Abstract
Alzheimer’s disease (AD) stands as a significant challenge in neurodegenerative research, with the apolipoprotein E (APOE) gene, particularly its 4 allele, emerging as a major genetic risk factor. This paper provides a comprehensive analysis of the role of APOE 4 in AD pathogenesis, focusing on its impact on amyloid-beta metabolism, neuroinflammation, synaptic dysfunction, neurovascular function, and neuronal survival. APOE 4 influences A aggregation, impairs clearance mechanisms, exacerbates neuroinflammatory responses, disrupts synaptic plasticity, compromises neurovascular function, and promotes neuronal damage within the brain. Studies using APOE-knockout (APOE-KO) mice were used to obtain results. Concerns and limitations regarding the translation of preclinical findings, the heterogeneity of APOE 4-associated phenotypes, and the complex interplay with other genetic and environmental factors are also discussed. Addressing these challenges is essential for developing targeted therapeutic interventions to mitigate the detrimental effects of APOE 4 and halt disease progression in AD. By elucidating the molecular mechanisms underlying APOE 4-mediated pathogenesis and identifying reliable biomarkers for risk prediction and disease monitoring, personalized therapeutic strategies can be developed to improve outcomes for individuals affected by AD.
Keywords: Alzheimer’s Disease (AD), APOE (2, 3, 4), A aggregation, peripheral APOE, brain APOE, apolipoproteins, tau pathology, neuroinflammation, cerebrovascular function, synaptic function, memory.
Introduction
Alzheimer’s disease (AD) stands as a profound challenge in the realm of neurodegenerative disorders and is characterized by progressive cognitive decline and memory impairment. As the leading cause of dementia, AD affected 6.5 million Americans 65 and older in 2022, and could grow to 13.8 million by 20601. Understanding the intricate mechanisms underlying its pathogenesis is crucial for developing effective therapeutic interventions2.
AD is a multifactorial disease with a complex mechanism of pathogenesis involving genetic, environmental, and lifestyle factors. At the cellular and tissue levels, AD manifests as progressive neuronal loss, synaptic dysfunction, and the accumulation of characteristic neuropathological hallmarks, including extracellular amyloid-beta (A) plaques and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein3.
These pathological changes primarily affect brain regions involved in memory and cognitive function, such as the hippocampus and neocortex, leading to the clinical symptoms observed in AD patients. Extracellular amyloid-beta plaques and intracellular neurofibrillary tangles disrupt cellular communication and function in the brain. Amyloid-beta aggregates lead to synaptic dysfunction and neuronal death, while neurofibrillary tangles compromise the structural integrity of neurons, impairing their ability to transmit signals effectively. These alterations contribute to the progressive cognitive decline observed in Alzheimer’s disease, highlighting the critical role of these pathological changes in the pathogenesis of the condition2.
One significant genetic risk factor implicated in the development of AD is the apolipoprotein E (APOE) gene, specifically its 4 allele. The APOE gene, located on chromosome 19, encodes a glycoprotein, named APOE primarily expressed in the liver and brain, where it plays a crucial role in lipid metabolism and transport. APOE exists in three common isoforms: 2, 3, and 4, which differ by single amino acid substitutions at positions 112 and 1584,5,6. The precise structure of native APOE is still unclear due to the protein’s tendency to aggregate into other isoforms7 Among these isoforms, APOE 3 is the most prevalent, while APOE 4 is associated with an increased risk of developing AD. Conversely, individuals carrying the APOE 2 allele exhibit a decreased risk of AD by almost half as compared to those with the 3 allele8. Individuals with APOE 2/4 and 3/4 genotypes have an odds ratio (OR) of 2.64 and 3.63, respectively, in comparison to a homozygous APOE 3/3 genotype (reference OR of 1). This implies that individuals carrying the 4 allele (whether in combination with 2 or 3) have a higher risk of developing AD compared to individuals with the 3/3 genotype. Notably, the 3/4 genotype shows a significantly higher risk among the heterozygous genotypes. Moreover, the homozygous 4/4 genotype dramatically increases the risk, with an OR of 14.49 compared to the 3/3 genotype. Conversely, the homozygous 2/2 genotype is the most protective against AD, with the 2 allele associated with a reduced risk (OR = 0.621) in comparison to the 3/3 genotype. The presence of one 4 allele can advance the onset of AD by 2-5 years, while two 4 alleles can advance it by 5-10 years. Ethnicity also influences the impact of the 4 allele, with a stronger association in Japanese individuals and a weaker association in African American and Hispanic populations9
The precise mechanisms by which the APOE 4 allele is associated with susceptibility to AD are not fully elucidated but are thought to involve multiple pathways contributing to neuronal dysfunction and neurodegeneration. Evidence suggests that APOE 4 may exacerbate A aggregation and deposition, promoting the formation of neurotoxic A oligomers and fibrils. Additionally, APOE 4 has been implicated in impairing A clearance pathways, such as the blood-brain barrier transport and microglial-mediated phagocytosis, thereby exacerbating A accumulation and neuroinflammation.
Moreover, APOE 4 has been linked to synaptic dysfunction and neuronal injury through mechanisms involving tau pathology, oxidative stress, and neuroinflammation. APOE isoforms differentially modulate synaptic plasticity and neuronal repair processes, with APOE 4 exerting detrimental effects on synaptic integrity and neuronal resilience in response to insults. Also, APOE 4 may influence neurovascular function and cerebral blood flow regulation, contributing to compromised brain perfusion and hypoxic conditions that exacerbate neuronal vulnerability in AD10
The APOE 4 allele represents a major genetic risk factor for AD, exerting pleiotropic effects on cellular and tissue processes implicated in disease pathogenesis. Understanding the key mechanisms underlying the association between APOE 4 and AD is critical for developing targeted therapeutic strategies to mitigate its detrimental effects and ultimately halt or delay disease progression11. This literature review aims to explore the current understanding of how APOE 4 influences the risk of developing AD at the cellular level with discussion at the tissue level to help focus on the underlying molecular mechanisms involved in its pathogenicity.
Methods
Inclusion Criteria
For this literature review, inclusion criteria were established to ensure the selection of relevant studies. Key words include: Alzheimer’s disease, APOE 4, amyloid-beta, neuroinflammation, synaptic dysfunction, neurovascular dysfunction, neuronal survival, pathogenesis, mechanisms, genetics, lipid metabolism, apolipoproteins, neurodegeneration, animal models, and neurological disorders. Peer-reviewed articles published in English-language journals were considered eligible for inclusion. Studies investigating the role of APOE 4 in Alzheimer’s disease pathogenesis at the cellular and tissue levels were included. Both experimental and observational studies, including in vitro, animal models, and clinical research, were considered. Studies focusing on the molecular mechanisms underlying the effects of APOE 4 on AD-related processes such as amyloid-beta metabolism, neuroinflammation, synaptic dysfunction, neurovascular function, and neuronal survival were prioritized for inclusion.
Data Extraction
Data extraction was performed systematically to gather relevant information from selected studies. Key data points extracted included study design, experimental methods, main findings, and conclusions. Information on APOE genotype distribution, outcomes related to AD pathology, and mechanistic insights into the role of APOE 4 at the cellular and tissue levels were prioritized for extraction. Data were extracted independently by two reviewers to ensure accuracy and consistency.
Synthesis Method
The synthesis of findings from the selected studies was conducted through a narrative approach. Data were organized thematically according to the various aspects of APOE 4-mediated pathogenesis in Alzheimer’s disease. Specifically, findings related to APOE 4’s effects on amyloid-beta metabolism, neuroinflammation, synaptic dysfunction, neurovascular function, and neuronal survival were synthesized and discussed in detail. Patterns, trends, and inconsistencies across studies were identified and critically analyzed to provide a comprehensive overview of the current understanding of APOE 4’s role in AD pathogenesis at the cellular and tissue levels.
Quality Assessment
Quality assessment of the included studies was conducted to evaluate the methodological rigor and risk of bias. Assessment criteria included study design, sample size, participant selection, outcome measurement, statistical analysis, and reporting quality. Studies deemed to have methodological limitations or a high risk of bias were noted, and their potential impact on the overall conclusions of the review was considered. Quality assessment was performed by two reviewers, myself and my mentor, Hamid Abuwarda.
Understanding APOE 4 and Apolipoproteins
The APOE 4 allele, a variant of the apolipoprotein E gene, has been consistently associated with an increased risk of developing Alzheimer’s disease (AD) alleles, Carrying one APOE 4 allele brings the onset of AD forward by 2–5 years, and carrying two APOE 4 alleles brings the onset forward by 5–10 years12 Apolipoproteins are a group of proteins involved in the metabolism and transport of lipids in the body. They play a crucial role in packaging lipids into lipoprotein particles, which are essential for transporting cholesterol, triglycerides, and other lipids through the bloodstream. Apolipoproteins serve as structural components of these particles and also act as ligands for cell surface receptors, facilitating the uptake and clearance of lipids by various tissues. Additionally, apolipoproteins participate in lipid metabolism, including the regulation of cholesterol synthesis, metabolism, and excretion13. Apolipoproteins, including APOE, play critical roles in lipid metabolism and transport, facilitating the transport of lipids and cholesterol within the body. In the central nervous system (CNS), APOE is primarily synthesized by astrocytes and microglia, which are immune cells in the CNS. It functions in lipid homeostasis, neuronal repair, and synaptic plasticity. APOE is known to interact with lipoprotein particles, such as high-density lipoprotein (HDL), low-density lipoprotein (LDL), and very-low-density lipoprotein (VLDL), serving as ligands for cell surface receptors involved in lipid uptake and clearance. Beyond its canonical role in lipoprotein metabolism, APOE has been implicated in neuroprotective and neurodegenerative processes, including modulation of neuroinflammation, antioxidant defense, and amyloid-beta (A) metabolism13,14,15. However, the precise mechanisms by which APOE isoforms influence AD pathogenesis remain incompletely understood, highlighting the need for further investigation into the intricate interplay between APOE genotype, lipid metabolism, and neurodegenerative processes in the context of AD.
Peripheral APOE 4 and its Implications in Alzheimer’s Disease
While the primary focus of APOE 4’s role in Alzheimer’s disease (AD) has centered on its functions within the CNS, emerging evidence suggests that APOE 4 in the peripheral nervous system (PNS) may also contribute to AD pathogenesis through systemic mechanisms. Although APOE is primarily synthesized in astrocytes and microglia in the brain, it is abundantly expressed in peripheral tissues, including the liver, spleen, and macrophages. Peripheral APOE plays crucial roles in lipid metabolism, cholesterol transport, and immune response regulation, all of which are implicated in AD pathology16.
The study by Liu et al. investigated the impact of peripheral expression of APOE isoforms, specifically APOE 3 and APOE 4, on brain function using mouse models. Through inducible expression in the liver, APOE 3 and APOE 4 mice (iE3 and iE4 mice) were examined to understand the effects on brain function while circumventing the presence of brain-specific APOE in the brain itself. Both APOE 3 and APOE 4 were confirmed to be expressed in the liver and secreted into the bloodstream, rescuing metabolic abnormalities observed in APOE knockout (KO) mice.
Behavioral and electrophysiological studies revealed significant differences in brain function between iE3/Cre+ and iE4/Cre+ mice. APOE 4 expression in the liver led to memory deficits as compared to controls. This is done through multiple mechanisms including impaired cholesterol metabolism, increased A deposition, compromised BBB integrity, inflammation, oxidative stress, and direct effects on neuronal function16. APOE 3 expression improved associative memory as shown in Figure 1. Anxiety behaviors, locomotor activity, and motor coordination remained unaffected by either isoform.
Notably, peripheral APOE 4 suppressed synaptic plasticity, as evidenced by reduced long-term potentiation (LTP), a process that strengthens synapses, leading to a long-lasting increase in signal transmission between neurons, whereas APOE 3 increased it17. These findings suggest that peripheral APOE 3 had a beneficial effect on brain function, while peripheral APOE 4 exhibited a gain-of-toxic effect, impairing brain function in the absence of brain-derived APOE. This dichotomy underscores the critical role of peripheral APOE variants in influencing brain health independently of brain-derived APOE, highlighting the significance of systemic APOE effects on neurological outcomes. APOE 4’s detrimental impact may stem from its known effects on lipid metabolism and neuroinflammatory processes, which disrupt synaptic integrity and contribute to cognitive decline.
Overall, this study underscores the differential impact of peripheral APOE isoforms on brain function, providing insights into the role of APOE in neurobiology and potentially informing therapeutic strategies targeting APOE-related disorders such as Alzheimer’s disease.
One aspect of peripheral APOE 4’s involvement in AD is its impact on systemic lipid metabolism. APOE isoforms exhibit differential effects on plasma lipid levels, with APOE 4 carriers often displaying alterations in lipid profiles characterized by elevated levels of circulating cholesterol and triglycerides. These dyslipidemic changes may exacerbate vascular dysfunction and atherosclerosis, contributing to compromised cerebral blood flow and increased susceptibility to cerebrovascular pathology in AD. Furthermore, peripheral APOE 4-mediated dyslipidemia may indirectly influence CNS cholesterol homeostasis and A metabolism, exacerbating amyloid deposition and neurodegeneration18.
Beyond its effects on lipid metabolism, peripheral APOE 4 may modulate systemic immune responses and inflammation, which are increasingly recognized as key contributors to AD pathogenesis. APOE is known to interact with immune cells, including macrophages and T lymphocytes, regulating their activation, cytokine production, and inflammatory signaling pathways. APOE 4 has been associated with dysregulated immune responses and heightened pro-inflammatory cytokine production, both peripherally and centrally. These inflammatory changes may exacerbate neuroinflammation, glial activation, and neuronal damage in AD, amplifying disease progression18.
Moreover, peripheral APOE 4 may influence systemic clearance pathways for A and other amyloidogenic proteins implicated in AD. APOE isoforms differentially modulate the uptake and clearance of A by peripheral macrophages and the blood-brain barrier (BBB) a system of endothelial cells that shield the brain from toxic substances in the blood, supply the brain with nutrients, and provide a filtration system for harmful substances to exit the brain, and go to the bloodstream9. This affects the balance of A levels in the CNS and PNS5. APOE 4 has been shown to impair A clearance mechanisms, both in the periphery and within the brain, promoting A accumulation and aggregation. Additionally, peripheral APOE 4 may influence the transport of peripheral A into the CNS via the glymphatic system or other clearance pathways, exacerbating amyloid pathology and neurodegeneration in AD18.
Furthermore, peripheral APOE 4 may contribute to systemic metabolic dysfunction and insulin resistance, which have been implicated in AD pathogenesis. APOE 4 carriers exhibit increased susceptibility to metabolic syndrome, type 2 diabetes, and obesity, all of which
are associated with elevated AD risk1. Peripheral insulin resistance and dysregulated insulin signaling pathways may promote neuroinflammation, tau hyperphosphorylation, and synaptic dysfunction in AD, exacerbating cognitive decline and disease progression18.
Understanding the role of peripheral APOE 4 in AD offers new insights into the complex interplay between genetic, metabolic, and immune factors contributing to disease susceptibility and progression. Targeting peripheral APOE 4-mediated pathways may represent a promising therapeutic strategy for mitigating AD risk and progression, highlighting the importance of comprehensive approaches to AD treatment and prevention.
The Role of Brain APOE 4 in Alzheimer’s Disease Pathogenesis
While APOE is expressed throughout the body, its expression within the brain is of particular interest due to its crucial roles in lipid metabolism, neuronal repair, synaptic plasticity, and amyloid-beta (A) metabolism. This paper will delve into the intricate functions of brain APOE 4 and its implications in the pathogenesis of AD.
A study conducted by Liu, et. al identified APOE 4’s influence on lipid pathways in the brain. These findings have profound implications for our understanding of Alzheimer’s disease, as dysregulation of lipid metabolism has been increasingly implicated in the pathogenesis of neurodegenerative disorders. The dysregulation of lipid metabolism affects the pathogenesis of AD due to its role in amyloid-beta peptide production and clearance, as well as in the formation of neurofibrillary tangles, contributing to the neurodegenerative processes characteristic of AD.
Recent studies have extensively explored the impact of APOE 4 on lipid pathways within neurons. For instance, Liu et al. identified significant influences of APOE 4 on lipid metabolism in neuronal cells, underscoring its potential role in Alzheimer’s disease (AD) pathogenesis19. Dysregulation of lipid metabolism has been increasingly implicated in neurodegenerative disorders, affecting processes such as amyloid-beta peptide production, clearance, and the formation of neurofibrillary tangles, hallmark features of AD19.Building upon these neuronal findings, Blanchard et al. delved into the role of oligodendrocytes, the myelin-producing cells of the central nervous system, in AD22. Their investigation revealed upregulation of cholesterol biosynthesis genes specifically in APOE 4 -expressing oligodendrocytes, suggesting potential disruptions in myelin maintenance and function22. This finding aligns with prior evidence indicating white matter abnormalities and myelin dysfunction in AD pathology.
This emerging research underscores a paradigm shift towards investigating the role of non-neuronal cell types in AD. By examining how APOE 4 influences lipid pathways in both neurons and oligodendrocytes, scientists are gaining deeper insights into the complex interplay between lipid metabolism and neurodegeneration
Blanchard et al. investigated the role of oligodendrocytes, the myelin-producing cells in the central nervous system, in Alzheimer’s disease (AD). Their study highlighted the upregulation of cholesterol biosynthesis genes in APOE 4 oligodendrocytes, suggesting disruptions in myelin maintenance and function. This finding supports previous evidence of white matter abnormalities and myelin dysfunction in AD pathology.
a) Co-Culture Myelination Analysis
– Description: Images of APOE4/4 and APOE3/3 oligodendroglia with NGN2-induced neurons after 6 weeks.
– Quantification: Myelin basic protein (MBP) quantified around neurofilament staining using Imaris software.
– Data: Each point is an average from 4 independent images. Scale bar: 50 ?m.
– Findings: APOE4/4 co-cultures show reduced myelination compared to APOE3/3 co-cultures, as indicated by lower MBP staining intensity. This suggests APOE genotype influences myelination efficiency.
b) Mix-and-Match Experiment
– Setup: Co-cultures in four combinations:
1. APOE3/3 with APOE3/3
2. APOE3/3 with APOE4/4
3. APOE4/4 with APOE4/4
4. APOE4/4 with APOE3/3
– Quantification: MBP around neurofilament staining analyzed using Imaris and ImageJ. Each point is an average from 4 independent images.
– Statistical Analysis: One-way ANOVA with Bonferroni correction.
– Findings: APOE4/4 oligodendrocytes exhibit reduced myelination when co-cultured with either APOE4/4 or APOE3/3 neurons compared to APOE3/3 oligodendrocytes. APOE4/4 neurons also show reduced myelination with APOE4/4 oligodendrocytes compared to APOE3/3 oligodendrocytes. These results highlight the complex interplay between APOE genotypes in myelination, with implications for neurodegenerative disorders associated with APOE4.
Additionally, the identification of elevated cholesteryl ester levels in APOE 4 carriers linked APOE 4 to dysregulated lipid metabolism in AD. The accumulation of cholesteryl esters, which are crucial for cholesterol storage, has been associated with neurodegenerative processes. This suggests a cholesterol-associated mechanism for APOE 4-mediated neuronal dysfunction and synaptic impairment, emphasizing the significance of lipid metabolism in AD18.
Overall, the study’s findings highlight the complex interplay between APOE 4, lipid metabolism, and Alzheimer’s disease pathogenesis. By elucidating the molecular mechanisms underlying APOE 4-mediated effects on lipid pathways in the brain, this research study provides critical insights into potential therapeutic targets for mitigating the detrimental effects of APOE 4 and ultimately halting the progression of Alzheimer’s disease.
Synaptic dysfunction is a hallmark of early AD pathogenesis, preceding widespread neuronal loss and cognitive decline. APOE 4 has been implicated in impairing synaptic plasticity and neuronal function within the brain, contributing to memory deficits and cognitive impairment in AD. APOE isoforms differentially modulate synaptic transmission, dendritic spine density, and long-term potentiation (LTP), with APOE 4 exerting detrimental effects on synaptic integrity and neuronal connectivity. Moreover, APOE 4 has been shown to impair neuronal survival pathways, increase susceptibility to excitotoxicity, and enhance tau phosphorylation, exacerbating neurodegenerative changes and cognitive decline in AD16.
Cerebrovascular dysfunction is increasingly recognized as a contributing factor to AD pathogenesis, with APOE 4 implicated in disrupting neurovascular function and BBB integrity. APOE 4 carriers exhibit alterations in cerebral blood flow regulation, compromised BBB permeability, and increased susceptibility to cerebrovascular pathology. These vascular changes may exacerbate neuronal vulnerability, promote neuroinflammation, and impair A clearance mechanisms within the brain, contributing to AD pathogenesis and cognitive decline16.
Brain APOE 4 plays a multifaceted role in the pathogenesis of Alzheimer’s disease, influencing A metabolism, neuroinflammation, synaptic dysfunction, neurovascular function, and neuronal survival. Elucidating the molecular mechanisms underlying the effects of APOE 4 on brain function and AD pathogenesis is essential for developing targeted therapeutic interventions aimed at mitigating its detrimental effects and ultimately halting disease progression16.
Discussion
While considerable progress has been made in elucidating the role of brain APOE 4 in Alzheimer’s disease (AD) pathogenesis, several concerns and limitations warrant consideration. Firstly, the multifactorial nature of AD suggests that APOE 4 may interact with other genetic, environmental, and lifestyle factors to modulate disease risk and progression. Thus, future studies should aim to elucidate the complex interplay between APOE 4 and other susceptibility genes, as well as environmental factors such as diet, exercise, and socioeconomic status, to better understand AD heterogeneity and inform personalized therapeutic interventions19
Moreover, the precise mechanisms underlying the effects of APOE 4 on AD pathogenesis remain incompletely understood. While APOE 4 has been implicated in modulating amyloid-beta (A) metabolism, neuroinflammation, synaptic dysfunction, neurovascular function, and neuronal survival, the specific molecular pathways mediating these effects require further investigation. Integrating multi-omic approaches, including genomics, transcriptomics, proteomics, and metabolomics, may provide comprehensive insights into the molecular underpinnings of APOE 4-mediated AD pathogenesis20. Future research efforts focused on understanding the complex interplay between APOE 4, amyloid-beta pathology, and neurodegenerative processes within the brain hold promise for advancing our understanding of AD and identifying novel therapeutic strategies for this devastating disorder.
Additionally, translating preclinical findings into clinically effective interventions poses significant challenges. While experimental studies in animal models, specifically mice, have elucidated key mechanisms underlying the effects of APOE 4 on AD pathogenesis, translating these findings to human patients remains a formidable task20. The development of APOE 4-targeted therapeutics necessitates rigorous preclinical validation, including the optimization of drug delivery strategies, assessment of pharmacokinetics and pharmacodynamics, and evaluation of long-term safety and efficacy in human clinical trials.
Furthermore, the heterogeneity of APOE 4-associated phenotypes and outcomes complicates therapeutic targeting and risk prediction in AD. Not all APOE 4 carriers develop AD, indicating the existence of additional genetic and environmental modifiers that modulate disease susceptibility. Therefore, identifying reliable biomarkers for stratifying APOE 4 carriers based on their risk of developing AD and monitoring disease progression remains a critical challenge in the field.
While significant progress has been made in elucidating the role of APOE 4 in AD, several concerns and limitations remain. These include the complex interplay between APOE 4 and other genetic and environmental factors, the need for comprehensive molecular characterization of APOE 4-mediated pathways, challenges in translating preclinical findings into clinically effective interventions, and the heterogeneity of APOE 4-associated phenotypes and outcomes. Addressing these concerns and limitations is essential for advancing our understanding of APOE 4-mediated AD pathogenesis and developing personalized therapeutic strategies for this devastating neurodegenerative disorder.
Moving forward, future research efforts should focus on elucidating the molecular mechanisms underlying the effects of APOE 4 on AD pathogenesis, identifying reliable biomarkers for risk prediction and disease monitoring, optimizing translational strategies for drug development, and exploring innovative therapeutic approaches targeting APOE 4-mediated pathways. By addressing these challenges and limitations, we can pave the way for the development of effective treatments that target the underlying pathogenic mechanisms of AD, ultimately improving outcomes for patients and their families affected by this debilitating disease.
In conclusion, the role of apolipoprotein E (APOE) 4 in Alzheimer’s disease (AD) pathogenesis is multifaceted, encompassing its effects on amyloid-beta metabolism, neuroinflammation, synaptic dysfunction, neurovascular function, and neuronal survival. APOE 4 represents a major genetic risk factor for AD, with carriers exhibiting a significantly increased risk of developing the disease compared to non-carriers. Understanding the molecular mechanisms underlying the effects of APOE 4 on AD pathogenesis is crucial for developing targeted therapeutic interventions aimed at mitigating its detrimental effects and ultimately halting disease progression.
Acknowledgments
I would like to thank Hamid Abuwarda, MD/PhD student at Yale University School of Medicine, for his guidance and insight during my research on this topic. I would also like to thank Lumiere Education for their support and resources during the undertaking of this project.
References
- “2022 Alzheimer’s Disease Facts and Figures.” Alzheimer’s & Dementia, vol. 18, no. 4, 14 Mar. 2022, pp. 700–789, pubmed.ncbi.nlm.nih.gov/35289055/, https://doi.org/10.1002/alz.12638. Accessed 29 Mar. 2024. [↩] [↩]
- Knopman, David S., et al. “Alzheimer Disease.” Nature Reviews Disease Primers, vol. 7, no. 1, May 2021, https://doi.org/10.1038/s41572-021-00269-y. [↩] [↩]
- Bu G. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat Rev Neurosci. 2009;10:333–44., Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol neuro- degeneration. 2020;15:40. [↩]
- Ruiz J, Kouiavskaia D, Migliorini M, Robinson S, Saenko EL, Gorlatova N, Li D, Lawrence D, Hyman BT, Weisgraber KH, Strickland DK. The apoE isoform bind- ing properties of the VLDL receptor reveal marked differences from LRP and the LDL receptor., J Lipid Res. 2005;46:1721–31. [↩]
- Hatters DM, Zhong N, Rutenber E, Weisgraber KH. Amino-terminal domain stability mediates apolipoprotein E aggregation into neurotoxic fibrils. J Mol Biol. 2006;361:932–44. [↩]
- Ma Q, Zhao Z, Sagare AP, Wu Y, Wang M, Owens NC, Verghese PB, Herz J, Holtzman DM, Zlokovic BV. Blood-brain barrier-associated pericytes internal- ize and clear aggregated amyloid-42 by LRP1-dependent apolipoprotein E isoform-specific mechanism. Mol neurodegeneration. 2018;13:57. [↩]
- 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;108:14813–8. [↩]
- Corder EH, Saunders AM, Risch NJ, Strittmatter WJ, Schmechel DE, Gaskell PC Jr, Rimmler JB, Locke PA, Conneally PM, Schmader KE, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet. 1994;7:180–4. [↩]
- Genin, E. et al. APOE and Alzheimer disease: a major gene with semi-dominant inheritance. Mol. Psychiatry 16, 903–907 (2011). [↩]
- Yamazaki, Yu, et al. “Apolipoprotein E and Alzheimer Disease: Pathobiology and Targeting Strategies.” Nature Reviews Neurology, vol. 15, no. 9, July 2019, https://doi.org/10.1038/s41582-019-0228-7. [↩]
- Bu G. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat Rev Neurosci. 2009;10:333–44. [↩]
- Yamazaki, Yu, et al. “Apolipoprotein E and Alzheimer Disease: Pathobiology and Targeting Strategies.” Nature Reviews Neurology, vol. 15, no. 9, July 2019, https://doi.org/10.1038/s41582-019-0228-7. [↩]
- Boyles JK, Pitas RE, Wilson E, Mahley RW, Taylor JM. Apolipoprotein E associ- ated with astrocytic glia of the central nervous system and with nonmyelin- ating glia of the peripheral nervous system. J Clin Investig. 1985;76:1501–13. [↩]
- Aoki K, Uchihara T, Sanjo N, Nakamura A, Ikeda K, Tsuchiya K, Wakayama Y. Increased expression of neuronal apolipoprotein E in human brain with cerebral infarction. Stroke. 2003;34:875–80. [↩]
- Mahley RW, Huang Y. Apolipoprotein e sets the stage: response to injury trig- gers neuropathology. Neuron. 2012;76:871–85. [↩]
- Liu, Chia-Chen, et al. “Peripheral ApoE4 Enhances Alzheimer’s Pathology and Impairs Cognition by Compromising Cerebrovascular Function.” Nature Neuroscience, vol. 25, no. 8, 1 Aug. 2022, pp. 1020–1033, www.nature.com/articles/s41593-022-01127-0, https://doi.org/10.1038/s41593-022-01127-0. Accessed 16 Mar. 2024.? [↩] [↩] [↩] [↩] [↩]
- Li, Jimmy, and Dènahin Hinnoutondji Toffa. “Magnesium Supplemental Therapy in Epileptogenesis and Ictogenesis.” Elsevier EBooks, 1 Jan. 2023, pp. 327–344, www.sciencedirect.com/topics/neuroscience/long-term-potentiation, https://doi.org/10.1016/b978-0-323-90052-2.00035-4. Accessed 24 Mar. 2024. [↩]
- Blanchard, Joel W, et al. “APOE4 Impairs Myelination via Cholesterol Dysregulation in Oligodendrocytes.” Nature, vol. 611, no. 7937, 16 Nov. 2022, pp. 769–779, www.nature.com/articles/s41586-022-05439-w, https://doi.org/10.1038/s41586-022-05439-w. Accessed 16 Mar. 2024. [↩] [↩] [↩] [↩] [↩]
- Tokgöz S, Claassen J. Exercise as Potential Therapeutic Target to Modulate Alzheimer’s Disease Pathology in APOE 4 Carriers: A Systematic Review. Cardiol therapy. 2021;10:67–88., Cancela-Carral JM, López-Rodríguez A, Mollinedo-Cardalda I. Effect of physi- cal exercise on cognitive function in older adults’ carriers versus noncarriers of apolipoprotein E4: systematic review and meta-analysis. J Exerc rehabilita- tion. 2021;17:69–80. [↩]
- Liu T, Zhu B, Liu Y, Zhang X, Yin J, Li X, Jiang L, Hodges AP, Rosenthal SB, Zhou L, et al: Multi-omic comparison of Alzheimer’s variants in human ESC-derived microglia reveals convergence at APOE. J Exp Med 2020, 217. [↩] [↩]