , Glass JD , Growdon JH , Hamilton RL , Harrell LE , Head E , Honig LS , Hulette CM , Hyman BT , Jicha GA , Jin L-W , Johnson N , Karlawish J , Karydas A , Kaye JA , Kim R , Koo EH , Kowall NW , Lah JJ , Levey AI , Lieberman AP , Lopez OL , Mack WJ , Marson DC , Martiniuk F , Mash DC , Masliah E , McCormick WC , McCurry SM , McDavid AN , McKee AC , Mesulam M , Miller BL , Miller CA , Miller JW , Parisi JE , Perl DP , Peskind E , Petersen RC , Poon WW , Quinn JF , Rajbhandary RA , Raskind M , Reisberg B , Ringman JM , Roberson ED , Rosenberg RN , Sano M , Schneider LS , Seeley W , Shelanski ML , Slifer MA , Smith CD , Sonnen JA , Spina S , Stern RA , Tanzi RE , Trojanowski JQ , Troncoso JC , Van Deerlin VM , Vinters HV , Vonsattel JP , Weintraub S , Welsh-Bohmer KA , Williamson J , Woltjer RL , Cantwell LB , Dombroski BA , Beekly D , Lunetta KL , Martin ER , Kamboh MI , Saykin AJ , Reiman EM , Bennett DA , Morris JC , Montine TJ , Goate AM , Blacker D , Tsuang DW , Hakonarson H , Kukull WA , Foroud TM , Haines JL , Mayeux R , Pericak-Vance MA , Farrer LA , Schellenberg GD ((2011) ) Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease.Nat Genet 43: , 436–441.

[8] 

Bis JC , Jian X , Kunkle BW , Chen Y , Hamilton-Nelson KL , Bush WS , Salerno WJ , Lancour D , Ma Y , Renton AE , Marcora E , Farrell JJ , Zhao Y , Qu L , Ahmad S , Amin N , Amouyel P , Beecham GW , Below JE , Campion D , Cantwell L , Charbonnier C , Chung J , Crane PK , Cruchaga C , Cupples LA , Dartigues JF , Debette S , Deleuze JF , Fulton L , Gabriel SB , Genin E , Gibbs RA , Goate A , Grenier-Boley B , Gupta N , Haines JL , Havulinna AS , Helisalmi S , Hiltunen M , Howrigan DP , Ikram MA , Kaprio J , Konrad J , Kuzma A , Lander ES , Lathrop M , Lehtimäki T , Lin H , Mattila K , Mayeux R , Muzny DM , Nasser W , Neale B , Nho K , Nicolas G , Patel D , Pericak-Vance MA , Perola M , Psaty BM , Quenez O , Rajabli F , Redon R , Reitz C , Remes AM , Salomaa V , Sarnowski C , Schmidt H , Schmidt M , Schmidt R , Soininen H , Thornton TA , Tosto G , Tzourio C , van der Lee SJ , van Duijn CM , Valladares O , Vardarajan B , Wang LS , Wang W , Wijsman E , Wilson RK , Witten D , Worley KC , Zhang X , Bellenguez C , Lambert JC , Kurki MI , Palotie A , Daly M , Boerwinkle E , Lunetta KL , Destefano AL , Dupuis J , Martin ER , Schellenberg GD , Seshadri S , Naj AC , Fornage M , Farrer LA ((2020) ) Whole exome sequencing study identifies novel rare and common Alzheimer’s-Associated variants involved in immune response and transcriptional regulation.Mol Psychiatry 25: , 1859–1875.

[9] 

Gagliano SA , Pouget JG , Hardy J , Knight J , Barnes MR , Ryten M , Weale ME ((2016) ) Genomics implicates adaptive and innate immunity in Alzheimer’s and Parkinson’s diseases.Ann Clin Transl Neurol 3: , 924–933.

