• Open access
  • Published: 09 May 2019

Alzheimer’s disease: risk factors and potentially protective measures

  • Marcos Vinícius Ferreira Silva 1 ,
  • Cristina de Mello Gomide Loures 1 ,
  • Luan Carlos Vieira Alves 1 ,
  • Leonardo Cruz de Souza 2 ,
  • Karina Braga Gomes Borges 1 &
  • Maria das Graças Carvalho 1  

Journal of Biomedical Science volume  26 , Article number:  33 ( 2019 ) Cite this article

91k Accesses

385 Citations

542 Altmetric

Metrics details

Alzheimer’s disease (AD) is the most common type of dementia and typically manifests through a progressive loss of episodic memory and cognitive function, subsequently causing language and visuospatial skills deficiencies, which are often accompanied by behavioral disorders such as apathy, aggressiveness and depression. The presence of extracellular plaques of insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFT) containing hyperphosphorylated tau protein (P-tau) in the neuronal cytoplasm is a remarkable pathophysiological cause in patients’ brains. Approximately 70% of the risk of developing AD can be attributed to genetics. However, acquired factors such as cerebrovascular diseases, diabetes, hypertension, obesity and dyslipidemia increase the risk of AD development. The aim of the present minireview was to summarize the pathophysiological mechanism and the main risk factors for AD. As a complement, some protective factors associated with a lower risk of disease incidence, such as cognitive reserve, physical activity and diet will also be addressed.

Introduction

Alzheimer’s disease (AD) is the most common type of dementia [ 1 ], affecting at least 27 million people and corresponding from 60 to 70% of all dementias cases [ 2 ]. The occurrence of this disease also has a huge impact on life of patient’s family, in addition to a high financial cost to society [ 3 ]. From an anatomopathological point of view, AD is characterized by two prototypical lesions: 1) senile plaques, composed of a nucleus of β-amyloid protein accumulation (Aβ42), as extra-cellular lesions and 2) neurofibrillary tangles composed of phosphorylated tau protein (P-tau) and which are intraneuronal findings [ 4 ]. Deposition of β-amyloid protein can also occur in capillaries walls, arteries and arterioles causing amyloid cerebral angiopathy leading to degeneration of vascular wall componentes and worsening of blood flow, besides predisposing to intraparenchymal hemorrhages [ 5 ].

AD typically manifests through a progressive loss of episodic memory and cognitive function, with later deficiency of language and visuospatial abilities. Such changes are often accompanied by behavioral disorders such as apathy, aggressiveness and depression [ 6 ]. It should be noted that there is an important subgroup of AD patients who do not present a typically amnestic picture, manifesting non-amnestic deficits from the onset of symptoms [ 7 ]. Structural neuroimaging, with a pattern of hippocampal and parietal atrophy in typical cases reinforces the diagnosis [ 8 ]. Patients who meet typical disease characteristics, excluding other causes such as vascular and fronto-temporal dementias, have a probable diagnosis of AD [ 6 ]. Definitive diagnosis of the disease is usually carried out only through postmortem examination, whose purpose is to demonstrate histologically the neurofibrillary tangles and the senile plaques [ 9 ].

Pathophysiology of Alzheimer’s disease

The presence of extracellular plaques of insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFT) of P-tau in neuronal cytoplasm is the hallmark of AD [ 10 ]. Although the mechanisms by which these changes lead to cognitive decline are still debated, these deposits are believed to lead to atrophy and death of neurons resulting from excitotoxicity processes [excessive stimulation of neurotransmitter receptors in neuronal membranes], collapse in calcium homeostasis, inflammation and depletion of energy and neuronal factors. As a result of this process, damage to neurons and synapses involved in memory processes, learning and other cognitive functions lead to the aforementioned cognitive decline [ 11 ].

According to amyloid cascade theory (one of the most accepted theories about AD pathogenesis, although still debated), the cerebral accumulation of Aβ peptide, resulting from the imbalance between production and clearance of this protein, is the main event causing the disease, being other events observed (including the formation of NFT) resulting from this process [ 12 ].

The Aβ peptide, which has 36 to 43 aminoacids, is derived from amyloid precursor protein (APP) enzymatic proteolysis, a physiologically produced protein that plays important roles in brain homeostasis [ 13 , 14 ]. The APP gene is located on chromosome 21, which explains the higher incidence of early-onset AD in individuals with 21 trisomy (Down Syndrome) and in individuals with APP gene locus duplication [a rare form of early onset of familial origin]. It is believed that overexpression of APP results in an increase of cerebral Aβ peptide, and consequently, in its deposition [ 15 ].

Two main pathways for APP processing are now recognized: a non-amyloidogenic α-secretase-mediated pathway and an amyloidogenic β-and γ-secretase-mediated pathway. Cleavage of APP by α-secretase results in a soluble molecule, sAPPα, which has probable neuroprotective function, playing important roles in the plasticity and survival of neurons and protection against excitotoxicity [ 16 , 17 ]. The Aβ peptide is produced by APP cleavage by a β-secretase (mainly BACE1 enzyme). In this pathway, APP is cleaved by β-secretase to give a APP soluble fragment (sAPPβ, a mediator related to neuronal death), and a carboxy-terminal complex linked to cell membrane. The latter is cleaved by a γ-secretase complex composed by 4 proteins: presenilin 1 or 2, nicastrin, APH-1 (formerly pharynx-defective-1) and and PEN-2 (presenilin enhancer-2), to give rise to the Aβ peptide. Aβ peptides ranging in size from 38 to 43 aminoacids are generated with predominance of the 40 aminoacid form (Aβ 40), followed by 42 (Aβ 42) [ 17 , 18 ]. In physiological conditions, the amyloidogenic and non-amyloidogenic pathways coexist in equilibrium, the latter being favored preferentially [ 19 ].

The Aβ42 peptide is more prone to aggregation than Aβ40. Immunohistochemical analyses indicate that Aβ42 is initially deposited and found at higher concentrations in the amyloid plaques observed in AD patients [ 20 ]. Several studies showed that CSF Aβ42 levels are surrogate markers of underlying brain amyloidosis [ 21 , 22 ]. On the contrary, the correlation between serum Aβ42 levels and cerebral amyloidosis is not yet demonstrated. A decrease in Aβ42 levels is observed in cerebrospinal fluid of AD subjects, which can be explained in part by higher deposition of β-amyloid plaques [ 23 ]. As additional evidence of Aβ42 peptide and the AD pathophysiology, it is further noted that mutations in APP and presenilin genes, which give rise to early-onset familial AD forms, lead to a relative increase in Aβ42 levels [ 20 ].

Aβ peptides, under physiological conditions, are produced primarily in monomeric forms with synapses protective function. However, the accumulation of this protein leads to formation of fibrils that accumulate in senile plaques. High levels of Aβ may lead to oligomeric products formation (dimers, trimers, tetramers) leading to neuronal toxicity and degeneration (both by interaction with cell membranes and their receptors, and by direct interference in intracellular processes), interfering with the function and survival of cholinergic, serotonergic, noradrenergic and dopaminergic neurons, reducing their control over the amyloidogenic pathway and favoring the accumulation of insoluble Aβ peptide [ 19 , 24 ].

The exact mechanism by which deposition of Aβ peptide promotes NFT formation of hyperphosphorylated tau protein is not known. Blurton-Jones & Laferla (2006) [ 25 ] suggest four basic mechanisms:

The Aβ peptide promotes the activation of specific kinases (GSK3β, e.g.) that catalyze the hyperphosphorylation of tau protein, leading to its conformation change and formation of NFT;

Neuroinflammation promoted by the deposition of Aβ peptide leads to the production of proinflammatory cytokines that stimulate the phosphorylation of tau protein;

Reduced capacity of degradation of tau protein by the proteasome, in a process induced by Aβ peptide;

Defects in axonal transport promoted by Aβ peptide lead to inadequate localization of tau protein and its messenger RNA, which can lead to hyperphosphorylation and aggregation in NFT.

Tau protein is a microtubule-associated protein, produced by alternative splicing of the MAPT gene, located on chromosome 17 (17q21). Six isoforms of tau protein are produced by this process [ 26 ]. The main known physiological functions of this protein are the stimulation of tubulin polymerization, microtubules stabilization and intracellular organelles transport by microtubules. Once hyperphosphorylated, the protein loses its functions in the synthesis and stabilization of microtubules, leading to neuronal damage and promoting cytotoxicity [ 27 ]. Histological analyses demonstrate that both the load and the distribution of NFT in brain tissue correlate better with the severity of cognitive deficit than the Aβ peptide deposits [ 28 ].

  • Genetic risk factors

AD can be classified by the age of onset of the first symptoms. Early-onset AD affects individuals under 65 years of age, accounting for about 4–6% of cases of AD, while the late form AD affects individuals aged 65 years or older. Besides the age of onset of symptoms, the early and late forms of AD differ in other clinical, neuropsychological, neuropathological and neuroimaging variables [ 29 ].

According to Ballard et al. (2011) [ 1 ] about 70% of the risk of developing AD can be attributed to genetics. Early AD usually occurs due to mutations in genes APP, PSEN1 and PSEN2 (genes of amyloid precursor protein, presenilin 1 and presenilin 2, respectively), whereas late-form AD is mainly associated with a polymorphism in APOE gene (apolipoprotein E gene), especially the presence of ε4 allele [ 30 , 31 ].

More than 30 dominant mutations have already been found in APP gene (located in chromosome 21q21) and are associated with about 15% of cases of early-onset autosomal dominant AD. Mutations in PSEN1 gene (located at 14q24.3) are associated with 80% of cases of early-onset AD, whereas 5% of cases are associated with PSEN2 mutations (located at 1q31-q42) [ 32 ]. Most of APP gene mutations, as well as PSEN1 mutations, lead to an increase in Aβ42: Aβ40 ratio, either by Aβ42 increased expression, reduction of Aβ40, or both. This deregulation favors early Aβ deposition in brain tissue favoring the amyloidogenic cascade [ 33 ]. It is believed that there are other genes besides APP, PSEN1 and PSEN2 involved in the pathogenesis of early-onset AD, as demonstrated by Campion et al. (1999) [ 34 ].

Apolipoprotein E (ApoE) is a protein involved in lipid metabolism encoded by APOE gene, located on chromosome 19. There are three APOE alleles described (ε2, ε3 and ε4, giving rise to apoE2, apoE3 and apoE4 isoforms), present in population at different frequencies (ε2: 5–10%, ε3: 65–70% and ε4: 15–20%). A study by Corbo and Scacchi (1999) [ 35 ] showed that there is a great variability in the APOE allele distribution among the different populations, with ε2 frequencies varying from 0.0 in some Native American populations up to 0.145 in Papuans. The ε 4 frequencies obtained by the authors range from 0.052 (Sardinians) to 0.407 (Pygmies). The ε4 allele is the main risk factor for late-onset AD. The presence of ε4 in heterozygosity increases 3-fold the risk of AD developing, whereas in homozygosis, the risk is increased 12-fold. Conversely, the presence of ε2 allele reduces the risk of AD developing [ 36 , 37 ].

The causes of the association between apoE are not yet fully understood, although some mechanisms have been proposed, and presented consistent results in clinical and in vitro studies. Among these studies, some show that apoE is able to bind to Aβ peptide. While the apoE4 isoform binds to Aβ peptide promoting its polymerization in fibrils and its deposition, apoE2 and apoE3 forms are more efficient in promoting the clearance of this peptide, reducing its deposition in brain tissue [ 38 ]. ApoE has neuroprotective effects and is able to act on neurons development, with apoE2 and apoE3 performing better than apoE4. Additionally, it is observed that protease-generated apoE fragments have toxic effects, which may lead to neuronal injury and favor Aβ peptide deposition [ 38 , 39 ].

More recently it was observed that rare alterations in the triggering receptor expressed on myeloid cells 2 ( TREM2 ) gene elevated the risk ratio by 2.9% for AD development [ 40 , 41 ]. The pathophysiological mechanism by which the deficiency in the gene increases the risk ratio for AD still needs to be better clarified. The gene is located on chromosome 6p21 [ 42 ] and the TREM2 protein is a highly expressed receptor on the surface of microglia, phagocytic cells of central nervous system, and has the function of modulating phagocytic and inflammatory responses in central nervous system [ 43 ]. Activation of microglia through the interaction of TREM2 and DAP12 stimulates the production of CCL19 and CCL21 chemokines and phagocytosis [ 44 ]. In knockout models for the TREM2 receptor it was observed that phagocytic capacity of apoptotic neuronal cell bodies was deficient [ 44 ]. Thus the accumulation of these cellular debris would promote a proinflammatory microenvironment [ 44 ]. Xiang et al. (2016) [ 45 ] observed that the removal capacity of Aβ peptide deposits is impaired in TREM2 receptor deficiency and would favor amyloid plaques accumulation.

  • Acquired risk factors

A number of acquired factors increase the risk of developing AD. Among those factors are cerebrovascular diseases (most commonly reported risk factor), diabetes, hypertension, obesity and dyslipidemia [ 46 ]. The association of these risk factors to AD development will be described in the following subsections, as well as some protective factors associated with a lower risk of disease incidence, such as cognitive reserve, physical activity and diet as reported by Mayeux & Stern (2012) [ 46 ].

Cerebrovascular diseases

Cerebrovascular diseases and AD share many risk factors, which often overlap. Cerebrovascular changes such as hemorrhagic infarcts, small and large ischemic cortical infarcts, vasculopathies, and changes in cerebral white matter are known to increase the risk of dementia. Postmortem analyses of the brains of patients with AD indicate the presence of parenchymal vascular disease (amyloid angiopathy by Aβ peptide and small vessels arteriolosclerotic disease), with hemorrhagic outbreaks and infarcts being found in more than 50% of them [ 47 , 48 ]. According to Liu et al., (2015) [ 49 ], neuropathological findings indicate that between 6 and 47% of individuals with dementia have a simultaneous occurrence of cerebrovascular disease. These observations point to the potential role of homeostatic mechanisms in AD and lead to question whether the dementias in which vascular processes are involved are fundamentally different from those related to accumulation of Aβ42 and tau proteins or if both pathological processes produce synergistic effects on cognitive function [ 9 ].

