Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Sep 22.
Published in final edited form as: J Alzheimers Dis. 2022;90(2):461–473. doi: 10.3233/JAD-220755

Cognitive Resilience in Brain Health and Dementia Research

Mahesh S Joshi 1, James E Galvin 1,*
PMCID: PMC10515194  NIHMSID: NIHMS1923767  PMID: 36093713

Abstract

With the expected rise in Alzheimer’s disease and related dementias (ADRD) in the coming decades due to the aging population and a lack of effective disease-modifying treatments, there is a need for preventive strategies that may tap into resilience parameters. A wide array of resilience strategies has been proposed including genetics, socioeconomic status, lifestyle modifications, behavioral changes, and management of comorbid disease. These different strategies can be broadly classified as distinguishing between modifiable and non-modifiable risk factors, some of which can be quantified so that their clinical intervention can be effectively accomplished. A clear shift in research focus from dementia risk to addressing disease resistance and resilience is emerging that has provided new potential therapeutic targets. Here we review and summarize the latest investigations of resilience mechanisms and methods of quantifying resilience for clinical research. These approaches include identifying genetic variants that may help identify novel pathways (e.g., lipid metabolism, cellular trafficking, synaptic function, inflammation) for therapeutic treatments and biomarkers for use in a precision medicine-like regimen. In addition, innovative structural and molecular neuroimaging analyses may assist in detecting and quantifying pathological changes well before the onset of clinical symptoms setting up the possibility of primary and secondary prevention trials. Lastly, we summarize recent studies demonstrating the study of resilience in caregivers of persons living with dementia may have direct and indirect impact on the quality of care and patient outcomes.

Keywords: Alzheimer’s disease, biomarkers, brain health, cognition, cognitive reserve, dementia, neuroimaging, resilience


With the US population living longer with a greater life expectancy, the number of cases of Alzheimer’s disease and related dementias (ADRD) are expected to increase, roughly doubling every 20 years [1, 2]. While many older adults subjectively report changes in cognitive functioning as they age, some of these individuals will go on to develop objective cognitive decline first as mild cognitive impairment (MCI) and then ADRD when progressively declining cognitive performance interferes with everyday functioning. However, the risk, extent, degree, and progression of cognitive decline varies widely among individuals, leading to the fundamental question of why some individuals develop ADRD and other individuals, even those with similar risk factors do not. This variation can be ascribed to the presence of a collection of factors that could potentially alleviate or lessen the expression in the clinical manifestation of symptoms despite the presence of underlying neurodegenerative pathology. In other words, cognitive resilience (CR) is the ability of the brain to resist the effects of pathology and preserve or maintain function. CR could explain why some individuals do not appear to develop ADRD despite the presence of brain amyloid or other pathologies. It could explain why some individuals with MCI do not “convert” to ADRD or may even reverse. It could also explain why some individuals with ADRD progress more slowly than others. With the growing interest in primary, secondary, and tertiary prevention studies [3, 4], understanding CR could open new avenues of research.

Many risk factors have been associated with ADRD prevention efforts that include both modifiable (e.g., exposures, lifestyle, and social habits) and nonmodifiable (e.g., age, sex, genetics) risk factors with modifiable risk factors explaining up to 40% of the attributable risk [5]. More specifically, CR factors have been reported to include years of education, gender, social connectedness, diet, mindfulness, cognitive and physical activity [6]. Another concept frequently used to estimate resilience is the construct of cognitive reserve. It is defined as the reserve that actively compensates for AD pathology by maximizing performance through differential recruitment of brain networks. Cognitive reserve is a latent variable usually measured in some combination of education, cognitive performance, and/or occupational attainment [7-11]. Educational attainment is frequently reported as the number of years of formal schooling completed. However, the number of years of schooling may not be representative of the quality of the educational experience, and opportunities for advanced education may not be equal for all individuals. Occupational attainment may provide a more accurate measure of an individual’s practical cognitive reserve, as job opportunities and advancement may not necessarily match educational attainment [7, 8]. Other potential measures of cognitive reserve include neuropsychological testing of cognitive performance which often include an assessment of verbal IQ [9], although questions remain as to whether IQ tests have racial, ethnic and social biases [10]. To address these challenges, we created the Cognitive Reserve Unit Scale (Box 1) combining weighted scores of major occupation and educational attainment with more weight being placed on occupation [11]. The Cognitive Reserve Unit Scale provides a continuous measurement of CR that can be used for statistical analyses and comparisons across different individuals of different social, racial, ethnic, and geographic backgrounds.

Box 1. Cognitive Reserve Unit Scale (CRUS).

Box 1

Other research methods to determine CR have focused on neuroimaging techniques such as magnetic resonance imaging (MRI) or positron emission tomography (PET) [12] providing quantitative measurements of cortical volume, thickness, and surface areas or measurements of amyloid or tau pathology. Another method of measuring resilience is to study the burden of pathology at autopsy in individuals with and without a prior clinical diagnosis of dementia [13, 14]. However, these extensive research methods may not be practical in all research projects and cannot be utilized in a clinical setting so that an appropriate dementia prevention and/or treatment regimen can be designed and implemented. This can include primary prevention limiting incidence of new cases, secondary prevention limiting prevalence of cases, and tertiary prevention limiting disability in existing cases. A clear understanding and definition of CR may help in preventing ADRD by modification of known risk factors, health promotion, healthful behavioral change, and management of comorbid medical conditions. In this review, we address existing measures of CR and summarize a recently described quantitative measure of resilience that can be used in multiple research and clinical settings [6].

MEASURES OF COGNITIVE RESILIENCE

To date, many measures of CR are qualitative rather than quantitative instruments, with some of the quantitative measurements relying on measurements of postmortem pathology. Stoner et al. [15] developed two methods for people with early-stage dementia; a positive psychology outcome measure and Engagement and Independence in Dementia Questionnaire, which included four themes—hope, resilience, a sense of independence, and social engagement. Both methods exhibited acceptable internal consistency and convergent validity, but the sample size used to acquire data was relatively small. Yao et al. [16] designed the AD Cognitive Resilience (AD-CR) score, which is defined as the difference between observed and expected cognition based on the AD pathology. It is a stand-alone, individual-level quantification of cognitive resilience validated by demonstrating strong associations with known factors related to CR. They also have proposed a framework for predicting whether an individual will have a high or low AD-CR score using measures collected during premortem assessments. A notable drawback of this method is that AD-CR score cannot be calculated during an individual’s lifetime since it uses postmortem pathology. Bowles et al. [17] analyzed data from well-characterized cases that came to autopsy which had intermediate to high levels of AD pathology. They reported that individuals who had a college education and/or the absence of non-AD pathology (e.g., vascular lesions, Lewy bodies) had higher resiliency. This data was supported by earlier studies demonstrating a clear link between higher education level and lower risk of dementia [18]. They assessed the data using CERAD (Consortium to Establish a Registry for Alzheimer’s Disease) score as none, sparse, moderate, or frequent. It was also observed that cognitively resilient people had higher average brain weights than those of non-resilient individuals. An apparent shortcoming of their study is that cognitively resilient people were categorized as such only after their death due to its reliance on neuropathology.

A more clinically relevant approach would be a continuous scale of resilience measurement considering processes that occur throughout aging. Hohman et al. [19] used a different approach to quantify resilience by leveraging AD biomarkers and baseline brain aging outcomes, including left hippocampal and right hippocampal volumes as primary outcome measurements from the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Cognitive reserve was measured using experiences like educational and occupational attainment [20]. This reserve was based on the proposal that past life experiences can enhance the brain reserve. However, there could be differences in efficiency and capacity of individuals’ cognitive networks. Positive lifestyles (diet, exercise, cognitive activity) and positive behaviors (mindfulness, optimism, self-efficacy) are examples of modifiable factors that have been shown to increase resilience [21-24]. Several studies have documented benefits of exposure to greenspaces in improved cognitive functions in older adults [25-27]. These studies are based on the hypothesis that spending time in nature in the form of greenspaces may slow the cognitive decline [28]. In a cross-sectional analysis of walkable neighborhoods, participants who are APOE ε4 carriers had worse cognition-related outcomes compared with non-carriers [29].

