10 Apr 2021

Differential effects of fats on gut–host health

Dietary fats are ubiquitous and essential, while their quantity and quality modulate health. Recently, effects on the gut microbiome are being revealed. This post explores their differential effects on the gut–host dialog and underlying mechanisms relevant to many diseases.

Dietary fats appear to differentially affect human physiology; and perhaps most notoriously in the case of cardiovascular disease (CVD), the leading cause of death globally. For instance, in large observational studies, substitution analyses suggest opposing effects of saturated vs. monounsaturated and polyunsaturated fatty acids (i.e. SFAs vs. MUFAs and PUFAs, respectively) on CVD 1–3; a relationship tested and supported by meta-analyses of randomised controlled trials (RCTs) 4, and referenced in many dietary guidelines. Further, in 3–4 week RCTs on healthy adults, adjusting the habitual palmitate/oleate ratio (i.e. the most abundant SFA/MUFA) affects blood/tissue lipids, alongside energy metabolism, immune activity and brain function 5–11. And even single meals with different fats can have markedly different effects on postprandial cardiometabolic biomarkers 12.

From a cellular perspective, lipids serve various fundamental roles (e.g. as membrane components, signalling molecules and calorie-dense fuel) requiring the properties of specific fatty acids and their spatiotemporal regulation. Consequently, while dietary fats vary widely in quantity/quality, cell membrane composition is highly regulated across organs and different diets 13, and perturbations can induce rapid lipidome remodelling to maintain biophysical properties and fitness 14. Nonetheless, such adaptability depends on differential modulation of lipid metabolism 14–16 and genetic/kinetic constraints therein (e.g. PUFA synthesis) 13,17, underlying dietary sensitivity. From a whole-body perspective, dietary fats may interact with many tissues to affect physiology; but notably, foods initially encounter the gut, a host–environment interface directly exposed to dietary extremes and potential perturbations, and home to a rich microbiome and much of the immune system. Accordingly, in human trials 18–22 and rodent models 23–31, high fat diets (i.e. 30–70% kcal; varied total) can induce heterogeneous changes to gut microbiome in association with host physiology (e.g. inflammation 19,21,24,26,28–31 and metabolic dysfunction 18–20,22,23,25,27; reviews 32,33). Crucially, such diets are typically high in SFAs (e.g. animal 20, lard 23,25,30 or dairy 18,26,27,29), MUFAs (e.g. lard or olive oil 27) or omega-6 PUFAs (e.g. soybean 19, corn 24,31 or sunflower oil 25), which can have differential effects (at same % kcal) 18,25–27,29,30; as can moderate omega-3 PUFAs (e.g. fish oil 24,25,31,34–36 or EPA/DHA 37; vs. lard 25, dairy 36,37 or corn oil 24,31). Hence this post explores some of the differential effects of (natural) fats on the gut–host dialog and underlying mechanisms relevant to many diseases and systemic health.

Gut microbiome

The gut microbiome contains trillions of microbes from hundreds of species, which populate specific metabolic and mucosal niches, thereby supplementing host metabolism and colonisation resistance, and underlying a mutual symbiosis shaped by host physiology and diet. In particular, in observational studies, dietary fats have (differentially) been associated with bacterial diversity 34,38,39, enterotypes 40 and species/strains 41–43; and as above, high fat diet interventions can modulate the gut microbiome. For instance, in perhaps the largest and longest RCT so far, conducted in China, with 217 healthy young adults on isocaloric 20, 30 or 40% fat diets for 6 months, higher fat consumption (mainly via soybean oil) lowered microbiota α-diversity, while modulating specific genera and metabolites 19,44. Notably, diet can induce significant gut microbiome changes within 1–5 days in young adults 20,21,40, although long-term diet may have stronger associations 40. Also, in animal models, high fat diets may further modulate susceptibility to exogenous pathogen infection/colonisation (e.g. Salmonella 45, C. difficile 46 and C. albicans 47).

How does fat shape the gut microbiome? In typical diets, most energy comes from carbs (4 kcal/g) and fats (9 kcal/g), which exist in balance; so generally higher fat = lower carb. This may underlie indirect effects, since gut microbes typically ferment non-digested carbs/fibre as a major energy source, resulting in production of short-chain fatty acids (SCFAs), which in turn fuel colonocytes, while lowering lumen pH and oxygen to inhibit pathogens 48–51. Accordingly, many studies use refined sources of fats/oils, with unmatched fibre 52, and high fat/low carb diets can decrease SCFAs in humans 21,22. Whereas whole nuts (i.e. fat with fibre, polyphenols, etc.) can increase SCFA-producing bacteria (review 53); as also occurred with overfeeding unsaturated fat 18, in part via nuts 54. However, other high fat diet studies affect the microbiome despite matched fibre 19,25,28,29,45 (and even carbs in hypercaloric studies 18,27), with different fats having different effects (refs above). Moreover, dietary and blood levels of omega-3/DHA positively correlated (independent of fibre) with bacterial diversity and Lachnospiraceae spp. (i.e. major SCFA-producers) in a large cohort of female twins (n=876) 42. Accordingly, a 6-week RCT with walnut (i.e. 65% fat; 9% α-linolenic acid—C18:3, n-3) or equivalent α-linolenic acid similarly increased Roseburia (family Lachnospiraceae) 55; and another with EPA/DHA increased Coprococcus (also family Lachnospiraceae), which correlated butyrate 56. Therefore, omega-3s may actually support SCFA/butyrate production.

As with carbs, a portion of fat intake also escapes digestion and can be utilised by gut microbes for growth, as demonstrated in vitro with a human colon simulator 57. Specifically, fat-only medium (resembling that in a typical western colon, Table 1) promoted growth of Alistipes, Bilophila and Gammaproteobacteria, while reducing glycan and protein degraders (e.g. Bacteroides, Clostridium and Roseburia spp.), SCFAs and antioxidants; i.e. similar to high fat diets (e.g. Table 3) 57. Also, culturing mouse microbiome with high fat diet media (i.e. lard-based) promoted growth of Enterobacteriaceae (class Gammaproteobacteria) 23, while EPA/DHA directly inhibited E. coli (family Enterobacteriaceae) and promoted C. glomerans (phylum Actinobacteria) 58; i.e. both replicating changes seen in mice 23,58. In the relatively anoxic environment of the gut, “lipophilic” bacteria may perform β-oxidation of fats via anaerobic respiration with various alternate terminal electron acceptors (e.g. sulfate, nitrate, fumarate, etc.) 57. Exogenous fatty acids may also modulate microbial membranes and signalling/metabolism 59–63. Notably, fungi can also metabolise fats 47, while PUFAs 64, MCFAs (i.e. coconut oil) 47 and SCFAs 65 can have antifungal activity.

Dietary fats also alter the intestinal environment, foremost by stimulating bile release, which aids digestion and also modulates the microbiome 66. Accordingly, faecal bile acids were >3x higher in omnivores than vegans (n=32/32), and associated with higher fat and lower fibre intake 67; while in young adults (n=297), habitual animal fat intake correlated lower bacterial diversity and SCFAs, alongside increased bile acids (ns) 39. Moreover, in the large Chinese RCT above, higher fat consumption (i.e. mainly soybean oil) induced several microbiota changes (i.e. ↓ α-diversity, Faecalibacterium, SCFAs; ↑ Alistipes, Bacteriodes) 19 in association with increased bile acids 44. Bile acids are detergents with antimicrobial properties, although some microbes are more resistant than others (e.g. Gammaproteobacteria; Fig. 2e) 45,66; so their administration can lower microbial diversity 66,68 and favour growth of bile-resistant microbes 26,45. Similarly, high fat diets can increase intestinal bile acids 21,26,44,45,69, while their manipulation replicates microbial changes 26,45,66,68. For instance, high fat diets promoted Salmonella infection via bile-mediated (i.e. CA or TCA) suppression of core microbiota, but only in mice lacking competitive E. coli (another Enterobacteriaceae) 45. A particularly susceptible bacterium was Prevotella copri 45, which may generally decline on high fat diets 21,27 and with Westernisation 70. Notably, animal fats also come with cholesterol (i.e. bile acid precursor), but a high cholesterol diet did not significantly affect gut bile or microbiome, suggesting dietary fat content is more important 71.

Regarding differential effects, the abundance of sulfide-producing bacteria in human colon biopsies (from 9 healthy subjects) over several months was positively associated with intake of dairy/SFAs and negatively with MUFAs/PUFAs 41. Accordingly, high SFA/lard diets seem to particularly increase Desulfovibrionaceae (i.e. Bilophila 18,20,21,26,30,33,68 and Desulfovibrio 25,27,72,73), which reduce sulfur compounds to hydrogen sulfide (H2S). With milk fat, expansion of Bilophila wadsworthia was linked to taurine-conjugated bile acids (i.e. Fig. 3a) 26, which provide organic sulfonate for sulfite respiration; and may be stimulated by long-chain SFAs (e.g. C16:0/18:0) in relation to their greater hydrophobicity 36,74,75 and slower absorption 76–78. Conversely, sulfide-producing bacteria did not increase with oils rich in omega-6 25,26, and were even suppressed by addition of moderate omega-3 25,36,37 (i.e. at 1–7%; not 21% 30). However, both butter and refined olive oil (i.e. high MUFA) similarly increased Desulfovibrio (a sulfate-reducing genus), but not extra virgin olive oil, thus implicating other minor constituents (e.g. additives or polyphenols) 27. Notably, low fibre diets can also increase Desulfovibrio 52, and specifically D. piger 79, along with degradation of the mucus layer (releases sulfate) and enhanced pathogen susceptibly 79. Conversely, prior infection may actually enhance colonisation resistance via taurine-induced sulfide production 80.