According to the “double-stroke” theory of AD, vascular risk factors (“first stroke”) lead to dysfunction in blood-brain barrier and reduction in cerebral blood flow, with decreased blood supply to the region (oligoemia). This event leads to neuronal damage by non-amyloidogenic and amyloidogenic pathways. Firstly, the dysfunction of blood-brain barrier leads to oligoemia and the accumulation of neurotoxic molecules, events associated with the occurrence of multiple focal ischemic infarcts and micro-injuries resulting from hypoxia, causing neuronal damage. In the amyloidogenic pathway, vascular injury leads to increased expression and processing of APP, resulting in an increase in Aβ peptide. In addition, damage to blood-brain barrier leads to decreased clearance of Aβ peptide. The accumulation of amyloid in brain (“second stroke”) amplifies neuronal dysfunction and speeds up neurodegeneration process. Both Aβ peptide accumulation and hypoperfusion lead to hyperphosphorylation of tau protein, promoting the formation of NFT [ 50 ].

Hypertension

A longitudinal study carried out by Skoog et al. (1996) demonstrated that hypertension is capable of leading to increased risk of developing AD [ 51 ]. Other studies have confirmed this association, indicating that hypertension, especially when present in middle age, negatively affects cognitive performance at more advanced ages, and this association becomes weaker with age [ 52 ]. Hypertension is capable of causing changes in the vascular walls which can lead to hypoperfusion, ischemia and cerebral hypoxia, contributing to trigger the development of AD. Studies demonstrate that cerebral ischemia is capable of leading to the accumulation of APP and Aβ, in addition to stimulating the expression of presenilin, involved in Aβ synthesis. Hypertension may also lead to dysfunction in the blood-brain barrier, an event associated with the genesis of AD by previously discussed mechanisms [ 53 ].

Type 2 diabetes

Epidemiological studies indicate a clear association between type 2 diabetes mellitus and the increased risk of developing AD. Several mechanisms for this association are suggested, including insulin resistance and insulin deficiency, impaired insulin receptor, toxicity of hyperglycemia, adverse effects due to advanced glycation end products, cerebrovascular damage, vascular inflammation and others [ 54 ].

The use of animal models was able to demonstrate that deficiency or resistance to insulin are able to stimulate the action of β and γ-secretases, besides promoting reduction of Aβ clearance, leading to its accumulation in brain tissue. Insulin resistance or deficiency are still capable of inducing hyperphosphorylation of tau protein, leading to NFT formation. Insulin and insulin-like growth factor bind to insulin receptor, leading to its autophosphorylation and activation. Activation of this receptor leads to phosphorylation of phosphoinositide 3-kinase (PI3K) enzyme, which in turn phosphorylates and inhibits glycogen synthase kinase 3β (GSK3β) enzyme, which is important for tau protein phosphorylation. Thus, insulin deficiency / resistance leads to GSK3β abnormal activation, and consequently, to an increase of p-tau formation [ 55 ].

In addition to the mechanisms discussed earlier, studies have reported that advanced glycation end products (AGEs) induce neuronal death through activation of cell death pathways, in addition to stimulating APP processing through increased expression of complexes β and γ-secretases (BACE and PSEN1), in a process involving reactive oxygen species generation [ 56 ]. In addition, Aβ peptide may undergo non-enzymatic glycation, making it an AGE more neurotoxic than its non-glycated form [ 57 ].

The role of obesity as a risk factor for AD development is still uncertain, with studies presenting rather heterogeneous results. According to a meta-analysis developed by Profenno, Porsteinsson, & Faraone (2010) [ 58 ], obesity (Body Mass Index - BMI ≥30 kg / m 2 ) is significantly and independently associated with AD developing risk. On the other hand, a meta-analysis conducted by Fitzpatrick et al. (2009) [ 59 ] indicated that obesity in middle age is a risk factor for dementia development (hazard ratio - HR: 1.39; 95% CI: 1.03–1.87), while in later stages of life, obesity is inversely correlated with the risk of dementia (HR: 0.63; 95% CI: 0.44–0.91). The same authors have also reported that below-ideal weight (BMI < 20 kg / m 2 ) is also associated with an increased risk of dementia (HR: 1.62, 95% CI: 1.02–2.64). Weight loss at more advanced ages occurs in concomitance to other comorbidities and is often indicative of poor health, and may even precede dementia onset within 10 years. Another meta-analysis conducted by Anstey et al. (2011) [ 60 ] indicated that both low weight and overweight as well as obesity in middle age are associated with a higher risk of developing AD in late life.

Dyslipidemia

Elevated cholesterol levels have been proposed as risk factors for the development of AD. Studies have already demonstrated 10% higher cholesterol levels in patients with AD, compared to healthy individuals [ 61 ]. Hypercholesterolemia is a risk factor both for atherosclerosis development and AD development as well as other neurodegenerative diseases [ 62 ].

Hypercholesterolemia increases AD risk primarily because of its effects on the blood-brain barrier. Studies have shown that elevated circulating cholesterol levels are capable of compromising integrity in blood-brain barrier [ 62 ], resulting in mechanisms previously discussed. In addition, experimental studies using animal models demonstrate that hypercholesterolemia is associated with increased Aβ peptide deposition, in addition to increased NFT formation, cognitive decline, neuroinflammation, dysfunction of cholinergic neurons and the presence of cerebral microhemorrhages compatible with AD [ 63 , 64 ].

In observational studies a beneficial effect was observed in the users of statins as the reduction in AD incidence or improvement in the disease progression [ 65 , 66 , 67 ]. However, clinical studies to date have not demonstrated benefit of statins treatment and protection against cognitive decline in AD patients at various stages of disease [ 68 , 69 , 70 , 71 , 72 ]. Contrary to meta-analysis findings conducted by Song et al. (2013) [ 73 ] who observed a lower risk of developing AD in statins users, a Cochrane meta-analysis [ 74 ] did not observe difference in disease outcome as well as alteration in mini-mental status examination (MMSE) in patients using or not statins. However, some important questions regarding the clinical studies are pointed out, i.e., whether treatment initiated in middle age prior disease onset would also have a beneficial effect in elderly, or whether in people with AD family history the treatment would be effective in comparison to those without this background.

Marital status, stress, depression and inadequate sleep

Widowhood status has been reported as an important risk factor AD. A cohort study by Håkansson et al. (2009) [ 75 ] shows that widowed individuals have an increased risk of developing AD compared to married or cohabiting individuals and that this effect is more pronounced in carriers of the APOE ε4 allele. Other studies, such as that by Fan et al. (2015) [ 76 ] demonstrated an association between the risk of all-cause dementia and widow status. A meta-analysis by Sommerlad et al. (2018) [ 77 ] reported an association between widowhood and all-cause dementia, but the same association was not found between widowhood and AD or vascular dementia.

Studies in animal models of AD have shown that stress, characterized as hyperactivation of the hypothalamic, pituitary and adrenal axis (HPA) leading to an increase in cortisol production, causes an increase in Aβ peptide deposition in regions of the brain such as hypothalamus and prefrontal cortex [ 78 , 79 , 80 ]. Carroll et al. (2011) [ 81 ] have observed that the prolonged stress caused by this hyperactivation also causes an increase in the accumulation of hyperphosphorylated tau and neurodegeneration in mice. In humans, increased levels of cortisol were observed in patients with AD compared to the control group [ 82 , 83 , 84 ]. Huang et al. (2009) [ 85 ] observed in a 2-year follow-up of patients with AD that the higher cortisol levels correlated with the faster progression of the disease, worsened in the MMSE and smaller volume of the hippocampus region when observed by resonance. The authors of this study argue that hippocampal atrophy causes a disinhibition effect on the HPA axis, which would cause elevation in cortisol levels as a consequence of the pathophysiological process of AD. Toledo et al. (2012) [ 86 ], observed in a sample of 26 patients with AD that the increase in cortisol levels is correlated with the deposition of the Aβ peptide observed by means of pittsburgh compound b-positron emission tomography (PiB-PET). Ennis et al. (2017) [ 87 ], in a 10-year longitudinal study with 1025 participants observed an increased risk of 1.31 for the development of AD and elevation in cortisol levels that were dosed in 24-h urine samples. However, this result contrasts with that observed in the Rotterdan study [ 88 ] in blood samples collected in the morning when there was no correlation between cortisol levels and AD or dementia in general.

Early adult depression is a risk factor for the development of dementia at more advanced age including AD [ 89 , 90 , 91 ]. Zverova et al. (2013) [ 83 ] observed a greater odds ratio for cognitive decline in the presence of cortisol levels and patients with AD and symptoms of depression. Wu et al. (2018) [ 92 ] observed in some patients with major depression in middle age hippocampal atrophy and Aβ peptide deposition observed by PET indicating that the protein metabolism may be altered in patients with depression.

According to a study published by Proserpio et al. (2018) [ 93 ], sleep disorders have a bidirectional relationship with AD: sleep disorders arise during the early stages of dementia and tend to worsen with the onset of dementia. Similarly, sleep disorders can lead to an increased risk of dementia. A meta-analysis by Shi et al. (2018) [ 94 ] demonstrated that individuals with sleep disorders have an increased risk of developing dementia. More specifically, individuals with insomnia are at high risk for developing AD but not for vascular dementia or other causes. Similarly, individuals with sleep disordered breathing had an increased risk of developing all-cause dementia, AD, and vascular dementia.

Smoking may affect the risk of developing AD by various mechanisms. It is known that it is able to raise the generation of free radicals, increasing oxidative stress, and to promote pro-inflammatory action in the immune system, leading to the activation of phagocytes and consequently, additional oxidative damage. In addition, smoking may lead to cerebrovascular diseases, which increase the risk of AD [ 95 , 96 ]. In a meta-analysis performed by Cataldo et al. (2010), an analysis of 8 case-control studies with affiliations with the tobacco industry suggested a protective effect of smoking in relation to AD (odds ratio (OR): 0.91, 95% CI 0.75–1, 10). In contrast, 14 cohort studies with no association with the tobacco industry demonstrated an increased relative risk for smokers (Relative Risk (RR): 1.45; 95% CI, 1.16–1.80) [ 97 ]. According to Durazzo et al. (2014), the sum of the evidence presented today in the literature is enough for the cessation of smoking to be recommended in order to reduce the incidence of dementia [ 96 ].

Protective factors

Cognitive reserve.

It has been observed in many cases a discrepancy between the degree of brain damage found in histopathological analyses and the severity of cognitive decline. To explain these findings, the theory of cognitive reserve was proposed, which postulates that the gap between brain injury and clinical manifestations is attributable to cognitive reserve capacity. This can be subdivided into two models: brain reserve model or threshold, and cognitive reserve model and / or compensation. The first is based on the amount of available neural substrate (eg, brain size, synapses density or dendritic branching), while the latter focuses on the more efficient ability to use the preexisting brain network in healthy individuals and on the recruitment of more resources to support normal functioning in presence of brain damage [ 98 ].

Several elements are associated with a greater cognitive reserve, such as educational level, occupational activities, leisure activities, physical activities and the integrity of relationships network [ 98 , 99 ]. A study conducted by Stern et al. (1994) [ 100 ] indicated that individuals with low level of schooling and low level of professional achievement had an approximately two-fold increased risk of developing dementia. Similarly, another study indicated that individuals with a higher level of leisure activities performance had a lower risk of developing dementia [ 101 ].

Physical activity

A meta-analysis developed by Hamer & Chida (2009) [ 102 ] indicated that physical activity practice is able to reduce AD risk by 45%. This protective effect is related to several mechanisms, such as reduction of blood pressure, obesity and proinflammatory activity besides the improvement in lipid profile and endothelial function. In addition, adaptations that occur in response to exercise can lead to a better cerebral blood flow and, consequently, better oxygenation of important areas for cognitive function [ 102 ]. It is also believed that physical activity is able to prevent AD by increasing neurotrophic factors such as BDNF (Brain Derived Neurotrophic Factor), IGF-1 (Insulin-Like Growth Factor), VEGF (Vascular Endothelial Growth Factor), stimulating neurogenesis and synaptic plasticity; and by the reduction of free radicals in the hippocampus, as well as increase of superoxide dismutase and eNOS (endothelial nitric oxide synthase) [ 103 ]. Studies have shown that the practice of physical activities is capable of promoting an increase in hippocampal volume, in addition to increasing plasma BDNF concentrations in healthy elderly, indicating a possible neuroprotective effect. It was also reported that in the AD elderly, practice of physical activities correlates positively with the levels of BDNF [ 104 ], which is a growth factor associated with the development and survival of neurons and synapses [ 105 ].

The relationship between the effects of diet and the risk of developing AD was based on certain patterns that were associated with lower or higher risk of developing AD [ 106 ]. As an example, Mediterranean diet is rich in unsaturated fats and antioxidants which confers a protection factor, as diets rich in saturated and trans fats and low levels of anti-oxidants are associated with higher risk of developing AD [ 106 , 107 ]. Some dietary components are essential for neurocognition protection such as dietary fatty acids, including fish oil; antioxidants, such as vitamins E and C; fruits and vegetables; vitamins B6, B12 (cobalamine) and folate, in addition to caloric restriction [ 108 ]. Antioxidants are able to prevent damage caused by reactive oxygen species in addition to stabilizing the membranes; docosahexaenoic acid (DHA) helps clear the Aβ peptide and, together with choline and uridine, aid in the synthesis of the neuronal membrane [ 106 ]. Phospholipid composition is essential in neuronal membrane function. Thus, adequate intake of DHA, eicosapentaenoic acid (EPA), uridine monophosphate, choline, folate, vitamins B6, B12, C, and E, and selenium contributes to a better synthesis of phospholipids and, consequently, to synaptic function preservation and against neurodegeneration [ 106 ]. Nerve synapses consist mainly of neuronal membranes, and neuronal and synaptic losses observed in AD have been related to degeneration and alteration in the composition of these membranes [ 109 ]. Brain aging associated with changes in lipid composition is well studied for treatment and prevention purposes with phospholipids such as phosphatidylcholine and phosphatidylserine that could favor cognitive improvement [ 110 ]. The OmegAD study (a set of double-blind, placebo-controlled clinical trials involving AD patients which evaluated the effects of omega-3 fatty acids (n − 3 FAs) daily supplementation in patients with mild to moderate AD) showed that after six months, DHA (1.7 g) and EPA (0.6 g) supplementation demonstrated benefits such as preservation of cognitive performance, increase in plasma and CSF (Cerebrospinal fluid) levels of n − 3 FAs, DHA and EPA (and negative correlation between DHA and total / phosphorylated tau levels in CSF), reduction in cytokine release pro-inflammatory by blood peripheral mononuclear cells (PBMC), modulation in the expression of genes involved in the regulation of inflammation in PBMC, elevation in transthyretin plasma levels (a protein that binds to AB and which may influence its deposition in the brain), and increase in body weight and BMI. However, the literature data do not support the benefits of 3-FA supplementation in preventing cognitive decline in elderly subjects [ 111 ].

