Int J Pharm Pharm Sci, Vol 8, Issue 2, 8-15Review Article


RECENT ADVANCES IN ALZHEIMER'S DISEASE: CAUSES AND TREATMENT

KIRANJIT KAUR*, RAJNEET KAUR, MANJINDER KAUR

G. H. G. Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana (India) 141104
Email: rajneet.manchanda@gmail.com

 Received: 27 Aug 2015 Revised and Accepted: 19 Dec 2015


ABSTRACT

Alzheimer’s disease (AD) is a destructive neurodegenerative disorder characterized by progressive memory defeat and impairment in behavior, language, and visuospatial skills. Neuropsychiatric symptoms such as apathy, depression, aggression, agitation, sleep disruption, and psychosis are now recognized as core symptoms of AD that are expressed to varying degrees throughout the course of disease. The neuro pathological features of AD comprise extracellular senile plaques constituted of β-amyloid (Aβ) pledges, intracellular neurofibrillary tangles (NFTs), and cerebral atrophy; others include apolipoprotein E, oxidative stress, mitochondrial dysfunction and cholinergic hypothesis. Anti-amyloid therapy is available for the treatment of Alzheimer’s disease, others are anticholinergic therapy, and therapy for mitochondrial dysfunction, γ-secretase inhibitors (GSI) and modulators (GSM), 𝛽-secretase (BACE1) inhibitors, Glial modulating drugs includes RAGE receptor antagonists, TNF-α antagonists, neuroprotective drugs such as antioxidants, phosphodiesterase inhibitors, PPARγ agonists, and anti-tau or tau modulators like microtubule stabilizers, kinase inhibitors. This review includes discussion on neurobiological mechanisms and newly developed compounds which have lesser side effects and are proving more efficient for treatment of Alzheimer’s disease.

Keywords: Alzheimer'sdisease, Alzheimer causes and treatment


INTRODUCTION

Alzheimer’s disease (AD) is a critical neurodegenerative illness characterized by memory loss and diminished performance, language, and visuospatial skills [1]. Epidemiological data show that the occurrence of AD increases with age and doubles every 5 y after 65 y of age [2, 3]. There were about 26.6 million cases of AD in the world in 2006 and it is predictable that the worldwide dominance of AD will grow fourfold to 106.8 million by the year 2050 [4]. The neuropathological features of AD involve extracellular senile plaques constituted of β-amyloid (Aβ) pledges, intracellular neurofibrillary tangles (NFTs), and cerebral atrophy [5]. Aβ, produced from the proteolytic administration of amyloid precursor protein (APP), has been projected to have a causative role in the generation of AD and this Accumulated Amyloid-β can provoke euro-toxicity by generating oxidative stress and inflammation in the brain [6, 7].

Causative factors

Both non-modifiable and modifiable risk factors are involved in AD. Non-modifiable factors include age which is one of the important risk factors, genes, family history, Down’s syndrome while modifiable factors include cognitive engagement, diet/nutritional supplement intake, physical activity level, type 2 diabetes, alcohol consumption, mood disorders, hypertension, hypercholesterolemia, and smoking [8-11].

Genetic factors

Alzheimer’s disease happens in both familial and an irregular form, also recognized as early onset or late onset Alzheimer's disease (LOAD), respectively [12]. Early-onset domestic AD is generally produced by autosomal dominant variations in the genes for amyloid precursor protein (APP), presenilin 1 and presenilin 2. This form of AD accounts for around 2–5% of all AD cases [13]. The apoE4 allele is the single verified heritable factor in the progress of together the early and late-onset practices of AD. This factor rises vulnerability to AD however it is neither essential nor adequate for the progress of this disease. In late-onset irregular form, greater the number of the apoE4 alleles, the greater the possibility of AD and the lesser the age of onset. The risk influence of the occurrence of the apoE4 allele declines with age. Usually, around 15-20% of AD cases could be recognized to this risk [14].

Vascular factors

Smoking

Smoking is a well-recognised cardiac risk factor and facilitates oxidative stress, inflammation and atherosclerosis which are identified as the risk factors for neurodegeneration [15]. Oxidative stress promotes augmented β-secretase cleavage of APP and abnormal tau phosphorylation. It may openly enable the amyloidogenic pathway involved in Aβ oligomer invention and extracellular fibrillar Aβ aggregation, as well as abnormal tau phosphorylation [16].

Alcohol

It is well predictable that alcohol abuse causes alcohol dementia. In heavy consumers, alcohol causes injuries in the brain. Light to adequate alcohol intake is found to be closely associated with brain atrophy and volume loss [17, 18].

Obesity

It has been showed that being obese contributes to dementia and cerebral decay. Obesity leads to dementia, cerebral damage and consistent neurological modifications [19].

Blood pressure and management of hypertension

High blood pressure is one of the most significant manageable risk factors for stroke, which in turn can result in vascular dementia. It has been reported that there is a close relationship between AD and hypertension [20]. It has been recently proved that antihypertensive drugs have a protective effect against the progress of dementia and AD [21-23]. The antihypertensive remedy may protect against dementia and AD by decaying the atherosclerotic process, reducing the number of atherosclerotic scratches and improving cerebral perfusion [24].

Diabetes mellitus

It has been reported that there is an increased risk of vascular dementia in persons with diabetes [25-27]. Binding of insulin or IGF-1 causes a conformational variation of the receptor leading to their autophosphorylation on definite tyrosine residues on the β-subunit resulting in activation of the insulin receptor substrate-1 (IRS-1) [28, 29]. The latter, in turn, stimulates two main signaling pathways: initially the PI3K pathway, which is involved in the preservation of synaptic plasticity and memory consolidation [30], Aβ-induced memory loss [31], and synthesis of nitric oxide (NO),which in turn plays a role in learning and memory practices[32]; and second pathway contains the MAPK cascade, which is accountable both for the induction of several genes essential for neuronal and synapse development, conservation and healing processes, as well as serving as a modulator of hippocampal synaptic plasticity that motivates learning and memory [33].

Hypercholesterolemia and statin therapy

High total serum cholesterol levels in medium age are involved to be a risk factor for the progress of AD at a later age [34, 35]. High total cholesterol in medium age is a risk factor for the progress of AD and additional dementias 20 y later but declining serum cholesterol levels in late medium age may be due to continuing disease progress and may characterize a sign for later AD and other dementias [36]. Statins have been reported to decline the production of β-amyloid. Statins also have several other effects that may be valuable for the CNS and thus, may smaller the risk of AD [37, 38].

