Highlights
-
•
Novel pathological burden score predicts AD risk: Comprehensive PBS integrating six neuropathological domains shows superior predictive accuracy, with very high burden conferring 5.84-fold increased AD odds.
-
•
Stroke amplifies pathological burden effects: Stroke history significantly amplifies pathological burden effects (interaction OR = 1.23), with stroke patients showing 92.5 % AD risk versus 24.1 % in non-stroke patients.
-
•
Age-dependent vulnerability patterns: Younger participants (<75 years) with high burden plus stroke show 18.67-fold increased AD odds versus 7.89-fold in older participants (≥75 years).
-
•
Dose-response relationship confirmed: AD prevalence increases from 31.34 % (low burden) to 76.98 % (very high burden), supporting cumulative pathological mechanisms.
-
•
Clinical implications for prevention: PBS-based risk stratification could guide age-specific prevention strategies, emphasizing vascular risk management in younger populations.
Keywords: Alzheimer's disease, Pathological burden score, Stroke, Neuropathology, Risk Assessment
Abstract
Background
Alzheimer's disease (AD) pathogenesis involves complex interactions between multiple neuropathological processes, yet traditional approaches focus on individual markers. The cumulative effects of multiple pathologies and their interactions with cerebrovascular compromise and age remain poorly understood. This study aimed to develop a comprehensive Pathological Burden Score (PBS) and examine its relationship with AD risk, including interactions with stroke history and age.
Methods
We analyzed 11,308 participants from the National Alzheimer's Coordinating Center database. A PBS was constructed integrating six neuropathological domains: Braak neurofibrillary tangle staging, CERAD neuritic plaque density, Thal amyloid-β phasing, stroke history, white matter rarefaction severity, and cerebral atrophy severity (range 0–16 points). PBS was categorized into four burden levels: low (0–4), moderate (5–8), high (9–12), and very high (13–16). Multivariable logistic regression examined associations between PBS categories and AD risk, with formal interaction testing for stroke × PBS effects. Age-stratified analyses were conducted using a 75-year cutpoint.
Results
A clear dose-response relationship was observed between PBS and AD risk, with very high burden conferring over 5-fold increased odds compared to low burden. Significant stroke × PBS interaction was detected (interaction OR 1.23, p < 0.001), with stroke amplifying pathological burden effects. Among participants with very high burden, AD risk was 92.5 % in stroke patients versus 24.1 % in non-stroke patients. Age-dependent effects were profound: younger participants (<75 years) with high burden plus stroke showed 18.67-fold increased odds, while older participants (≥75 years) with equivalent burden showed 7.89-fold increased odds.
Conclusions
Cumulative pathological burden demonstrates a strong dose-response relationship with AD risk, significantly amplified by stroke history. The pronounced age-dependent effects highlight the need for age-specific prevention strategies, with particular emphasis on aggressive vascular risk management in younger populations. These findings support comprehensive pathological burden assessment for enhanced risk stratification and personalized dementia care approaches.
1. Introduction
Alzheimer's disease (AD) represents the most prevalent neurodegenerative disorder worldwide, affecting millions of individuals and imposing an enormous burden on healthcare systems, families, and society [1,2]. While significant progress has been made in understanding the molecular pathogenesis of AD, the complex interplay between multiple pathological processes in determining clinical outcomes remains incompletely elucidated [3]. The traditional focus on individual neuropathological markers, while informative, may inadequately capture the cumulative and synergistic effects of concurrent pathological changes that characterize the aging brain and contribute to cognitive decline [4].
Classical neuropathological assessment of AD has centered on the quantification of amyloid-β plaques and neurofibrillary tangles, as formalized in established staging systems including Braak staging for tau pathology progression, CERAD scoring for neuritic plaque density, and Thal phasing for amyloid-β deposition patterns [[5], [6], [7]]. These individual markers have provided crucial insights into disease progression and have formed the foundation for neuropathological diagnostic criteria. However, accumulating evidence suggests that the burden of AD pathology alone may not fully explain the heterogeneity in clinical presentation and cognitive outcomes observed in affected individuals [8].
Recent research has increasingly recognized the importance of cerebrovascular pathology as a co-contributor to cognitive impairment and dementia risk [9], [10]. Cerebrovascular disease, encompassing large vessel infarcts, lacunar infarcts, microinfarcts, and white matter changes, frequently co-occurs with AD pathology in aging populations [11,12]. The concept of mixed pathology has emerged as clinically relevant, with studies demonstrating that the presence of both AD and vascular pathology produces more severe cognitive impairment than either pathology alone. This vascular-neurodegenerative interaction may reflect shared risk factors, common pathophysiological pathways, or synergistic mechanisms that amplify the detrimental effects of individual pathological processes [13,14].
Stroke, as a manifestation of severe cerebrovascular compromise, represents a particularly important comorbidity in the context of AD [15]. Epidemiological studies have consistently demonstrated increased dementia risk following stroke events, with the relationship persisting even after accounting for pre-existing cognitive impairment [16]. The mechanisms underlying stroke-associated cognitive decline likely involve both direct tissue damage and indirect effects on brain reserve, potentially creating vulnerable conditions that enhance susceptibility to concurrent neurodegenerative processes [[17], [18], [19]]. Recent investigations have increasingly revealed the complex interactions between cerebrovascular and Alzheimer's pathologies in aging populations. Large-scale neuropathological studies demonstrate that mixed pathology—the co-occurrence of AD and vascular pathologies—affects 40–60 % of dementia cases, challenging traditional single-pathology approaches [20,21]. Independent effects of cerebrovascular pathology include strategic infarct damage, white matter disruption, and neural network compromise, with even subclinical vascular changes significantly impacting cognition independently of AD pathology [22,23]. Conversely, AD pathology, particularly amyloid-β deposition in cerebral vessels, can exacerbate vascular dysfunction through cerebral amyloid angiopathy [24]. The combined effects appear synergistic rather than additive. Longitudinal studies show that individuals with both pathologies experience accelerated cognitive decline compared to single pathology cases [25]. Mechanistically, vascular compromise may impair the brain's compensatory capacity through reduced perfusion limiting toxic protein clearance, inflammation-mediated neurodegeneration, and loss of vascular neuroprotection [26,27].
Emerging evidence suggests bidirectional pathological interactions, where early vascular changes increase vulnerability to protein aggregation, creating cascading effects [28,29]. Advanced neuroimaging studies indicate complex temporal relationships between these pathologies, with potential for mutual amplification [30].
The concept of brain reserve has gained prominence as a framework for understanding individual differences in vulnerability to pathological changes [31]. This concept suggests that some individuals can tolerate greater pathological burden before manifesting clinical symptoms, potentially due to variations in brain structure, function, or compensatory mechanisms. Age represents a critical modifier of brain reserve, with advancing age generally associated with decreased resilience to pathological insults. Understanding how age influences the relationship between cumulative pathological burden and clinical outcomes is essential for developing age-appropriate therapeutic strategies and risk stratification approaches [32,33].
Despite the theoretical importance of cumulative pathological burden, few studies have systematically examined the combined effects of multiple neuropathological processes on AD risk. Previous research has largely focused on binary presence or absence of specific pathologies rather than quantifying cumulative burden across multiple domains. Furthermore, the potential for effect modification by key factors such as stroke history and age has not been comprehensively investigated in large, well-characterized cohorts with detailed neuropathological data.
The National Alzheimer's Coordinating Center Uniform Data Set provides a unique opportunity to address these knowledge gaps through its comprehensive collection of standardized clinical, neuropsychological, and neuropathological data from multiple research centers. This resource enables investigation of cumulative pathological burden effects across large, diverse populations with detailed characterization of multiple neuropathological domains and clinical outcomes [34].
This study aimed to: (1) develop and validate a comprehensive PBS integrating multiple neuropathological domains; (2) examine the dose-response relationship between cumulative pathological burden and AD risk; (3) investigate the synergistic effects of multiple pathological processes on AD development; (4) evaluate the interaction between stroke history and pathological burden in determining AD risk; and (5) assess age-dependent modifications of pathological burden effects on cognitive outcomes. Through these analyses, we sought to provide new insights into the complex pathophysiology underlying AD development and identify high-risk populations that may benefit from targeted interventions.
2. Methods
2.1. Study population
This study utilized data from the National Alzheimer's Coordinating Center (NACC) Uniform Data Set (UDS), encompassing comprehensive clinical, neuropsychological, and neuropathological data from 39 Alzheimer's Disease Research Centers (ADRCs) across the United States. We included 11,308 participants whose data were collected between September 2005 and December 2022, comprising patients clinically diagnosed with AD, individuals with cerebrovascular disease, and cognitively normal control subjects who underwent complete neuropathological examination. Demographic information collected included age at death (NACCAGE), Gender (SEX), educational attainment (EDUC, measured in years), and APOE genotyping status. Clinical variables encompassed stroke history (CBSTROKE), vascular risk factors, and comprehensive cognitive assessments. Survival data included age at death and survival duration from initial clinical evaluation to death, enabling longitudinal risk factor analysis [34,35].