Epidemiological studies have observed a relationship between serum levels of vitamin D reduction, especially 25-hydroxyvitamin D, and AD development [ 112 , 113 , 114 ]. Vitamin D is an important steroid hormone that acts on calcium metabolism and bone regulation, and has some functions in central nervous system, such as regulation of neurotrophic factors, calcium homeostasis, acts on oxidative stress mechanisms, immune system modulation and inflammation [ 115 ]. In the case of inflammation, vitamin D deficiency causes an increase in the amyloidogenic pathway due to elevation of BACE1 and APP cleavage and decrease of Aβ degradation [ 116 ]. Briones & Darwish (2012) [ 117 ] reported a BACE1 and Aβ peptide reduction after vitamin D supplementation in elderly rats. It has also been observed that vitamin D acts on macrophages in order to promote clearence of Aβ peptide [ 118 , 119 ]. In AD patients mutations were also observed in vitamin D receptor (VDR) gene, which would favor the onset of the disease [ 120 ]. To date, no large randomized clinical trial has been conducted on the effect of vitamin D supplementation on the cognition of AD patients. However, in smaller or cohort studies, the results of using high doses of vitamin D and cognitive improvement are divergent [ 121 , 122 , 123 , 124 ]. Vitamin D deficiency should be screened and supplemented in the elderly population due to its high prevalence, but this treatment is not specific for cognitive improvement.

Estrogen (hormone replacement therapy)

Estrogen roles in sex organs are well understood, but it has recently been observed that local production of estrogen plays specific roles in tissues in which it is produced, with or without dependence on circulating estrogen [ 125 ]. Estrogen, especially estradiol, is able to prevent mitochondrial dysfunction in nerve cells, neuroinflammation and assist in DNA repair mechanisms [ 126 ], thus presenting neuroprotective effect [ 126 , 127 ]. The results observed in epidemiological studies are inconsistent [ 128 , 129 ]. Some studies have not observed a beneficial effect of hormone replacement therapy, estrogen or combination therapy on the risk of developing AD [ 130 , 131 ]. Other studies reported a beneficial effect on cognition protection in women receiving hormone replacement therapy at different ages after the onset of menopause [ 132 , 133 , 134 , 135 ]. Inconsistent epidemiological findings, in addition to other factors such as increased risk of deep venous thrombosis, hormone replacement therapy is not recommended in order to prevent cognitive decline and AD development [ 136 ].

Other relevant factors and conclusion

The main pathophysiological mechanisms of AD are amyloidosis and tau-related neurodegeneration, and have specific topographical and chronological pathways. For instance, brain amyloidosis starts in neocortical regions and then affects subcortical structures [ 137 ]. On the other hand, neurodegeneration first appear on locus coeruleus and then spreads through transentorrinal area and neocortical regions [ 137 ]. Cognitive and behavioral features of AD are significantly correlated to the topographical distribution of neurofibrillary tangles.

There is great variability in topographical patterns of pathological findings in AD, causing great phenotypical variability [ 7 ], with atypical presentations of the disease [ 138 ]. It is not clear how risk and beneficial factors may modulate the topographical progression of amyloidosis and neurodegeneration.

The effects of modifiable risk factors on cross-sectional cognition have been the target of multiple WRAP (The Wisconsin Registry for Alzheimer’s Prevention) investigations. This study has investigated risk factors for AD in middle age, since this phase of life is less studied in relation to the later stages of aging. However, this is a critical time because it is when the Alzheimer’s pathology begins and thus, when its trajectory can be modified through pharmacological approaches and / or lifestyle changes. Within this context, the WRAP study, reported by Johnson et al. (2018), suggest that social engagement, physical and cognitive activities, glucose regulation, stress and sleep, in addition to cardiovascular and metabolic risks are interventional parameters that may improve brain health and reduce the likelihood and severity of AD pathology. These authors conclude that a good health and a salutary lifestyle are factors associated not only with better cognition and brain structure but also the lower AD pathophysiologic burden [ 139 ].

The studies of genetic risk factors are important to better elucidate the pathophysiological processes in the development of AD. However, such factors are not passible to any intervention until now. Faced to this scenario, modifiable risk factors such as diabetes, hypertension and dyslipidemia and others previously mentioned should be closely monitored to prevent complications favoring cognitive decline or even to improve the quality of life of patients with AD. In this context, it should also be emphasized that factors considered protective, such as physical exercise, diet and cognitive stimuli should be strongly and widely encouraged, so that such theoretically preventive measures can be adopted by the population contributing to reduce risk of this disease. Since no current drug intervention can modify the pathophysiological mechanisms related to the development of this devastating disease, adoption of these measures constitutes an important strategy for clinical management in order to prevent or postpone cognitive decline.

Abbreviations

  • Alzheimer’s disease

Advanced glycation end products

Apolipoprotein E (ApoE)

Amyloid precursor protein

β-amyloid peptide

Brain Derived Neurotrophic Factor

Body Mass Index

Cerebrospinal fluid

Docosahexaenoic acid

Endothelial nitric oxide synthase

Eicosapentaenoic acid

Hypothalamic, pituitary and adrenal axis

Hazard Ratio

Insulin-Like Growth Factor

Omega-3 fatty acids

Neurofibrillary tangles

Peripheral blood mononuclear cells

Phosphorylated tau protein

Vitamin D receptor

Vascular Endothelial Growth Factor

Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer's disease. Lancet. 2011;377(9770):1019–31.

Article   PubMed   Google Scholar  

Ferreira D, Perestelo-Perez L, Westman E, Wahlund LO, Sarria A, Serrano-Aguilar P. Meta-review of CSF Core biomarkers in Alzheimer's disease: the state-of-the-art after the new revised diagnostic criteria. Front Aging Neurosci. 2014;6:47.

PubMed   PubMed Central   Google Scholar  

IDd A, FHdEdM G, Forlenza OV, UdS P, HLd B, et al. Alzheimer disease: correlation between memory and autonomy. Rev psiquiatr clín. 2005;32(3):131–6.

Article   Google Scholar  

Shinohara M, Sato N, Shimamura M, Kurinami H, Hamasaki T, Chatterjee A, et al. Possible modification of Alzheimer's disease by statins in midlife: interactions with genetic and non-genetic risk factors. Front Aging Neurosci. 2014;6:71.

Article   PubMed   PubMed Central   Google Scholar  

Cortes-Canteli M, Paul J, Norris EH, Bronstein R, Ahn HJ, Zamolodchikov D, et al. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer's disease. Neuron. 2010;66(5):695–709.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Bateman RJ, Xiong C, Benzinger TLS, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, Holtzman DM, Santacruz A, Buckles V, Oliver A, Moulder K, Aisen PS, Ghetti B, Klunk WE, McDade E, Martins RN, Masters CL, Mayeux R, Ringman JM, Rossor MN, Schofield PR, Sperling RA, Salloway S, Morris JC. Clinical and Biomarker Changes in Dominantly Inherited Alzheimer's Disease. N Engl J Med. 2012;367:795–804.

Murray ME, Graff-Radford NR, Ross OA, Petersen RC, Duara R, Dickson DW. Neuropathologically defined subtypes of Alzheimer's disease with distinct clinical characteristics: a retrospective study. Lancet Neurol. 2011;10(9):785–96.

de Souza LC, Sarazin M, Teixeira-Junior AL, Caramelli P, Santos AE, Dubois B. Biological markers of Alzheimer's disease. Arq Neuropsiquiatr. 2014;72(3):227–31.

Kling MA, Trojanowski JQ, Wolk DA, Lee VM, Arnold SE. Vascular disease and dementias: paradigm shifts to drive research in new directions. Alzheimers Dement. 2013;9(1):76–92.

Kang S, Lee YH, Lee JE. Metabolism-centric overview of the pathogenesis of Alzheimer's disease. Yonsei Med J. 2017;58(3):479–88.

Mucke L. Neuroscience: Alzheimer's disease. Nature. 2009;461:895–7.

Article   CAS   PubMed   Google Scholar  

Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–6.

Querfurth HW, LaFerla FM. Alzheimer's disease. N Engl J Med. 2010;362(4):329–44.

Muller UC, Deller T, Korte M. Not just amyloid: physiological functions of the amyloid precursor protein family. Nat Rev Neurosci. 2017;18(5):281–98.

Doran E, Keator D, Head E, Phelan MJ, Kim R, Totoiu M, et al. Down syndrome, partial trisomy 21, and absence of Alzheimer's disease: the role of APP. J Alzheimers Dis. 2017;56(2):459–70.

Gupta A, Goyal R. Amyloid beta plaque: a culprit for neurodegeneration. Acta Neurol Belg. 2016;116(4):445–50.

Zhang YW, Thompson R, Zhang H, Xu H. APP processing in Alzheimer's disease. Mol Brain. 2011;4:3.

Zhang H, Ma Q, Zhang YW, Xu H. Proteolytic processing of Alzheimer's beta-amyloid precursor protein. J Neurochem. 2012;120(Suppl 1):9–21.

Martorana A, Di Lorenzo F, Belli L, Sancesario G, Toniolo S, Sallustio F, et al. Cerebrospinal fluid Abeta42 levels: when physiological become pathological state. CNS Neurosci Ther. 2015;21(12):921–5.

Naslund J, Haroutunian V, Mohs R, Davis KL, Davies P, Greengard P, et al. Correlation between elevated levels of amyloid beta-peptide in the brain and cognitive decline. Jama. 2000;283(12):1571–7.

Grimmer T, Riemenschneider M, Forstl H, Henriksen G, Klunk WE, Mathis CA, et al. Beta amyloid in Alzheimer's disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid. Biol Psychiatry. 2009;65(11):927–34.

Tapiola T, Alafuzoff I, Herukka SK, Parkkinen L, Hartikainen P, Soininen H, et al. Cerebrospinal fluid {beta}-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. Arch Neurol. 2009;66(3):382–9.

Blennow K, Hampel H. CSF markers for incipient Alzheimer's disease. Lancet Neurol. 2003;2(10):605–13.

Lee SJ, Nam E, Lee HJ, Savelieff MG, Lim MH. Towards an understanding of amyloid-beta oligomers: characterization, toxicity mechanisms, and inhibitors. Chem Soc Rev. 2017;46(2):310–23.

Blurton-Jones M, Laferla FM. Pathways by which Abeta facilitates tau pathology. Curr Alzheimer Res. 2006;3(5):437–48.

Kovacs GG. Invited review: neuropathology of tauopathies: principles and practice. Neuropathol Appl Neurobiol. 2015;41(1):3–23.

Khan SS, Bloom GS. Tau: the Center of a Signaling Nexus in Alzheimer's disease. Front Neurosci. 2016;10:31.

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(5):362–81.

Mendez MF. Early-onset Alzheimer disease. Neurol Clin. 2017;35(2):263–81.

Giri M, Zhang M, Lu Y. Genes associated with Alzheimer's disease: an overview and current status. Clin Interv Aging. 2016;11:665–81.

Cacace R, Sleegers K, Van Broeckhoven C. Molecular genetics of early-onset Alzheimer's disease revisited. Alzheimers Dement. 2016;12(6):733–48.

Calero M, Gómez-Ramos A, Calero O, Soriano E, Avila J, Medina M. Additional mechanisms conferring genetic susceptibility to Alzheimer’s disease. Front Cell Neurosci. 2015;9:138.

Bekris LM, Yu CE, Bird TD, Tsuang DW. Genetics of Alzheimer disease. J Geriatr Psychiatry Neurol. 2010;23(4):213–27.

Campion D, Dumanchin C, Hannequin D, Dubois B, Belliard S, Puel M, et al. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet. 1999;65:664–70.

Corbo RM, Scacchi R. Apolipoprotein E (APOE) allele distribution in the world. Is APOE*4 a ‘thrifty’ allele? Ann Hum Genet. 1999;63(Pt 4):301–10.

Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry. 2015;77(1):43–51.

Mahley RW. Apolipoprotein E: from cardiovascular disease to neurodegenerative disorders. J Mol Med (Berl). 2016;94:739–46.

Article   CAS   Google Scholar  

Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer’s disease. Neuron. 2009;63(3):287–303.

Hauser PS, Narayanaswami V, Ryan RO. Apolipoprotein E: from lipid transport to neurobiology. Prog Lipid Res. 2011;50(1):62–74.

Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, et al. Variant of TREM2 associated with the risk of Alzheimer's disease. N Engl J Med. 2013;368(2):107–16.

Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al. TREM2 variants in Alzheimer's disease. N Engl J Med. 2013;368(2):117–27.

Allcock RJ, Barrow AD, Forbes S, Beck S, Trowsdale J. The human TREM gene cluster at 6p21.1 encodes both activating and inhibitory single IgV domain receptors and includes NKp44. Eur J Immunol. 2003;33(2):567–77.

Jiang T, Zhang YD, Gao Q, Ou Z, Gong PY, Shi JQ, et al. TREM2 ameliorates neuronal tau pathology through suppression of microglial inflammatory response. Inflammation. 2018;41(3):811–23.

Mecca C, Giambanco I, Donato R, Arcuri C. Microglia and Aging: The Role of the TREM2-DAP12 and CX3CL1-CX3CR1 Axes. Int J Mol Sci. 2018;19(1):318.

Article   CAS   PubMed Central   Google Scholar  

Xiang X, Werner G, Bohrmann B, Liesz A, Mazaheri F, Capell A, et al. TREM2 deficiency reduces the efficacy of immunotherapeutic amyloid clearance. EMBO Mol Med. 2016;8:992–1004.

Mayeux R, Stern Y. Epidemiology of Alzheimer Disease. Cold Spring Harb Perspect Med. 2012;2:a006239.

Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol. 2011;7(3):137–52.

Love S, Miners JS. Cerebrovascular disease in ageing and Alzheimer's disease. Acta Neuropathol. 2016;131(5):645–58.

