Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Nov 1.
Published in final edited form as: Neurol Clin. 2016 Aug 4;34(4):941–953. doi: 10.1016/j.ncl.2016.06.009

Alzheimer Disease and its Growing Epidemic: Risk Factors, Biomarkers and the Urgent Need for Therapeutics

RA Hickman 1,*, A Faustin 1,2,4,*, T Wisniewski 1,2,3,4,
PMCID: PMC5116320  NIHMSID: NIHMS793069  PMID: 27720002

SYNOPSIS

Alzheimer disease represents one of the greatest medical challenges that face this century; the condition is becoming increasingly prevalent worldwide and as yet, no effective treatments have been developed for this terminal disease. Since the disease manifests at a late stage after a long period of clinically silent neurodegeneration, knowledge of the modifiable risk factors and the implementation of biomarkers is crucial in the primary prevention of the disease and pre-symptomatic detection of AD, respectively. This review discusses the growing epidemic of AD and antecedent risk factors in the disease process. Disease biomarkers are discussed and the implications that this may have for the treatment of this currently incurable disease.

Keywords: Alzheimer disease, risk factors, biomarkers, epidemiology

INTRODUCTION

Alzheimer disease represents one of the greatest medical challenges that face this century; the condition is becoming increasingly prevalent worldwide and as yet, no effective treatments have been developed for this terminal disease. In the United States in 2015, over five million people suffered with AD, costing over 170 billion dollars. Since the disease manifests at a late stage after a long period of clinically silent neurodegeneration, knowledge of the modifiable risk factors and the implementation of biomarkers is crucial in the primary prevention of the disease and pre-symptomatic detection of AD, respectively. This review discusses the growing epidemic of AD and antecedent risk factors in the disease process. Disease biomarkers are discussed and the implications that this may have for the treatment of this currently incurable disease.

EPIDEMIOLOGY

Alzheimer disease (AD) is the most common dementia in the elderly and is a growing epidemic across the globe. Although the risks associated with developing AD are multifactorial, the greatest risk factor by far is aging1. The age-specific risk of AD dramatically increases as individuals get older; findings from the Framingham study in the early 1990s showed that the incidence doubles every five years up to the ages of 89 years2. Age-dependent increases have been seen in other studies35. Unsurprisingly therefore, with global reductions in fertility and extended life expectancies, the number of patients with AD is expected to increase as populations age6. In the United States, it is estimated that approximately 5.3 million people had AD in 2015; 5.1 million people being 65 years and older and approximately 200,000 people under the age of 65 years with early onset AD (EOAD) 79. It is estimated that the number of new cases of AD and other dementias will at least double by 2050 and substantially increase the socioeconomic burden worldwide7, 10.

In 2010, it was estimated that dementia afflicted 35.6 million people worldwide, many of which will have AD, with the projection that this figure will double every twenty years11. The incidence of AD is generally lower in many less economically developed countries than in North America and Europe, however, sharp rises in prevalence have been predicted and seen in China, India and Latin America12, 13.

The effect of this increasing dementia has obvious socioeconomic consequences for each country affected, through costs of hospital care and also of caregivers. In the USA, the total payments were estimated at $226 billion of which Medicare and Medicaid provided 68% 7, 14, whilst out-of-pocket expenses for patients and their families were expected to be $44 billion7.

CLASSIFICATION AND STAGING

Revised criteria and guidelines by the National Institute on Aging and the Alzheimer Association published in 2011 (NIA-AA) have recognized three stages of Alzheimer disease: preclinical Alzheimer disease, mild cognitive impairment (MCI) due to Alzheimer disease and dementia due to Alzheimer disease8, 15. These are described as follows:

  1. Preclinical AD: pre-symptomatic of AD with early AD-related brain changes as detected by neuroimaging or other biomarker studies;

  2. Mild cognitive impairment (MCI) due to AD: mild cognitive decline but still able to perform activities of daily living;

  3. Dementia due to AD: cognitive decline is more pronounced and interferes with activities of daily living7.

With this classification in mind, it follows that the actual number of individuals with active disease are gross underestimates because they are based on approximations of diagnosed symptomatic patients and largely ignores the vast number of individuals who are preclinical, in whom the disease process is active but asymptomatic16. This long pre-clinical phase of AD is characterized by progressive neuronal loss, the formation of neurofibrillary tangles (NFT) and the deposition of amyloid plaques within the brain1720. Although the exact pathogenesis of AD is debated, the prevailing hypothesis is that the neurodegeneration is the result of the amyloid cascade, in which aberrant digestion and processing of the amyloid precursor protein (APP) results in the accumulation of neurotoxic Aβ oligomeric proteins2124. These proteins aggregate to form the insoluble amyloid plaques that are seen at microscopic examination of autopsy brains of AD patients.

BIOMARKERS

It is widely believed therefore that future therapeutics should be introduced during the preclinical and MCI stages of the disease course so as to preserve the existing functioning neural networks7, 25. In order to provide effective recognition of preclinical AD, there will likely need to be widespread implementation of disease biomarkers, such as in national screening programs targeting specific age groups and other high-risk categories. Such screening may prove popular as many patients are keen to know their disease status at an earlier time point2628.

Current disease biomarkers focus on indicators of cerebral amyloidosis or synaptic dysfunction. Markers of brain amyloidosis include reduced CSF Aβ42 concentrations and increased amyloid tracer uptake on positron emission tomography (PET) 29. These changes are followed after a period of time with markers of neuronal injury, notably increased CSF tau levels and brain atrophy on magnetic resonance imaging (MRI) 30, 31. PET tau imaging has great promise as a biomarker and may be able to provide estimates of pathological disease stage32. Validation is needed prior to introduction of these tests into the clinical setting, but would certainly be useful in providing a more complete overview of the current number of patients with AD and also in evaluating pre-clinical therapeutic response.

