Next Article in Journal
Identification of Serum Cytokine Biomarkers Associated with Multidrug Resistant Tuberculosis (MDR-TB)
Previous Article in Journal
Recent Advances in Understanding the Role of TIGIT+ Follicular Helper T Cells in IgG4-Related Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of APOE and NF-κB in Alzheimer’s Disease

Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB R2H 2A6, Canada
Immuno 2021, 1(4), 391-399; https://doi.org/10.3390/immuno1040027
Submission received: 2 September 2021 / Revised: 13 October 2021 / Accepted: 26 October 2021 / Published: 28 October 2021
(This article belongs to the Section Neuroimmunology)

Abstract

:
Apolipoprotein E (APOE) has three different isoforms, with APOE4 carriers representing a major risk factor for the development of Alzheimer’s disease (AD). AD is the most common form of dementia, and is a relentlessly progressive disorder that afflicts the aged, characterized by severe memory loss. Presently, AD does not have a cure, increasing the urgency for the development of novel therapeutics for the prevention/treatment of AD. The APOE4 isoform is associated with many pathological mechanisms, such as increased neuroinflammation and a reduction in β-amyloid (Aβ) clearance. The accumulation of Aβ plaques in the brain is a hallmark of AD. The presence of APOE4 can increase neuroinflammation via overactivation of the nuclear factor kappa B (NF-κB) pathway. The NF-κB pathway is a family of transcription factors involved with regulating over 400 genes involved with inflammation. AD is associated with sustained inflammation and an overactivation of the NF-κB pathway. Therefore, targeting the APOE4 isoform and suppressing the NF-κB pathway using anti-inflammatory compounds may result in the development of novel therapeutics for the prevention/treatment of AD.

1. Introduction

The global population is reaching unprecedented levels of advanced age, generating a large demographic vulnerable to brain insults, thus creating enormous healthcare, economic and social impacts. Alzheimer’s disease (AD) is a relentlessly progressive disorder that afflicts the aged, and is the sixth leading cause of death in the United States (Centers for Disease Control and Prevention). Two hallmarks of AD include β-amyloid (Aβ) peptides that are extracellularly deposited, and neurofibrillary tangles (NFTs), composed of intracellular filamentous aggregates of hyperphosphorylated tau protein [1,2,3,4]. In June 2021, Aducanumab was approved as a disease-modifying therapeutic via the removal of Aβ plaques in the brain [5]. However, the presence of Aβ plaques does not always result in AD, which has led to controversy over the use of aducanumab. Other therapeutics used for AD do not alter the course of the disease, which increases the urgency of developing novel therapeutics for AD.
Apolipoprotein E (APOE) has emerged as a therapeutic target for the treatment of AD. APOE is a cholesterol carrier that is involved with lipid transportation and repair in the brain [6]. APOE is a polymorphic protein with three isoforms, APOE4, APOE3, and APOE2, which differ from each other by two amino acid substitutions (arg/arg, cys/arg, and cys/cys, respectively), resulting in different tertiary structures and likely altering APOE function. Despite the similarity among the isoforms, they present major differences in the risk of developing AD, with APOE4 being a strong risk factor for the development of AD. APOE3 is the most common isoform and does not affect risk of developing AD, and APOE2 is associated with a decreased risk of the development of AD. The worldwide frequency of APOE4 is 13.7%, that of APOE3 is 77.9%, and that of APOE2 is 8.4% [7]. The frequency of APOE4 in AD is approximately 40% [7]. Genome-wide association studies have shown that APOE4 is the strongest genetic risk factor for early-onset AD and late-onset AD [8,9].