Liu W, Wong A, Law AC, Mok VC. Cerebrovascular disease, amyloid plaques, and dementia. Stroke. 2015;46(5):1402–7.

Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12(12):723–38.

Skoog I, Lernfelt B, Landahl S, Palmertz B, Andreasson LA, Nilsson L, et al. 15-year longitudinal study of blood pressure and dementia. Lancet. 1996;347(9009):1141–5.

Staessen JA, Richart T, Birkenhager WH. Less atherosclerosis and lower blood pressure for a meaningful life perspective with more brain. Hypertension. 2007;49(3):389–400.

Skoog I, Gustafson D. Update on hypertension and Alzheimer's disease. Neurol Res. 2006;28(6):605–11.

Li X, Song D, Leng SX. Link between type 2 diabetes and Alzheimer’s disease: from epidemiology to mechanism and treatment. Clin Interv Aging. 2015;10:549–60.

Kimura N. Diabetes mellitus induces Alzheimer's disease pathology: histopathological evidence from animal models. Int J Mol Sci. 2016;17(4):503.

Ko SY, Ko HA, Chu KH, Shieh TM, Chi TC, Chen HI, et al. The Possible Mechanism of Advanced Glycation End Products (AGEs) for Alzheimer’s Disease. PLoS One. 2015;10.

Li XH, Du LL, Cheng XS, Jiang X, Zhang Y, Lv BL, et al. Glycation exacerbates the neuronal toxicity of beta-amyloid. Cell Death Dis. 2013;4:e673.

Profenno LA, Porsteinsson AP, Faraone SV. Meta-analysis of Alzheimer's disease risk with obesity, diabetes, and related disorders. Biol Psychiatry. 2010;67(6):505–12.

Fitzpatrick AL, Kuller LH, Lopez OL, Diehr P, O'Meara ES, Longstreth WT Jr, et al. Midlife and late-life obesity and the risk of dementia: cardiovascular health study. Arch Neurol. 2009;66(3):336–42.

Anstey KJ, Cherbuin N, Budge M, Young J. Body mass index in midlife and late-life as a risk factor for dementia: a meta-analysis of prospective studies. Obes Rev. 2011;12(5):e426–37.

Popp J, Meichsner S, Kolsch H, Lewczuk P, Maier W, Kornhuber J, et al. Cerebral and extracerebral cholesterol metabolism and CSF markers of Alzheimer's disease. Biochem Pharmacol. 2013;86(1):37–42.

Xue-shan Z, Juan P, Qi W, Zhong R, Li-hong P, Zhi-han T, et al. Imbalanced cholesterol metabolism in Alzheimer's disease. Clin Chim Acta. 2016;456:107–14.

Ricciarelli R, Canepa E, Marengo B, Marinari UM, Poli G, Pronzato MA, et al. Cholesterol and Alzheimer's disease: a still poorly understood correlation. IUBMB Life. 2012;64(12):931–5.

Ullrich C, Pirchl M, Humpel C. Hypercholesterolemia in rats impairs the cholinergic system and leads to memory deficits. Mol Cell Neurosci. 2010;45:408–17.

Hendrie HC, Hake A, Lane K, Purnell C, Unverzagt F, Smith-Gamble V, et al. Statin use, incident dementia and Alzheimer disease in elderly African Americans. Ethn Dis. 2015;25(3):345–54.

Haag MD, Hofman A, Koudstaal PJ, Stricker BH, Breteler MM. Statins are associated with a reduced risk of Alzheimer disease regardless of lipophilicity. The Rotterdam study. J Neurol Neurosurg Psychiatry. 2009;80(1):13–7.

Lin FC, Chuang YS, Hsieh HM, Lee TC, Chiu KF, Liu CK, et al. Early statin use and the progression of Alzheimer disease: a Total population-based case-control study. Medicine (Baltimore). 2015;94(47):e2143.

Simons M, Schwarzler F, Lutjohann D, von Bergmann K, Beyreuther K, Dichgans J, et al. Treatment with simvastatin in normocholesterolemic patients with Alzheimer's disease: a 26-week randomized, placebo-controlled, double-blind trial. Ann Neurol. 2002;52(3):346–50.

Sano M, Bell KL, Galasko D, Galvin JE, Thomas RG, van Dyck CH, et al. A randomized, double-blind, placebo-controlled trial of simvastatin to treat Alzheimer disease. Neurology. 2011;77(6):556–63.

Feldman HH, Doody RS, Kivipelto M, Sparks DL, Waters DD, Jones RW, et al. Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease: LEADe. Neurology. 2010;74(12):956–64.

Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe SM, et al. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet. 2002;360(9346):1623–30.

Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):7–22.

Song Y, Nie H, Xu Y, Zhang L, Wu Y. Association of statin use with risk of dementia: a meta-analysis of prospective cohort studies. Geriatr Gerontol Int. 2013;13(4):817–24.

McGuinness B, Craig D, Bullock R, Passmore P. Statins for the prevention of dementia. Cochrane Database Syst Rev. 2016;(1):Cd003160. https://doi.org/10.1002/14651858.CD003160.pub3 .

Håkansson K, Rovio S, Helkala EL, Vilska AR, Winblad B, Soininen H, et al. Association between mid-life marital status and cognitive function in later life: population based cohort study. BMJ. 2009;339:b2462

Fan LY, Sun Y, Lee HJ, Yang SC, Chen TF, Lin KN, et al. Marital status, lifestyle and dementia: a Nationwide survey in Taiwan. PLoS One. 2015;10(9):e0139154.

Sommerlad A, Ruegger J, Singh-Manoux A, Lewis G, Livingston G. Marriage and risk of dementia: systematic review and meta-analysis of observational studies. J Neurol Neurosurg Psychiatry. 2018;89(3):231–8.

Ray B, Gaskins DL, Sajdyk TJ, Spence JP, Fitz SD, Shekhar A, et al. Restraint stress and repeated corticotrophin-releasing factor receptor activation in the amygdala both increase amyloid-beta precursor protein and amyloid-beta peptide but have divergent effects on brain-derived neurotrophic factor and pre-synaptic proteins in the prefrontal cortex of rats. Neuroscience. 2011;184:139–50.

Lesuis SL, Maurin H, Borghgraef P, Lucassen PJ, Van Leuven F, Krugers HJ. Positive and negative early life experiences differentially modulate long term survival and amyloid protein levels in a mouse model of Alzheimer's disease. Oncotarget. 2016;7(26):39118–35.

Justice NJ, Huang L, Tian JB, Cole A, Pruski M, Hunt AJ Jr, et al. Posttraumatic stress disorder-like induction elevates beta-amyloid levels, which directly activates corticotropin-releasing factor neurons to exacerbate stress responses. J Neurosci. 2015;35(6):2612–23.

Carroll JC, Iba M, Bangasser DA, Valentino RJ, James MJ, Brunden KR, et al. Chronic stress exacerbates tau pathology, neurodegeneration, and cognitive performance through a corticotropin-releasing factor receptor-dependent mechanism in a transgenic mouse model of tauopathy. J Neurosci. 2011;31(40):14436–49.

Lara VP, Caramelli P, Teixeira AL, Barbosa MT, Carmona KC, Carvalho MG, et al. High cortisol levels are associated with cognitive impairment no-dementia (CIND) and dementia. Clin Chim Acta. 2013;423:18–22.

Zverova M, Fisar Z, Jirak R, Kitzlerova E, Hroudova J, Raboch J. Plasma cortisol in Alzheimer's disease with or without depressive symptoms. Med Sci Monit. 2013;19:681–9.

Wang LY, Raskind MA, Wilkinson CW, Shofer JB, Sikkema C, Szot P, et al. Associations between CSF cortisol and CSF norepinephrine in cognitively normal controls and patients with amnestic MCI and AD dementia. Int J Geriatr Psychiatry. 2018;33(5):763–8.

Huang CW, Lui CC, Chang WN, Lu CH, Wang YL, Chang CC. Elevated basal cortisol level predicts lower hippocampal volume and cognitive decline in Alzheimer's disease. J Clin Neurosci. 2009;16(10):1283–6.

Toledo JB, Toledo E, Weiner MW, Jack CR, Jagust W, Lee VMY, et al. Cardiovascular risk factors, cortisol, and amyloid-β deposition in Alzheimer’s Disease Neuroimaging Initiative. Alzheimers Dement. 2012;8(6):483–9.

Ennis GE, An Y, Resnick SM, Ferrucci L, O'Brien RJ, Moffat SD. Long-term cortisol measures predict Alzheimer disease risk. Neurology. 2017;88:371–8.

Schrijvers EM, Direk N, Koudstaal PJ, Kirschbaum C, Hofman A, Tiemeier H, et al. Associations of serum cortisol with cognitive function and dementia: the Rotterdam study. J Alzheimers Dis. 2011;25(4):671–7.

Byers AL, Yaffe K. Depression and risk of developing dementia. Nat Rev Neurol. 2011;7(6):323–31.

Ricci S, Fuso A, Ippoliti F, Businaro R. Stress-induced cytokines and neuronal dysfunction in Alzheimer's disease. J Alzheimers Dis. 2012;28(1):11–24.

Vilalta-Franch J, Lopez-Pousa S, Llinas-Regla J, Calvo-Perxas L, Merino-Aguado J, Garre-Olmo J. Depression subtypes and 5-year risk of dementia and Alzheimer disease in patients aged 70 years. Int J Geriatr Psychiatry. 2013;28(4):341–50.

Wu KY, Lin KJ, Chen CH, Chen CS, Liu CY, Huang SY, et al. Diversity of neurodegenerative pathophysiology in nondemented patients with major depressive disorder: Evidence of cerebral amyloidosis and hippocampal atrophy. Brain Behav. 2018;8(7):e01016.

Proserpio P, Arnaldi D, Nobili F, Nobili L. Integrating sleep and Alzheimer's disease pathophysiology: hints for sleep disorders management. J Alzheimers Dis. 2018;63(3):871–86.

Shi L, Chen SJ, Ma MY, Bao YP, Han Y, Wang YM, et al. Sleep disturbances increase the risk of dementia: a systematic review and meta-analysis. Sleep Med Rev. 2018;40:4–16.

Traber MG, van der Vliet A, Reznick AZ, Cross CE. Tobacco-related diseases. Is there a role for antioxidant micronutrient supplementation? Clin Chest Med. 2000;21(1):173–87 x.

Durazzo TC, Mattsson N, Weiner MW. Smoking and increased Alzheimer's disease risk: a review of potential mechanisms. Alzheimers Dement. 2014;10(3 Suppl):S122–45.

Cataldo JK, Prochaska JJ, Glantz SA. Cigarette smoking is a risk factor for Alzheimer's disease: an analysis controlling for tobacco industry affiliation. J Alzheimers Dis. 2010;19(2):465–80.

Xu W, Yu JT, Tan MS, Tan L. Cognitive reserve and Alzheimer's disease. Mol Neurobiol. 2015;51(1):187–208.

Tucker AM, Stern Y. Cognitive reserve in aging. Curr Alzheimer Res. 2011;8(4):354–60.

Stern Y, Gurland B, Tatemichi TK, Tang MX, Wilder D, Mayeux R. Influence of education and occupation on the incidence of Alzheimer's disease. Jama. 1994;271(13):1004–10.

Scarmeas N, Levy G, Tang MX, Manly J, Stern Y. Influence of leisure activity on the incidence of Alzheimer’s disease. Neurology. 2001;57(12):2236–42.

Hamer M, Chida Y. Physical activity and risk of neurodegenerative disease: a systematic review of prospective evidence. Psychol Med. 2009;39(1):3–11.

Mendiola-Precoma J, Berumen LC, Padilla K, Garcia-Alcocer G. Therapies for prevention and treatment of Alzheimer's disease. Biomed Res Int. 2016;2016:2589276.

Paillard T, Rolland Y, de Souto Barreto P. Protective effects of physical exercise in Alzheimer's disease and Parkinson's disease: a narrative review. J Clin Neurol. 2015;11(3):212–9.

Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function*. Annu Rev Neurosci. 2001;24:677–736.

de Wilde MC, Vellas B, Girault E, Yavuz AC, Sijben JW. Lower brain and blood nutrient status in Alzheimer's disease: Results from meta-analyses. Alzheimers Dement (N Y). 2017;3:416–31.

Google Scholar  

Scarmeas N, Stern Y, Mayeux R, Manly JJ, Schupf N, Luchsinger JA. Mediterranean diet and mild cognitive impairment. Arch Neurol. 2009;66(2):216–25.

Smith PJ, Blumenthal JA. Diet and Neurocognition: review of evidence and methodological considerations. Curr Aging Sci. 2010;3(1):57–66.

van Wijk N, Broersen LM, de Wilde MC, Hageman RJ, Groenendijk M, Sijben JW, et al. Targeting synaptic dysfunction in Alzheimer's disease by administering a specific nutrient combination. J Alzheimers Dis. 2014;38(3):459–79.

Olivera-Pueyo J, Pelegrin-Valero C. Dietary supplements for cognitive impairment. Actas Esp Psiquiatr. 2017;45(Supplement):37–47.

PubMed   Google Scholar  

Fraga VG, Carvalho MDG, Caramelli P, de Sousa LP, Gomes KB. Resolution of inflammation, n-3 fatty acid supplementation and Alzheimer disease: a narrative review. J Neuroimmunol. 2017;310:111–9.

Knekt P, Saaksjarvi K, Jarvinen R, Marniemi J, Mannisto S, Kanerva N, et al. Serum 25-hydroxyvitamin d concentration and risk of dementia. Epidemiology. 2014;25(6):799–804.

Shen L, Ji HF. Vitamin D deficiency is associated with increased risk of Alzheimer's disease and dementia: evidence from meta-analysis. Nutr J. 2015;14:76.

Licher S, de Bruijn R, Wolters FJ, Zillikens MC, Ikram MA, Ikram MK. Vitamin D and the risk of dementia: the Rotterdam study. J Alzheimers Dis. 2017;60(3):989–97.

Landel V, Annweiler C, Millet P, Morello M, Féron F. Vitamin D, Cognition and Alzheimer’s Disease: The Therapeutic Benefit is in the D-Tails. J Alzheimers Dis. 2016;53:419–44.

Grimm MOW, Thiel A, Lauer AA, Winkler J, Lehmann J, Regner L, et al. Vitamin D and Its Analogues Decrease Amyloid-β (Aβ) Formation and Increase Aβ-Degradation. Int J Mol Sci. 2017;18(12):2764.