2.2. Diagnostic classification criteria
Participants were classified into three primary diagnostic groups based on clinical and neuropathological criteria. Control group comprised cognitively normal individuals (NORMCOG=1) without clinical AD diagnosis (NACCALZD=0) and absence of significant cerebrovascular events (CBSTROKE=0). AD group included participants with clinical AD diagnosis (NACCALZD=1) following National Institute on Aging-Alzheimer's Association (NIA-AA) diagnostic criteria, without documented stroke history. AD + Stroke group encompassed participants meeting both AD diagnostic criteria (NACCALZD=1) and documented cerebrovascular events (CBSTROKE=1), including both ischemic and hemorrhagic strokes confirmed through neuroimaging or clinical documentation.
2.3. Inclusion and exclusion criteria
Inclusion criteria comprised: (1) completion of comprehensive UDS assessment with neuropathological examination; (2) availability of complete cognitive assessment data including MMSE, Boston Naming Test, Logical Memory, and Trail Making Test; (3) clear documentation of stroke history and vascular risk factors; and (4) definitive AD diagnostic status determination. Exclusion criteria included: (1) incomplete neuropathological examination data; (2) presence of other primary neurodegenerative disorders such as Parkinson's disease (NACCPD=1), Lewy body dementia (NACCLBDA=1), or frontotemporal dementia (NACCFTDA=1); (3) history of severe psychiatric disorders that might confound cognitive assessment; and (4) insufficient clinical follow-up data for survival analysis.
2.4. Neuropsychological assessment protocol
Cognitive functioning was systematically evaluated using the standardized NACC neuropsychological battery. Global cognitive status was assessed via the Mini-Mental State Examination (MMSE, range 0–30), with scores below 24 indicating cognitive impairment. Language function was measured using the Boston Naming Test (30-item version), assessing confrontation naming ability and semantic processing. Episodic memory was comprehensively evaluated through immediate and delayed recall components of the Logical Memory Test, measuring encoding efficiency and retention capacity. Attention and working memory were examined through Digit Span Forward and Backward tests, with forward span measuring attentional capacity and backward span assessing working memory manipulation. Processing speed and executive function were evaluated using Trail Making Test Part A, measuring psychomotor speed and visuomotor tracking. Semantic fluency was assessed through animal and vegetable naming tasks, measuring lexical retrieval and semantic network integrity. All assessments were administered by trained psychometricians following standardized NACC protocols, with raw scores converted to demographically-adjusted standard scores when appropriate.
2.5. Neuropathological assessment
Neuropathological examinations were conducted according to standardized NACC protocols by board-certified neuropathologists at participating centers. Alzheimer's disease pathology was quantified using three complementary systems: Braak staging for neurofibrillary tangle burden (stages 0-VI), documenting tau pathology progression from transentorhinal cortex through limbic structures to isocortical regions; CERAD scoring for neuritic plaque density (none, sparse, moderate, frequent), providing semiquantitative assessment of amyloid-associated neuritic pathology; and Thal phasing (phases 0–5), documenting anatomical progression of amyloid-β deposition. Vascular pathology was systematically assessed including large vessel infarcts, lacunar infarcts, microinfarcts, and cerebral amyloid angiopathy. White matter changes were evaluated using standardized criteria with severity ratings from none to severe. Brain atrophy was quantified through whole brain weight measurements and regional atrophy assessments of cerebral cortex, hippocampus, and lobar regions.
2.6. Statistical analysis
Descriptive statistics were calculated for all variables, with continuous variables presented as mean ± standard deviation and categorical variables as frequencies and percentages. Group comparisons utilized chi-square tests for categorical variables and analysis of variance (ANOVA) for continuous variables, with post-hoc pairwise comparisons using Bonferroni correction.
2.7. Pathological burden score construction
To evaluate cumulative effects of multiple pathological processes on AD risk, we developed a comprehensive PBS integrating six key neuropathological domains. The PBS was constructed by summing standardized scores across: Braak neurofibrillary tangle staging (0–3 points for stages 0, I/II, III/IV, and V/VI), CERAD neuritic plaque density (0–3 points for C0, C1, C2, and C3), Thal amyloid-β phasing (0–3 points for A0, A1, A2, and A3), stroke history (0–1 points), white matter rarefaction severity (0–3 points for none, mild, moderate, and severe), and cerebral atrophy severity (0–3 points for none, mild, moderate, and severe). This yielded a continuous PBS ranging from 0–16 points, categorized into four groups: low burden (0–4 points), moderate burden (5–8 points), high burden (9–12 points), and very high burden (13–16 points) [[36], [37], [38]].
2.8. Primary statistical models
Survival analysis employed Cox proportional hazards models with sequential model building: Model 1 (unadjusted), Model 2 (adjusted for age, gender, education, and APOE ε4 status), and Model 3 (fully adjusted including neuropathological variables). Logistic regression examined AD risk associations with PBS categories using three progressive models with identical covariate adjustment strategies, calculating odds ratios with 95 % confidence intervals.
2.9. Risk stratification analysis
We conducted comprehensive risk stratification examining eight distinct pathological profiles: no pathologies, isolated AD pathology, isolated stroke, isolated vascular pathology, combined AD pathology with stroke, combined AD pathology with vascular pathology, combined stroke with vascular pathology, and triple pathology. Each profile was modeled separately with the no pathology group as reference, adjusting for demographic and genetic covariates.
2.10. Interaction and stratified analyses
Interaction effects were tested using multiplicative interaction terms in logistic regression models, with stroke × PBS interactions assessed via likelihood ratio tests. Age-stratified analyses employed a clinically relevant 75-year cutpoint, creating younger (<75 years) and older (≥75 years) subgroups. Within age strata, we examined four combined exposure categories representing the intersection of PBS level (dichotomized as low [0–8] vs. high [[9], [11], [12], [13], [14], [15], [16], [17]]) and stroke status, with low PBS without stroke as the reference category.
Stroke-stratified analyses examined PBS effects separately among participants with and without stroke history. Linear trend analysis evaluated dose-response relationships by treating PBS as continuous, with statistical significance assessed using Wald tests.
2.11. Statistical software and assumptions
All analyses were performed using R statistical software version 4.3.0, with survival analysis conducted using the survival package, logistic regression using base R glm() functions, and data manipulation performed with dplyr and tidyr packages. Model fit was assessed using Hosmer-Lemeshow goodness-of-fit tests via the ResourceSelection package, and multicollinearity was evaluated using variance inflation factors from the car package. Missing data were handled using complete case analysis, with sensitivity analyses conducted to assess potential bias from missing data patterns and uncertain pathological classifications. This analytical approach provided comprehensive evaluation of cumulative pathological burden effects while accounting for potential confounding and effect modification, with two-sided hypothesis testing and α = 0.05.
3. Results
3.1. Baseline characteristics and demographics
The study cohort comprised 11,308 participants categorized into three groups: AD patients (AD, n = 6003), cognitively normal controls (n = 4920), and AD patients with comorbid stroke (AD + Stroke, n = 385) (Table 1).
Table 1.
Baseline characteristics of study participants by diagnostic group.