Nutritional factors

Consumption of antioxidants such as vitamin E and vitamin C decreases the risk of developing AD. It has been reported that diet rich in saturated fats and cholesterol increases the risk of AD [39-41]. Antioxidants in turn decrease the oxidative stress and amyloid beta-peptide (A𝛽) accumulation [42, 43]. Antioxidants, vitamins, polyphenol, polyunsaturated fatty acids, fish, fruits, vegetables, tea, and light to moderate intake of alcohol are useful for AD, while trans-fatty acids, saturated fatty acids, carbohydrates, and whole-fat dairy are harmful to AD[44].

Psychosocial factors

It was found that psychosocial factors and a vigorous lifestyle during life may decline the risk of dementias including AD [45].

Education and socioeconomic status

The risk for dementia is minor for those with higher education, occupational achievement, intelligence or IQ and cerebrally exciting leisure events [46]. Lower education is tied to enlarged risk of dementia and AD [47, 48]. It has been reported that a poor social network or a lack of social commitment are related with reduced cognitive functions and dementia [49]. The risk of dementia is also higher in elderly individuals with increased social segregation and less numerous and inadequate contacts with families and friends. Individuals with low neuroticism joined with high extraversion had a lower risk of dementia [50]. Low levels of social commitment in late life and declined social commitment from medium to late life are associated with a two-fold increase in the risk of the progress of dementia and AD later in life [51, 52].

Physical and mental activity

It has been reported that midlife physical activity may be linked with a reduced risk of AD or vascular dementia in later life [53]. Physical activity is assumed to enrich cognitive function by increasing cardiac suitability and cerebral perfusion and probably by stimulating neurogenesis [54]. Numerous activities necessitating mental exertion such as reading, social and traditional activities, indicate a defensive effect against dementia and AD [55, 56]. It has been proved recently that complex cerebral activity across lifecycle leads to reduced hippocampal atrophy [57].

Pathophysiology of AD

Two of the symbol neuro pathologic outcomes in AD are extracellular amyloid and neuritic plaques and intracellular neurofibrillary tangles.

Amyloid-β and tau proteins

According to the amyloid theory, A𝛽 peptide massing in patient’s brain is the crucial event leading to the progress of AD. It has been reported in the in-vitro studies that the incubation of the Aβ peptide with cells in culture persuades a neurotoxic consequence characterized by oxidative stress, apoptosis, and injury to membrane and cytoplasmic proteins, mitochondrial DNA, and lipids [58, 59]. Amyloid plaques are mainly formed by accretion of insoluble Aβ peptides, while neurotic plaques are composed of insoluble Aβ peptides in deteriorated neurites (dendrites, axons, or telodendria), and some comprise hyper phosphorylated tau proteins (p-tau) [60].

In AD, the tau protein undertakes oligomerization and forms paired helical filaments (PHFs), which then leads to the progress of NFT [61]. It has been reported that tau persuades mitochondrial dysfunction, leading to severe energy dysfunction and the generation of ROS and reactive nitrogen species (RNS) [62], disturb the integrity of biological membranes and leads to synaptic failure [63], which is characterized by synaptic degradation, and neuronal loss [64, 65].

Apolipoprotein E

The Apolipoprotein E (Apo E) is a significant protein involved in preserving the structural and functional integrity of synapses and membranes [66]. There are three iso forms: ApoE2 (Cys112, Cys158), ApoE3 (Cys112, Arg158), and ApoE4 (Arg112, Arg158) [67]. The E4 allele confers an augmented risk; however the e2 allele decreases the risk. ApoE4 may increase the intracellular reprocessing of APP, which could raise A𝛽 production [68]. Modifications in complement signaling pathways may affect microglia function resulting in reduced capability to phagocytize apoptotic cells and clear beta-amyloid [69].

Fig. 1: Effect of oxidative stress in lipid peroxidation and protein modification in AD [82]

Role of oxidative stress in AD

Oxidative stress results due to an imbalance between the production and quenching of free radicals from oxygen species [70]. Enhanced ROS assembly or decreased antioxidant system leads to oxidative imbalance and cause ROS overproduction [71-73]. The process of aging is also related with augmented oxidative stress [74]. It has been reported that oxidative stress plays a vital role in the pathogenesis of AD leading to neuronal dysfunction and cell loss [75]. As lipid peroxidation is significantly augmented in AD. Lipid peroxidation is the process in which lipids are attacked by ROS through a free radical chain reaction mechanism to produce lipid peroxidation products. It has been reported that the 4-hydroxynonal levels are considerably raised in the hippocampus, entorhinal cortex, temporal cortex, amygdala, parahippocampal gyrus in AD. Oxidative amendments in proteins occur which are the outcome of direct ROS attack or from reaction with glycation, glycoxidation, and lipid peroxidation product binding [76, 77]. Oxidative impairment of DNA/RNA has been reported in AD. Oxidative impairment of DNA can cause DNA double strand disruptions, DNA/DNA or DNA/protein crosslinking, and base alteration (see in figure1). It has been reported that high levels of DNA breakdowns were established in the hippocampus and cerebral cortex in AD [78,79]. It has been reported that an increase of oxidative biomolecule products and reduction in antioxidant levels or antioxidant enzyme action leads to AD [80]. The decreased plasma levels of antioxidants such as albumin, bilirubin, uric acid, lycopene, vitamin A, vitamin C, and vitamin E has been reported in AD patients [81].

Mitochondrial dysfunction

Mitochondria are responsible for ATP production, signaling pathway and cell activities. They are also involved in apoptosis and in dynamic movements essential for precise respiratory activity and metabolic efficiency through fusion/fission [83]. Dysfunctional mitochondria are less effective producers of ATP but more effective producers of ROS, signifying the main source of oxidative imbalance as detected in AD [84, 85]. It has been reported that mitochondrial dysfunction is a prominent and primary feature of AD [86]. Furthermore, mitochondrial dysfunction results in diminished features of mitochondrial function in AD [87]. Dysfunctional mitochondria cause high levels of ROS that may be noxious for neurons [88]. Moreover, ROS treats mitochondria as a target triggering oxidation of its constituents such as mtDNA, lipids, and proteins which finally results in mitochondrial worsening [89]. Declined calcium uptake and increased calcium burden leads to calcium deregulation and enlarged intracellular calcium in the brain [90-92]. Furthermore, mitochondrial dysfunction has been reported to modify the levels of numerous enzymes which contain pyruvate dehydrogenase and 𝛼-ketoglutarate dehydrogenase ATP-citrate lyase, and acetoacetyl-CoA thiolase. Reduced levels of these enzymes results in diminish production of acetyl-coA which causes deficit cholinergic expressions in AD patients [93-100]. ROS action on mitochondria generate toxic products like hydroxynonenal which facilitate the self-assembly of tau proteins into helical filaments which are reported to be present in AD [101].