Briones TL, Darwish H. Vitamin D mitigates age-related cognitive decline through the modulation of pro-inflammatory state and decrease in amyloid burden. J Neuroinflammation. 2012;9:244.

Mizwicki MT, Menegaz D, Zhang J, Barrientos-Duran A, Tse S, Cashman JR, et al. Genomic and nongenomic signaling induced by 1alpha,25(OH)2-vitamin D3 promotes the recovery of amyloid-beta phagocytosis by Alzheimer's disease macrophages. J Alzheimers Dis. 2012;29(1):51–62.

Masoumi A, Goldenson B, Ghirmai S, Avagyan H, Zaghi J, Abel K, et al. 1alpha,25-dihydroxyvitamin D3 interacts with curcuminoids to stimulate amyloid-beta clearance by macrophages of Alzheimer's disease patients. J Alzheimers Dis. 2009;17(3):703–17.

Gezen-Ak D, Dursun E, Bilgic B, Hanagasi H, Ertan T, Gurvit H, et al. Vitamin D receptor gene haplotype is associated with late-onset Alzheimer's disease. Tohoku J Exp Med. 2012;228(3):189–96.

Annweiler C, Herrmann FR, Fantino B, Brugg B, Beauchet O. Effectiveness of the combination of memantine plus vitamin D on cognition in patients with Alzheimer disease: a pre-post pilot study. Cogn Behav Neurol. 2012;25(3):121–7.

Annweiler C, Rolland Y, Schott AM, Blain H, Vellas B, Herrmann FR, et al. Higher vitamin D dietary intake is associated with lower risk of alzheimer's disease: a 7-year follow-up. J Gerontol A Biol Sci Med Sci. 2012;67(11):1205–11.

Stein MS, Scherer SC, Ladd KS, Harrison LC. A randomized controlled trial of high-dose vitamin D2 followed by intranasal insulin in Alzheimer's disease. J Alzheimers Dis. 2011;26(3):477–84.

Miller BJ, Whisner CM, Johnston CS. Vitamin D supplementation appears to increase plasma Abeta40 in vitamin D insufficient older adults: a pilot randomized controlled trial. J Alzheimers Dis. 2016;52(3):843–7.

Li R, Cui J, Shen Y. Brain sex matters: estrogen in cognition and Alzheimer’s disease. Mol Cell Endocrinol. 2014;389(0):13–21.

Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci. 2017;9:430.

Depypere H, Vierin A, Weyers S, Sieben A. Alzheimer's disease, apolipoprotein E and hormone replacement therapy. Maturitas. 2016;94:98–105.

Brinton RD. Investigative models for determining hormone therapy-induced outcomes in brain: evidence in support of a healthy cell bias of estrogen action. Ann N Y Acad Sci. 2005;1052:57–74.

Dye RV, Miller KJ, Singer EJ, Levine AJ. Hormone replacement therapy and risk for neurodegenerative diseases. Int J Alzheimers Dis. 2012;2012:258454.

Imtiaz B, Taipale H, Tanskanen A, Tiihonen M, Kivipelto M, Heikkinen AM, et al. Risk of Alzheimer's disease among users of postmenopausal hormone therapy: a nationwide case-control study. Maturitas. 2017;98:7–13.

Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ockene JK, et al. Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women's Health Initiative memory study: a randomized controlled trial. Jama. 2003;289(20):2651–62.

Wharton W, Baker LD, Gleason CE, Dowling M, Barnet JH, Johnson S, et al. Short-term hormone therapy with transdermal estradiol improves cognition for postmenopausal women with Alzheimer's disease: results of a randomized controlled trial. J Alzheimers Dis. 2011;26(3):495–505.

Henderson VW, Benke KS, Green RC, Cupples LA, Farrer LA. Postmenopausal hormone therapy and Alzheimer's disease risk: interaction with age. J Neurol Neurosurg Psychiatry. 2005;76(1):103–5.

Espeland MA, Rapp SR, Manson JE, Goveas JS, Shumaker SA, Hayden KM, et al. Long-term effects on cognitive trajectories of postmenopausal hormone therapy in two age groups. J Gerontol A Biol Sci Med Sci. 2017;72(6):838–45.

Fox M, Berzuini C, Knapp LA. Cumulative estrogen exposure, number of menstrual cycles, and Alzheimer's risk in a cohort of British women. Psychoneuroendocrinology. 2013;38(12):2973–82.

Marjoribanks J, Farquhar C, Roberts H, Lethaby A, Lee J. Long-term hormone therapy for perimenopausal and postmenopausal women. Cochrane Database Syst Rev. 2017;1:Cd004143.

Mattei TA. Is it all about contact? Neurodegeneration as a “protein freeze tag game” inside the central nervous system. Front Neurol. 2013;4:75.

Warren JD, Fletcher PD, Golden HL. The paradox of syndromic diversity in Alzheimer disease. Nat Rev Neurol. 2012;8(8):451–64.

Johnson SC, Koscik RL, Jonaitis EM, Clark LR, Mueller KD, Berman SE, et al. The Wisconsin registry for Alzheimer's prevention: a review of findings and current directions. Alzheimers Dement (Amst). 2018;10:130–42.

Download references

Acknowledgements

Not applicable

CNPq, CAPES and Fapemig

Availability of data and materials

Author information, authors and affiliations.

Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 – Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil

Marcos Vinícius Ferreira Silva, Cristina de Mello Gomide Loures, Luan Carlos Vieira Alves, Karina Braga Gomes Borges & Maria das Graças Carvalho

Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, Minas Gerais, 30130-100, Brazil

Leonardo Cruz de Souza

You can also search for this author in PubMed   Google Scholar

Contributions

MVFS, CGML and LCVA drafted the manuscript. LCS, KBGB and MGC reviewed the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Marcos Vinícius Ferreira Silva .

Ethics declarations

Ethics approval and consent to participate, consent for publication, competing interests.

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/ ), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Cite this article.

Silva, M.V.F., Loures, C.d.M.G., Alves, L.C.V. et al. Alzheimer’s disease: risk factors and potentially protective measures. J Biomed Sci 26 , 33 (2019). https://doi.org/10.1186/s12929-019-0524-y

Download citation

Received : 19 January 2019

Accepted : 18 April 2019

Published : 09 May 2019

DOI : https://doi.org/10.1186/s12929-019-0524-y

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Journal of Biomedical Science

ISSN: 1423-0127

  • Submission enquiries: Access here and click Contact Us
  • General enquiries: [email protected]

how to do a research paper on alzheimer's disease

A comprehensive research setup for monitoring Alzheimer’s disease using EEG, fNIRS, and Gait analysis

  • Original Article
  • Published: 09 August 2023
  • Volume 14 , pages 13–21, ( 2024 )

Cite this article

how to do a research paper on alzheimer's disease

  • Minhee Kim 1 ,
  • Sehyeon Jang 2 ,
  • Donjung Lee 3 ,
  • Seungchan Lee 4 ,
  • Jeonghwan Gwak 5 ,
  • Sung Chan Jun 2 &
  • Jae Gwan Kim   ORCID: orcid.org/0000-0002-1010-7712 1  

631 Accesses

7 Altmetric

Explore all metrics

Alzheimer’s disease (AD) has a detrimental impact on brain function, affecting various aspects such as cognition, memory, language, and motor skills. Previous research has dominantly used electroencephalography (EEG) and functional near-infrared spectroscopy (fNIRS) to individually measure brain signals or combine the two methods to target specific brain functions. However, comprehending Alzheimer’s disease requires monitoring various brain functions rather than focusing on a single function. This paper presents a comprehensive research setup for a monitoring platform for AD. The platform incorporates a 32-channel dry electrode EEG, a custom-built four-channel fNIRS, and gait monitoring using a depth camera and pressure sensor. Various tasks are employed to target multiple brain functions. The paper introduced the detailed instrumentation of the fNIRS system, which measures the prefrontal cortex, outlines the experimental design targeting various brain functioning programmed in BCI2000 for visualizing EEG signals synchronized with experimental stimulation, and describes the gait monitoring hardware and software and protocol design. The ultimate goal of this platform is to develop an easy-to-perform brain and gait monitoring method for elderly individuals and patients with Alzheimer’s disease.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price includes VAT (Russian Federation)

Instant access to the full article PDF.

Rent this article via DeepDyve

Institutional subscriptions

how to do a research paper on alzheimer's disease

Similar content being viewed by others

how to do a research paper on alzheimer's disease

Advancements in Measuring Cognition Using EEG and fNIRS

how to do a research paper on alzheimer's disease

Emerging Non-invasive Brain–Computer Interface Technologies and Their Clinical Applications

Mf F. “Mini–mental state’’ A practical method for grading the cognitive state of patients for the clinician. J Psychiatr res. 1975;12(3):189.

Google Scholar  

Kang Y, Na D, Hahn S. Seoul neuropsychological screening battery. Incheon: Human brain research & consulting co. 2003.

Devenney E, Hodges JR. The mini–mental state examination: pitfalls and limitations. Pract Neurol. 2017;17(1):79–80.

Article   Google Scholar  

Carnero-Pardo C. Should the mini–mental state examination be retired? Neurol (Engl Ed). 2014;29(8):473–81.

Han C, Jo SA, Jo I, Kim E, Park MH, Kang Y. An adaptation of the Korean mini–mental state examination (K-MMSE) in elderly Koreans: demographic influence and population-based norms (the AGE study). Arch Gerontol Geriatr. 2008;47(3):302–10.

Delpy DT, Cope M, van der Zee P, Arridge S, Wray S, Wyatt J. Estimation of optical pathlength through tissue from direct time of flight measurement. Phys Med Biol. 1988;33(12):1433.

Kim M, Nguyen T, Gwak J, Lee JJ, Choi KY, Lee KH, et al. Investigation of cerebral hemodynamic changes in mild cognitive impairment due to Alzheimer’s disease during a verbal fluency task. In: 7th international conference on the development of biomedical engineering in Vietnam (BME7) translational health science and technology for developing countries 7. Springer; 2020. pp. 379–382.

Nguyen T, Kim M, Gwak J, Lee JJ, Choi KY, Lee KH, et al. Investigation of brain functional connectivity in patients with mild cognitive impairment: a functional near-infrared spectroscopy (fNIRS) study. J Biophoton. 2019;12(9): e201800298.

Ho TKK, Kim M, Jeon Y, Kim BC, Kim JG, Lee KH, et al. Deep learning-based multilevel classification of Alzheimer’s disease using non-invasive functional near-infrared spectroscopy. Front Aging Neurosci. 2022;14:810125.

Park J, Jang S, Gwak J, Kim BC, Lee JJ, Choi KY, et al. Individualized diagnosis of preclinical Alzheimer’s disease using deep neural networks. Exp Syst Appl. 2022;210: 118511.

Jeon Y, Kang J, Kim BC, Lee KH, Song JI, Gwak J. Early Alzheimer’s disease diagnosis using wearable sensors and multilevel gait assessment: a machine learning ensemble approach. IEEE Sens J. 2023;23(9):10041–53.

Mccarthy G, Luby M, Gore J, Goldman-Rakic P. Infrequent events transiently activate human prefrontal and parietal cortex as measured by functional MRI. J Neurophysiol. 1997;77(3):1630–4.

Proulx N, Samadani AA, Chau T. Quantifying fast optical signal and event-related potential relationships during a visual oddball task. Neuroimage. 2018;178:119–28.

Fraga FJ, Mamani GQ, Johns E, Tavares G, Falk TH, Phillips NA. Early diagnosis of mild cognitive impairment and Alzheimer’s with event-related potentials and event-related desynchronization in N-back working memory tasks. Comput Meth Progr Biomed. 2018;164:1–13.

Herff C, Heger D, Fortmann O, Hennrich J, Putze F, Schultz T. Mental workload during n-back task-quantified in the prefrontal cortex using fNIRS. Front Human Neurosci. 2014;7:935.

Yang D, Hong KS, Yoo SH, Kim CS. Evaluation of neural degeneration biomarkers in the prefrontal cortex for early identification of patients with mild cognitive impairment: an fNIRS study. Front Human Neurosci. 2019;13:317.

Heinzel S, Metzger FG, Ehlis AC, Korell R, Alboji A, Haeussinger FB, et al. Aging-related cortical reorganization of verbal fluency processing: a functional near-infrared spectroscopy study. Neurobiol Aging. 2013;34(2):439–50.

Yeung MK, Sze SL, Woo J, Kwok T, Shum DH, Yu R, et al. Altered frontal lateralization underlies the category fluency deficits in older adults with mild cognitive impairment: a near-infrared spectroscopy study. Front Aging Neurosci. 2016;8:59.

Arai H, Takano M, Miyakawa K, Ota T, Takahashi T, Asaka H, et al. A quantitative near-infrared spectroscopy study: a decrease in cerebral hemoglobin oxygenation in Alzheimer’s disease and mild cognitive impairment. Brain and cognition. 2006;61(2):189–94.

Tang TB, Chan YL, Functional connectivity analysis on mild Alzheimer’s disease, mild cognitive impairment and normal aging using fNIRS. In: 40th annual international conference of the IEEE engineering in medicine and biology society (EMBC). IEEE. 2018;2018, pp. 17–20

Yap KH, Ung WC, Ebenezer EG, Nordin N, Chin PS, Sugathan S, et al. Visualizing hyperactivation in neurodegeneration based on prefrontal oxygenation: a comparative study of mild Alzheimer’s disease, mild cognitive impairment, and healthy controls. Front Aging Neurosci. 2017;9:287.

Brickman AM, Paul RH, Cohen RA, Williams LM, MacGregor KL, Jefferson AL, et al. Category and letter verbal fluency across the adult lifespan: relationship to EEG theta power. Arch Clin Neuropsychol. 2005;20(5):561–73.

Gras LZ, Kanaan SF, McDowd JM, Colgrove YM, Burns J, Pohl PS. Balance and gait of adults with very mild Alzheimer’s disease. J Geriat Phys Ther (2001). 2015;38(1):1.

Li R, Nguyen T, Potter T, Zhang Y. Dynamic cortical connectivity alterations associated with Alzheimer’s disease: an EEG and fNIRS integration study. NeuroImage: Clin. 2019;21:101622.

