Skip to main content
The Journal of Prevention of Alzheimer's Disease logoLink to The Journal of Prevention of Alzheimer's Disease
. 2025 May 29;12(8):100209. doi: 10.1016/j.tjpad.2025.100209

Cerebrovascular disease in Alzheimer's disease: Brain structure as a critical mediator of cognitive decline

Chao Tang 1,, Yaqi Ding 1,, Jiaxin Yang 1,, Dian He 1,
PMCID: PMC12413723  PMID: 40447488

Highlights

  • Cerebrovascular disease substantially increases Alzheimer's disease risk.

  • Hippocampal atrophy serves as the strongest mediator between vascular pathology and cognitive decline.

  • Concomitant AD +CVD pathology produces a distinctive hybrid pattern of brain-cognition relationships.

  • Vascular factors show strong correlations with both tau and amyloid pathologies.

  • Findings emphasize vascular health as an integral component in Alzheimer's disease prevention and treatment.

Keywords: Alzheimer's disease, Cerebrovascular disease, Hippocampal atrophy, Cognitive decline, Structural mediation

Abstract

Background

The co-occurrence of Alzheimer's disease and cerebrovascular disease is increasingly prevalent in aging populations, yet the mechanisms of their interaction remain incompletely understood. This study aims to investigate the associations between CVD and AD and their composite effects on cognitive function, identifying key mediating pathways in these relationships.

Methods

Participants underwent standardized clinical evaluations, detailed neuropsychological testing, and comprehensive neuropathological examinations. Structural equation modeling with multiple mediation analyses was employed to disentangle direct and indirect effects of vascular pathology on cognition and identify key mediating pathways. Relationships between specific cognitive domain assessments and whole brain and hippocampal volumes were analyzed, while interactions between traditional AD biomarkers (amyloid, tau) and vascular factors were examined.

Results

CVD substantially increased AD risk. Structural equation modeling revealed that vascular factors influence cognitive performance primarily through hippocampal atrophy, APOE genotype, and cerebral atrophy. Participants with concomitant AD +CVD pathology displayed a distinctive hybrid pattern of brain-cognition relationships, with stronger correlations between hippocampal atrophy and cognitive performance compared to pure AD or CVD cases. Pathway-specific analysis demonstrated that hippocampal atrophy served as the strongest mediator of vascular effects on cognition, followed by cerebral atrophy and APOE genotype.

Conclusion

Our findings demonstrate that cerebrovascular disease significantly increases the risk of Alzheimer's disease and substantially influences its clinical expression through multiple pathways, with structural brain changes serving as critical mediators of vascular effects on cognition. These results highlight the importance of addressing vascular health as an integral component of strategies to prevent and treat Alzheimer's disease and related cognitive disorders.

1. Introduction

Alzheimer's disease (AD) represents the most common cause of dementia worldwide, characterized by progressive cognitive decline and distinctive neuropathological features including Aβ plaques and neurofibrillary tangles [1]. Cerebrovascular disease (CVD), meanwhile, constitutes another major contributor to cognitive impairment in aging populations [2]. While traditionally viewed as distinct pathological entities, accumulating evidence suggests substantial overlap and potential interaction between AD and cerebrovascular pathologies [3], with population-based autopsy studies revealing that mixed pathologies are more common than pure presentations in elderly individuals with dementia [4].

The relationship between cerebrovascular factors and AD pathogenesis remains only partially understood [5], but recent studies have significantly advanced our knowledge of this complex interplay. Multiple pathways have been proposed through which vascular factors can influence neurodegeneration related to AD [6,7]. For instance, chronic hypoperfusion can lead to insufficient blood flow to critical brain regions, which can exacerbate neuronal damage and cognitive decline [8,9]. Additionally, blood-brain barrier dysfunction allows potentially harmful substances to enter the brain, triggering neuroinflammation, which is increasingly recognized as a key contributor to AD pathology [10]. Recent research has identified specific inflammatory markers, such as cytokines and chemokines, that relate closely to both cerebrovascular disease and neurodegenerative processes [11,12]. Furthermore, oxidative stress resulting from vascular injury has been implicated in accelerating amyloid-beta accumulation and tau phosphorylation, two hallmark features of AD [13,14].

Recent studies have shown that cerebrovascular pathology may lower the clinical threshold for AD symptoms, leading to earlier manifestation and greater cognitive impairment in individuals with concomitant vascular dysfunction [15]. For example, a 2022 study demonstrated that patients with both vascular lesions and AD pathology exhibited significantly worse cognitive outcomes compared to those with AD alone [16]. This increasing body of evidence underscores the critical importance of addressing cerebrovascular health in understanding, preventing, and treating Alzheimer's disease. By bridging the gap between cerebrovascular factors and AD, future interventions may enhance cognitive resilience and improve clinical outcomes for at-risk populations.

The potential mediating mechanisms linking cerebrovascular disease to cognitive outcomes in the context of AD remain poorly characterized. While traditional AD biomarkers including amyloid and tau pathologies represent well-established correlates of cognitive decline, the relative contributions of these markers versus structural brain changes in mediating vascular effects on cognition have not been comprehensively assessed [17]. Moreover, the potential synergistic interactions between vascular factors and white matter lesions—a common manifestation of small vessel cerebrovascular disease—warrant further investigation.

The National Alzheimer's Coordinating Center (NACC) database provides a unique opportunity to investigate these complex relationships through its comprehensive collection of standardized clinical, neuropsychological, and neuropathological data from Alzheimer's Disease Research Centers across the United States [16]. This resource enables robust analysis of multiple pathological features and their relationships to cognitive outcomes in well-characterized cohorts [18,19].

While the relationship between CVD and AD is increasingly recognized, significant research gaps remain. Existing studies have inadequately explored the complex interactions between vascular pathologies and neurodegeneration, often examining these factors in isolation. Our research addresses these limitations by employing advanced modeling techniques to comprehensively investigate the intricate mechanisms linking CVD and AD progression, providing novel insights into cognitive decline.

To this end, we aimed to determine the direct and indirect effects of cerebrovascular pathology on cognitive performance in AD, identifying key mediating pathways through structural equation modeling with multiple mediation analysis. Specifically, we examined whether vascular factors and white matter lesions impact cognitive outcomes through traditional AD pathological markers (amyloid and tau pathology), structural brain changes (hippocampal and cerebral atrophy, brain volume), or genetic factors (APOE genotype) [2,20]. By elucidating these mediating mechanisms, we sought to enhance understanding of the complex interplay between cerebrovascular and AD pathologies and potentially identify novel targets for therapeutic intervention aimed at preserving cognitive function in individuals with mixed pathologies.

2. Methods

2.1. Data source and participants

Data for this study were obtained from the NACC database, established in 1999 to facilitate collaborative research on AD and related disorders. The NACC database aggregates standardized clinical and neuropathological data collected by Alzheimer's Disease Research Centers (ADRCs) across the United States, funded by the National Institute on Aging (NIA). The database includes comprehensive information on participants' demographics, medical history, neurological examinations, neuropsychological test results, functional assessments, clinical diagnoses, APOE genotyping, neuroimaging findings, and neuropathological evaluations for those with available autopsy data.

For the current investigation, we included participants from the NACC database who had completed standardized clinical evaluations, cognitive assessments, and had available neuropathological data. All participants underwent comprehensive clinical assessments during life and subsequent neuropathological examinations at death. Only participants with complete data for cognitive measures, cerebrovascular pathology variables, and AD-related neuropathological markers were included in the analysis. For the etiologic diagnosis of cognitive disorders, Alzheimer's disease was identified using the code NACCALZD, where NACCALZD = 1 indicates the presence of Alzheimer's disease as the presumptive etiologic diagnosis of the cognitive disorder, while NACCALZD = 0 indicates its absence. Cerebrovascular disease was identified using the code CVD, where CVD = 1 indicates the presence of vascular brain injury (VBI) as an etiologic diagnosis, while CVD = 0 indicates no VBI as an etiologic diagnosis. The NACC database used in this study has received all necessary ethical approvals, and the research process strictly adheres to the principles of the Declaration of Helsinki and the guidelines of the ethics committees of all participating institutions. NACC ensures that informed consent was obtained from all participants or their legal representatives.

2.2. Inclusion and exclusion criteria

2.2.1. Inclusion criteria

  • 1. Diagnosis Confirmation: Participants must be diagnosed with AD using the NACC code NACCALZD = 1

  • 2. Clinical Assessment: Participants should have undergone comprehensive clinical evaluations, including cognitive and functional assessments, as well as neuropathological examinations at death.

  • 3. Cerebrovascular Disease Assessment: Diagnosis of cerebrovascular disease must be confirmed with CVD = 1, indicating vascular brain injury.

  • 4. Complete Data: Participants must have comprehensive data for all cognitive measures, cerebrovascular pathology variables, and AD-related neuropathological markers.

2.2.2. Exclusion criteria

  • 1. Incomplete Data: Participants with missing or incomplete cognitive or neuropathological data will be excluded.

  • 2. Non-completers: Individuals who did not finish required evaluations or assessments will be excluded.

  • 3. Non-AD Diagnosis: Exclude participants diagnosed with other dementias or cognitive disorders not meeting AD or CVD criteria.

  • 4. Lack of Autopsy: Participants must have neuropathological data from an autopsy; those without it will be excluded.

2.3. Cognitive assessment

Cognitive functioning was comprehensively evaluated using standardized neuropsychological assessments from the NACC Uniform Data Set. The Clinical Dementia Rating (CDR) scale, a semi-structured interview with both participants and informants, assessed impairment across six domains (memory, orientation, judgment/problem solving, community affairs, home/hobbies, and personal care), providing Global scores (0–3) and Sum of Boxes scores (0–18) to quantify dementia severity. Global cognitive status was measured using the Mini-Mental State Examination (MMSE, 0–30 points), assessing orientation, attention, calculation, recall, language, and visuospatial abilities, with lower scores indicating greater impairment. Language function was evaluated through the Boston Naming Test (confrontation naming of objects) and category fluency tasks (generating animal and vegetable names within one minute), which assess semantic knowledge, word retrieval, and cognitive processing speed. Memory capabilities were quantified via immediate and delayed recall tests, measuring both encoding and retrieval processes critical for episodic memory function. Attention and executive functioning were assessed through Digit Span Forward (assessing attention span by repeating increasingly long number sequences), Digit Span Backward (evaluating working memory by repeating number sequences in reverse order), and Trail Making Test differential scores (TMT B-TMT A, isolating executive control by accounting for processing speed), providing a comprehensive profile of frontal-subcortical cognitive functions often affected in both AD and cerebrovascular disease.