Cholinergic hypothesis

There is a prominent and inconsistent insufficiency of acetylcholine. The functional basis of the cholinergic shortage is the atrophy and worsening of subcortical cholinergic neurons, mainly those in the basal forebrain (nucleus basalis of Meynert), that provide cholinergic innervation to the entire cerebral cortex. The selective insufficiency of acetylcholine in AD, results in dementia in AD, has given rise to “cholinergic hypothesis,” which recommends that a lack of acetylcholine is critical in the origin of the signs of AD. In AD, there is not only damage of cholinergic neurons but also the cortical and hippocampus areas that receive cholinergic input [102].

Signs and symptoms of AD

Neuropsychiatric symptoms such as apathy, depression, violence, anxiety, sleep interruption, and psychosis are now recognized as the main symptoms of AD that are expressed to variable degrees during the progression of the disease [103] and finally results in Alzheimer’s dementia.

Diagnosis of AD

The occurrence and distribution of amyloid plaques and NFT in the brain is used to found the finding of ‘definitive’ AD and stage the illness [104]. In clinical settings, the diagnosis of AD, is mainly based on medical history, somatic and neural examinations, and neuropsychological evaluation [105]. Patients with minor cognitive damage should be monitored for cognitive and functional decay because of their increased risk for dementia. The NINCDS-ADRDA conditions for the diagnosis of possible AD or the DSM-III-R conditions for dementia of the Alzheimer type should regularly be used. Structural neuroimaging and magnetic resonance imaging are suitable in the initial evaluation of patients with dementia. [106-108].

Recent advances in treatment of AD

Anti-amyloid therapy

Anti-amyloid therapy involves the uses of drugs (see in table 1) with a different mechanism of actions: (i) enhance the clearance of Aβ; (ii) Prevent the production of Aβ; or (iii) Inhibit the accumulation of Aβ [109]. Active and passive immunization results in decreased levels of intracerebral Aβ burden by inducing humoral reaction against the Aβ peptide leading to its clearance from the brain [110].

Table 1: Newer compounds targeted to anti-beta-amyloid treatment [111-120]

Compound

Target/Treatment

Current phase

ANI-1792

Vaccine-active immunization

Interrupted at phase I (severe side effects such as meningoencephalitis)

CAD-106

Vaccine-active immunization

Phase I (ongoing)

Bapineuzumab

Beta-amyloid monoclonal antibody

Phase III (ongoing)

Solanezumab

Beta-amyloid monoclonal antibody

Phase III (ongoing)

Ponezumab

Beta-amyloid monoclonal antibody

Interrupted at phase II (no efficacy)

Gantenerzumab

Beta-amyloid monoclonal antibody

Phase I (ongoing)

Crenezumab

Beta-amyloid monoclonal antibody

Phase I (ongoing)

Semagacestat

Gamma-secretase inhibitor

Interrupted at phase III (no efficacy and risk for skin cancer)

Avagacestat

Gamma-secretase inhibitor

Phase II (ongoing)

GRL-834

Beta-secretase inhibitor

Ongoing

TAK-070

Beta-secretase inhibitor

Ongoing

CHF-5074

Non-steroid anti-inflammatory agent

Ongoing

DAPT

Prototypal Gamma-secretase inhibitor

Ongoing

Curcumin

Anti-amyloid aggregator

Ongoing


𝛽-Secretase (BACE1) inhibitor

Beta-site APP-cleaving enzyme 1 (BACE1) is a protease responsible for cleavage of APP, resulting in generation of assembly of neurotoxic irregular A𝛽[121,122]. Nuclear peroxisome proliferator activated receptor gamma (PPAR𝛾) functions as a transcription factor which regulates gene expression [123], promotes microglia-mediated A𝛽endocytosis. Also it reduces inflammation response and causes decreased cytokine excretion [124]. Thiazolidinedione can induce PPAR𝛾 to inhibit 𝛽-secretase and stimulate ubiquitination to worsen amyloid burden [125]. It has been also reported that PPAR𝛾 agonist i.e. thiazolidinedione derivatives like rosiglitazone and pioglitazone worsens AD neuropathology by reducing insulin sensitivity which helps in A𝛽proteolysis [126].

γ-Secretase inhibitors (GSI) and modulators (GSM)

γ-secretase is a transmembrane protease responsible for cleavage of amyloid precursor protein (APP) to produce Aβ [127,128]. Different GSIs such as DAPT, L685458 andMRK-560 131have been recently developed [129, 130]. While different (GSM) such as avagacestat (BMS-708163), begacestat and NIC5-15 are under clinical trials [131].

Kinase inhibitors

The first class of tau inhibitors which helps in targeting tau phosphorylation and reduces tau phosphorylation by decreasing the activity of kinase enzyme. Interaction between glycogen synthase kinase 3 beta (GSK3𝛽) and protein phosphate 2 (PP2A) augments tau hyper phosphorylation and NFT generation [132]. Lithium, valproate, NP-031112 (NP-12) and epothilone D (BMS-241027) decreases tau phosphorylation and prevent reversed features of tauopathy [133-136].

Therapy for mitochondrial dysfunction

Latrepirdine (DIMEBON), an antihistamine which preserves mitochondrial structure and function and protects against A𝛽 induced apoptosis is under investigation [137,138]. Its combination with donepezil is also under investigation [139]. AC-1204 is considered to improve mitochondrial metabolism by inducing chronic ketosis, thereby releasing regional cerebral hypometabolism presented in early Alzheimer’s disease, and this agent is also under investigation [140].

Anticholinergic therapy

Anticholinergic therapy includes administration of cholinesterase inhibitors to treat the cholinergic deficit associated with AD. The drugs include tacrine (COGNEXS), donepezil (ARICEPTS), rivastigmine (EXELON), and galantamine (REMINYLS) [141].

