ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Epidemiology, pathology, and pathogenesis of Alzheimer disease

Epidemiology, pathology, and pathogenesis of Alzheimer disease
Literature review current through: Jan 2024.
This topic last updated: Aug 23, 2022.

INTRODUCTION — Alzheimer disease (AD) is the most common cause of dementia and one of the leading sources of morbidity and mortality in the aging population.

The hallmark neuropathologic changes of AD are diffuse and neuritic plaques, marked by extracellular amyloid beta deposition, and neurofibrillary tangles, comprised of the intracellular accumulation of hyperphosphorylated tau (p-tau) protein. The epidemiologic study of AD is being transformed by the availability of new biomarker technologies to measure such neuropathologic changes in vivo. Large randomized clinical trials are evaluating anti-amyloid and other disease-modifying therapies for the treatment and prevention of AD utilizing these imaging or cerebrospinal fluid (CSF) biomarkers [1].

The study of AD has focused on three interrelated hypotheses [2]:

Extracellular amyloid plaques (primarily composed of amyloid beta peptides) are a unique genetic and lifestyle disease due to increased production of amyloid beta in younger, genetically high-risk individuals and reduced metabolism and removal among older individuals.

Vascular disease is an independent determinant of vascular dementia but also of increased amyloid deposition and neurodegeneration.

Dementia in older individuals is a syndrome that derives most commonly from an individually varying mixture of diseases that afflict the brain. The most prevalent of these is AD, but it commonly occurs along with other diseases of the brain, vascular brain injury, Lewy body disease, and hippocampal sclerosis (HS).

This topic will focus on the incidence, prevalence, and risk factors for AD specifically. Risk factors for cognitive decline and all-cause dementia more generally, derived primarily from studies that define dementia according to a Mini-Mental State Examination score cutoff or similarly nonspecific case definition, are discussed separately. Protective factors and prevention of dementia are also discussed separately.

(See "Risk factors for cognitive decline and dementia".)

(See "Prevention of dementia".)

PATHOLOGY — The neuropathologic assessment of AD includes both evaluation of neuropathologic changes and correlation with clinical, neuropsychologic, neuroimaging, and other laboratory data [3,4]. Essential neuropathologic changes of AD include the following:

Extracellular deposits of amyloid beta peptides (image 1)

Neuritic plaques, associated with neuronal injury and characterized by amyloid formed from amyloid beta with dystrophic neurites that frequently have phospho-tau immunoreactivity [5-8]

Neurofibrillary degeneration, best exemplified by neurofibrillary tangles (image 1)

AD neuropathologic change is ranked along three parameters: amyloid beta plaque distribution score [9], neurofibrillary tangle distribution stage [10,11], and neuritic plaque density score [12]. An overall assessment of low, intermediate, or high levels of AD neuropathologic change is obtained based on these three parameters.

In addition to these essential features, several other pathologic changes are commonly observed in association with AD.

Cerebral amyloid angiopathy is often found in cases with parenchymal amyloid beta deposits [13,14].

Inclusions of abnormal alpha-synuclein accumulation, known as Lewy bodies, are common in the setting of intermediate-to-high levels of AD neuropathologic change [15,16], including some early-onset familial AD cases [17,18]. Lewy bodies in AD often include extensive deposition in the amygdala. (See "Epidemiology, pathology, and pathogenesis of dementia with Lewy bodies", section on 'Pathology'.)

Vascular brain injury is encountered commonly in brains with AD. Pathologic changes of vascular brain injury are caused by oligemia, hypoxemia, or ischemia involving different caliber vessels in different regions of brain. (See "Etiology, clinical manifestations, and diagnosis of vascular dementia".)

Hippocampal sclerosis (HS) in AD is defined by pyramidal cell loss and gliosis in the hippocampal formation that is out of proportion to AD neuropathologic change [19]. HS can be observed alone or in the context of AD, frontotemporal lobar degeneration, or vascular brain injury. A similar lesion also can be observed in chronic epilepsy and other processes that injure the hippocampus. (See "Focal epilepsy: Causes and clinical features", section on 'Hippocampal sclerosis'.)

Immunoreactive inclusions of TAR DNA binding protein 43 (TDP-43), as seen in most cases of amyotrophic lateral sclerosis and in a subset of frontotemporal lobar degeneration cases, are also commonly observed in cases with AD neuropathologic change [20]. With the exception of individuals with specific mutations that cause frontotemporal dementia, it is not clear whether TDP-43-immunoreactive inclusions in these other neurodegenerative diseases are a primary, secondary, or coincidental event [21]. Some evidence suggests that hippocampal TDP-43 deposition is associated with faster clinical progression and hippocampal atrophy among patients with AD [20,22,23]. (See "Epidemiology and pathogenesis of amyotrophic lateral sclerosis", section on 'Intracellular inclusions' and "Frontotemporal dementia: Epidemiology, pathology, and pathogenesis", section on 'FTLD-TDP'.)

PATHOGENESIS — While the pathogenesis of AD remains unclear, all forms of AD appear to share overproduction and/or decreased clearance of amyloid beta peptides. Amyloid beta peptides are produced by the endoproteolytic cleavage of mature protein translated from the amyloid beta precursor protein (APP) gene and cleaved by beta-secretase and gamma-secretase. Presenilin forms part of the gamma-secretase complex, and mutations in presenilin 1 (PSEN1) or presenilin 2 (PSEN2) appear to favor production of amyloid beta overall, or more neurotoxic forms of amyloid beta. The ultimate neurotoxin in AD is debated, but experimental evidence highlights small aggregates of amyloid beta peptides called oligomers, as opposed to larger aggregates called fibrils [24]. (See "Genetics of Alzheimer disease", section on 'Presenilin 1' and "Genetics of Alzheimer disease", section on 'Presenilin 2'.)

The pathogenesis of AD also involves a second protein, tau. Tau is a microtubule-associated protein that aids in microtubule assembly and stabilization. In AD, tau becomes hyperphosphorylated and aggregates to form paired helical filament (PHF) tau, a major component of neurofibrillary tangles within the neuronal cytoplasm. The accumulation of this altered protein is toxic to neurons in experimental models. In addition, transmission of pathologic forms of tau between neurons has been proposed to account for the spread of AD in the brain, which follows a distinct progression across brain regions as AD advances [25-27].

There are several other important and potentially overlapping pathways likely involved in AD pathogenesis, many of which have been implicated by the various risk genes that have been identified. As an example, the human apolipoprotein E (APOE) gene is a pleiotropic lipoprotein involved in multiple cellular processes including cholesterol transport, development, synaptic plasticity, and immune regulation, among others. There are three alleles of APOE, called epsilon 2 (e2), e3, and e4, and their encoded isoforms also vary in several activities. At least one mechanism by which inheritance of APOE e4 may increase AD risk is impairment of amyloid beta clearance from cerebrum [28]. (See "Genetics of Alzheimer disease", section on 'Apolipoprotein E'.)

CASE DEFINITION — A definitive diagnosis of AD requires histopathologic examination, but most studies of AD rely on clinical criteria to define cases. The clinical criteria for a diagnosis of probable AD have changed over time, and much of the older literature has either not distinguished between AD and other forms of dementia or utilized relatively small case-control-type studies in selected populations. Current clinical criteria for the diagnosis of AD are discussed separately. (See "Clinical features and diagnosis of Alzheimer disease", section on 'Diagnosis'.)

Studies will continue to evolve with the adoption of more sensitive and specific clinical criteria for AD as well as the incorporation of techniques for in vivo measurement of neuropathology such as amyloid positron emission tomography (PET), which allows for antemortem evaluation of cerebral amyloid accumulation. In addition, new PET tracers are under development to assess hyperphosphorylated tau (p-tau) to further characterize pathologic peptides in the living brain. The cost of these new technologies poses challenges for the development of large-scale epidemiologic studies, however, and may limit sample sizes [2,29,30]. (See "Clinical features and diagnosis of Alzheimer disease", section on 'Neuroimaging'.)

An additional consideration is that the pathologic changes of AD are often not found in isolation; many patients who die with dementia have evidence of mixed pathology. Based on information from several large, population-based clinicopathologic studies in older adults, the following observations can be made about the relationship between brain pathology and antemortem risk factors for AD [31-40]:

There is a very high correlation between a clinical antemortem diagnosis of AD and postmortem measures of AD.

Approximately one-third of individuals over the age of 85 years who die without antemortem diagnoses of dementia or AD have the pathologic changes of AD.

Cerebrovascular disease is very common among persons who die with dementia, and the presence of AD pathologic changes and cerebrovascular disease together is common among individuals who die with dementia.

Pathologic changes of multiple diseases, known as mixed dementia, are the most common finding in older individuals with clinical diagnosis of probable AD. (See 'Cerebrovascular disease' below.)

INCIDENCE AND PREVALENCE — The overall burden of AD is substantial worldwide [41-46]. Globally, an estimated 47 million people are affected by dementia [47].

