Next Article in Journal
Perinatal Gram-Positive Bacteria Exposure Elicits Distinct Cytokine Responses In Vitro
Next Article in Special Issue
Regulation of the CoA Biosynthetic Complex Assembly in Mammalian Cells
Previous Article in Journal
Deletion of KLF10 Leads to Stress-Induced Liver Fibrosis upon High Sucrose Feeding
Previous Article in Special Issue
Exploring Yeast as a Study Model of Pantothenate Kinase-Associated Neurodegeneration and for the Identification of Therapeutic Compounds
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Monounsaturated Fatty Acids in Obesity-Related Inflammation

CERMO-FC Research Center, Molecular Metabolism of Lipids Laboratory, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC H3C 3P8, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(1), 330; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010330
Submission received: 3 December 2020 / Revised: 23 December 2020 / Accepted: 25 December 2020 / Published: 30 December 2020
(This article belongs to the Special Issue CoA in Health and Disease)

Abstract

:
Obesity is an important aspect of the metabolic syndrome and is often associated with chronic inflammation. In this context, inflammation of organs participating in energy homeostasis (such as liver, adipose tissue, muscle and pancreas) leads to the recruitment and activation of macrophages, which secrete pro-inflammatory cytokines. Interleukin-1β secretion, sustained C-reactive protein plasma levels and activation of the NLRP3 inflammasome characterize this inflammation. The Stearoyl-CoA desaturase-1 (SCD1) enzyme is a central regulator of lipid metabolism and fat storage. This enzyme catalyzes the generation of monounsaturated fatty acids (MUFAs)—major components of triglycerides stored in lipid droplets—from saturated fatty acid (SFA) substrates. In this review, we describe the molecular effects of specific classes of fatty acids (saturated and unsaturated) to better understand the impact of different diets (Western versus Mediterranean) on inflammation in a metabolic context. Given the beneficial effects of a MUFA-rich Mediterranean diet, we also present the most recent data on the role of SCD1 activity in the modulation of SFA-induced chronic inflammation.

1. Inflammation in the Metabolic Syndrome

Obesity is the main factor responsible for the development of the metabolic syndrome, which is characterized by metabolic complications including visceral adiposity, hypertension, high circulating cholesterol and elevated glycemia [1,2,3]. This pathological combination often leads to insulin resistance and type 2 diabetes and is associated with a sustained inflammation profile [4,5]. In North America, people with a body mass index (BMI) superior to 30 are considered obese. This represents approximatively 36% of the population of North America and 13% worldwide [6].
Obesity is characterized by an excessive accumulation of lipids in adipose tissue. This accumulation becomes deleterious when it occurs in visceral fat [7]. In fact, waist circumference (as an indirect measure of visceral fat accumulation) is correlated with the development of specific metabolic disorders including cardiovascular diseases, hypercholesterolemia and type 2 diabetes [8]. When excessive lipid accumulation in adipose tissues occurs, ectopic accumulation (steatosis) appears in other tissues such as liver and muscle [8,9,10]. Saturated adipocytes release free fatty acids into the blood through the action of the Fatty acid translocase (FAT/CD36), the plasmatic Fatty acid binding protein (FABPpm) and the Fatty acid transport proteins (FATPs). These circulating free fatty acids are then captured by other organs, especially the liver and muscle, which gives rise to steatosis [11,12]. Accumulation of long chain fatty acids in non-adipose cells leads to the formation of toxic lipids such as ceramides and cholesterol esters [13]. These lipids induce lipotoxicity, leading to deleterious metabolic consequences including endoplasmic reticulum (ER) stress and inflammation [14,15].
Several population studies reveal that a low-grade and chronic inflammation is often developed in obese patients [16]. This is characterized by increased circulating levels of pro-inflammatory cytokines—especially Interleukin-6 (IL-6)—and of the chemokine MCP-1, both produced by the adipose tissue. Consequently, monocytes are recruited to the adipose tissue, inducing the secretion of other cytokines such as IL-1β and amplifying the inflammatory state [17,18]. In response to elevated cytokines levels, the liver secretes C-reactive protein (CRP), a key marker of inflammation associated with several metabolic diseases including type 2 diabetes and cardiovascular diseases [19,20,21,22]. CRP also aggravates disease development by activating the NF-κB signaling pathway, which is directly implicated in the expression of pro-inflammatory cytokines [23].

2. The Molecular Mechanisms of Inflammation

There are two main types of inflammation: acute and chronic. Acute inflammation appears in response to infections or injuries. This type of inflammation involves polynucleolar neutrophils and is characterized by apparition of swelling and heat around the damaged tissues. Activation of Toll-like receptors (TLRs) triggers the expression of inflammation effectors such as cytokines, prostaglandins, platelet activation factors, inflammasomes complexes, CRP, as well as NF-κB [24]. The resolution of this inflammation requires several conditions: destruction of the cause of inflammation, neutralization of pro-inflammatory markers (cytokines and prostaglandins) and clearance of neutrophils. These events typically occur in a few days, making this type of inflammation transient by nature [25].
The second type of inflammation, chronic inflammation, is sustained over time and is more deleterious for health. It often appears in individuals with poor feeding habits and a sedentary lifestyle, features strongly correlated with obesity development [26,27]. It is also present in different pathologies such as Alzheimer disease and asthma, and in several diseases associated with unbalanced metabolism such as atherosclerosis, cardiovascular diseases and type 2 diabetes [28,29,30,31]. Often named microinflammation or metabolic inflammation, it entails a complex mechanism involving crosstalk between various tissues (such as liver and adipose tissue) across the entire body. In general, this low-grade inflammation appears when cellular stress is recognized by the immune system [32]. Consequently, monocytes are recruited and infiltrate the tissues, becoming macrophages [24].
In inflammatory conditions such as obesity, two distinct macrophage subpopulations can be found in the affected organs. These are associated with different functions. The so-called M1 macrophages display an extreme pro-inflammatory state. They express high levels of pro-inflammatory receptors such as TLRs, Tumor necrosis factor receptors (TNFRs) and Interleukin-1 receptor (IL-1R), and exhibit a powerful activation of the NF-κB transcription factor necessary for the expression of pro-inflammatory cytokines. Conversely, the Μ2 macrophages are anti-inflammatory and are characterized by a higher expression of the Interleukin-4 receptor (IL-4R) whose activation downregulates inflammatory mediators such as TNF-α and IL-6. They also display an activation of the transcription factors PPARγ and PPARδ, which leads to higher expression of anti-inflammatory cytokines such as IL-10 [33]. The inflammation level present in tissues is therefore dependent on the balance between infiltrated M1 and M2 macrophages. This balance can be modulated by diet and hormonal status and is regulated by the PPARγ transcription factor [34].
A number of potential inflammation triggers have been identified in the context of chronic inflammation. TLR4 is activated by circulating long chain saturated fatty acids [35]. Consequently, the IKK-IκB signaling cascade leads to NF-κB nuclear translocation, where it activates the transcription of several pro-inflammatory cytokines and interleukins [36]. High circulating levels of pro-inflammatory cytokines such as TNF-α, MCP-1, TGF-β and IFN-γ, as well as of interleukins IL-6, IL-1β, IL-18, and IL-8, are observed in patients presenting an inflammatory state [37]. TLR4 activation is also linked to the increased expression of several proteins involved in the formation of inflammasomes, multiprotein complexes responsible for the activation of inflammatory responses. In particular for NLRP3 (NOD-like receptor family, pyrine domain containing 3), an inflammasome complex involved in several diseases associated with chronic and low-grade inflammation [38,39].
NLRP3 is considered an intracellular receptor responsible for the activation of inflammatory responses. Several factors can activate NLRP3 including elevated concentration of intracellular ATP, reactive oxygen species (ROS), mitochondrial oxidized DNA, and lysosomal destabilisation [40]. It can also be activated by low intracellular potassium or high calcium concentrations, which arise in response to cellular stress [40]. As NLRP3 is activated, the caspase 1 subunit of the NLRP3 complex cleaves pro-interleukins into mature IL-1β and IL-18, key circulating markers of low-grade inflammation [41]. NLRP3 is considered as a key factor responsible for the induction and progression of chronic inflammation. In fact, disruption of NLRP3 in adipose tissues decreases the concentration of pro-inflammatory cytokines and restores insulin sensitivity in obese mice [42].
Another mechanism involved in the development of chronic inflammation involves excessive storage of triglyceride (TG) lipids within adipose tissues. Sedentary lifestyles and poor eating habits aggravate this unbalanced TG storage. In mice, excessive TG storage in white adipose tissue (WAT) induces secretion of pro-inflammatory adipokines such as IL-1β, TNF-α, MCP-1, and IL-6, triggering systemic metabolic inflammation [43]. In addition, excessive TG storage feeds lipolysis and increases the amount of intracellular and circulating free fatty acids (FFAs) (Figure 1). These fatty acids act as stress-inducing molecules which, captured by TLR4, induce activation of NF-κB and, in turn, induce NLRP3 expression in macrophages (Figure 1). In addition, intracellular FFAs can impair mitochondria and lysosome integrity, generating ROS (Figure 1) [44]. FFAs can also inactivate the serine-threonine kinase AMPK, an intracellular energy sensor. In this situation, secretion of IL-1β (via activation of the NLRP3 inflammasome) is increased and leads to lower insulin sensitivity [45]. Several authors even suggest that activation of AMPK can be considered as an anti-inflammatory marker in the context of metabolic inflammation [46,47].

3. Overview of Lipid Metabolism

Fatty acid molecules are structurally very diverse and, accordingly, are involved in several different biological functions. For example, phospholipids are an integral part of cell membranes while TGs are mainly involved in energy storage. There are two sources of lipids in the organism: dietary intake and de novo synthesis.
In humans, dietary lipids such as cholesterol, TGs, as well as long-chain saturated and unsaturated fatty acids are absorbed in the form of micelles by the intestinal enterocytes. Meanwhile, short and medium-chain fatty acids (2 to 10 carbon chain length) can directly cross enterocyte membranes and reach the bloodstream [48,49]. Enterocytes secrete lipids into the lymphatic and blood circulation in the form of chylomicrons. The liver then captures part of the chylomicrons, using the extracted lipids to assemble very low-density lipoproteins (VLDLs) containing Apolipoprotein B-100 (apoB-100). Secreted, circulating VLDLs transfer their lipids to the rest of the organism, becoming low-density lipoproteins (LDLs) in the process. In parallel to this system, enterocytes and hepatocytes secrete Apolipoprotein A-I (apoA-I) which, in complex with the uncaptured chylomicrons, forms high-density lipoproteins (HDLs) [50]. The main know function of HDLs is to sequester the cholesterol coming from peripheral organs and bring it to the liver [51].
Several mechanisms allow the intake of lipids into cells. Cholesterol is captured via the transmembrane Scavenger Receptor class B type I (SRB1) [52], while TG integrated into lipoproteins are hydrolyzed by the Lipoprotein lipase at the surface of epithelial cells. The FFAs generated are then absorbed by cells through different transporters such as the Fatty acid transport proteins (FATPs) and the Fatty acid translocase (FAT/CD36). The internalized FFAs are rapidly esterified into fatty acid-CoA, which can be then transformed back into TG. This esterification process involves various fatty acyltransferases such as GPAT (Glycerol-3-phosphate acyltransferase) and DGAT (Diacylglycerol O-acyltransferase). Newly formed TG are subsequently integrated into intracellular lipid droplets (LDs) where they are stored [53]. LDs are present in all eukaryotic cells. In normal conditions, lipids are preferentially stored into adipocytes, forming very large LDs. Under conditions where adipocytes are saturated (like obesity), lipids can be stored in other cells such as hepatocytes and myocytes, forming much smaller LDs [54]. This ectopic storage often leads to metabolic disorders and their associated inflammation.
The other source of lipids in the organism comes from de novo lipid synthesis, also termed lipogenesis. This process occurs in most cells but, in humans, it principally occurs in hepatocytes (Figure 2) and adipocytes [55]. Lipogenesis synthesizes long-chain saturated fatty acids (palmitate) from acetyl-CoA generated by glucose hydrolysis. This synthesis is catalysed by the combined actions of Acetyl-CoA carboxylase (ACC) and Fatty acid synthase (FAS). Subsequently, saturated fatty acids (SFAs) are elongated by Fatty acid elongases (ELOVLs) [56] and/or desaturated by Stearoyl-CoA desaturases (SCDs), forming monounsaturated fatty acids (MUFAs) [57].
SCDs are the rate-limiting enzymes of MUFA formation. They are integrated into the ER membrane and are highly regulated by nutritional status and by hormonal regulators of appetite such as insulin [58,59]. SCDs introduce a delta-9 desaturation in SFAs stearate (C18:0) and palmitate (C16:0), forming the MUFAs oleate (C18:1n-9) and palmitoleate (C16:1n-7), respectively. These MUFAs are the main components of TGs (fatty acids that are preferentially stored) [60], cholesterol esters (cellular membrane components, precursors to steroid hormones and biliary acids) [61] and wax esters (compounds preventing evaporative water loss) [62]. They also constitute a large proportion of phospholipids comprising cellular membranes [57]. As such, SCDs are considered key regulators of lipid homeostasis, especially in liver and adipose tissue where lipogenesis is predominant. Modulation of SCD activity has been implicated in the development of the metabolic syndrome and its associated inflammatory state. Therefore, several studies have suggested targeting SCDs in order to treat various aspects of the metabolic syndrome, including type 2 diabetes and cardiovascular diseases [63,64,65].
In humans, there are two SCD isoforms, SCD1 and SCD5. SCD5 is mainly expressed in the brain, while SCD1 is more ubiquitously expressed [66,67]. In mice, the situation is more complex as four isoforms have been characterized (SCD1-4). They all share 85% amino acids homology with human SCD1, while SCD5 appears to be specific to primates. Mouse SCD1 is mainly expressed in lipogenic organs such as liver and adipose tissues. SCD2 is chiefly expressed in the brain, while SCD3 is found in the harderian, preputial and sebaceous glands. SCD4 expression has only be reported in the heart [68,69,70,71,72].