In a systematic review and meta-analysis, Meng and colleagues identified a positive link between mid-life vascular risk factors and significantly increased risk of AD [30]. Using a mixed random effects regression model, Li et al. demonstrated that subjects with vascular risk factors had increased AD dementia [31] and active intervention for vascular risk factors may slow the progression of cognitive decline. Negative lifestyle choices (e.g., smoking, alcohol consumption) and behaviors (e.g., depression, anxiety) are examples of modifiable factors which adversely affect cognitive resilience. In a meta-analysis of prospective cohort studies, as compared to non-smokers, current smokers showed an increased risk of all-cause dementia and there was 34% increased risk for every 20 cigarettes per day [32]. Excessive alcohol consumption (≥14 drinks per week) was associated with lower cognitive scores in older adults with and without cognitive impairment [33, 34]. A recent interesting meta-analysis shed light on the effect of binge drinking on the health of oral microbiome, which in turn enhance AD pathogenesis. The enhanced pathogenesis appears to be mediated through the blood-brain barrier leakage [35]. In a study of socio-economic impact on dementia in southern Taiwan, Liu et al. have observed that illiteracy and being female was associated with increased risk of dementia [36]. Being older and perceived insufficient income [37], age and education [38] had strong impact on dementia.

NEUROIMAGING STUDIES OF RESILIENCE

Studies conducted using the ADNI data have contributed significantly to the understanding of AD progression and contribution of resilience and immune response. Since the significance of recording continuous pathophysiological changes that occur several years prior to symptom appearance was well appreciated, imaging these changes was helpful in determining spatial and temporal ordering of biomarker changes. Insel et al. [39] estimated the trajectories of regional amyloid-β (Aβ) and tau PET uptake as a function of time, and observed Aβ deposition in the posterior cingulate cortex and precuneus 5–10 years before the onset of symptoms. Further, early tau PET uptake was noted in inferior temporal lobes, amygdala, and inferior parietal lobes. Libowitz et al. [40] demonstrated that abnormal cerebrospinal fluid (CSF) Aβ42 was associated with smaller brain volumes in MRI, particularly in hippocampus and prefrontal cortex region.

Cerebrovascular diseases, manifested as white matter lesions, microinfarcts, and microhemorrhages, are increasingly believed to mediate not just vascular forms of dementia but also prominent in the brains of individuals with AD, and Lewy body dementia [41, 42]. The risk factors for cerebrovascular disease are also risk factors for ADRD, including hypertension [43, 44] and Type-2 diabetes mellitus [42]. Cerebrovascular disease and vascular risk factors may also directly affect cognition by hippocampal atrophy [45]. While neuroimaging can provide important clues to risk of ADRD and assist in differential diagnosis, at the present time there are no specific neuroimaging markers of CR.

FLUID BIOMARKER STUDIES OF RESILIENCE

Increasingly, fluid biomarkers such as Aβ42, total tau (t-tau), and phosphorylated tau (p-tau) can be measured in CSF and blood to aid in developing personalized diagnosis and treatments for ADRD [46-48]. The deposition of Aβ occurs over decades but well before the symptom appearance leading to the occurrence of individuals with amyloid deposition but normal cognition known as preclinical AD [49]. What is particularly interesting is that although many individuals will eventually progress to MCI and clinical AD, not all do. This suggests that there may be resilience factors that offer protection. As amyloid levels do not appear to specify clinical dementia and several conditions (e.g., vascular dementia, Lewy body dementia) can have amyloid deposition as a comorbid pathology, ratios of t-tau/Aβ42 and p-tau/Aβ42 may outperform individual biomarkers in distinguishing AD from healthy controls and other forms of dementia [50]. In addition, other fluid biomarkers, particularly those that involve synaptic and intracellular processes such as SNAP-25, YKL-40, TREM-2, BACE-1, IP-10, α-synuclein, and TDP-43 may help identify ADRD (Table 1). Many of these are known to mediate inflammatory pathways, which in turn activate Aβ synthesis and tau phosphorylation [51]. Recently, Tible et al. [52] studied CSF biomarkers to differentiate between AD and non-AD disorders. Among those biomarkers, SNAP-25aa40 had maximal discriminative power and could be a potential candidate to aid in the clinical diagnosis of ADRD. Because CSF requires an invasive procedure, blood samples may offer an easier and more readily acceptable way to collect and analyze biomarkers. However, at the present time, most plasma assays are still early in the validation process. For example, tau monoclonal antibody method was developed to detect very low levels of tau present in plasma of AD patients [53]. Since then, several other groups have confirmed sensitivity of immunoassay for the plasma tau measurements [54, 55]. More recently, a blood-based biomarker combining plasma Aβ40, Aβ42, Apolipoprotein E (APOE) proteotypes was developed by a commercial company (C2N, St Louis, MO) to provide an amyloid prediction score to distinguish brain amyloid positive individuals from brain amyloid negative individuals. Despite these advances in fluid biomarkers, there are no markers that currently define CR.

Table 1.

Proteins postulated to participate in cognitive resilience

Protein Chromosome Role in resilience Ref
α-Synuclein 4 Aggregation in neurons [88]
ATP binding cassette transporter (ABCA1) 9 Aβ deposition [89]
Phosphatidylinositol-binding clathrin assembly protein (PICALM) 11 Aβ clearance [90]
Ras-related protein Rab-10 (RAB10) 2 Lipid droplet degradation [91]
Disks large-associated protein 2 (DLGAP2) 8 Slower cognitive decline [60]
Klotho 13 Longer lifespan [92]
Restrictive element-1 silencing transcription factor (REST) 4 Repressor of pro-apoptotic genes [93]
ATPase phospholipid transporting 8A (ATP8B) 18 Bile acid homeostasis [57]
Sortillin related receptor 1 (SORL1) 11 Aβ degradation [62]
Triggering receptor expressed on myeloid cell like 2 (TREM2) 17 Inflammation attenuation [94]

GENETIC FACTORS ASSOCIATED WITH RESILIENCE

A large proportion (~40–50%) of cognitively normal individuals who die and come to autopsy have evidence of amyloid deposition in their brains suggesting that CR factors may come into play that offers protection from manifestation of clinical symptoms. The genetic factors contributing to the resilience to development of pathology are not well understood (Table 2). Identifying these factors may provide novel therapeutic targets for clinically treating AD and related diseases. Protective genes may function by attenuating effects of accumulating neuropathological burden, providing higher cognitive baseline, or mediating delayed onset of clinical disease. An example is the study showing presence of rare coding variants PLCG2, ABI3, and TREM 2 implicating innate immunity response observed in late onset AD [56]. Several genome wide association studies (GWAS) were conducted to identify protective or resilience genes. One of these studies [57] was performed by employing harmonized resilience metrices across cross-sectional A4 study and 3 longitudinal cohort studies of AD. They observed significant locus among participants with unimpaired cognition on the chromosome 18 upstream of ATP8B1, indicating a novel resilience gene along the bile acid metabolic pathway.

Table 2.