[10] 

Gerring ZF , Gamazon ER , White A , Derks EM ((2021) ) Integrative network-based analysis reveals gene networks and novel drug repositioning candidates for Alzheimer disease.Neurol Genet 7: , e622.

[11] 

Busse S , Hoffmann J , Michler E , Hartig R , Frodl T , Busse M ((2021) ) Dementia-associated changes of immune cell composition within the cerebrospinal fluid.Brain Behav Immun Health 14: , 100218.

[12] 

Gate D , Saligrama N , Leventhal O , Yang AC , Unger MS , Middeldorp J , Chen K , Lehallier B , Channappa D , De Los Santos MB , McBride A , Pluvinage J , Elahi F , Tam GK , Kim Y , Greicius M , Wagner AD , Aigner L , Galasko DR , Davis MM , Wyss-Coray T ((2020) ) Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease.Nature 577: , 399–404.

[13] 

Busse M , Michler E , von Hoff F , Dobrowolny H , Hartig R , Frodl T , Busse S ((2017) ) Alterations in the peripheral immune system in dementia.J Alzheimers Dis 58: , 1303–1313.

[14] 

Richartz-Salzburger E , Batra A , Stransky E , Laske C , Köhler N , Bartels M , Buchkremer G , Schott K ((2007) ) Altered lymphocyte distribution in Alzheimer’s disease.J Psychiatr Res 41: , 174–178.

[15] 

Park JC , Han SH , Mook-Jung I ((2020) ) Peripheral inflammatory biomarkers in Alzheimer’s disease: A brief review.BMB Rep 53: , 10–19.

[16] 

Togo T , Akiyama H , Iseki E , Kondo H , Ikeda K , Kato M , Oda T , Tsuchiya K , Kosaka K ((2002) ) Occurrence of T cells in the brain of Alzheimer’s disease and other neurological diseases.J Neuroimmunol 124: , 83–92.

[17] 

Cao W , Zheng H ((2018) ) Peripheral immune system in aging and Alzheimer’s disease.Mol Neurodegener 13: , 51.

[18] 

Joshi C , Sivaprakasam K , Christley S , Ireland S , Rivas J , Zhang W , Sader D , Logan R , Lambracht-Washington D , Rosenberg R , Cullum M , Hitt B , Li QZ , Barber R , Greenberg B , Cowell L , Zhang R , Stowe A , Huebinger R , Kelley B , Monson N ((2022) ) CSF-derived CD4(+) T-cell diversity is reduced in patients with Alzheimer clinical syndrome.Neurol Neuroimmunol Neuroinflamm 9: ,e1106.

[19] 

Zhang Y , Fung ITH , Sankar P , Chen X , Robison LS , Ye L , D’Souza SS , Salinero AE , Kuentzel ML , Chittur SV , Zhang W , Zuloaga KL , Yang Q ((2020) ) Depletion of NK cells improves cognitive function in the Alzheimer disease mouse model.J Immunol 205: , 502–510.

[20] 

Solana C , Tarazona R , Solana R ((2018) ) Immunosenescence of natural killer cells, inflammation, and Alzheimer’s disease.Int J Alzheimers Dis 2018: , 3128758.

[21] 

Araga S , Kagimoto H , Funamoto K , Takahashi K ((1991) ) Reduced natural killer cell activity in patients with dementia of the Alzheimer type.Acta Neurol Scand 84: , 259–263.

[22] 

Pellicanò M , Larbi A , Goldeck D , Colonna-Romano G , Buffa S , Bulati M , Rubino G , Iemolo F , Candore G , Caruso C , Derhovanessian E , Pawelec G ((2012) ) Immune profiling of Alzheimer patients.J Neuroimmunol 242: , 52–59.

[23] 

Larbi A , Pawelec G , Witkowski JM , Schipper HM , Derhovanessian E , Goldeck D , Fulop T ((2009) ) Dramatic shifts in circulating CD4 but not CD8 T cell subsets in mild Alzheimer’s disease.J Alzheimers Dis 17: , 91–103.