4. Stearoyl-CoA Desaturase-1

SCD1 is the most characterized SCD isoform. SCD1 transforms 85% of stearate and 51% of palmitate (from both dietary and lipogenesis origin) into MUFA [68]. Many studies have been performed in SCD1 knockout mice to better understand its role in metabolic processes. Global SCD1 knockout mice, in which every cell of the organism is SCD1 deficient, present with lack of sebum secretion and of lacrimal surfactant [73]. The lack of sebum gives rise to very dry skin with less hair and has led to the consideration of topical SCD1 inhibition as a potential treatment for acne.
Global SCD1 knockout mice are protected against obesity [74], insulin resistance [75] and fatty liver disease [61], as induced by both high-carbohydrate diet (HCD) [76] and high-fat diet (HFD) [74,75]. These mice display increased levels of plasma ketone bodies while the levels of circulating insulin and leptin are reduced [75]. Glycemia is also improved, as determined by a glucose tolerance test. The metabolic profiles of global knockout mice are more beneficial than their wildtype counterpart, as seen through the upregulation of lipid oxidation and the downregulation of lipid synthesis genes [74,76]. Because of the difference of global knockout mice, mice with a specific deletion of SCD1 in the liver are only protected from the deleterious effects of HCD (and not HFD). Under HCD, liver-specific knockout mice show a reduction of hepatic lipogenic enzyme gene expression as well as a reduction of plasmatic TG relative to controls [76]. As could be expected, these mice display a decrease of hepatic steatosis and associated metabolic complications such as hypercholesterolemia. This is consistent with diminished activation of SREBP-1 (as measured by protein maturation and nuclear localization levels) and with increased protein expression of the lipolysis transcription factor PPARα and the mitochondrial uptake acyl transporter Carnitine O-palmitoyl transferase 1 (CPT1) in the liver of global SCD1-deficient mice [77]. However, under HFD, liver-specific knockout mice develop hepatic steatosis and insulin resistance [78]. The steatotic effect of HFD on liver-specific knockout mice is probably due to the presence of SFA in the diet, which can be desaturated and integrated into TG, and, subsequently, into chylomicrons by enterocytes that still express SCD1. The chylomicrons can then be captured by the liver leading to hepatic steatosis and associated hepatic dysfunctions [76,79].
SCD1 expression is chiefly controlled by the lipogenic transcription factor SREBP-1c [77,80]. Under post-prandial conditions, the rise of lipemia and glycemia induce insulin secretion, one of the most important lipid anabolic hormones. Insulin activates the PI3K-PKB-mTORC1 signaling pathway, which induces the nuclear translocation of SREBP-1c and activates expression of enzymes involved in lipogenesis, including SCD1 [81]. There are others lipogenic transcription factors activated by dietary and hormonal factors such as insulin and glucose. Expression of lipogenic genes such as SCD1, FAS and ELOVL6 is triggered by the Liver X receptor (LXR), which is activated by insulin and by the Carbohydrate response element binding protein (ChREBP), itself activated by glucose [82]. One of the main LXR targets in lipid metabolism (especially of the LXRα isoform) is SREBP-1c, driving the expression of SCD1 [83]. Furthermore, MUFA (products of SCD activity) can regulate lipogenesis through AMPK phosphorylation [84,85]. Phosphorylated AMPK inhibits the mTORC1 complex [86], reducing the nuclear translocation of SREBP-1c and the expression of lipogenic genes like SCD1.

5. Role of Saturated Fatty Acids in Inflammation

5.1. Human Studies—Effect of Dietary SFAs

The type of lipids present in animal organisms is strongly influenced by diet [87]. Dietary SFAs are deleterious to metabolic health as they play an important role in the development of obesity, metabolic syndrome and chronic inflammation [88]. In fact, high SFA levels in the diet can be considered a pro-inflammatory factor in itself. Several studies have described clear correlations between the consumption of Western diets, rich in SFA, and the presence of obesity, hepatic steatosis and type 2 diabetes in humans [89,90,91]. Acute intake of SFA-rich diets triggers the development of an inflammatory profile in human subcutaneous adipose tissues, which includes increased expression of several genes involved in the synthesis of pro-inflammatory chemokines and cytokines [92]. In addition, compared to unsaturated fatty acid-rich diets, SFA-rich diets increase lipid storage within adipose tissues [90]. The adipocytes develop larger LDs and, therefore, contain more TGs. This increased intracellular TG pool leads to increased leptin secretion by adipocytes [93]. Furthermore, high circulating leptin is correlated with increased macrophage secretion of IL-1β, IL-6 and TNF-α [94,95]. A clinical trial has shown that a single 1000 kcal meal containing 60% fat (mainly SFA) leads to elevated plasmatic IL-6 concentrations [96]. This type of systemic inflammation is associated with vascular damages leading to coronary heart disease [96].

5.2. Animal Studies—Effect of Dietary SFAs

In agreement with observations made in humans, feeding rodents with diets rich in saturated fat increases hepatic and plasmatic TG levels and raises circulating IL-6 concentration [97,98]. Animals also develop glucose intolerance while macrophage recruitment in the liver is increased [97,99]. This suggests that inflammation is a consequence of diet-induced metabolic changes. Indeed, mice fed during 15 weeks with a HFD containing a majority of SFAs display an increased expression of hepatic TLR4 [98]. These animals also show elevated plasmatic concentration of IL-6, TNF-α and MCP-1, and lowered plasmatic concentration of the anti-inflammatory cytokine IL-10 [98].
Mice under SFA-rich HFD develop muscle steatosis due to accumulation of palmitate and stearate [100]. SFAs can also induce inflammation in the central nervous system. Brains of mice fed during 8 weeks with a HFD (composed mainly of SFA) display high concentrations of inflammatory markers (IL-6, IL-1β and TNF-α) and low levels of IL-10 [101]. Mice on a SFA-rich diet for as little as 4 weeks show elevated activation of NF-κB and, through TLR4 activation in the hypothalamus, expression of inflammatory markers (IL-1β, TNF-α and IFN-γ) in the brain as well as in the plasma [102,103]. This inflammation can even contribute to the development of obesity, at least in mice. Sustained HFD-induced inflammation in the arcuate nucleus, a specific region of the hypothalamus that regulates energy homeostasis, triggers microglia recruitment and fosters the death of satiety neurons [104].

5.3. Cellular Models—Effect of Exogenous SFAs

In vivo studies are realized with diets containing a mix of several types of fatty acids, which are at least partially transformed during the digestion process. This complicates interpretation of the results of these studies. Therefore, treatment of cultured cells with exogenous fatty acids has been used to determine the effect of specific SFAs expected to be found in post-prandial circulation.
Adipocyte cell models can provide insight into the in vivo mechanisms taking place within adipose tissue. Incubation of 3T3-L1 preadipocytes and rat primary epididymal adipocytes with palmitate for 24 h induces TNF-α and IL-6 secretion [105]. This treatment also increases the release of Monocyte chemoattractant protein-1 (MCP-1) [106,107], which has the potential to induce the recruitment of macrophages in vivo as well as their polarization into a M1 pro-inflammatory state. Exposition of pancreatic β cells (1.1B4 human cell line and rat primary cells) to palmitate increases secretion of IL-6 and IL-8, as well as ROS production. It is also associated with impaired insulin secretion [108,109]. This process has the potential to explain, at least in part, why saturated fat-rich diets lead to the development of type 2 diabetes.
In mouse microglia BV2 cells, palmitate treatment during 4 h induces IL-1β, IL-6 and TLR4 gene expression, as well as NF-κB induction [103]. In the RAW 264.7 mouse macrophage cell line, lauric acid (a 12-carbon chain SFA) can directly bind TLR4 and activate the nuclear translocation of NF-κB. This subsequently activates the expression of pro-inflammatory cytokines, especially TNF-α [110,111]. Treatment of RAW 264.7 cells with palmitate inhibits the expression of the transcription factor PGC-1β, which indirectly activates the nuclear translocation of NF-κB [112]. This leads to increased secretion of inflammatory cytokines TNF-α and IL-1β in the medium. Interestingly, when this medium is added to cultured 3T3-L1 preadipocytes, activation of the PI3K-PKB pathway is impaired, suggesting a decrease in insulin sensitivity [113].
The effect of SFAs on muscle cells has also been studied in vitro. Treatment of C2C12 mouse myotube cells with palmitate increases lipid storage as observed via lipid droplet size [114]. As for other cell types, this intracellular lipid accumulation causes lipotoxicity (elevated ROS and ER stress) and insulin resistance (disruption in PKP signaling). It also triggers NF-κB nuclear translocation, leading to the expression of pro-inflammatory cytokines such as TNF-α [114].