Older adults — AD is increasingly prevalent with advancing age. In the United States in 2011, there were an estimated 4.5 million individuals over the age of 65 years living with clinical AD; this included 0.7 million people ages 65 to 74 years, 2.3 million ages 75 to 84, and 1.8 million 85 years and older [46]. This figure is projected to rise to 13.8 million in the United States and >130 million worldwide by 2050. The age-standardized prevalence of dementia ranges from 5 to 7 percent in most developed countries [42,45,47-50].

Estimates of the incidence of dementia vary across studies and depend heavily on age. In general, the incidence of dementia doubles every 10 years after age 60 years [44]. In the longitudinal Cardiovascular Health Study-Cognition Study (CHS-CS), only 19 of 160 participants alive at age 93 years remained free of mild cognitive impairment or dementia [51]. There is very little sex difference in incidence and prevalence of dementia or AD, although by absolute numbers there are more females than males with the disease, particularly over the age of 85 years, due to differences in life expectancy.

In 24 longitudinal studies, the age-specific incidence of dementia ranged from as low as 5 of 1000 for individuals ages 65 to 70 years and as high as 60 to 80 of 1000 for those age 85 years and older [45]. Examples of individual studies include the following:

The CHS-CS followed over 3000 adults who were free of dementia at baseline over a five-year period and captured cases of AD or a mixture of AD and vascular dementia. Incidence estimates varied from 32 of 1000 person-years ages 75 to 79 at entry to 96 of 1000 person-years age ≥85 at entry, with little sex difference [52].

In a population-based study in Rotterdam, the short-term incidence of dementia in the year 2000 was 1 of 1000 for ages 60 to 69 years, 6.4 of 1000 for ages 70 to 79 years, and 26 of 1000 for ages 80 to 89 years [53].

Dementia is a common cause of death in older adults. In a Medicare survey of 22,896 adults age 65 years and older, 15 diseases accounted for 70 percent of all deaths [54]. Dementia was second to heart failure as a leading cause of mortality, accounting for 19 percent of the deaths. Traditional mortality statistics are not a good measure of deaths attributed to dementia, however; these figures underestimate the contribution of dementia to mortality [54,55]. While individuals do not die of AD per se, advanced disease increases vulnerability for other disorders, commonly infections, which ultimately lead to death.

Younger adults — AD is also prevalent in younger adults with dementia, although there are fewer population-based studies in individuals under the age of 65 years [56,57].

A study from the United Kingdom estimated that the incidence of dementia in individuals ages 30 to 65 years was approximately 54 per 100,000 person-years [58]. The most common causes of dementia in these patients were AD (34 percent), vascular dementia (18 percent), frontotemporal dementia (12 percent), dementia with Lewy bodies (7 percent), alcohol-related dementia (10 percent), and others (19 percent). A separate population-based study in England reported an incidence of AD of 4.2 cases per 100,000 person-years among individuals ages 45 to 64 years [59].

Temporal trends — Multiple studies have reported that the prevalence of all-cause dementia may be declining over time in high-income countries [41,60-66]. As an example, a population-based study in Rotterdam reported a decreased incidence of dementia in all age groups between 1990 and 2000 as well as an increase in brain volume as a percent of intracranial volume [53]. In another study using data from the Framingham Heart Study in the United States, the cumulative five-year hazard rate of dementia fell from 3.6 per 100 persons during the late 1970s to 2.0 per 100 persons in the late 2000s [67]. The magnitude of the decline was greater for vascular dementia than for AD dementia, and rates of hypertension, atrial fibrillation, and stroke also fell over the same time period. Possible reasons for such a trend include improved educational levels of the more recent birth cohorts who are reaching the age of dementia risk or improved prevention and treatment of risk factors for vascular disease. By contrast, age-adjusted mortality rates from AD specifically have risen in the United States over the last decade [68].

It remains possible that the apparent decline in overall dementia is an artifact introduced by the definition of dementia as a specific level of cognitive impairment, especially memory, and related disability, rather than a relative change. Better-educated, cognitively higher-functioning individuals can tolerate a greater extent of neuropathologic change prior to reaching the threshold of dementia (this concept is termed "cognitive reserve") [69]. If dementia is defined as a change in cognition to below an absolute level and education improves over time, the prevalence of dementia may appear to decline when the only thing that has changed is the initial (higher) level of education. In support of this, a population-based study in the northern United States found no change in the incidence of dementia over time in a relatively homogenous higher-educated population [41].

PRESYMPTOMATIC PERIOD — There is a long presymptomatic period between the onset of pathophysiologic processes in the brain and the development of clinical symptoms of AD, suggesting that long-term epidemiologic studies beginning at an early age are needed to properly study the gene-lifestyle environmental determinants of amyloid vascular disease and neurodegeneration. In the Rotterdam Study, for example, the average age at diagnosis of dementia was 82 years, and memory complaints in these patients began 16 years before diagnosis [70].

The long presymptomatic period has also been demonstrated in families with dominantly inherited AD due to genetic mutations in amyloid beta precursor protein (APP), presenilin 1 (PSEN1), or presenilin 2 (PSEN2) [71,72]. Such mutations carry a nearly 100 percent risk of early-onset AD, which is typically diagnosed between 35 and 50 years of age, depending on the family. In one study, 88 carriers were evaluated at a mean age of 39 years [71]. Parental age at onset of AD in the index cases was used to estimate the relationship between disease biomarkers and the onset of clinical AD. A decline in amyloid beta 1 through 42 in cerebrospinal fluid (CSF) appeared to precede disease onset by 25 years; amyloid in the brain measured by Pittsburgh compound-B (PiB) retention on positron emission tomography (PET) scan, phospho-tau in CSF, and magnetic resonance imaging (MRI) measures of brain atrophy appear to be present approximately 15 years prior to onset of disease. Measurable cognitive impairment appears to occur approximately five years prior to clinical diagnosis.

These biomarkers appear to identify a presymptomatic period for AD in the general population as well. Neuroimaging findings including amyloid positivity on PET, as well as white matter lesions, neurodegeneration (cortical thickness), and small hippocampal volumes measured on MRI, have been found to be associated with cognitive decline and incident dementia, particularly when these findings occur in combination [73-76].

GENETIC RISK FACTORS — Aside from age, the most clearly established risk factors for AD are a family history of dementia, rare dominantly inherited mutations in genes that impact amyloid in the brain, and the apolipoprotein E (APOE) epsilon 4 (e4) allele.

Early-onset Alzheimer disease — The genetic basis for AD is understood most clearly in the early-onset form, which accounts for less than 1 percent of cases. Early-onset AD follows an autosomal dominant inheritance pattern related to mutations in genes that alter amyloid beta protein production, aggregation, or clearance, including amyloid beta precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2). Such mutations are highly penetrant, meaning that carriers have a nearly 100 percent chance of developing the disease in their lifetime.

Other early-onset cases without mutations can present clinically prior to 65 years of age. While these individuals have higher than expected inheritance of APOE e4, not all do, and this early-onset group is not fully understood [77]. (See "Genetics of Alzheimer disease", section on 'Early-onset Alzheimer disease'.)

Late-onset Alzheimer disease — The genetic basis of late-onset AD is more complex, with susceptibility likely conferred by a variety of more common but less penetrant genetic factors interacting with environmental and epigenetic influences. The most firmly established genetic risk factor for late-onset AD is APOE; carriers of one e4 allele are at two- to threefold increased odds of developing AD compared with noncarriers, and those with two e4 alleles are at approximately 8- to 12-fold increased odds. The strength of the association may be modified by several factors, including sex, race, and vascular risk factors. (See "Genetics of Alzheimer disease", section on 'Apolipoprotein E'.)

Many more candidate susceptibility genes have been identified through genome-wide association studies and other techniques. The average allelic summary odds ratios for these candidates are modest, ranging from approximately 1.1 to 1.5 in most cases. (See "Genetics of Alzheimer disease", section on 'Other candidate genes'.)

Family history — Family history of dementia is a risk factor for the development of AD; patients with a first-degree relative with dementia have a 10 to 30 percent increased risk of developing the disorder [78]. Individuals in families with two or more affected siblings with late-onset AD have a threefold increased risk of AD compared with the general population [79].

One study found that the increased risk in first-degree relatives was lower if the patient developed AD later in life (age 85 or older); their risk was similar to that of a control group [80]. Additionally, the excess risk of AD in relatives of patients with early-onset, nonmutation-associated disease was highest when the relatives were younger and diminished as the relatives aged: The hazard rate ratio of developing AD in relatives of a patient with early-onset disease was 19.7 when the relative was ages 50 to 54 but decreased to 1.2 by ages 90 to 94.

First-degree relatives of African Americans with AD appear to have a higher cumulative risk of dementia than relatives of White Americans. This was illustrated in an epidemiologic study of 17,639 first-degree biologic relatives of 2339 White probands and 2281 first-degree biologic relatives of 255 African American probands with AD [81]. The cumulative risk of the disorder in the African American and White relatives was 43.7 and 26.9 percent, respectively (relative risk [RR] 1.6, 95% CI 1.4-1.9).