[24] 

Rosenkranz D , Weyer S , Tolosa E , Gaenslen A , Berg D , Leyhe T , Gasser T , Stoltze L ((2007) ) Higher frequency of regulatory T cells in the elderly and increased suppressive activity in neurodegeneration.J Neuroimmunol 188: , 117–127.

[25] 

Saresella M , Calabrese E , Marventano I , Piancone F , Gatti A , Calvo MG , Nemni R , Clerici M ((2010) ) PD1 negative and PD1 positive CD4+T regulatory cells in mild cognitive impairment and Alzheimer’s disease.J Alzheimers Dis 21: , 927–938.

[26] 

Ciccocioppo F , Lanuti P , Pierdomenico L , Simeone P , Bologna G , Ercolino E , Buttari F , Fantozzi R , Thomas A , Onofrj M , Centonze D , Miscia S , Marchisio M ((2019) ) The characterization of regulatory T-cell profiles in Alzheimer’s disease and multiple sclerosis.Sci Rep 9: , 8788.

[27] 

Oberstein TJ , Taha L , Spitzer P , Hellstern J , Herrmann M , Kornhuber J , Maler JM ((2018) ) Imbalance of circulating T(h)17 and regulatory T cells in Alzheimer’s disease: A case control study.Front Immunol 9: , 1213.

[28] 

Illouz T , Madar R , Hirsh T , Biragyn A , Okun E ((2021) ) Induction of an effective anti-Amyloid-β humoral response in aged mice.Vaccine 39: , 4817–4829.

[29] 

Martorana A , Bulati M , Buffa S , Pellicanò M , Caruso C , Candore G , Colonna-Romano G ((2012) ) Immunosenescence, inflammation and Alzheimer’s disease.Longev Healthspan 1: , 8.

[30] 

Tramutola A , Abate G , Lanzillotta C , Triani F , Barone E , Iavarone F , Vincenzoni F , Castagnola M , Marziano M , Memo M , Garrafa E , Butterfield DA , Perluigi M , Di Domenico F , Uberti D ((2018) ) Protein nitration profile of CD3(+) lymphocytes from Alzheimer disease patients: Novel hints on immunosenescence and biomarker detection.Free Radic Biol Med 129: , 430–439.

[31] 

Fried LP , Borhani NO , Enright P , Furberg CD , Gardin JM , Kronmal RA , Kuller LH , Manolio TA , Mittelmark MB , Newman A , O’Leary DH , Psaty B , Rautaharju P , Tracy RP , Weiler PG ((1991) ) The Cardiovascular Health Study: Design and rationale.Ann Epidemiol 1: , 263–276.

[32] 

Olson NC , Sitlani CM , Doyle MF , Huber SA , Landay AL , Tracy RP , Psaty BM , Delaney JA ((2020) ) Innate and adaptive immune cell subsets as risk factors for coronary heart disease in two population-based cohorts.Atherosclerosis 300: , 47–53.

[33] 

Tracy RP , Doyle MF , Olson NC , Huber SA , Jenny NS , Sallam R , Psaty BM , Kronmal RA ((2013) ) T-helper type 1 bias in healthy people is associated with cytomegalovirus serology and atherosclerosis: The Multi-Ethnic Study of Atherosclerosis.J Am Heart Assoc 2: , e000117.

[34] 

Arnold AM , Newman AB , Dermond N , Haan M , Fitzpatrick A ((2009) ) Using telephone and informant assessments to estimate missing Modified Mini-Mental State Exam scores and rates of cognitive decline. The cardiovascular health study.Neuroepidemiology 33: , 55–65.

[35] 

Odden MC , Koh WJH , Arnold AM , Rawlings AM , Psaty BM , Newman AB ((2019) ) Trajectories of nonagenarian health: Sex, age, and period effects.Am J Epidemiol 188: , 382–388.

