Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2022

Open Access 01.12.2022 | Perspective

APOE targeting strategy in Alzheimer’s disease: lessons learned from protective variants

verfasst von: Guojun Bu

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2022

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Amyloid-β
AD
Alzheimer’s disease
ApoE
Apolipoprotein E
Ch
Christchurch
HSPG
Heparan sulfate proteoglycan
Jac
Jacksonville (Jac)
LDL
Low-density lipoprotein
The APOE gene is the strongest genetic risk factor for Alzheimer’s disease (AD) with the ε4 allele increasing the risk by 2-4 folds compared to the most common ε3 allele [1]. The high prevalence of APOE4 allele frequency (~ 14% in Caucasian non-demented versus ~ 38% in AD) and carrier frequency (~ 20-30% in Caucasian non-demented versus 50-70% in AD) in AD patients presents a strong rationale to target APOE for AD prevention and therapy [1]. More interestingly, carriers of the APOE2 allele have a significantly reduced risk for AD [2], further attracting interests to target this strong AD risk gene.
The apolipoprotein E (apoE) protein encoded by the APOE gene is a major lipid carrier in both periphery and the brain transporting and distributing lipids across cells and tissues with recognized roles in homeostasis and injury repair [1]. In AD, strong clinical and preclinical evidence suggests that the main pathway by which APOE4 drives AD risk is to promote earlier and more abundant amyloid-β (Aβ) deposition, likely by inhibiting Aβ clearance and accelerating Aβ aggregation and amyloid seeding [1]. Although Aβ plaque is a defining pathological feature of AD, it is not sufficient to cause dementia. Efforts in targeting Aβ has generated mostly disappointing clinical outcomes which is not surprising considering that Aβ deposition occurs up to two decades before the clinical onset and that the amounts of Aβ plaque typically do not correlate with dementia. As such, targeting Aβ might need to be explored as a prevention rather than a treatment strategy. Because Aβ deposition triggers or accelerates additional pathogenic events including microglia-mediated immune response and tau tangle spread, effective strategy targeting Aβ will likely be primary prevention prior to plaque development.
ApoE isoforms also have differential effects on multiple other AD-related pathogenic pathways highlighted by endocytic trafficking, immune response, cerebrovascular integrity, tau-mediated neurodegeneration, and energy metabolism [1, 3], some might be Aβ-independent. Indeed, APOE genotype also has differential effects on age-related cognitive decline and Lewy body dementia parallel to their risk profile for AD. How apoE isoforms that differ at only two amino acids (apoE2: Cys112 and Cys158; apoE3: Cys112 and Arg158; apoE4: Arg112 and Arg158) (Fig. 1) have such profound effects on risk for AD and other age-related dementias has puzzled the field for decades. Recently, an ApoE cascade hypothesis was proposed where it emphasizes the structural and related biochemical differences including lipidation, protein levels, receptor binding, and oligomerization, among the three isoforms as drivers of downstream effects at the cellular and phenotypical levels [4]. As such, targeting the biochemical features of apoE will likely yield greater and broader effects on multiple AD-related pathways.
Although targeting strategies developed against apoE4 or learned from apoE4 are emerging including down-regulation of apoE4 levels [5], the apoE field still struggles with nominating alternative, perhaps more effective ways of targeting apoE in general. Towards this, lessons learned from studying apoE2 has provided significant insights [2]. Despite limited attention to apoE2 compared to apoE4 in preclinical studies, emerging evidence suggests that a major pathway by which apoE2 protects against AD and promotes healthy brain aging is by enhancing lipid efflux and related lipid metabolism. ApoE2 is associated with greater lipidation in CSF and higher apoE protein levels in both periphery and the brain. Interestingly, the higher apoE levels and lower cholesterol in brain parenchyma are associated with better memory performance [6] and longevity [7] in human apoE-targeted replacement mice. ApoE2 is also associated with unique lipidomic and metabolomic profiles in plasma [8, 9]. This knowledge of apoE2 highlights the potential beneficial effects of enhancing lipid efflux and promoting lipid metabolism as a lead strategy to treat AD and related dementia.