Table 2: Overall pharmacologic treatments other than anti-amyloid therapy under research for Alzheimer’s disease [142-144]

Pharmacologic treatment

Agent under research

Neurotransmitter-based

 

Acetylcholine

ST 101, AF 267B, ABT 089, AZD 3480, MEM 3454, EVP-6124, Posiphen, Huperzine

Serotonin

5-HT4 partial agonists, 5-HT1A agonists/antagonists, 5-HT6 antagonists

Norepinephrine/Dopamine:

MAO A and MAO B inhibitors

GABA:

GABA-B antagonists

Glutamate:

AMPA potentiator

Glycine:

Partial agonists

Glial modulating drugs:

 

Direct glial target:

G and GM CSF, Nitro flurbiprofen, ONO-2506, Tacrolimus

RAGE receptor antagonist:

TTP 488

TNF-α antagonist:

Enbrel

Neuroprotection:

 

Antioxidants:

Vitamin C and E, alpha lipoic acid, CoQ10

Miscellaneous:

PDE inhibitors, PPARγ agonists and insulin, SIRT1 activators, Growth factors (BDNF and NGF), Dimebon

Anti-tau or tau modulators:

 

Microtubule stabilizers:

NAP (AL-108) and Methylene blue (Rember)

Kinase inhibitors (GSK-3α, GSK-3β, CDK 5)

Lithium, AZD-1080, Minocycline

 

PDE-4 inhibitors, immunotherapies

[GABA, gamma-amino butyric acid; RAGE, receptor for advanced glycation end products; TNF, tumor necrosis factor; GSK, glycogen synthase kinase; CDK, cyclic-dependent kinase; 5-HT (4, 1A and 6), 5-hydroxytryptamine (receptor subtypes); MAO (A and B), monoamine oxidase (A and B subtypes); AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; PPAR, peroxisome proliferator-activated receptors; SIRT1, sirtuin (silent mating type information regulation 2 homolog)-1; BDNF, brain-derived neurotrophic factor; NGF, nerve growth factor; NAP, neuronal microtubule-interacting agent (a peptide of eight amino acids, NAPVSIPQ); PDE, phosphodiesterase]


CONCLUSION

Herein, we have made an effort to review recent trends in AD. The molecular heredities of AD and the role of key proteins, oxidative damage, mitochondrial dysfunction and the cholinergic hypothesis that are assumed to contribute in AD pathogenesis are significant fields for advanced research. There is significant active investigation ongoing in the development of new inhibitors for BACE, kinase, and γ-secretase as targets for treatment of AD. Thus, it is hoped that all these lines of ongoing research, combined, should lead to a deeper understanding of the progressions that happen in the AD brain to permit us to preclude efficiently their incidence. Thus, we conclude that these categories of drugs discussed in this review can be potentially targeted for research and development for the treatment of AD.

ACKNOWLEDGEMENT

The authors are grateful to Department of Pharmaceutical Sciences, G. H. G Khalsa College of Pharmacy, Gurusar Sadhar, and Ludhiana for providing funding and other technical facilities for work.

ABBREVIATION

AD= Alzheimer disease, NFTs= Neurofibrillary tangles, GSI= Gamma secretase inhibitors, GSM= Gamma secretase modulators, BACE1=Beta-site APP-cleaving Enzyme 1, RAGE=Receptors for Advanced Glycation End products, PPARγ=Peroxisome Proliferator-activated Receptor γ, DSM-III-R = Diagnostic and Statistical Manual, 3rd edition, revised; NINCDS-ADRDA = National Institute of Neurologic, Communicative Disorders and Stroke–Alzheimer’s Disease and Related Disorders Association.