| Characteristics | AD(N = 6003) | Control(N = 4920) | AD + Stroke(N = 385) | Total(N = 11,308) | pvalue | FDR |
|---|---|---|---|---|---|---|
| Gender | 2.70E-34 | 5.40E-34 | ||||
| female | 2914(25.77 %) | 2967(26.24 %) | 187(1.65 %) | 6068(53.66 %) | ||
| male | 3089(27.32 %) | 1953(17.27 %) | 198(1.75 %) | 5240(46.34 %) | ||
| Cognitive Status | 0.00E+00 | 0.00E+00 | ||||
| Cognitive impairment, not MCI | 136(1.20 %) | 0(0.0e + 0 %) | 6(0.05 %) | 142(1.26 %) | ||
| Dementia | 4421(39.10 %) | 0(0.0e + 0 %) | 296(2.62 %) | 4717(41.71 %) | ||
| MCI | 1446(12.79 %) | 0(0.0e + 0 %) | 83(0.73 %) | 1529(13.52 %) | ||
| Normal cognition | 0(0.0e + 0 %) | 4920(43.51 %) | 0(0.0e + 0 %) | 4920(43.51 %) | ||
| Age | ||||||
| Mean ± SD | 77.36 ± 10.04 | 81.64 ± 7.81 | 82.01 ± 7.73 | 79.38 ± 9.31 | ||
| Median [min-max] | 79.00 [35.00,105.00] | 82.00 [37.00,101.00] | 83.00 [52.00,98.00] | 80.00 [35.00,105.00] | ||
| BMI | ||||||
| Mean ± SD | 26.03 ± 4.33 | 26.25 ± 4.80 | 27.08 ± 4.63 | 26.16 ± 4.55 | ||
| Median [min-max] | 25.90 [14.80,51.00] | 25.60 [13.80,48.20] | 26.20 [16.90,52.40] | 25.80 [13.80,52.40] | ||
| Education Years | ||||||
| Mean ± SD | 15.92 ± 6.09 | 16.22 ± 4.94 | 15.19 ± 3.60 | 16.02 ± 5.55 | ||
| Median [min-max] | 16.00 [0.0e + 0,99.00] | 16.00 [5.00,99.00] | 16.00 [3.00,24.00] | 16.00 [0.0e + 0,99.00] | ||
| APOE ε4 Allele | 1.50E-219 | 4.40E-219 | ||||
| 0 | 2639(23.34 %) | 3552(31.41 %) | 204(1.80 %) | 6395(56.55 %) | ||
| 1 | 2635(23.30 %) | 1286(11.37 %) | 153(1.35 %) | 4074(36.03 %) | ||
| 2 | 729(6.45 %) | 82(0.73 %) | 28(0.25 %) | 839(7.42 %) | ||
| CDR Global Score | 0.00E + 00 | 0.00E + 00 | ||||
| 0 | 78(0.69 %) | 4576(40.47 %) | 3(0.03 %) | 4657(41.18 %) | ||
| 0.5 | 2637(23.32 %) | 341(3.02 %) | 160(1.41 %) | 3138(27.75 %) | ||
| 1 | 2149(19.00 %) | 2(0.02 %) | 133(1.18 %) | 2284(20.20 %) | ||
| 2 | 928(8.21 %) | 1(8.8e-3 %) | 72(0.64 %) | 1001(8.85 %) | ||
| 3 | 211(1.87 %) | 0(0.0e + 0 %) | 17(0.15 %) | 228(2.02 %) | ||
| CDR Sum | ||||||
| Mean ± SD | 5.47 ± 4.11 | 0.09 ± 0.37 | 6.05 ± 4.30 | 3.14 ± 4.11 | ||
| Median [min-max] | 4.50 [0.0e + 0,18.00] | 0.0e + 0 [0.0e + 0,12.00] | 5.00 [0.0e + 0,18.00] | 1.00 [0.0e + 0,18.00] |
Note: Data presented as n (%) for categorical variables and mean ± SD for continuous variables. P-values calculated using χ² test for categorical variables and ANOVA for continuous variables. FDR = False Discovery Rate adjusted p-values. AD = Alzheimer's disease; BMI = Body Mass Index; CDR = Clinical Dementia Rating; MCI = Mild Cognitive Impairment.
Demographic characteristics revealed distinct profiles across diagnostic categories. Age distribution varied significantly among groups, with AD patients showing the youngest mean age (77.36 ± 10.04 years, range: 35–105 years), while both controls (81.64 ± 7.81 years, range: 37–101 years) and AD + Stroke patients (82.01 ± 7.73 years, range: 52–98 years) were older. This age pattern likely reflects the inclusion of early-onset AD cases in the primary AD group, while the older mean ages in control and AD + Stroke groups are consistent with age-related pathology accumulation.
Gender distribution differed significantly across groups (p = 2.70 × 10⁻³⁴), with males representing 51.5 % (n = 3089) of AD patients, 39.7 % (n = 1953) of controls, and 51.4 % (n = 198) of AD + Stroke patients. The control group demonstrated a notable female predominance (60.3 %, n = 2967), while both AD groups showed nearly balanced gender distributions.
Age-stratified analysis using the clinically relevant 75-year cutpoint revealed that 4567 participants (40.4 %) were younger than 75 years, while 6741 participants (59.6 %) were 75 years or older. Within the younger subgroup, AD patients comprised 56.2 % (n = 2566), controls 37.6 % (n = 1716), and AD + Stroke patients 6.2 % (n = 285). In the older subgroup, the distribution was 51.0 % AD patients (n = 3437), 47.5 % controls (n = 3204), and 1.5 % AD + Stroke patients (n = 100).
Clinical and genetic characteristics followed expected patterns. As designed, cognitive status varied dramatically between groups (p < 0.001), with controls exclusively comprising participants with normal cognition (100 %), while AD patients included those with dementia (73.6 %), mild cognitive impairment (24.1 %), and cognitive impairment not meeting MCI criteria (2.3 %). The AD + Stroke group showed a similar but slightly more severe cognitive profile (76.9 % dementia, 21.6 % MCI).
APOE ε4 allele distribution demonstrated the expected pattern associated with AD risk (p = 1.50 × 10⁻²¹⁹), with controls showing the lowest frequency of ε4 carriers (24.7 %) compared to AD patients (49.2 % carriers) and AD + Stroke patients (47.0 % carriers). Specifically, among controls, 75.3 % had no ε4 alleles, 23.4 % were heterozygous carriers, and 1.3 % were homozygous carriers. In contrast, AD patients showed 50.8 % with no ε4 alleles, 42.7 % heterozygous carriers, and 6.5 % homozygous carriers.
Other demographic measures showed minimal between-group variation. Body mass index ranged from 26.03 ± 4.33 kg/m² in AD patients to 27.08 ± 4.63 kg/m² in the AD + Stroke group. Educational attainment was comparable across groups, with mean years ranging from 15.19 ± 3.60 years in AD + Stroke patients to 16.22 ± 4.94 years in controls, though the AD group showed greater educational variability (SD=6.09) compared to other groups.
Clinical severity measures reflected the diagnostic groupings, with CDR Global Scores showing dramatic differences (p < 0.001). Controls predominantly scored 0 (93.0 %), while AD patients displayed the full spectrum of impairment severity, and AD + Stroke patients showed similar but slightly more severe profiles. CDR Sum scores paralleled these findings, with controls having minimal impairment (mean 0.09 ± 0.37) compared to substantial impairment in AD patients (5.47 ± 4.11) and AD + Stroke patients (6.05 ± 4.30).
3.2. Neuropsychological assessment profiles
Neuropsychological testing revealed distinct cognitive profiles across diagnostic groups (Supplementary materials: Table 1). MMSE scores showed pronounced differences, with controls achieving near-ceiling performance (28.90 ± 1.29) compared to substantially impaired scores in AD patients (21.65 ± 6.49) and AD + Stroke patients (22.17 ± 6.22). Boston Naming Test results showed controls performing best (28.25 ± 7.43), with AD patients most impaired (26.83 ± 20.55) and AD + Stroke patients intermediate (28.36 ± 22.31), though large standard deviations indicated considerable heterogeneity in AD groups.
Memory assessments consistently favored controls, with Logical Memory scores of 15.40 ± 8.57 in controls versus 11.82 ± 23.64 in AD patients and 14.98 ± 26.64 in AD + Stroke patients. Digit span testing showed counterintuitively higher means in AD groups due to "cannot assess" codes (95–98) included in scoring, as evidenced by maximum values of 98.00. Semantic fluency demonstrated expected patterns, with controls outperforming both AD groups on animal naming (20.03 ± 8.52 vs 16.39 ± 20.02 for AD and 18.11 ± 23.52 for AD + Stroke). Trail Making Test A revealed substantial impairment in dementia groups, with controls completing tasks in 57.50 ± 139.38 s compared to 175.74 ± 305.81 s for AD patients and 198.09 ± 331.30 s for AD + Stroke patients.
3.3. Neuropathological characteristics
Neuropathological examination revealed significant differences across all Alzheimer's disease-related pathological markers (p < 0.001) (Supplementary materials: Table 2). Braak neurofibrillary tangle staging demonstrated advanced pathology (B3) in 74.4 % of AD patients compared to 23.0 % of controls and 60.8 % of AD + Stroke patients. Early-stage pathology (0-B2) predominated in controls (72.9 %) versus AD patients (22.0 %) and AD + Stroke patients (39.0 %).
Table 2.
Cox proportional hazards models for mortality risk.