Genes postulated to mediate cognitive resilience

Gene Chromosome Role in resilience Ref
PLCG2 (Phospholipase C Gamma 2) 16 Immune cell function [95]
ABI3 (ABI gene family member 3) 17 Inflammation attenuation [96]
TREM2 (triggering receptor expressed on myeloid cells-2) 17 Inflammation attenuation [97]
Kdm6a (Lysine demethylase 6A) X Slower cognitive decline [58]
SORL1 (Sortilin Related Receptor 1) 11 Aβ degradation [98]
ABCA1 (ATP binding cassette subfamily A member 1) 9 Neutralizes Aβ aggregation [99, 100]
KL-VS (Lifespan extending variant) 13 Slower cognitive decline [101, 102]
BDNF (Brain derived neurotrophic factor) 11 Learning and memory [103, 104]
DLGAP2 (Disk large-associated protein 2) 8 Slower cognitive decline [60]
NLRP3 (NLR family pyrin domain containing 3) 1 Inflammasome [105]
MS4A6A (Membrane Spanning 4-Domains A6A) 11 Braak plaques [106]
RAB10 (member RAS oncogene family) 2 Lipid droplet degradation [91]

Male patients with AD tend to exhibit more extensive cognitive deficits and may die early than women [1]. In an interesting mouse model of AD expressing the human amyloid precursor protein (hAPP), the X chromosome affects AD-related vulnerability [58]. XY-hAPP mice which were genetically modified to develop either testicles or ovaries had worse cognitive deficits and greater mortality than XX-hAPP mice. Further, mice with an X chromosome with or without a Y chromosome (e.g., XY or XO) had worse outcomes than XX mice. The observed resilience was hypothesized to be due to the candidate gene Kdm6a that resists X-linked inactivation and may attenuate Aβ neurotoxicity. Genetic variation in human Kdm6a gene was associated with less cognitive decline in aging. Seto et al. [59] provide an exhaustive review of resilient genes that focus on genes involved in lipid metabolism, synaptic function and inflammation. For example, Dlgap2 (disk large associated protein 2) is a protective candidate gene in a genetically diverse mouse model of AD [60]. DLGAP 2 protein, that is part of DLGAP 2 family, has been linked to neurological and psychiatric disorders including schizophrenia, AD, and Parkinson’s disease [61]. A variant of SORL1 (sortillin-related receptor 1) gene was found to be protective and is a receptor for APOE [62] (Table 2).

QUANTIFYING RESILIENCE IN BRAIN HEALTH AND ADRD

We recently developed a resilience index (RI) to address the need for a brief, easy to score quantifiable measure of brain health [6] based on six modifiable lifestyle factors including physical activity, cognitive activity, dietary patterns, social engagements, mindfulness, and cognitive reserve that each individually were demonstrated to have effects on cognitive performance and risk of disease. The RI demonstrated moderate-to-strong correlations with clinical and cognitive measures and provided very good discrimination between healthy controls, MCI, and ADRD. Further, within group analyses demonstrated that healthy controls, MCI, and ADRD cases with high RI had better cognitive, functional, behavioral, and global outcomes than those with low RI. Lower RI scores could be used as a guidance for referring patients for a more extensive evaluation of modifiable factors and development of a personalized prevention plan. While the initial application was in a cross-sectional study of individuals at a tertiary academic center, a quantifiable measure of resilience could assist with the discovery and validation of imaging, fluid, and genetic biomarkers of resilience as well as providing a basis for primary, secondary, and tertiary prevention programs.

RESILIENCE IN CAREGIVERS OF PEOPLE LIVING WITH DEMENTIA

With the expected increase in number of people with dementia in the coming decades, there will be corresponding increased demand for caregivers. Secondly, with the progressive worsening of disease, the person is more dependent on the assistance of caregiver and thus the caregiver may undergo increased stress and decline in their mental health. The caregiving activities may include assistance with chores of daily living, medical care, transportation, management of behaviors, and concerns related to cognitive decline. They have reported high levels of stress, depression, and experiencing significantly increased suicidal thoughts. These ill effects on caregivers may in turn negatively impact the quality of care for individuals living with dementia. However, not all caregivers are impacted to the same level by the ill effects of caregiving, and not all aspects of caregiving are perceived as negative. Caregivers can report positive feelings about caregiving (e.g., family togetherness and the satisfaction of helping others). Other perceived benefits associated with caregiving may include the opportunity to give back; improved relationships; feeling good about the quality of care; serving as a role model for others; increased self-esteem; an enhanced sense of purpose; and feelings of pleasure and satisfaction. Individuals with higher positive views of caregiving may report less burden and depression. We created the positive and negative appraisals of caregiving (PANAC) scale to better understand the caregiving experience [63]. Of note, caregivers with more positive appraisals had higher resilience including mindfulness and social engagement. Therefore, it is important to document their CR with systematic quantitative measurement of its parameters not just for their own brain health but also to potentially impact the cognitive health of the individuals they care for. The significance of quantifying CR is appreciated considering existing tools based on self-reported outcomes from caregivers, that could be influenced by depression, caregiver strain or burden, of reduced quality of life. The outcomes of these systematic resilience studies will help in developing intervention strategies so that caregivers are more effective in providing care for people living with MCI and ADRD.

Diest and Greef [64] conducted a caregiver study in which adult children were caring for a parent with dementia. It used a cross-sectional survey research design and analyzed both quantitative and qualitative data. The quantitative data were acquired using Family Attachment Changeability Index (FACI8). The family adaptations as an indicator of resilience were optimal in those families receiving community support. In addition, financial stability had a positive impact on family adaptation. Most participants identified factors that promoted family connectedness for a sustained resilience.

Several studies, using qualitative analysis, have focused on CR domains that are beneficial to caregivers, namely personal mastery, self-efficacy, and coping. These resilience domains had direct effect on the physical health outcomes with beneficial effects on biomarkers and clinical markers of disease including blood pressure. For example, in a sample of 100 spousal AD caregivers [65] higher levels of self-efficacy were associated with significantly lower mean arterial pressure. Another study [66] examined the protective effects of personal mastery on the relationship between stress and plasma levels of Plasminogen activator inhibitor-1 (PAI-1), which is implicated in development of cardiovascular disease. The data analysis showed that when the mastery was low, the stress was markedly related to PAI-1 levels in in-home spousal caregivers of AD patients.

Recently, Zhou et al. [67] have proposed a comprehensive framework for promoting CR in caregivers as part of intervention, based on prior qualitative and quantitative studies by other investigators. The framework focuses on strengths rather than hardships and burden faced by caregivers. They have carried out thematic analysis and proposed 4 domains as part of the framework: 1) problem response behaviors; 2) self-growth behaviors; 3) help related behaviors; and 4) learning related behaviors. The framework provides an insight into how dementia caregivers learn and develop resilience related behaviors. However, there is a clear gap in the studies that acquire and analyze physiological parameters like, blood pressure, immunity, heart rate, cortisol, and gene expression profiles. Han et al. [68] using resilience framework have attempted to identify challenges and resilience opportunities in a group of caregivers for hospice patients with dementia. The study identified unique challenges including difficulty in treatment decision making, prolonged caregiving burden, death with dignity, and lack of public awareness. Resilience parameters identified were self-control and appraisal, self-care, using visual materials, and option to choose good care facilities. The authors anticipate their findings help in design of supportive interventions for caregivers or families caring for hospice patients with ADRD.

PROMOTING AND ENHANCING RESILIENCE

With limited availability of treatments for dementia, prevention studies have gained importance in recent years. A diverse array of interventions—mindfulness, performing arts, and nutrition—are being proposed and tested in both persons living with dementia and family caregivers with most focusing on potentially modifiable CR factors. Several studies document the benefits of mindfulness-based exercises. It is a skill that could be developed at any age with a minimal training regimen. An 8-week Mindfulness-Based Stress Reduction training was provided to 7 dyads (an early-stage dementia patient and caregiver) [69]. The analyzed data showed a noticeable increase in quality of life for caregivers and all participants exhibited large increases in mindfulness. Another review of mind-body interventions such as mindfulness, Tai Chi, and Qigong, in MCI [70] showed improved cognitive function, reduction in depression and lowered dementia risk in older adults. Multi-modal performing arts intervention in caregivers of mild to moderately severe dementia patients [71] demonstrated significant decline in burden. However, the benefits declined at the end of the study showing the need for continued participation in the intervention program.