2. Neuroinflammation and Alzheimer’s Disease

Immunological processes in the brain contribute to AD progression and severity [10]. Microglia are the resident macrophages of the central nervous system (CNS) that detect the presence of pathogens and cellular debris while providing tissue maintenance. In addition, microglia contribute to synaptic plasticity through phagocytosis, which removes select synapses [11]. Therefore, sustained inflammation may cause synaptic dysfunction. The depletion of microglia is associated with learning and memory impairments [12]. Microglia become active once exposed to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). Microglia bind to soluble and fibrillar forms of Aβ in different ways [13]. Microglia bind to soluble forms of Aβ via endocytosis and the low-density lipoprotein (LDL) receptor-related protein pathway. The fibrillar form of Aβ binds to the cell surface of the innate immune receptor complex, which results in phagocytosis. Other reviews have examined the role of microglia in AD in more detail [10].
Nuclear factor kappa B (NF-κB) is a family of transcription factors involved with the regulation of inflammation [14,15]. The NF-κB family includes NF-κBp50/105, NF-κBp52/100 (RelB), NF-κBp65 (RelA), and NF-κBp75 (c-Rel), with dimers bound by the inhibitory protein IκB. The NF-κB proteins are often located in the cytoplasm in an inactive state [16]. The activation of NF-κB results in two signaling pathways, the canonical and nonconical pathways, which are both important for the modulation of inflammatory and immune responses [17,18]. The canonical NF-κB pathway is activated when binding occurs with immune receptors, including tumor necrosis factor receptor (TNFR), interlukin-1 receptor (IL-1R), toll-like receptors (TLR), and lipopolysaccharide (LPS), which activates the IκB kinase (IKK) trimeric complex (IKKα, IKKβ, and IKKγ (NEMO)) and phosphorylates IκBα [19]. The NF-κB p50/RelA dimers translocate from the cytoplasm into the nucleus and bind to IκB, which regulates NF-κB-associated genes [20]. The noncanonical NF-κB pathway is activated by the B-cell activating factor belonging to the TNF family receptor (BAFFR) and the receptor activator for NF-κB (RANK), which involves interactions with IKKα and the phosphorylation of NF-κB p100. Once NF-κB p100 is degraded, NF-κB p52/RelB dimers translocate from the cytoplasm into the nucleus to modulate NF-κB genes [19].
NF-κB dysregulation is associated with various neurological conditions, including ischemic stroke, multiple sclerosis, AD, and other neurodegenerative disorders [21,22,23,24,25]. In AD, the transmembrane aspartic protease beta site amyloid precursor protein cleaving enzyme 1 (BACE1) initiates the production of Aβ with gamma-secretase, which cleaves a portion of the extracellular amyloid precursor protein (APP) [26]. This results in an accumulation of Aβ that can form plaques between neurons, which disrupts the neuronal signaling and leads to dysfunctional synaptic plasticity and neuroinflammation [27]. The NF-κB pathway is involved with the modulation of BACE1 expression, and therefore the production of Aβ. The inhibition of the functional binding site IkappaB kinase of Nf-κB prevents Aβ-induced BACE1 promoter transactivation, suggesting that this pathway could modulate an Aβ-associated phenotype [28]. NF-κB activation upregulates the transcription of BACE1 and increases the expression of Aβ [29,30]. Aβ infusions into mice brains and nonhuman primates increased IKKβ/NF-κB signaling in the hypothalamus [31]. In drosophila, the genetic overexpression of NF-κB in neurons and glial leads to an AD phenotype with increased neurodegeneration [32]. AD is associated with sustained inflammation, which in part may be due to the Receptor for Advanced Glycation End-Products (RAGE)/NF-κB axis, which activates an autoregulatory loop that further amplifies neuroinflammation [33]. The Advanced Glycation End-Products (AGEs)/RAGE complex upregulates BACE1 via the activation of the NF-κB pathway [34]. The immunoreactivity of p65 was increased near Aβ plaques in AD patients, suggesting that NF-κB is active in neurons surrounding Aβ plaques [24]. The regulation of NF-κB by Aβ may result in glial cell activation, as demonstrated by experiments showing that Aβ activated the NF- κB pathway and increased nitric oxide in astrocytes [35]. Tanshinone I inhibits the NF-κB pathway in LPS-induced microglia [36]. Phytochemicals inhibit the NF-κB pathway by preventing phosphorylation and ubiquitination, which inhibits the degradation of IκB, thereby preventing the translocation of NF-κB into the nucleus [37]. Given the detrimental effects that an overactive NF-κB pathway has on neurodegeneration, attenuating the NF-κB pathway may improve neurodegenerative conditions, such as AD.

3. APOE and Inflammation

APOE and its receptors play important roles in inflammatory responses that modulate the clearance of Aβ. APOE genotypes affect Aβ plaque deposition and can cause cerebral amyloid angiopathy (CAA) [38]. APOE is deposited in the plaques of patients with AD, and is more abundant in APOE4 carriers compared with non-APOE4 carriers [39,40,41,42]. PET scanning using Pittsburgh compound B (PiB) showed that fibrillar aggregates of Aβ were more common in people who are APOE4 carriers [43,44]. APOE4 carriers have high rates of fibrillar Aβ in frontal, posterior cingulate-precuneus, temporal, parietal, and basal ganglia regions of the brain [45]. Cognitively normal APOE4 carriers received PiB PET imaging that indicated fibrillar aggregates of Aβ at approximately 56 years of age, compared to approximately 76 years of age in non-APOE4 carriers [46]. These results have led to speculation that increases in fibrillar Aβ in APOE4 carriers in cognitively normal people may result in an increased risk of developing mild cognitive impairment (MCI) and/or AD in the future [47].
Since the 1990s, transgenic mice have been used to model AD in the presence of mouse APOE. As previously mentioned, APOE has three different isoforms; however, mice express a single isoform, and it differs from the human APOE isoforms by approximately 100–300 amino acids [48]. The difference between human APOE and mouse APOE has led researchers to investigate the role of transcription factors in the expression of APOE, such as the LXRE consensus sequences in human and mouse APOE [49,50,51]. APOE knock-out mice were crossed with a transgenic mouse model of AD overexpressing a human mutant APP gene, resulting in PDAPP+/+; APOE −/− mice, which exhibited reduced Aβ deposits compared to PDAPP+/+; APOE +/+ mice [52]. PDAPP+/+; APOE −/− mice that expressed either human APOE2, APOE3, or APOE4 showed a reduction in Aβ40 in plasma as they aged for each isoform [53]. However, levels of Aβ40 and Aβ42 increased in the brain as the mice aged, regardless of the APOE isoform. Hippocampal insoluble Aβ40 and Aβ42 levels increased in an APOE isoform-dependent manner, with the highest levels in the APOE4 mice and the lowest levels in APOE2 mice.
APOE4 mice had increased levels of microglia, astrocytes, and invading T-cells after a brain infusion of LPS. In addition, the APOE4 mice had increased cytokine responses compared with APOE2 and APOE3 mice [54,55]. Cortical levels of IL-1β and the microglial reactivity in cortical plaques of APOE4 mice were increased compared to APOE3 mice [56]. Experimental autoimmune encephalomyelitis impaired learning and memory in APOE4 knock-in mice, suggesting that neuroinflammation affects learning and memory in APOE4 carriers [57]. Intravenous LPS administration increased pro-inflammatory cytokines, TNFα, and IL-6 in APOE4 mice compared to APOE3 mice [58]. Additionally, the administration of APOE mimetic peptide from the receptor-binding region decreased systemic and brain pro-inflammatory responses after administration with LPS. The APOE peptide was associated with the decreased activation of c-Jun N-terminal kinase (JNK) signaling [59]. The microglial lipoprotein receptors regulate JNK activity, and are necessary for APOE’s regulation of inflammation. The APOE mimetic peptide crosses the blood–brain barrier (BBB), and using peptides that can cross the BBB may be a novel therapeutic strategy for the treatment of AD [60].
While there is mounting evidence that APOE4 is associated with pro-inflammatory responses, an in vitro study using cultured rat glia found that APOE4 decreased the oligomeric Aβ production of nitric oxide synthase and cyclo-oxygenase-2, suggesting that APOE4 has anti-inflammatory properties [61]. APOE has anti-inflammatory effects in isolated macrophages via the APOE receptor-2, which result in macrophage conversion from pro-inflammatory M1 to the anti-inflammatory M2 [62]. Endogenous APOE from glial cell cultures inhibits microglial nitric oxide production [63]. The inhibition of inflammatory signaling increased APOE expression, which indicates that inflammation and APOE levels are involved in a negative feedback loop [64]. APOE deletion upregulates TLR4 and TLR2, and increases TLR activation [65,66]. The APOE protein is involved with an anti-inflammatory state, with APOE4 being the least anti-inflammatory, APOE2 being the most anti-inflammatory, and APOE3 being in the middle of the anti-inflammatory scale [67].