In addition to the common variants, recent studies have identified several APOE rare protective variants that offer new insights on apoE properties linked to their protective effects (Fig. 1). Top of the list is the APOE3-Christchurch (R136S) variant where a homozygous carrier with a PSEN1 mutation had a delayed onset of mild cognitive impairment by three decades [10]. Brain imaging studies detected high amyloid load but limited tau pathology and neurodegeneration, suggesting a protective mechanism that is resilient against amyloid. One known effect of the R136S mutation is the reduced binding to the low-density lipoprotein (LDL) receptor and heparan sulfate proteoglycan (HSPG). Interestingly, several pathogenic molecules in AD including Aβ, tau, α-synuclein, and apoE bind to cell surface HSPG which has been implicated in proteinopathy in neurodegenerative diseases by modulating protein trafficking, aggregation, and propagation. How does reduced apoE binding to HSPG lightens the risk to AD is still unclear but given the detrimental effects of HSPG in pathogenic events, it is possible that targeting apoE-HSPG interaction can be beneficial to reducing dementia-related outcomes. One caution for lessons learned with the APOE3-Christchurch variant is that all information to date on APOE3-Christchurch is from study of one individual with incomplete information on neuropathological, biomarker, and other outcomes. Preclinical studies using animal models and cellular models are needed to address specific effects of this variant on neuropathological features (i.e., amyloid plaques and tau tangles), immune response, lipid metabolism, vascular integrity and function, and other AD-related pathways. It is interesting to note that both APOE3-Christchurch and APOE2 homozygosity are associated with Type III hyperlipoproteinemia. Additionally, apoE2 is deficient in binding to the LDL receptor and has the lowest affinity to heparin compared to apoE3 and apoE4. As such, it is templating to speculate that the protective mechanisms of APOE3-Christchurch and APOE2 are likely related in pathways relevant to receptor binding and lipid metabolism.
Another APOE protective variant is APOE3-Jacksonville (V236E) with the mutation localizing in the apoE C-terminal domain, which is best known for apoE lipid-binding function. The protective effect was first reported in 2014 [11] but only recently the underlying mechanism was revealed as reducing apoE self-aggregation [12]. Specifically, the apoE3-Jacksonville mutation reduces the tendency to aggregate both in mammalian cells and when produced in bacteria. More importantly, reduced apoE aggregation is associated with greater ability to promote cholesterol efflux, a process thought to require the apoE monomeric form. ApoE3-Jacksonville is also associated with enhanced lipid-association and when expressed in an amyloid mouse model, reduces amyloid load and related toxicity [12]. These mechanistic insights suggest that reducing apoE self-aggregation can be an alternative apoE targeting strategy to enhance apoE protective effect against AD. Reduced apoE aggregation likely alleviates the detrimental effect of apoE in amyloid seeding and promotes endocytic trafficking of apoE-interacting receptors such as apoE receptor 2 (ApoER2/LRP8), glutamate receptors, insulin receptor, and lipid-efflux transporter ABCA1. More importantly, as intracellular lipid accumulation has been increasingly recognized as a major pathogenic event in aging and AD [13], the potentially enhanced function of apoE in lipid efflux will likely promote repair and healthy brain aging.
More recently, Le Guen et al reported genetic identification of two rare APOE protective variants [14]. In addition to confirming the protective effect of APOE3-Jacksonville, they discovered a new protective variant APOE4-R251G which co-segregates with the APOE4 allele. By analyzing multiple cohorts and combining data for meta-analysis, the genetic data are in general convincing despite the typical difficulties in genetic association studies on rare variants. As with APOE3-Jacksonville, the APOE4-R251G is also localized in the C-terminal domain of apoE (Fig. 1). Specifically, the R251G mutation is located within the lipid-binding region, perhaps just outside the area where apoE initially binds lipid. How this specific mutation is protective against AD risk is not clear, but two possibilities worth exploring. First, the change from a hydrophilic residue to a non-polar residue could enhance lipid binding thus promoting apoE function in lipid metabolism as it relates to brain homeostasis and injury repair. Second, the R251G mutation could potentially change the apoE4 structure such that it either reduces its harmful effects or enhances its physiological functions [1]. Towards this, it is interesting to note that the R251 residue was proposed to form a salt bridge with the Q98 residue as part of apoE domain-domain interaction in a structural study of a mutated apoE3 [15]. Collectively, the APOE3-Jacksonville and APOE4-R251G protective variants offer new clues as to how apoE C-terminal domain impacts AD and related dementias and how knowledge learned from them can guide new apoE-targeting strategies.
Although APOE common variants, APOE2 and APOE4, continue to teach us how apoE-related outcomes contribute to AD pathogenesis, functional rare variants such as APOE3-Christchurch, APOE3-Jacksonville, and APOE4-R251G can provide additional insights. The fact that these rare variants carrying mutations in different regions of apoE offers additional opportunities to explore how structural and related biochemical properties of apoE impact its pathophysiology in aging and AD. Along with the common APOE2 variant, these APOE rare protective variants can teach us how to target apoE as we seek effective ways to prevent and cure AD and other age-related dementias.