Modifiable risk factors play a vital role in prevention of dementia. Dietary modification is one such risk factor that may facilitate lowering the dementia. Accumulating evidence from both observational and clinical trials point to a protective association between certain nutrients or food groups and cognitive outcomes. Benefits were observed with B vitamins [72], antioxidants [73], curcumin [74], food groups such as seafood [75], fruits and vegetables [76], and nuts [77]. A literature review [78] was conducted of elevated plasma homocysteine as a risk factor for development of cognitive decline, dementia, and AD. Intervention in a trial using homocysteine lowering treatment with B vitamins markedly slowed the cognitive decline. Regular physical activity and exercise may serve as non-pharmacological intervention in dementia and AD. In several studies, physical exercise enhanced resilience in people with dementia [79-81]. A combined exercise and cognitive training system were effective in enhancing concentration and improving cognitive function in 100 dementia patients [82].

In recent years, many systematic trials are being undertaken to study the effectiveness of preventive measures for cognitive impairment and disability. An example is the FINGER study (Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability), which is a multicenter randomized clinical trial comprising of 1200 independently living persons in Finland [83]. The intervention comprised of 4 components: 1) nutritional guidance; 2) physical exercise; 3) cognitive training and social activity; and 4) intensive monitoring and management of metabolic and cardiovascular risk factors. After 2 years, significant intervention effects were observed on the primary outcome (overall cognition), main cognitive secondary outcomes (executive functioning and processing speed) [84]. Their findings suggested a multidomain intervention may improve cognitive functioning in at-risk older population. The encouraging data from the FINGER study has prompted the launching of worldwide FINGERS network (WW-FINGERS), a consortium of 25 countries, which is the global network of multidomain lifestyle intervention trials for dementia risk reduction and prevention [85]. This ambitious undertaking may facilitate adaptation of the interventions globally and establish opportunities for joint initiatives. It aims to test initially whether FINGER-based protocols are feasible and effective in various populations with diverse cultural contexts. The study is based on the premise that interventions targeting several risk factors simultaneously may provide optimal outcomes in preventing ADRD. Another trial called US POINTER (The US Study to Protect Brain Health Through Lifestyle Intervention to Reduce Risk) is a multisite randomized clinical trial designed to evaluate whether different lifestyle interventions protect cognitive function over a 2-year period in older adults at increased risk for cognitive impairment and dementia [86]. The study is designed to follow a global composite score that will allow harmonization with FINGER study and its associated trials through WW-FINGERS. An ancillary study, The POINTER Imaging, was initiated [87] where participants undergo PET imaging to measure Aβ and tau burden, and MRI to measure brain morphometry, white matter hyperintensities, and microstructure and cerebral blood flow. The outcomes from these exhaustive intervention trials are still awaited.

CONCLUDING COMMENTS

There are several avenues available to measure CR in older adults including genetic data, neuroimaging, biomarkers, and clinical tools (i.e., Resilience Index) that can used in clinical research and potentially in clinical practice. While each of these tools provides a “different peek into a different window”, combining these different instruments may afford a unique opportunity to study CR and design prevention studies. This review described an array of factors that may positively and negatively influence cognitive resilience (Fig. 1). Many resilience parameters are quantifiable and can be used in a clinical setting so that clinicians can develop personalized dementia prevention regimens to support brain health. An abundance of research in the coming years—delineating modifiable and nonmodifiable factors—may pave the way for effective strategies addressing ever increasing need for ADRD treatment and prevention.

Fig. 1.

Fig. 1.

Factors with positive and negative impact on cognitive resilience. The positive factors include cognitive reserve (education and occupation), positive lifestyles (diet, exercise, and cognitive activity), positive behaviors (mindfulness, optimism, and self-sufficiency), genetics (kdm6a, TREM2), greenspace (early and mid-life exposure to natural greenery), and socio-demographics (younger and high income). The negative factors are vascular risk factors (hypertension, diabetes, and obesity), negative lifestyles (head injury, smoking, and alcohol consumption), negative behaviors (depression and anxiety), genetics (MS4A6A, BDNF), co-morbidities (multiple medical conditions), and socio-demographics (low income, old age, racial/ethnic differences).

ACKNOWLEDGMENTS

This study was supported by grants from the National Institute on Aging (R01 AG071514, R01 AG071514-S1, R56 AG074889, R01 NS101483, and R01 NS101483-S1), the Harry T. Mangurian Foundation, and the Leo and Anne Albert Charitable Trust. The funders had no role in decision to publish, or preparation of the manuscript.

Dr. Galvin and the University of Miami Miller School of Medicine are the creators and copyright holders for the Resilience Index, Quick Physical Activity Rating, Cognitive & Leisure Activity Scale, and Cognitive Reserve Unit Scale described in this review.

Authors’ disclosures available online (https://www.j-alz.com/manuscript-disclosures/22-0755r1).