Gut barrier

The gut microbiome is separated from the body by the gut barrier, which is demarcated by a single outer layer of epithelial cells and the mucus they exude. Diet greatly influences barrier permeability to microbes and their antigens, such as lipopolysaccharides (LPS, aka. endotoxin) from Gram-negative bacteria. In humans and animals, even single high energy/fat meals can induce a postprandial (0–5hr) increase in blood LPS 81–83 (with inflammation and oxidative stress 84–87), which is blocked by adding orange juice, polyphenols or fibre 88–90.

Moreover, many studies specifically implicate saturated fat (e.g. postprandial 85,87,91,92 and longer 18,23,25,30,54,93; reviews 81,83). For instance, in healthy adults, blood LPS was increased by 30g of cream (70% sat fat; low LPS content), but not orange juice or glucose (i.e. Fig. 3) 85. In pigs and humans, blood LPS was increased by a porridge-based meal made with 16g coconut oil (86% sat fat), but decreased with fish oil (500mg DHA), while there was no significant effect with other oils (i.e. olive, vegetable and grapeseed oil) 91,92; and no meal affected blood inflammatory markers 92. Further, in mice, 1 week of a high fat diet induced translocation of live commensal intestinal bacteria into blood and mesenteric adipose 94; while in a subsequent study, 8 weeks of a high saturated fat diet increased colonic permeability and translocation of bacterial DNA, whereas omega-6 did not and omega-3 was ameliorative 25. However, other mouse models show an elevated omega-6:3 ratio (e.g. 20:1) can increase blood LPS 24,31,95; and in people at risk of colon cancer, the dietary omega-6:3 ratio correlated blood LPS-binding protein (LBP) 96.

Modulation of intestinal permeability may occur via transcellular or paracellular routes (i.e. through or between cells, respectively) and various mechanisms (reviews 81,97). Firstly, fat absorption and incorporation into chylomicrons can promote auxiliary transport of LPS (a fat-containing molecule) into peripheral blood 81,98,99. However, the acute differential effects of saturated vs. omega-3 fats on LPS above were linked to modulation of intestinal membrane lipid rafts, which may represent an initial gateway 91. And recently, fat was shown to promote rapid transcellular transport of LPS from small intestine into portal vein via a lipid raft and CD36-dependent pathway, with slower and smaller transport into lymph via the chylomicron pathway 100.

Fat digestion also liberates free fatty acids (FFAs), which increase on a high fat diet and can induce lipotoxicity 101,102. In particular, saturated FFAs (e.g. palmitate) can induce ER/oxidative stress in intestinal cells, which was associated with inflammation and an impaired mucosal barrier 102. In addition, fat-induced changes to faecal bile acid profile (i.e. DCA:UDCA ratio) were linked to barrier dysfunction 103; and suggested to involve DCAs surfactant-related ability to disrupt lipid bilayers (e.g. cell membranes) 103. In an ex vivo study, fat-induced permeability mainly occurred in intestinal crypts, possibly due to the presence of immature cells lacking detergent-resistant lipid rafts with increased susceptibility to bile and FFAs 104. Notably, diets high in soybean oil or lard (40% fat) similarly decreased colon tight junction expression and increased paracellular permeability in relation to secondary bile acids 105; whereas saturated fat-specific barrier dysfunction has been linked to H2S-producing bacteria 25,37,69. Accordingly, B. wadsworthia can induce host bile and lower faecal butyrate, a barrier-supportive SCFA 69; while H2S can further impair SCFA metabolism by colon cells 33,106, and reduce disulfide bonds in the mucus network, thus increasing exposure of bacteria to the epithelium 51,107.

Host immunity

The gut also contains the largest collection of immune cells in the body (i.e. GALT), which support barrier function and interact reciprocally with microbes. As such, dietary fats can modulate immune activity via the gut microbiome. For instance, LPS binds to toll-like receptor 4 (TLR4) expressed on the surface of many cells to modulate inflammatory signalling. In the human gut, LPS largely originates from the Gram-negative Bacteroidetes phylum and has anti-inflammatory effects, as compared to that from Proteobacteria 108. However, high fat diets can alter the Firmicutes to Bacteroidetes ratio 19,21,23,26, and especially induce growth of Proteobacteria (i.e. Desulfovibrionaceae 18,25,27 or Enterobacteriaceae 23,31,95); as well as LPS biosynthesis 19,23,28, translocation (see above) and TLR-related inflammation in blood 84,85,88–90 and organs 23,28,30.

In addition, fats may have more direct effects. In 3-week RCTs on healthy adults, lowering the dietary palmitate/oleate ratio lowered (fasting) blood and tissue fatty acid ratios in relation to cytokine release (to LPS in vitro) 9,10 and muscle NLRP3 inflammasome (not ER stress) 10. In healthy humans, fat-induced postprandial inflammation has been suggested to involve saturated FFAs, rather than LPS 109. And in mouse models, high fat diets may impair intestinal immunity 101 and mucosal barrier via FFAs 102, and induce loss of gut neurons and dysmotility via both LPS and saturated FFA-mediated TLR4 signalling 28. Accordingly, fats make up cell membranes, including bacterial LPS—so named because it contains fat (lipo-) and carbohydrate (polysaccharide). In particular, saturated fats acylated in the lipid A portion of LPS are essential for signalling via TLR4, as well as lipopeptide signalling via TLR2 110. Further, cell membranes are heterogeneous and contain microdomains known as lipid rafts, which result from associations between cholesterol, sphingolipids and other saturated lipids, while proteins can be recruited via palmitoylations 111. Stimulation of cells with LPS triggers formation of a TLR4–lipid raft complex, which requires activation of sphingomyelinase to produce ceramide 112 (also produced de novo from palmitate). Moreover, saturated FFAs (e.g. lauric and palmitic) can also stimulate TLR4/2 signalling, via (redox-dependent 113) receptor dimerization and translocation into lipid rafts, which can be inhibited by DHA/omega-3 110,113–116. Note, intestinal enterocytes also mediate uptake of LPS via TLR4 117,118, which may mediate the differential effects of fats (and antioxidants) on intestinal permeability above 91 (e.g. figure below); while omega-3s were further shown to enrich the microdomains of tight junctions with unsaturated fats and prevent intestinal permeability induced by inflammatory cytokines 119,120. Inside cells, palmitate can induce synthesis of saturated glycerolipids/ceramide, lower ER membrane fluidity 121, and activate ER stress and NF-kB/NLRP3 inflammasome pathways 122–124, all of which is inhibited by unsaturated fats (i.e. oleate and DHA). Note however, some potential methodological issues with saturated fats in cell studies 125,126.

Omega-6 and 3 PUFAs broadly serve as precursors to pro- and anti-inflammatory mediators (i.e. oxylipins/eicosanoids), respectively, which may be paralleled by corresponding regulation of pro- and anti-inflammatory bacteria 24,25,31,35,37,95,127. For instance, in a 6-month RCT, higher dietary fat (i.e. 20 vs. 40% kcals; mostly via soybean oil—rich in C18:2, n-6) induced proinflammatory changes, including bacterial LPS biosynthesis and arachidonic acid (AA; i.e. C20:4, n-6) metabolism, while changes in faecal AA correlated its derivatives (i.e. PGE2 and TXB2) and CRP in plasma 19. An omega-3 trial also revealed a modest post-treatment inverse relation between bacterial diversity and colonic PGE2 34. In mice, systemic administration of resolvin D1 (derived from omega-3/DHA) ameliorated saturated fat-induced gut dysbiosis, permeability and inflammation 25. Further, in transgenic mouse models, a high tissue omega-6:3 ratio promoted systemic inflammation and gut dysbiosis characterised by overgrowth of Enterobacteriaceae and suppression of Bifidobacteria 31,95. This was linked to modulation of intestinal alkaline phosphatase (IAP) activity; where omega-3 induction of IAP reversed gut dysbiosis and decreased LPS production, translocation and inflammation 31. However, while a high omega-6 diet exacerbated infectious colitis, addition of omega-3 promoted sepsis, alongside suppression of immune/IAP responses 24; thus illustrating the potential for harm from excess (reviews 128,129). Notably, both saturated 23,25 and omega-6 fats 24,95 can promote intestinal inflammation, which itself can induce luminal release of respiratory acceptors (e.g. nitrate) for Enterobacteriaceae allowing them to bloom 50,130.

