17 Feb 2024

From saturated fat to arterial plaque: cholesterol and beyond

Cardiovascular diseases (CVDs) are still the leading cause of death globally (OWID), but vary markedly by demographics and in relation to various lifestyle factors. In particular, in the second half of the 20th century, the seminal Seven Countries Study illuminated associations with diet, especially saturated fatty acids (SFAs), which at 50-year follow-up remain strongly associated with coronary heart disease mortality (n=16 cohorts, r=0.92) 1. Many other studies have further probed this relationship, and while some heterogeneity emerged, so has context. For instance, some prospective cohort studies and meta-analyses thereof fail to find independent associations with SFA intake 2; however, studies performing substitution analyses generally report that replacing sources of SFAs with (plant and marine-sourced) unsaturated fatty acids (UFAs) or complex carbohydrates lowers CVD risk and mortality (e.g. US 3–8, Europe 9–11 and pooled 12,13). Importantly, in the US it was also revealed that SFAs are typically replaced by refined grains and added sugars, which are also associated with CVD, potentially explaining prior null findings 4. Further, dietary SFAs are also highly correlated with animal-sourced MUFAs (r=>0.8), which may have obscured favourable associations seen only with plant sources 7,8.

Paralleling the observational research, a number of heterogeneous randomised controlled trials (RCTs) have also been conducted. In the latest Cochrane meta-analysis of such RCTs lowering SFAs significantly lowered CVD events, mainly being driven by replacement with various PUFAs (n=8, RR=0.79, 95% CI 0.62–1.0) and carbs (n=5, RR=0.84, 95% CI 0.67–1.06), but not mortality (albeit with 75% less death vs. event data) 14. Further supporting this, a diverse literature of shorter RCTs show replacement of SFAs can favourably modulate various biomarkers of risk and pathophysiology; e.g. lipids/lipoproteins 15,16, immune-inflammation 17,18, glucose-insulin homeostasis 19, liver fat 20, endothelial integrity and platelet activity 21,22.

Accordingly, lowering/replacing SFAs is a cornerstone of dietary guidelines worldwide, along with refined grains and added sugars (FAO). On the other hand, not everyone agrees 23–26. As well as issues with food/nutrient replacements above, other nuances include the specific SFAs/PUFAs, food source/matrix, dietary patterns and endogenous biosynthesis (i.e. de novo lipogenesis, DNL), all of which may confer differential effects. These factors in mind, this post explores some of the major lipid-based pathways which may mediate the association of dietary SFAs with CVD, as guided by qualitative comparisons with UFAs in humans, and supported by preclinical and mechanistic models.

Cholesterol

CVD events are typically precipitated by atherosclerosis—the build-up of arterial plaque. Atherosclerosis starts in youth and progresses insidiously, from initially superficial intima–media thickening, which increasingly protrudes the lumen and reduces blood flow, to eventual plaque rupture triggering thrombosis and infarction (e.g. heart attack or stroke). Importantly, atherosclerosis is a systemic disease and systemic imaging of general populations suggests subclinical plaque (i.e. stenosis or calcification) is present in around half of asymptomatic individuals by midlife (e.g. US 27, Scotland 28,29, Spain 30,31 and Egypt 32), affecting many arteries (esp. aorta–iliac) and correlating brain hypometabolism 30. At the cellular–molecular level, necropsy studies show the progression from initial fatty lesion to advanced plaque involves an increasing content of lipids, chiefly cholesterol (i.e. free and esterified) and phospholipids (i.e. phosphatidylcholines and sphingolipids) 33, along with increasing leukocytes (esp. macrophages), necrosis, fibrosis and calcification 34. The lipids are present in foam cells (i.e. as lipid droplets) and extracellular deposits (e.g. lipoprotein aggregates and crystals), while the simultaneous presence of apolipoprotein B (apoB) implicates plasma lipoproteins as a source 33,35–37.

Low-density lipoprotein cholesterol (LDL-c) in particular has a long association with CVD 38; even in explicitly low risk populations LDL-c has been independently and linearly associated with subclinical atherosclerosis and CVD mortality 31,39,40. The causal role of LDL is supported by drug trials (i.e. late life lipid-lowering) and more recent Mendelian randomisation studies (i.e. lifetime lipids) 38,41. However, CVD risk generally tracks better with apoB, which reflects the particle count of all atherogenic lipoproteins—LDL being most abundant (typically >90%)—and can be discordant from LDL-c (and non-HDL-c) due to variations in particle size/content, especially in metabolic disorders with abundant small-dense LDL particles (i.e. pattern B) 42,43. A recent Mendelian study further suggests the risk from apoB is actually mediated by non-HDL-c, which reflects the total cholesterol content of all apoB-lipoproteins 44. Here the apoB100 lineage dominates and is secreted by the liver and metabolised systemically, where VLDL undergoes lipolysis (via LpL) to IDL and LDL, while HDL transfers (via CETP) cholesteryl esters (CEs), thereby generating increasingly triglyceride-poor and cholesterol-rich particles. LDL is the major cholesterol carrier and has a long plasma residence (in the order of days) before being endocytosed via the LDL receptor (LDLR) and largely cleared by the liver 45, where it subserves (indirect) reverse cholesterol transport; i.e. ~70% of HDL-CEs are transferred to VLDL/LDL before hepatic uptake 46. However, the majority of peripheral cholesterol clearance may occur more rapidly via direct uptake of HDL-derived free cholesterol 47,48.

Conversely, in early atherogenesis lipids and lipoproteins accumulate in arterial intima at susceptible sites (e.g. branches and bifurcations) 49–51, and prior to macrophages 37. These sites are typically exposed to turbulent blood flow and shear stress and often exhibit increased lipoprotein permeability and/or retention 52. Normally apoB-lipoproteins up to ~70nm in diameter (i.e. VLDL–LDLs and chylomicron remnants) can cross an intact endothelium, wherein binding to extracellular matrix proteoglycans (via electrostatic interaction) and LpL (acting as a bridge) promotes their retention 37,49,50, while exposure to various enzymes and oxidants promote modifications, ultimately resulting in lipoprotein aggregation, fusion and cholesterol crystallisation 36,53,54. Notably, among plasma lipoproteins small-dense LDL exhibits various qualities which may render it particularly atherogenic 55, albeit while carrying a smaller cholesterol load 42; indeed when controlling for particle count, large and small LDL similarly relate to CVD risk 56. A characteristic event of atherogenesis is the formation of foam cells, which contain abundant CE-rich lipid droplets, giving them their ‘foamy’ appearance. Lipoprotein aggregates isolated from human plaques induce accelerated macrophage uptake, greater cholesterol esterification 57 and inflammasome activation 36. This reflects a dichotomy: foam cells remove and digest harmful extracellular deposits liberating free cholesterol, with potential for efflux to HDL and out of arteries, but when overloaded can themselves be a site of crystallisation, inflammation and cytotoxicity 51,58. Cholesterol crystals may also form early in experimental atherosclerosis (i.e. pre-macrophage) via endothelial cells, which also process LDL but seem less able to upregulate efflux 59. Such crystals can impair the endothelial barrier (to leukocytes) 59, and activate endothelial 60 and macrophage NLRP3 inflammasomes—a pathway required for atherogenesis, and which releases cytokines recruiting more immune cells 61.

Beyond the arterial wall, plasma lipids also modulate important extravascular pathways. For instance, hypercholesterolemia suppresses and lipid-lowering restores bone marrow-derived endothelial progenitor cells (EPCs), which mediate vascular repair 62,63. Hypercholesterolemia also acts on bone marrow and peripheral cells to favour platelet biogenesis and activation 64, as well as monocyte skewing and activation 65, thereby priming the precursor to tissue macrophage foam cells 66. Furthermore, circulating monocytes can also internalise CE-rich lipoproteins, become foamy and inflammatory, and subsequently infiltrate nascent lesions 67.

In the Cochrane analysis of SFA-lowering trials above, meta-regression of various factors showed that the greater the reduction in serum cholesterol, the greater the reduction in CVD events, accounting for 99% of between-trial variation 14. Notably, of these trials, STARS also measured angiographic progression of CAD, which correlated intake of animal-sourced trans fatty acids (TFAs) and SFAs (i.e. C14–18:0), and which was also partially mediated via plasma cholesterol 68. Indeed it’s well established that isocaloric replacement of complex carbohydrates with common dietary SFAs (i.e. C12–16:0) raises total/LDL cholesterol, whereas typical plant-source PUFAs (i.e. C18:2/3) lower it, as tested in 100s of metabolic ward studies 15 and formularised since the 1950s 69. This dichotomy extends to low carb/keto diets 70–73; although more extreme hypercholesterolemia may relate to leanness and greater TG/VLDL turnover 74 (even when favouring MUFAs 75). The effect of SFAs may be accentuated by dietary cholesterol 76,77, attenuated in the case of cheese (vs. butter) 78 and modulated by APOE genotype (i.e. E4 79,80 and -219G/T 81). SFA-rich diets similarly increase apoB 78,82–85 (meta-regression in 16); more specifically, dairy fat/SFAs can increase all VLDL–LDL particles (vs. seed oils/n-6 PUFAs) 82,86, large LDL (vs. MUFAs) 84, or in people with pattern B, medium–small LDL particles (vs. MUFAs) 85. Mechanistically, human and animal studies suggest SFA-rich diets suppress LDL catabolism 87 and LDLR expression 82,83, thereby prolonging LDL residence in blood 88, but in a manner dependent upon dietary cholesterol and UFAs 89,90. However, even mice made prone to atherosclerosis via genetically impaired (hepatic) uptake exhibit differential effects. For instance, in APOE–/– or LDLR–/– mice a coconut oil/SFA-rich diet (vs. fish oil/n-3 PUFAs) greatly increased arterial uptake of whole LDL and CEs, paralleling macrophage LpL and lesion development 91–93, while in LDLR–/– mice replacing SFAs with MUFAs (i.e. dairy fat with extra-virgin olive oil and nuts, respectively) decreased circulating foamy-inflammatory monocytes and atherosclerosis 94.

SFAs may also affect atherogenesis via modulation of triglyceride-rich lipoproteins (TRLs) 95, which constitute non-LDL/HDL-associated ‘remnant cholesterol’ 96. The effect of different dietary fats on postprandial lipemia in humans so far seems equivocal (reviewed in 97). However, in healthy adults high fat/SFA meals (vs. low fat) can induce foamy-activated monocytes in association with postprandial TRLs 98–101, while in vitro SFAs (vs. MUFAs and PUFAs) induced greater monocyte lipid droplet formation 100. As intestine-derived apoB48-chylomicrons are depleted of their triglycerides they become remnant particles, which are rich in cholesterol and associated with atherogenesis (i.e. analogous to conversion of VLDL to LDL). In preclinical studies enrichment of chylomicron remnants with SFAs (vs. various UFAs) reduces hepatic uptake but induces macrophage lipid accumulation 102. Further, in humans SFA-rich meals (i.e. palm oil and cocoa butter vs. various UFAs) induced apoE expression in TRLs which in vitro increased hepatocyte LDLR binding resulting in competitive inhibition of LDL uptake 103; this effect was even greater in those with an APOE4 allele, which is itself associated with both Alzheimer’s and CVD 80. Notably, in mice a cocoa butter/SFA-rich diet (not MUFAs or fish oil/n-3 PUFAs) also increased amyloid-beta (Aβ) in gut enterocytes and plasma TRLs 104,105, and induced blood–brain barrier (BBB) dysfunction and Aβ transport to the brain 106; potentially underlying the association between SFA intake and Alzheimer’s (reviewed in 107). Reciprocally, dementia and plasma Aβ40 are associated with CVD 108, while Aβ40/42 binding to native or modified LDL enhanced foam cell formation in vitro 109.

It’s generally thought that lipoproteins must first be modified in some way to promote atherogenesis. However, this may not be strictly true since atherogenic concentrations of native LDL in vitro can induce endothelial dysfunction 110 and permeability 111 (e.g. reviews 58,63), which was recently linked to LDL uptake and cholesterol crystallisation 59. Further, high levels of native LDL can dose-dependently induce macrophage foam cell formation in a non-saturable manner via receptor-independent fluid-phase pinocytosis 112 and selective CE uptake 113; as well as CCL20 secretion by vascular smooth muscle cells, causing lymphocyte migration 114. Regardless, lipoprotein modifications have important amplifying and auxiliary effects…

Oxidation

Oxidised LDL (oxLDL) is present in plaques and plasma where it’s associated with CVD 115,116, although not always independently of apoB (e.g. CHD 117 and MetS 118), likely due to 4E6 antibody cross-reactivity 119. On the other hand, oxidised phospholipids on apoB100 (oxPL–apoB) are independently associated with CVD and mainly carried by Lp(a), an LDL variant; indeed oxLDL donates its oxPL to Lp(a) in vitro 119. OxLDL normally represents a very small fraction of plasma LDL 120 and increases preceding progression and regression of experimental atherosclerosis, suggesting exchange with plaque 121; similar to oxPL–apoB 119. Oxidation of LDL in vitro is typically achieved by incubation with copper (Cu2+) sulfate solution or arterial cells cultured in media containing transition metals, which induce 1-electron oxidations via Fenton chemistry. This enhances its pro-atherogenic endothelial/inflammatory effects (e.g. eNOS 58, CCL20 114, EPCs 63 and HSPCs 122) and induces macrophage uptake via scavenger receptors 116. Here it induces lipid droplets, which may be limited by defective lysosomal processing 112, and also lysosomal crystals and NLRP3 activation 61,123. As the major transition metal in vivo, iron (Fe2+) dysregulation may particularly promote oxidation during plaque haemolysis 124. Moreover, many studies also report that human plaques have increased expression of inducible nitric oxide synthase (iNOS) 125–129 and myeloperoxidase (MPO) 130 (which employ a central heme-iron active site to generate oxidants), while recovered LDL and apoA-I/HDL are highly enriched in their 2-electron protein oxidation products (i.e. nitrotyrosine and 3-chlorotyrosine, respectively) 131,132, implicating immune-dependant redox modifications 133. These oxidations are not blocked by serum (unlike copper oxidation) and resulting NO2–LDL stimulates macrophage uptake and cholesterol loading via scavenger receptor CD36 134,135. Note however, in early studies LDL isolated from human aortic fatty streaks and plaques was not sufficiently oxidised for receptor-mediated uptake, which instead was increased in a non-saturable manner attributable to aggregates 57. Indeed LDL aggregation greatly increases macrophage uptake by receptor-independent endocytosis 57,113 and CE accumulation beyond native or oxLDL 136,137. Further, mildly oxidised LDL inhibits native LDL-induced foam cell formation 138, although such particles tend to aggregate 53 and still induce macrophage crystals 61,123, so may contribute in this way 57.

Of lipids PUFAs are particularly susceptible to oxidation, of which linolenic acid (LA; C18:2, n-6) is the major form in LDL and plaque CEs. A substantial proportion of plaque CE-LA is oxidised 139, despite normal levels of the major lipophilic antioxidant α-tocopherol (aka. vitamin E) 140,141; i.e. contrasting typical conditions in vitro and consistent with failed antioxidant trials 133. Further, under strong copper-oxidising conditions α-tocopherol acts as a chain-breaking antioxidant and underlies the lipid oxidation lag phase, whereas under more mildly oxidising conditions the α-tocopherol radical can initiate lipid oxidation 142, or when there are insufficient regenerative co-antioxidants (e.g. CoQ10 and carotenoids) 143; and α-tocopherol does not block iNOS/MPO-derived oxidants 140. Importantly, human aortic lesions from early to end-stage disease had accumulation of non-oxidised cholesterol and CEs before their oxidised derivatives 144, consistent with the ‘response-to-retention’ model 49,50. More recent high-resolution imaging of advanced carotid plaques has further revealed that oxidised CEs co-localise with sphingomyelin in the necrotic core 145. Of potential relevance, oxidation of LDL-c generates 7-ketocholesterol, which in macrophages inhibits lysosomal sphingomyelinase (SMase) causing accumulation of sphingomyelin–cholesterol particles 146, and also dose-dependently induces cholesterol crystals 147.