CONFLICT OF INTERESTS

Declared none

REFERENCES

  1. Selkoe DJ. Alzheimer's disease: genes, proteins, and therapy. Physiol Rev 2001;81:741-66.
  2. Querfurth HW, LaFerla FM. Alzheimer's disease. N Engl J Med 2010;362:329-44.
  3. Mayeux R. Clinical practice. early Alzheimer's disease. N Engl J Med 2010;362:2194-201.
  4. Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimer’s Dementia 2007;3:186-91.
  5. Findeis MA. The role of amyloid beta peptide-42 in Alzheimer's disease. Pharmacol Ther 2007;116:266-86.
  6. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 2002;297:353-6.
  7. Pimplikar SW. Reassessing the amyloid cascade hypothesis of Alzheimer's disease. Int J Biochem Cell Biol 2009;41:1261-8.
  8. Daviglus ML, Plassman BL, Pirzada A, Bell CC, Bowen PE, Burke JR, et al. Risk factors and preventive interventions for Alzheimer disease: state of the science. Arch Neurol 2011;68:1185-90.
  9. Barnes DE, Yaffe K. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol 2011;10:819–28.
  10. Ersche KD, Barnes A, Jones PS, Morein-Zamir S, Robbins TW, Bullmore ET. Abnormal structure of front striatal brain systems is associated with aspects of impulsivity and compulsivity in cocaine dependence. Brain 2011;134:2013–24.
  11. Etgen T, Sander D, Bickel H, Forstl H. Mild cognitive impairment and dementia: the importance of modifiable risk factors. Deutsches Ärzteblatt 2011;108:743–50.
  12. Bateman RJ, Aisen PS, De Strooper B, Fox NC, Lemere CA. Autosomal-dominant Alzheimer’s disease: a review and proposal for the prevention of Alzheimer’s disease. Alzheimer's Res Ther 2011;3:1.
  13. Blennow K, de Leon MJ, Zetterberg H. Alzheimer’s disease. Lancet 2006;368:387-403.
  14. Qiu C, Kivipelto M, Agüero-Torres H, Winblad B, Fratiglioni L. Risk and protective effects of the APOE gene towards Alzheimer’s disease in the Kungsholmen project: variation by age and sex. J Neurol Neurosurg Psychiatry 2004;75:828-33.
  15. Swan, Lee Y. Systematic review of behavioral health risks and cognitive health in older adults. Int Psychogeriatrics 2010;20:174.
  16. Giunta B, Deng J, Jin J, Sadic E, Rum S, Zhou H, et al. Evaluation of how cigarette smoke is a direct risk factor for Alzheimer’s disease. Technol Innovation 2012;14:39–48.
  17. Ding J, Eigenbrodt ML, Hutchinson RG, Folsom AR, Harris TB, Nieto FJ, et al. Alcohol intake and cerebral abnormalities on magnetic resonance imaging in a community-based population of middle-aged adults: the atherosclerosis risk in communities (ARIC) study. Stroke 2004;35:16-21.
  18. Paul CA, Au R, Fredman L, Massaro JM, Seshadri S, Decarli C, et al. Association of alcohol consumption with brain volume in the framingham study. Arch Neurol 2008;65:1363-7.
  19. Cognitive Function and Decline in Obesity in Press; 2012.
  20. Povova J, Pastucha D, Sery O, Matejkova M, andJanout V. Is cardiovascular disease a risk factor for Alzheimer’s disease? Exp Clin Cardiol 2014;20:1-6.
  21. Qiu C, Winblad B, Fratiglioni L. The age-dependent relation of blood pressure to cognitive function and dementia. Lancet Neurol 2005;4:487-99.
  22. Yasar S, Corrada M, Broo kmeyer R, Kawas C. Calcium-channel blockers and risk of AD: the baltimore longitudinal study of aging. Neurobiol Aging 2005;26:157-63.
  23. Khachaturian AS, Zandi PP, Lyketsos CG, Hayden KM, Skoog I, Norton MC, et al. Antihypertensive medication use and incident Alzheimer disease: the cache county study. Arch Neurol 2006;63:686-92.
  24. Beishon LC, Harrison JK, Harwood RH, Robinson TG, Gladman JR, Conroy SP. The evidence for treating hypertension in older people with dementia: a systematic review. J Hum Hypertens 2014;28:283-7.
  25. Arvanitakis Z, Wilson RS, Bienias JL, Evans DA, Bennett DA. Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. Arch Neurol 2004;61:661-6.
  26. Akomolafe A, Beiser A, Meigs JB, Au R, Green RC, Farrer LA, et al. Diabetes mellitus and risk of developing Alzheimer disease: results from the framingham study. Arch Neurol 2006;63:1551-5.
  27. Irie F, Fitzpatrick AL, Lopez OL, Kuller LH, Peila R, Newman AB, et al. Enhanced risk for Alzheimer disease in persons with type 2 diabetes and APOE e4: the cardiovascular health study cognition study. Arch Neurol 2008;65:89-93.
  28. Freude S, Schilbach K, Schubert M. The role of IGF-1 receptor and insulin receptor signaling for the pathogenesis of Alzheimer's disease: from model organisms to human disease. Curr Alzheimer Res 2009;6:213–23.
  29. Plum L, Schubert M, Bruning JC. The role of insulin receptor signaling in the brain. Trends Endocrinol Metab 2005;16:59–65.
  30. Horwood JM, Dufour F, Laroche S, Davis S. Signaling mechanisms mediated by the phosphoinositide 3-kinase/Akt cascade in synaptic plasticity and memory in the rat. Eur J Neurosci 2006;23;3375-84.
  31. Chiang HC, Wang L, Xie Z, Yau A, Zhong Y. PI3 kinase signaling is involved in Abeta-induced memory loss in Drosophila. Proc Natl Acad Sci USA 2010;107:7060-5.
  32. Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield DA, Stella AM. Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat Rev Neurosci 2007;8:766-75.
  33. Akter K, Lanza EA, Martin SA, Myronyuk N, Rua M, Raffa RB. Diabetes mellitus and Alzheimer's disease: shared pathology and treatment. Br J Clin Pharmacol 2011;71:365-76.
  34. Karen R, Craig WR, Kristine Y, Ingmar S, Nikolaos S. Is late-onset Alzheimer’s disease really a disease of midlife? Alzheimer’s Dementia 2015;1:122-30.
  35. Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology 2005;64:277-81.
  36. Solomon A, Kareholt I, Ngandu T, Winblad B, Tuomilehto J, Soininen H, et al. Serum cholesterol changes after midlife and late-life cognition: twenty-one-year follow-up study. Neurology 2007;68:751-6.
  37. Rea TD, Breitner JC, Psaty BM, Fitzpatrick AL, Lopez OL, Newman AB, et al. Statin use and the risk of incident dementia: the cardiovascular health study. Arch Neurol 2005;62:1047-51.
  38. Zandi PP, Sparks DL, Khachaturian AS, Khachaturian AS, Tschanz J, Norton M, et al. Do statins reduce the risk of incident dementia and Alzheimer disease? The cache county study. Arch Gen Psychiatry 2005;62:217-24.