REFERENCES

  • [1].(2022) 2022 Alzheimer’s disease facts and figures. Alzheimers Dement 18, 700–789. [DOI] [PubMed] [Google Scholar]
  • [2].Prince M, Guerchet M, Prina M (2013) The Global Impact of Dementia 2013-2050. Alzheimer’s Disease International, London. [Google Scholar]
  • [3].Galvin JE (2017) Prevention of Alzheimer’s disease: Lessons learned and applied. J Am Geriatr Soc 65, 2128–2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Hodes JF, Oakley CI, O’Keefe JH, Lu P, Galvin JE, Saif N, Bellara S, Rahman A, Kaufman Y, Hristov H, Rajji TK, Fosnacht Morgan AM, Patel S, Merrill DA, Kaiser S, Melendez-Cabrero J, Melendez JA, Krikorian R, Isaacson RS (2018) Alzheimer’s “prevention” vs. “risk reduction": Transcending semantics for clinical practice. Front Neurol 9, 1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A, Cohen-Mansfield J, Cooper C, Costafreda SG, Dias A, Fox N, Gitlin LN, Howard R, Kales HC, Kivimaki M, Larson EB, Ogunniyi A, Orgeta V, Ritchie K, Rockwood K, Sampson EL, Samus Q, Schneider LS, Selbaek G, Teri L, Mukadam N (2020) Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 396, 413–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Galvin JE, Kleiman MJ, Chrisphonte S, Cohen I, Disla S, Galvin CB, Greenfield KK, Moore C, Rawn S, Riccio ML, Rosenfeld A, Simon J, Walker M, Tolea MI (2021) The Resilience Index: A quantifiable measure of brain health and risk of cognitive impairment and dementia. J Alzheimers Dis 84, 1729–1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Avila JF, Renteria MA, Jones RN, Vonk JMJ, Turney I, Sol K, Seblova D, Arias F, Hill-Jarrett T, Levy SA, Meyer O, Racine AM, Tom SE, Melrose RJ, Deters K, Medina LD, Carrion CI, Diaz-Santos M, Byrd DR, Chesebro A, Colon J, Igwe KC, Maas B, Brickman AM, Schupf N, Mayeux R, Manly JJ (2021) Education differentially contributes to cognitive reserve across racial/ethnic groups. Alzheimers Dement 17, 70–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Zahodne LB, Mayeda ER, Hohman TJ, Fletcher E, Racine AM, Gavett B, Manly JJ, Schupf N, Mayeux R, Brickman AM, Mungas D (2019) The role of education in a vascular pathway to episodic memory: Brain maintenance or cognitive reserve? Neurobiol Aging 84, 109–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Chapko D, McCormack R, Black C, Staff R, Murray A (2018) Life-course determinants of cognitive reserve (CR) in cognitive aging and dementia - a systematic literature review. Aging Ment Health 22, 915–926. [DOI] [PubMed] [Google Scholar]
  • [10].Reynolds CR, Altmann RA, Allen DN (2021) The problem of bias in psychological assessment. In Mastering Modern Psychological Testing, Reynolds CR, Altmann RA, Allen DN, eds. Springer International Publishing, Cham, pp. 573–613. [Google Scholar]
  • [11].Galvin JE, Tolea MI, Moore C, Chrisphonte S (2020) The Number Symbol Coding Task: A brief measure of executive function to detect dementia and cognitive impairment. PLoS One 15, e0242233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Busatto GF, de Gobbi Porto FH, Faria DP, Squarzoni P, Coutinho AM, Garcez AT, Rosa PGP, da Costa NA, Carvalho CL, Torralbo L, de Almeida Hernandes JR, Ono CR, Brucki SMD, Nitrini R, Buchpiguel CA, Souza Duran FL, Forlenza OV (2020) In vivo imaging evidence of poor cognitive resilience to Alzheimer’s disease pathology in subjects with very low cognitive reserve from a low-middle income environment. Alzheimers Dement (Amst) 12, e12122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Bennett DA, Wilson RS, Boyle PA, Buchman AS, Schneider JA (2012) Relation of neuropathology to cognition in persons without cognitive impairment. Ann Neurol 72, 599–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Boyle PA, Yu L, Wilson RS, Schneider JA, Bennett DA (2013) Relation of neuropathology with cognitive decline among older persons without dementia. Front Aging Neurosci 5, 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Stoner CR, Orrell M, Long M, Csipke E, Spector A (2017) The development and preliminary psychometric properties of two positive psychology outcome measures for people with dementia: The PPOM and the EID-Q. BMC Geriatr 17, 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Yao T, Sweeney E, Nagorski J, Shulman JM, Allen GI (2020) Quantifying cognitive resilience in Alzheimer’s disease: The Alzheimer’s Disease Cognitive Resilience Score. PLoS One 15, e0241707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Aiello Bowles EJ, Crane PK, Walker RL, Chubak J, LaCroix AZ, Anderson ML, Rosenberg D, Keene CD, Larson EB (2019) Cognitive resilience to Alzheimer’s disease pathology in the human brain. J Alzheimers Dis 68, 1071–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Foverskov E, Glymour MM, Mortensen EL, Osler M, Okholm GT, Lund R (2020) Education and adolescent cognitive ability as predictors of dementia in a cohort of Danish men. PLoS One 15, e0235781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Hohman TJ, McLaren DG, Mormino EC, Gifford KA, Libon DJ, Jefferson AL, Alzheimer’s Disease Neuroimaging Initiative (2016) Asymptomatic Alzheimer disease: Defining resilience. Neurology 87, 2443–2450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Stern Y, Albert S, Tang MX, Tsai WY (1999) Rate of memory decline in AD is related to education and occupation: Cognitive reserve? Neurology 53, 1942–1947. [DOI] [PubMed] [Google Scholar]
  • [21].Andel R, Crowe M, Pedersen NL, Fratiglioni L, Johansson B, Gatz M (2008) Physical exercise at midlife and risk of dementia three decades later: A population-based study of Swedish twins. J Gerontol A Biol Sci Med Sci 63, 62–66. [DOI] [PubMed] [Google Scholar]
  • [22].Begdache L, Chaar M, Sabounchi N, Kianmehr H (2019) Assessment of dietary factors, dietary practices and exercise on mental distress in young adults versus matured adults: A cross-sectional study. Nutr Neurosci 22, 488–498. [DOI] [PubMed] [Google Scholar]
  • [23].Johnson DC, Thom NJ, Stanley EA, Haase L, Simmons AN, Shih PA, Thompson WK, Potterat EG, Minor TR, Paulus MP (2014) Modifying resilience mechanisms in at-risk individuals: A controlled study of mindfulness training in Marines preparing for deployment. Am J Psychiatry 171, 844–853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Kuszewski JC, Howe PRC, Wong RHX (2020) Evaluation of cognitive performance following fish-oil and curcumin supplementation in middle-aged and older adults with overweight or obesity. J Nutr 150, 3190–3199. [DOI] [PubMed] [Google Scholar]
  • [25].Brown SC, Perrino T, Lombard J, Wang K, Toro M, Rundek T, Gutierrez CM, Dong C, Plater-Zyberk E, Nardi MI, Kardys J, Szapocznik J (2018) Health disparities in the relationship of neighborhood greenness to mental health outcomes in 249,405 U.S. Medicare Beneficiaries. Int J Environ Res Public Health 15, 430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Cherrie MPC, Shortt NK, Mitchell RJ, Taylor AM, Redmond P, Thompson CW, Starr JM, Deary IJ, Pearce JR (2018) Green space and cognitive ageing: A retrospective life course analysis in the Lothian Birth Cohort 1936. Soc Sci Med 196, 56–65. [DOI] [PubMed] [Google Scholar]
  • [27].de Keijzer C, Tonne C, Basagana X, Valentin A, Singh-Manoux A, Alonso J, Anto JM, Nieuwenhuijsen MJ, Sunyer J, Dadvand P (2018) Residential surrounding greenness and cognitive decline: A 10-year follow-up of the Whitehall II Cohort. Environ Health Perspect 126, 077003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Hartig T, Mitchell R, de Vries S, Frumkin H (2014) Nature and health. Annu Rev Public Health 35, 207–228. [DOI] [PubMed] [Google Scholar]