2.4. Neuropathological assessment

Neuropathological evaluations were conducted by expert neuropathologists at NACC-affiliated centers following standardized protocols to ensure consistency across sites. Alzheimer's disease pathology was systematically quantified using Braak staging for neurofibrillary tangles (0=absent, I/II=transentorhinal, III/IV=limbic, V/VI=isocortical), reflecting the topographical progression of tau pathology from medial temporal structures to association cortices. Neuritic plaque burden was assessed using CREAD scores (0–3, reflecting none to frequent), providing a standardized measure of amyloid-associated neuritic degeneration. Thal phases (0–3) documented the anatomical progression of Aβ deposition from neocortex to brainstem structures. Cerebrovascular pathology evaluation included documentation of large vessel disease, small vessel disease, microinfarcts, and hemorrhages, with overall cerebrovascular disease categorized as present or absent based on established neuropathological criteria. White matter damage was specifically quantified through assessment of white matter rarefaction (classified as None, Mild, Moderate, or Severe), reflecting the extent of myelin loss and axonal damage associated with small vessel cerebrovascular disease. Brain structural integrity was assessed through whole brain weight measurements (in grams) and detailed regional atrophy evaluations of cerebral cortex, hippocampus, and specific lobar regions (frontal, temporal, parietal, occipital), each rated as None, Mild, Moderate, or Severe based on macroscopic examination. APOE genotyping was performed on DNA extracted from blood or brain tissue, with participants classified according to specific allele combinations (ε2/ε2, ε2/ε3, ε2/ε4, ε3/ε3, ε3/ε4, ε4/ε4) and carrier status, given the established role of the ε4 allele as a significant risk factor for both AD pathology and cerebrovascular disease.

2.5. Statistical analysis

Statistical analyses were performed to examine relationships between cerebrovascular pathology, AD pathological markers, and cognitive outcomes. Participants were initially categorized into four groups based on neuropathological findings: AD (N = 298), CVD (N = 17), AD+CVD (N = 81), and controls (N = 309). This classification was used for baseline characteristic comparisons and correlation analyses, with between-group differences evaluated using analysis of variance (ANOVA) for continuous variables and chi-square tests for categorical variables. For risk factor assessment and mediation analyses, subjects were reclassified into two groups: AD group and non-AD group, allowing focused investigation of cerebrovascular disease as a potential risk factor for AD pathology.

Two primary cognitive outcomes were analyzed: MMSE as a measure of global cognitive function (with lower scores indicating greater impairment) and CDR Sum as an assessment of dementia severity (with higher scores indicating more severe impairment). Primary exposure variables included vascular factors (dichotomized as present or absent) and white matter lesions (rated as mild, moderate, or severe). An interaction term between vascular factors and white matter lesions was created to assess potential synergistic effects on cognitive outcomes and neuropathological markers.

We employed structural equation modeling (SEM) to conduct multiple mediation analyses examining seven potential mediating variables: tau pathology (Braak staging: 0 = none, 1 = stages I/II, 3 = stages III/IV, 5 = stages V/VI), amyloid pathology (CREAD score: 0–3), amyloid distribution (Thal phase: 0–3), brain volume (total cerebral volume), cerebral atrophy (mild/moderate/severe), hippocampal atrophy (mild/moderate/severe), and APOE genotype (coded as number of ε4 alleles: 0, 1, or 2). All analyses were adjusted for age and education level as established covariates influencing cognitive performance and disease risk.

For the mediation analysis, comprehensive models were constructed for each cognitive outcome that incorporated: exposure-mediator paths (a paths) representing effects of vascular factors, white matter lesions, and their interaction on each potential mediator; mediator-outcome paths (b paths) representing effects of each mediator on cognitive outcomes; and direct effect paths (c' paths) representing direct effects of exposures on outcomes independent of mediators. Specific indirect effects were computed as products of corresponding a and b coefficients, with total indirect effects calculated as sums of all specific indirect effects for each exposure. Total effects were determined by combining direct and total indirect effects. Full Information Maximum Likelihood estimation was employed to handle missing data, allowing inclusion of all available data points without listwise deletion. All statistical analyses were conducted using R (version 4.3.3) with specialized packages for structural equation modeling (lavaan), data manipulation (dplyr), visualization (ggplot2), and descriptive statistics (psych). Model fit was assessed using chi-square test and comparative fit indices, with significance levels set at p < 0.05 for all analyses.

3. Results

3.1. Participant demographics and group distribution

As shown in Table 1, the study included 705 participants: 298 (42.3 %) with AD, 17 (2.4 %) with CVD, 81 (11.5 %) with AD+CVD, and 309 (43.8 %) controls. Gender distribution differed significantly between groups (p = 8.60E-04, FDR = 1.70E-03), with more males in the AD group (54.7 %) than in controls (39.2 %). The mean age of all participants was 86.30 ± 7.86 years, with the AD +CVD group being oldest (88.23 ± 5.95 years). Educational attainment was similar across groups (overall mean: 15.80 ± 4.16 years). Cognitive status showed marked differences between groups (p = 1.70E-147, FDR = 2.10E-146), with most AD patients having dementia (70.8 %) or MCI (28.9 %), while all CVD and control participants had no cognitive impairment. In the concomitant group, 81.5 % had dementia and 18.5 % had MCI. APOE ε4 allele presence varied significantly (p = 1.20E-10, FDR = 5.90E-10), being more frequent in AD (48.7 %) and concomitant AD/CVD (38.3 %) groups compared to controls (22.0 %) and CVD patients (5.9 %). Clinical Dementia Rating scores also differed significantly (p = 2.20E-125, FDR = 2.40E-124), with most control (93.2 %) and CVD (94.1 %) participants scoring 0 (no dementia), while AD and concomitant groups showed predominantly higher scores indicating various dementia severity levels.

Table 1.

Demographic and clinical characteristics of study participants.

Characteristics AD (N = 298) CVD (N = 17) AD with CVD (N = 81) Control (N = 309) Total (N = 705) P-value FDR
Gender, N (%) 8.60E-04 1.70E-03
female 135 (19.15 %) 10 (1.42 %) 37 (5.25 %) 188 (26.67 %) 370 (52.48 %)
male 163 (23.12 %) 7 (0.99 %) 44 (6.24 %) 121 (17.16 %) 335 (47.52 %)
Age (years) 85.30 ± 8.77 87.82 ± 8.03 88.23 ± 5.95 86.67 ± 7.23 86.30 ± 7.86
Years of education (years) 15.52 ± 2.78 16.18 ± 2.27 16.20 ± 2.72 15.94 ± 5.46 15.80 ± 4.16
Cognitive Status, N (%) 1.70E-147 2.10E-146
Cognitive impairment (non-MCI) 1 (0.14 %) 0 0 0 1 (0.14 %)
Dementia 211 (29.93 %) 0 66 (9.36 %) 0 277 (39.29 %)
MCI 86 (12.20 %) 0 15 (2.13 %) 0 101 (14.33 %)
NO cognition 0 17 (2.41 %) 0 309 (43.83 %) 326 (46.24 %)
APOE ε4 allele, N (%) 1.20E-10 5.90E-10
0 153 (21.70 %) 16 (2.27 %) 50 (7.09 %) 241 (34.18 %) 460 (65.25 %)
1 128 (18.16 %) 1 (0.14 %) 25 (3.55 %) 64 (9.08 %) 218 (30.92 %)
2 17 (2.41 %) 0 6 (0.85 %) 4 (0.57 %) 27 (3.83 %)
CDR Global, N (%) 2.20E-125 2.40E-124
0 1 (0.14 %) 16 (2.27 %) 2 (0.28 %) 288 (40.85 %) 307 (43.55 %)
0.5 109 (15.46 %) 1 (0.14 %) 21 (2.98 %) 20 (2.84 %) 151 (21.42 %)
1 117 (16.60 %) 0 36 (5.11 %) 1 (0.14 %) 154 (21.84 %)
2 62 (8.79 %) 0 18 (2.55 %) 0 80 (11.35 %)
3 9 (1.28 %) 0 4 (0.57 %) 0 13 (1.84 %)
CDR Sum 6.06 ± 4.13 0.06 ± 0.17 6.77 ± 4.16 0.13 ± 0.53 3.40 ± 4.30

Abbreviations: AD: Alzheimer's Disease, CVD: Cerebrovascular Disease, MCI: Mild Cognitive Impairment, APOE: Apolipoprotein E, CDR: Clinical Dementia Rating.

3.2. Cognitive assessment scores by group

Neuropsychological test performance varied markedly across diagnostic groups (Table 2). The AD group demonstrated the most impaired cognitive profile with the lowest MMSE scores (21.77 ± 5.85), compared to control (28.60 ± 1.43) and CVD (29.18 ± 0.95) groups, while the AD+CVD group showed similar impairment (22.53 ± 5.75). Memory function was severely affected in both AD and concomitant groups, with immediate recall scores of 6.63 ± 5.17 and 7.57 ± 5.23, and delayed recall scores of 4.94 ± 5.33 and 5.69 ± 5.10, respectively, substantially lower than CVD (immediate: 16.00 ± 3.61; delayed: 15.12 ± 3.87) and control groups (immediate: 14.92 ± 3.99; delayed: 13.89 ± 4.16). Working memory, as measured by digit span tests, showed similar patterns with AD and concomitant groups performing worse than CVD and control groups in both forward and backward conditions. Semantic fluency was notably impaired in AD (animals: 11.13 ± 5.53; vegetables: 6.97 ± 3.94) and concomitant groups (animals: 12.70 ± 4.93; vegetables: 7.27 ± 3.57) compared to CVD (animals: 16.59 ± 5.65; vegetables: 10.53 ± 3.57) and control groups (animals: 18.12 ± 4.89; vegetables: 12.51 ± 3.75). Executive function, assessed by TMT B-TMT A, showed greater impairment in AD (61.99 ± 35.09) and concomitant (58.42 ± 30.44) groups compared to CVD (37.44 ± 7.20) and control groups (40.70 ± 16.55), indicating poorer cognitive flexibility and set-shifting abilities in patients with AD pathology.

Table 2.

Neuropsychological test performance across diagnostic groups.