4. APOE and NF-κB

APOE is shown to modulate neuroinflammation via the NF-κB pathway. APOE4 mice showed increased NF-κB-regulated genes compared to APOE3 mice [68]. However, another study found that both APOE3 and APOE4 downregulated the NF-κB pathway [69]. These discrepant results may be due to the differences in methodology used between the two studies, with the in vivo experiment showing an increase in NF-κB activity and the in vitro experiment showing a decrease in NF- κB activity. APOE knock-out mice had increased inflammation and oxidative stress via activation of the NF-κB pathway [70]. PDAPP+/+; APOE −/− mice have reduced Aβ deposits in the cortex and hippocampus, and future studies should examine the NF-κB pathway in PDAPP+/+; APOE −/− mice [52]. Administration of the anti-inflammatory compounds Tanshinone IIA and Astragaloside IV in APOE knock-out mice suppressed the TLR4/NF-κB signaling pathway in vivo and in vitro [71]. Brain infusions of LPS in APOE4 mice were associated with the increased activation of the NF-κB pathway compared to APOE3 mice [72]. APOE4 mice showed increased nuclear translocation of NF-κB and increased IL-1β. The activation of the NF-κB pathway was increased after traumatic brain injury (TBI) in APOE4 mice compared to APOE3 mice [73]. APOE3 may inhibit the NF-κB pathway after TBI to alleviate BBB impairment. Schwann cells from APOE4 and APOE2 mice showed impaired cytokine production, which may have resulted from activation of the NF-κB pathway [74]. APOE activates the NF-κB pathway, inducing the expression of immunosuppressive chemokines Cxcl1 and Cxcl5 in tumor cells [75].
A high-fat diet and sedentary lifestyle can affect many medical conditions, including AD. APOE knock-out mice fed a high-fat diet had hypothalamic inflammation, glial cells activation, and cognition decline, which were reversed with diet control and exercise [76]. The diet control and exercise resulted in increased expressions of SIRT1 and the inhibition of the NF-κB pathway. Chronic stress is another important lifestyle factor that can influence various health conditions. Chronic unpredictable mild stress in APOE knock-out mice upregulated TLR4/NF-κB expression [77], and the administration of an NF-κB inhibitor downregulated the NF-κB pathway [78]. APOE knock-out mice that received AGEs via injection displayed increased Aβ formation and NF-κB p65 expression [79]. The statin medication atorvastatin decreased Aβ formation and suppressed AGEs-induced NF-κB p65 expression. PDAPP+/+; APOE −/− mice have decreased Aβ deposits, which contrasts with the previously mentioned experiment with increased Aβ formation. PDAPP+/+ mice are generated using a platelet-derived growth factor promoter with a human APP gene mutation associated with AD. Modafinil is prescribed for narcolepsy patients to increase wakefulness, and has anti-inflammatory effects. In APOE knock-out mice, modafinil inhibited the NF-κB pathway [80].
Vascular defects occurred in APOE4 mice before the neurodegenerative impairments occurred [81]. Astrocytes that secreted APOE3 and APOE2 but not APOE4 inhibited the cyclophilin A (CypA)-NF-κB–matrix-metalloproteinase-9 (MMP-9) pathway in pericytes, suggesting that APOE4 is a key target for the treatment of neurovascular conditions [82]. CypA has a variety of roles, including protein folding, trafficking and T cell activation, and is secreted from cells in response to inflammation [83]. MMP-9 is a type of enzyme in the zinc-metalloproteinases family involved with the breakdown of the extracellular matrix in both normal and pathological processes, including neurodegeneration [84]. The pro-inflammatory CypA–NF-κB–MMP-9 pathway causes BBB impairment via the MMP-9 degradation of tight junction proteins, which is associated with the onset of neurodegenerative disorders [81]. Additionally, astrocytes that secrete APOE3 and APOE2 have high binding affinities with lipoprotein receptor-related protein 1 (LRP1) [81]. However, astrocytes that secrete APOE4 have a low binding affinity with LRP1. In pericytes, the weak binding affinity of APOE4 to LRP1 leads to a reduction in Aβ clearance and a subsequent Aβ accumulation, resulting in neurodegeneration (see Figure 1). The inhibition of the CypA–NF-κB–MMP-9 pathway in APOE4 mice increased the coverage of tight junction proteins, prevented the loss of neurons and axon density, and improved cognitive function [85]. However, the inhibition of the CypA–NF-κB–MMP-9 pathway does not protect against Aβ accumulation.

5. Conclusions

APOE4 and the NF-κB pathway have emerged as targets for the prevention/treatment of AD. Targeting the APOE4 isoform may suppress the NF-κB pathway, given that the weak binding of APOE4 to LRP1 leads to increased NF-κB activity. Novel therapeutic approaches focus on removing APOE4 with antibodies and modifying the structure of the APOE4 protein to shift to the APOE3 and APOE2 isoforms [86]. Using CRISPR in cell culture showed that APOE4 can be converted into APOE3, and does not affect the expression of APOE3 [87,88]. However, in vivo experiments converting APOE4 to APOE3 have not been reported. The overexpression of the NF-κB pathway in AD has gained increased recognition, making it a therapeutic target. The NF-κB pathway triggers a cascade of pro-inflammatory cytokines and chemokines, and compounds including resveratrol, indomethacin, quercetin, adiponectin, caffeic acid, aspirin, and sodium salicylate may suppress the NF-κB pathway, leading to the development of a novel therapeutic for AD [89]. Future research should examine the interactions among the APOE–LRP1–CypA–NF-κB–MMP-9 pathway for the development of novel therapeutics to prevent/treat AD.

