Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder that is growing in prevalence globally. It is the only major cause of death without any effective pharmacological means to treat or slow progression. Inheritance of the ε4 allele of the Apolipoprotein (APO) E gene is the strongest genetic risk factor for late-onset AD. The interaction between APOE and amyloid β (Aβ) plays a key role in AD pathogenesis. The APOE-Aβ interaction regulates Aβ aggregation and clearance and therefore directly influences the development of amyloid plaques, congophilic amyloid angiopathy and subsequent tau related pathology. Relatively few AD therapeutic approaches have directly targeted the APOE-Aβ interaction thus far. Here we review the critical role of APOE in the pathogenesis of AD and some of the most promising therapeutic approaches that focus on the APOE-Aβ interaction.
Keywords: Apolipoprotein E, Immunomodulation, Oligomers, Early onset AD, Therapy, Peptoids, Pathological chaperone, Beta amyloid, Interaction
1. The critical role of apolipoprotein E in the pathogenesis of Alzheimer’s disease
Alzheimer’s disease (AD) is a devastating age associated neurodegenerative disorder. It is the sixth leading cause of death in the USA, with its prevalence expected to grow rapidly as the average age of the world’s population increases (Alzheimer’s Association, 2019; Long and Holtzman, 2019). AD is the only cause of death among the top ten causes of death globally for which no effective pharmaceutical agents exist to halt or slow disease progression. AD is defined neuropathologically by the accumulation of amyloid β (Aβ) into extracellular plaques in the brain parenchyma and in the vasculature (known as congophilic [or cerebral] amyloid angiopathy [CAA]), and abnormally phosphorylated tau that accumulates intraneuronally to form neurofibrillary tangles (NFTs) (Long and Holtzman, 2019; Rabinovici, 2019; Wisniewski and Drummond, 2019). In our early studies, we discovered that apolipoprotein E (APOE) was a new amyloid-associated protein that was abundantly present in amyloid plaques. We hypothesized that APOE was a “pathological chaperone” that directly promoted aggregation/fibrillization of Aβ (Wisniewski and Frangione, 1992;Wisniewski et al., 1993). Subsequent work by Dr. Allen Roses and his group identified the ε4 allele of the APOE gene as a key genetic risk factor for AD (Strittmatter et al., 1993a; Corder et al., 1993; Schmechel et al., 1993). This seminal observation was confirmed in other populations in the US, Europe and Australia (Saunders et al., 1996; Laws et al., 2002; Martins et al., 1995). Many confirmatory studies have now shown that the APOE4 genotype is the most common and potent AD genetic risk factor identified thus far (Potter and Wisniewski, 2012; Martins et al., 2018; Belloy et al., 2019; Roda et al., 2019; Tzioras et al., 2019). Our work and that of others showed that APOE4 particularly promoted Aβ oligomerization/fibrillization in comparison to APOE2 or APOE3 (Wisniewski et al., 1993; Golabek et al., 1996; Wisniewski and Frangione, 1996; Wisniewski et al., 1995a; Castano et al., 1995; Wisniewski et al., 1994a; Wisniewski et al., 1994b; Sanan et al., 1994; Potter et al., 2001; Ma et al., 1994). This effect was associated with an earlier onset of disease and greater amyloid plaque and CAA burden, in both patients and AD model transgenic mice expressing different human APOE genotypes (APOE4 > APOE3 > APOE2) (Martins et al., 2018; Strittmatter et al., 1993b; Hyman et al., 1995; Fryer et al., 2003; Bales et al., 2009; Holtzman et al., 2000; Resnick et al., 2015; Lim et al., 2017; Morris et al., 2010).
Phosphorylated tau and Aβ pathologically aggregate in a sequential process. Monomers first aggregate into oligomers intraneuronally, which then further aggregate into the fibrils observed in amyloid plaques and NFTs. This pathology then spreads throughout the brain in a characteristic manner that is distinct for NFTs and plaques (Scheltens et al., 2016; Selkoe and Hardy, 2016; Braak and Braak, 1991; Thal et al., 2002). This process develops over two to three decades resulting in a long preclinical period when AD associated neuropathology is present in the brain without any associated cognitive impairment (Martins et al., 2018; Belloy et al., 2019; Dubois et al., 2016; Pletnikova et al., 2018). Significant evidence indicates that oligomers of Aβ and tau are the most neurotoxic species in AD as levels of oligomers correlate better with cognitive decline compared to the burden of plaques or NFTs (Selkoe and Hardy, 2016; Viola and Klein, 2015; Sengupta et al., 2016; Lane et al., 2018). APOE genotype determines the age of onset for Alzheimer’s disease (APOE4 < APOE3 < APOE2) and directly influences the pathological aggregation of Aβ. Specifically, the Aβ-APOE interaction plays a key role in stabilizing toxic oligomers of Aβ, with APOE4 having a particularly pathological influence (Koffe et al., 2012; Hashimoto et al., 2012; Cerf et al., 2011).
In familial early onset AD (EOAD) there is either increased production of soluble Aβ (sAβ) or the production of more aggregation prone species, while in sporadic late onset AD (LOAD) there is impaired clearance of soluble Aβ (Selkoe and Hardy, 2016). EOAD pathology is also universal in Down syndrome (DS), which is attributed to the increased production of Aβ in DS due to the presence of three copies of the amyloid precursor protein (Head et al., 2016; Hartley et al., 2015).Autosomal dominant mutations in presenilin 1, presenilin 2 (PSEN1 and PSEN2) or the amyloid precursor protein (APP) account for only ~10% of all EOAD cases (~1% of all AD cases), leaving the cause of the majority of EOAD unexplained (Cacace et al., 2016; Guerreiro and Hardy, 2014; Wingo et al., 2012; Pimenova et al., 2017). LOAD afficts > 95% of patients with AD and is related to both genetic and environmental factors (Guerreiro and Hardy, 2014; Karch et al., 2014; Bertram and Tanzi, 2012; Kim et al., 2014; Kim, 2018). A combination of genome-wide association studies (GWAS), linkage, and whole genome/exome sequencing have identified over 30 loci that confer increased risk for LOAD, including genes involved in innate immunity, cholesterol metabolism and synaptic/neuronal membrane function, suggesting that the pathogenesis of LOAD has considerable heterogeneity (Guerreiro and Hardy, 2014; Pimenova et al., 2017; Kim, 2018; Cuyvers and Sleegers, 2016; Karch and Goate, 2015; Jansen et al., 2019; Kunkle et al., 2019). Of these, APOE is the strongest identified genetic risk factor for LOAD (Potter and Wisniewski, 2012; Belloy et al., 2019; Huang and Mahley, 2014; Zhao et al., 2018). Variants of another gene that encodes the triggering receptor expressed on myeloid cells 2 (TREM2) have also been reported as a significant risk factor for LOAD, with an odds ratio approaching that of APOE4; however, these TREM2 variants are uncommon (Ulrich et al., 2017; Efthymiou and Goate, 2017; Shi and Holtzman, 2018).
This genetic diversity that drives AD pathogenesis suggests that AD is a syndrome associated with a neuropathological signature of Aβ and tau oligomer/fibril accumulation, where neither amyloid plaques or NFTs necessarily have a causative role. Our understanding of these complex pathways has greatly increased in recent years; however, despite this expanding knowledge base there has been a very high failure rate of ~99.6% of AD targeting clinical trials (Cummings et al., 2014; Banik et al., 2015; Schneider et al., 2014). There are many reasons for this high failure rate, which have been reviewed elsewhere (Long and Holtzman, 2019; Wisniewski and Drummond, 2019; Herline et al., 2018; Elmaleh et al., 2019); however, one factor associated with this lack of success is that relatively few therapeutic studies are targeting the critical role of APOE in AD. Even in EOAD and DS, where AD pathology is primarily driven by overproduction of Aβ, expression of the APOE4 genotype has an additive detrimental effect and lowers the age of onset, while APOE2 has a protective effect (van Duijn et al., 1994; Sorbi et al., 1995; Velez et al., 2016; Wijsman et al., 2005; Coppus et al., 2008; Wisniewski et al., 1995b; Royston et al., 1996). The paramount role APOE in AD pathogenesis is particularly highlighted in a recent report of a patient with the Colombia kindred PSEN1 E280A mutation, which typically causes the clinical onset of mild cognitive impairment (MCI) and dementia at the median ages of 44 and 49 years respectively (Arboleda-Velasquez et al., 2019; Zalocusky et al., 2019). This study found that the additional homozygous expression of the rare Christchurch APOE3 mutation of R154S (APOE3ch) resulted in resistance to the effects of the PS1 mutation; the patient did not develop MCI until her seventies, with very little tau pathology being detected by PET tracers (Arboleda-Velasquez et al., 2019). The APOE3ch variant protein has a significantly reduced ability to promote Aβ 1–42 peptide aggregation compared to wild-type APOE3, which was comparable to the effect of APOE2. The R154S mutation is in the region of APOE involved in lipoprotein receptor and heparin sulfate proteoglycan binding (Hashimoto et al., 2012). In addition, this region is involved in APOE binding to Aβ. Aβ binds to APOE both at the lipid binding region of residues 244–272 and at the N-terminal domain (Liu et al., 2011; Deroo et al., 2015; Luo et al., 2010). This case illustrates the dramatic effect that altering the APOE-Aβ interaction can have in vivo, indicating that abrogation of this interaction is highly protective against the subsequent development of dementia, even in settings with abundant Aβ deposition. Interestingly, another rare APOE variant p.V236E in the lipid-binding, C-terminal domain of APOE (also involved in Aβ binding) is associated with a markedly reduced risk of AD (Medway et al., n.d.). The key role of APOE in AD pathogenesis may also explain why AD is a uniquely human disease (Drummond and Wisniewski, 2017; Walker and Jucker, 2017). The most biologically proximate animals to humans are non-human primates (NHP). It is well established that many NHP species develop abundant age dependent amyloid plaques and/or vascular amyloid deposition, yet none of these species develop significant (or any) NFT pathology or AD like dementia (Walker and Jucker, 2017; Devinsky et al., 2018; Heuer et al., 2012). All NHP are homozygous for APOE4, in that NHP APOE has an arginine at positions 112 and 158 (Walker and Jucker, 2017; Morelli et al., 1996). However, NHP APOE has a threonine instead of an arginine at amino acid position 61, causing it to function biologically in a manner similar to APOE3 (Walker and Jucker, 2017; Morelli et al., 1996). Hence, NHP lack a biological APOE4 like protein to interact with Aβ and drive the subsequent steps needed for the emergence of the AD phenotype. These data suggests that the APOE-Aβ interaction could be a highly effective therapeutic target.
