Skip to main content
Redox Biology logoLink to Redox Biology
. 2021 Mar 10;41:101938. doi: 10.1016/j.redox.2021.101938

COVID-19 enters the expanding network of apolipoprotein E4-related pathologies

Kalliopi Gkouskou a,b,∗∗, Theodora Vasilogiannakopoulou a, Evangelos Andreakos c, Nikolaos Davanos d, Maria Gazouli a, Despina Sanoudou c,e,f, Aristides G Eliopoulos a,c,f,
PMCID: PMC7943392  PMID: 33730676

Abstract

COVID-19 incidence and case fatality rates (CFR) differ among ethnicities, stimulating efforts to pinpoint genetic factors that could explain these phenomena. In this regard, the multiallelic apolipoprotein E (APOE) gene has recently been interrogated in the UK biobank cohort, demonstrating associations of the APOE ε4/ε4 genotype with COVID-19 severity and mortality. The frequency of the ε4 allele and thus the distribution of APOE ε4/ε4 genotype may differ among populations. We have assessed APOE genotypes in 1638 Greek individuals, based on haplotypes derived from SNP rs7412 and rs429358 and found reduced frequency of ε4/ε4 compared to the British cohort. Herein we discuss this finding in relation to CFR and hypothesize on the potential mechanisms linking APOE ε4/ε4 to severe COVID-19. We postulate that the metabolic deregulation ensued by APOE4, manifested by elevated cholesterol and oxidized lipoprotein levels, may be central to heightened pneumocyte susceptibility to infection and to exaggerated lung inflammation associated with the ε4/ε4 genotype. We also discuss putative dietary and pharmacological approaches for the prevention and management of COVID-19 in APOE ε4/ε4 individuals.

Keywords: COVID-19, SARS-CoV-2, APOE4, ε4/ε4, Genetics, Cholesterol, HDL, Inflammation, Diet, Nutrition, Statins

1. Introduction

Coronavirus disease 2019 (COVID-19) is a respiratory infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Infection is mediated by the proteolytic priming of the viral spike (S) envelope glycoprotein by the cellular transmembrane serine protease 2 (TMPRSS2) and binding to angiotensin-converting enzyme 2 (ACE2) receptor expressed at the plasma membrane of several cell types, including pneumocytes, intestinal epithelial cells, macrophages and endothelial cells [1].

Severe COVID-19 is associated with damage of the alveolar capillaries interfering with the gas exchange function and leading to the development of acute respiratory distress syndrome (ARDS). ARDS is characterized by bilateral pulmonary infiltrates, severe hypoxemia and an aberrant immune response that may culminate to septic shock, multi-organ dysfunction and death. The immunological mechanisms involved in severe COVID-19 are not fully elucidated but it has been shown that SARS-CoV-2 infection may not trigger the conventional route of antiviral immunity: instead of activating first the antiviral response followed by pro-inflammatory processes as a second line of defense, it triggers the pro-inflammatory response long before interferon-mediated antiviral defenses are induced - if at all [2]. Inflammation is likely to be initiated by infected pneumocytes as a result of NLRP3-dependent inflammasome activation and intracellular Toll-like receptor signaling by viral proteins and RNA. This, in turn, induces pneumocyte pyroptosis and release of cytokines and damage-associated molecular patterns (DAMPs) such as HMGB1 and ATP [3] as a result of lytic cell death [4]. DAMPs are recognized by neighboring epithelial cells, endothelial cells and alveolar macrophages, triggering NLRP3-dependent inflammasome activation and other signaling pathways that culminate to the production and secretion of several pro-inflammatory cytokines and chemokines [4]. These molecules attract neutrophils, monocytes, macrophages and T cells to the site of infection, amplifying inflammation and lung tissue damage. The resulting “cytokine storm” ensues systemic effects, aggravating disease symptoms and occasionally leading to multi-organ failure.

The clinical outcomes of SARS-CoV-2 infection are influenced by several factors, related to both the host and the virus. Older age, obesity, diabetes, hypertension, chronic obstructive pulmonary disease (COPD), as well as viral load kinetics are risk factors for the development of severe disease and mortality [1]. These risk factors, however, do not fully explain why some people have mild or no symptoms whereas others develop severe responses. In addition, the case fatality rate (CFR) of COVID-19 differs among ethnicities (Fig. 1). These differences have sparked considerable interest in defining genetic factors impacting clinical outcomes of SARS-CoV-2 infection. Herein we discuss recently published evidence implicating the multiallelic Apolipoprotein E (APOE) gene in COVID-19 severity and mortality [5,6].

Fig. 1.

Fig. 1

Case fatality rates (CFR) of COVID-19, defined as the number of confirmed deaths divided by the number of confirmed cases, in various European countries till November 15, 2020. Data were extracted from the European Centre for Disease Prevention and Control (ECDC) web site, https://www.ecdc.europa.eu/en.

2. Apolipoprotein E in physiology and disease

APOE is a member of the apolipoprotein family that controls organismal lipid and cholesterol homeostasis [7]. APOE participates in cellular cholesterol efflux from hepatic and non-hepatic tissues, cholesterol transport, and clearance of lipoprotein remnants in the plasma [8]. The protein is mainly associated with triglyceride-rich Very Low Density Lipoprotein (VLDL) particles to mediate extra-hepatic lipid supply and, secondarily, with High Density Lipoprotein (HDL) particles for reverse cholesterol transport (RCT), a process that transfers excess cholesterol from non-hepatic tissues back to the liver for excretion. Delivery of lipids to cells by VLDL is mediated via binding of APOE to membrane Low Density Lipoprotein Receptors (LDLR) followed by receptor-mediated lipoprotein endocytosis. Cholesterol efflux entails the transfer of cholesterol to the lipoprotein via ATP-binding cassette transporter ABCA1.

Most of APOE in the plasma is derived from hepatocytes, but 20–40% of total APOE is produced in non-hepatic tissues, including macrophages, adipocytes, glial cells, neurons and some epithelial cell types. Expression patterns are of interest as they may point to potential physiological functions and/or tissue-specific pathological manifestations of APOE deregulation. For example, APOE is detected in several cell types in the lung, including alveolar macrophages, type I and type II alveolar epithelial cells and pulmonary artery smooth muscle cells [9]; studies in the mouse have shown that ablation of apoe results in defects in lung development typified by impaired alveologenesis, increased airway resistance and accelerated loss of lung recoil during aging [10]. Likewise, hepatic ablation of apoe leads to accelerated liver aging [11] and steatohepatitis when mice are fed a high cholesterol diet [12]. APOE also contributes to cholesterol efflux from lipid-laden macrophages (foam cells) reducing formation of lesions in the arterial wall [13,14]. Beyond these important biological functions that directly relate to lipid metabolism, APOE mediates anti-inflammatory and immune-regulatory effects [15,16], in common with other apolipoproteins [17].

Although these knock-out studies in the mouse are informative, they do not fully reflect the complexity of the effects of human APOE. This is because unlike rodents, the APOE gene is polymorphic in humans. The most prominent variants are single nucleotide polymorphisms (SNPs) rs429358 [C/T] and rs7412 [C/T], both in exon 4 of the gene, which is located on chromosome 19q13.2 (Fig. 2). Three haplotypes emerge, ε2, ε3 and ε4 translating to three protein isoforms, E2, E3 and E4, and six combinations of variants, ε2/ε2, ε2/ε3, ε2/ε4, ε3/ε3, ε3/ε4 and ε4/ε4 can be found [18,19]. Among them, ε4/ε4 has attracted most attention because of indisputable epidemiological and experimental evidence that links it to several human pathologies [7].

Fig. 2.

Fig. 2

Schematic representation of the human APOE gene and positions of SNPs rs7412 and rs429358 in exon 4 of APOE. Depending on the SNP combinations, 3 haplotypes arise for each allele termed ε2, ε3, ε4, resulting in different amino acid combinations at residues 112 and 158.

Indeed, the APOE ε4 variant is a genetic risk factor for cardiovascular disease (CVD) and E4 expression is associated with increased carotid intima-media thickness and higher total blood and LDL-bound cholesterol [20]. In fact, variation at the APOE locus accounts for ≈7% of the population variance in total and LDL-cholesterol concentrations [21]. The ε4/ε4 genotype is also linked to both sporadic and familial late onset Alzheimer's disease (AD), reportedly increasing disease risk by up to tenfold, compared to ε3/ε3, whereas ε2/ε2 is associated with reduced risk [22]. A recent phenome-wide association study of APOE genotypes with 950 disease outcomes registered in the UK biobank confirmed the elevated risk of cardiovascular diseases, AD and hypercholesterolaemia associated with ε4 (ε3/ε4 or ε4/ε4) compared to the ε3/ε3 genotype [23].

APOE is also linked to cellular and organismal responses to infection by viruses and pathogenic microorganisms. Notably, the APOE ε4/ε4 genotype is associated with increased susceptibility to HIV-1 infection and aggravated disease course of AIDS [24], and to susceptibility for HSV-1 related herpes labialis and neuronal invasiveness of HSV-1 compared to other APOE variants [25].