  • [29].Cerin E, Rainey-Smith SR, Ames D, Lautenschlager NT, Macaulay SL, Fowler C, Robertson JS, Rowe CC, Maruff P, Martins RN, Masters CL, Ellis KA (2017) Associations of neighborhood environment with brain imaging outcomes in the Australian Imaging, Biomarkers and Lifestyle cohort. Alzheimers Dement 13, 388–398. [DOI] [PubMed] [Google Scholar]
  • [30].Meng XF, Yu JT, Wang HF, Tan MS, Wang C, Tan CC, Tan L (2014) Midlife vascular risk factors and the risk of Alzheimer’s disease: A systematic review and meta-analysis. J Alzheimers Dis 42, 1295–1310. [DOI] [PubMed] [Google Scholar]
  • [31].Li J, Wang YJ, Zhang M, Xu ZQ, Gao CY, Fang CQ, Yan JC, Zhou HD, Chongqing Ageing Study G (2011) Vascular risk factors promote conversion from mild cognitive impairment to Alzheimer disease. Neurology 76, 1485–1491. [DOI] [PubMed] [Google Scholar]
  • [32].Zhong G, Wang Y, Zhang Y, Guo JJ, Zhao Y (2015) Smoking is associated with an increased risk of dementia: A meta-analysis of prospective cohort studies with investigation of potential effect modifiers. PLoS One 10, e0118333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Koch M, Fitzpatrick AL, Rapp SR, Nahin RL, Williamson JD, Lopez OL, DeKosky ST, Kuller LH, Mackey RH, Mukamal KJ, Jensen MK, Sink KM (2019) Alcohol consumption and risk of dementia and cognitive decline among older adults with or without mild cognitive impairment. JAMA Network Open 2, e1910319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Lao Y, Hou L, Li J, Hui X, Yan P, Yang K (2021) Association between alcohol intake, mild cognitive impairment and progression to dementia: A dose-response meta-analysis. Aging Clin Exp Res 33, 1175–1185. [DOI] [PubMed] [Google Scholar]
  • [35].Yussof A, Yoon P, Krkljes C, Schweinberg S, Cottrell J, Chu T, Chang SL (2020) A meta-analysis of the effect of binge drinking on the oral microbiome and its relation to Alzheimer’s disease. Sci Rep 10, 19872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Liu CK, Lai CL, Tai CT, Lin RT, Yen YY, Howng SL (1998) Incidence and subtypes of dementia in southern Taiwan: Impact of socio-demographic factors. Neurology 50, 1572–1579. [DOI] [PubMed] [Google Scholar]
  • [37].Chaaya M, Phung K, Atweh S, El Asmar K, Karam G, Khoury RM, Ghandour L, Ghusn H, Assaad S, Prince M, Waldemar G (2018) Socio-demographic and cardiovascular disease risk factors associated with dementia: Results of a cross-sectional study from Lebanon. Prev Med Rep 9, 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Gavrila D, Antunez C, Tormo MJ, Carles R, Garcia Santos JM, Parrilla G, Fortuna L, Jimenez J, Salmeron D, Navarro C (2009) Prevalence of dementia and cognitive impairment in Southeastern Spain: The Ariadna study. Acta Neurol Scand 120, 300–307. [DOI] [PubMed] [Google Scholar]
  • [39].Insel PS, Mormino EC, Aisen PS, Thompson WK, Donohue MC (2020) Neuroanatomical spread of amyloid beta and tau in Alzheimer’s disease: Implications for primary prevention. Brain Commun 2, fcaa007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Libowitz MR, Wei K, Tran T, Chu K, Moncrieffe K, Harrington MG, King K (2021) Regional brain volumes relate to Alzheimer’s disease cerebrospinal fluid biomarkers and neuropsychometry: A cross-sectional, observational study. PLoS One 16, e0254332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].De Reuck J, Deramecourt V, Cordonnier C, Leys D, Pasquier F, Maurage CA (2013) Prevalence of cerebrovascular lesions in patients with Lewy body dementia: A neuropathological study. Clin Neurol Neurosurg 115, 1094–1097. [DOI] [PubMed] [Google Scholar]
  • [42].Moran C, Beare R, Wang W, Callisaya M, Srikanth V (2019) Type 2 diabetes mellitus, brain atrophy, and cognitive decline. Neurology 92, e823–e830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Skoog I, Lernfelt B, Landahl S, Palmertz B, Andreasson LA, Nilsson L, Persson G, Oden A, Svanborg A (1996) 15-year longitudinal study of blood pressure and dementia. Lancet 347, 1141–1145. [DOI] [PubMed] [Google Scholar]
  • [44].Skoog I, Gustafson D (2006) Update on hypertension and Alzheimer’s disease. Neurol Res 28, 605–611. [DOI] [PubMed] [Google Scholar]
  • [45].Wisse LEM, Das SR, Davatzikos C, Dickerson BC, Xie SX, Yushkevich PA, Wolk DA (2018) Defining SNAP by cross-sectional and longitudinal definitions of neurodegeneration. Neuroimage Clin 18, 407–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Dore V, Fowler C, Li QX, Martins R, Rowe C, Tomita T, Matsuzaki K, Ishii K, Ishii K, Arahata Y, Iwamoto S, Ito K, Tanaka K, Masters CL, Yanagisawa K (2018) High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature 554, 249–254. [DOI] [PubMed] [Google Scholar]
  • [47].Dage JL, Wennberg AMV, Airey DC, Hagen CE, Knopman DS, Machulda MM, Roberts RO, Jack CR Jr., Petersen RC, Mielke MM (2016) Levels of tau protein in plasma are associated with neurodegeneration and cognitive function in a population-based elderly cohort. Alzheimers Dement 12, 1226–1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Degerman Gunnarsson M, Ingelsson M, Blennow K, Basun H, Lannfelt L, Kilander L (2016) High tau levels in cerebrospinal fluid predict nursing home placement and rapid progression in Alzheimer’s disease. Alzheimers Res Ther 8, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Sperling RA, Jack CR Jr., Black SE, Frosch MP, Greenberg SM, Hyman BT, Scheltens P, Carrillo MC, Thies W, Bednar MM, Black RS, Brashear HR, Grundman M, Siemers ER, Feldman HH, Schindler RJ (2011) Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: Recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement 7, 367–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Fagan AM, Shaw LM, Xiong C, Vanderstichele H, Mintun MA, Trojanowski JQ, Coart E, Morris JC, Holtzman DM (2011) Comparison of analytical platforms for cerebrospinal fluid measures of beta-amyloid 1-42, total tau, and p-tau181 for identifying Alzheimer disease amyloid plaque pathology. Arch Neurol 68, 1137–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Bronzuoli MR, Iacomino A, Steardo L, Scuderi C (2016) Targeting neuroinflammation in Alzheimer’s disease. J Inflamm Res 9, 199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Tible M, Sandelius A, Hoglund K, Brinkmalm A, Cognat E, Dumurgier J, Zetterberg H, Hugon J, Paquet C, Blennow K (2020) Dissection of synaptic pathways through the CSF biomarkers for predicting Alzheimer disease. Neurology 95, e953–e961. [DOI] [PubMed] [Google Scholar]
  • [53].Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J, Hansson O (2013) Plasma tau levels in Alzheimer’s disease. Alzheimers Res Ther 5, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Karikari TK, Pascoal TA, Ashton NJ, Janelidze S, Benedet Aa L, Rodriguez JL, Chamoun M, Savard M, Kang MS, Therriault J (2020) Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: A diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol 19, 422–433. [DOI] [PubMed] [Google Scholar]
