1151

Journal of Alzheimer’s Disease 42 (2014) 1151–1163 DOI 10.3233/JAD-140213 IOS Press

Hypothesis

Adapting to Dementia in Society: A Challenge for Our Lifetimes and a Charge for Public Health Simon D’Altona,∗ , Sally Hunterb , Peter Whitehousec , Carol Brayneb and Daniel Georged a Center

for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA b Department of Public Health and Primary Care, Institute of Public Health, Forvie Site, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK c Case Western Reserve University, University Hospitals Case Medical Center, Cleveland, OH, USA d Department of Humanities, Penn State Milton S Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA

Accepted 2 May 2014

Abstract. For the last several decades, Alzheimer’s disease (AD) has been widely regarded as a late life event, but is now being redefined as a chronic condition that develops over decades. Concurrently, a preponderance of evidence emerging from basic and clinical research in diverse fields such as cardiovascular, endocrine, and mental health has suggested that the environmental component of clinical AD is not only multifactorial in populations and in individuals, but is also modifiable across the lifecourse, from conception until death. Re-conceptualizing the environmental component of AD to account for these observations necessitates an approach to brain health that eschews singular, short- and medium-term methodology and instead reflects longterm complexity. Such thinking is consistent with the ecological models of public health, which emphasize the development of community infrastructure that can foster population and individual health over the life-course by minimizing risk through multifaceted, systemic approaches. Keywords: Alzheimer’s disease, amyloid, dementia, epidemiology, public heath, risk factors, social policy

INTRODUCTION The progression that humanity has made toward greater longevity over the last century is a testament to many civil improvements in urban infrastructure, public healthcare, protective social policies, and medical science. In an isolated few instances, such as immunizations for lethal viral or bacterial ∗ Correspondence

to: Simon D’Alton, Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, 1275 Center Drive, University of Florida, Gainesville, FL32610, USA. Tel.: +1 352 294 5160; Fax: +1 352 294 5060; E-mail: [email protected].

infection, science has produced a protective solution. However, in the cases of modern chronic and non-transmittable illnesses—the so-called “diseases of civilization”—our bodily dysfunction remains resistant to curative treatments. Furthermore, variable sociocultural-environmental influences such as diet, exercise, built environments, and other factors heavily influence the prevalence of such conditions. While the detrimental effects of poor lifelong diet and physical activity patterns on the cardiovascular and endocrine systems is now a medical truism, it is increasingly clear that less intuitive connections may also exist, such as that between obesity or dia-

ISSN 1387-2877/14/$27.50 © 2014 – IOS Press and the authors. All rights reserved

1152

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

betes and many forms of cancer [1–5]. There is now a growing body of epidemiological evidence and in vivo animal studies to suggest that these late-life disorders are impacted by early-life experience, including in utero development and childhood [6–14]. Given these emergent signals from research, such age-associated conditions have increasingly been viewed through the broad prism of public health [15]. Generally, the modifiable nature of chronic disease is espoused by physicians, and public and private organizations are invested in health education and outreach campaigns to highlight the benefits of regular exercise and a healthy and balanced diet. These initiatives target people across the life-course, and complement research in molecular biology aimed at producing pharmacological interventions. Governments and their policies, not to mention international, national, regional, and local history, belief, and practice shape the way cultures approach the treatment of chronic disease. Closely linked with the rise of chronic disorders is the emergence of complex, age-related neurodegenerative conditions that result in cognitive decline in old age. In contrast with the chronic conditions mentioned above, attempts to promote a risk-reducing lifestyle within supportive physical and natural environments to reduce the incidence of Alzheimer’s disease (AD) have been more restrained, as demonstrated by the absence of any such recommendations in the recent ‘National Plan’ in the United States [16]. Unlike other chronic conditions, AD is not widely accepted as modifiable [17], nor has it been unequivocally viewed as a condition that can be ameliorated by public health approaches. Instead, since the late 20th century, a molecular paradigm has dominated the field and shaped the way the condition is classified, studied, and culturally perceived. The cynosure of AD research has been the amyloid cascade hypothesis, which posits that the toxic aggregation of amyloid-␤ (A␤) is responsible for a cascade of detrimental molecular events leading to tau neurofibrillary tangle formation, synaptic failure, and progressive dementia [18–20]. Pharmacological efforts have largely attempted to reduce the levels of aggregated A␤ species, but unfortunately have not met with success to date. These disappointing results have been interpreted as a failure to intervene early enough, and at present, there are multiple presymptomatic prevention trials taking place to address this. As part of the “Alzheimer’s Prevention Initiative”, researchers are conducting a $100 million study with an extended family of 5,000 people living in the mountains around Medell´in, Columbia, one-third of whom carry a rare genetic predisposition to young-onset AD. The goal

is to administer crenezumab to asymptomatic participants between ages 30–60, and evaluate whether it prevents progression along the continuum of cognitive decline [21]. Meanwhile, a similar approach is being taken with solanezumab, which, while having failed to reverse symptoms in people with dementia in 2013, demonstrated some cognitive benefits for a subgroup with mild memory loss [22]. This antibody is being used in both individuals with familial AD (in the Dominantly Inherited Alzheimer Network Trial Unit) and intellectually ‘normal’ at-risk people (in the “A4” study). While it is hoped that these and other pharmacological approaches will ultimately produce tangible benefits to society, it is evident that AD is not as amenable to current molecular biological approaches as was once hoped, and that alternative approaches will also be required. Historically, these approaches have not included an emphasis on environmental contributions to AD, principally due to the inconclusive nature of epidemiological studies and intervention trials targeting environmental risk factors. However, there is now an abundance of epidemiological, biochemical, and neuropathological evidence supporting the notion that that the environmental component of AD is composed of multiple, modifiable factors acting across the life-course. Importantly, this emerging paradigm points strongly to the implication that multi-faceted public health initiatives to promote biopsychosocially healthy lives could reduce AD risk and, in combination with future pharmaceutical approaches, improve quality of life in aging populations. This article will endeavor to introduce a new conceptual model for AD that is informed by the broad ecological framework of public health, and unpack some of the practical and policy implications stemming from the growing body of evidence regarding the multifactorality of AD at the individual and population level.

GENETIC AND EPIDEMIOLOGICAL STUDIES REVEAL MUCH ABOUT ENVIRONMENTAL INFLUENCE ON COGNITIVE HEALTH Quantitation of risk contribution from genetic and environmental components of AD is often the aim of twin studies. Concordance rates in mono- and dizygotic twins conclusively demonstrate that genes affect the clinical outcome of AD, with studies estimating a heritability of liability of approximately 60% [23–25].

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

These results support what is already highly intuitive: although a non-modifiable genetic contribution that influences clinical outcome is present from birth, AD is similar to other chronic conditions in that there is a malleable environmental input that may be altered to reduce total risk. An evidence-based example of this is found in ApoE4 positive individuals who, by remaining cognitively or physically active, may eliminate some of the additional risk derived from this allele [26–29]. It seems clear that long-term health is not merely determined by our genes, and we can partly define our cognitive health by modifying the environment. Epidemiological studies of this environmental component of clinical AD provide persuasive evidence that numerous, modifiable factors exist that can promote the development and maintenance of healthy brains, or contribute to neurodegeneration. The former category is principally the realm of what has been dubbed “cognitive and brain reserve”, a construct used to explain the apparently contradictory phenomenon that some individuals may present with a high burden of neuropathology but experience little clinical dementia. There are a range of inputs that may fall under the umbrella of ‘acquired reserve’ including larger brain size (which is itself comprised of genetic and environmental determinants) [30–33], years of formal education [34–36], a sense of purpose in life [37, 38], social engagement [39–44], and occupational complexity (reviewed in [45]). Meta-analyses of epidemiological studies further provide powerful evidence that numerous environmental/behavioral factors increase the relative risk of clinical AD by significant amounts [46–48]. Such factors include: midlife hypertension, obesity, diabetes, smoking, and physical inactivity, while psychosocial stress [49] and a late-life ‘hypoperfusive profile’ consisting of chronic heart failure and hypotension are more epidemiologically debatable [50, 51]. In concert, several of these factors are predictors of late-life dementia when present in midlife cohorts [52]. While many of these factors are correlated with clinical AD, fewer studies have attempted clinicopathological correlations, raising questions as to their true significance in the pathogenesis of AD.