2. Targeting the major genetic risk factor for AD: Apolipoprotein E4
APOE has pleiotropic functions in the CNS that include being the major CNS cholesterol and other lipid carrier. In addition, it is involved in vascular integrity, synaptic plasticity, glucose metabolism and mitochondrial function (Belloy et al., 2019; Huang and Mahley, 2014; Zhao et al., 2018). Hence, one important consideration when targeting the role of APOE in AD is that these normal functions should not be compromised. In addition, APOE is highly expressed outside of the brain, therefore off target effects must be considered when developing a therapeutic approach. Furthermore, APOE concurrently influences both the clearance and aggregation of Aβ in an isotype specific manner (Potter and Wisniewski, 2012; Belloy et al., 2019; Zhao et al., 2018;Hudry et al., 2019; Huynh et al., 2017; Bell et al., 2007; Han et al., 2016); hence, therapeutic approaches need to carefully balance these potentially opposing roles. In AD, it is still unclear whether the pathogenic role of APOE4 results from a toxic gain of function or loss of protective function. Transgenic mouse studies suggest that APOE4 has a toxic gain of function specifically with regards to its interaction with Aβ, while other pathological efects of APOE4 (e.g. astrocyte activation and synaptic loss) may result from loss of protective function (Safieh et al., 2019).
APOE enhances aggregation of Aβ in the order of APOE4 > APOE3 > APOE2 (Wisniewski and Frangione, 1992;Castano et al., 1995; Wisniewski et al., 1994b; Ma et al., 1994; Hori et al., 2015). APOE isotype specific effects have also been observed with regards to the stabilization of Aβ oligomers, where APOE4 was again shown to have the greatest influence (Hashimoto et al., 2012; Garai et al., 2014). In physiological conditions, there is relatively minor interaction between APOE and soluble Aβ (Verghese et al., 2013).
Instead, APOJ is the major CNS Aβ binding protein (Calero et al., 2000; Matsubara et al., 1995). However, a greater interaction between APOE and Aβ is observed in AD as the aggregate state of Aβ shifts (Wisniewski et al., 1995a; Han et al., 2016; Golabek et al., 1995). The finding that APOE4 was less effective in clearing Aβ than APOE3 (Castellano et al., 2011) led to the initial hypothesis that blocking the interaction between Aβ and APOE could decrease the clearance of Aβ and therefore increase the formation of Aβ plaques. However, pivotal in vivo studies show that this does not occur. Blocking the Aβ/APOE interaction instead results in enhanced Aβ clearance from the brain and decreased plaque deposition (Sadowski et al., 2006; Sadowski et al., 2004; Yang et al., 2011; Liu et al., 2014; Pankiewicz et al., 2014). This is consistent with the finding that eliminating APOE greatly reduces the amount of amyloid plaque and vessel pathology in AD mouse models (Bales et al., 1997; Miao et al., 2005). Also supporting the hypothesis that APOE is an AD pathology promoter, rather than APOE4 having a loss of normal Aβ brain clearance function, is human data from a rare individual who lacks APOE due to an ablative APOE frameshift mutation. Despite complete absence of APOE this individual had no cognitive deficits, had normal brain MRI findings and normal CSF levels of Aβ and tau proteins (Mak et al., 2014). Multiple approaches for therapeutically targeting APOE in AD have been explored. Major examples of these are listed below.
2.1. Blocking the APOE/Aβ interaction
We have shown that treatment with a peptide that interferes with the Aβ/APOE interaction significantly decreased the amount of both parenchymal and vascular Aβ in three AD transgenic mouse models (Sadowski et al., 2006; Sadowski et al., 2004; Yang et al., 2011; Liu et al., 2014). This peptide (Aβ12–28P) is homologous to the APOE binding domain of Aβ, and therefore inhibits the Aβ-APOE interaction (Ma et al., 1996). The peptide was synthesized with D-amino acids and a proline substitution of valine at residue 18 of Aβ, which improved its resistance to proteolysis and ensures that it is non-toxic and non-fibrillogenic (Sadowski et al., 2006; Sadowski et al., 2004). Treatment of 3xTg mice with this peptide reduced both Aβ and tau pathology, while in TgSwDI mice with extensive CAA, it reduced vascular amyloid pathology (Yang et al., 2011; Liu et al., 2014). Hence, as illustrated in Fig. 1, altering the APOE-Aβ interaction can reduce all major AD related pathological lesions, in the absence of toxicity such as increased inflammation or microhemorrhages.
Corroborating results from another group have found that Aβ12–28P treatment caused a similar reduction of Aβ oligomer and plaque deposition in amyloid Tg mice with both an APOE2-targeted replacement (TR) or APOE4-TR mouse background, showing that the inhibition of the Aβ/APOE interaction is therapeutically beneficial regardless of the APOE isoform (Pankiewicz et al., 2014). More recently, we have enhanced our approach for potential future clinical application. We have designed and screened a large group of linear and cyclic peptoid compounds that block the Aβ/APOE interaction in a similar manner, to try to identify a compound that has higher effcacy and safety (Liu et al., 2017). The most promising candidate, CPO_Aβ17–21P, inhibited the APOE4/Aβ42 binding at a 2:1 M ratio and virtually blocked all binding at a 8:1 M ratio (peptoid:APOE4). This new candidate also had a significantly improved half-maximal inhibition (IC50) in comparison to Aβ12–28P (Sadowski et al., 2004). The Aβ residues between 17 and 21 appear to be the critical region for Aβ binding to APOE, with the lysine at residue 16 being particularly important (Liu et al., 2011; Deroo et al., 2015), therefore it is consistent that a peptoid conforming to this sequence is an effective inhibitor of the Aβ/APOE interaction. Treatment of APP/PS1 Tg mice with CPO_Aβ17–21P resulted in significant reduction of soluble and insoluble Aβ peptide/oligomer levels in brain, lower numbers of amyloid plaques and significantly improved cognitive function (Liu et al., 2017). Importantly, all of these effects were observed after treatment with a 7.5 fold lower dose than required for the Aβ12–28P studies (Liu et al., 2014; Liu et al., 2017), indicating that CPO_Aβ17–21P has improved bioavailability/biostability over Aβ12–28P. Importantly, treatment of APP/PS1 AD transgenic mice with CPO_Aβ17–21P did not increase the soluble Aβ pool. Additionally, there was no evidence of increased brain inflammation after treatment (Liu et al., 2017), which has been another possible concern with therapeutic strategies that target Aβ. We are in the process of using an innovative “scaffold hopping” medicinal chemistry approach (Hu et al., 2017) from CPO_Aβ17–21P to drug-like small molecule candidates, along with a receptor-based scaffold-independent approach to further refine this therapeutic approach. Our preclinical studies in multiple AD models have demonstrated that blocking the APOE/Aβ interaction is a promising therapeutic approach that is capable of reducing AD associated neuropathology and improving cognitive performance, in the absence of toxicity.