ACQUIRED RISK FACTORS — A variety of polygenic or acquired factors influence risk for dementia and perhaps specifically the risk for AD, including hypertension, dyslipidemia, cerebrovascular disease, altered glucose metabolism, and brain trauma. Many of these risk factors appear to be most relevant when they are present in midlife [82,83]. In one study, individuals with two or more vascular risk factors in midlife had nearly threefold higher odds of brain amyloid deposition in late life as measured by amyloid positron emission tomography (PET) [82].

As such, aggressive management of vascular risk factors during midlife represents a key strategy for reducing risk, progression, and severity of AD and other forms of dementia [84,85]. It has been estimated that up to one-third of AD cases worldwide might be attributable to modifiable risk factors such as diabetes, midlife hypertension, and physical inactivity [86].

The discussion of risk factors for dementia (not specific to AD) is provided separately. (See "Risk factors for cognitive decline and dementia".)

Hypertension — Midlife hypertension has been consistently associated with risk of dementia and AD in cross-sectional and longitudinal cohort studies [83,87-91]. The risk is probably mediated through cerebrovascular disease and the long-term effects of elevated blood pressure, smoking, and diabetes [83,92-94]. The association may be stronger in females than males, although this finding has not been consistent [91].

Arteriolosclerosis and blood pressure variability may play an important role in the relationship between blood pressure and the risk of AD [95-97]. Cerebral arteriolosclerosis increases with age, blood pressure levels, and possibly diabetes and smoking [98]. In a cohort of dementia-free older adults ages 83 to 96 years, for example, arterial stiffness was directly related to amyloid beta plaque deposition, independent of blood pressure and apolipoprotein E (APOE) genotype [99,100]. Other studies have shown similar associations [101].

While observational studies suggest that treatment of hypertension reduces the risk of dementia, clinical trials of antihypertensive drug therapies have not consistently demonstrated a reduction in the risk of dementia. However, most trials have measured short-term outcomes, and it remains possible that long-term treatment of hypertension, particularly beginning in midlife, alters AD risk later in life. Studies evaluating antihypertensive treatment in the prevention of dementia are discussed separately. (See "Prevention of dementia", section on 'Antihypertensive therapy'.)

Dyslipidemia — While some epidemiologic studies have reported an association between total or low-density lipoprotein cholesterol (LDL-C) levels, especially in midlife, and risk of AD, the interpretation of these studies is complex. Peripheral blood LDL-C does not cross the blood-brain barrier unless the barrier is damaged by factors such as vascular disease. Most of the cholesterol in the brain is synthesized by astrocytes and neurons and provided to brain cells by high-density lipoprotein (HDL) complexes, and there is little or no LDL-C in the brain [102-106].

Nonetheless, observational data generally support an association between elevated LDL-C or total cholesterol at midlife and risk of AD. In one longitudinal cohort study, high total cholesterol at midlife (mean age 50 years) conferred threefold higher odds of AD, independent of APOE genotype, education level, smoking status, and alcohol consumption [107]. Other studies have reported similar results [87,108]. When late-life cholesterol levels are examined, results have been mixed, with some studies finding a positive association between cholesterol and AD or dementia risk [109-111], and others finding either no correlation [112] or an inverse relationship [113,114]. Elevated total cholesterol and LDL-C have also been associated with higher rates of cognitive decline in patients with AD [109].

It is speculated that brain cholesterol may increase the risk of AD by enhancing the formation and/or deposition of amyloid beta, or that it may affect nonamyloid factors such as cerebrovascular risk, local inflammation, or tau metabolism [115].

Based on these observed associations, there has been considerable interest in cholesterol-lowering therapy as a potential preventive therapy for AD. Observational data have been mixed, however, and randomized trials in patients with cardiovascular disease or at higher risk of cardiovascular disease have not shown a protective effect of statins for cognition (although these studies were not designed to identify incident dementia as a primary endpoint). Oral statins also do not affect the rate of decline in mild to moderate AD.

Cerebrovascular disease — Cerebrovascular disease and AD frequently coexist, and the co-occurrence of AD pathologic changes and vascular brain injury is a common form of mixed dementia. Cerebral small vessel disease is common in aging, does not occur in isolation, and is often comorbid with AD. Risk factors include hypertension, diabetes, smoking, obesity, low physical activity, and hyperlipidemia [116].

Decreased blood flow before beta amyloid deposition has been observed in both mouse models of AD and human studies and has been proposed to contribute directly to amyloid accumulation, probably by impairment of clearance of amyloid. Vascular factors may also contribute to the breakdown of the blood-brain barrier [117]. In a population-based cohort study, decreased cerebral blood flow (CBF) was associated with an increased risk of AD, particularly in combination with white matter abnormalities [118].

In autopsy series, vascular brain injury is found in 34 to 50 percent of those with AD pathologic changes [119-127]. Conversely, approximately one-third of patients diagnosed with vascular dementia will have AD pathologic changes at autopsy [122]. Using the consensus definitions of AD pathologic changes and vascular brain injury, patients with a diagnosis of dementia are more likely to have combined pathologic features rather than either AD or vascular brain injury in isolation [128]. One large autopsy study found that coexistence of lacunar and large infarcts was associated with higher likelihood of a clinical diagnosis of AD dementia only when the AD pathology burden was low [129].

The APOE epsilon 4 (e4) allele is associated with an increased risk of cardiovascular disease and with AD [120,130]. In an autopsy study of 99 individuals, the severity of coronary artery disease was significantly associated with the density of AD neuropathologic features, primarily in APOE e4 carriers [131]. In another autopsy series, APOE e4 was associated with subcortical microvascular changes but not gross cerebrovascular pathology in patients with AD [132].

Cerebrovascular disease has been associated with worse cognitive performance in patients with AD, and clinicopathologic studies suggest that cerebrovascular disease lowers the threshold for clinical dementia in patients with a neuropathologic diagnosis of AD [74,119,133-141]. Numerous studies have found an increased risk of AD in association with various neuroimaging or pathologic markers of cerebrovascular disease, including atherosclerosis in the circle of Willis [142-145], periventricular white matter lesions [138,146-148], cerebral microbleeds [149], and cortical infarcts [138,139,150]. From these and other studies, some conclude that a vascular mechanism may be a primary etiologic factor in AD [151].

Peripheral and cardiovascular atherosclerosis — Markers of peripheral atherosclerotic disease burden, including measures of carotid intima media thickness and coronary artery calcification, have also been associated with an increased risk of dementia and AD. It is possible that such associations are modulated by arteriosclerosis and vascular brain disease, especially white matter lesions and microinfarcts in the brain [152,153].

Type 2 diabetes and obesity — Obesity and type 2 diabetes have been associated with an approximately 1.5-fold increased risk of AD [82,154,155]. As with other vascular risk factors, the data are strongest for midlife obesity and more mixed for late-life body mass index. (See "Risk factors for cognitive decline and dementia", section on 'Obesity and body mass index'.)

The direct effects of hyperinsulinemia and insulin resistance in the brain, as well as a possible relationship between insulin and amyloid beta metabolism, are areas of active investigation [155-164]. Some studies have suggested a role of insulin-degrading enzyme (IDE), which metabolizes both amyloid beta and insulin, in the accumulation of oligomeric beta amyloid [165]. Other studies are investigating a pathogenic role for the accumulation of advanced glycation end products within brain tissue [166].

Small pilot studies of intranasal insulin and dietary manipulation in patients with mild cognitive impairment or AD showing improved cognitive function or biochemical profiles provide further support for the potential link between brain insulin and AD pathogenesis [167-169]. Larger follow-up studies are underway. (See "Mild cognitive impairment: Prognosis and treatment", section on 'Others'.)

Additional data supporting these associations in dementia more broadly are discussed separately. (See "Risk factors for cognitive decline and dementia", section on 'Cardiometabolic risk factors' and "Estrogen and cognitive function".)

Lifestyle and activity — Physically active individuals have lower incidence and prevalence of cognitive decline and dementia, including AD [170-173]. A meta-analysis of 16 prospective studies suggested a 28 percent reduction in overall dementia and a 45 percent reduction in AD among those who were physically active compared with those who were less active, even after adjusting for confounding variables [174].

The Alzheimer's Association and the World Dementia Council reviewed the evidence for modifiable risk factors for cognitive decline and dementia and concluded that sufficient evidence supports the link between regular physical activity and management of cardiovascular risk factors (diabetes, obesity, smoking, and hypertension) and reduced risk of cognitive decline and possibly dementia [175]. They also found strong evidence to conclude that a healthy diet and lifelong learning (cognitive training) may also reduce the risk of cognitive decline.

Small clinical trials have also shown that exercise improves cognitive function and may also increase hippocampal and total brain volume [171]. Most of these trials were of short duration, however, and no trials have evaluated the effects of sustained exercise on the long-term risk of AD [176].

Studies supporting an association between other lifestyle factors and dementia, potential mechanisms by which lifestyle factors might influence risk, and prevention strategies based on these data are discussed separately. (See "Risk factors for cognitive decline and dementia", section on 'Lifestyle and activity' and "Prevention of dementia", section on 'Lifestyle and activity'.)