  39. Zandi PP, Anthony JC, Khachaturian AS, Stone SV, Gustafson D, Tschanz JT, et al. Reduced risk of Alzheimer disease in users of antioxidant vitamin supplements: the cache county study. Arch Neurol 2004;61:82-8.
  40. Barberger-Gateau P, Raffaitin C, Letenneur L, Berr C, Tzourio C, Dartigues JF, et al. Dietary patterns and risk of dementia: the three-city cohort study. Neurology 2007;69:1921-30.
  41. Laitinen MH, Ngandu T, Rovio S, Helkala EL, Uusitalo U, Viitanen M, et al. Fat intake at midlife and risk of dementia and Alzheimer’s disease: a population based study. Dementia Geriatr Cognit Disord 2006;22:99-107.
  42. Clark TA, Lee HP, Rolston RK. Oxidative stress and its implications for future treatments and management of Alzheimer disease. Int J Biomed Sci 2010;6:225-7.
  43. Ono K, Yamada M. Vitamin A and Alzheimer’s disease. Geriatrics Gerontol Int 2012;12:180-8.
  44. Lawlor DA, Smith GD, Kundu D, Bruckdorfer KR, Ebrahim S. Those confounded vitamins: what can we learn from the differences between observational versus randomized trial evidence? Lancet 2004;363:1724-7.
  45. Fratiglioni L, Paillard-Borg S, Winblad B. An active and socially integrated lifestyle in late life might protect against dementia. Lancet Neurol 2004;3:343-53.
  46. Barnes DE, Yaffe K. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol 2011;10:819-28.
  47. Qiu C, Bäckman L, Winblad B, Agüero-Torres H, Fratiglioni L. The influence of education on clinically diagnosed dementia: incidence and mortality data from the kungsholmen project. Arch Neurol 2001;58:2034-9.
  48. Ngandu T, Von Strauss E, Helkala EL, Winblad B, Nissinen A, Tuomilehto J, et al. Education and dementia: what lies behind the association? Neurology 2007;69:1442-50.
  49. Fratiglioni L, Wang HX, Ericsson K, Maytan M, Winblad B. Influence of social network on occurrence of dementia: a community-based longitudinal study. Lancet 2000;355:1315-9.
  50. Wang HX, Karp A, Herlitz A, Crowe M, Kareholt I, Winblad B, et al. Personality and lifestyle in relation to dementia incidence. Neurology 2009;72:253-9.
  51. Wang HX, Karp A, Winblad B, Fratiglioni L. Late-life engagement in social and leisure activities is associated with a decreased risk of dementia: a longitudinal study from the Kungsholmen project. Am J Epidemiol 2002;155:1081-7.
  52. Serrani D. Narcissism vulnerability as a risk factor for Alzheimer´s disease-a prospective study. Austin J Clin Neurol 2015;2:1057.
  53. Saczynski JS, Pfeifer LA, Masaki K, Korf ES, Laurin D, White L, et al. The effect of social engagement on incident dementia: the honolulu-asia aging study. Am J Epidemiol 2006;163:433-40.
  54. Lee Y. Systematic review of health behavioral risks and cognitive health in older adults. Int Psychogeriatrics 2010;22:174.
  55. Crowe M, Andel R, Pedersen NL, Johansson B, Gatz M. Does participation in leisure activities lead to reduced risk of Alzheimer’s disease? A prospective study of swedish twins. J Gerontol B Psychol Sci Soc Sci 2003;58:249-55.
  56. Biessels GJ, Staekenborg S, Brunner E, Brayne C, Scheltens P. Risk of dementia in diabetes mellitus: a systematic review. Lancet Neurol 2006;5:64-74.
  57. Valenzuela MJ, Sachdev P, Wen W, Chen X, Brodaty H. Lifespan mental activity predicts diminished rate of hippocampal atrophy. PLoS One 2008;3:2598.
  58. Reddy PH, Beal MF. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 2008;14:45–53.
  59. Butterfield DA, Boyd-Kimball D. The critical role of methionine 35 in Alzheimer’s amyloid 𝛽-peptide (1-42)-induced oxidative stress and neurotoxicity. Biochim Biophys Acta 2005;1703:149-56.
  60. Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, Cairns NJ, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol 2012;71:362–81.
  61. Karen C, Edward HK. Emerging therapeutics for Alzheimer’s disease in Insel, Amara, Blaschkeed. Ann Rev Pharmacol Toxicol 2014;54:381-406.
  62. Rapoport SI. Coupled reductions in brain oxidative phosphorylation and synaptic function can be quantified and staged in the course of Alzheimer disease. Neurotoxic Res 2003;5:385-97.
  63. Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Clos AL, Jackson GR, Kayed R. Tau oligomers impair memory and induce synaptic and mitochondrial dysfunction in wild-type mice. Mol Neurodegener 2011;6:39.
  64. Hyman BT. Amyloid-dependent and amyloid-independent stages of Alzheimer disease. Arch Neurol 2011;68:1062-4.
  65. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002;297:353-6.
  66. Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science 1988;240:622-30.
  67. Hatters DM, Peters-Libeu CA, Weisgraber KH. Apolipoprotein E structure: insights into function. Trends Biochem Sci 2006;31:445-54.
  68. Ye S, Huang Y, Mullendorff K, Dong L, Giedt G, Meng EC, et al. Apolipoprotein (apo) E4 enhance amyloid-β peptide production in cultured neuronal cells: ApoE structure as a potential therapeutic target. Proc Natl Acad Sci USA 2005;102:18700-5.
  69. Crehan H, Hardy J, Pocock J. Microglia, Alzheimer’s disease, and complement. Int J Alzheimer’s Disase 2012:9:836-40.
  70. Enns GM, Kinsman SL, Perlman SL, Spicer KM, Abdenur JE, et al. Initial experience in the treatment of inherited mitochondrial disease with EPI-743. Mol Genet Metab 2012;105:91-102.
  71. Lovell MA, Ehmann WD, Butler SM, Markesbery WR. Elevated thiobarbituric acid reactive substances and antioxidant enzyme activity in the brain in Alzheimer's disease. Neurology 1995;45:1594-601.
  72. Markesbery WR, Lovell MA. Four-hydroxy nonanal, a product of lipid peroxidation, is increased in the brain in Alzheimer's disease. Neurobiol Aging 1998;19:33-6.
  73. Montine KS, Reich E, Neely MD, Sidell KR, Olson SJ, Markesbery WR, et al. Distribution of reducible 4-hydroxynonenal adduct immunoreactivity in Alzheimer disease is associated with APOE genotype. J Neuropathol Exp Neurol 1998;57:415-25.
  74. Lu T, Aron L, Zullo J, Pan Y, Kim H, Chen Y, et al. REST and stress resistance in aging and Alzheimer’s disease. Nature 2014;507:448-54.
  75. Jomova K, Vondrakova D, Lawson M, Valko M. Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem 2010;345:91-104.
  76. Hensley K, Hall N, Subramaniam R, Cole P, Harris M, Aksenov M, et al. Brain regional correspondence between Alzheimer's disease histopathology and biomarkers of protein oxidation. J Neurochem 1995;65:2146-56.