6. Role of Monounsaturated Fatty Acids in Inflammation

6.1. Human Studies—Effect of Dietary MUFAs

While SFAs increase inflammation, unsaturated fatty acids often have the opposite effect. Polyunsaturated fatty acids (PUFAs), especially the omega-3 class, have favorable effects on health. Several population studies have indeed demonstrated that, compared to SFA-rich Western diets, diets rich in omega-3 PUFAs exert beneficial metabolic effects at least in part by decreasing inflammation [115,116,117]. The effects of MUFAs on inflammation are less documented, but more and more evidences link MUFAs to anti-inflammatory states [92].
Dietary lipids are assimilated in the gut and then transported throughout the entire organism where they influence organ metabolism. Higher MUFA consumption increases MUFA levels, and reduces both SFA and PUFA, throughout the body [118]. The type of lipids present in our body can therefore be modulated through nutrition.
The impact of the Mediterranean diet has been studied in humans, including in several randomized crossover studies (Table 1) [119,120,121]. This diet is characterized by a high consumption of fish, olive oil, fruits and vegetables, and whole grains. In this type of diet, fat constitutes one third of the total kcal absorbed with almost 60% MUFA and 20% SFA [122]. For comparison, the Western diet has a similar amount of total fat but with a much lower proportion of MUFA (36% MUFA and 33% SFA) [119]. Compared to other diets, the Mediterranean diet is associated with lower blood pressure, as well as improved glucose and lipid blood profiles [123,124,125]. The Mediterranean diet lowers cardiovascular disease risk and even leads to beneficial gut microbiome changes: increasing bacteroides, prevotella and faecalibacterium genera, which are known to improve general metabolic health and prevent atherosclerosis and thrombosis (Table 1) [121,126]. In fact, olive oil, one of the main components of the Mediterranean diet, has been characterized as a prebiotic improving the host-microbial ecosystem (Table 1) [120].
Interestingly, supplementation of food with olive oil (an oil that is naturally enriched with the SCD1 product oleate) correlates with low occurrences of obesity and metabolic syndrome, and therefore, less chronic inflammation and mortality [127,128]. Furthermore, people consuming a Mediterranean diet generally show lower levels of the systemic inflammation profile that often appears when Western or carbohydrate-rich diets are consumed (Table 1) [129,130,131,132]. Consumption of Mediterranean diet for 3 to 4 weeks is also correlated with increased secretion of adiponectin, an adipokine with anti-inflammatory effects [94,133]. Similar observations on inflammation are made when subjects are fed with olive oil (Table 1) [131,134,135]. Subjects fed with a diet rich in olive oil for a period ranging from 3 weeks to 2 years display lower levels of circulating mononuclear cells (monocytic cells involved in the inflammatory response). In addition, their plasmatic pro-inflammatory cytokine levels (such as TNF-α, MCP-1, IFN-γ, CRP, IL-18, and IL-6) are lower when compared to subjects on a Western diet for the same period of time [131,136,137,138]. Compared to a one-time oral dose of a fat emulsion containing cow’s milk cream (25% oleate and 26% palmitate), an emulsion of olive oil (70% oleate and 15% palmitate) generates a more favorable lipid plasmatic profile, including a higher plasmatic concentration of MUFA-rich TG. Interestingly, in the same study, the authors incubated mouse BV2 microglia cells with purified plasmatic lipoproteins from these subjects. Upon treatment, the incubated cells switched from a M1 pro-inflammatory state to a M2 anti-inflammatory state in the presence of MUFA-rich TG (Table 1) [139]. This observation has been confirmed in another study on isolated human blood monocytes [140].
Anti-inflammatory effects of MUFA have been reported when MUFA are part of a dietary intervention. However, increased MUFA levels in vivo do not always have positive impacts on inflammation. In patients with chronic kidney disease, an elevated MUFA/SFA ratio in blood lipids—presumably reflecting increased SCD1 activity—is correlated with high levels of circulating CRP, suggesting an aggravation of inflammation [141]. In obese patients that underwent bariatric surgery, the concentration of lipids in SAT (subcutaneous adipose tissue) and VAT (visceral adipose tissue) was measured by gas chromatography. The MUFA proportion in these SAT and VAT samples is negatively correlated with inflammation and obesity-related conditions such as insulin resistance and type 2 diabetes, as measured by gene expression [91].
Though the beneficial effects of the Mediterranean diet cannot simply be attributed to its high olive oil content, as it also contains many omega-3 fatty acids, these population studies strongly suggest that dietary MUFA have anti-inflammatory effects, especially compared to SFA-rich diets such as the Western diet.

6.2. Animal Studies—Effect of Dietary MUFAs

To further investigate the effects of MUFA in the diet, several studies have been performed on HFD-fed mice. These animals allow for measurements of metabolic and inflammatory markers throughout the organism, rather than only in the blood or on surgical samples. Mice raised for 4 weeks on a diet rich in olive oil show elevated plasmatic concentration of MUFA with no change in hepatic SCD1 gene expression [142]. As in humans then, MUFA-rich diets can be used to study the effects of MUFA on systemic responses in rodents.
Studies performed in mice raised on a MUFA-rich diet for 15 weeks show higher circulating levels of anti-inflammatory markers (IL-4 and IL-10) and lower levels of pro-inflammatory markers (IL-6, MCP-1, IL-1β and TNF-α) compared to mice fed with a SFA-rich diet [98]. Even in obese and hypercholesterolemic mouse models, a MUFA-rich 8-week-long diet improves metabolic features, increasing the expression of anti-inflammatory markers such as IL-4, IL-10 and PPARγ. In addition, a decrease in circulating level of pro-inflammatory IL-6, MCP-1, TNF-α, and IL-1β, and a larger proportion of M2 macrophages (compared to M1) is observed in adipose tissues [143]. In a study performed in male Wistar rats, animals were raised 12 weeks on HFDs (35% kcal from fat) with different SFA/MUFA/PUFA ratios [144]. Compared to a diet containing a higher proportion of SFA, increasing the proportion of MUFA improves insulin sensitivity and induces expression of the anti-inflammatory cytokine adiponectin, especially in subcutaneous adipose tissue. However, MUFAs are less effective than PUFAs in inducing the expression of the adiponectin gene. Higher MUFA or PUFA proportions are also correlated with lower circulating LDL-cholesterol levels [145]. The effects of fatty acids on inflammation were studied in mice fed for 15 weeks with isocaloric diets rich in either SFA or MUFA [98]. Compared to the SFA group, liver analysis of mice fed with MUFA shows less macrophage infiltration as well as a decrease in TG content and lipid peroxidation (measured via thiobarbituric acid reactive substances). The plasmatic lipid profile is improved, as well as insulin sensitivity (as measured by HOMA-IR). The levels of circulating pro-inflammatory CRP and MCP-1 are also decreased [98]. Interestingly, a very recent study has shown that switching mice from a SFA-based HFD to a MUFA-based HFD partially attenuates the progression of hyperglycemia, diminishing pancreatic inflammation and ameliorating β cell function [146]. Macrophage infiltration in the pancreas was lower in MUFA-HFD fed mice. The authors suggest that this effect is mediated by AMPK [147]. Interestingly, when compared to a diet rich in n-6 PUFA, an olive oil-rich 24-month-long diet protects cardiac mitochondria from age-related damages in rats [148]. A very promising study has recently shown that, compared to SFAs, dietary MUFAs reduce the pro-inflammatory profile in the mouse brain (and in human blood), stimulating M2 macrophage polarization. The authors even propose to use olive oil in nutraceutical strategies to treat diseases associated with a neuro-inflammatory profile [139].

6.3. Cellular Models—Effect of Exogenous MUFAs

Oleate protects HepG2 cells (a human hepato-carcinoma cell line) against SFA-induced lipotoxicity, decreasing ER stress, ROS production, and activation of inflammation markers (NLRP3, IL-6, MCP-1 and IL-1β) [149]. In primary murine hepatocytes, intracellular LD-derived MUFAs bind to SIRT1 (NAD-dependent protein deacylase sirtuin-1/silent information regulator 1), resulting in the activation of PPARα via PGC-1α. Oleate is also a direct PPARα agonist [150]. These mechanisms inhibit NF-κB activity (Figure 3) [151,152], explaining, at least in part, the resorption of hepatic inflammation by MUFAs.
In the 3T3-L1 murine preadipocyte cell line, oleate treatment increases expression of the adiponectin gene [153], probably through PPARγ activation [154,155]. Adiponectin induces IL-10 secretion and inhibits IL-6 and TNF-α secretion [153], which has the potential to reduce local inflammation in vivo. Adiponectin can also reduce peripheral inflammation by enhancing M2 macrophage polarization (Figure 3) [154,155,156,157,158,159,160,161].
Bone marrow-derived macrophages prepared from HFD-fed mice present a pro-inflammatory profile including macrophage M1 polarization and elevated secretion of IL-6 and TNF-α (Figure 3) [162]. The treatment of these macrophages with palmitoleate can switch the polarization of macrophages to M2 (Figure 3) [162]. Palmitoleate also activates AMPK, leading to a decrease of NF-κB nuclear translocation (Figure 3). This increases the expression of several anti-inflammatory factors such as MGL2, IL-10, TGFβ1, and MRC1 [162,163]. Incubation of mouse adipose stromal vascular fraction and bone marrow primary cultures with oleate inhibits LPS-induced IL-1β secretion [45,164]. In this situation, AMPK is activated, which in turn inhibits NLRP3 activation (responsible for IL-1β maturation) (Figure 3) [45,164]. Similar observations were reported on primary rat pancreatic islet cells [165].
MUFA also display protective effects in several other cell lines. For instance, oleate protects mouse muscle C2C12 cells from palmitate-induced insulin resistance and ER stress [166]. In mouse podocyte cells, derived from kidney epithelium, SFAs activate the cell death pathways associated with ER stress. This effect is reversed by oleate [167]. In the human endothelial EAHy926 cell line, palmitoleate decreases pro-inflammatory IL-6, IL-8 and MCP-1 secretion, and downregulates NF-κB (via PPARγ stimulation), as compared to palmitate [168].

7. The Role of Stearoyl-CoA Desaturase-1 in Inflammation

7.1. Human Correlation Studies

Given that SCD1 is the major enzyme involved in MUFA synthesis, several authors have hypothesized that an increase in expression and/or activity of SCD1 could be correlated with an improvement of patient inflammatory profile.
In a study performed on young adults [169], a clear correlation was observed between the rs2060792 (A/G) single nucleotide polymorphism (SNP) upstream of the SCD1 gene and the level of circulating SFAs palmitate and stearate. European women bearing the major allele present with higher palmitate and lower stearate concentrations. Interestingly, this SNP was positively associated with obesity and a higher level of the circulating pro-inflammatory factor CRP, especially in women. In a study analysing surgical samples from human visceral adipose tissue of obese individuals, an enrichment of histone methylation (H3K4me3) in the SCD1 and IL-6 promoters was correlated with increased BMI. This histone methylation enrichment pattern was associated with lower SCD1 expression and higher pro-inflammatory TNF-α and IL-6 expression [170].
However, in overweight adults, high palmitoleate plasma concentrations, reflecting high SCD1 activity, is correlated with the occurrence of inflammatory fatty liver disease [171]. This increased SCD1 activity could be due to a compensatory mechanism triggered by high circulating concentrations of its substrate palmitate [20,172].
The results obtained in these human studies have not always shown a strict correlation between SCD1 activity and inflammation. This suggests that the level of endogenous synthesis is not the only factor behind the modulation of inflammatory state by MUFA.

7.2. Animal Genetic Models

Both human and animal dietary studies clearly argue for a beneficial role of MUFA on the inflammatory status. Given that MUFAs are a product of SCD1 activity, the deletion of this enzyme should reduce the availability of MUFA (and increase SFA accumulation), leading to increased inflammation.
SCD1-deficient mice are a useful tool to study the effect of endogenous MUFA synthesis on lipid metabolism and inflammation processes. The asebia mouse model is deficient for SCD1 due to a naturally occurring genomic deletion. As in the SCD1 knockout mice, asebia animals display eye inflammation, a lack of sebaceous glands, and an absence of hair within scarred dermis [173,174]. In skin-specific SCD1 knockout mice, expression of pro-inflammatory genes IL-6, TNF-α and IL-1β is increased around hair follicles [175,176]. By inducing follicle cell death, this inflammation contributes to hair loss [177].
Like SCD1 knockout mice, asebia mice are protected from HFD-induced obesity, hepatic steatosis and glucose intolerance [178,179,180]. However, compared to wildtype mice, they exhibit a complex inflammatory profile including circulating pro-inflammatory markers such as IL-6 and IL-1β [181]. Adipose tissue-specific SCD1 knockout mice are protected against Western diet-induced obesity and fatty liver disease [74]. Their WAT displays a lower concentration of MCP-1 and TNF-α compared to WAT from wildtype mice, even when they are raised on HFD (60% kcal fat, mainly lard).
Enterocyte-specific SCD1 knockout mice display an increase in pro-inflammatory markers IL-6 and TLR4 within their colon and ileum [182]. Interestingly, these enterocyte-specific effects can be rescued by an oleate-rich diet [183]. Intriguingly, enterocyte-specific SCD1 knockout mice show diminished expression of the TLR4 receptor in the jejunum, suggesting a protection against inflammation [182].
Liver-specific SCD1 knockout mice display an increase in pro-inflammatory markers IL-1β and TNF-α within their liver [184]. These knockout mice models exhibit a reduction in the expression of lipogenic markers ACC, FAS and SREBP-1c. This potential for diminished palmitate synthesis could attenuate the inflammatory effects of SCD1 depletion.