Acknowledgements

The author thanks Danielle Feathers, Hongmei Li, Lucy Job, Chia-Chen Liu, and Na Zhao for critical reading of the manuscript.

Declarations

Not applicable.
All authors read and approved the final manuscript.

Competing interests

G.B. consults for SciNeuro, has consulted for AbbVie, E-Scape, Eisai, Vida Ventures, Lexeo, and is on the scientific advisory board for Kisbee.
Open AccessThis 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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15(9):501–18.CrossRef Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15(9):501–18.CrossRef
2.
Zurück zum Zitat Li Z, Shue F, Zhao N, Shinohara M, Bu G. APOE2: protective mechanism and therapeutic implications for Alzheimer's disease. Mol Neurodegener. 2020;15(1):63.CrossRef Li Z, Shue F, Zhao N, Shinohara M, Bu G. APOE2: protective mechanism and therapeutic implications for Alzheimer's disease. Mol Neurodegener. 2020;15(1):63.CrossRef
3.
Zurück zum Zitat Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol Neurodegener. 2020;15(1):40.CrossRef Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol Neurodegener. 2020;15(1):40.CrossRef
4.
Zurück zum Zitat Martens YA, Zhao N, Liu CC, Kanekiyo T, Yang AJ, Goate AM, et al. ApoE Cascade hypothesis in the pathogenesis of Alzheimer's disease and related dementias. Neuron. 2022;110(8):1304–17.CrossRef Martens YA, Zhao N, Liu CC, Kanekiyo T, Yang AJ, Goate AM, et al. ApoE Cascade hypothesis in the pathogenesis of Alzheimer's disease and related dementias. Neuron. 2022;110(8):1304–17.CrossRef
5.
Zurück zum Zitat Li Y, Macyczko JR, Liu CC, Bu G. ApoE4 reduction: an emerging and promising therapeutic strategy for Alzheimer's disease. Neurobiol Aging. 2022;115:20–8.CrossRef Li Y, Macyczko JR, Liu CC, Bu G. ApoE4 reduction: an emerging and promising therapeutic strategy for Alzheimer's disease. Neurobiol Aging. 2022;115:20–8.CrossRef
6.
Zurück zum Zitat Shinohara M, Kanekiyo T, Yang L, Linthicum D, Fu Y, Price L, Frisch-Daiello JL, Han X, Fryer JD, Bu G. APOE2 eases cognitive decline during Aging: Clinical and preclinical evaluations. Ann Neurol. 2016;79(5):758-74. https://doi.org/10.1002/ana.24628. Epub 2016 Mar 29. Shinohara M, Kanekiyo T, Yang L, Linthicum D, Fu Y, Price L, Frisch-Daiello JL, Han X, Fryer JD, Bu G. APOE2 eases cognitive decline during Aging: Clinical and preclinical evaluations. Ann Neurol. 2016;79(5):758-74. https://​doi.​org/​10.​1002/​ana.​24628. Epub 2016 Mar 29.
7.
8.
Zurück zum Zitat Wang T, Huynh K, Giles C, Mellett NA, Duong T, Nguyen A, Lim WLF, Smith AA, Olshansky G, Cadby G, et al. APOE ε2 resilience for Alzheimer's disease is mediated by plasma lipid species: Analysis of three independent cohort studies. Alzheimers Dement. 2022. https://doi.org/10.1002/alz.12538. Online ahead of print Wang T, Huynh K, Giles C, Mellett NA, Duong T, Nguyen A, Lim WLF, Smith AA, Olshansky G, Cadby G, et al. APOE ε2 resilience for Alzheimer's disease is mediated by plasma lipid species: Analysis of three independent cohort studies. Alzheimers Dement. 2022. https://​doi.​org/​10.​1002/​alz.​12538. Online ahead of print
9.
Zurück zum Zitat Zhao N, Ren Y, Yamazaki Y, Qiao W, Li F, Felton LM, et al. Alzheimer's risk factors age, APOE genotype, and sex drive distinct molecular pathways. Neuron. 2020;106(5):727–742.e726.CrossRef Zhao N, Ren Y, Yamazaki Y, Qiao W, Li F, Felton LM, et al. Alzheimer's risk factors age, APOE genotype, and sex drive distinct molecular pathways. Neuron. 2020;106(5):727–742.e726.CrossRef
10.
Zurück zum Zitat Arboleda-Velasquez JF, Lopera F, O'Hare M, Delgado-Tirado S, Marino C, Chmielewska N, et al. Resistance to autosomal dominant Alzheimer's disease in an APOE3 Christchurch homozygote: a case report. Nat Med. 2019;25(11):1680–3.CrossRef Arboleda-Velasquez JF, Lopera F, O'Hare M, Delgado-Tirado S, Marino C, Chmielewska N, et al. Resistance to autosomal dominant Alzheimer's disease in an APOE3 Christchurch homozygote: a case report. Nat Med. 2019;25(11):1680–3.CrossRef
11.
Zurück zum Zitat Medway CW, Abdul-Hay S, Mims T, Ma L, Bisceglio G, Zou F, Pankratz S, Sando SB, Aasly JO, Barcikowska M, et al. ApoE variant p.V236E is associated with markedly reduced risk of Alzheimer's disease. Mol Neurodegener. 2014;9:11. https://doi.org/10.1186/1750-1326-9-11. Medway CW, Abdul-Hay S, Mims T, Ma L, Bisceglio G, Zou F, Pankratz S, Sando SB, Aasly JO, Barcikowska M, et al. ApoE variant p.V236E is associated with markedly reduced risk of Alzheimer's disease. Mol Neurodegener. 2014;9:11. https://​doi.​org/​10.​1186/​1750-1326-9-11.
12.
Zurück zum Zitat Liu CC, Murray ME, Li X, Zhao N, Wang N, Heckman MG, et al. APOE3-Jacksonville (V236E) variant reduces self-aggregation and risk of dementia. Sci Transl Med. 2021;13(613):eabc9375.CrossRef Liu CC, Murray ME, Li X, Zhao N, Wang N, Heckman MG, et al. APOE3-Jacksonville (V236E) variant reduces self-aggregation and risk of dementia. Sci Transl Med. 2021;13(613):eabc9375.CrossRef
13.
Zurück zum Zitat Raulin AC, Martens YA, Bu G. Lipoproteins in the central nervous system: from biology to pathobiology. Annu Rev Biochem. 2022;91:731–59.CrossRef Raulin AC, Martens YA, Bu G. Lipoproteins in the central nervous system: from biology to pathobiology. Annu Rev Biochem. 2022;91:731–59.CrossRef
14.
Zurück zum Zitat Le Guen Y, Belloy ME, Grenier-Boley B, de Rojas I, Castillo-Morales A, Jansen I, Nicolas A, Bellenguez C, Dalmasso C, Küçükali F, et al. Association of Rare APOE Missense Variants V236E and R251G With Risk of Alzheimer Disease. JAMA Neurol. 2022;79(7):652-63. https://doi.org/10.1001/jamaneurol.2022.1166. Le Guen Y, Belloy ME, Grenier-Boley B, de Rojas I, Castillo-Morales A, Jansen I, Nicolas A, Bellenguez C, Dalmasso C, Küçükali F, et al. Association of Rare APOE Missense Variants V236E and R251G With Risk of Alzheimer Disease. JAMA Neurol. 2022;79(7):652-63. https://​doi.​org/​10.​1001/​jamaneurol.​2022.​1166.
15.
Zurück zum Zitat Chen J, Li Q, Wang J. Topology of human apolipoprotein E3 uniquely regulates its diverse biological functions. Proc Natl Acad Sci U S A. 2011;108(36):14813–8.CrossRef Chen J, Li Q, Wang J. Topology of human apolipoprotein E3 uniquely regulates its diverse biological functions. Proc Natl Acad Sci U S A. 2011;108(36):14813–8.CrossRef
Metadaten
Titel
APOE targeting strategy in Alzheimer’s disease: lessons learned from protective variants
verfasst von
Guojun Bu
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2022
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-022-00556-6

Weitere Artikel der Ausgabe 1/2022

Molecular Neurodegeneration 1/2022 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Schwindelursache: Massagepistole lässt Otholiten tanzen

14.05.2024 Benigner Lagerungsschwindel Nachrichten

Wenn jüngere Menschen über ständig rezidivierenden Lagerungsschwindel klagen, könnte eine Massagepistole der Auslöser sein. In JAMA Otolaryngology warnt ein Team vor der Anwendung hochpotenter Geräte im Bereich des Nackens.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.