Regarding diet, in various short-term trials a lower total fat/SFA or higher olive oil/MUFA intake can lower LDL susceptibility to copper oxidation and monocyte adhesion in vitro, correlating the LDL oleate/linoleate (i.e. MUFA/PUFA) ratio 148–150; conversely, these effects increased most with a mixed n-6/n-3 PUFA diet 148. However, the relevance of this is questionable since it doesn’t parallel favourable associations between PUFAs and hard outcomes, or consider changes to pathways in vivo which initiate oxidation (e.g. iron and inflammation) and increase apoB-lipoproteins (i.e. total substrate) 150. Importantly, the omega-3 content of advanced carotid plaques is increased by supplementation and correlates greater stability and lower inflammation, consistent with anti-inflammatory effects 151,152. Furthermore, the food matrix is also important. For instance, in healthy adults a PUFA-rich walnut meal (i.e. 59g fat, 42g PUFA, 34g LA) increased blood antioxidants and lowered oxLDL at 2 hours (i.e. Fig. 1) 153, while longer trials show enrichment of PUFAs with preservation of oxidation status 154,155, alongside many other cardio-protective effects (reviewed in 156). Conversely, red meat and heme-iron intake are associated with CVD 157–160 and can promote lipid oxidation during digestion 161. For instance, in humans and mice red meat ingestion induced postprandial plasma lipid oxidation (i.e. 3x MDA) and corresponding LDL modification, which was greatly inhibited by polyphenols 162,163; and in gastric models olive oil/MUFAs inhibited red meat/iron-induced lipid peroxidation, opposite to fish oil/n-3 PUFAs 164. Notably, in a unique RCT comparing oxidised vs. high quality fish oil, only the latter lowered apoB-lipoproteins 165.

In preclinical studies HDL has many anti-atherogenic activities, while several human studies report that cholesterol efflux capacity from macrophage to HDL predicts CVD risk better than HDL-c 46. Conversely, apoA-I/HDL isolated from human plaques is lipid-poor and pro-inflammatory as a result of various modifications 166, including chlorination, as above. In particular, the first stage of reverse cholesterol transport involves ABCA1-dependent efflux to apoA-I forming nascent HDL 46, which is inactivated by MPO 132,167. These pathways may be modulated by dietary fats. For instance, healthy adults (n=122) with higher insulin resistance or SFA intake (>10% kcal) had lower ABCA1-dependent efflux (independent of HDL-c) 168, while in mice SFA-rich diets lowered liver­–faeces cholesterol transport and enriched HDL with hepatic-derived acute-phase (inflammatory) proteins (i.e. palm oil/SFAs vs. sunflower oil/MUFAs) 168, and impaired HDL antioxidant activity (i.e. dairy fat/SFAs vs. soybean oil/n-6 PUFAs) 169. Accordingly, in young healthy adults a single coconut oil/SFA-rich meal (vs. safflower oil/n-6 PUFAs) suppressed HDL anti-inflammatory activity and flow-mediated dilation (FMD) 170. Further, a high fat milkshake (vs. low fat) induced pathological RBC remodelling and foamy monocytes, while elevating plasma and RBC-bound MPO in association with impaired FMD and chlorination of HDL 98; and in vitro cow milk fatty acids (i.e. oleate or palmitate) induced MPO release by monocytes and uptake by porcine arteries 98.

Sphingolipids

Alongside cholesterol, another significant component of plaque is sphingolipids 33. Here various sphingomyelins and ceramides are present and associated with plaque inflammation and apoptosis 171, while serum ceramides (esp. Cer16:0, Cer18:0 and Cer24:1) predict CVD risk independent of conventional risk factors (incl. apoB) 172. Moreover, in rodent models high fat diets induce de novo sphingolipid synthesis (i.e. via SPT) and salvage pathway turnover, increasing the generation of long-chain ceramides in liver, plasma and elsewhere, while the SPT inhibitor myriocin ameliorates atherosclerosis (reviewed in 173). Serum ceramides are particularly elevated in obesity and type-2 diabetes (T2D)—itself a major CVD risk factor 172. In particular, LDL ceramides were specifically elevated in T2D (not obesity) and in preclinical models induce macrophage activation and muscle insulin resistance 174, which involves mitochondrial dysfunction (reviewed in 175). Such insulin resistance promotes hyperglycaemia, which is also associated with CVD 176, while a 1-year RCT with insulin-stimulating drugs induced regression of carotid intima-media thickness (cIMT) in relation to postprandial glucose 177. Note, even in the nondiabetic PESA cohort HbA1c (i.e. monthly glucose control) independently correlated the presence of subclinical atherosclerosis 31. Accordingly, hyperglycaemia can induce non-specific glycation, oxidative-inflammatory activity and endothelial dysfunction 176; in particular, glycation of LDL increases arterial proteoglycan binding 178,179. Insulin itself also promotes hepatic apoB metabolism 180 and may affect many atherogenic cells 181,182.

In people spanning the range of insulin sensitivity (i.e. athletes–lean–obese–T2D), intramuscular accumulation and subcellular localisation (i.e. sarcolemma and organelles) of saturated triglycerides 183 and sphingolipids/ceramides 184 correlate insulin resistance, while SFA intake was increased in T2D 183. Accordingly, in some trials on healthy adults a higher palmitate/SFA intake (vs. oleate/MUFA) for 2–3 weeks 185,186, or as a single bolus 187, induced blood/muscle sphingolipids/ceramides and suppressed glucose metabolism and insulin sensitivity. Further, in trials on overweight adults over-feeding SFAs from mixed sources (vs. UFAs or sugars) for 3 weeks 188, or palm oil/SFAs (vs. sunflower oil/n-6 PUFAs) for 8 weeks 189, increases multiple plasma/LDL sphingolipid species (opposite to PUFAs), paralleling insulin resistance and liver fat (i.e. intrahepatic triglycerides). Conversely, in obese adolescents on a 1-year multidisciplinary intervention SFA reduction correlated a decreased insulin and increased adiponectin/leptin ratio, which itself negatively correlated cIMT 190. Notably, in mice and human cells palm oil/SFAs (vs. olive oil/MUFAs) were also reported to induce intestinal insulin resistance via ceramide, thereby impairing the ability of insulin to inhibit triglyceride secretion and linking to postprandial hypertriglyceridemia 191.

Besides glucose-insulin homeostasis, sphingolipids may have more direct effects on atherogenesis. For instance, LDL can deliver ceramide to endothelial cells 192, where it can mediate apoptosis 192, suppress nitric oxide (i.e. eNOS) 193 and increase the uptake and retention of oxLDL 194. Moreover, aggregated LDL from human plaques was highly enriched in ceramide, a product of sphingomyelin cleavage by SMase, which promotes LDL aggregation and fusion in vitro 53. Accordingly, LDL susceptibility to aggregation, as induced by human secretory sphingomyelinase (S-SMase), was associated with CVD death independent of traditional markers (e.g. LDL-c) and activated macrophages and T cells in vitro (contrasting oxLDL) 195. LDL aggregation was also related to the surface/core lipidome (esp. sphingolipids/ceramides vs. phospholipids) which was favourably modified by a ‘Healthy Nordic diet’ (incl. PUFAs; Fig. S5) or lipid-lowering drug (i.e. PCSK9 inhibition) 195. In a subsequent trial on overweight adults over-eating SFAs from mixed sources for 3 weeks induced LDL sphingolipids and aggregation, while UFAs (i.e. 57% MUFAs, 22% PUFAs) lowered LDL proteoglycan binding (and apoE) and sugars were without effect 196. A further study including liver biopsies reported that LDL aggregation and lipid composition correlates the liver lipidome, implicating hepatic sphingolipid metabolism in LDL composition 197. Also, in LDLR–/– mice a dairy fat/SFA and cholesterol-rich diet induced macrophage S-SMase which acts on serum LDL to increase ceramide and susceptibility to aggregation and oxidation 198.

Lipopolysaccharides

In 1999 the prospective Bruneck study of older Italians (n=516; age 50–79 at baseline) published the first evidence of an association between circulating levels of lipopolysaccharide (LPS), an outer membrane component of Gram-negative bacteria, and early carotid atherosclerosis, which was independent of traditional vascular risk factors (incl. apoB) except smoking 199. Nowadays many studies support a link with cardiometabolic disease and athero-thrombosis (e.g. reviews 200–202). LPS is the canonical ligand for toll-like receptor 4 (TLR4), which stimulates innate immunity and primes the NLRP3 pathway 36,61,123; although depending on source/structure, it can act as an agonist or antagonist (e.g. E. coli and Bacteroides, respectively) 203. In mouse models of endotoxemia, platelet TLR4 triggers neutrophil extracellular traps (NETs) to ensnare bacteria in liver sinusoids and pulmonary capillaries 204,205, but which also promote monocyte recruitment and aortic atherosclerosis 206,207, and increase carotid plaque MPO and instability 208, consistent with human samples 207,208. In particular, E. coli-LPS was present in human carotid plaques (esp. necrotic core; Fig. 1A) and associated with enlarged macrophages, while plaque levels tested in vitro induced monocyte NADPH oxidase 2 (Nox2) and oxLDL, implicating TLR4-mediated oxidative stress 209. Further, carotid LPS correlated plasma LPS (r=0.668), which correlated soluble TLR4 and serum zonulin (a marker of intestinal permeability) 209; with similar blood marker relationships reported in other populations 210–212. Accordingly, several gut microbiome studies on people with CVD find elevated Gram-negative Enterobacteriaceae (e.g. E. coli and Klebsiella) and decreased butyrate-producing bacteria (e.g. Roseburia and Faecalibacterium) 213–215. Butyrate is the archetypal beneficial short-chain fatty acid (SCFA) and ameliorates atherosclerosis in APOE–/– mice by lowering gut permeability and endotoxemia 216 and inducing ABCA1-dependent cholesterol efflux 217. Several human studies also find depletion of Bacteroides spp. 213,218,219, which when administered to APOE–/– mice also lowered atherosclerosis and gut/blood LPS 218.

In another prospective study on people with atrial fibrillation (n=912; mean age 73.5), blood LPS was associated with major CVD events, platelet activation and LDL-c, and negatively with Mediterranean diet scores (esp. fruit and legumes) 220. In blood both LPS and Gram-positive lipoteichoic acid (LTA), as exogenous lipid-based molecules, are largely bound to lipoproteins 221, transferred to LDL (via PLTP) 222,223 and removed predominantly via the hepatic LDLR (in humans) 224,225, suggesting dietary fats may also differentially regulate LPS clearance. A single LDL particle can bind many LPS molecules with only minor changes to its composition 226. While such lipoprotein binding sequesters the lipid A region of LPS within the phospholipid monolayer and hepatic uptake is apparently non-toxic 224,225, chylomicrons and LDL may still carry bioactive LPS across other tissues to elicit inflammation (e.g. lymph nodes 227, endothelium 228, adipose 229 and brain 230). In particular, LDL–LPS complexes formed in vitro acquire a negative charge and have increased binding and accumulation in arterial wall and macrophages 231. LPS also induces smooth muscle cell synthesis of elongated proteoglycans (equal to traditional agonists) 232, increases LDL susceptibility to oxidation (by copper, endothelial and smooth muscle cells) 233, and stimulates macrophage oxLDL uptake and foam cell formation 234; while at concentrations in CAD, LPS and indoxyl sulfate (a microbial metabolite of tryptophan) exhibited co-toxicity on endothelial cells 219. Moreover, in humans and rodents LPS impairs total (i.e. macrophage–faeces) reverse cholesterol transport at multiple steps 235; in part via induction of MPO/SAA 236 and suppression of ABCA1 237.

Regarding dietary fats, in a 3-week trial on overweight adults (similar to those above 188,196), over-feeding SFAs increased faecal Gram-negative Proteobacteria, while UFAs increased butyrate-producing bacteria (i.e. Lachnospira, Roseburia and Ruminococcaceae spp.) 238. Similarly, in a double-blind crossover trial, replacement of butter/SFAs with margarine/PUFAs (i.e. seed oils/n-6 PUFAs) for just 3 days induced Lachnospiraceae and Bifidobacteria, with the former negatively correlating total cholesterol (r=­–0.511) 239. While not generally altered in CVD 213, SFA-rich diets also associate with elevated bile-resistant, sulfide-producing genera (i.e. esp. Bilophila; in faeces 238,240 and mucosa 241), which in mice results from increased secretion of taurine-conjugated bile acids 242 and lowers colonic butyrate and barrier function 242–244. Moreover, in humans SFA-rich meals can increase postprandial plasma LPS (systematic review 18). For instance, dairy fat (vs. carbs, MUFAs and n-3 PUFAs) increased postprandial LPS, paralleling immune cell activation and endothelial adhesion markers 245–247; an effect quicker in obesity 247 and present after 12 weeks of a SFA-rich diet 246. A porridge-based meal with coconut oil/SFAs (vs. fish oil/n-3 PUFAs) also increased postprandial LPS, but not serum cytokines 248. In people with impaired fasting glucose, postprandial LPS correlated apoB48 (i.e. chylomicrons) and oxLDL, which was suppressed by substituting cheese/SFAs for extra-virgin olive oil/MUFAs and inversely correlated plasma polyphenols; while plasma LPS concentrations incubated with platelets in vitro induced TLR4-dependent Nox2 and oxLDL 249. Accordingly, in preclinical studies fat-induced LPS absorption occurs in the small intestine via transcellular chylomicron and portal vein pathways (i.e. lymph and liver, respectively) 227,250. Short-term controlled-feeding crossover trials in healthy adults also report that lowering typical dietary SFA/MUFA ratios lowers LPS-induced cytokine secretion 251,252, suggesting SFA-rich diets may further increase LPS sensitivity.

In human and experimental fatty liver, hepatocyte LPS is also increased, associated with immune-inflammatory markers 253 and implicated in pathogenesis (e.g. reviewed in 254). Various short-term trials show that SFAs in particular can induce liver fat, under isocaloric (i.e. butter/SFAs vs. sunflower oil/n-6 PUFAs 255) or hypercaloric conditions (i.e. SFAs vs. UFAs 189,256 and sugars 188,238); and in association with adipose lipolysis and inflammation, endotoxemia (i.e. LBP/CD14 ratio) and baseline levels of faecal Bilophila 188,238. Accordingly, in mice Bilophila wadsworthia aggravates dairy fat/SFA-induced metabolic dysfunctions and steatosis, while suppressing microbial butyrate and promoting LPS biosynthesis and translocation 243. Also, in healthy adults a single bolus of palm oil/SFAs acutely induced whole-body/muscle/liver insulin resistance and intrahepatic triglycerides, where a parallel mouse study revealed transcriptomic evidence of hepatic LPS/TLR signalling 257. Notably, the intestine secretes HDL3 into the portal blood which can bind LPS and restrain high fat diet-induced liver injury and fat storage 258. In humans experimental endotoxemia induces peripheral inflammation, oxidative stress and lipolysis (i.e. free fatty acids), the latter 2 of which were particularly inhibited by co-infusion of insulin 259. Endotoxemia also induces ceramides in VLDL and LDL 260, which in rodents is accompanied by activation of S-SMase in serum and de novo sphingolipid biosynthesis (i.e. SPT) in liver 260,261. Therefore LPS may mediate some of the changes induced by SFAs above, and consequently differential effects to UFAs. Notably, in other postprandial trials cream-induced liver fat was attenuated by co-administration of 50g glucose (consistent with the ability of insulin to inhibit lipolysis) but not fructose 262, which itself may also be capable of inducing endotoxemia 263, DNL and ceramides 264.

Microdomains

The body is an aqueous environment and lipids are stored and transported in amphipathic membranes which exhibit heterogeneous biophysical properties in relation to their specific compositions. Generally, the plasma membrane exists in a state of liquid-disorder (Ld) with distinct liquid-order (Lo) microdomains (aka. lipid rafts) which are characteristically detergent-resistant and enriched in cholesterol and saturated sphingolipids 265. Such rafts may serve as functional platforms to assemble proteins subserving cell signalling and endocytosis 266, which can be modulated by exogenous lipids. In particular, free cholesterol in the lipoprotein monolayer is in equilibrium exchange with cell membranes 267, while VLDL and LDL were reported to preferentially interact with membrane raft regions, consistent with the high affinity of apoB100 for cholesterol (and contrasting triglyceride-rich chylomicrons) 268. Further, the hepatic LDLR is associated with both clathrin and caveolae-rich membrane regions 269 (which correspond to non-raft and raft regions, respectively 266), and treatment with LDL or cholesterol induced translocation to caveolae coinciding with reduced LDL uptake 269. In addition, internalised LDL-c travels from lysosomes to plasma membrane first, then endoplasmic reticulum (ER) regulatory domains, which can suppress SREBP and thereby cholesterol uptake and synthesis, to maintain homeostasis 270. While in the other direction, the ABCA1 transporter may associate with cholesterol-rich lipid rafts 271 to mediate efflux to apoA-I/HDL 272; indeed the composition of nascent HDL resembles lipid rafts 272. Therefore conditions of high membrane cholesterol may favour reduced uptake and increased efflux.

Membrane fluidity is also determined by lipid saturation, with Ld regions containing phospholipids enriched in UFAs. Challenging cells with PUFAs results in plasma membrane incorporation and compensatory induction of saturated lipids and cholesterol to maintain biophysical homeostasis 273. Conversely, SFAs can induce saturated glycerolipids in ER and solid phase (i.e. solid-order, So) microdomains, in a manner correlating SFA chain length and offset by UFAs 274. In rodent studies SFAs (i.e. C12–16:0) are also relatively poor substrates for ACAT-dependent cholesterol esterification, which may increase free cholesterol in the ER to inhibit SREBP-dependent LDLR expression, thereby increasing plasma cholesterol 89, whereas UFAs (C18:1 > C18:2) stimulate greater hepatic CE synthesis and secretion, as well as LDLR activity, thereby increasing plasma cholesterol turnover 275. In addition, human plasma CEs are especially rich in n-6 PUFAs (i.e. C18:2 > C18:1 > C16:0), even on a high SFA diet 71, consistent with the phospholipid fatty acid preference of LCAT, which mediates esterification in plasma lipoproteins, especially HDL. Accordingly, in humans and rats dietary fat saturation modulates LCAT activity (i.e. n-6 PUFAs > MUFAs > SFAs), which itself correlates plasma linoleate and inversely with oleate 276–278, and in one case also total cholesterol 278. Further, LCAT overexpression in transgenic rabbits elevates HDL-c and lowers LDL-c via the LDLR 279. In this respect, HDL-CEs are transferred to VLDL/LDL 46 and UFAs promote LDL–LDLR interaction. For instance, in men with hypercholesterolemia a 6-week walnut-rich diet increased LDL PUFA content and hepatocyte association in vitro, especially in those showing lower LDL-c and correlating C18:3 in core lipids (i.e. TGs + CEs) 280. In monkeys corn oil/n-6 PUFAs (vs. coconut oil/SFAs) increased LDL uptake by PBMCs which correlated membrane fluidity and lower plasma cholesterol 281; and enrichment of hepatocytes in various fatty acids (i.e. n-6 PUFAs > MUFAs > SFAs) affected LDL binding/metabolism and membrane fluidity in a highly correlated manner, without altering total or esterified cholesterol 282. Similarly, in hamsters decreasing the dietary fat saturation also increased HDL–liver membrane binding 283.

Lipid microdomains are directly implicated in vascular function and atherogenesis. For instance, LDL and hypercholesterolemia inhibit endothelial nitric oxide synthase (eNOS) via translocation to caveolae rafts (reviewed in 58). Similarly, in human atherosclerotic plaques the oxLDL receptor LOX-1 is associated with caveolae and dissociated by statins or MβCD (which extracts membrane cholesterol), thereby abrogating oxLDL-induced apoptosis 284. As above, LDL can also deliver ceramide to endothelial cells 192, where endogenous ceramide promotes the uptake and retention of oxLDL via regulation of transcytosis-related and raft-associated proteins, including LOX-1 194. Further, conditions of hypercholesterolemia and 7-ketocholesterol induce endothelial A-SMase/ceramide-dependent membrane raft redox signalling platforms linked to NLRP3 activation 285. Notably, electronegative LDL also possesses intrinsic SMase activity associated with apoB100 serine O-glycosylation 286; this may be outward-facing so as to engage plasma membrane sphingomyelin, generating ceramide-based microdomains and endocytic vesicles 287,288. Moreover, arterial SMase can hydrolyse sphingomyelin within lipoproteins themselves generating ceramide-rich domains, which may act as nonpolar spots promoting aggregation via hydrophobic interaction 53, as well as displacement and release of cholesterol to neighbouring vesicles 289. Atherosclerotic plaques were also reported to contain membranes enriched in free cholesterol and crystalline domains 58. In preclinical studies plaque crystals co-associated with cholesterol microdomains 290, which can be shed from macrophage membranes 291. Rapid loading of macrophages via phagocytosis of large lipid droplets induces lysosomal free cholesterol and extracellular crystals 51; inhibition of esterification also induces crystals and cytotoxicity offset by extracellular acceptors (i.e. apoA-I/E) mediating efflux of cholesterol from the plasma membrane 292,293. Further, lipid oxidation induces crystalline domains in model membranes under conditions of hyperglycaemia, which can be inhibited by n-3 PUFAs, especially EPA 294. More specifically, smooth muscle cells grown in the presence of 7-ketocholesterol produced extracellular crystals via formation of distinct membrane microdomains due to reduced intercalation with phospholipids 295.

Statins have well-known anti-inflammatory effects. In particular, in an RCT on obese individuals, dual lipid-lowering therapy for 6 weeks markedly lowered apoB and lipids (e.g. LDL-c 141–73mg/dl), as well as fasting and cream/SFA-induced LPS and immune cell activation; pushing postprandial MNC TLR2/4 expression even below baseline (i.e. Fig. 3G/H) 247. In macrophages TLR4 activation requires cholesterol biosynthesis (via FASN) to enter lipid rafts 296, while ABCA1-dependent cholesterol efflux suppresses raft-associated TLR/inflammatory signalling in macrophages 297,298 and endothelial cells 299. Furthermore, in various human cohorts total plasma cholesterol was independently associated with NK cell cytotoxicity, which was lowered by statins or MβCD in vitro 300; and LDL-c correlated a haematopoietic monocyte skewing (vs. granulocytes) in blood 65 and proinflammatory macrophage phenotype in adipose 301, which were suppressed by statins in vivo. Accordingly, in LDLR–/– mice administration of lipid-free apoA-I reduced aortic cholesterol and macrophage deposition, as well as systemic CD131+ immune cells and their CE content, while in vitro LDL and apoA-I oppositely regulated monocyte membrane cholesterol shifting CD131 (i.e. the common β subunit of GM-CSF and IL-3 receptors) between raft and non-raft fractions, respectively 302.

Importantly, lipid rafts are involved in endocytosis 266 and represent a common entry point for many viral, bacterial and fungal pathogens 303–305. For instance, in the colon butyrate may inhibit enteric pathogen invasion via depletion of cholesterol and increased membrane fluidity 306. Similarly, in porcine ileum samples SFA-induced LPS permeability was abrogated by MβCD, implicating lipid rafts 307. Intriguingly, intestinal enterocytes were reported to phagocytose and translocate E. coli/LPS via TLR4 308, which in macrophages is recruited to lipid rafts in response LPS, and also SFAs, but inhibited by n-3 PUFAs 309 (i.e. paralleling effects on postprandial LPS 248,307). Accordingly, bacterial LPS and lipopeptides are acylated with chains of SFAs which engage TLR4 and TLR2, respectively 309. For instance, in E. coli LPS the lipid A region is typically hexa-acylated with C14/12 SFAs which interact with MD2 lipid domains inducing TLR4 dimerization, whereas hypo-acylation or incorporation of UFAs result in antagonist activity 309,310. Indeed total gut LPS silences TLR signalling due to hypo-acylated lipid A in Bacteroidales 203. Consequently, plasma free SFAs have been suggested to mediate LPS-associated postprandial inflammation 311. However, palmitate may not act as a direct ligand for TLR4, but facilitate activation via metabolic reprogramming 312. Of potential relevance, LPS and oxidative stress-induced TLR4 complex formation also requires A-SMase-derived ceramide 313,314 (which has structural similarities to lipid A 315), while palmitate augments LPS inflammatory responses via SMase and de novo ceramide synthesis 316–319. Free SFAs (i.e. palmitate and stearate) can also activate macrophage NLRP3 inflammasomes via flux into phosphatidylcholine 320 and crystallisation 321, which are offset by UFAs.

Ecology

Evidence of atherosclerosis has been reported in ancient humans spanning 4000 years and from diverse locations suggesting a basic predisposition 322; patterns of systemic vascular calcification were even similar between ancient and modern Egyptians, appearing in aorta–iliac beds almost a decade prior to event-related coronary and carotid beds 32. These populations may have been exposed to various enduring risk factors, including diet, smoke and infections, although this remains speculative 323. However, there are some notable examples of extant pre-industrial people with divergent health outcomes, suggesting post-industrial changes are also important 324,325. Foremost, the Tsimane forager-farmers of Bolivia are a tropical subsistence population with a high infectious/inflammatory burden, yet some of the lowest ever reported coronary artery calcium (CAC) scores throughout life 326; as well as atrial fibrillation 327, age-related brain atrophy 328 and dementia 329.

Lipids likely play a fundamental role in our susceptibility to CVD. Indeed many other mammals are relatively resistant to atherosclerosis and their use as experimental models requires ‘humanisation’ of the lipid profile, by genetically increasing apoB-lipoproteins (e.g. APOE–/– or LDLR–/– mice) and/or feeding a high cholesterol/high fat diet 330. Such experimental atherosclerosis requires ACAT2 (aka. SOAT2), which is expressed in intestine and liver where it synthesises MUFA-rich CEs for apoB-lipoproteins 331,332, and mediates LDL proteoglycan binding 333 and aggregation 195. However, in humans a high oleic oil diet enriched CEs but lowered LDL biglycan binding 333, suggesting further differences; of note, other mammals may express higher ACAT2 334 and are often fed higher dietary cholesterol 89,333. Experimental atherosclerosis also requires NLRP3, which links microbial and/or sterile cell stress to innate immunity 61. Today many infections are associated with atherosclerosis and the acute-phase response encompasses mutual changes supporting immunometabolism and defence reminiscent of those implicated in CVD 335,336. For instance, infections/inflammation induce macrophage aerobic glycolysis and accumulation of lipid droplets with antimicrobial activity 337,338, while also modulating systemic insulin sensitivity (i.e. glucose metabolism) 339,340, lipid metabolism (e.g. lipolysis, cholesterol and sphingolipids) and lipoprotein modifications (e.g. oxLDL) 336. Induction of vascular retention and LDL oxidation might even support bacterial sequestration (e.g. LPS­–TLR4 204,232 and M. tuberculosis 50) and phagocyte clearance (e.g. LPS 233,234). These mechanisms could therefore support acute survival, while persistent stimulation exacerbates vascular disease hastening late-life mortality 336, as a form of antagonistic pleiotropy. Indeed while CVD is currently the leading cause of death, ancestrally it was likely injury and infections, underscoring the evolutionary priority.

Many bacterial pathogens are Gram-negative with a distinct outer membrane coated in LPS, which confers barrier function and protection from antibiotics 341. The specific composition of LPS varies between bacteria and is modulated by environmental factors 341. In pathogenic bacteria the lipid A region is typically hexa-acylated with SFAs 310, which may impart a gel-like state and low permeability, while also mediating activation of innate immunity in mammalian cells via Lo microdomains, which are similarly enriched in SFAs 342. As above, SFA-rich diets may act on gut and liver to increase circulating immunogenic lipids (e.g. cholesterol, ceramides and LPS) which converge on membrane microdomains. Further, in healthy adults SFA intake and SCD (aka. 9-desaturase) activity also correlated the kynurenine/tryptophan ratio (a surrogate of IFNg/Th1 activity), which itself correlated CRP 343. In mice SFA-rich diets (i.e. C12/16:0) exacerbated central autoimmunity by increasing Th1/17 activity via the small intestine 344,345. Th17 cells express particularly high levels of TLR4, and LPS directly induces Th17 differentiation in vitro 346. Accordingly, Th17 lymphocytes are part of type-3 immunity which mediates antimicrobial responses to extracellular pathogens (e.g. Gram-negative bacteria), although when dysregulated also autoimmunity 347. In addition, in Rag1–/– mice lacking adaptive immunity, a SFA-based ketogenic formula enhanced clearance of C. albicans, while simultaneously conferring susceptibility to endotoxemia, which involved palmitate and ceramide, persisted for 7 days post-exposure, and was reversible with oleate 348. This was consistent with SFA induction of innate immune memory (i.e. ‘trained immunity’), itself another double-edged sword 349. As above, plasma cholesterol may similarly stimulate immune cells, and while treatment of human hypercholesterolemia with statins normalised monocyte skewing and lipid droplets, an activated phenotype persisted, again implicating trained immunity 65. Dietary cholesterol may also affect the pathophysiology of infectious and autoimmune disease (reviewed in 350).

The ability of exogenous lipids to differentially regulate endogenous lipids and physiology may arise from several key factors. Foremost, the dependence of cell membrane biophysics and signalling on specific fatty acids means lipid saturation must be tightly regulated; in particular, excess long-chain SFAs can induce ER stress/inflammasome pathways offset by UFAs 274,320,321. Accordingly, the ER-associated enzyme SCD1 mediates endogenous desaturation of SFAs to MUFAs and limits experimental atherosclerosis and macrophage TLR4 hypersensitivity, potentially via suppression of Lo microdomains, albeit at the cost of obesity-related metabolic disease 351. However, mammalian fatty acid metabolism is imperfect and especially limited at PUFA synthesis, underlying the essential fatty acids (i.e. C18:2/3) and modulation by diet. Similar issues pertain to cholesterol, which is also fundamental to membrane biophysics, but in excess can precipitate as crystals and exert toxicity. While most cells can synthesise cholesterol its catabolism is limited to steroidogenic and hepatic tissues, so excess must be esterified for safe storage or undergo efflux and transport to the liver and intestine for biliary and direct excretion, respectively. Fatty acid and cholesterol metabolism directly converge on the formation of cholesteryl esters, which have a preference for UFAs (i.e. ACAT 89,275 and LCAT 276–278), suggesting their availability may influence cholesterol turnover and therein processes of uptake/efflux, transport and crystallisation. Further, the role of lipids and lipoproteins in immunity may superimpose another layer of regulation mostly concerned with acute survival 336.

Consequently, these metabolic constraints and connections create a susceptibility to different foods and associated environments, which themselves may confer adaptive or maladaptive effects on short-term fitness or long-term health, in concert with genomic variation. How might this play out through our evolution? The other great apes from which we diverged are highly plant-based (i.e. fruits and leaves), after which our diet became increasingly diverse and animal-based with our spread to colder environments and eventually pastoralism 352. An increase in dietary cholesterol and fats from animals (i.e. land and marine) and plants (esp. nuts/seeds) may have modulated plasma cholesterol in relation to the SFA/PUFA ratio 76,77,84,89,90. Notably, since meat can deteriorate quickly, this might have also increased exposure to pathogens, consistent with our relatively low stomach acid pH (i.e. similar to scavengers) 352 and animal food aversion during early pregnancy (i.e. morning sickness), a relatively immune-suppressed period 353,354. The advent of cooking would support sterilisation and may be reinforced by the appealing sensory qualities of advanced glycation end products (AGEs) 355, albeit at the potential cost of cardiometabolic dysregulation, as seen in modern humans 356. Despite these factors native populations often exhibit relative cardiometabolic health 324,325. In particular, our genus and species emerged in Africa, where remaining hunter-gatherer exemplars such as the Hadza of Northern Tanzania maintain a low body weight, blood pressure and plasma lipids throughout life, with a relatively low fat intake (i.e. median ~18% kcals) from plants and lean meats 352. Similarly, Tsimane vascular health is accompanied by low plasma/LDL cholesterol 326, with a very low fat and SFA intake (i.e. men: 15.1% and 3.7% kcals, respectively) from a plant-dominant diet with moderate fish/meat 357. Furthermore, in this energy-limited and pathogenically diverse context, the ancestral APOE4 allele is actually associated with better cognition in those infected with parasites 358, and slightly increased lipids (i.e. cholesterol +2.8% and oxLDL +3.9%) and lower innate immune markers (e.g. CRP –21.6%), which were inversely associated in those with a lower BMI only 359, suggesting it may not have the same deleterious effects as in post-industrial populations, but instead support cognition and immunity. Notably, in a modern UK population, carriers of the APOE4 allele (vs. E3/E3 and E2) had higher LDL-c and apoB, but which was the most responsive to lowering when replacing SFAs with low GI carbs, not MUFAs 79.

Industrialization has led to another nutrition transition, wherein major SFA sources are now grain-fed meats, which are richer in total fat and specific fatty acids 360, along with concentrated animal/dairy and tropical fats (as used in SFA trials herein), in the context of a diet high in ultra-processed foods/calories, with a high n-6/3 PUFA ratio, and low in micro-/phytonutrients, implicating evolutionary mismatch in SFA-associated diseases 325. Indeed many nutritional and physiological factors may modulate the effects of dietary SFAs today. For instance, SFA-induced postprandial inflammation may especially occur in obesity 247,249,361, but be blunted by lipid-lowering therapy 247 or co-ingestion of phytochemicals (e.g. polyphenols 362, spices 363 and fibre 364), which can also accompany UFAs (e.g. olive oil and nuts). Long-chain n-3 PUFA status may also be important via anti-LPS/TLR effects 248,307,309; cellular levels are also highly responsive to diet at the expense of n-6 PUFAs 365 (see supplement). SFA-induced liver fat may especially be promoted by overfeeding 188,189,238,256, and exacerbated by excess fructose 262. Conversely, low carb diets increase fat oxidation and may mitigate the differential effects of SFAs (vs. UFAs) on insulin sensitivity and inflammation, but not cholesterol and ketones 70–73. Further, ketosis may have inherent anti-inflammatory effects 366, and a 3-day isocaloric ketogenic diet suppressed LPS/palmitate-induced inflammasome activation in macrophages ex vivo 367.

While many factors may contribute to atherogenesis, as a condition of arterial lipid accumulation, a lipid-threshold may ultimately govern its progression 368,369; indeed atherosclerosis rarely occurs in mammals and humans with an LDL-c <80mg/dl 50,267. Accordingly, in the PESA study of a healthy middle-aged Spanish cohort 30, systemic subclinical atherosclerosis became increasingly rare and undetectable at low LDL-c values, albeit with an increasingly small sample size 31. Moreover, decades of research on experimental atherosclerosis shows that most aspects of advanced plaques can regress/reverse, including necrotic and crystalline material, in association with dramatic lipid lowering and improved HDL function 60,370,371. Similarly, in humans athero-regression can be induced with intensive lifestyle changes 372 (with <SFAs 68,190) and/or lipid-lowering drugs 373 (with LDL-c <80mg/dl 374,375); and some extreme cases have been reported 376,377. In fact when considering other mammals, newborn humans and native populations, these low cholesterol levels may even be physiologically normal 267,324; in which case athero-regression could simply reflect a return to the natural homeostatic state. So despite its ubiquity, perhaps atherosclerotic CVD is not inevitable, at least not without a chronic deviation from physiological homeostasis, as a result of relatively rare mutations (i.e. familial hypercholesterolemia), or more generally, a maladaptive environment.

References

1.           Kromhout, D. et al. Comparative ecologic relationships of saturated fat, sucrose, food groups, and a Mediterranean food pattern score to 50-year coronary heart disease mortality rates among 16 cohorts of the Seven Countries Study. Eur. J. Clin. Nutr. 72, 1103–1110 (2018).

2.           Siri-Tarino, P. W., Sun, Q., Hu, F. B. & Krauss, R. M. Meta-analysis of prospective cohort studies evaluating the association of saturated fat with cardiovascular disease. Am. J. Clin. Nutr. 91, 535–46 (2010).

3.           Wang, D. D. et al. Association of Specific Dietary Fats With Total and Cause-Specific Mortality. JAMA Intern. Med. 176, 1134–45 (2016).

4.           Li, Y. et al. Saturated Fats Compared With Unsaturated Fats and Sources of Carbohydrates in Relation to Risk of Coronary Heart Disease: A Prospective Cohort Study. J. Am. Coll. Cardiol. 66, 1538–1548 (2015).

5.           Chen, M. et al. Dairy fat and risk of cardiovascular disease in 3 cohorts of US adults. Am. J. Clin. Nutr. 104, 1209–1217 (2016).

6.           Guasch-Ferré, M. et al. Consumption of Olive Oil and Risk of Total and Cause-Specific Mortality Among U.S. Adults. J. Am. Coll. Cardiol. 79, 101–112 (2022).

7.           Guasch-Ferré, M. et al. Associations of Monounsaturated Fatty Acids From Plant and Animal Sources With Total and Cause-Specific Mortality in Two US Prospective Cohort Studies. Circ. Res. 124, 1266–1275 (2019).

8.           Zong, G. et al. Monounsaturated fats from plant and animal sources in relation to risk of coronary heart disease among US men and women. Am. J. Clin. Nutr. 107, 445–453 (2018).

9.           Jakobsen, M. U. et al. Intake of carbohydrates compared with intake of saturated fatty acids and risk of myocardial infarction: importance of the glycemic index. Am. J. Clin. Nutr. 91, 1764–8 (2010).

10.        Guasch-Ferré, M. et al. Dietary fat intake and risk of cardiovascular disease and all-cause mortality in a population at high risk of cardiovascular disease. Am. J. Clin. Nutr. 102, 1563–73 (2015).

11.        Mölenberg, F. J. M. et al. Dietary fatty acid intake after myocardial infarction: a theoretical substitution analysis of the Alpha Omega Cohort. Am. J. Clin. Nutr. 106, 895–901 (2017).

12.        Jakobsen, M. U. et al. Major types of dietary fat and risk of coronary heart disease: a pooled analysis of 11 cohort studies. Am. J. Clin. Nutr. 89, 1425–32 (2009).

13.        Farvid, M. S. et al. Dietary linoleic acid and risk of coronary heart disease: a systematic review and meta-analysis of prospective cohort studies. Circulation 130, 1568–78 (2014).

14.        Hooper, L. et al. Reduction in saturated fat intake for cardiovascular disease. Cochrane database Syst. Rev. 5, CD011737 (2020).

15.        Clarke, R., Frost, C., Collins, R., Appleby, P. & Peto, R. Dietary lipids and blood cholesterol: quantitative meta-analysis of metabolic ward studies. BMJ 314, 112–7 (1997).

16.        Mensink, R. P. Effects of saturated fatty acids on serum lipids and lipoproteins: a systematic review and regression analysis. (World Health Organization., 2016).

17.        Rocha, D. M., Bressan, J. & Hermsdorff, H. H. The role of dietary fatty acid intake in inflammatory gene expression: a critical review. Sao Paulo Med. J. 135, 157–168 (2017).

18.        Cândido, T. L. N. et al. Effects of dietary fat quality on metabolic endotoxaemia: a systematic review. Br. J. Nutr. 124, 654–667 (2020).

19.        Imamura, F. et al. Effects of Saturated Fat, Polyunsaturated Fat, Monounsaturated Fat, and Carbohydrate on Glucose-Insulin Homeostasis: A Systematic Review and Meta-analysis of Randomised Controlled Feeding Trials. PLoS Med. 13, e1002087 (2016).

20.        Hydes, T., Alam, U. & Cuthbertson, D. J. The Impact of Macronutrient Intake on Non-alcoholic Fatty Liver Disease (NAFLD): Too Much Fat, Too Much Carbohydrate, or Just Too Many Calories? Front. Nutr. 8, 640557 (2021).

21.        Weech, M. et al. Replacement of dietary saturated fat with unsaturated fats increases numbers of circulating endothelial progenitor cells and decreases numbers of microparticles: findings from the randomized, controlled Dietary Intervention and VAScular function (DIVAS) s. Am. J. Clin. Nutr. 107, 876–882 (2018).

22.        Jakubowski, J. A. & Ardlie, N. G. Modification of human platelet function by a diet enriched in saturated or polyunsaturated fat. Atherosclerosis 31, 335–44 (1978).

23.        Diamond, D. M., Bikman, B. T. & Mason, P. Statin therapy is not warranted for a person with high LDL-cholesterol on a low-carbohydrate diet. Curr. Opin. Endocrinol. Diabetes. Obes. 29, 497–511 (2022).

24.        Okuyama, H. et al. A Critical Review of the Consensus Statement from the European Atherosclerosis Society Consensus Panel 2017. Pharmacology 101, 184–218 (2018).

25.        Astrup, A. et al. Saturated Fats and Health: A Reassessment and Proposal for Food-Based Recommendations: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 76, 844–857 (2020).

26.        Ravnskov, U. et al. The questionable benefits of exchanging saturated fat with polyunsaturated fat. Mayo Clin. Proc. 89, 451–3 (2014).

27.        Allison, M. A., Criqui, M. H. & Wright, C. M. Patterns and risk factors for systemic calcified atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 24, 331–6 (2004).

28.        Lambert, M. A. et al. Prevalence and Distribution of Atherosclerosis in a Low- to Intermediate-Risk Population: Assessment with Whole-Body MR Angiography. Radiology 287, 795–804 (2018).

29.        Leiner, T. Whole-Body MR Angiography: Assessing the Global Burden of Cardiovascular Disease. Radiology 287, 805–807 (2018).

30.        Ibanez, B. et al. Progression of Early Subclinical Atherosclerosis (PESA) Study: JACC Focus Seminar 7/8. J. Am. Coll. Cardiol. 78, 156–179 (2021).

31.        Fernández-Friera, L. et al. Normal LDL-Cholesterol Levels Are Associated With Subclinical Atherosclerosis in the Absence of Risk Factors. J. Am. Coll. Cardiol. 70, 2979–2991 (2017).

32.        Allam, A. H. et al. Atherosclerosis in ancient and modern Egyptians: the Horus study. Glob. Heart 9, 197–202 (2014).

33.        Small, D. M. George Lyman Duff memorial lecture. Progression and regression of atherosclerotic lesions. Insights from lipid physical biochemistry. Arteriosclerosis 8, 103–29 (1988).

34.        Melaku, L. & Dabi, A. The cellular biology of atherosclerosis with atherosclerotic lesion classification and biomarkers. Bull. Natl. Res. Cent. 45, (2021).

35.        Baumer, Y., Mehta, N. N., Dey, A. K., Powell-Wiley, T. M. & Boisvert, W. A. Cholesterol crystals and atherosclerosis. Eur. Heart J. 41, 2236–2239 (2020).

36.        Lehti, S. et al. Extracellular Lipids Accumulate in Human Carotid Arteries as Distinct Three-Dimensional Structures and Have Proinflammatory Properties. Am. J. Pathol. 188, 525–538 (2018).

37.        Nakashima, Y., Wight, T. N. & Sueishi, K. Early atherosclerosis in humans: role of diffuse intimal thickening and extracellular matrix proteoglycans. Cardiovasc. Res. 79, 14–23 (2008).

38.        Ference, B. A. et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 38, 2459–2472 (2017).

39.        Masrouri, S., Tamehri Zadeh, S. S., Shapiro, M. D., Khalili, D. & Hadaegh, F. Impact of optimal cholesterol levels on subclinical atherosclerosis in the absence of risk factors in young adults. Atherosclerosis 117520 (2024). doi:10.1016/j.atherosclerosis.2024.117520

40.        Abdullah, S. M. et al. Long-Term Association of Low-Density Lipoprotein Cholesterol With Cardiovascular Mortality in Individuals at Low 10-Year Risk of Atherosclerotic Cardiovascular Disease. Circulation 138, 2315–2325 (2018).

41.        Postmus, I. et al. LDL cholesterol still a problem in old age? A Mendelian randomization study. Int. J. Epidemiol. 44, 604–12 (2015).

42.        Sniderman, A. D. et al. Apolipoprotein B Particles and Cardiovascular Disease: A Narrative Review. JAMA Cardiol. 4, 1287–1295 (2019).

43.        Glavinovic, T. et al. Physiological Bases for the Superiority of Apolipoprotein B Over Low-Density Lipoprotein Cholesterol and Non-High-Density Lipoprotein Cholesterol as a Marker of Cardiovascular Risk. J. Am. Heart Assoc. 11, e025858 (2022).

44.        Helgadottir, A. et al. Cholesterol not particle concentration mediates the atherogenic risk conferred by apolipoprotein B particles: a Mendelian randomization analysis. Eur. J. Prev. Cardiol. 29, 2374–2385 (2022).

45.        Goldstein, J. L. & Brown, M. S. The LDL receptor. Arterioscler. Thromb. Vasc. Biol. 29, 431–8 (2009).

46.        Ogura, M. HDL, cholesterol efflux, and ABCA1: Free from good and evil dualism. J. Pharmacol. Sci. 150, 81–89 (2022).

47.        Rosales, C., Gillard, B. K., Xu, B., Gotto, A. M. & Pownall, H. J. Revisiting Reverse Cholesterol Transport in the Context of High-Density Lipoprotein Free Cholesterol Bioavailability. Methodist Debakey Cardiovasc. J. 15, 47–54 (2019).

48.        Schwartz, C. C., VandenBroek, J. M. & Cooper, P. S. Lipoprotein cholesteryl ester production, transfer, and output in vivo in humans. J. Lipid Res. 45, 1594–607 (2004).

49.        Williams, K. J. & Tabas, I. The response-to-retention hypothesis of early atherogenesis. Arterioscler. Thromb. Vasc. Biol. 15, 551–61 (1995).

50.        Borén, J. & Williams, K. J. The central role of arterial retention of cholesterol-rich apolipoprotein-B-containing lipoproteins in the pathogenesis of atherosclerosis: a triumph of simplicity. Curr. Opin. Lipidol. 27, 473–83 (2016).

51.        Guyton, J. R. & Klemp, K. F. Development of the lipid-rich core in human atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 16, 4–11 (1996).

52.        Nielsen, L. B. Transfer of low density lipoprotein into the arterial wall and risk of atherosclerosis. Atherosclerosis 123, 1–15 (1996).

53.        Oörni, K., Pentikäinen, M. O., Ala-Korpela, M. & Kovanen, P. T. Aggregation, fusion, and vesicle formation of modified low density lipoprotein particles: molecular mechanisms and effects on matrix interactions. J. Lipid Res. 41, 1703–14 (2000).

54.        Öörni, K. & Kovanen, P. T. Aggregation Susceptibility of Low-Density Lipoproteins-A Novel Modifiable Biomarker of Cardiovascular Risk. J. Clin. Med. 10, (2021).

55.        Borén, J. et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease: pathophysiological, genetic, and therapeutic insights: a consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 41, 2313–2330 (2020).

56.        Mora, S. Advanced lipoprotein testing and subfractionation are not (yet) ready for routine clinical use. Circulation 119, 2396–404 (2009).

57.        Steinbrecher, U. P. & Lougheed, M. Scavenger receptor-independent stimulation of cholesterol esterification in macrophages by low density lipoprotein extracted from human aortic intima. Arterioscler. Thromb.  a J. Vasc. Biol. 12, 608–25 (1992).

58.        Mason, R. P., Walter, M. F. & Jacob, R. F. Effects of HMG-CoA reductase inhibitors on endothelial function: role of microdomains and oxidative stress. Circulation 109, II34-41 (2004).

59.        Baumer, Y. et al. Hyperlipidemia-induced cholesterol crystal production by endothelial cells promotes atherogenesis. Nat. Commun. 8, 1129 (2017).

60.        Zhang, Y. et al. Coronary endothelial dysfunction induced by nucleotide oligomerization domain-like receptor protein with pyrin domain containing 3 inflammasome activation during hypercholesterolemia: beyond inflammation. Antioxid. Redox Signal. 22, 1084–96 (2015).

61.        Duewell, P. et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464, 1357–61 (2010).

62.        Altabas, V. & Biloš, L. S. K. The Role of Endothelial Progenitor Cells in Atherosclerosis and Impact of Anti-Lipemic Treatments on Endothelial Repair. Int. J. Mol. Sci. 23, (2022).

63.        Rodríguez, C. et al. Modulation of endothelium and endothelial progenitor cell function by low-density lipoproteins: implication for vascular repair, angiogenesis and vasculogenesis. Pathobiology 76, 11–22 (2009).

64.        Wang, N. & Tall, A. R. Cholesterol in platelet biogenesis and activation. Blood 127, 1949–53 (2016).

65.        Stiekema, L. C. A. et al. Impact of cholesterol on proinflammatory monocyte production by the bone marrow. Eur. Heart J. 42, 4309–4320 (2021).

66.        Soehnlein, O. & Swirski, F. K. Hypercholesterolemia links hematopoiesis with atherosclerosis. Trends Endocrinol. Metab. 24, 129–36 (2013).

67.        Xu, L. et al. Foamy monocytes form early and contribute to nascent atherosclerosis in mice with hypercholesterolemia. Arterioscler. Thromb. Vasc. Biol. 35, 1787–97 (2015).

68.        Watts, G. F., Jackson, P., Burke, V. & Lewis, B. Dietary fatty acids and progression of coronary artery disease in men. Am. J. Clin. Nutr. 64, 202–9 (1996).

69.        Müller, H., Kirkhus, B. & Pedersen, J. I. Serum cholesterol predictive equations with special emphasis on trans and saturated fatty acids. an analysis from designed controlled studies. Lipids 36, 783–91 (2001).

70.        Lundsgaard, A.-M. et al. Mechanisms Preserving Insulin Action during High Dietary Fat Intake. Cell Metab. 29, 50-63.e4 (2019).

71.        Forsythe, C. E. et al. Limited effect of dietary saturated fat on plasma saturated fat in the context of a low carbohydrate diet. Lipids 45, 947–62 (2010).

72.        Fuehrlein, B. S. et al. Differential metabolic effects of saturated versus polyunsaturated fats in ketogenic diets. J. Clin. Endocrinol. Metab. 89, 1641–5 (2004).

73.        Nathan, J., Bailur, S., Datay, K., Sharma, S. & Khedekar Kale, D. A Switch to Polyunsaturated Fatty Acid Based Ketogenic Diet Improves Seizure Control in Patients with Drug-resistant Epilepsy on the Mixed Fat Ketogenic Diet: A Retrospective Open Label Trial. Cureus 11, e6399 (2019).

74.        Soto-Mota, A. et al. Increased LDL-cholesterol on a low-carbohydrate diet in adults with normal but not high body weight: a meta-analysis. Am. J. Clin. Nutr. (2024). doi:10.1016/j.ajcnut.2024.01.009

75.        Norwitz, N. G., Soto-Mota, A., Feldman, D., Parpos, S. & Budoff, M. Case Report: Hypercholesterolemia “Lean Mass Hyper-Responder” Phenotype Presents in the Context of a Low Saturated Fat Carbohydrate-Restricted Diet. Front. Endocrinol. (Lausanne). 13, (2022).

76.        Fielding, C. J. et al. Effects of dietary cholesterol and fat saturation on plasma lipoproteins in an ethnically diverse population of healthy young men. J. Clin. Invest. 95, 611–8 (1995).

77.        Schonfeld, G. et al. Effects of dietary cholesterol and fatty acids on plasma lipoproteins. J. Clin. Invest. 69, 1072–80 (1982).

78.        Brassard, D. et al. Comparison of the impact of SFAs from cheese and butter on cardiometabolic risk factors: a randomized controlled trial. Am. J. Clin. Nutr. 105, 800–809 (2017).

79.        Griffin, B. A. et al. 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 10, 1524 (2018).

80.        Calabuig-Navarro, M. V et al. A randomized trial and novel SPR technique identifies altered lipoprotein-LDL receptor binding as a mechanism underlying elevated LDL-cholesterol in APOE4s. Sci. Rep. 7, 44119 (2017).

81.        Moreno, J. A. et al. Apolipoprotein E gene promoter -219G->T polymorphism increases LDL-cholesterol concentrations and susceptibility to oxidation in response to a diet rich in saturated fat. Am. J. Clin. Nutr. 80, 1404–9 (2004).

82.        Ulven, S. M. et al. Using metabolic profiling and gene expression analyses to explore molecular effects of replacing saturated fat with polyunsaturated fat-a randomized controlled dietary intervention study. Am. J. Clin. Nutr. 109, 1239–1250 (2019).

83.        Mustad, V. A. et al. Reducing saturated fat intake is associated with increased levels of LDL receptors on mononuclear cells in healthy men and women. J. Lipid Res. 38, 459–68 (1997).

84.        Bergeron, N., Chiu, S., Williams, P. T., M King, S. & Krauss, R. M. Effects of red meat, white meat, and nonmeat protein sources on atherogenic lipoprotein measures in the context of low compared with high saturated fat intake: a randomized controlled trial. Am. J. Clin. Nutr. 110, 24–33 (2019).

85.        Chiu, S., Williams, P. T. & Krauss, R. M. Effects of a very high saturated fat diet on LDL particles in adults with atherogenic dyslipidemia: A randomized controlled trial. PLoS One 12, 1–14 (2017).

86.        Dias, C. B. et al. Improvement of the omega 3 index of healthy subjects does not alter the effects of dietary saturated fats or n-6PUFA on LDL profiles. Metabolism. 68, 11–19 (2017).

87.        Shepherd, J. et al. Effects of saturated and polyunsaturated fat diets on the chemical composition and metabolism of low density lipoproteins in man. J. Lipid Res. 21, 91–9 (1980).

88.        Fernandez, M. L. & West, K. L. Mechanisms by which dietary fatty acids modulate plasma lipids. J. Nutr. 135, 2075–8 (2005).

89.        Dietschy, J. M. Dietary fatty acids and the regulation of plasma low density lipoprotein cholesterol concentrations. J. Nutr. 128, 444S-448S (1998).

90.        Hayes, K. C., Khosla, P., Hajri, T. & Pronczuk, A. Saturated fatty acids and LDL receptor modulation in humans and monkeys. Prostaglandins. Leukot. Essent. Fatty Acids 57, 411–8 (1997).

91.        Seo, T. et al. Saturated fat-rich diet enhances selective uptake of LDL cholesteryl esters in the arterial wall. J. Clin. Invest. 115, 2214–22 (2005).

92.        Chang, C. L., Torrejon, C., Jung, U. J., Graf, K. & Deckelbaum, R. J. Incremental replacement of saturated fats by n-3 fatty acids in high-fat, high-cholesterol diets reduces elevated plasma lipid levels and arterial lipoprotein lipase, macrophages and atherosclerosis in LDLR-/- mice. Atherosclerosis 234, 401–9 (2014).

93.        Sudheendran, S., Chang, C. C. & Deckelbaum, R. J. N-3 vs. saturated fatty acids: effects on the arterial wall. Prostaglandins. Leukot. Essent. Fatty Acids 82, 205–9 (2010).

94.        Lian, Z. et al. Replacing Saturated Fat With Unsaturated Fat in Western Diet Reduces Foamy Monocytes and Atherosclerosis in Male Ldlr-/- Mice. Arterioscler. Thromb. Vasc. Biol. 40, 72–85 (2020).

95.        Peng, X. & Wu, H. Inflammatory Links Between Hypertriglyceridemia and Atherogenesis. Curr. Atheroscler. Rep. 24, 297–306 (2022).

96.        Stürzebecher, P. E., Katzmann, J. L. & Laufs, U. What is ‘remnant cholesterol’? Eur. Heart J. 2, 1–3 (2023).

97.        Desmarchelier, C., Borel, P., Lairon, D., Maraninchi, M. & Valéro, R. Effect of Nutrient and Micronutrient Intake on Chylomicron Production and Postprandial Lipemia. Nutrients 11, (2019).

98.        Benson, T. W. et al. A single high-fat meal provokes pathological erythrocyte remodeling and increases myeloperoxidase levels: implications for acute coronary syndrome. Lab. Invest. 98, 1300–1310 (2018).

99.        Varela, L. M. et al. A high-fat meal promotes lipid-load and apolipoprotein B-48 receptor transcriptional activity in circulating monocytes. Am. J. Clin. Nutr. 93, 918–25 (2011).

100.      den Hartigh, L. J., Connolly-Rohrbach, J. E., Fore, S., Huser, T. R. & Rutledge, J. C. Fatty acids from very low-density lipoprotein lipolysis products induce lipid droplet accumulation in human monocytes. J. Immunol. 184, 3927–36 (2010).

101.      Gower, R. M. et al. CD11c/CD18 expression is upregulated on blood monocytes during hypertriglyceridemia and enhances adhesion to vascular cell adhesion molecule-1. Arterioscler. Thromb. Vasc. Biol. 31, 160–6 (2011).

102.      Botham, K. M., Moore, E. H., De Pascale, C. & Bejta, F. The induction of macrophage foam cell formation by chylomicron remnants. Biochem. Soc. Trans. 35, 454–8 (2007).

103.      Jackson, K. G., Maitin, V., Leake, D. S., Yaqoob, P. & Williams, C. M. Saturated fat-induced changes in Sf 60-400 particle composition reduces uptake of LDL by HepG2 cells. J. Lipid Res. 47, 393–403 (2006).

104.      Galloway, S. et al. The differential effects of fatty acids on enterocytic abundance of amyloid-beta. Lipids Health Dis. 18, 209 (2019).

105.      Galloway, S., Takechi, R., Pallebage-Gamarallage, M. M. S., Dhaliwal, S. S. & Mamo, J. C. L. Amyloid-beta colocalizes with apolipoprotein B in absorptive cells of the small intestine. Lipids Health Dis. 8, 46 (2009).

106.      Takechi, R. et al. Differential effects of dietary fatty acids on the cerebral distribution of plasma-derived apo B lipoproteins with amyloid-beta. Br. J. Nutr. 103, 652–62 (2010).

107.      D’Alonzo, Z. J. et al. Peripheral metabolism of lipoprotein-amyloid beta as a risk factor for Alzheimer’s disease: potential interactive effects of APOE genotype with dietary fats. Genes Nutr. 18, (2023).

108.      Wolters, F. J. et al. Plasma amyloid-β40 in relation to subclinical atherosclerosis and cardiovascular disease: A population-based study. Atherosclerosis 348, 44–50 (2022).

109.      Schulz, B. et al. Beta-amyloid (Abeta40, Abeta42) binding to modified LDL accelerates macrophage foam cell formation. Biochim. Biophys. Acta 1771, 1335–44 (2007).

110.      Martínez-González, J., Raposo, B., Rodríguez, C. & Badimon, L. 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition prevents endothelial NO synthase downregulation by atherogenic levels of native LDLs: balance between transcriptional and posttranscriptional regulation. Arterioscler. Thromb. Vasc. Biol. 21, 804–9 (2001).

111.      Rodríguez, C., Raposo, B., Martínez-González, J., Casaní, L. & Badimon, L. Low density lipoproteins downregulate lysyl oxidase in vascular endothelial cells and the arterial wall. Arterioscler. Thromb. Vasc. Biol. 22, 1409–14 (2002).

112.      Kruth, H. S. Fluid-phase pinocytosis of LDL by macrophages: a novel target to reduce macrophage cholesterol accumulation in atherosclerotic lesions. Curr. Pharm. Des. 19, 5865–72 (2013).

113.      Meyer, J. M., Ji, A., Cai, L. & van der Westhuyzen, D. R. High-capacity selective uptake of cholesteryl ester from native LDL during macrophage foam cell formation. J. Lipid Res. 53, 2081–2091 (2012).

114.      Calvayrac, O. et al. CCL20 is increased in hypercholesterolemic subjects and is upregulated by LDL in vascular smooth muscle cells: role of NF-κB. Arterioscler. Thromb. Vasc. Biol. 31, 2733–41 (2011).

115.      Gao, S. & Liu, J. Association between circulating oxidized low-density lipoprotein and atherosclerotic cardiovascular disease. Chronic Dis. Transl. Med. 3, 89–94 (2017).

116.      Ishigaki, Y., Oka, Y. & Katagiri, H. Circulating oxidized LDL: a biomarker and a pathogenic factor. Curr. Opin. Lipidol. 20, 363–9 (2009).

117.      Wu, T. et al. Is plasma oxidized low-density lipoprotein, measured with the widely used antibody 4E6, an independent predictor of coronary heart disease among U.S. men and women? J. Am. Coll. Cardiol. 48, 973–9 (2006).

118.      Koskinen, J. et al. Apolipoprotein B, oxidized low-density lipoprotein, and LDL particle size in predicting the incidence of metabolic syndrome: the Cardiovascular Risk in Young Finns study. Eur. J. Prev. Cardiol. 19, 1296–303 (2012).

119.      Tsimikas, S. & Witztum, J. L. Oxidized phospholipids in cardiovascular disease. Nat. Rev. Cardiol. (2023). doi:10.1038/s41569-023-00937-4

120.      Shoji, T. et al. Inverse relationship between circulating oxidized low density lipoprotein (oxLDL) and anti-oxLDL antibody levels in healthy subjects. Atherosclerosis 148, 171–7 (2000).

121.      Itabe, H., Obama, T. & Kato, R. The Dynamics of Oxidized LDL during Atherogenesis. J. Lipids 2011, 418313 (2011).

122.      van der Valk, F. M. et al. Increased haematopoietic activity in patients with atherosclerosis. Eur. Heart J. 38, 425–432 (2017).

123.      Sheedy, F. J. et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 14, 812–20 (2013).

124.      Cornelissen, A., Guo, L., Sakamoto, A., Virmani, R. & Finn, A. V. New insights into the role of iron in inflammation and atherosclerosis. EBioMedicine 47, 598–606 (2019).

125.      Baker, C. S. et al. Cyclooxygenase-2 is widely expressed in atherosclerotic lesions affecting native and transplanted human coronary arteries and colocalizes with inducible nitric oxide synthase and nitrotyrosine particularly in macrophages. Arterioscler. Thromb. Vasc. Biol. 19, 646–55 (1999).

126.      Cromheeke, K. M. et al. Inducible nitric oxide synthase colocalizes with signs of lipid oxidation/peroxidation in human atherosclerotic plaques. Cardiovasc. Res. 43, 744–54 (1999).

127.      Depre, C., Havaux, X., Renkin, J., Vanoverschelde, J. L. & Wijns, W. Expression of inducible nitric oxide synthase in human coronary atherosclerotic plaque. Cardiovasc. Res. 41, 465–72 (1999).

128.      Ohishi, M. et al. Increased expression and co-localization of ACE, angiotensin II AT(1) receptors and inducible nitric oxide synthase in atherosclerotic human coronary arteries. Int. J. Physiol. Pathophysiol. Pharmacol. 2, 111–24 (2010).

129.      Buttery, L. D. et al. Inducible nitric oxide synthase is present within human atherosclerotic lesions and promotes the formation and activity of peroxynitrite. Lab. Invest. 75, 77–85 (1996).

130.      Nadel, J., Jabbour, A. & Stocker, R. Arterial myeloperoxidase in the detection and treatment of vulnerable atherosclerotic plaque: a new dawn for an old light. Cardiovasc. Res. (2022). doi:10.1093/cvr/cvac081

131.      Marsche, G., Stadler, J. T., Kargl, J. & Holzer, M. Understanding Myeloperoxidase-Induced Damage to HDL Structure and Function in the Vessel Wall: Implications for HDL-Based Therapies. Antioxidants (Basel, Switzerland) 11, (2022).

132.      Nicholls, S. J. & Hazen, S. L. Myeloperoxidase, modified lipoproteins, and atherogenesis. J. Lipid Res. 50, (2009).

133.      Stocker, R. & Keaney, J. F. Role of oxidative modifications in atherosclerosis. Physiol. Rev. 84, 1381–478 (2004).

134.      Podrez, E. A., Schmitt, D., Hoff, H. F. & Hazen, S. L. Myeloperoxidase-generated reactive nitrogen species convert LDL into an atherogenic form in vitro. J. Clin. Invest. 103, 1547–60 (1999).

135.      Podrez, E. A. et al. Macrophage scavenger receptor CD36 is the major receptor for LDL modified by monocyte-generated reactive nitrogen species. J. Clin. Invest. 105, 1095–108 (2000).

136.      Sanda, G. M. et al. Aggregated LDL turn human macrophages into foam cells and induce mitochondrial dysfunction without triggering oxidative or endoplasmic reticulum stress. PLoS One 16, e0245797 (2021).

137.      Asmis, R. & Jelk, J. Large variations in human foam cell formation in individuals: a fully autologous in vitro assay based on the quantitative analysis of cellular neutral lipids. Atherosclerosis 148, 243–53 (2000).

138.      Meyer, J. M., Ji, A., Cai, L. & van der Westhuyzen, D. R. Minimally oxidized LDL inhibits macrophage selective cholesteryl ester uptake and native LDL-induced foam cell formation. J. Lipid Res. 55, 1648–56 (2014).

139.      Hutchins, P. M., Moore, E. E. & Murphy, R. C. Electrospray MS/MS reveals extensive and nonspecific oxidation of cholesterol esters in human peripheral vascular lesions. J. Lipid Res. 52, 2070–83 (2011).

140.      Kontush, A., Chapman, M. J. & Stocker, R. Vitamin E is not deficient in human atherosclerotic plaques. Arterioscler. Thromb. Vasc. Biol. 24, e139-40; author reply e141-2 (2004).

141.      Kathir, K. et al. Equivalent lipid oxidation profiles in advanced atherosclerotic lesions of carotid endarterectomy plaques obtained from symptomatic type 2 diabetic and nondiabetic subjects. Free Radic. Biol. Med. 49, 481–6 (2010).

142.      Thomas, S. R. & Stocker, R. Molecular action of vitamin E in lipoprotein oxidation: implications for atherosclerosis. Free Radic. Biol. Med. 28, 1795–805 (2000).

143.      Ohkawa, S. et al. Pro-oxidative effect of alpha-tocopherol in the oxidation of LDL isolated from co-antioxidant-depleted non-diabetic hemodialysis patients. Atherosclerosis 176, 411–8 (2004).

144.      Upston, J. M. et al. Disease stage-dependent accumulation of lipid and protein oxidation products in human atherosclerosis. Am. J. Pathol. 160, 701–10 (2002).

145.      Moerman, A. M. et al. Lipid signature of advanced human carotid atherosclerosis assessed by mass spectrometry imaging. J. Lipid Res. 62, 100020 (2021).

146.      Maor, I., Mandel, H. & Aviram, M. Macrophage uptake of oxidized LDL inhibits lysosomal sphingomyelinase, thus causing the accumulation of unesterified cholesterol-sphingomyelin-rich particles in the lysosomes. A possible role for 7-Ketocholesterol. Arterioscler. Thromb. Vasc. Biol. 15, 1378–87 (1995).

147.      Geng, Y.-J., Phillips, J. E., Mason, R. P. & Casscells, S. W. Cholesterol crystallization and macrophage apoptosis: implication for atherosclerotic plaque instability and rupture. Biochem. Pharmacol. 66, 1485–92 (2003).

148.      Mata, P. et al. Effect of dietary fat saturation on LDL oxidation and monocyte adhesion to human endothelial cells in vitro. Arterioscler. Thromb. Vasc. Biol. 16, 1347–55 (1996).

149.      Hargrove, R. L., Etherton, T. D., Pearson, T. A., Harrison, E. H. & Kris-Etherton, P. M. Low fat and high monounsaturated fat diets decrease human low density lipoprotein oxidative susceptibility in vitro. J. Nutr. 131, 1758–63 (2001).

150.      Yu-Poth, S. et al. Lowering dietary saturated fat and total fat reduces the oxidative susceptibility of LDL in healthy men and women. J. Nutr. 130, 2228–37 (2000).

151.      Cawood, A. L. et al. Eicosapentaenoic acid (EPA) from highly concentrated n-3 fatty acid ethyl esters is incorporated into advanced atherosclerotic plaques and higher plaque EPA is associated with decreased plaque inflammation and increased stability. Atherosclerosis 212, 252–9 (2010).

152.      Thies, F. et al. Association of n-3 polyunsaturated fatty acids with stability of atherosclerotic plaques: a randomised controlled trial. Lancet (London, England) 361, 477–85 (2003).

153.      Haddad, E. H., Gaban-Chong, N., Oda, K. & Sabaté, J. Effect of a walnut meal on postprandial oxidative stress and antioxidants in healthy individuals. Nutr. J. 13, 4 (2014).

154.      McKay, D. L. et al. Chronic and acute effects of walnuts on antioxidant capacity and nutritional status in humans: a randomized, cross-over pilot study. Nutr. J. 9, 21 (2010).

155.      Zambón, D. et al. Substituting walnuts for monounsaturated fat improves the serum lipid profile of hypercholesterolemic men and women. A randomized crossover trial. Ann. Intern. Med. 132, 538–46 (2000).

156.      Kris-Etherton, P. M. Walnuts decrease risk of cardiovascular disease: a summary of efficacy and biologic mechanisms. J. Nutr. 144, 547S-554S (2014).

157.      Al-Shaar, L. et al. Red meat intake and risk of coronary heart disease among US men: prospective cohort study. BMJ 371, m4141 (2020).

158.      Fang, X. et al. Dietary intake of heme iron and risk of cardiovascular disease: a dose-response meta-analysis of prospective cohort studies. Nutr. Metab. Cardiovasc. Dis. 25, 24–35 (2015).

159.      Han, M. et al. Dietary iron intake and risk of death due to cardiovascular diseases: A systematic review and dose-response meta-analysis of prospective cohort studies. Asia Pac. J. Clin. Nutr. 29, 309–321 (2020).

160.      Etemadi, A. et al. Mortality from different causes associated with meat, heme iron, nitrates, and nitrites in the NIH-AARP Diet and Health Study: population based cohort study. BMJ 357, j1957 (2017).

161.      Kanner, J. et al. Redox homeostasis in stomach medium by foods: The Postprandial Oxidative Stress Index (POSI) for balancing nutrition and human health. Redox Biol. 12, 929–936 (2017).

162.      Kanner, J., Gorelik, S., Roman, S. & Kohen, R. Protection by polyphenols of postprandial human plasma and low-density lipoprotein modification: the stomach as a bioreactor. J. Agric. Food Chem. 60, 8790–6 (2012).

163.      Sirota, R., Gorelik, S., Harris, R., Kohen, R. & Kanner, J. Coffee polyphenols protect human plasma from postprandial carbonyl modifications. Mol. Nutr. Food Res. 57, 916–9 (2013).

164.      Tirosh, O., Shpaizer, A. & Kanner, J. Lipid Peroxidation in a Stomach Medium Is Affected by Dietary Oils (Olive/Fish) and Antioxidants: The Mediterranean versus Western Diet. J. Agric. Food Chem. 63, 7016–23 (2015).

165.      Rundblad, A., Holven, K. B., Ottestad, I., Myhrstad, M. C. & Ulven, S. M. High-quality fish oil has a more favourable effect than oxidised fish oil on intermediate-density lipoprotein and LDL subclasses: a randomised controlled trial. Br. J. Nutr. 117, 1291–1298 (2017).

166.      Rosenson, R. S. et al. Dysfunctional HDL and atherosclerotic cardiovascular disease. Nat. Rev. Cardiol. 13, 48–60 (2016).

167.      Huang, Y. et al. An abundant dysfunctional apolipoprotein A1 in human atheroma. Nat. Med. 20, 193–203 (2014).

168.      O’Reilly, M. et al. High-Density Lipoprotein Proteomic Composition, and not Efflux Capacity, Reflects Differential Modulation of Reverse Cholesterol Transport by Saturated and Monounsaturated Fat Diets. Circulation 133, 1838–50 (2016).

169.      Cedó, L. et al. Consumption of polyunsaturated fat improves the saturated fatty acid-mediated impairment of HDL antioxidant potential. Mol. Nutr. Food Res. 59, 1987–96 (2015).

170.      Nicholls, S. J. et al. Consumption of saturated fat impairs the anti-inflammatory properties of high-density lipoproteins and endothelial function. J. Am. Coll. Cardiol. 48, 715–20 (2006).

171.      Edsfeldt, A. et al. Sphingolipids Contribute to Human Atherosclerotic Plaque Inflammation. Arterioscler. Thromb. Vasc. Biol. 36, 1132–40 (2016).

172.      Meeusen, J. W. et al. Ceramides improve atherosclerotic cardiovascular disease risk assessment beyond standard risk factors. Clin. Chim. Acta 511, 138–142 (2020).

173.      Choi, S. & Snider, A. J. Sphingolipids in High Fat Diet and Obesity-Related Diseases. Mediators Inflamm. 2015, 520618 (2015).

174.      Boon, J. et al. Ceramides contained in LDL are elevated in type 2 diabetes and promote inflammation and skeletal muscle insulin resistance. Diabetes 62, 401–10 (2013).

175.      Roszczyc-Owsiejczuk, K. & Zabielski, P. Sphingolipids as a Culprit of Mitochondrial Dysfunction in Insulin Resistance and Type 2 Diabetes. Front. Endocrinol. (Lausanne). 12, 1–15 (2021).

176.      Hanssen, N. M. J. et al. Postprandial Glucose Spikes, an Important Contributor to Cardiovascular Disease in Diabetes? Front. Cardiovasc. Med. 7, 570553 (2020).

177.      Esposito, K., Giugliano, D., Nappo, F., Marfella, R. & Campanian Postprandial Hyperglycemia Study Group. Regression of carotid atherosclerosis by control of postprandial hyperglycemia in type 2 diabetes mellitus. Circulation 110, 214–9 (2004).

178.      Jayaraman, S. et al. Binding to heparin triggers deleterious structural and biochemical changes in human low-density lipoprotein, which are amplified in hyperglycemia. Biochim. Biophys. acta. Mol. cell Biol. lipids 1865, 158712 (2020).

179.      Rabbani, N. et al. Glycation of LDL by methylglyoxal increases arterial atherogenicity: a possible contributor to increased risk of cardiovascular disease in diabetes. Diabetes 60, 1973–80 (2011).

180.      Haas, M. E., Attie, A. D. & Biddinger, S. B. The regulation of ApoB metabolism by insulin. Trends Endocrinol. Metab. 24, 391–7 (2013).

181.      Defronzo, R. A. Is insulin resistance atherogenic? Possible mechanisms. Atheroscler. Suppl. 7, 11–5 (2006).

182.      Park, Y. M., R Kashyap, S., A Major, J. & Silverstein, R. L. Insulin promotes macrophage foam cell formation: potential implications in diabetes-related atherosclerosis. Lab. Invest. 92, 1171–80 (2012).

183.      Kahn, D. et al. Subcellular localisation and composition of intramuscular triacylglycerol influence insulin sensitivity in humans. Diabetologia 64, 168–180 (2021).

184.      Perreault, L. et al. Intracellular localization of diacylglycerols and sphingolipids influences insulin sensitivity and mitochondrial function in human skeletal muscle. JCI insight 3, (2018).

185.      Kien, C. L. et al. A lipidomics analysis of the relationship between dietary fatty acid composition and insulin sensitivity in young adults. Diabetes 62, 1054–63 (2013).

186.      Tuccinardi, D. et al. An extra virgin olive oil-enriched chocolate spread positively modulates insulin-resistance markers compared with a palm oil-enriched one in healthy young adults: A double-blind, cross-over, randomised controlled trial. Diabetes. Metab. Res. Rev. 38, e3492 (2022).

187.      Sarabhai, T. et al. Dietary palmitate and oleate differently modulate insulin sensitivity in human skeletal muscle. Diabetologia 65, 301–314 (2022).

188.      Luukkonen, P. K. et al. Saturated Fat Is More Metabolically Harmful for the Human Liver Than Unsaturated Fat or Simple Sugars. Diabetes Care 41, 1732–1739 (2018).

189.      Rosqvist, F. et al. Overeating Saturated Fat Promotes Fatty Liver and Ceramides Compared With Polyunsaturated Fat: A Randomized Trial. J. Clin. Endocrinol. Metab. 104, 6207–6219 (2019).

190.      Masquio, D. C. L. et al. Reduction in saturated fat intake improves cardiovascular risks in obese adolescents during interdisciplinary therapy. Int. J. Clin. Pract. 69, 560–70 (2015).

191.      Tran, T. T. T. et al. Short Term Palmitate Supply Impairs Intestinal Insulin Signaling via Ceramide Production. J. Biol. Chem. 291, 16328–38 (2016).

192.      Boyanovsky, B., Karakashian, A., King, K., Giltiay, N. & Nikolova-Karakashian, M. Uptake and metabolism of low density lipoproteins with elevated ceramide content by human microvascular endothelial cells: implications for the regulation of apoptosis. J. Biol. Chem. 278, 26992–9 (2003).

193.      Xiao-Yun, X. et al. Ceramide mediates inhibition of the AKT/eNOS signaling pathway by palmitate in human vascular endothelial cells. Med. Sci. Monit. 15, BR254-61 (2009).

194.      Li, W. et al. Endogenous ceramide contributes to the transcytosis of oxldl across endothelial cells and promotes its subendothelial retention in vascular wall. Oxid. Med. Cell. Longev. 2014, (2014).

195.      Ruuth, M. et al. Susceptibility of low-density lipoprotein particles to aggregate depends on particle lipidome, is modifiable, and associates with future cardiovascular deaths. Eur. Heart J. 39, 2562–2573 (2018).

196.      Ruuth, M. et al. Overfeeding Saturated Fat Increases LDL (Low-Density Lipoprotein) Aggregation Susceptibility While Overfeeding Unsaturated Fat Decreases Proteoglycan-Binding of Lipoproteins. Arterioscler. Thromb. Vasc. Biol. 41, 2823–2836 (2021).

197.      Lahelma, M. et al. The human liver lipidome is significantly related to the lipid composition and aggregation susceptibility of low-density lipoprotein (LDL) particles. Atherosclerosis 363, 22–29 (2022).

198.      Deevska, G. M., Sunkara, M., Morris, A. J. & Nikolova-Karakashian, M. N. Characterization of secretory sphingomyelinase activity, lipoprotein sphingolipid content and LDL aggregation in ldlr-/- mice fed on a high-fat diet. Biosci. Rep. 32, 479–90 (2012).

199.      Wiedermann, C. J. et al. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: prospective results from the Bruneck Study. J. Am. Coll. Cardiol. 34, 1975–81 (1999).

200.      Forkosh, E. & Ilan, Y. The heart-gut axis: New target for atherosclerosis and congestive heart failure therapy. Open Hear. 6, 1–6 (2019).

201.      Gorabi, A. M. et al. Implications for the role of lipopolysaccharide in the development of atherosclerosis. Trends Cardiovasc. Med. 32, 525–533 (2022).

202.      Violi, F. et al. Gut-derived low-grade endotoxaemia, atherothrombosis and cardiovascular disease. Nat. Rev. Cardiol. 20, 24–37 (2023).

203.      d’Hennezel, E., Abubucker, S., Murphy, L. O. & Cullen, T. W. Total Lipopolysaccharide from the Human Gut Microbiome Silences Toll-Like Receptor Signaling. mSystems 2, (2017).

204.      Clark, S. R. et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 13, 463–9 (2007).

205.      McDonald, B., Urrutia, R., Yipp, B. G., Jenne, C. N. & Kubes, P. Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis. Cell Host Microbe 12, 324–33 (2012).

206.      Schumski, A. et al. Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion. Circulation 143, 254–266 (2021).

207.      Saito, Y. et al. Structural differences in bacterial lipopolysaccharides determine atherosclerotic plaque progression by regulating the accumulation of neutrophils. Atherosclerosis 358, 1–11 (2022).

208.      Mawhin, M.-A. et al. Neutrophils recruited by leukotriene B4 induce features of plaque destabilization during endotoxaemia. Cardiovasc. Res. 114, 1656–1666 (2018).

209.      Carnevale, R. et al. Localization of lipopolysaccharide from Escherichia Coli into human atherosclerotic plaque. Sci. Rep. 8, 3598 (2018).

210.      Loffredo, L. et al. Is There an Association Between Atherosclerotic Burden, Oxidative Stress, and Gut-Derived Lipopolysaccharides? Antioxid. Redox Signal. (2020). doi:10.1089/ars.2020.8109

211.      Loffredo, L. et al. Oxidative Stress and Gut-Derived Lipopolysaccharides in Neurodegenerative Disease: Role of NOX2. Oxid. Med. Cell. Longev. 2020, 8630275 (2020).

212.      Loffredo, L. et al. Low-grade endotoxemia and NOX2 in patients with coronary microvascular angina. Kardiol. Pol. 80, 911–918 (2022).

213.      Jie, Z. et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 8, 845 (2017).

214.      Liu, H. et al. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome 7, 68 (2019).

215.      Zhu, Q. et al. Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol. Genomics 50, 893–903 (2018).

216.      Kasahara, K. et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 3, 1461–1471 (2018).

217.      Du, Y. et al. Butyrate protects against high-fat diet-induced atherosclerosis via up-regulating ABCA1 expression in apolipoprotein E-deficiency mice. Br. J. Pharmacol. 177, 1754–1772 (2020).

218.      Yoshida, N. et al. Bacteroides vulgatus and Bacteroides dorei Reduce Gut Microbial Lipopolysaccharide Production and Inhibit Atherosclerosis. Circulation 138, 2486–2498 (2018).

219.      Choroszy, M. et al. Co-toxicity of Endotoxin and Indoxyl Sulfate, Gut-Derived Bacterial Metabolites, to Vascular Endothelial Cells in Coronary Arterial Disease Accompanied by Gut Dysbiosis. Nutrients 14, (2022).

220.      Pastori, D. et al. Gut-Derived Serum Lipopolysaccharide is Associated With Enhanced Risk of Major Adverse Cardiovascular Events in Atrial Fibrillation: Effect of Adherence to Mediterranean Diet. J. Am. Heart Assoc. 6, (2017).

221.      Rehues, P. et al. Characterization of the LPS and 3OHFA Contents in the Lipoprotein Fractions and Lipoprotein Particles of Healthy Men. Biomolecules 12, (2021).

222.      Levels, J. H. M. et al. Lipopolysaccharide is transferred from high-density to low-density lipoproteins by lipopolysaccharide-binding protein and phospholipid transfer protein. Infect. Immun. 73, 2321–6 (2005).

223.      Levels, J. H. M., Abraham, P. R., van Barreveld, E. P., Meijers, J. C. M. & van Deventer, S. J. H. Distribution and kinetics of lipoprotein-bound lipoteichoic acid. Infect. Immun. 71, 3280–4 (2003).

224.      Grin, P. M. et al. Low-density lipoprotein (LDL)-dependent uptake of Gram-positive lipoteichoic acid and Gram-negative lipopolysaccharide occurs through LDL receptor. Sci. Rep. 8, 1–11 (2018).

225.      Topchiy, E. et al. Lipopolysaccharide Is Cleared from the Circulation by Hepatocytes via the Low Density Lipoprotein Receptor. PLoS One 11, e0155030 (2016).

226.      Victorov, A. V et al. Composition and structure of lipopolysaccharide-human plasma low density lipoprotein complex. Analytical ultracentrifugation, 31P-NMR, ESR and fluorescence spectroscopy studies. Biochim. Biophys. Acta 984, 119–27 (1989).

227.      Ghoshal, S., Witta, J., Zhong, J., de Villiers, W. & Eckhardt, E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J. Lipid Res. 50, 90–7 (2009).

228.      Navab, M., Hough, G. P., Van Lenten, B. J., Berliner, J. A. & Fogelman, A. M. Low density lipoproteins transfer bacterial lipopolysaccharides across endothelial monolayers in a biologically active form. J. Clin. Invest. 81, 601–5 (1988).

229.      Clemente-Postigo, M. et al. Metabolic endotoxemia promotes adipose dysfunction and inflammation in human obesity. Am. J. Physiol. Endocrinol. Metab. 316, E319–E332 (2019).

230.      Vargas-Caraveo, A. et al. Lipopolysaccharide enters the rat brain by a lipoprotein-mediated transport mechanism in physiological conditions. Sci. Rep. 7, 13113 (2017).

231.      Schwartz, Y. S. & Dushkin, M. I. In vitro accumulation of complexes of endotoxin and low-density lipoproteins by macrophages and arterial wall. Bull. Exp. Biol. Med. 147, 189–92 (2009).

232.      Afroz, R. et al. Toll-like Receptor 4 Stimulates Gene Expression via Smad2 Linker Region Phosphorylation in Vascular Smooth Muscle Cells. ACS Pharmacol. Transl. Sci. 3, 524–534 (2020).

233.      Maziere, C., Conte, M. A., Dantin, F. & Maziere, J. C. Lipopolysaccharide enhances oxidative modification of low density lipoprotein by copper ions, endothelial and smooth muscle cells. Atherosclerosis 143, 75–80 (1999).

234.      An, D. et al. JNK1 Mediates Lipopolysaccharide-Induced CD14 and SR-AI Expression and Macrophage Foam Cell Formation. Front. Physiol. 8, 1075 (2017).

235.      Feingold, K. R. & Grunfeld, C. The acute phase response inhibits reverse cholesterol transport. J. Lipid Res. 51, 682–4 (2010).

236.      Annema, W. et al. Myeloperoxidase and serum amyloid A contribute to impaired in vivo reverse cholesterol transport during the acute phase response but not group IIA secretory phospholipase A(2). J. Lipid Res. 51, 743–54 (2010).

237.      McGillicuddy, F. C. et al. Inflammation impairs reverse cholesterol transport in vivo. Circulation 119, 1135–45 (2009).

238.      Jian, C., Luukkonen, P., Sädevirta, S., Yki-Järvinen, H. & Salonen, A. Impact of short-term overfeeding of saturated or unsaturated fat or sugars on the gut microbiota in relation to liver fat in obese and overweight adults. Clin. Nutr. 40, 207–216 (2021).

239.      Telle-Hansen, V. H. et al. Replacing saturated fatty acids with polyunsaturated fatty acids increases the abundance of Lachnospiraceae and is associated with reduced total cholesterol levels-a randomized controlled trial in healthy individuals. Lipids Health Dis. 21, 92 (2022).

240.      Zhu, C. et al. Human gut microbiome composition and tryptophan metabolites were changed differently by fast food and Mediterranean diet in 4 days: a pilot study. Nutr. Res. 77, 62–72 (2020).

241.      Wolf, P., Cummings, P., Shah, N., Gaskins, H. R. & Mutlu, E. Sulfidogenic Bacteria Abundance in Colonic Mucosa is Positively Correlated with Milk and Animal Fat Intake and Negatively Correlated with Mono and Polyunsaturated Fatty Acids. FASEB J. 29, (2015).

242.      Devkota, S. et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature 487, 104–8 (2012).

243.      Natividad, J. M. et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat. Commun. 9, 2802 (2018).

244.      Lam, Y. Y. et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obesity (Silver Spring). 23, 1429–39 (2015).

245.      Deopurkar, R. et al. Differential effects of cream, glucose, and orange juice on inflammation, endotoxin, and the expression of Toll-like receptor-4 and suppressor of cytokine signaling-3. Diabetes Care 33, 991–7 (2010).

246.      López-Moreno, J. et al. Effect of Dietary Lipids on Endotoxemia Influences Postprandial Inflammatory Response. J. Agric. Food Chem. 65, 7756–7763 (2017).

247.      Ghanim, H. et al. Ezetimibe and simvastatin combination inhibits and reverses the pro-inflammatory and pro-atherogenic effects of cream in obese patients. Atherosclerosis 263, 278–286 (2017).

248.      Lyte, J. M., Gabler, N. K. & Hollis, J. H. Postprandial serum endotoxin in healthy humans is modulated by dietary fat in a randomized, controlled, cross-over study. Lipids Health Dis. 15, 186 (2016).

249.      Carnevale, R. et al. Gut-derived lipopolysaccharides increase post-prandial oxidative stress via Nox2 activation in patients with impaired fasting glucose tolerance: effect of extra-virgin olive oil. Eur. J. Nutr. 58, 843–851 (2019).

250.      Akiba, Y. et al. Lipopolysaccharides transport during fat absorption in rodent small intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 318, G1070–G1087 (2020).

251.      Kien, C. L. et al. Lipidomic evidence that lowering the typical dietary palmitate to oleate ratio in humans decreases the leukocyte production of proinflammatory cytokines and muscle expression of redox-sensitive genes. J. Nutr. Biochem. 26, 1599–606 (2015).

252.      Dumas, J. A. et al. Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metabolism. 65, 1582–8 (2016).

253.      Carpino, G. et al. Increased Liver Localization of Lipopolysaccharides in Human and Experimental NAFLD. Hepatology 72, 470–485 (2020).

254.      An, L. et al. The Role of Gut-Derived Lipopolysaccharides and the Intestinal Barrier in Fatty Liver Diseases. J. Gastrointest. Surg. 26, 671–683 (2022).

255.      Bjermo, H. et al. Effects of n-6 PUFAs compared with SFAs on liver fat, lipoproteins, and inflammation in abdominal obesity: a randomized controlled trial. Am. J. Clin. Nutr. 95, 1003–12 (2012).

256.      Rosqvist, F. et al. Overfeeding polyunsaturated and saturated fat causes distinct effects on liver and visceral fat accumulation in humans. Diabetes 63, 2356–68 (2014).

257.      Hernández, E. Á. et al. Acute dietary fat intake initiates alterations in energy metabolism and insulin resistance. J. Clin. Invest. 127, 695–708 (2017).

258.      Han, Y.-H. et al. Enterically derived high-density lipoprotein restrains liver injury through the portal vein. Science 373, eabe6729 (2021).

259.      Dandona, P. et al. Insulin suppresses endotoxin-induced oxidative, nitrosative, and inflammatory stress in humans. Diabetes Care 33, 2416–23 (2010).

260.      Lightle, S. et al. Elevation of ceramide in serum lipoproteins during acute phase response in humans and mice: role of serine-palmitoyl transferase. Arch. Biochem. Biophys. 419, 120–8 (2003).

261.      Memon, R. A. et al. Endotoxin and cytokines increase hepatic sphingolipid biosynthesis and produce lipoproteins enriched in ceramides and sphingomyelin. Arterioscler. Thromb. Vasc. Biol. 18, 1257–65 (1998).

262.      Kovar, J. et al. Acute responses of hepatic fat content to consuming fat, glucose and fructose alone and in combination in non-obese non-diabetic individuals with non-alcoholic fatty liver disease. J. Physiol. Pharmacol. 72, (2021).

263.      Nier, A., Brandt, A., Rajcic, D., Bruns, T. & Bergheim, I. Short-Term Isocaloric Intake of a Fructose- but not Glucose-Rich Diet Affects Bacterial Endotoxin Concentrations and Markers of Metabolic Health in Normal Weight Healthy Subjects. Mol. Nutr. Food Res. 63, e1800868 (2019).

264.      Olson, E. et al. Effects of Isocaloric Fructose Restriction on Ceramide Levels in Children with Obesity and Cardiometabolic Risk: Relation to Hepatic De Novo Lipogenesis and Insulin Sensitivity. Nutrients 14, (2022).

265.      Levental, I., Levental, K. R. & Heberle, F. A. Lipid Rafts: Controversies Resolved, Mysteries Remain. Trends Cell Biol. 30, 341–353 (2020).

266.      El-Sayed, A. & Harashima, H. Endocytosis of gene delivery vectors: from clathrin-dependent to lipid raft-mediated endocytosis. Mol. Ther. 21, 1118–30 (2013).

267.      Brown, M. S. & Goldstein, J. L. A receptor-mediated pathway for cholesterol homeostasis. Science 232, 34–47 (1986).

268.      Sapuri-Butti, A. R., Wang, L., Tetali, S. D., Rutledge, J. C. & Parikh, A. N. Interactions of different lipoproteins with supported phospholipid raft membrane (SPRM) patterns to understand similar in-vivo processes. Biochim. Biophys. acta. Biomembr. 1863, 183535 (2021).

269.      Ivaturi, S., Wooten, C. J., Nguyen, M. D., Ness, G. C. & Lopez, D. Distribution of the LDL receptor within clathrin-coated pits and caveolae in rat and human liver. Biochem. Biophys. Res. Commun. 445, 422–7 (2014).

270.      Infante, R. E. & Radhakrishnan, A. Continuous transport of a small fraction of plasma membrane cholesterol to endoplasmic reticulum regulates total cellular cholesterol. Elife 6, (2017).

271.      Raducka-Jaszul, O. et al. Molecular Diffusion of ABCA1 at the Cell Surface of Living Cells Assessed by svFCS. Membranes (Basel). 11, (2021).

272.      Sorci-Thomas, M. G. et al. Nascent high density lipoproteins formed by ABCA1 resemble lipid rafts and are structurally organized by three apoA-I monomers. J. Lipid Res. 53, 1890–909 (2012).

273.      Levental, K. R. et al. Lipidomic and biophysical homeostasis of mammalian membranes counteracts dietary lipid perturbations to maintain cellular fitness. Nat. Commun. 11, 1339 (2020).

274.      Shen, Y. et al. Metabolic activity induces membrane phase separation in endoplasmic reticulum. Proc. Natl. Acad. Sci. 114, 13394–13399 (2017).

275.      Xie, C., Woollett, L. A., Turley, S. D. & Dietschy, J. M. Fatty acids differentially regulate hepatic cholesteryl ester formation and incorporation into lipoproteins in the liver of the mouse. J. Lipid Res. 43, 1508–19 (2002).

276.      Baudet, M. F. & Jacotot, B. Dietary fats and lecithin-cholesterol acyltransferase activity in healthy humans. Ann. Nutr. Metab. 32, 352–9 (1988).

277.      Romijn, D., Wiseman, S. A., Scheek, L. M., de Fouw, N. J. & van Tol, A. A linoleic acid enriched diet increases serum cholesterol esterification by lecithin:cholesterol acyltransferase in meal-fed rats. Ann. Nutr. Metab. 42, 244–50 (1998).

278.      BUDIJANTO, S. et al. Dietary Fatty Acid Ethyl Esters and Lecithin-Cholesterol Acyltransferase Activity in Rats. J. Clin. Biochem. Nutr. 14, 183–193 (1993).

279.      Brousseau, M. E. et al. LCAT modulates atherogenic plasma lipoproteins and the extent of atherosclerosis only in the presence of normal LDL receptors in transgenic rabbits. Arterioscler. Thromb. Vasc. Biol. 20, 450–8 (2000).

280.      Muñoz, S. et al. Walnut-enriched diet increases the association of LDL from hypercholesterolemic men with human HepG2 cells. J. Lipid Res. 42, 2069–76 (2001).

281.      Kuo, P. C., Rudd, M. A., Nicolosi, R. & Loscalzo, J. Effect of dietary fat saturation and cholesterol on low density lipoprotein degradation by mononuclear cells of Cebus monkeys. Arteriosclerosis 9, 919–27 (1989).

282.      Kuo, P., Weinfeld, M. & Loscalzo, J. Effect of membrane fatty acyl composition on LDL metabolism in Hep G2 hepatocytes. Biochemistry 29, 6626–32 (1990).

283.      Terpstra, A. H. M., van den Berg, P., Jansen, H., Beynen, A. C. & van Tol, A. Decreasing dietary fat saturation lowers HDL-cholesterol and increases hepatic HDL binding in hamsters. Br. J. Nutr. 83, 151–9 (2000).

284.      Matarazzo, S. et al. Cholesterol-lowering drugs inhibit lectin-like oxidized low-density lipoprotein-1 receptor function by membrane raft disruption. Mol. Pharmacol. 82, 246–54 (2012).

285.      Yuan, X. et al. Endothelial Acid Sphingomyelinase Promotes NLRP3 Inflammasome and Neointima Formation During Hypercholesterolemia. J. Lipid Res. 63, 100298 (2022).

286.      Ke, L.-Y. et al. Enhanced Sphingomyelinase Activity Contributes to the Apoptotic Capacity of Electronegative Low-Density Lipoprotein. J. Med. Chem. 59, 1032–40 (2016).

287.      Holopainen, J. M., Medina, O. P., Metso, A. J. & Kinnunen, P. K. Sphingomyelinase activity associated with human plasma low density lipoprotein. J. Biol. Chem. 275, 16484–9 (2000).

288.      Kinnunen, P. K. J. & Holopainen, J. M. Sphingomyelinase activity of LDL: a link between atherosclerosis, ceramide, and apoptosis? Trends Cardiovasc. Med. 12, 37–42 (2002).

289.      Guarino, A. J., Lee, S. P. & Wrenn, S. P. Interactions between sphingomyelin and cholesterol in low density lipoproteins and model membranes. J. Colloid Interface Sci. 293, 203–12 (2006).

290.      Baumer, Y. et al. Ultramorphological analysis of plaque advancement and cholesterol crystal formation in Ldlr knockout mouse atherosclerosis. Atherosclerosis 287, 100–111 (2019).

291.      Jin, X. et al. Macrophages Shed Excess Cholesterol in Unique Extracellular Structures Containing Cholesterol Microdomains. Arterioscler. Thromb. Vasc. Biol. 38, 1504–1518 (2018).

292.      Kellner-Weibel, G. et al. Crystallization of free cholesterol in model macrophage foam cells. Arterioscler. Thromb. Vasc. Biol. 19, 1891–8 (1999).

293.      Kellner-Weibel, G., Luke, S. J. & Rothblat, G. H. Cytotoxic cellular cholesterol is selectively removed by apoA-I via ABCA1. Atherosclerosis 171, 235–43 (2003).

294.      Sherratt, S. C. R., Juliano, R. A. & Mason, R. P. Eicosapentaenoic acid (EPA) has optimal chain length and degree of unsaturation to inhibit oxidation of small dense LDL and membrane cholesterol domains as compared to related fatty acids in vitro. Biochim. Biophys. acta. Biomembr. 1862, 183254 (2020).

295.      Phillips, J. E., Geng, Y. J. & Mason, R. P. 7-Ketocholesterol forms crystalline domains in model membranes and murine aortic smooth muscle cells. Atherosclerosis 159, 125–35 (2001).

296.      Carroll, R. G. et al. An unexpected link between fatty acid synthase and cholesterol synthesis in proinflammatory macrophage activation. J. Biol. Chem. 293, 5509–5521 (2018).

297.      Zhu, X. et al. Macrophage ABCA1 reduces MyD88-dependent Toll-like receptor trafficking to lipid rafts by reduction of lipid raft cholesterol. J. Lipid Res. 51, 3196–206 (2010).

298.      Chowdhury, S. M. et al. Proteomic Analysis of ABCA1-Null Macrophages Reveals a Role for Stomatin-Like Protein-2 in Raft Composition and Toll-Like Receptor Signaling. Mol. Cell. Proteomics 14, 1859–70 (2015).

299.      Stamatikos, A. et al. ABCA1 Overexpression in Endothelial Cells In Vitro Enhances ApoAI-Mediated Cholesterol Efflux and Decreases Inflammation. Hum. Gene Ther. 30, 236–248 (2019).

300.      Hillyard, D. Z. et al. Statins inhibit NK cell cytotoxicity by membrane raft depletion rather than inhibition of isoprenylation. Atherosclerosis 191, 319–25 (2007).

301.      Kauerova, S. et al. Statins Directly Influence the Polarization of Adipose Tissue Macrophages: A Role in Chronic Inflammation. Biomedicines 9, 211 (2021).

302.      Kaul, S. et al. Lipid-Free Apolipoprotein A-I Reduces Progression of Atherosclerosis by Mobilizing Microdomain Cholesterol and Attenuating the Number of CD131 Expressing Cells: Monitoring Cholesterol Homeostasis Using the Cellular Ester to Total Cholesterol Ratio. J. Am. Heart Assoc. 5, (2016).

303.      Vieira, F. S., Corrêa, G., Einicker-Lamas, M. & Coutinho-Silva, R. Host-cell lipid rafts: a safe door for micro-organisms? Biol. cell 102, 391–407 (2010).

304.      Kulkarni, R., Wiemer, E. A. C. & Chang, W. Role of Lipid Rafts in Pathogen-Host Interaction - A Mini Review. Front. Immunol. 12, 815020 (2021).

305.      de Turris, V. et al. Candida albicans Targets a Lipid Raft/Dectin-1 Platform to Enter Human Monocytes and Induce Antigen Specific T Cell Responses. PLoS One 10, e0142531 (2015).

306.      Das, O. et al. Butyrate driven raft disruption trots off enteric pathogen invasion: possible mechanism of colonization resistance. Gut Pathog. 15, 19 (2023).

307.      Mani, V., Hollis, J. H. & Gabler, N. K. Dietary oil composition differentially modulates intestinal endotoxin transport and postprandial endotoxemia. Nutr. Metab. (Lond). 10, 6 (2013).

308.      Neal, M. D. et al. Enterocyte TLR4 mediates phagocytosis and translocation of bacteria across the intestinal barrier. J. Immunol. 176, 3070–9 (2006).

309.      Hwang, D. H., Kim, J.-A. & Lee, J. Y. Mechanisms for the activation of Toll-like receptor 2/4 by saturated fatty acids and inhibition by docosahexaenoic acid. Eur. J. Pharmacol. 785, 24–35 (2016).

310.      Saha, S., Pupo, E., Zariri, A. & van der Ley, P. Lipid A heterogeneity and its role in the host interactions with pathogenic and commensal bacteria. microLife 3, 1–13 (2022).

311.      Mo, Z., Huang, S., Burnett, D. J., Rutledge, J. C. & Hwang, D. H. Endotoxin May Not Be the Major Cause of Postprandial Inflammation in Adults Who Consume a Single High-Fat or Moderately High-Fat Meal. J. Nutr. 150, 1303–1312 (2020).

312.      Lancaster, G. I. et al. Evidence that TLR4 Is Not a Receptor for Saturated Fatty Acids but Mediates Lipid-Induced Inflammation by Reprogramming Macrophage Metabolism. Cell Metab. 27, 1096-1110.e5 (2018).

313.      Cuschieri, J., Bulger, E., Billgrin, J., Garcia, I. & Maier, R. V. Acid sphingomyelinase is required for lipid Raft TLR4 complex formation. Surg. Infect. (Larchmt). 8, 91–106 (2007).

314.      Tawadros, P. S. et al. Oxidative Stress Increases Surface Toll-Like Receptor 4 Expression in Murine Macrophages Via Ceramide Generation. Shock 44, 157–65 (2015).

315.      Joseph, C. K. et al. Bacterial lipopolysaccharide has structural similarity to ceramide and stimulates ceramide-activated protein kinase in myeloid cells. J. Biol. Chem. 269, 17606–10 (1994).

316.      Lu, Z., Liu, S., Lopes-Virella, M. F. & Wang, Z. LPS and palmitic acid Co-upregulate microglia activation and neuroinflammatory response. Compr. psychoneuroendocrinology 6, 100048 (2021).

317.      Jin, J. et al. Docosahexaenoic acid antagonizes the boosting effect of palmitic acid on LPS inflammatory signaling by inhibiting gene transcription and ceramide synthesis. PLoS One 13, e0193343 (2018).

318.      Li, Y. et al. Saturated fatty acid combined with lipopolysaccharide stimulates a strong inflammatory response in hepatocytes in vivo and in vitro. Am. J. Physiol. - Endocrinol. Metab. 315, E745–E757 (2018).

319.      Schilling, J. D. et al. Palmitate and Lipopolysaccharide Trigger Synergistic Ceramide Production in Primary Macrophages. J. Biol. Chem. 288, 2923–2932 (2013).

320.      Robblee, M. M. et al. Saturated Fatty Acids Engage an IRE1α-Dependent Pathway to Activate the NLRP3 Inflammasome in Myeloid Cells. Cell Rep. 14, 2611–23 (2016).

321.      Karasawa, T. & Takahashi, M. Saturated fatty acid-crystals activate NLRP3 inflammasome. Aging (Albany. NY). 11, 1613–1614 (2019).

322.      Thompson, R. C. et al. Atherosclerosis across 4000 years of human history: the Horus study of four ancient populations. Lancet 381, 1211–22 (2013).

323.      Heagerty, A. M. Scanning ancient history for evidence of modern diseases. Lancet (London, England) 381, 1165–6 (2013).

324.      O’Keefe, J. H., Cordain, L., Harris, W. H., Moe, R. M. & Vogel, R. Optimal low-density lipoprotein is 50 to 70 mg/dl: lower is better and physiologically normal. J. Am. Coll. Cardiol. 43, 2142–6 (2004).

325.      Cordain, L., Eaton, S. B., Miller, J. B., Mann, N. & Hill, K. The paradoxical nature of hunter-gatherer diets: meat-based, yet non-atherogenic. Eur. J. Clin. Nutr. 56 Suppl 1, S42-52 (2002).

326.      Kaplan, H. et al. Coronary atherosclerosis in indigenous South American Tsimane: a cross-sectional cohort study. Lancet (London, England) 389, 1730–1739 (2017).

327.      Rowan, C. J. et al. Very Low Prevalence and Incidence of Atrial Fibrillation among Bolivian Forager-Farmers. Ann. Glob. Heal. 87, 18 (2021).

328.      Irimia, A. et al. The Indigenous South American Tsimane Exhibit Relatively Modest Decrease in Brain Volume With Age Despite High Systemic Inflammation. J. Gerontol. A. Biol. Sci. Med. Sci. 76, 2147–2155 (2021).

329.      Gatz, M. et al. Prevalence of dementia and mild cognitive impairment in indigenous Bolivian forager-horticulturalists. Alzheimers. Dement. 19, 44–55 (2023).

330.      Gisterå, A., Ketelhuth, D. F. J., Malin, S. G. & Hansson, G. K. Animal Models of Atherosclerosis-Supportive Notes and Tricks of the Trade. Circ. Res. 130, 1869–1887 (2022).

331.      Lee, R. G. et al. Plasma cholesteryl esters provided by lecithin:cholesterol acyltransferase and acyl-coenzyme a:cholesterol acyltransferase 2 have opposite atherosclerotic potential. Circ. Res. 95, 998–1004 (2004).

332.      Bell, T. A., Kelley, K., Wilson, M. D., Sawyer, J. K. & Rudel, L. L. Dietary fat-induced alterations in atherosclerosis are abolished by ACAT2-deficiency in ApoB100 only, LDLr-/- mice. Arterioscler. Thromb. Vasc. Biol. 27, 1396–402 (2007).

333.      Jones, P. J. H. et al. High-oleic canola oil consumption enriches LDL particle cholesteryl oleate content and reduces LDL proteoglycan binding in humans. Atherosclerosis 238, 231–8 (2015).

334.      Parini, P. et al. ACAT2 is localized to hepatocytes and is the major cholesterol-esterifying enzyme in human liver. Circulation 110, 2017–23 (2004).

335.      Peters, A. The energy request of inflammation. Endocrinology 147, 4550–2 (2006).

336.      Feingold, K. R. & Grunfeld, C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. Endotext (2019).

337.      Bosch, M. et al. Mammalian lipid droplets are innate immune hubs integrating cell metabolism and host defense. Science 370, (2020).

338.      Knight, M., Braverman, J., Asfaha, K., Gronert, K. & Stanley, S. Lipid droplet formation in Mycobacterium tuberculosis infected macrophages requires IFN-γ/HIF-1α signaling and supports host defense. PLoS Pathog. 14, e1006874 (2018).

339.      van der Crabben, S. N. et al. Early endotoxemia increases peripheral and hepatic insulin sensitivity in healthy humans. J. Clin. Endocrinol. Metab. 94, 463–8 (2009).

340.      Mehta, N. N. et al. Experimental endotoxemia induces adipose inflammation and insulin resistance in humans. Diabetes 59, 172–81 (2010).

341.      Bertani, B. & Ruiz, N. Function and Biogenesis of Lipopolysaccharides. EcoSal Plus 8, (2018).

342.      Nikaido, H. Molecular basis of bacterial outer membrane permeability revisited. Microbiol. Mol. Biol. Rev. 67, 593–656 (2003).

343.      Berg, J., Seyedsadjadi, N. & Grant, R. Saturated Fatty Acid Intake Is Associated With Increased Inflammation, Conversion of Kynurenine to Tryptophan, and Delta-9 Desaturase Activity in Healthy Humans. Int. J. Tryptophan Res. 13, 1178646920981946 (2020).

344.      Haghikia, A. et al. Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. Immunity 43, 817–829 (2015).

345.      Haase, S. et al. Propionic Acid Rescues High-Fat Diet Enhanced Immunopathology in Autoimmunity via Effects on Th17 Responses. Front. Immunol. 12, 701626 (2021).

346.      Park, J.-H., Jeong, S.-Y., Choi, A.-J. & Kim, S.-J. Lipopolysaccharide directly stimulates Th17 differentiation in vitro modulating phosphorylation of RelB and NF-κB1. Immunol. Lett. 165, 10–9 (2015).

347.      Annunziato, F., Romagnani, C. & Romagnani, S. The 3 major types of innate and adaptive cell-mediated effector immunity. J. Allergy Clin. Immunol. 135, 626–35 (2015).

348.      Seufert, A. L. et al. Enriched dietary saturated fatty acids induce trained immunity via ceramide production that enhances severity of endotoxemia and clearance of infection. Elife 11, (2022).

349.      Seufert, A. L. & Napier, B. A. A new frontier for fat: dietary palmitic acid induces innate immune memory. Immunometabolism 5, e00021 (2023).

350.      Andersen, C. J. Impact of Dietary Cholesterol on the Pathophysiology of Infectious and Autoimmune Disease. Nutrients 10, (2018).

351.      Brown, J. M. et al. Inhibition of stearoyl-coenzyme A desaturase 1 dissociates insulin resistance and obesity from atherosclerosis. Circulation 118, 1467–75 (2008).

352.      Pontzer, H. & Wood, B. M. Effects of Evolution, Ecology, and Economy on Human Diet: Insights from Hunter-Gatherers and Other Small-Scale Societies. Annu. Rev. Nutr. 41, 363–385 (2021).

353.      Flaxman, S. M. & Sherman, P. W. Morning Sickness: A Mechanism for Protecting Mother and Embryo. Q. Rev. Biol. 75, 113–148 (2000).

354.      Fessler, D. M. T. Reproductive Immunosuppression and Diet. Curr. Anthropol. 43, 19–61 (2002).

355.      Uribarri, J. et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J. Am. Diet. Assoc. 110, 911–16.e12 (2010).

356.      Baye, E., Kiriakova, V., Uribarri, J., Moran, L. J. & de Courten, B. Consumption of diets with low advanced glycation end products improves cardiometabolic parameters: meta-analysis of randomised controlled trials. Sci. Rep. 7, 2266 (2017).

357.      Kraft, T. S. et al. Nutrition transition in 2 lowland Bolivian subsistence populations. Am. J. Clin. Nutr. 108, 1183–1195 (2018).

358.      Trumble, B. C. et al. Apolipoprotein E4 is associated with improved cognitive function in Amazonian forager-horticulturalists with a high parasite burden. FASEB J. 31, 1508–1515 (2017).

359.      Garcia, A. R. et al. APOE4 is associated with elevated blood lipids and lower levels of innate immune biomarkers in a tropical Amerindian subsistence population. Elife 10, (2021).

360.      Nogoy, K. M. C. et al. Fatty Acid Composition of Grain- and Grass-Fed Beef and Their Nutritional Value and Health Implication. Food Sci. Anim. Resour. 42, 18–33 (2022).

361.      González, F., Considine, R. V, Abdelhadi, O. A. & Acton, A. J. Saturated Fat Ingestion Promotes Lipopolysaccharide-Mediated Inflammation and Insulin Resistance in Polycystic Ovary Syndrome. J. Clin. Endocrinol. Metab. 104, 934–946 (2019).

362.      Ghanim, H. et al. A resveratrol and polyphenol preparation suppresses oxidative and inflammatory stress response to a high-fat, high-carbohydrate meal. J. Clin. Endocrinol. Metab. 96, 1409–14 (2011).

363.      Oh, E. S., Petersen, K. S., Kris-Etherton, P. M. & Rogers, C. J. Spices in a High-Saturated-Fat, High-Carbohydrate Meal Reduce Postprandial Proinflammatory Cytokine Secretion in Men with Overweight or Obesity: A 3-Period, Crossover, Randomized Controlled Trial. J. Nutr. 150, 1600–1609 (2020).

364.      Ghanim, H. et al. Antiinflammatory and ROS Suppressive Effects of the Addition of Fiber to a High-Fat High-Calorie Meal. J. Clin. Endocrinol. Metab. 102, 858–869 (2017).

365.      Ramsden, C. E. et al. Dietary alteration of n-3 and n-6 fatty acids for headache reduction in adults with migraine: randomized controlled trial. BMJ 374, n1448 (2021).

366.      Neudorf, H. & Little, J. P. Impact of fasting & ketogenic interventions on the NLRP3 inflammasome: A narrative review. Biomed. J. 47, 100677 (2023).

367.      Kim, E. R. et al. Short Term Isocaloric Ketogenic Diet Modulates NLRP3 Inflammasome Via B-hydroxybutyrate and Fibroblast Growth Factor 21. Front. Immunol. 13, 843520 (2022).

368.      Roberts, W. C. The cause of atherosclerosis. Nutr. Clin. Pract. 23, 464–7 (2008).

369.      Williams, K. J., Tabas, I. & Fisher, E. A. How an artery heals. Circ. Res. 117, 909–13 (2015).

370.      Chistiakov, D. A., Myasoedova, V. A., Revin, V. V., Orekhov, A. N. & Bobryshev, Y. V. The phenomenon of atherosclerosis reversal and regression: Lessons from animal models. Exp. Mol. Pathol. 102, 138–145 (2017).

371.      Feig, J. E., Feig, J. L. & Dangas, G. D. The role of HDL in plaque stabilization and regression: basic mechanisms and clinical implications. Coron. Artery Dis. 27, 592–603 (2016).

372.      Jhamnani, S. et al. Meta-analysis of the effects of lifestyle modifications on coronary and carotid atherosclerotic burden. Am. J. Cardiol. 115, 268–75 (2015).

373.      Parsons, C., Agasthi, P., Mookadam, F. & Arsanjani, R. Reversal of coronary atherosclerosis: Role of life style and medical management. Trends Cardiovasc. Med. 28, 524–531 (2018).

374.      Dawson, L. P., Lum, M., Nerleker, N., Nicholls, S. J. & Layland, J. Coronary Atherosclerotic Plaque Regression: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 79, 66–82 (2022).

375.      Gragnano, F. & Calabrò, P. Role of dual lipid-lowering therapy in coronary atherosclerosis regression: Evidence from recent studies. Atherosclerosis 269, 219–228 (2018).

376.      Chockalingam, P., Vinayagam, N. S., Chockalingam, V. & Chockalingam, A. Remarkable regression of coronary atherosclerosis: An interplay of pharmacotherapeutic and lifestyle factors. Indian Heart J. 68, 188–9 (2016).

377.      Najjar, R. S. & Montgomery, B. D. A defined, plant-based diet as a potential therapeutic approach in the treatment of heart failure: A clinical case series. Complement. Ther. Med. 45, 211–214 (2019).

 

No comments:

Post a Comment