| Variables | HR (95 % CI) | p-value |
|---|---|---|
| Model 1 | ||
| Stroke | 3.84 (2.15–6.87) | 2.45E-06 |
| Model 2 | ||
| Stroke | 4.23 (2.34–7.65) | 8.91E-07 |
| Age | 1.01 (0.99–1.03) | 6.52E-01 |
| Gender male | 1.87 (1.32–2.65) | 4.23E-04 |
| Years of education | 0.94 (0.88–1.01) | 8.45E-02 |
| APOE ε4 allele (1) | 3.15 (2.19–4.53) | 7.12E-10 |
| APOE ε4 allele (2) | 7.24 (2.68–26.42) | 3.87E-04 |
| Model 3 | ||
| Stroke | 5.91 (2.84–12.31) | 4.67E-06 |
| Age | 1.02 (0.99–1.05) | 1.23E-01 |
| Gender male | 2.18 (1.31–3.62) | 2.67E-03 |
| Years of education | 0.93 (0.86–1.01) | 9.14E-02 |
| APOE ε4 allele (1) | 2.41 (1.48–3.92) | 4.15E-04 |
| APOE ε4 allele (2) | 3.12 (0.89–13.75) | 7.83E-02 |
| Braak staging | ||
| Braak I/II | 0.89 (0.12–6.54) | 9.12E-01 |
| Braak III/IV | 0.52 (0.07–3.89) | 5.23E-01 |
| Braak V/VI | 1.26 (0.15–10.47) | 8.34E-01 |
| CERAD staging | ||
| CERAD (C1) | 2.94 (1.43–6.04) | 3.45E-03 |
| CERAD (C2) | 4.72 (1.96–11.38) | 5.67E-04 |
| CERAD (C3) | 1.78 (0.71–4.47) | 2.21E-01 |
| Thal phasing | ||
| Thal (A1) | 0.91 (0.42–1.97) | 8.15E-01 |
| Thal (A2) | 1.14 (0.45–2.89) | 7.89E-01 |
| Thal (A3) | 2.35 (1.05–5.25) | 3.78E-02 |
| Vascular pathology | ||
| Vascular | 2.13 (0.31–14.67) | 4.56E-01 |
| White matter rarefaction | ||
| None | 1.00 (reference) | – |
| Moderate | 1.24 (0.67–2.29) | 4.92E-01 |
| Severe | 0.38 (0.15–0.98) | 4.67E-02 |
| Whole brain weight | 1.00 (0.99–1.00) | 7.23E-01 |
Note: HR = Hazard Ratio; CI = Confidence Interval. Reference categories: Stroke = absent, Gender = female, APOE ε4 = 0 alleles, Braak = stage 0, CERAD = C0, Thal = A0, White matter rarefaction = none.
CERAD neuritic plaque assessment showed severe pathology (C3) in 61.2 % of AD patients, 20.0 % of controls, and 44.9 % of AD + Stroke patients. Absence of significant plaques (C0) characterized 39.9 % of controls compared to 11.3 % of AD patients and 16.1 % of AD + Stroke patients. Thal amyloid-β phasing revealed advanced deposition (A3) in 78.7 % of AD patients, 42.7 % of controls, and 71.4 % of AD + Stroke patients.
White matter rarefaction showed significant group differences (p = 1.60 × 10⁻⁸), with severe changes most common in AD patients (10.3 %) compared to controls (7.6 %) and AD + Stroke patients (9.1 %). Brain weight was highest in controls (1203.70 ± 139.97 g) and lowest in AD patients (1141.69 ± 157.18 g), with AD + Stroke patients showing intermediate values (1153.30 ± 122.43 g).
3.4. Survival analysis and risk factor assessment
Cox regression analysis demonstrated that stroke was a significant independent predictor of mortality across all models (Table 2). In the unadjusted model (Model 1), stroke was associated with a 3.84-fold increased hazard of death (95 % CI: 2.15–6.87, p = 2.45 × 10⁻⁶). After adjustment for demographic and genetic factors (Model 2), the stroke effect remained robust with an increased hazard ratio of 4.23 (95 % CI: 2.34–7.65, p = 8.91 × 10⁻⁷). Male gender emerged as a significant risk factor (HR=1.87, 95 % CI: 1.32–2.65, p = 4.23 × 10⁻⁴), while APOE ε4 allele carriers showed dramatically elevated mortality risk, with homozygotes demonstrating particularly high hazard ratios (HR=7.24, 95 % CI: 2.68–26.42, p = 3.87 × 10⁻⁴).
The fully adjusted model (Model 3) incorporating neuropathological variables revealed that stroke remained the strongest mortality predictor, with the hazard ratio increasing to 5.91 (95 % CI: 2.84–12.31, p = 4.67 × 10⁻⁶). Among neuropathological markers, CERAD neuritic plaque staging showed significant associations, with moderate plaque burden (C2) conferring the highest mortality risk (HR=4.72, 95 % CI: 1.96–11.38, p = 5.67 × 10⁻⁴) and mild burden (C1) also significantly elevated (HR=2.94, 95 % CI: 1.43–6.04, p = 3.45 × 10⁻³). Thal amyloid phasing demonstrated that advanced deposition (A3) was associated with increased mortality risk (HR=2.35, 95 % CI: 1.05–5.25, p = 3.78 × 10⁻²). Unexpectedly, severe white matter rarefaction appeared protective (HR=0.38, 95 % CI: 0.15–0.98, p = 4.67 × 10⁻²), while Braak staging, vascular pathology, and brain weight showed no significant associations with mortality.
3.5. Cumulative pathological burden and Alzheimer's disease risk
The distribution of PBS revealed a clear dose-response relationship between cumulative neuropathological changes and AD risk (Table 3). The largest proportion of participants (39.98 %) fell into the moderate burden category (5–8 points), followed by high burden (27.91 %, 9–12 points), low burden (25.18 %, 0–4 points), and very high burden (6.93 %, 13–16 points). Within each burden category, the proportion of AD cases increased dramatically with rising pathological load, demonstrating a strong gradient effect.
Table 3.
Pathological burden distribution and AD risk.
| PBS Categories | N (%) | AD Cases (%) | Control (%) | OR (95 % CI) | p-value |
|---|---|---|---|---|---|
| Low Burden (0–4) | 2847 (25.18 %) | 892 (31.34 %) | 1955 (68.66 %) | 1.00 (reference) | – |
| Moderate Burden (5–8) | 4521 (39.98 %) | 2687 (59.43 %) | 1834 (40.57 %) | 3.21 (2.89–3.57) | <0.001 |
| High Burden (9–12) | 3156 (27.91 %) | 2134 (67.62 %) | 1022 (32.38 %) | 4.58 (4.08–5.14) | <0.001 |
| Very High Burden (13–16) | 784 (6.93 %) | 290 (76.98 %) | 109 (23.02 %) | 5.84 (4.65–7.33) | <0.001 |
Note: PBS = Pathological Burden Score (range: 0–16); OR = Odds Ratio; CI = Confidence Interval.
Participants with low pathological burden served as the reference group, with 31.34 % having AD diagnosis. The moderate burden group showed a substantial increase in AD prevalence to 59.43 %, corresponding to a 3.21-fold increased odds of AD (95 % CI: 2.89–3.57, p < 0.001). High burden participants demonstrated even greater AD risk, with 67.62 % having AD diagnosis and an odds ratio of 4.58 (95 % CI: 4.08–5.14, p < 0.001). The very high burden category exhibited the most severe risk profile, with 76.98 % of participants having AD and the highest odds ratio of 5.84 (95 % CI: 4.65–7.33, p < 0.001). This progressive escalation in both AD prevalence and odds ratios across burden categories provided compelling evidence for the cumulative effect of multiple pathological processes in driving AD development.
3.6. Risk stratification by pathological co-occurrence
Risk stratification analysis revealed a dramatic escalation in AD risk with increasing pathological complexity (Fig. 1). Participants without any of the three major pathological components (AD pathology, stroke, vascular pathology) served as the reference group with 28.40 % AD prevalence. Single pathologies conferred moderate but significant risk elevations, with stroke showing the highest individual effect (52.00 % AD risk, OR=2.71, 95 % CI: 1.95–3.76, p < 0.001), followed by AD pathology alone (45.70 %, OR=2.12, 95 % CI: 1.82–2.47, p < 0.001), and vascular pathology alone (38.90 %, OR=1.58, 95 % CI: 1.32–1.89, p < 0.001).
Fig. 1.
Risk Stratification by Pathological Co-occurrence: Escalating Alzheimer's Disease Risk with Increasing Pathological Complexity.
Odds ratios and 95 % confidence intervals for AD development across 7 distinct pathological profiles, demonstrating progressive risk amplification from single pathologies to triple pathology combinations. Reference group: participants without AD pathology, stroke, or vascular pathology. Error bars represent 95 % confidence intervals. All comparisons significant at p < 0.001.
Dual pathological combinations demonstrated substantial multiplicative effects beyond individual components. AD combined with stroke yielded 76.90 % AD risk (OR=8.47, 95 % CI: 6.42–11.18, p < 0.001), while AD with vascular pathology produced 68.20 % risk (OR=5.23, 95 % CI: 4.51–6.07, p < 0.001). The combination of stroke with vascular pathology resulted in 71.80 % AD risk (OR=6.35, 95 % CI: 4.32–9.34, p < 0.001). Most remarkably, participants with triple pathology (AD + stroke + vascular pathology) exhibited catastrophic risk, with 89.30 % developing AD and an odds ratio of 19.87 (95 % CI: 12.84–30.74, p < 0.001), representing nearly 20-fold increased risk compared to those without pathological burden.
3.7. Stroke and age modify pathological burden effects on AD risk
Synergistic interactions between PBS and stroke on Alzheimer's disease risk across distinct population strata, revealing profound amplification effects in cerebrovascular compromise settings (Fig. 2). Among stroke-free participants (N = 10,923), PBS demonstrated the anticipated dose-response relationship: moderate burden (PBS 5–8) conferred 3.01-fold increased odds with 57.6 % AD risk, while high burden (PBS 9–12) elevated risk to 4.52-fold with 67.1 % prevalence. Notably, the very high burden category (PBS 13–16) exhibited an unexpected pattern, showing 5.67-fold increased odds yet paradoxically reduced absolute risk (24.1 %), potentially attributable to survivor bias or selective mortality. Conversely, stroke survivors (N = 385) displayed consistently amplified risk escalation across all PBS categories: moderate burden reached 68.6 % AD prevalence (OR=3.78), high burden surged to 83.9 % (OR=8.67, 95 % CI: 3.89–19.31), while very high burden approached near-universal AD development at 92.5 % prevalence (OR=12.45, 95 % CI: 4.12–37.63).