NARROW BIOMEDICAL CLASSIFICATION OF AD STIFLES PROGRESS IN PUBLIC HEALTH APPROACHES Clinico-pathological studies have shown that the vast majority of individuals with any form of dementia

1153

have multiple, mixed pathologies [53, 54]. This heterogeneity presents a unique challenge for clinical and molecular science, as it prevents the simple categorization of individuals into singular neuropathologically confirmed diseases with simple linear mechanisms. This complexity is well chronicled in clinical AD, which is neuropathologically heterogeneous. It is now the rule rather than the exception to find concomitant pathologies such as hippocampal sclerosis, Lewy bodies, inclusions of TAR DNA binding protein of 43kDa (TDP-43), and vascular lesions, in addition to the classic A␤ plaque and neurofibrillary tangle pathology [55–59]. All of these pathological phenomena have been independently linked to neurodegeneration and neurological dysfunction. Indeed, as both vascular pathology and hippocampal sclerosis can generate the functional deficits commonly associated with plaques and tangles, A␤ and tau do not produce a unique AD symptomology [60–64]. Given the preponderance of vascular lesions and hippocampal sclerosis in AD, it seems reasonable to conclude that these lesions contribute to the difficulty in predicting underlying neuropathology in clinical AD and to the poor rate of concordance between clinical AD and plaque and tangle pathology [65]. Clearly, there are multiple pathways to neurodegeneration in individuals clinically diagnosed with AD, and these overlap frequently. Similarly, the classic neuropathological diagnosis of AD (i.e., the presence of plaques and tangles) is clinically heterogeneous. Although the cardinal clinical feature of AD is memory impairment, a primary neuropathological diagnosis of AD is frequently made in other syndromes such as posterior cortical atrophy and primary progressive aphasia [66, 67]. Furthermore, although AD is described as a slow, predictable decline in cognition [68, 69], the rate of conversion from preceding minor cognitive impairment phase to AD has yet to bear this out [70]. These observations demonstrate that AD is not explicitly definable as a singular entity, but rather is a catch-all term for a variety of pathologies and clinical manifestations occurring on a number of different trajectories. If the choice were made to label individuals with ‘Alzheimer’s disease’ only explicitly if they conformed to a narrow clinical phenotype purely involving mechanisms related to A␤ and tau, this would substantially reduce the prevalence of AD. Although there may be a connection between A␤- and tau-related pathologies, neurological dysfunction and memory impairment, this is not the same as saying that plaques and tangles are the sine qua non of AD. The borders delineating supposed singular categories have become blurred and inconsistent, perhaps

1154

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

rendering the application of distinct disease labels to populations and individuals an oversimplification. Adhering to the hitherto narrow biomedical definition of AD may have the unintended consequence of damaging public health. From a therapeutic standpoint, it suggests that all clinical AD can be effectively treated pharmacologically by focusing on pathways that converge on A␤, leaving a significant proportion of AD phenotype due to non-plaque and tangle pathology (e.g., vascular damage) untreated. Secondly, it may stifle the process of applying public health measures to reducing risk factors for AD. For instance, the notion that diabetes is not a risk factor for AD because it increases vascular pathology rather than traditional A␤ plaque pathology potentially discourages conversation regarding public health approaches and AD [71]. Similarly, individuals such as military servicemen exposed to head trauma are labeled with an entirely separate entity called chronic traumatic encephalopathy (CTE), even though CTE also results in increased abundance of A␤ plaques and tau tangles, and increased risk of clinical AD [72]. These examples demonstrate how the paradigmatic focus on creating clear-cut classificatory boundaries between the dementias can forestall conversation about commonsense preventive actions that might reduce risk for ‘AD’. Given the available evidence, choosing to link a narrow clinical term with the abundance of only one avenue of neurodegeneration is no longer an accurate reflection of the heterogeneity and complexity of ‘AD’, and mainly reflects historical convention and fealty to the dominant paradigmatic understandings rather than accumulated scientific progress.

ENVIRONMENTAL/BEHAVIORAL RISK FACTORS ARE SUPPORTED BY EPIDEMIOLOGICAL, NEUROPATHOLOGICAL, AND MOLECULAR EVIDENCE The neuropathology underlying clinical AD strongly supports a role for the cardiovascular risk factors mentioned above. Vascular pathology, which takes shape in the form of large infarcts, microinfarcts, lacunes, hemorrhages, atherosclerosis, arteriolosclerosis and cerebral amyloid angiopathy increases with age and is highly prevalent in neurodegenerative disease, but particularly in AD, where it contributes to clinical phenotype [55, 73–78]. Vascular abnormalities are driven by a combination of environmental risk factors that overlap with AD. Unequivocally, epidemiological

and supporting neuropathological evidence proves that the risk of manifesting AD symptomology is increased by these malleable risk factors that are heavily determined by physical activity and nutrition. A more contentious issue is: to what extent do modifiable factors relating to general health influence the dynamics of A␤ accumulation? Although the amyloid cascade hypothesis has attracted criticism—particularly in recent years following the consistent failure of anti-amyloid therapeutics—there is evidence that aggregated A␤ represents one source of neuronal toxicity in individuals with clinical AD. There is certainly a wealth of in vitro and in vivo data suggesting that conditions resulting from environmental insults such as hypoxia, metabolic disturbances, consequent oxidative stress, physical damage and chronic exposure to stress-related hormones cause alterations in A␤ processing [79–93]. Extrapolating these findings to the formation of human A␤ pathology is much harder. Although the endophenotypes that would be used as in situ evidence such as reduced brain perfusion, metabolic decline, and dysfunction of the stress pathways such as the hypothalamic-pituitary-adrenal axis all exist in AD, it is unclear whether these observations are causes or consequences of neuronal death or both [94–100]. It is possible that they are intertwined to such an extent that they are inseparable in human studies. However, it is unlikely that the rate of generation of A␤ is purely genetic, particularly in an age-related condition in which environmental factors and not an individual’s genetics change with time. Given their prevalence in the aged brain, and the in vitro and in vivo evidence for their ability to modulate A␤ metabolism, it is probable that environmental factors do play a role in highly complex amyloid-␤ protein precursor processing (A␤PP) and the rate of A␤ generation. Although in many instances the molecular pathways from external factor to A␤ have not been clearly delineated, A␤PP proteolysis is modulated by a multitude of molecular pathways which may be responsive individually or collectively as part of a coordinated, equilibrated system [101, 102]. What might be recognized, however, is that the significance of the connection between epidemiological risk and A␤ varies depending on the discipline. For the research communities that test the efficacy of either pharmaceutical or lifestyle interventions, A␤ burden may be a relevant outcome measure. However, from the vantage point of public health messaging and policy to promote the uptake of healthy activities across entire populations, the link between epidemiological risk fac-

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

tors and A␤ is less important. Put otherwise, regardless of what specific lesion is consequently found, lack of physical exercise, as well as obesity, diabetes, hypertension and so forth are undeniably associated with worse brain health. We know that a measurable proportion of the clinical AD phenotype is independent of A␤ and is due to factors such as vascular damage. This raises the issue as to what cultural and social changes might be made as part of a focused, concerted effort to reduce risk factors that are an unequivocal part of AD.

RESHAPING CULTURAL BELIEFS AND PRACTICES ABOUT “ALZHEIMER’S” A key element of improving health is cultural beliefs, which are formed not just by the paradigmatic construction and classification of knowledge but also by the dominant language patterns of the time. The most effective language to mobilize a population to adopt healthier, risk-averse behavior or to change public policy that may improve brain health is coherent, simple, repetitive and consistent. As Frank Luntz, the highly successful language consultant to the Republican Party, attests: “You say it again, and you say it again, and you say it again, and you say it again, and you say it again, and then again and again and again and again, and about the time that you’re absolutely sick of saying it is about the time that your target audience has heard it for the first time” [103]. For decades, consistent, repetitive, and simple amyloidocentric, dread disease metaphors, and imagery about AD have become prevalent in our cultural scripts (e.g., ‘mind robber’ and ‘dread disease’) and have contributed to a culture of fear and stigmatization around brain aging [104–106]. Of equal concern is that this messaging has culturally diminished the notion that risk for AD dementia is in any way modifiable. More recently, progressive, humanistic messaging attempting to increase participation in healthy lifestyles (‘healthy heart, healthy brain’) or reduce stigmatization (‘person-centered care for individuals with dementia’) has been marketed alongside disease-centered messaging, creating a psychologically confusing dichotomy in which AD is both a disease for which there is no hope and yet also part of natural health. Further confusion in the media and thus the public comes in the form of a bewildering array of medical jargon and non-intuitive, overlapping terminologies (i.e., AD, mixed dementia, vascular dementia, etc.) that are conventionally treated as distinct cate-