Characteristics AD (N = 298) CVD (N = 17) AD with CVD (N = 81) Control (N = 309) Total (N = 705)
MMSE 21.77 ± 5.85 29.18 ± 0.95 22.53 ± 5.75 28.60 ± 1.43 25.03 ± 5.51
Boston Naming 21.90 ± 7.03 27.18 ± 2.40 23.74 ± 5.21 27.52 ± 2.31 24.85 ± 5.68
Immediate recall 6.63 ± 5.17 16.00 ± 3.61 7.57 ± 5.23 14.92 ± 3.99 10.92 ± 6.16
Delayed recall 4.94 ± 5.33 15.12 ± 3.87 5.69 ± 5.10 13.89 ± 4.16 9.57 ± 6.49
Digit span forw. – trials 7.24 ± 2.09 8.94 ± 2.30 7.21 ± 1.96 8.11 ± 1.88 7.69 ± 2.04
Digit span forw. – length 6.09 ± 1.12 7.06 ± 1.03 6.00 ± 1.06 6.51 ± 1.08 6.30 ± 1.12
Digit span backw. – trials 4.81 ± 2.02 7.71 ± 2.23 5.00 ± 2.03 6.47 ± 1.87 5.68 ± 2.14
Digit span backw. – length 3.83 ± 1.25 5.24 ± 1.25 3.92 ± 1.16 4.78 ± 1.08 4.32 ± 1.25
Animals 11.13 ± 5.53 16.59 ± 5.65 12.70 ± 4.93 18.12 ± 4.89 14.69 ± 6.14
Vegetables 6.97 ± 3.94 10.53 ± 3.57 7.27 ± 3.57 12.51 ± 3.75 9.68 ± 4.66
TMT B-TMT A 61.99 ± 35.09 37.44 ± 7.20 58.42 ± 30.44 40.70 ± 16.55 50.53 ± 28.37

3.3. Pathological findings across various assessment tools

Significant neuropathological differences were observed across diagnostic groups (Table 3). Braak staging revealed marked differences (p = 1.80E-22), with AD patients showing predominantly high-stage pathology (Braak V/VI: 52.3 % of AD cases) compared to controls, where most cases exhibited moderate pathology (Braak III/IV: 63.4 %). The CREAD neuritic plaque assessment similarly demonstrated significant differences (p = 1.80E-19), with AD patients showing higher scores (CREAD 2–3: 59.1 %) while controls predominantly had minimal plaque pathology (CREAD 0: 51.5 %). Thal amyloid phase distribution also varied significantly (p = 3.50E-17), with 67.8 % of AD cases showing advanced phase 3 amyloid deposition compared to only 31.7 % of controls.

Table 3.

Neuropathological findings across diagnostic groups.

Characteristics AD (N = 298) CVD (N = 17) AD with CVD (N = 81) Control (N = 309) Total (N = 705) p-value FDR
Braak, N (%) 1.80E-22 1.60E-21
0 6(0.85 %) 2(0.28 %) 0 2(0.28 %) 10(1.42 %)
Braak I/ II 35(4.96 %) 2(0.28 %) 12(1.70 %) 67(9.50 %) 116(16.45 %)
Braak III/IV 101(14.33 %) 12(1.70 %) 34(4.82 %) 196(27.80 %) 343(48.65 %)
Braak V/VI 156(22.13 %) 1(0.14 %) 35(4.96 %) 42(5.96 %) 234(33.19 %)
CREAD, N (%) 1.80E-19 1.30E-18
0 52(7.38 %) 12(1.70 %) 28(3.97 %) 159(22.55 %) 251(35.60 %)
1 70(9.93 %) 0 17(2.41 %) 67(9.50 %) 154(21.84 %)
2 87(12.34 %) 2(0.28 %) 22(3.12 %) 23(3.26 %) 134(19.01 %)
3 89(12.62 %) 3(0.43 %) 14(1.99 %) 60(8.51 %) 166(23.55 %)
Thal, N (%) 3.50E-17 2.10E-16
0 23(3.26 %) 6(0.85 %) 13(1.84 %) 87(12.34 %) 129(18.30 %)
1 37(5.25 %) 4(0.57 %) 18(2.55 %) 62(8.79 %) 121(17.16 %)
2 36(5.11 %) 5(0.71 %) 13(1.84 %) 62(8.79 %) 116(16.45 %)
3 202(28.65 %) 2(0.28 %) 37(5.25 %) 98(13.90 %) 339(48.09 %)
vascular, N (%) 0.63 0.63
NO 1(0.14 %) 0 0 3(0.43 %) 4(0.57 %)
YES 297(42.13 %) 17(2.41 %) 81(11.49 %) 306(43.40 %) 701(99.43 %)
White matter rarefaction, N (%) 2.00E-04 5.90E-04
Mild 149(21.13 %) 10(1.42 %) 30(4.26 %) 124(17.59 %) 313(44.40 %)
Moderate 48(6.81 %) 1(0.14 %) 17(2.41 %) 51(7.23 %) 117(16.60 %)
None 89(12.62 %) 3(0.43 %) 18(2.55 %) 104(14.75 %) 214(30.35 %)
Severe 12(1.70 %) 3(0.43 %) 16(2.27 %) 30(4.26 %) 61(8.65 %)
Whole brain weight, N (%) 1176.74 ± 137.14 1134.76 ± 139.33 1181.82 ± 152.93 1202.00 ± 134.35 1187.33 ± 138.33
cerebral cortex atrophy, N (%) 1.10E-20 8.50E-20
Mild 141(20.00 %) 15(2.13 %) 23(3.26 %) 197(27.94 %) 376(53.33 %)
Moderate 118(16.74 %) 1(0.14 %) 41(5.82 %) 63(8.94 %) 223(31.63 %)
None 8(1.13 %) 1(0.14 %) 8(1.13 %) 49(6.95 %) 66(9.36 %)
Severe 31(4.40 %) 0 9(1.28 %) 0 40(5.67 %)
hippocampus atrophy, N (%) 1.40E-24 1.40E-23
Mild 105(14.89 %) 7(0.99 %) 29(4.11 %) 144(20.43 %) 285(40.43 %)
Moderate 105(14.89 %) 6(0.85 %) 27(3.83 %) 48(6.81 %) 186(26.38 %)
None 30(4.26 %) 4(0.57 %) 7(0.99 %) 111(15.74 %) 152(21.56 %)
Severe 58(8.23 %) 0 18(2.55 %) 6(0.85 %) 82(11.63 %)
lobar atrophy, N (%) 1.20E-04 5.00E-04
NO 252(35.74 %) 17(2.41 %) 69(9.79 %) 293(41.56 %) 631(89.50 %)
YES 46(6.52 %) 0 12(1.70 %) 16(2.27 %) 74(10.50 %)

While vascular pathology was nearly universal across all groups (99.43 % overall), white matter rarefaction severity differed significantly (p = 2.00E-04), with AD +CVD showing the highest proportion of severe rarefaction (19.8 %). Mean brain weight was lowest in the CVD group (1134.76 ± 139.33 g) and highest in controls (1202.00 ± 134.35 g). Cerebral cortical atrophy patterns varied significantly (p = 1.10E-20), with moderate-to-severe atrophy observed in 50.0 % of AD cases and 61.7 % of concomitant cases, compared to only 20.4 % of controls. Hippocampal atrophy showed the most significant group differences (p = 1.40E-24), with severe atrophy in 19.5 % of AD cases and 22.2 % of concomitant cases, but only 1.9 % of controls. Lobar atrophy was significantly more common (p = 1.20E-04) in AD (15.4 %) and concomitant groups (14.8 %) compared to controls (5.2 %), illustrating the extensive structural brain changes associated with Alzheimer's disease pathology.

3.4. Association between cerebrovascular pathology and Alzheimer's risk

CVD was significantly associated with increased risk of AD across all models (Supplementary materials: Table S1). In the unadjusted analysis (Model 1), CVD was associated with nearly 5-fold higher risk of AD (HR = 4.94, 95 % CI: 2.93-8.80, p = 1.01E-08). After adjustment for demographic factors and APOE genotype (Model 2), the association strengthened (HR = 5.71, 95 % CI: 3.31–10.39, p = 1.94E-09), with male gender (HR = 1.99, p = 8.99E-05) and APOE ε4 allele presence (one allele: HR = 3.20, p = 4.67E-10; two alleles: HR = 6.93, p = 6.80E-04) emerging as significant risk factors, while education showed a modest protective effect (HR = 0.93, p = 3.17E-02).

In the fully adjusted model (Model 3), which accounted for all demographic, genetic, and neuropathological variables, the association between CVD and AD risk further increased (HR = 7.72, 95 % CI: 3.88–16.16, p = 1.77E-08). This model revealed that beyond CVD, significant independent predictors of AD included age (HR = 1.03, p = 4.86E-02), male gender (HR=2.44, p = 5.54E-04), APOE ε4 carrier status (one allele: HR = 2.25, p = 9.27E-04), neuritic plaque burden (CREAD C1: HR = 2.76, p = 6.04E-03; CREAD C2: HR=4.37, p = 1.11E-03), moderate cerebral cortex atrophy (HR = 3.16, p = 2.04E-04), and severe hippocampal atrophy (HR = 8.91, p = 2.10E-05). Notably, the effect of APOE ε4 homozygosity was attenuated and no longer significant in the fully adjusted model, suggesting its effects may be mediated through the included neuropathological variables. These findings demonstrate that CVD remains a robust and significant risk factor for AD even after comprehensive adjustment for established demographic, genetic, and neuropathological factors, with the association strengthening rather than weakening with more extensive adjustment.

3.5. Correlation analysis of pathological markers and cognitive test performance

This comprehensive correlation analysis reveals distinct patterns of association between neuropathological markers and cognitive performance across diagnostic groups (Fig. 1). In the AD group, APOE ε4 allele showed the strongest correlations with cognitive measures (Eta = 0.937 for Boston Naming, p < 0.001), followed by significant associations between Braak staging and global cognitive status (CDR Global: Eta = 0.021, p < 0.001; CDR Sum: Eta = 0.026, p < 0.000). Amyloid pathology markers (CREAD, Thal) demonstrated modest but significant correlations with multiple cognitive domains. Structural measures including cerebral cortical atrophy (CDR Sum: Eta = 0.021, p < 0.001) and hippocampal atrophy (MMSE: Eta = 0.021, p < 0.001) were significantly associated with cognitive performance. Notably, whole brain weight correlated significantly with all cognitive measures in the AD group.

Fig. 1.

Fig 1

Relationship between cognitive test scores and neuropathological features in Alzheimer's disease and cerebrovascular disease.