7.3. Cellular Models

Several studies address the specific role of SCD1 in cellular models of inflammation. Silencing or inactivation of the SCD1 gene in the murine preadipocyte 3T3-L1 cell line exacerbates the effects of SFAs, increasing the expression of the pro-inflammatory markers TGF-β, IL-6 and MCP-1, and decreasing the anti-inflammatory IL-10 [185,186]. Similar results are observed in the EndoC-βH1 human pancreatic β cell line. Silencing SCD1 aggravates the lipotoxic effect of palmitate on inflammatory marker expression and, interestingly, oleate and palmitoleate treatments rescue these effects [187]. Incubating RAW 264.7 macrophages with a conditioned media obtained from primary adipocytes isolated from global SCD1 knockout mice decreases expression of both TNF-α and IL-1β pro-inflammatory cytokines [188]. SCD1 silencing in mouse primary macrophages renders the TLR4 receptor hypersensitive, which exacerbates the gene expression of pro-inflammatory cytokine (IL-1β, MCP-1 and IL-6) [189]. TLR4 hypersensitivity is thought to stem from increased SFA proportions within membrane phospholipids [189].
Other technical approaches allow for insight into the effect of SCD1 overexpression. In primary human myotube cells, overexpression of SCD1 prevents palmitate-induced ER stress and IL-8 gene expression [190]. Mesenchymal stromal cells (MSC) can be prepared from posterior iliac crest bone marrow extracted from patients [191]. When these MSC cells are treated with T0901317 (an LXR agonist), SCD1 and LXRα expression are increased. This treatment reduces palmitate-induced Caspase 3/7 activation and expression of pro-inflammatory IL-6 and IL-8. When MSC cells are incubated with the specific SCD1 inhibitor CAY 10566, the effect of the LXR agonist is abrogated. This suggests that, at least in bone marrow stromal cells from these patients, SCD1 is involved in the prevention of inflammation and apoptosis induced by palmitate [191].
More recently, a study has been performed using primary hepatic cell isolated from G-protein coupled receptor 120 (GPR120) deficient mice. This receptor interacts with MUFA, especially palmitoleate [192]. The activation of GPR120 by palmitoleate, is involved in the resolution of palmitate-induced inflammation through a reduction of NF-κB activity. Interestingly, in these cells, a correlation between SCD1 expression and GPR120 activity is observed [193].
Inhibiting SCD1 in cells leads to increased inflammation. This is probably due to a combination of lower intracellular MUFA concentration and, undoubtedly, higher intracellular SFA concentration.

8. Conclusions

As presented throughout this text, dietary fat intake has an undeniable impact on inflammation. There is evidence that chronic low-grade inflammation can be prevented by lifestyle interventions. The SFA-rich Western diet can induce chronic inflammation and increase the risk of developing obesity-related metabolic disorders such as cardiovascular diseases, type 2 diabetes, and hepatic steatosis. At the opposite, a Mediterranean diet especially rich in oleate is favorable to an anti-inflammatory state and is associated with a decreased risk of metabolic syndrome development. Indeed, both human and animal diet studies have shown that substitution of SFA by MUFA activates beneficial anti-inflammatory mechanisms (M2 macrophage polarization, adipocyte IL-10 secretion, inhibition of NLRP3 inflammasome) and reverses the deleterious effect of SFAs on adipose tissues, hepatic tissue and β cells. Many mechanisms presented here can account for the protective effects of dietary oleate and high levels of circulating MUFAs. The addition of MUFA in diets can therefore be a potential nutraceutical avenue to decrease chronic inflammation and, subsequently, to ameliorate the general metabolic profile. In accordance with the beneficial effects of dietary MUFAs, some studies have shown that inhibiting SCD1 aggravated the deleterious effects of SFAs. This is probably due to an increase of SFA levels (SCD1 substrates). Thus, SCD1 is an interesting therapeutic target to decrease intracellular SFA concentration in favour of MUFA. However, other studies have shown that SCD1 inhibition can have favourable outcomes. SCD1 deletion protects mice against the deleterious effects of SFA-rich HFD and even improves the metabolic profile of humans and animals. In this case, the protective effects of SCD1 deletion cannot be attributed to MUFA activity in the organism. In fact, we and others have shown that SCD1 deletion inhibits lipogenesis [74,76,77,79,182]. This can be attributed to inhibition of SREBP-1c oleylation, decreasing its transcriptional activity [77]. This aspect of SCD1 activity deserves to be further investigated to better understand its specific role in inflammation.

Author Contributions

G.R. and A.L. wrote the manuscript. K.-F.B. and C.M. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

G.R. was supported by the foundation Lueur d’espoir pour Ayden and A.L. by a NSREC grant.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACCAcetyl-CoA carboxylase
AGPATAcylglycerol-3-phosphate-O-acyltransferase
AMPKAMP-activated protein kinase
apoA-IApolipoprotein A-I
apoB-100Apolipoprotein B-100
ATPAdenosine Triphosphate
BMIBody mass index
ChREBPCarbohydrate-responsive element-bonding protein
CPT-1Carnitine palmitoyltrasnferase-1
CRPC-reactive protein
DGATDiglyceride acyltransferase
DNADesoxyribonucleic acid
ELOVLElongation of very long chain fatty acid
EREndoplasmic reticulum
FABPFatty acid binding protein
FASFatty acid synthase
FAT/CD36Fatty acid translocase/cluster of differentiation 36
FATPFatty acid transport protein
FFAFree fatty acid
GPATGlycerol-3-phosphate acyltransferase
GPR120G-protein-coupled receptor 120
HCDHigh carbohydrate diet
HDLHigh density lipoprotein
HFDHigh fat diet
hMSCHuman mesenchymal stromal cells
HOMA-IRHomeostatic model assessment of insulin resistance
IFN-γInterferon-gamma
IKK-IκBIκB kinase-inhibitor of nuclear factor kappaB
IL-1βInterleukin-1 beta
IL-10Interleukin-10
IL-18Interleukin-18
IL-1RInterleukin-1 receptor
IL-4RInterleukin-4 receptor
IL-6Interleukin-6
IL-8Interleukin-8
LDLipid droplet
LDLLow density lipoprotein
LPSLipopolysaccharide
LXRLiver X receptor
MCP-1Monocyte chemoattractant protein-1
MGL2Macrophage galactose N-acetyl-galactosamine specific lectin 2
MRC1Macrophage mannose receptor 1 precursor
mTORC1Mammalian target of rapamycin complex 1
MUFAMonounsaturated fatty acids
NF-κBNuclear factor-kappaB
NLRP3NOD-like receptor family, pyrin domain
PGC-1βPeroxysome proliferator-activated receptor 1-beta
PI3KPhosphoinisitide-3-kinase
PKBProtein kinase B
PPARαPeroxysome proliferator-activated receptor alpha
PPARδPeroxysome proliferator-activated receptor delta
PPARγPeroxysome proliferator-activated receptor gamma
PUFAPolyunsaturated fatty acid
ROSReactive oxygen species
SATSubcutaneous adipose tissue
SCDStearoyl-CoA desaturase
SFASaturated fatty acid
SNPSingle nucleotide polymorphism
SRB1Scavenger receptor class B type 1
SREBP-1Sterol regulatory element-binding protein-1
TGTriglycerides
TGF-βTransforming growth factor-beta
TLRToll-Like receptor
TNF-αTumor necrosis factor-alpha
TNFRTumor necrosis factor receptor
VATVisceral adipose tissue
VLDLVery low-density lipoprotein
WATWhite adipose tissue

