4 Oct 2021

Solid vs. liquid fat—a biophysical perspective

As reviewed previously, dietary fats have differential effects on the body in relation to various mechanisms. This post explores why from a more fundamental perspective.

The body is largely an aqueous environment, compartmentalised by amphipathic lipid barriers/membranes containing specific hydrophobic fatty acids; and similarly, lipids are transported in amphipathic lipoproteins and metabolised by water-soluble enzymes (e.g. lipases). However, dietary fats have diverse structures and physiochemical properties. Foremost, unsaturated fatty acids (UFAs) are liquid at body temperature (37°C), while saturated fatty acids (SFAs) have higher melting points, which increase with chain length, resulting in short–medium chain fatty acids (e.g. C3–11:0) being liquid and longer chains solid; with a parallel relationship to water insolubility (Wiki). Could these basic characteristics underlie some of their differential effects?

Differential metabolism

Most well-known is the ability of dietary fats to modulate blood lipids. In particular, saturated fats increase cholesterol (esp. total/LDL 1; even eucaloric low-carb 2,3); the extent of which is related to apoE genotype 4–6. This effect is mainly attributed to the more prevalent C12–16:0 SFAs (relative potency: C14>16>12) 7,8, while stearic acid (C18:0) is considered neutral 9. However, some less prevalent SFAs may also elevate cholesterol 10–12; for instance, medium-chain fats (i.e. C8:0/10:0) 13 and behenic acid (C22:0) 8 had lower and greater potency than C16:0, respectively, and both increased endogenous C12–18:0 SFAs, suggesting indirect effects 8,13. Replacement of SFAs with UFAs lowers cholesterol, but PUFAs/C18:2 more so than MUFAs/C18:1 1,7,14; and of PUFAs, linoleic acid (C18:2, n-6) and α-linolenic acid (C18:3, n-3) may be similar 15,16. Whereas supplementing long-chain omega-3 (typically fish oil) may have no effect 17, or increase cholesterol in relation to oil oxidation 18, DHA (C22:6, n-3) 19–21 and apoE genotype 19. Fat modulation of blood lipids extends to lipoprotein turnover; for instance, diets rich in typical SFAs decrease the LDL fractional catabolic rate (FCR) and LDL receptor (LDLR) expression/activity 12,22–25 (even postprandially 26), and therefore tissue uptake; also affected by apoE genotype 4,19. But why would fats differentially affect lipoprotein metabolism?

Fundamentally, fats are the most calorie-dense macronutrient, and can essentially be oxidised immediately for energy, or stored as esters in more complex lipids (e.g. phospholipids, triglycerides and cholesteryl esters) and used in other functions (e.g. signalling and membranes). Different fatty acids have different metabolic fates (reviews 27,28). For instance, in human tracer studies, whole-body oxidation of specific fats decreases with chain length (e.g. C12:0>16:0>18:0 29 and C18:3>22:6 30) and saturation (e.g. C16:0/18:0 vs. C18:1/18:2/18:3; Fig. 4) 9,29,31. Short-term trials also show overfeeding with SFAs (vs. UFAs) specifically induces greater liver fat 32–34, while low carb diets favouring UFAs (vs. SFAs) induce higher blood ketones (e.g. after 5 days 35 or 6 weeks 2,3). Tissue studies further highlight differential and interactive effects (e.g. intestine 36, hepatocytes 37,38 and myotubes 39–42). For instance, in myotubes, medium-chain fats (i.e. C10:0/12:0 vs. C16:0/18:1/18:2) induced the highest mitochondrial oxidative capacity and lowest ROS and lipid accumulation 43; while palmitate (C16:0) generally had higher oxidation and esterification than oleate (C18:1), which increased with co-administration of UFAs (i.e. C16:0 + C18:1 or 20:5) 39–42. In hepatocytes, stearate (i.e. C18:0 vs. C16:0/18:0/18:1/18:2) had particularly low oxidation and incorporation into various lipids 28, remaining relatively unmetabolised 37,38. Coincidently, oxidation of long-chain fats may induce ROS via LCAD (esp. in liver), where stearate had the fastest H2O2 production kinetics 44. Long-chain UFAs (vs. SFAs) may also have greater liver metabolism to ketones 24,30,37,45; a suggested mechanism by which PUFAs may lower production of triglycerides and VLDL/LDL-c 45. Note however, medium-chain C8–12 SFAs have high oxidation rates yet can elevate blood triglycerides and cholesterol (vs. UFAs) 11–13; replacing long-chain SFAs with UFAs may have no effect on triglycerides, unlike cholesterol 46; whereas supplementing long-chain omega-3s can dose-dependently lower blood triglycerides, by lowering VLDL production and increasing fatty acid uptake/oxidation 19,47, with discordant effects on cholesterol, as above.

However, the first stage of metabolism is digestion. Notably, in the human tracer studies above, SFAs were dissolved in liquids which facilitate absorption, while oxidation was far lower with capsules 29. Accordingly, digestion of (long-chain) fats initially involves emulsification by bile acids, forming micelles which increase surface area and facilitate hydrolysis by pancreatic lipase. Diets high in SFAs especially seem to favour secretion of taurine-conjugated bile acid (i.e. taurocholate) 48, and related outgrowth of sulphite-reducing bacteria 48–51, which are inhibited by omega-3s 52–54; while taurine supplementation can particularly enhance absorption of SFAs, in relation to their chain length and hydrophobicity 55,56. Moreover, in animal models, even micellar palmitic acid (the most common dietary SFA) has a relatively slow absorption (vs. UFAs), requiring more length of intestine and taurocholate 57–59. This was linked to differential metabolism by intestinal enterocytes, where palmitic vs. linoleic acid was more slowly re-esterified to triglyceride (esp. with lower taurocholate) 58. Further, while the intestine mainly secretes triglycerides via chylomicrons into lymph, palmitate also induced some smaller VLDL-like particles (i.e. lower triglyceride/protein ratio) 36,60. Subsequently, similar differential effects were reported on hepatic VLDL secretion (e.g. palmitic vs. oleic 61 or palmitoleic acids 62).

Interestingly, while diet can alter blood fatty acids 2,3,8,13,14,63–68, there is still a predominance of SFAs (esp. C16:0), MUFAs (esp. C18:1) and PUFAs (esp. C18:2), in plasma phospholipids, triglycerides and cholesteryl esters, respectively; even with ~60% fat diets favouring SFAs or UFAs 3. This suggests esterification is highly regulated and may be limited by fatty acid availability (e.g. FFA/albumin ratios) 38. Accordingly, in cells, palmitate accumulates in phospholipids, glycerides and ceramides, while oleate and EPA support channelling into triglycerides (and β-oxidation), via regulation of DGAT2 40–42, which meditates the final step of triglyceride synthesis (i.e. DAG–TAG). UFAs also favour cholesterol esterification by ACAT, while SFAs (C12–16:0) suppress esterification and increase free cholesterol (in the ER), which may inhibit hepatic LDLR expression 69 and VLDL-c secretion 70. Note, the blood cholesterol-lowering ability of common fatty acids parallels their cholesteryl ester content (i.e. C18:2>18:1>16:0) 3,14,68; while stearate may lower liver cholesterol and increase faecal excretion 71,72, by lowering secondary bile acids and cholesterol solubility/absorption 73–75. Moreover, dietary fats also have differential effects on lipogenesis. While eucaloric low-carb diets favouring SFAs or PUFAs can lower lipogenesis 2, diets favouring MUFAs and PUFAs specifically lower desaturation (i.e. SCD activity) 2,65,68,76, and high dietary SFAs or PUFAs can upregulate desaturation 77–79 or saturation 76, respectively (alongside cholesterol 76–78). Thus cells regulate their fatty acid balances (e.g. via differential regulation of SCD-1 78–81, PGC-1β 77,78 and SREBPs 22,76,82,83), which may maintain preferred substrates for esterification. Accordingly, DGAT2 co-localises with SCD-1 (in the ER) 84, which mediates production of the major MUFAs (C16:1 and 18:1), and supports their esterification 85,86. Also, blood SCD indices correlated triglycerides, but negatively with several PUFAs (LA and EPA/DHA) 87,88, which can suppress SCD-1/SREBP-1c expression in hepatocytes 88 and macrophages 81,89. Therefore, the storage and utilisation of dietary fats may involve counter-regulation of lipid saturation; but what principles underlie the bodies preference for specific fatty acid profiles?