In the CVD group, the pattern of associations differed substantially, with whole brain weight emerging as the strongest correlate of cognitive performance (Eta values 0.214–0.446, all p < 0.001). White matter rarefaction uniquely showed significant correlations with global cognitive measures (CDR Global: Eta = 0.070, p < 0.001; CDR Sum: Eta = 0.079, p < 0.001) and attention measures (digit span forward trials: Eta = 0.041, p = 0.001). Traditional AD markers showed minimal associations with cognition in the CVD group, except for CREAD with global measures (CDR Global: Eta = 0.032, p = 0.00392).

The concomitant AD +CVD group displayed a hybrid pattern with remarkably strong correlations between hippocampal atrophy and cognitive performance (CDR Sum: Eta = 0.173, p < 0.001; MMSE: Eta = 0.115, p < 0.001), cerebral cortical atrophy and global cognition (CDR Sum: Eta = 0.165, p < 0.001), and extraordinarily strong correlations between whole brain weight and all cognitive measures (Eta values 0.232–0.528, all p < 0.001). Both AD-specific pathology markers (Braak, CREAD) and vascular pathology measures (white matter rarefaction) showed significant correlations with cognition in this group.

3.6. Mediation analysis of cerebrovascular factors on cognitive performance in AD pathology

Vascular factors demonstrated substantial indirect effects on cognitive performance, with a negative impact on MMSE (−1.865) and positive effect on CDR Sum (1.502), indicating that cerebrovascular pathology primarily influences cognition through mediating mechanisms rather than direct pathways. White matter lesions showed parallel but weaker effects (MMSE: −0.298; CDR Sum: 0.223), with interaction effects mirroring white matter lesion patterns exactly, suggesting overlapping pathophysiological processes or synergistic mechanisms.

The analysis of exposure-mediator relationships (Supplementary materials: Table S2) revealed that vascular factors most strongly associated with amyloid pathology (CREAD: 1.378), followed by APOE genotype (0.565), hippocampal atrophy (0.482), and Thal amyloid plaques (0.456). This pattern supports significant cerebrovascular contributions to multiple AD pathological features. White matter lesions primarily impacted brain volume (−11.171), with considerably weaker associations with amyloid (CREAD: 0.230) and tau pathology (Braak: 0.165), indicating differential mechanisms between vascular factors and white matter pathology.

Examination of mediator-outcome relationships (Supplementary materials: Table S3) identified hippocampal atrophy as the strongest determinant of cognitive performance (MMSE: −1.333; CDR Sum: 1.083), followed by cerebral atrophy (MMSE: −0.821; CDR Sum: 1.108) and APOE genotype (MMSE: −0.950; CDR Sum: 0.692). Traditional AD biomarkers showed moderate (Braak, Thal) to minimal (CREAD) cognitive effects, suggesting structural brain changes may more directly influence cognitive outcomes than molecular pathology markers.

Pathway-specific analysis (Table 4) demonstrated that vascular factors impacted cognition predominantly through hippocampal atrophy (MMSE: −0.656; CDR Sum: 0.522), APOE genotype (MMSE: −0.536; CDR Sum: 0.391), and cerebral atrophy (MMSE: −0.294; CDR Sum: 0.397). White matter lesions operated through similar pathways but with substantially weaker effects. Notably, structural brain changes served as stronger mediators than traditional AD pathological markers, particularly for white matter lesions, highlighting the critical role of neuroanatomical integrity in the cerebrovascular-cognitive relationship.

Table 4.

Specific indirect effects of cerebrovascular pathology on cognitive outcomes through multiple mediating pathways.

Mediating pathway MMSE vascular indirect effect MMSE white matter indirect effect MMSE interaction indirect effect CDR sum vascular indirect effect CDR sum white matter indirect effect CDR sum interaction indirect effect
Through Braak Score −0.097 −0.091 −0.091 0.031 0.029 0.029
Through CREAD Score −0.113 −0.019 −0.019 −0.041 −0.007 −0.007
Through Thal Score −0.17 −0.042 −0.042 0.202 0.05 0.05
Through Brain Volume 0 −0.049 −0.049 0 0.041 0.041
Through Brain Atrophy −0.294 −0.061 −0.061 0.397 0.081 0.081
Through Hippocampal Atrophy −0.656 −0.063 −0.063 0.522 0.048 0.048
Through APOE −0.536 0.027 0.027 0.391 −0.02 −0.02
Total Indirect Effect −1.865 −0.298 −0.298 1.502 0.223 0.223

Our mediation analysis revealed complex relationships between vascular factors and cognitive outcomes (Supplementary materials: Table S4). For MMSE scores, vascular factors showed complete mediation (100.20 %) primarily through hippocampal atrophy (35.2 %), APOE genotype (28.8 %), and brain atrophy (15.8 %), while white matter lesions exhibited negative mediation via hippocampal and brain atrophy. Regarding CDR Sum outcomes, vascular factors' effects were substantially mediated (96.60 %) through hippocampal atrophy (34.7 %), brain atrophy (26.4 %), and APOE (26.0 %), whereas white matter lesions and their interaction with vascular factors displayed pronounced mediation effects (518.60 %), mainly through brain atrophy (188.4 %) and hippocampal atrophy (111.6 %). These findings highlight that vascular pathology influences cognitive function predominantly by inducing structural brain changes and interacting with genetic factors, with the substantial mediation percentages for CDR Sum outcomes emphasizing the crucial role of brain structural alterations in translating vascular pathology to clinical dementia severity.

4. Discussion

Our study demonstrates significant interactions between CVD and AD, with CVD substantially increasing AD risk. Structural equation modeling revealed that vascular factors influence cognitive performance primarily through hippocampal atrophy, APOE genotype, and cerebral atrophy. Participants with concomitant AD +CVD pathology displayed a distinctive hybrid pattern of brain-cognition relationships, with stronger correlations between hippocampal atrophy and cognitive performance compared to pure AD or CVD cases.

Recent clinical research has increasingly recognized the significance of mixed cerebrovascular and AD pathologies in aging populations [21,22]. Some study demonstrated that cerebrovascular pathology co-occurs with primary AD pathology in most cases of late-onset dementia, with neuroimaging studies showing that cerebrovascular lesions directly impact disease progression and cognitive expression [23]. This phenomenon may explain the variance in cognitive symptoms among individuals with similar amyloid and tau burdens [24,25]. A recent study further reinforced this relationship, showing that vascular burden interacts with Aβ accumulation to accelerate cognitive decline, particularly in individuals in early disease stages [26]. Their longitudinal analysis revealed that those with both vascular pathology and amyloid positivity showed faster progression to dementia than those with either pathology alone [27].

A study analyzed data from the National Alzheimer's Coordinating Center to examine associations between cerebrovascular disease and AD pathology with cognitive decline. They found that white matter hyperintensities, a common marker of small vessel cerebrovascular disease, independently predicted cognitive trajectory even after controlling for AD biomarkers [28]. Similarly, Morales et al. (2024) demonstrated that small vessel cerebrovascular disease was significantly associated with cognitive impairment in prospective Alzheimer's clinical trial participants [17], further supporting our findings that vascular pathology represents an important and potentially modifiable risk factor for AD-related cognitive decline.

The mechanistic relationships between CVD and AD pathologies appear to involve complex interactions between vascular factors, genetic susceptibility, and traditional AD biomarkers [29,30]. Our study found that the APOE ε4 allele is significantly more prevalent in both AD and concomitant AD +CVD groups compared to controls and pure CVD patients, suggesting shared genetic vulnerability. Recent evidence indicates that APOE plays a crucial role in both disorders through multiple mechanisms [31]. APOE ε4 leads to blood-brain barrier dysfunction preceding cognitive decline, establishing a potential vascular pathway through which this genetic variant increases AD risk [32,33]. Tau and apolipoprotein E modulate cerebrovascular tight junction integrity independent of cerebral amyloid angiopathy, suggesting that these proteins affect vascular function through multiple pathways [34,35].

The structural integrity of the hippocampus is critical for cognitive function, and its atrophy is considered a central feature of cognitive decline in AD and related disorders [36,37]. Existing studies have indicated that hippocampal atrophy is not only associated with the worsening of AD pathologies but is also closely related to cerebrovascular health [38,39]. Research shows that chronic ischemia and insufficient cerebral blood flow may lead to neuronal damage in the hippocampal region, thereby accelerating cognitive decline [40,41]. Moreover, cerebrovascular damage can promote neuroinflammation and oxidative stress, leading to neurodegeneration in the hippocampus [42,43].

As evidenced in this study, AD patients with cerebrovascular pathology often experience more significant hippocampal atrophy and its negative impact on cognitive performance. Hippocampal atrophy predicts declines in cognitive ability and suggests that hippocampal atrophy may be a primary mediating mechanism for the indirect effects of cerebrovascular pathology on cognition [44]. Our analyses further support this notion, indicating that hippocampal atrophy plays a particularly significant role in the relationship between cerebrovascular factors and cognitive decline. Changes in the structure of the hippocampus could serve as a pivotal target for future therapeutic interventions.

Individuals carrying the APOE ε4 allele may experience blood-brain barrier dysfunction, leading to increased risks of neuroinflammation and neuronal damage [45,46]. This phenomenon may cause these individuals to exhibit more pronounced symptoms in the early stages of cognitive decline [47]. Epidemiological studies have shown that APOE ε4 is associated with a more rapid rate of cognitive decline, and individuals with one or two APOE ε4 alleles typically demonstrate more significant cognitive deterioration in clinical presentations compared to those with other genotypes [48,49].

Additionally, research has found that the presence of APOE ε4 may affect the interactions with other pathological markers in the brain, such as beta-amyloid and tau proteins [50,51]. Specifically, APOE ε4 carriers may be more susceptible to structural brain damage related to vascular injury and Alzheimer's disease (e.g., hippocampal atrophy), which further affects their cognitive performance [52,53]. Therefore, when analyzing the relationship between small vessel disease and cognitive function, it is essential to consider the genotype of APOE ε4. This genotype is not only a hereditary risk factor for Alzheimer's disease but may also modulate the effects of other pathological markers on cognitive abilities by influencing brain structure and function.

The interaction between Aβ and cerebrovascular function represents another critical mechanism [54]. Aβ directly contributes to neurovascular dysfunction by impairing blood vessel reactivity, promoting inflammation, and accelerating vascular degeneration [55]. Aβ oligomers induce cerebrovascular dysfunction through pericyte-mediated endothelial damage, establishing a direct mechanistic link between this hallmark AD protein and vascular pathology [56]. Notably, this relationship appears bidirectional, as studies of cerebrovascular phenotypes in mouse models of AD reveal vascular alterations that not only precede but potentially accelerate both amyloid and tau deposition [57].