Cicalese PA, Li R, Ahmadi MB, Wang C, Francis JT, Selvaraj S, et al. An EEG–fNIRS hybridization technique in the four-class classification of Alzheimer’s disease. J Neurosci Meth. 2020;336:108618.

Perpetuini D, Chiarelli AM, Filippini C, Cardone D, Croce P, Rotunno L, et al. Working memory decline in Alzheimer’s disease is detected by complexity analysis of multimodal EEG–fNIRS. Entropy. 2020;22(12):1380.

Chiarelli AM, Perpetuini D, Croce P, Filippini C, Cardone D, Rotunno L, et al. Evidence of neurovascular un-coupling in mild Alzheimer’s disease through multimodal EEG–fNIRS and multivariate analysis of resting-state data. Biomedicines. 2021;9(4):337.

Download references

Acknowledgements

This work was supported by Brain Research Program, National Research Foundation of Korea (NRF-2016M3C7A1905475) and Healthcare AI Convergence Research & Development Program through the National IT Industry Promotion Agency of Korea (NIPA) funded by the Ministry of Science and ICT (No. S1601-20-1016)

Author information

Authors and affiliations.

Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea

Minhee Kim & Jae Gwan Kim

School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea

Sehyeon Jang & Sung Chan Jun

Korea Photonics Technology Institute, Gwangju, 61007, Republic of Korea

Donjung Lee

Department of Medical Device, Korea Institute of Machinery & Materials, Daegu, 42994, Republic of Korea

Seungchan Lee

Department of Software, Korea National University of Transportation, Chungju, 27469, Republic of Korea

Jeonghwan Gwak

You can also search for this author in PubMed   Google Scholar

Contributions

Conceptualization: JGK, SCJ, JG; Methodology: MK, SJ, DL, SL; Formal analysis and investigation: MK, SJ; Writing–original draft preparation: MK; Writing—review and editing: MK, JGK; Funding acquisition: JGK, SCJ, JG; Supervision: JGK, SCJ.

Corresponding author

Correspondence to Jae Gwan Kim .

Ethics declarations

Conflict of interest.

The authors declare that there’s no conflict of interest related to this study.

Ethical approval

This study was approved by the Institutional Review Board at the Gwangju Institute of Science and Technology.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file 1 (png 104 KB)

Supplementary file 2 (png 941 kb), supplementary file 3 (657 kb), supplementary file 4 (pdf 1637 kb), rights and permissions.

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Kim, M., Jang, S., Lee, D. et al. A comprehensive research setup for monitoring Alzheimer’s disease using EEG, fNIRS, and Gait analysis. Biomed. Eng. Lett. 14 , 13–21 (2024). https://doi.org/10.1007/s13534-023-00306-7

Download citation

Received : 03 April 2023

Revised : 10 June 2023

Accepted : 12 July 2023

Published : 09 August 2023

Issue Date : January 2024

DOI : https://doi.org/10.1007/s13534-023-00306-7

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Brain monitoring system
  • Electroencephalogram
  • Functional near-infrared spectroscopy
  • Gait monitoring
  • Alzheimer’s disease
  • Find a journal
  • Publish with us
  • Track your research

Masks Strongly Recommended but Not Required in Maryland, Starting Immediately

Due to the downward trend in respiratory viruses in Maryland, masking is no longer required but remains strongly recommended in Johns Hopkins Medicine clinical locations in Maryland. Read more .

  • Vaccines  
  • Masking Guidelines
  • Visitor Guidelines  

News & Publications

Research aims to uncover the mysteries of alzheimer’s disease.

Physicians often detect early warning signs of dementia in their aging patients from the stories shared by a patient’s family members. They learn about situations that are out of character, such as paying a bill multiple times, misplacing keys and repeating stories. Often a patient has become more irritable and anxious.

When the clinical examination also suggests that a longtime patient’s behavior or disposition has changed, the next step is to rule out possible causes such as medication issues, poor sleep or traumatic life events, according to Esther Oh , co-director of the Johns Hopkins Memory and Alzheimer’s Treatment Center.

“When there’s clearly some functional impairment going on, I try to figure out why,” she says. “How older patients experience cognitive problems is complex.”

If physicians suspect a patient may have mild cognitive impairment due to Alzheimer’s disease or a related brain disease, they can order a variety of labs including blood tests, MRIs and CT scans, and may refer the patient to Oh and her colleagues in psychiatry, geriatrics or neurology at the Memory and Alzheimer’s Treatment Center.

There, patients can receive individualized treatment based on their needs, including medications targeting memory and cognition and treatments for mood, behavioral and sleep changes. Additionally, patients, caregivers and family members can get guidance and support that helps improve their quality of life, plan for the future and manage inevitable crises.

Much is still unknown about the disease. At this time, there’s no cure for Alzheimer’s, no proven way of slowing down its progression and no treatment available to reverse the deterioration that occurs in the brain.

Best practices for managing the disease include physical and social activity, healthy lifestyle and diet, and a well-structured environment. Older adults who engage in these behaviors appear to have less risk of cognitive and functional decline.

While the diagnosis is grim, Johns Hopkins clinicians and researchers are broadening Alzheimer’s and dementia research and expanding treatment options in a number of ways: searching for biological markers (biomarkers) that could predict Alzheimer’s; determining how to target certain proteins that are present in the brains of patients with the disease; defining the different kinds of Alzheimer’s to tailor future treatment and research; developing new drugs; and piloting a home-based care program.

Dome Ad

Defining and Refining

Dementia is a general term that refers to memory loss and decline of other cognitive abilities that limit independence in day-to-day function. Alzheimer’s is the most common brain disease that causes dementia among older adults, accounting for 60%–80% of cases. It affects an estimated one in nine people age 65 and older — 6.2 million Americans. This number is projected to grow to 12.7 million by 2050, according to the Alzheimer’s Association.

Almost two-thirds of the cases are in women, and people of color are at a higher risk of developing Alzheimer’s.

Alzheimer’s is a progressive, neurodegenerative disease that occurs when nerve cells in the brain die. It affects memory, thinking and behavior. But, unlike other forms of dementia, it does not affect patients’ motor function until late stages of the disease.

Alzheimer’s experts think individuals may experience different versions of the disease.

“It’s probably not one kind of Alzheimer’s disease, it’s probably many,” says psychiatrist Paul Rosenberg , co-director of the Johns Hopkins Memory and Alzheimer’s Treatment Center. “What we want to do is find the subtypes so we can find better treatments.”

Cancer treatment, for example, is specific to the kind and subtype of cancer. Breast cancer is treated differently from colon cancer, and within breast cancer, different subtypes mean different treatments. This is the direction in which Johns Hopkins researchers hope to move Alzheimer’s treatment.

Rosenberg and his colleagues, including Memory and Alzheimer’s Treatment Center director Kostas Lyketsos , are crunching data in the Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease to do just that. The goal is to find characteristics that can allow physicians to predict which patients will develop Alzheimer’s, as well as determine what clinical data is necessary to differentiate subtypes of the disease.

The Precision Medicine Center’s patient registry includes more than 130,000 medical records that researchers hope can help define clinical subgroups of patients with dementia, determine when symptoms first develop and when diagnoses occur, among other factors. Additionally, center researchers are developing a collection of unique blood biomarkers that could help target future treatments to subgroups of patients.

Other projects include:

  • Analyzing hundreds of MRIs from patients with dementia to look for variations in the size of different structures that could indicate subgroups.
  • Studying changes in biomarkers over time to try to measure the progression of dementia.
  • Using person-specific stem cells — which are made using a person’s own blood — to create different brain cells in the lab with the potential to predict response to specific medications that may have a role in improving cognitive decline.
  • Treating brain vascular disease — an important contributor to dementia — by repurposing an existing drug, atorvastatin, which is typically used to lower cholesterol. Researchers are studying the drug’s effect on the brain’s circulatory system using a new MRI technique.

Infographic of a brain and alzheimers facts. Left side: 1 in 9 Americans age 65 and older has Alzheimer’s disease.Right side: 6.2 million Americans age 65 and older have Alzheimer’s. The number is projected to reach 12.7 million by 2050.

New Drugs in Development

Other Johns Hopkins researchers are testing a drug that has the potential to slow the progression of Alzheimer’s. Marilyn Albert , director of the Johns Hopkins Alzheimer’s Disease Research Center , Michela Gallagher , a professor of neuroscience at the Johns Hopkins University School of Medicine, and Arnold Bakker , director of the Johns Hopkins Psychiatric Neuroimaging Core , hope to receive FDA approval for the drug for patients in the earliest stages of the disease.

The drug targets two proteins: tau, which serves many functions in healthy neurons but can accumulate into tangles in cells, and amyloid, which forms plaques between cells. Gallagher theorized that periods of hyperactivity in the brain, such as seizures, push those proteins around the brain, spreading more tangles and plaques. She identified a compound, levetiracetam, that calms these hyperactive periods and is now approved by the FDA to treat seizures in patients with epilepsy when used with other medications. She and Albert are testing a time-release capsule version of levetiracetam taken at breakfast by patients in early stages of the disease.

The study , sponsored by AgeneBio , involves more than 164 people taking the drug for 78 weeks at 27 research sites across the country. Preliminary results are expected in fall 2022.

In addition to Albert and Gallagher’s drug, referred to as AGB101, there are more than 100 other Alzheimer’s drugs being tested at Johns Hopkins and elsewhere, according to the Alzheimer’s Association.

Universal Screening

As researchers wrestle with how to catch the disease in its early stages, the question of whether to do universal screening for cognitive impairment remains. Such screening is required as part of the Medicare Annual Wellness visit, initiated in 2011 as part of the Affordable Care Act. While primary care providers must perform this screening for Medicare patients, specialists at Johns Hopkins believe this kind of test can be useful in more targeted ways.

“There could be a place for targeted universal screening, but we have to be very careful,” Oh says. “The question remains, what are you going to do with that information?”

In some scenarios, screening may be appropriate, she says. For example, a patient undergoing surgery who has possible cognitive impairment could be screened in order to make sure they understand the procedure as well as post-op instructions. It could help providers manage possible complications, such as delirium after surgery, which occurs more commonly in individuals with dementia.

The Alzheimer’s Association recommends evaluation for people with memory concerns or cognitive complaints as well as for those with non-memory triggers, including personality change, depression, deterioration of chronic disease without explanation, and falls or balance issues. It is also recommended if a family member or loved one reports cognitive impairment.

Like Oh, Paul Rosenberg and Kostas Lyketsos don’t see a role for universal screening as there’s no demonstrated benefit from it.

“Until we have a safe and effective therapy that people can afford, it is just not ethical to do the tests,” Rosenberg says.

Related Reading

Alzheimer’s disease: frustration and hope.

While Johns Hopkins clinicians help patients with Alzheimer's disease maximize quality of life, researchers are gaining insights that could lead to better treatment.

Alzheimers Disease Frustration and Hope_edit

Stress, Alzheimer’s and Aging

New research suggests that if you are caring for a spouse with Alzheimer’s, you have a higher risk of developing the disease yourself.

DOM1701039 Alzhiemers

Preventing and Treating Alzheimer's Disease and Related Dementias: Promising Research and Opportunities to Accelerate Progress: Proceedings of a Workshop—in Brief

  • PMID: 38713791
  • Bookshelf ID: NBK603367
  • DOI: 10.17226/27784

In response to a request from Congress, the National Institutes of Health asked the National Academies to conduct a study to assess the current state of research on Alzheimers Disease and Related Dementias (AD/ADRD) prevention and treatment, recommend research priorities, and identify strategies for overcoming barriers that impede scientific advancement. The resulting committee held a public workshop in January 2024 to explore promising areas of research that could catalyze scientific breakthroughs or accelerate the translation of discoveries into effective prevention and treatment strategies, as well as to discuss barriers to the advancement of research. The committee final report will be released in December 2024.

Copyright 2024 by the National Academy of Sciences. All rights reserved.

Publication types

Grants and funding.

  • Share full article

Advertisement

Supported by

Study Suggests Genetics as a Cause, Not Just a Risk, for Some Alzheimer’s

People with two copies of the gene variant APOE4 are almost certain to get Alzheimer’s, say researchers, who proposed a framework under which such patients could be diagnosed years before symptoms.

A colorized C.T. scan showing a cross-section of a person's brain with Alzheimer's disease. The colors are red, green and yellow.

By Pam Belluck

Scientists are proposing a new way of understanding the genetics of Alzheimer’s that would mean that up to a fifth of patients would be considered to have a genetically caused form of the disease.

Currently, the vast majority of Alzheimer’s cases do not have a clearly identified cause. The new designation, proposed in a study published Monday, could broaden the scope of efforts to develop treatments, including gene therapy, and affect the design of clinical trials.

It could also mean that hundreds of thousands of people in the United States alone could, if they chose, receive a diagnosis of Alzheimer’s before developing any symptoms of cognitive decline, although there currently are no treatments for people at that stage.

The new classification would make this type of Alzheimer’s one of the most common genetic disorders in the world, medical experts said.

“This reconceptualization that we’re proposing affects not a small minority of people,” said Dr. Juan Fortea, an author of the study and the director of the Sant Pau Memory Unit in Barcelona, Spain. “Sometimes we say that we don’t know the cause of Alzheimer’s disease,” but, he said, this would mean that about 15 to 20 percent of cases “can be tracked back to a cause, and the cause is in the genes.”

The idea involves a gene variant called APOE4. Scientists have long known that inheriting one copy of the variant increases the risk of developing Alzheimer’s, and that people with two copies, inherited from each parent, have vastly increased risk.

The new study , published in the journal Nature Medicine, analyzed data from over 500 people with two copies of APOE4, a significantly larger pool than in previous studies. The researchers found that almost all of those patients developed the biological pathology of Alzheimer’s, and the authors say that two copies of APOE4 should now be considered a cause of Alzheimer’s — not simply a risk factor.

The patients also developed Alzheimer’s pathology relatively young, the study found. By age 55, over 95 percent had biological markers associated with the disease. By 65, almost all had abnormal levels of a protein called amyloid that forms plaques in the brain, a hallmark of Alzheimer’s. And many started developing symptoms of cognitive decline at age 65, younger than most people without the APOE4 variant.

“The critical thing is that these individuals are often symptomatic 10 years earlier than other forms of Alzheimer’s disease,” said Dr. Reisa Sperling, a neurologist at Mass General Brigham in Boston and an author of the study.