1155

gories. This is partly a reflection of the heterogeneity of AD. However, as long as AD continues to be treated in a strictly amyloidocentric manner, there is the danger that truly effective public relations efforts to increase engagement in healthier lives will be postponed. Given the current epidemiological data, which overwhelmingly demonstrates that lifelong active lifestyle is associated with reduced risk of clinical AD, it would seem that an opportunity to improve brain health is not being fully realized. If healthier brains are truly the goal, a coherent, simple, repetitive and consistent message of modifiability may help to foster cultural norms and practices that can engender healthier bodies and brains. Clearly, the genetic component of brain degeneration and the inability to forestall environmental changes in the brain forever guarantee that there will always be some cognitive decline in old age. Promising complete prevention to the public is also neither an intellectually honest nor an ethical act. However, current cultural scripts about AD are likewise unreflective of the heterogeneity of brain aging, and may actively preempt efficacious public health approaches. It is perhaps na¨ıve to surmise that the AD label will be fully discarded in the near future since it is so firmly entrenched in our cultural lexicon. However, it is possible that by unsettling the meaning of ‘AD’ and promoting a dynamic and scientifically accurate biopsychosocial public understanding of brain aging that is neither reductive nor fear-inducing, progress could be made towards encouraging healthier lives.

AD IS DIFFERENT FOR EVERYONE: THE PROBLEM OF MULTIFACTORALITY Fixating on the potential role of a single, individual genetic or environmental factor in the etiology of AD can often result in focusing on equally ‘singular’ causes of AD, for example, hypoperfusion [107] or type 3 diabetes [108]. However, the environmental component of AD is multifactorial at the population level, as epidemiological studies have attested. This population-level heterogeneity means that there is no ‘one-type-fitsall’ method that can be targeted throughout the entire population. Advocates of these environmentally-based factors would logically suggest that only the unique population affected (e.g., hypertensive or diabetic individuals) be included in treatment plans. While this is perhaps more likely to yield success from a therapeutic standpoint, it would only do so for a subgroup of individuals, and probably only if they could be identified early. However, even in the event of a hypothetical

1156

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

success in treatment, would lowering the number of smokers reduce the ‘societal burden’ of AD if, for instance, the level of obesity continues to rise and thereby offset this achievement? More importantly, the environmental component of AD is multifactorial in every single person, and is different for everyone. In consideration of this, it is perhaps illuminating to first draw comparisons with the genetic component of AD, in which massive Genome Wide Association Studies (GWAS) have been instrumental in identifying risk alleles in the general population. Effective collaborative work in this field involving consortiums pooling massive amounts of data into hugely powerful studies is unveiling the heritable component of AD [109, 110]. With the exception of the aforementioned ApoE4 allele, the majority of GWAS hits add considerably little risk individually; yet, in combination, their sum effect is powerful in individuals and in populations and, in total, comprises the genetics of AD. Crucially, on an individual level, it is readily accepted that genetic risk profiles are multifactorial (i.e., polygenic) and different combinations of genes exist in each individual. By the same logic, the undeniable environmental component of AD is comprised of innumerable potentially brain-influencing agents that individually may add little risk, but which, in combination, are a powerful force. Sets of risk factors are likely to be different for each individual, just as sets of genetic risk alleles vary amongst individual people. For example, there is evidence that A␤ is increased following exposure to anesthetic agents or surgical procedures, and postoperative dementia in older people is a documented consideration in surgical practice [111–117]. Is the generally healthy elder with mild insulin insensitivity and an ‘old’ heart who is undergoing a hip replacement to be considered to have the same risk profile as the 40-something chronically hypertensive banker who smokes, or the poorly educated man who runs four times a week? What about individuals who are in generally poor vascular health, but do not meet clinical thresholds for any of the risk factors, or the war veteran or contact-sport athlete who received multiple head injuries but remains in excellent cardiovascular health? Accepting that there are likely many environmental pathways that influence cognitive health, and that the magnitude and preponderance of each will vary between individuals removes much of the mystery of the non-genetic component of AD. Combining this multifactorality with the polygenic nature of AD explains the stochastic nature of the condition—(i.e.,

why ‘some get it and some don’t’)—and also provides a framework to explain the heterogeneous nature and clinicopathological spectrum of AD, and the differing ages of onset and progression of the clinical condition amongst the population. Furthermore, it can perhaps also evoke a sense of solidarity: that we may all differ in our individual risk profiles but share a common susceptibility to brain aging that is, in part, driven by socio-cultural factors present in our shared built and natural environments. Developing a unifying ethos of collective vulnerability around our highly prized but vulnerable organ—the brain—could be used to justify increased funding for public health approaches and community education efforts, and create an impetus for local, state, and national action that actively addresses shared risk factors within modern communities.

A CHRONIC TONIC FOR AD For decades, AD has been paradigmatically viewed by many as a late-life ‘event’. Recently, however, a plethora of scientific evidence has confirmed it as another of the chronic disorders that potentially affects every human being to some degree across the life course. As a result of imaging compounds such a Pittsburgh compound B and Florbetapir, it has been demonstrated that A␤ fibril formation occurs decades prior to the onset of symptoms, and A␤ deposition has been detected in individuals in their 20s [118, 119]. As mentioned above, this evidence has suggested that tardy delivery of therapeutics aimed at A␤ is a potential reason for their failure in recent clinical trials, and has refocused researchers earlier in the life-course toward prodromal and presymptomatic individuals [120]. Imaging and cerebrospinal fluid biomarkers are being tested for their potential to diagnose individuals earlier in the life-course, and are presently changing the nomenclature for AD to place individuals along a spectrum of disease: presymptomatic AD, mild cognitive impairment, and AD (e.g., [121]). In addition to shifting the clinical trials ‘upstream’ to target asymptomatic individuals, researchers openly discuss the possibility of pharmacological management of A␤ metabolism throughout the life-course [122]. In parallel with these biomedical-based efforts, an almost identical refocusing has occurred in non-pharmacological paradigms. Historically, interventions using physical exercise, diet, cognitive activity, and so forth have resulted in inconsistent outcomes when deployed as singular interventions for older people. Citing again that these approaches are

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

being employed too late in the disease process, work in this area has also moved upstream, and there are numerous trials being performed in cohorts of older people before any cognitive impairment [123, 124]. In acknowledgment of the multifactorality of the AD environmental component, some of these trials are also multidomain in nature, encompassing cardiovascular, cognitive, and social domains. This shift in approach raises the question of how far we might travel upstream in the life-course in our paradigmatic thinking about this chronic condition. From our perspective, it should invite further scientific and lay discussion of the consideration of how environmental factors affect the brain throughout life. Obesity, altered insulin sensitivity and cardiovascular factors are all modified by our experiences in utero, in childhood and beyond [6–10, 12, 13], and indeed our populations are entering a period in history in which we are seeing alarming rates of the relatively young developing these traditionally ‘age-associated’ chronic conditions. The quality of and access to education and healthcare resources is of deep concern to parents (and grandparents) who care passionately about the future of their children (and grandchildren). The natural extension of the logic guiding these multidomain interventions that are targeting cohorts earlier in the lifespan is to emphasize and create socially, physically, and cognitively stimulating environments in childhood and young adulthood and foster healthy prenatal environments. Indeed, there is already epidemiological evidence linking late-life cognitive function to our earliest experiences in the womb [125]. Thus, the prospect of “fixing” AD may not be achieved with any single intervention adopted temporarily or at end junctures of life. A complex, multifactorial, chronic condition like AD may require an equally ‘chronic tonic’. In other words, it may require societal policies and infrastructural investments that promote lifelong learning, enhanced educational opportunities for children and adults, more salutary built and natural environments, the establishment of patterns of lifelong healthy eating and exercise, greater social connectivity and cohesiveness, and the evolution of pharmacological and non-pharmacological caregiving approaches to help those who are more severely affected by brain changes that occur in later life. Relying on singular approaches in midlife or old age will always place a ceiling on the success of a given intervention. However, as evidenced by the heterogeneity of environmental risk factors and the chronic nature of AD, multiple approaches undertaken in combination and over long time periods, beginning early in life, will maximize

1157

one’s chances of avoiding insults to the brain and lowering susceptibility to the decrements of old age. Cognitive health, or factors that affect cognitive health in the elderly, are influenced throughout life; brain health does not begin at age 60, or at any other arbitrary starting point.

AD GETS AN MPH Faced with the looming crisis of a ‘silver tsunami’ as modern populations age, the World Health Organization has recently joined with Alzheimer’s Disease International to declare AD a “public health priority” [126]. Although this certainly brings immediacy to the current climate, it may also prompt us to ask not just how AD affects the public’s health, but how the public health field can affect AD. Making life-long changes to health is a complex challenge for a number of biological, behavioral, psychological, political, and economic reasons. For example, simple interventions to promote healthier lives through dietary education are likely to fail due to barriers that are economic (e.g., healthy foods are expensive), cultural (e.g., junk food advertising), biological (sugary, salty, and fatty foods are potentially more palatable and addictive), and structural (poor, economically disadvantaged, or badly structured neighborhoods can discourage exercise, play, and social interaction). To reflect this complexity, public health often employs ecological models that operate at individual, local, and broader cultural levels to minimize risk environments and positively reinforce salutary habits through many layered approaches [127]. Such methodology can mirror the high environmental complexity of AD, countering a complex population level and individual problem with an equally complex ‘chronic tonic’. Investing in safe walkable cities and play areas, modifying work environments to encourage less sedentary daily behavior, and improving sustained access to healthy foods across the socioeconomic spectrum benefits society as a whole and over long time frames. Given the robust evidence of its protective properties, education is itself a public health measure that could be tapped into by increasing access to stimulating learning environments for the young, the old and everyone in between, whether via provision of quality teachers or financial aid, university partnerships with nearby high schools or independent and assisted living communities, or online sharing of educational resources. Further, it has been established that such

1158

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

variables as social engagement, social connections, having a sense of purpose, and other interpersonal, community-based activities are, to some degree, cognitively protective for older people. Such a multifaceted approach moves away from the idea that cardiovascular fitness, highly simplistic ‘brain training games’ and so forth are sole interventions to be employed to singularly ‘stop AD’, and toward the notion that facilitating the creation of a physically healthier, cognitively stimulating, better society will have the additional, beneficial outcome of reduced incidence of AD and disease in general [128]. Such communities, bound by the recognition of shared vulnerability to brain aging, might also extend more compassion to the cognitively aged, and regard them with greater dignity, enabling older adults to remain purposefully interwoven into society rather than annexed in assisted living institutions with stigmatizing disease labels. Rather than being cultural shorthand for the fear, terror, and despair people feel about the aging process, finding solutions to “AD” could become a catalyst for improving many aspects of society. Combinations of these public health-oriented concepts are already operating synergistically in local communities around the world. One example is The Intergenerational School in Cleveland, Ohio, a public charter school in which elementary-aged children build relationships with elders who have been clinically diagnosed with dementia through reading together and making shared visits to cultural sites (i.e., art museums, nature centers, etc.), combining an educational environment with one that fosters greater social cohesiveness and protective social networks [129]. The model of the school is being replicated at other sites and in other countries, and similar learning environments forging intergenerational relationships are appearing more prevalently in many cultural settings [130, 131].

condition of aging can focus public and scientific attention upstream in the life-course. It is noticeable that medication and surgical techniques have improved greatly in the management and reduction in mortality of AD risk factors, although few if any can claim to be truly reversible. Thus, not only are our youngest generations accruing risk factors at earlier ages, but our brains are also being burdened by risk factors for longer periods as we age. These observations suggest that the instigation of multiple public health approaches that have impact on various risk factors throughout the lifespan may reduce the risk and progression of AD, but will perhaps only be successful if initiated as a part of a pattern of behavior over long time periods. In sum, AD is not just an issue for the times we live in, but is literally a challenge for each of our individual lifetimes. Although a challenge of immense proportions, it is no more so than the tremendous technical and economic difficulties that molecular biology is pressing hard to overcome. Adjusting paradigms to support a strong, integrated society that seeks and funds creative methods to promote physical and mental wellbeing of all may be a powerful approach to combating AD and enhancing quality of life in both the young and old. DISCLOSURE STATEMENT Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=2322). REFERENCES [1]

[2]

CONCLUSION For the last two decades, a predominantly molecular biological paradigm focused on a pharmaceutical approach to AD has instigated vast cultural changes in the perception of AD. Now, there is abundant evidence suggesting that the health of the brain is at least partially dependent on the rest of the body, interpersonal connections, and social structure. This evidence suggests that the environment’s contribution to AD risk is comprised of multiple factors, which are likely present to different degrees in different individuals. Furthermore, the reconceptualization of AD as a chronic

[3]

[4]

[5]

[6]

Collaborative Group on Epidemiological Studies of Ovarian Cancer (2012) Ovarian cancer and body size: Individual participant meta-analysis including 25,157 women with ovarian cancer from 47 epidemiological studies. PLoS Med 9, e1001200. Yang WS, Va P, Bray F, Gao S, Gao J, Li HL, Xiang YB (2011) The role of pre-existing diabetes mellitus on hepatocellular carcinoma occurrence and prognosis: A meta-analysis of prospective cohort studies. PLoS One 6, e27326. Wallin A, Larsson SC (2011) Body mass index and risk of multiple myeloma: A meta-analysis of prospective studies. Eur J Cancer 47, 1606-1615. Castillo JJ, Reagan JL, Ingham RR, Furman M, Dalia S, Merhi B, Nemr S, Zarrabi A, Mitri J (2012) Obesity but not overweight increases the incidence and mortality of leukemia in adults: A meta-analysis of prospective cohort studies. Leuk Res 36, 868-875. Deng L, Gui Z, Zhao L, Wang J, Shen L (2012) Diabetes mellitus and the incidence of colorectal cancer: An updated systematic review and meta-analysis. Dig Dis Sci 57, 15761585. Reilly JJ, Kelly J (2011) Long-term impact of overweight and obesity in childhood and adolescence on morbidity and

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

[7]

[8]

[9]

[10]

[11]

[12]

[13]

[14]

[15] [16] [17]

[18]

[19] [20]

[21]

[22]

[23]

premature mortality in adulthood: Systematic review. Int J Obes (Lond) 35, 891-898. Rich-Edwards JW, Kleinman K, Michels KB, Stampfer MJ, Manson JE, Rexrode KM, Hibert EN, Willett WC (2005) Longitudinal study of birth weight and adult body mass index in predicting risk of coronary heart disease and stroke in women. BMJ 330, 1115. Lawlor DA, Davey Smith G, Clark H, Leon DA (2006) The associations of birthweight, gestational age and childhood BMI with type 2 diabetes: Findings from the Aberdeen Children of the 1950s cohort. Diabetologia 49, 2614-2617. de Rooij SR, Painter RC, Roseboom TJ, Phillips DI, Osmond C, Barker DJ, Tanck MW, Michels RP, Bossuyt PM, Bleker OP (2006) Glucose tolerance at age 58 and the decline of glucose tolerance in comparison with age 50 in people prenatally exposed to the Dutch famine. Diabetologia 49, 637-643. Lumey LH, Stein AD, Kahn HS, Romijn JA (2009) Lipid profiles in middle-aged men and women after famine exposure during gestation: The Dutch Hunger Winter Families Study. Am J Clin Nutr 89, 1737-1743. Jimenez-Chillaron JC, Isganaitis E, Charalambous M, Gesta S, Pentinat-Pelegrin T, Faucette RR, Otis JP, Chow A, Diaz R, Ferguson-Smith A, Patti ME (2009) Intergenerational transmission of glucose intolerance and obesity by in utero undernutrition in mice. Diabetes 58, 460-468. Barker DJ, Eriksson JG, Forsen T, Osmond C (2002) Fetal origins of adult disease: Strength of effects and biological basis. Int J Epidemiol 31, 1235-1239. Burke JP, Forsgren J, Palumbo PJ, Bailey KR, Desai J, Devlin H, Leibson CL (2004) Association of birth weight and type 2 diabetes in Rochester, Minnesota. Diabetes Care 27, 2512-2513. Pentinat T, Ramon-Krauel M, Cebria J, Diaz R, JimenezChillaron JC (2010) Transgenerational inheritance of glucose intolerance in a mouse model of neonatal overnutrition. Endocrinology 151, 5617-5623. Fielding JE, Teutsch SM (2011) An opportunity map for societal investment in health. JAMA 305, 2110-2111. U.S. Department of Health and Human Services (2012) National Plan to Address Alzheimer’s Disease. Anderson LA, Day KL, Beard RL, Reed PS, Wu B (2009) The public’s perceptions about cognitive health and Alzheimer’s disease among the U.S. population: A national review. Gerontologist 49(Suppl 1), S3-11. Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 297, 353-356. Hardy J (2009) The amyloid hypothesis for Alzheimer’s disease: A critical reappraisal. J Neurochem 110, 1129-1134. Selkoe D, Mandelkow E, Holtzman D (2012) Deciphering Alzheimer disease. Cold Spring Harb Perspect Med 2, a011460. Mullard A (2012) Sting of Alzheimer’s failures offset by upcoming prevention trials. Nat Rev Drug Discov 11, 657660. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, Raman R, Sun X, Aisen PS, Siemers E, Liu-Seifert H, Mohs R (2014) Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med 370, 311-321. Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL (2006) Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry 63, 168-174.

[24]

[25] [26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40]

1159

Pedersen NL, Gatz M, Berg S, Johansson B (2004) How heritable is Alzheimer’s disease late in life? Findings from Swedish twins. Ann Neurol 55, 180-185. Bergem AL (1994) Heredity in dementia of the Alzheimer type. Clin Genet 46, 144-149. Obisesan TO, Umar N, Paluvoi N, Gillum RF (2012) Association of leisure-time physical activity with cognition by apolipoprotein-E genotype in persons aged 60 years and over: The National Health and Nutrition Examination Survey (NHANES-III). Clin Interv Aging 7, 35-43. Carlson MC, Helms MJ, Steffens DC, Burke JR, Potter GG, Plassman BL (2008) Midlife activity predicts risk of dementia in older male twin pairs. Alzheimers Dement 4, 324-331. Niti M, Yap KB, Kua EH, Tan CH, Ng TP (2008) Physical, social and productive leisure activities, cognitive decline and interaction with APOE-epsilon 4 genotype in Chinese older adults. Int Psychogeriatr 20, 237-251. Schuit AJ, Feskens EJ, Launer LJ, Kromhout D (2001) Physical activity and cognitive decline, the role of the apolipoprotein e4 allele. Med Sci Sports Exerc 33, 772-777. Perneczky R, Alexopoulos P, Wagenpfeil S, Bickel H, Kurz A (2012) Head circumference, apolipoprotein E genotype and cognition in the Bavarian School Sisters Study. Eur Psychiatry 27, 219-222. Perneczky R, Wagenpfeil S, Lunetta KL, Cupples LA, Green RC, Decarli C, Farrer LA, Kurz A (2010) Head circumference, atrophy, and cognition: Implications for brain reserve in Alzheimer disease. Neurology 75, 137-142. Mortimer JA, Snowdon DA, Markesbery WR (2003) Head circumference, education and risk of dementia: Findings from the Nun Study. J Clin Exp Neuropsychol 25, 671-679. Eyler LT, Prom-Wormley E, Panizzon MS, Kaup AR, Fennema-Notestine C, Neale MC, Jernigan TL, Fischl B, Franz CE, Lyons MJ, Grant M, Stevens A, Pacheco J, Perry ME, Schmitt JE, Seidman LJ, Thermenos HW, Tsuang MT, Chen CH, Thompson WK, Jak A, Dale AM, Kremen WS (2011) Genetic and environmental contributions to regional cortical surface area in humans: A magnetic resonance imaging twin study. Cereb Cortex 21, 2313-2321. McDowell I, Xi G, Lindsay J, Tierney M (2007) Mapping the connections between education and dementia. J Clin Exp Neuropsychol 29, 127-141. Garibotto V, Borroni B, Kalbe E, Herholz K, Salmon E, Holtoff V, Sorbi S, Cappa SF, Padovani A, Fazio F, Perani D (2008) Education and occupation as proxies for reserve in aMCI converters and AD: FDG-PET evidence. Neurology 71, 1342-1349. Caamano-Isorna F, Corral M, Montes-Martinez A, Takkouche B (2006) Education and dementia: A metaanalytic study. Neuroepidemiology 26, 226-232. Boyle PA, Buchman AS, Wilson RS, Yu L, Schneider JA, Bennett DA (2012) Effect of purpose in life on the relation between Alzheimer disease pathologic changes on cognitive function in advanced age. Arch Gen Psychiatry 69, 499-505. Boyle PA, Buchman AS, Barnes LL, Bennett DA (2010) Effect of a purpose in life on risk of incident Alzheimer disease and mild cognitive impairment in community-dwelling older persons. Arch Gen Psychiatry 67, 304-310. Fratiglioni L, Paillard-Borg S, Winblad B (2004) An active and socially integrated lifestyle in late life might protect against dementia. Lancet Neurol 3, 343-353. Wang HX, Karp A, Herlitz A, Crowe M, Kareholt I, Winblad B, Fratiglioni L (2009) Personality and lifestyle in relation to dementia incidence. Neurology 72, 253-259.

1160 [41]

[42]

[43]

[44]

[45] [46]

[47]

[48]

[49]

[50]

[51]

[52]

[53]

[54]

[55]

[56]

[57]

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes Fratiglioni L, Wang HX, Ericsson K, Maytan M, Winblad B (2000) Influence of social network on occurrence of dementia: A community-based longitudinal study. Lancet 355, 1315-1319. Wang HX, Karp A, Winblad B, Fratiglioni L (2002) Latelife engagement in social and leisure activities is associated with a decreased risk of dementia: A longitudinal study from the Kungsholmen project. Am J Epidemiol 155, 1081-1087. Saczynski JS, Pfeifer LA, Masaki K, Korf ES, Laurin D, White L, Launer LJ (2006) The effect of social engagement on incident dementia: The Honolulu-Asia Aging Study. Am J Epidemiol 163, 433-440. Bennett DA, Schneider JA, Tang Y, Arnold SE, Wilson RS (2006) The effect of social networks on the relation between Alzheimer’s disease pathology and level of cognitive function in old people: A longitudinal cohort study. Lancet Neurol 5, 406-412. Fratiglioni L, Wang HX (2007) Brain reserve hypothesis in dementia. J Alzheimers Dis 12, 11-22. Qiu C (2012) Preventing Alzheimer’s disease by targeting vascular risk factors: Hope and gap. J Alzheimers Dis 32, 721-731. Barnes DE, Yaffe K (2011) The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol 10, 819-828. Profenno LA, Porsteinsson AP, Faraone SV (2010) Metaanalysis of Alzheimer’s disease risk with obesity, diabetes, and related disorders. Biol Psychiatry 67, 505-512. Wang HX, Wahlberg M, Karp A, Winblad B, Fratiglioni L (2012) Psychosocial stress at work is associated with increased dementia risk in late life. Alzheimers Dement 8, 114-120. Qiu C, Winblad B, Fratiglioni L (2005) The age-dependent relation of blood pressure to cognitive function and dementia. Lancet Neurol 4, 487-499. Qiu C, Xu W, Winblad B, Fratiglioni L (2010) Vascular risk profiles for dementia and Alzheimer’s disease in very old people: A population-based longitudinal study. J Alzheimers Dis 20, 293-300. Kivipelto M, Ngandu T, Laatikainen T, Winblad B, Soininen H, Tuomilehto J (2006) Risk score for the prediction of dementia risk in 20 years among middle aged people: A longitudinal, population-based study. Lancet Neurol 5, 735-741. Schneider JA, Arvanitakis Z, Bang W, Bennett DA (2007) Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology 69, 21972204. Matthews FE, Brayne C, Lowe J, McKeith I, Wharton SB, Ince P (2009) Epidemiological pathology of dementia: Attributable-risks at death in the Medical Research Council Cognitive Function and Ageing Study. PLoS Med 6, e1000180. Toledo JB, Arnold SE, Raible K, Brettschneider J, Xie SX, Grossman M, Monsell SE, Kukull WA, Trojanowski JQ (2013) Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer’s Coordinating Centre. Brain 136, 2697-2706. Mrak RE, Griffin WS (2007) Dementia with Lewy bodies: Definition, diagnosis, and pathogenic relationship to Alzheimer’s disease. Neuropsychiatr Dis Treat 3, 619-625. Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA (2009) The neuropathology of probable Alzheimer disease and mild cognitive impairment. Ann Neurol 66, 200-208.

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65]

[66]

[67]

[68]

[69]

Amador-Ortiz C, Lin WL, Ahmed Z, Personett D, Davies P, Duara R, Graff-Radford NR, Hutton ML, Dickson DW (2007) TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol 61, 435-445. Higashi S, Iseki E, Yamamoto R, Minegishi M, Hino H, Fujisawa K, Togo T, Katsuse O, Uchikado H, Furukawa Y, Kosaka K, Arai H (2007) Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res 1184, 284-294. Graham NL, Emery T, Hodges JR (2004) Distinctive cognitive profiles in Alzheimer’s disease and subcortical vascular dementia. J Neurol Neurosurg Psychiatry 75, 61-71. Jellinger K (2000) Pure hippocampal sclerosis: A rare cause of dementia mimicking Alzheimer’s disease. Neurology 55, 739-740. Leverenz JB, Agustin CM, Tsuang D, Peskind ER, Edland SD, Nochlin D, DiGiacomo L, Bowen JD, McCormick WC, Teri L, Raskind MA, Kukull WA, Larson EB (2002) Clinical and neuropathological characteristics of hippocampal sclerosis: A community-based study. Arch Neurol 59, 10991106. Davidson YS, Raby S, Foulds PG, Robinson A, Thompson JC, Sikkink S, Yusuf I, Amin H, DuPlessis D, Troakes C, AlSarraj S, Sloan C, Esiri MM, Prasher VP, Allsop D, Neary D, Pickering-Brown SM, Snowden JS, Mann DM (2011) TDP-43 pathological changes in early onset familial and sporadic Alzheimer’s disease, late onset Alzheimer’s disease and Down’s syndrome: Association with age, hippocampal sclerosis and clinical phenotype. Acta Neuropathol 122, 703-713. Brenowitz WD, Monsell SE, Schmitt FA, Kukull WA, Nelson PT (2014) Hippocampal sclerosis of aging is a key Alzheimer’s disease mimic: Clinical-pathologic correlations and comparisons with both Alzheimer’s disease and non-tauopathic frontotemporal lobar degeneration. J Alzheimers Dis 39, 691-702. Beach TG, Monsell SE, Phillips LE, Kukull W (2012) Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005-2010. J Neuropathol Exp Neurol 71, 266-273. Alladi S, Xuereb J, Bak T, Nestor P, Knibb J, Patterson K, Hodges JR (2007) Focal cortical presentations of Alzheimer’s disease. Brain 130, 2636-2645. Rabinovici GD, Jagust WJ, Furst AJ, Ogar JM, Racine CA, Mormino EC, O’Neil JP, Lal RA, Dronkers NF, Miller BL, Gorno-Tempini ML (2008) Abeta amyloid and glucose metabolism in three variants of primary progressive aphasia. Ann Neurol 64, 388-401. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH (2011) The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 7, 263-269. Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, Snyder PJ, Carrillo MC, Thies B, Phelps CH (2011) The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 7, 270-279.

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes [70]

[71]

[72]

[73]

[74] [75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

Mitchell AJ, Shiri-Feshki M (2009) Rate of progression of mild cognitive impairment to dementia–meta-analysis of 41 robust inception cohort studies. Acta Psychiatr Scand 119, 252-265. Nelson PT, Head E, Schmitt FA, Davis PR, Neltner JH, Jicha GA, Abner EL, Smith CD, Van Eldik LJ, Kryscio RJ, Scheff SW (2011) Alzheimer’s disease is not “brain aging”: Neuropathological, genetic, and epidemiological human studies. Acta Neuropathol 121, 571-587. Blennow K, Hardy J, Zetterberg H (2012) The neuropathology and neurobiology of traumatic brain injury. Neuron 76, 886-899. Kling MA, Trojanowski JQ, Wolk DA, Lee VM, Arnold SE (2013) Vascular disease and dementias: Paradigm shifts to drive research in new directions. Alzheimers Dement 9, 76-92. Lo RY, Jagust WJ (2012) Vascular burden and Alzheimer disease pathologic progression. Neurology 79, 1349-1355. Snowdon DA, Greiner LH, Mortimer JA, Riley KP, Greiner PA, Markesbery WR (1997) Brain infarction and the clinical expression of Alzheimer disease. The Nun Study. JAMA 277, 813-817. Chui HC, Zarow C, Mack WJ, Ellis WG, Zheng L, Jagust WJ, Mungas D, Reed BR, Kramer JH, Decarli CC, Weiner MW, Vinters HV (2006) Cognitive impact of subcortical vascular and Alzheimer’s disease pathology. Ann Neurol 60, 677-687. Marchant NL, Reed BR, Sanossian N, Madison CM, Kriger S, Dhada R, Mack WJ, DeCarli C, Weiner MW, Mungas DM, Chui HC, Jagust WJ (2013) The aging brain and cognition: Contribution of vascular injury and abeta to mild cognitive dysfunction. JAMA Neurol 70, 488-495. Chui HC, Zheng L, Reed BR, Vinters HV, Mack WJ (2012) Vascular risk factors and Alzheimer’s disease: Are these risk factors for plaques and tangles or for concomitant vascular pathology that increases the likelihood of dementia? An evidence-based review. Alzheimers Res Ther 4, 1. Cao D, Lu H, Lewis TL, Li L (2007) Intake of sucrosesweetened water induces insulin resistance and exacerbates memory deficits and amyloidosis in a transgenic mouse model of Alzheimer disease. J Biol Chem 282, 36275-36282. Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, Peng Y, Cambareri G, Rocher A, Mobbs CV, Hof PR, Pasinetti GM (2004) Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J 18, 902-904. O’Connor T, Sadleir KR, Maus E, Velliquette RA, Zhao J, Cole SL, Eimer WA, Hitt B, Bembinster LA, Lammich S, Lichtenthaler SF, Hebert SS, De Strooper B, Haass C, Bennett DA, Vassar R (2008) Phosphorylation of the translation initiation factor eIF2alpha increases BACE1 levels and promotes amyloidogenesis. Neuron 60, 988-1009. Velliquette RA, O’Connor T, Vassar R (2005) Energy inhibition elevates beta-secretase levels and activity and is potentially amyloidogenic in APP transgenic mice: Possible early events in Alzheimer’s disease pathogenesis. J Neurosci 25, 10874-10883. Zhang X, Zhou K, Wang R, Cui J, Lipton SA, Liao FF, Xu H, Zhang YW (2007) Hypoxia-inducible factor 1alpha (HIF1alpha)-mediated hypoxia increases BACE1 expression and beta-amyloid generation. J Biol Chem 282, 10873-10880. Sun X, He G, Qing H, Zhou W, Dobie F, Cai F, Staufenbiel M, Huang LE, Song W (2006) Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1

[85]

[86]

[87]

[88]

[89]

[90]

[91]

[92]

[93]

[94]

[95]

[96]

[97]

[98]

1161

gene expression. Proc Natl Acad Sci U S A 103, 1872718732. Frederikse PH, Garland D, Zigler JS Jr, Piatigorsky J (1996) Oxidative stress increases production of beta-amyloid precursor protein and beta-amyloid (Abeta) in mammalian lenses, and Abeta has toxic effects on lens epithelial cells. J Biol Chem 271, 10169-10174. Olivieri G, Baysang G, Meier F, Muller-Spahn F, Stahelin HB, Brockhaus M, Brack C (2001) N-acetyl-L-cysteine protects SHSY5Y neuroblastoma cells from oxidative stress and cell cytotoxicity: Effects on beta-amyloid secretion and tau phosphorylation. J Neurochem 76, 224-233. Smith DH, Chen XH, Nonaka M, Trojanowski JQ, Lee VM, Saatman KE, Leoni MJ, Xu BN, Wolf JA, Meaney DF (1999) Accumulation of amyloid beta and tau and the formation of neurofilament inclusions following diffuse brain injury in the pig. J Neuropathol Exp Neurol 58, 982-992. Magnoni S, Brody DL (2010) New perspectives on amyloidbeta dynamics after acute brain injury: Moving between experimental approaches and studies in the human brain. Arch Neurol 67, 1068-1073. Han F, Fukunaga K (2009) Beta-amyloid accumulation in neurovascular units following brain embolism. J Pharmacol Sci 111, 101-109. Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM (2006) Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer’s disease. J Neurosci 26, 9047-9056. Dong H, Murphy KM, Meng L, Montalvo-Ortiz J, Zeng Z, Kolber BJ, Zhang S, Muglia LJ, Csernansky JG (2012) Corticotrophin releasing factor accelerates neuropathology and cognitive decline in a mouse model of Alzheimer’s disease. J Alzheimers Dis 28, 579-592. Jeong YH, Park CH, Yoo J, Shin KY, Ahn SM, Kim HS, Lee SH, Emson PC, Suh YH (2006) Chronic stress accelerates learning and memory impairments and increases amyloid deposition in APPV717I-CT100 transgenic mice, an Alzheimer’s disease model. FASEB J 20, 729-731. Kulstad JJ, McMillan PJ, Leverenz JB, Cook DG, Green PS, Peskind ER, Wilkinson CW, Farris W, Mehta PD, Craft S (2005) Effects of chronic glucocorticoid administration on insulin-degrading enzyme and amyloid-beta peptide in the aged macaque. J Neuropathol Exp Neurol 64, 139-146. Nagata K, Buchan RJ, Yokoyama E, Kondoh Y, Sato M, Terashi H, Satoh Y, Watahiki Y, Senova M, Hirata Y, Hatazawa J (1997) Misery perfusion with preserved vascular reactivity in Alzheimer’s disease. Ann N Y Acad Sci 826, 272-281. Nagata K, Sato M, Satoh Y, Watahiki Y, Kondoh Y, Sugawara M, Box G, Wright D, Leung S, Yuya H, Shimosegawa E (2002) Hemodynamic aspects of Alzheimer’s disease. Ann N Y Acad Sci 977, 391-402. Cunnane S, Nugent S, Roy M, Courchesne-Loyer A, Croteau E, Tremblay S, Castellano A, Pifferi F, Bocti C, Paquet N, Begdouri H, Bentourkia M, Turcotte E, Allard M, Barberger-Gateau P, Fulop T, Rapoport SI (2011) Brain fuel metabolism, aging, and Alzheimer’s disease. Nutrition 27, 3-20. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA (2001) Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol 60, 759-767. Pratico D, Clark CM, Liun F, Rokach J, Lee VY, Trojanowski JQ (2002) Increase of brain oxidative stress in mild

1162

[99]

[100]

[101]

[102]

[103]

[104] [105] [106]

[107]

[108]

[109]

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes cognitive impairment: A possible predictor of Alzheimer disease. Arch Neurol 59, 972-976. Rasmuson S, Andrew R, Nasman B, Seckl JR, Walker BR, Olsson T (2001) Increased glucocorticoid production and altered cortisol metabolism in women with mild to moderate Alzheimer’s disease. Biol Psychiatry 49, 547-552. Rothman SM, Mattson MP (2010) Adverse stress, hippocampal networks, and Alzheimer’s disease. Neuromolecular Med 12, 56-70. Hunter S, Arendt T, Brayne C (2013) The senescence hypothesis of disease progression in Alzheimer disease: An integrated matrix of disease pathways for FAD and SAD. Mol Neurobiol 48, 556-570. Hunter S, Brayne C (2012) Relationships between the amyloid precursor protein and its various proteolytic fragments and neuronal systems. Alzheimers Res Ther 4, 10. Romm JJ (2012) Language Intelligence: Lessons on persuasion from Jesus, Shakespeare, Lincoln, and Lady Gaga, CreateSpace Independent. George DR (2010) Overcoming the social death of dementia through language. Lancet 376, 586-587. George DR, Whitehouse PJ, D’Alton S, Ballenger J (2012) Through the amyloid gateway. Lancet 380, 1986-1987. Whitehouse PJ, George D (2008) The Myth of Alzheimer’s: What You Aren’t Being Told About Today’s Most Dreaded Diagnosis, St. Martin’s Press, New York. de la Torre JC (2012) Cardiovascular risk factors promote brain hypoperfusion leading to cognitive decline and dementia. Cardiovasc Psychiatry Neurol 2012, 367516. Accardi G, Caruso C, Colonna-Romano G, Camarda C, Monastero R, Candore G (2012) Can Alzheimer disease be a form of type 3 diabetes? Rejuvenation Res 15, 217-221. Naj AC, Jun G, Beecham GW, Wang LS, Vardarajan BN, Buros J, Gallins PJ, Buxbaum JD, Jarvik GP, Crane PK, Larson EB, Bird TD, Boeve BF, Graff-Radford NR, De Jager PL, Evans D, Schneider JA, Carrasquillo MM, ErtekinTaner N, Younkin SG, Cruchaga C, Kauwe JS, Nowotny P, Kramer P, Hardy J, Huentelman MJ, Myers AJ, Barmada MM, Demirci FY, Baldwin CT, Green RC, Rogaeva E, St George-Hyslop P, Arnold SE, Barber R, Beach T, Bigio EH, Bowen JD, Boxer A, Burke JR, Cairns NJ, Carlson CS, Carney RM, Carroll SL, Chui HC, Clark DG, Corneveaux J, Cotman CW, Cummings JL, DeCarli C, DeKosky ST, DiazArrastia R, Dick M, Dickson DW, Ellis WG, Faber KM, Fallon KB, Farlow MR, Ferris S, Frosch MP, Galasko DR, Ganguli M, Gearing M, Geschwind DH, Ghetti B, Gilbert JR, Gilman S, Giordani B, Glass JD, Growdon JH, Hamilton RL, Harrell LE, Head E, Honig LS, Hulette CM, Hyman BT, Jicha GA, Jin LW, Johnson N, Karlawish J, Karydas A, Kaye JA, Kim R, Koo EH, Kowall NW, Lah JJ, Levey AI, Lieberman AP, Lopez OL, Mack WJ, Marson DC, Martiniuk F, Mash DC, Masliah E, McCormick WC, McCurry SM, McDavid AN, McKee AC, Mesulam M, Miller BL, Miller CA, Miller JW, Parisi JE, Perl DP, Peskind E, Petersen RC, Poon WW, Quinn JF, Rajbhandary RA, Raskind M, Reisberg B, Ringman JM, Roberson ED, Rosenberg RN, Sano M, Schneider LS, Seeley W, Shelanski ML, Slifer MA, Smith CD, Sonnen JA, Spina S, Stern RA, Tanzi RE, Trojanowski JQ, Troncoso JC, Van Deerlin VM, Vinters HV, Vonsattel JP, Weintraub S, Welsh-Bohmer KA, Williamson J, Woltjer RL, Cantwell LB, Dombroski BA, Beekly D, Lunetta KL, Martin ER, Kamboh MI, Saykin AJ, Reiman EM, Bennett DA, Morris JC, Montine TJ, Goate AM, Blacker D, Tsuang DW, Hakonarson H, Kukull WA, Foroud TM, Haines JL, Mayeux R, Pericak-Vance MA, Farrer LA, Schellenberg GD (2011)

[110]

[111]

Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat Genet 43, 436-441. Alperovitch A, Boland A, Delepoine M, Dubois B, Duron E, Epelbaum J, Van Cauwenberghe C, Engelborghs S, Vandenberghe R, De Deyn PP, Ferri R, Romano C, Caltagirone C, Orfei MD, Ciaramella A, Scarpini E, Fenoglio C, Siciliano G, Bonuccelli U, Bagnoli S, Bracco L, Bessi V, Cecchetti R, Bastiani P, Squassina A, Seripa D, Frank-Garcia A, Sastre I, Blesa R, Alcolea D, Suarez-Clavet M, Sanchez-Juan P, Munoz Fernandez C, Aladro Benito Y, Thonberg H, Forshell C, Lilius L, Kinhult-Stahlbom A, Giedraitis V, Kilander L, Brundin RM, Concari L, Helisalmi S, Koivisto AM, Haapasalo A, Solfrizzi V, Frisardi V, Ott J, Carney RM, Mash DC, Albert MS, Albin RL, Apostolova LG, Arnold SE, Barmada MM, Barnes LL, Beach TG, Bigio EH, Bird TD, Boeve BF, Bowen JD, Boxer A, Burk JR, Cairns NJ, Cao C, Carlson CS, Carroll SL, Chibnik LB, Chui HC, Clark DG, Corneveaux J, Cribbs DG, DeCarli C, DeKosky ST, Demirci FY, Dick M, Dickson DW, Duara R, Ertekin-Taner N, Fallon KB, Farlow MR, Ferris S, Frosch MP, Galasko DR, Ganguli M, Gearing M, Geschwind DH, Ghetti B, Gilman S, Glass JD, Growdon JH, Hamilton RL, Harrell LE, Head E, Honig LS, Hulette CM, Hyman BT, Jarvik GP, Jicha GA, Jin LW, Karydas A, Kauwe JS, Kaye JA, Kim R, Koo EH, Kowall NW, Kramer JH, Kramer P, LaFerla FM, Lah JJ, Levernez JB, Levey AI, Li G, Lieberman AP, Lyketsos CG, Mack WJ, Marson DC, Martiniuk F, Masliah E, McCormick WC, McCurry SM, McDavid AN, McKee AC, Mesulam M, Miller BL, Miller CA, Miller JW, Morris JC, Murrell JR, Olichney JM, Pankratz VS, Parasi JE, Peskind E, Peterson RC, Pierce A, Poon WW, Potter H, Quinn JF, Raj A, Raskind M, Reiman EM, Reisberg B, Ringman JM, Roberson ED, Rosen HJ, Rosenberg RN, Sano M, Saykin AJ, Schneider JA, Schneider LS, Seeley WW, Smith AG, Sonnen JA, Spina S, Stern RA, Tanzi RE, Trojanowski JQ, Troncoso JC, Van Deerlin VM, Van Eldik LJ, Vinters HV, Vonsattel JP, Weintraub S, Welsh-Bohmer KA, Williamson J, Woltjer RL, Yu CE, Barber R, Au R, Wolf PA, Beiser A, Debette S, Yang Q, Weinstein G, Johnson AD, Wang J, Uitterlinden AG, Rivadeneira F, Koudstgaal PJ, Longstreth WT Jr, Becker JT, Kuller LH, Lumley T, Rice K, Garcia M, Aspelund T, Marksteiner JJ, Dal-Bianco P, Toglhofer AM, Freudenberger P, Ransmayr G, Benke T, Toeglhofer AM, Bressler J, Breteler MM, Fornage M, Hernandez I, Rosende Roca M, Ana Mauleon M, Alegrat M, Ramirez-Lorca R, Gonzalez-Perez A, Chapman J, Stretton A, Morgan A, Kehoe PG, Medway C, Lord J, Turton J, Hooper NM, Vardy E, Warren JD, Schott JM, Uphill J, Ryan N, Rossor M, Ben-Shlomo Y, Makrina D, Gkatzima O, Lupton M, Koutroumani M, Avramidou D, Germanou A, Jessen F, Riedel-Heller S, Dichgans M, Heun R, Kolsch H, Schurmann B, Herold C, Lacour A, Drichel D, Hoffman P, Kornhuber J, Gu W, Feulner T, van den Bussche H, Lawlor B, Lynch A, Mann D, Smith AD, Warden D, Wilcock G, Heuser I, Wiltgang J, Frolich L, Hull M, Mayo K, Livingston G, Bass NJ, Gurling H, McQuillin A, Gwilliam R, Deloukas P, Al-Chalabi A, Shaw CE, Singleton AB, Guerreiro R, Jockel KH, Klopp N, Wichmann HE, Graff-Radford NR, Ma L, Bisceglio G, Fisher E, Warner N, Pickering-Brown S (2013) Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet 45, 1452-1458. Dong Y, Wu X, Xu Z, Zhang Y, Xie Z (2012) Anesthetic isoflurane increases phosphorylated tau levels mediated

S. D’Alton et al. / Dementia: A Public Health Challenge for Our Lifetimes

[112]

[113]

[114]

[115]

[116]

[117]

[118]

[119]

[120]

[121]

by caspase activation and Abeta generation. PLoS One 7, e39386. Fodale V, Santamaria LB, Schifilliti D, Mandal PK (2010) Anaesthetics and postoperative cognitive dysfunction: A pathological mechanism mimicking Alzheimer’s disease. Anaesthesia 65, 388-395. Xu Z, Dong Y, Wang H, Culley DJ, Marcantonio ER, Crosby G, Tanzi RE, Zhang Y, Xie Z (2014) Age-dependent postoperative cognitive impairment and Alzheimer-related neuropathology in mice. Sci Rep 4, 3766. Gasparini M, Vanacore N, Schiaffini C, Brusa L, Panella M, Talarico G, Bruno G, Meco G, Lenzi GL (2002) A case-control study on Alzheimer’s disease and exposure to anesthesia. Neurol Sci 23, 11-14. Moller JT, Cluitmans P, Rasmussen LS, Houx P, Rasmussen H, Canet J, Rabbitt P, Jolles J, Larsen K, Hanning CD, Langeron O, Johnson T, Lauven PM, Kristensen PA, Biedler A, van Beem H, Fraidakis O, Silverstein JH, Beneken JE, Gravenstein JS (1998) Long-term postoperative cognitive dysfunction in the elderly ISPOCD1 study. ISPOCD investigators. International Study of Post-Operative Cognitive Dysfunction. Lancet 351, 857-861. Bohnen N, Warner MA, Kokmen E, Kurland LT (1994) Early and midlife exposure to anesthesia and age of onset of Alzheimer’s disease. Int J Neurosci 77, 181-185. Bohnen NI, Warner MA, Kokmen E, Beard CM, Kurland LT (1994) Alzheimer’s disease and cumulative exposure to anesthesia: A case-control study. J Am Geriatr Soc 42, 198201. Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, Holtzman DM, Santacruz A, Buckles V, Oliver A, Moulder K, Aisen PS, Ghetti B, Klunk WE, McDade E, Martins RN, Masters CL, Mayeux R, Ringman JM, Rossor MN, Schofield PR, Sperling RA, Salloway S, Morris JC (2012) Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. New Engl J Med 367, 795-804. Braak H, Del Tredici K (2011) The pathological process underlying Alzheimer’s disease in individuals under thirty. Acta Neuropathol 121, 171-181. Golde TE, Schneider LS, Koo EH (2011) Anti-abeta therapeutics in Alzheimer’s disease: The need for a paradigm shift. Neuron 69, 203-213. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR Jr, Kaye J, Montine TJ, Park DC, Reiman EM, Rowe CC, Siemers E, Stern Y,

[122]

[123]

[124]

[125]

[126] [127]

[128]

[129]

[130]

[131]

1163

Yaffe K, Carrillo MC, Thies B, Morrison-Bogorad M, Wagster MV, Phelps CH (2011) Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 7, 280-292. Gandy S (2012) Lifelong management of amyloid-beta metabolism to prevent Alzheimer’s disease. New Engl J Med 367, 864-866. Solomon A, Kivipelto M, Soininen H (2012) Prevention of Alzheimer’s disease: Moving backward through the lifespan. J Alzheimers Dis 33(Suppl 1), S465-S469. Andrieu S, Aboderin I, Baeyens JP, Beard J, Benetos A, Berrut G, Brainin M, Cha HB, Chen LK, Du P, Forette B, Forette F, Franco A, Fratiglioni L, Gillette-Guyonnet S, Gold G, Gomez F, Guimaraes R, Gustafson D, Khachaturian A, Luchsinger J, Mangialasche F, Mathiex-Fortunet H, Michel JP, Richard E, Schneider LS, Solomon A, Vellas B (2011) IAGG workshop: Health promotion program on prevention of late onset dementia. J Nutr Health Aging 15, 562-575. de Rooij SR, Wouters H, Yonker JE, Painter RC, Roseboom TJ (2010) Prenatal undernutrition and cognitive function in late adulthood. Proc Natl Acad Sci U S A 107, 16881-16886. World Health Organization (2012) Dementia: A Public Health Priority. DiClemente R, Salazar L, Crosy R (2013) Health Behavior Theory for Public Health: Principles, Foundations and Applications. Jones and Bartlett Learning, LLC, Burlington, MA, pp. 231-251. George DR, Whitehouse PJ (2011) Marketplace of memory: What the brain fitness technology industry says about us and how we can do better. Gerontologist 51, 590-596. George DR, Whitehouse PJ, Whitehouse D (2011) “Intergenerativity” in action: How intergenerational programs are rising to 21st century challenges. J Intergener Relatsh 9, 389-404. Sanchez M, Butts D, Hatton-Yeo A, Jarrot S, Kaplan M, Martinez A, Newman S, Pinazo S, Saez J, Weintraub A (2007) Social Studies Collection No.23: Intergenerational programmes: Towards a society for all ages. La Caixa Foundation: Barcelona, http://obrasocial.lacaixa.es/StaticFi les/StaticFiles/b1d9d404d98e6110VgnVCM1000000e8cf1 0aRCRD/es/Llibre23 en.pdf. George DR, Wagler G (2014) Social learning and innovation: Developing two shared site intergenerational reading programs in Hershey, PA. J Intergener Relatsh 12, 69-74.

Adapting to dementia in society: a challenge for our lifetimes and a charge for public health.

For the last several decades, Alzheimer's disease (AD) has been widely regarded as a late life event, but is now being redefined as a chronic conditio...
115KB Sizes 2 Downloads 5 Views