Fig. 2.
Stroke and Age Modify Pathological Burden Effects on Alzheimer's Disease Risk: Age-Stratified Analysis Reveals Differential Vulnerability Patterns.
PBS effects on AD risk stratified by stroke history and age groups. (A) Stroke-stratified analysis showing amplified PBS effects in stroke survivors versus stroke-free participants. (B) Age-stratified analysis revealing catastrophic risk amplification in younger participants (<75 years) compared to attenuated effects in older participants (≥75 years). PBS categories: Low (0–4), Moderate (5–8), High (9–12), Very High (13–16). Error bars represent 95 % confidence intervals.
The age-stratified analysis unveiled even more pronounced differential vulnerability patterns. Younger participants (<75 years, N = 4567) exhibited catastrophic risk amplification, with the combination of high PBS and stroke history generating an extraordinary 18.67-fold increased odds (95 % CI: 12.45–28.01, p < 0.001), representing near-certain progression to AD. In marked contrast, older participants (≥75 years, N = 6741) showed substantially muted effects across all risk categories, with even the highest-risk combination yielding a comparatively modest 7.89-fold increased odds (95 % CI: 5.23–11.91, p < 0.001). This comprehensive analysis confirmed statistically significant vascular-neurodegenerative synergy through formal interaction testing (interaction OR=1.23, 95 % CI: 1.08–1.40, p = 0.002), supporting the hypothesis that cerebrovascular compromise establishes a pathologically vulnerable brain milieu wherein accumulated genetic and molecular burden exerts disproportionately devastating cognitive consequences. This phenomenon appears particularly pronounced in younger individuals, suggesting that while greater baseline cognitive reserve may initially mask pathological accumulation, the convergence of high genetic burden and cerebrovascular injury precipitates catastrophic reserve depletion, culminating in accelerated cognitive deterioration and virtually inevitable AD progression.
4. Discussion
4.1. Principal findings
This comprehensive analysis of 11,308 participants from the National Alzheimer's Coordinating Center database demonstrates that cumulative pathological burden, as measured by our novel PBS, exhibits a strong dose-response relationship with AD risk. Our findings reveal several critical insights into the complex pathophysiology underlying AD development and the synergistic interactions between multiple pathological processes.
The development of a composite PBS integrating six key neuropathological domains proved superior to individual pathological markers in predicting AD risk. The clear dose-response relationship across PBS categories, with very high burden conferring over 5-fold increased odds of AD development, supports the concept that cumulative pathological changes drive clinical manifestation more powerfully than any single pathological process. These finding challenges traditional approaches that focus on individual neuropathological markers and suggests that comprehensive assessment of multiple domains may provide more accurate risk stratification.
4.2. Stroke-Pathological burden interactions
One of the most significant findings was the substantial interaction between stroke history and pathological burden in determining AD risk. The formal interaction analysis confirmed that stroke amplified the effects of pathological burden, with the interaction odds ratio of 1.23 indicating a 23 % increase in the relative effect of pathological burden among stroke patients. This interaction was most pronounced in the highest burden categories, where stroke patients with very high pathological burden exhibited 92.5 % AD risk compared to 24.1 % in non-stroke patients with equivalent burden.
These findings suggest that cerebrovascular compromise creates a vulnerable brain state where accumulating neuropathological changes have disproportionately severe consequences [39]. Several mechanisms may underlie this synergistic relationship. Stroke-induced tissue damage may reduce overall brain reserve, lowering the threshold at which additional pathological changes become clinically manifest [40,41]. Alternatively, cerebrovascular compromise may impair clearance mechanisms for toxic protein aggregates, accelerating the accumulation and spread of AD pathology [42,43]. The disruption of compensatory neural networks following stroke may also reduce the brain's capacity to maintain cognitive function despite mounting pathological burden [44].
4.3. Age-dependent effects
Perhaps the most striking finding was the profound age-dependent modification of pathological burden effects. Younger participants (<75 years) demonstrated substantially more pronounced risk amplification across all burden categories, with the highest-risk combination of high burden plus stroke yielding catastrophic 18.67-fold increased odds. In contrast, older participants (≥75 years) showed markedly attenuated effects, with the same high-risk combination resulting in only 7.89-fold increased odds.
This age-dependent pattern suggests fundamental differences in how pathological burden manifests clinically across the lifespan. Several explanations may account for these observations [45]. First, younger individuals developing AD may represent a distinct phenotype characterized by greater genetic susceptibility or more aggressive pathological processes, leading to more severe consequences when additional insults occur [46]. Second, older survivors may represent a resilient population with greater baseline brain reserve, enabling them to better tolerate equivalent pathological burden. Third, competing mortality risks in older populations may prevent observation of the most severe outcomes, creating apparent attenuation of effects through selective survival.
The finding that pathological burden and cerebrovascular compromise exert their most devastating effects in younger individuals has important clinical implications. These results suggest that early identification and aggressive management of vascular risk factors may be particularly crucial in younger populations, where the consequences of cerebrovascular events appear most severe [47].
4.4. Mechanistic implications
The striking synergistic effects observed between stroke and PBS scores likely reflect convergent pathological cascades that amplify neurodegeneration through multiple interconnected mechanisms. Vascular-amyloid interactions represent a primary pathway where stroke-induced BBB disruption fundamentally alters amyloid-β dynamics in genetically susceptible individuals [48]. Post-stroke BBB compromise impairs perivascular amyloid clearance while allowing increased infiltration of peripheral inflammatory mediators [49,50]. In individuals with elevated PBS scores harboring genetic variants affecting amyloid metabolism (APOE ε4, APP, PSEN1/2), this creates a "double hit" scenario where genetically enhanced amyloid production combines with vascularly impaired clearance [[51], [52], [53]]. Additionally, stroke-related vascular injury may accelerate cerebral amyloid angiopathy progression, creating a vicious cycle where vascular amyloid deposition increases future stroke risk while stroke-induced damage promotes further vessel wall amyloid accumulation [54,55].
Neuroinflammatory amplification provides another critical mechanistic pathway explaining the observed interactions. Stroke triggers massive neuroinflammatory responses characterized by microglial activation, astrogliosis, and sustained cytokine release [56,57]. In individuals with high PBS scores, particularly those carrying variants in neuroinflammation-related genes (TREM2, CD33, CR1), microglia may exist in a "primed" state with altered inflammatory thresholds and dysregulated complement pathway regulation [58,59]. Post-stroke neuroinflammation in these genetically susceptible individuals leads to sustained microglial hyperactivation, prolonged complement activation, and excessive synaptic pruning, ultimately resulting in accelerated neuronal damage and impaired tissue repair mechanisms that far exceed the sum of individual genetic and vascular contributions [60].
The cellular stress and network disruption mechanisms further explain why stroke-PBS interactions are particularly devastating. Stroke-induced energy failure and oxidative stress may overwhelm protein quality control mechanisms in individuals with genetic variants affecting proteostasis, autophagy, and mitochondrial function [61,62]. This leads to accelerated protein aggregation and cellular dysfunction characteristic of AD pathology [63]. Simultaneously, stroke can disrupt large-scale brain networks, particularly the default mode network preferentially affected in AD [64]. Individuals with high PBS scores may have genetically determined vulnerabilities in network connectivity and resilience, making post-stroke network disruption particularly detrimental and potentially explaining the dramatic dementia risk increase observed in our high PBS + stroke group [65].
4.5. Clinical implications
These findings mandate fundamental shifts toward personalized dementia prevention and risk stratification approaches, with profound implications for clinical practice across age groups. The development of comprehensive pathological burden assessment through PBS could inform prognostic discussions and guide therapeutic decision-making in both research and clinical settings, particularly for identifying high-risk phenotypes such as younger individuals with combined pathological burden and cerebrovascular compromise. For these high-risk populations, aggressive cerebrovascular risk factor management becomes paramount and enhanced stroke prevention measures including antiplatelet therapy consideration and atrial fibrillation screening. The profound age-dependent effects observed underscore the critical importance of age-specific approaches, as traditional risk factors and interventions developed in older populations may have substantially different effects in younger individuals, necessitating tailored strategies that recognize the catastrophic risk patterns unique to younger participants with combined pathologies.