All these pathways further regulate T and B cells, and thus adaptive immunity. For instance, LPS–TLR4 signalling promotes Th17 activity and worsens experimental autoimmune encephalomyelitis (EAE) 131, an animal model of brain inflammation and multiple sclerosis (MS). Similarly, another study showed that dietary long-chain saturated fats (i.e. palmitic and lauric acid) promoted Th1/17 activity and worsened EAE, via suppression of intestinal SCFAs which induce Tregs 132. Interestingly, in humans, Dr. Swank published repeatedly on a link between animal/saturated fat and MS, including the long-term effect of a low fat diet on disease activity (YouTube). More recently, a low cholesterol diet (i.e. low animal fat?) was also shown to lower autoimmune potential (i.e. Th17/Treg balance) in people with HCV 133. Notably, compared to typical high fat diets, ketogenic diets (e.g. 80% fat, 5% carb) have distinct effects on the microbiome, mediated by the antimicrobial activity of ketones, resulting in lower intestinal Th17 cells 134.

Host energy

A primary function of the gut is to digest and absorb nutrients, where a symbiotic relationship with microbes extends and modulates systemic metabolism 135–138. As a macronutrient, dietary fat also fundamentally shapes metabolism, both directly and via microbes 33,139. For instance, putting healthy young, but slightly overweight, men on eucaloric diets high in SFAs or PUFAs (64% kcal) for 6 weeks induced extensive metabolic adaptation, with maintenance of insulin sensitivity 140. However, excessive fat intake (i.e. SFAs, MUFAs or PUFAs) can rapidly induce metabolic dysfunction 141–146, with saturated fat seeming most detrimental 54,141,147. For instance, in humans and mice, a single high dose of palm oil (50% sat fat) initiated whole-body, liver and adipose insulin resistance, alongside liver LPS signalling (not blood inflammation) 143.

Importantly, the small intestine also becomes insulin resistant early in human obesity 148, while itself regulating systemic glucose control (e.g. whole-body 149 and brain 150), as revealed by gastric bypass. This may involve intestinal gluconeogenesis and portal glucose, a signal sensed by nerves regulating gut–brain signalling and systemic energy homeostasis 151. In the post-absorptive period, this signal may be sustained by protein or fibre, which serve as reservoirs of gluconeogenic substrates, promoting satiety 151. Conversely, a high fat diet (36% safflower oil—rich in C18:2, n-6) impaired small intestinal glucose uptake and gluconeogenesis 151,152. In human obesity, enterocyte GLUT2 location is altered (i.e. apical and endosomal), in relation to insulin resistance, intestinal inflammation (i.e. CD8αβ T cells) and dietary calories/macros 153,154, while high fat diet models support causality 153. In fasting hyperglycaemic mice, apical GLUT2 increased blood–lumen glucose flux, with potential consequence for gut microbiome 153; note, small intestinal bacterial overgrowth (SIBO) is increased in metabolic disease 81 and obesity in relation to refined carb intake 155. Insulin also normally suppresses enterocyte lipid secretion (via chylomicrons), which was inhibited in mice fed palm vs. olive oil, which may promote postprandial hyperlipidaemia (a risk factor for CVD) 156.

In the colon, bacterial fermentation of carbs/fibre generates SCFAs and succinate which fuel intestinal metabolism and mediate gut–host signalling to regulate systemic energy metabolism (reviews 137,138). For instance, in overweight men, acute colonic infusion of SCFAs induced fasting fat oxidation 157; and in mice on a high fat diet, butyrate prevented insulin resistance and obesity, while increasing energy expenditure and mitochondrial function 158. Accordingly, high fat diets may lower SCFAs (as above) and suppress Prevotella copri 21,27,40,45,70, a succinate-producer with metabolic benefits 159 and human health association (n=1098) 160, while promoting overgrowth of pathobionts with detrimental effects. For instance, transplantation of specific Enterobacteriaceae from obese humans to mice fed a high fat diet induced endotoxin-dependant obesity, insulin resistance 161 and fatty liver 162. Also, in overweight adults, overfeeding saturated fat increased Proteobacteria and liver fat 54, which was associated with baseline Bilophila 18. In mice, B. wadsworthia aggravated metabolic dysfunctions, via inflammation-dependent and independent pathways 69. Similarly, in other studies, high fat diets induced gut microbiome changes which promoted inflammation and metabolic dysfunction, but largely independently 23,25,30. For instance, both high saturated and omega-6 fat diets induced weight gain; while only saturated fat induced H2S-producing bacteria (i.e. Desulfovibrio and Bilophila), leaky gut, inflammation and insulin resistance; and omega-3 ameliorated gut changes, but not insulin resistance 25. Omega-6 fats are also precursors to endocannabinoids which regulate gut–host physiology 163; while low–high fat diets with a high omega-6:3 ratio induced endocannabinoids and adiposity (not insulin) 164,165.

In humans and mice, acute high fat diet-induced muscle insulin resistance was causally linked to increased mtROS and oxidation of GSH redox 145. Accordingly, compared to carbs, fat oxidation generates more ROS, although this was lower with medium vs. long-chain fats (i.e. C10:0/12:0 vs. C16:0/18:1/18:2) 166 and may involve LCAD 167. In human tracer studies, dietary fat oxidation also decreases with chain length (e.g. C12:0>16:0>18:0 168 and C18:3>22:6 169) and saturation (e.g. C16:0/18:0 vs. C18:1/18:2/18:3; Fig. 4) 168,170,171. Further, in a series of 3–4 week RCTs on healthy adults, lowering the habitual dietary palmitate/oleate ratio (i.e. C16:0/18:1) lowered cytokines 9,10 and raised systemic energy/lipid metabolism 5–8,11. And in humans spanning the range of insulin sensitivity (i.e. athletes–lean–obese–T2D), there were inverse associations with intramuscular saturated fats and sphingolipids 172,173 (and SFA intake was increased in T2D 172); while in myotubes, palmitate (not oleate) oxidation was impaired in T2D 174. Blood FFAs are also elevated in metabolic diseases and ageing, with their saturated fat content (e.g. palmitate) linked to systemic lipotoxic effects (e.g. inflammation, insulin resistance, endothelial dysfunction, etc.) 114,175,176 and unsaturated fat (e.g. oleate and omega-3s) to protection 177–180. For instance, plasma palmitate fuels intramuscular ceramide synthesis in vivo 181,182, and induces ceramide-dependent insulin resistance in vitro (e.g. gut 156, vascular 175,183 and muscle cells 184–186), via TLR4-dependent ceramide biosynthesis 185 and mtROS 184, which can be ameliorated by oleate 184,186. Moreover, cerebrospinal palmitate is also increased in cardiometabolic disease; while palmitate (not oleate) infusion impaired memory in mice and neuronal insulin signalling in vitro via TNFα 187.

Host lipids

Importantly, fatty acids are also synthesised endogenously via de novo lipogenesis (from sugars), while FFAs are released by triglyceride lipolysis (inhibited by insulin). Consequently, eucaloric low carb/high fat diets can decrease lipogenesis and triglycerides 140,188, whereas overfeeding simple carbs increased lipogenesis and triglyceride saturated fat % 54. On the other hand, an Atkins-style low carb/high fat weight loss diet decreased postprandial insulin and increased FFAs 189, while overfeeding saturated fat (not unsaturated fat or simple carbs) increased lipolysis and blocked insulin-dependent suppression of FFAs 54; alongside increased endotoxemia, ceramides and insulin resistance 54. Notably, in people injected with LPS, insulin infusion also completely suppressed LPS-induced nitro-oxidative stress and FFAs 190. Low carb diets high in saturated fat also increase cholesterol (esp. LDL-c 140,188,191, which correlated FFAs 189), while those favouring unsaturated fat can induce lower blood lipids (e.g. lipogenesis/desaturation 140, triglycerides 191, SFA/PUFA ratios 140,188 and LDL-c 140,188,191) with higher ketones (e.g. after 5 days 191 or 6 weeks 140,188). Notably, fats and ketogenic diets can also differentially affect insulin sensitivity 191, while hepatic insulin signalling regulates lipid/lipoprotein metabolism 192–194; although differential lipid responses are still present even with preservation of insulin sensitivity 140,188. Saturated fats may favour more lipogenesis–desaturation to produce MUFAs (e.g. C16:1/18:1) 15,195, which support storage (as triglycerides) and maintain cell physiology (e.g. membrane fluidity and insulin sensitivity), buffering toxicity 15; whereas unsaturated fats lower desaturation/SCD activity 140,196,197. Further, the greater oxidation rate of long-chain unsaturated vs. saturated fats may favour more ketogenesis 169,198; and ketones can modulate gut microbiome and immunity 134. Long-chain omega-3s in particular can also lower triglycerides 199, by lowering VLDL production and increasing fatty acid uptake/oxidation 200,201. Moreover, in a 6-week RCT, omega-3 supplementation increased Coprococcus, which correlated butyrate (+) and VLDL-TGs (–), suggesting a role for the microbiome 56.