2.2. APOE immunotherapy
Immunotherapy using anti-APOE antibodies is also a potential therapeutic strategy for AD. Initial studies showed that treatment with an antibody against endogenous mouse APOE (HJ6.3) significantly reduced Aβ pathology in an AD transgenic mouse model (Kim et al., 2012;Liao et al., 2014). Recently, this work has been extended to show that treatment with an antibody against human APOE (HAE-4) also significantly reduces Aβ plaque pathology in an AD transgenic mouse model expressing human APOE4 (APPPS1–21/APOE4 mice). This antibody recognizes both APOE3 and APOE4 and preferentially binds to non-lipidated, aggregated APOE (Liao et al., 2018). While the exact mechanism of action is still unclear, it was proposed that the binding of anti-APOE antibodies to APOE present in amyloid plaques stimulated microglial activation via their Fc domain, which therefore resulted in plaque clearance. Further testing of this approach is underway; however, one potential problem is that off-target antibody binding may detrimentally interfere with the physiological functions of APOE.
2.3. Altering APOE4 conformation
Another means of ameliorating the pathological interaction between APOE4 and Aβ is to make it behave more like APOE3. The point mutations present in APOE4 result in a unique conformation due to a specific interaction between its amino-terminal and carboxyl terminal domains. Small molecule inhibitors have been developed to interfere with this interaction and therefore alter the conformation of APOE4 so that it resembles that of APOE3 or APOE2 (Mahley and Huang, 2012; Chen et al., 2012; Brodbeck et al., 2011). One of these structure correctors has been shown to reduce the APOE4 effects on Aβ, tau phosphorylation and neurodegeneration in human iPSC derived neurons expressing APOE4 (Wang et al., 2018). This approach is being further developed and tested as it is still unclear whether it has therapeutic benefits in vivo.
2.4. Expression of APOE2
Another elegant method of targeting the AD pathology promoting effects of APOE4 expression is to genetically drive production of “protective” APOE2 expression. Early studies showed that viral vector mediated expression of APOE2 in AD transgenic mouse models expressing endogenous mouse APOE resulted in significantly reduced amyloid plaque burden, significantly reduced levels of insoluble Aβ42 and Aβ40, and significantly reduced synapse loss around plaques in comparison to viral vector mediated expression of APOE4 (Hudry et al., 2013; Dodart et al., 2005). Importantly, it was then shown that viral vector mediated expression of APOE2 partially countered the detrimental effects of human APOE4 expression in AD transgenic mouse models if given at early stages of pathology development: AAV-APOE2 treatment of APP.PS1/APOE-TR mice resulted in significantly decreased levels of insoluble Aβ42 and Aβ40 in the brain (Zhao et al., 2016).
A more recent study tested the delivery of AAVrh.10hAPOE2-HA, an AAVrh.10 serotype coding for an HA-tagged human APOE2 by intraparenchymal, intracisternal, and intraventricular routes of delivery to the CNS of African Green monkeys (Rosenberg et al., 2018). The data showed that while all three routes are capable of mediating APOE2 expression in AD relevant regions, intracisternal delivery of AAVrh.10hAPOE2-HA safely mediated wide distribution of APOE2 with the least invasive surgery, thus potentially providing the optimal strategy to deliver vector-mediated human APOE2 to the CNS. A phase I clinical trial is now on-going in AD APOE4 homozygous patients of AAVrh-10APOE2 delivered by intracisternal injection (Crystal et al., 2019).
2.5. Gene editing APOE4 to APOE3
Direct conversion of APOE4 to APOE2 or APOE3 using a gene editing strategy such as CRISPR could be a potentially straightforward therapeutic approach. Proof-of-concept studies in vitro using iPSCs and organoids has shown that this approach has potential; conversion of APOE4 to APOE3 in these models was shown to decrease Aβ, and to reduce tau phosphorylation and neurodegeneration (Wang et al., 2018;Lin et al., 2018). However, using gene editing as a therapeutic approach is still in its infancy and it is not yet known whether this would work in vivo.
3. Summary
There is extensive evidence that APOE alleles differentially modulate AD pathogenesis by varying effects on net Aβ oligomerization/aggregation versus clearance. The APOE4 isoform remains the most important genetic risk factor for AD. Recent studies have shown strikingly dramatic APOE mediated effects even in the setting of an EOAD PS1 mutation (Arboleda-Velasquez et al., 2019; Zalocusky et al., 2019). The road to the discovery of effective AD therapies has been marked by a long string of failures, at least in part, due to relatively few investigators directly targeting the critical role of APOE in AD. In this review, we summarize a few of what we feel are the most promising on-going approaches to overcome this shortcoming in the field. These approaches have great potential for both the prevention and/or treatment of AD in the absence of significant toxicity.
Acknowledgments
This manuscript was supported the NIH grants NS073502 and AG008051 and by funding from the Bluesand Foundation, the Edward and Della L. Thome Memorial Foundation and Dementia Australia. We thank Geoffrey Pires for his assistance with figure preparation.
References
- Alzheimer’s Association, 2019. 2019 Alzheimer’s disease facts and figures. AlzheimersDement 15 (3), 321–387. [Google Scholar]
- Arboleda-Velasquez JF, Lopera F, O’Hare M, Delgado-Tirado S, Marino C,Chmielewska N, Saez-Torres KL, Amarnani D, Schultz AP, Sperling RA, Leyton-Cifuentes D, Chen K, Baena A, Aguillon D, Rios-Romenets S, Giraldo M, Guzmán-Vélez E, Norton DJ, Pardilla-Delgado E, Artola A, Sanchez JS, Acosta-Uribe J, Lalli M, Kosik KS, Huentelman MJ, Zetterberg H, Blennow K, Reiman RA, Luo J, Chen Y, Thiyyagura P, Su Y, Jun GR, Naymik M, Gai X, Bootwalla M, Ji J, Shen L, Miller JB, Kim LA, Tariot PN, Johnson KA, Reiman EM, Quiroz YT, 2019. Resistance to autosomal dominant Alzheimer’s disease in an APOE3 Christchurch homozygote: a case report. Nat. Med 10.1038/s41591-019-0611-3. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bales KR, Liu F, Wu S, Lin S, Koger D, DeLong C, Hansen JC, Sullivan PM, Paul SM, 2009. Human APOE isoform-dependent effects on brain beta-amyloid levels in PDAPP transgenic mice. J. Neurosci 29 (21), 6771–6779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bales KR, Verina T, Dodel RC, Du YS, Altstiel L, Bender M, Hyslop P,Johnstone EM, Little SP, Cummins DJ, Piccardo P, Ghetti B, Paul SM, 1997. Lack of apolipoprotein E dramatically reduces amyloid β-peptide deposition. Nat.Gen 17 (3), 263–264. [DOI] [PubMed] [Google Scholar]
- Banik A, Brown RE, Bamburg J, Lahiri DK, Khurana D, Friedland RP, Chen W,Ding Y, Mudher A, Padjen AL, Mukaetova-Ladinska E, Ihara M, Srivastava S, Padma Srivastava MV, Masters CL, Kalaria RN, Anand A, 2015. Translation of pre-clinical studies into successful clinical trials for Alzheimer’s disease: what are the roadblocks and how can they be overcome? J. Alzheimers Dis 47 (4), 815–843. 10.3233/JAD-150136. [DOI] [PubMed] [Google Scholar]
- Bell RD, Sagare AP, Friedman AE, Bedi GS, Holtzman DM, Deane R, Zlokovic BV, 2007. Transport pathways for clearance of human Alzheimer’s amyloid beta-peptide and apolipoproteins E and J in the mouse central nervous system. J. Cereb.Blood Flow Metab 27 (5), 909–918. 10.1038/sj.jcbfm.9600419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belloy ME, Napolioni V, Greicius MD, 2019. A quarter century of APOE and Alzheimer’s disease: progress to date and the path forward. Neuron 101 (5), 820–838. Epub 2019/03/08. 10.1016/j.neuron.2019.01.056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertram L, Tanzi RE, 2012. The genetics of Alzheimer’s disease. Prog. Mol. Biol.Transl. Sci 107, 79–100. [DOI] [PubMed] [Google Scholar]
- Braak H, Braak E, 1991. Neuropathological staging of Alzheimer-related changes. ActaNeuropathol 82, 239–259. [DOI] [PubMed] [Google Scholar]
- Brodbeck J, McGuire J, Liu Z, Meyer-Franke A, Balestra ME, Jeong DE, Pleiss M, McComas C, Hess F, Witter D, Peterson S, Childers M, Goulet M, Liverton N, Hargreaves R, Freedman S, Weisgraber KH, Mahley RW, Huang Y, 2011. Structure-dependent impairment of intracellular apolipoprotein E4 traffcking and its detrimental effects are rescued by small-molecule structure correctors. J. Biol. Chem 286 (19), 17217–17226. 10.1074/jbc.M110.217380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cacace R, Sleegers K, Van Broeckhoven C, 2016. Molecular genetics of early-onset Alzheimer’s disease revisited. Alzheimers Dement 12 (6), 733–748. 10.1016/j.jalz.2016.01.012. [DOI] [PubMed] [Google Scholar]
- Calero M, Rostagno A, Matsubara E, Zlokovic B, Frangione B, Ghiso J, 2000. Apolipoprotein J (clusterin) and Alzheimer’s disease. Microsc. Res. Tech 50 (4), 305–315. . [DOI] [PubMed] [Google Scholar]
- Castano EM, Prelli F, Wisniewski T, Golabek A, Kumar RA, Soto C, Frangione B, 1995. Fibrillogenesis in Alzheimer’s disease of amyloid beta peptides and apolipoprotein E. Biochem. J 306 (Pt 2), 599–604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, Fagan AM, Morris JC, Mawuenyega KG, Cruchaga C, Goate AM, Bales KR, Paul SM, Bateman RJ, Holtzman DM, 2011. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci. Transl. Med 3 (89), 89ra57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerf E, Gustot A, Goormaghtigh E, Ruysschaert JM, Raussens V, 2011. High ability of apolipoprotein E4 to stabilize amyloid-beta peptide oligomers, the pathological entities responsible for Alzheimer’s disease. FASEB J 25 (5), 1585–1595. Epub 2011/01/27. 10.1096/fj.10-175976. [DOI] [PubMed] [Google Scholar]
- Chen HK, Liu Z, Meyer-Franke A, Brodbeck J, Miranda RD, McGuire JG, Pleiss MA, Ji ZS, Balestra ME, Walker DW, Xu Q, Jeong DE, Budamagunta MS, Voss JC, Freedman SB, Weisgraber KH, Huang Y, Mahley RW, 2012. Small molecule structure correctors abolish detrimental effects of apolipoprotein E4 in cultured neurons. J. Biol. Chem 287 (8), 5253–5266. Epub 2011/12/14. 10.1074/jbc.M111.276162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coppus AM, Evenhuis HM, Verberne GJ, Visser FE, Arias-Vasquez A, Sayed-Tabatabaei FA, Vergeer-Drop J, Eikelenboom P, van Gool WA, van Duijn CM, 2008. The impact of apolipoprotein E on dementia in persons with Down’s syndrome. Neurobiol. Aging 29 (6), 828–835. Epub 2007/01/26. 10.1016/j.neurobiolaging.2006.12.013. [DOI] [PubMed] [Google Scholar]
- Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA, 1993. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261 (5123), 921–923. [DOI] [PubMed] [Google Scholar]
- Crystal RG, Gandy S, Sano M, 2019. Gene Therapy for APOE4 Homozygote of Alzheimer’s Disease Available from: https://clinicaltrials.gov/ct2/show/NCT03634007.