  • [55].Tatebe H, Kasai T, Ohmichi T, Kishi Y, Kakeya T, Waragai M, Kondo M, Allsop D, Tokuda T (2017) Quantification of plasma phosphorylated tau to use as a biomarker for brain Alzheimer pathology: Pilot case-control studies including patients with Alzheimer’s disease and down syndrome. Mol Neurodegener 12, 63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, Kunkle BW, Boland A, Raybould R, Bis JC, et al. (2017) Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet 49, 1373–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Dumitrescu L, Mahoney ER, Mukherjee S, Lee ML, Bush WS, Engelman CD, Lu Q, Fardo DW, Trittschuh EH, Mez J (2020) Genetic variants and functional pathways associated with resilience to Alzheimer’s disease. Brain 143, 2561–2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Davis EJ, Broestl L, Abdulai-Saiku S, Worden K, Bonham LW, Minones-Moyano E, Moreno AJ, Wang D, Chang K, Williams G, Garay BI, Lobach I, Devidze N, Kim D, Anderson-Bergman C, Yu G-Q, White CC, Harris JA, Miller BL, Bennett DA, Arnold AP, Jager PLD, Palop JJ, Panning B, Yokoyama JS, Mucke L, Dubal DB (2020) A second X chromosome contributes to resilience in a mouse model of Alzheimer’s disease. Sci Trans Med 12, eaaz5677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Seto M, Weiner RL, Dumitrescu L, Hohman TJ (2021) Protective genes and pathways in Alzheimer’s disease: Moving towards precision interventions. Mol Neurodegener 16, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Ouellette AR, Neuner SM, Dumitrescu L, Anderson LC, Gatti DM, Mahoney ER, Bubier JA, Churchill G, Peters L Huentelman MJ, Herskowitz JH, Yang HS, Smith AN, Reitz C, Kunkle BW, White CC, De Jager PL, Schneider JA, Bennett DA, Seyfried NT, Alzheimer’s Disease Genetics Consortium, Chesler EJ, Hadad N, Hohman TJ, Kaczorowski CC (2020) Cross-species analyses identify Dlgap2 as a regulator of age-related cognitive decline and Alzheimer’s dementia. Cell Rep 32, 108091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Rasmussen AH, Rasmussen HB, Silahtaroglu A (2017) The DLGAP family: Neuronal expression, function and role in brain disorders. Mol Brain 10, 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Taira K, Bujo H, Hirayama S, Yamazaki H, Kanaki T, Takahashi K, Ishii I, Miida T, Schneider WJ, Saito Y (2001) LR11, a mosaic LDL receptor family member, mediates the uptake of ApoE-rich lipoproteins in vitro. Arterioscler Thromb Vasc Biol 21, 1501–1506. [DOI] [PubMed] [Google Scholar]
  • [63].Galvin JE, Tolea MI, Chrisphonte S (2020) The positive and negative appraisals of caregiving (PANAC) scale: A new measure to examine the caregiving experience in Alzheimer’s disease and related dementias. Alzheimers Dement (N Y) 6, e12104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Deist M, Greeff AP (2017) Living with a parent with dementia: A family resilience study. Dementia (London) 16, 126–141. [DOI] [PubMed] [Google Scholar]
  • [65].Harmell AL, Mausbach BT, Roepke SK, Moore RC, von Kanel R, Patterson TL, Dimsdale JE, Mills PJ, Ziegler MG, Allison MA, Ancoli-Israel S, Grant I (2011) The relationship between self-efficacy and resting blood pressure in spousal Alzheimer’s caregivers. Br J Health Psychol 16, 317–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Mausbach BT, von Kanel R, Patterson TL, Dimsdale JE, Depp CA, Aschbacher K, Mills PJ, Ancoli-Israel S, Grant I (2008) The moderating effect of personal mastery and the relations between stress and Plasminogen Activator Inhibitor-1 (PAI-1) antigen. Health Psychol 27, S172–179. [DOI] [PubMed] [Google Scholar]
  • [67].Zhou Y, Ishado E, O’Hara A, Borson S, Sadak T (2021) Developing a unifying model of resilience in dementia caregiving: A scoping review and content analysis. J Appl Gerontol 40, 1377–1388. [DOI] [PubMed] [Google Scholar]
  • [68].Han S, Chi NC, Han C, Oliver DP, Washington K, Demiris G (2019) Adapting the resilience framework for family caregivers of hospice patients with dementia. Am J Alzheimers Dis Other Demen 34, 399–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Berk L, Warmenhoven F, Stiekema APM, van Oorsouw K, van Os J, de Vugt M, van Boxtel M (2019) Mindfulness-based intervention for people with dementia and their partners: Results of a mixed-methods study. Front Aging Neurosci 11, 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Farhang M, Miranda-Castillo C, Rubio M, Furtado G (2019) Impact of mind-body interventions in older adults with mild cognitive impairment: A systematic review. Int Psychogeriatr 31, 643–666. [DOI] [PubMed] [Google Scholar]
  • [71].McManus K, Tao H, Jennelle PJ, Wheeler JC, Anderson GA (2022) The effect of a performing arts intervention on caregivers of people with mild to moderately severe dementia. Aging Ment Health 26, 735–744. [DOI] [PubMed] [Google Scholar]
  • [72].Hooshmand B, Mangialasche F, Kalpouzos G, Solomon A, Kareholt I, Smith AD, Refsum H, Wang R, Muhlmann M, Ertl-Wagner B, Laukka EJ, Backman L, Fratiglioni L, Kivipelto M (2016) Association of vitamin B12, folate, and sulfur amino acids with brain magnetic resonance imaging measures in older adults: A longitudinal population-based study. JAMA Psychiatry 73, 606–613. [DOI] [PubMed] [Google Scholar]
  • [73].Summers WK, Martin RL, Liu Y, Pena B, Marsh GM (2018) Complex antioxidants in a randomized single-blinded study of memory in seniors. Aging Clin Exp Res 30, 395–405. [DOI] [PubMed] [Google Scholar]
  • [74].Voulgaropoulou SD, van Amelsvoort T, Prickaerts J, Vingerhoets C (2019) The effect of curcumin on cognition in Alzheimer’s disease and healthy aging: A systematic review of pre-clinical and clinical studies. Brain Res 1725, 146476. [DOI] [PubMed] [Google Scholar]
  • [75].Bakre AT, Chen R, Khutan R, Wei L, Smith T, Qin G, Danat IM, Zhou W, Schofield P, Clifford A, Wang J, Verma A, Zhang C, Ni J (2018) Association between fish consumption and risk of dementia: A new study from China and a systematic literature review and meta-analysis. Public Health Nutr 21, 1921–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Glabska D, Guzek D, Groele B, Gutkowska K (2020) Fruit and vegetable intake and mental health in adults: A systematic review. Nutrients 12, 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Theodore LE, Kellow NJ, McNeil EA, Close EO, Coad EG, Cardoso BR (2021) Nut consumption for cognitive performance: A systematic review. Adv Nutr 12, 777–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Smith AD, Refsum H, Bottiglieri T, Fenech M, Hooshmand B, McCaddon A, Miller JW, Rosenberg IH, Obeid R (2018) Homocysteine and dementia: An international consensus statement. J Alzheimers Dis 62, 561–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Lamb SE, Sheehan B, Atherton N, Nichols V, Collins H, Mistry D, Dosanjh S, Slowther AM, Khan I, Petrou S, Lall R, DAPA Trial Investigators (2018) Dementia And Physical Activity (DAPA) trial of moderate to high intensity exercise training for people with dementia: Randomised controlled trial. BMJ 361, k1675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Lamb SE, Mistry D, Alleyne S, Atherton N, Brown D, Copsey B, Dosanjh S, Finnegan S, Fordham B, Griffiths F, Hennings S, Khan I, Khan K, Lall R, Lyle S, Nichols V, Petrou S, Zeh P, Sheehan B (2018) Aerobic and strength training exercise programme for cognitive impairment in people with mild to moderate dementia: The DAPA RCT. Health Technol Assess 22, 1–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Bajwa RK, Goldberg SE, Van der Wardt V, Burgon C, Di Lorito C, Godfrey M, Dunlop M, Logan P, Masud T, Gladman J, Smith H, Hood-Moore V, Booth V, Das Nair R, Pollock K, Vedhara K, Edwards RT, Jones C, Hoare Z, Brand A, Harwood RH (2019) A randomised controlled trial of an exercise intervention promoting activity, independence and stability in older adults with mild cognitive impairment and early dementia (PrAISED) - A Protocol. Trials 20, 815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Okamura H, Otani M, Shimoyama N, Fujii T (2018)Combined exercise and cognitive training system for dementia patients: A randomized controlled trial. Dement Geriatr Cogn Disord 45, 318–325. [DOI] [PubMed] [Google Scholar]