The blood cholesterol-raising effect of saturated fats is mainly attributable to C12–16:0 (not C18:0/stearic) 170 and related to lower LDL receptor expression (i.e. hepatic LDL uptake) 16,202,203; and potentially differential effects on cellular lipid homeostasis (e.g. via SREBPs) 16,195,204,205 and systemic turnover (e.g. intestinal absorption 206,207 and bile/excretion 16,202). Further, diets high in saturated fat can also promote lipoprotein oxidative/inflammatory modifications (e.g. vs. low fat 208–210, MUFAs 210–212 or PUFAs 213,214). In particular, in mice, a high saturated fat diet (vs. MUFA diet) increased body weight and HDL-c but impaired reverse cholesterol transport (RCT) from liver–faeces, alongside increased liver/adipose cholesterol and inflammation, and enrichment of HDL with acute-phase proteins 212. A similar diet also enriched liver and LDL with sphingomyelin (SM) and ceramide 208, which can mediate LDL aggregation, oxidation 208, inflammatory and apoptotic activity 215; and deliver ceramide to macrophages and muscle inducing insulin resistance 216. In humans, LDL aggregation was independently associated with CVD mortality and reduced by a ‘healthy Nordic diet’ in relation to increased PUFA intake and LDL-PC/SM ratios (i.e. Fig. S5B) 217. Note, both aggregated and oxidised LDL can induce foam cell formation (an early stage of atherosclerosis) 218, via TLR4 219 and in synergy with LPS 220.

As discussed earlier, dietary fat can modulate LPS translocation via portal and lymph-chylomicron pathways 98,100. The former would deliver LPS to the liver, where it may induce inflammation 100 (even in absence of blood inflammation 143); albeit limited by enteric release of HDL3, which is complexed with LBP and can bind and deactivate LPS 221. Similarly, the affinity of chylomicrons (aka. ULDL) for LPS is mediated by LBP and prevents endotoxin activity 222; although LPS-containing chylomicrons still activated lymph nodes 98, and may be delivered to other tissues such as adipose 223,224 and endothelium 225. In blood, LPS and Gram-positive LTA have affinity for all lipoproteins 226, especially HDL 227, and are subsequently transferred to LDL 228,229, while the hepatic LDL receptor plays a major role in their clearance 230,231 (e.g. figure below). Therefore lipoproteins (forward–reverse) transport both host and microbial lipids, subserving lipid homeostasis and innate immunity 232. Further, systemic infections/LPS induce many changes to lipid pathways, including lipid biosynthesis (e.g. fatty acids and sphingolipids), lipoprotein modifications and suppression of RCT, which may acutely support immunity, but if prolonged become harmful by promoting atherosclerosis 226,232,233; while diet can also chronically modulate these pathways. Accordingly, in a meta-analysis of 3 Finnish cohorts (n=7178), blood LPS was associated with an adverse metabolic profile (incl. lipoprotein sizes, fatty acids, glycation and amino acids), and more so with increased BMI and MetS 234. Further, compared to healthy weight people, metabolic disorders (e.g. obesity 99 and T2D 235) may be even more sensitive to saturated fat, with greater increases in postprandial LPS (and chylomicron enrichment 99), alongside decreasing LDL-c and HDL-c (not total cholesterol) 235; while in people with coronary disease (CORDIOPREV cohort), a high postprandial LPS predicted later development of T2D 236. This sensitivity might involve differences in gut LPS 18,108,161,162 and blood transport/clearance 99,235. Also, in human obesity, a subtly altered small intestinal barrier was exacerbated by lipid challenge (in vitro) and correlated intestinal/systemic inflammation and T2D 237; while postprandial LPS in chylomicrons correlated triglyceride responses 99,238, which may relate to intestinal insulin resistance, as above.

Dietary fats may also differentially affect metabolism of amyloid-beta (Aβ)—the major protein in brain plaques characterising Alzheimer’s disease (AD). In humans, brain amyloid burden is highly correlated with blood Aβ peptides 239,240, which largely associate with triglyceride-rich lipoproteins originating from gut and liver (i.e. chylomicrons and VLDL, respectively), also detectable in human CSF 241,242. In mice, a diet high in saturated fat (not MUFAs or PUFAs) increased Aβ production in enterocytes (concomitant with apoB-48 chylomicrons 243) and accumulation in blood 244; and induced disruption of blood–brain barrier allowing delivery of plasma proteins, including apoB-lipoproteins enriched with Aβ, into the brain 245. High (saturated) fat diet-induced fatty liver was also linked to AD 246; and Aβ production restricted to liver was shown to induce neurovascular dysfunction and inflammation with parenchymal extravasation 247. Moreover, both apoB-lipoproteins 248 and Aβ 249,250 are cleared by hepatic LDLR and LRP-1, while their induction lowers brain amyloid 249,251,252; further supporting a lipoprotein-Aβ vascular–brain axis 243,247,253. Interestingly, people with Alzheimer’s had increased (3-fold) postprandial chylomicrons, which may increase exposure to peripheral Aβ 241. In addition, the gut microbiome also likely plays an important role in AD 254,255; and in particular, gut Proteobacteria/SCFAs and blood LPS/SCFAs/cytokines have correlated CSF/brain amyloid 255–257, and can be modulated by diet as above. Further, LPS may be elevated in the brain and induce AD-like pathology 254,258, while Aβ may serve as an antimicrobial peptide in innate immunity 259.

Fats may also modulate systemic health via the meta-organismal TMAO pathway; where gut bacteria convert dietary choline/carnitine to TMA, which host FMO3 converts to TMAO. In humans, a 5-day high fat diet (50% sat fat) increased postprandial TMAO 260, while 4 weeks of a high (animal) fat Atkins diet increased fasting TMAO 261. Also, in a transgenic mouse model, an increased tissue omega-6:3 ratio elevated both faecal and blood TMAO 95. Fats may influence TMAO at various levels. Firstly, saturated fats can come with TMAO precursors in animal foods 260. Also, TMAO increases with age 262, which in mice was accompanied by increased Proteobacteria and Desulfovibrio (capable of producing TMA) 263, which can also be induced by fats (as above). Further, FMO3 is also under complex genetic and dietary regulation, including by bile–FXR signalling 264; and itself controls cholesterol balance 265 and obesity 266.

Perspective

In summary, dietary fats may differentially affect gut–host health; with higher levels of saturated (vs. unsaturated) and omega-6 (vs. omega-3) fats having unfavourable effects at multiple levels (i.e. gut microbiome and barrier; host immunity and metabolism). Mechanistically, the different physiochemical properties of saturated and unsaturated fats may require different bile acid responses for digestion and metabolic responses for homeostasis, while specific fatty acids serve as mediators/precursors regulating specific metabolic-immune pathways, with imbalances in any of these factors promoting dysfunction. In particular, as the first organ exposed to diet, the gut may represent ‘ground zero’ for dietary perturbations, while gut–host reciprocity may modulate systemic pathways (e.g. LPS/TLR signalling) involved in many chronic diseases (e.g. NAFLD, MetS, T2D, CVD, AD, etc.) and ageing, contributing to the differential effects of fats on systemic health.

In accord, dietary guidelines typically advise lowering saturated fats (e.g. <10% kcals) 212 and favouring foods richer in unsaturated fats and omega-3s (FAO). Some further considerations also seem noteworthy. Dietary fat content varies widely around the world, and generally increases with latitude, wealth and Westernisation 19,70,267. In 2013, in the UK and US respectively, 36% and 39% of calories came from fats, and much from refined sources such as oils (OWID). Compared to natural wholefoods, these sources isolate the fat/calories and fat-soluble nutrients, at the expense of everything else. For instance, compared to extra virgin olive oil (100% fat), whole olives are ~10–15% fat, which comes with gut-supporting fibre and >5x more polyphenols 268 (e.g. oleuropein and hydroxytyrosol); although curing does add salt. Moreover, the contemporary increase in oil consumption may have particularly increased omega-6 intake, modulating endogenous omega-3 levels and 6:3 ratios 17,165,269,270, and chronic inflammation/disease 13,19,95,165,271. On the other hand, with the popularisation of fish oil, its excessive consumption also has potential to be harmful, especially during infections 128,129. PUFAs are also prone to oxidation, which may be promoted by oil isolation (i.e. removal of protective food matrix/antiox), heating and even digestion; the latter being catalysed by co-ingestion of heme-iron (in red meat) and inhibited by polyphenols (in plants) 272. This may impact bioactivity, since oxidised fish oil had less favourable effects on lipoproteins in humans 273, and worsened gut dysbiosis in an animal model 274. Notably, both animal and plant fat sources (esp. dry heated) can also be high in advanced glycation end-products (AGEs) 275, which may also affect gut–host health 276 and blood lipids (incl. omega-3) 277.

Finally, PUFA status is not only related dietary content, but also fatty acid metabolism and genetic variation therein, supporting personalised nutrition 17. Strong relative correlations have been reported between changes to the blood omega-3 index (i.e. RBC EPA + DHA) and gastrointestinal tissues, suggesting it could serve as a surrogate measure 278.