Brain trauma — History of severe brain trauma with loss of consciousness of 30 minutes or more has been inconsistently associated with an increased risk of AD [177-180]. A causal relationship has been suggested by the finding of increased amyloid in the brain shortly after severe brain trauma [179]. However, at least two well-designed cohort studies have not confirmed an association between traumatic brain injury (TBI) and risk of AD or AD biomarkers [181]. In one autopsy study examining 1589 brains, no association was found between self-reported TBI and neuropathologic changes consistent with AD nor with incident AD [180]. Similarly, A Veterans Affairs study did not find a relationship between TBI and PET amyloid deposition [182].

Repeated milder forms of head trauma have been associated with neuropathologic changes of a tauopathy that is distinct from AD. (See "Sequelae of mild traumatic brain injury", section on 'Chronic traumatic encephalopathy'.)

The association of TBI with cognitive decline and dementia more generally is discussed separately. (See "Risk factors for cognitive decline and dementia", section on 'Head trauma'.)

Medications — Multiple studies have found an association between use of certain medication classes (eg, benzodiazepines, anticholinergics, antihistamines, opioids) and cognitive impairment in older adults, but the effects have been presumed to be transient and reversible [183]. Long-term exposure has been linked to an elevated risk of AD or all-cause dementia in several large studies, however, raising the possibility that cognitive effects may not be reversible in some patients or that these exposures somehow enhance the penetrance of presymptomatic AD.

Benzodiazepines – Available data on the association between benzodiazepine use and dementia risk are conflicting. In a nested case-control study of nearly 2000 older adults enrolled in a public drug plan in Canada, use of benzodiazepines for >180 days was associated with a 1.5-fold increase in risk of AD after adjusting for multiple potential confounders, including anxiety, depression, and insomnia [184]. A dose-response effect was observed, with longer exposure and longer half-life drugs associated with increased risk. It remains possible that benzodiazepines were used to treat prodromal symptoms of dementia, however, even though the study limited exposure to prescriptions begun at least five years before the diagnosis of AD [185,186].

Other large observational studies have failed to find a significant association between long-term benzodiazepine use and incident dementia; these are discussed separately. (See "Risk factors for cognitive decline and dementia", section on 'Medications'.)

Anticholinergics – The case for anticholinergics increasing the risk of irreversible effects is probably stronger [187,188] and is plausible given the prominence of cholinergic deficits in AD. Studies associating these medications with incident dementia are discussed separately. (See "Risk factors for cognitive decline and dementia", section on 'Medications'.)

Proton pump inhibitors – There are conflicting data on the association between proton pump inhibitors (PPIs) and risk of AD. In one longitudinal study, PPI use had similar associations with all-cause dementia (hazard ratio [HR] 1.3) and with AD dementia (HR 1.4) [189].

By contrast, two other observational studies, including a large case-control study with over 70,000 cases of AD and over 280,000 age-, sex-, and region-matched controls, found no association between PPI use and AD, nor evidence of a dose-response relationship in either dose or duration of use [190,191].

Limited preclinical data suggest a possible interaction between PPIs and both amyloid and tau [192-194]. Alternatively, malabsorption of vitamin B12 or other nutrients due to long-term PPI use could play a role [195]. On the other hand, the association may reflect residual confounding by factors related to both use of PPIs and the development of dementia, and more studies are needed [196].

ENVIRONMENTAL RISK FACTORS — As large-scale genetic studies have yielded limited predictive risk alleles beyond apolipoprotein E (APOE) epsilon 4 (e4), there is a renewed interest in environmental and toxic exposures as potential risk factors for AD [197]. Areas of investigation include the following:

Secondhand smoke – In a cross-sectional study of 2692 never-smokers aged 60 years and older in China, secondhand smoke exposure was associated with an increased risk of AD (adjusted relative risk [RR] 2.28, 95% CI 1.82-2.84) [198]. Risk was higher for in-home exposure compared with at-work exposure.

Air pollution – Animal studies [199,200] and limited human epidemiologic data provide support for air pollution as a potential risk factor for AD. An increase in diffuse amyloid plaques and inflammation in the olfactory bulb, hippocampus, and frontal lobes have been described in brain autopsy specimens from adults living in Mexico City and Monterrey, areas known for high levels of air pollution, compared with adults living in smaller cities in Mexico with lower levels of air pollution [201]. Similar findings have also been reported in children and young adults [202].

Pesticides – Several studies have implicated occupational or environmental exposure to pesticides as a risk factor for AD [203,204]. As an example, a case-control study examined serum dichlorodiphenyldichloroethylene (DDE) levels in 86 pathologically confirmed AD cases and 79 controls and found that levels of DDE were 3.8-fold higher in those with AD [204].

SUMMARY

Hallmark neuropathologic features – Neuropathologic changes of Alzheimer disease (AD) include neuritic plaques, extracellular deposits of amyloid beta, and neurofibrillary degeneration. (See 'Pathology' above.)

Pathogenesis – While the pathogenesis of AD remains unclear, all forms of AD appear to share overproduction and/or decreased clearance of a family of proteins known as amyloid beta peptides. The pathogenesis of AD also involves tau, a microtubule-associated protein that aids in microtubule assembly. (See 'Pathogenesis' above.)

Case definition – A definitive diagnosis of AD requires histopathologic examination. Clinical criteria for the diagnosis of AD have evolved over time, and the ability to accurately diagnose AD has improved with the development of techniques for in vivo measurement of pathophysiologic features of AD. The diagnosis of AD requires a decline in both cognition, especially memory, and function, as well as specific neuropathology. (See 'Case definition' above.)

Presymptomatic disease – There is a long presymptomatic period between the onset of biochemical changes in the brain and the development of clinical symptoms of AD, suggesting that long-term epidemiologic studies beginning at an early age are needed to properly study the gene-lifestyle environmental determinants of amyloid vascular disease and neurodegeneration. (See 'Presymptomatic period' above.)

Epidemiology and risk factors – AD is increasingly prevalent with advancing age, and the overall burden of AD is substantial worldwide. It has been estimated that the global prevalence of dementia will rise to >100 million by 2050.

Advanced age – The age-standardized prevalence of dementia ranges from 5 to 7 percent in most regions of the world. (See 'Incidence and prevalence' above.)

Genetic factors Aside from age, the most clearly established risk factors for AD are a family history of dementia, rare dominantly inherited mutations in genes that impact amyloid in the brain, and the apolipoprotein E (APOE) epsilon 4 (e4) allele. (See 'Genetic risk factors' above.)

Vascular risk factors Risk factors for vascular disease, including hypertension, obesity, and diabetes, increase the risk of dementia and may impact AD, particularly when they are present in midlife. The pathogenic mechanisms linking various cardiometabolic factors and AD are not well understood, and both vascular and nonvascular mechanisms may be involved. (See 'Hypertension' above and 'Type 2 diabetes and obesity' above.)

Brain cholesterol metabolism may be an important determinant of AD. The relationship between diet, genetics, blood lipoproteins levels, and AD is complex and inconsistent. (See 'Dyslipidemia' above.)

Cerebrovascular disease and AD frequently coexist. Hypertension is the primary risk factor for vascular brain injury. Cerebrovascular disease is associated with worse cognitive performance in patients with AD, and clinicopathologic studies suggest that cerebrovascular disease lowers the threshold for clinical dementia in patients with a neuropathologic diagnosis of AD. (See 'Cerebrovascular disease' above.)

Lifestyle risk factors – Accumulating data suggest that social, cognitive, and physical activities are inversely associated with the risk of AD and other forms of dementia, and there is considerable interest in their potential as preventive strategies. (See "Risk factors for cognitive decline and dementia", section on 'Lifestyle and activity' and "Prevention of dementia", section on 'Lifestyle and activity'.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Lewis H Kuller, MD, DrPH (deceased), who contributed to earlier versions of this topic review.