References

  1. Goodpaster, B.H.; Krishnaswami, S.; Harris, T.B.; Katsiaras, A.; Kritchevsky, S.B.; Simonsick, E.M.; Nevitt, M.; Holvoet, P.; Newman, A.B. Obesity, regional body fat distribution, and the metabolic syndrome in older men and women. Arch. Intern. Med. 2005, 165, 777–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Sherling, D.H.; Perumareddi, P.; Hennekens, C.H. Metabolic Syndrome. J. Cardiovasc. Pharm. 2017, 22, 365–367. [Google Scholar] [CrossRef] [PubMed]
  3. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lee, B.C.; Lee, J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim. Biophys. Acta 2014, 1842, 446–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Grandl, G.; Wolfrum, C. Hemostasis, endothelial stress, inflammation, and the metabolic syndrome. Semin. Immunopathol. 2018, 40, 215–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Arroyo-Johnson, C.; Mincey, K.D. Obesity Epidemiology Worldwide. Gastroenterol. Clin. N. Am. 2016, 45, 571–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Antonopoulos, A.S.; Tousoulis, D. The molecular mechanisms of obesity paradox. Cardiovasc. Res. 2017, 113, 1074–1086. [Google Scholar] [CrossRef]
  8. Tchernof, A.; Despres, J.P. Pathophysiology of human visceral obesity: An update. Physiol. Rev. 2013, 93, 359–404. [Google Scholar] [CrossRef]
  9. Despres, J.P.; Lemieux, I. Abdominal obesity and metabolic syndrome. Nature 2006, 444, 881–887. [Google Scholar] [CrossRef]
  10. Engin, A.B. What Is Lipotoxicity? Adv. Exp. Med. Biol. 2017, 960, 197–220. [Google Scholar] [CrossRef]
  11. Bonen, A.; Luiken, J.J.; Arumugam, Y.; Glatz, J.F.; Tandon, N.N. Acute regulation of fatty acid uptake involves the cellular redistribution of fatty acid translocase. J. Biol. Chem. 2000, 275, 14501–14508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ason, B.; Castro-Perez, J.; Tep, S.; Stefanni, A.; Tadin-Strapps, M.; Roddy, T.; Hankemeier, T.; Hubbard, B.; Sachs, A.B.; Michael Flanagan, W.; et al. ApoB siRNA-induced liver steatosis is resistant to clearance by the loss of fatty acid transport protein 5 (Fatp5). Lipids 2011, 46, 991–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Unger, R.H. Lipotoxic diseases. Annu. Rev. Med. 2002, 53, 319–336. [Google Scholar] [CrossRef] [PubMed]
  14. Summers, S.A. Ceramides in insulin resistance and lipotoxicity. Prog. Lipid Res. 2006, 45, 42–72. [Google Scholar] [CrossRef] [PubMed]
  15. Van Herpen, N.A.; Schrauwen-Hinderling, V.B. Lipid accumulation in non-adipose tissue and lipotoxicity. Physiol. Behav. 2008, 94, 231–241. [Google Scholar] [CrossRef]
  16. Saltiel, A.R.; Olefsky, J.M. Inflammatory mechanisms linking obesity and metabolic disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef]
  17. Xydakis, A.M.; Case, C.C.; Jones, P.H.; Hoogeveen, R.C.; Liu, M.Y.; Smith, E.O.; Nelson, K.W.; Ballantyne, C.M. Adiponectin, inflammation, and the expression of the metabolic syndrome in obese individuals: The impact of rapid weight loss through caloric restriction. J. Clin. Endocrinol. Metab. 2004, 89, 2697–2703. [Google Scholar] [CrossRef] [Green Version]
  18. Karczewski, J.; Sledzinska, E.; Baturo, A.; Jonczyk, I.; Maleszko, A.; Samborski, P.; Begier-Krasinska, B.; Dobrowolska, A. Obesity and inflammation. Eur. Cytokine Netw. 2018, 29, 83–94. [Google Scholar] [CrossRef]
  19. Weiss, R.; Dziura, J.; Burgert, T.S.; Tamborlane, W.V.; Taksali, S.E.; Yeckel, C.W.; Allen, K.; Lopes, M.; Savoye, M.; Morrison, J.; et al. Obesity and the metabolic syndrome in children and adolescents. N. Engl. J. Med. 2004, 350, 2362–2374. [Google Scholar] [CrossRef] [Green Version]
  20. Nieuwdorp, M.; Stroes, E.S.; Meijers, J.C.; Buller, H. Hypercoagulability in the metabolic syndrome. Curr. Opin. Pharm. 2005, 5, 155–159. [Google Scholar] [CrossRef]
  21. Dandona, P.; Aljada, A.; Bandyopadhyay, A. Inflammation: The link between insulin resistance, obesity and diabetes. Trends Immunol. 2004, 25, 4–7. [Google Scholar] [CrossRef] [PubMed]
  22. Dandona, P.; Aljada, A.; Chaudhuri, A.; Mohanty, P.; Garg, R. Metabolic syndrome: A comprehensive perspective based on interactions between obesity, diabetes, and inflammation. Circulation 2005, 111, 1448–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tang, Y.; Fung, E.; Xu, A.; Lan, H.Y. C-reactive protein and ageing. Clin. Exp. Pharmacol. Physiol. 2017, 44, 9–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Gabay, C.; Kushner, I. Acute-phase proteins and other systemic responses to inflammation. N. Engl. J. Med. 1999, 340, 448–454. [Google Scholar] [CrossRef] [PubMed]
  25. Serhan, C.N. Resolution phase of inflammation: Novel endogenous anti-inflammatory and proresolving lipid mediators and pathways. Annu. Rev. Immunol. 2007, 25, 101–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Rodier, F.; Campisi, J. Four faces of cellular senescence. J. Cell Biol. 2011, 192, 547–556. [Google Scholar] [CrossRef] [PubMed]
  27. Cevenini, E.; Monti, D.; Franceschi, C. Inflamm-ageing. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 14–20. [Google Scholar] [CrossRef]
  28. Weissmann, G. It’s complicated: Inflammation from Metchnikoff to Meryl Streep. Faseb J. 2010, 24, 4129–4132. [Google Scholar] [CrossRef]
  29. Murdoch, J.R.; Lloyd, C.M. Chronic inflammation and asthma. Mutat. Res. 2010, 690, 24–39. [Google Scholar] [CrossRef]
  30. Chambers, J.C.; Eda, S.; Bassett, P.; Karim, Y.; Thompson, S.G.; Gallimore, J.R.; Pepys, M.B.; Kooner, J.S. C-reactive protein, insulin resistance, central obesity, and coronary heart disease risk in Indian Asians from the United Kingdom compared with European whites. Circulation 2001, 104, 145–150. [Google Scholar] [CrossRef]
  31. Donath, M.Y.; Shoelson, S.E. Type 2 diabetes as an inflammatory disease. Nat. Rev. Immunol. 2011, 11, 98–107. [Google Scholar] [CrossRef] [PubMed]
  32. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  33. Murray, P.J. Macrophage Polarization. Annu. Rev. Physiol. 2017, 79, 541–566. [Google Scholar] [CrossRef] [PubMed]
  34. Chinetti-Gbaguidi, G.; Staels, B. Macrophage polarization in metabolic disorders: Functions and regulation. Curr. Opin. Lipidol. 2011, 22, 365–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Huang, S.; Rutkowsky, J.M.; Snodgrass, R.G.; Ono-Moore, K.D.; Schneider, D.A.; Newman, J.W.; Adams, S.H.; Hwang, D.H. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J. Lipid Res. 2012, 53, 2002–2013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sun, S.C. The non-canonical NF-kappaB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef]
  37. Kirwan, A.M.; Lenighan, Y.M.; O’Reilly, M.E.; McGillicuddy, F.C.; Roche, H.M. Nutritional modulation of metabolic inflammation. Biochem. Soc. Trans. 2017, 45, 979–985. [Google Scholar] [CrossRef]
  38. Ahechu, P.; Zozaya, G.; Marti, P.; Hernandez-Lizoain, J.L.; Baixauli, J.; Unamuno, X.; Fruhbeck, G.; Catalan, V. NLRP3 Inflammasome: A Possible Link Between Obesity-Associated Low-Grade Chronic Inflammation and Colorectal Cancer Development. Front. Immunol. 2018, 9, 2918. [Google Scholar] [CrossRef]
  39. Mastrocola, R.; Aragno, M.; Alloatti, G.; Collino, M.; Penna, C.; Pagliaro, P. Metaflammation: Tissue-Specific Alterations of the NLRP3 Inflammasome Platform in Metabolic Syndrome. Curr. Med. Chem. 2018, 25, 1294–1310. [Google Scholar] [CrossRef]
  40. Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  41. Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.; Brickey, W.J.; Ting, J.P. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Vandanmagsar, B.; Youm, Y.H.; Ravussin, A.; Galgani, J.E.; Stadler, K.; Mynatt, R.L.; Ravussin, E.; Stephens, J.M.; Dixit, V.D. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 2011, 17, 179–188. [Google Scholar] [CrossRef] [PubMed]
  43. Kanda, H.; Tateya, S.; Tamori, Y.; Kotani, K.; Hiasa, K.; Kitazawa, R.; Kitazawa, S.; Miyachi, H.; Maeda, S.; Egashira, K.; et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Investig. 2006, 116, 1494–1505. [Google Scholar] [CrossRef] [PubMed]
  44. Ly, L.D.; Xu, S.; Choi, S.K.; Ha, C.M.; Thoudam, T.; Cha, S.K.; Wiederkehr, A.; Wollheim, C.B.; Lee, I.K.; Park, K.S. Oxidative stress and calcium dysregulation by palmitate in type 2 diabetes. Exp. Mol. Med. 2017, 49, e291. [Google Scholar] [CrossRef]
  45. Finucane, O.M.; Lyons, C.L.; Murphy, A.M.; Reynolds, C.M.; Klinger, R.; Healy, N.P.; Cooke, A.A.; Coll, R.C.; McAllan, L.; Nilaweera, K.N.; et al. Monounsaturated fatty acid-enriched high-fat diets impede adipose NLRP3 inflammasome-mediated IL-1beta secretion and insulin resistance despite obesity. Diabetes 2015, 64, 2116–2128. [Google Scholar] [CrossRef] [Green Version]
  46. Lyons, C.L.; Roche, H.M. Nutritional Modulation of AMPK-Impact upon Metabolic-Inflammation. Int. J. Mol. Sci. 2018, 19, 3092. [Google Scholar] [CrossRef] [Green Version]
  47. Berg, A.H.; Scherer, P.E. Adipose tissue, inflammation, and cardiovascular disease. Circ. Res. 2005, 96, 939–949. [Google Scholar] [CrossRef] [Green Version]
  48. Greenberger, N.J.; Skillman, T.G. Medium-chain triglycerides. N. Engl. J. Med. 1969, 280, 1045–1058. [Google Scholar] [CrossRef] [Green Version]
  49. Hu, J.; Lin, S.; Zheng, B.; Cheung, P.C.K. Short-chain fatty acids in control of energy metabolism. Crit. Rev. Food Sci. Nutr. 2018, 58, 1243–1249. [Google Scholar] [CrossRef]
  50. Ouimet, M.; Barrett, T.J.; Fisher, E.A. HDL and Reverse Cholesterol Transport. Circ. Res. 2019, 124, 1505–1518. [Google Scholar] [CrossRef]
  51. Tosheska Trajkovska, K.; Topuzovska, S. High-density lipoprotein metabolism and reverse cholesterol transport: Strategies for raising HDL cholesterol. Anatol. J. Cardiol. 2017, 18, 149–154. [Google Scholar] [CrossRef] [PubMed]
  52. Li, C.; Ge, X.; Su, B.; Fu, Q.; Wang, B.; Liu, X.; Ren, Y.; Song, L.; Yang, N. Characterization of class B scavenger receptor type 1 (SRB1) in turbot (Scophthalmus maximus L.). Fish. Shellfish. Immunol. 2020, 100, 358–367. [Google Scholar] [CrossRef] [PubMed]
  53. Walther, T.C.; Chung, J.; Farese, R.V., Jr. Lipid Droplet Biogenesis. Annu. Rev. Cell Dev. Biol. 2017, 33, 491–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Thiam, A.R.; Beller, M. The why, when and how of lipid droplet diversity. J. Cell Sci. 2017, 130, 315–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ameer, F.; Scandiuzzi, L.; Hasnain, S.; Kalbacher, H.; Zaidi, N. De novo lipogenesis in health and disease. Metabolism 2014, 63, 895–902. [Google Scholar] [CrossRef] [PubMed]
  56. Matsuzaka, T.; Kuba, M.; Koyasu, S.; Yamamoto, Y.; Motomura, K.; Arulmozhiraja, S.; Ohno, H.; Sharma, R.; Shimura, T.; Okajima, Y.; et al. Hepatocyte ELOVL Fatty Acid Elongase 6 Determines Ceramide Acyl-Chain Length and Hepatic Insulin Sensitivity in Mice. Hepatology 2020, 71, 1609–1625. [Google Scholar] [CrossRef]
  57. Shi, X.; Li, J.; Zou, X.; Greggain, J.; Rodkaer, S.V.; Faergeman, N.J.; Liang, B.; Watts, J.L. Regulation of lipid droplet size and phospholipid composition by stearoyl-CoA desaturase. J. Lipid Res. 2013, 54, 2504–2514. [Google Scholar] [CrossRef] [Green Version]