Tau pathology, while traditionally examined primarily within the context of AD, demonstrates significant and meaningful interactions with vascular factors. Research has identified distinct patterns of Aβ, tau, and APOE accumulation when comparing AD-tau with other tauopathies, with cerebrovascular factors actively modulating these complex relationships [[58], [59], [60]]. Our mediation analyses strongly support these observations, demonstrating that vascular factors correlate robustly with both tau (Braak staging) and amyloid (CREAD, Thal) pathologies. These findings collectively suggest that cerebrovascular disease may serve as a critical mediator of both core pathological processes underlying Alzheimer's disease progression [61].

Our findings highlight the critical role of structural brain changes, particularly hippocampal atrophy and generalized cerebral atrophy, in mediating the effects of vascular pathology on cognitive outcomes [62]. Specifically, our pathway-specific analysis demonstrated that hippocampal atrophy served as the strongest mediator of vascular effects on cognition, followed by cerebral atrophy and APOE genotype. These findings align with recent research demonstrating that blood-brain barrier dysfunction in AD leads to increased neuroinflammation, accelerated neurodegeneration, and consequent brain atrophy [63,64].

The preferential involvement of the hippocampus in these pathological processes explains the prominent memory deficits observed in our concomitant AD +CVD group. Hippocampal integrity serves as a critical mediator between various pathological processes and cognitive function, with both AD and cerebrovascular pathologies converging on this vulnerable structure [62,65]. The strong correlation between whole brain weight and all cognitive measures in our concomitant AD +CVD group further underscores the importance of preserving overall brain structural integrity in the face of mixed pathologies.

Neuroimaging studies have offered insights into these relationships. Multiple neuroimaging techniques in AD research and highlighted that structural changes detectable through MRI often precede clinical symptoms by several years [66,67], with mixed vascular and AD pathologies showing distinctive patterns of regional atrophy and white matter damage [68]. These structural changes represent the result of multiple upstream pathological processes, including amyloid deposition, tau aggregation, vascular damage, and neuroinflammation, making them powerful integrative biomarkers of disease progression and cognitive outcomes [[69], [70], [71]].

Our study has notable strengths, including a comprehensive assessment of participants through standardized clinical evaluations, neuropsychological testing, and neuropathological examinations, which provided valuable insights into the links between vascular factors and AD. We employed advanced statistical methods, such as structural equation modeling, to clarify the effects of vascular pathology on cognition, and we focused on specific cognitive domains to better understand these impacts. Additionally, the inclusion of traditional AD biomarkers and structural brain integrity measures allowed for a thorough evaluation of relevant pathways.

However, our research has limitations, such as a cross-sectional design that restricts causal inferences and temporal relationships, necessitating longitudinal studies. The small sample size for pure CVD cases (n = 17) relative to the AD (n = 298) and control (n = 309) groups may have limited statistical power. Moreover, our reliance on categorical assessments rather than continuous measures may have reduced sensitivity to detect subtle differences. Also, we did not assess relevant factors like inflammation and specific vascular biomarkers. Lastly, findings from a clinical cohort may not fully generalize to the broader aging population with mixed cerebrovascular and AD pathologies.

Our findings highlight a significant link between CVD and AD, emphasizing the need for an integrated clinical approach. Vascular factors can worsen cognitive decline, particularly through hippocampal atrophy. Clinicians should integrate cardiovascular assessments in cognitive evaluations and personalize treatment plans based on factors like APOE genotype. Lifestyle changes—such as better diet, physical activity, and smoking cessation—along with vascular-targeting medications, are crucial for neuroprotection. Ongoing neuropsychological assessments for at-risk patients will allow timely treatment adjustments. Understanding the interplay between CVD and AD enables healthcare providers to implement proactive strategies to prevent cognitive decline and improve the quality of life for aging individuals.

Future research should prioritize longitudinal designs to capture the dynamic relationships among vascular function, AD biomarkers, and cognitive performance over time. By conducting repeated assessments, researchers can elucidate the temporal sequence of events and their interactions within the context of aging. Incorporating additional biomarkers that reflect inflammation, oxidative stress, and blood-brain barrier integrity is crucial, as these factors may significantly influence the pathophysiology of AD and its association with vascular health. Understanding how these biological processes interconnect can provide deeper insights into potential interventions. Furthermore, implementing therapeutic studies focused on vascular health is essential to determine whether improving vascular function can alter the trajectory of AD progression. Such studies should evaluate the effects of lifestyle modifications, pharmacologic treatments, and other interventions on cognitive outcomes and AD biomarkers. Ultimately, this multifaceted approach will enhance our understanding of the mechanisms linking vascular health and neurodegeneration, paving the way for more effective strategies in preventing and managing AD.

5. Conclusion

Our findings demonstrate that cerebrovascular disease significantly increases the risk of Alzheimer's disease and substantially influences its clinical expression through multiple pathways, with structural brain changes serving as critical mediators of vascular effects on cognition. These results highlight the importance of addressing vascular health as an integral component of strategies to prevent and treat Alzheimer's disease and related cognitive disorders.

Abbreviation List

AD

Alzheimer's Disease

CVD

Cerebrovascular Disease

MCI

Mild Cognitive Impairment

APOE

Apolipoprotein E

CDR

Clinical Dementia Rating

CDR Global

Clinical Dementia Rating Global Score

CDR Sum

Clinical Dementia Rating Sum Score

MMSE

Mini-Mental State Examination

NACC

National Alzheimer's Coordinating Center

CREAD

Neuritic Plaque Assessment Score

Thal

Amyloid Deposition Staging

Amyloid Beta

SEM

Structural Equation Modeling

TMT

Trail Making Test

HR

Hazard Ratio

FDR

False Discovery Rate

CI

Confidence Interval

Declarations

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 STI2023-Major Projects (2021ZD0201801) and the Key and Dominant Discipline Construction Project of the Health Commission of Guizhou Province in 2023, China.

CRediT authorship contribution statement

Chao Tang: Data curation, Conceptualization. Yaqi Ding: Formal analysis, Data curation. Jiaxin Yang: Conceptualization. Dian He: Writing – review & editing, Conceptualization.

Declaration of competing interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgements

The NACC database is funded by NIA/NIH Grant U24 AG072122. NACC data are contributed by the NIA-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), 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.2025.100209.

Appendix. Supplementary materials

mmc1.docx (21.3KB, docx)