- Cummings JL, Morstorf T, Zhong K, 2014. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res. Ther 6 (4), 37. 10.1186/alzrt269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cuyvers E, Sleegers K, 2016. Genetic variations underlying Alzheimer’s disease: evidence from genome-wide association studies and beyond. Lancet Neurol 15 (8), 857–868. 10.1016/S1474-4422(16)00127-7. [DOI] [PubMed] [Google Scholar]
- Deroo S, Stengel F, Mohammadi A, Henry N, Hubin E, Krammer EM, Aebersold R, Raussens V, 2015. Chemical cross-linking/mass spectrometry maps the amyloid beta peptide binding region on both apolipoprotein E domains. ACS Chem. Biol 10(4), 1010–1016. 10.1021/cb500994j. [DOI] [PubMed] [Google Scholar]
- Devinsky O, Boesch JM, Cerda-Gonzalez S, Coffey B, Davis K, Friedman D, Hainline B, Houpt K, Lieberman D, Perry P, Prüss H, Samuels MA, Small GW, Volk H, Summerfield A, Vite C, Wisniewski T, Natterson-Horowitz B, 2018. A cross-species approach to disorders affecting brain and behaviour. Nat. Rev. Neurol 10.1038/s41582-018-0074-z. in press. [DOI] [PubMed] [Google Scholar]
- Dodart JC, Marr RA, Koistinaho M, Gregersen BM, Malkani S, Verma IM, Paul SM, 2005. Gene delivery of human apolipoprotein E alters brain Abeta burden in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. U. S. A 102 (4), 1211–1216. Epub 2005/01/20. 10.1073/pnas.0409072102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drummond E, Wisniewski T, 2017. Alzheimer’s disease: experimental models and reality. Acta Neuropathol 133 (2), 155–175. 10.1007/s00401-016-1662-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues JF, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert MO, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O’Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R,Teichmann M, Carrillo MC, Cummings J, Jack CR Jr., 2016. Proceedings of the Meeting of the International Working G, the American Alzheimer’s Association on The Preclinical State of AD, July, Washington DC USA. Preclinical Alzheimer’s disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 12 (3), 292–323. 10.1016/j.jalz.2016.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Duijn CM, de Knijff P, Cruts M, Wehnert A, Havekes LM, Hofman A, Van Broeckhoven C, 1994. Apolipoprotein E4 allele in a population-based study of early-onset Alzheimer’s disease. Nat. Gen 7 (1), 74–78. [DOI] [PubMed] [Google Scholar]
- Efthymiou AG, Goate AM, 2017. Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk. Mol. Neurodegener 12 (1), 43. 10.1186/s13024-017-0184-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmaleh DR, Farlow MR, Conti PS, Tompkins RG, Kundakovic L, Tanzi RE,2019. Developing effective Alzheimer’s disease therapies: clinical experience and future directions. J. Alzheimers Dis 71 (3), 715–732. Epub 2019/09/03. 10.3233/JAD-190507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fryer JD, Taylor JW, DeMattos RB, Bales KR, Paul SM, Parsadanian M,Holtzman DM, 2003. Apolipoprotein E markedly facilitates age-dependent cerebral amyloid angiopathy and spontaneous hemorrhage in amyloid precursor protein transgenic mice. J. Neurosci 23 (21), 7889–7896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garai K, Verghese PB, Baban B, Holtzman DM, Frieden C, 2014. The binding of apolipoprotein E to oligomers and fibrils of amyloid-beta alters the kinetics of amyloid aggregation. Biochemistry 53 (40), 6323–6331. 10.1021/bi5008172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golabek A, Marques MA, Lalowski M, Wisniewski T, 1995. Amyloid beta binding proteins in vitro and in normal human cerebrospinal fluid. Neurosci. Lett 191 (1–2), 79–82. [DOI] [PubMed] [Google Scholar]
- Golabek AA, Soto C, Vogel T, Wisniewski T, 1996. The interaction between apolipoprotein E and Alzheimer’s amyloid β-peptide is dependent on β-peptide conformation. J. Biol. Chem 271, 10602–10606. [DOI] [PubMed] [Google Scholar]
- Guerreiro R, Hardy J, 2014. Genetics of Alzheimer’s disease. Neurotherapeutics 11 (4),432–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han SH, Park JC, Mook-Jung I, 2016. Amyloid beta-interacting partners in Alzheimer’s disease: from accomplices to possible therapeutic targets. Prog. Neurobiol 137, 17–38. 10.1016/j.pneurobio.2015.12.004. [DOI] [PubMed] [Google Scholar]
- Hartley D, Blumenthal T, Carrillo M, DiPaolo G, Esralew L, Gardiner K, Granholm AC, Iqbal K, Krams M, Lemere CA, Lott I, Mobley WC, Ness S, Nixon R, Potter H, Reeves R, Sabbagh M, Silverman W, Tycko B, Whitten M, Wisniewski T, 2015. Down syndrome and Alzheimer’s Disease: common pathways, common goals. Alzheimer’s Dementia 11 (6), 700–709 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashimoto T, Serrano-Pozo A, Hori Y, Adams KW, Takeda S, Banerji AO, Mitani A, Joyner D, Thyssen DH, Bacskai BJ, Frosch MP, Spires-Jones TL, Finn MB, Holtzman DM, Hyman BT, 2012. Apolipoprotein E, especially apolipoprotein E4, increases the oligomerization of amyloid beta peptide. J. Neurosci 32 (43), 15181–15192. 10.1523/JNEUROSCI.1542-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Head E, Lott IT, Wilcock DM, Lemere CA, 2016. Aging in Down syndrome and the development of Alzheimer’s disease neuropathology. Curr. Alzheimer Res 13 (1), 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herline K, Drummond E, Wisniewski T, 2018. Recent advancements toward therapeutic vaccines against Alzheimer’s disease. Expert Rev. Vaccin 17 (8), 707–721. Epub 2018/07/15. 10.1080/14760584.2018.1500905. [DOI] [PubMed] [Google Scholar]
- Heuer E, Rosen RF, Cintron A, Walker LC, 2012. Nonhuman primate models of Alzheimer-like cerebral proteopathy. Curr. Pharm. Des 18 (8), 1159–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holtzman DM, Bales KR, Tenkova T, Fagan AM, Parsadanian M, Sartorius LJ, Mackey B, Olney J, McKeel D, Wozniak D, Paul SM, 2000. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. PNAS 97, 2892–2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hori Y, Hashimoto T, Nomoto H, Hyman BT, Iwatsubo T, 2015. Role ofApolipoprotein E in beta-Amyloidogenesis: isoform-specific effects on protofibril to fibril conversion of abeta in vitro and brain abeta deposition in vivo. J. Biol. Chem 290 (24), 15163–15174. Epub 2015/04/29. 10.1074/jbc.M114.622209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Y, Stumpfe D, Bajorath J, 2017. Recent advances in scaffold hopping. J. Med. Chem 60 (4), 1238–1246. 10.1021/acs.jmedchem.6b01437. [DOI] [PubMed] [Google Scholar]