  58. Waters, K.M.; Ntambi, J.M. Polyunsaturated fatty acids inhibit hepatic stearoyl-CoA desaturase-1 gene in diabetic mice. Lipids 1996, 31, S33–S36. [Google Scholar] [CrossRef]
  59. Ntambi, J.M. Cellular differentiation and dietary regulation of gene expression. Prostaglandins Leukot. Essent. Fat. Acids 1995, 52, 117–120. [Google Scholar] [CrossRef]
  60. 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. 2006, 47, 1928–1939. [Google Scholar] [CrossRef] [Green Version]
  61. Miyazaki, M.; Kim, Y.C.; Gray-Keller, M.P.; Attie, A.D.; Ntambi, J.M. The biosynthesis of hepatic cholesterol esters and triglycerides is impaired in mice with a disruption of the gene for stearoyl-CoA desaturase 1. J. Biol. Chem. 2000, 275, 30132–30138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Miyazaki, M.; Man, W.C.; Ntambi, J.M. Targeted disruption of stearoyl-CoA desaturase1 gene in mice causes atrophy of sebaceous and meibomian glands and depletion of wax esters in the eyelid. J. Nutr. 2001, 131, 2260–2268. [Google Scholar] [CrossRef] [PubMed]
  63. Lounis, M.A.; Escoula, Q.; Veillette, C.; Bergeron, K.F.; Ntambi, J.M.; Mounier, C. SCD1 deficiency protects mice against ethanol-induced liver injury. Biochim. Biophys. Acta 2016, 1861, 1662–1670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cedernaes, J.; Alsio, J.; Vastermark, A.; Riserus, U.; Schioth, H.B. Adipose tissue stearoyl-CoA desaturase 1 index is increased and linoleic acid is decreased in obesity-prone rats fed a high-fat diet. Lipids Health Dis. 2013, 12, 2. [Google Scholar] [CrossRef] [Green Version]
  65. Liu, X.L.; Cao, H.X.; Wang, B.C.; Xin, F.Z.; Zhang, R.N.; Zhou, D.; Yang, R.X.; Zhao, Z.H.; Pan, Q.; Fan, J.G. miR-192-5p regulates lipid synthesis in non-alcoholic fatty liver disease through SCD-1. World J. Gastroenterol. 2017, 23, 8140–8151. [Google Scholar] [CrossRef]
  66. Nagao, K.; Murakami, A.; Umeda, M. Structure and Function of Delta9-Fatty Acid Desaturase. Chem. Pharm. Bull. 2019, 67, 327–332. [Google Scholar] [CrossRef] [Green Version]
  67. Igal, R.A.; Sinner, D.I. Stearoyl-CoA desaturase 5 (SCD5), a Delta-9 fatty acyl desaturase in search of a function. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158840. [Google Scholar] [CrossRef]
  68. Miyazaki, M.; Bruggink, S.M.; Ntambi, J.M. Identification of mouse palmitoyl-coenzyme A Delta9-desaturase. J. Lipid Res. 2006, 47, 700–704. [Google Scholar] [CrossRef] [Green Version]
  69. Zhang, S.; Yang, Y.; Shi, Y. Characterization of human SCD2, an oligomeric desaturase with improved stability and enzyme activity by cross-linking in intact cells. Biochem. J. 2005, 388, 135–142. [Google Scholar] [CrossRef] [Green Version]
  70. Kaestner, K.H.; Ntambi, J.M.; Kelly, T.J., Jr.; Lane, M.D. Differentiation-induced gene expression in 3T3-L1 preadipocytes. A second differentially expressed gene encoding stearoyl-CoA desaturase. J. Biol. Chem. 1989, 264, 14755–14761. [Google Scholar]
  71. Lee, S.H.; Dobrzyn, A.; Dobrzyn, P.; Rahman, S.M.; Miyazaki, M.; Ntambi, J.M. Lack of stearoyl-CoA desaturase 1 upregulates basal thermogenesis but causes hypothermia in a cold environment. J. Lipid Res. 2004, 45, 1674–1682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Miyazaki, M.; Jacobson, M.J.; Man, W.C.; Cohen, P.; Asilmaz, E.; Friedman, J.M.; Ntambi, J.M. Identification and characterization of murine SCD4, a novel heart-specific stearoyl-CoA desaturase isoform regulated by leptin and dietary factors. J. Biol. Chem. 2003, 278, 33904–33911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Miyazaki, M.; Gomez, F.E.; Ntambi, J.M. Lack of stearoyl-CoA desaturase-1 function induces a palmitoyl-CoA Delta6 desaturase and represses the stearoyl-CoA desaturase-3 gene in the preputial glands of the mouse. J. Lipid Res. 2002, 43, 2146–2154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ntambi, J.M.; Miyazaki, M.; Stoehr, J.P.; Lan, H.; Kendziorski, C.M.; Yandell, B.S.; Song, Y.; Cohen, P.; Friedman, J.M.; Attie, A.D. Loss of stearoyl-CoA desaturase-1 function protects mice against adiposity. Proc. Natl. Acad. Sci. USA 2002, 99, 11482–11486. [Google Scholar] [CrossRef] [Green Version]
  75. Miyazaki, M.; Sampath, H.; Liu, X.; Flowers, M.T.; Chu, K.; Dobrzyn, A.; Ntambi, J.M. Stearoyl-CoA desaturase-1 deficiency attenuates obesity and insulin resistance in leptin-resistant obese mice. Biochem. Biophys. Res. Commun. 2009, 380, 818–822. [Google Scholar] [CrossRef] [Green Version]
  76. Miyazaki, M.; Flowers, M.T.; Sampath, H.; Chu, K.; Otzelberger, C.; Liu, X.; Ntambi, J.M. Hepatic stearoyl-CoA desaturase-1 deficiency protects mice from carbohydrate-induced adiposity and hepatic steatosis. Cell Metab. 2007, 6, 484–496. [Google Scholar] [CrossRef] [Green Version]
  77. Lounis, M.A.; Bergeron, K.F.; Burhans, M.S.; Ntambi, J.M.; Mounier, C. Oleate activates SREBP-1 signaling activity in SCD1-deficient hepatocytes. Am. J. Physiol. Endocrinol. Metab. 2017, 313, E710–E720. [Google Scholar] [CrossRef]
  78. Softic, S.; Cohen, D.E.; Kahn, C.R. Role of Dietary Fructose and Hepatic De Novo Lipogenesis in Fatty Liver Disease. Dig. Dis. Sci. 2016, 61, 1282–1293. [Google Scholar] [CrossRef] [Green Version]
  79. Lounis, M.A.; Lalonde, S.; Rial, S.A.; Bergeron, K.F.; Ralston, J.C.; Mutch, D.M.; Mounier, C. Hepatic BSCL2 (Seipin) Deficiency Disrupts Lipid Droplet Homeostasis and Increases Lipid Metabolism via SCD1 Activity. Lipids 2017, 52, 129–150. [Google Scholar] [CrossRef] [Green Version]
  80. Mauvoisin, D.; Mounier, C. Hormonal and nutritional regulation of SCD1 gene expression. Biochimie 2011, 93, 78–86. [Google Scholar] [CrossRef]
  81. Mauvoisin, D.; Rocque, G.; Arfa, O.; Radenne, A.; Boissier, P.; Mounier, C. Role of the PI3-kinase/mTor pathway in the regulation of the stearoyl CoA desaturase (SCD1) gene expression by insulin in liver. J. Cell Commun. Signal. 2007, 1, 113–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ortega-Prieto, P.; Postic, C. Carbohydrate Sensing Through the Transcription Factor ChREBP. Front. Genet. 2019, 10, 472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yao, D.W.; Luo, J.; He, Q.Y.; Xu, H.F.; Li, J.; Shi, H.B.; Wang, H.; Chen, Z.; Loor, J.J. Liver X receptor alpha promotes the synthesis of monounsaturated fatty acids in goat mammary epithelial cells via the control of stearoyl-coenzyme A desaturase 1 in an SREBP-1-dependent manner. J. Dairy Sci. 2016, 99, 6391–6402. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, G.; Kuang, S.; Cao, R.; Wang, J.; Peng, Q.; Sun, C. Sorafenib kills liver cancer cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids via the ATP-AMPK-mTOR-SREBP1 signaling pathway. Faseb J. 2019, 33, 10089–10103. [Google Scholar] [CrossRef] [Green Version]
  85. De Souza, C.O.; Teixeira, A.A.S.; Biondo, L.A.; Lima Junior, E.A.; Batatinha, H.A.P.; Rosa Neto, J.C. Palmitoleic Acid Improves Metabolic Functions in Fatty Liver by PPARalpha-Dependent AMPK Activation. J. Cell Physiol. 2017, 232, 2168–2177. [Google Scholar] [CrossRef]
  86. Kwon, B.; Querfurth, H.W. Palmitate activates mTOR/p70S6K through AMPK inhibition and hypophosphorylation of raptor in skeletal muscle cells: Reversal by oleate is similar to metformin. Biochimie 2015, 118, 141–150. [Google Scholar] [CrossRef]
  87. Da Silva-Santi, L.G.; Antunes, M.M.; Caparroz-Assef, S.M.; Carbonera, F.; Masi, L.N.; Curi, R.; Visentainer, J.V.; Bazotte, R.B. Liver Fatty Acid Composition and Inflammation in Mice Fed with High-Carbohydrate Diet or High-Fat Diet. Nutrients 2016, 8, 682. [Google Scholar] [CrossRef] [Green Version]
  88. Calder, P.C.; Ahluwalia, N.; Brouns, F.; Buetler, T.; Clement, K.; Cunningham, K.; Esposito, K.; Jonsson, L.S.; Kolb, H.; Lansink, M.; et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br. J. Nutr. 2011, 106, S5–S78. [Google Scholar] [CrossRef]
  89. Cnop, M. Fatty acids and glucolipotoxicity in the pathogenesis of Type 2 diabetes. Biochem. Soc. Trans. 2008, 36, 348–352. [Google Scholar] [CrossRef] [Green Version]
  90. Johnson, L.; Mander, A.P.; Jones, L.R.; Emmett, P.M.; Jebb, S.A. Energy-dense, low-fiber, high-fat dietary pattern is associated with increased fatness in childhood. Am. J. Clin. Nutr. 2008, 87, 846–854. [Google Scholar] [CrossRef] [Green Version]
  91. Vaittinen, M.; Mannisto, V.; Kakela, P.; Agren, J.; Tiainen, M.; Schwab, U.; Pihlajamaki, J. Interorgan cross talk between fatty acid metabolism, tissue inflammation, and FADS2 genotype in humans with obesity. Obesity 2017, 25, 545–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Rocha, D.M.; Bressan, J.; Hermsdorff, H.H. The role of dietary fatty acid intake in inflammatory gene expression: A critical review. Sao Paulo Med. J. 2017, 135, 157–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhang, Y.; Chua, S., Jr. Leptin Function and Regulation. Compr. Physiol. 2017, 8, 351–369. [Google Scholar] [CrossRef] [PubMed]
  94. Carbone, F.; La Rocca, C.; Matarese, G. Immunological functions of leptin and adiponectin. Biochimie 2012, 94, 2082–2088. [Google Scholar] [CrossRef]
  95. Jimenez-Gomez, Y.; Lopez-Miranda, J.; Blanco-Colio, L.M.; Marin, C.; Perez-Martinez, P.; Ruano, J.; Paniagua, J.A.; Rodriguez, F.; Egido, J.; Perez-Jimenez, F. Olive oil and walnut breakfasts reduce the postprandial inflammatory response in mononuclear cells compared with a butter breakfast in healthy men. Atherosclerosis 2009, 204, e70–e76. [Google Scholar] [CrossRef]
  96. Lundman, P.; Boquist, S.; Samnegard, A.; Bennermo, M.; Held, C.; Ericsson, C.G.; Silveira, A.; Hamsten, A.; Tornvall, P. A high-fat meal is accompanied by increased plasma interleukin-6 concentrations. Nutr. Metab. Cardiovasc. Dis. 2007, 17, 195–202. [Google Scholar] [CrossRef]
  97. Zaki, S.M.; Fattah, S.A.; Hassan, D.S. The differential effects of high-fat and high- -fructose diets on the liver of male albino rat and the proposed underlying mechanisms. Folia Morphol. (Warsz.) 2019, 78, 124–136. [Google Scholar] [CrossRef] [Green Version]
  98. Tamer, F.; Ulug, E.; Akyol, A.; Nergiz-Unal, R. The potential efficacy of dietary fatty acids and fructose induced inflammation and oxidative stress on the insulin signaling and fat accumulation in mice. Food Chem. Toxicol. 2020, 135, 110914. [Google Scholar] [CrossRef]
  99. Feillet-Coudray, C.; Fouret, G.; Vigor, C.; Bonafos, B.; Jover, B.; Blachnio-Zabielska, A.; Rieusset, J.; Casas, F.; Gaillet, S.; Landrier, J.F.; et al. Long-Term Measures of Dyslipidemia, Inflammation, and Oxidative Stress in Rats Fed a High-Fat/High-Fructose Diet. Lipids 2019, 54, 81–97. [Google Scholar] [CrossRef] [Green Version]
  100. Antunes, M.M.; Godoy, G.; de Almeida-Souza, C.B.; da Rocha, B.A.; da Silva-Santi, L.G.; Masi, L.N.; Carbonera, F.; Visentainer, J.V.; Curi, R.; Bazotte, R.B. A high-carbohydrate diet induces greater inflammation than a high-fat diet in mouse skeletal muscle. Braz. J. Med. Biol. Res. 2020, 53, e9039. [Google Scholar] [CrossRef]