References

  • 1.Jack CR, Jr, Bennett D.A., Blennow K., Carrillo M.C., Dunn B., Haeberlein S.B., Holtzman D.M., Jagust W., Jessen F., Karlawish J., Liu E., Molinuevo J.L., Montine T., Phelps C., Rankin K.P., Rowe C.C., Scheltens P., Siemers E., Snyder H.M., Sperling R. NIA-AA research framework: toward a biological definition of Alzheimer's disease. Alzheimer's dement: J Alzheimer's Assoc. 2018;14(4):535–562. doi: 10.1016/j.jalz.2018.02.018. eng. Epub 2018/04/15Cited in: Pubmed; PMID 29653606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Montagne A., Nation D.A., Sagare A.P., Barisano G., Sweeney M.D., Chakhoyan A., Pachicano M., Joe E., Nelson A.R., D'Orazio L.M., Buennagel D.P., Harrington M.G., Benzinger T.L.S., Fagan A.M., Ringman J.M., Schneider L.S., Morris J.C., Reiman E.M., Caselli R.J., Chui H.C., Tcw J., Chen Y., Pa J., Conti P.S., Law M., Toga A.W., Zlokovic B.V. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature. 2020;581(7806):71–76. doi: 10.1038/s41586-020-2247-3. eng. Epub 2020/05/08Cited in: Pubmed; PMID 32376954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lao P., Young C.B., Ezeh C., Lacayo B., Seblova D., Andrews R.M., Gibbons L., Kraal A.Z., Turney I., Deters K.D., Dotson V., Manly J.J., Barnes L.L., Zahodne L.B. Loneliness, cerebrovascular and Alzheimer's disease pathology, and cognition. Alzheimer's dement: J Alzheimer's Assoc. 2024;20(10):7113–7123. doi: 10.1002/alz.14196. eng. Epub 2024/09/05Cited in: Pubmed; PMID 39234651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chin K.S., Holper S., Loveland P., Churilov L., Yassi N., Watson R. Prevalence of cerebral microbleeds in Alzheimer's disease, dementia with Lewy bodies and Parkinson's disease dementia: a systematic review and meta-analysis. Neurobiol Aging. 2024;134:74–83. doi: 10.1016/j.neurobiolaging.2023.11.006. eng. Epub 2023/11/26Cited in: Pubmed; PMID 38006706. [DOI] [PubMed] [Google Scholar]
  • 5.Bonomi C.G., Motta C., Di Donna M.G., Poli M., Nuccetelli M., Bernardini S., Mercuri N.B., Koch G., Martorana A. Age of onset moderates the effects of vascular risk factors on neurodegeneration, blood-brain-barrier permeability, and cognitive decline in Alzheimer's disease. Alzheimers Res Ther. 2024;16(1):248. doi: 10.1186/s13195-024-01617-2. eng. Epub 2024/11/17Cited in: Pubmed; PMID 39550595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kumar Nelson V., Jha N.K., Nuli M.V., Gupta S., Kanna S., Gahtani R.M., Hani U., Singh A.K., Abomughaid M.M., Abomughayedh A.M., Almutary A.G., Iqbal D., Al Othaim A., Begum S.S., Ahmad F., Mishra P.C., Jha S.K., Ojha S. Unveiling the impact of aging on BBB and Alzheimer's disease: factors and therapeutic implications. Ageing Res Rev. 2024;98 doi: 10.1016/j.arr.2024.102224. eng. Epub 2024/02/13Cited in: Pubmed; PMID 38346505. [DOI] [PubMed] [Google Scholar]
  • 7.Eisenmenger L.B., Peret A., Famakin B.M., Spahic A., Roberts G.S., Bockholt J.H., Johnson K.M., Paulsen J.S. Vascular contributions to Alzheimer's disease. Transl Res: J Lab Clin Med. 2023;254:41–53. doi: 10.1016/j.trsl.2022.12.003. eng. Epub 2022/12/19Cited in: Pubmed; PMID 36529160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chun M.Y., Lee T., Kim S.H., Lee H.S., Kim Y.J., Lee P.H., Sohn Y.H., Jeong Y., Chung S.J. Hypoperfusion in Alzheimer's disease-prone regions and dementia conversion in Parkinson's disease. Clin Nucl Med. 2024;49(6):521–528. doi: 10.1097/rlu.0000000000005211. eng. Epub 2024/04/08Cited in: Pubmed; PMID 38584352. [DOI] [PubMed] [Google Scholar]
  • 9.Fu P., Chen Y., Wu M., Bao B., Yin X., Chen Z., Zhang M. Effect of ferroptosis on chronic cerebral hypoperfusion in vascular dementia. Exp Neurol. 2023;370 doi: 10.1016/j.expneurol.2023.114538. eng. Epub 2023/09/15Cited in: Pubmed; PMID 37709116. [DOI] [PubMed] [Google Scholar]
  • 10.Andjelkovic A.V., Situ M., Citalan-Madrid A.F., Stamatovic S.M., Xiang J., Keep R.F. Blood-brain barrier dysfunction in normal aging and neurodegeneration: mechanisms, Impact, and treatments. Stroke. 2023;54(3):661–672. doi: 10.1161/strokeaha.122.040578. eng. Epub 2023/02/28Cited in: Pubmed; PMID 36848419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Valletta M., Vetrano D.L., Rizzuto D., Winblad B., Canevelli M., Andersson S., Dale M., Fredolini C., Fratiglioni L., Grande G. Blood biomarkers of Alzheimer's disease in the community: variation by chronic diseases and inflammatory status. Alzheimer's Dement: J Alzheimer's Assoc. 2024;20(6):4115–4125. doi: 10.1002/alz.13860. eng. Epub 2024/05/08.Cited in: Pubmed; PMID 38717935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jung E., Kim Y.E., Jeon H.S., Yoo M., Kim M., Kim Y.M., Koh S.H., Choi Y.K. Chronic hypoxia of endothelial cells boosts HIF-1α-NLRP1 circuit in Alzheimer's disease. Free Radic Biol Med. 2023;204:385–393. doi: 10.1016/j.freeradbiomed.2023.05.011. eng. Epub 2023/05/29Cited in: Pubmed; PMID 37245530. [DOI] [PubMed] [Google Scholar]
  • 13.Ishikawa H., Shindo A., Mizutani A., Tomimoto H., Lo E.H., Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab: Off J Int Soc Cereb Blood Flow Metab. 2023;43(2_suppl):18–36. doi: 10.1177/0271678x231154597. eng. Epub 2023/03/09Cited in: Pubmed; PMID 36883344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Youwakim J., Vallerand D., Girouard H. Neurovascular coupling in hypertension is impaired by IL-17A through oxidative stress. Int J Mol Sci. 2023;24(4) doi: 10.3390/ijms24043959. eng. Epub 2023/02/26Cited in: Pubmed; PMID 36835372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Arenaza-Urquijo E.M., Boyle R., Casaletto K., Anstey K.J., Vila-Castelar C., Colverson A., Palpatzis E., Eissman J.M., Kheng Siang Ng T., Raghavan S., Akinci M., Vonk J.M.J., Machado L.S., Zanwar P.P., Shrestha H.L., Wagner M., Tamburin S., Sohrabi H.R., Loi S., Bartrés-Faz D., Dubal D.B., Vemuri P., Okonkwo O., Hohman T.J., Ewers M., Buckley R.F. Sex and gender differences in cognitive resilience to aging and Alzheimer's disease. Alzheimer's Dement: J Alzheimer's Assoc. 2024;20(8):5695–5719. doi: 10.1002/alz.13844. eng. Epub 2024/07/05Cited in: Pubmed; PMID 38967222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chatterjee A., Lee S., Diaz V., Saloner R., Sanderson-Cimino M., deCarli C., Maillard P., Hinman J., Vossel K., Casaletto K.B., Staffaroni A.M., Paolillo E.W., Kramer J.H. Associations of cerebrovascular disease and Alzheimer's disease pathology with cognitive decline: analysis of the national Alzheimer's coordinating center uniform data set. Neurobiol Aging. 2024;142:1–7. doi: 10.1016/j.neurobiolaging.2024.06.002. eng. Epub 2024/07/19Cited in: Pubmed; PMID 39024720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Morales C.D., Cotton-Samuel D., Lao P.J., Chang J.F., Pyne J.D., Alshikho M.J., Lippert R.V., Bista K., Hale C., Edwards N.C., Igwe K.C., Deters K., Zimmerman M.E., Brickman A.M. Small vessel cerebrovascular disease is associated with cognition in prospective Alzheimer's clinical trial participants. Alzheimers Res Ther. 2024;16(1):25. doi: 10.1186/s13195-024-01395-x. eng. Epub 2024/02/03Cited in: Pubmed; PMID 38308344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chen S., Guo D., Zhu Y., Xiao S., Xie J., Zhang Z., Hu Y., Huang J., Ma X., Ning Z., Cao L., Cheng J., Tang Y. Amyloid β oligomer induces cerebral vasculopathy via pericyte-mediated endothelial dysfunction. Alzheimers Res Ther. 2024;16(1):56. doi: 10.1186/s13195-024-01423-w. eng. Epub 2024/03/13Cited in: Pubmed; PMID 38475929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Prosser L., Macdougall A., Sudre C.H., Manning E.N., Malone I.B., Walsh P., Goodkin O., Pemberton H., Barkhof F., Biessels G.J., Cash D.M., Barnes J. Predicting cognitive decline in older adults using baseline metrics of AD pathologies, cerebrovascular disease, and neurodegeneration. Neurology. 2023;100(8):e834–e845. doi: 10.1212/wnl.0000000000201572. eng. Epub 2022/11/11Cited in: Pubmed; PMID 36357185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.La Joie R., Visani A.V., Lesman-Segev O.H., Baker S.L., Edwards L., Iaccarino L., Soleimani-Meigooni D.N., Mellinger T., Janabi M., Miller Z.A., Perry D.C., Pham J., Strom A., Gorno-Tempini M.L., Rosen H.J., Miller B.L., Jagust W.J., Rabinovici G.D. Association of APOE4 and clinical variability in alzheimer disease with the pattern of Tau- and Amyloid-PET. Neurology. 2021;96(5):e650–e661. doi: 10.1212/wnl.0000000000011270. eng. Epub 2020/12/03Cited in: Pubmed; PMID 33262228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Badji A., Westman E. Cerebrovascular pathology in Alzheimer's disease: hopes and gaps. Psychiatry Res Neuroimaging. 2020;306 doi: 10.1016/j.pscychresns.2020.111184. eng. Epub 2020/09/21Cited in: Pubmed; PMID 32950333. [DOI] [PubMed] [Google Scholar]
  • 22.Biessels G.J. Alzheimer's disease, cerebrovascular disease and dementia: lump, split or integrate? Brain: J Neurol. 2022;145(8):2632–2634. doi: 10.1093/brain/awac228. eng. Epub 2022/07/19Cited in: Pubmed; PMID 35848864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fiford C.M., Sudre C.H., Young A.L., Macdougall A., Nicholas J., Manning E.N., Malone I.B., Walsh P., Goodkin O., Pemberton H.G., Barkhof F., Alexander D.C., Cardoso M.J., Biessels G.J., Barnes J. Presumed small vessel disease, imaging and cognition markers in the Alzheimer's disease neuroimaging initiative. Brain Commun. 2021;3(4):fcab226. doi: 10.1093/braincomms/fcab226. eng. Epub 2021/10/19Cited in: Pubmed; PMID 34661106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rennie A., Ekman U., Shams S., Rydén L., Samuelsson J., Zettergren A., Kern S., Oppedal K., Blanc F., Hort J., Garcia-Ptacek S., Antonini A., Lemstra A.W., Padovani A., Kramberger M.G., Rektorová I., Walker Z., Snædal J., Pardini M., Taylor J.P., Bonanni L., Granberg T., Aarsland D., Skoog I., Wahlund L.O., Kivipelto M., Westman E., Ferreira D. Cerebrovascular and Alzheimer's disease biomarkers in dementia with Lewy bodies and other dementias. Brain Commun. 2024;6(5):fcae290. doi: 10.1093/braincomms/fcae290. eng. Epub 2024/09/18Cited in: Pubmed; PMID 39291165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hijazi Z., Yassi N., O'Brien J.T., Watson R. The influence of cerebrovascular disease in dementia with Lewy bodies and Parkinson's disease dementia. Eur J Neurol. 2022;29(4):1254–1265. doi: 10.1111/ene.15211. eng. Epub 2021/12/20Cited in: Pubmed; PMID 34923713. [DOI] [PubMed] [Google Scholar]