Differential biophysics

A major fate of fats is incorporation into membranes, which compartmentalise cells and regulate biomolecule transport and signalling. Cell membranes are heterogeneous, containing hundreds of lipid species associated with distinct biophysical phases, such as liquid disorder (Ld) and liquid order (Lo), which may support specific processes 90,91. Fundamentally, membrane amphipathicity is mediated by polar lipids (e.g. phospholipids and sphingolipids), which typically consist of a hydrophilic head attached to a hydrophobic tail (acyl chain) of fatty acids, which determine physiochemical properties, lipid–sterol interactions and membrane biophysics (e.g. fluidity/viscosity, elasticity/compressibility, permeability, fusion, flip-flop, etc.) 92–94. For instance, SFAs are straight molecules which confer order and rigidity, while the double-bonds of UFAs introduce disorder and flexibility. An extreme example is DHA (C22:6, n-3), which contains many double-bonds and confers a loosely packed and dynamic membrane, with high fluidity, compressibility and permeability 92. Generally, plasma membrane bilayers are asymmetric; the inner leaflet possessing greater phospholipid unsaturation and fluidity, and outer leaflet being particularly rich in saturated sphingolipids, which can support Lo nanodomains termed ‘lipid rafts’ 91. However, membrane composition varies by organelle and cell/tissue, likely reflecting specific biophysical requirements; e.g. DHA being especially high in brain, retina and sperm.

While dietary fats vary widely in quantity/quality, plasma and organ phospholipid (i.e. membrane) fatty acid composition is highly regulated across different diets 95; although is susceptible (e.g. human muscle 65–67), and especially to PUFAs, given their limited biosynthesis 95. Such regulation involves crosstalk between membranes and metabolism, as exemplified by homeoviscous adaptation 76,96. For instance, exogenous PUFAs can be rapidly incorporated into membranes, and at high doses, upregulate SFA and cholesterol synthesis to maintain biophysical properties and fitness 76. Accordingly, cholesterol preferentially interacts with saturated lipids and supports Lo phase 90,92,97, while membrane fluidity is linked to cholesterol biosynthesis via HMG-CoA reductase 98–102. As above, SFA loading can induce lipogenesis and SCD/desaturation to produce MUFAs 78–80, which may support their oxidation and storage (as triglyceride vs. DAG/ceramide), and maintain cell physiology (e.g. membrane fluidity and insulin sensitivity) 40,41,80. In particular, plasma membrane raft regions are especially rich in cholesterol and saturated sphingolipids 90; where exogenous saturated and omega-3 fats oppositely regulated raft-dependant TLR signalling 103 and intestinal permeability 104; and omega-3s enriched tight junction microdomains preventing disruption by inflammation 105,106. Further, inside cells, metabolism of SFAs can even induce solid phase separation in ER membrane (and eventually cell death), in relation to the chain length (e.g. C12<14<16<18) and phase transition temperature (Tm) of their metabolites, which was offset by UFAs (i.e. C18:1/22:6) 107. Note, compared to plasma membrane, the ER is cholesterol-poor, while cholesterol eliminates solid phase behaviour 107. On the other hand, excess free cholesterol can cause membrane/organelle dysfunction and precipitate into crystals, triggering inflammasome activation 108,109. Similarly, free SFAs were also reported to induce crystallisation and inflammation in macrophages, which was inhibited by UFAs (i.e. C18:1/18:2/18:3), alongside accumulation of triglycerides 110,111. Taken together, these studies link cellular SFA/UFA balances to lipid biophysics, attesting the importance of (and rationale for) their tight regulation.

Similar principles may apply during (neutral) lipid storage and transport. As above, excess free fatty acids (FFAs) and free cholesterol (FC) are cytotoxic, but can be esterified and encapsulated within a phospholipid monolayer for intracellular storage in lipid droplets or intercellular transport in lipoproteins, the composition of which affects biophysical properties. In particular, cholesteryl esters can exist in liquid–crystalline states dependant on acyl chain length and unsaturation 112–114; where esterification with C18:2/PUFA may support fluidity 114–117, as may the presence of MUFA-based triglyceride (i.e. triolein) 117–120. As discussed earlier, SFAs can induce secretion of denser VLDL particles, with a lower triglyceride/cholesterol ratio 58,60–62; suggested to result from slower triglyceride esterification 58 and compensate insolubility 61. Higher fat and saturated fat diets (incl. SFAs vs. MUFAs 121) may also particularly increase large-buoyant LDL 122,123 (i.e. rich in PL/CE/FC, not TGs 124,125). Note, conditions of lower triglyceride availability are thought to promote formation of large LDL, while elevated levels favour small-dense LDL 122,126. Diets favouring n-6 PUFAs over SFAs can lower many VLDL–LDL particle sizes and lipids 24,127; as may high-quality vs. oxidised fish oil 18. This may involve modulation of lipoprotein turnover 22,23, in association with biophysics. For instance, high-PUFA diets have increased LDL PUFA content, alongside LDL fluidity 128 and LDLR binding 64; and similarly, in vitro, linoleate most increased hepatocyte membrane fluidity and LDL uptake (i.e. C18:2>18:1>Ctrl>18:0/16:0), which were highly correlated 129. In addition, LDL aggregation potential was related to surface sphingolipid content, and lowered by a ‘Healthy Nordic diet’ (incl. higher PUFA/SFA ratio) or lipid-lowering therapy 130, whereas overfeeding saturated fat had the opposite effect 131.

Lipid saturation also determines redox sensitivity; where the multiple double-bonds of PUFAs renders them particularly susceptible to oxidation (e.g. in supplements 18, stomach 132,133 and LDL 134). Both blood 3,14,68 and LDL lipids 23,63,64,128 are high in linoleic acid (C18:2, n-6), especially in cholesteryl esters (e.g. PL/TG ~20%, CE ~50% 3). Accordingly, trials with higher MUFAs 135–138 or lower total/saturated fat 5,135,139,140 have lowered lipoprotein oxidation (i.e. MDA/dienes in vitro/vivo) or plasma isoprostanes 138, in relation to LDL quality (i.e. C18:1/18:2 ratio) 135,136,138,140 or quantity (e.g. LDL-c/apoB) 5,139,140. Also, while LDL particle size and free cholesterol were associated with lower oxidative susceptibility 125, in vivo LDL oxidation (antibody method) and in vitro proteoglycan-binding strongly correlated many apoB-100 lipoproteins and their lipids, especially in smaller VLDLs (e.g. Fig. 4) 131, unlike LDL aggregation (which correlates PC/SM, as above) 130,131,141. Moreover, all PUFAs may not be equal; habitual intake and LDL content of omega-6 and 3 (esp. C18:2 and 22:6) correlated higher and lower LDL oxidation sensitivity, respectively 63. Further, RCTs with omega-3s generally improve blood redox markers 142; and even in advanced atherosclerotic plaques, omega-3 (esp. EPA) content is increased with supplementation and correlates greater stability and lower inflammation 143,144. Accordingly, compared to various fatty acids, omega-3s (esp. EPA) had the greatest antioxidant activity on micelles 145 and small-dense LDL 146, and EPA/DHA best suppressed ROS in endothelial cells 145. Also, oxidation of omega-6 and 3s generates different products 134,147 with different effects; e.g. DHA-derived 4-HHE can activate the Nrf2/HO-1 antioxidant pathway in multiple organs 148, while AA-derived 4-HNE associates more with inflammation 149, consistent with typical immunomodulatory paradigms. Ultimately, since omega-3s are particularly sensitive to oxidation, this may make them ideal redox sensors, coupling antioxidant metabolism to preservation of PUFAs and membrane biophysics 150.