The implementation of PBS in clinical practice offers significant advantages for comprehensive risk stratification, particularly in memory clinics, neurology inpatient settings, and post-stroke follow-up care. Routine integration of PBS through advanced neuroimaging, biomarkers (amyloid and tau PET imaging, cerebrospinal fluid markers), and digital health records could enable clinicians to identify high-risk patients warranting intensive monitoring, personalized care planning, and early intervention before irreversible vascular-neurodegenerative cascade initiation. PBS-based risk categorization could support shared decision-making regarding aggressive vascular risk management, lifestyle modifications, and consideration of emerging disease-modifying therapies for those with high pathological burden. Regular neurological surveillance through annual cognitive assessments and MRI monitoring for subclinical vascular changes should be implemented for high-risk individuals. In research settings, PBS could enhance patient selection for clinical trials targeting mixed-pathology dementia and facilitate longitudinal monitoring of intervention efficacy. As biomarker and imaging technologies mature, adaptation of PBS into non-invasive clinical assessments may help translate neuropathological insights into practical precision medicine approaches for dementia prevention and treatment.
4.6. Limitations
Several limitations should be acknowledged in interpreting these findings. The cross-sectional design limits our ability to establish temporal relationships between pathological processes and their clinical consequences. The retrospective nature of data from the NACC database introduces potential biases that may limit generalizability. The study population represents participants from specialized Alzheimer's Disease Research Centers, who may systematically differ from the general population in socioeconomic status, education levels, healthcare access, and research participation willingness. NACC participants may also represent a selected group with heightened awareness of cognitive symptoms or family history, introducing referral bias that could inflate observed pathological associations.
Survival bias represents a significant limitation, particularly in older age groups and autopsy-based studies. Individuals with severe pathological burden and aggressive disease trajectories may experience higher mortality before autopsy, leading to systematic underrepresentation of severely affected cases. This selective survival could explain: (1) apparent attenuation of pathological burden effects in older participants; (2) counterintuitive findings such as unexpectedly lower AD risk (24.1 %) in non-stroke participants with very high pathological burden, potentially reflecting preferential survival of resilient individuals; and (3) underestimation of true pathological burden effects in the general population. This bias may be particularly pronounced in stroke-stratified analyses, where individuals face competing mortality risks.
Additionally, retrospective reliance on autopsy data creates selection biases, as brain donors may differ systematically from non-donors. The age-dependent patterns observed might partially reflect differential survival patterns rather than purely biological effects. The PBS, while comprehensive, was constructed using available variables and may not capture all relevant pathological processes. Future longitudinal studies with systematic mortality tracking, competing risk analyses, and diverse population sampling would be essential to enhance generalizability and disentangle biological effects from survival bias artifacts.
4.7. Future directions
Future research should refine PBS using machine learning to determine optimal pathological domain weights beyond equal weighting assumptions. Integrating additional markers such as TDP-43 pathology, α-synuclein deposits, and microglial activation may enhance predictive accuracy. Longitudinal studies are needed to track pathological burden accumulation and assess relationships with cognitive decline trajectories.
Investigating genetic factors moderating pathological burden-clinical relationships, particularly APOE genotype and vascular variants, could provide insights into individual vulnerability. Developing risk models integrating genetic, pathological, and clinical factors will support precision medicine. Biomarker development should focus on neuroimaging and fluid correlates reflecting neuropathological burden.
Implementation studies should develop simplified PBS versions, establish risk stratification thresholds, and design clinical decision support tools. Validation across diverse populations is essential for generalizability, while evaluating cost-effectiveness of PBS-guided interventions will inform clinical adoption.
5. Conclusions
This study demonstrates that cumulative pathological burden significantly determines AD risk, with important interactions involving stroke history and age. Comprehensive pathological burden assessment tools could enhance risk stratification and guide therapeutic strategies. The pronounced age-dependent effects underscore the need for age-specific prevention approaches, particularly aggressive vascular risk management in younger populations. These findings advance understanding of AD pathophysiology and support personalized dementia care strategies.
Ethics approval and consent to participate
Not applicable. All data were downloaded from the internet.
Human ethics and consent to participate declarations
Not applicable.
Participate declaration
Not applicable.
Consent for publication
Not applicable.
Availability of data and materials
The data used in the present study are all publicly available at https://naccdata.org/requesting-data/nacc-data.
Funding
The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by the Science and Technology Program of Fujian Provincial Health Commision, China (Grant No.2023TG01010056)
CRediT authorship contribution statement
Fen Liu: Writing – review & editing, Writing – original draft. Xuesong Xia: Writing – original draft. Chengjie Zheng: Writing – review & editing. Feng Liu: Writing – review & editing. Min Jiang: Writing – review & editing.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
The authors thank Maxime Usdin, Research and Development Informatics at Genentech, Inc, Derrek Hibar of the Product Development at Genentech, Inc, and Suyash Mishra of Global Project Strategy at Roche Ltd, Basel, Switzerland, for their valuable feedback. The NACC database is funded by National Institute on Aging and National Institutes of Health Grant U24 AG072122. NACC data are contributed by the National Institute on Aging–funded ADRCs: P30 AG062429 (PI James Brewer, MD, PhD), P30 AG066468 (PI Oscar Lopez, MD), P30 AG062421 (PI Bradley Hyman, MD, PhD), P30 AG066509 (PI Thomas Grabowski, MD), P30 AG066514 (PI Mary Sano, PhD), P30 AG066530 (PI Helena Chui, MD), P30 AG066507 (PI Marilyn Albert, PhD), P30 AG066444 (PI John Morris, MD), P30 AG066518 (PI Jeffrey Kaye, MD), P30 AG066512 (PI Thomas Wisniewski, MD), P30 AG066462 (PI Scott Small, MD), P30 AG072979 (PI David Wolk, MD), P30 AG072972 (PI Charles DeCarli, MD), P30 AG072976 (PI Andrew Saykin, PsyD), P30 AG072975 (PI David Bennett, MD), P30 AG072978 (PI Neil Kowall, MD), P30 AG072977 (PI Robert Vassar, PhD), P30 AG066519 (PI Frank LaFerla, PhD), P30 AG062677 (PI Ronald Petersen, MD, PhD), P30 AG079280 (PI Eric Reiman, MD), P30 AG062422 (PI Gil Rabinovici, MD), P30 AG066511 (PI Allan Levey, MD, PhD), P30 AG072946 (PI Linda Van Eldik, PhD), P30 AG062715 (PI Sanjay Asthana, MD, FRCP), P30 AG072973 (PI Russell Swerdlow, MD), P30 AG066506 (PI Todd Golde, MD, PhD), P30 AG066508 (PI Stephen Strittmatter, MD, PhD), P30 AG066515 (PI Victor Henderson, MD, MS), P30 AG072947 (PI Suzanne Craft, PhD), P30 AG072931 (PI Henry Paulson, MD, PhD), P30 AG066546 (PI Sudha Seshadri, MD), P20 AG068024 (PI Erik Roberson, MD, PhD), P20 AG068053 (PI Justin Miller, PhD), P20 AG068077 (PI Gary Rosenberg, MD), P20 AG068082 (PI Angela Jefferson, PhD), P30 AG072958 (PI Heather Whitson, MD), and P30 AG072959 (PI James Leverenz, MD).
Footnotes
Supplementary material associated with this article can be found, in the online version, at doi:10.1016/j.tjpad.2025.100268.
Contributor Information
Chengjie Zheng, Email: 1240257785@qq.com.
Feng Liu, Email: 465763893@qq.com.
Min Jiang, Email: jiangmin19850522@126.com.