  1. Gandy S, DeKosky ST. Toward the treatment and prevention of Alzheimer's disease: rational strategies and recent progress. Annu Rev Med 2013; 64:367.
  2. Kuller LH, Lopez OL. Dementia and Alzheimer's disease: a new direction.The 2010 Jay L. Foster Memorial Lecture. Alzheimers Dement 2011; 7:540.
  3. Montine TJ, Phelps CH, Beach TG, et al. National Institute on Aging-Alzheimer's Association guidelines for the neuropathologic assessment of Alzheimer's disease: a practical approach. Acta Neuropathol 2012; 123:1.
  4. Hyman BT, Phelps CH, Beach TG, et al. National Institute on Aging-Alzheimer's Association guidelines for the neuropathologic assessment of Alzheimer's disease. Alzheimers Dement 2012; 8:1.
  5. Masliah E, Terry RD, Mallory M, et al. Diffuse plaques do not accentuate synapse loss in Alzheimer's disease. Am J Pathol 1990; 137:1293.
  6. Masliah E, Mallory M, Deerinck T, et al. Re-evaluation of the structural organization of neuritic plaques in Alzheimer's disease. J Neuropathol Exp Neurol 1993; 52:619.
  7. Terry RD, Masliah E, Salmon DP, et al. Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol 1991; 30:572.
  8. DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer's disease: correlation with cognitive severity. Ann Neurol 1990; 27:457.
  9. Thal DR, Rüb U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology 2002; 58:1791.
  10. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991; 82:239.
  11. Braak H, Alafuzoff I, Arzberger T, et al. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol 2006; 112:389.
  12. Mirra SS, Heyman A, McKeel D, et al. The Consortium to Establish a Registry for Alzheimer's Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer's disease. Neurology 1991; 41:479.
  13. Vonsattel JP, Myers RH, Hedley-Whyte ET, et al. Cerebral amyloid angiopathy without and with cerebral hemorrhages: a comparative histological study. Ann Neurol 1991; 30:637.
  14. Thal DR, Griffin WS, de Vos RA, Ghebremedhin E. Cerebral amyloid angiopathy and its relationship to Alzheimer's disease. Acta Neuropathol 2008; 115:599.
  15. Hamilton RL. Lewy bodies in Alzheimer's disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry. Brain Pathol 2000; 10:378.
  16. Uchikado H, Lin WL, DeLucia MW, Dickson DW. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol 2006; 65:685.
  17. Lippa CF, Duda JE, Grossman M, et al. DLB and PDD boundary issues: diagnosis, treatment, molecular pathology, and biomarkers. Neurology 2007; 68:812.
  18. Leverenz JB, Fishel MA, Peskind ER, et al. Lewy body pathology in familial Alzheimer disease: evidence for disease- and mutation-specific pathologic phenotype. Arch Neurol 2006; 63:370.
  19. Amador-Ortiz C, Dickson DW. Neuropathology of hippocampal sclerosis. Handb Clin Neurol 2008; 89:569.
  20. James BD, Wilson RS, Boyle PA, et al. TDP-43 stage, mixed pathologies, and clinical Alzheimer's-type dementia. Brain 2016.
  21. Wilson AC, Dugger BN, Dickson DW, Wang DS. TDP-43 in aging and Alzheimer's disease - a review. Int J Clin Exp Pathol 2011; 4:147.
  22. Josephs KA, Dickson DW, Tosakulwong N, et al. Rates of hippocampal atrophy and presence of post-mortem TDP-43 in patients with Alzheimer's disease: a longitudinal retrospective study. Lancet Neurol 2017; 16:917.
  23. Josephs KA, Whitwell JL, Knopman DS, et al. Abnormal TDP-43 immunoreactivity in AD modifies clinicopathologic and radiologic phenotype. Neurology 2008; 70:1850.
  24. Gremer L, Schölzel D, Schenk C, et al. Fibril structure of amyloid-β(1-42) by cryo-electron microscopy. Science 2017; 358:116.
  25. Guo JL, Lee VM. Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J Biol Chem 2011; 286:15317.
  26. Iba M, Guo JL, McBride JD, et al. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer's-like tauopathy. J Neurosci 2013; 33:1024.
  27. Medina M, Avila J. The role of extracellular Tau in the spreading of neurofibrillary pathology. Front Cell Neurosci 2014; 8:113.
  28. Castellano JM, Kim J, Stewart FR, et al. Human apoE isoforms differentially regulate brain amyloid-β peptide clearance. Sci Transl Med 2011; 3:89ra57.
  29. Lopez OL, Becker JT, Kuller LH. Patterns of compensation and vulnerability in normal subjects at risk of Alzheimer's disease. J Alzheimers Dis 2013; 33 Suppl 1:S427.
  30. Kuller LH, Lopez OL. Commentary on "Developing a national strategy to prevent dementia: Leon Thal Symposium 2009." Is dementia among older individuals 75+ a unique disease? Alzheimers Dement 2010; 6:142.
  31. Bennett DA, Launer LJ. Longitudinal epidemiologic clinical-pathologic studies of aging and Alzheimer's disease. Curr Alzheimer Res 2012; 9:617.
  32. Bennett DA, Schneider JA, Arvanitakis Z, Wilson RS. Overview and findings from the religious orders study. Curr Alzheimer Res 2012; 9:628.
  33. Bennett DA, Schneider JA, Buchman AS, et al. Overview and findings from the rush Memory and Aging Project. Curr Alzheimer Res 2012; 9:646.
  34. Corrada MM, Berlau DJ, Kawas CH. A population-based clinicopathological study in the oldest-old: the 90+ study. Curr Alzheimer Res 2012; 9:709.
  35. Gabel MJ, Foster NL, Heidebrink JL, et al. Validation of consensus panel diagnosis in dementia. Arch Neurol 2010; 67:1506.
  36. Mortimer JA. The Nun Study: risk factors for pathology and clinical-pathologic correlations. Curr Alzheimer Res 2012; 9:621.
  37. Murray ME, Graff-Radford NR, Ross OA, et al. Neuropathologically defined subtypes of Alzheimer's disease with distinct clinical characteristics: a retrospective study. Lancet Neurol 2011; 10:785.
  38. Richardson K, Stephan BC, Ince PG, et al. The neuropathology of vascular disease in the Medical Research Council Cognitive Function and Ageing Study (MRC CFAS). Curr Alzheimer Res 2012; 9:687.
  39. Zaccai J, Ince P, Brayne C. Population-based neuropathological studies of dementia: design, methods and areas of investigation--a systematic review. BMC Neurol 2006; 6:2.
  40. Gelber RP, Launer LJ, White LR. The Honolulu-Asia Aging Study: epidemiologic and neuropathologic research on cognitive impairment. Curr Alzheimer Res 2012; 9:664.
  41. Rocca WA, Petersen RC, Knopman DS, et al. Trends in the incidence and prevalence of Alzheimer's disease, dementia, and cognitive impairment in the United States. Alzheimers Dement 2011; 7:80.
  42. Ganguli M, Hendrie HC. Screening for cognitive impairment and depression in ethnically diverse older populations. Alzheimer Dis Assoc Disord 2005; 19:275.
  43. Llibre Rodriguez JJ, Ferri CP, Acosta D, et al. Prevalence of dementia in Latin America, India, and China: a population-based cross-sectional survey. Lancet 2008; 372:464.
  44. Prince M, Bryce R, Albanese E, et al. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement 2013; 9:63.
  45. Sosa-Ortiz AL, Acosta-Castillo I, Prince MJ. Epidemiology of dementias and Alzheimer's disease. Arch Med Res 2012; 43:600.
  46. Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010-2050) estimated using the 2010 census. Neurology 2013; 80:1778.
  47. World Alzheimer Report 2015: The Global Impact of Dementia http://www.alz.co.uk/research/world-report-2015 (Accessed on October 30, 2015).
  48. Yuan J, Zhang Z, Wen H, et al. Incidence of dementia and subtypes: A cohort study in four regions in China. Alzheimers Dement 2016; 12:262.
  49. Mehta KM, Yeo GW. Systematic review of dementia prevalence and incidence in United States race/ethnic populations. Alzheimers Dement 2017; 13:72.
  50. Mayeda ER, Glymour MM, Quesenberry CP, Whitmer RA. Inequalities in dementia incidence between six racial and ethnic groups over 14 years. Alzheimers Dement 2016; 12:216.
  51. Kuller LH, Lopez OL, Becker JT, et al. Risk of dementia and death in the long-term follow-up of the Pittsburgh Cardiovascular Health Study-Cognition Study. Alzheimers Dement 2016; 12:170.
  52. Fitzpatrick AL, Kuller LH, Ives DG, et al. Incidence and prevalence of dementia in the Cardiovascular Health Study. J Am Geriatr Soc 2004; 52:195.
  53. Schrijvers EM, Verhaaren BF, Koudstaal PJ, et al. Is dementia incidence declining?: Trends in dementia incidence since 1990 in the Rotterdam Study. Neurology 2012; 78:1456.
  54. Tinetti ME, McAvay GJ, Murphy TE, et al. Contribution of individual diseases to death in older adults with multiple diseases. J Am Geriatr Soc 2012; 60:1448.