RISK FACTORS

The modifiable and non-modifiable risk factors of AD are important because they provide insight into the predispositions of the disease process prior to onset and also provides stratification of individuals who may be at increased risk. Besides aging, which, as discussed previously, is the most significant risk factor; other determinants of AD include genetic risk factors, and non-genetic, modifiable risk factors.

NON-MODIFIABLE GENETIC RISK FACTORS

Recent genome wide association studies (GWAS) have revealed many new genes that increase the risk of developing AD33. This review, however, considers the most commonly discussed genetic influences on the disease, notably mutations in ApoE, APP and presenilin mutations.

ApoE

Of all of the mutations identified in AD, genome wide association studies have demonstrated that it is the ε4 allele of the APOE gene that poses the greatest risk for AD3438. ApoE is a 34 kDa astrocytic protein that is encoded on chromosome 19q13. The apoE gene has three alleles that result in the production of ε2, ε3 and ε4 isoforms. One of its principal functions within the CNS is the delivery of cholesterol to neurons via the ApoE receptor39. ApoE3 is the most common variant, present in approximately 60% of the population and is regarded as having no altered risk in AD7, 35, 36. However, the next most common allele is the ε4 followed by the ε2 allele. ApoE heterozygosity with ApoE4/E3 or ApoE4 homozygosity confers a significantly risk of developing AD, from three-fold to eight-twelve fold, respectively. In approximately 40% of cases of AD, ApoE4 is identified 40. Furthermore, patients with ApoE4 have poorer cognitive performance in childhood and tend to develop the disease significantly earlier than those with ApoE341. In a population based study, patients who had suffered a head injury and carried the ApoE4 allele had a ten times increased risk of developing AD, unlike those without the allele who were at two-fold increased risk42. The MIRAGE study demonstrated that patients who have suffered head injury are at markedly increased risk of developing AD43. Curiously, the ε2 isoform bestows a decreased risk of AD than the ε3 allele36. It comes as no surprise therefore that the ε2 allele is over-represented in centenarians44.

Triggering Receptor on Myeloid Cells 2

Discovery of the triggering receptor on myeloid cells 2 (TREM2) allele as a rare genetic predisposition for AD has sparked interest because of its role in inflammation45. TREM2 is a receptor found on microglia that is important in phagocytosis and dampening the CNS immune response46. Mutation in TREM2 is rare, however, the most common receptor mutation (R47H) increases the risk of LOAD by approximately twofold. Furthermore, mutations in TREM2 are associated with more severe degrees of atrophy in AD than those without47. Mutations in TREM2 that increase the risk and severity of AD may result from derangements in neuroinflammation and amyloid clearance.

APP and Presenilin Mutations

Early onset familial AD (EOAD), which usually begins in patients younger than 65 years of age, represents less than 1% of cases of AD48. EOAD is often caused by autosomal dominant mutations such as mutations in amyloid precursor protein (APP), presenilin-1, and presenilin-2 genes48.

Mutations in proteins that are involved in the synthesis of Aβ result in downstream overproduction of the pathological Aβ. APP is encoded on chromosome 21q21.3 and comprises 3 transcript variants, the most common of which protein within the CNS is 695 amino acids long49. Over 30 coding APP mutations have been identified that usually result in an autosomal dominant EOAD because of increased Aβ production, shifts in synthesis of pathologic Aβ1–42, or production of Aβ that may have increased susceptibility to aggregation50. Of interest, not all APP mutations result in AD preponderance; actually, one mutation was found to be protective50. Presenilin is one of the proteins that constitute the active site of γ-secretase and therefore mutations alter the efficacy of this enzyme increasing the amount of Aβ1–42 production. Presenilin mutations account for the majority of cases of familial AD48.

Down Syndrome

Down syndrome (DS) is the most common chromosomal abnormality with an incidence of 1 per 733 live births and is characterized by trisomy 2149. Since APP is encoded on chromosome 21q21.3, this results in three copies of the APP protein. This increased abundance of APP expression, production of Aβ is considered to be one of the mechanisms as to why many of these patients develop EOAD. Given that the lifespan of patients with Down’s syndrome is now 55–60 years of age, approximately 70% of patients will suffer from AD51.

Cardiovascular Health

A large body of evidence suggests that cardiovascular disease increases the risk of dementia. Studies that have investigated patients with clinical and subclinical cardiovascular disease have poorer cognitive function than those without52, 53. Cortical ischemic changes can increase the risk of dementia54. However, studying the role of cardiovascular disease and AD is complicated by several issues, notably that extensive cardiovascular disease and dementia may preclude from a clinical diagnosis of AD and may instead favor a diagnosis of multi-infarct dementia55.

Studies have shown mixed results with regard to the influence of hypertension and this is in part due to differences in study design54, 5658. Observational studies however have generally shown that increased hypertension are associated with cognitive decline and an increased likelihood of developing AD, possibly through blood vessel injury, protein extravasation, neuronal injury and subsequent Aβ accumulation59.

Diabetes Mellitus

Diabetes mellitus (DM) is associated with an increased risk of cognitive decline and AD later in life. Observational studies of type 2 DM (T2DM) have found that T2DM nearly doubles the risk of AD6062. In the Religious Orders Study, 824 individuals who were older than 55 years of age, were evaluated for cognitive decline and AD and found that those with DM had a 65% increased risk of developing AD after a mean 5.5 year period63. The cognitive decline was found to be mainly in perceptual speed. Several meta-analyses have further confirmed an increased risk of AD in DM. The biological mechanism for this association may relate to competition of Aβ and insulin for insulin degrading enzyme, thereby reducing Aβ clearance. Alternatively, increased Aβ aggregation has been demonstrated through increased age-related glycation end-products that can occur in DM.

Anti-diabetic therapies in patients with DM and cognitive impairment and also in patients with AD have shown improvement in cognition, which may be related to the anti-inflammatory properties of these drugs.

Traumatic Brain Injury

Traumatic brain injury (TBI) is a growing public health concern worldwide because the incidence is rising and it carries a significant healthcare and socioeconomic burden for society6468. For patients who survive TBI, the average life expectancy is considerably shortened and many cases of TBI suffer chronic neurological and psychological morbidity that reduces quality of life 6972. More data is now showing that there are ongoing chronic changes within the brain following TBI and that these ensuing processes may result in further damage with possible neurodegenerative sequelae73. The first documentation of a syndrome that directed attention towards a neurodegenerative phenomenon after head injury was ‘punch drunk syndrome’. This syndrome described degenerative changes after repeated episodes of sub-lethal head injuries in professional boxers 74. This condition is now termed chronic traumatic encephalopathy (CTE) and afflicts a diverse range of people including professional and amateur players of contact sports as well as veterans 7578. CTE has pathological features that overlap with AD and TBI is recognized to shorten the time to onset of AD79. Furthermore, it is now considered that TBI is the most significant environmental risk factor for AD76, 80.

Recent data has demonstrated that in both long-standing TBI and AD there is chronic inflammation within the brain parenchyma and this persistent inflammatory milieu within the brain parenchyma could be where the pathophysiology of TBI and AD converge81, 82.

Following TBI the amyloid levels increase due to several factors. Firstly, APP expression is noticeably increased post-TBI83, 84. APP is particularly prominent at the axon terminals where there has been axonal transection and axonal transection is known to occur even in mild cases of TBI 85, 86. Secondly, β-secretase and γ-secretase, enzymes that both contribute to the digestion of APP and formation of Aβ are also upregulated8789. These increases in both substrate and enzymes, results in increased deposition of amyloid at the axon bulbs and offers one explanation as to how the risk of AD is increased after TBI 90.

The Influence of Neprilysin and TBI

Removal of cerebral amyloid is likely to be multifactorial, involving partly passive diffusion of soluble amyloid, active transport mechanisms and cellular digestion91, 92. The degree of amyloid pathology post-TBI and in AD is particularly influenced by neprilysin. Neprilysin is a membrane zinc metalloprotease that is capable of digesting Aβ peptide and thus has the capability of reducing the amyloid load within the brain93. Neprilysin knockout mice demonstrate increased amyloid burden in a gene-dose dependent correlation94. Johnson and colleagues demonstrated that in post-mortem subjects, the degree of amyloid burden was most in patients who had more than 41 GT repeats in the promoter region of the neprilysin gene, which was considered to be related to defective amyloid clearance 95. Curiously, neprilysin expression increases post-TBI and this may be a mechanism by which Aβ and amyloid plaques are cleared months after injury, despite increased intra-axonal APP and presenilin-1 expression 96. With age-related reduction in neprilysin, the balance between formation of amyloid and its breakdown may shift towards accumulation of amyloid and this may be responsible for the preponderance to AD post-TBI97.

There is also likely to be a contribution to amyloid breakdown from microglial activation. Neprilysin, metalloproteinase-9 and several other factors that are released by healthy microglia, digest Aβ98. There is a heightened neuro-inflammatory response following TBI that persists, and the activation of microglia most likely releases factors that will assist in the digestion of Aβ. However, with aging, the efficacy of microglial breakdown is likely to be lost, and may even accentuate the accumulation, thereby causing a gradual shift toward accumulation of amyloid in the dynamic Aβ turnover99.

Previous Amyloid Exposure

One of the most concerning developments over the past few years has been the accumulation of evidence that suggests infectivity of amyloid in a prion-like fashion. In a recent case series of iatrogenic CJD, a proportion of patients who received homogenized human pituitaries for growth hormone replacement were found to have significant cerebral amyloid angiopathy at autopsy, to an extent that was inconsistent with age100. Given that pituitaries may have amyloid deposits, there is the possibility that amyloid could seed through peripheral injection with proteopathic spread over subsequent decades100, 101. The proteopathic spread of amyloid in the brain has been demonstrated in numerous animal models and in human AD pathological staging102.

The main fear that stem from these findings is that iatrogenic infection may occur from re-used surgical instruments, since amyloid is difficult to remove from metal devices103. Further research is needed in this area in order to gauge the significance of these findings on amyloid infectivity.

Protective Factors

In general, environmental influences that are anti-inflammatory appear to be beneficial at reducing the likelihood of developing AD. Low calorie diets that are sustained for a protracted period of time are associated with reduced free radical production and increased brain neurogenesis and BDNF concentrations, all of which are recognized to promote healthier brain aging44, 104. Data regarding diets that are rich in antioxidants and polyunsaturated fatty acids (PUFA) have proved inconclusive with some studies demonstrating a reduction in the risk of AD, whilst others showing no such association105107.

Other protective influences include cognitive stimulation and a high educational achievement, which improves cognitive reserve108, 109. Physical exercise does appear to reduce the risk of developing dementia and can show improvements in cognition in patients with dementia110114.

Perspectives

While the incidence of AD appears to be increasing worldwide, the age-specific risk of developing AD in high income countries may be decreasing. Improvements in diet, exercise, education and management of chronic conditions, such as DM, appear to be improving the individual age-specific risk of AD within the USA115. However, in view of longer life expectancies and worldwide increases in the prevalence of other risk factors, such as obesity and DM, the incidence of AD is most likely set to increase considerably with significant socioeconomic impact.

Future work on the development and validation of biomarkers and on the introduction of therapeutics into the preclinical phase of AD has the greatest promise in the effective treatment of this otherwise incurable disease.

KEY POINTS.

  • Alzheimer disease is increasing in prevalence worldwide

  • Many individuals have preclinical AD without symptoms

  • Biomarkers are currently in development for detecting preclinical AD that may be amenable to novel therapies

  • Addressing modifiable risk factors should also help to reduce the prevalence of AD in the future

Footnotes

None of the authors have anything to disclose.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Esiri MM, Chance SA. Cognitive reserve, cortical plasticity and resistance to Alzheimer’s disease. Alzheimer’s research & therapy. 2012;4(2):7. doi: 10.1186/alzrt105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bachman DL, Wolf PA, Linn RT, et al. Incidence of dementia and probable Alzheimer’s disease in a general population: the Framingham Study. Neurology. 1993;43(3 Pt 1):515–519. doi: 10.1212/wnl.43.3_part_1.515. [DOI] [PubMed] [Google Scholar]
  • 3.Vardarajan BN, Faber KM, Bird TD, et al. Age-specific incidence rates for dementia and Alzheimer disease in NIA-LOAD/NCRAD and EFIGA families: National Institute on Aging Genetics Initiative for Late-Onset Alzheimer Disease/National Cell Repository for Alzheimer Disease (NIA-LOAD/NCRAD) and Estudio Familiar de Influencia Genetica en Alzheimer (EFIGA) JAMA neurology. 2014;71(3):315–323. doi: 10.1001/jamaneurol.2013.5570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Edland SD, Rocca WA, Petersen RC, et al. Dementia and Alzheimer disease incidence rates do not vary by sex in Rochester, Minn. Archives of neurology. 2002;59(10):1589–1593. doi: 10.1001/archneur.59.10.1589. [DOI] [PubMed] [Google Scholar]
  • 5.Ott A, Breteler MM, van Harskamp F, et al. Prevalence of Alzheimer’s disease and vascular dementia: association with education. The Rotterdam study Bmj. 1995;310(6985):970–973. doi: 10.1136/bmj.310.6985.970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Division UNDoEaSAP. World Population Ageing 2013. 2013. [Google Scholar]
  • 7.2015 Alzheimer’s disease facts and figures. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2015;11(3):332–384. doi: 10.1016/j.jalz.2015.02.003. [DOI] [PubMed] [Google Scholar]
  • 8.Hebert LE, Weuve J, Scherr PA, et al. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology. 2013;80(19):1778–1783. doi: 10.1212/WNL.0b013e31828726f5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Association As. Early-onset dementia: a national challenge, a future crisis. Washington, DC: Alzheimer’s Association; 2006. [Google Scholar]
  • 10.Brookmeyer R, Johnson E, Ziegler-Graham K, et al. Forecasting the global burden of Alzheimer’s disease. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2007;3(3):186–191. doi: 10.1016/j.jalz.2007.04.381. [DOI] [PubMed] [Google Scholar]
  • 11.Prince M, Bryce R, Albanese E, et al. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2013;9(1):63–75. e62. doi: 10.1016/j.jalz.2012.11.007. [DOI] [PubMed] [Google Scholar]
  • 12.Kalaria RN, Maestre GE, Arizaga R, et al. Alzheimer’s disease and vascular dementia in developing countries: prevalence, management, and risk factors. Lancet neurology. 2008;7(9):812–826. doi: 10.1016/S1474-4422(08)70169-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ferri CP, Prince M, Brayne C, et al. Global prevalence of dementia: a Delphi consensus study. Lancet (London, England) 2005;366(9503):2112–2117. doi: 10.1016/S0140-6736(05)67889-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hurd MD, Martorell P, Langa KM. Monetary costs of dementia in the United States. The New England journal of medicine. 2013;369(5):489–490. doi: 10.1056/NEJMc1305541. [DOI] [PubMed] [Google Scholar]
  • 15.Jack CR, Jr, Albert MS, Knopman DS, et al. Introduction to the recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2011;7(3):257–262. doi: 10.1016/j.jalz.2011.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34(7):939–944. doi: 10.1212/wnl.34.7.939. [DOI] [PubMed] [Google Scholar]
  • 17.Nelson PT, Alafuzoff I, Bigio EH, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012;71(5):362–381. doi: 10.1097/NEN.0b013e31825018f7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. The National Institute on Aging, and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease. Neurobiol Aging. 1997;18(4 Suppl):S1–2. [PubMed] [Google Scholar]
  • 19.Brier MR, McCarthy JE, Benzinger TL, et al. Local and distributed PiB accumulation associated with development of preclinical Alzheimer’s disease. Neurobiology of aging. 2016;38:104–111. doi: 10.1016/j.neurobiolaging.2015.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Saito S, Yamamoto Y, Ihara M. Mild Cognitive Impairment: At the Crossroad of Neurodegeneration and Vascular Dysfunction. Current Alzheimer research. 2015;12(6):507–512. doi: 10.2174/1567205012666150530202508. [DOI] [PubMed] [Google Scholar]
  • 21.Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends in pharmacological sciences. 1991;12(10):383–388. doi: 10.1016/0165-6147(91)90609-v. [DOI] [PubMed] [Google Scholar]
  • 22.Tanzi RE, Bertram L. Twenty years of the Alzheimer’s disease amyloid hypothesis: a genetic perspective. Cell. 2005;120(4):545–555. doi: 10.1016/j.cell.2005.02.008. [DOI] [PubMed] [Google Scholar]
  • 23.Wisniewski T, Goni F. Immunotherapeutic approaches for Alzheimer’s disease. Neuron. 2015;85(6):1162–1176. doi: 10.1016/j.neuron.2014.12.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Di Paolo G, Kim TW. Linking lipids to Alzheimer’s disease: cholesterol and beyond. Nature reviews Neuroscience. 2011;12(5):284–296. doi: 10.1038/nrn3012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bloudek LM, Spackman DE, Blankenburg M, et al. Review and meta-analysis of biomarkers and diagnostic imaging in Alzheimer’s disease. Journal of Alzheimer’s disease: JAD. 2011;26(4):627–645. doi: 10.3233/JAD-2011-110458. [DOI] [PubMed] [Google Scholar]
  • 26.Wikler EM, Blendon RJ, Benson JM. Would you want to know? Public attitudes on early diagnostic testing for Alzheimer’s disease. Alzheimer’s research & therapy. 2013;5(5):43. doi: 10.1186/alzrt206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Neumann PJ, Cohen JT, Hammitt JK, et al. Willingness-to-pay for predictive tests with no immediate treatment implications: a survey of US residents. Health economics. 2012;21(3):238–251. doi: 10.1002/hec.1704. [DOI] [PubMed] [Google Scholar]
  • 28.Kopits IM, Chen C, Roberts JS, et al. Willingness to pay for genetic testing for Alzheimer’s disease: a measure of personal utility. Genetic testing and molecular biomarkers. 2011;15(12):871–875. doi: 10.1089/gtmb.2011.0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Jack CR, Jr, Knopman DS, Jagust WJ, et al. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet neurology. 2010;9(1):119–128. doi: 10.1016/S1474-4422(09)70299-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Toledo JB, Xie SX, Trojanowski JQ, et al. Longitudinal change in CSF Tau and Abeta biomarkers for up to 48 months in ADNI. Acta neuropathologica. 2013;126(5):659–670. doi: 10.1007/s00401-013-1151-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Vemuri P, Wiste HJ, Weigand SD, et al. Serial MRI and CSF biomarkers in normal aging, MCI, and AD. Neurology. 2010;75(2):143–151. doi: 10.1212/WNL.0b013e3181e7ca82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.James OG, Doraiswamy PM, Borges-Neto S. PET Imaging of Tau Pathology in Alzheimer’s Disease and Tauopathies. Frontiers in neurology. 2015;6:38. doi: 10.3389/fneur.2015.00038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Karch CM, Goate AM. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biological psychiatry. 2015;77(1):43–51. doi: 10.1016/j.biopsych.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261(5123):921–923. doi: 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
  • 35.Qiu C, Kivipelto M, von Strauss E. Epidemiology of Alzheimer’s disease: occurrence, determinants, and strategies toward intervention. Dialogues Clin Neurosci. 2009;11(2):111–128. doi: 10.31887/DCNS.2009.11.2/cqiu. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Raber J, Huang Y, Ashford JW. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiology of aging. 2004;25(5):641–650. doi: 10.1016/j.neurobiolaging.2003.12.023. [DOI] [PubMed] [Google Scholar]
  • 37.Holtzman DM, Herz J, Bu G. Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease. Cold Spring Harbor perspectives in medicine. 2012;2(3):a006312. doi: 10.1101/cshperspect.a006312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Spinney L. Alzheimer’s disease: The forgetting gene. Nature. 2014;510(7503):26–28. doi: 10.1038/510026a. [DOI] [PubMed] [Google Scholar]
  • 39.Bu G. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nature reviews Neuroscience. 2009;10(5):333–344. doi: 10.1038/nrn2620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Farrer LA, Cupples LA, Haines JL, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. Jama. 1997;278(16):1349–1356. [PubMed] [Google Scholar]
  • 41.Deary IJ, Whiteman MC, Pattie A, et al. Apolipoprotein e gene variability and cognitive functions at age 79: a follow-up of the Scottish mental survey of 1932. Psychology and aging. 2004;19(2):367–371. doi: 10.1037/0882-7974.19.2.367. [DOI] [PubMed] [Google Scholar]
  • 42.Mayeux R, Ottman R, Maestre G, et al. Synergistic effects of traumatic head injury and apolipoprotein-epsilon 4 in patients with Alzheimer’s disease. Neurology. 1995;45(3 Pt 1):555–557. doi: 10.1212/wnl.45.3.555. [DOI] [PubMed] [Google Scholar]
  • 43.Guo Z, Cupples LA, Kurz A, et al. Head injury and the risk of AD in the MIRAGE study. Neurology. 2000;54(6):1316–1323. doi: 10.1212/wnl.54.6.1316. [DOI] [PubMed] [Google Scholar]
  • 44.Esiri MM. Ageing and the brain. The Journal of pathology. 2007;211(2):181–187. doi: 10.1002/path.2089. [DOI] [PubMed] [Google Scholar]
  • 45.Guerreiro R, Wojtas A, Bras J, et al. TREM2 variants in Alzheimer’s disease. The New England journal of medicine. 2013;368(2):117–127. doi: 10.1056/NEJMoa1211851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rohn TT. The triggering receptor expressed on myeloid cells 2: “TREM-ming” the inflammatory component associated with Alzheimer’s disease. Oxidative medicine and cellular longevity. 2013;2013:860959. doi: 10.1155/2013/860959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rajagopalan P, Hibar DP, Thompson PM. TREM2 and neurodegenerative disease. The New England journal of medicine. 2013;369(16):1565–1567. doi: 10.1056/NEJMc1306509#SA3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Blennow K, de Leon MJ, Zetterberg H. Alzheimer’s disease. Lancet (London, England) 2006;368(9533):387–403. doi: 10.1016/S0140-6736(06)69113-7. [DOI] [PubMed] [Google Scholar]
  • 49.Bayer TA, Cappai R, Masters CL, et al. It all sticks together--the APP-related family of proteins and Alzheimer’s disease. Molecular psychiatry. 1999;4(6):524–528. doi: 10.1038/sj.mp.4000552. [DOI] [PubMed] [Google Scholar]
  • 50.Jonsson T, Atwal JK, Steinberg S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96–99. doi: 10.1038/nature11283. [DOI] [PubMed] [Google Scholar]
  • 51.Hartley D, Blumenthal T, Carrillo M, et al. Down syndrome and Alzheimer’s disease: Common pathways, common goals. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2015;11(6):700–709. doi: 10.1016/j.jalz.2014.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Breteler MM, Claus JJ, Grobbee DE, et al. Cardiovascular disease and distribution of cognitive function in elderly people: the Rotterdam Study. BMJ. 1994;308(6944):1604–1608. doi: 10.1136/bmj.308.6944.1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kuller LH, Shemanski L, Manolio T, et al. Relationship between ApoE, MRI findings, and cognitive function in the Cardiovascular Health Study. Stroke. 1998;29(2):388–398. doi: 10.1161/01.str.29.2.388. [DOI] [PubMed] [Google Scholar]
  • 54.Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nature reviews Neurology. 2011;7(3):137–152. doi: 10.1038/nrneurol.2011.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Stampfer MJ. Cardiovascular disease and Alzheimer’s disease: common links. Journal of internal medicine. 2006;260(3):211–223. doi: 10.1111/j.1365-2796.2006.01687.x. [DOI] [PubMed] [Google Scholar]
  • 56.Kivipelto M, Helkala EL, Hanninen T, et al. Midlife vascular risk factors and late-life mild cognitive impairment: A population-based study. Neurology. 2001;56(12):1683–1689. doi: 10.1212/wnl.56.12.1683. [DOI] [PubMed] [Google Scholar]
  • 57.Launer LJ, Masaki K, Petrovitch H, et al. The association between midlife blood pressure levels and late-life cognitive function. The Honolulu-Asia Aging Study. Jama. 1995;274(23):1846–1851. [PubMed] [Google Scholar]
  • 58.Swan GE, Carmelli D, Larue A. Systolic blood pressure tracking over 25 to 30 years and cognitive performance in older adults. Stroke. 1998;29(11):2334–2340. doi: 10.1161/01.str.29.11.2334. [DOI] [PubMed] [Google Scholar]
  • 59.Kalaria RN. Vascular basis for brain degeneration: faltering controls and risk factors for dementia. Nutrition reviews. 2010;68(Suppl 2):S74–87. doi: 10.1111/j.1753-4887.2010.00352.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Leibson CL, Rocca WA, Hanson VA, et al. The risk of dementia among persons with diabetes mellitus: a population-based cohort study. Annals of the New York Academy of Sciences. 1997;826:422–427. doi: 10.1111/j.1749-6632.1997.tb48496.x. [DOI] [PubMed] [Google Scholar]
  • 61.Luchsinger JA, Tang MX, Stern Y, et al. Diabetes mellitus and risk of Alzheimer’s disease and dementia with stroke in a multiethnic cohort. American journal of epidemiology. 2001;154(7):635–641. doi: 10.1093/aje/154.7.635. [DOI] [PubMed] [Google Scholar]
  • 62.Ott A, Stolk RP, van Harskamp F, et al. Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology. 1999;53(9):1937–1942. doi: 10.1212/wnl.53.9.1937. [DOI] [PubMed] [Google Scholar]
  • 63.Arvanitakis Z, Wilson RS, Bienias JL, et al. Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. Archives of neurology. 2004;61(5):661–666. doi: 10.1001/archneur.61.5.661. [DOI] [PubMed] [Google Scholar]
  • 64.Roozenbeek B, Maas AI, Menon DK. Changing patterns in the epidemiology of traumatic brain injury. Nature reviews Neurology. 2013;9(4):231–236. doi: 10.1038/nrneurol.2013.22. [DOI] [PubMed] [Google Scholar]
  • 65.Maas AI, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet neurology. 2008;7(8):728–741. doi: 10.1016/S1474-4422(08)70164-9. [DOI] [PubMed] [Google Scholar]
  • 66.Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact of traumatic brain injury: a brief overview. The Journal of head trauma rehabilitation. 2006;21(5):375–378. doi: 10.1097/00001199-200609000-00001. [DOI] [PubMed] [Google Scholar]
  • 67.Coronado VG, McGuire LC, Sarmiento K, et al. Trends in Traumatic Brain Injury in the U.S. and the public health response: 1995–2009. Journal of safety research. 2012;43(4):299–307. doi: 10.1016/j.jsr.2012.08.011. [DOI] [PubMed] [Google Scholar]
  • 68.Bruns J, Jr, Hauser WA. The epidemiology of traumatic brain injury: a review. Epilepsia. 2003;44(Suppl 10):2–10. doi: 10.1046/j.1528-1157.44.s10.3.x. [DOI] [PubMed] [Google Scholar]
  • 69.Dutton RP, Stansbury LG, Leone S, et al. Trauma mortality in mature trauma systems: are we doing better? An analysis of trauma mortality patterns, 1997–2008. J Trauma. 2010;69(3):620–626. doi: 10.1097/TA.0b013e3181bbfe2a. [DOI] [PubMed] [Google Scholar]
  • 70.Selassie AW, McCarthy ML, Ferguson PL, et al. Risk of posthospitalization mortality among persons with traumatic brain injury, South Carolina 1999–2001. The Journal of head trauma rehabilitation. 2005;20(3):257–269. doi: 10.1097/00001199-200505000-00008. [DOI] [PubMed] [Google Scholar]
  • 71.Ferguson PL, Smith GM, Wannamaker BB, et al. A population-based study of risk of epilepsy after hospitalization for traumatic brain injury. Epilepsia. 2010;51(5):891–898. doi: 10.1111/j.1528-1167.2009.02384.x. [DOI] [PubMed] [Google Scholar]
  • 72.Fazel S, Wolf A, Pillas D, et al. Suicide, Fatal Injuries, and Other Causes of Premature Mortality in Patients With Traumatic Brain Injury: A 41-Year Swedish Population Study. JAMA Psychiatry. 2014 doi: 10.1001/jamapsychiatry.2013.3935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Masel BE, DeWitt DS. Traumatic brain injury: a disease process, not an event. J Neurotrauma. 2010;27(8):1529–1540. doi: 10.1089/neu.2010.1358. [DOI] [PubMed] [Google Scholar]
  • 74.Martland HS. Punch drunk. JAMA. 1928;91(15):1103–1107. [Google Scholar]
  • 75.McKee AC, Stern RA, Nowinski CJ, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain: a journal of neurology. 2013;136(Pt 1):43–64. doi: 10.1093/brain/aws307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Shively S, Scher AI, Perl DP, et al. Dementia resulting from traumatic brain injury: what is the pathology? Archives of neurology. 2012;69(10):1245–1251. doi: 10.1001/archneurol.2011.3747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Goldstein LE, Fisher AM, Tagge CA, et al. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Science translational medicine. 2012;4(134):134ra160. doi: 10.1126/scitranslmed.3003716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. Journal of neuropathology and experimental neurology. 2009;68(7):709–735. doi: 10.1097/NEN.0b013e3181a9d503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Plassman BL, Havlik RJ, Steffens DC, et al. Documented head injury in early adulthood and risk of Alzheimer’s disease and other dementias. Neurology. 2000;55(8):1158–1166. doi: 10.1212/wnl.55.8.1158. [DOI] [PubMed] [Google Scholar]
  • 80.Fleminger S, Oliver DL, Lovestone S, et al. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. Journal of neurology, neurosurgery, and psychiatry. 2003;74(7):857–862. doi: 10.1136/jnnp.74.7.857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Johnson VE, Stewart JE, Begbie FD, et al. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain: a journal of neurology. 2013;136(Pt 1):28–42. doi: 10.1093/brain/aws322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Smith C, Gentleman SM, Leclercq PD, et al. The neuroinflammatory response in humans after traumatic brain injury. Neuropathology and applied neurobiology. 2013;39(6):654–666. doi: 10.1111/nan.12008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pierce JE, Trojanowski JQ, Graham DI, et al. Immunohistochemical characterization of alterations in the distribution of amyloid precursor proteins and beta-amyloid peptide after experimental brain injury in the rat. The Journal of neuroscience: the official journal of the Society for Neuroscience. 1996;16(3):1083–1090. doi: 10.1523/JNEUROSCI.16-03-01083.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Van den Heuvel C, Blumbergs PC, Finnie JW, et al. Upregulation of amyloid precursor protein messenger RNA in response to traumatic brain injury: an ovine head impact model. Experimental neurology. 1999;159(2):441–450. doi: 10.1006/exnr.1999.7150. [DOI] [PubMed] [Google Scholar]
  • 85.Gentleman SM, Nash MJ, Sweeting CJ, et al. Beta-amyloid precursor protein (beta APP) as a marker for axonal injury after head injury. Neuroscience letters. 1993;160(2):139–144. doi: 10.1016/0304-3940(93)90398-5. [DOI] [PubMed] [Google Scholar]
  • 86.Oppenheimer DR. Microscopic lesions in the brain following head injury. Journal of neurology, neurosurgery, and psychiatry. 1968;31(4):299–306. doi: 10.1136/jnnp.31.4.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cribbs DH, Chen LS, Cotman CW, et al. Injury induces presenilin-1 gene expression in mouse brain. Neuroreport. 1996;7(11):1773–1776. doi: 10.1097/00001756-199607290-00016. [DOI] [PubMed] [Google Scholar]
  • 88.Nadler Y, Alexandrovich A, Grigoriadis N, et al. Increased expression of the gamma-secretase components presenilin-1 and nicastrin in activated astrocytes and microglia following traumatic brain injury. Glia. 2008;56(5):552–567. doi: 10.1002/glia.20638. [DOI] [PubMed] [Google Scholar]
  • 89.Blasko I, Beer R, Bigl M, et al. Experimental traumatic brain injury in rats stimulates the expression, production and activity of Alzheimer’s disease beta-secretase (BACE-1) Journal of neural transmission. 2004;111(4):523–536. doi: 10.1007/s00702-003-0095-6. [DOI] [PubMed] [Google Scholar]
  • 90.Clinton J, Roberts GW, Gentleman SM, et al. Differential pattern of beta-amyloid protein deposition within cortical sulci and gyri in Alzheimer’s disease. Neuropathology and applied neurobiology. 1993;19(3):277–281. doi: 10.1111/j.1365-2990.1993.tb00438.x. [DOI] [PubMed] [Google Scholar]
  • 91.Deane R, Sagare A, Zlokovic BV. The role of the cell surface LRP and soluble LRP in blood-brain barrier Abeta clearance in Alzheimer’s disease. Current pharmaceutical design. 2008;14(16):1601–1605. doi: 10.2174/138161208784705487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Turner AJ, Fisk L, Nalivaeva NN. Targeting amyloid-degrading enzymes as therapeutic strategies in neurodegeneration. Annals of the New York Academy of Sciences. 2004;1035:1–20. doi: 10.1196/annals.1332.001. [DOI] [PubMed] [Google Scholar]
  • 93.Iwata N, Tsubuki S, Takaki Y, et al. Identification of the major Abeta1–42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition. Nature medicine. 2000;6(2):143–150. doi: 10.1038/72237. [DOI] [PubMed] [Google Scholar]