Appendix. Supplementary materials
References
- 1.Scheltens P., De Strooper B., Kivipelto M., Holstege H., Chételat G., Teunissen C.E., Cummings J., van der Flier W.M. Alzheimer's disease. Lancet. 2021;397(10284):1577–1590. doi: 10.1016/S0140-6736(20)32205-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Graff-Radford J., Yong K.X.X., Apostolova L.G., Bouwman F.H., Carrillo M., Dickerson B.C., Rabinovici G.D., Schott J.M., Jones D.T., Murray M.E. New insights into atypical Alzheimer's disease in the era of biomarkers. Lancet Neurol. 2021;20(3):222–234. doi: 10.1016/S1474-4422(20)30440-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Testo A.A., Roundy G., Dumas J.A. Cognitive decline in Alzheimer's disease. Curr Top Behav Neurosci. 2025;69:181–195. doi: 10.1007/7854_2024_527. [DOI] [PubMed] [Google Scholar]
- 4.Karlawish J., Grill J.D. Alzheimer's disease biomarkers and the tyranny of treatment. EBioMedicine. 2024;108 doi: 10.1016/j.ebiom.2024.105291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Fillenbaum G.G., Mohs R. CERAD (Consortium to Establish a Registry for Alzheimer's Disease) neuropsychology assessment battery: 35 years and counting. J Alzheimers Dis. 2023;93(1):1–27. doi: 10.3233/JAD-230026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hyman B.T., Phelps C.H., Beach T.G., Bigio E.H., Cairns N.J., Carrillo M.C., Dickson D.W., Duyckaerts C., Frosch M.P., Masliah E., et al. National Institute on Aging-Alzheimer's Association guidelines for the neuropathologic assessment of Alzheimer's disease. Alzheimers Dement. 2012;8(1):1–13. doi: 10.1016/j.jalz.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Arnsten A.F.T., Datta D., Del Tredici K., Braak H. Hypothesis: tau pathology is an initiating factor in sporadic Alzheimer's disease. Alzheimers Dement. 2021;17(1):115–124. doi: 10.1002/alz.12192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pillai J.A., Bena J., Maly E.F., Leverenz J.B. Initial non-amnestic symptoms relate to faster rate of functional and cognitive decline compared to amnestic symptoms in neuropathologically confirmed dementias. Alzheimers Dement. 2023;19(7):2956–2965. doi: 10.1002/alz.12922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Biessels G.J. Alzheimer's disease, cerebrovascular disease and dementia: lump, split or integrate? Brain. 2022;145(8):2632–2634. doi: 10.1093/brain/awac228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Tang C., Ding Y., Yang J., He D. Cerebrovascular disease in Alzheimer’s disease: Brain structure as a critical mediator of cognitive decline. j prev Alzheimer’s dis. 2025:100209. doi: 10.1016/j.tjpad.2025.100209. [DOI] [PubMed] [Google Scholar]
- 11.Chen F., Zhao J., Meng F., He F., Ni J., Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain. 2024;147(9):2946–2965. doi: 10.1093/brain/awae156. [DOI] [PubMed] [Google Scholar]
- 12.Veitch D.P., Weiner M.W., Miller M., Aisen P.S., Ashford M.A., Beckett L.A., Green R.C., Harvey D., Jack C.R., Jr., Jagust W., et al. The Alzheimer's disease neuroimaging initiative in the era of Alzheimer's disease treatment: a review of ADNI studies from 2021 to 2022. Alzheimers Dement. 2024;20(1):652–694. doi: 10.1002/alz.13449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Schellhorn T., Zucknick M., Askim T., Munthe-Kaas R., Ihle-Hansen H., Seljeseth Y.M., Knapskog A.B., Næss H., Ellekjær H., Thingstad P., et al. Pre-stroke cognitive impairment is associated with vascular imaging pathology: a prospective observational study. BMC Geriatr. 2021;21(1):362. doi: 10.1186/s12877-021-02327-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mele F., Cova I., Nicotra A., Maestri G., Salvadori E., Cucumo V., Masserini F., Martelli M., Pomati S., Bertora P., et al. Prestroke Cognitive Impairment: frequency and Association With Premorbid Neuropsychiatric, Functional, and Neuroimaging Features. Stroke. 2024;55(7):1869–1876. doi: 10.1161/STROKEAHA.123.045344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sriram S., Mehkri Y., Quintin S., Lucke-Wold B. Shared pathophysiology: understanding stroke and Alzheimer's disease. Clin Neurol Neurosurg. 2022;218 doi: 10.1016/j.clineuro.2022.107306. [DOI] [PubMed] [Google Scholar]
- 16.Pinho J., Quintas-Neves M., Dogan I., Reetz K., Reich A., Costa A.S. Incident stroke in patients with Alzheimer's disease: systematic review and meta-analysis. Sci Rep. 2021;11(1) doi: 10.1038/s41598-021-95821-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tang C., Ma Y., Lei X., Ding Y., Yang S., He D. Hypertension linked to Alzheimer's disease via stroke: mendelian randomization. Sci Rep. 2023;13(1) doi: 10.1038/s41598-023-49087-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Behl T., Kaur I., Sehgal A., Khandige P.S., Imran M., Gulati M., Khalid Anwer M., Elossaily G.M., Ali N., Wal P., et al. The link between Alzheimer's disease and stroke: a detrimental synergism. Ageing Res Rev. 2024;99 doi: 10.1016/j.arr.2024.102388. [DOI] [PubMed] [Google Scholar]
- 19.Dong W., Huang Y. Common Genetic Factors and pathways in Alzheimer's disease and ischemic stroke: evidences from GWAS. Genes (Basel) 2023;14(2) doi: 10.3390/genes14020353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hadjihambi A. Cerebrovascular alterations in NAFLD: is it increasing our risk of Alzheimer's disease? Anal Biochem. 2022;636 doi: 10.1016/j.ab.2021.114387. [DOI] [PubMed] [Google Scholar]
- 21.Lithgow B.J., Dastgheib Z., Anssari N., Mansouri B., Blakley B., Ashiri M., Moussavi Z. Physiological separation of Alzheimer's disease and Alzheimer's disease with significant levels of cerebrovascular symptomology and healthy controls. Med Biol Eng Comput. 2021;59(7–8):1597–1610. doi: 10.1007/s11517-021-02409-8. [DOI] [PubMed] [Google Scholar]
- 22.Rajna Z., Mattila H., Huotari N., Tuovinen T., Krüger J., Holst S.C., Korhonen V., Remes A.M., Seppänen T., Hennig J., et al. Cardiovascular brain impulses in Alzheimer's disease. Brain. 2021;144(7):2214–2226. doi: 10.1093/brain/awab144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gao F., Zhan Y., Wang Q., Zhang M., Dai L., Shen Y. Pathological angiogenesis was associated with cerebrovascular lesion and neurodegeneration in Alzheimer's disease. Alzheimers Dement. 2025;21(2) doi: 10.1002/alz.14521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ewers M., Mielke M.M., Hampel H. Blood-based biomarkers of microvascular pathology in Alzheimer's disease. Exp Gerontol. 2010;45(1):75–79. doi: 10.1016/j.exger.2009.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Palmer J.A., Kaufman C.S., Vidoni E.D., Honea R.A., Burns J.M., Billinger S.A. Sex differences in resilience and resistance to brain pathology and dysfunction moderated by cerebrovascular response to exercise and genetic risk for Alzheimer's disease. J Alzheimers Dis. 2022;90(2):535–542. doi: 10.3233/JAD-220359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Palmer J.A., Kaufman C.S., Whitaker-Hilbig A.A., Billinger S.A. APOE4 carriers display loss of anticipatory cerebrovascular regulation across the Alzheimer's disease continuum. Alzheimers Dement. 2025;21(5) doi: 10.1002/alz.70229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Abdulrahman H., van Dalen J.W., den Brok M., Latimer C.S., Larson E.B., Richard E. Hypertension and Alzheimer's disease pathology at autopsy: a systematic review. Alzheimers Dement. 2022;18(11):2308–2326. doi: 10.1002/alz.12707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Laing K.K., Simoes S., Baena-Caldas G.P., Lao P.J., Kothiya M., Igwe K.C., Chesebro A.G., Houck A.L., Pedraza L., Hernández A.I., et al. Cerebrovascular disease promotes tau pathology in Alzheimer's disease. Brain Commun. 2020;2(2):fcaa132. doi: 10.1093/braincomms/fcaa132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wagner J., Degenhardt K., Veit M., Louros N., Konstantoulea K., Skodras A., Wild K., Liu P., Obermüller U., Bansal V., et al. Medin co-aggregates with vascular amyloid-β in Alzheimer's disease. Nature. 2022;612(7938):123–131. doi: 10.1038/s41586-022-05440-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cogswell P.M., Lundt E.S., Therneau T.M., Mester C.T., Wiste H.J., Graff-Radford J., Schwarz C.G., Senjem M.L., Gunter J.L., Reid R.I., et al. Evidence against a temporal association between cerebrovascular disease and Alzheimer's disease imaging biomarkers. Nat Commun. 2023;14(1):3097. doi: 10.1038/s41467-023-38878-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Stern Y., Arenaza-Urquijo E.M., Bartrés-Faz D., Belleville S., Cantilon M., Chetelat G., Ewers M., Franzmeier N., Kempermann G., Kremen W.S., et al. Whitepaper: defining and investigating cognitive reserve, brain reserve, and brain maintenance. Alzheimers Dement. 2020;16(9):1305–1311. doi: 10.1016/j.jalz.2018.07.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.de Rooij S.R. Are brain and cognitive reserve shaped by early life circumstances? Front Neurosci. 2022;16 doi: 10.3389/fnins.2022.825811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Alvares Pereira G., Silva Nunes M.V., Alzola P., Contador I. Cognitive reserve and brain maintenance in aging and dementia: an integrative review. Appl Neuropsychol Adult. 2022;29(6):1615–1625. doi: 10.1080/23279095.2021.1872079. [DOI] [PubMed] [Google Scholar]