  55. James BD, Leurgans SE, Hebert LE, et al. Contribution of Alzheimer disease to mortality in the United States. Neurology 2014; 82:1045.
  56. Nordström P, Nordström A, Eriksson M, et al. Risk factors in late adolescence for young-onset dementia in men: a nationwide cohort study. JAMA Intern Med 2013; 173:1612.
  57. Rossor MN, Fox NC, Mummery CJ, et al. The diagnosis of young-onset dementia. Lancet Neurol 2010; 9:793.
  58. Harvey RJ, Skelton-Robinson M, Rossor MN. The prevalence and causes of dementia in people under the age of 65 years. J Neurol Neurosurg Psychiatry 2003; 74:1206.
  59. Mercy L, Hodges JR, Dawson K, et al. Incidence of early-onset dementias in Cambridgeshire, United Kingdom. Neurology 2008; 71:1496.
  60. Larson EB, Yaffe K, Langa KM. New insights into the dementia epidemic. N Engl J Med 2013; 369:2275.
  61. Christensen K, Thinggaard M, Oksuzyan A, et al. Physical and cognitive functioning of people older than 90 years: a comparison of two Danish cohorts born 10 years apart. Lancet 2013; 382:1507.
  62. Matthews FE, Arthur A, Barnes LE, et al. A two-decade comparison of prevalence of dementia in individuals aged 65 years and older from three geographical areas of England: results of the Cognitive Function and Ageing Study I and II. Lancet 2013; 382:1405.
  63. Matthews FE, Stephan BC, Robinson L, et al. A two decade dementia incidence comparison from the Cognitive Function and Ageing Studies I and II. Nat Commun 2016; 7:11398.
  64. Kövari E, Herrmann FR, Bouras C, Gold G. Amyloid deposition is decreasing in aging brains: an autopsy study of 1,599 older people. Neurology 2014; 82:326.
  65. Grasset L, Brayne C, Joly P, et al. Trends in dementia incidence: Evolution over a 10-year period in France. Alzheimers Dement 2016; 12:272.
  66. Ahmadi-Abhari S, Guzman-Castillo M, Bandosz P, et al. Temporal trend in dementia incidence since 2002 and projections for prevalence in England and Wales to 2040: modelling study. BMJ 2017; 358:j2856.
  67. Satizabal CL, Beiser AS, Chouraki V, et al. Incidence of Dementia over Three Decades in the Framingham Heart Study. N Engl J Med 2016; 374:523.
  68. Taylor CA, Greenlund SF, McGuire LC, et al. Deaths from Alzheimer's Disease - United States, 1999-2014. MMWR Morb Mortal Wkly Rep 2017; 66:521.
  69. Amieva H, Mokri H, Le Goff M, et al. Compensatory mechanisms in higher-educated subjects with Alzheimer's disease: a study of 20 years of cognitive decline. Brain 2014; 137:1167.
  70. Verlinden VJ, van der Geest JN, de Bruijn RF, et al. Trajectories of decline in cognition and daily functioning in preclinical dementia. Alzheimers Dement 2016; 12:144.
  71. Bateman RJ, Xiong C, Benzinger TL, et al. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N Engl J Med 2012; 367:795.
  72. Bassett SS, Yousem DM, Cristinzio C, et al. Familial risk for Alzheimer's disease alters fMRI activation patterns. Brain 2006; 129:1229.
  73. Roberts RO, Aakre JA, Kremers WK, et al. Prevalence and Outcomes of Amyloid Positivity Among Persons Without Dementia in a Longitudinal, Population-Based Setting. JAMA Neurol 2018; 75:970.
  74. Vemuri P, Lesnick TG, Przybelski SA, et al. Vascular and amyloid pathologies are independent predictors of cognitive decline in normal elderly. Brain 2015; 138:761.
  75. Roberts RO, Knopman DS, Syrjanen JA, et al. Weighting and standardization of frequencies to determine prevalence of AD imaging biomarkers. Neurology 2017; 89:2039.
  76. Lopez OL, Becker JT, Chang Y, et al. Amyloid deposition and brain structure as long-term predictors of MCI, dementia, and mortality. Neurology 2018; 90:e1920.
  77. Chen Y, Sillaire AR, Dallongeville J, et al. Low Prevalence and Clinical Effect of Vascular Risk Factors in Early-Onset Alzheimer's Disease. J Alzheimers Dis 2017; 60:1045.
  78. van Duijn CM, Clayton D, Chandra V, et al. Familial aggregation of Alzheimer's disease and related disorders: a collaborative re-analysis of case-control studies. Int J Epidemiol 1991; 20 Suppl 2:S13.
  79. Vardarajan BN, Faber KM, Bird TD, et al. Age-specific incidence rates for dementia and Alzheimer disease in NIA-LOAD/NCRAD and EFIGA families: National Institute on Aging Genetics Initiative for Late-Onset Alzheimer Disease/National Cell Repository for Alzheimer Disease (NIA-LOAD/NCRAD) and Estudio Familiar de Influencia Genetica en Alzheimer (EFIGA). JAMA Neurol 2014; 71:315.
  80. Silverman JM, Smith CJ, Marin DB, et al. Familial patterns of risk in very late-onset Alzheimer disease. Arch Gen Psychiatry 2003; 60:190.
  81. Green RC, Cupples LA, Go R, et al. Risk of dementia among white and African American relatives of patients with Alzheimer disease. JAMA 2002; 287:329.
  82. Gottesman RF, Schneider AL, Zhou Y, et al. Association Between Midlife Vascular Risk Factors and Estimated Brain Amyloid Deposition. JAMA 2017; 317:1443.
  83. Gottesman RF, Albert MS, Alonso A, et al. Associations Between Midlife Vascular Risk Factors and 25-Year Incident Dementia in the Atherosclerosis Risk in Communities (ARIC) Cohort. JAMA Neurol 2017; 74:1246.
  84. Smith AD, Yaffe K. Dementia (including Alzheimer's disease) can be prevented: statement supported by international experts. J Alzheimers Dis 2014; 38:699.
  85. Banerjee S. Good news on dementia prevalence--we can make a difference. Lancet 2013; 382:1384.
  86. Norton S, Matthews FE, Barnes DE, et al. Potential for primary prevention of Alzheimer's disease: an analysis of population-based data. Lancet Neurol 2014; 13:788.
  87. Whitmer RA, Sidney S, Selby J, et al. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology 2005; 64:277.
  88. Freitag MH, Peila R, Masaki K, et al. Midlife pulse pressure and incidence of dementia: the Honolulu-Asia Aging Study. Stroke 2006; 37:33.
  89. Alonso A, Mosley TH Jr, Gottesman RF, et al. Risk of dementia hospitalisation associated with cardiovascular risk factors in midlife and older age: the Atherosclerosis Risk in Communities (ARIC) study. J Neurol Neurosurg Psychiatry 2009; 80:1194.
  90. Skoog I, Lernfelt B, Landahl S, et al. 15-year longitudinal study of blood pressure and dementia. Lancet 1996; 347:1141.
  91. Gilsanz P, Mayeda ER, Glymour MM, et al. Female sex, early-onset hypertension, and risk of dementia. Neurology 2017; 89:1886.
  92. Beason-Held LL, Thambisetty M, Deib G, et al. Baseline cardiovascular risk predicts subsequent changes in resting brain function. Stroke 2012; 43:1542.
  93. Kaffashian S, Dugravot A, Elbaz A, et al. Predicting cognitive decline: a dementia risk score vs. the Framingham vascular risk scores. Neurology 2013; 80:1300.
  94. Debette S, Seshadri S, Beiser A, et al. Midlife vascular risk factor exposure accelerates structural brain aging and cognitive decline. Neurology 2011; 77:461.
  95. Oishi E, Ohara T, Sakata S, et al. Day-to-Day Blood Pressure Variability and Risk of Dementia in a General Japanese Elderly Population: The Hisayama Study. Circulation 2017; 136:516.
  96. Poels MM, Zaccai K, Verwoert GC, et al. Arterial stiffness and cerebral small vessel disease: the Rotterdam Scan Study. Stroke 2012; 43:2637.
  97. Rabkin SW. Arterial stiffness: detection and consequences in cognitive impairment and dementia of the elderly. J Alzheimers Dis 2012; 32:541.
  98. Cavalcante JL, Lima JA, Redheuil A, Al-Mallah MH. Aortic stiffness: current understanding and future directions. J Am Coll Cardiol 2011; 57:1511.
  99. Hughes TM, Kuller LH, Barinas-Mitchell EJ, et al. Pulse wave velocity is associated with β-amyloid deposition in the brains of very elderly adults. Neurology 2013; 81:1711.
  100. Hughes TM, Kuller LH, Barinas-Mitchell EJ, et al. Arterial stiffness and β-amyloid progression in nondemented elderly adults. JAMA Neurol 2014; 71:562.
  101. Nation DA, Edmonds EC, Bangen KJ, et al. Pulse pressure in relation to tau-mediated neurodegeneration, cerebral amyloidosis, and progression to dementia in very old adults. JAMA Neurol 2015; 72:546.
  102. Leduc V, Jasmin-Bélanger S, Poirier J. APOE and cholesterol homeostasis in Alzheimer's disease. Trends Mol Med 2010; 16:469.
  103. Beel AJ, Sakakura M, Barrett PJ, Sanders CR. Direct binding of cholesterol to the amyloid precursor protein: An important interaction in lipid-Alzheimer's disease relationships? Biochim Biophys Acta 2010; 1801:975.
  104. Martin M, Dotti CG, Ledesma MD. Brain cholesterol in normal and pathological aging. Biochim Biophys Acta 2010; 1801:934.