Pathological biophysics

Many diseases are associated with dysregulated lipid metabolism and biophysics, foremost cardiovascular disease (CVD) 109. CVD is typically driven by atherosclerosis, which largely involves accumulation of lipids (esp. cholesterol) in the artery wall (esp. intimal foam cells), alongside lipoprotein particles (esp. apoB) 114,151–153. In animal models, a SFA-rich diet greatly increases arterial uptake of whole LDL and cholesteryl esters (i.e. selective uptake), in association with plasma cholesterol 154 and (macrophage) lipoprotein lipase 155, and in contrast to n-3 PUFAs 155,156. As above, SFAs (vs. UFAs) may also favour LDL aggregation and proteoglycan-binding, and therefore arterial retention 131. Exposure of cells to apoB-lipoproteins can induce ‘foamy’ cells with abundant lipid droplets of cholesteryl esters; e.g. macrophages incubated with native (high level) 157,158, aggregated 159,160 or multi-modified LDL 161,162; and perhaps less so (heavily) oxidised LDL 157,159–161,163, which induces lysosomal accumulation of cholesterol-sphingomyelin particles, due to inhibition of sphingomyelinase by 7-ketocholesterol 164. Importantly, the presence of cholesterol crystals is also a hallmark of atheroma 114,152, in association with extracellular lipoprotein particles and foam cells 153,165. In particular, in human aorta, transition from fatty streak to fibroatheroma (i.e. early–late stage) was associated with smooth muscle cell crystallisation and death 165. Further, in models of diet-induced atherosclerosis, crystals appeared early in endothelium 166,167 and increasingly macrophages 168, and caused lysosomal destabilisation and NLRP3 inflammasome activation 169–171; thus linking hyperlipidaemia to inflammation. Moreover, sustained exposure to LDL induces endothelial crystallisation 166, which is exacerbated by synergy between inflammatory cytokines (i.e. LDL + IFNg/TNFα), alongside suppression of esterification 167; while examples of higher peripheral inflammation with lower atherosclerosis attest a dependence on other risk factors/lipids 172,173. Oxidised LDL also induces macrophage phago-lysosomal crystals and NLRP3 activation 163,170,174; while in model membranes cholesterol crystal domains are induced by oxidation and inhibited by omega-3s (esp. EPA/DPA) 146. Notably, plaque crystals co-associated with cholesterol microdomains 168, which may be shed from macrophage plasma membranes 175; while excess free cholesterol in macrophages induced extracellular crystals and cytotoxicity offset by extracellular acceptors (i.e. apoA-1/E) mediating efflux of cholesterol from plasma membrane 176,177. In particular, the ABCA1 transporter may associate with cholesterol-rich lipid rafts 178 to mediate efflux to nascent HDL 179 and suppress raft-associated TLR/inflammatory signalling 180–182.

In humans and animals, intensive lipid-lowering can induce athero-regression 183, which may involve a net efflux of cholesterol via reverse cholesterol transport 184,185. Notably, trials with various unsaturated oils have lowered carotid thickening (i.e. cIMT) 186 and increased cholesterol efflux (in vitro) 14 in relation to plasma 16 or HDL fluidity 187; with some trends for omega-3 superiority 14,186. A rate-limiting step for efflux is cholesteryl ester hydrolysis to free cholesterol, which is faster with liquid-state (isotropic) esters 188 and supported by lysosomal delivery of triglycerides (via VLDL or triolein) 189. Accordingly, diet can increase omega-3 content in blood/LDL cholesteryl esters 3,14,63,64, which in triglyceride-depleted macrophages associated with increased fluidity, hydrolysis and efflux 188. Incorporation of omega-3s into membrane phospholipids may also move cholesterol from the inner to outer leaflet 81,188, an optimal position for efflux 190. Moreover, serum from people on omega-3 inhibited lipogenic genes in monocytes (i.e. SCD and FADS2) 89 and foam cells (i.e. SCD-1) 81; and α-linolenic acid (C18:3, n-3) modulation of an FXR–SCD-1 pathway decreased cholesterol storage and increased efflux 81,191. Also, chylomicron remnants loaded with triglycerides rich in n-6 or n-3 PUFAs (vs. MUFAs/SFAs) best suppressed macrophage NF-kB/COX-2 and increased cholesterol efflux, which may be linked 192. Note, in the dietary fat trials above, cholesterol efflux seemed unaffected by increased LDL oxidation 14,136 (these studies = same design), but was markedly blunted with higher BMI 16 or CRP 81; while immune-associated and myeloperoxidase-induced HDL oxidation has been linked to CVD and HDL dysfunction, including impaired cholesterol efflux via ABCA1 193.

Importantly, beyond CVD, cholesterol accumulation and crystallisation are implicated in many other tissues/diseases 109. For instance, hepatic cholesterol is elevated in NAFLD, while the presence of crystals in lipid droplets is specifically associated with NASH 194. Accordingly, in preclinical models, hepatocyte lipid droplets represent the major repository of cholesterol, where excess free cholesterol can precipitate and crystallise 194,195, resulting in cell death and inflammation, underlying the appearance of ‘crown-like’ structures and foam cells, and transition from NAFLD to NASH 108,194,195. In these models crystallisation is induced by the combination of dietary fat and cholesterol 108,195,196, and exacerbated by PCSK9 deletion (increases LDLR) 197, or can be induced in vitro with oleic acid and sustained LDL exposure 194,195, indicating a synergistic effect of fat and cholesterol, as with LDLR regulation above 69. Note, crown-like structures were originally reported in adipose during obesity, where crystals may also be implicated 198. Also, age-related thymic involution is associated with lipid accumulation, adipogenesis and NLRP3 activation, all of which was linked to macrophage-associated ceramides and free cholesterol 199; while macrophage crystals have also been reported (i.e. Fig. 4) 200. Conversely, a 2-year RCT with 14% caloric restriction improved thymopoiesis, in association with mobilisation of ectopic lipid and suppression of adipose PLA2G7 targeting NLRP3 201. In the CNS, myelin is particularly rich in cholesterol, which may be liberated by injury. Accordingly, in the aged mouse, myelin damage induced a defective remyelination response, accompanied by sustained immune infiltration and foam cells with lipid droplets and cholesterol crystals, which was restored by stimulating reverse cholesterol transport 202. And in the eye, early stages of age-related macular degeneration (AMD) are characterised by accumulation of lipid-rich drusen, where the presence of crystals (i.e. the ‘onion sign’) may be indicative of progression to vision loss (i.e. drusen end-stages) 203. Conversely, a small trial with high-dose statins induced regression of drusen with vision gain in some people 204.

The brain and retina are particularly rich in lipids and PUFAs, the balances of which may be disrupted in ageing and disease, and of consequence to membrane physiology 150,205–208. For instance, in human orbitofrontal cortex, an age-related decline in PUFAs (esp. AA and DHA) correlated increased SCD activity (i.e. mRNA and SFA/MUFA ratios) 209; which may also occur in Alzheimer’s disease (AD) 210. Further, frontal cortex lipid rafts may undergo gender-specific changes during ageing, which was generally associated with decreasing cholesterol and n-6 PUFA content 211. And in early-stage sporadic AD, frontal and entorhinal lipid rafts had many deficits, including lower cholesterol, sphingomyelin, unsaturation (n-9 and n-3) and peroxidability indexes 212; alongside greater liquid-order and viscosity, which correlated decreased n-3 PUFAs and increased BACE1­–APP interaction, potentially facilitating amyloid-β (Aβ) production 212,213. Preclinical models suggest lipid rafts increase in viscosity and size with ageing, which is exacerbated in familial AD, potentially favouring coalescence of amyloidogenic machinery, and offset by cholesterol and PUFAs 214. In particular, DHA modulates membrane cholesterol/phase separation 93,94, neuro-receptor partitioning 215 and Aβ processing 216. Conversely, in familial AD, elevated cellular and lipid raft cholesterol may promote APP localisation 217. Moreover, the most important risk factor for sporadic AD is genetic variation in apoE, which employs astrocyte-derived cholesterol to traffic APP in and out of neuronal lipid rafts, controlling production of neurotoxic Aβ vs. neuroprotective sAPP-α, respectively 218; the former being favoured by apoE4 219. A potential reconciliation to these divergent findings is that lipid raft APP localisation/processing is ultimately tied to viscosity, which can be increased by low unsaturation or high cholesterol 213,214. Notably, retinal PUFAs and Aβ are also implicated in AMD, suggesting overlapping pathology 220.

Finally, the gut microbiome is a potential source of inflammation in ageing and disease. As above, dietary SFAs promote and omega-3s suppress postprandial translocation of bacterial LPS from gut to blood 221, potentially via differential modulation of enterocyte lipid raft-associated TLR signalling 104. Similarly, SFAs promote and omega-3s suppress bile-related outgrowth of sulphite-reducing (Gram-negative) Bilophila 54, along with gut inflammation and permeability 52–54; and in association with SFA-induced liver fat 222. Microbial lipids can be transported to blood via portal­­ or lymph pathways 223. In blood, Gram-negative LPS and Gram-positive LTA have affinity for all lipoproteins 224, especially HDL 225, and are subsequently transferred to LDL 226,227, while the hepatic LDLR plays a major role in their clearance 228,229. LPS binding to lipoproteins suppresses acute toxicity, although may still transport bioactive LPS into tissues, including lymph nodes 230, adipose 231 and endothelium 232, where it may induce inflammation. In particular, LPS acts as a priming signal for inflammasomes, facilitating activation by cholesterol crystals 153,165; and also promotes LDL oxidation by copper ions, endothelial and smooth muscle cells 233. Furthermore, in animals, dietary SFAs (not MUFAs or PUFAs) also induce intestinal (enterocyte) and plasma Aβ 234, where it associates with triglyceride-rich lipoproteins (potentially as a regulating apolipoprotein 235) and is capable of inducing neurovascular dysfunction 236,237. Notably, Aβ also increases macrophage uptake of LDL, intracellular free and esterified cholesterol, and foam cell formation 238. Conversely, peripheral Aβ is cleared by liver LDLR and LRP1, which may also facilitate clearance of brain Aβ 239–241.

Perspective

In summary, dietary fats have differential effects on the body, attributable to various pathways and their regulation—but under what forces? This post explored a biophysical perspective. Accordingly, fatty acids have diverse physiochemical properties which underlie specific biophysical behaviours in aqueous and lipid compartments of the body, including that of lipid-based biomolecules mediating storage/transport and structure/function (e.g. lipoproteins and membranes, respectively). As such, cells may require tight regulation of specific fatty acid balances to maintain lipid behaviours conducive to normal physiology; while the physiochemical heterogeneity of dietary fats may present a constant homeostatic challenge, requiring rapid/dynamic adaptive responses to maintain short-term fitness, and modulating long-term health across environments and genetic adaptations. Ultimately, differential effects may arise from modulation of fatty acid levels and counter-regulatory pathways serving to maintain biophysical homeostasis; a perspective which might help explain prominent effects on gut and liver (i.e. 2 initial diet buffers), and downstream cardiovascular and neurological consequences, amongst others.

Most notoriously, saturated fat elevates blood cholesterol—but why, and does it matter? As above, perhaps this could involve suppression of LDLR activity via free cholesterol and membrane physiology, and represent extracellular backlog from a constrained effort to maintain the lipid unsaturation and fluidity favoured by evolution, which long-term promotes ectopic lipid accumulation and age-related biophysical dysfunction and disease?

References

1.           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).

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

3.           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).

4.           Griffin, B. 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).

5.           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).

6.           Yang, Y., Ruiz-Narvaez, E., Kraft, P. & Campos, H. Effect of apolipoprotein E genotype and saturated fat intake on plasma lipids and myocardial infarction in the Central Valley of Costa Rica. Hum. Biol. 79, 637–47 (2007).

7.           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).

8.           Cater, N. B. & Denke, M. A. Behenic acid is a cholesterol-raising saturated fatty acid in humans. Am. J. Clin. Nutr. 73, 41–4 (2001).

9.           Rodríguez-Morató, J., Galluccio, J., Dolnikowski, G. G., Lichtenstein, A. H. & Matthan, N. R. Comparison of the Postprandial Metabolic Fate of U-13C Stearic Acid and U-13C Oleic Acid in Postmenopausal Women. Arterioscler. Thromb. Vasc. Biol. 40, 2953–2964 (2020).

10.        Denke, M. A. Dietary fats, fatty acids, and their effects on lipoproteins. Curr. Atheroscler. Rep. 8, 466–71 (2006).

11.        McKenzie, K. M., Lee, C. M., Mijatovic, J., Haghighi, M. M. & Skilton, M. R. Medium-Chain Triglyceride Oil and Blood Lipids: A Systematic Review and Meta-Analysis of Randomized Trials. J. Nutr. (2021). doi:10.1093/jn/nxab220

12.        Tsai, Y. H., Park, S., Kovacic, J. & Snook, J. T. Mechanisms mediating lipoprotein responses to diets with medium-chain triglyceride and lauric acid. Lipids 34, 895–905 (1999).

13.        Cater, N. B., Heller, H. J. & Denke, M. A. Comparison of the effects of medium-chain triacylglycerols, palm oil, and high oleic acid sunflower oil on plasma triacylglycerol fatty acids and lipid and lipoprotein concentrations in humans. Am. J. Clin. Nutr. 65, 41–5 (1997).

14.        Montoya, M. T. et al. Fatty acid saturation of the diet and plasma lipid concentrations, lipoprotein particle concentrations, and cholesterol efflux capacity. Am. J. Clin. Nutr. 75, 484–91 (2002).

15.        Goyens, P. L. L. & Mensink, R. P. The dietary alpha-linolenic acid to linoleic acid ratio does not affect the serum lipoprotein profile in humans. J. Nutr. 135, 2799–804 (2005).

16.        Liu, X. et al. Diets Low in Saturated Fat with Different Unsaturated Fatty Acid Profiles Similarly Increase Serum-Mediated Cholesterol Efflux from THP-1 Macrophages in a Population with or at Risk for Metabolic Syndrome: The Canola Oil Multicenter Intervention Trial. J. Nutr. 148, 721–728 (2018).

17.        Abdelhamid, A. S. et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane database Syst. Rev. 3, CD003177 (2020).

18.        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).

19.        Oscarsson, J. & Hurt-Camejo, E. Omega-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid and their mechanisms of action on apolipoprotein B-containing lipoproteins in humans: A review. Lipids Health Dis. 16, 1–13 (2017).

20.        Ishida, T. et al. Distinct regulation of plasma LDL cholesterol by eicosapentaenoic acid and docosahexaenoic acid in high fat diet-fed hamsters: participation of cholesterol ester transfer protein and LDL receptor. Prostaglandins. Leukot. Essent. Fatty Acids 88, 281–8 (2013).

21.        Allaire, J. et al. High-Dose DHA Has More Profound Effects on LDL-Related Features Than High-Dose EPA: The ComparED Study. J. Clin. Endocrinol. Metab. 103, 2909–2917 (2018).

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

23.        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).

24.        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).

25.        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, 459–468 (1997).

26.        Øyri, L. K. L. et al. Postprandial changes in gene expression of cholesterol influx and efflux mediators after intake of SFA compared with n-6 PUFA in subjects with and without familial hypercholesterolaemia: secondary outcomes of a randomised controlled trial. J. Nutr. Sci. 8, e27 (2019).

27.        Bergouignan, A., Momken, I., Schoeller, D. A., Simon, C. & Blanc, S. Metabolic fate of saturated and monounsaturated dietary fats: the Mediterranean diet revisited from epidemiological evidence to cellular mechanisms. Prog. Lipid Res. 48, 128–47 (2009).

28.        Sampath, H. & Ntambi, J. M. The fate and intermediary metabolism of stearic acid. Lipids 40, 1187–91 (2005).

29.        DeLany, J. P., Windhauser, M. M., Champagne, C. M. & Bray, G. A. Differential oxidation of individual dietary fatty acids in humans. Am. J. Clin. Nutr. 72, 905–11 (2000).

30.        Freemantle, E. et al. Omega-3 fatty acids, energy substrates, and brain function during aging. Prostaglandins Leukot. Essent. Fat. Acids 75, 213–220 (2006).

31.        Schmidt, D. E., Allred, J. B. & Kien, C. L. Fractional oxidation of chylomicron-derived oleate is greater than that of palmitate in healthy adults fed frequent small meals. J. Lipid Res. 40, 2322–32 (1999).

32.        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).

33.        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).

34.        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).

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

36.        Gangl, A. & Ockner, R. K. Intestinal metabolism of lipids and lipoproteins. Gastroenterology 68, 167–86 (1975).

37.        Pai, T. & Yeh, Y. Y. Stearic acid unlike shorter-chain saturated fatty acids is poorly utilized for triacylglycerol synthesis and beta-oxidation in cultured rat hepatocytes. Lipids 31, 159–64 (1996).

38.        Kvilekval, K., Lin, J., Cheng, W. & Abumrad, N. Fatty acids as determinants of triglyceride and cholesteryl ester synthesis by isolated hepatocytes: kinetics as a function of various fatty acids. J. Lipid Res. 35, 1786–94 (1994).

39.        Gaster, M., Rustan, A. C. & Beck-Nielsen, H. Differential utilization of saturated palmitate and unsaturated oleate: evidence from cultured myotubes. Diabetes 54, 648–56 (2005).

40.        Capel, F. et al. Oleate dose-dependently regulates palmitate metabolism and insulin signaling in C2C12 myotubes. Biochim. Biophys. Acta 1861, 2000–2010 (2016).

41.        Henique, C. et al. Increased mitochondrial fatty acid oxidation is sufficient to protect skeletal muscle cells from palmitate-induced apoptosis. J. Biol. Chem. 285, 36818–27 (2010).

42.        Wensaas, A. J. et al. Fatty acid incubation of myotubes from humans with type 2 diabetes leads to enhanced release of beta-oxidation products because of impaired fatty acid oxidation: effects of tetradecylthioacetic acid and eicosapentaenoic acid. Diabetes 58, 527–35 (2009).

43.        Montgomery, M. K. et al. Contrasting metabolic effects of medium- versus long-chain fatty acids in skeletal muscle. J. Lipid Res. 54, 3322–33 (2013).

44.        Zhang, Y., Bharathi, S. S., Beck, M. E. & Goetzman, E. S. The fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase can be a source of mitochondrial hydrogen peroxide. Redox Biol. 26, 101253 (2019).

45.        Beynen, A. C. & Katan, M. B. Why do polyunsaturated fatty acids lower serum cholesterol? Am. J. Clin. Nutr. 42, 560–3 (1985).

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

47.        Shearer, G. C., Savinova, O. V & Harris, W. S. Fish oil -- how does it reduce plasma triglycerides? Biochim. Biophys. Acta 1821, 843–51 (2012).

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

49.        David, L. a et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–63 (2014).

50.        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).

51.        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).

52.        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).

53.        Zhuang, P. et al. Eicosapentaenoic and Docosahexaenoic Acids Differentially Alter Gut Microbiome and Reverse High-Fat Diet–Induced Insulin Resistance. Mol. Nutr. Food Res. 64, 1900946 (2020).

54.        Devkota, S. & Chang, E. B. Interactions between Diet, Bile Acid Metabolism, Gut Microbiota, and Inflammatory Bowel Diseases. Dig. Dis. 33, 351–6 (2015).

55.        Galeano, N. F. et al. Taurine supplementation of a premature formula improves fat absorption in preterm infants. Pediatr. Res. 22, 67–71 (1987).

56.        Darling, P. B., Lepage, G., Leroy, C., Masson, P. & Roy, C. C. Effect of taurine supplements on fat absorption in cystic fibrosis. Pediatr. Res. 19, 578–82 (1985).

57.        Miura, S. et al. Studies on the difference of lymphatic absorption between saturated and unsaturated long chain fatty acids in rats--particularly in reference with the effect of puromycin and colchicine. Keio J. Med. 28, 121–30 (1979).

58.        Ockner, R. K., Pittman, J. P. & Yager, J. L. Differences in the intestinal absorption of saturated and unsaturated long chain fatty acids. Gastroenterology 62, 981–92 (1972).

59.        Gallagher, N. D. & Playoust, M. R. Absorption of saturated and unsaturated fatty acids by rat jejunum and ileum. Gastroenterology 57, 9–18 (1969).

60.        Ockner, R. K. & Jones, A. L. An electron microscopic and functional study of very low density lipoproteins in intestinal lymph. J. Lipid Res. 11, 284–292 (1970).

61.        Heimberg, M. & Wilcox, H. G. The effect of palmitic and oleic acids on the properties and composition of the very low density lipoprotein secreted by the liver. J. Biol. Chem. 247, 875–80 (1972).

62.        Wilcox, H. G., Dunn, G. D. & Heimberg, M. Effects of a mixture of a saturated with an unsaturated fatty acid on secretion of the very low density lipoprotein by the liver. Biochem. Biophys. Res. Commun. 73, 733–40 (1976).

63.        de Ruiz, G. J. C. et al. Habitual fish intake is associated with decreased LDL susceptibility to ex vivo oxidation. Lipids 37, 333–41 (2002).

64.        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).

65.        Vessby, B., Gustafsson, I.-B., Tengblad, S. & Berglund, L. Indices of fatty acid desaturase activity in healthy human subjects: effects of different types of dietary fat. Br. J. Nutr. 110, 871–9 (2013).

66.        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).

67.        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).

68.        Warensjö, E. et al. Effects of saturated and unsaturated fatty acids on estimated desaturase activities during a controlled dietary intervention. Nutr. Metab. Cardiovasc. Dis. 18, 683–90 (2008).

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

70.        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).

71.        Rao, R. & Lokesh, B. R. TG containing stearic acid, synthesized from coconut oil, exhibit lipidemic effects in rats similar to those of cocoa butter. Lipids 38, 913–8 (2003).

72.        Imaizumi, K., Abe, K., Kuroiwa, C. & Sugano, M. Fat containing stearic acid increases fecal neutral steroid excretion and catabolism of low density lipoproteins without affecting plasma cholesterol concentration in hamsters fed a cholesterol-containing diet. J. Nutr. 123, 1693–702 (1993).

73.        Meng, H. et al. Comparison of diets enriched in stearic, oleic, and palmitic acids on inflammation, immune response, cardiometabolic risk factors, and fecal bile acid concentrations in mildly hypercholesterolemic postmenopausal women-randomized crossover trial. Am. J. Clin. Nutr. 110, 305–315 (2019).

74.        Jesch, E. D. & Carr, T. P. Food Ingredients That Inhibit Cholesterol Absorption. Prev. Nutr. food Sci. 22, 67–80 (2017).

75.        Cohn, J. S., Kamili, A., Wat, E., Chung, R. W. S. & Tandy, S. Reduction in intestinal cholesterol absorption by various food components: mechanisms and implications. Atheroscler. Suppl. 11, 45–8 (2010).

76.        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).

77.        Lin, J. et al. Hyperlipidemic effects of dietary saturated fats mediated through PGC-1beta coactivation of SREBP. Cell 120, 261–73 (2005).

78.        Oosterveer, M. H. et al. High fat feeding induces hepatic fatty acid elongation in mice. PLoS One 4, e6066 (2009).

79.        Sampath, H., Miyazaki, M., Dobrzyn, A. & Ntambi, J. M. Stearoyl-CoA desaturase-1 mediates the pro-lipogenic effects of dietary saturated fat. J. Biol. Chem. 282, 2483–93 (2007).

80.        Collins, J. M., Neville, M. J., Hoppa, M. B. & Frayn, K. N. De novo lipogenesis and stearoyl-CoA desaturase are coordinately regulated in the human adipocyte and protect against palmitate-induced cell injury. J. Biol. Chem. 285, 6044–52 (2010).

81.        Zhang, J. et al. Walnut oil increases cholesterol efflux through inhibition of stearoyl CoA desaturase 1 in THP-1 macrophage-derived foam cells. Nutr. Metab. (Lond). 8, 61 (2011).

82.        Gu, Y. & Yin, J. Saturated fatty acids promote cholesterol biosynthesis: Effects and mechanisms. Obes. Med. 18, 100201 (2020).

83.        Jiang, T., Li, J. & Levi, M. Dietary saturated fats cause acute upregulation of transcriptional factors that modulate lipid synthetic pathways in the kidney. FASEB J. 20, (2006).

84.        Man, W. C., Miyazaki, M., Chu, K. & Ntambi, J. Colocalization of SCD1 and DGAT2: implying preference for endogenous monounsaturated fatty acids in triglyceride synthesis. J. Lipid Res. 47, 1928–39 (2006).

85.        Ntambi, J. M. & Miyazaki, M. Regulation of stearoyl-CoA desaturases and role in metabolism. Prog. Lipid Res. 43, 91–104 (2004).

86.        Flowers, M. T. & Ntambi, J. M. Role of stearoyl-coenzyme A desaturase in regulating lipid metabolism. Curr. Opin. Lipidol. 19, 248–56 (2008).

87.        Telle-Hansen, V. H. et al. Daily intake of cod or salmon for 2 weeks decreases the 18:1n-9/18:0 ratio and serum triacylglycerols in healthy subjects. Lipids 47, 151–60 (2012).

88.        Velliquette, R. A. et al. Regulation of human stearoyl-CoA desaturase by omega-3 and omega-6 fatty acids: Implications for the dietary management of elevated serum triglycerides. J. Clin. Lipidol. 3, 281–8 (2009).

89.        Gillies, P. J. et al. Regulation of inflammatory and lipid metabolism genes by eicosapentaenoic acid-rich oil. J. Lipid Res. 53, 1679–89 (2012).

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

91.        Lorent, J. H. et al. Plasma membranes are asymmetric in lipid unsaturation, packing and protein shape. Nat. Chem. Biol. 16, 644–652 (2020).

92.        Wassall, S. R. & Stillwell, W. Polyunsaturated fatty acid-cholesterol interactions: domain formation in membranes. Biochim. Biophys. Acta 1788, 24–32 (2009).

93.        Shaikh, S. R., Kinnun, J. J., Leng, X., Williams, J. A. & Wassall, S. R. How polyunsaturated fatty acids modify molecular organization in membranes: insight from NMR studies of model systems. Biochim. Biophys. Acta 1848, 211–9 (2015).

94.        Wassall, S. R. et al. Docosahexaenoic acid regulates the formation of lipid rafts: A unified view from experiment and simulation. Biochim. Biophys. acta. Biomembr. 1860, 1985–1993 (2018).

95.        Abbott, S. K., Else, P. L., Atkins, T. A. & Hulbert, A. J. Fatty acid composition of membrane bilayers: importance of diet polyunsaturated fat balance. Biochim. Biophys. Acta 1818, 1309–17 (2012).

96.        Ernst, R., Ejsing, C. S. & Antonny, B. Homeoviscous Adaptation and the Regulation of Membrane Lipids. J. Mol. Biol. 428, 4776–4791 (2016).

97.        Yang, J., Martí, J. & Calero, C. Pair interactions among ternary DPPC/POPC/cholesterol mixtures in liquid-ordered and liquid-disordered phases. Soft Matter 12, 4557–61 (2016).

98.        Sinensky, M. & Kleiner, J. The effect of reagents that increase membrane fluidity on the activity of 3-hydroxyl-3-methyl glutaryl coenzyme A reductase in the CHO-K1 cell. J. Cell. Physiol. 108, 309–16 (1981).

99.        Richert, L. et al. Growth-rate-related and hydroxysterol-induced changes in membrane fluidity of cultured hepatoma cells: correlation with 3-hydroxy-3-methyl glutaryl CoA reductase activity. Biochem. Biophys. Res. Commun. 120, 192–8 (1984).

100.      Morita, I., Sato, I., Ma, L. & Murota, S. Enhancement of membrane fluidity in cholesterol-poor endothelial cells pre-treated with simvastatin. Endothelium 5, 107–13 (1997).

101.      Davis, P. J. & Poznansky, M. J. Modulation of 3-hydroxy-3-methylglutaryl-CoA reductase by changes in microsomal cholesterol content or phospholipid composition. Proc. Natl. Acad. Sci. U. S. A. 84, 118–21 (1987).

102.      Rabini, R. A. et al. Effect of HMG-CoA reductase inhibitors on the erythrocyte membrane. Boll. Soc. Ital. Biol. Sper. 67, 129–35 (1991).

103.      Wong, S. W. et al. Fatty acids modulate Toll-like receptor 4 activation through regulation of receptor dimerization and recruitment into lipid rafts in a reactive oxygen species-dependent manner. J. Biol. Chem. 284, 27384–92 (2009).

104.      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).

105.      Zhao, S. et al. Study on the effect of eicosapentaenoic acid on phospholipids composition in membrane microdomains of tight junctions of epithelial cells by liquid chromatography/electrospray mass spectrometry. J. Pharm. Biomed. Anal. 47, 343–50 (2008).

106.      Li, Q. et al. N-3 Polyunsaturated Fatty Acids Prevent Disruption of Epithelial Barrier Function Induced By Proinflammatory Cytokines. Mol. Immunol. 45, 1356–1365 (2008).

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

108.      Horn, C. L., Morales, A. L., Savard, C., Farrell, G. C. & Ioannou, G. N. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol. Commun. 6, 12–35 (2022).

109.      Baumer, Y., McCurdy, S. G. & Boisvert, W. A. Formation and Cellular Impact of Cholesterol Crystals in Health and Disease. Adv. Biol. 5, e2100638 (2021).

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

111.      Karasawa, T. et al. Saturated Fatty Acids Undergo Intracellular Crystallization and Activate the NLRP3 Inflammasome in Macrophages. Arterioscler. Thromb. Vasc. Biol. 38, 744–756 (2018).

112.      Ginsburg, G. S., Atkinson, D. & Small, D. M. Physical properties of cholesteryl esters. Prog. Lipid Res. 23, 135–67 (1984).

113.      Guo, W. & Hamilton, J. A. Phase behavior and crystalline structures of cholesteryl ester mixtures: a C-13 MASNMR study. Biophys. J. 68, 2376–86 (1995).

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

115.      Dorset, D. L. Eutectic interactions between saturated and unsaturated chain cholesteryl esters: comparison of calculated and observed phase diagrams. Biochim. Biophys. Acta 1046, 195–201 (1990).

116.      Guo, W. & Hamilton, J. A. 13C MAS NMR studies of crystalline cholesterol and lipid mixtures modeling atherosclerotic plaques. Biophys. J. 71, 2857–68 (1996).

117.      Veloski, C. A., McCann, R. A. & Snow, J. W. An analytical model for the phase behavior of cholesteryl esters in intracellular inclusions. Biochim. Biophys. Acta 1213, 183–92 (1994).

118.      Shimobayashi, S. F. & Ohsaki, Y. Universal phase behaviors of intracellular lipid droplets. Proc. Natl. Acad. Sci. U. S. A. 116, 25440–25445 (2019).

119.      Tall, A. R., Small, D. M., Atkinson, D. & Rudel, L. L. Studies on the structure of low density lipoproteins isolated from Macaca fascicularis fed an atherogenic diet. J. Clin. Invest. 62, 1354–63 (1978).

120.      Croll, D. H., Small, D. M. & Hamilton, J. A. Molecular motions and thermotropic phase behavior of cholesteryl esters with triolein. Biochemistry 24, 7971–80 (1985).

121.      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).

122.      Froyen, E. The effects of fat consumption on low-density lipoprotein particle size in healthy individuals: a narrative review. Lipids Health Dis. 20, 1–21 (2021).

123.      DiNicolantonio, J. J. & O’Keefe, J. H. Effects of dietary fats on blood lipids: a review of direct comparison trials. Open Hear. 5, e000871 (2018).

124.      Capell, W. H., Zambon, A., Austin, M. A., Brunzell, J. D. & Hokanson, J. E. Compositional differences of LDL particles in normal subjects with LDL subclass phenotype A and LDL subclass phenotype B. Arterioscler. Thromb. Vasc. Biol. 16, 1040–6 (1996).

125.      Ohmura, H. et al. Lipid compositional differences of small, dense low-density lipoprotein particle influence its oxidative susceptibility: possible implication of increased risk of coronary artery disease in subjects with phenotype B. Metabolism. 51, 1081–7 (2002).

126.      Ivanova, E. A., Myasoedova, V. A., Melnichenko, A. A., Grechko, A. V & Orekhov, A. N. Small Dense Low-Density Lipoprotein as Biomarker for Atherosclerotic Diseases. Oxid. Med. Cell. Longev. 2017, 1273042 (2017).

127.      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).

128.      Berlin, E., Judd, J. T., Marshall, M. W. & Kliman, P. G. Dietary linoleate increases fluidity and influences chemical composition of plasma low density lipoprotein in adult men. Atherosclerosis 66, 215–25 (1987).

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

130.      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).

131.      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).

132.      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).

133.      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).

134.      Deleanu, M., Sanda, G. M., Stancu, C. S., Popa, M. E. & Sima, A. V. Profiles of fatty acids and the main lipid peroxidation products of human atherogenic low density lipoproteins. Rev. Chim. 67, 8–12 (2016).

135.      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).

136.      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).

137.      Egert, S., Kratz, M., Kannenberg, F., Fobker, M. & Wahrburg, U. Effects of high-fat and low-fat diets rich in monounsaturated fatty acids on serum lipids, LDL size and indices of lipid peroxidation in healthy non-obese men and women when consumed under controlled conditions. Eur. J. Nutr. 50, 71–9 (2011).

138.      Cicero, A. F. G. et al. Changes in LDL fatty acid composition as a response to olive oil treatment are inversely related to lipid oxidative damage: The EUROLIVE study. J. Am. Coll. Nutr. 27, 314–20 (2008).

139.      McEneny, J. et al. Does a diet high or low in fat influence the oxidation potential of VLDL, LDL and HDL subfractions? Nutr. Metab. Cardiovasc. Dis. 23, 612–8 (2013).

140.      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).

141.      Manninen, S. et al. The effect of intakes of fish and Camelina sativa oil on atherogenic and anti-atherogenic functions of LDL and HDL particles: A randomized controlled trial. Atherosclerosis 281, 56–61 (2019).

142.      Heshmati, J. et al. Omega-3 fatty acids supplementation and oxidative stress parameters: A systematic review and meta-analysis of clinical trials. Pharmacol. Res. 149, 104462 (2019).

143.      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).

144.      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).

145.      Richard, D., Kefi, K., Barbe, U., Bausero, P. & Visioli, F. Polyunsaturated fatty acids as antioxidants. Pharmacol. Res. 57, 451–5 (2008).

146.      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).

147.      Frankel, E. N. et al. Effect of n-3 fatty acid-rich fish oil supplementation on the oxidation of low density lipoproteins. Lipids 29, 233–6 (1994).

148.      Nakagawa, F. et al. 4-Hydroxy hexenal derived from dietary n-3 polyunsaturated fatty acids induces anti-oxidative enzyme heme oxygenase-1 in multiple organs. Biochem. Biophys. Res. Commun. 443, 991–6 (2014).

149.      Yang, B. et al. Yin-Yang Mechanisms Regulating Lipid Peroxidation of Docosahexaenoic Acid and Arachidonic Acid in the Central Nervous System. Front. Neurol. 10, 642 (2019).

150.      Díaz, M., Mesa-Herrera, F. & Marín, R. DHA and Its Elaborated Modulation of Antioxidant Defenses of the Brain: Implications in Aging and AD Neurodegeneration. Antioxidants (Basel, Switzerland) 10, (2021).

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

152.      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).

153.      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).

154.      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).

155.      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).

156.      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).

157.      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).

158.      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).

159.      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).

160.      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).

161.      Orekhov, A. N. LDL and foam cell formation as the basis of atherogenesis. Curr. Opin. Lipidol. 29, 279–284 (2018).

162.      Orekhov, A. N. & Myasoedova, V. A. Low density lipoprotein-induced lipid accumulation is a key phenomenon of atherogenesis at the arterial cell level. Vessel Plus 2019, (2019).

163.      Klinkner, A. M., Waites, C. R., Kerns, W. D. & Bugelski, P. J. Evidence of foam cell and cholesterol crystal formation in macrophages incubated with oxidized LDL by fluorescence and electron microscopy. J. Histochem. Cytochem. 43, 1071–8 (1995).

164.      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).

165.      Ho-Tin-Noé, B. et al. Cholesterol crystallization in human atherosclerosis is triggered in smooth muscle cells during the transition from fatty streak to fibroatheroma. J. Pathol. 241, 671–682 (2017).

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

167.      Baumer, Y. et al. Hyperlipidaemia and IFNgamma/TNFalpha Synergism are associated with cholesterol crystal formation in Endothelial cells partly through modulation of Lysosomal pH and Cholesterol homeostasis. EBioMedicine 59, 102876 (2020).

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

169.      Rajamäki, K. et al. Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: a novel link between cholesterol metabolism and inflammation. PLoS One 5, e11765 (2010).

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

171.      Freigang, S. et al. Nrf2 is essential for cholesterol crystal-induced inflammasome activation and exacerbation of atherosclerosis. Eur. J. Immunol. 41, 2040–51 (2011).

172.      Machado, R. M. et al. Omega-6 polyunsaturated fatty acids prevent atherosclerosis development in LDLr-KO mice, in spite of displaying a pro-inflammatory profile similar to trans fatty acids. Atherosclerosis 224, 66–74 (2012).

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

174.      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).

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

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

177.      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).

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

179.      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).

180.      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).

181.      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).

182.      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).

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

184.      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).

185.      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).

186.      de Oliveira, P. A. et al. Unsaturated Fatty Acids Improve Atherosclerosis Markers in Obese and Overweight Non-diabetic Elderly Patients. Obes. Surg. 27, 2663–2671 (2017).

187.      Fernández-Castillejo, S. et al. Determinants of HDL Cholesterol Efflux Capacity after Virgin Olive Oil Ingestion: Interrelationships with Fluidity of HDL Monolayer. Mol. Nutr. Food Res. 61, (2017).

188.      Lada, A. T., Rudel, L. L. & St Clair, R. W. Effects of LDL enriched with different dietary fatty acids on cholesteryl ester accumulation and turnover in THP-1 macrophages. J. Lipid Res. 44, 770–9 (2003).

189.      Ullery-Ricewick, J. C., Cox, B. E., Griffin, E. E. & Jerome, W. G. Triglyceride alters lysosomal cholesterol ester metabolism in cholesteryl ester-laden macrophage foam cells. J. Lipid Res. 50, 2014–26 (2009).

190.      Schroeder, F. et al. Caveolin, sterol carrier protein-2, membrane cholesterol-rich microdomains and intracellular cholesterol trafficking. Subcell. Biochem. 51, 279–318 (2010).

191.      Zhang, J. et al. Alpha-linolenic acid increases cholesterol efflux in macrophage-derived foam cells by decreasing stearoyl CoA desaturase 1 expression: evidence for a farnesoid-X-receptor mechanism of action. J. Nutr. Biochem. 23, 400–9 (2012).

192.      De Pascale, C., Graham, V., Fowkes, R. C., Wheeler-Jones, C. P. D. & Botham, K. M. Suppression of nuclear factor-kappaB activity in macrophages by chylomicron remnants: modulation by the fatty acid composition of the particles. FEBS J. 276, 5689–702 (2009).

193.      Jin, Z., Zhou, L., Tian, R. & Lu, N. Myeloperoxidase Targets Apolipoprotein A-I for Site-Specific Tyrosine Chlorination in Atherosclerotic Lesions and Generates Dysfunctional High-Density Lipoprotein. Chem. Res. Toxicol. 34, 1672–1680 (2021).

194.      Ioannou, G. N. et al. Cholesterol Crystals in Hepatocyte Lipid Droplets Are Strongly Associated With Human Nonalcoholic Steatohepatitis. Hepatol. Commun. 3, 776–791 (2019).

195.      Ioannou, G. N. et al. Cholesterol crystallization within hepatocyte lipid droplets and its role in murine NASH. J. Lipid Res. 58, 1067–1079 (2017).

196.      Püschel, G. P. & Henkel, J. Dietary cholesterol does not break your heart but kills your liver. Porto Biomed. J. 3, e12 (2018).

197.      Ioannou, G. N. et al. Pcsk9 Deletion Promotes Murine Nonalcoholic Steatohepatitis and Hepatic Carcinogenesis: Role of Cholesterol. Hepatol. Commun. (2021). doi:10.1002/hep4.1858

198.      Giordano, A. et al. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J. Lipid Res. 54, 2423–36 (2013).

199.      Youm, Y. H. et al. The NLRP3 Inflammasome Promotes Age-Related Thymic Demise and Immunosenescence. Cell Rep. 1, 56–68 (2012).

200.      Goldberg, E. L. & Dixit, V. D. Drivers of age-related inflammation and strategies for healthspan extension. Immunol. Rev. 265, 63–74 (2015).

201.      Spadaro, O. et al. Caloric restriction in humans reveals immunometabolic regulators of health span. Science 375, 671–677 (2022).

202.      Cantuti-Castelvetri, L. et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science 359, 684–688 (2018).

203.      Fragiotta, S. et al. The Fate and Prognostic Implications of Hyperreflective Crystalline Deposits in Nonneovascular Age-Related Macular Degeneration. Invest. Ophthalmol. Vis. Sci. 60, 3100–3109 (2019).

204.      Vavvas, D. G. et al. Regression of Some High-risk Features of Age-related Macular Degeneration (AMD) in Patients Receiving Intensive Statin Treatment. EBioMedicine 5, 198–203 (2016).

205.      Mesa-Herrera, F., Taoro-González, L., Valdés-Baizabal, C., Diaz, M. & Marín, R. Lipid and Lipid Raft Alteration in Aging and Neurodegenerative Diseases: A Window for the Development of New Biomarkers. Int. J. Mol. Sci. 20, (2019).

206.      Skowronska-Krawczyk, D. & Budin, I. Aging membranes: Unexplored functions for lipids in the lifespan of the central nervous system. Exp. Gerontol. 131, 110817 (2020).

207.      Weiser, M. J., Butt, C. M. & Mohajeri, M. H. Docosahexaenoic Acid and Cognition throughout the Lifespan. Nutrients 8, 99 (2016).

208.      Liu, A., Chang, J., Lin, Y., Shen, Z. & Bernstein, P. S. Long-chain and very long-chain polyunsaturated fatty acids in ocular aging and age-related macular degeneration. J. Lipid Res. 51, 3217–3229 (2010).

209.      McNamara, R. K., Liu, Y., Jandacek, R., Rider, T. & Tso, P. The aging human orbitofrontal cortex: decreasing polyunsaturated fatty acid composition and associated increases in lipogenic gene expression and stearoyl-CoA desaturase activity. Prostaglandins. Leukot. Essent. Fatty Acids 78, 293–304 (2008).

210.      Astarita, G. et al. Elevated stearoyl-CoA desaturase in brains of patients with Alzheimer’s disease. PLoS One 6, e24777 (2011).

211.      Díaz, M., Fabelo, N., Ferrer, I. & Marín, R. ‘Lipid raft aging’ in the human frontal cortex during nonpathological aging: gender influences and potential implications in Alzheimer’s disease. Neurobiol. Aging 67, 42–52 (2018).

212.      Fabelo, N. et al. Altered lipid composition in cortical lipid rafts occurs at early stages of sporadic Alzheimer’s disease and facilitates APP/BACE1 interactions. Neurobiol. Aging 35, 1801–12 (2014).

213.      Díaz, M. et al. Biophysical alterations in lipid rafts from human cerebral cortex associate with increased BACE1/AβPP interaction in early stages of Alzheimer’s disease. J. Alzheimers. Dis. 43, 1185–98 (2015).

214.      Santos, G., Díaz, M. & Torres, N. V. Lipid Raft Size and Lipid Mobility in Non-raft Domains Increase during Aging and Are Exacerbated in APP/PS1 Mice Model of Alzheimer’s Disease. Predictions from an Agent-Based Mathematical Model. Front. Physiol. 7, 90 (2016).

215.      Javanainen, M. et al. Reduced level of docosahexaenoic acid shifts GPCR neuroreceptors to less ordered membrane regions. PLoS Comput. Biol. 15, e1007033 (2019).

216.      Grimm, M. O. W. et al. Docosahexaenoic acid reduces amyloid beta production via multiple pleiotropic mechanisms. J. Biol. Chem. 286, 14028–39 (2011).

217.      Cho, Y. Y., Kwon, O.-H., Park, M. K., Kim, T.-W. & Chung, S. Elevated cellular cholesterol in Familial Alzheimer’s presenilin 1 mutation is associated with lipid raft localization of β-amyloid precursor protein. PLoS One 14, e0210535 (2019).

218.      Wang, H. et al. Regulation of beta-amyloid production in neurons by astrocyte-derived cholesterol. Proc. Natl. Acad. Sci. U. S. A. 118, (2021).

219.      Lee, S.-I. et al. APOE4-carrying human astrocytes oversupply cholesterol to promote neuronal lipid raft expansion and Aβ generation. Stem cell reports 16, 2128–2137 (2021).

220.      Wang, L. & Mao, X. Role of Retinal Amyloid-β in Neurodegenerative Diseases: Overlapping Mechanisms and Emerging Clinical Applications. Int. J. Mol. Sci. 22, (2021).

221.      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).

222.      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).

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

224.      Khovidhunkit, W. et al. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J. Lipid Res. 45, 1169–96 (2004).

225.      Meilhac, O., Tanaka, S. & Couret, D. High-Density Lipoproteins Are Bug Scavengers. Biomolecules 10, 598 (2020).

226.      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).

227.      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).

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

229.      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).

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

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

232.      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).

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

235.      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).

236.      Takechi, R., Galloway, S., Pallebage-Gamarallage, M. M. S., Lam, V. & Mamo, J. C. L. Dietary fats, cerebrovasculature integrity and Alzheimer’s disease risk. Prog. Lipid Res. 49, 159–70 (2010).

237.      Lam, V. et al. Synthesis of human amyloid restricted to liver results in an Alzheimer disease–like: Neurodegenerative phenotype. PLoS Biol. 19, 1–26 (2021).

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

239.      Guo, Y.-X. et al. 1,25-Dihydroxyvitamin D3 regulates expression of LRP1 and RAGE in vitro and in vivo, enhancing Aβ1-40 brain-to-blood efflux and peripheral uptake transport. Neuroscience 322, 28–38 (2016).

240.      Sehgal, N. et al. Withania somnifera reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc. Natl. Acad. Sci. U. S. A. 109, 3510–5 (2012).

241.      Yao, L. et al. Nanoformulated alpha-mangostin ameliorates Alzheimer’s disease neuropathology by elevating LDLR expression and accelerating amyloid-beta clearance. J. Control. Release 226, 1–14 (2016).


No comments:

Post a Comment