Funding

This research received no external funding.

Acknowledgments

I gratefully acknowledge the Provost’s Postdoctoral Fellowship for Black and Indigenous Scholars at York University.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Glabe, C.C. Amyloid accumulation and pathogensis of Alzheimer’s disease: Significance of monomeric, oligomeric and fibrillar Abeta. Subcell. Biochem. 2005, 38, 167–177. [Google Scholar] [CrossRef] [PubMed]
  2. Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Muralidar, S.; Ambi, S.V.; Sekaran, S.; Thirumalai, D.; Palaniappan, B. Role of tau protein in Alzheimer’s disease: The prime pathological player. Int. J. Biol. Macromol. 2020, 163, 1599–1617. [Google Scholar] [CrossRef]
  4. Adlimoghaddam, A.; Albensi, B.C. The nuclear factor kappa B (NF-kappaB) signaling pathway is involved in ammonia-induced mitochondrial dysfunction. Mitochondrion 2020, 57, 63–75. [Google Scholar] [CrossRef] [PubMed]
  5. Mukhopadhyay, S.; Banerjee, D. A primer on the evolution of aducanumab: The first antibody approved for treatment of alzheimer’s disease. J. Alzheimers Dis. 2021, 83, 1537–1552. [Google Scholar] [CrossRef]
  6. Mahley, R.W.; Rall, S.C., Jr. Apolipoprotein E: Far more than a lipid transport protein. Annu. Rev. Genom. Hum. Genet. 2000, 1, 507–537. [Google Scholar] [CrossRef]
  7. Farrer, L.A.; Cupples, L.A.; Haines, J.L.; Hyman, B.; Kukull, W.A.; Mayeux, R.; Myers, R.H.; Pericak-Vance, M.A.; Risch, N.; van Duijn, C.M. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and alzheimer disease meta analysis consortium. JAMA 1997, 278, 1349–1356. [Google Scholar] [CrossRef]
  8. Harold, D.; Abraham, R.; Hollingworth, P.; Sims, R.; Gerrish, A.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; Dowzell, K.; Williams, A.; et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1088–1093. [Google Scholar] [CrossRef] [Green Version]
  9. Lambert, J.C.; Heath, S.; Even, G.; Campion, D.; Sleegers, K.; Hiltunen, M.; Combarros, O.; Zelenika, D.; Bullido, M.J.; Tavernier, B.; et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1094–1099. [Google Scholar] [CrossRef] [PubMed]
  10. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [Green Version]
  11. Morris, G.P.; Clark, I.A.; Zinn, R.; Vissel, B. Microglia: A new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol. Learn. Mem. 2013, 105, 40–53. [Google Scholar] [CrossRef]
  12. Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lee, C.Y.; Landreth, G.E. The role of microglia in amyloid clearance from the AD brain. J. Neural. Transm. 2010, 117, 949–960. [Google Scholar] [CrossRef] [Green Version]
  14. Lawrence, T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 2009, 1, a001651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Eshraghi, M.; Adlimoghaddam, A.; Mahmoodzadeh, A.; Sharifzad, F.; Yasavoli-Sharahi, H.; Lorzadeh, S.; Albensi, B.C.; Ghavami, S. Alzheimer’s disease pathogenesis: Role of autophagy and mitophagy focusing in microglia. Int. J. Mol. Sci. 2021, 22, 3330. [Google Scholar] [CrossRef] [PubMed]
  16. Hayden, M.S.; Ghosh, S. Signaling to NF-kappaB. Genes Dev. 2004, 18, 2195–2224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Sun, S.C. The non-canonical NF-kappaB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef] [PubMed]
  18. Vallabhapurapu, S.; Karin, M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu. Rev. Immunol. 2009, 27, 693–733. [Google Scholar] [CrossRef]
  19. Li, Q.; Verma, I.M. NF-kappaB regulation in the immune system. Nat. Rev. Immunol. 2002, 2, 725–734. [Google Scholar] [CrossRef] [PubMed]
  20. Albensi, B.C. What Is Nuclear Factor Kappa B (NF-kappaB) doing in and to the mitochondrion? Front. Cell Dev. Biol. 2019, 7, 154. [Google Scholar] [CrossRef]
  21. Camandola, S.; Mattson, M.P. NF-kappa B as a therapeutic target in neurodegenerative diseases. Expert Opin. Ther. Targets 2007, 11, 123–132. [Google Scholar] [CrossRef]
  22. Kaltschmidt, B.; Baeuerle, P.A.; Kaltschmidt, C. Potential involvement of the transcription factor NF-kappa B in neurological disorders. Mol. Aspects Med. 1993, 14, 171–190. [Google Scholar] [CrossRef]
  23. Singh, S.; Singh, T.G. Role of nuclear factor kappa B (NF-kappaB) signalling in neurodegenerative diseases: An Mechanistic approach. Curr. Neuropharmacol. 2020, 18, 918–935. [Google Scholar] [CrossRef] [PubMed]
  24. O’Neill, L.A.; Kaltschmidt, C. NF-kappa B: A crucial transcription factor for glial and neuronal cell function. Trends Neurosci. 1997, 20, 252–258. [Google Scholar] [CrossRef]
  25. Zhou, Y.; Cui, C.; Ma, X.; Luo, W.; Zheng, S.G.; Qiu, W. Nuclear Factor kappaB (NF-kappaB)-mediated inflammation in multiple sclerosis. Front. Immunol. 2020, 11, 391. [Google Scholar] [CrossRef]
  26. O’Brien, R.J.; Wong, P.C. Amyloid precursor protein processing and Alzheimer’s disease. Annu. Rev. Neurosci. 2011, 34, 185–204. [Google Scholar] [CrossRef] [Green Version]
  27. Heneka, M.T.; McManus, R.M.; Latz, E. Inflammasome signalling in brain function and neurodegenerative disease. Nat. Rev. Neurosci. 2018, 19, 610–621. [Google Scholar] [CrossRef]
  28. Buggia-Prevot, V.; Sevalle, J.; Rossner, S.; Checler, F. NFkappaB-dependent control of BACE1 promoter transactivation by Abeta42. J. Biol. Chem. 2008, 283, 10037–10047. [Google Scholar] [CrossRef] [Green Version]
  29. Chen, C.H.; Zhou, W.; Liu, S.; Deng, Y.; Cai, F.; Tone, M.; Tone, Y.; Tong, Y.; Song, W. Increased NF-kappaB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int. J. Neuropsychopharmacol. 2012, 15, 77–90. [Google Scholar] [CrossRef] [Green Version]
  30. Bourne, K.Z.; Ferrari, D.C.; Lange-Dohna, C.; Rossner, S.; Wood, T.G.; Perez-Polo, J.R. Differential regulation of BACE1 promoter activity by nuclear factor-kappaB in neurons and glia upon exposure to beta-amyloid peptides. J. Neurosci. Res. 2007, 85, 1194–1204. [Google Scholar] [CrossRef] [PubMed]
  31. Clarke, J.R.; Lyra, E.S.N.M.; Figueiredo, C.P.; Frozza, R.L.; Ledo, J.H.; Beckman, D.; Katashima, C.K.; Razolli, D.; Carvalho, B.M.; Frazao, R.; et al. Alzheimer-associated Abeta oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol. Med. 2015, 7, 190–210. [Google Scholar] [CrossRef]
  32. Kounatidis, I.; Chtarbanova, S.; Cao, Y.; Hayne, M.; Jayanth, D.; Ganetzky, B.; Ligoxygakis, P. NF-kappaB immunity in the brain determines fly lifespan in healthy aging and age-related neurodegeneration. Cell Rep. 2017, 19, 836–848. [Google Scholar] [CrossRef]
  33. Bortolotto, V.; Grilli, M. Every cloud has a silver lining: Proneurogenic effects of abeta oligomers and HMGB-1 via activation of the RAGE-NF-kappaB Axis. CNS Neurol. Disord. Drug Targets 2017, 16, 1066–1079. [Google Scholar] [CrossRef]
  34. Guglielmotto, M.; Aragno, M.; Tamagno, E.; Vercellinatto, I.; Visentin, S.; Medana, C.; Catalano, M.G.; Smith, M.A.; Perry, G.; Danni, O.; et al. AGEs/RAGE complex upregulates BACE1 via NF-kappaB pathway activation. Neurobiol. Aging 2012, 33, e113–e127. [Google Scholar] [CrossRef] [PubMed]
  35. Mattson, M.P.; Camandola, S. NF-kappaB in neuronal plasticity and neurodegenerative disorders. J. Clin. Investig. 2001, 107, 247–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Thawkar, B.S.; Kaur, G. Inhibitors of NF-kappaB and P2X7/NLRP3/Caspase 1 pathway in microglia: Novel therapeutic opportunities in neuroinflammation induced early-stage Alzheimer’s disease. J. Neuroimmunol. 2019, 326, 62–74. [Google Scholar] [CrossRef]
  37. Seo, E.J.; Fischer, N.; Efferth, T. Phytochemicals as inhibitors of NF-kappaB for treatment of Alzheimer’s disease. Pharmacol. Res. 2018, 129, 262–273. [Google Scholar] [CrossRef]
  38. Ellis, R.J.; Olichney, J.M.; Thal, L.J.; Mirra, S.S.; Morris, J.C.; Beekly, D.; Heyman, A. Cerebral amyloid angiopathy in the brains of patients with Alzheimer’s disease: The CERAD experience, Part XV. Neurology 1996, 46, 1592–1596. [Google Scholar] [CrossRef]
  39. Namba, Y.; Tomonaga, M.; Kawasaki, H.; Otomo, E.; Ikeda, K. Apolipoprotein E immunoreactivity in cerebral amyloid deposits and neurofibrillary tangles in Alzheimer’s disease and kuru plaque amyloid in Creutzfeldt-Jakob disease. Brain Res. 1991, 541, 163–166. [Google Scholar] [CrossRef]
  40. Kok, E.; Haikonen, S.; Luoto, T.; Huhtala, H.; Goebeler, S.; Haapasalo, H.; Karhunen, P.J. Apolipoprotein E-dependent accumulation of Alzheimer disease-related lesions begins in middle age. Ann. Neurol. 2009, 65, 650–657. [Google Scholar] [CrossRef] [PubMed]
  41. Polvikoski, T.; Sulkava, R.; Haltia, M.; Kainulainen, K.; Vuorio, A.; Verkkoniemi, A.; Niinisto, L.; Halonen, P.; Kontula, K. Apolipoprotein E, dementia, and cortical deposition of beta-amyloid protein. N. Engl. J. Med. 1995, 333, 1242–1247. [Google Scholar] [CrossRef]
  42. Schmechel, D.E.; Saunders, A.M.; Strittmatter, W.J.; Crain, B.J.; Hulette, C.M.; Joo, S.H.; Pericak-Vance, M.A.; Goldgaber, D.; Roses, A.D. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 9649–9653. [Google Scholar] [CrossRef] [Green Version]
  43. Klunk, W.E.; Engler, H.; Nordberg, A.; Wang, Y.; Blomqvist, G.; Holt, D.P.; Bergstrom, M.; Savitcheva, I.; Huang, G.F.; Estrada, S.; et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann. Neurol. 2004, 55, 306–319. [Google Scholar] [CrossRef] [PubMed]
  44. Barthel, H.; Gertz, H.J.; Dresel, S.; Peters, O.; Bartenstein, P.; Buerger, K.; Hiemeyer, F.; Wittemer-Rump, S.M.; Seibyl, J.; Reininger, C.; et al. Cerebral amyloid-beta PET with florbetaben (18F) in patients with Alzheimer’s disease and healthy controls: A multicentre phase 2 diagnostic study. Lancet Neurol. 2011, 10, 424–435. [Google Scholar] [CrossRef]
  45. Reiman, E.M.; Chen, K.; Liu, X.; Bandy, D.; Yu, M.; Lee, W.; Ayutyanont, N.; Keppler, J.; Reeder, S.A.; Langbaum, J.B.; et al. Fibrillar amyloid-beta burden in cognitively normal people at 3 levels of genetic risk for Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2009, 106, 6820–6825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Fleisher, A.S.; Chen, K.; Liu, X.; Ayutyanont, N.; Roontiva, A.; Thiyyagura, P.; Protas, H.; Joshi, A.D.; Sabbagh, M.; Sadowsky, C.H.; et al. Apolipoprotein E epsilon4 and age effects on florbetapir positron emission tomography in healthy aging and Alzheimer disease. Neurobiol. Aging 2013, 34, 1–12. [Google Scholar] [CrossRef]
  47. Liu, C.C.; Liu, C.C.; Kanekiyo, T.; Xu, H.; Bu, G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat. Rev. Neurol. 2013, 9, 106–118. [Google Scholar] [CrossRef] [Green Version]
  48. Balu, D.; Karstens, A.J.; Loukenas, E.; Maldonado Weng, J.; York, J.M.; Valencia-Olvera, A.C.; LaDu, M.J. The role of APOE in transgenic mouse models of AD. Neurosci. Lett. 2019, 707, 134285. [Google Scholar] [CrossRef]
  49. Chen, X.F.; Zhang, Y.W.; Xu, H.; Bu, G. Transcriptional regulation and its misregulation in Alzheimer’s disease. Mol. Brain 2013, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  50. Mandrekar-Colucci, S.; Landreth, G.E. Nuclear receptors as therapeutic targets for Alzheimer’s disease. Expert Opin. Ther. Targets 2011, 15, 1085–1097. [Google Scholar] [CrossRef]
  51. Zannis, V.I.; Kan, H.Y.; Kritis, A.; Zanni, E.; Kardassis, D. Transcriptional regulation of the human apolipoprotein genes. Front. Biosci. 2001, 6, D456–D504. [Google Scholar] [CrossRef] [PubMed]
  52. Bales, K.R.; Verina, T.; Dodel, R.C.; Du, Y.; Altstiel, L.; Bender, M.; Hyslop, P.; Johnstone, E.M.; Little, S.P.; Cummins, D.J.; et al. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat. Genet. 1997, 17, 263–264. [Google Scholar] [CrossRef]
  53. Bales, K.R.; Liu, F.; Wu, S.; Lin, S.; Koger, D.; DeLong, C.; Hansen, J.C.; Sullivan, P.M.; Paul, S.M. Human APOE isoform-dependent effects on brain beta-amyloid levels in PDAPP transgenic mice. J. Neurosci. 2009, 29, 6771–6779. [Google Scholar] [CrossRef] [PubMed]
  54. Rebeck, G.W. The role of APOE on lipid homeostasis and inflammation in normal brains. J. Lipid. Res. 2017, 58, 1493–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhu, Y.; Nwabuisi-Heath, E.; Dumanis, S.B.; Tai, L.M.; Yu, C.; Rebeck, G.W.; LaDu, M.J. APOE genotype alters glial activation and loss of synaptic markers in mice. Glia 2012, 60, 559–569. [Google Scholar] [CrossRef] [Green Version]
  56. Rodriguez, G.A.; Tai, L.M.; LaDu, M.J.; Rebeck, G.W. Human APOE4 increases microglia reactivity at Abeta plaques in a mouse model of Abeta deposition. J. Neuroinflamm. 2014, 11, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Tu, J.L.; Zhao, C.B.; Vollmer, T.; Coons, S.; Lin, H.J.; Marsh, S.; Treiman, D.M.; Shi, J. APOE 4 polymorphism results in early cognitive deficits in an EAE model. Biochem. Biophys. Res. Commun. 2009, 384, 466–470. [Google Scholar] [CrossRef] [PubMed]
  58. Lynch, J.R.; Tang, W.; Wang, H.; Vitek, M.P.; Bennett, E.R.; Sullivan, P.M.; Warner, D.S.; Laskowitz, D.T. APOE genotype and an ApoE-mimetic peptide modify the systemic and central nervous system inflammatory response. J. Biol. Chem. 2003, 278, 48529–48533. [Google Scholar] [CrossRef] [Green Version]
  59. Pocivavsek, A.; Burns, M.P.; Rebeck, G.W. Low-density lipoprotein receptors regulate microglial inflammation through c-Jun N-terminal kinase. Glia 2009, 57, 444–453. [Google Scholar] [CrossRef] [Green Version]
  60. Lynch, J.R.; Wang, H.; Mace, B.; Leinenweber, S.; Warner, D.S.; Bennett, E.R.; Vitek, M.P.; McKenna, S.; Laskowitz, D.T. A novel therapeutic derived from apolipoprotein E reduces brain inflammation and improves outcome after closed head injury. Exp. Neurol. 2005, 192, 109–116. [Google Scholar] [CrossRef] [PubMed]
  61. Guo, L.; LaDu, M.J.; Van Eldik, L.J. A dual role for apolipoprotein e in neuroinflammation: Anti-and pro-inflammatory activity. J. Mol. Neurosci. 2004, 23, 205–212. [Google Scholar] [CrossRef]
  62. Baitsch, D.; Bock, H.H.; Engel, T.; Telgmann, R.; Muller-Tidow, C.; Varga, G.; Bot, M.; Herz, J.; Robenek, H.; von Eckardstein, A.; et al. Apolipoprotein E induces antiinflammatory phenotype in macrophages. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1160–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Laskowitz, D.T.; Matthew, W.D.; Bennett, E.R.; Schmechel, D.; Herbstreith, M.H.; Goel, S.; McMillian, M.K. Endogenous apolipoprotein E suppresses LPS-stimulated microglial nitric oxide production. Neuroreport 1998, 9, 615–618. [Google Scholar] [CrossRef]
  64. Pocivavsek, A.; Rebeck, G.W. Inhibition of c-Jun N-terminal kinase increases apoE expression in vitro and in vivo. Biochem. Biophys. Res. Commun. 2009, 387, 516–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Goldklang, M.; Golovatch, P.; Zelonina, T.; Trischler, J.; Rabinowitz, D.; Lemaitre, V.; D’Armiento, J. Activation of the TLR4 signaling pathway and abnormal cholesterol efflux lead to emphysema in ApoE-deficient mice. Am. J. Physiol. Lung Cell Mol. Physiol. 2012, 302, L1200–L1208. [Google Scholar] [CrossRef] [Green Version]
  66. Michelsen, K.S.; Doherty, T.M.; Shah, P.K.; Arditi, M. TLR signaling: An emerging bridge from innate immunity to atherogenesis. J. Immunol. 2004, 173, 5901–5907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Malik, M.; Parikh, I.; Vasquez, J.B.; Smith, C.; Tai, L.; Bu, G.; LaDu, M.J.; Fardo, D.W.; Rebeck, G.W.; Estus, S. Genetics ignite focus on microglial inflammation in Alzheimer’s disease. Mol. Neurodegener. 2015, 10, 52. [Google Scholar] [CrossRef] [Green Version]
  68. Ophir, G.; Amariglio, N.; Jacob-Hirsch, J.; Elkon, R.; Rechavi, G.; Michaelson, D.M. Apolipoprotein E4 enhances brain inflammation by modulation of the NF-kappaB signaling cascade. Neurobiol. Dis. 2005, 20, 709–718. [Google Scholar] [CrossRef]
  69. Noguchi, T.; Ebina, K.; Hirao, M.; Otsuru, S.; Guess, A.J.; Kawase, R.; Ohama, T.; Yamashita, S.; Etani, Y.; Okamura, G.; et al. Apolipoprotein E plays crucial roles in maintaining bone mass by promoting osteoblast differentiation via ERK1/2 pathway and by suppressing osteoclast differentiation via c-Fos, NFATc1, and NF-kappaB pathway. Biochem. Biophys. Res. Commun. 2018, 503, 644–650. [Google Scholar] [CrossRef] [PubMed]
  70. Yang, X.; Chen, S.; Shao, Z.; Li, Y.; Wu, H.; Li, X.; Mao, L.; Zhou, Z.; Bai, L.; Mei, X.; et al. Apolipoprotein E deficiency exacerbates spinal cord injury in mice: Inflammatory response and oxidative stress mediated by NF-kappaB signaling pathway. Front. Cell Neurosci. 2018, 12, 142. [Google Scholar] [CrossRef]
  71. Wang, N.; Zhang, X.; Ma, Z.; Niu, J.; Ma, S.; Wenjie, W.; Chen, J. Combination of tanshinone IIA and astragaloside IV attenuate atherosclerotic plaque vulnerability in ApoE(−/−) mice by activating PI3K/AKT signaling and suppressing TRL4/NF-kappaB signaling. Biomed. Pharmacother. 2020, 123, 109729. [Google Scholar] [CrossRef]
  72. Ophir, G.; Mizrahi, L.; Michaelson, D.M. Enhanced activation of NF-kB signaling by apolipoprotein E4. In Advances in Alzheimer’s and Parkinson’s Disease; Springer: Boston, MA, USA, 2008. [Google Scholar]
  73. Teng, Z.; Guo, Z.; Zhong, J.; Cheng, C.; Huang, Z.; Wu, Y.; Tang, S.; Luo, C.; Peng, X.; Wu, H.; et al. ApoE influences the blood-brain barrier through the NF-kappaB/MMP-9 pathway after traumatic brain injury. Sci. Rep. 2017, 7, 6649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, K.J.; Zhang, H.L.; Zhang, X.M.; Zheng, X.Y.; Quezada, H.C.; Zhang, D.; Zhu, J. Apolipoprotein E isoform-specific effects on cytokine and nitric oxide production from mouse Schwann cells after inflammatory stimulation. Neurosci. Lett. 2011, 499, 175–180. [Google Scholar] [CrossRef]
  75. Kemp, S.B.; Carpenter, E.S.; Steele, N.G.; Donahue, K.L.; Nwosu, Z.C.; Pacheco, A.; Velez-Delgado, A.; Menjivar, R.E.; Lima, F.; The, S.; et al. Apolipoprotein E promotes immune suppression in pancreatic cancer through NF-kappaB-mediated production of CXCL1. Cancer Res. 2021, 81, 4305–4318. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, X.; Yang, J.; Lu, T.; Zhan, Z.; Wei, W.; Lyu, X.; Jiang, Y.; Xue, X. The effect of swimming exercise and diet on the hypothalamic inflammation of ApoE−/− mice based on SIRT1-NF-kappaB-GnRH expression. Aging 2020, 12, 11085–11099. [Google Scholar] [CrossRef] [PubMed]
  77. Gu, H.; Tang, C.; Peng, K.; Sun, H.; Yang, Y. Effects of chronic mild stress on the development of atherosclerosis and expression of toll-like receptor 4 signaling pathway in adolescent apolipoprotein E knockout mice. J. Biomed. Biotechnol. 2009, 2009, 613879. [Google Scholar] [CrossRef]
  78. Tang, Y.L.; Jiang, J.H.; Wang, S.; Liu, Z.; Tang, X.Q.; Peng, J.; Yang, Y.Z.; Gu, H.F. TLR4/NF-kappaB signaling contributes to chronic unpredictable mild stress-induced atherosclerosis in ApoE−/− mice. PLoS ONE 2015, 10, e0123685. [Google Scholar] [CrossRef] [Green Version]
  79. Li, Z.; Yang, P.; Feng, B. Effect of atorvastatin on AGEs-induced injury of cerebral cortex via inhibiting NADPH oxidase -NF-kappaB pathway in ApoE(−/−) mice. Mol. Biol. Rep. 2020, 47, 9479–9488. [Google Scholar] [CrossRef]
  80. Han, J.; Chen, D.; Liu, D.; Zhu, Y. Modafinil attenuates inflammation via inhibiting Akt/NF-kappaB pathway in apoE-deficient mouse model of atherosclerosis. Inflammopharmacology 2018, 26, 385–393. [Google Scholar] [CrossRef]
  81. Zlokovic, B.V. Cerebrovascular effects of apolipoprotein E: Implications for Alzheimer disease. JAMA Neurol. 2013, 70, 440–444. [Google Scholar] [CrossRef]
  82. Bell, R.D.; Winkler, E.A.; Singh, I.; Sagare, A.P.; Deane, R.; Wu, Z.; Holtzman, D.M.; Betsholtz, C.; Armulik, A.; Sallstrom, J.; et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 2012, 485, 512–516. [Google Scholar] [CrossRef]
  83. Nigro, P.; Pompilio, G.; Capogrossi, M.C. Cyclophilin A: A key player for human disease. Cell Death Dis. 2013, 4, e888. [Google Scholar] [CrossRef] [Green Version]
  84. Kaplan, A.; Spiller, K.J.; Towne, C.; Kanning, K.C.; Choe, G.T.; Geber, A.; Akay, T.; Aebischer, P.; Henderson, C.E. Neuronal matrix metalloproteinase-9 is a determinant of selective neurodegeneration. Neuron 2014, 81, 333–348. [Google Scholar] [CrossRef] [Green Version]
  85. Montagne, A.; Nikolakopoulou, A.M.; Huuskonen, M.T.; Sagare, A.P.; Lawson, E.J.; Lazic, D.; Rege, S.; Grond, A.; Zuniga, E.; Zlokovic, B.V.; et al. APOE4 accelerates advanced-stage vascular and neurodegenerative disorder in old Alzheimer’s mice via cyclophilin A independently of amyloid-β. Nat. Aging 2021, 1, 506–520. [Google Scholar] [CrossRef]
  86. Safieh, M.; Korczyn, A.D.; Michaelson, D.M. ApoE4: An emerging therapeutic target for Alzheimer’s disease. BMC Med. 2019, 17, 64. [Google Scholar] [CrossRef] [Green Version]
  87. Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Offen, D.; Rabinowitz, R.; Michaelson, D.; Ben-Zur, T. Towards gene-editing treatment for alzheimer’s disease: ApoE4 allele-specific knockout using CRISPR cas9 variant. Cytotherapy 2018, 20, S18. [Google Scholar] [CrossRef]
  89. Jha, N.K.; Jha, S.K.; Kar, R.; Nand, P.; Swati, K.; Goswami, V.K. Nuclear factor-kappa beta as a therapeutic target for Alzheimer’s disease. J. Neurochem. 2019, 150, 113–137. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The proposed pathways involved with the interaction of apolipoprotein E (APOE) and nuclear factor kappa B (NF-κB). APOE4 protein secreted from astrocytes has a low binding affinity with the low-density lipoprotein receptor-related protein 1 (LRP1) and results in an increase in the cyclophilin A (CypA)–NF-κB–matrix metalloproteinase 9 (MMP-9) pathway (as represented by the red line). This also results in blood–brain barrier (BBB) damage via degradation of tight junctions. In contrast, the APOE3 and APOE2 (APOE3/2) protein has a high binding affinity to LRP1, which suppresses the CypA–NF-κB–MMP-9 pathway as represented with the black line. Damage to the BBB is also associated with the accumulation of β-amyloid (Aβ). The weak binding affinity of APOE4 to LRP1 results in reduced Aβ clearance. The high binding affinity of APOE3/2 to LRP1 results in greater Aβ clearance.
Figure 1. The proposed pathways involved with the interaction of apolipoprotein E (APOE) and nuclear factor kappa B (NF-κB). APOE4 protein secreted from astrocytes has a low binding affinity with the low-density lipoprotein receptor-related protein 1 (LRP1) and results in an increase in the cyclophilin A (CypA)–NF-κB–matrix metalloproteinase 9 (MMP-9) pathway (as represented by the red line). This also results in blood–brain barrier (BBB) damage via degradation of tight junctions. In contrast, the APOE3 and APOE2 (APOE3/2) protein has a high binding affinity to LRP1, which suppresses the CypA–NF-κB–MMP-9 pathway as represented with the black line. Damage to the BBB is also associated with the accumulation of β-amyloid (Aβ). The weak binding affinity of APOE4 to LRP1 results in reduced Aβ clearance. The high binding affinity of APOE3/2 to LRP1 results in greater Aβ clearance.
Immuno 01 00027 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Davies, D.A. The Role of APOE and NF-κB in Alzheimer’s Disease. Immuno 2021, 1, 391-399. https://doi.org/10.3390/immuno1040027

AMA Style

Davies DA. The Role of APOE and NF-κB in Alzheimer’s Disease. Immuno. 2021; 1(4):391-399. https://doi.org/10.3390/immuno1040027

Chicago/Turabian Style

Davies, Don A. 2021. "The Role of APOE and NF-κB in Alzheimer’s Disease" Immuno 1, no. 4: 391-399. https://doi.org/10.3390/immuno1040027

Article Metrics

Back to TopTop