  • [83].Kivipelto M, Solomon A, Ahtiluoto S, Ngandu T, Lehtisalo J, Antikainen R, Backman L, Hanninen T, Jula A, Laatikainen T, Lindstrom J, Mangialasche F, Nissinen A, Paajanen T, Pajala S, Peltonen M, Rauramaa R, Stigsdotter-Neely A, Strandberg T, Tuomilehto J, Soininen H (2013) The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER): Study design and progress. Alzheimers Dement 9, 657–665. [DOI] [PubMed] [Google Scholar]
  • [84].Ngandu T, Lehtisalo J, Solomon A, Levalahti E, Ahtiluoto S, Antikainen R, Backman L, Hanninen T, Jula A, Laatikainen T, Lindstrom J, Mangialasche F, Paajanen T, Pajala S, Peltonen M, Rauramaa R, Stigsdotter-Neely A, Strandberg T, Tuomilehto J, Soininen H, Kivipelto M (2015) A2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): A randomised controlled trial. Lancet 385, 2255–2263. [DOI] [PubMed] [Google Scholar]
  • [85].Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H, Baker L, Belleville S, Brodaty H, Brucki SM, Calandri I, Caramelli P, Chen C, Chertkow H, Chew E, Choi SH, Chowdhary N, Crivelli L, Torre R, Du Y, Dua T, Espeland M, Feldman HH, Hartmanis M, Hartmann T, Heffernan M, Henry CJ, Hong CH, Hakansson K, Iwatsubo T, Jeong JH, Jimenez-Maggiora G, Koo EH, Launer LJ, Lehtisalo J, Lopera F, Martinez-Lage P, Martins R, Middleton L, Molinuevo JL, Montero-Odasso M, Moon SY, Morales-Perez K, Nitrini R, Nygaard HB, Park YK, Peltonen M, Qiu C, Quiroz YT, Raman R, Rao N, Ravindranath V, Rosenberg A, Sakurai T, Salinas RM, Scheltens P, Sevlever G, Soininen H, Sosa AL, Suemoto CK, Tainta-Cuezva M, Velilla L, Wang Y, Whitmer R, Xu X, Bain LJ, Solomon A, Ngandu T, Carrillo MC (2020) World-Wide FINGERS Network: A global approach to risk reduction and prevention of dementia. Alzheimers Dement 16, 1078–1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Baker LD, Beavers DP, Cleveland M, Day CE, Decarli C, Espeland MA, Tomaszewski-Farias SE, Jimenez-Maggiora G, Katula J, Kivipelto M, Lambert K, Leng XI, Morris MC, Ngandu T, Papp KV, Raman R, Robertson J, Rushing S, Snyder HM, Solomon A, Su J, Ventrelle J, Williams B, Williamson JD, Whitmer RA, Woolard N, Carrillo MC (2019) O4-11-03:U.S. Pointer: Study Design and Launch. Alzheimers Dement 15, P1262–P1263. [Google Scholar]
  • [87].Lockhart SN, Baker LD, Carrillo MC, DeCarli C, Espeland MA, Harvey DJ, Jack CR, Jagust WJ, Jung Y, Koeppe RA, Lovato L, Snyder HM, Toga AW, Vemuri P, Woolard N, Landau SM (2020) U.S. POINTER Imaging: Study design and launch. Alzheimers Dement 16, e038414. [Google Scholar]
  • [88].Ghosh D, Mehra S, Sahay S, Singh PK, Maji SK (2017) alpha-synuclein aggregation and its modulation. Int J Biol Macromol 100, 37–54. [DOI] [PubMed] [Google Scholar]
  • [89].Fitz NF, Cronican AA, Saleem M, Fauq AH, Chapman R, Lefterov I, Koldamova R (2012) Abca1 deficiency affects Alzheimer’s disease-like phenotype in human ApoE4 but not in ApoE3-targeted replacement mice. J Neurosci 19, 13125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Xu W, Tan L, Yu J-T (2015) The role of PICALM in Alzheimer’s disease. Mol Neurobiol 52, 399–413. [DOI] [PubMed] [Google Scholar]
  • [91].Tavana JP, Rosene M, Jensen NO, Ridge PG, Kauwe JS, Karch CM (2019) RAB10: An Alzheimer’s disease resilience locus and potential drug target. Clin Interv Aging 14, 73–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP, Kuro-o M (2005) Suppression of aging in mice by the hormone Klotho. Science 309, 1829–1833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lu T, Aron L, Zullo J, Pan Y, Kim H, Chen Y, Yang T-H, Kim H-M, Drake D, Liu XS (2014) REST and stress resistance in ageing and Alzheimer’s disease. Nature 507, 448–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Benitez BA, Jin SC, Guerreiro R, Graham R, Lord J, Harold D, Sims R, Lambert JC, Gibbs JR, Bras J, Sassi C, Harari O, Bertelsen S, Lupton MK, Powell J, Bellenguez C, Brown K, Medway C, Haddick PC, van der Brug MP, Bhangale T, Ortmann W, Behrens T, Mayeux R, Pericak-Vance MA, Farrer LA, Schellenberg GD, Haines JL, Turton J, Braae A, Barber I, Fagan AM, Holtzman DM, Morris JC; 3C Study Group; EADI consortium; Alzheimer’s Disease Genetic Consortium (ADGC); Alzheimer’s Disease Neuroimaging Initiative (ADNI); GERAD Consortium, Williams J, Kauwe JS, Amouyel P, Morgan K, Singleton A, Hardy J, Goate AM, Cruchaga C (2014) Missense variant in TREML2 protects against Alzheimer’s disease. Neurobiol Aging 35, 1510 e1519–e1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Magno L, Lessard CB, Martins M, Lang V, Cruz P, Asi Y, Katan M, Bilsland J, Lashley T, Chakrabarty P, Golde TE, Whiting PJ (2019) Alzheimer’s disease phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. Alzheimers Res Ther 11, 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Karahan H, Smith DC, Kim B, Dabin LC, Al-Amin MM, Wijeratne HRS, Pennington T, Viana di Prisco G, McCord B, Lin PB, Li Y, Peng J, Oblak AL, Chu S, Atwood BK, Kim J (2021) Deletion of Abi3 gene locus exacerbates neuropathological features of Alzheimer’s disease in a mouse model of Abeta amyloidosis. Sci Adv 7, eabe3954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Raha AA, Henderson JW, Stott SR, Vuono R, Foscarin S, Friedland RP, Zaman SH, Raha-Chowdhury R (2017) Neuroprotective effect of TREM-2 in aging and Alzheimer’s disease model. J Alzheimers Dis 55, 199–217. [DOI] [PubMed] [Google Scholar]
  • [98].Yin RH, Yu JT, Tan L (2015) The role of SORL1 in Alzheimer’s disease. Mol Neurobiol 51, 909–918. [DOI] [PubMed] [Google Scholar]
  • [99].Elali A, Rivest S (2013) The role of ABCB1 and ABCA1 in beta-amyloid clearance at the neurovascular unit in Alzheimer’s disease. Front Physiol 4, 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Feher A, Giricz Z, Juhasz A, Pakaski M, Janka Z, Kalman J (2018) ABCA1 rs2230805 and rs2230806 common gene variants are associated with Alzheimer’s disease. Neurosci Lett 664, 79–83. [DOI] [PubMed] [Google Scholar]
  • [101].de Vries CF, Staff RT, Harris SE, Chapko D, Williams DS, Reichert P, Ahearn T, McNeil CJ, Whalley LJ, Murray AD (2017) Klotho, APOEepsilon4, cognitive ability, brain size, atrophy, and survival: A study in the Aberdeen Birth Cohort of 1936. Neurobiol Aging 55, 91–98. [DOI] [PubMed] [Google Scholar]
  • [102].Dubal DB, Yokoyama JS, Zhu L, Broestl L, Worden K, Wang D, Sturm VE, Kim D, Klein E, Yu GQ, Ho K, Eilertson KE, Yu L, Kuro-o M, De Jager PL, Coppola G, Small GW, Bennett DA, Kramer JH, Abraham CR, Miller BL, Mucke L (2014) Life extension factor klotho enhances cognition. Cell Rep 7, 1065–1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Cunha C, Brambilla R, Thomas KL (2010) A simple role for BDNF in learning and memory? Front Mol Neurosci 3, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].de Pins B, Cifuentes-Diaz C, Farah AT, Lopez-Molina L, Montalban E, Sancho-Balsells A, Lopez A, Gines S, Delgado-Garcia JM, Alberch J, Gruart A, Girault JA, Giralt A (2019) Conditional BDNF delivery from astrocytes rescues memory deficits, spine density, and synaptic properties in the 5xFAD mouse model of Alzheimer disease. J Neurosci 39, 2441–2458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Eren E, Ozoren N (2019) The NLRP3 inflammasome: A new player in neurological diseases. Turk J Biol 43, 349–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Karch CM, Jeng AT, Nowotny P, Cady J, Cruchaga C, Goate AM (2012) Expression of novel Alzheimer’s disease risk genes in control and Alzheimer’s disease brains. PLoS One 7, e50976. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES