Take a look at the Recent articles

Alzheimer’s disease syndrome – Recognizing the complexity of dementia

Tyler A. Kokjohn

Department of Microbiology, Midwestern University, Glendale, AZ, USA

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

Geidy E. Serrano

Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA

Alex E. Roher

Division of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA

Division of Clinical Education, Midwestern University Glendale, AZ, USA

DOI: 10.15761/NDT.1000116

Article
Article Info
Author Info
Figures & Data

Abstract

Therapies designed to disrupt amyloid plaque deposits or prevent their formation have yielded disappointing results against cognitive failure, suggesting that both our mechanistic models of dementia and interventions must become more nuanced. These treatments targeted Aβ40/42, but the amyloid accumulated in Alzheimer’s disease patients is modified extensively and far more structurally diverse. Despite intensive work, the structure and physical state of the most toxic amyloid species remains mysterious.  In addition, multiple lines of evidence suggest the genesis and progression of dementia is more complicated than the accumulation of senile plaques beyond a tolerable threshold. If amyloid plays key, but non-exclusive, roles in dementia pathogenesis this hypothesis must be addressed through investigations that are more holistic in scope. New imaging methods provide investigators unprecedented capabilities to detect and classify neuropathology in living subjects. Interpreting future clinical trial outcomes will hinge on correlating effects against dementia in subject cohorts that are precisely differentiated with respect to neuropathology and neurochemistry. Improving understanding of the comorbidities and the environmental/lifestyle factors foreshadowing cognitive failure will aid in the interpretation of clinical trials. Most important, as we seek an elusive cure for AD, these findings may be rapidly translatable into concrete and achievable public health improvements.

Key words

Alzheimer’s disease, late-onset Alzheimer’s disease, early-onset Alzheimer’s disease, amyloid, amyloid precursor protein, dementia, immunotherapy, systemic co-morbidities, non-amyloid pathology

Introduction

The notable accumulation of amyloid-β (Aβ) peptides in Alzheimer’s disease (AD) brains led to the postulation of  the amyloid cascade hypothesis. Profuse amyloid plaques in demented subjects and studies of early-onset Alzheimer disease (EOAD) genes provided strong experimental supporting evidence for the amyloid cascade hypothesis. However, disrupting Aβ deposits or preventing their formation have, so far, not yielded commensurate impacts against dementia in clinical trials. The almost exclusive focus on the amyloid cascade hypothesis inadvertently left systematic studies of comorbid conditions, environmental, lifestyle and psychosocial influences on dementia comparatively unexplored.

The amyloid cascade hypothesis has dominated the field for over two decades. Although amyloid plays an important role in the pathogenesis and clinical course of AD, interventions against Aβ have had only limited impacts against dementia. Despite the translational frustrations, the thorough investigation of amyloidosis in the ambit of AD has provided profound insights into the understanding of the mechanistic impacts of Aβ on dementia development. We offer a succinct description of the present status of late onset Alzheimer’s disease (LOAD) research and its nosology in relation to the amyloid hypothesis.  In addition, we make reference to synergistic and/or alternative groups of abnormal pathological options in view of the disappointing therapeutic attempts to interfere with the clinical course of LOAD. At present, some of these pathogenic alternatives have not been sufficiently studied to enable the development of robust hypotheses. Such is the case of environmental, lifestyle and psychosocial factors which have been mainly based on correlative observations but have potential for future preventative and therapeutic interventions. On the other hand, the contribution of cardiovascular system dysfunctions and the endocrine biochemical disturbances created by diabetes type-2 have been sufficiently investigated to warrant their participation in the pathogenesis and pathophysiology of LOAD.

While amyloid seems to play critical role(s) in LOAD, there are multiple routes to dementia that do not necessarily involve amyloid deposition.  We call for a more expansive view of dementia pathogenesis and its multifactorial instigators and suggest that studies of these factors are about to be augmented by a better understanding of age related comorbidities and exogenous risks factors impacting LOAD etiology. Advances in imaging technologies as well as improved knowledge of potential biochemical pathways leading to neurodegeneration coupled with data processing innovations promise to revolutionize our understanding of the intricate and complex pathogenesis of LOAD. New methods may ultimately confer capabilities to distinguish dementia arising through distinct pathways enabling their more refined nosological classification and the development of precision medicine.

Viewing dementia in broader context

Clearly defined mutational events in APP, PSEN1, PSEN2, tau, α-synuclein and APP gene duplications are directly responsible for genetically determined neurological disorders. Late onset Alzheimer’s neurodegeneration, on the other hand, results from the summation of genetic predisposition, epigenetic modulation and multiple brain and aging-associated systemic declines as well as from the negative contribution of environmental toxins, damaging lifestyles and negative psychosocial influences. Mutations or age-related molecular conformational changes that propagate misfolded molecules add to the long list of age-associated pathological events terminating in neuronal degeneration and brain vascular malfunction. The participation of a mélange of pathological events have been primarily classified on the bases of their clinical manifestations and most prominent pathological lesions. However, the neuropathological lesions associated with neurodegeneration are present in variable densities and unique combinations that complicate their classification. It is also evident that besides the presence of amyloid there are other independent systemic and cellular mechanisms in LOAD pathogenesis [1].

Diverse routes to dementia

After 25 years of research guided by the amyloid cascade hypothesis we cannot explain why some individuals with LOAD harbor profuse amyloid deposits while others either entirely avoid this situation or have a limited number of these lesions. What unique history triggers these outcomes? Is amyloid deposition an essential rescue operation that ensures brain survival and prolongs life or is it an accident of nature that disseminates a rogue molecule that spreads and destroys the brain? These pressing and obvious questions have not yet been entirely resolved.  

The variable clinical signs and symptoms of LOAD dementia may be the consequences of a collection of conditions and neuropathologically distinct diseases [2]. Although LOAD is formally confirmed after postmortem evaluation of brain amyloid plaque deposition and intracellular neurofibrillary tangle (NFT) accumulation, demented subjects often harbor heterogeneous conglomerations of cerebrovascular and other pathological lesions [3-10]. Advancing age is recognized to be the most significant risk factor for LOAD, but the relationship between neuropathology and dementia in the elderly is not a simple one [11].  Nonagenarian subjects harboring heavy amyloid plaque burdens offer an important insight into dementia etiology. The indistinct demarcation between LOAD and non-demented oldest-old groups in terms of Aβ-related pathological and biochemical features suggests involvement of other significant factors in neurodegeneration and cognitive failure [12].

Recognizing the relatively weak correlations between fibrillar amyloid levels and dementia, investigators have postulated the most neurotoxic entities are soluble Aβ oligomers [13-17]. However, the mechanism producing a transition to toxicity is unclear as Aβ peptides are normal constituents of brain, cerebrospinal fluid, plasma and other peripheral tissues. Moreover, Aβ42 toxicity is only evident in vitro at concentrations thousands of fold greater than present in vivo [13]. Detailed examination of the molecular constitution of the Aβ species present in LOAD exposes additional complexities. These amphipathic highly reactive Aβ molecules are subjected to extensive posttranslational modifications [18-37]  and proteolytic degradation with altered solubility, chemical reactivity and biophysical properties. These alterations also lead to a propensity for Aβ to dimerize and form larger oligomers [38-53]. In addition, subtle posttranslational conformational alterations can produce distinct Aβ strains with toxic prion-like properties [54]. Analogous to prion diseases, although non-infectious, AD amyloid pathology has long been recognized to be transmissible through Aβ seeding protocols into experimental rodents [55,56].

Several authors have challenged the supremacy of the amyloid cascade hypothesis implying that the presence of plaques is neither necessary nor sufficient to yield dementia [15,57-62]. Although Aβ is linked to LOAD, it may be an error to view it as the sole causative agent [57,63]. The prevailing view of LOAD dementia as caused by a succeeding series of consequential events initially precipitated by amyloid accumulation [64] seems poised to become more nuanced and mechanistically complex.

Less attention has been devoted to the possibility that other APP proteolytic fragments, which are presumably also elevated such as the Aη-α proteolytic fragment of APP that inhibits hippocampal neuronal activity, may play a prominent role in the pathogenesis and progression of LOAD [65]. In addition, the APP N-terminal peptide/DR6 receptor interaction [66], APP δ-ɣ fragment [67,68], APP C-terminal peptides CT99/CT83 [69], Jcasp, P31 and the Aβ-related P3 [70] are neurotoxic or provoke neuronal apoptosis. Our preliminary observations suggest that in LOAD putatively toxic CT99/CT83 peptides, rather than Aβ, accumulate abundantly in cellular membranes as if they were not processed by the ɣ-secretase. Furthermore, increased levels of AICD can cause hippocampal mossy fiber sprouting, neuronal hypersensitivity to stress and silent seizures [71]. Duplication of the APP gene results in ~50% increased production of Aβ causing EOAD with cerebral amyloid angiopathy [72] similar to the situation observed in Down’s syndrome [73]. Elevation of the multiple APP-derived peptides may have some disadvantageous metabolic effects and participate in neurodegeneration through pathways independent from amyloid accumulation. 

Expanding the nosology of AD related neurodegeneration 

New imaging methods have drawn attention to complexities in the relationship of neuropathology to dementia. Although persuasive evidence suggests amyloid positivity is correlated with dementia development [74] other observations indicate separate pathways to neurodegeneration exist [75] which do not necessarily involve amyloid deposition. Studies of younger Dominantly Inherited Alzheimer Network (DIAN) EOAD mutation carriers revealed amyloid deposition is an early event in the progression of neurodegeneration [76]. However, the situations in subjects with spontaneously emerging neurodegeneration and dementia are more complicated.  Imaging studies have revealed a class of cognitively normal subjects exhibiting biomarkers of neurodegeneration in the absence of detectable amyloid deposits designated suspected non-amyloid pathology (SNAP) [77]. A significant minority (25%) of the cognitively normal subjects examined in a population-based sample of elderly individuals were categorized as SNAP. Furthermore, a substantial number of elderly individuals labelled as primary age-related tauopathy (PART) harbor frequent NFT in the medial temporal lobe, basal forebrain, brainstem, olfactory bulbs and cortex without amyloid plaque deposits. Clinically these PART individuals exhibited cognitive function ranging from normal to severely impaired [78]. Additional longitudinal studies will be required to establish whether all PART subjects are destined to develop amyloid deposits and LOAD with the passage of sufficient time.

It is possible that imaging methods are comparatively insensitive to incipient amyloid deposits.  However, a study of subjects clinically diagnosed with mild to moderate LOAD [79] revealed a SNAP-like subgroup with cortical soluble and insoluble Aβ levels that were low or fell below ELISA detection limits. These results undermine attempts to explain LOAD dementia as a simple and direct consequence of the presence of soluble or aggregated toxic amyloid species. Further, they reveal that the standard model of LOAD progression [80] in which amyloid accumulates relentlessly for years and precedes the appearance of NFT and dementia is not universal. The data confirm the earliest phases of LOAD, the point deemed critical for preventative therapeutic intervention, is both poorly defined and neuropathologically complex.

The recent discovery that brain hypometabolism and atrophy sometimes precede amyloid deposition suggest that normalcy and LOAD are more precisely differentiated by the presence of biomarkers of neurodegeneration . A comprehensive reassessment of discreet AD neuropathological and neurodegeneration biomarkers in the context of NIA-AA clinical criteria has been recently published . The new scheme opens the possibility for the selection of more precisely defined cohorts for future clinical trials. The addition of other biomarkers such as those involving vascular, endocrine and immune disturbances will further refine the nosology of what we currently designate as ADfunction may help clarify whether anti-amyloid therapy would be most beneficial to only a subclass of dementia patients.

Transgenic mice do not produce the biochemically complex pathological events exhibited in human AD [83-85], perhaps explaining why the identical therapeutic interventions have been consistently far more effective in animals than cognitively impaired trial participants. Moreover, these observations reveal the pathophysiology and clinical changes observed in LOAD are the culmination of unique and complicated human disease processes, impossible to model fully in experimental animals.

The experiences of AD interventions

Immunotherapy and other amyloid diminution strategies that proved successful in transgenic mice [86] have yielded limited beneficial outcomes against AD in clinical trials. For a detailed description of these clinical trials and their results the reader is referred to the comprehensive reviews published in the Alzforum database (http://www.alzforum.org/therapeutics). The combined body of evidence from these disappointing efforts to halt AD dementia has led to concerns that the amyloid hypothesis [87,88] must be reevaluated [89] or is channeling the field into unproductive therapeutic directions [15,62].  

Some of the strongest evidence directly implicating amyloid in dementia instigation is genetic; mutations in PSEN and APP genes, as well as APP gene duplications, lead to what we defined as EOAD. Despite these links and detailed molecular characterizations, it is not possible to specify the role(s) played by amyloid and other APP fragments in AD [90-91] nor, given the enormous list of critical ɣ-secretase substrates, is it clear how PSEN mutations produce neurodegeneration. Presenilin gene conditional knockout mice exhibit reduced Aβ production and increased inflammatory reactions [92] and detailed examination of PSEN mutations has revealed unexpected diversity in the corresponding impacts on Aβ production [19,93-98]. It is now clear that the pathologic and clinical effects of these mutations are impossible to match with simple notions of enhanced production of Aβ42 or altered Aβ40/Aβ42 ratios. Noting that amyloid production is not necessarily required for AD prompted the proposal of the PSEN hypothesis [99] in which the partial loss of PSEN gene function provokes neurodegeneration and AD pathology.  This model predicts that inhibition of ɣ-secretase would actually aggravate dementia [100]. 

It is noteworthy that a large-scale clinical trial of the ɣ-secretase inhibitor semagacestat was terminated when analysis revealed the subjects were experiencing accelerated cognitive function declines and other serious adverse events such as cancer [101]. Although failing to achieve the desired clinical endpoints, the trial might provide fresh insights into the mechanism of dementia production through the biochemical activities of PSEN. Postmortem neuropathological and neurochemical evaluations of trial participants may help clarify the nature of treatment responses and expose the existence of any common patterns.  A comprehensive results report [101] revealed 26 subjects receiving semagacestat expired during the clinical trial. Detailed postmortem biochemical investigations of this participant subgroup, in particular, will be extraordinarily important, but 5 years after the trial termination only a single assessment has been published [102]. Other clinical trials involving the ɣ-secretase inhibitors were also halted due to undesirable side effects [103,104]. In spite of these frustrations controlling Aβ production by modulating the activity of the β- and ɣ- secretases is being actively pursued (see Alzforum database: http://www.alzforum.org/therapeutics).

Immunotherapy interventions to mitigate AD dementia have been predicated on the assumption that unmodified Aβ deposits are the prime targets. The clear biochemical complexity of Aβ peptides in AD and the comparatively vague understanding of their functions raise the unsettling prospect that interventions have not succeeded in changing the clinical course of dementia because they were targeted inappropriately or the clinical trials were incorrectly designed [105-108]. However, a precise deconvolution of immunotherapy clinical trial data revealed some encouraging results that were initially obscured due to grouping together subjects in the treatment cohort with different levels of individual clinical conditions coupled with large patient dropouts from the placebo group. However, recent results indicate that the use of monoclonal antibodies in large phase-3 clinical trials comprising individuals with mild LOAD failed to meet their primary end-points (http://www.alzforum.org/therapeutics). In general, these observations suggest that independent of amyloid accumulation other pleiotropic dysfunctional molecules, perturbed biochemical pathways and/or systemic comorbidities are involved in the evolution of LOAD neurodegeneration.

The primary amino acid sequence of APP has been highly conserved through evolutionary history suggesting an underlying functional importance for both the intact molecules and its proteolytically processed derivatives [109-111].  Although AD is defined on the basis of plaque and tangle neuropathology, considered in a broader evolutionary context it is difficult to assign unambiguous pathologic effects to amyloid deposits [112]. Brain amyloid deposition is linked to aging and dementia in humans as well as other mammals including canines and primates, but these animals lack NFT and are incomplete LOAD models [113].

Several investigators speculate that soluble oligomeric Aβ represent the most neurotoxic species in AD while more massive amyloid deposits are comparatively benign [13-17]. This suggests the sudden and complete disruption of amyloid deposits may be detrimental by unleashing toxic species as well as eliciting vasogenic edema and microhemorrhages [114,115] aggravating neuroinflammation. Examination of the aftermath following active immunization suggests that mobilized Aβ remained trapped in the brain [116-119]. Passive immunization with humanized monoclonal antibodies against Aβ exhibited adverse effects, which were exacerbated in subjects with APOE ε4 genotypes [120]. These deleterious effects may result from disruption of vascular amyloid deposited to seal breaches in the blood-brain barrier [90], blocking the presumed antimicrobial activity of Aβ [121,122], interference with the heavy metal chelating ability of Aβ   [123-124], meddling with the vasoconstriction induced by Aβ/endothelin-1 activity [125,126] or disturbing Aβ-mediated glucose-fatty acid energy metabolism [127]. Fresh approaches toward the design of Aβ active vaccinations, that include enhanced immunogenicity, may provide effective and affordable AD mitigation [128]. Besides assessing age, gender, personal and family clinical history, it may be necessary to adopt more holistic diagnostic and enrollment criteria which include general evaluation of cardiovascular, endocrine, immune and other systemic and metabolic dysfunctions as well as environmental and lifestyle factors [129-131].

Cardiovascular dysfunction and diabetes in AD pathogenesis

The pathogenesis and pathophysiology of LOAD may be complicated by systemic diseases and environmental factors such as cardiovascular dysregulation. Cardiovascular system disease or natural aging of the heart and vessels, and brain microcirculation may result in regional or global brain perfusion insufficiency with consequent negative effects on cognitive capacities [132].  The brain consumes disproportionate amounts of oxygen and has no energy reserves making an adequate blood supply crucial to proper function [133]. Regional brain activity levels may be inferred by detecting rapid changes in blood oxygenation levels (BOLD) which are presumed to reflect neuronal and associated tissue metabolic responses to stimulation [134]. Recent analyses have revealed an association between intracranial atherosclerosis and LOAD dementia [3-10], and significant correlations between neuropathology and cognition and cardiovascular disease as well as with age-associated cardiovascular performance decline [135-144]. Observations of LOAD subject brain activities reveal marked region-specific metabolic reductions compared to subjects without dementia suggesting both baseline perfusion level declines as well as a loss of surge capacity in response to stimulation [145-149]. An overwhelming body of epidemiologic evidence confirms that LOAD and cerebrovascular disease share common risk factors [150]. New imaging studies have indicated that in some elderly individuals, white matter rarefaction (a surrogate of cerebrovascular insufficiency), brain infarcts and amyloid exert additive effects on cognitive impairment [151]. Intriguingly, what appears to differentiate the oldest-old high pathology control subjects from age-matched LOAD cases is that the former have significantly less white matter rarefaction, less cerebral amyloid angiopathy and fewer NFT than the latter, which suggests less vascular compromise [12]. Recently, an integrative multifactorial data-driven model of LOAD suggested that vascular dysregulation plays a preeminent role in the pathogenesis and progression of this dementia and apparently precedes Aβ manifestations [152]. 

A substantial fraction of dementia cases harbors a heterogeneous blend of neuropathological lesions and vascular malfunction described as ‘mixed’ pathology [3,107-110] [3, 153-156]. Although putatively independent of each other [157] these pathological changes do not exist in isolation and using cognitive impairment as a benchmark, it is clear coexisting cerebrovascular malfunction amplifies the deleterious effects of LOAD pathology and accelerates dementia progression [150,157] with important therapeutic implications [158]. Impaired circulation will yield commensurate energy production failure and overall metabolic disturbances and these will have significant consequences for neural function [127]. In addition, the age-associated brain metabolic fueling pathway shifts from glycolysis to ketone body oxidation, reminiscent of hibernation [127,159]. This adjustment may represent an alternative rescue or adaptive function enabling vital neural activities to continue at nominal levels despite suboptimal perfusion and physiological conditions. The complexity of brain physiology coupled with the highly heterogeneous nature of neuropathology may have hindered attempts to mitigate AD dementia using therapeutics focused on amyloid deposit disruption [153].

If circulatory system insufficiency induces or exacerbates AD, restoring blood flow should improve cognitive function [132]. Aggressive medical therapy, artery bypass and stenting methods have been successfully attempted as a means to restore brain blood flow [160-162]. Systematic exploration, of respiratory and cardiovascular disorders related to dysfunctional breathing, lung and heart maladies, hemodynamic dysregulation, arterial stiffness and cerebrovascular disease as well as assessment of the blood-brain barrier (BBB) conditions in relation to cognitive evaluations and treatment of memory impaired patients is long overdue  [132,163-168]. New devices to monitor cardiovascular function and blood chemistry on a continuous basis together with imaging-based determinations and ultrasound studies of extant or unfolding AD pathology and cognitive function may offer the potential to undertake proactive dementia preventative and effective measures. A capacity to detect and mitigate the subtle changes heralding increased risk of cognitive failure would be a cost-effective adjunct to the standard treatment approaches.

Subjects with type 2 diabetes are at elevated risk for dementia. This tenet is based on studies suggesting decreased brain insulin and insulin receptors, including insulin-like growth factor and reduced CSF insulin levels. Although these observations have been controversial [169], brain insulin administration enhances memory and cognition in humans [170]. Brain insulin resistance appears to be responsible for grave alterations in energy metabolism and mitochondria dysregulation which could explain amyloid and NFT deposition as well as white matter atrophy, brain microvascular disease, neuroinflammation and glial and neuronal demise [171]. Recent investigations have demonstrated that long lasting insulin (‘insulin detemir’) administered intranasally improved cognition in individuals with MCI and AD [172]. An ongoing Study of Nasal Insulin to Fight Forgetfulness (SNIFF) is evaluating the effects of insulin on amnestic MCI and early AD individuals. It remains unclear whether insulin dysregulation and hyperglycemia actually cause the pathologic changes of AD [173].  However, vascular damage is prominent in both conditions and chronic diabetes may promote malfunction of the blood-brain barrier associated with AD [174].  Alzheimer disease treatments have focused on eliminating amyloid deposits, but the intricate interrelationship between dementia and diabetes suggests that successful long term mitigation will also necessitate careful management of insulin and glucose levels.

Between now and then – modifying the trajectory of AD dementia while seeking an elusive cure

The long effort toward unraveling the pathologic mechanisms of AD has not yielded therapeutic successes commensurate with the enormous investment of funds and effort. Mindful of oft-repeated ominous demographic trends [175] new approaches are needed. However, while no miracle cure is in the offing, we are not on an inevitable arc toward disaster. Perhaps the message that LOAD risk is potentially modifiable by multiple interventions has been lost in the pursuit of a universal remedy.

Alzheimer’s disease research investigations are often harmonized with the amyloid cascade hypothesis. However, we may need to move past the widely accepted assumptions that Aβ species or their soluble/oligomeric/fibrillary forms are the only culprits underlying the pathogenesis of neurodegeneration and clinical manifestations of AD. A crucial first experimental step, capable of validating or refuting the amyloid hypothesis, will be to clearly establish the molecular forms of Aβ that are toxic to the brain and investigate their biophysical properties. Validation of this hypothesis may demand biopsies rather than experimental cell lines or animal models and will present a challenge due to the kaleidoscopic heterogeneity of Aβ. However, the ultimate validation of the amyloid cascade hypothesis will enable precisely targeted therapy which may produce significant results against dementia.

The route to dementia is varied and complex and we have simply equated EOAD and LOAD on the bases of their most striking neuropathological lesions [176]. Although the current mechanistic model is economical, exploring biochemical, biophysical and biomarker differences will lead to a better nosological understanding. Analogous to the situation in cancer research, the passage of time and accumulation of additional data may force an appreciation for the underlying biochemical heterogeneity of dementia and its implications  [177-179].

New technologies embracing the use of human pluripotent stem cells and 3-dimensional human cell culture systems, which mimic the biochemistry and physiology of the human brain, may offer suitable models to better understand the dynamics of dementia than phylogenetically distant animal paradigms. The use of human brain biopsies [180] will be an indispensable approach that will provide information on the typical sequence of events preceding or precipitating the distinct classes of dementia. They will also afford appraisals of protein, lipid, carbohydrate and metabolite pools with state-of-the-art mass spectrometry and ultrasensitive immunoassays as well as ultramicroscopic assessments of structural cellular scaffolds, organelles and membrane pathology. The availability of biopsied brain tissue will also enable the discovery of uniquely time-dependent epigenetic changes and disturbed metabolic pathways involved in the pathogenesis and evolution of the different types of dementia. Furthermore, brain biopsies will also allow harvesting of autologous replacement cells that may restore cognitive function [181]. Neuroimaging advances will supply further information on specific quantitative imaging-detectable neuropathology such as Aβ, tau, α-synuclein, TDP-43 and other deviant molecular forms enabling improved selection and cohorting of patients for clinical trials. Neuroimaging innovations will also clarify the temporal sequence of neuronal and glial degeneration including myelin loss, defective connectivity configurations, synaptic density, brain microvascular disease, BBB status and detailed quantitation of chronic hypoperfusion and its functional consequences. In addition, improved computational models that integrate molecular network dynamics and data processing analysis will be pivotal in understanding the origin and dynamic evolution of the complex chain of inter-related events that lead to different forms of neurodegeneration. 

Multiple lines of biochemical and clinical data confirm that LOAD is not inevitable and strongly suggest several important medical and behavioral risk factors are modifiable [182, 183]. The incidence of dementia may have stabilized or decreased in some high-income countries over three decades [184-185]. Although the exact causes behind these trends are unknown, they may reflect improved provision of medical services, enhanced educational levels and better control of cardiovascular diseases in addition to healthier diets and increased physical activity. Regrettably, a concurrent upsurge in obesity, diabetes and hypertension in the general population as well as the increases in life expectancy and population numbers may reverse these trends in the foreseeable future. 

The Religious Order Study [156] and other investigations [186,187] reveal LOAD development risk may be recognizable early and imply the trajectory toward dementia could be altered through several intervention strategies. Late onset Alzheimer’s disease emergence is clearly correlated with readily recognized and prevalent exacerbating medical issues such as diabetes  [188, 189], mid-life hypertension [166, 190, 191] cardiovascular diseases [2, 166, 168, 192], depression [193-196] and anxiety [197] that are potentially subject to direct medicinal mitigation. In addition, practical behavioral interventions such as stress reduction strategies [198], regular exercise regimens [199-202], social engagement [203-206], intellectual attainment ] [207-210], spirituality [211-215] and weight control [216-218] that are under the personal management of subjects may exert positive influences against dementia development risks. The vast unexplored and simultaneously underexploited territory of LOAD is the impact of the environment [219]. Nearly 25 years ago, Martin and Fukuchi [220] noted the complex interrelations between environmental factors and intrinsic aging processes that sometimes culminates in dementia.

It is clear that amyloid plays a pivotal role in the pathogenesis and pathophysiology of AD. The combined investment of research effort by academia and the pharmaceutical industry in search of an effective AD treatment has expanded understanding of dementia development and fundamental brain chemistry and physiology.  Although there seem to be several pathways to dementia, it is important to recognize the contributions to healthy aging that active lifestyle interventions and education offer. Ultimately, as new imaging studies have confirmed, a substantial fraction of dementia cases may have etiologies distinct from what we presently designate as LOAD. Perhaps cancers sparked by an assortment of interacting genetic, epigenetic, physiologic and environmental factors, provide an apt analogy for dementia therapy. Vanquishing the multifactorial disorder, we know as Alzheimer’s disease may oblige the implementation of several strategies because there are many ways to cross the Rubicon of dementia.

Acknowledgements

We express our gratitude to Dr. Vincent Marchesi, Dr. Walter Kalback and Ms. Chera Maarouf for critical review of the manuscript. The present study was supported in part by Midwestern University.

The authors declare no competing interests.

References

  1. Pimplikar SW, Nixon RA, Robakis NK, Shen J, Tsai LH (2010) Amyloid-independent mechanisms in Alzheimer's disease pathogenesis. J Neurosci 30: 14946-14954.[Crossref]
  2. Lock M (2013) The Alzheimer Conundrum: Entanglements of Dementia and Aging. Princeton, New Jersey: Princeton University Press.
  3. Schneider JA, Bennett DA (2010) Where vascular meets neurodegenerative disease. Stroke 41: S144-146. [Crossref]
  4. Crystal HA, SchneiderJA, Bennett DA, Leurgans S, Levine SR (2014) Associations of cerebrovascular and Alzheimer's disease pathology with brain atrophy. Curr Alzheimer Res 11: 309-316. [Crossref]
  5. Hunter JM, Kwan J, Malek-Ahmadi M, Maarouf CL, Kokjohn TA, et al.  (2012) Morphological and pathological evolution of the brain microcirculation in aging and Alzheimer's disease. PLoS One 7: e36893. [Crossref]
  6. Roher AE, Tyas SL, Maarouf CL, Daugs ID, Kokjohn TA, et al. (2011) Intracranial atherosclerosis as a contributing factor to Alzheimer's disease dementia. Alzheimers Dement 7: 436-444. [Crossref]
  7. Roher AE, Garami Z, Tyas SL, Maarouf CL, Kokjohn TA, et al. (2011) Transcranial Doppler ultrasound blood flow velocity and pulsatility index as systemic indicators for Alzheimer's disease. Alzheimers Dement 7: 445-455. [Crossref]
  8. Beach TG, Wilson JR, Sue LI, Newell A, Poston M, et al. (2007) Circle of Willis atherosclerosis: association with Alzheimer's disease, neuritic plaques and neurofibrillary tangles. Acta Neuropathol 113: 13-21. [Crossref]
  9. Roher AE, Esh C, Rahman A, Kokjohn TA, Beach TG (2004) Atherosclerosis of cerebral arteries in Alzheimer disease. Stroke 35: 2623-2627. [Crossref]
  10. Kalback W, Esh C, Castano EM, Rahman A, Kokjohn T, et al. (2004) Atherosclerosis, vascular amyloidosis and brain hypoperfusion in the pathogenesis of sporadic Alzheimer's disease. Neurol Res 2004; 26: 525-539. [Crossref]
  11. Le Couteur DG, Doust J, Creasey H, Brayne C (2013) Political drive to screen for pre-dementia: not evidence based and ignores the harms of diagnosis. BMJ 347: f5125. [Crossref]
  12. Maarouf CL, Daugs ID, Kokjohn TA, Walker DG, Hunter JM, et al. (2011) Alzheimer's disease and non-demented high pathology control nonagenarians: comparing and contrasting the biochemistry of cognitively successful aging. PLoS One 6: e27291. [Crossref]
  13. Robakis NK (2010) Are Abeta and its derivatives causative agents or innocent bystanders in AD? Neurodegener Dis 7: 32-37. [Crossref]
  14. Lesne SE, Sherman MA, Grant M, Kuskowski M, Schneider JA, et al. (2013) Brain amyloid-β oligomers in ageing and Alzheimer's disease. Brain 136: 1383-1398. [Crossref]
  15. Morris GP, Clark IA, Vissel B (2014) Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer's disease. Acta Neuropathol Commun 2: 135. [Crossref]
  16. Cerasoli E, Ryadnov MG, Austen BM (2015) The elusive nature and diagnostics of misfolded Aβ oligomers. Front Chem 3: 17. [Crossref]
  17. Lesne SE (2014) Toxic oligomer species of amyloid- β in Alzheimer's disease, a timing issue. Swiss Med Wkly 144: w14021. [Crossref]
  18. Roher AE, Lowenson JD, Clarke S, Wolkow C, Wang R, et al. (1993) Structural alterations in the peptide backbone of beta-amyloid core protein may account for its deposition and stability in Alzheimer's disease. J Biol Chem 268: 3072-3083. [Crossref]
  19. Miravalle L, Calero M, Takao M, Roher AE, Ghetti B, et al. (2005) Amino-terminally truncated Abeta peptide species are the main component of cotton wool plaques. Biochemistry 44: 10810-10821. [Crossref]
  20. Fukuda H, Shimizu T, Nakajima M, Mori H, Shirasawa T (1999) Synthesis, aggregation, and neurotoxicity of the Alzheimer's Abeta1-42 amyloid peptide and its isoaspartyl isomers. Bioorg Med Chem Lett 9: 953-956. [Crossref]
  21. Shimizu T, Watanabe A, Ogawara M, Mori H, Shirasawa T (2000) Isoaspartate formation and neurodegeneration in Alzheimer's disease. Arch Biochem Biophys 381: 225-234. [Crossref]
  22. Shimizu T, Matsuoka Y, Shirasawa T (2005) Biological significance of isoaspartate and its repair system. Biol Pharm Bull 28: 1590-1596. [Crossref]
  23. Shin Y, Cho HS, Fukumoto H, Shimizu T, Shirasawa T, et al. (2003) Abeta species, including IsoAsp23 Abeta, in Iowa-type familial cerebral amyloid angiopathy. Acta Neuropathol 105: 252-258. [Crossref]
  24. Tomidokoro Y, Rostagno A, Neubert TA, Lu Y, Rebeck GW, et al. (2010) Iowa variant of familial Alzheimer's disease: accumulation of posttranslationally modified AbetaD23N in parenchymal and cerebrovascular amyloid deposits. Am J Pathol 176: 1841-1854. [Crossref]
  25. Iwatsubo T, Saido TC, Mann DM, Lee VM, Trojanowski JQ (1996) Full-length amyloid-beta (1-42(43)) and amino-terminally modified and truncated amyloid-beta 42(43) deposit in diffuse plaques. Am J Pathol 149: 1823-1830. [Crossref]
  26. Mori H, Takio K, Ogawara M, Selkoe DJ (1992) Mass spectrometry of purified amyloid beta protein in Alzheimer's disease. J Biol Chem 267: 17082-17086. [Crossref]
  27. Kuo YM, Emmerling MR, Woods AS, Cotter RJ, Roher AE (1997) Isolation, chemical characterization, and quantitation of A beta 3-pyroglutamyl peptide from neuritic plaques and vascular amyloid deposits. Biochem Biophys Res Commun 237:188-191. [Crossref]
  28. Saido TC, Iwatsubo T, Mann DM, Shimada H, Ihara Y, et al. (1995) Dominant and differential deposition of distinct beta-amyloid peptide species, A beta N3(pE), in senile plaques. Neuron 14: 457-466. [Crossref]
  29. He W, Barrow CJ (1999) The A beta 3-pyroglutamyl and 11-pyroglutamyl peptides found in senile plaque have greater beta-sheet forming and aggregation propensities in vitro than full-length A beta. Biochemistry 38: 10871-10877. [Crossref]
  30. Harigaya Y, Saido TC, Eckman CB, Prada CM, Shoji M, et al. (2000) Amyloid beta protein starting pyroglutamate at position 3 is a major component of the amyloid deposits in the Alzheimer's disease brain. Biochem Biophys Res Commun 276: 422-427. [Crossref]
  31. Russo C, Violani E, Salis S, Venezia V, Dolcini V (2002) Pyroglutamate-modified amyloid beta-peptides--AbetaN3(pE)--strongly affect cultured neuron and astrocyte survival. J Neurochem 82: 1480-1489. [Crossref]
  32. Gunn AP, Masters CL, Cherny RA (2010) Pyroglutamate-Aβ: role in the natural history of Alzheimer's disease. Int J Biochem Cell Biol 42: 1915-1918. [Crossref]
  33. Kumar S, Rezaei-Ghaleh N, Terwel D, Thal DR, Richard M, et al. (2011) Extracellular phosphorylation of the amyloid beta-peptide promotes formation of toxic aggregates during the pathogenesis of Alzheimer's disease. EMBO J 30: 2255-2265. [Crossref]
  34. Kumar S, Wirths O, Theil S, Gerth J, Bayer TA, et al. (2013) Early intraneuronal accumulation and increased aggregation of phosphorylated Abeta in a mouse model of Alzheimer's disease. Acta Neuropathol 125: 699-709. [Crossref]
  35. Butterfield DA, Sultana R (2011) Methionine-35 of aβ (1-42): importance for oxidative stress in Alzheimer disease. J Amino Acids 2011: 198430. [Crossref]
  36. Butterfield DA, Kanski J (2002) Methionine residue 35 is critical for the oxidative stress and neurotoxic properties of Alzheimer's amyloid beta-peptide 1-42. Peptides 23: 1299-1309. [Crossref]
  37. Ali FE, Separovic F, Barrow CJ, Cherny RA, Fraser F, et al. (2005) Methionine regulates copper/hydrogen peroxide oxidation products of Abeta. J Pept Sci 11: 353-360. [Crossref]
  38. Kuo YM, Emmerling MR, Vigo-Pelfrey C, Kasunic TC, Kirkpatrick JB, et al. (1996) Water-soluble Abeta (N-40, N-42) oligomers in normal and Alzheimer disease brains. J Biol Chem 271: 4077-4081. [Crossref]
  39. Kuo YM, Webster S, Emmerling MR, De Lima N, Roher AE (1998) Irreversible dimerization/tetramerization and post-translational modifications inhibit proteolytic degradation of A beta peptides of Alzheimer's disease. Biochim Biophys Acta 1406: 291-298. [Crossref]
  40. Masters CL, Selkoe DJ (2012) Biochemistry of amyloid β-protein and amyloid deposits in Alzheimer disease. Cold Spring Harb Perspect Med 2: a006262. [Crossref]
  41. Roher AE, Baudry J, Chaney MO, Kuo YM, Stine WB, et al. (2000) Oligomerizaiton and fibril asssembly of the amyloid-beta protein. Biochim Biophys Acta 1502: 31-43.
  42. Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, et al. (2003) Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300: 486-489. [Crossref]
  43. Kokubo H, Kayed R, Glabe CG, Yamaguchi H (2005) Soluble Abeta oligomers ultrastructurally localize to cell processes and might be related to synaptic dysfunction in Alzheimer's disease brain. Brain Res 1031: 222-228. [Crossref]
  44. Garzon-Rodriguez W, Sepulveda-Becerra M, Milton S, Glabe CG (1997) Soluble amyloid Abeta-(1-40) exists as a stable dimer at low concentrations. J Biol Chem 272: 21037-21044. [Crossref]
  45. Deshpande A, Mina E, Glabe C, Busciglio J (2006) Different conformations of amyloid beta induce neurotoxicity by distinct mechanisms in human cortical neurons. J Neurosci 26: 6011-6018. [Crossref]
  46. Jin M, Shepardson N, Yang T, Chen G, Walsh D, et al. (2011) Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A 108: 5819-5824. [Crossref]
  47. Mc Donald JM, Savva GM, Brayne C, Welzel AT, Forster G, et al. (2010) The presence of sodium dodecyl sulphate-stable Abeta dimers is strongly associated with Alzheimer-type dementia. Brain 133: 1328-1341. [Crossref]
  48. Watt AD, Perez KA, Rembach A, Sherrat NA, Hung LW, et al. (2013) Oligomers, fact or artefact? SDS-PAGE induces dimerization of beta-amyloid in human brain samples. Acta Neuropathol 2013; 125:549-564. [Crossref]
  49. Hung LW, Ciccotosto GD, Giannakis E, Tew DJ, Perez K, et al. (2008) Amyloid-beta peptide (Abeta) neurotoxicity is modulated by the rate of peptide aggregation: Abeta dimers and trimers correlate with neurotoxicity. J Neurosci 28: 11950-11958. [Crossref]
  50. Hong S, Ostaszewski BL, Yang T, O'Malley TT, Jin M, et al. (2014) Soluble Abeta Oligomers Are Rapidly Sequestered from Brain ISF In Vivo and Bind GM1 Ganglioside on Cellular Membranes. Neuron 82:308-319. [Crossref]
  51. Crouch PJ, Blake R, Duce JA, Ciccotosto GD, Li QX (2005) Copper-dependent inhibition of human cytochrome c oxidase by a dimeric conformer of amyloid-beta1-42. J Neurosci 25: 672-679. [Crossref]
  52. Kim BH, Palermo NY, Lovas S, Zaikova T, Keana JF, et al. (2011) Single-molecule atomic force microscopy force spectroscopy study of Abeta-40 interactions. Biochemistry 50: 5154-5162. [Crossref]
  53. Lv Z, Roychaudhuri R, Condron MM, Teplow DB, Lyubchenko YL (2013) Mechanism of amyloid beta-protein dimerization determined using single-molecule AFM force spectroscopy. Sci Rep 3: 2880.
  54. Watts JC, Condello C, Stohr J, Oehler A, Lee J, et al. (2014) Serial propagation of distinct strains of Aβ prions from Alzheimer's disease patients. Proc Natl Acad Sci U S A 111: 10323-10328. [Crossref]
  55. Kane MD, Lipinski WJ, Callahan MJ, Bian F, DurhamvRA, et al. (2000) Evidence for seeding of beta -amyloid by intracerebral infusion of Alzheimer brain extracts in beta -amyloid precursor protein-transgenic mice. J Neurosci 20: 3606-3611.
  56. Jucker M, Walker LC (2013) Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 501: 45-51. [Crossref]
  57. Pimplikar SW (2009) Reassessing the amyloid cascade hypothesis of Alzheimer's disease. Int J Biochem Cell Biol 41: 1261-1268. [Crossref]
  58. Armstrong RA (2011) The pathogenesis of Alzheimer's disease: a reevaluation of the "amyloid cascade hypothesis". Int J Alzheimers Dis 2011: 630865. [Crossref]
  59. Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, et al. (2012) Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol 71: 362-381. [Crossref]
  60. Castellani RJ1, Zhu X, Lee HG, Moreira PI, Perry G, et al. (2007) Neuropathology and treatment of Alzheimer disease: did we lose the forest for the trees? Expert Rev Neurother 7: 473-485. [Crossref]
  61.  [No authors listed] (2011) The amyloid cascade hypothesis has misled the pharmaceutical industry. Biochem Soc Trans 39: 920-923. [Crossref]
  62. Mullane K, Williams M (2013) Alzheimer's therapeutics: continued clinical failures question the validity of the amyloid hypothesis-but what lies beyond? Biochem Pharmacol 2013; 85:289-305. [Crossref]
  63. Drachman DA (2014) The amyloid hypothesis, time to move on: Amyloid is the downstream result, not cause, of Alzheimer's disease. Alzheimers Dement 10: 372-380. [Crossref]
  64. Musiek ES, Holtzman DM (2015) Three dimensions of the amyloid hypothesis: time, space and 'wingmen'. Nat Neurosci 18: 800-806. [Crossref]
  65. Willem M, Tahirovic S, Busche MA, Ovsepian SV, Chafai M, et al. (2015) eta-Secretase processing of APP inhibits neuronal activity in the hippocampus. Nature 526: 443-447. [Crossref]
  66. Nikolaev A, McLaughlin T, O'Leary DD, Tessier-Lavigne M (2009) APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 457: 981-989. [Crossref]
  67. Simons M, De SB, Multhaup G, Tienari PJ, Dotti CG, et al. (1996) Amyloidogenic processing of the human amyloid precursor protein in primary cultures of rat hippocampal neurons. J Neurosci 16: 899-908. [Crossref]
  68. Zhang Z, Song M, Liu X, Su KS, Duong DM, et al. (2015) Delta-secretase cleaves amyloid precursor protein and regulates the pathogenesis in Alzheimer's disease. Nat Commun 6: 8762. [Crossref]
  69. Berger-Sweeney J, McPhie DL, Arters JA, Greenan J, Oster-Granite ML, et al. (1999) Impairments in learning and memory accompanied by neurodegeneration in mice transgenic for the carboxyl-terminus of the amyloid precursor protein. Brain Res Mol Brain Res 66:150-162. [Crossref]
  70. Nhan HS, Chiang K, Koo EH (2015) The multifaceted nature of amyloid precursor protein and its proteolytic fragments: friends and foes. Acta Neuropathol 129: 1-19. [Crossref]
  71. Ghosal K, Pimplikar SW (2011) Aging and excitotoxic stress exacerbate neural circuit reorganization in amyloid precursor protein intracellular domain transgenic mice. Neurobiol Aging 32: 2320-2329. [Crossref]
  72. Rovelet-Lecrux A, Hannequin D, Raux G, Le Meur N, Laquerriere A, et al. (2006) Campion, APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet 38: 24-26. [Crossref]
  73. Oyama F, Cairns NJ, Shimada H, Oyama R, Titani K, et al. (1994) Down's syndrome: up-regulation of beta-amyloid protein precursor and tau mRNAs and their defective coordination. J Neurochem 62: 1062-1066. [Crossref]
  74. Selkoe DJ, Hardy J2 (2016) The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med 8: 595-608. [Crossref]
  75. Jack CR, Wiste HJ, Weigand SD, Knopman DS, Lowe V, et al, (2013) Amyloid-first and neurodegeneration-first profiles characterize incident amyloid PET positivity. Neurology 81: 1732-1740. [Crossref]
  76. Benzinger TL, Blazey T, Jack CR, Jr., Koeppe RA, Su Y, et al. (2013) Regional variability of imaging biomarkers in autosomal dominant Alzheimer's disease. Proc Natl Acad Sci U S A 2013; 110: E4502-E4509. [Crossref]
  77. Jack CR, Jr., Knopman DS, Weigand SD, Wiste HJ, Vemuri P, et al. (2012) An operational approach to National Institute on Aging-Alzheimer's Association criteria for preclinical Alzheimer disease. Ann Neurol 71:765-775. [Crossref]
  78. Crary JF, Trojanowski JQ, Schneider JA, Abisambra JF, Abner EL, et al. (2014) Primary age-related tauopathy (PART): a common pathology associated with human aging. Nelson. Acta Neuropathol 128: 755-766. [Crossref]
  79. Monsell SE, Kukull WA, Roher AE, Maarouf CL, Serrano G, et al. (2015) Characterizing Apolipoprotein E ε4 Carriers and Noncarriers With the Clinical Diagnosis of Mild to Moderate Alzheimer Dementia and Minimal β-Amyloid Peptide Plaques. JAMA Neurol 72: 1124-1131. [Crossref]
  80. Jack CR, Knopman DS, Jagust WJ, Petersen RC, Weiner MW, et al. (2013) Tracking pathophysiological processes in Alzheimer's disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol 12:207-216. [Crossref]
  81. Jack CR, Wiste HJ, Weigand SD, Knopman DS, Mielke MM, et al. (2015) Different definitions of neurodegeneration produce similar amyloid/neurodegeneration biomarker group findings. Brain 138: 3747-3759. [Crossref]
  82. Jack CR, Bennett DA, Blennow K, Carrillo MC, Feldman HH, et al. (2016) A/T/N: an unbias descriptive classification scheme for Alzheimer disease biomarkers. Neurology 87: 539-547. [Crossref]
  83. Kuo YM, Kokjohn TA, Beach TG, Sue LI, Brune D, et al. (2001) Comparative analysis of amyloid-beta chemical structure and amyloid plaque morphology of transgenic mouse and Alzheimer's disease brains. J Biol Chem 276: 12991-12998. [Crossref]
  84. Kalback W, Watson MD, Kokjohn TA, Kuo YM, Weiss N, et al. (2002) APP transgenic mice Tg2576 accumulate Abeta peptides that are distinct from the chemically modified and insoluble peptides deposited in Alzheimer's disease senile plaques. Biochemistry 41: 922-928. [Crossref]
  85. Kokjohn TA, Roher AE (2009) Amyloid precursor protein transgenic mouse models and Alzheimer's disease: understanding the paradigms, limitations, and contributions. Alzheimers Dement 5: 340-347. [Crossref]
  86. Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, et al. (1999) Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400: 173-177. [Crossref]
  87. Masters CL (1984) Etiology and pathogenesis of Alzheimer's disease. Pathology 16: 233-234. [Crossref]
  88. Hardy JA, Higgins GA (1992) Alzheimer's disease: the amyloid cascade hypothesis. Science 256: 184-185. [Crossref]
  89. Hardy J (2009) The amyloid hypothesis for Alzheimer's disease: a critical reappraisal. J Neurochem 110: 1129-1134. [Crossref]
  90. Kokjohn TA, Maarouf CL, Roher AE (2012) Is Alzheimer's disease amyloidosis the result of a repair mechanism gone astray? Alzheimers Dement 8: 574-583. [Crossref]
  91. Roher AE, Esh CL, Kokjohn TA, Castano EM, Van Vickle GD, et al. (2009) Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer's disease. Alzheimers Dement 5: 18-29. [Crossref]
  92. Beglopoulos V, Sun X, Saura CA, Lemere CA, Kim RD, et al. (2004) Reduced beta-amyloid production and increased inflammatory responses in presenilin conditional knock-out mice. J Biol Chem 279: 46907-46914. [Crossref]
  93. Steiner H, Revesz T, Neumann M, Romig H, Grim MG, et al. (2001) A pathogenic presenilin-1 deletion causes abberrant Abeta 42 production in the absence of congophilic amyloid plaques. J Biol Chem 276: 7233-7239.
  94. Roher AE, Maarouf CL, Malek-Ahmadi M, Wilson J, Kokjohn TA, et al. (2013) Subjects harboring presenilin familial Alzheimer's disease mutations exhibit diverse white matter biochemistry alterations. Am J Neurodegener Dis 2: 187-207. [Crossref]
  95. Heilig EA, Xia W, Shen J, Kelleher RJ III (2010) A presenilin-1 mutation identified in familial Alzheimer disease with cotton wool plaques causes a nearly complete loss of gamma-secretase activity. J Biol Chem 285: 22350-22359. [Crossref]
  96. Maarouf CL, Daugs ID, Spina S, Vidal R, Kokjohn TA, et al. (2008) Histopathological and molecular heterogeneity among individuals with dementia associated with Presenilin mutations. Mol Neurodegener 2008; 3:20. [Crossref]
  97. Van Vickle GD, Esh CL, Kalback WM, Patton RL, Luehrs DC, et al. (2007) TgCRND8 amyloid precursor protein transgenic mice exhibit an altered gamma-secretase processing and an aggressive, additive amyloid pathology subject to immunotherapeutic modulation. Biochemistry 46: 10317-10327. [Crossref]
  98. Roher AE, Kokjohn TA, Esh C, Weiss N, Childress J, et al. (2004) The human amyloid-beta precursor protein770 mutation V717F generates peptides longer than amyloid-beta-(40-42) and flocculent amyloid aggregates. J Biol Chem 279: 5829-5836. [Crossref]
  99. Shen J, Kelleher RJ 3rd (2007) The presenilin hypothesis of Alzheimer's disease: evidence for a loss-of-function pathogenic mechanism. Proc Natl Acad Sci U S A 104: 403-409. [Crossref]
  100. Kelleher RJ 3rd, Sh2021 Copyright OAT. All rights reserv and human disease. Science 330: 1055-1056. [Crossref]
  101. Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, et al. (2013) A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Engl J Med 369: 341-350. [Crossref]
  102. Roher AE, Maarouf CL, Kokjohn TA, Whiteside CM, Kalback WM, et al. (2014) Neuropathological and biochemical assessments of an Alzheimer's disease patient treated with the gamma-secretase inhibitor semagacestat. Am J Neurodegener Dis 3:115-133. [Crossref]
  103. Hall A, Patel TR (2014) γ- Secretase modulators: current status and future directions. Prog Med Chem 53: 101-145. [Crossref]
  104. Coric V, Salloway S, Van Dyck CH, Dubois B, Andreasen N, et al. (2015) Targeting Prodromal Alzheimer Disease With Avagacestat: A Randomized Clinical Trial. JAMA Neurol 72: 1324-1333. [Crossref]
  105. Robakis NK (2011) Mechanisms of AD neurodegeneration may be independent of Aβ and its derivatives. Neurobiol Aging 32: 372-379. [Crossref]
  106. Castellani RJ, Perry G (2014) The complexities of the pathology-pathogenesis relationship in Alzheimer disease. Biochem Pharmacol 88: 671-676. [Crossref]
  107. Pimplikar SW, Ghosal K (2011) Amyloid precursor protein: more than just neurodegeneration. Stem Cell Res Ther 2: 39. [Crossref]
  108. Karran E, Hardy J (2014) A critique of the drug discovery and phase 3 clinical programs targeting the amyloid hypothesis for Alzheimer disease. Ann Neurol 76:185-205. [Crossref]
  109. Yamada T, Sasaki H, Furuya H, Miyata T, Goto I, et al. (1987) Complementary DNA for the mouse homolog of the human amyloid beta protein precursor. Biochem Biophys Res Commun 149: 665-671. [Crossref]
  110. Shivers BD, Hilbich C, Multhaup G, Salbaum M, Beyreuther K, et al. (1988) Alzheimer's disease amyloidogenic glycoprotein: expression pattern in rat brain suggests a role in cell contact. EMBO J 7:1365-1370. [Crossref]
  111. Coulson EJ, Paliga K, Beyreuther K, Masters CL (2000) What the evolution of the amyloid protein precursor supergene family tells us about its function. Neurochem Int 36: 175-184. [Crossref]
  112. Castellani RJ, Lee HG, Siedlak SL, Nunomura A, Hayashi T, et al. (2009) Reexamining Alzheimer's disease: evidence for a protective role for amyloid-beta protein precursor and amyloid-beta. J Alzheimers Dis 18: 447-452. [Crossref]
  113. Selkoe DJ, Bell DS, Podlisny MB, Price DL, Cork LC (1987) Conservation of brain amyloid proteins in aged mammals and humans with Alzheimer's disease. Science 235: 873-877. [Crossref]
  114. Sperling R, Salloway S, Brooks DJ, Tampieri D, Barakos J, et al. (2012) Amyloid-related imaging abnormalities in patients with Alzheimer's disease treated with bapineuzumab: a retrospective analysis. Lancet Neurol 11: 241-249. [Crossref]
  115. Barakos J, Sperling R, Salloway S, Jack C, Gass A, et al. (2013) MR Imaging Features of Amyloid-Related Imaging Abnormalities. AJNR Am J Neuroradiol 34: 1958-1965. [Crossref]
  116. Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, et al. (2003) Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med 9: 448-452. [Crossref]
  117. Patton RL, Kalback WM, Esh CL, Kokjohn TA, Van Vickle GD, et al. (2006) Amyloid-beta peptide remnants in AN-1792-immunized Alzheimer's disease patients: a biochemical analysis. Am J Pathol 169: 1048-1063. [Crossref]
  118. Holmes C, Boche B, Wilkinson D, Yadegarfar G, Hopkins V, et al. (2008) Long-term effects of Abeta42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet 372: 216-223. [Crossref]
  119. Maarouf CL, Daugs ID, Kokjohn TA, Kalback WM, Patton RL, et al. (2010) The biochemical aftermath of anti-amyloid immunotherapy. Mol Neurodegener 5: 39. [Crossref]
  120. Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, et al. (2014) Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N Engl J Med 370: 322-333. [Crossref]
  121. Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, et al. (2010) The Alzheimer's disease-associated amyloid beta-protein is an antimicrobial peptide. PLoS One 5: e9505. [Crossref]
  122. Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Tucker S, et al. (2016) Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci Transl Med 8: 340ra72.
  123. Bush AI, Masters CL, Tanzi RE (2003) Copper, beta-amyloid, and Alzheimer's disease: tapping a sensitive connection. Proc Natl Acad Sci U S A 100: 11193-11194. [Crossref]
  124. Atwood CS, Obrenovich ME, Liu T, Chan H, Perry G, et al. (2003) Amyloid-beta: a chameleon walking in two worlds: a review of the trophic and toxic properties of amyloid-beta. Brain Res Brain Res Rev 43: 1-16. [Crossref]
  125. Paris D, Town T, Mori T, Parker TA, Humphrey J, et al. (2000) Soluble beta-amyloid peptides mediate vasoactivity via activation of a pro-inflammatory pathway. Neurobiol Aging 21: 183-197. [Crossref]
  126. Townsend KP, Obregon D, Quadros A, Patel N, Volmar C, et al. (2002) Proinflammatory and vasoactive effects of Abeta in the cerebrovasculature. Ann N Y Acad Sci 977: 65-76.
  127. Heininger K1 (2000) A unifying hypothesis of Alzheimer's disease. IV. Causation and sequence of events. Rev Neurosci 11 Spec No: 213-328. [Crossref]
  128. Agadjanyan MG, Petrovsky N, Ghochikyan A (2015) A fresh perspective from immunologists and vaccine researchers: active vaccination strategies to prevent and reverse Alzheimer's disease. Alzheimers Dement 11: 1246-1259. [Crossref]
  129. Bredesen DE, John V (2013) Next generation therapeutics for Alzheimer's disease. EMBO Mol Med 5: 795-798. [Crossref]
  130. Bredesen DE (2014) Reversal of cognitive decline: a novel therapeutic program. Aging (Albany NY) 6: 707-717. [Crossref]
  131. Bredesen DE (2015) Metabolic profiling distinguishes three subtypes of Alzheimer's disease. Aging (Albany NY) 7:  595-600. [Crossref]
  132. Picano E, Bruno RM, Ferrari GF, Bonuccelli U (2014) Cognitive impairment and cardiovascular disease: so near, so far. Int J Cardiol 175: 21-29. [Crossref]
  133. Lorthois S, Cassot F, Lauwers F (2011) Simulation study of brain blood flow regulation by intra-cortical arterioles in an anatomically accurate large human vascular network: Part I: methodology and baseline flow. Neuroimage 54: 1031-1042. [Crossref]
  134. Mullinger KJ, Mayhew SD, Bagshaw AP, Bowtell R, Francis ST (2013) Poststimulus undershoots in cerebral blood flow and BOLD fMRI responses are modulated by poststimulus neuronal activity. Proc Natl Acad Sci U S A 110: 13636-13641.
  135. Kumar R, Yadav SK2, Palomares JA2, Park B2, Joshi SH3, et al. (2015) Reduced regional brain cortical thickness in patients with heart failure. PLoS One 10: e0126595. [Crossref]
  136. Kalaria RN (2012) Risk factors and neurodegenerative mechanisms in stroke related dementia. Panminerva Med 54: 139-148. [Crossref]
  137. Pires PW, Dams Ramos CM, Matin N, Dorrance AM (2013) The effects of hypertension on the cerebral circulation. Am J Physiol Heart Circ Physiol 304: H1598-1614. [Crossref]
  138. Neumann JT, Cohan CH, Dave KR, Wright CB, Perez-Pinzon MA (2013) Global cerebral ischemia: synaptic and cognitive dysfunction. Curr Drug Targets 14: 20-35. [Crossref]
  139. Farkas E, De Wilde MC, Kiliaan AJ, Luiten PG (2002) Chronic cerebral hypoperfusion-related neuropathologic changes and compromised cognitive status: window of treatment. Drugs Today (Barc) 38: 365-376. [Crossref]
  140. Chmayssani M, Festa JR, Marshall RS (2007) Chronic ischemia and neurocognition. Neuroimaging Clin N Am 17: 313-324, viii. [Crossref]
  141. Anderson CA, Arciniegas DB (2010) Cognitive sequelae of hypoxic-ischemic brain injury: a review. NeuroRehabilitation 26: 47-63. [Crossref]
  142. Jefferson AL, Beiser AS, Himali JJ, Seshadri S, O'Donnell CJ, et al. (2015) Low cardiac index is associated with incident dementia and Alzheimer disease: the Framingham Heart Study. Circulation 131: 1333-1339. [Crossref]
  143. Jefferson AL (2010) Cardiac output as a potential risk factor for abnormal brain aging. J Alzheimers Dis 20: 813-821. [Crossref]
  144. Jefferson AL, Himali JJ, Beiser AS, Au R, Massaro JM (2010) Cardiac index is associated with brain aging: the Framingham Heart Study. Circulation 122: 690-697. [Crossref]
  145. Kapogiannis D, Mattson MP (2011) Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer's disease. Lancet Neurol 10: 187-198. [Crossref]
  146. Verclytte S, Lopes R, Lenfant P, Rollin A, Semah F, et al. (2015) Cerebral Hypoperfusion and Hypometabolism Detected by Arterial Spin Labeling MRI and FDG-PET in Early-Onset Alzheimer's Disease. J Neuroimaging 26: 207-212. [Crossref]
  147. Zou JX, Wang MJ, Lei XJ, Chen XG (2014) 3.0 T MRI arterial spin labeling and magnetic resonance spectroscopy technology in the application of Alzheimer's disease. Exp Gerontol 60: 31-36. [Crossref]
  148. Wierenga CE, Hays CC, Zlatar ZZ (2014) Cerebral blood flow measured by arterial spin labeling MRI as a preclinical marker of Alzheimer's disease. J Alzheimers Dis 42 Suppl 4: S411-419. [Crossref]
  149. Bangen KJ, Restom K, Liu TT, Wierenga CE, Jak AJ, et al. (2012) Assessment of Alzheimer's disease risk with functional magnetic resonance imaging: an arterial spin labeling study. J Alzheimers Dis 31 Suppl 3: S59-S74. [Crossref]
  150. Iadecola C (2010) The overlap between neurodegenerative and vascular factors in the pathogenesis of dementia. Acta Neuropathol 120: 287-296. [Crossref]
  151. Vemuri P, Lesnick TG, Przybelski SA, Knopman DS, Preboske GM, et al. (2015) Vascular and amyloid pathologies are independent predictors of cognitive decline in normal elderly. Brain 138: 761-771. [Crossref]
  152. Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Perez JM, Evans AC (2016) Early role of vascular dysregulation on late-onset Alzheimer's disease based on multifactorial data-driven analysis. Nat Commun 7: 11934. [Crossref]
  153. Kalaria RN (2003) Comparison between Alzheimer's disease and vascular dementia: implications for treatment. Neurol Res 25: 661-664. [Crossref]
  154. Jellinger KA, Attems J (2007) Neuropathological evaluation of mixed dementia. J Neurol Sci 257: 80-87. [Crossref]
  155. Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA (2009) The neuropathology of probable Alzheimer disease and mild cognitive impairment. Ann Neurol 66: 200-208. [Crossref]
  156. Bennett DA, Wilson RS, Arvanitakis Z, Boyle PA, de Toledo-Morrell L, et al. (2013) Selected findings from the Religious Orders Study and Rush Memory and Aging Project. J Alzheimers Dis 33 Suppl 1: S397-S403. [Crossref]
  157. Launer LJ, Petrovitch H, Ross GW, Markesbery W, White LR (2008) AD brain pathology: vascular origins? Results from the HAAS autopsy study. Neurobiol Aging 29: 1587-1590. [Crossref]
  158. Fotuhi M, Hachinski V, Whitehouse PJ (2009) Changing perspectives regarding late-life dementia. Nat Rev Neurol 5: 649-658. [Crossref]
  159. Perry G, Taddeo MA, Nunomura A, Zhu X, Zenteno-Savin T, et al. (2002) Comparative biology and pathology of oxidative stress in Alzheimer and other neurodegenerative diseases: beyond damage and response. Comp Biochem Physiol C Toxicol Pharmacol 133: 507-513. [Crossref]
  160. Reynolds MR, Derdeyn CP, Grubb RL, Jr. Powers WJ, Zipfel GJ (2014) Extracranial-intracranial bypass for ischemic cerebrovascular disease: what have we learned from the Carotid Occlusion Surgery Study? Neurosurg Focus 36: E9. [Crossref]
  161. Low SW, Teo K, Lwin S, Yeo LL, Paliwal PR, et al. (2015) Improvement in cerebral hemodynamic parameters and outcomes after superficial temporal artery-middle cerebral artery bypass in patients with severe stenoocclusive disease of the intracranial internal carotid or middle cerebral arteries. J Neurosurg 123: 662-669. [Crossref]
  162. van der Wijngaard IR, Holswilder G, van Walderveen MAA, Algra A, Wermer MJH, et al. (2016) Treatment and Imaging of intracranial atherosclerotic stenosis: current perspectives and future directions. Brain and Behaviour 6: e00536. [Crossref]
  163. Pan W, Kastin AJ (2014) Can sleep apnea cause Alzheimer's disease? Neurosci Biobehav Rev 47: 656-669. [Crossref]
  164. Daulatzai MA (2015) Evidence of neurodegeneration in obstructive sleep apnea: Relationship between obstructive sleep apnea and cognitive dysfunction in the elderly. J Neurosci Res 93: 1778-1794. [Crossref]
  165. Gorelick PB, Scuteri A, Black SE, DeCarli C, Greenberg SM, et al. (2011) Vascular contributions to cognitive impairment and dementia: a statement for healthcare professionals from the american heart association/american stroke association. Stroke 42: 2672-2713. [Crossref]
  166. Moretti R, Torre P, Antonello RM, Manganaro D, Vilotti C, et al. (2008) Risk factors for vascular dementia: hypotension as a key point. Vasc Health Risk Manag 4: 395-402. [Crossref]
  167. Cermakova P, Eriksdotter M, Lund LH, Winblad B, Religa P, et al. (2015) Heart failure and Alzheimer's disease. J Intern Med 277: 406-425. [Crossref]
  168. Nelson L, Gard P, Tabet N (2014) Hypertension and inflammation in Alzheimer's disease: close partners in disease development and progression! J Alzheimers Dis 41: 331-343. [Crossref]
  169. Freiherr J, Hallschmid M, Frey WH, Brunner TF, Chapman CD, et al. (2013) Intranasal insulin as a treatment for Alzheimer's disease: a review of basic research and clinical evidence. CNS Drugs 27: 505-514.  [Crossref]
  170. Benedict C, Hallschmid M, Schultes B, Born J, Kern W (2007) Intranasal insulin to improve memory function in humans. Neuroendocrinology 86: 136-142. [Crossref]
  171. de la Monte SM (2017) Insulin Resistance and Neurodegeneration: Progress Towards the Development of New Therapeutics for Alzheimer's Disease. Drugs 77: 47-65. [Crossref]
  172. Claxton A, Baker LD, Hanson A, Trittschuh EH, Cholerton B, et al. (2015) Long-acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment on early-stage Alzheimer's disease dementia. J Alzheimers Dis 45: 1269-1270. [Crossref]
  173. Mayeda ER, Whitmer RA, Yaffe K (2015) Diabetes and cognition. Clin Geriatr Med 31: 101-115, ix. [Crossref]
  174. Goldwaser EL, Acharya NK, Sarkar A, Godsey G, Nagele RG (2016) Breakdown of the cerebrovasculature anf blood-brain barrier: A mechanistic link between diabetes mellitus and Alzheimer's disease. J Alzheimer’s disease 54: 445-456. [Crossref]
  175. Prince M, Comas-Herrera AA, Knapp M, Guerchet M, Karaglannidou M (2016) World Alzheimer Report. Alzheimer's Disease International, London
  176. Roher AE, Maarouf CL, Kokjohn TA (2016) Familial Presenilin Mutations and Sporadic Alzheimer's Disease Pathology: Is the Assumption of Biochemical Equivalence Justified? J Alzheimers Dis 50: 645-658. [Crossref]
  177. Lyman GH, Moses HL (2016) Biomarker Tests for Molecularly Targeted Therapies-The Key to Unlocking Precision Medicine. N Engl J Med 375: 4-6.
  178. Collins FS, Varmus H (2015) A new initiative on precision medicine. N Engl J Med 372: 793-795. [Crossref]
  179. Jameson JL, Longo DL (2015) Precision medicine--personalized, problematic, and promising. N Engl J Med 372: 2229-2234. [Crossref]
  180. Serrano GE, Intorcia A, Carew J, Chiarolanza G, Hidalgo JA, et al. (2015) Detection of Characteristic Proteinopathies of Neurodegenerative Diseases. J Neuropathol Exp Neurol 74: 934-942.
  181. Bloch J, Brunet JF, McEntire CR, Redmond DE (2014) Primate adult brain cell autotransplantation produces behavioral and biological recovery in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonian St. Kitts monkeys. J Comp Neurol 522: 2729-2740. [Crossref]
  182. Norton S, Matthews FE, Barnes DE, Yaffe K, Brayne C (2014) Potential for primary prevention of Alzheimer's disease: an analysis of population-based data. Lancet Neurol 13: 788-794. [Crossref]
  183. Larson EB, Yaffe K, Langa KM (2013) New insights into the dementia epidemic. N Engl J Med 369: 2275-2277. [Crossref]
  184. Satizabal CL, Beiser AS, Chouraki V, Chene G, Dufouil C, et al. (2016) Incidence of Dementia over Three Decades in the Framingham Heart Study. N Engl J Med 374: 523-532.
  185. Wu YT, Fratiglioni L, Matthews FE, Lobo A, Breteler MM, et al. (2016) Brayne, Dementia in western Europe: epidemiological evidence and implications for policy making. Lancet Neurol 15: 116-124. [Crossref]
  186. Sindi S, Mangialasche F, Kivipelto M (2015) Advances in the prevention of Alzheimer's Disease. F1000Prime Rep 7: 50. [Crossref]
  187. Solomon A, Mangialasche F, Richard E, Andrieu S, Bennett DA, et al. (2014) Advances in the prevention of Alzheimer's disease and dementia. J Intern Med 275: 229-250. [Crossref]
  188. de la Monte SM, Tong M (2014) Brain metabolic dysfunction at the core of Alzheimer's disease. Biochem Pharmacol 88: 548-559. [Crossref]
  189. Cholerton B, Baker LD, Craft S (2013) Insulin, cognition, and dementia. Eur J Pharmacol 719: 170-179. [Crossref]
  190. Deckers K, van Boxtel MP, Schiepers OJ, De VM, Munoz Sanchez JL, et al. (2015) Target risk factors for dementia prevention: a systematic review and Delphi consensus study on the evidence from observational studies. Int J Geriatr Psychiatry 30: 234-246. [Crossref]
  191. Joas E, Bäckman K, Gustafson D, Ostling S, Waern M, et al. (2012) Blood pressure trajectories from midlife to late life in relation to dementia in women followed for 37 years. Hypertension 59: 796-801. [Crossref]
  192. Roher AE (2015) Cardiovascular system participation in Alzheimer's disease pathogenesis. J Intern Med 277: 426-428. [Crossref]
  193. Gutzmann H, Qazi A (2015) Depression associated with dementia. Z Gerontol Geriatr 48: 305-311. [Crossref]
  194. Garcez ML, Falchetti AC, Mina F, Budni J (2015) Alzheimer's Disease associated with Psychiatric Comorbidities. An Acad Bras Cienc 87: 1461-1473. [Crossref]
  195. Bennett S, Thomas AJ (2014) Depression and dementia: cause, consequence or coincidence? Maturitas 79: 184-190. [Crossref]
  196. Kitching D (2015) Depression in dementia. Aust Prescr 38: 209-2011. [Crossref]
  197. Levenson RW, Sturm VE, Haase CM (2014) Emotional and behavioral symptoms in neurodegenerative disease: a model for studying the neural bases of psychopathology. Annu Rev Clin Psychol 10: 581-606. [Crossref]
  198. Khalsa DS (2015) Stress, Meditation, and Alzheimer's Disease Prevention: Where The Evidence Stands. J Alzheimers Dis 48: 1-12. [Crossref]
  199. Paillard T, Rolland Y, de Souto Barreto P (2015) Protective Effects of Physical Exercise in Alzheimer's Disease and Parkinson's Disease: A Narrative Review. J Clin Neurol 11: 212-219. [Crossref]
  200. Baumgart M, Snyder HM, Carrillo MC, Fazio S, Kim H, et al. Summary of the evidence on modifiable risk factors for cognitive decline and dementia: A population-based perspective. Alzheimers Dement 11: 718-726. [Crossref]
  201. Loprinzi PD, Herod SM, Cardinal BJ, Noakes TD (2013) Physical activity and the brain: a review of this dynamic, bi-directional relationship. Brain Res 1539: 95-104. [Crossref]
  202. Brown BM, Peiffer JJ, Martins RN (2013) Multiple effects of physical activity on molecular and cognitive signs of brain aging: can exercise slow neurodegeneration and delay Alzheimer's disease? Mol Psychiatry 18: 864-874. [Crossref]
  203. Henderson VW (2014) Three midlife strategies to prevent cognitive impairment due to Alzheimer's disease. Climacteric 17 Suppl 2:38-46. [Crossref]
  204. Savica R, Petersen RC (2011) Prevention of dementia. Psychiatr Clin North Am 34: 127-145. [Crossref]
  205. Qiu C, Kivipelto M, Von SE (2009) Epidemiology of Alzheimer's disease: occurrence, determinants, and strategies toward intervention. Dialogues Clin Neurosci 11: 111-128. [Crossref]
  206. Middleton LE, Yaffe K (2009) Promising strategies for the prevention of dementia. Arch Neurol 66: 1210-1215. [Crossref]
  207. Meng X, D'Arcy C (2012) Education and dementia in the context of the cognitive reserve hypothesis: a systematic review with meta-analyses and qualitative analyses. PLoS One 7: e38268. [Crossref]
  208. Rizzi L, Rosset I, Roriz-Cruz M (2014) Global epidemiology of dementia: Alzheimer's and vascular types. Biomed Res Int 2014: 908915. [Crossref]
  209. Scarmeas N, Stern Y (2004) Cognitive reserve: implications for diagnosis and prevention of Alzheimer's disease. Curr Neurol Neurosci Rep 4: 374-380. [Crossref]
  210. Stern Y, Alexander GE, Prohovnik I, Stricks L, Link B, et al. (1995) Relationship between lifetime occupation and parietal flow: implications for a reserve against Alzheimer's disease pathology. Neurology 45: 55-60. [Crossref]
  211. Beuscher L, Beck C (2008) A literature review of spirituality in coping with early-stage Alzheimer's disease. J Clin Nurs 17: 88-97. [Crossref]
  212. Beuscher L, Grando VT (2009) Using spirituality to cope with early-stage Alzheimer's disease. West J Nurs Res 31: 583-598. [Crossref]
  213. Newberg AB, Wintering N, Khalsa DS, Roggenkamp H, Waldman MR (2012) Meditation effects on cognitive function and cerebral blood flow in subjects with memory loss: a preliminary study. Ann Neurosci 2012; 19: 81. [Crossref]
  214. Kaufman Y, Anaki D, Binns M, Freedman M (2007) Cognitive decline in Alzheimer disease: Impact of spirituality, religiosity, and QOL. Neurology 68: 1509-1514. [Crossref]
  215. Post SG, Whitehouse PJ (1999) Spirituality, religion, and Alzheimer's disease. J Health Care Chaplain 8: 45-57. [Crossref]
  216. Barrett-Connor E (2007) An introduction to obesity and dementia. Curr Alzheimer Res 4: 97-101. [Crossref]
  217. Hildreth KL, Van Pelt RE, Schwartz RS (2012) Obesity, insulin resistance, and Alzheimer's disease. Obesity (Silver Spring) 20: 1549-1557. [Crossref]
  218. Ashrafian H, Harling L, Darzi A, Athanasiou T (2013) Neurodegenerative disease and obesity: what is the role of weight loss and bariatric interventions? Metab Brain Dis 28: 341-353. [Crossref]
  219. Stein J, Schettler T, Rohrer B, Valenti M (2008) Environmental Factors in the Development of Dementia: Focus on Alzheimer's Disease and Cognitive Decline. In Environmental Threats to Healthy Aging: With a Closer Look at Alzheimer's & Parkinson's Diseases, Myers N, Eds. Greater Boston Physicians for Social Responsibility and Science and Environmental Health Network, Boston, pp. 97-144
  220. Martin GM, Fukuchi KI (1992) Pathogenetic mechanisms in dementias of the Alzheimer's type. Current Science 63: 410-416. 

Editorial Information

Editor-in-Chief

Gary W. Jay
University of North Carolina

Article Type

Research Article

Publication history

Received date: July 15, 2017
Accepted date: August 09, 2017
Published date: August 12, 2017

Copyright

© 2017 Kokjohn TA. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Kokjohn TA, Serrano GE, Roher AE (2017) Alzheimer’s disease syndrome – Recognizing the complexity of dementia. Neurol Disord Therap 1: DOI: 10.15761/NDT.1000116

Corresponding author

Alex E. Roher

Division of Clinical Education, Midwestern University Glendale AZ 85308 And Division of Neurobiology, Barrow Neurological Institute, Phoenix AZ 85013, USA.

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

No Figures.