Resources

References

1.           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).

2.           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).

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

5.           Kien, C. L., Bunn, J. Y. & Ugrasbul, F. Increasing dietary palmitic acid decreases fat oxidation and daily energy expenditure. Am. J. Clin. Nutr. 82, 320–6 (2005).

6.           Børsheim, E., Kien, C. L. & Pearl, W. M. Differential effects of dietary intake of palmitic acid and oleic acid on oxygen consumption during and after exercise. Metabolism. 55, 1215–21 (2006).

7.           Kien, C. L. & Bunn, J. Y. Gender alters the effects of palmitate and oleate on fat oxidation and energy expenditure. Obesity (Silver Spring). 16, 29–33 (2008).

8.           Kien, C. L. et al. Substituting dietary monounsaturated fat for saturated fat is associated with increased daily physical activity and resting energy expenditure and with changes in mood. Am. J. Clin. Nutr. 97, 689–97 (2013).

9.           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).

10.        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).

11.        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).

12.        Dimina, L. & Mariotti, F. The Postprandial Appearance of Features of Cardiometabolic Risk: Acute Induction and Prevention by Nutrients and Other Dietary Substances. Nutrients 11, 1–23 (2019).

13.        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).

14.        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).

15.        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).

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

17.        Chilton, F. H. et al. Precision Nutrition and Omega-3 Polyunsaturated Fatty Acids: A Case for Personalized Supplementation Approaches for the Prevention and Management of Human Diseases. Nutrients 9, 1–26 (2017).

18.        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).

19.        Wan, Y. et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: a 6-month randomised controlled-feeding trial. Gut 68, 1417–1429 (2019).

20.        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).

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

22.        Russell, W. R. et al. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am. J. Clin. Nutr. 93, 1062–72 (2011).

23.        Kim, K.-A., Gu, W., Lee, I.-A., Joh, E.-H. & Kim, D.-H. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One 7, e47713 (2012).

24.        Ghosh, S. et al. Fish oil attenuates omega-6 polyunsaturated fatty acid-induced dysbiosis and infectious colitis but impairs LPS dephosphorylation activity causing sepsis. PLoS One 8, e55468 (2013).

25.        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).

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

27.        Martínez, N. et al. Refined versus Extra Virgin Olive Oil High-Fat Diet Impact on Intestinal Microbiota of Mice and Its Relation to Different Physiological Variables. Microorganisms 7, (2019).

28.        Reichardt, F. et al. Western diet induces colonic nitrergic myenteric neuropathy and dysmotility in mice via saturated fatty acid- and lipopolysaccharide-induced TLR4 signalling. J. Physiol. 595, 1831–1846 (2017).

29.        Huang, E. Y. et al. Composition of dietary fat source shapes gut microbiota architecture and alters host inflammatory mediators in mouse adipose tissue. JPEN. J. Parenter. Enteral Nutr. 37, 746–54 (2013).

30.        Caesar, R., Tremaroli, V., Kovatcheva-Datchary, P., Cani, P. D. & Bäckhed, F. Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab. 22, 658–68 (2015).

31.        Kaliannan, K., Wang, B., Li, X.-Y., Kim, K.-J. & Kang, J. X. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci. Rep. 5, 11276 (2015).

32.        Wolters, M. et al. Dietary fat, the gut microbiota, and metabolic health – A systematic review conducted within the MyNewGut project. Clin. Nutr. 38, 2504–2520 (2019).

33.        Shen, W., Gaskins, H. R. & McIntosh, M. K. Influence of dietary fat on intestinal microbes, inflammation, barrier function and metabolic outcomes. J. Nutr. Biochem. 25, 270–280 (2014).

34.        Djuric, Z. et al. Increases in colonic bacterial diversity after ω-3 fatty acid supplementation predict decreased colonic prostaglandin E2 concentrations in healthy adults. J. Nutr. 149, 1170–1179 (2019).

35.        Vetrani, C. et al. Diets naturally rich in polyphenols and/or long-chain n-3 polyunsaturated fatty acids differently affect microbiota composition in high-cardiometabolic-risk individuals. Acta Diabetol. 57, 853–860 (2020).

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

37.        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).

38.        Claesson, M. J. et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 488, 178–84 (2012).

39.        Wan, Y. et al. Habitual animal fat consumption in shaping gut microbiota and microbial metabolites. Food Funct. 10, 7973–7982 (2019).

40.        Wu, G. D. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105–8 (2011).

41.        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).

42.        Menni, C. et al. Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women. Sci. Rep. 7, 11079 (2017).

43.        De Filippis, F. et al. Distinct Genetic and Functional Traits of Human Intestinal Prevotella copri Strains Are Associated with Different Habitual Diets. Cell Host Microbe 25, 444-453.e3 (2019).

44.        Wan, Y. et al. Unconjugated and secondary bile acid profiles in response to higher-fat, lower-carbohydrate diet and associated with related gut microbiota: A 6-month randomized controlled-feeding trial. Clin. Nutr. 39, 395–404 (2020).

45.        Wotzka, S. Y. et al. Escherichia coli limits Salmonella Typhimurium infections after diet shifts and fat-mediated microbiota perturbation in mice. Nat. Microbiol. 4, 2164–2174 (2019).

46.        Mefferd, C. C. et al. A High-Fat/High-Protein, Atkins-Type Diet Exacerbates Clostridioides (Clostridium) difficile Infection in Mice, whereas a High-Carbohydrate Diet Protects. mSystems 5, (2020).

47.        Gunsalus, K. T. W., Tornberg-Belanger, S. N., Matthan, N. R., Lichtenstein, A. H. & Kumamoto, C. A. Manipulation of Host Diet To Reduce Gastrointestinal Colonization by the Opportunistic Pathogen Candida albicans. mSphere 1, e00020-15 (2016).

48.        Sun, Y. & O’Riordan, M. X. D. Regulation of bacterial pathogenesis by intestinal short-chain Fatty acids. Adv. Appl. Microbiol. 85, 93–118 (2013).

49.        Jacobson, A. et al. A Gut Commensal-Produced Metabolite Mediates Colonization Resistance to Salmonella Infection. Cell Host Microbe 24, 296-307.e7 (2018).

50.        Rivera-Chávez, F., Lopez, C. A. & Bäumler, A. J. Oxygen as a driver of gut dysbiosis. Free Radic. Biol. Med. 105, 93–101 (2017).

51.        Teigen, L. M. et al. Dietary Factors in Sulfur Metabolism and Pathogenesis of Ulcerative Colitis. Nutrients 11, 1–20 (2019).

52.        Morrison, K. E., Jašarević, E., Howard, C. D. & Bale, T. L. It’s the fiber, not the fat: significant effects of dietary challenge on the gut microbiome. Microbiome 8, 15 (2020).

53.        Creedon, A. C., Hung, E. S., Berry, S. E. & Whelan, K. Nuts and their Effect on Gut Microbiota, Gut Function and Symptoms in Adults: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Nutrients 12, (2020).

54.        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).

55.        Tindall, A. M., McLimans, C. J., Petersen, K. S., Kris-Etherton, P. M. & Lamendella, R. Walnuts and Vegetable Oils Containing Oleic Acid Differentially Affect the Gut Microbiota and Associations with Cardiovascular Risk Factors: Follow-up of a Randomized, Controlled, Feeding Trial in Adults at Risk for Cardiovascular Disease. J. Nutr. 150, 806–817 (2020).

56.        Vijay, A., Astbury, S., Le Roy, C., Spector, T. D. & Valdes, A. M. The prebiotic effects of omega-3 fatty acid supplementation: A six-week randomised intervention trial. Gut Microbes 13, 1–11 (2021).

57.        Agans, R. et al. Dietary Fatty Acids Sustain the Growth of the Human Gut Microbiota. Appl. Environ. Microbiol. 84, (2018).

58.        Zhuang, P. et al. Eicosapentaenoic and docosahexaenoic acids attenuate hyperglycemia through the microbiome-gut-organs axis in db/db mice. Microbiome 9, 185 (2021).

59.        Emiola, A., Andrews, S. S., Heller, C. & George, J. Crosstalk between the lipopolysaccharide and phospholipid pathways during outer membrane biogenesis in Escherichia coli. Proc. Natl. Acad. Sci. U. S. A. 113, 3108–3113 (2016).

60.        Kankaanpää, P., Yang, B., Kallio, H., Isolauri, E. & Salminen, S. Effects of polyunsaturated fatty acids in growth medium on lipid composition and on physicochemical surface properties of lactobacilli. Appl. Environ. Microbiol. 70, 129–36 (2004).

61.        Eder, A. E. et al. Exogenous polyunsaturated fatty acids (PUFAs) alter phospholipid composition, membrane permeability, biofilm formation and motility in Acinetobacter baumannii. Microbiology 163, 1626–1636 (2017).

62.        Herndon, J. L. et al. Exogenous polyunsaturated fatty acids (PUFAs) promote changes in growth, phospholipid composition, membrane permeability and virulence phenotypes in Escherichia coli. BMC Microbiol. 20, 305 (2020).