3. A new association of APOE with COVID-19 clinical outcomes

Two recent studies have added COVID-19 to the panel of pathologies associated with the APOE ε4/ε4 genotype [5,6]. Kuo et al. analyzed genetic and clinical data registered in the UK Biobank (>450,000 European-ancestry participants) and uncovered a 2-fold higher risk of severe COVID-19 of people carrying the ε4/ε4 versus the ε3/ε3 genotype [6] and a 4-fold increase in mortality after testing positive for COVID-19 [5]. These associations were independent of preexisting dementia, cardiovascular disease and type-2 diabetes.

Given previous evidence suggesting that the frequency of ε4/ε4 carriers may vary among populations [26], the findings by Kuo et al. could, potentially, also explain some of the reported differences in SARS-CoV-2 disease outcomes in different ethnicities. The lower CFR in Greece compared to the UK which is rated among the European countries with the highest COVID-19 mortality (Fig. 1), prompted us to compare the frequencies of APOE ε4/ε4 genotype in Greeks versus British. We assessed APOE genotypes in 1638 Greek individuals, based on haplotypes derived from SNPs rs7412 and rs429358 and compared the results with the APOE genotyping data extracted from the UK biobank, as reported elsewhere [23]. The distribution of APOE genotypes was found to significantly differ between the two populations (Table 1). Of note, ε4/ε4 is found in 2.42% of European ancestry British but only in 1% of the Greek population cohort with the frequency of the ε4 allele being 0.1563 and 0.0919, respectively (Table 2). Our data is in agreement with and largely extend a previously reported ε4 frequency of 0.1020 in a cohort of 240 healthy middle-age Greeks based on a PCR-RFLP method [27]. These observations warrant further studies to confirm the findings of the UK Biobank [5,6] in other populations. Further investigations are also needed to dissect the biological mechanisms linking APOE genotypes to COVID-19 severity.

Table 1.

APOE genotypes were determined in 1638 individuals across Greece, based on haplotypes derived from SNPs rs7412 and rs429358, using Taqman-based assays on a StepOnePlusTM real time PCR engine (all from Applied Biosystems, USA). The percentage (%) distribution and number of individuals per APOE genotype (count) are shown. The data concerning European descent British were extracted from Ref. [23].

APOE Genotype COUNT
% distribution
BRITISH GREEK BRITISH GREEK
ε4ε4 8179 20 2,42% 1,00%
ε3ε4 80499 242 23,85% 15,00%
ε2ε4 8616 19 2,55% 1,00%
ε3ε3 196306 1203 58,17% 73,00%
ε2ε3 41695 142 12,36% 9,00%
ε2ε2 2172 12 0,64% 1,00%
Total 337467 1638 100% 100%

Table 2.

Allele frequency was calculated on the basis of the analysis described in Table 1 and of the data concerning European descent British, extracted from Ref. [23].

Allele, %
ε2 ε3 ε4
GREEK 5,65 85,16 9,19
BRITISH 8,10 76,28 15,63

4. Mechanisms that may underpin the association of APOE with the clinical outcomes of COVID-19

Scrutinizing some of the known molecular roles of different APOE isoforms provides putative mechanistic clues into the association of APOE ε4/ε4 with COVID-19 severity. Cholesterol may be central to it (Fig. 3).

Fig. 3.

Fig. 3

Graphical representation of the main mechanisms by which APOE E4/E4 may impact COVID-19 severity and mortality. The APOE ε4/ε4 genotype is associated with elevated levels of circulating and tissue cholesterol and oxidized LDL (oxLDL). Their intracellular accumulation in pneumocytes, lung macrophages and endothelial cells increases the density of ACE2/TMPRSS2 in ceramide and cholesterol-enriched membrane microdomains (lipid rafts), resulting in heightened susceptibility to SARS-CoV-2 infection (right panel) relative to ε3/ε3 cells (left panel). This, in turn, leads to heightened NLRP3 inflammasome activation, pyroptosis and release of DAMPs. We also postulate that the elevated levels of oxLDL in ε4/ε4 carriers result in basal inflammasome activation in alveolar and recruited monocyte-derived macrophages through LOX-1 – mediated oxLDL internalization, production of reactive oxygen species (ROS) and the generation of oxidized lipids and cholesterol crystals which serve as endogenous DAMPs. These macrophages are thus subject to inflammasome hyperactivation and to amplified pyroptosis in response to lung infection by SARS-CoV-2, unleashing a “cytokine storm” that ensues recruitment of immune cells, pulmonary edema and severe systemic effects.

4.1. APOE4 may influence SARS-COV-2 infectivity by modulating intracellular cholesterol levels

APOE isoforms differentially affect intracellular and circulating cholesterol levels. A meta-analysis of 14,799 individuals from 17 different ethnicities has confirmed that carriers of the ε4 allele have higher levels of plasma cholesterol than individuals with the ε3/ε3 genotype [28], a phenomenon that is mirrored in the APOE4 targeted replacement mouse fed a western type diet [29].

Chronic administration of high-fat diet increases cholesterol levels in mouse lung tissue by 40% [30] which is exaggerated in the absence of apoe [31] and most likely reflects changes in the circulating levels of cholesterol. Indeed, lipid tracing experiments in rodents have shown that approximately 83% of lung cholesterol is derived from the plasma with the remainder coming from synthesis by lung-resident cells [32]. It is thus likely that beyond elevated cholesterol levels in the plasma, the ε4 allele may also be associated with excess cholesterol in pneumocytes.

What could be the consequences of elevated intracellular cholesterol levels for COVID-19? Several observational studies have shown that the severity of COVID-19 is associated with reduced serum HDL which indicates defects in the reverse cholesterol transport (RCT) pathway responsible for lipid homeostasis in peripheral tissues [33,34]. Along these lines, abnormal pulmonary accumulation of lipid-laden macrophages has been noted in COVID-19 [35]. Intracellular cholesterol also accumulates with age in several tissues [36,37], including pneumocytes [38], which correlates with increased COVID-19 severity in older people.

Beyond these correlations, a recent pre-print report directly links high cellular cholesterol to increased SARS-CoV-2 infectivity [30]. Loading cells with cholesterol increases ACE2 trafficking to the endocytic entry site and doubles the endocytic entry points of the virus [30]. Cholesterol may also impact the production of more infectious virions with improved binding to the ACE2 receptor [30]. The spike (S) glycoprotein of SARS-CoV-2 contains the sequence PRRAR which is cleaved by furin proteases that are abundant in the respiratory tract. Cholesterol optimally positions the S glycoprotein furin cleavage site for proteolysis upon exit from epithelial cells and consequently the virus may infect other cells with increased efficiency [30]. It has also been reported that compared with ε3/ε3 subjects, monocytes from APOE ε3/ε4 individuals display an increase in cholesterol-enriched membrane microdomains (lipid rafts) [39] which represent sites for initial binding, internalization and cell-to-cell transmission of SARS-CoV-2 [40]. Therefore, APOE4 may influence SARS-CoV-2 infectivity by affecting intracellular cholesterol levels.

4.2. APOE4 may enhance SARS-CoV-2 induced inflammation by priming the NLRP3 inflammasome

In humans, APOE4 has been associated with a heightened innate immune response to bacteremia [41], including higher production of pro-inflammatory cytokines IL-6 and TNF by activated macrophages [42], prothrombotic properties of endothelial cells [43], as well as neuroinflammation characterized by increased expression of CCL3 in the brain [44]. Although a detailed characterization of the cytokines and chemokines affected by APOE4 in different pathological conditions and tissues is still missing, IL-6, TNF and CCL3 represent some of the cytokine storm markers of severe COVID-19 [4,39]. Moreover, whilst the mechanisms underlying these APOE4 effects are not entirely understood, we hypothesize that deregulated lipid metabolic processes may bridge APOE4 with heightened pro-inflammatory responses to SARS-CoV-2.

The exaggerated inflammation observed in severe COVID-19 has largely been attributed to over-activation of NLRP3 inflammasome that leads to overproduction of IL-1β, a clinical target for COVID-19. Currently, cholesterol and fatty acids are emerging as important regulators of NLRP3 signaling through various routes [45,46]. It has been shown that saturated fatty acids palmitic and stearic undergo intracellular crystallization in lipid-laden macrophages and activate the NLRP3 inflammasome in vitro and in vivo by serving as endogenous DAMPs [47]. Cholesterol crystals, detected in the lungs of heavy smokers [48], idiopathic pulmonary fibrosis [49] and lipoid pneumonia [50], are also potent inducers of NLRP3 [51]. It could thus be envisaged that by exaggerating cholesterol accumulation in the lung [30], APOE4 may prime epithelial cells, macrophages and endothelial cells towards heightened inflammasome activation in response to SARS-CoV-2 infection (Fig. 3). This concept is compatible with the higher basal activity of NLRP3 in mouse models and patients with obesity [52,53] that constitutes a risk factor for severe COVID-19. Likewise, inflammasome activation parallels the low-grade inflammation associated with ageing [54], which also constitutes a risk factor for severe COVID-19 [1,54]. In addition to functioning as intracellular DAMPs, cholesterol crystals trigger neutrophils to release neutrophil extracellular traps (NETs) which are found in cholesterol-rich atherosclerotic plaques in apoe−/- mice and prime macrophages to release cytokines [55]. Notably, elevated serum markers of NETosis are observed in severe cases of COVID-19 [56].