She added, “By the time they are picked up and clinically diagnosed, because they’re often younger, they have more pathology.”

People with two copies, known as APOE4 homozygotes, make up 2 to 3 percent of the general population, but are an estimated 15 to 20 percent of people with Alzheimer’s dementia, experts said. People with one copy make up about 15 to 25 percent of the general population, and about 50 percent of Alzheimer’s dementia patients.

The most common variant is called APOE3, which seems to have a neutral effect on Alzheimer’s risk. About 75 percent of the general population has one copy of APOE3, and more than half of the general population has two copies.

Alzheimer’s experts not involved in the study said classifying the two-copy condition as genetically determined Alzheimer’s could have significant implications, including encouraging drug development beyond the field’s recent major focus on treatments that target and reduce amyloid.

Dr. Samuel Gandy, an Alzheimer’s researcher at Mount Sinai in New York, who was not involved in the study, said that patients with two copies of APOE4 faced much higher safety risks from anti-amyloid drugs.

When the Food and Drug Administration approved the anti-amyloid drug Leqembi last year, it required a black-box warning on the label saying that the medication can cause “serious and life-threatening events” such as swelling and bleeding in the brain, especially for people with two copies of APOE4. Some treatment centers decided not to offer Leqembi, an intravenous infusion, to such patients.

Dr. Gandy and other experts said that classifying these patients as having a distinct genetic form of Alzheimer’s would galvanize interest in developing drugs that are safe and effective for them and add urgency to current efforts to prevent cognitive decline in people who do not yet have symptoms.

“Rather than say we have nothing for you, let’s look for a trial,” Dr. Gandy said, adding that such patients should be included in trials at younger ages, given how early their pathology starts.

Besides trying to develop drugs, some researchers are exploring gene editing to transform APOE4 into a variant called APOE2, which appears to protect against Alzheimer’s. Another gene-therapy approach being studied involves injecting APOE2 into patients’ brains.

The new study had some limitations, including a lack of diversity that might make the findings less generalizable. Most patients in the study had European ancestry. While two copies of APOE4 also greatly increase Alzheimer’s risk in other ethnicities, the risk levels differ, said Dr. Michael Greicius, a neurologist at Stanford University School of Medicine who was not involved in the research.

“One important argument against their interpretation is that the risk of Alzheimer’s disease in APOE4 homozygotes varies substantially across different genetic ancestries,” said Dr. Greicius, who cowrote a study that found that white people with two copies of APOE4 had 13 times the risk of white people with two copies of APOE3, while Black people with two copies of APOE4 had 6.5 times the risk of Black people with two copies of APOE3.

“This has critical implications when counseling patients about their ancestry-informed genetic risk for Alzheimer’s disease,” he said, “and it also speaks to some yet-to-be-discovered genetics and biology that presumably drive this massive difference in risk.”

Under the current genetic understanding of Alzheimer’s, less than 2 percent of cases are considered genetically caused. Some of those patients inherited a mutation in one of three genes and can develop symptoms as early as their 30s or 40s. Others are people with Down syndrome, who have three copies of a chromosome containing a protein that often leads to what is called Down syndrome-associated Alzheimer’s disease .

Dr. Sperling said the genetic alterations in those cases are believed to fuel buildup of amyloid, while APOE4 is believed to interfere with clearing amyloid buildup.

Under the researchers’ proposal, having one copy of APOE4 would continue to be considered a risk factor, not enough to cause Alzheimer’s, Dr. Fortea said. It is unusual for diseases to follow that genetic pattern, called “semidominance,” with two copies of a variant causing the disease, but one copy only increasing risk, experts said.

The new recommendation will prompt questions about whether people should get tested to determine if they have the APOE4 variant.

Dr. Greicius said that until there were treatments for people with two copies of APOE4 or trials of therapies to prevent them from developing dementia, “My recommendation is if you don’t have symptoms, you should definitely not figure out your APOE status.”

He added, “It will only cause grief at this point.”

Finding ways to help these patients cannot come soon enough, Dr. Sperling said, adding, “These individuals are desperate, they’ve seen it in both of their parents often and really need therapies.”

Pam Belluck is a health and science reporter, covering a range of subjects, including reproductive health, long Covid, brain science, neurological disorders, mental health and genetics. More about Pam Belluck

The Fight Against Alzheimer’s Disease

Alzheimer’s is the most common form of dementia, but much remains unknown about this daunting disease..

How is Alzheimer’s diagnosed? What causes Alzheimer’s? We answered some common questions .

A study suggests that genetics can be a cause of Alzheimer’s , not just a risk, raising the prospect of diagnosis years before symptoms appear.

Determining whether someone has Alzheimer’s usually requires an extended diagnostic process . But new criteria could lead to a diagnosis on the basis of a simple blood test .

The F.D.A. has given full approval to the Alzheimer’s drug Leqembi. Here is what to know about i t.

Alzheimer’s can make communicating difficult. We asked experts for tips on how to talk to someone with the disease .

Posted for Davos Alzheimer’s Collaborative

May 14, 2024

How to Lower Your Risk for Alzheimer’s

Genes and age play a big role in susceptibility to the disease, but choices make a difference, too

Colorful illustration showing a brain, a group of elderly people and a city on different levels.

Joelle Bolt

Scientific American Custom Media Logo

This article was produced in partnership with the Davos Alzheimer’s Collaborative by Scientific American Custom Media, a division separate from the magazine’s board of editors.

S cientists are still learning about the causes of Alzheimer’s, but most agree that many factors contribute to an individual’s risk of getting the disease. Some risk factors, such as age and genes, are inescapable. Others can be modified with lifestyle changes, such as exercise and nutrition—in one study, participants were able to reduce their risk of getting Alzheimer’s by 60 percent. Although most risk studies focus on dementia in general, here are important findings about the known risk factors as they apply to Alzheimer’s.

Graphic illustration showing risk factors for Alzheimer's based on genetics, brain health, age, environment, lifestyle and body health

Katie Peek and Joelle Bolt; Sources: Risk factor relative importance: G. Livingston et al., The Lancet , 396, 413 (2020); normal brain aging: “Alzheimer’s Disease and Other Dementias,” Mayo Clinic, 2020

This article is part of The New Age of Alzheimer’s , a special report on the advances fueling hope for ending this devastating disease.

Learn more here about the innovation ecosystem that Davos Alzheimer’s Collaborative is building to speed breakthroughs and end Alzheimer’s disease. Explore the transforming landscape of Alzheimer’s in this special report .

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts

Volume 629 Issue 8011, 9 May 2024

Fossil fuels supply most of the world’s energy and are the basis of many key products used in everyday life, but they are also a major source of carbon dioxide emissions. Although renewable energy has the potential to replace fossil-fuel-generated energy, there will still be a need for the transport fuels and chemicals produced by oil refineries. In this week’s issue, Eelco Vogt and Bert Weckhuysen examine ways in which oil refineries could be reinvented to be completely fossil-free. They note that with sufficient long-term commitment and support, the science and technology for such a refinery could be developed, and they sketch a roadmap towards this goal.

Cover image: Jasiek Krzysztofiak/Nature

Reinvent oil refineries for a net-zero future

From petrol to plastics, oil-derived products define modern life. A bold plan to change that comes with huge costs — but researchers and policymakers should take it seriously.

Advertisement

Advertisement

Expat grants won’t fix Brazilian research

Permanent jobs and fairer hiring practices would encourage overseas scientists to return.

  • Juliano Morimoto

Research Highlights

A magnetic liquid makes for an injectable sensor in living tissue.

Fluid studded with microscopic magnetic particles can be inserted into the body and later retrieved.

Pandemic lockdowns were less of a shock for people with fewer ties

During periods of enforced isolation, life satisfaction for older adults took less of a hit in those who were already socially isolated.

Not just truffles: dogs can sniff out surpassingly rare native fungus

Daisy, a member of a breed used to find fungal delicacies, detected a critically endangered Australian fungus faster than a trained human could.

Never mind little green men: life on other planets might be purple

Bacteria that make food using a compound other than chlorophyll could paint other planets in a wide range of colours.

News in Focus

What china’s mission to collect rocks from the moon’s far side could reveal.

The Chang’e-6 mission aims to land in the Moon’s oldest and largest crater, where it will collect rocks to bring back to Earth.

First fetus-to-fetus transplant demonstrated in rats

The tissue developed into functioning kidneys and produced urine.

  • Smriti Mallapaty

Superconductivity hunt gets boost from China’s $220 million physics ‘playground’

From extreme cold to strong magnets and high pressures, the Synergetic Extreme Condition User Facility (SECUF) provides conditions for researching potential wonder materials.

  • Gemma Conroy

Scientists tried to give people COVID — and failed

Researchers deliberately infect participants with SARS-CoV-2 in ‘challenge’ trials — but high levels of immunity complicate efforts to test vaccines and treatments.

  • Ewen Callaway

Epic blazes threaten Arctic permafrost. Can firefighters save it?

Some scientists argue that it’s time to rethink the blanket policy of letting blazes burn themselves out in northern wildernesses.

  • Jeff Tollefson

How reliable is this research? Tool flags papers discussed on PubPeer

Browser plug-in alerts users when studies — or their references — have been posted on a site known for raising integrity concerns.

  • Dalmeet Singh Chawla

‘ChatGPT for CRISPR’ creates new gene-editing tools

Some of the AI-designed gene editors could be more versatile than those found in nature.

Dark energy is tearing the Universe apart. What if the force is weakening?

The first set of results from a pioneering cosmic-mapping project hints that the repulsive force known as dark energy has changed over 11 billion years, which would alter ideas about how the Universe has evolved and what its future will be.

  • Davide Castelvecchi

Hacking the immune system could slow ageing — here’s how

Our immune system falters over time, which could explain the negative effects of ageing.

  • Alison Abbott

Books & Arts

Book review, smarty plants controversial plant-intelligence studies explored in new book.

A deep dive into plant behaviour and consciousness asks why the topic has been taboo for so long, and whether botanists are changing their minds about plants’ cognitive abilities.

  • Beronda L. Montgomery

How rich is too rich?

Where should society draw the line on extreme wealth? A fresh account sets out the logic and suggests how to redress inequality.

  • Lucas Chancel

From multiverses to cities: Books in brief

Andrew Robinson reviews five of the best science picks.

  • Andrew Robinson

Allen J. Bard obituary: electrochemist whose techniques underpin clinical diagnostics, materials discovery and more

Innovator who pioneered scanning electrochemical microscopy, bioassays and solar fuels.

  • Michael Rose
  • Henry S. White

Correspondence

Beware of graphene’s huge and hidden environmental costs.

  • Qingyuan Ding

Japan can embrace open science — but flexible approaches are key

Countering extreme wildfires with prescribed burning can be counterproductive.

  • David Lindenmayer
  • Philip Zylstra

Finding millennia-old ‘monumental’ corals could unlock secrets of climate resilience

  • Simone Montano
  • Federica Siena
  • Giovanni Strona

Hunger on campus: why US PhD students are fighting over food

Graduate students are relying on donated and discounted food in the struggle to make ends meet.

  • Laurie Udesky

Technology Feature

Powerful ‘nanopore’ dna sequencing method tackles proteins too.

Latest methods bring the speed, portability, and long read lengths of nanopore sequencing to proteomics.

  • Caroline Seydel

Collection:

Where I Work

I make 3d models of conifer needles to explore their climate effects.

Jan Pisek seeks a better understanding of how forests absorb sunlight, carbon and heat.

  • Nic Fleming

News & Views

Ancient dna traces family lines and political shifts in the avar empire.

Genetic pedigrees spanning nine generations uncover the social organization of a nomadic empire that dominated much of central and eastern Europe from the sixth to the early ninth century.

  • Lara M. Cassidy

Vibration isolation could boost performance of near-infrared organic LEDs

The development of high-performance organic LEDs and other devices that emit near-infrared light has been hindered by seemingly fundamental features of the light-emitting molecules. A potential solution has been identified.

  • Margherita Maiuri

Streamlined skull helps foxes take a nosedive

Some fox species leap up and pounce head first into snow to capture prey that they hear below the surface. An analysis of the forces involved reveals how the shape of the skull has evolved to minimize damage from this behaviour.

  • Mary Abraham

Coupled neural activity controls working memory in humans

How does the human brain temporarily store information without losing track of it? Neuroscientists have discovered that neurons in the frontal and temporal lobes work together to hold information in working memory.

  • Ziv Williams

Bioengineered ‘mini-colons’ shed light on cancer progression

Cells grown on a 3D scaffold have generated a ‘mini-colon’ that mimics key features of the organ. Controlled expression of cancer-associated genes in the system offers a way to examine tumour formation over space and time.

  • Nicolò Riggi
  • Felipe de Sousa e Melo
  • Cancer at Nature Portfolio

Perspective

The refinery of the future.

Efforts to find renewable alternatives to fossil fuels that might enable a carbon-neutral society by 2050 are described, as well as outlining a possible roadmap towards a refinery of the future and evaluating its requirements.

  • Eelco T. C. Vogt
  • Bert M. Weckhuysen

Venus water loss is dominated by HCO + dissociative recombination

Water loss to space late in Venus history is shown to be more active than previously thought, with unmeasured HCO + dissociative recombination dominating present-day H loss.

  • M. S. Chaffin
  • E. M. Cangi

An atomic boson sampler

Boson sampling using ultracold atoms in a two-dimensional, tunnel-coupled optical lattice is enabled by high-fidelity programmable control with optical tweezers of a large number of atoms trapped in an optical lattice.

  • Aaron W. Young
  • Shawn Geller
  • Adam M. Kaufman

Observation of Nagaoka polarons in a Fermi–Hubbard quantum simulator

Emergence of Nagaoka polarons and kinetic magnetism is observed in a Hubbard system realized with strongly interacting fermions trapped in a triangular optical lattice.

  • Martin Lebrat
  • Markus Greiner

Directly imaging spin polarons in a kinetically frustrated Hubbard system

A triangular-lattice Hubbard system realized with ultracold atoms is used to directly image spin polarons, revealing ferromagnetic correlations around a charge dopant, a manifestation of the Nagaoka effect.

  • Max L. Prichard
  • Benjamin M. Spar
  • Waseem S. Bakr

All-optical subcycle microscopy on atomic length scales

All-optical subcycle microscopy is achieved on atomic length scales, with picometric spatial and femtosecond temporal resolution.