63.        Zheng, C. J. et al. Fatty acid synthesis is a target for antibacterial activity of unsaturated fatty acids. FEBS Lett. 579, 5157–62 (2005).

64.        Thibane, V. S. et al. Polyunsaturated fatty acids cause apoptosis in C. albicans and C. dubliniensis biofilms. Biochim. Biophys. Acta 1820, 1463–8 (2012).

65.        Nguyen, L. N., Lopes, L. C. L., Cordero, R. J. B. & Nosanchuk, J. D. Sodium butyrate inhibits pathogenic yeast growth and enhances the functions of macrophages. J. Antimicrob. Chemother. 66, 2573–80 (2011).

66.        Yokota, A. et al. Is bile acid a determinant of the gut microbiota on a high-fat diet? Gut Microbes 3, 455–9 (2012).

67.        Trefflich, I. et al. Associations between Dietary Patterns and Bile Acids—Results from a Cross-Sectional Study in Vegans and Omnivores. Nutrients 12, 47 (2019).

68.        Zheng, X. et al. Bile acid is a significant host factor shaping the gut microbiome of diet-induced obese mice. BMC Biol. 15, 120 (2017).

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

70.        Tett, A. et al. The Prevotella copri Complex Comprises Four Distinct Clades Underrepresented in Westernized Populations. Cell Host Microbe 26, 666-679.e7 (2019).

71.        Dimova, L. G., Zlatkov, N., Verkade, H. J., Uhlin, B. E. & Tietge, U. J. F. High-cholesterol diet does not alter gut microbiota composition in mice. Nutr. Metab. (Lond). 14, 15 (2017).

72.        Zhang, C. et al. Structural resilience of the gut microbiota in adult mice under high-fat dietary perturbations. ISME J. 6, 1848–57 (2012).

73.        Onishi, J. C. et al. Bacterial communities in the small intestine respond differently to those in the caecum and colon in mice fed low- and high-fat diets. Microbiology 163, 1189–1197 (2017).

74.        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).

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

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

77.        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).

78.        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).

79.        Desai, M. S. et al. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 167, 1339-1353.e21 (2016).

80.        Stacy, A. et al. Infection trains the host for microbiota-enhanced resistance to pathogens. Cell 184, 615-627.e17 (2021).

81.        Moreira, A. P. B., Texeira, T. F. S., Ferreira, A. B., Peluzio, M. D. C. G. & Alfenas, R. D. C. G. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br. J. Nutr. 108, 801–9 (2012).

82.        Pendyala, S., Walker, J. M. & Holt, P. R. A high-fat diet is associated with endotoxemia that originates from the gut. Gastroenterology 142, 1100-1101.e2 (2012).

83.        Kelly, C. J., Colgan, S. P. & Frank, D. N. Of microbes and meals: the health consequences of dietary endotoxemia. Nutr. Clin. Pract. 27, 215–25 (2012).

84.        Ghanim, H. et al. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance. Diabetes Care 32, 2281–7 (2009).

85.        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).

86.        Dandona, P. et al. Decreased insulin secretion and incretin concentrations and increased glucagon concentrations after a high-fat meal when compared with a high-fruit and -fiber meal. Am. J. Physiol. Endocrinol. Metab. 308, E185-91 (2015).

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

88.        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).

89.        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).

90.        Ghanim, H. et al. Orange juice neutralizes the proinflammatory effect of a high-fat, high-carbohydrate meal and prevents endotoxin increase and Toll-like receptor expression. Am. J. Clin. Nutr. 91, 940–9 (2010).

91.        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).

92.        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).

93.        Guevara-Cruz, M. et al. Improvement of Lipoprotein Profile and Metabolic Endotoxemia by a Lifestyle Intervention That Modifies the Gut Microbiota in Subjects With Metabolic Syndrome. J. Am. Heart Assoc. 8, e012401 (2019).

94.        Amar, J. et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol. Med. 3, 559–72 (2011).

95.        Kaliannan, K. et al. Multi-omic analysis in transgenic mice implicates omega-6/omega-3 fatty acid imbalance as a risk factor for chronic disease. Commun. Biol. 2, 276 (2019).

96.        Umoh, F. I. et al. Markers of systemic exposures to products of intestinal bacteria in a dietary intervention study. Eur. J. Nutr. 55, 793–798 (2016).

97.        Rohr, M. W., Narasimhulu, C. A., Rudeski-Rohr, T. A. & Parthasarathy, S. Negative Effects of a High-Fat Diet on Intestinal Permeability: A Review. Adv. Nutr. 11, 77–91 (2020).

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

99.        Vors, C. et al. Postprandial Endotoxemia Linked With Chylomicrons and Lipopolysaccharides Handling in Obese Versus Lean Men: A Lipid Dose-Effect Trial. J. Clin. Endocrinol. Metab. 100, 3427–35 (2015).

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

101.      Tanaka, S. et al. High-fat diet-derived free fatty acids impair the intestinal immune system and increase sensitivity to intestinal epithelial damage. Biochem. Biophys. Res. Commun. 522, 971–977 (2020).

102.      Gulhane, M. et al. High Fat Diets Induce Colonic Epithelial Cell Stress and Inflammation that is Reversed by IL-22. Sci. Rep. 6, 28990 (2016).

103.      Stenman, L. K., Holma, R., Eggert, A. & Korpela, R. A novel mechanism for gut barrier dysfunction by dietary fat: epithelial disruption by hydrophobic bile acids. Am. J. Physiol. Gastrointest. Liver Physiol. 304, G227-34 (2013).

104.      Danielsen, E. M., Hansen, G. H., Rasmussen, K. & Niels-Christiansen, L.-L. Permeabilization of enterocytes induced by absorption of dietary fat. Mol. Membr. Biol. 30, 261–72 (2013).

105.      Murakami, Y., Tanabe, S. & Suzuki, T. High-fat Diet-induced Intestinal Hyperpermeability is Associated with Increased Bile Acids in the Large Intestine of Mice. J. Food Sci. 81, H216-22 (2016).

106.      Carbonero, F., Benefiel, A. C., Alizadeh-Ghamsari, A. H. & Gaskins, H. R. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front. Physiol. 3, 448 (2012).

107.      Ijssennagger, N., van der Meer, R. & van Mil, S. W. C. Sulfide as a Mucus Barrier-Breaker in Inflammatory Bowel Disease? Trends Mol. Med. 22, 190–199 (2016).

108.      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).

109.      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).

110.      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).

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

112.      Ruysschaert, J. M. & Lonez, C. Role of lipid microdomains in TLR-mediated signalling. Biochim. Biophys. Acta - Biomembr. 1848, 1860–1867 (2015).

113.      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).

114.      Li, B., Leung, J. C. K., Chan, L. Y. Y., Yiu, W. H. & Tang, S. C. W. A global perspective on the crosstalk between saturated fatty acids and Toll-like receptor 4 in the etiology of inflammation and insulin resistance. Progress in Lipid Research 77, 101020 (2020).

115.      Rocha, D. M., Caldas, A. P., Oliveira, L. L., Bressan, J. & Hermsdorff, H. H. Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis 244, 211–215 (2016).

116.      Gupta, S., Knight, A. G., Gupta, S., Keller, J. N. & Bruce-Keller, A. J. Saturated long-chain fatty acids activate inflammatory signaling in astrocytes. J. Neurochem. 120, 1060–71 (2012).

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

118.      Hornef, M. W., Frisan, T., Vandewalle, A., Normark, S. & Richter-Dahlfors, A. Toll-like receptor 4 resides in the Golgi apparatus and colocalizes with internalized lipopolysaccharide in intestinal epithelial cells. J. Exp. Med. 195, 559–70 (2002).

119.      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).

120.      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).

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

122.      Korbecki, J. & Bajdak-Rusinek, K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm. Res. 68, 915–932 (2019).

123.      Snodgrass, R. G. et al. Docosahexaenoic acid and palmitic acid reciprocally modulate monocyte activation in part through endoplasmic reticulum stress. J. Nutr. Biochem. 32, 39–45 (2016).

124.      Sui, Y.-H., Luo, W.-J., Xu, Q.-Y. & Hua, J. Dietary saturated fatty acid and polyunsaturated fatty acid oppositely affect hepatic NOD-like receptor protein 3 inflammasome through regulating nuclear factor-kappa B activation. World J. Gastroenterol. 22, 2533–44 (2016).

125.      Ono-Moore, K. D., Blackburn, M. L. & Adams, S. H. Is palmitate truly proinflammatory? Experimental confounders and context-specificity. Am. J. Physiol. Endocrinol. Metab. 315, E780–E794 (2018).

126.      Huang, S. et al. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J. Lipid Res. 53, 2002–13 (2012).

127.      Watson, H. et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 67, 1974–1983 (2018).

128.      Fenton, J. I., Hord, N. G., Ghosh, S. & Gurzell, E. a. Immunomodulation by dietary long chain omega-3 fatty acids and the potential for adverse health outcomes. Prostaglandins. Leukot. Essent. Fatty Acids 89, 379–90 (2013).

129.      Husson, M.-O. et al. Modulation of host defence against bacterial and viral infections by omega-3 polyunsaturated fatty acids. J. Infect. 73, 523–535 (2016).

130.      Winter, S. E. & Bäumler, A. J. Dysbiosis in the inflamed intestine: chance favors the prepared microbe. Gut Microbes 5, 71–3 (2014).

131.      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).

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

133.      Maggio, R. et al. Normocaloric low cholesterol diet modulates Th17/Treg balance in patients with chronic hepatitis C virus infection. PLoS One 9, e112346 (2014).

134.      Ang, Q. Y. et al. Ketogenic Diets Alter the Gut Microbiome Resulting in Decreased Intestinal Th17 Cells. Cell 181, 1263-1275.e16 (2020).

135.      Velagapudi, V. R. et al. The gut microbiota modulates host energy and lipid metabolism in mice. J. Lipid Res. 51, 1101–12 (2010).

136.      Visconti, A. et al. Interplay between the human gut microbiome and host metabolism. Nat. Commun. 10, 4505 (2019).

137.      Sukkar, A. H., Lett, A. M., Frost, G. & Chambers, E. S. Regulation of energy expenditure and substrate oxidation by short-chain fatty acids. J. Endocrinol. 242, R1–R8 (2019).

138.      de Vadder, F. & Mithieux, G. Gut-brain signaling in energy homeostasis: The unexpected role of microbiota-derived succinate. J. Endocrinol. 236, R105–R108 (2018).

139.      Geurts, L., Neyrinck,  a. M., Delzenne, N. M., Knauf, C. & Cani, P. D. Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: Novel insights into molecular targets and interventions using prebiotics. Benef. Microbes 5, 3–17 (2014).

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

141.      Sarabhai, T. et al. Monounsaturated fat rapidly induces hepatic gluconeogenesis and whole-body insulin resistance. JCI insight 5, (2020).

142.      Parry, S., Woods, R., Hodson, L. & Hulston, C. A Single Day of Excessive Dietary Fat Intake Reduces Whole-Body Insulin Sensitivity: The Metabolic Consequence of Binge Eating. Nutrients 9, 818 (2017).

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

144.      Nowotny, B. et al. Mechanisms underlying the onset of oral lipid-induced skeletal muscle insulin resistance in humans. Diabetes 62, 2240–2248 (2013).

145.      Anderson, E. J. et al. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J. Clin. Invest. 119, 573–81 (2009).

146.      Lundsgaard, A. M. et al. Opposite regulation of insulin sensitivity by dietary lipid versus carbohydrate excess. Diabetes 66, 2583–2595 (2017).

147.      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).

148.      Mäkinen, J. et al. Obesity-associated intestinal insulin resistance is ameliorated after bariatric surgery. Diabetologia 58, 1055–62 (2015).

149.      Angelini, G. et al. Small intestinal metabolism is central to whole-body insulin resistance. Gut gutjnl-2020-322073 (2020). doi:10.1136/gutjnl-2020-322073

150.      Li, N. et al. Duodenal-Jejunal Bypass Ameliorates Type 2 Diabetes Mellitus by Activating Insulin Signaling and Improving Glucose Utilization in the Brain. Obes. Surg. 30, 279–289 (2020).

151.      Soty, M., Gautier-Stein, A., Rajas, F. & Mithieux, G. Gut-Brain Glucose Signaling in Energy Homeostasis. Cell Metabolism 25, 1231–1242 (2017).

152.      Mithieux, G., Rajas, F. & Zitoun, C. Glucose utilization is suppressed in the gut of insulin-resistant high fat-fed rats and is restored by metformin. Biochem. Pharmacol. 72, 1757–62 (2006).

153.      Ait-Omar, A. et al. GLUT2 accumulation in enterocyte apical and intracellular membranes: a study in morbidly obese human subjects and ob/ob and high fat-fed mice. Diabetes 60, 2598–607 (2011).

154.      Monteiro-Sepulveda, M. et al. Jejunal T Cell Inflammation in Human Obesity Correlates with Decreased Enterocyte Insulin Signaling. Cell Metab. 22, 113–24 (2015).

155.      Ierardi, E. et al. Macronutrient intakes in obese subjects with or without small intestinal bacterial overgrowth: an alimentary survey. Scand. J. Gastroenterol. 51, 277–80 (2016).

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

157.      Canfora, E. E. et al. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci. Rep. 7, 2360 (2017).

158.      Gao, Z. et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58, 1509–1517 (2009).

159.      De Vadder, F. et al. Microbiota-Produced Succinate Improves Glucose Homeostasis via Intestinal Gluconeogenesis. Cell Metab. 24, 151–7 (2016).

160.      Asnicar, F. et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat. Med. in press, (2021).

161.      Fei, N. & Zhao, L. An opportunistic pathogen isolated from the gut of an obese human causes obesity in germfree mice. ISME J. 7, 880–4 (2013).

162.      Fei, N. et al. Endotoxin producers overgrowing in human gut microbiota as the causative agents for nonalcoholic fatty liver disease. MBio 11, (2020).

163.      Forte, N., Fernández-Rilo, A. C., Palomba, L., Di Marzo, V. & Cristino, L. Obesity Affects the Microbiota-Gut-Brain Axis and the Regulation Thereof by Endocannabinoids and Related Mediators. Int. J. Mol. Sci. 21, 1–25 (2020).

164.      Alvheim, A. R. et al. Dietary linoleic acid elevates the endocannabinoids 2-AG and anandamide and promotes weight gain in mice fed a low fat diet. Lipids 49, 59–69 (2014).

165.      Alvheim, A. R. et al. Dietary linoleic acid elevates endogenous 2-AG and anandamide and induces obesity. Obesity (Silver Spring). 20, 1984–94 (2012).

166.      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).

167.      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).

168.      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).

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

170.      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).

171.      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).

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

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

174.      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).

175.      Symons, J. D. & Abel, E. D. Lipotoxicity contributes to endothelial dysfunction: a focus on the contribution from ceramide. Rev. Endocr. Metab. Disord. 14, 59–68 (2013).

176.      Estadella, D. et al. Lipotoxicity: effects of dietary saturated and transfatty acids. Mediators Inflamm. 2013, 137579 (2013).

177.      Ertunc, M. E. & Hotamisligil, G. S. Lipid signaling and lipotoxicity in metaflammation: indications for metabolic disease pathogenesis and treatment. J. Lipid Res. 57, 2099–2114 (2016).

178.      Kwon, B. & Querfurth, H. W. Opposite effects of saturated and unsaturated free fatty acids on intracellular signaling and metabolism in neuronal cells. Inflamm. Cell Signal. 1, 10-14800/ics.200 (2014).

179.      Pararasa, C., Bailey, C. J. & Griffiths, H. R. Ageing, adipose tissue, fatty acids and inflammation. Biogerontology 16, 235–48 (2015).

180.      Acosta-Montaño, P. & García-González, V. Effects of Dietary Fatty Acids in Pancreatic Beta Cell Metabolism, Implications in Homeostasis. Nutrients 10, (2018).

181.      Chung, J. O., Koutsari, C., Blachnio-Zabielska, A. U., Hames, K. C. & Jensen, M. D. Intramyocellular Ceramides: Subcellular Concentrations and Fractional De Novo Synthesis in Postabsorptive Humans. Diabetes 66, 2082–2091 (2017).

182.      Zabielski, P., Błachnio-Zabielska, A. U., Wójcik, B., Chabowski, A. & Górski, J. Effect of plasma free fatty acid supply on the rate of ceramide synthesis in different muscle types in the rat. PLoS One 12, e0187136 (2017).

183.      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).

184.      Yuzefovych, L., Wilson, G. & Rachek, L. Different effects of oleate vs. palmitate on mitochondrial function, apoptosis, and insulin signaling in L6 skeletal muscle cells: role of oxidative stress. Am. J. Physiol. Endocrinol. Metab. 299, E1096-105 (2010).

185.      Holland, W. L. et al. Lipid-induced insulin resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. J. Clin. Invest. 121, 1858–70 (2011).

186.      Sabin, M. A. et al. Fatty acid-induced defects in insulin signalling, in myotubes derived from children, are related to ceramide production from palmitate rather than the accumulation of intramyocellular lipid. J. Cell. Physiol. 211, 244–52 (2007).

187.      Melo, H. M. et al. Palmitate Is Increased in the Cerebrospinal Fluid of Humans with Obesity and Induces Memory Impairment in Mice via Pro-inflammatory TNF-α. Cell Rep. 30, 2180-2194.e8 (2020).

188.      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).

189.      Hernandez, T. L. et al. Lack of suppression of circulating free fatty acids and hypercholesterolemia during weight loss on a high-fat, low-carbohydrate diet. Am. J. Clin. Nutr. 91, 578–85 (2010).

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

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

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

193.      Ormazabal, V. et al. Association between insulin resistance and the development of cardiovascular disease. Cardiovasc. Diabetol. 17, 122 (2018).

194.      Ramakrishnan, G., Arjuman, A., Suneja, S., Das, C. & Chandra, N. C. The association between insulin and low-density lipoprotein receptors. Diabetes Vasc. Dis. Res. 9, 196–204 (2012).

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

196.      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).

197.      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).

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

199.      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).

200.      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).

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

202.      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).

203.      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).

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

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

206.      Yang, F. et al. Fatty acids modulate the expression levels of key proteins for cholesterol absorption in Caco-2 monolayer. Lipids Health Dis. 17, 32 (2018).

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

208.      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).

209.      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).

210.      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).

211.      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).

212.      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).

213.      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).

214.      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).

215.      Puig, N., Estruch, M., Jin, L., Sanchez-Quesada, J. L. & Benitez, S. The Role of Distinctive Sphingolipids in the Inflammatory and Apoptotic Effects of Electronegative LDL on Monocytes. Biomolecules 9, (2019).

216.      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).

217.      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).

218.      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).

219.      Singh, R. K. et al. TLR4 (Toll-Like Receptor 4)-Dependent Signaling Drives Extracellular Catabolism of LDL (Low-Density Lipoprotein) Aggregates. Arterioscler. Thromb. Vasc. Biol. 40, 86–102 (2020).

220.      Howell, K. W. et al. Toll-like Receptor 4 Mediates Oxidized LDL-Induced Macrophage Differentiation to Foam Cells. J. Surg. Res. 171, e27–e31 (2011).

221.      Han, Y.-H. et al. Enterically derived high-density lipoprotein restrains liver injury through the portal vein. Science (80-. ). 373, eabe6729 (2021).

222.      Vreugdenhil, A. C. E. et al. Lipopolysaccharide (LPS)-binding protein mediates LPS detoxification by chylomicrons. J. Immunol. 170, 1399–405 (2003).

223.      Hersoug, L.-G., Møller, P. & Loft, S. Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: implications for inflammation and obesity. Obes. Rev. 17, 297–312 (2016).

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

225.      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).

226.      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).

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

228.      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).

229.      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).

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

231.      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).

232.      Azzam, K. M. & Fessler, M. B. Crosstalk between reverse cholesterol transport and innate immunity. Trends Endocrinol. Metab. 23, 169–178 (2012).

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

234.      Määttä, A.-M. et al. Endotoxemia is associated with an adverse metabolic profile. Innate Immun. 27, 3–14 (2021).

235.      Harte, A. L. et al. High fat intake leads to acute postprandial exposure to circulating endotoxin in type 2 diabetic subjects. Diabetes Care 35, 375–382 (2012).

236.      Camargo, A. et al. Postprandial endotoxemia may influence the development of type 2 diabetes mellitus: From the CORDIOPREV study. Clin. Nutr. 38, 529–538 (2019).

237.      Genser, L. et al. Increased jejunal permeability in human obesity is revealed by a lipid challenge and is linked to inflammation and type 2 diabetes. J. Pathol. 246, 217–230 (2018).

238.      Clemente-Postigo, M. et al. Endotoxin increase after fat overload is related to postprandial hypertriglyceridemia in morbidly obese patients. J. Lipid Res. 53, 973–978 (2012).

239.      Schindler, S. E. et al. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology 93, e1647–e1659 (2019).

240.      Nakamura, A. et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature 554, 249–254 (2018).

241.      Mamo, J. C. L. et al. Plasma lipoprotein beta-amyloid in subjects with Alzheimer’s disease or mild cognitive impairment. Ann. Clin. Biochem. 45, 395–403 (2008).

242.      James, A. P., Pal, S., Gennat, H. C., Vine, D. F. & Mamo, J. C. L. The incorporation and metabolism of amyloid-beta into chylomicron-like lipid emulsions. J. Alzheimers. Dis. 5, 179–88 (2003).

243.      Takechi, R., Galloway, S., Pallebage-Gamarallage, M. M. S. & Mamo, J. C. L. Chylomicron amyloid-beta in the aetiology of Alzheimer’s disease. Atheroscler. Suppl. 9, 19–25 (2008).

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

245.      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).

246.      Kim, D.-G. et al. Non-alcoholic fatty liver disease induces signs of Alzheimer’s disease (AD) in wild-type mice and accelerates pathological signs of AD in an AD model. J. Neuroinflammation 13, 1 (2016).

247.      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).

248.      Frazier-Wood, A. C. et al. The association between LRP-1 variants and chylomicron uptake after a high fat meal. Nutr. Metab. Cardiovasc. Dis. 23, 1154–8 (2013).

249.      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).

250.      Tamaki, C., Ohtsuki, S. & Terasaki, T. Insulin facilitates the hepatic clearance of plasma amyloid beta-peptide (1 40) by intracellular translocation of low-density lipoprotein receptor-related protein 1 (LRP-1) to the plasma membrane in hepatocytes. Mol. Pharmacol. 72, 850–5 (2007).

251.      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).

252.      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).

253.      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).

254.      Zhan, X., Stamova, B. & Sharp, F. R. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer’s Disease Brain: A Review. Front. Aging Neurosci. 10, 42 (2018).

255.      Nagpal, R., Neth, B. J., Wang, S., Craft, S. & Yadav, H. Modified Mediterranean-ketogenic diet modulates gut microbiome and short-chain fatty acids in association with Alzheimer’s disease markers in subjects with mild cognitive impairment. EBioMedicine 47, 529–542 (2019).

256.      Cattaneo, A. et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 49, 60–68 (2017).

257.      Marizzoni, M. et al. Short-Chain Fatty Acids and Lipopolysaccharide as Mediators Between Gut Dysbiosis and Amyloid Pathology in Alzheimer’s Disease. J. Alzheimers. Dis. 78, 683–697 (2020).

258.      Brown, G. C. The endotoxin hypothesis of neurodegeneration. J. Neuroinflammation 2019 161 16, 1–10 (2019).

259.      Moir, R. D., Lathe, R. & Tanzi, R. E. The antimicrobial protection hypothesis of Alzheimer’s disease. Alzheimers. Dement. 14, 1602–1614 (2018).

260.      Boutagy, N. E. et al. Short-term high-fat diet increases postprandial trimethylamine-N-oxide in humans. Nutr. Res. 35, 858–64 (2015).

261.      Park, J. E., Miller, M., Rhyne, J., Wang, Z. & Hazen, S. L. Differential effect of short-term popular diets on TMAO and other cardio-metabolic risk markers. Nutr. Metab. Cardiovasc. Dis. 29, 513–517 (2019).

262.      Brunt, V. E. et al. Trimethylamine-N-Oxide Promotes Age-Related Vascular Oxidative Stress and Endothelial Dysfunction in Mice and Healthy Humans. Hypertension 76, 101–112 (2020).

263.      Brunt, V. E. et al. Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice. J. Physiol. 597, 2361–2378 (2019).

264.      Bennett, B. J. et al. Trimethylamine-N-Oxide, a Metabolite Associated with Atherosclerosis, Exhibits Complex Genetic and Dietary Regulation. Cell Metab. 17, 49–60 (2013).

265.      Warrier, M. et al. The TMAO-Generating Enzyme Flavin Monooxygenase 3 Is a Central Regulator of Cholesterol Balance. Cell Rep. 10, 326–338 (2015).

266.      Schugar, R. C. et al. The TMAO-Producing Enzyme Flavin-Containing Monooxygenase 3 Regulates Obesity and the Beiging of White Adipose Tissue. Cell Rep. 19, 2451–2461 (2017).

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

268.      Pérez-Jiménez, J., Neveu, V., Vos, F. & Scalbert, A. Identification of the 100 richest dietary sources of polyphenols: An application of the Phenol-Explorer database. Eur. J. Clin. Nutr. 64, S112–S120 (2010).

269.      Wood, K. E., Mantzioris, E., Gibson, R. A., Ramsden, C. E. & Muhlhausler, B. S. The effect of modifying dietary LA and ALA intakes on omega-3 long chain polyunsaturated fatty acid (n-3 LCPUFA) status in human adults: a systematic review and commentary. Prostaglandins. Leukot. Essent. Fatty Acids 95, 47–55 (2015).

270.      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).

271.      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).

272.      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).

273.      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).

274.      Feng, R. et al. Oxidation of fish oil exacerbates alcoholic liver disease by enhancing intestinal dysbiosis in mice. Commun. Biol. 3, 481 (2020).

275.      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).

276.      Snelson, M. & Coughlan, M. T. Dietary Advanced Glycation End Products: Digestion, Metabolism and Modulation of Gut Microbial Ecology. Nutrients 11, (2019).

277.      Birlouez-Aragon, I. et al. A diet based on high-heat-treated foods promotes risk factors for diabetes mellitus and cardiovascular diseases. Am. J. Clin. Nutr. 91, 1220–6 (2010).

278.      Gurzell, E. A. et al. Is the omega-3 index a valid marker of intestinal membrane phospholipid EPA+DHA content? Prostaglandins. Leukot. Essent. Fatty Acids 91, 87–96 (2014).

 

No comments:

Post a Comment