  77. Aksenov MY, Aksenova MV, Butterfield DA, Geddes JW, Markesbery WR. Protein oxidation in the brain in Alzheimer's disease. Neuroscience 2001;103:373-83.
  78. Anderson AJ, Su JH, Cotman CW. DNA damage and apoptosis in Alzheimer's disease: colocalization with c-Jun immune-reactivity, relationship to brain area, and effect of postmortem delay. J Neurosci 1996;16:1710-9.
  79. Mullaart E, Boerrigter ME, Ravid R, Swaab DF, Vijg J. Increased levels of DNA breaks in the cerebral cortex of Alzheimer's disease patients. Neurobiol Aging 1990;11:169-73.
  80. Foy CJ, Passmore AP, Vahidassr MD, Young IS, Lawson JT. Plasma chain breaking antioxidants in Alzheimer's disease, vascular dementia and Parkinson's disease. Q J Med 1999;92:39-45.
  81. Kim TS, Pae CU, Yoon SJ, Lee NJ, Kim JJ, Lee SJ, et al. Decreased plasma antioxidants in patients with Alzheimer's disease. Int J Geriatric Psychiatry 2006;21:344-8.
  82. Alejandro Gella, Nuria Durany. Oxidative stress in Alzheimer disease. Cell Adhesion Migration 2009;3:1, 88-93.
  83. Karbowski M. Mitochondria on guard: role of mitochondrial fusion and fission in the regulation of apoptosis. Adv Exp Med Biol 2010;687:131–42.
  84. Castellani R, Hirai K, Aliev G, Drew KL, Nunomura A, Takeda A, et al. Role of mitochondrial dysfunction in Alzheimer's disease. J Neurosci Res 2002;70:357-60.
  85. Gibson GE, Sheu KF, Blass JP. Abnormalities of mitochondrial enzymes in Alzheimer disease. J Neural Transm 1998;105:855-70.
  86. Wang X, Su B, Zheng L, Perry G, Smith MA, Zhu X. The role of abnormal mitochondrial dynamics in the pathogenesis of Alzheimer's disease. J Neurochem 2009;109:153-9.
  87. Schmitt K, Grimm A, Kazmierczak A, Strosznajder JB, otz JG, Eckert A. Insights into mitochondrial dysfunction: aging, amyloid-𝛽, and tau-A deleterious trio. Antioxid Redox Signaling 2012;16:1456-66.
  88. Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G. Mitochondrial dysfunction is a trigger of Alzheimer’s disease pathophysiology. Biochim Biophys Acta 2010;1802:2-10.
  89. Hauptmann S, Scherping I, Drose S, Brandt U, Schulz KL, Jendrach M, et al. Mitochondrial dysfunction: an early event in Alzheimer pathology accumulates with age in AD transgenic mice. Neurobiol Aging 2009;30:1574-86.
  90. Gibson GE, Chen HL, Xu H, Qiu L, Xu Z, Denton TT, et al. Deficits in the mitochondrial enzyme alpha-ketoglutarate dehydrogenase leads to Alzheimer’s disease like calcium dysregulation. Neurobiol Aging 2012;33:1113-24.
  91. Peterson C, Gibson GE, Blass JP. Altered calcium uptake in cultured skin fibroblasts from patients with Alzheimer's disease. N Engl J Med 1985;312:1063-5.
  92. Ito E, Oka K, Etcheberrigaray R, Nelson TJ, McPhie DL, Tofel-Grehl B, et al. Internal Ca2+mobilization is altered in fibroblasts from patients with Alzheimer disease. Proc Natl Acad Sci USA 1994;91:534-8.
  93. Olichon A, Landes T, Arnaune-Pelloquin L, Emorine LJ, Mils V, Guichet A, et al. Effects of OPA1 mutations on mitochondrial morphology and apoptosis: relevance to ADOA pathogenesis. J Cell Physiol 2007;211:423–30.
  94. Perry EK, Perry RH, Tomlinson BE. Coenzyme a acetylating enzymes in Alzheimer’s disease: possible cholinergic compartment of pyruvate dehydrogenase. Neurosci Lett 1980;18:105-10.
  95. Gibson GE, Sheu KF, Blass JP. Abnormalities of mitochondrial enzymes in Alzheimer disease. J Neural Transm 1998;105:855-70.
  96. Chandrasekaran K, Giordano T, Brady DR, Stoll J, Martin LJ, Rapoport SI. Impairment in mitochondrial cytochrome oxidase gene expression in Alzheimer disease. Brain Res Mol Brain Res 1994;24:336-40.
  97. Cottrell DA, Blakely EL, Johnson MA, Ince PG, Turnbull DM. Mitochondrial enzyme-deficient hippocampal neurons and choroidal cells in AD. Neurology 2001;57:260-4.
  98. Maurer I, Zierz S, Moller HJ. A selective defect of cytochrome c oxidase is present in the brain of Alzheimer disease patients. Neurobiol Aging 2000;21:455-62.
  99. Nagy Z, Esiri MM, LeGris M, Matthews PM. Mitochondrial enzyme expression in the hippocampus in relation to Alzheimer-type pathology. Acta Neuropathol 1999;97:346-54.
  100. Parker WD, Mahr NJ, Filley CM, Parks JK, Hughes D, Young DA, et al. Reduced platelet cytochrome c oxidase activity in Alzheimer's disease. Neurology 1994;44:1086-90.
  101. Ko LW, Sheu KFR, Thaler HT, Markesbery WR, Blass JP. Selective loss of KGDHC-enriched neurons in Alzheimer temporal cortex: does mitochondrial variation contribute to selective vulnerability? J Mol Neurosci 2001;17:361-9.
  102. Perry EK. The cholinergic hypothesis ten years on. Br Med Bull 1986;42:63-9.
  103. Lyketsos CG, Carrillo MC, Ryan JM. Neuropsychiatric symptoms in Alzheimer’s disease. Alzheimer’s Dementia 2011;7:532-9.
  104. Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 y. J Neuropathol Exp Neurol 2011;70:960-9.
  105. Beach TG, Monsell SE, Phillips LE, Kukull W. Accuracy of the clinical diagnosis of Alzheimer disease at the national institute on aging Alzheimer disease centers. J Neuropathol Exp Neurol 2012;71:266-73.
  106. Petersen RC, Stevens JC, Ganguli M, Tangalos EG, Cummings JL, Dekosky ST. Practice parameter: early detection of dementia: mild cognitive impairment (an evidence-based review). Neurology 2001;56:1133-42.
  107. Knopman DS, Dekosky ST, Cummings JL. Practice parameter: diagnosis of dementia (an evidenced-based review). Neurology 2001;56:1143-53.
  108. Doody RS, Stevens JC, Beck C. Practice parameter: management of dementia (an evidenced-based review). Neurology 2001;56:1154-66.
  109. Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New pharmacological strategies for treatment of Alzheimer’s disease: focus on disease modifying drugs. Br J Clin Pharmacol 2012;73:504-17.
  110. Panza F, Frisardi V, Imbimbo BP, Seripa D, Paris F, Santamato A, et al. Anti-β-amyloid immunotherapy for Alzheimer’s disease: focus on bapineuzumab. Curr Alzheimer Res 2011;8:808-17.
  111. Aprahamian I, Florindo S, Orestes, Forlenza V. New treatment strategies for Alzheimer’s disease: is there hope? Indian J Med Res 2013;449-60.
  112. Bolognesi ML, Bartolini M, Tarozzi A, Morroni F, Lizzi F, Milelli A, et al. Multitargeted drugs discovery: balancing anti-amyloid and anticholinesterase capacity in a single chemical entity. Bioorg Med Chem Lett 2011;21:2655-8.
  113. Nagaraja Prasad S, Jagadeesh K, Vedavathi H, Shreenivas P. Alzheimer disease: therapeutic targets and recent developments in treatment. Sch Acad J Pharm 2015;4:222-5.
  114. Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S, Jacobson LH, et al. The second-generation active Aβ immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. J Neurosci 2011;31:9323-31.
  115. Landen JW, Zhao Q, Cohen S, Borrie M, Woodward M, Billing CB, et al. Safety and pharmacology of a single intravenous dose of ponezumab in subjects with mild-to-moderate Alzheimer disease: a phase I, randomized, placebo-controlled, double-blind, dose-escalation study. Clin Neuropharmacol 2013;36:14-23.
  116. Burstein AH, Zhao Q, Ross J, Styren S, Landen JW, Ma WW, et al. Safety and pharmacology of ponezumab (PF-04360365) after a single 10-minute intravenous infusion in subjects with mild to moderate Alzheimer disease. Clin Neuropharmacol 2013;36:8-13.
  117. Barakos J, Carlson C, Estergard W, Oh J, Suhy J, Jack CR, et al. Vasogenic edema in the setting of β-amyloid lowering therapy, adverse event: what is it and how is it detected? Alzheimers Dement 2011;7(Suppl):75.
  118. Chang WP, Huang X, Downs D, Cirrito JR, Koelsch G, Holtzman DM, et al. Beta-secretase inhibitor GRL-8234 rescues age-related cognitive decline in APP transgenic mice. FASEB J 2011;25:775-84.
  119. Hamada Y, Kiso Y. Advances in the identification of β-secretase inhibitors. Expert Opin Drug Discovery 2013;8:709-31.
  120. Sivilia S, Lorenzini L, Giuliani A, Gusciglio M, Fernandez M, Baldassarro VA, et al. Multitarget action of the novel anti-Alzheimer compound CHF5074: in vivo study of long-term treatment in T-2576 mice. BMC Neurosci 2013;14:44.
  121. Vassar R, Bennett BD, Babu-Khan S. 𝛽-Secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 1999;286:735-41.
  122. Yan R, Blenkowski MJ, Shuck ME. Membrane anchored aspartyl protease with Alzheimer’s disease 𝛽-secretase activity. Nature1999;402:533-7.
  123. Michalik L, Auwerx J, Berger JP. International union of pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev 2006;58:726-41.
  124. Mandrekar-Colucci S, Colleen Karlo J, Landreth GE. Mechanisms underlying the rapid peroxisome proliferator activated receptor-γ mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer’s disease.  J Neurosci 2012;32:10117-28.
  125. Landreth G, Jiang Q, Mandrekar S, Heneka M. PPAR agonists as therapeutics for the treatment of Alzheimer’s disease. Neurotherapeutics 2008;5:481-9.
  126. Craft S. The role of metabolic disorders in Alzheimer disease and vascular dementia: two roads converged. Arch Neurol 2009;66:300-5.
  127. Shoji M, Golde TE, Ghiso J. Production of the Alzheimer amyloid 𝛽 protein by normal proteolytic processing. Science 1992;258:126-9.
  128. Haass C, Schlossmacher MG, Hung AY, Vigo-Pelfrey C, Mellon A, Ostaszewski BL, et al. Amyloid beta-peptide is produced by cultured cells during normal metabolism. Nature 1992;359:322-5.
  129. Borgegcard T, Gustavsson S, Nilsson C. Alzheimers disease: presenilin 2-sparing γ-secretase inhibition is a tolerable peptide-lowering strategy.  J Neurosci 2012;32:17297-305.
  130. Zhao B, Yu M, Neitzel M, Marugg J, Jagodzinski J, Lee M, et al. Identification of γ-secretase inhibitor potency determinants on presenilin. J Biol Chem 2008;283:2927-38.
  131. Crump CJ, Castro SV, Wang F, Pozdnyakov N, Ballard TE, Sisodia SS, et al. BMS-708,163 targets presenilin and lacks notch-sparing activity. Biochemistry 2012;51:7209-11.
  132. Pei JJ, Ogren MS, Winblad B. Neurofibrillary degeneration in Alzheimer’s disease: from molecular mechanisms to identification of drug targets. Curr Opinion Psychiatry 2008;21:555-61.
  133. QiutianJia, Yulin Deng, Hong Qing. Potential therapeutic strategies for Alzheimer’s disease targeting or beyond 𝛽-Amyloid: Insights from clinical trials. Bio Med Res Int 2014. doi.org/10.1155/2014/837157. [Article in Press]
  134. Seren L, Coma M, Rodr M, Guez L. A novel GSK-3α inhibitor reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol Dis 2009;35:359-67.
  135. Hurtado DE, Molina-Porcel L, Iba M. A𝛽 accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am J Pathol 2010;177:1977-88.
  136. Zhang B, Carroll J, Trojanowski JQ. The microtubule-stabilizing agent, epothilone D, reduces axonal dysfunction, neurotoxicity, cognitive deficits, and Alzheimer's-like pathology in an interventional study with aged tau transgenic mice. J Neurosci 2012;32:3601-11.
  137. Swerdlow RH, Khan SM. A mitochondrial cascade hypothesis for sporadic Alzheimer’s disease. Med Hypotheses 2004; 63:8–20.
  138. Doody RS, Gavrilova SI, Sano M, Thomas RG, Aisen PS, Bachurin SO, et al. Effect of dimebon on cognition, activities of daily living, behavior, and global function in patients with mild-to-moderate Alzheimer’s disease: a randomized, double-blind, placebo-controlled study. Lancet 2008;372:207-15.
  139. Tariot P, Sabbagh M, Flitman S, Reyes P, Taber L, andSeely L. A safety, tolerability and pharmacokinetic study of dimebon in patients with Alzheimer’s disease already receiving donepezil. Alzheimer’s Dementia 2009;5:P251.
  140. Reiman EM, Chen K, Alexander GE. Functional brain abnormalities in young adults at genetic risk for late-onset Alzheimer's dementia. Proc Natl Acad Sci USA 2004; 101:284-9.
  141. Nordberg A, Darreh-Shori T, Svenson A, Guan Z. AChE and BuChE activities in CSF of mild AD patients following 12 mo of rivastigmine treatment. J Neurol Sci 2001;187:P0144.
  142. Hampel H. Current insights into the pathophysiology of Alzheimer’s disease: selecting targets for early therapeutic intervention. Int Psychogeriatr 2012;24(Suppl 1):S10-7.
  143. Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New pharmacological strategies for treatment of Alzheimer’s disease: focus on disease modifying drugs. Br J Clin Pharmacol 2012;73:504-17.
  144. Hanger DP, Anderton BH, Noble W. Tau phosphorylation: the therapeutic challenge for neurodegenerative disease. Trends Mol Med 2009;15:112-9.