  • 94.Iwata N, Tsubuki S, Takaki Y, et al. Metabolic regulation of brain Abeta by neprilysin. Science. 2001;292(5521):1550–1552. doi: 10.1126/science.1059946. [DOI] [PubMed] [Google Scholar]
  • 95.Johnson VE, Stewart W, Graham DI, et al. A neprilysin polymorphism and amyloid-beta plaques after traumatic brain injury. Journal of neurotrauma. 2009;26(8):1197–1202. doi: 10.1089/neu.2008.0843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Chen XH, Johnson VE, Uryu K, et al. A lack of amyloid beta plaques despite persistent accumulation of amyloid beta in axons of long-term survivors of traumatic brain injury. Brain pathology. 2009;19(2):214–223. doi: 10.1111/j.1750-3639.2008.00176.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hellstrom-Lindahl E, Ravid R, Nordberg A. Age-dependent decline of neprilysin in Alzheimer’s disease and normal brain: inverse correlation with A beta levels. Neurobiology of aging. 2008;29(2):210–221. doi: 10.1016/j.neurobiolaging.2006.10.010. [DOI] [PubMed] [Google Scholar]
  • 98.Yan P, Hu X, Song H, et al. Matrix metalloproteinase-9 degrades amyloid-beta fibrils in vitro and compact plaques in situ. The Journal of biological chemistry. 2006;281(34):24566–24574. doi: 10.1074/jbc.M602440200. [DOI] [PubMed] [Google Scholar]
  • 99.Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2008;28(33):8354–8360. doi: 10.1523/JNEUROSCI.0616-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Jaunmuktane Z, Mead S, Ellis M, et al. Evidence for human transmission of amyloid-beta pathology and cerebral amyloid angiopathy. Nature. 2015;525(7568):247–250. doi: 10.1038/nature15369. [DOI] [PubMed] [Google Scholar]
  • 101.Irwin DJ, Abrams JY, Schonberger LB, et al. Evaluation of potential infectivity of Alzheimer and Parkinson disease proteins in recipients of cadaver-derived human growth hormone. JAMA neurology. 2013;70(4):462–468. doi: 10.1001/jamaneurol.2013.1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Eisele YS, Obermuller U, Heilbronner G, et al. Peripherally applied Abeta-containing inoculates induce cerebral beta-amyloidosis. Science. 2010;330(6006):980–982. doi: 10.1126/science.1194516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Prusiner SB. Biology and genetics of prions causing neurodegeneration. Annual review of genetics. 2013;47:601–623. doi: 10.1146/annurev-genet-110711-155524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mattson MP, Maudsley S, Martin B. BDNF and 5-HT: a dynamic duo in age-related neuronal plasticity and neurodegenerative disorders. Trends in neurosciences. 2004;27(10):589–594. doi: 10.1016/j.tins.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 105.Kalmijn S, Launer LJ, Ott A, et al. Dietary fat intake and the risk of incident dementia in the Rotterdam Study. Ann Neurol. 1997;42(5):776–782. doi: 10.1002/ana.410420514. [DOI] [PubMed] [Google Scholar]
  • 106.Roberts RO, Cerhan JR, Geda YE, et al. Polyunsaturated fatty acids and reduced odds of MCI: the Mayo Clinic Study of Aging. Journal of Alzheimer’s disease: JAD. 2010;21(3):853–865. doi: 10.3233/JAD-2010-091597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Engelhart MJ, Geerlings MI, Ruitenberg A, et al. Diet and risk of dementia: Does fat matter?: The Rotterdam Study. Neurology. 2002;59(12):1915–1921. doi: 10.1212/01.wnl.0000038345.77753.46. [DOI] [PubMed] [Google Scholar]
  • 108.Carlson MC, Helms MJ, Steffens DC, et al. Midlife activity predicts risk of dementia in older male twin pairs. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2008;4(5):324–331. doi: 10.1016/j.jalz.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Fratiglioni L, Wang HX. Brain reserve hypothesis in dementia. Journal of Alzheimer’s disease: JAD. 2007;12(1):11–22. doi: 10.3233/jad-2007-12103. [DOI] [PubMed] [Google Scholar]
  • 110.Groot C, Hooghiemstra AM, Raijmakers PG, et al. The effect of physical activity on cognitive function in patients with dementia: A meta-analysis of randomized control trials. Ageing research reviews. 2016;25:13–23. doi: 10.1016/j.arr.2015.11.005. [DOI] [PubMed] [Google Scholar]
  • 111.Abbott RD, White LR, Ross GW, et al. Walking and dementia in physically capable elderly men. Jama. 2004;292(12):1447–1453. doi: 10.1001/jama.292.12.1447. [DOI] [PubMed] [Google Scholar]
  • 112.Fratiglioni L, Paillard-Borg S, Winblad B. An active and socially integrated lifestyle in late life might protect against dementia. Lancet neurology. 2004;3(6):343–353. doi: 10.1016/S1474-4422(04)00767-7. [DOI] [PubMed] [Google Scholar]
  • 113.Rovio S, Kareholt I, Helkala EL, et al. Leisure-time physical activity at midlife and the risk of dementia and Alzheimer’s disease. Lancet neurology. 2005;4(11):705–711. doi: 10.1016/S1474-4422(05)70198-8. [DOI] [PubMed] [Google Scholar]
  • 114.Akbaraly TN, Portet F, Fustinoni S, et al. Leisure activities and the risk of dementia in the elderly: results from the Three-City Study. Neurology. 2009;73(11):854–861. doi: 10.1212/WNL.0b013e3181b7849b. [DOI] [PubMed] [Google Scholar]
  • 115.Langa KM. Is the risk of Alzheimer’s disease and dementia declining? Alzheimer’s research & therapy. 2015;7(1):34. doi: 10.1186/s13195-015-0118-1. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES