Skip to main content

APOE4 is associated with cognitive and pathological heterogeneity in patients with Alzheimer’s disease: a systematic review

Abstract

Possession of the ε4 allele of apolipoprotein E (APOE) is the primary genetic risk factor for the sporadic form of Alzheimer’s disease (AD). While researchers have extensively characterized the impact that APOE ε4 (APOE4) has on the susceptibility of AD, far fewer studies have investigated the phenotypic differences of patients with AD who are APOE4 carriers vs. those who are non-carriers. In order to understand these differences, we performed a qualitative systematic literature review of the reported cognitive and pathological differences between APOE4-positive (APOE4+) vs. APOE4-negative (APOE4−) AD patients. The studies performed on this topic to date suggest that APOE4 is not only an important mediator of AD susceptibility, but that it likely confers specific phenotypic heterogeneity in AD presentation, as well. Specifically, APOE4+ AD patients appear to possess more tau accumulation and brain atrophy in the medial temporal lobe, resulting in greater memory impairment, compared to APOE4− AD patients. On the other hand, APOE4− AD patients appear to possess more tau accumulation and brain atrophy in the frontal and parietal lobes, resulting in greater impairment in executive function, visuospatial abilities, and language, compared to APOE4+ AD patients. Although more work is necessary to validate and interrogate these findings, these initial observations of pathological and cognitive heterogeneity between APOE4+ vs. APOE4− AD patients suggest that there is a fundamental divergence in AD manifestation related to APOE genotype, which may have important implications in regard to the therapeutic treatment of these two patient populations.

Introduction

APOE4 carriers have an increased risk of developing AD

In 1993, Roses and colleagues first reported that an individual’s risk of developing Alzheimer’s disease (AD) is increased if they carry the ε4 allele of apolipoprotein E (APOE) [1,2,3], an important apolipoprotein that had primarily been studied for its role in transporting cholesterol and other lipids through the periphery and within the brain [4,5,6]. Since that time, the link between APOE4 and AD susceptibility has been extensively validated and characterized. A 1997 meta-analysis by Farrer et al. nicely summarizes the general associations between APOE genotype and AD susceptibility [7], which has remained relatively consistent in future studies [8,9,10]. For example, while the APOE2, APOE3, and APOE4 alleles are present in cognitively normal Caucasians at a relative frequency of about 8%, 78%, and 14%, respectively, APOE4 has an allele frequency of about 37% in Caucasian AD patients [7]. When broken down by the specific genotype frequencies, APOE3/4 individuals represent about 21% of the cognitively normal Caucasian population, vs. about 41% of Caucasian AD patients (odds ratio [OR] 3.2), whereas APOE4/4 individuals have a genotype frequency of about 2% in the cognitively normal Caucasian population, vs. about 15% in the AD-affected Caucasian population (OR 14.9) [7]. Furthermore, while possession of the APOE2 allele is protective against AD [2, 10, 11], with Caucasian individuals who possess either the APOE2/2 or the APOE2/3 genotype having an OR of 0.6, this protective effect is overtaken by the risk effect of the APOE4 allele in APOE2/4 individuals (OR 2.6) [7].

These numbers shift, however, when the APOE4-associated risk of AD is stratified by traits such as age, gender, and ancestry. For example, the effects of the APOE4 allele on AD risk are greatest in younger individuals, with the risk of AD among Caucasian APOE3/4 individuals peaking at age 65 (OR ~ 4) and the risk of AD among Caucasian APOE4/4 individuals peaking at age 60 (OR ~ 15.5) [7]. In terms of gender, numerous studies have found that the effects of APOE4 on AD susceptibility are greater in women than in men [7, 12, 13], although these gender differences appear to decrease after age 75 [7, 8, 14]. For example, in the Farrer et al. meta-analysis, the authors reported that a 65-year-old Caucasian woman with an APOE3/4 genotype has an OR of developing AD of over 4, whereas a 65-year-old Caucasian man with the same genotype has an OR of less than 2 [7]. Perhaps the most intriguing differences in APOE4’s effect size, however, are seen in individuals with different ancestral backgrounds. For example, individuals from African-ancestry populations, such as African Americans, have a higher general frequency of APOE4 (APOE4 allele frequency ~ 19%) than Caucasian populations, but these individuals are at a relatively lower risk of developing AD (APOE3/4 OR 1.1; APOE4/4 OR 5.7) [7]. However, the opposite appears to be true for East-Asian populations; for example, individuals of Japanese ancestry have a relatively low APOE4 allele frequency (~ 9%), but a relatively high APOE4-related risk (APOE3/4 OR 5.6; APOE4/4 OR 33.1) [7].

It is clear from these studies that the APOE4 allele is a strong genetic risk factor for developing AD, even though the disease penetrance varies greatly with regard to age, gender, and ancestry. However, while extensive studies have characterized the role of APOE4 in conferring AD risk, far fewer studies have investigated the effects of APOE4 on the cognitive and pathological manifestation of the disease in individuals who have already converted to AD. In order to understand how a patient’s APOE genotype affects their disease presentation, we have performed a qualitative systematic literature review of the human studies that have been published to date examining the cognitive and pathological differences between APOE4-positive (APOE4+) vs. APOE4-negative (APOE4−) AD patients. Interestingly, these studies suggest that possession of APOE4 does in fact result in phenotypic differences between APOE4+ vs. APOE4− AD patients, with APOE4+ AD patients appearing to possess relatively more tau accumulation and brain atrophy in the medial temporal lobe, resulting in greater memory impairment, than APOE4− AD patients, while APOE4− AD patients appear to possess relatively more fronto-parietal lobe tau accumulation and brain atrophy, resulting in greater impairment in executive function, visuospatial abilities, and language, than APOE4+ AD patients (Fig. 1).

Fig. 1
figure 1

Cognitive and pathological heterogeneity in APOE4+ vs. APOE4− AD patients. A representation of the heterogeneity reported in APOE4+ vs. APOE4− AD patients. APOE4+ AD patients possess relatively more tau accumulation and brain atrophy in their medial temporal lobe, resulting in greater memory impairment, compared to APOE4− AD patients. On the other hand, APOE4− AD patients possess relatively more tau accumulation and brain atrophy in their fronto-parietal lobes, resulting in greater impairment in executive function, visuospatial abilities, and language, compared to APOE4+ AD patients. The level of tau accumulation (brown) represents the levels observed in AD brains during Braak stages V–VI

Methods

The data and information utilized in this qualitative systematic review were obtained from literature published between January 1, 1993, and June 1, 2020. A literature search using both electronic and manual search components was performed, with the goal of identifying all studies published during this time period that specifically compared AD presentation in APOE4 carriers vs. APOE4 non-carriers who were diagnosed with AD using standard methods. In order to accomplish this, PubMed was exhaustively searched to help identify articles containing a combination of keywords: Apoliprotein E, APOE, APOE4, APOE4-positive, APOE4-negative, Alzheimer’s disease, AD, and patients, which was followed by a first level screening of the articles’ titles and abstracts to identify studies that directly investigated our review topic. These searches were limited to studies with human subjects that were published in the English language. A manual reference check of the bibliographies of the relevant studies was also performed in order to identify additional articles that were not identified by the electronic search.

The full article of each identified study on this topic was downloaded and stored in a single folder, at which time a second level of screening of the full text was performed to confirm that each article directly compared the cognitive and/or pathological characteristics of APOE4+ vs. APOE4− AD patients. Lastly, each study was sorted into one or more of the following diagnostic categories: rate of cognitive decline (17 studies), neuropsychological profile (12 studies), brain atrophy (13 studies), Aß pathology (7 studies), or tau pathology (6 studies). Those studies that did not fit into one of these five categories were not included in the primary review.

We did not exclude studies based on any patient demographic characteristics or any specific methodologies employed. This broad inclusion criterion was utilized in order to provide the scientific community with a comprehensive record of the studies that have investigated the cognitive and pathological differences between APOE4+ vs. APOE4− AD patients to date. However, the demographic and methodological differences between studies were carefully considered in our overall conclusions, as is discussed throughout the review.

Results

APOE4+ AD patients do not appear to differ in their overall rates of cognitive decline compared to APOE4− AD patients

Although the heterogeneity between APOE4+ vs. APOE4− AD patients is an understudied phenomenon, one question that has been repeatedly investigated over the years is whether or not APOE4+ AD patients undergo an accelerated rate of cognitive decline as compared to APOE4− AD patients. However, the results of these studies have been decidedly mixed. While numerous groups have reported that APOE4+ AD patients do in fact experience a more accelerated cognitive decline compared to APOE4− AD patients [15,16,17,18,19,20], other studies have shown either no APOE genotype-associated differences in the rate of cognitive decline in AD patients [21,22,23,24,25,26,27] or slower cognitive decline in APOE4+ vs. APOE4− AD patients [28,29,30] (Table 1).

Table 1 Studies investigating the effects of APOE4 on the rate of cognitive decline in AD patients

In general, these discrepancies between the reported rates of cognitive decline in APOE4+ vs. APOE4− AD patients highlight the difficulty of trying to determine a consensus about the contributions of one single trait, such as APOE genotype, on the overall presentation of AD. Adding to this difficulty, each of the studies that we have cited in this review utilizes different methods for their analysis, and the patient populations that they assessed often vary widely in their demographic characteristics. As discussed in the “Introduction” section, differences in age, gender, and ancestral background are known to affect AD susceptibility among APOE4 carriers; therefore, it is likely that these differences also affect AD presentation among APOE4 carriers. Furthermore, in some of the studies we have cited, the authors have utilized a relatively small number of AD patients for their analysis; because of this, it is possible that type II statistical errors may affect the conclusions that these authors reported (i.e., a study's small sample size may have resulted in no differences being observed between APOE genotype groups, even if actual differences exist).

It should also be noted that a large number of the studies investigating the effects APOE genotype on AD presentation have focused on “probable” AD patients. Probable AD is classified using standardized cognitive screening tools and robust neuropsychological tests, and must follow a strict criteria, such as those described by the NINCDS-ADRDA workgroup in 1984 [32], or an updated criteria described by the NIA-AA workgroup in 2011 [33]. However, the utilization of cognitive profiles alone (or, likewise, the utilization of pathological markers alone) cannot give a 100% confident diagnosis of AD. Given this information, it is possible that some probable AD patients included in the studies cited in this review were misdiagnosed. Notably, it has been reported that APOE4− individuals make up the majority of AD-diagnosed patients who are later found to be Aß-negative by PET or at autopsy [34, 35]. For this reason, it is possible that the sole reliance on probable AD diagnosis in some of these studies could result in type I statistical errors that may affect their findings (i.e., a study's potential inclusion of non-AD patients, especially if this was weighted towards the APOE4− individuals, may have resulted in significant differences being observed between APOE genotype groups, even if none exist). Rather than excluding such studies, however, we chose to include them, but to take their limitations into account in our overall, qualitative assessment of the data.

In regard to APOE4’s effects on the rate of cognitive decline in AD, assessing the demographic and methodological differences between the studies listed above does provide some clarity. For example, in many of these studies, the authors had access to a relatively small number of AD patients. One potential approach to assess the findings, therefore, is to focus only on the studies with a relatively large number of participants. Interestingly, when we only include the studies that meet a conservative threshold of n > 100 AD patients, there are three studies that reported accelerated cognitive decline in APOE4+ vs. APOE4− AD patients [15,16,17], four that reported no difference [21, 23, 26, 27], and only one that reported slower cognitive decline in APOE4+ vs. APOE4− AD patients [30]. Importantly, in the three largest studies from this group, a study by Kleiman et al. that analyzed 366 patients with probable AD [21], a study by Farlow et al. that analyzed 374 placebo-treated AD clinical-trial participants [26], and a study by Aerssens et al. that analyzed 504 placebo-treated AD clinical-trial participants [27], the authors did not find any APOE4-associated differences in the rate of cognitive decline in AD. These studies suggest that, when analyzed in a broad fashion, AD patients who carry the APOE4 allele do not appear to possess a more aggressive form of the disease.

However, more work is needed to determine if APOE genotype may have a significant effect on the rate of cognitive decline in specific subsets of AD patients, such as within a given age group or gender or ancestry. For example, two of the highly powered studies referenced above, by Cosentino et al. and Craft et al., reported significant APOE4-associated increases in the rates of cognitive decline when looking specifically at incident (i.e., newly diagnosed) AD cases [15, 17]. This suggests that APOE4 may accelerate cognitive decline at the earliest stages of AD diagnosis, but that these effects may dissipate with increasing disease severity. This possibility would be in line with what occurs prior to AD diagnosis, where APOE4 carriers show increased conversion from mild cognitive impairment (MCI) to AD compared to non-carriers [36,37,38]. Similarly, non-demented elderly APOE4 carriers have also been reported to undergo increased cognitive decline compared to non-demented elderly non-carriers [39, 40], especially when these APOE4 carriers are positive for Aß [41,42,43].

APOE4+ AD patients have a more amnestic cognitive profile than APOE4− AD patients

Another factor that deserves critical attention is the multi-faceted nature of the cognitive presentation of AD. For example, AD patients are not only prone to the characteristic amnestic symptoms commonly associated with the disease; they are also prone to deficits in other cognitive domains, such as executive function, visuospatial abilities, and language [44]. Indeed, some atypical AD patients present with distinct non-amnestic cognitive phenotypes, including corticobasal syndrome (CBS), where patients present with movement impairment; frontal variant Alzheimer’s disease (fvAD), where patients present with behavioral/executive function impairment; logopenic variant primary progressive aphasia (lvPPA), where patients present with language impairment; and posterior cortical atrophy (PCA), where patients present with visual impairment. Furthermore, even within the overarching concept of memory, there is significant complexity that must be considered during the neuropsychological assessment of AD patients. For example, poor performance on immediate recall, delayed recall, and delayed recognition is typically suggestive of amnesia [45, 46]. However, difficulties on immediate and delayed recall, in the absence of reduced performance on delayed recognition, are suggestive of problems with lexical access, a task that is associated with significant frontal lobe involvement [47].

In order to assess whether APOE genotype may alter the cognitive profile of AD patients, a number of studies have utilized neuropsychological assessment tools—including cognitive screening tools, such as the Mini-Mental State Examination (MMSE); brief neuropsychological tests, such as the Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-Cog); or more in-depth neuropsychological tests, such as the California Verbal Learning Test (CVLT)—in an attempt to parse out the potential divergence in cognitive deficits between APOE4+ vs. APOE4− AD patients [48,49,50,51,52,53,54,55,56,57,58]. Interestingly, the majority of these studies have reported that APOE4+ AD patients possess relatively more pronounced memory deficits than APOE4− AD patients [49,50,51,52,53, 58, 59], although a few studies did not find an association between APOE genotype and memory function [54, 55, 57]. In addition, a number of these studies have also reported that APOE4− AD patients possess relatively more pronounced deficits in non-memory cognitive domains, such as executive function, visuospatial abilities, and language, than APOE4+ AD patients [48, 51,52,53,54,55,56,57,58], with a greater effect observed in younger APOE4− vs. APOE4+ AD patients [54, 58] (Table 2).

Table 2 Studies investigating the effects of APOE4 on cognitive profiles in AD patients

Although these studies utilized different methodological approaches, the results were generally consistent. For example, Scheltens et al. combined four large probable AD cohorts using a neuropsychologically derived cluster analysis and found two distinct groups—a memory-impaired group and a non-memory-impaired group, with the non-memory-impaired group comprised primarily of younger, APOE4− AD patients, as compared to the memory-impaired group [48]. Kim et al. recruited 846 South Korean patients diagnosed with probable AD and categorized them into three groups with respect to their age (< 65, 65–74, and ≥ 75 years old). The authors discovered that younger (< 65 years old) APOE4− AD patients performed worse on executive function tasks compared to younger APOE4+ AD patients, while intermediate (65–74 year-old) APOE3/4 AD patients performed worse on visuospatial tasks compared to intermediate APOE4/4 AD patients, and older (≥ 75 years old) APOE4/4 AD patients performed worse on verbal memory compared to older APOE4− AD patients [58]. Finally, Wolk et al. compared cognitive differences in 67 APOE4+ vs. 24 APOE4− patients diagnosed with mild AD and possessing CSF biomarker profiles consistent with AD [51]. APOE4+ AD patients performed worse on memory retention, while APOE4− AD patients were more impaired on tests of working memory, executive function, and lexical access, but not on confrontational naming.

These results suggest that AD patients likely diverge in their cognitive presentations based on their APOE genotype, with APOE4+ AD patients presenting with relatively more pronounced amnestic deficits than APOE4− AD patients, and APOE4− AD patients presenting with relatively more non-memory deficits than APOE4+ AD patients. This conclusion is also consistent with the reported observation that AD patients presenting with atypical phenotypes, such as CBS, fvAD, lvPPA, and PCA, are less likely to be APOE4 carriers [60, 61].

Interestingly, possession of the APOE4 allele has also been associated with decreased memory performance in non-demented elderly individuals [62,63,64,65], as well as with increased incidence of amnestic MCI vs. non-amnestic MCI [66, 67]. This suggests that possession of the APOE4 allele may confer increased memory deficits throughout the aging to AD continuum, although it should be noted that APOE4 carriers have also been found to be at an increased risk of developing several non-AD dementias, including vascular dementia (VaD) [68,69,70,71], Lewy body dementia (LBD) [72,73,74], and frontotemporal dementia (FTD) [75, 76], which often do not present with a predominantly amnestic phenotype.

APOE4+ AD patients have more atrophy in the medial temporal lobe than APOE4− AD patients

The cognitive deficits observed in AD patients are a direct result of the pathological abnormalities that occur in a patient’s brain during the course of the disease. AD pathology is characterized by the hallmark accumulation of Aß-containing amyloid plaques and hyperphosphorylated tau-containing neurofibrillary tangles (NFTs). Amyloid plaques are extracellular and accumulate in the brain in a rather diffuse manner, typically starting in the neocortex (Thal phase 1), followed by the entorhinal cortex, hippocampus, and insular cortex (Thal phase 2), and eventually accumulating in subcortical regions such as the basal forebrain and brainstem (Thal phases 3–5) [77]. On the other hand, NFTs are intracellular and accumulate in the brain in a more localized and regionally conserved manner, typically occurring first in the transentorhinal and entorhinal cortex regions (Braak stages I–II), followed by the hippocampus and neighboring neocortical regions (Braak stages III–IV), and eventually accumulating throughout the remainder of the neocortex (Braak stages V–IV) [78]. The third major pathological feature of AD is “brain atrophy,” as measured by volumetric reduction or cortical thinning observed during magnetic resonance imaging (MRI). In general, the atrophy observed in the brains of AD patients has been found to follow along the same regional path as NFTs, with the first signs of volumetric loss observed in the medial temporal lobe during the MCI phase, followed by the neocortical portions of the temporal lobe, then the parietal lobe, and finally the frontal lobe during the course of MCI and AD progression [79]. In addition to these three distinctive features of AD pathology, other important pathological events that also occur during the course of the disease include neuroinflammation, deficits in cellular metabolism, cholinergic dysfunction, aberrant network activity, and cerebrovascular pathology [80].

With respect to APOE genotype effects on AD pathology, the most compelling results published to date describe the differing regional patterns of brain atrophy observed in APOE4+ vs. APOE4− AD patients. While a couple of studies have not observed any differences in brain volume or cortical thickness between APOE4+ vs. APOE4− AD patients [81, 82], the vast majority of the studies that have investigated this topic to date have found that APOE4+ AD patients possess greater volumetric loss or cortical thinning in the medial temporal lobe than APOE4− AD patients [51, 57, 58, 83,84,85,86,87,88,89], with many reporting that APOE4+ vs. APOE4− AD patients display volumetric decreases in specific medial temporal lobe structures, such as the hippocampus [57, 83, 86, 87, 89], the amygdala [57, 83, 86, 87], and the entorhinal cortex [84, 89]. Furthermore, many of these studies also reported that APOE4− AD patients possess greater volumetric loss or cortical thinning in their frontal and parietal lobes than APOE4+ AD patients [51, 58, 85, 89, 90] (Table 3).

Table 3 Studies investigating the effects of APOE4 on brain atrophy in AD patients

Importantly, a number of these studies noted a direct correlation between the regional brain atrophy patterns that they observed between APOE4+ vs. APOE4− AD patients and the differences in cognitive profile that they observed in these same patients [51, 57, 58, 84, 87, 90]. For example, in the Scheltens et al. study, the authors also analyzed MRI data from their four large probable AD cohorts and observed that in their non-memory-impaired group, which was enriched for APOE4− AD patients, there was less hippocampal volume loss and more posterior cortex volume loss than in the memory-impaired group [48]. And in the Kim et al. study, the authors measured cortical thinning using MRI in their 846 South Korean probable AD patients and found that in the younger (< 65 years old) APOE4− AD patients, who performed worse on executive function tasks, there was increased bilateral cortical thinning in their lateral frontal, medial frontal, and perisylvian areas compared to the younger APOE4− AD patients, whereas in the older (≥ 75 years old) APOE4+ AD patients, who performed worse on verbal memory tasks, there was increased bilateral cortical thinning in their medial temporal areas compared to the older APOE4− AD patients [58]. Finally, in the Wolk et al. study, the authors used MRI to measure brain volume and cortical thickness in their mild AD patients and found that APOE4+ AD patients, who performed worse on memory retention, displayed greater hippocampal volume loss than APOE4− AD patients, whereas APOE4− AD patients, who performed worse on working memory, executive function, and lexical access, displayed decreased cortical thickness in their superior parietal lobule, precuneus, and angular gyrus than APOE4+ AD patients.

APOE4+ AD patients do not appear to have higher Aß levels than APOE4− AD patients

As noted above, the regional brain atrophy that is observed in AD patients is thought to be a direct result of the tau accumulation that progressively occurs in neurons within these brain regions. And it is believed that this tau accumulation and the regional progression of NFTs likely occur downstream of the Aß accumulation/amyloid plaque deposition that begins early in AD pathogenesis. Given this information, it is important to determine whether the presentation of these two hallmark pathologies also displays heterogeneity in APOE4+ vs. APOE4− AD patients, and how this presentation may relate to the differences in brain atrophy and cognitive deficits that are observed in these patients. In terms of Aß, it has been well documented that individuals who carry the APOE4 allele accumulate Aß in their brains at an earlier age than non-carriers, and that this occurs long before the onset of AD. For example, a 2015 meta-analysis by Jansen et al. revealed that by the time APOE4/4 carriers turn 40 years old, about 15% of them will already be positive for cerebral Aß (as detected by PET or CSF), whereas this threshold is not reached until 55 years of age for APOE3/4 carriers and 65 years of age for APOE3/3 carriers [91]. However, Aß levels have been shown to plateau before the clinical diagnosis of AD [92], so any differences in Aß levels associated with APOE genotype are not expected to be as dramatic once a patient converts to AD as it is during the linear phase of Aß accumulation. For this reason, it is perhaps not surprising that the handful of studies that have compared the levels of Aß in APOE4+ vs. APOE4− AD patients have shown conflicting results, with some studies reporting increased Aß levels in the brains of APOE4+ AD patients compared to APOE4− AD patients [81, 93, 94], some reporting no changes in Aß levels between these two groups [95, 96], and some reporting decreased Aß levels in the brains of APOE4+ AD patients compared to APOE4− AD patients [97, 98] (Table 4).

Table 4 Studies investigating the effects of APOE4 on amyloid plaques in AD patients

Looking closely at these studies, it is difficult to make a conclusive statement about how exactly APOE genotype affects Aß levels or amyloid plaque distribution in the brains of AD patients. For example, the studies by Drzezga et al. (32 patients with moderate AD) [81], Rowe et al. (53 patients with mild AD) [95], and Lehmann et al. (52 patients with probable AD) [98] each utilized Pittsburgh Compound B (PIB) PET analysis on age- and cognition-matched AD patients who were confirmed to be Aß-positive, but with each study arriving at a different conclusion about the relative levels of Aß in APOE4+ vs. APOE4− AD patients. Perhaps future work on this topic will reveal more regionally specific differences in how Aß is distributed in the brains of APOE4+ vs. APOE4− AD patients. This is hinted at by the Lehmann et al. study, where the observed decrease in Aß in APOE4+ AD patients was primarily localized to the right lateral frontotemporal regions of the brain [98].

Of course, it is also important to note that amyloid plaques are only one manifestation of Aß pathology that can occur in the brain. Aß can also build up in the walls of arteries (cerebral amyloid angiopathy; CAA) or inside of neurons (intraneuronal Aß). Interestingly, several studies have reported that APOE4+ AD patients have a more frequent CAA comorbidity than APOE4− AD patients [94, 99,100,101]. As for intraneuronal Aß, while one study did report that post-mortem brains from APOE4+ AD patients possess higher levels of intraneuronal Aß than those from APOE4− AD patients [102], much more investigation is required before any conclusive statements can be made on this topic.

APOE4+ AD patients appear to develop more tau pathology in their medial temporal lobe than APOE4− AD patients

As with Aß, there have been numerous reports that APOE4 carriers possess higher levels of tau pathology than non-carriers prior to AD onset, although this effect on preclinical tau pathology does not seem to be nearly as robust as it is with APOE4’s effects on preclinical Aß levels. For example, in a study where Braak and colleagues analyzed autopsied brain tissues specifically from individuals who reached Braak stage I (transentorhinal cortex only) at a relatively young age (less than 47 years old), the authors reported a significant increase in the percentage of APOE4 carriers in this group (36%) vs. the percentage of APOE4 carriers in the control group (16%) [103]. A later, more generalized autopsy study from Braak and colleagues also observed that women who were APOE4 carriers met the criteria for Braak stages II (entorhinal cortex) and III (hippocampus) 3 years earlier than non-carriers [104]. Several more recent studies have also reported a female-dominant effect of APOE genotype on tau levels prior to AD diagnosis [12, 105, 106]. In each of these studies, the authors reported that APOE4 possession increases CSF tau levels specifically in female APOE4 carriers, with two of the studies reporting that this APOE4-associated effect on CSF tau levels was only present when the women were positive for Aß pathology [105, 106].

In regard to APOE4+ vs. APOE4− AD patients, tau pathology also appears to differ according to APOE genotype, although the primary differences here appear to revolve around the regional pattern of NFT distribution, as opposed to the overall levels (Table 5). For example, Murray et al. have reported that, when AD autopsy cases were divided into three distinct groups based on the regional pattern of the NFT pathology observed (“hippocampal-sparing,” “typical,” and “limbic predominant”), there was a trend towards fewer APOE4 carriers in the “hippocampal-sparing” AD group, and there were significantly more late-onset (greater than 65 years old at diagnosis) APOE4 carriers vs. non-carriers in the “limbic predominant” AD group [61]. Although a more recent study failed to replicate this finding in a set of AD autopsy cases enriched for atypical presentation (in which APOE4 carriers were underrepresented), there did appear to be a trend (p = 0.0992) towards more APOE4 carriers among “limbic predominant” AD cases and fewer APOE4 carriers among “hippocampal-sparing” AD cases [107]. Interestingly, a recent follow-up paper by Murray and colleagues also reported that APOE4+ “typical” AD patients, as compared to APOE4− “typical” AD patients, possess more NFT pathology in their nucleus basalis of Meynert (nbM), the major source of cholinergic innervation in the brain [110].

Table 5 Studies investigating the effects of APOE4 on neurofibrillary tangles in AD patients

To interrogate this correlation between APOE genotype and tau pathology in living individuals, researchers have begun utilizing recently developed tau PET imaging ligands to compare tau levels in APOE4+ vs. APOE4− AD patients. For example, a small study by Ossenkoppele et al. utilizing the 18F-AV-1451 tau PET ligand in 20 individuals diagnosed with MCI or AD found increased uptake of the PET ligand in bilateral medial temporal and right temporoparietal cortex of APOE4+ patients, as compared to APOE4− patients [108]. And in a study by Whitwell et al. employing 18F-AV-1451 to investigate tau deposition in 62 amyloid-positive AD patients with a mix of typical and atypical AD presentations, the authors separated their subjects into three groups (ECLo/CLo, ECLo/CHi, and ECHi/CHi) based on the amount of tau deposition they observed in the entorhinal cortex (EC), as compared to the whole cortex (C) [109]. The authors found that the APOE4 frequency was significantly lower in the ECLo/CHi group, suggesting that APOE4− AD patients have less relative tau accumulation in the entorhinal cortex region than APOE4+ AD patients in the context of high cortical tau load. Finally, in a study by Mattsson et al. that also utilized the 18F-AV-1451 tau PET ligand, again on a mixed group of MCI and AD patients (65 patients total), the authors reported an increased tau load in the entorhinal cortex (relative to the whole cortex) of APOE4+ patients compared to APOE4− patients, whereas the tau load in the parietal and occipital lobes was higher in APOE4− patients compared to APOE4+ patients [90].

To be clear, these studies on tau pathology in APOE4+ vs. APOE4− AD patients are still somewhat preliminary, with additional work required to confidently answer this question. Specifically, additional tau PET imaging ligand studies are required in pure AD populations, and with larger sample sizes. Also, as with the other studies on this topic, analysis of specific subgroupings needs to be performed with respect to age, gender, and ancestral background. However, based on these early results, it does appear that APOE4+ AD patients may possess relatively more NFTs in the medial temporal lobe, most notably in the entorhinal cortex, while APOE4− AD patients may possess more NFTs in other cortical regions, such as the frontal and parietal lobes.

Conclusions

Primary findings and key limitations

The majority of the previous research investigating the relationship between APOE4 and AD has focused on elucidating the patterns and mechanisms associated with the increased risk of developing AD among APOE4 carriers. And for good reason, after all, APOE4 is the primary genetic risk factor for sporadic AD. However, the possibility that APOE4 may also affect the cognitive and pathological presentation of AD deserves significant attention, as this possibility may elucidate differing pathogenic mechanisms between APOE4+ vs. APOE4− AD patients, both before and after disease onset, and may have important implications for how we should therapeutically treat APOE4+ vs. APOE4− AD patients.

Overall, the studies that have been performed on this topic to date suggest that APOE4+ vs. APOE4− AD patients do appear to possess both cognitive and pathological heterogeneity in their presentation of the disease, as depicted in Fig. 1. Specifically, the neuropsychological studies outlined above show that APOE4+ AD patients appear to possess relatively more pronounced memory deficits than APOE4− AD patients, while APOE4− AD patients appear to possess relatively more pronounced non-memory deficits (particularly deficits in executive function, visuospatial abilities, and language) than APOE4+ AD patients. The literature also points to divergent pathological underpinnings that likely explain the differences in cognitive profiles related to an AD patient’s APOE genotype. Most notably, APOE4+ AD patients appear to possess relatively more brain atrophy in their medial temporal lobe than APOE4− AD patients, while APOE4− AD patients appear to possess relatively more brain atrophy in their frontal and parietal lobes than APOE4+ AD patients. The literature also suggests that the upstream trigger of these regional brain atrophy differences is likely to be the observed differences in the regional distribution of NFTs in APOE4+ vs. APOE4− AD patients, with APOE4+ AD patients possessing a greater relative accumulation of NFTs in their medial temporal lobe (particularly in the entorhinal cortex) than APOE4− AD patients, and APOE4− AD patients possessing relatively more NFTs in their frontal and parietal lobes than APOE4+ AD patients. However, due to the limited number of studies performed using recently developed tau PET imaging ligands, this last conclusion is particularly unresolved.

It should also be noted that an AD patient’s APOE genotype may affect the presentation of several additional brain pathologies not covered in the “Results” section of this review. For example, in the previously discussed Lehmann et al. study, where the authors observed regional decreases in Aß deposition in APOE4+ vs. APOE4− AD patients, the authors also reported regional differences in glucose metabolism (as measured by FDG-PET), with APOE4+ AD patients displaying more hypometabolism in bilateral medial temporal and right lateral temporal regions than APOE4− AD patients, while APOE4− AD patients displayed more hypometabolism in other cortical areas, including supplementary motor cortex and superior frontal gyrus [98]. Furthermore, autopsied brains from APOE4+ vs. APOE4−AD patients have also been reported to possess increased levels of two pathological comorbidities commonly associated with AD: TDP-43 [111,112,113] and Lewy bodies [114, 115].

As noted throughout this review, there are a number of limitations in the studies we cited, which decreases the overall confidence with which we can assert that there is a definitive difference in disease presentation between APOE4+ vs. APOE4− AD patients. For example, some of the studies performed on this topic utilized relatively small sample sizes, which may result in type II ("false-negative") statistical errors. In addition, many of the studies we cited utilized “probable AD” for their AD diagnosis, which may result in type I ("false-positive") statistical errors. Lastly, inherent differences among APOE4 carriers, like age, gender, and ancestral background, are likely to modulate the effects of APOE genotype on AD presentation, a possibility that requires much more investigation.

In order to address these issues, we propose that additional studies comparing the cognitive and pathological presentation of AD in APOE4+ vs. APOE4− AD patients should include the following criteria: (1) Comprehensive neuropsychological testing, or numerous cognitive tests measuring multiple cognitive domains, should be utilized to diagnose AD patients. Cognitive screeners, such as the MMSE, are helpful in identifying individuals who require more comprehensive assessment, but robust neuropsychological tests are far more capable of making accurate diagnoses and clear determinations of the severity of a patient’s cognitive impairment. (2) Pathological diagnosis should be confirmed using established biomarkers such PET tracers or CSF measurements, or histology on post-mortem tissues if the subjects are deceased. (3) Large, diverse cohorts of AD patients should be utilized. These cohorts should include hundreds of participants with different ages, genders, and ancestral backgrounds. Power analysis should be performed not only for the cohort as a whole, but also for the individual demographic subgroupings, in order to allow for statistically significant results from each independent subgroup. (4) All three of the first three criteria should be utilized in tandem in order to carefully match the APOE4+ AD patients to the APOE4− AD patients with which they are being compared. Matched APOE genotype groups should possess similar neuropsychological profiles, similar pathology levels, and similar demographics, although variations may be necessary depending on the specific question being tested. We anticipate that these robust future studies will definitively determine whether APOE4+ vs. APOE4− AD patients possess the cognitive and pathological heterogeneity that the initial studies on this topic suggest.

Why disease heterogeneity is important

In recent years, disease heterogeneity has gained increased attention in AD research, with numerous publications reporting on divergent aspects of AD such as atypical neuropsychological profiles and mixed pathologies in AD patients [116,117,118,119,120,121]. One reason why the topic of disease heterogeneity is so important in AD research is that it suggests a previously unappreciated complexity that may make therapeutic treatment of AD more difficult (and could also help to explain past clinical trial failures). If AD is not the single, uniform disease that researchers once believed it to be, then a single therapeutic strategy may not be able to help all AD patients equally. In respect to APOE genotype, disease heterogeneity may even point to divergent pathological mechanisms that will be particularly important to understand when attempting to treat APOE4 carriers vs. non-carriers.

On that topic, there have been numerous examples of therapies showing efficacy in APOE4 carriers, but not in non-carriers, or vice-versa. For example, currently approved acetylcholinesterase inhibitors have often been reported as having differential effects on APOE4+ vs. APOE4− AD patients, although these results have been mixed [122,123,124,125]. Investigations of intranasal insulin as an AD treatment have also shown mixed results, with an acute insulin treatment showing memory improvement in APOE4− MCI and AD patients, but not APOE4+ MCI and AD patients [126], while a chronic insulin treatment has shown memory improvement in APOE4+ MCI and AD patients, but not APOE4− MCI and AD patients [127]. Differential APOE genotype effects have also been reported for the treatment of mild-to-moderate AD patients using the diabetes drug rosiglitazone, with APOE4− AD patients, but not APOE4+ AD patients, showing cognitive improvement [128]. Furthermore, the retinoid x receptor (RXR) agonist bexarotene has been shown to reduce Aß levels in APOE4− AD patients, but not in APOE4+ AD patients [129]. And in a phase 3 clinical trial of the anti-Aß antibody bapaineuzumab for mild-to-moderate AD, reductions of both Aß and tau levels were observed in APOE4+ AD patients, but not in APOE4− AD patients [130].

Given these potential differences in treatment efficacy for APOE4+ vs. APOE4− AD patients, it is important not only to elucidate any overall cognitive and pathological heterogeneity between these two groups, but also to understand the underlying mechanisms that may drive this heterogeneity. Indeed, the discovery of divergent pathological mechanisms between APOE4+ vs. APOE4− AD patients would not only point to important treatment differences for these two patient groups, but it could also help clarify the mechanism of AD pathogenesis in general. The majority of AD research has focused on Aß and tau accumulation, the pathological hallmarks of the disease. However, understanding the ways in which differential isoform expression of APOE, which primarily plays a role in cholesterol and lipid trafficking, mediates AD presentation would add important context to how AD develops and progresses.

Potential mechanisms

For the most part, the studies referenced in this review do not attempt to pinpoint the underlying mechanism(s) responsible for the heterogeneity that they report between APOE4+ vs. APOE4− AD patients. However, it is worth discussing the potential mechanisms that may be responsible for this heterogeneity. First off, although the pathological differences that occur in APOE4+ vs. APOE4− AD patients appear to center around tau pathology and the resulting brain atrophy, it is still quite possible that the earlier onset of Aß pathology that occurs in APOE4 carriers vs. non-carriers may play a direct role in the pathological differences that appear to occur in APOE4+ vs. APOE4− AD patients. If tau aggregation is in fact a direct result of Aß pathology, as is proposed by the amyloid cascade hypothesis, it is probable that the earlier increases in Aß accumulation that are observed in the brains of APOE4 carriers would lead to an early and prolonged accumulation of tau pathology within the entorhinal cortex and hippocampus, the brain regions where NFTs are first observed. This possibility is hinted at by the recent studies showing that women who are positive for both APOE4 and Aß have higher levels of CSF tau compared to other groups [105, 106], even in the absence of any cognitive decline [106]. That said, it would be anticipated that this increased early accumulation of tau pathology in the medial temporal lobe of APOE4 carriers would also translate to increased tau pathology in the fronto-parietal lobes as the disease progresses. However, these initial studies have observed the exact opposite, with decreased tau pathology and neurodegeneration occurring in the fronto-parietal lobes of APOE4+ vs. APOE4− AD patients.

Alternatively, it is possible that the mechanisms responsible for this observed cognitive and pathological heterogeneity in APOE4+ vs. APOE4− AD patients are independent of APOE4’s effects on Aß. To that end, it is important to note that APOE4 expression has been found to have a deleterious effect on numerous Aß-independent pathways within the brain, including cholesterol/lipid metabolism [131,132,133], endosomal-lysosomal processing [134,135,136,137,138,139,140,141,142], energy metabolism [143,144,145,146,147,148], neuroinflammation [149,150,151], and cerebrovascular integrity [152,153,154,155]. Furthermore, APOE appears to be highly expressed in the medial temporal lobe compared to other brain regions, as shown in this spatial modeling of APOE mRNA levels derived from Allen Brain Atlas mRNA expression data (http://www.meduniwien.ac.at/neuroimaging/lib/dlpage.php?value=348&name=apolipoprotein%20E) [156]. Therefore, cells in the medial temporal lobe of APOE4 carriers may be particularly susceptible to deficits in the biological pathways listed above.

Clearly, there are still many questions left to be answered with regard to this apparent heterogeneity in APOE4+ vs. APOE4− AD patients, which we anticipate future studies will help to elucidate. We believe that validating and interrogating this APOE4-associated heterogeneity will yield important information for how best to treat AD patients based on their specific APOE genotype. In addition, uncovering the biological mechanism(s) responsible for this apparent heterogeneity may pave the way for the discovery of novel therapeutic strategies for treating or preventing AD in general.

Availability of data and materials

Not applicable

References

  1. Strittmatter WJ, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A. 1993;90:1977–81.

    Article  CAS  Google Scholar 

  2. Corder EH, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.

    Article  CAS  Google Scholar 

  3. Saunders AM, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology. 1993;43:1467–72.

    Article  CAS  Google Scholar 

  4. Mahley RW, Rall SC Jr. Apolipoprotein E: far more than a lipid transport protein. Annu Rev Genomics Hum Genet. 2000;1:507–37.

    Article  CAS  Google Scholar 

  5. Han X. The role of apolipoprotein E in lipid metabolism in the central nervous system. Cell Mol Life Sci. 2004;61:1896–906.

    Article  CAS  Google Scholar 

  6. Holtzman DM, Herz J, Bu G. Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2:a006312.

    Google Scholar 

  7. Farrer LA, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–56.

    Article  CAS  Google Scholar 

  8. Neu SC, et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis. JAMA Neurol. 2017;74:1178–89.

    Article  Google Scholar 

  9. Belloy ME, Napolioni V, Greicius MD. A quarter century of APOE and Alzheimer’s disease: progress to date and the path forward. Neuron. 2019;101:820–38.

    Article  CAS  Google Scholar 

  10. Reiman EM, et al. Exceptionally low likelihood of Alzheimer’s dementia in APOE2 homozygotes from a 5,000-person neuropathological study. Nat Commun. 2020;11:667.

    Article  CAS  Google Scholar 

  11. Corder EH, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet. 1994;7:180–4.

    Article  CAS  Google Scholar 

  12. Altmann A, Tian L, Henderson VW, Greicius MD, I. Alzheimer’s Disease Neuroimaging Initiative. Sex modifies the APOE-related risk of developing Alzheimer disease. Ann Neurol. 2014;75:563–73.

    Article  CAS  Google Scholar 

  13. Payami H, et al. Alzheimer’s disease, apolipoprotein E4, and gender. JAMA. 1994;271:1316–7.

    Article  CAS  Google Scholar 

  14. Davidson Y, et al. Apolipoprotein E epsilon4 allele frequency and age at onset of Alzheimer’s disease. Dement Geriatr Cogn Disord. 2007;23:60–6.

    Article  CAS  Google Scholar 

  15. Cosentino S, et al. APOE epsilon 4 allele predicts faster cognitive decline in mild Alzheimer disease. Neurology. 2008;70:1842–9.

    Article  CAS  Google Scholar 

  16. Martins CA, Oulhaj A, de Jager CA, Williams JH. APOE alleles predict the rate of cognitive decline in Alzheimer disease: a nonlinear model. Neurology. 2005;65:1888–93.

    Article  CAS  Google Scholar 

  17. Craft S, et al. Accelerated decline in apolipoprotein E-epsilon4 homozygotes with Alzheimer’s disease. Neurology. 1998;51:149–53.

    Article  CAS  Google Scholar 

  18. Hirono N, Hashimoto M, Yasuda M, Kazui H, Mori E. Accelerated memory decline in Alzheimer’s disease with apolipoprotein epsilon4 allele. J Neuropsychiatry Clin Neurosci. 2003;15:354–8.

    Article  CAS  Google Scholar 

  19. Kanai M, et al. Apolipoprotein E4 accelerates dementia and increases cerebrospinal fluid tau levels in Alzheimer’s disease. Neurosci Lett. 1999;267:65–8.

    Article  CAS  Google Scholar 

  20. Chang YL, et al. APOE interacts with age to modify rate of decline in cognitive and brain changes in Alzheimer’s disease. Alzheimers Dement. 2014;10:336–48.

    Article  Google Scholar 

  21. Kleiman T, et al. Apolipoprotein E epsilon4 allele is unrelated to cognitive or functional decline in Alzheimer’s disease: retrospective and prospective analysis. Dement Geriatr Cogn Disord. 2006;22:73–82.

    Article  Google Scholar 

  22. Growdon JH, Locascio JJ, Corkin S, Gomez-Isla T, Hyman BT. Apolipoprotein E genotype does not influence rates of cognitive decline in Alzheimer’s disease. Neurology. 1996;47:444–8.

    Article  CAS  Google Scholar 

  23. Holmes C, Levy R, McLoughlin DM, Powell JF, Lovestone S. Apolipoprotein E: non-cognitive symptoms and cognitive decline in late onset Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1996;61:580–3.

    Article  CAS  Google Scholar 

  24. Kurz A, et al. Apolipoprotein E epsilon 4 allele, cognitive decline, and deterioration of everyday performance in Alzheimer’s disease. Neurology. 1996;47:440–3.

    Article  CAS  Google Scholar 

  25. Basun H, Grut M, Winblad B, Lannfelt L. Apolipoprotein epsilon 4 allele and disease progression in patients with late-onset Alzheimer’s disease. Neurosci Lett. 1995;183:32–4.

    Article  CAS  Google Scholar 

  26. Farlow MR, et al. Metrifonate treatment of AD: influence of APOE genotype. Neurology. 1999;53:2010–6.

    Article  CAS  Google Scholar 

  27. Aerssens J, et al. APOE genotype: no influence on galantamine treatment efficacy nor on rate of decline in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2001;12:69–77.

    Article  CAS  Google Scholar 

  28. Stern Y, et al. The absence of an apolipoprotein epsilon4 allele is associated with a more aggressive form of Alzheimer’s disease. Ann Neurol. 1997;41:615–20.

    Article  CAS  Google Scholar 

  29. Frisoni GB, et al. Gene dose of the epsilon 4 allele of apolipoprotein E and disease progression in sporadic late-onset Alzheimer’s disease. Ann Neurol. 1995;37:596–604.

    Article  CAS  Google Scholar 

  30. Hoyt BD, Massman PJ, Schatschneider C, Cooke N, Doody RS. Individual growth curve analysis of APOE epsilon 4-associated cognitive decline in Alzheimer disease. Arch Neurol. 2005;62:454–9.

    Article  Google Scholar 

  31. Murphy GM Jr, Taylor J, Kraemer HC, Yesavage J, Tinklenberg JR. No association between apolipoprotein E epsilon 4 allele and rate of decline in Alzheimer’s disease. Am J Psychiatry. 1997;154:603–8.

    Article  Google Scholar 

  32. McKhann G, et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34:939–44.

    Article  CAS  Google Scholar 

  33. McKhann GM, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.

    Article  Google Scholar 

  34. Monsell SE, et al. Characterizing apolipoprotein E epsilon4 carriers and noncarriers with the clinical diagnosis of mild to moderate Alzheimer dementia and minimal beta-amyloid peptide plaques. JAMA Neurol. 2015;72:1124–31.

    Article  Google Scholar 

  35. Landau SM, Horng A, Fero A, Jagust WJ, I. Alzheimer’s Disease Neuroimaging. Amyloid negativity in patients with clinically diagnosed Alzheimer disease and MCI. Neurology. 2016;86:1377–85.

    Article  CAS  Google Scholar 

  36. Elias-Sonnenschein LS, Viechtbauer W, Ramakers IH, Verhey FR, Visser PJ. Predictive value of APOE-epsilon4 allele for progression from MCI to AD-type dementia: a meta-analysis. J Neurol Neurosurg Psychiatry. 2011;82:1149–56.

    Article  Google Scholar 

  37. Fleisher AS, et al. Clinical predictors of progression to Alzheimer disease in amnestic mild cognitive impairment. Neurology. 2007;68:1588–95.

    Article  CAS  Google Scholar 

  38. Petersen RC, et al. Apolipoprotein E status as a predictor of the development of Alzheimer’s disease in memory-impaired individuals. JAMA. 1995;273:1274–8.

    Article  CAS  Google Scholar 

  39. Caselli RJ, et al. Longitudinal modeling of age-related memory decline and the APOE epsilon4 effect. N Engl J Med. 2009;361:255–63.

    Article  CAS  Google Scholar 

  40. Caselli RJ, et al. Cognitive domain decline in healthy apolipoprotein E epsilon4 homozygotes before the diagnosis of mild cognitive impairment. Arch Neurol. 2007;64:1306–11.

    Article  Google Scholar 

  41. Kantarci K, et al. APOE modifies the association between Abeta load and cognition in cognitively normal older adults. Neurology. 2012;78:232–40.

    Article  CAS  Google Scholar 

  42. Mormino EC, et al. Amyloid and APOE epsilon4 interact to influence short-term decline in preclinical Alzheimer disease. Neurology. 2014;82:1760–7.

    Article  CAS  Google Scholar 

  43. Lim YY, et al. Association of beta-amyloid and apolipoprotein E epsilon4 with memory decline in preclinical Alzheimer disease. JAMA Neurol. 2018;75:488–94.

    Article  Google Scholar 

  44. Weintraub S, Wicklund AH, Salmon DP. The neuropsychological profile of Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2:a006171.

    Article  Google Scholar 

  45. Davis KL, Price CC, Kaplan E, Libon DJ. Error analysis of the nine-word California Verbal Learning Test (CVLT-9) among older adults with and without dementia. Clin Neuropsychol. 2002;16:81–9.

    Article  Google Scholar 

  46. Libon DJ, et al. A nine—word dementia version of the California Verbal Learning Test. Clin Neuropsychol. 1996;10:237–44.

    Article  Google Scholar 

  47. McCabe DP, Roediger HL, McDaniel MA, Balota DA, Hambrick DZ. The relationship between working memory capacity and executive functioning: evidence for a common executive attention construct. Neuropsychology. 2010;24:222–43.

    Article  Google Scholar 

  48. Scheltens NME, et al. Cognitive subtypes of probable Alzheimer’s disease robustly identified in four cohorts. Alzheimers Dement. 2017;13:1226–36.

    Article  Google Scholar 

  49. Marra C, et al. Apolipoprotein E epsilon4 allele differently affects the patterns of neuropsychological presentation in early- and late-onset Alzheimer’s disease patients. Dement Geriatr Cogn Disord. 2004;18:125–31.

    Article  CAS  Google Scholar 

  50. Snowden JS, et al. Cognitive phenotypes in Alzheimer’s disease and genetic risk. Cortex. 2007;43:835–45.

    Article  Google Scholar 

  51. Wolk DA, Dickerson BC, I. Alzheimer's Disease Neuroimaging. Apolipoprotein E (APOE) genotype has dissociable effects on memory and attentional-executive network function in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2010;107:10256–61.

    Article  CAS  Google Scholar 

  52. van der Vlies AE, et al. Cognitive impairment in Alzheimer’s disease is modified by APOE genotype. Dement Geriatr Cogn Disord. 2007;24:98–103.

    Article  CAS  Google Scholar 

  53. Lehtovirta M, et al. Clinical and neuropsychological characteristics in familial and sporadic Alzheimer’s disease: relation to apolipoprotein E polymorphism. Neurology. 1996;46:413–9.

    Article  CAS  Google Scholar 

  54. Smits LL, et al. Early onset APOE E4-negative Alzheimer’s disease patients show faster cognitive decline on non-memory domains. Eur Neuropsychopharmacol. 2015;25:1010–7.

    Article  CAS  Google Scholar 

  55. Davidson JE, et al. An exploration of cognitive subgroups in Alzheimer’s disease. J Int Neuropsychol Soc. 2010;16:233–43.

    Article  Google Scholar 

  56. Schott JM, et al. Apolipoprotein e genotype modifies the phenotype of Alzheimer disease. Arch Neurol. 2006;63:155–6.

    Article  Google Scholar 

  57. Hashimoto M, et al. Apolipoprotein E epsilon 4 and the pattern of regional brain atrophy in Alzheimer’s disease. Neurology. 2001;57:1461–6.

    Article  CAS  Google Scholar 

  58. Kim J, et al. The impact of APOE varepsilon4 in Alzheimer’s disease differs according to age. J Alzheimers Dis. 2018;61:1377–85.

    Article  CAS  Google Scholar 

  59. Weintraub S, et al. APOE is a correlate of phenotypic heterogeneity in Alzheimer disease in a national cohort. Neurology. 2020;94:e607–12.

    Article  Google Scholar 

  60. van der Flier WM, Pijnenburg YA, Fox NC, Scheltens P. Early-onset versus late-onset Alzheimer’s disease: the case of the missing APOE varepsilon4 allele. Lancet Neurol. 2011;10:280–8.

    Article  CAS  Google Scholar 

  61. Murray ME, et al. Neuropathologically defined subtypes of Alzheimer’s disease with distinct clinical characteristics: a retrospective study. Lancet Neurol. 2011;10:785–96.

    Article  Google Scholar 

  62. Lind J, et al. Reduced hippocampal volume in non-demented carriers of the apolipoprotein E epsilon4: relation to chronological age and recognition memory. Neurosci Lett. 2006;396:23–7.

    Article  CAS  Google Scholar 

  63. Nilsson LG, et al. The influence of APOE status on episodic and semantic memory: data from a population-based study. Neuropsychology. 2006;20:645–57.

    Article  Google Scholar 

  64. Caselli RJ, et al. Longitudinal changes in cognition and behavior in asymptomatic carriers of the APOE e4 allele. Neurology. 2004;62:1990–5.

    Article  CAS  Google Scholar 

  65. Bondi MW, et al. Episodic memory changes are associated with the APOE-epsilon 4 allele in nondemented older adults. Neurology. 1995;45:2203–6.

    Article  CAS  Google Scholar 

  66. Michaud TL, Su D, Siahpush M, Murman DL. The risk of incident mild cognitive impairment and progression to dementia considering mild cognitive impairment subtypes. Dement Geriatr Cogn Dis Extra. 2017;7:15–29.

    Article  Google Scholar 

  67. Knopman DS, et al. Association of prior stroke with cognitive function and cognitive impairment: a population-based study. Arch Neurol. 2009;66:614–9.

    Article  Google Scholar 

  68. Rohn TT. Is apolipoprotein E4 an important risk factor for vascular dementia? Int J Clin Exp Pathol. 2014;7:3504–11.

    Google Scholar 

  69. Yin YW, et al. Association between apolipoprotein E gene polymorphism and the risk of vascular dementia: a meta-analysis. Neurosci Lett. 2012;514:6–11.

    Article  CAS  Google Scholar 

  70. Baum L, et al. Apolipoprotein E epsilon4 allele is associated with vascular dementia. Dement Geriatr Cogn Disord. 2006;22:301–5.

    Article  CAS  Google Scholar 

  71. Frisoni GB, et al. Association of apolipoprotein E E4 with vascular dementia. JAMA. 1994;271:1317.

    Article  CAS  Google Scholar 

  72. Tsuang D, et al. APOE epsilon4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. 2013;70:223–8.

    Article  Google Scholar 

  73. Borroni B, et al. APOE genotype and cholesterol levels in Lewy body dementia and Alzheimer disease: investigating genotype-phenotype effect on disease risk. Am J Geriatr Psychiatry. 2006;14:1022–31.

    Article  Google Scholar 

  74. Dickson DW, et al. APOE epsilon4 is associated with severity of Lewy body pathology independent of Alzheimer pathology. Neurology. 2018;91:e1182–95.

    Article  CAS  Google Scholar 

  75. Koriath C, et al. ApoE4 lowers age at onset in patients with frontotemporal dementia and tauopathy independent of amyloid-beta copathology. Alzheimers Dement (Amst). 2019;11:277–80.

    Article  Google Scholar 

  76. Seripa D, et al. The APOE gene locus in frontotemporal dementia and primary progressive aphasia. Arch Neurol. 2011;68:622–8.

    Article  Google Scholar 

  77. Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791–800.

    Article  Google Scholar 

  78. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82:239–59.

    Article  CAS  Google Scholar 

  79. Whitwell JL, et al. 3D maps from multiple MRI illustrate changing atrophy patterns as subjects progress from mild cognitive impairment to Alzheimer’s disease. Brain. 2007;130:1777–86.

    Article  Google Scholar 

  80. Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med. 2011;1:a006189.

    Article  CAS  Google Scholar 

  81. Drzezga A, et al. Effect of APOE genotype on amyloid plaque load and gray matter volume in Alzheimer disease. Neurology. 2009;72:1487–94.

    Article  CAS  Google Scholar 

  82. Jack CR Jr, et al. Hippocampal atrophy and apolipoprotein E genotype are independently associated with Alzheimer’s disease. Ann Neurol. 1998;43:303–10.

    Article  Google Scholar 

  83. Filippini N, et al. Anatomically-distinct genetic associations of APOE epsilon4 allele load with regional cortical atrophy in Alzheimer’s disease. NeuroImage. 2009;44:724–8.

    Article  Google Scholar 

  84. Juottonen K, Lehtovirta M, Helisalmi S, Riekkinen PJ Sr, Soininen H. Major decrease in the volume of the entorhinal cortex in patients with Alzheimer’s disease carrying the apolipoprotein E epsilon4 allele. J Neurol Neurosurg Psychiatry. 1998;65:322–7.

    Article  CAS  Google Scholar 

  85. Pievani M, et al. Mapping the effect of APOE epsilon4 on gray matter loss in Alzheimer’s disease in vivo. NeuroImage. 2009;45:1090–8.

    Article  CAS  Google Scholar 

  86. Lehtovirta M, et al. SPECT and MRI analysis in Alzheimer’s disease: relation to apolipoprotein E epsilon 4 allele. J Neurol Neurosurg Psychiatry. 1996;60:644–9.

    Article  CAS  Google Scholar 

  87. Lehtovirta M, et al. Volumes of hippocampus, amygdala and frontal lobe in Alzheimer patients with different apolipoprotein E genotypes. Neuroscience. 1995;67:65–72.

    Article  CAS  Google Scholar 

  88. Tanaka S, et al. Inferior temporal lobe atrophy and APOE genotypes in Alzheimer’s disease. X-ray computed tomography, magnetic resonance imaging and Xe-133 SPECT studies. Dement Geriatr Cogn Disord. 1998;9:90–8.

    Article  CAS  Google Scholar 

  89. Geroldi C, et al. APOE-epsilon4 is associated with less frontal and more medial temporal lobe atrophy in AD. Neurology. 1999;53:1825–32.

    Article  CAS  Google Scholar 

  90. Mattsson N, et al. Greater tau load and reduced cortical thickness in APOE epsilon4-negative Alzheimer’s disease: a cohort study. Alzheimers Res Ther. 2018;10:77.

    Article  CAS  Google Scholar 

  91. Jansen WJ, et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA. 2015;313:1924–38.

    Article  Google Scholar 

  92. Jack CR Jr, et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013;12:207–16.

    Article  CAS  Google Scholar 

  93. Tiraboschi P, et al. Impact of APOE genotype on neuropathologic and neurochemical markers of Alzheimer disease. Neurology. 2004;62:1977–83.

    Article  CAS  Google Scholar 

  94. Berg L, et al. Clinicopathologic studies in cognitively healthy aging and Alzheimer’s disease: relation of histologic markers to dementia severity, age, sex, and apolipoprotein E genotype. Arch Neurol. 1998;55:326–35.

    Article  CAS  Google Scholar 

  95. Rowe CC, et al. Amyloid imaging results from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol Aging. 2010;31:1275–83.

    Article  Google Scholar 

  96. Landen M, Thorsell A, Wallin A, Blennow K. The apolipoprotein E allele epsilon 4 does not correlate with the number of senile plaques or neurofibrillary tangles in patients with Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1996;61:352–6.

    Article  CAS  Google Scholar 

  97. Ossenkoppele R, et al. Differential effect of APOE genotype on amyloid load and glucose metabolism in AD dementia. Neurology. 2013;80:359–65.

    Article  CAS  Google Scholar 

  98. Lehmann M, et al. Greater medial temporal hypometabolism and lower cortical amyloid burden in ApoE4-positive AD patients. J Neurol Neurosurg Psychiatry. 2014;85:266–73.

    Article  Google Scholar 

  99. Shinohara M, et al. Impact of sex and APOE4 on cerebral amyloid angiopathy in Alzheimer’s disease. Acta Neuropathol. 2016;132:225–34.

    Article  CAS  Google Scholar 

  100. Thal DR, Ghebremedhin E, Orantes M, Wiestler OD. Vascular pathology in Alzheimer disease: correlation of cerebral amyloid angiopathy and arteriosclerosis/lipohyalinosis with cognitive decline. J Neuropathol Exp Neurol. 2003;62:1287–301.

    Article  Google Scholar 

  101. Ringman JM, et al. Clinical predictors of severe cerebral amyloid angiopathy and influence of APOE genotype in persons with pathologically verified Alzheimer disease. JAMA Neurol. 2014;71:878–83.

    Article  Google Scholar 

  102. Christensen DZ, Schneider-Axmann T, Lucassen PJ, Bayer TA, Wirths O. Accumulation of intraneuronal Abeta correlates with ApoE4 genotype. Acta Neuropathol. 2010;119:555–66.

    Article  CAS  Google Scholar 

  103. Ghebremedhin E, Schultz C, Braak E, Braak H. High frequency of apolipoprotein E epsilon4 allele in young individuals with very mild Alzheimer’s disease-related neurofibrillary changes. Exp Neurol. 1998;153:152–5.

    Article  CAS  Google Scholar 

  104. Corder EH, et al. The biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphism. Ann N Y Acad Sci. 2004;1019:24–8.

    Article  CAS  Google Scholar 

  105. Hohman TJ, et al. Sex-specific association of apolipoprotein E with cerebrospinal fluid levels of tau. JAMA Neurol. 2018;75:989–98.

    Article  Google Scholar 

  106. Buckley RF, et al. Associations between baseline amyloid, sex, and APOE on subsequent tau accumulation in cerebrospinal fluid. Neurobiol Aging. 2019;78:178–85.

    Article  CAS  Google Scholar 

  107. Petersen C, et al. Alzheimer’s disease clinical variants show distinct regional patterns of neurofibrillary tangle accumulation. Acta Neuropathol. 2019;138:597–612.

    Article  CAS  Google Scholar 

  108. Ossenkoppele R, et al. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain. 2016;139:1551–67.

    Article  Google Scholar 

  109. Whitwell JL, et al. [(18) F]AV-1451 clustering of entorhinal and cortical uptake in Alzheimer’s disease. Ann Neurol. 2018;83:248–57.

    Article  CAS  Google Scholar 

  110. Hanna Al-Shaikh FS, et al. Selective vulnerability of the nucleus basalis of Meynert among neuropathologic subtypes of Alzheimer disease. JAMA Neurol. 2019;77:225–33.

  111. Wennberg AM, Tosakulwong N, Lesnick TG, Murray ME, Whitwell JL, Liesinger AM, et al. Association of Apolipoprotein E ε4 With Transactive Response DNA-Binding Protein 43. JAMA Neurol. 2018;75(11):1347-1354. https://doi.org/10.1001/jamaneurol.2018.3139. PMID: 30422173; PMCID: PMC6248121.

  112. Yang HS, et al. Evaluation of TDP-43 proteinopathy and hippocampal sclerosis in relation to APOE epsilon4 haplotype status: a community-based cohort study. Lancet Neurol. 2018;17:773–81.

    Article  CAS  Google Scholar 

  113. Josephs KA, et al. TDP-43 is a key player in the clinical features associated with Alzheimer’s disease. Acta Neuropathol. 2014;127:811–24.

    Article  CAS  Google Scholar 

  114. Bayram E, Shan G, Cummings JL. Associations between comorbid TDP-43, Lewy body pathology, and neuropsychiatric symptoms in Alzheimer’s disease. J Alzheimers Dis. 2019;69:953–61.

    Article  Google Scholar 

  115. Chung EJ, et al. Clinical features of Alzheimer disease with and without Lewy bodies. JAMA Neurol. 2015;72:789–96.

    Article  Google Scholar 

  116. Emrani S, et al. Alzheimer’s/vascular spectrum dementia: classification in addition to diagnosis. J Alzheimers Dis. 2020;73:63–71.

    Article  Google Scholar 

  117. Barnes LL, et al. Mixed pathology is more likely in black than white decedents with Alzheimer dementia. Neurology. 2015;85:528–34.

    Article  Google Scholar 

  118. James BD, Bennett DA, Boyle PA, Leurgans S, Schneider JA. Dementia from Alzheimer disease and mixed pathologies in the oldest old. JAMA. 2012;307:1798–800.

    Article  CAS  Google Scholar 

  119. James BD, et al. TDP-43 stage, mixed pathologies, and clinical Alzheimer’s-type dementia. Brain. 2016;139:2983–93.

    Article  Google Scholar 

  120. Boyle PA, et al. Person-specific contribution of neuropathologies to cognitive loss in old age. Ann Neurol. 2018;83:74–83.

    Article  CAS  Google Scholar 

  121. Schneider JA, Arvanitakis Z, Bang W, Bennett DA. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology. 2007;69:2197–204.

    Article  Google Scholar 

  122. Poirier J, et al. Apolipoprotein E4 allele as a predictor of cholinergic deficits and treatment outcome in Alzheimer disease. Proc Natl Acad Sci U S A. 1995;92:12260–4.

    Article  CAS  Google Scholar 

  123. Waring JF, et al. APOE-varepsilon4 carrier status and donepezil response in patients with Alzheimer’s disease. J Alzheimers Dis. 2015;47:137–48.

    Article  CAS  Google Scholar 

  124. Bizzarro A, et al. Apolipoprotein E epsilon4 allele differentiates the clinical response to donepezil in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2005;20:254–61.

    Article  CAS  Google Scholar 

  125. Petersen RC, et al. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med. 2005;352:2379–88.

    Article  CAS  Google Scholar 

  126. Reger MA, et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging. 2006;27:451–8.

    Article  CAS  Google Scholar 

  127. Claxton A, et al. Long acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment or early-stage Alzheimer’s disease dementia. J Alzheimers Dis. 2015;45:1269–70.

    Article  Google Scholar 

  128. Risner ME, et al. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharmacogenomics J. 2006;6:246–54.

    Article  CAS  Google Scholar 

  129. Cummings JL, et al. Double-blind, placebo-controlled, proof-of-concept trial of bexarotene Xin moderate Alzheimer’s disease. Alzheimers Res Ther. 2016;8:4.

    Article  CAS  Google Scholar 

  130. Salloway S, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370:322–33.

    Article  CAS  Google Scholar 

  131. Hamanaka H, et al. Altered cholesterol metabolism in human apolipoprotein E4 knock-in mice. Hum Mol Genet. 2000;9:353–61.

    Article  CAS  Google Scholar 

  132. Chouinard-Watkins R, Plourde M. Fatty acid metabolism in carriers of apolipoprotein E epsilon 4 allele: is it contributing to higher risk of cognitive decline and coronary heart disease? Nutrients. 2014;6:4452–71.

    Article  CAS  Google Scholar 

  133. Gong JS, et al. Novel action of apolipoprotein E (ApoE): ApoE isoform specifically inhibits lipid-particle-mediated cholesterol release from neurons. Mol Neurodegener. 2007;2:9.

    Article  CAS  Google Scholar 

  134. DeKroon RM, Armati PJ. The endosomal trafficking of apolipoprotein E3 and E4 in cultured human brain neurons and astrocytes. Neurobiol Dis. 2001;8:78–89.

    Article  CAS  Google Scholar 

  135. Heeren J, et al. Impaired recycling of apolipoprotein E4 is associated with intracellular cholesterol accumulation. J Biol Chem. 2004;279:55483–92.

    Article  CAS  Google Scholar 

  136. Ji ZS, et al. Apolipoprotein E4 potentiates amyloid beta peptide-induced lysosomal leakage and apoptosis in neuronal cells. J Biol Chem. 2002;277:21821–8.

    Article  CAS  Google Scholar 

  137. Ye S, et al. Apolipoprotein (apo) E4 enhances amyloid beta peptide production in cultured neuronal cells: apoE structure as a potential therapeutic target. Proc Natl Acad Sci U S A. 2005;102:18700–5.

    Article  CAS  Google Scholar 

  138. Li J, et al. Differential regulation of amyloid-beta endocytic trafficking and lysosomal degradation by apolipoprotein E isoforms. J Biol Chem. 2012;287:44593–601.

    Article  CAS  Google Scholar 

  139. Yamauchi K, et al. Isoform-specific effect of apolipoprotein E on endocytosis of beta-amyloid in cultures of neuroblastoma cells. Ann Clin Lab Sci. 2002;32:65–74.

    CAS  Google Scholar 

  140. Rellin L, Heeren J, Beisiegel U. Recycling of apolipoprotein E is not associated with cholesterol efflux in neuronal cells. Biochim Biophys Acta. 2008;1781:232–8.

    Article  CAS  Google Scholar 

  141. Chen Y, Durakoglugil MS, Xian X, Herz J. ApoE4 reduces glutamate receptor function and synaptic plasticity by selectively impairing ApoE receptor recycling. Proc Natl Acad Sci U S A. 2010;107:12011–6.

    Article  CAS  Google Scholar 

  142. Nuriel T, et al. The endosomal-lysosomal pathway is dysregulated by APOE4 expression in vivo. Front Neurosci. 2017;11:702.

    Article  Google Scholar 

  143. Ong QR, Chan ES, Lim ML, Cole GM, Wong BS. Reduced phosphorylation of brain insulin receptor substrate and Akt proteins in apolipoprotein-E4 targeted replacement mice. Sci Rep. 2014;4:3754.

    Article  CAS  Google Scholar 

  144. Zhao N, et al. Apolipoprotein E4 impairs neuronal insulin signaling by trapping insulin receptor in the endosomes. Neuron. 2017;96:115–129 e115.

    Article  CAS  Google Scholar 

  145. Johnson LA, et al. Apolipoprotein E4 mediates insulin resistance-associated cerebrovascular dysfunction and the post-prandial response. J Cereb Blood Flow Metab. 2017;39:770–81.

  146. Johnson LA, Torres ER, Impey S, Stevens JF, Raber J. Apolipoprotein E4 and insulin resistance interact to impair cognition and alter the epigenome and metabolome. Sci Rep. 2017;7:43701.

    Article  Google Scholar 

  147. Keeney JT, Ibrahimi S, Zhao L. Human ApoE isoforms differentially modulate glucose and amyloid metabolic pathways in female brain: evidence of the mechanism of neuroprotection by ApoE2 and implications for Alzheimer’s disease prevention and early intervention. J Alzheimers Dis. 2015;48:411–24.

    Article  CAS  Google Scholar 

  148. Wu L, Zhang X, Zhao L. Human ApoE isoforms differentially modulate brain glucose and ketone body metabolism: implications for Alzheimer’s disease risk reduction and early intervention. J Neurosci. 2018;38:6665–81.

    Article  CAS  Google Scholar 

  149. Huebbe P, Lodge JK, Rimbach G. Implications of apolipoprotein E genotype on inflammation and vitamin E status. Mol Nutr Food Res. 2010;54:623–30.

    Article  CAS  Google Scholar 

  150. Rodriguez GA, Tai LM, LaDu MJ, Rebeck GW. Human APOE4 increases microglia reactivity at Abeta plaques in a mouse model of Abeta deposition. J Neuroinflammation. 2014;11:111.

    Article  Google Scholar 

  151. Shi Y, et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature. 2017;549:523–7.

    Article  CAS  Google Scholar 

  152. Bell RD, et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature. 2012;485:512–6.

    Article  CAS  Google Scholar 

  153. Tai LM, et al. The role of APOE in cerebrovascular dysfunction. Acta Neuropathol. 2016;131:709–23.

    Article  CAS  Google Scholar 

  154. Mielke MM, et al. Interaction between vascular factors and the APOE epsilon4 allele in predicting rate of progression in Alzheimer’s disease. J Alzheimers Dis. 2011;26:127–34.

    Article  CAS  Google Scholar 

  155. Montagne A, et al. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature. 2020;581:71–6.

    Article  CAS  Google Scholar 

  156. Gryglewski G, et al. Spatial analysis and high resolution mapping of the human whole-brain transcriptome for integrative analysis in neuroimaging. NeuroImage. 2018;176:259–67.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge Drs. Karen Duff and Mark Millan for their editing and advice on this review.

Funding

This work was supported by a career development award from the NIA to T.N. (AG061264).

Author information

Authors and Affiliations

Authors

Contributions

This review was written by S.E. and T.N., with additional editing and input by H.A.A. and C.D.M. The figure was created by H.A.A., and the tables were created by C.D.M. The authors read and approved the final manuscript.

Authors’ information

S.E. is a clinical psychology Ph.D. candidate who trained in the lab of Dr. David Libon at Rowan University. T.N. is an Assistant Professor at Columbia University whose research focuses on APOE4’s role in AD.

Corresponding author

Correspondence to Tal Nuriel.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Additional information

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 licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Emrani, S., Arain, H.A., DeMarshall, C. et al. APOE4 is associated with cognitive and pathological heterogeneity in patients with Alzheimer’s disease: a systematic review. Alz Res Therapy 12, 141 (2020). https://doi.org/10.1186/s13195-020-00712-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13195-020-00712-4

Keywords