  101. Gimenez da Silva-Santi, L.; Masetto Antunes, M.; Mori, M.A.; Biesdorf de Almeida-Souza, C.; Vergilio Visentainer, J.; Carbonera, F.; Rabello Crisma, A.; Nunes Masi, L.; Massao Hirabara, S.; Curi, R.; et al. Brain Fatty Acid Composition and Inflammation in Mice Fed with High-Carbohydrate Diet or High-Fat Diet. Nutrients 2018, 10, 1277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Decarie-Spain, L.; Sharma, S.; Hryhorczuk, C.; Issa-Garcia, V.; Barker, P.A.; Arbour, N.; Alquier, T.; Fulton, S. Nucleus accumbens inflammation mediates anxiodepressive behavior and compulsive sucrose seeking elicited by saturated dietary fat. Mol. Metab. 2018, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
  103. Tu, T.H.; Kim, H.; Yang, S.; Kim, J.K.; Kim, J.G. Linoleic acid rescues microglia inflammation triggered by saturated fatty acid. Biochem. Biophys. Res. Commun. 2019, 513, 201–206. [Google Scholar] [CrossRef] [PubMed]
  104. Tran, D.Q.; Tse, E.K.; Kim, M.H.; Belsham, D.D. Diet-induced cellular neuroinflammation in the hypothalamus: Mechanistic insights from investigation of neurons and microglia. Mol. Cell Endocrinol. 2016, 438, 18–26. [Google Scholar] [CrossRef] [PubMed]
  105. Guo, W.; Wong, S.; Xie, W.; Lei, T.; Luo, Z. Palmitate modulates intracellular signaling, induces endoplasmic reticulum stress, and causes apoptosis in mouse 3T3-L1 and rat primary preadipocytes. Am. J. Physiol. Endocrinol. Metab. 2007, 293, E576–E586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Kennedy, A.; Martinez, K.; Chuang, C.C.; LaPoint, K.; McIntosh, M. Saturated fatty acid-mediated inflammation and insulin resistance in adipose tissue: Mechanisms of action and implications. J. Nutr. 2009, 139, 1–4. [Google Scholar] [CrossRef] [PubMed]
  107. Namgaladze, D.; Brune, B. Macrophage fatty acid oxidation and its roles in macrophage polarization and fatty acid-induced inflammation. Biochim. Et Biophys. Acta 2016, 1861, 1796–1807. [Google Scholar] [CrossRef]
  108. Gehrmann, W.; Wurdemann, W.; Plotz, T.; Jorns, A.; Lenzen, S.; Elsner, M. Antagonism Between Saturated and Unsaturated Fatty Acids in ROS Mediated Lipotoxicity in Rat Insulin-Producing Cells. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2015, 36, 852–865. [Google Scholar] [CrossRef]
  109. Nemecz, M.; Constantin, A.; Dumitrescu, M.; Alexandru, N.; Filippi, A.; Tanko, G.; Georgescu, A. The Distinct Effects of Palmitic and Oleic Acid on Pancreatic Beta Cell Function: The Elucidation of Associated Mechanisms and Effector Molecules. Front. Pharmacol. 2018, 9, 1554. [Google Scholar] [CrossRef] [Green Version]
  110. Lee, J.Y.; Ye, J.; Gao, Z.; Youn, H.S.; Lee, W.H.; Zhao, L.; Sizemore, N.; Hwang, D.H. Reciprocal modulation of Toll-like receptor-4 signaling pathways involving MyD88 and phosphatidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids. J. Biol. Chem. 2003, 278, 37041–37051. [Google Scholar] [CrossRef] [Green Version]
  111. Wong, S.W.; Kwon, M.J.; Choi, A.M.; Kim, H.P.; Nakahira, K.; Hwang, D.H. 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. 2009, 284, 27384–27392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Eisele, P.S.; Salatino, S.; Sobek, J.; Hottiger, M.O.; Handschin, C. The peroxisome proliferator-activated receptor gamma coactivator 1alpha/beta (PGC-1) coactivators repress the transcriptional activity of NF-kappaB in skeletal muscle cells. J. Biol. Chem. 2013, 288, 2246–2260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Chen, H.; Liu, Y.; Li, D.; Song, J.; Xia, M. PGC-1beta suppresses saturated fatty acid-induced macrophage inflammation by inhibiting TAK1 activation. Iubmb Life 2016, 68, 145–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Bosquet, A.; Girona, J.; Guaita-Esteruelas, S.; Heras, M.; Saavedra-Garcia, P.; Martinez-Micaelo, N.; Masana, L.; Rodriguez-Calvo, R. FABP4 inhibitor BMS309403 decreases saturated-fatty-acid-induced endoplasmic reticulum stress-associated inflammation in skeletal muscle by reducing p38 MAPK activation. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2018, 1863, 604–613. [Google Scholar] [CrossRef] [PubMed]
  115. Shahidi, F.; Ambigaipalan, P. Omega-3 Polyunsaturated Fatty Acids and Their Health Benefits. Annu Rev. Food Sci. Technol. 2018, 9, 345–381. [Google Scholar] [CrossRef]
  116. Costantini, L.; Molinari, R.; Farinon, B.; Merendino, N. Impact of Omega-3 Fatty Acids on the Gut Microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef] [Green Version]
  117. Wysoczanski, T.; Sokola-Wysoczanska, E.; Pekala, J.; Lochynski, S.; Czyz, K.; Bodkowski, R.; Herbinger, G.; Patkowska-Sokola, B.; Librowski, T. Omega-3 Fatty Acids and their Role in Central Nervous System-A Review. Curr. Med. Chem. 2016, 23, 816–831. [Google Scholar] [CrossRef]
  118. Raatz, S.K.; Conrad, Z.; Jahns, L.; Belury, M.A.; Picklo, M.J. Modeled replacement of traditional soybean and canola oil with high-oleic varieties increases monounsaturated fatty acid and reduces both saturated fatty acid and polyunsaturated fatty acid intake in the US adult population. Am. J. Clin. Nutr. 2018, 108, 594–602. [Google Scholar] [CrossRef]
  119. Shively, C.A.; Appt, S.E.; Vitolins, M.Z.; Uberseder, B.; Michalson, K.T.; Silverstein-Metzler, M.G.; Register, T.C. Mediterranean versus Western Diet Effects on Caloric Intake, Obesity, Metabolism, and Hepatosteatosis in Nonhuman Primates. Obesity 2019, 27, 777–784. [Google Scholar] [CrossRef]
  120. Martin-Pelaez, S.; Mosele, J.I.; Pizarro, N.; Farras, M.; de la Torre, R.; Subirana, I.; Perez-Cano, F.J.; Castaner, O.; Sola, R.; Fernandez-Castillejo, S.; et al. Effect of virgin olive oil and thyme phenolic compounds on blood lipid profile: Implications of human gut microbiota. Eur. J. Nutr. 2017, 56, 119–131. [Google Scholar] [CrossRef]
  121. Wang, Z.; Zhao, Y. Gut microbiota derived metabolites in cardiovascular health and disease. Protein Cell 2018, 9, 416–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Trichopoulou, A.; Katsouyanni, K.; Gnardellis, C. The traditional Greek diet. Eur. J. Clin. Nutr. 1993, 47, S76–S81. [Google Scholar] [PubMed]
  123. De Pergola, G.; D’Alessandro, A. Influence of Mediterranean Diet on Blood Pressure. Nutrients 2018, 10, 1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Esposito, K.; Maiorino, M.I.; Bellastella, G.; Panagiotakos, D.B.; Giugliano, D. Mediterranean diet for type 2 diabetes: Cardiometabolic benefits. Endocrine 2017, 56, 27–32. [Google Scholar] [CrossRef]
  125. Tuttolomondo, A.; Simonetta, I.; Daidone, M.; Mogavero, A.; Ortello, A.; Pinto, A. Metabolic and Vascular Effect of the Mediterranean Diet. Int. J. Mol. Sci. 2019, 20, 4716. [Google Scholar] [CrossRef] [Green Version]
  126. De Filippis, F.; Pellegrini, N.; Vannini, L.; Jeffery, I.B.; La Storia, A.; Laghi, L.; Serrazanetti, D.I.; Di Cagno, R.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef]
  127. Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef] [Green Version]
  128. Cesari, F.; Sofi, F.; Molino Lova, R.; Vannetti, F.; Pasquini, G.; Cecchi, F.; Marcucci, R.; Gori, A.M.; Macchi, C. Aging process, adherence to Mediterranean diet and nutritional status in a large cohort of nonagenarians: Effects on endothelial progenitor cells. Nutr. Metab. Cardiovasc. Dis. NMCD 2018, 28, 84–90. [Google Scholar] [CrossRef]
  129. Galland, L. Diet and inflammation. Nutr. Clin. Pract. Off. Publ. Am. Soc. Parenter. Enter. Nutr. 2010, 25, 634–640. [Google Scholar] [CrossRef]
  130. Casas, R.; Castro-Barquero, S.; Estruch, R.; Sacanella, E. Nutrition and Cardiovascular Health. Int. J. Mol. Sci. 2018, 19, 3988. [Google Scholar] [CrossRef] [Green Version]
  131. Schwingshackl, L.; Christoph, M.; Hoffmann, G. Effects of Olive Oil on Markers of Inflammation and Endothelial Function-A Systematic Review and Meta-Analysis. Nutrients 2015, 7, 7651–7675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Candido, F.G.; Valente, F.X.; Grzeskowiak, L.M.; Moreira, A.P.B.; Rocha, D.; Alfenas, R.C.G. Impact of dietary fat on gut microbiota and low-grade systemic inflammation: Mechanisms and clinical implications on obesity. Int. J. Food Sci. Nutr. 2018, 69, 125–143. [Google Scholar] [CrossRef] [PubMed]
  133. Paniagua, J.A.; Gallego de la Sacristana, A.; Romero, I.; Vidal-Puig, A.; Latre, J.M.; Sanchez, E.; Perez-Martinez, P.; Lopez-Miranda, J.; Perez-Jimenez, F. Monounsaturated fat-rich diet prevents central body fat distribution and decreases postprandial adiponectin expression induced by a carbohydrate-rich diet in insulin-resistant subjects. Diabetes Care 2007, 30, 1717–1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Casas, R.; Estruch, R.; Sacanella, E. The Protective Effects of Extra Virgin Olive Oil on Immune-mediated Inflammatory Responses. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 23–35. [Google Scholar] [CrossRef] [PubMed]
  135. Wongwarawipat, T.; Papageorgiou, N.; Bertsias, D.; Siasos, G.; Tousoulis, D. Olive Oil-related Anti-inflammatory Effects on Atherosclerosis: Potential Clinical Implications. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 51–62. [Google Scholar] [CrossRef] [PubMed]
  136. Konstantinidou, V.; Covas, M.I.; Sola, R.; Fito, M. Up-to date knowledge on the in vivo transcriptomic effect of the Mediterranean diet in humans. Mol. Nutr. Food Res. 2013, 57, 772–783. [Google Scholar] [CrossRef]
  137. Konstantinidou, V.; Covas, M.I.; Munoz-Aguayo, D.; Khymenets, O.; de la Torre, R.; Saez, G.; Tormos Mdel, C.; Toledo, E.; Marti, A.; Ruiz-Gutierrez, V.; et al. In vivo nutrigenomic effects of virgin olive oil polyphenols within the frame of the Mediterranean diet: A randomized controlled trial. Faseb J. 2010, 24, 2546–2557. [Google Scholar] [CrossRef] [Green Version]
  138. Esposito, K.; Marfella, R.; Ciotola, M.; Di Palo, C.; Giugliano, F.; Giugliano, G.; D’Armiento, M.; D’Andrea, F.; Giugliano, D. Effect of a mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: A randomized trial. JAMA 2004, 292, 1440–1446. [Google Scholar] [CrossRef] [Green Version]
  139. Toscano, R.; Millan-Linares, M.C.; Lemus-Conejo, A.; Claro, C.; Sanchez-Margalet, V.; Montserrat-de la Paz, S. Postprandial triglyceride-rich lipoproteins promote M1/M2 microglia polarization in a fatty-acid-dependent manner. J. Nutr. Biochem. 2020, 75, 108248. [Google Scholar] [CrossRef]
  140. Montserrat-de la Paz, S.; Rodriguez, D.; Cardelo, M.P.; Naranjo, M.C.; Bermudez, B.; Abia, R.; Muriana, F.J.G.; Lopez, S. The effects of exogenous fatty acids and niacin on human monocyte-macrophage plasticity. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef]