- Huang Y, Mahley RW, 2014. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. Neurobiol. Dis 72 (Pt A), 3–12. 10.1016/j.nbd.2014.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hudry E, Dashkoff J, Roe AD, Takeda S, Koffie RM, Hashimoto T, Scheel M, Spires-Jones T, Arbel-Ornath M, Betensky R, Davidson BL, Hyman BT, 2013. Gene transfer of human Apoe isoforms results in differential modulation of amyloid deposition and neurotoxicity in mouse brain. Sci. Transl. Med 5 (212). 10.1126/scitranslmed.3007000.212ra161.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hudry E, Klickstein J, Cannavo C, Jackson R, Muzikansky A, Gandhi S, Urick D, Sargent T, Wrobleski L, Roe AD, Hou SS, Kuchibhotla KV, Betensky RA, Spires-Jones T, Hyman BT, 2019. Opposing roles of apolipoprotein E in aging and neurodegeneration. Life Sci Alliance 2 (1). 10.26508/lsa.201900325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huynh TV, Davis AA, Ulrich JD, Holtzman DM, 2017. Apolipoprotein E and Alzheimer disease: the influence of apoE on amyloid-beta and other amyloidogenic proteins. J. Lipid Res 10.1194/jlr.R075481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hyman BT, West HL, Rebeck GW, Buldyrev SV, Mantegna RN, Ukleja M,Havlin S, Stanley HE, 1995. Quantitative analysis of senile plaques in Alzheimer disease: observation of log-normal size distribution and molecular epidemiology of differences associated with apolipoprotein E genotype and trisomy 21 (Down syndrome). Proc. Natl. Acad. Sci. U. S. A 92 (8), 3586–3590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jansen IE, Savage JE, Watanabe K, Bryois J, Williams DM, Steinberg S, Sealock J, Karlsson IK, Hagg S, Athanasiu L, Voyle N, Proitsi P, Witoelar A, Stringer S, Aarsland D, Almdahl IS, Andersen F, Bergh S, Bettella F, Bjornsson S, Braekhus A, Brathen G, de Leeuw C, Desikan RS, Djurovic S, Dumitrescu L, Fladby T, Hohman TJ, Jonsson PV, Kiddle SJ, Rongve A, Saltvedt I, Sando SB, Selbaek G, Shoai M, Skene NG, Snaedal J, Stordal E, Ulstein ID, Wang Y, White LR, Hardy J, Hjerling-Leffer J, Sullivan PF, van der Flier WM, Dobson R, Davis LK, Stefansson H, Stefansson K, Pedersen NL, Ripke S, Andreassen OA, Posthuma D, 2019. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet 51 (3), 404–413. Epub 2019/01/09. 10.1038/s41588-018-0311-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karch CM, Cruchaga C, Goate AM, 2014. Alzheimer’s disease genetics: from the bench to the clinic. Neuron 83 (1), 11–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karch CM, Goate AM, 2015. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol. Psychiatry 77 (1), 43–51. 10.1016/j.biopsych.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim DH, Yeo SH, Park JM, Choi JY, Lee TH, Park SY, Ock MS, Eo J, Kim HS, Cha HJ, 2014. Genetic markers for diagnosis and pathogenesis of Alzheimer’s disease. Gene 545 (2), 185–193. [DOI] [PubMed] [Google Scholar]
- Kim J, Eltorai AE, Jiang H, Liao F, Verghese PB, Kim J, Stewart FR, Basak JM, Holtzman DM, 2012. Anti-apoE immunotherapy inhibits amyloid accumulation in a transgenic mouse model of Abeta amyloidosis. J. Exp. Med 209 (12), 2149–2156. 10.1084/jem.20121274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JH, 2018. Genetics of Alzheimer’s disease. Dement. Neurocogn. Disord 17 (4), 131–136. Epub 2019/03/25. 10.12779/dnd.2018.17.4.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koffe RM, Hashimoto T, Tai HC, Kay KR, Serrano-Pozo A, Joyner D, Hou S, Kopeikina KJ, Frosch MP, Lee VM, Holtzman DM, Hyman BT, Spires-Jones TL, 2012. Apolipoprotein E4 effects in Alzheimer’s disease are mediated by synaptotoxic oligomeric amyloid-beta. Brain 135 (Pt 7), 2155–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, Naj AC, Boland A, Vronskaya M, van der Lee SJ, Amie-Wolf A, Bellenguez C, Frizatti A, Chouraki V, Martin ER, Sleegers K, Badarinarayan N, Jakobsdottir J, Hamilton-Nelson KL, Moreno-Grau S, Olaso R, Raybould R, Chen Y, Kuzma AB, Hiltunen M, Morgan T, Ahmad S, Vardarajan BN, Epelbaum J, Hoffmann P, Boada M, Beecham GW, Garnier JG, Harold D, Fitzpatrick AL, Valladares O, Moutet ML, Gerrish A, Smith AV, Qu L, Bacq D, Denning N, Jian X, Zhao Y, Del Zompo M, Fox NC, Choi SH, Mateo I, Hughes JT, Adams HH, Malamon J, Sanchez-Garcia F, Patel Y, Brody JA, Dombroski BA, MCD N, Daniilidou M, Eiriksdottir G, Mukherjee S, Wallon D, Uphill J, Aspelund T, Cantwell LB, Garzia F, Galimberti D, Hofer E, Butkiewicz M, Fin B, Scarpini E, Sarnowski C, Bush WS, Meslage S, Kornhuber J, White CC, Song Y, Barber RC, Engelborghs S, Sordon S, Voijnovic D, Adams PM, Vandenberghe R, Mayhaus M, Cupples LA, Albert MS, De Deyn PP, Gu W, Himali JJ, Beekly D, Squassina A, Hartmann AM, Orellana A, Blacker D, Rodriguez-Rodriguez E, Lovestone S, Garcia ME, Doody RS, Munoz-Fernadez C, Sussams R, Lin H, Fairchild TJ, Benito YA, Holmes C, Karamujic-Comic H, Frosch MP, Thonberg H, Maier W, Roschupkin G, Ghetti B, Giedraitis V, Kawalia A, Li S, Huebinger RM, Kilander L, Moebus S, Hernandez I, Kamboh MI, Brundin R, Turton J, Yang Q, Katz MJ, Concari L, Lord J, Beiser AS, Keene CD, Helisalmi S, Kloszewska I, Kukull WA, Koivisto AM, Lynch A, Tarraga L, Larson EB, Haapasalo A, Lawlor B, Mosley TH, Lipton RB, Solfrizzi V, Gill M, Longstreth WT Jr., Montine TJ, Frisardi V, Diez-Fairen M, Rivadeneira F, Petersen RC, Deramecourt V, Alvarez I, Salani F, Ciaramella A, Boerwinkle E, Reiman EM, Fievet N, Rotter JI, Reisch JS, Hanon O, Cupidi C, Andre Uitterlinden AG, Royall DR, Dufouil C, Maletta RG, de Rojas I, Sano M, Brice A, Cecchetti R, George-Hyslop PS, Ritchie K, Tsolaki M, Tsuang DW, Dubois B, Craig D, Wu CK, Soininen H, Avramidou D, Albin RL, Fratiglioni L, Germanou A, Apostolova LG, Keller L, Koutroumani M, Arnold SE, Panza F, Gkatzima O, Asthana S, Hannequin D, Whitehead P, Atwood CS, Caffarra P, Hampel H, Quintela I, Carracedo A, Lannfelt L, Rubinsztein DC, Barnes LL, Pasquier F, Frolich L, Barral S, McGuinness B, Beach TG, Johnston JA, Becker JT, Passmore P, Bigio EH, Schott JM, Bird TD, Warren JD, Boeve BF, Lupton MK, Bowen JD, Proitsi P, Boxer A, Powell JF, Burke JR, JSK K, Burns JM, Mancuso M, Buxbaum JD, Bonuccelli U, Cairns NJ, McQuillin A, Cao C, Livingston G, Carlson CS, Bass NJ, Carlsson CM, Hardy J, Carney RM, Bras J, Carrasquillo MM, Guerreiro R, Allen M, Chui HC, Fisher E, Masullo C, Crocco EA, DeCarli C, Bisceglio G, Dick M, Ma L, Duara R, Graff-Radford NR, Evans DA, Hodges A, Faber KM, Scherer M, Fallon KB, Riemenschneider M, Fardo DW, Heun R, Farlow MR, Kolsch H, Ferris S, Leber M, Foroud TM, Heuser I, Galasko DR, Giegling I, Gearing M, Hull M, Geschwind DH, Gilbert JR, Morris J, Green RC, Mayo K, Growdon JH, Feulner T, Hamilton RL, Harrell LE, Drichel D, Honig LS, Cushion TD, Huentelman MJ, Hollingworth P, Hulette CM, Hyman BT, Marshall R, Jarvik GP, Meggy A, Abner E, Menzies GE, llas B, Kramer JH, Vardy E, FM L, Jockel KH, Lah JJ, Dichgans M, Leverenz JB, Mann D, Levey AI, Pickering-Brown S, Lieberman AP, Klopp N, Lunetta KL, Wichmann HE, Lyketsos CG, Morgan K, Marson DC, Brown K, Martiniuk F, Medway C, Mash DC, Nothen MM, Masliah E, Hooper NM, McCormick WC, Daniele A, SM M, Bayer A, AN M, Gallacher J, AC M, van den Bussche H, Mesulam M, Brayne C, Miller BL, Riedel-Heller S, Miller CA, Miller JW, Al-Chalabi A, Morris JC, Shaw CE, Myers AJ, Wiltfang J, O’Bryant S, Olichney JM, Alvarez V, Parisi JE, Singleton