[36] 

Fitzpatrick AL , Kuller LH , Ives DG , Lopez OL , Jagust W , Breitner JCS , Jones B , Lyketsos C , Dulberg C ((2004) ) Incidence and prevalence of dementia in the Cardiovascular Health Study.J Am Geriatr Soc 52: , 195–204.

[37] 

Kuller LH , Lopez OL , Jagust WJ , Becker JT , DeKosky ST , Lyketsos C , Kawas C , Breitner JCS , Fitzpatrick A , Dulberg C ((2005) ) Determinants of vascular dementia in the Cardiovascular Health Cognition Study.Neurology 64: , 1548–1552.

[38] 

Lopez OL , Kuller LH , Fitzpatrick A , Ives D , Becker JT , Beauchamp N ((2003) ) Evaluation of dementia in the Cardiovascular Health Cognition Study.Neuroepidemiology 22: , 1–12.

[39] 

Newman AB , Sachs MC , Arnold AM , Fried LP , Kronmal R , Cushman M , Psaty BM , Harris TB , Robbins JA , Burke GL , Kuller LH , Lumley T ((2009) ) Total and cause-specific mortality in the cardiovascular health study.J Gerontol A Biol Sci Med Sci 64: , 1251–1261.

[40] 

Diehr P , Williamson J , Burke GL , Psaty BM ((2002) ) The aging and dying processes and the health of older adults.J Clin Epidemiol 55: , 269–278.

[41] 

Diehr PH , Thielke SM , Newman AB , Hirsch C , Tracy R ((2013) ) Decline in health for older adults: Five-year change in 13 key measures of standardized health.J Gerontol A Biol Sci Med Sci 68: , 1059–1067.

[42] 

Firbank MJ , Allan LM , Burton EJ , Barber R , O’Brien JT , Kalaria RN ((2012) ) Neuroimaging predictors of death and dementia in a cohort of older stroke survivors.J Neurol Neurosurg Psychiatry 83: , 263–267.

[43] 

Schulz R , Beach SR , Ives DG , Martire LM , Ariyo AA , Kop WJ ((2000) ) Association between depression and mortality in older adults: The Cardiovascular Health Study.Arch Intern Med 160: , 1761–1768.

[44] 

Fried LP , Kronmal RA , Newman AB , Bild DE , Mittelmark MB , Polak JF , Robbins JA , Gardin JM ((1998) ) Risk factors for 5-year mortality in older adults: The Cardiovascular Health Study.JAMA 279: , 585–592.

[45] 

Robins JM , Finkelstein DM ((2000) ) Correcting for noncompliance and dependent censoring in an AIDS Clinical Trial with inverse probability of censoring weighted (IPCW) log-rank tests.Biometrics 56: , 779–788.

[46] 

Faridar A , Thome AD , Zhao W , Thonhoff JR , Beers DR , Pascual B , Masdeu JC , Appel SH ((2020) ) Restoring regulatory T-cell dysfunction in Alzheimer’s disease through ex vivoexpansion.Brain Commun 2: , fcaa112.

[47] 

Cunningham C , Hennessy E ((2015) ) Co-morbidity and systemic inflammation as drivers of cognitive decline: New experimental models adopting a broader paradigm in dementia research.Alzheimers Res Ther 7: , 33.

[48] 

Xu H , Jia J ((2021) ) Single-cell RNA sequencing of peripheral blood reveals immune cell signatures in Alzheimer’s disease.Front Immunol 12: , 645666.

[49] 

Sommer A , Winner B , Prots I ((2017) ) The Trojan horse - neuroinflammatory impact of T cells in neurodegenerative diseases.Mol Neurodegener 12: , 78.

[50] 

Batchu S ((2020) ) In silico analysis of the immunological landscape of hippocampi in Alzheimer’s disease.Dement Geriatr Cogn Disord 49: , 252–254.

[51] 

Tuppo EE , Arias HR ((2005) ) The role of inflammation in Alzheimer’s disease.Int J Biochem Cell Biol 37: , 289–305.