Cholesterol also impacts T cell receptor (TcR) signaling through membrane raft formation that enables TcR signal initiation, higher sensitivity to TcR ligands and T cell proliferation [57]. Intracellular sterols also promote T cell differentiation towards Th17 [58]. This differentiation may explain the increased numbers of Th17 cells in high cholesterol-fed atherosclerotic apoe−/- mice [59]. Interestingly, humans expressing the APOE4 isoform (ε3/4, ε4/4) have increased circulating numbers of activated T cells [60] and patients affected by COVID-19 pneumonia display marked T cell activation and skewing toward the Th17 phenotype [61]. Based on these observations we speculate that the metabolic deregulation ensued by APOE4 may prime pneumocytes, macrophages and endothelial cells towards heightened inflammasome activation and cytokine production and amplify tissue and systemic effects of SARS-CoV-2 on T cell numbers and differentiation (Fig. 3).

4.3. APOE4 may enhance the pathogenic effects of SARS-CoV-2 through LDL lipid oxidation

LDL oxidation is caused by enzymatic or non-enzymatic oxidative modifications of LDL lipids and apolipoproteins [62]. Lipid oxidation is driven by excessive production of reactive oxygen species (ROS) that are generated as a result of vascular oxidative stress in hypercholesterolemia, diabetes mellitus, hypertension, smoking and other pathologies [63]. The end product of lipid oxidation is the formation of oxidized LDL (oxLDL) that contains several hundred bioactive phospholipid, triglyceride and cholesterol products [62].

OxLDL is a key pathogenic factor in the development of atherosclerosis, because it is recognized and taken up by scavenger receptors expressed in macrophages leading to the formation of foam cells that are found in atherosclerotic lesions [63]. OxLDL also exerts pro-inflammatory properties by binding to lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) and the heterotrimer of CD36/TLR4/TLR6 expressed on monocytes/macrophages and endothelial cells. The binding of oxLDL to LOX-1 results in its internalization, causing intracellular lipid accumulation and the activation of several pro-inflammatory and pro-oxidative pathways [62]. For example, oxLDL has been shown to stimulate the production of ROS and of pro-inflammatory lipids, including isoprostanes generated through free radical-induced peroxidation of arachidonic acid [64]. Oxidized phospholipids (oxPLs) are a major component of oxLDL and serve as DAMPs that activate NLRP3, leading to the secretion of IL-1β/IL-18 [65]. The relevance of LDL oxidation to lung diseases is highlighted by the observation that intranasal administration of human oxLDL to mice initiates an inflammatory response that mirrors the characteristics of cigarette smoke-induced lung inflammation [60].

Several lines of evidence indicate a possible link between oxLDL and COVID-19. First, beyond atherosclerosis, elevated levels of circulating oxLDL are detected in obesity, metabolic syndrome and COPD [[66], [67], [68], [69]], all of which are risk factors for severe COVID-19 [1]. Second, the pro-inflammatory pathways triggered by oxLDL have common elements with those activated by SARS-CoV-2, and involve DAMP-orchestrated inflammation. Third, oxLDL increases the expression levels of ACE2 and TMPRSS2 [70] and the density of cholesterol and ceramide-enriched lipid rafts [71] that contribute to enhanced virus infectivity [40,72]. Finally, oxPLs are detected in the inflammatory exudates overlaying pneumocytes and macrophages in the injured air spaces of patients infected with SARS-CoV [73]. Based on these observations, we hypothesize that oxLDL may increase host cell susceptibility to the pathogenic effects of SARS-CoV-2.

APOE confers protection against oxidative stress in the lung. Ablation of apoe in the mouse exacerbates acute lung injury, causing an ARDS-like condition due to elevated oxLDL and IL-6 levels [74]. APOE4 has the least effective antioxidant properties compared with other APOE isoforms [75] and is less effective in inhibiting LDL oxidation in vitro [76,77]. APOE4 is also positively associated with markers of oxidative stress. Thus, APOE ε4/ε3 individuals have elevated levels of lipid peroxides [78], and smokers who are carriers of the ε4 allele have a 26.7% increase in the serum levels of oxLDL compared to other APOE genotypes [79]. Collectively, these findings suggest that APOE4 is linked to elevated levels of oxLDL, particularly under conditions of chronic or acute inflammation, which may amplify susceptibility to SARS-CoV-2 infection in the lung.

5. Putative pharmacological and dietary approaches for the prevention and management of COVID-19 in APOE ε4/ε4 individuals

We have herein reasoned that elevated cholesterol and oxidized lipoproteins may serve as important mediators of the detrimental effects of APOE ε4/ε4 genotype on the severity and mortality of COVID-19. Thus, it might be possible to mitigate the consequences of COVID-19 infection by pharmacological and dietary strategies that reduce cholesterol burden in individuals with the APOE ε4/ε4 genotype.

Statins are cholesterol-lowering drugs targeting HMG CoA reductase, the enzyme controlling the rate-limiting step of cholesterol biosynthesis pathway. Administration of statins, in addition to the reduction in LDL-cholesterol, also reduces circulating oxLDL [80,81]. Statins have demonstrated beneficial effects on several viral-induced pathologies, including a significantly reduced risk of influenza death among moderate-dose statin users [82], improved outcomes of anti-retroviral therapy in HIV-positive patients [83] and reduced mortality of Ebola virus disease patients [84]. Notwithstanding the pleiotropic functions of statins that include systemic immunomodulatory properties, a direct in vitro effect of lovastatin on reducing production of infectious Ebola virus has been reported [85].

The clinical utility of statins for the management of COVID-19 has been debated [86]. However, a recently published meta-analysis of 4 clinical studies involving a total of 8990 COVID-19 patients concluded that statins reduce the hazard for severe or fatal COVID-19 cases [87]. There have been some concerns that treatment with statins may increase the risk of type 2 diabetes, especially in obese patients [88]. Notably, although statin users without the APOE ε4 risk allele have greater insulin resistance, APOE ε4 carriers have improved insulin function [89]. It would thus be of interest to examine if the beneficial effects of statins are more pronounced in ε4/ε4 COVID-19 patients compared to other APOE genotypes. Biological agents such as APOE mimetic peptides and antibodies that neutralize oxPLs present in oxLDL are being evaluated in the context of atherosclerosis [90] and may offer additional opportunities for the management of ε4/ε4 COVID-19 patients.

The APOE4 isoform is considered generally insensitive to dietary interventions. For example, plant sterols reduce blood cholesterol levels in hypercholesterolemic patients with APOE ε3/ε3 genotype but not ε4 carriers [91]. However, specific dietary plans for APOE4 isoform carriers are both feasible and effective [92]. Thus, a 24 week diet that replaces saturated with monounsaturated fats or with low glycaemic index carbohydrates has been found to benefit APOE4 carriers to a greater extent compared to other APOE isoforms in reducing plasma cholesterol [93]. A similar hypotriacylglycerolaemic effect specifically on APOE4 carriers has been reported following a 4 week healthy diet supplemented with daily consumption of two kiwifruit presumably by increasing intake of lipid-lowering anti-oxidant vitamins and polyphenols [94].

Chronic consumption of fish-derived omega-3 polyunsaturated fatty acids and of docosahexaenoic acid in the form of lysophosphatidylcholine have also been reported to benefit APOE ε4 carriers by lowering their blood triglyceride concentrations [92] and possibly promoting resolution of inflammation through the production of proresolving lipid mediators such as resolvins and protectins [95]. The potential benefit of this dietary approach for modulation of lung inflammation is illustrated by experimental data showing that accumulation of pro-inflammatory eicosanoids in the lungs of APOE-deficient mice fed a diet with reduced omega-3 fatty acids is reversed by supplementation with omega-3 fatty acids [96]. Interestingly, dexamethasone which now constitutes standard care for the treatment of severe COVID-19 patients in need of oxygen support or mechanical ventilation [97], has also been shown to induce proresolving lipid mediators such as protectin D1 [98,99]. APOE ε4 carriers may also have a slightly increased demand for vitamin E [91,100], which confers anti-oxidant and immunoregulatory effects that are beneficial to viral respiratory tract infections [101].

6. Discussion

APOE is a multifaceted protein that has been implicated in several pathologies and displays isoform-dependent effects. Homozygous carriers of the E4 isoform are particularly vulnerable to cardiovascular disease, hypercholesterolaemia, stroke, Alzheimer's disease and virus-induced pathologies. COVID-19 has recently joined the network of APOE ε4-related diseases with a significant association between APOE ε4/ε4 genotype and COVID-19 severity and mortality [5,6]. Recent experimental evidence supports the heightened sensitivity of ε4/ε4 neurons and astrocytes to SARS-CoV-2 infection [102].

The frequency of the ε4 allele and thus the distribution of APOE ε4/ε4 genotype may differ among ethnicities [103]. Data presented herein demonstrate that the frequency of ε4/ε4 genotype in a Greek cohort is significantly lower than in European ancestry UK biobank British cohort, which parallels the lower COVID-19 CFR in Greece as compared to the UK. Notwithstanding the complexity of responses to SARS-CoV-2 that are likely to be influenced by a multitude of host, virus and environmental factors, this observation suggests that putative associations between the genetic make-up of ethnic groups with the incidence, severity and mortality of SARS-CoV-2 infection should be explored, along the lines of a recently published GWAS [104].

Further investigations are also needed for the elucidation of the biological mechanisms linking APOE genotypes to COVID-19 severity. Increased levels of intracellular and systemic cholesterol typify the metabolic function of APOE E4/E4 [28], and may be central to this association (Fig. 3). We propose that the accumulation of cholesterol and oxLDL in pneumocytes ensued by ε4 homozygocity may result in greater susceptibility and severity to SARS-CoV-2 infection. This is likely to be caused by quantitative and qualitative changes in lipid rafts and a high oxidative status in the cells lining the lung airways. Under these conditions, there is increased expression of ACE2 and TMPRSS2 at the plasma membrane that leads to increased virus binding, internalization and cell-to-cell transmission (Fig. 3). Beyond SARS-CoV-2, lipid rafts play a prominent role as the entry points of several viruses [105], including HIV-1 the infectious cycle of which is also influenced by the APOE ε4/ε4 genotype [24].

Compared to other isoforms, APOE4 may promote basal NLRP3 inflammasome activation that exaggerates inflammatory responses in the lung, pertinent to the “cytokine storm” of severe COVID-19 (Fig. 3). The detrimental clinical effects of APOE4 on COVID-19 [5,6] may depend on or be amplified by the co-occurrence of additional factors, such as smoking, obesity, asthma or COPD, as previously recorded for CVD risk [79,106].

In favor of the aforementioned model implicating lipid metabolism at the core of the APOE4 – COVID-19 connection, statins provide clinical benefit to COVID-19 patients. We reason that APOE ε4/ε4 individuals, who are at increased risk of severe COVID-19, could particularly benefit from preventive dietary changes and/or therapeutic use of moderate-doses of statins. The major influence of APOE4 on lipid metabolism, however, does not exclude the possibility that other APOE4 functions in the lung impact the clinical outcome of SARS-CoV-2 infection. In this regard, the APOE ε4 allele has been associated with reduced lung respiratory capacity in the elderly irrespective of lipid levels [107], and apoe ablation in the mouse is associated with the development of severe pulmonary hypertension [108], which increases the risk of death in COVID-19 patients.

Since its discovery in 1973 by Havel and Kane [109], APOE continues to surprise us with new associations with several diseases and represents a fertile ground for future research.

Author contributions

Conceptualization, K.G. and A.G.E.; methodology, materials and formal analysis, K.G, T.V., N.D., M.G., D.S.; writing—original draft preparation, A.G.E. and D.S.; writing—review and editing, all authors; funding acquisition, D.S. All authors have read and agreed to the published version of the manuscript.

Funding

CURE-PLaN, a grant from the Leducq Foundation for Cardiovascular Research.

Declaration of competing interest

The authors declare no conflict of interest with the exception of K. Gkouskou who is founder and CEO of Embiodiagnostics S.A.

Contributor Information

Kalliopi Gkouskou, Email: gkouskoukal@med.uoa.gr.

Aristides G. Eliopoulos, Email: eliopag@med.uoa.gr.

References

  • 1.Cevik M., Kuppalli K., Kindrachuk J., Peiris M. Virology, transmission, and pathogenesis of SARS-CoV-2. BMJ. 2020;371:m3862. doi: 10.1136/bmj.m3862. [DOI] [PubMed] [Google Scholar]
  • 2.Galani I.E., Rovina N., Lampropoulou V., Triantafyllia V., Manioudaki M., Pavlos E., Koukaki E., Fragkou P.C., Panou V., Rapti V., Koltsida O., Mentis A., Koulouris N., Tsiodras S., Koutsoukou A., Andreakos E. Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 2021;22(1):32–40. doi: 10.1038/s41590-020-00840-x. [DOI] [PubMed] [Google Scholar]
  • 3.Kepp O., Senovilla L., Vitale I., Vacchelli E., Adjemian S., Agostinis P., Apetoh L., Aranda F., Barnaba V., Bloy N., Bracci L., Breckpot K., Brough D., Buque A., Castro M.G., Cirone M., Colombo M.I., Cremer I., Demaria S., Dini L., Eliopoulos A.G., Faggioni A., Formenti S.C., Fucikova J., Gabriele L., Gaipl U.S., Galon J., Garg A., Ghiringhelli F., Giese N.A., Guo Z.S., Hemminki A., Herrmann M., Hodge J.W., Holdenrieder S., Honeychurch J., Hu H.M., Huang X., Illidge T.M., Kono K., Korbelik M., Krysko D.V., Loi S., Lowenstein P.R., Lugli E., Ma Y., Madeo F., Manfredi A.A., Martins I., Mavilio D., Menger L., Merendino N., Michaud M., Mignot G., Mossman K.L., Multhoff G., Oehler R., Palombo F., Panaretakis T., Pol J., Proietti E., Ricci J.E., Riganti C., Rovere-Querini P., Rubartelli A., Sistigu A., Smyth M.J., Sonnemann J., Spisek R., Stagg J., Sukkurwala A.Q., Tartour E., Thorburn A., Thorne S.H., Vandenabeele P., Velotti F., Workenhe S.T., Yang H., Zong W.X., Zitvogel L., Kroemer G., Galluzzi L. Consensus guidelines for the detection of immunogenic cell death. OncoImmunology. 2014;3(9) doi: 10.4161/21624011.2014.955691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tay M.Z., Poh C.M., Renia L., MacAry P.A., Ng L.F.P. The trinity of COVID-19: immunity, inflammation and intervention. Nat. Rev. Immunol. 2020;20(6):363–374. doi: 10.1038/s41577-020-0311-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kuo C.L., Pilling L.C., Atkins J.L., Masoli J.A.H., Delgado J., Kuchel G.A., Melzer D. ApoE e4e4 Genotype and Mortality With COVID-19 in UK Biobank. J Gerontol A Biol Sci Med Sci. 2020;75(9):1801–1803. doi: 10.1093/gerona/glaa169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kuo C.L., Pilling L.C., Atkins J.L., Masoli J.A.H., Delgado J., Kuchel G.A., Melzer D. APOE e4 Genotype Predicts Severe COVID-19 in the UK Biobank Community Cohort. J Gerontol A Biol Sci Med Sci. 2020;75(11):2231–2232. doi: 10.1093/gerona/glaa131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Martinez-Martinez A.B., Torres-Perez E., Devanney N., Del Moral R., Johnson L.A., Arbones-Mainar J.M. Beyond the CNS: the many peripheral roles of APOE. Neurobiol. Dis. 2020;138 doi: 10.1016/j.nbd.2020.104809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Marais A.D. Apolipoprotein E in lipoprotein metabolism, health and cardiovascular disease. Pathology. 2019;51(2):165–176. doi: 10.1016/j.pathol.2018.11.002. [DOI] [PubMed] [Google Scholar]
  • 9.Lin C.T., Xu Y.F., Wu J.Y., Chan L. Immunoreactive apolipoprotein E is a widely distributed cellular protein. Immunohistochemical localization of apolipoprotein E in baboon tissues. J. Clin. Invest. 1986;78(4):947–958. doi: 10.1172/JCI112685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Massaro D., Massaro G.D. Apoetm1Unc mice have impaired alveologenesis, low lung function, and rapid loss of lung function. Am. J. Physiol. Lung Cell Mol. Physiol. 2008;294(5):L991–L997. doi: 10.1152/ajplung.00013.2008. [DOI] [PubMed] [Google Scholar]
  • 11.Bonomini F., Rodella L.F., Moghadasian M., Lonati C., Rezzani R. Apolipoprotein E deficiency and a mouse model of accelerated liver aging. Biogerontology. 2013;14(2):209–220. doi: 10.1007/s10522-013-9424-9. [DOI] [PubMed] [Google Scholar]
  • 12.Tous M., Ferre N., Camps J., Riu F., Joven J. Feeding apolipoprotein E-knockout mice with cholesterol and fat enriched diets may be a model of non-alcoholic steatohepatitis. Mol. Cell. Biochem. 2005;268(1–2):53–58. doi: 10.1007/s11010-005-2997-0. [DOI] [PubMed] [Google Scholar]
  • 13.Hasty A.H., Linton M.F., Brandt S.J., Babaev V.R., Gleaves L.A., Fazio S. Retroviral gene therapy in ApoE-deficient mice: ApoE expression in the artery wall reduces early foam cell lesion formation. Circulation. 1999;99(19):2571–2576. doi: 10.1161/01.cir.99.19.2571. [DOI] [PubMed] [Google Scholar]
  • 14.Bellosta S., Mahley R.W., Sanan D.A., Murata J., Newland D.L., Taylor J.M., Pitas R.E. Macrophage-specific expression of human apolipoprotein E reduces atherosclerosis in hypercholesterolemic apolipoprotein E-null mice. J. Clin. Invest. 1995;96(5):2170–2179. doi: 10.1172/JCI118271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Baitsch D., Bock H.H., Engel T., Telgmann R., Muller-Tidow C., Varga G., Bot M., Herz J., Robenek H., von Eckardstein A., Nofer J.R. Apolipoprotein E induces antiinflammatory phenotype in macrophages. Arterioscler. Thromb. Vasc. Biol. 2011;31(5):1160–1168. doi: 10.1161/ATVBAHA.111.222745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yin C., Ackermann S., Ma Z., Mohanta S.K., Zhang C., Li Y., Nietzsche S., Westermann M., Peng L., Hu D., Bontha S.V., Srikakulapu P., Beer M., Megens R.T.A., Steffens S., Hildner M., Halder L.D., Eckstein H.H., Pelisek J., Herms J., Roeber S., Arzberger T., Borodovsky A., Habenicht L., Binder C.J., Weber C., Zipfel P.F., Skerka C., Habenicht A.J.R. ApoE attenuates unresolvable inflammation by complex formation with activated C1q. Nat. Med. 2019;25(3):496–506. doi: 10.1038/s41591-018-0336-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gkouskou K.K., Ioannou M., Pavlopoulos G.A., Georgila K., Siganou A., Nikolaidis G., Kanellis D.C., Moore S., Papadakis K.A., Kardassis D., Iliopoulos I., McDyer F.A., Drakos E., Eliopoulos A.G. Apolipoprotein A-I inhibits experimental colitis and colitis-propelled carcinogenesis. Oncogene. 2016;35(19):2496–2505. doi: 10.1038/onc.2015.307. [DOI] [PubMed] [Google Scholar]
  • 18.Zannis V.I., Breslow J.L. Human very low density lipoprotein apolipoprotein E isoprotein polymorphism is explained by genetic variation and posttranslational modification. Biochemistry. 1981;20(4):1033–1041. doi: 10.1021/bi00507a059. [DOI] [PubMed] [Google Scholar]
  • 19.Zannis V.I., Just P.W., Breslow J.L. Human apolipoprotein E isoprotein subclasses are genetically determined. Am. J. Hum. Genet. 1981;33(1):11–24. [PMC free article] [PubMed] [Google Scholar]
  • 20.Graner M., Kahri J., Varpula M., Salonen R.M., Nyyssonen K., Jauhiainen M., Nieminen M.S., Syvanne M., Taskinen M.R. Apolipoprotein E polymorphism is associated with both carotid and coronary atherosclerosis in patients with coronary artery disease. Nutr. Metabol. Cardiovasc. Dis. 2008;18(4):271–277. doi: 10.1016/j.numecd.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 21.Wilson P.W., Schaefer E.J., Larson M.G., Ordovas J.M. Apolipoprotein E alleles and risk of coronary disease. A meta-analysis. Arterioscler. Thromb. Vasc. Biol. 1996;16(10):1250–1255. doi: 10.1161/01.atv.16.10.1250. [DOI] [PubMed] [Google Scholar]
  • 22.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(2):106–118. doi: 10.1038/nrneurol.2012.263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lumsden A.L., Mulugeta A., Zhou A., Hypponen E., Apolipoprotein E.(A.P.O.E. genotype-associated disease risks: a phenome-wide, registry-based, case-control study utilising the UK Biobank. EBioMedicine. 2020;59 doi: 10.1016/j.ebiom.2020.102954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Burt T.D., Agan B.K., Marconi V.C., He W., Kulkarni H., Mold J.E., Cavrois M., Huang Y., Mahley R.W., Dolan M.J., McCune J.M., Ahuja S.K. Apolipoprotein (apo) E4 enhances HIV-1 cell entry in vitro, and the APOE epsilon4/epsilon4 genotype accelerates HIV disease progression. Proc. Natl. Acad. Sci. U. S. A. 2008;105(25):8718–8723. doi: 10.1073/pnas.0803526105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Itzhaki R.F., Wozniak M., Dobson C., Lin W.R. ApoE-viral interactions. Nat. Med. 1998;4(12):1344. doi: 10.1038/3908. [DOI] [PubMed] [Google Scholar]
  • 26.Howard B.V., Gidding S.S., Liu K. Association of apolipoprotein E phenotype with plasma lipoproteins in african-American and white young adults. The CARDIA study. Coronary artery risk development in young adults. Am. J. Epidemiol. 1998;148(9):859–868. doi: 10.1093/oxfordjournals.aje.a009711. [DOI] [PubMed] [Google Scholar]
  • 27.Kolovou G., Yiannakouris N., Hatzivassiliou M., Malakos J., Daskalova D., Hatzigeorgiou G., Cariolou M.A., Cokkinos D.V. Association of apolipoprotein E polymorphism with myocardial infarction in Greek patients with coronary artery disease. Curr. Med. Res. Opin. 2002;18(3):118–124. doi: 10.1185/030079902125000444. [DOI] [PubMed] [Google Scholar]
  • 28.Dallongeville J., Lussier-Cacan S., Davignon J. Modulation of plasma triglyceride levels by apoE phenotype: a meta-analysis. J. Lipid Res. 1992;33(4):447–454. [PubMed] [Google Scholar]
  • 29.Knouff C., Hinsdale M.E., Mezdour H., Altenburg M.K., Watanabe M., Quarfordt S.H., Sullivan P.M., Maeda N. Apo E structure determines VLDL clearance and atherosclerosis risk in mice. J. Clin. Invest. 1999;103(11):1579–1586. doi: 10.1172/JCI6172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang H., Yuan Z., Pavel M.A., Hansen S.B. The role of high cholesterol in age-related COVID19 lethality. bioRxiv. 2020 [Google Scholar]
  • 31.Ouyang Q., Huang Z., Lin H., Ni J., Lu H., Chen X., Wang Z., Lin L. Apolipoprotein E deficiency and high-fat diet cooperate to trigger lipidosis and inflammation in the lung via the toll-like receptor 4 pathway. Mol. Med. Rep. 2015;12(2):2589–2597. doi: 10.3892/mmr.2015.3774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Turley S.D., Andersen J.M., Dietschy J.M. Rates of sterol synthesis and uptake in the major organs of the rat in vivo. J. Lipid Res. 1981;22(4):551–569. [PubMed] [Google Scholar]
  • 33.Wang G., Zhang Q., Zhao X., Dong H., Wu C., Wu F., Yu B., Lv J., Zhang S., Wu G., Wu S., Wang X., Wu Y., Zhong Y. Low high-density lipoprotein level is correlated with the severity of COVID-19 patients: an observational study. Lipids Health Dis. 2020;19(1):204. doi: 10.1186/s12944-020-01382-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hu X., Chen D., Wu L., He G., Ye W. Declined serum high density lipoprotein cholesterol is associated with the severity of COVID-19 infection. Clin. Chim. Acta. 2020;510:105–110. doi: 10.1016/j.cca.2020.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cai Y., Hao Z., Gao Y., Ping W., Wang Q., Peng S., Zhao B., Sun W., Zhu M., Li K., Han Y., Kuang D., Chu Q., Fu X., Zhang N. Coronavirus disease 2019 in the perioperative period of lung resection: a brief report from a single thoracic surgery department in wuhan, people's Republic of China. J. Thorac. Oncol. 2020;15(6):1065–1072. doi: 10.1016/j.jtho.2020.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Adams C.W., Bayliss O.B., Baker R.W., Abdulla Y.H., Hunter-Craig C.J. Lipid deposits in ageing human arteries, tendons and fascia. Atherosclerosis. 1974;19(3):429–440. doi: 10.1016/s0021-9150(74)80007-9. [DOI] [PubMed] [Google Scholar]
  • 37.Sene A., Khan A.A., Cox D., Nakamura R.E., Santeford A., Kim B.M., Sidhu R., Onken M.D., Harbour J.W., Hagbi-Levi S., Chowers I., Edwards P.A., Baldan A., Parks J.S., Ory D.S., Apte R.S. Impaired cholesterol efflux in senescent macrophages promotes age-related macular degeneration. Cell Metabol. 2013;17(4):549–561. doi: 10.1016/j.cmet.2013.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Angelidis I., Simon L.M., Fernandez I.E., Strunz M., Mayr C.H., Greiffo F.R., Tsitsiridis G., Ansari M., Graf E., Strom T.M., Nagendran M., Desai T., Eickelberg O., Mann M., Theis F.J., Schiller H.B. An atlas of the aging lung mapped by single cell transcriptomics and deep tissue proteomics. Nat. Commun. 2019;10(1):963. doi: 10.1038/s41467-019-08831-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Coperchini F., Chiovato L., Croce L., Magri F., Rotondi M. The cytokine storm in COVID-19: an overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev. 2020;53:25–32. doi: 10.1016/j.cytogfr.2020.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sviridov D., Miller Y.I., Ballout R.A., Remaley A.T., Bukrinsky M. Targeting lipid rafts-A potential therapy for COVID-19. Front. Immunol. 2020;11 doi: 10.3389/fimmu.2020.574508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gale S.C., Gao L., Mikacenic C., Coyle S.M., Rafaels N., Murray Dudenkov T., Madenspacher J.H., Draper D.W., Ge W., Aloor J.J., Azzam K.M., Lai L., Blackshear P.J., Calvano S.E., Barnes K.C., Lowry S.F., Corbett S., Wurfel M.M., Fessler M.B. APOepsilon4 is associated with enhanced in vivo innate immune responses in human subjects. J. Allergy Clin. Immunol. 2014;134(1):127–134. doi: 10.1016/j.jaci.2014.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jofre-Monseny L., Loboda A., Wagner A.E., Huebbe P., Boesch-Saadatmandi C., Jozkowicz A., Minihane A.M., Dulak J., Rimbach G. Effects of apoE genotype on macrophage inflammation and heme oxygenase-1 expression. Biochem. Biophys. Res. Commun. 2007;357(1):319–324. doi: 10.1016/j.bbrc.2007.03.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rieker C., Migliavacca E., Vaucher A., Baud G., Marquis J., Charpagne A., Hegde N., Guignard L., McLachlan M., Pooler A.M. Apolipoprotein E4 expression causes gain of toxic function in isogenic human induced pluripotent stem cell-derived endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2019;39(9):e195–e207. doi: 10.1161/ATVBAHA.118.312261. [DOI] [PubMed] [Google Scholar]
  • 44.Cudaback E., Yang Y., Montine T.J., Keene C.D. APOE genotype-dependent modulation of astrocyte chemokine CCL3 production. Glia. 2015;63(1):51–65. doi: 10.1002/glia.22732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tall A.R., Westerterp M. Inflammasomes, neutrophil extracellular traps, and cholesterol. J. Lipid Res. 2019;60(4):721–727. doi: 10.1194/jlr.S091280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Anand P.K. Lipids, inflammasomes, metabolism, and disease. Immunol. Rev. 2020;297(1):108–122. doi: 10.1111/imr.12891. [DOI] [PubMed] [Google Scholar]
  • 47.Karasawa T., Kawashima A., Usui-Kawanishi F., Watanabe S., Kimura H., Kamata R., Shirasuna K., Koyama Y., Sato-Tomita A., Matsuzaka T., Tomoda H., Park S.Y., Shibayama N., Shimano H., Kasahara T., Takahashi M. Saturated fatty acids undergo intracellular crystallization and activate the NLRP3 inflammasome in macrophages. Arterioscler. Thromb. Vasc. Biol. 2018;38(4):744–756. doi: 10.1161/ATVBAHA.117.310581. [DOI] [PubMed] [Google Scholar]
  • 48.Corrin B., Soliman S.S. Cholesterol in the lungs of heavy cigarette smokers. Thorax. 1978;33(5):565–568. doi: 10.1136/thx.33.5.565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fireman E., Spitzer S., Grief J., Kivity S., Topilsky M. Cholesterol crystals in BAL fluid from patients with idiopathic pulmonary fibrosis. Respir. Med. 1996;90(6):361–363. doi: 10.1016/s0954-6111(96)90134-1. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang M., Tie H.T., Wang C.L., Wu Q.C. Pulmonary interstitial cholesterol crystals associated with diffuse lung cysts in adult: a case report and literature review. J. Cardiothorac. Surg. 2016;11:11. doi: 10.1186/s13019-016-0397-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Duewell P., Kono H., Rayner K.J., Sirois C.M., Vladimer G., Bauernfeind F.G., Abela G.S., Franchi L., Nunez G., Schnurr M., Espevik T., Lien E., Fitzgerald K.A., Rock K.L., Moore K.J., Wright S.D., Hornung V., Latz E. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464(7293):1357–1361. doi: 10.1038/nature08938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vandanmagsar B., Youm Y.H., Ravussin A., Galgani J.E., Stadler K., Mynatt R.L., Ravussin E., Stephens J.M., Dixit V.D. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 2011;17(2):179–188. doi: 10.1038/nm.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Rheinheimer J., de Souza B.M., Cardoso N.S., Bauer A.C., Crispim D. Current role of the NLRP3 inflammasome on obesity and insulin resistance: a systematic review. Metabolism. 2017;74:1–9. doi: 10.1016/j.metabol.2017.06.002. [DOI] [PubMed] [Google Scholar]
  • 54.Latz E., Duewell P. NLRP3 inflammasome activation in inflammaging. Semin. Immunol. 2018;40:61–73. doi: 10.1016/j.smim.2018.09.001. [DOI] [PubMed] [Google Scholar]
  • 55.Warnatsch A., Ioannou M., Wang Q., Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349(6245):316–320. doi: 10.1126/science.aaa8064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kalyanaraman B. Do free radical NETwork and oxidative stress disparities in African Americans enhance their vulnerability to SARS-CoV-2 infection and COVID-19 severity? Redox Biol. 2020;37 doi: 10.1016/j.redox.2020.101721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fessler M.B. The intracellular cholesterol landscape: dynamic integrator of the immune response. Trends Immunol. 2016;37(12):819–830. doi: 10.1016/j.it.2016.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Soroosh P., Wu J., Xue X., Song J., Sutton S.W., Sablad M., Yu J., Nelen M.I., Liu X., Castro G., Luna R., Crawford S., Banie H., Dandridge R.A., Deng X., Bittner A., Kuei C., Tootoonchi M., Rozenkrants N., Herman K., Gao J., Yang X.V., Sachen K., Ngo K., Fung-Leung W.P., Nguyen S., de Leon-Tabaldo A., Blevitt J., Zhang Y., Cummings M.D., Rao T., Mani N.S., Liu C., McKinnon M., Milla M.E., Fourie A.M., Sun S. Oxysterols are agonist ligands of RORgammat and drive Th17 cell differentiation. Proc. Natl. Acad. Sci. U. S. A. 2014;111(33):12163–12168. doi: 10.1073/pnas.1322807111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gao Q., Jiang Y., Ma T., Zhu F., Gao F., Zhang P., Guo C., Wang Q., Wang X., Ma C., Zhang Y., Chen W., Zhang L. A critical function of Th17 proinflammatory cells in the development of atherosclerotic plaque in mice. J. Immunol. 2010;185(10):5820–5827. doi: 10.4049/jimmunol.1000116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bonacina F., Coe D., Wang G., Longhi M.P., Baragetti A., Moregola A., Garlaschelli K., Uboldi P., Pellegatta F., Grigore L., Da Dalt L., Annoni A., Gregori S., Xiao Q., Caruso D., Mitro N., Catapano A.L., Marelli-Berg F.M., Norata G.D. Myeloid apolipoprotein E controls dendritic cell antigen presentation and T cell activation. Nat. Commun. 2018;9(1):3083. doi: 10.1038/s41467-018-05322-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.De Biasi S., Meschiari M., Gibellini L., Bellinazzi C., Borella R., Fidanza L., Gozzi L., Iannone A., Lo Tartaro D., Mattioli M., Paolini A., Menozzi M., Milic J., Franceschi G., Fantini R., Tonelli R., Sita M., Sarti M., Trenti T., Brugioni L., Cicchetti L., Facchinetti F., Pietrangelo A., Clini E., Girardis M., Guaraldi G., Mussini C., Cossarizza A. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat. Commun. 2020;11(1):3434. doi: 10.1038/s41467-020-17292-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Miller Y.I., Shyy J.Y. Context-dependent role of oxidized lipids and lipoproteins in inflammation. Trends Endocrinol. Metabol. 2017;28(2):143–152. doi: 10.1016/j.tem.2016.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Forstermann U., Xia N., Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ. Res. 2017;120(4):713–735. doi: 10.1161/CIRCRESAHA.116.309326. [DOI] [PubMed] [Google Scholar]
  • 64.Lara-Guzman O.J., Gil-Izquierdo A., Medina S., Osorio E., Alvarez-Quintero R., Zuluaga N., Oger C., Galano J.M., Durand T., Munoz-Durango K. Oxidized LDL triggers changes in oxidative stress and inflammatory biomarkers in human macrophages. Redox Biol. 2018;15:1–11. doi: 10.1016/j.redox.2017.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Que X., Hung M.Y., Yeang C., Gonen A., Prohaska T.A., Sun X., Diehl C., Maatta A., Gaddis D.E., Bowden K., Pattison J., MacDonald J.G., Yla-Herttuala S., Mellon P.L., Hedrick C.C., Ley K., Miller Y.I., Glass C.K., Peterson K.L., Binder C.J., Tsimikas S., Witztum J.L. Oxidized phospholipids are proinflammatory and proatherogenic in hypercholesterolaemic mice. Nature. 2018;558(7709):301–306. doi: 10.1038/s41586-018-0198-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Weinbrenner T., Schroder H., Escurriol V., Fito M., Elosua R., Vila J., Marrugat J., Covas M.I. Circulating oxidized LDL is associated with increased waist circumference independent of body mass index in men and women. Am. J. Clin. Nutr. 2006;83(1):30–35. doi: 10.1093/ajcn/83.1.30. [DOI] [PubMed] [Google Scholar]
  • 67.Holvoet P., Lee D.H., Steffes M., Gross M., Jacobs D.R., Jr. Association between circulating oxidized low-density lipoprotein and incidence of the metabolic syndrome. J. Am. Med. Assoc. 2008;299(19):2287–2293. doi: 10.1001/jama.299.19.2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ehara S., Ueda M., Naruko T., Haze K., Itoh A., Otsuka M., Komatsu R., Matsuo T., Itabe H., Takano T., Tsukamoto Y., Yoshiyama M., Takeuchi K., Yoshikawa J., Becker A.E. Elevated levels of oxidized low density lipoprotein show a positive relationship with the severity of acute coronary syndromes. Circulation. 2001;103(15):1955–1960. doi: 10.1161/01.cir.103.15.1955. [DOI] [PubMed] [Google Scholar]
  • 69.Shen Y., Yang T., Guo S., Li X., Chen L., Wang T., Wen F. Increased serum ox-LDL levels correlated with lung function, inflammation, and oxidative stress in COPD. Mediat. Inflamm. 2013;2013 doi: 10.1155/2013/972347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gkogkou E., Barnasas G., Vougas K., Trougakos I.P. Expression profiling meta-analysis of ACE2 and TMPRSS2, the putative anti-inflammatory receptor and priming protease of SARS-CoV-2 in human cells, and identification of putative modulators. Redox Biol. 2020;36 doi: 10.1016/j.redox.2020.101615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wallner S., Grandl M., Liebisch G., Peer M., Orso E., Sigruner A., Sobota A., Schmitz G. oxLDL and eLDL induced membrane microdomains in human macrophages. PloS One. 2016;11(11) doi: 10.1371/journal.pone.0166798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Carpinteiro A., Edwards M.J., Hoffmann M., Kochs G., Gripp B., Weigang S., Adams C., Carpinteiro E., Gulbins A., Keitsch S., Sehl C., Soddemann M., Wilker B., Kamler M., Bertsch T., Lang K.S., Patel S., Wilson G.C., Walter S., Hengel H., Pohlmann S., Lang P.A., Kornhuber J., Becker K.A., Ahmad S.A., Fassbender K., Gulbins E. Pharmacological inhibition of acid sphingomyelinase prevents uptake of SARS-CoV-2 by epithelial cells. Cell Rep Med. 2020;1(8) doi: 10.1016/j.xcrm.2020.100142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Imai Y., Kuba K., Neely G.G., Yaghubian-Malhami R., Perkmann T., van Loo G., Ermolaeva M., Veldhuizen R., Leung Y.H., Wang H., Liu H., Sun Y., Pasparakis M., Kopf M., Mech C., Bavari S., Peiris J.S., Slutsky A.S., Akira S., Hultqvist M., Holmdahl R., Nicholls J., Jiang C., Binder C.J., Penninger J.M. Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell. 2008;133(2):235–249. doi: 10.1016/j.cell.2008.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yamashita C.M., Fessler M.B., Vasanthamohan L., Lac J., Madenspacher J., McCaig L., Yao L., Wang L., Puntorieri V., Mehta S., Lewis J.F., Veldhuizen R.A. Apolipoprotein E-deficient mice are susceptible to the development of acute lung injury. Respiration. 2014;87(5):416–427. doi: 10.1159/000358438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Miyata M., Smith J.D. Apolipoprotein E allele-specific antioxidant activity and effects on cytotoxicity by oxidative insults and beta-amyloid peptides. Nat. Genet. 1996;14(1):55–61. doi: 10.1038/ng0996-55. [DOI] [PubMed] [Google Scholar]
  • 76.Pham T., Kodvawala A., Hui D.Y. The receptor binding domain of apolipoprotein E is responsible for its antioxidant activity. Biochemistry. 2005;44(20):7577–7582. doi: 10.1021/bi0472696. [DOI] [PubMed] [Google Scholar]
  • 77.Wehr H., Bednarska-Makaruk M., Graban A., Kunicki P.K., Lojkowska W., Rodo M., Ryglewicz D. Apolipoprotein E polymorphism and low density lipoprotein (LDL) oxidation in patients with dementia. Folia Neuropathol. 2003;41(2):65–68. [PubMed] [Google Scholar]
  • 78.Smith J.D., Miyata M., Poulin S.E., Neveux L.M., Craig W.Y. The relationship between apolipoprotein E and serum oxidation-related variables is apolipoprotein E phenotype dependent. Int. J. Clin. Lab. Res. 1998;28(2):116–121. doi: 10.1007/s005990050030. [DOI] [PubMed] [Google Scholar]
  • 79.Talmud P.J., Stephens J.W., Hawe E., Demissie S., Cupples L.A., Hurel S.J., Humphries S.E., Ordovas J.M. The significant increase in cardiovascular disease risk in APOEepsilon4 carriers is evident only in men who smoke: potential relationship between reduced antioxidant status and ApoE4. Ann. Hum. Genet. 2005;69(Pt 6):613–622. doi: 10.1111/j.1529-8817.2005.00205.x. [DOI] [PubMed] [Google Scholar]
  • 80.Ky B., Burke A., Tsimikas S., Wolfe M.L., Tadesse M.G., Szapary P.O., Witztum J.L., FitzGerald G.A., Rader D.J. The influence of pravastatin and atorvastatin on markers of oxidative stress in hypercholesterolemic humans. J. Am. Coll. Cardiol. 2008;51(17):1653–1662. doi: 10.1016/j.jacc.2008.01.026. [DOI] [PubMed] [Google Scholar]
  • 81.Hogue J.C., Lamarche B., Tremblay A.J., Bergeron J., Gagne C., Couture P. Differential effect of atorvastatin and fenofibrate on plasma oxidized low-density lipoprotein, inflammation markers, and cell adhesion molecules in patients with type 2 diabetes mellitus. Metabolism. 2008;57(3):380–386. doi: 10.1016/j.metabol.2007.10.014. [DOI] [PubMed] [Google Scholar]
  • 82.Frost F.J., Petersen H., Tollestrup K., Skipper B. Influenza and COPD mortality protection as pleiotropic, dose-dependent effects of statins. Chest. 2007;131(4):1006–1012. doi: 10.1378/chest.06-1997. [DOI] [PubMed] [Google Scholar]
  • 83.Funderburg N.T., Mehta N.N. Lipid abnormalities and inflammation in HIV inflection. Curr. HIV AIDS Rep. 2016;13(4):218–225. doi: 10.1007/s11904-016-0321-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Fedson D.S., Rordam O.M. Treating Ebola patients: a 'bottom up' approach using generic statins and angiotensin receptor blockers. Int. J. Infect. Dis. 2015;36:80–84. doi: 10.1016/j.ijid.2015.04.019. [DOI] [PubMed] [Google Scholar]
  • 85.Shrivastava-Ranjan P., Flint M., Bergeron E., McElroy A.K., Chatterjee P., Albarino C.G., Nichol S.T., Spiropoulou C.F. Statins suppress Ebola virus infectivity by interfering with glycoprotein processing. mBio. 2018;9(3) doi: 10.1128/mBio.00660-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ganjali S., Bianconi V., Penson P.E., Pirro M., Banach M., Watts G.F., Sahebkar A. Commentary: statins, COVID-19, and coronary artery disease: killing two birds with one stone. Metabolism. 2020;113 doi: 10.1016/j.metabol.2020.154375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kow C.S., Hasan S.S. Meta-analysis of effect of statins in patients with COVID-19. Am. J. Cardiol. 2020;134:153–155. doi: 10.1016/j.amjcard.2020.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ahmadizar F., Ochoa-Rosales C., Glisic M., Franco O.H., Muka T., Stricker B.H. Associations of statin use with glycaemic traits and incident type 2 diabetes. Br. J. Clin. Pharmacol. 2019;85(5):993–1002. doi: 10.1111/bcp.13898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.VanFossen B.T., Watson G.S., Baker L.D., Rhoads K.W., Cholerton B.A., Reger M.A., Plymate S.R., Schellenberg G., Craft S. Statin users without an APOE-epsilon4 allele have increased insulin resistance. J Alzheimers Dis. 2010;19(4):1149–1153. doi: 10.3233/JAD-2010-1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Valanti E.K., Dalakoura-Karagkouni K., Siasos G., Kardassis D., Eliopoulos A.G., Sanoudou D. Advances in biological therapies for dyslipidemias and atherosclerosis. Metabolism. 2020;116 doi: 10.1016/j.metabol.2020.154461. [DOI] [PubMed] [Google Scholar]
  • 91.Sanchez-Muniz F.J., Maki K.C., Schaefer E.J., Ordovas J.M. Serum lipid and antioxidant responses in hypercholesterolemic men and women receiving plant sterol esters vary by apolipoprotein E genotype. J. Nutr. 2009;139(1):13–19. doi: 10.3945/jn.108.090969. [DOI] [PubMed] [Google Scholar]
  • 92.Gkouskou Kk G.M., Vlastos I., Sanoudou D., Eliopoulos A.G. Nutr Rev in press; 2020. Genotype-guided Dietary Supplementation in Precision Nutrition. [DOI] [PubMed] [Google Scholar]
  • 93.Griffin B.A., Walker C.G., Jebb S.A., Moore C., Frost G.S., Goff L., Sanders T.A.B., Lewis F., Griffin M., Gitau R., Lovegrove J.A. APOE4 genotype exerts greater benefit in lowering plasma cholesterol and apolipoprotein B than wild type (E3/E3), after replacement of dietary saturated fats with low glycaemic index carbohydrates. Nutrients. 2018;10(10) doi: 10.3390/nu10101524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gammon C.S., Kruger R., Minihane A.M., Conlon C.A., von Hurst P.R., Stonehouse W. Kiwifruit consumption favourably affects plasma lipids in a randomised controlled trial in hypercholesterolaemic men. Br. J. Nutr. 2013;109(12):2208–2218. doi: 10.1017/S0007114512004400. [DOI] [PubMed] [Google Scholar]
  • 95.Serhan C.N., Levy B.D. Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J. Clin. Invest. 2018;128(7):2657–2669. doi: 10.1172/JCI97943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sorokin A.V., Yang Z.H., Vaisman B.L., Thacker S., Yu Z.X., Sampson M., Serhan C.N., Remaley A.T. Addition of aspirin to a fish oil-rich diet decreases inflammation and atherosclerosis in ApoE-null mice. J. Nutr. Biochem. 2016;35:58–65. doi: 10.1016/j.jnutbio.2016.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Group R.C., Horby P., Lim W.S., Emberson J.R., Mafham M., Bell J.L., Linsell L., Staplin N., Brightling C., Ustianowski A., Elmahi E., Prudon B., Green C., Felton T., Chadwick D., Rege K., Fegan C., Chappell L.C., Faust S.N., Jaki T., Jeffery K., Montgomery A., Rowan K., Juszczak E., Baillie J.K., Haynes R., Landray M.J. Dexamethasone in hospitalized patients with covid-19 - preliminary report. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2021436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pyrillou K., Chairakaki A.D., Tamvakopoulos C., Andreakos E. Dexamethasone induces omega3-derived immunoresolvents driving resolution of allergic airway inflammation. J. Allergy Clin. Immunol. 2018;142(2):691–695 e4. doi: 10.1016/j.jaci.2018.04.004. [DOI] [PubMed] [Google Scholar]
  • 99.Andreakos E., Papadaki M., Serhan C.N. 2020. Dexamethasone, Pro-resolving Lipid Mediators and Resolution of Inflammation in COVID-19. Allergy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Huebbe P., Lodge J.K., Rimbach G. Implications of apolipoprotein E genotype on inflammation and vitamin E status. Mol. Nutr. Food Res. 2010;54(5):623–630. doi: 10.1002/mnfr.200900398. [DOI] [PubMed] [Google Scholar]
  • 101.Jovic T.H., Ali S.R., Ibrahim N., Jessop Z.M., Tarassoli S.P., Dobbs T.D., Holford P., Thornton C.A., Whitaker I.S. Could vitamins help in the fight against COVID-19? Nutrients. 2020;12(9) doi: 10.3390/nu12092550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wang C., Zhang M., Garcia G., Jr., Tian E., Cui Q., Chen X., Sun G., Wang J., Arumugaswami V., Shi Y. ApoE-isoform-dependent SARS-CoV-2 neurotropism and cellular response. Cell Stem Cell. 2021;28(2):331–342 e5. doi: 10.1016/j.stem.2020.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Scacchi R., De Bernardini L., Mantuano E., Donini L.M., Vilardo T., Corbo R.M. Apolipoprotein E (APOE) allele frequencies in late-onset sporadic Alzheimer's disease (AD), mixed dementia and vascular dementia: lack of association of epsilon 4 allele with AD in Italian octogenarian patients. Neurosci. Lett. 1995;201(3):231–234. doi: 10.1016/0304-3940(95)12190-0. [DOI] [PubMed] [Google Scholar]
  • 104.Severe Covid G.G., Ellinghaus D., Degenhardt F., Bujanda L., Buti M., Albillos A., Invernizzi P., Fernandez J., Prati D., Baselli G., Asselta R., Grimsrud M.M., Milani C., Aziz F., Kassens J., May S., Wendorff M., Wienbrandt L., Uellendahl-Werth F., Zheng T., Yi X., de Pablo R., Chercoles A.G., Palom A., Garcia-Fernandez A.E., Rodriguez-Frias F., Zanella A., Bandera A., Protti A., Aghemo A., Lleo A., Biondi A., Caballero-Garralda A., Gori A., Tanck A., Carreras Nolla A., Latiano A., Fracanzani A.L., Peschuck A., Julia A., Pesenti A., Voza A., Jimenez D., Mateos B., Nafria Jimenez B., Quereda C., Paccapelo C., Gassner C., Angelini C., Cea C., Solier A., Pestana D., Muniz-Diaz E., Sandoval E., Paraboschi E.M., Navas E., Garcia Sanchez F., Ceriotti F., Martinelli-Boneschi F., Peyvandi F., Blasi F., Tellez L., Blanco-Grau A., Hemmrich-Stanisak G., Grasselli G., Costantino G., Cardamone G., Foti G., Aneli S., Kurihara H., ElAbd H., My I., Galvan-Femenia I., Martin J., Erdmann J., Ferrusquia-Acosta J., Garcia-Etxebarria K., Izquierdo-Sanchez L., Bettini L.R., Sumoy L., Terranova L., Moreira L., Santoro L., Scudeller L., Mesonero F., Roade L., Ruhlemann M.C., Schaefer M., Carrabba M., Riveiro-Barciela M., Figuera Basso M.E., Valsecchi M.G., Hernandez-Tejero M., Acosta-Herrera M., D'Angio M., Baldini M., Cazzaniga M., Schulzky M., Cecconi M., Wittig M., Ciccarelli M., Rodriguez-Gandia M., Bocciolone M., Miozzo M., Montano N., Braun N., Sacchi N., Martinez N., Ozer O., Palmieri O., Faverio P., Preatoni P., Bonfanti P., Omodei P., Tentorio P., Castro P., Rodrigues P.M., Blandino Ortiz A., de Cid R., Ferrer R., Gualtierotti R., Nieto R., Goerg S., Badalamenti S., Marsal S., Matullo G., Pelusi S., Juzenas S., Aliberti S., Monzani V., Moreno V., Wesse T., Lenz T.L., Pumarola T., Rimoldi V., Bosari S., Albrecht W., Peter W., Romero-Gomez M., D'Amato M., Duga S., Banales J.M., Hov J.R., Folseraas T., Valenti L., Franke A., Karlsen T.H. Genomewide association study of severe covid-19 with respiratory failure. N. Engl. J. Med. 2020;383(16):1522–1534. doi: 10.1056/NEJMoa2020283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Fecchi K., Anticoli S., Peruzzu D., Iessi E., Gagliardi M.C., Matarrese P., Ruggieri A. Coronavirus interplay with lipid rafts and autophagy unveils promising therapeutic targets. Front. Microbiol. 2020;11:1821. doi: 10.3389/fmicb.2020.01821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Humphries S.E., Talmud P.J., Hawe E., Bolla M., Day I.N., Miller G.J. Apolipoprotein E4 and coronary heart disease in middle-aged men who smoke: a prospective study. Lancet. 2001;358(9276):115–119. doi: 10.1016/S0140-6736(01)05330-2. [DOI] [PubMed] [Google Scholar]
  • 107.Kulminski A.M., Barochia A.V., Loika Y., Raghavachari N., Arbeev K.G., Wojczynski M.K., Thyagarajan B., Vardarajan B.N., Christensen K., Yashin A.I., Levine S.J. The APOE epsilon4 allele is associated with a reduction in FEV1/FVC in women: a cross-sectional analysis of the Long Life Family Study. PloS One. 2018;13(11) doi: 10.1371/journal.pone.0206873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lawrie A., Hameed A.G., Chamberlain J., Arnold N., Kennerley A., Hopkinson K., Pickworth J., Kiely D.G., Crossman D.C., Francis S.E. Paigen diet-fed apolipoprotein E knockout mice develop severe pulmonary hypertension in an interleukin-1-dependent manner. Am. J. Pathol. 2011;179(4):1693–1705. doi: 10.1016/j.ajpath.2011.06.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Havel R.J., Kane J.P. Primary dysbetalipoproteinemia: predominance of a specific apoprotein species in triglyceride-rich lipoproteins. Proc. Natl. Acad. Sci. U. S. A. 1973;70(7):2015–2019. doi: 10.1073/pnas.70.7.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Redox Biology are provided here courtesy of Elsevier

RESOURCES