AB, Paulson HL, Collinge J, Perry WR, Mead S, Peskind E, Cribbs DH, Rossor M, Pierce A, Ryan NS, Poon WW, Nacmias B, Potter H, Sorbi S, Quinn JF, Sacchinelli E, Raj A, Spalletta G, Raskind M, Caltagirone C, Bossu P, Orfei MD, Reisberg B, Clarke R, Reitz C, Smith AD, Ringman JM, Warden D, Roberson ED, Wilcock G, Rogaeva E, Bruni AC, Rosen HJ, Gallo M, Rosenberg RN, Ben-Shlomo Y, Sager MA, Mecocci P, Saykin AJ, Pastor P, Cuccaro ML, Vance JM, Schneider JA, Schneider LS, Slifer S, Seeley WW, Smith AG, Sonnen JA, Spina S, Stern RA, Swerdlow RH, Tang M, Tanzi RE, Trojanowski JQ, Troncoso JC, Van Deerlin VM, Van Eldik LJ, Vinters HV, Vonsattel JP, Weintraub S, Welsh-Bohmer KA, Wilhelmsen KC, Williamson J, Wingo TS, Woltjer RL, Wright CB, Yu CE, Yu L, Saba Y,Alzheimer Disease Genetics C, European Alzheimer’s Disease I, Cohorts for H, Aging Research in Genomic Epidemiology C, Genetic,Environmental Risk in Ad/Defining Genetic P, Environmental Risk for Alzheimer’sDisease C,Pilotto A, Bullido MJ, Peters O, Crane PK, Bennett D, Bosco P, Coto E, Boccardi V, De Jager PL, Lleo A, Warner N, Lopez OL, Ingelsson M, Deloukas P, Cruchaga C, Graff C, Gwilliam R, Fornage M, Goate AM, Sanchez-Juan P, Kehoe PG, Amin N, Ertekin-Taner N, Berr C, Debette S, Love S, Launer LJ, Younkin SG, Dartigues JF, Corcoran C, Ikram MA, Dickson DW, Nicolas G, Campion D, Tschanz J, Schmidt H, Hakonarson H, Clarimon J, Munger R, Schmidt R, Farrer LA, Van Broeckhoven C, COD M, AL D, Jones L, Haines JL, Deleuze JF, Owen MJ, Gudnason V, Mayeux R, Escott-Price V, Psaty BM, Ramirez A, Wang LS, Ruiz A, van Duijn CM, Holmans PA, Seshadri S, Williams J, Amouyel P, Schellenberg GD, Lambert JC, Pericak-Vance MA, 2019. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Abeta, tau, immunity and lipid processing. Nat.Genet 51 (3), 414–430. Epub 2019/03/02. 10.1038/s41588-019-0358-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lane CA, Hardy J, Schott JM, 2018. Alzheimer’s disease. Eur. J. Neurol 25 (1),59–70. 10.1111/ene.13439. [DOI] [PubMed] [Google Scholar]
- Laws SM, Clarnette RM, Taddei K, Martins G, Paton A, Hallmayer J, Almeida OP, Groth DM, Gandy SE, Forstl H, Martins RN, 2002. APOE-epsilon4 and APOE −491A polymorphisms in individuals with subjective memory loss. Mol. Psychiatry 7 (7), 768–775. Epub 2002/08/23. 10.1038/sj.mp.4001083. [DOI] [PubMed] [Google Scholar]
- Liao F, Hori Y, Hudry E, Bauer AQ, Jiang H, Mahan TE, Lefton KB, Zhang TJ, Dearborn JT, Kim J, Culver JP, Betensky R, Wozniak DF, Hyman BT, Holtzman DM, 2014. Anti-ApoE antibody given after plaque onset decreases Abeta accumulation and improves brain function in a mouse model of Abeta amyloidosis. J. Neurosci 34 (21), 7281–7292. Epub 2014/05/23. 10.1523/JNEUROSCI.0646-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liao F, Li A, Xiong M, Bien-Ly N, Jiang H, Zhang Y, Finn MB, Hoyle R, Keyser J, Lefton KB, Robinson GO, Serrano JR, Silverman AP, Guo JL, Getz J, Henne K, Leyns CE, Gallardo G, Ulrich JD, Sullivan PM, Lerner EP, Hudry E, Sweeney ZK, Dennis MS, Hyman BT, Watts RJ, Holtzman DM, 2018. Targeting of nonlipidated, aggregated apoE with antibodies inhibits amyloid accumulation. J. Clin. Invest 10.1172/JCI96429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim YY, Mormino EC, Alzheimer’s Disease Neuroimaging I, 2017. APOE genotype and early beta-amyloid accumulation in older adults without dementia. Neurology 89(10), 1028–1034. Epub 2017/08/11. 10.1212/WNL.0000000000004336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin YT, Seo J, Gao F, Feldman HM, Wen HL, Penney J, Cam HP, Gjoneska E, Raja WK, Cheng J, Rueda R, Kritskiy O, Abdurrob F, Peng Z, Milo B, Yu CJ, Elmsaouri S, Dey D, Ko T, Yankner BA, Tsai LH, 2018. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 98 (6), 1294. Epub 2018/06/29. 10.1016/j.neuron.2018.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Q, Wu WH, Fang CL, Li RW, Liu P, Lei P, Hu J, Sun X, Zheng YZ, Zhao YF, Li YM, 2011. Mapping ApoE/Abeta binding regions to guide inhibitor discovery. Mol. BioSyst 7 (5), 1693–1700. [DOI] [PubMed] [Google Scholar]
- Liu S, Breitbart A, Sun Y, Mehta PD, Boutajangout A, Scholtzova H, Wisniewski T, 2014. Blocking the apolipoprotein E/amyloid β-interaction in triple transgenic mice ameliorates Alzheimer’s disease related amyloid β and tau pathology. J.Neurochem 128 (577), 591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S, Park S, Allington G, Prelli F, Sun Y, Martá-Ariza M, Scholtzova H, Biswas G, Brown B, Verghese PB, Mehta PD, Kwon Y-U, Wisniewski T, 2017. Targeting apolipoprotein E/amyloid β binding by peptoid CPO_Aβ17–21P ameliorates Alzheimer’s disease related pathology and cognitive decline. Sci. Rep 7 (1), 8009. 10.1038/s41598-017-08604-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Long JM, Holtzman DM, 2019. Alzheimer disease: An update on pathobiology and treatment strategies. Cell 10.1016/j.cell.2019.09.001. Epub 2019/10/01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo J, Marechal JD, Warmlander S, Graslund A, Peralvarez-Marin A, 2010. In silico analysis of the apolipoprotein E and the amyloid beta peptide interaction: mis-folding induced by frustration of the salt bridge network. PLoS Comput. Biol 6(2). 10.1371/journal.pcbi.1000663.e1000663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma J, Brewer HB, Potter H, 1996. Alzheimer Aβ neurotoxicity: promotion by antic-hymotrypsin, ApoE4; inhibition by Aβ-related peptides. Neurobiol. Aging 17 (5), 773–780. 10.1016/0197-4580(96)00112-1. [DOI] [PubMed] [Google Scholar]
- Ma J, Yee A, Brewer HB Jr., Das S, Potter H, 1994. Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature 372 (6501), 92–94. [DOI] [PubMed] [Google Scholar]
- Mahley RW, Huang Y, 2012. Small-molecule structure correctors target abnormal protein structure and function: structure corrector rescue of apolipoprotein E4-associated neuropathology. J. Med. Chem 55 (21), 8997–9008. Epub 2012/09/28. 10.1021/jm3008618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mak AC, Pullinger CR, Tang LF, Wong JS, Deo RC, Schwarz JM, Gugliucci A, Movsesyan I, Ishida BY, Chu C, Poon A, Kim P, Stock EO, Schaefer EJ, Asztalos BF, Castellano JM, Wyss-Coray T, Duncan JL, Miller BL, Kane JP, Kwok PY, Malloy MJ, 2014. Effects of the absence of apolipoprotein e on lipo-proteins, neurocognitive function, and retinal function. JAMA Neurol 71 (10), 1228–1236. Epub 2014/08/12. 10.1001/jamaneurol.2014.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martins RN, Clarnette R, Fisher C, Broe GA, Brooks WS, Montgomery P, Gandy SE, 1995. ApoE genotypes in Australia: roles in early and late onset Alzheimer’s disease and Down’s syndrome. Neuroreport 6 (11), 1513–1516. [PubMed] [Google Scholar]