  • 26.Rabin J.S., Pruzin J., Scott M., Yang H.S., Hampton O., Hsieh S., Schultz A.P., Buckley R.F., Hedden T., Rentz D., Johnson K.A., Sperling R.A., Chhatwal J.P. Association of β-Amyloid and vascular risk on longitudinal patterns of brain atrophy. Neurology. 2022;99(3):e270–e280. doi: 10.1212/wnl.0000000000200551. eng. Epub 2022/04/28Cited in: Pubmed; PMID 35473760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dark H.E., An Y., Duggan M.R., Joynes C., Davatzikos C., Erus G., Lewis A., Moghekar A.R., Resnick S.M., Walker K.A. Alzheimer's and neurodegenerative disease biomarkers in blood predict brain atrophy and cognitive decline. Alzheimers Res Ther. 2024;16(1):94. doi: 10.1186/s13195-024-01459-y. eng. Epub 2024/05/01Cited in: Pubmed; PMID 38689358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nagaraja N., DeKosky S., Duara R., Kong L., Wang W.E., Vaillancourt D., Albayram M. Imaging features of small vessel disease in cerebral amyloid angiopathy among patients with Alzheimer's disease. Neuroimage Clin. 2023;38 doi: 10.1016/j.nicl.2023.103437. eng. Epub 2023/05/29Cited in: Pubmed; PMID 37245492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Alkhalifa A.E., Al-Ghraiybah N.F., Odum J., Shunnarah J.G., Austin N., Kaddoumi A. Blood-brain barrier breakdown in Alzheimer's disease: mechanisms and targeted strategies. Int J Mol Sci. 2023;24(22) doi: 10.3390/ijms242216288. eng. Epub 2023/11/25Cited in: Pubmed; PMID 38003477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Love S., Miners J.S. Cerebrovascular disease in ageing and Alzheimer's disease. Acta Neuropathol. 2016;131(5):645–658. doi: 10.1007/s00401-015-1522-0. eng. Epub 2015/12/30Cited in: Pubmed; PMID 26711459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Liu C.C., Zhao J., Fu Y., Inoue Y., Ren Y., Chen Y., Doss S.V., Shue F., Jeevaratnam S., Bastea L., Wang N., Martens Y.A., Qiao W., Wang M., Zhao N., Jia L., Yamazaki Y., Yamazaki A., Rosenberg C.L., Wang Z., Kong D., Li Z., Kuchenbecker L.A., Trottier Z.A., Felton L., Rogers J., Quicksall Z.S., Linares C., Knight J., Chen Y., Kurti A., Kanekiyo T., Fryer J.D., Asmann Y.W., Storz P., Wang X., Peng J., Zhang B., Kim B.Y.S., Bu G. Peripheral apoE4 enhances Alzheimer's pathology and impairs cognition by compromising cerebrovascular function. Nat Neurosci. 2022;25(8):1020–1033. doi: 10.1038/s41593-022-01127-0. eng. Epub 2022/08/02Cited in: Pubmed; PMID 35915180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Blanchard J.W., Bula M., Davila-Velderrain J., Akay L.A., Zhu L., Frank A., Victor M.B., Bonner J.M., Mathys H., Lin Y.T., Ko T., Bennett D.A., Cam H.P., Kellis M., Tsai L.H. Reconstruction of the human blood-brain barrier in vitro reveals a pathogenic mechanism of APOE4 in pericytes. Nat Med. 2020;26(6):952–963. doi: 10.1038/s41591-020-0886-4. eng. Epub 2020/06/10Cited in: Pubmed; PMID 32514169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kirchner K., Garvert L., Kühn L., Bonk S., Grabe H.J., Van der Auwera S. Detrimental effects of ApoE ε4 on Blood-brain barrier integrity and their potential implications on the pathogenesis of Alzheimer's disease. Cells. 2023;12(21) doi: 10.3390/cells12212512. eng. Epub 2023/11/10Cited in: Pubmed; PMID 37947590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sun Y.Y., Wang Z., Huang H.C. Roles of ApoE4 on the pathogenesis in Alzheimer's disease and the potential therapeutic approaches. Cell Mol Neurobiol. 2023;43(7):3115–3136. doi: 10.1007/s10571-023-01365-1. eng. Epub 2023/05/25Cited in: Pubmed; PMID 37227619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liu C.C., Yamazaki Y., Heckman M.G., Martens Y.A., Jia L., Yamazaki A., Diehl N.N., Zhao J., Zhao N., DeTure M., Davis M.D., Felton L.M., Qiao W., Li Y., Li H., Fu Y., Wang N., Wren M., Aikawa T., Holm M.L., Oue H., Linares C., Allen M., Carrasquillo M.M., Murray M.E., Petersen R.C., Ertekin-Taner N., Dickson D.W., Kanekiyo T., Bu G. Tau and apolipoprotein E modulate cerebrovascular tight junction integrity independent of cerebral amyloid angiopathy in Alzheimer's disease. Alzheimer's Dement: J Alzheimer's Assoc. 2020;16(10):1372–1383. doi: 10.1002/alz.12104. eng. Epub 2020/08/23Cited in: Pubmed; PMID 32827351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chevalier C., Marizzoni M., Quattrini G., Lopizzo N., Albani D., Jovicich J., Cattaneo A., Frisoni G.B. Peripheral inflammation and AD-related hippocampal neurodegeneration in prodromal AD patients [Article] Alzheimer's Dement: J Alzheimer's Assoc. 2021;1215(17) doi: 10.1002/alz.053579. Suppl 3. [DOI] [Google Scholar]
  • 37.Xiao Y., Hu Y., Huang K. Atrophy of hippocampal subfields relates to memory decline during the pathological progression of Alzheimer's disease [Article] Front Aging Neurosci. 2023;0615(15) doi: 10.3389/fnagi.2023.1287122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Manco C., Cortese R., Leoncini M., Plantone D., Gentile G., Luchetti L., Zhang J., Di Donato I., Salvadori E., Poggesi A., Cosottini M., Mascalchi M., Federico A., Dotti M.T., Battaglini M., Inzitari D., Pantoni L., De Stefano N. Hippocampal atrophy and white matter lesions characteristics can predict evolution to dementia in patients with vascular mild cognitive impairment [Article] J Neurol Sci. 2024;0915(464) doi: 10.1016/j.jns.2024.123163. [DOI] [PubMed] [Google Scholar]
  • 39.Herawati A.A., Habibi A.S.Y., Pohan R.A. Hippocampal atrophy and white matter lesions as predictors of the transition from VMCI to vascular dementia: implications for early intervention [Article] J Neurol Sci. 2024;1215(467) doi: 10.1016/j.jns.2024.123307. [DOI] [PubMed] [Google Scholar]
  • 40.Ponirakis G., Elsotouhy A., Al Hamad H., Vattoth S., Petropoulos I.N., Khan A., Gad H., Al-Khayat F., Chandran M., Ramadan M., Elorrabi M., Gadelseed M., Tosino R., Gawhale P.V., Alobaidi M., Khan S., Manikoth P., Abdelrahim Y.H.M., Thodi N., Almuhannadi H., Al-Mohannadi S. Association of cerebral ischemia with corneal nerve loss and brain atrophy in MCI and dementia [Article] Front Neurosci. 2021;0615(15) doi: 10.3389/fnins.2021.690896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hase Y., Jobson D., Cheong J., Gotama K., Maffei L., Hase M., Hamdan A., Ding R., Polivkoski T., Horsburgh K., Kalaria R.N. Hippocampal capillary pericytes in post-stroke and vascular dementias and Alzheimer's disease and experimental chronic cerebral hypoperfusion [Article] Acta Neuropathol Commun. 2024;12(1):29. doi: 10.1186/s40478-024-01737-8. 0215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kang Y.C., Zhang L., Su Y., Li Y., Ren W.L., Wei W.S. MicroRNA-26b regulates the microglial inflammatory response in hypoxia/ischemia and affects the development of vascular cognitive impairment [Article] Front Cell Neurosci. 2018;0615(12):154. doi: 10.3389/fncel.2018.00154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shen X., Li M., Shao K., Li Y., Ge Z. Post-ischemic inflammatory response in the brain: targeting immune cell in ischemic stroke therapy [Article] Front Mol Neurosci. 2023;0615(16) doi: 10.3389/fnmol.2023.1076016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zhao W., Zhao L., Chang X., Lu X., Tu Y. Elevated dementia risk, cognitive decline, and hippocampal atrophy in multisite chronic pain. Proc Natl Acad Sci U.S.A. 2023;120(9) doi: 10.1073/pnas.2215192120. eng. Epub 2023/02/22Cited in: Pubmed; PMID 36802440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ding Y., Palecek S.P., Shusta E.V. iPSC-derived blood-brain barrier modeling reveals APOE isoform-dependent interactions with amyloid beta [Article] Fluids Barriers CNS. 2024;21(1):79. doi: 10.1186/s12987-024-00580-2. 1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dias I.H., Taiwo R., Ma D. The blood-brain barrier models to study apolipoprotein E genotypes in Alzheimer's disease [Article] Neural Regen Res. 2022;17(9):1973–1974. doi: 10.4103/1673-5374.331538. 0915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Montagne A., Nation D.A., Sagare A.P., Barisano G., Sweeney M.D., Chakhoyan A., Pachicano M., Joe E., Nelson A.R., D'Orazio L.M., Buennagel D.P., Harrington M.G., Benzinger T.L.S., Fagan A.M., Ringman J.M., Schneider L.S., Morris J.C., Reiman E.M., Caselli R.J., Chui H.C., Tcw J. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline [Article] Nature. 2020;581(7806):71–76. doi: 10.1038/s41586-020-2247-3. 0515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Merritt V.C., Lange R.T., Lippa S.M., Brickell T.A., Soltis A.R., Dalgard C.L., Gill J.M., French L.M. Apolipoprotein e (APOE) ε4 genotype influences memory performance following remote traumatic brain injury in U.S. military service members and veterans [Article] Brain Cogn. 2021;1115(154) doi: 10.1016/j.bandc.2021.105790. [DOI] [PubMed] [Google Scholar]
  • 49.Wooten T., Brown E., Sullivan D.R., Logue M.W., Fortier C.B., Fonda J.R., DeGutis J., Salat D.H., McGlinchey R., Milberg W., Esterman M. Apolipoprotein E (APOE) ε4 moderates the relationship between c-reactive protein, cognitive functioning, and white matter integrity [Article] Brain Behav Immun. 2021;0715(95):84–95. doi: 10.1016/j.bbi.2021.02.016. [DOI] [PubMed] [Google Scholar]