Multi-project wafers for flexible thin-film electronics by independent foundries

The iconic 6502 microprocessor designed in two key thin-film transistor technologies by independent foundries is used to demonstrate and expand the multi-project wafer approach for flexible electronics.

  • Hikmet Çeliker
  • Wim Dehaene

Chemical short-range disorder in lithium oxide cathodes

The introduction of chemical short-range disorder substantially affects the crystal structure of layered lithium oxide cathodes, leading to improved charge transfer and structural stability.

  • Zhenpeng Yao
  • Chenglong Zhao

Growth of diamond in liquid metal at 1 atm pressure

Diamond crystals and polycrystalline diamond films can be grown using liquid metal at standard pressure and high temperature instead of conventional high pressure and high temperature.

  • Rodney S. Ruoff

Decoupling excitons from high-frequency vibrations in organic molecules

A molecular design strategy for reducing the vibration-induced non-radiative losses in emissive organic semiconductors is realized by decoupling excitons from high-frequency vibrations.

  • Pratyush Ghosh
  • Antonios M. Alvertis

Copper-catalysed dehydrogenation or lactonization of C( sp 3 )–H bonds

Use of N -methoxyamides as oxidants enables controllable, redox-neutral, green catalysis of bimodal dehydrogenation/lactonization reactions with methanol as the only by-product.

  • Shupeng Zhou
  • Zi-Jun Zhang
  • Jin-Quan Yu

Ghost roads and the destruction of Asia-Pacific tropical forests

An effort to map roads in the Asia-Pacific region finds that there are 3.0–6.6 times more roads than other sources suggest, and that unmapped ‘ghost roads’ are a major contributor to tropical forest loss.

  • Jayden E. Engert
  • Mason J. Campbell
  • William F. Laurance

Network of large pedigrees reveals social practices of Avar communities

Analysis of ancient DNA from 424 individuals in the Avar period, from the sixth to the ninth century AD, reveals population movement from the steppe and the prolonged existence of a steppe nomadic descent system centred around patrilineality and female exogamy in central Europe.

  • Guido Alberto Gnecchi-Ruscone
  • Zsófia Rácz
  • Zuzana Hofmanová

Cell-type-resolved mosaicism reveals clonal dynamics of the human forebrain

Using mosaic variant barcode analysis, clonal dynamics of specific cell types are deconvolved in the human forebrain.

  • Changuk Chung
  • Xiaoxu Yang
  • Joseph G. Gleeson

Control of working memory by phase–amplitude coupling of human hippocampal neurons

Hippocampal theta–gamma phase–amplitude coupling integrates cognitive control and working memory storage across brain areas in humans.

  • Jonathan Daume
  • Jan Kamiński
  • Ueli Rutishauser

Control of neuronal excitation–inhibition balance by BMP–SMAD1 signalling

Signalling by the developmental morphogen BMP2 through the transcription factor SMAD1 has a key role in controlling the glutamatergic innervation of parvalbumin-expressing interneurons and maintaining the balance between excitation and inhibition in the mammalian cortex.

  • Zeynep Okur
  • Nadia Schlauri
  • Peter Scheiffele

DNA glycosylases provide antiviral defence in prokaryotes

A screen utilizing an environmental DNA library in Escherichia coli is used to identify Brig1, a previously unknown anti-phage defence system with homologues across distinct clades of bacteria.

  • Amer A. Hossain
  • Ying Z. Pigli
  • Luciano A. Marraffini

PGE 2 limits effector expansion of tumour-infiltrating stem-like CD8 + T cells

Tumour-derived prostaglandin E 2 , signaling through its receptors EP 2 and EP 4 , is shown to restrain the responses of tumour-infiltrating stem-like TCF1 + CD8 + T lymphocytes, and modulation of T cell EP 2 and EP 4 can restore anticancer immunity.

  • Sebastian B. Lacher
  • Janina Dörr
  • Jan P. Böttcher

PGE 2 inhibits TIL expansion by disrupting IL-2 signalling and mitochondrial function

Prostaglandin E2 from the tumour microenvironment impairs interleukin-2 sensing by tumour-infiltrating lymphocytes, restricting proliferative response and promoting T cell death via metabolic impairment and ferroptosis. 

  • Matteo Morotti
  • Alizee J. Grimm
  • George Coukos

Chemoproteomic discovery of a covalent allosteric inhibitor of WRN helicase

VVD-133214, a clinical-stage, covalent allosteric inhibitor of the helicase WRN, was well tolerated in mice and led to robust tumour regression in multiple microsatellite-instability-high colorectal cancer cell lines and patient-derived xenograft models.

  • Kristen A. Baltgalvis
  • Kelsey N. Lamb
  • Todd M. Kinsella

Discovery of WRN inhibitor HRO761 with synthetic lethality in MSI cancers

HRO761 is a potent, selective, allosteric WRN inhibitor that binds at the interface of the D1 and D2 helicase domains, locking WRN in an inactive conformation.

  • Stephane Ferretti
  • Jacques Hamon
  • Marta Cortés-Cros

Spatiotemporally resolved colorectal oncogenesis in mini-colons ex vivo

Topobiologically complex mini-colons—which enable the faithful in vitro recapitulation of colorectal cancer tumorigenesis and its environmental determinants—offer the possibility to reduce animal use in a wide range of experimental applications.

  • L. Francisco Lorenzo-Martín
  • Tania Hübscher
  • Matthias P. Lutolf

Single-cell analysis reveals context-dependent, cell-level selection of mtDNA

A new method for tracking single-cell heteroplasmy, called SCI-LITE, is combined with mitochondrial DNA base editing to reveal principles of heteroplasmy dynamics in dividing cells.

  • Anna V. Kotrys
  • Timothy J. Durham
  • Vamsi K. Mootha

Structures and activation mechanism of the Gabija anti-phage system

Structures of complexes containing GajA and GajB proteins of the prokaryotic Gabija anti-phage defence system reveal the mechanism of its activation after DNA cleavage upon ATP depletion.

  • Longfei Wang

Ligand efficacy modulates conformational dynamics of the µ-opioid receptor

Studies on the µ-opioid receptor using fluorescent labelling of intracellular residues and energy transfer experiments in the presence of different ligands with or without G-protein binding reveals conformational changes that correlate to ligand efficacy.

  • Jiawei Zhao
  • Matthias Elgeti
  • Chunlai Chen

Promiscuous G-protein activation by the calcium-sensing receptor

Structures of the human calcium-sensing receptor can be bound into complex with G proteins from three different Gα subtypes while maintaining G-protein-binding specificity.

  • Jinseo Park
  • Qing R. Fan

Amendments & Corrections

Author correction: targeting swi/snf atpases in enhancer-addicted prostate cancer.

  • Abhijit Parolia
  • Arul M. Chinnaiyan

Author Correction: Interim analyses of a first-in-human phase 1/2 mRNA trial for propionic acidaemia

  • Dwight Koeberl
  • Andreas Schulze
  • Stephanie Grunewald

Publisher Correction: FOXO1 is a master regulator of memory programming in CAR T cells

  • Alexander E. Doan
  • Katherine P. Mueller
  • Evan W. Weber

Advertisement

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

how to do a research paper on alzheimer's disease

IMAGES

  1. Informative Essay

    how to do a research paper on alzheimer's disease

  2. Alzheimer’s Disease Research Paper Free Essay Example

    how to do a research paper on alzheimer's disease

  3. (PDF) Early Detection of Alzheimer’s Disease Using Magnetic Resonance

    how to do a research paper on alzheimer's disease

  4. Understanding Alzheimer's Disease and Its Impact on Memory Free Essay

    how to do a research paper on alzheimer's disease

  5. ⇉Alzheimers Disease research paper Essay Example

    how to do a research paper on alzheimer's disease

  6. (PDF) ALZHEIMER DISEASE: A REVIEW

    how to do a research paper on alzheimer's disease

VIDEO

  1. H. Pylori: A Hidden Risk for Alzheimer's?

  2. Steps for Systematic Literature review

  3. Learn About Breakthroughs in Dementia Research at AAIC

  4. New Study: 29% Improvement In Alzheimer’s Disease?!

  5. How Alzheimer’s Disease Affects Your Brain Cells

COMMENTS

  1. Comprehensive Review on Alzheimer's Disease: Causes and Treatment

    1. Introduction. Alzheimer's disease (AD) (named after the German psychiatric Alois Alzheimer) is the most common type of dementia and can be defined as a slowly progressive neurodegenerative disease characterized by neuritic plaques and neurofibrillary tangles (Figure 1) as a result of amyloid-beta peptide's (Aβ) accumulation in the most affected area of the brain, the medial temporal ...

  2. Progress with Treatments for Alzheimer's Disease

    The inexorable progression of Alzheimer's disease exerts a huge toll on patients, families, and society, costing approximately $1 trillion annually, an amount that is likely to increase with the ...

  3. (PDF) Alzheimer's Disease: A Comprehensive Review of its Causes

    Abstract. Alzheimer's Disease (AD) is a progressive neurodegenerative disease that results in the loss of memory, motor function, ability to think, and other basic functions required for day-to ...

  4. Alzheimer's disease

    New data confirm that APOE4 homozygosity is a major genetic cause of Alzheimer's disease, warranting the development of specialized research strategies, treatment approaches and clinical trials.

  5. The Amyloid-β Pathway in Alzheimer's Disease

    Similarly, in Western Europe, dementia affects ~2.5% of people aged 65-69 years, escalating to about 40% of those aged 90-94 years , and by 2050, there will likely be up to 18.9 million ...

  6. In-depth insights into Alzheimer's disease by using ...

    Alzheimer's disease is still a field of research with lots of open questions. The complexity of the disease prevents the early diagnosis before visible symptoms regarding the individual's ...

  7. (PDF) ALZHEIMER DISEASE: A REVIEW

    Alzheimer's disease is a progressive neurodegenerative disease that causes brain cells to waste away and die. It is characterized by progressive cognitive deterioration and continuous decline in ...

  8. Alzheimer's disease: risk factors and potentially protective measures

    Alzheimer's disease (AD) is the most common type of dementia [], affecting at least 27 million people and corresponding from 60 to 70% of all dementias cases [].The occurrence of this disease also has a huge impact on life of patient's family, in addition to a high financial cost to society [].From an anatomopathological point of view, AD is characterized by two prototypical lesions: 1 ...

  9. Comprehensive Review on Alzheimer's Disease: Causes and Treatment

    Alzheimer's disease (AD) is a disorder that causes degeneration of the cells in the brain and it is the main cause of dementia, which is characterized by a decline in thinking and independence in personal daily activities. AD is considered a multifactorial disease: two main hypotheses were proposed as a cause for AD, cholinergic and amyloid hypotheses. Additionally, several risk factors such ...

  10. A comprehensive research setup for monitoring Alzheimer's disease using

    Alzheimer's disease (AD) has a detrimental impact on brain function, affecting various aspects such as cognition, memory, language, and motor skills. Previous research has dominantly used electroencephalography (EEG) and functional near-infrared spectroscopy (fNIRS) to individually measure brain signals or combine the two methods to target specific brain functions. However, comprehending ...

  11. Research on Alzheimer's Disease and Related Dementias

    The federal government's Alzheimer's and related dementias research strategy focuses on engaging a cross-disciplinary team of geneticists, epidemiologists, gerontologists, behavioral scientists, disease and structural biologists, pharmacologists, clinical researchers, and others to bring the greatest and most diverse expertise to the field.

  12. Is Alzheimer disease a disease?

    Extensive research has resulted in no cure for what is perceived as the most common dementing illness: Alzheimer disease (AD). AD is defined by specific brain abnormalities — amyloid-β plaques ...

  13. Research Aims to Uncover the Mysteries of Alzheimer's Disease

    Dementia is a general term that refers to memory loss and decline of other cognitive abilities that limit independence in day-to-day function. Alzheimer's is the most common brain disease that causes dementia among older adults, accounting for 60%-80% of cases. It affects an estimated one in nine people age 65 and older — 6.2 million ...

  14. Preventing and Treating Alzheimer's Disease and Related Dementias

    In response to a request from Congress, the National Institutes of Health asked the National Academies to conduct a study to assess the current state of research on Alzheimers Disease and Related Dementias (AD/ADRD) prevention and treatment, recommend research priorities, and identify strategies for overcoming barriers that impede scientific advancement.

  15. Researchers call for a major rethink of how Alzheimer's ...

    In a paper published last year 2, when he was working for Danish firm Novo Nordisk, in a lab just outside Copenhagen, Raket took a similar approach to calculating treatment effects in terms of ...

  16. Study Suggests Genetics as a Cause, Not Just a Risk, for Some Alzheimer

    May 6, 2024 Updated 12:19 p.m. ET. Scientists are proposing a new way of understanding the genetics of Alzheimer's that would mean that up to a fifth of patients would be considered to have a ...

  17. Analysis of the hippocampal neural network activity in vivo by

    The miniscope technique appears to be an effective tool for identifying shifts in neuronal networks during the progression of neurodegenerative diseases, such as Alzheimer's disease, and may also aid in detecting possible changes following neurological complications related to viral infections. Miniature fluorescent microscopy is a method that enables neurobiologists to visualize and record ...

  18. How to Lower Your Risk for Alzheimer's

    S cientists are still learning about the causes of Alzheimer's, but most agree that many factors contribute to an individual's risk of getting the disease. Some risk factors, such as age and ...

  19. Diagnosis and Management of Dementia: A Review

    Dementia is an acquired loss of cognition in multiple cognitive domains sufficiently severe to affect social or occupational function. In the US, Alzheimer's disease (AD) affects 5.8 million people. However, dementia is commonly associated with more than one neuropathology, usually AD with cerebrovascular pathology.

  20. Molecular Mechanism of Alzheimer's Disease

    Molecular Mechanism of Alzheimer's Disease. Neurodegenerative disorders are a major public health concern. Despite decades of research, we are still seeking an efficient cure for these pathologies. Alzheimer's disease (AD), which is closely associated with aging, is the most common neurodegenerative disease and the leading cause of dementia ...

  21. Volume 629 Issue 8011, 9 May 2024

    Efforts to find renewable alternatives to fossil fuels that might enable a carbon-neutral society by 2050 are described, as well as outlining a possible roadmap towards a refinery of the future ...