- Martins RN, Villemagne V, Sohrabi HR, Chatterjee P, Shah TM, Verdile G,Fraser P, Taddei K, Gupta VB, Rainey-Smith SR, Hone E, Pedrini S, Lim WL, Martins I, Frost S, Gupta S, O’Bryant S, Rembach A, Ames D, Ellis K, Fuller SJ, Brown B, Gardener SL, Fernando B, Bharadwaj P, Burnham S, Laws SM, Barron AM, Goozee K, Wahjoepramono EJ, Asih PR, Doecke JD, Salvado O, Bush AI, Rowe CC, Gandy SE, Masters CL, 2018. Alzheimer’s disease: a journey from amyloid peptides and oxidative stress, to biomarker technologies and disease prevention strategies-gains from AIBL and DIAN cohort studies. J. Alzheimers Dis 62 (3), 965–992. Epub 2018/03/23. 10.3233/JAD-171145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsubara E, Frangione B, Ghiso J, 1995. Characterization of apolipoprotein JAlzheimer’s A beta interaction. J. Biol. Chem 270 (13), 7563–7567. [DOI] [PubMed] [Google Scholar]
- Medway CW, Abdul-Hay S, Mims T, Ma L, Bisceglio G, Zou F, Pankratz S, Sando SB, Aasly JO, Barcikowska M, Siuda J, Wszolek ZK, Ross OA, Carrasquillo M, Dickson DW, Graff-Radford N, Petersen RC, Ertekin-Taner N, Morgan K, Bu G, Younkin SG. ApoE variant p.V236E is associated with markedly reduced risk of Alzheimer’s disease. Mol. Neurodegener 2014;9:11. Epub 2014/03/13. doi: 10.1186/1750-1326-9-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miao J, Vitek MP, Xu F, Previti ML, Davis J, Van Nostrand WE, 2005. Reducing cerebral microvascular amyloid-beta protein deposition diminishes regional neuroinflammation in vasculotropic mutant amyloid precursor protein transgenic mice. J. Neurosci 25 (27), 6271–6277. 10.1523/JNEUROSCI.1306-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morelli L, Wei L, Amorim A, McDermid J, Abee CR, Frangione B, Walker LC, Levy E, 1996. Cerebrovascular amyloidosis in squirrel monkeys and rhesus monkeys: apolipoprotein E genotype. FEBS Lett 379 (2), 132–134. Epub 1996/01/29. 10.1016/0014-5793(95)01491-8. [DOI] [PubMed] [Google Scholar]
- Morris JC, Roe CM, Xiong C, Fagan AM, Goate AM, Holtzman DM, Mintun MA, 2010. APOE predicts amyloid-beta but not tau Alzheimer pathology in cognitively normal aging. Ann. Neurol 67 (1), 122–131. 10.1002/ana.21843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pankiewicz JE, Guridi M, Kim J, Asuni AA, Sanchez S, Sullivan PM, Holtzman DM, Sadowski MJ, 2014. Blocking the apoE/Abeta interaction ameliorates Abeta-related pathology in APOE epsilon2 and epsilon4 targeted replacement Alzheimer model mice. Acta Neuropathol. Commun 2, 75. 10.1186/s40478-014-0075-0.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pimenova AA, Raj T, Goate AM, 2017. Untangling genetic risk for Alzheimer’s disease. Biol. Psychiatry 10.1016/j.biopsych.2017.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pletnikova O, Kageyama Y, Rudow G, LaClair KD, Albert M, Crain BJ, Tian J, Fowler D, Troncoso JC, 2018. The spectrum of preclinical Alzheimer’s disease pathology and its modulation by ApoE genotype. Neurobiol. Aging 71, 72–80. (Epub 2018/08/14). 10.1016/j.neurobiolaging.2018.07.007 [DOI] [PubMed] [Google Scholar]
- Potter H, Wefes IM, Nilsson LN, 2001. The inflammation-induced pathological chaperones ACT and apo-E are necessary catalysts of Alzheimer amyloid formation. Neurobiol. Aging 22 (6), 923–930. [DOI] [PubMed] [Google Scholar]
- Potter H, Wisniewski T, 2012. Apolipoprotein E: essential catalyst of the Alzheimer amyloid cascade. Int. J. Alzheimers Dis 489428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rabinovici GD, 2019. Late-onset Alzheimer disease. Continuum (Minneap Minn) 25(1), 14–33. Epub 2019/02/02. 10.1212/CON.0000000000000700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Resnick SM, Bilgel M, Moghekar A, An Y, Cai Q, Wang MC, Thambisetty M, Prince JL, Zhou Y, Soldan A, Wong DF, O’Brien RJ, Ferrucci L, Albert MS, 2015. Changes in Abeta biomarkers and associations with APOE genotype in 2 longitudinal cohorts. Neurobiol. Aging 36 (8), 2333–2339. Epub 2015/05/26. 10.1016/j.neurobiolaging.2015.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roda AR, Montoliu-Gaya L, Villegas S, 2019. The role of apolipoprotein E isoforms in Alzheimer’s disease. J. Alzheimers Dis 68 (2), 459–471. Epub 2019/02/19. 10.3233/JAD-180740. [DOI] [PubMed] [Google Scholar]
- Rosenberg JB, Kaplitt MG, De BP, Chen A, Flagiello T, Salami C, Pey E, Zhao L, Ricart Arbona RJ, Monette S, Dyke JP, Ballon DJ, Kaminsky SM, Sondhi D, Petsko GA, Paul SM, Crystal RG, 2018. AAVrh.10-mediated APOE2 central nervous system gene therapy for APOE4-associated Alzheimer’s disease. Hum. Gene. Ther. Clin. Dev 29 (1), 24–47. Epub 2018/02/08. 10.1089/humc.2017.231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Royston MC, Mann D, Pickering-Brown S, Owen F, Perry R, Ragbavan R, Khin-Nu C, Tyner S, Day K, Crook R, Hardy J, Roberts GW, 1996. ApoE2 allele, Down’s syndrome, and dementia. Ann. N. Y. Acad. Sci 777, 255–259. Epub 1996/01/17. 10.1111/j.1749-6632.1996.tb34428.x. [DOI] [PubMed] [Google Scholar]
- Sadowski M, Pankiewicz J, Scholtzova H, Mehta P, Prelli F, Quartermain D,Wisniewski T, 2006. Blocking the apolipoproteinE/Amyloid β interaction reduces the parenchymal and vascular amyloid-β deposition and prevents memory deficit in AD transgenic mice. PNAS 103 (49), 18787–18792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sadowski M, Pankiewicz J, Scholtzova H, Ripellino JA, Li Y, Schmidt SD, Mathews PM, Fryer JD, Holtzman DM, Sigurdsson EM, Wisniewski T, 2004. A synthetic peptide blocking the apolipoprotein E/beta-amyloid binding mitigates beta-amyloid toxicity and fibril formation in vitro and reduces beta-amyloid plaques in transgenic mice. Am. J. Pathol 165 (3), 937–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Safieh M, Korczyn AD, Michaelson DM, 2019. ApoE4: an emerging therapeutic target for Alzheimer’s disease. BMC Med 17 (1), 64. Epub 2019/03/21. 10.1186/s12916-019-1299-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanan DA, Weisgraber KH, Russell SJ, Mahley RW, Huang D, Saunders A,Schmechel D, Wisniewski T, Frangione B, Roses AD, Strittmatter WJ, 1994. Apolipoprotein E associates with beta amyloid peptide of Alzheimer’s disease to form novel monofibrils. Isoform apoE4 associates more efficiently than apoE3. J. Clin. Invest 94 (2), 860–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saunders AM, Hulette C, Welsh-Bohmer KA, Schmechel DE, Crain B, Burke JR, Alberts MJ, Strittmatter WJ, Breitner JCS, Rosenberg C, Scott SV, Gaskell PC Jr., Pericak-Vance MA, Roses AD, 1996. Specificity, sensitivity, and predictive value of apolipoprotein- E genotyping for sporadic Alzheimer’s disease. Lancet 348 (9020), 90–93. [DOI] [PubMed] [Google Scholar]
- Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, Van der Flier WM, 2016. Alzheimer’s disease. Lancet 388 (10043), 505–517. 10.1016/S0140-6736(15)01124-1. [DOI] [PubMed] [Google Scholar]