  • 50.Young C.B., Johns E., Kennedy G., Belloy M.E., Insel P.S., Greicius M.D., Sperling R.A., Johnson K.A., Poston K.L., Mormino E.C. APOE effects on regional tau in preclinical Alzheimer's disease [Article] Mol Neurodegener. 2023;18(1):1. doi: 10.1186/s13024-022-00590-4. 0104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lozupone M., Panza F. Impact of apolipoprotein E isoforms on sporadic Alzheimer's disease: beyond the role of amyloid beta [Article] Neural Regen Res. 2024;19(1):80–83. doi: 10.4103/1673-5374.375316. 0115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Marques V.B., Leal M.A.S., Mageski J.G.A., Fidelis H.G., Nogueira B.V., Vasquez E.C., Meyrelles S.D.S., Simões M.R., Dos Santos L. Chronic iron overload intensifies atherosclerosis in apolipoprotein E deficient mice: role of oxidative stress and endothelial dysfunction [Article] Life Sci. 2019;0915(233) doi: 10.1016/j.lfs.2019.116702. [DOI] [PubMed] [Google Scholar]
  • 53.Chandio B.Q., Villalon-Reina J.E., Nir T.M., Thomopoulos S.I., Feng Y., Benavidez S., Jahanshad N., Harezlak J., Garyfallidis E., Thompson P.M. Amyloid, tau, and APOE in Alzheimer's disease: impact on white matter tracts [Article] bioRxiv. 2024;0806 doi: 10.1101/2024.08.05.606560. null. [DOI] [PubMed] [Google Scholar]
  • 54.Greenberg S.M., Bacskai B.J., Hernandez-Guillamon M., Pruzin J., Sperling R., van Veluw S.J. Cerebral amyloid angiopathy and Alzheimer disease - one peptide, two pathways. Nat Rev Neurol. 2020;16(1):30–42. doi: 10.1038/s41582-019-0281-2. eng. Epub 2019/12/13Cited in: Pubmed; PMID 31827267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hong H., Hong L., Luo X., Zeng Q., Li K., Wang S., Jiaerken Y., Zhang R., Yu X., Zhang Y., Lei C., Liu Z., Chen Y., Huang P., Zhang M. The relationship between amyloid pathology, cerebral small vessel disease, glymphatic dysfunction, and cognition: a study based on Alzheimer's disease continuum participants. Alzheimers Res Ther. 2024;16(1):43. doi: 10.1186/s13195-024-01407-w. eng. Epub 2024/02/21Cited in: Pubmed; PMID 38378607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Taylor X., Clark I.M., Fitzgerald G.J., Oluoch H., Hole J.T., DeMattos R.B., Wang Y., Pan F. Amyloid-β (Aβ) immunotherapy induced microhemorrhages are associated with activated perivascular macrophages and peripheral monocyte recruitment in Alzheimer's disease mice. Mol Neurodegener. 2023;18(1):59. doi: 10.1186/s13024-023-00649-w. eng. Epub 2023/08/31Cited in: Pubmed; PMID 37649100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bryant A.G., Hu M., Carlyle B.C., Arnold S.E., Frosch M.P., Das S., Hyman B.T., Bennett R.E. Cerebrovascular senescence is associated with tau pathology in Alzheimer's disease. Front Neurol. 2020;11 doi: 10.3389/fneur.2020.575953. eng. Epub 2020/10/13Cited in: Pubmed; PMID 33041998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Han S.L., Ou Y.N., Han B.L., Guo H.H., Chi H.C., Huang Y.M., Wang H.F., Tan L. Total tau protein mediates the association of ischemic cerebrovascular disease with cognitive decline. J Alzheimer's dis: JAD. 2024;98(3):1133–1143. doi: 10.3233/jad-231093. eng. Epub 2024/04/05Cited in: Pubmed; PMID 38578896. [DOI] [PubMed] [Google Scholar]
  • 59.Koivumäki M., Ekblad L., Lantero-Rodriguez J., Ashton N.J., Karikari T.K., Helin S., Parkkola R., Lötjönen J., Zetterberg H., Blennow K., Rinne J.O., Snellman A. Blood biomarkers of neurodegeneration associate differently with amyloid deposition, medial temporal atrophy, and cerebrovascular changes in APOE ε4-enriched cognitively unimpaired elderly. Alzheimers Res Ther. 2024;16(1):112. doi: 10.1186/s13195-024-01477-w. eng. Epub 2024/05/19Cited in: Pubmed; PMID 38762725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lorenzini L., Maranzano A., Ingala S., Collij L.E., Tranfa M., Blennow K., Di Perri C., Foley C., Fox N.C., Frisoni G.B., Haller S., Martinez-Lage P., Mollison D., O'Brien J., Payoux P., Ritchie C., Scheltens P., Schwarz A.J., Sudre C.H., Tijms B.M., Verde F., Ticozzi N., Silani V., Visser P.J., Waldman A., Wolz R., Chételat G., Ewers M., Wink A.M., Mutsaerts H., Gispert J.D., Wardlaw J.M., Barkhof F. Association of vascular risk factors and cerebrovascular pathology with Alzheimer disease pathologic changes in individuals without dementia. Neurology. 2024;103(7) doi: 10.1212/wnl.0000000000209801. eng. Epub 2024/09/17 23:43Cited in: Pubmed; PMID 39288341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Menze I., Bernal J., Kaya P., Aki Ç., Pfister M., Geisendörfer J., Yakupov R., Coello R.D., Valdés-Hernández M.D.C., Heneka M.T., Brosseron F., Schmid M.C., Glanz W., Incesoy E.I., Butryn M., Rostamzadeh A., Meiberth D., Peters O., Preis L., Lammerding D., Gref D., Priller J., Spruth E.J., Altenstein S., Lohse A., Hetzer S., Schneider A., Fliessbach K., Kimmich O., Vogt I.R., Wiltfang J., Bartels C., Schott B.H., Hansen N., Dechent P., Buerger K., Janowitz D., Perneczky R., Rauchmann B.S., Teipel S., Kilimann I., Goerss D., Laske C., Munk M.H., Sanzenbacher C., Hinderer P., Scheffler K., Spottke A., Roy-Kluth N., Lüsebrink F., Neumann K., Wardlaw J., Jessen F., Schreiber S., Düzel E., Ziegler G. Perivascular space enlargement accelerates in ageing and Alzheimer's disease pathology: evidence from a three-year longitudinal multicentre study. Alzheimer's Res Ther. 2024;16(1):242. doi: 10.1186/s13195-024-01603-8. eng. Epub 2024/11/04Cited in: Pubmed; PMID 39482759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Johnson A.C. Hippocampal vascular supply and its role in vascular cognitive impairment. Stroke. 2023;54(3):673–685. doi: 10.1161/strokeaha.122.038263. eng. Epub 2023/02/28Cited in: Pubmed; PMID 36848422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Inoue Y., Shue F., Bu G., Kanekiyo T. Pathophysiology and probable etiology of cerebral small vessel disease in vascular dementia and Alzheimer's disease. Mol Neurodegener. 2023;18(1):46. doi: 10.1186/s13024-023-00640-5. eng. Epub 2023/07/12Cited in: Pubmed; PMID 37434208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cicognola C., Mattsson-Carlgren N., van Westen D., Zetterberg H., Blennow K., Palmqvist S., Ahmadi K., Strandberg O., Stomrud E., Janelidze S., Hansson O. Associations of CSF PDGFRβ with aging, blood-brain barrier damage, neuroinflammation, and alzheimer disease pathologic changes. Neurology. 2023;101(1):e30–e39. doi: 10.1212/wnl.0000000000207358. eng. Epub 2023/05/04Cited in: Pubmed; PMID 37137722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lee H., Fu J.F., Gaudet K., Bryant A.G., Price J.C., Bennett R.E., Johnson K.A., Hyman B.T., Hedden T., Salat D.H., Yen Y.F., Huang S.Y. Aberrant vascular architecture in the hippocampus correlates with tau burden in mild cognitive impairment and Alzheimer's disease. J Cereb Blood Flow Metab: Off J Int Soc Cereb Blood Flow Metab. 2024;44(5):787–800. doi: 10.1177/0271678x231216144. eng. Epub 2023/11/24Cited in: Pubmed; PMID 38000018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Albrecht D., Isenberg A.L., Stradford J., Monreal T., Sagare A., Pachicano M., Sweeney M., Toga A., Zlokovic B., Chui H., Joe E., Schneider L., Conti P., Jann K., Pa J. Associations between vascular function and tau PET are associated with global cognition and amyloid. J Neurosci: Off J Soc Neurosci. 2020;40(44):8573–8586. doi: 10.1523/jneurosci.1230-20.2020. eng. Epub 2020/10/14Cited in: Pubmed; PMID 33046556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Eloyan A., Thangarajah M., An N., Borowski B.J., Reddy A.L., Aisen P., Dage J.L., Foroud T., Ghetti B., Griffin P., Hammers D., Iaccarino L., Jack CR, Jr, Kirby K., Kramer J., Koeppe R., Kukull W.A., La Joie R., Mundada N.S., Murray M.E., Nudelman K., Rumbaugh M., Soleimani-Meigooni D.N., Toga A., Touroutoglou A., Atri A., Day G.S., Duara R., Graff-Radford N.R., Honig L.S., Jones D.T., Masdeu J., Mendez M.F., Musiek E., Onyike C.U., Rogalski E., Salloway S., Sha S., Turner R.S., Wingo T.S., Wolk D.A., Womack K., Beckett L., Gao S., Carrillo M.C., Rabinovici G., Apostolova L.G., Dickerson B., Vemuri P. White matter hyperintensities are higher among early-onset Alzheimer's disease participants than their cognitively normal and early-onset nonAD peers: longitudinal Early-onset Alzheimer's Disease Study (LEADS) Alzheimer's Dement: J Alzheimer's Assoc. 2023;19(Suppl 9):S89–s97. doi: 10.1002/alz.13402. Suppl 9eng. Epub 2023/07/26Cited in: Pubmed; PMID 37491599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhang J., Chen H., Wang J., Huang Q., Xu X., Wang W., Xu W., Guan Y., Liu J., Wardlaw J.M., Deng Y., Xie F., Li B. Linking white matter hyperintensities to regional cortical thinning, amyloid deposition, and synaptic density loss in Alzheimer's disease. Alzheimer's Dement: J Alzheimer's Assoc. 2024;20(6):3931–3942. doi: 10.1002/alz.13845. eng. Epub 2024/04/23Cited in: Pubmed; PMID 38648354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Preis L., Villringer K., Brosseron F., Düzel E., Jessen F., Petzold G.C., Ramirez A., Spottke A., Fiebach J.B., Peters O. Assessing blood-brain barrier dysfunction and its association with Alzheimer's pathology, cognitive impairment and neuroinflammation. Alzheimer's Res Ther. 2024;16(1):172. doi: 10.1186/s13195-024-01529-1. eng. Epub 2024/08/01Cited in: Pubmed; PMID 39085945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lin Z., Sur S., Liu P., Li Y., Jiang D., Hou X., Darrow J., Pillai J.J., Yasar S., Rosenberg P., Albert M., Moghekar A., Lu H. Blood-brain barrier breakdown in relationship to Alzheimer and vascular disease. Ann Neurol. 2021;90(2):227–238. doi: 10.1002/ana.26134. eng. Epub 2021/05/28Cited in: Pubmed; PMID 34041783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Moon Y., Jeon H.J., Han S.H., Min-Young N., Kim H.J., Kwon K.J., Moon W.J., Kim S.H. Blood-brain barrier breakdown is linked to tau pathology and neuronal injury in a differential manner according to amyloid deposition. J Cereb Blood Flow Metab: Off J Int Soc Cereb Blood Flow Metab. 2023;43(11):1813–1825. doi: 10.1177/0271678x231180035. eng. Epub 2023/06/07Cited in: Pubmed; PMID 37283062. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

mmc1.docx (21.3KB, docx)

Data Availability Statement

The data used in the present study are all publicly available at https://naccdata.org/requesting-data/nacc-data.


Articles from The Journal of Prevention of Alzheimer's Disease are provided here courtesy of Elsevier

RESOURCES