  105. Shobab LA, Hsiung GY, Feldman HH. Cholesterol in Alzheimer's disease. Lancet Neurol 2005; 4:841.
  106. Grösgen S, Grimm MO, Friess P, Hartmann T. Role of amyloid beta in lipid homeostasis. Biochim Biophys Acta 2010; 1801:966.
  107. Kivipelto M, Helkala EL, Laakso MP, et al. Apolipoprotein E epsilon4 allele, elevated midlife total cholesterol level, and high midlife systolic blood pressure are independent risk factors for late-life Alzheimer disease. Ann Intern Med 2002; 137:149.
  108. Solomon A, Kåreholt I, Ngandu T, et al. Serum cholesterol changes after midlife and late-life cognition: twenty-one-year follow-up study. Neurology 2007; 68:751.
  109. Helzner EP, Luchsinger JA, Scarmeas N, et al. Contribution of vascular risk factors to the progression in Alzheimer disease. Arch Neurol 2009; 66:343.
  110. Notkola IL, Sulkava R, Pekkanen J, et al. Serum total cholesterol, apolipoprotein E epsilon 4 allele, and Alzheimer's disease. Neuroepidemiology 1998; 17:14.
  111. Yaffe K, Barrett-Connor E, Lin F, Grady D. Serum lipoprotein levels, statin use, and cognitive function in older women. Arch Neurol 2002; 59:378.
  112. Reitz C, Tang MX, Manly J, et al. Plasma lipid levels in the elderly are not associated with the risk of mild cognitive impairment. Dement Geriatr Cogn Disord 2008; 25:232.
  113. Reitz C, Tang MX, Luchsinger J, Mayeux R. Relation of plasma lipids to Alzheimer disease and vascular dementia. Arch Neurol 2004; 61:705.
  114. Mielke MM, Zandi PP, Sjögren M, et al. High total cholesterol levels in late life associated with a reduced risk of dementia. Neurology 2005; 64:1689.
  115. Shepardson NE, Shankar GM, Selkoe DJ. Cholesterol level and statin use in Alzheimer disease: I. Review of epidemiological and preclinical studies. Arch Neurol 2011; 68:1239.
  116. Snyder HM, Corriveau RA, Craft S, et al. Vascular contributions to cognitive impairment and dementia including Alzheimer's disease. Alzheimers Dement 2015; 11:710.
  117. Ighodaro ET, Abner EL, Fardo DW, et al. Risk factors and global cognitive status related to brain arteriolosclerosis in elderly individuals. J Cereb Blood Flow Metab 2017; 37:201.
  118. Wolters FJ, Zonneveld HI, Hofman A, et al. Cerebral Perfusion and the Risk of Dementia: A Population-Based Study. Circulation 2017; 136:719.
  119. Chui HC, Zarow C, Mack WJ, et al. Cognitive impact of subcortical vascular and Alzheimer's disease pathology. Ann Neurol 2006; 60:677.
  120. Langa KM, Foster NL, Larson EB. Mixed dementia: emerging concepts and therapeutic implications. JAMA 2004; 292:2901.
  121. Massoud F, Devi G, Stern Y, et al. A clinicopathological comparison of community-based and clinic-based cohorts of patients with dementia. Arch Neurol 1999; 56:1368.
  122. Kalaria RN, Ballard C. Overlap between pathology of Alzheimer disease and vascular dementia. Alzheimer Dis Assoc Disord 1999; 13 Suppl 3:S115.
  123. Schneider JA, Arvanitakis Z, Bang W, Bennett DA. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology 2007; 69:2197.
  124. Kalaria RN. Cerebrovascular disease and mechanisms of cognitive impairment: evidence from clinicopathological studies in humans. Stroke 2012; 43:2526.
  125. Saczynski JS, Sigurdsson S, Jonsdottir MK, et al. Cerebral infarcts and cognitive performance: importance of location and number of infarcts. Stroke 2009; 40:677.
  126. Debette S, Beiser A, DeCarli C, et al. Association of MRI markers of vascular brain injury with incident stroke, mild cognitive impairment, dementia, and mortality: the Framingham Offspring Study. Stroke 2010; 41:600.
  127. Weinstein G, Beiser AS, Decarli C, et al. Brain imaging and cognitive predictors of stroke and Alzheimer disease in the Framingham Heart Study. Stroke 2013; 44:2787.
  128. Neuropathology Group. Medical Research Council Cognitive Function and Aging Study. Pathological correlates of late-onset dementia in a multicentre, community-based population in England and Wales. Neuropathology Group of the Medical Research Council Cognitive Function and Ageing Study (MRC CFAS). Lancet 2001; 357:169.
  129. Dodge HH, Zhu J, Woltjer R, et al. Risk of incident clinical diagnosis of Alzheimer's disease-type dementia attributable to pathology-confirmed vascular disease. Alzheimers Dement 2017; 13:613.
  130. Song Y, Stampfer MJ, Liu S. Meta-analysis: apolipoprotein E genotypes and risk for coronary heart disease. Ann Intern Med 2004; 141:137.
  131. Beeri MS, Rapp M, Silverman JM, et al. Coronary artery disease is associated with Alzheimer disease neuropathology in APOE4 carriers. Neurology 2006; 66:1399.
  132. Yip AG, McKee AC, Green RC, et al. APOE, vascular pathology, and the AD brain. Neurology 2005; 65:259.
  133. Snowdon DA, Greiner LH, Mortimer JA, et al. Brain infarction and the clinical expression of Alzheimer disease. The Nun Study. JAMA 1997; 277:813.
  134. Bennett DA, Wilson RS, Arvanitakis Z, et al. Selected findings from the Religious Orders Study and Rush Memory and Aging Project. J Alzheimers Dis 2013; 33 Suppl 1:S397.
  135. Toledo JB, Arnold SE, Raible K, et al. Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer's Coordinating Centre. Brain 2013; 136:2697.
  136. Troncoso JC, Zonderman AB, Resnick SM, et al. Effect of infarcts on dementia in the Baltimore longitudinal study of aging. Ann Neurol 2008; 64:168.
  137. Riekse RG, Leverenz JB, McCormick W, et al. Effect of vascular lesions on cognition in Alzheimer's disease: a community-based study. J Am Geriatr Soc 2004; 52:1442.
  138. Kövari E, Gold G, Herrmann FR, et al. Cortical microinfarcts and demyelination affect cognition in cases at high risk for dementia. Neurology 2007; 68:927.
  139. Schneider JA, Boyle PA, Arvanitakis Z, et al. Subcortical infarcts, Alzheimer's disease pathology, and memory function in older persons. Ann Neurol 2007; 62:59.
  140. Brickman AM, Honig LS, Scarmeas N, et al. Measuring cerebral atrophy and white matter hyperintensity burden to predict the rate of cognitive decline in Alzheimer disease. Arch Neurol 2008; 65:1202.
  141. Xie S, Xiao JX, Gong GL, et al. Voxel-based detection of white matter abnormalities in mild Alzheimer disease. Neurology 2006; 66:1845.
  142. Roher AE, Esh C, Rahman A, et al. Atherosclerosis of cerebral arteries in Alzheimer disease. Stroke 2004; 35:2623.
  143. Honig LS, Kukull W, Mayeux R. Atherosclerosis and AD: analysis of data from the US National Alzheimer's Coordinating Center. Neurology 2005; 64:494.
  144. Yarchoan M, Xie SX, Kling MA, et al. Cerebrovascular atherosclerosis correlates with Alzheimer pathology in neurodegenerative dementias. Brain 2012; 135:3749.
  145. Dolan H, Crain B, Troncoso J, et al. Atherosclerosis, dementia, and Alzheimer disease in the Baltimore Longitudinal Study of Aging cohort. Ann Neurol 2010; 68:231.
  146. de Leeuw FE, Korf E, Barkhof F, Scheltens P. White matter lesions are associated with progression of medial temporal lobe atrophy in Alzheimer disease. Stroke 2006; 37:2248.
  147. Behl P, Bocti C, Swartz RH, et al. Strategic subcortical hyperintensities in cholinergic pathways and executive function decline in treated Alzheimer patients. Arch Neurol 2007; 64:266.
  148. Knopman DS, Griswold ME, Lirette ST, et al. Vascular imaging abnormalities and cognition: mediation by cortical volume in nondemented individuals: atherosclerosis risk in communities-neurocognitive study. Stroke 2015; 46:433.
  149. Akoudad S, Wolters FJ, Viswanathan A, et al. Association of Cerebral Microbleeds With Cognitive Decline and Dementia. JAMA Neurol 2016; 73:934.
  150. Schneider JA, Wilson RS, Bienias JL, et al. Cerebral infarctions and the likelihood of dementia from Alzheimer disease pathology. Neurology 2004; 62:1148.
  151. de la Torre JC. Alzheimer disease as a vascular disorder: nosological evidence. Stroke 2002; 33:1152.
  152. van Oijen M, de Jong FJ, Witteman JC, et al. Atherosclerosis and risk for dementia. Ann Neurol 2007; 61:403.
  153. Bos D, Ikram MA, Elias-Smale SE, et al. Calcification in major vessel beds relates to vascular brain disease. Arterioscler Thromb Vasc Biol 2011; 31:2331.
  154. Profenno LA, Porsteinsson AP, Faraone SV. Meta-analysis of Alzheimer's disease risk with obesity, diabetes, and related disorders. Biol Psychiatry 2010; 67:505.
  155. Biessels GJ, Staekenborg S, Brunner E, et al. Risk of dementia in diabetes mellitus: a systematic review. Lancet Neurol 2006; 5:64.
  156. Peila R, Rodriguez BL, White LR, Launer LJ. Fasting insulin and incident dementia in an elderly population of Japanese-American men. Neurology 2004; 63:228.
  157. Craft S, Watson GS. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol 2004; 3:169.
  158. Geroldi C, Frisoni GB, Paolisso G, et al. Insulin resistance in cognitive impairment: the InCHIANTI study. Arch Neurol 2005; 62:1067.
  159. Fishel MA, Watson GS, Montine TJ, et al. Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults. Arch Neurol 2005; 62:1539.
  160. Kulstad JJ, Green PS, Cook DG, et al. Differential modulation of plasma beta-amyloid by insulin in patients with Alzheimer disease. Neurology 2006; 66:1506.
  161. Rönnemaa E, Zethelius B, Sundelöf J, et al. Impaired insulin secretion increases the risk of Alzheimer disease. Neurology 2008; 71:1065.
  162. Matsuzaki T, Sasaki K, Tanizaki Y, et al. Insulin resistance is associated with the pathology of Alzheimer disease: the Hisayama study. Neurology 2010; 75:764.
  163. Schrijvers EM, Witteman JC, Sijbrands EJ, et al. Insulin metabolism and the risk of Alzheimer disease: the Rotterdam Study. Neurology 2010; 75:1982.
  164. Burns JM, Donnelly JE, Anderson HS, et al. Peripheral insulin and brain structure in early Alzheimer disease. Neurology 2007; 69:1094.
  165. Farris W, Mansourian S, Chang Y, et al. Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc Natl Acad Sci U S A 2003; 100:4162.
  166. Yaffe K, Lindquist K, Schwartz AV, et al. Advanced glycation end product level, diabetes, and accelerated cognitive aging. Neurology 2011; 77:1351.
  167. Hanson AJ, Bayer-Carter JL, Green PS, et al. Effect of apolipoprotein E genotype and diet on apolipoprotein E lipidation and amyloid peptides: randomized clinical trial. JAMA Neurol 2013; 70:972.
  168. Bayer-Carter JL, Green PS, Montine TJ, et al. Diet intervention and cerebrospinal fluid biomarkers in amnestic mild cognitive impairment. Arch Neurol 2011; 68:743.
  169. Craft S, Baker LD, Montine TJ, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol 2012; 69:29.
  170. Rolland Y, Abellan van Kan G, Vellas B. Physical activity and Alzheimer's disease: from prevention to therapeutic perspectives. J Am Med Dir Assoc 2008; 9:390.
  171. Ahlskog JE, Geda YE, Graff-Radford NR, Petersen RC. Physical exercise as a preventive or disease-modifying treatment of dementia and brain aging. Mayo Clin Proc 2011; 86:876.
  172. Vemuri P, Knopman DS, Lesnick TG, et al. Evaluation of Amyloid Protective Factors and Alzheimer Disease Neurodegeneration Protective Factors in Elderly Individuals. JAMA Neurol 2017; 74:718.
  173. Livingston G, Sommerlad A, Orgeta V, et al. Dementia prevention, intervention, and care. Lancet 2017; 390:2673.
  174. Hamer M, Chida Y. Physical activity and risk of neurodegenerative disease: a systematic review of prospective evidence. Psychol Med 2009; 39:3.
  175. Baumgart M, Snyder HM, Carrillo MC, et al. Summary of the evidence on modifiable risk factors for cognitive decline and dementia: A population-based perspective. Alzheimers Dement 2015; 11:718.
  176. Lautenschlager NT, Cox KL, Flicker L, et al. Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: a randomized trial. JAMA 2008; 300:1027.
  177. Shively S, Scher AI, Perl DP, Diaz-Arrastia R. Dementia resulting from traumatic brain injury: what is the pathology? Arch Neurol 2012; 69:1245.
  178. Moretti L, Cristofori I, Weaver SM, et al. Cognitive decline in older adults with a history of traumatic brain injury. Lancet Neurol 2012; 11:1103.
  179. Hong YT, Veenith T, Dewar D, et al. Amyloid imaging with carbon 11-labeled Pittsburgh compound B for traumatic brain injury. JAMA Neurol 2014; 71:23.
  180. Crane PK, Gibbons LE, Dams-O'Connor K, et al. Association of Traumatic Brain Injury With Late-Life Neurodegenerative Conditions and Neuropathologic Findings. JAMA Neurol 2016; 73:1062.
  181. Weiner MW, Crane PK, Montine TJ, et al. Traumatic brain injury may not increase the risk of Alzheimer disease. Neurology 2017; 89:1923.
  182. Weiner MW, Harvey D, Hayes J, et al. Effects of traumatic brain injury and posttraumatic stress disorder on development of Alzheimer's disease in Vietnam Veterans using the Alzheimer's Disease Neuroimaging Initiative: Preliminary Report. Alzheimers Dement (N Y) 2017; 3:177.
  183. Tannenbaum C, Paquette A, Hilmer S, et al. A systematic review of amnestic and non-amnestic mild cognitive impairment induced by anticholinergic, antihistamine, GABAergic and opioid drugs. Drugs Aging 2012; 29:639.
  184. Billioti de Gage S, Moride Y, Ducruet T, et al. Benzodiazepine use and risk of Alzheimer's disease: case-control study. BMJ 2014; 349:g5205.
  185. Koyama A, Steinman M, Ensrud K, et al. Ten-year trajectory of potentially inappropriate medications in very old women: importance of cognitive status. J Am Geriatr Soc 2013; 61:258.
  186. Koyama A, Steinman M, Ensrud K, et al. Long-term cognitive and functional effects of potentially inappropriate medications in older women. J Gerontol A Biol Sci Med Sci 2014; 69:423.
  187. Gray SL, Anderson ML, Dublin S, et al. Cumulative use of strong anticholinergics and incident dementia: a prospective cohort study. JAMA Intern Med 2015; 175:401.
  188. Risacher SL, McDonald BC, Tallman EF, et al. Association Between Anticholinergic Medication Use and Cognition, Brain Metabolism, and Brain Atrophy in Cognitively Normal Older Adults. JAMA Neurol 2016; 73:721.
  189. Haenisch B, von Holt K, Wiese B, et al. Risk of dementia in elderly patients with the use of proton pump inhibitors. Eur Arch Psychiatry Clin Neurosci 2015; 265:419.
  190. Taipale H, Tolppanen AM, Tiihonen M, et al. No Association Between Proton Pump Inhibitor Use and Risk of Alzheimer's Disease. Am J Gastroenterol 2017; 112:1802.
  191. Goldstein FC, Steenland K, Zhao L, et al. Proton Pump Inhibitors and Risk of Mild Cognitive Impairment and Dementia. J Am Geriatr Soc 2017; 65:1969.
  192. Rojo LE, Alzate-Morales J, Saavedra IN, et al. Selective interaction of lansoprazole and astemizole with tau polymers: potential new clinical use in diagnosis of Alzheimer's disease. J Alzheimers Dis 2010; 19:573.
  193. Badiola N, Alcalde V, Pujol A, et al. The proton-pump inhibitor lansoprazole enhances amyloid beta production. PLoS One 2013; 8:e58837.
  194. Fallahzadeh MK, Borhani Haghighi A, Namazi MR. Proton pump inhibitors: predisposers to Alzheimer disease? J Clin Pharm Ther 2010; 35:125.
  195. Lam JR, Schneider JL, Zhao W, Corley DA. Proton pump inhibitor and histamine 2 receptor antagonist use and vitamin B12 deficiency. JAMA 2013; 310:2435.
  196. Kuller LH. Do Proton Pump Inhibitors Increase the Risk of Dementia? JAMA Neurol 2016; 73:379.
  197. Dekosky ST, Gandy S. Environmental exposures and the risk for Alzheimer disease: can we identify the smoking guns? JAMA Neurol 2014; 71:273.
  198. Chen R. Association of environmental tobacco smoke with dementia and Alzheimer's disease among never smokers. Alzheimers Dement 2012; 8:590.
  199. Kim SH, Knight EM, Saunders EL, et al. Rapid doubling of Alzheimer's amyloid-β40 and 42 levels in brains of mice exposed to a nickel nanoparticle model of air pollution. F1000Res 2012; 1:70.
  200. Davis DA, Akopian G, Walsh JP, et al. Urban air pollutants reduce synaptic function of CA1 neurons via an NMDA/NȮ pathway in vitro. J Neurochem 2013; 127:509.
  201. Calderón-Garcidueñas L, Reed W, Maronpot RR, et al. Brain inflammation and Alzheimer's-like pathology in individuals exposed to severe air pollution. Toxicol Pathol 2004; 32:650.
  202. Calderón-Garcidueñas L, Solt AC, Henríquez-Roldán C, et al. Long-term air pollution exposure is associated with neuroinflammation, an altered innate immune response, disruption of the blood-brain barrier, ultrafine particulate deposition, and accumulation of amyloid beta-42 and alpha-synuclein in children and young adults. Toxicol Pathol 2008; 36:289.
  203. Hayden KM, Norton MC, Darcey D, et al. Occupational exposure to pesticides increases the risk of incident AD: the Cache County study. Neurology 2010; 74:1524.
  204. Richardson JR, Roy A, Shalat SL, et al. Elevated serum pesticide levels and risk for Alzheimer disease. JAMA Neurol 2014; 71:284.
Topic 16575 Version 19.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