- Schmechel DE, Saunders AM, Strittmatter WJ, Crain BJ, Hulette CM, Joo SH, Pericak-Vance MA, Goldgaber D, Roses AD, 1993. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc. Natl. Acad. Sci. U. S. A 90 (20), 9649–9653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider LS, Mangialasche F, Andreasen N, Feldman H, Giacobini E, Jones R, Mantua V, Mecocci P, Pani L, Winblad B, Kivipelto M, 2014. Clinical trials and late-stage drug development for Alzheimer’s disease: an appraisal from 1984 to 2014. J. Intern. Med 275 (3), 251–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selkoe DJ, Hardy J, 2016. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med 8 (6), 595–608. 10.15252/emmm.201606210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sengupta U, Nilson AN, Kayed R, 2016. The role of amyloid-beta oligomers in toxi-city, propagation, and immunotherapy. EBioMedicine 6, 42–49. 10.1016/j.ebiom.2016.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Holtzman DM, 2018. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat. Rev. Immunol 10.1038/s41577-018-0051-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sorbi S, Nacmias B, Forleo P, Piacentini S, Latorraca S, Amaducci L, 1995. Epistatic effect of APP717 mutation and apolipoprotein E genotype in familial Alzheimer’s disease. Ann. Neurol 38 (1), 124–127. [DOI] [PubMed] [Google Scholar]
- Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J,Salvesen GS, Roses AD, 1993a. 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 90 (5), 1977–1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strittmatter WJ, Weisgraber KH, Huang DY, Dong LM, Salvesen GS, Pericak-Vance M, Schmechel D, Saunders AM, Goldgaber D, Roses AD, 1993b. Binding of human apolipoprotein E to synthetic amyloid beta peptide: isoform-specific effects and implications for late-onset Alzheimer disease. Proc. Natl. Acad. Sci.U. S. A 90 (17), 8098–8102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thal DR, Rub U, Orantes M, Braak H, 2002. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology 58 (12), 1791–1800. [DOI] [PubMed] [Google Scholar]
- Tzioras M, Davies C, Newman A, Jackson R, Spires-Jones T, 2019. Invited Review: APOE at the interface of inflammation, neurodegeneration and pathological protein spread in Alzheimer’s disease. Neuropathol. Appl. Neurobiol 45 (4), 327–346. Epub 2018/11/06. 10.1111/nan.12529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ulrich JD, Ulland TK, Colonna M, Holtzman DM, 2017. Elucidating the role of TREM2 in Alzheimer’s disease. Neuron 94 (2), 237–248. 10.1016/j.neuron.2017.02.042. [DOI] [PubMed] [Google Scholar]
- Velez JI, Lopera F, Sepulveda-Falla D, Patel HR, Johar AS, Chuah A, Tobon C, Rivera D, Villegas A, Cai Y, Peng K, Arkell R, Castellanos FX, Andrews SJ, Silva Lara MF, Creagh PK, Easteal S, de Leon J, Wong ML, Licinio J, Mastronardi CA, Arcos-Burgos M, 2016. APOE*E2 allele delays age of onset in PSEN1 E280A Alzheimer’s disease. Mol. Psychiatry 21 (7), 916–924. Epub 2015/12/02. 10.1038/mp.2015.177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, Bu G, Frieden C, Holtzman DM, 2013. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc. Natl. Acad. Sci. U.S. A 110 (19) E1807–E16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viola KL, Klein WL, 2015. Amyloid beta oligomers in Alzheimer’s disease pathogenesis, treatment, and diagnosis. Acta Neuropathol 129 (2), 183–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walker LC, Jucker M, 2017. The exceptional vulnerability of humans to Alzheimer’s disease. Trends Mol. Med 23 (6), 534–545. Epub 2017/05/10. 10.1016/j.molmed.2017.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Najm R, Xu Q, Jeong DE, Walker D, Balestra ME, Yoon SY, Yuan H, Li G, Miller ZA, Miller BL, Malloy MJ, Huang Y, 2018. Gain of toxic apolipoprotein E4 effects in human iPSC-derived neurons is ameliorated by a small-molecule structure corrector. Nat. Med 24 (5), 647–657. Epub 2018/04/11. 10.1038/s41591-018-0004-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wijsman EM, Daw EW, Yu X, Steinbart EJ, Nochlin D, Bird TD, Schellenberg GD, 2005. APOE and other loci affect age-at-onset in Alzheimer’s disease families with PS2 mutation. Am. J. Med. Genet. B Neuropsychiatr. Genet 132B (1), 14–20. Epub 2004/09/25. 10.1002/ajmg.b.30087. [DOI] [PubMed] [Google Scholar]
- Wingo TS, Lah JJ, Levey AI, Cutler DJ, 2012. Autosomal recessive causes likely in early-onset Alzheimer disease. Arch. Neurol 69 (1), 59–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wisniewski T, Castano EM, Golabek A, Vogel T, Frangione B, 1994b. Acceleration of Alzheimer’s fibril formation by apolipoprotein E in vitro. Am. J. Pathol 145 (5), 1030–1035. [PMC free article] [PubMed] [Google Scholar]
- Wisniewski T, Drummond E, 2019. Future horizons in Alzheimer’s disease research. Prog. Mol. Biol. Transl. Sci 168, 223–241. Epub 2019/11/09. 10.1016/bs.pmbts.2019.08.001. [DOI] [PubMed] [Google Scholar]
- Wisniewski T, Frangione B, 1992. Apolipoprotein E: a pathological chaperone protein in patients with cerebral and systemic amyloid. Neurosci. Lett 135 (2), 235–238. [DOI] [PubMed] [Google Scholar]
- Wisniewski T, Frangione B, 1996. Apolipoprotein E, amyloidosis and Alzheimer’s disease. Dementia 10, 171–183. [Google Scholar]
- Wisniewski T, Ghiso J, Frangione B, 1994a. Alzheimer’s disease and soluble Aβ. Neurobiol. Aging 15 (2), 143–152. [DOI] [PubMed] [Google Scholar]
- Wisniewski T, Golabek A, Matsubara E, Ghiso J, Frangione B, 1993. Apolipoprotein E: binding to soluble Alzheimer’s beta-amyloid. Biochem. Biophys. Res. Commun 192 (2), 359–365. Epub 1993/04/30. 10.1006/bbrc.1993.1423. [DOI] [PubMed] [Google Scholar]
- Wisniewski T, Lalowski M, Golabek AA, Vogel T, Frangione B, 1995a. Is Alzheimer’s disease an apolipoprotein E amyloidosis? Lancet 345 (8955), 956–958. [DOI] [PubMed] [Google Scholar]
- Wisniewski T, Morelli L, Wegiel J, Levy E, Wisniewski HM, Frangione B, 1995b. The influence of apolipoprotein E isotypes on Alzheimer’s disease pathology in 40 cases of Down’s syndrome. Ann. Neurol 37 (1), 136–138. 10.1002/ana.410370127. [DOI] [PubMed] [Google Scholar]
- Yang J, Ji Y, Mehta P, Bates KA, Sun Y, Wisniewski T, 2011. Blocking the apolipoprotein E/amyloid β interaction reduces fibrillar vascular amyloid deposition and cerebral microhemorrhages in TgSwDI mice. J. Alzheimers Dis 24 (2), 269–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zalocusky KA, Nelson MR, Huang Y, 2019. An Alzheimer’s-disease-protective APOE mutation. Nat. Med 10.1038/s41591-019-0634-9. in press. [DOI] [PubMed] [Google Scholar]
- Zhao L, Gottesdiener AJ, Parmar M, Li M, Kaminsky SM, Chiuchiolo MJ, Sondhi D, Sullivan PM, Holtzman DM, Crystal RG, Paul SM, 2016. Intracerebral adeno-associated virus gene delivery of apolipoprotein E2 markedly reduces brain amyloid pathology in Alzheimer’s disease mouse models. Neurobiol. Aging 44, 159–172. Epub 2016/06/19. 10.1016/j.neurobiolaging.2016.04.020. [DOI] [PubMed] [Google Scholar]
- Zhao N, Liu CC, Qiao W, Bu G, 2018. Apolipoprotein E, receptors, and modulation of Alzheimer’s disease. Biol. Psychiatry 83 (4), 347–357. 10.1016/j.biopsych.2017.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]