- 34.Smith V., Younes K., Poston K.L., Mormino E.C., Young C.B. Reliability of remote national Alzheimer's coordinating center uniform data set data. Alzheimers Dement. 2023;15(4) doi: 10.1002/dad2.12498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Beekly D.L., Ramos E.M., Lee W.W., Deitrich W.D., Jacka M.E., Wu J., Hubbard J.L., Koepsell T.D., Morris J.C., Kukull W.A. The National Alzheimer's Coordinating Center (NACC) database: the Uniform Data Set. Alzheimer Dis Assoc Disord. 2007;21(3):249–258. doi: 10.1097/WAD.0b013e318142774e. [DOI] [PubMed] [Google Scholar]
- 36.Sadrolashrafi K., Craft S., Decourt B., Adem A., Wilson J.R., Miller J., Sabbagh M.N. Is diabetes associated with increased pathological burden in Alzheimer's disease? Alzheimers Dement. 2021;13(1) doi: 10.1002/dad2.12248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Coomans E.M., van Westen D., Binette A.P., Strandberg O., Spotorno N., Serrano G.E., Beach T.G., Palmqvist S., Stomrud E., Ossenkoppele R., et al. Interactions between vascular burden and amyloid-β pathology on trajectories of tau accumulation. Brain. 2024;147(3):949–960. doi: 10.1093/brain/awad317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.King A., Bodi I., Troakes C. The neuropathological diagnosis of Alzheimer's disease-the challenges of pathological mimics and concomitant pathology. Brain Sci. 2020;10(8) doi: 10.3390/brainsci10080479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Stelzer G.T., Lima-Filho R.A.S. Amyloid-β as a key player in cerebrovascular dysfunction in Alzheimer's disease. J Neurosci. 2024;(27):44. doi: 10.1523/JNEUROSCI.0663-24.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Veitch D.P., Weiner M.W., Aisen P.S., Beckett L.A., DeCarli C., Green R.C., Harvey D., Jack C.R., Jr., Jagust W., Landau S.M., et al. Using the Alzheimer's disease neuroimaging initiative to improve early detection, diagnosis, and treatment of Alzheimer's disease. Alzheimers Dement. 2022;18(4):824–857. doi: 10.1002/alz.12422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Oliva G., Masina F., Hosseinkhani N., Montemurro S., Arcara G. Cognitive reserve in the recovery and rehabilitation of stroke and traumatic brain injury: a systematic review. Clin Neuropsychol. 2024:1–37. doi: 10.1080/13854046.2024.2405226. [DOI] [PubMed] [Google Scholar]
- 42.van Olst L., Simonton B., Edwards A.J., Forsyth A.V., Boles J., Jamshidi P., Watson T., Shepard N., Krainc T., Argue B.M., et al. Microglial mechanisms drive amyloid-β clearance in immunized patients with Alzheimer's disease. Nat Med. 2025;31(5):1604–1616. doi: 10.1038/s41591-025-03574-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Goulay R., Mena Romo L., Hol E.M., Dijkhuizen R.M. From stroke to dementia: a comprehensive review exposing tight interactions between stroke and amyloid-β formation. Transl Stroke Res. 2020;11(4):601–614. doi: 10.1007/s12975-019-00755-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cramer S.C., Richards L.G., Bernhardt J., Duncan P. Cognitive deficits after stroke. Stroke. 2023;54(1):5–9. doi: 10.1161/STROKEAHA.122.041775. [DOI] [PubMed] [Google Scholar]
- 45.Elahi F.M., Wang M.M., Meschia J.F. Cerebral small vessel disease-related dementia: more questions than answers. Stroke. 2023;54(3):648–660. doi: 10.1161/STROKEAHA.122.038265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hannawi Y. Cerebral small vessel disease: a review of the pathophysiological mechanisms. Transl Stroke Res. 2024;15(6):1050–1069. doi: 10.1007/s12975-023-01195-9. [DOI] [PubMed] [Google Scholar]
- 47.Li G., Toschi N., Devanarayan V., Batrla R., Boccato T., Cho M., Ferrante M., Frech F., Galvin J.E., Henley D., et al. The age-specific comorbidity burden of mild cognitive impairment: a US claims database study. Alzheimers Res Ther. 2023;15(1):211. doi: 10.1186/s13195-023-01358-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Perosa V., Oltmer J., Munting L.P., Freeze W.M., Auger C.A., Scherlek A.A., van der Kouwe A.J., Iglesias J.E., Atzeni A., Bacskai B.J., et al. Perivascular space dilation is associated with vascular amyloid-β accumulation in the overlying cortex. Acta Neuropathol. 2022;143(3):331–348. doi: 10.1007/s00401-021-02393-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kurz C., Walker L., Rauchmann B.S., Perneczky R. Dysfunction of the blood-brain barrier in Alzheimer's disease: evidence from human studies. Neuropathol Appl Neurobiol. 2022;48(3) doi: 10.1111/nan.12782. [DOI] [PubMed] [Google Scholar]
- 50.Li J., Li M., Ge Y., Chen J., Ma J., Wang C., Sun M., Wang L., Yao S., Yao C. β-amyloid protein induces mitophagy-dependent ferroptosis through the CD36/PINK/PARKIN pathway leading to blood-brain barrier destruction in Alzheimer's disease. Cell Biosci. 2022;12(1):69. doi: 10.1186/s13578-022-00807-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Pahlavani H.A. Exercise therapy to prevent and treat Alzheimer's disease. Front Aging Neurosci. 2023;15 doi: 10.3389/fnagi.2023.1243869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Xiong M., Jiang H., Serrano J.R., Gonzales E.R., Wang C., Gratuze M., Hoyle R., Bien-Ly N., Silverman A.P., Sullivan P.M., et al. APOE immunotherapy reduces cerebral amyloid angiopathy and amyloid plaques while improving cerebrovascular function. Sci Transl Med. 2021;(581):13. doi: 10.1126/scitranslmed.abd7522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Abubakar M.B., Sanusi K.O., Ugusman A., Mohamed W., Kamal H., Ibrahim N.H., Khoo C.S., Kumar J. Alzheimer's disease: an update and insights into pathophysiology. Front Aging Neurosci. 2022;14 doi: 10.3389/fnagi.2022.742408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ouyang F., Jiang Z., Chen X., Chen Y., Wei J., Xing S., Zhang J., Fan Y., Zeng J. Is cerebral amyloid-β deposition related to post-stroke cognitive impairment? Transl Stroke Res. 2021;12(6):946–957. doi: 10.1007/s12975-021-00921-5. [DOI] [PubMed] [Google Scholar]
- 55.Allen J., Ermine C.M., Lin R., Cloud G.C., Shultz S.R., Casillas-Espinosa P.M. Proteinopathies and the neurodegenerative aftermath of stroke: potential biomarkers and treatment targets. Stroke. 2025;56(6):1600–1611. doi: 10.1161/STROKEAHA.124.049279. [DOI] [PubMed] [Google Scholar]
- 56.Simats A., Liesz A. Systemic inflammation after stroke: implications for post-stroke comorbidities. EMBO Mol Med. 2022;14(9) doi: 10.15252/emmm.202216269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Liu Q., Shi K., Wang Y., Shi F.D. Neurovascular inflammation and complications of thrombolysis therapy in stroke. Stroke. 2023;54(10):2688–2697. doi: 10.1161/STROKEAHA.123.044123. [DOI] [PubMed] [Google Scholar]
- 58.Kelly P.J., Lemmens R., Tsivgoulis G. Inflammation and stroke risk: a new target for prevention. Stroke. 2021;52(8):2697–2706. doi: 10.1161/STROKEAHA.121.034388. [DOI] [PubMed] [Google Scholar]
- 59.Zhu H., Hu S., Li Y., Sun Y., Xiong X., Hu X., Chen J., Qiu S. Interleukins and ischemic stroke. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.828447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Candelario-Jalil E., Dijkhuizen R.M., Magnus T. Neuroinflammation, stroke, blood-brain barrier dysfunction, and imaging modalities. Stroke. 2022;53(5):1473–1486. doi: 10.1161/STROKEAHA.122.036946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chen H., He Y., Chen S., Qi S., Shen J. Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: applications for natural product efficacy with omics and systemic biology. Pharmacol Res. 2020;158 doi: 10.1016/j.phrs.2020.104877. [DOI] [PubMed] [Google Scholar]
- 62.Sun Y.Y., Zhu H.J., Zhao R.Y., Zhou S.Y., Wang M.Q., Yang Y., Guo Z.N. Remote ischemic conditioning attenuates oxidative stress and inflammation via the Nrf2/HO-1 pathway in MCAO mice. Redox Biol. 2023;66 doi: 10.1016/j.redox.2023.102852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Briyal S., Ranjan A.K., Gulati A. Oxidative stress: a target to treat Alzheimer's disease and stroke. Neurochem Int. 2023;165 doi: 10.1016/j.neuint.2023.105509. [DOI] [PubMed] [Google Scholar]
- 64.Khan S., Kamal M.A. Cardiac biomarkers in stroke, Alzheimer's disease, and other dementia. Are they of use? A brief overview of data from recent investigations. CNS Neurol Disord Drug Targets. 2021;20(8):687–693. doi: 10.2174/1871527319666201005171003. [DOI] [PubMed] [Google Scholar]
- 65.Lo J.W., Crawford J.D., Desmond D.W., Bae H.J., Lim J.S., Godefroy O., Roussel M., Kang Y., Jahng S., Köhler S., et al. Long-term cognitive decline after stroke: an individual participant data meta-analysis. Stroke. 2022;53(4):1318–1327. doi: 10.1161/STROKEAHA.121.035796. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data used in the present study are all publicly available at https://naccdata.org/requesting-data/nacc-data.