  141. Mika, A.; Sikorska-Wisniewska, M.; Malgorzewicz, S.; Stepnowski, P.; Debska-Slizien, A.; Sledzinski, T.; Chmielewski, M. Potential contribution of monounsaturated fatty acids to cardiovascular risk in chronic kidney disease. Pol. Arch. Intern. Med. 2018, 128, 755–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Weiss-Hersh, K.; Garcia, A.L.; Marosvolgyi, T.; Szklenar, M.; Decsi, T.; Ruhl, R. Saturated and monounsaturated fatty acids in membranes are determined by the gene expression of their metabolizing enzymes SCD1 and ELOVL6 regulated by the intake of dietary fat. Eur. J. Nutr. 2020, 59, 2759–2769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Montserrat-de la Paz, S.; Naranjo, M.C.; Millan-Linares, M.C.; Lopez, S.; Abia, R.; Biessen, E.A.L.; Muriana, F.J.G.; Bermudez, B. Monounsaturated Fatty Acids in a High-Fat Diet and Niacin Protect from White Fat Dysfunction in the Metabolic Syndrome. Mol. Nutr. Food Res. 2019, 63, e1900425. [Google Scholar] [CrossRef] [PubMed]
  144. Yang, Z.H.; Pryor, M.; Noguchi, A.; Sampson, M.; Johnson, B.; Pryor, M.; Donkor, K.; Amar, M.; Remaley, A.T. Dietary Palmitoleic Acid Attenuates Atherosclerosis Progression and Hyperlipidemia in Low-Density Lipoprotein Receptor-Deficient Mice. Mol. Nutr. Food Res. 2019, 63, e1900120. [Google Scholar] [CrossRef]
  145. Yang, X.; Zhang, Y.; Lin, J.; Pen, A.; Ying, C.; Cao, W.; Mao, L. A lower proportion of dietary saturated/monounsaturated/polyunsaturated fatty acids reduces the expression of adiponectin in rats fed a high-fat diet. Nutr. Res. 2012, 32, 285–291. [Google Scholar] [CrossRef]
  146. Acosta-Montano, P.; Rodriguez-Velazquez, E.; Ibarra-Lopez, E.; Frayde-Gomez, H.; Mas-Oliva, J.; Delgado-Coello, B.; Rivero, I.A.; Alatorre-Meda, M.; Aguilera, J.; Guevara-Olaya, L.; et al. Fatty Acid and Lipopolysaccharide Effect on Beta Cells Proteostasis and its Impact on Insulin Secretion. Cells 2019, 8, 884. [Google Scholar] [CrossRef] [Green Version]
  147. Diakogiannaki, E.; Welters, H.J.; Morgan, N.G. Differential regulation of the endoplasmic reticulum stress response in pancreatic beta-cells exposed to long-chain saturated and monounsaturated fatty acids. J. Endocrinol. 2008, 197, 553–563. [Google Scholar] [CrossRef]
  148. Quiles, J.L.; Pamplona, R.; Ramirez-Tortosa, M.C.; Naudi, A.; Portero-Otin, M.; Araujo-Nepomuceno, E.; Lopez-Frias, M.; Battino, M.; Ochoa, J.J. Coenzyme Q addition to an n-6 PUFA-rich diet resembles benefits on age-related mitochondrial DNA deletion and oxidative stress of a MUFA-rich diet in rat heart. Mech. Ageing Dev. 2010, 131, 38–47. [Google Scholar] [CrossRef]
  149. Chen, X.; Li, L.; Liu, X.; Luo, R.; Liao, G.; Li, L.; Liu, J.; Cheng, J.; Lu, Y.; Chen, Y. Oleic acid protects saturated fatty acid mediated lipotoxicity in hepatocytes and rat of non-alcoholic steatohepatitis. Life Sci. 2018, 203, 291–304. [Google Scholar] [CrossRef]
  150. Patil, R.; Mohanty, B.; Liu, B.; Chandrashekaran, I.R.; Headey, S.J.; Williams, M.L.; Clements, C.S.; Ilyichova, O.; Doak, B.C.; Genissel, P.; et al. A ligand-induced structural change in fatty acid-binding protein 1 is associated with potentiation of peroxisome proliferator-activated receptor alpha agonists. J. Biol. Chem. 2019, 294, 3720–3734. [Google Scholar] [CrossRef]
  151. Najt, C.P.; Khan, S.A.; Heden, T.D.; Witthuhn, B.A.; Perez, M.; Heier, J.L.; Mead, L.E.; Franklin, M.P.; Karanja, K.K.; Graham, M.J.; et al. Lipid Droplet-Derived Monounsaturated Fatty Acids Traffic via PLIN5 to Allosterically Activate SIRT1. Mol. Cell 2020, 77, 810–824.e818. [Google Scholar] [CrossRef] [PubMed]
  152. Kauppinen, A.; Suuronen, T.; Ojala, J.; Kaarniranta, K.; Salminen, A. Antagonistic crosstalk between NF-kappaB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell. Signal. 2013, 25, 1939–1948. [Google Scholar] [CrossRef] [PubMed]
  153. Granados, N.; Amengual, J.; Ribot, J.; Palou, A.; Bonet, M.L. Distinct effects of oleic acid and its trans-isomer elaidic acid on the expression of myokines and adipokines in cell models. Br. J. Nutr. 2011, 105, 1226–1234. [Google Scholar] [CrossRef] [PubMed]
  154. Rosen, E.D.; Spiegelman, B.M. PPARgamma: A nuclear regulator of metabolism, differentiation, and cell growth. J. Biol. Chem. 2001, 276, 37731–37734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Iwaki, M.; Matsuda, M.; Maeda, N.; Funahashi, T.; Matsuzawa, Y.; Makishima, M.; Shimomura, I. Induction of adiponectin, a fat-derived antidiabetic and antiatherogenic factor, by nuclear receptors. Diabetes 2003, 52, 1655–1663. [Google Scholar] [CrossRef] [Green Version]
  156. Fang, H.; Judd, R.L. Adiponectin Regulation and Function. Compr. Physiol. 2018, 8, 1031–1063. [Google Scholar] [CrossRef]
  157. Dietze-Schroeder, D.; Sell, H.; Uhlig, M.; Koenen, M.; Eckel, J. Autocrine action of adiponectin on human fat cells prevents the release of insulin resistance-inducing factors. Diabetes 2005, 54, 2003–2011. [Google Scholar] [CrossRef] [Green Version]
  158. Ertunc, M.E.; Hotamisligil, G.S. Lipid signaling and lipotoxicity in metaflammation: Indications for metabolic disease pathogenesis and treatment. J. Lipid Res. 2016, 57, 2099–2114. [Google Scholar] [CrossRef] [Green Version]
  159. Lo Verme, J.; Fu, J.; Astarita, G.; La Rana, G.; Russo, R.; Calignano, A.; Piomelli, D. The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol. Pharmacol. 2005, 67, 15–19. [Google Scholar] [CrossRef]
  160. Kang, K.; Reilly, S.M.; Karabacak, V.; Gangl, M.R.; Fitzgerald, K.; Hatano, B.; Lee, C.H. Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity. Cell Metab. 2008, 7, 485–495. [Google Scholar] [CrossRef] [Green Version]
  161. Lovren, F.; Pan, Y.; Quan, A.; Szmitko, P.E.; Singh, K.K.; Shukla, P.C.; Gupta, M.; Chan, L.; Al-Omran, M.; Teoh, H.; et al. Adiponectin primes human monocytes into alternative anti-inflammatory M2 macrophages. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H656–H663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Chan, K.L.; Pillon, N.J.; Sivaloganathan, D.M.; Costford, S.R.; Liu, Z.; Theret, M.; Chazaud, B.; Klip, A. Palmitoleate Reverses High Fat-induced Proinflammatory Macrophage Polarization via AMP-activated Protein Kinase (AMPK). J. Biol. Chem. 2015, 290, 16979–16988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Yang, Z.; Kahn, B.B.; Shi, H.; Xue, B.Z. Macrophage alpha1 AMP-activated protein kinase (alpha1AMPK) antagonizes fatty acid-induced inflammation through SIRT1. J. Biol. Chem. 2010, 285, 19051–19059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Cimen, I.; Kocaturk, B.; Koyuncu, S.; Tufanli, O.; Onat, U.I.; Yildirim, A.D.; Apaydin, O.; Demirsoy, S.; Aykut, Z.G.; Nguyen, U.T.; et al. Prevention of atherosclerosis by bioactive palmitoleate through suppression of organelle stress and inflammasome activation. Sci. Transl. Med. 2016, 8, 358ra126. [Google Scholar] [CrossRef] [Green Version]
  165. Ralston, J.C.; Nguyen-Tu, M.S.; Lyons, C.L.; Cooke, A.A.; Murphy, A.M.; Falvey, A.; Finucane, O.M.; McGillicuddy, F.C.; Rutter, G.A.; Roche, H.M. Dietary substitution of SFA with MUFA within high-fat diets attenuates hyperinsulinaemia and pancreatic islet dysfunction. Br. J. Nutr. 2020, 124, 247–255. [Google Scholar] [CrossRef] [Green Version]
  166. Hu, W.; Ross, J.; Geng, T.; Brice, S.E.; Cowart, L.A. Differential regulation of dihydroceramide desaturase by palmitate versus monounsaturated fatty acids: Implications for insulin resistance. J. Biol. Chem. 2011, 286, 16596–16605. [Google Scholar] [CrossRef] [Green Version]
  167. Sieber, J.; Lindenmeyer, M.T.; Kampe, K.; Campbell, K.N.; Cohen, C.D.; Hopfer, H.; Mundel, P.; Jehle, A.W. Regulation of podocyte survival and endoplasmic reticulum stress by fatty acids. Am. J. Physiol. Ren. Physiol. 2010, 299, F821–F829. [Google Scholar] [CrossRef] [Green Version]
  168. De Souza, C.O.; Valenzuela, C.A.; Baker, E.J.; Miles, E.A.; Rosa Neto, J.C.; Calder, P.C. Palmitoleic Acid has Stronger Anti-Inflammatory Potential in Human Endothelial Cells Compared to Oleic and Palmitic Acids. Mol. Nutr. Food Res. 2018, 62, e1800322. [Google Scholar] [CrossRef]
  169. Stryjecki, C.; Roke, K.; Clarke, S.; Nielsen, D.; Badawi, A.; El-Sohemy, A.; Ma, D.W.; Mutch, D.M. Enzymatic activity and genetic variation in SCD1 modulate the relationship between fatty acids and inflammation. Mol. Genet. Metab. 2012, 105, 421–427. [Google Scholar] [CrossRef]
  170. Castellano-Castillo, D.; Denechaud, P.D.; Fajas, L.; Moreno-Indias, I.; Oliva-Olivera, W.; Tinahones, F.; Queipo-Ortuno, M.I.; Cardona, F. Human adipose tissue H3K4me3 histone mark in adipogenic, lipid metabolism and inflammatory genes is positively associated with BMI and HOMA-IR. PLoS ONE 2019, 14, e0215083. [Google Scholar] [CrossRef] [Green Version]
  171. Anty, R.; Gual, P. Pathogenesis of non-alcoholic fatty liver disease. Presse Med. 2019, 48, 1468–1483. [Google Scholar] [CrossRef] [PubMed]
  172. Lee, J.J.; Lambert, J.E.; Hovhannisyan, Y.; Ramos-Roman, M.A.; Trombold, J.R.; Wagner, D.A.; Parks, E.J. Palmitoleic acid is elevated in fatty liver disease and reflects hepatic lipogenesis. Am. J. Clin. Nutr. 2015, 101, 34–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Oran, A.; Marshall, J.S.; Kondo, S.; Paglia, D.; McKenzie, R.C. Cyclosporin inhibits intercellular adhesion molecule-1 expression and reduces mast cell numbers in the asebia mouse model of chronic skin inflammation. Br. J. Dermatol. 1997, 136, 519–526. [Google Scholar] [CrossRef] [PubMed]
  174. Zheng, Y.; Eilertsen, K.J.; Ge, L.; Zhang, L.; Sundberg, J.P.; Prouty, S.M.; Stenn, K.S.; Parimoo, S. Scd1 is expressed in sebaceous glands and is disrupted in the asebia mouse. Nat. Genet. 1999, 23, 268–270. [Google Scholar] [CrossRef] [PubMed]
  175. Dumas, S.N.; Guo, C.A.; Kim, J.K.; Friedline, R.H.; Ntambi, J.M. Interleukin-6 derived from cutaneous deficiency of stearoyl-CoA desaturase-1 may mediate metabolic organ crosstalk among skin, adipose tissue and liver. Biochem. Biophys. Res. Commun. 2019, 508, 87–91. [Google Scholar] [CrossRef] [PubMed]
  176. Flowers, M.T.; Paton, C.M.; O’Byrne, S.M.; Schiesser, K.; Dawson, J.A.; Blaner, W.S.; Kendziorski, C.; Ntambi, J.M. Metabolic changes in skin caused by Scd1 deficiency: A focus on retinol metabolism. PLoS ONE 2011, 6, e19734. [Google Scholar] [CrossRef] [Green Version]
  177. Ji, J.; Ho, B.S.; Qian, G.; Xie, X.M.; Bigliardi, P.L.; Bigliardi-Qi, M. Aging in hair follicle stem cells and niche microenvironment. J. Dermatol. 2017, 44, 1097–1104. [Google Scholar] [CrossRef]
  178. Lu, Y.; Bu, L.; Zhou, S.; Jin, M.; Sundberg, J.P.; Jiang, H.; Qian, M.; Shi, Y.; Zhao, G.; Kong, X.; et al. Scd1ab-Xyk: A new asebia allele characterized by a CCC trinucleotide insertion in exon 5 of the stearoyl-CoA desaturase 1 gene in mouse. Mol. Genet. Genom. Mgg. 2004, 272, 129–137. [Google Scholar] [CrossRef]
  179. Schneider, M.R. Fifty years of the asebia mouse: Origins, insights and contemporary developments. Exp. Dermatol. 2015, 24, 340–341. [Google Scholar] [CrossRef] [Green Version]
  180. Sampath, H.; Flowers, M.T.; Liu, X.; Paton, C.M.; Sullivan, R.; Chu, K.; Zhao, M.; Ntambi, J.M. Skin-specific deletion of stearoyl-CoA desaturase-1 alters skin lipid composition and protects mice from high fat diet-induced obesity. J. Biol. Chem. 2009, 284, 19961–19973. [Google Scholar] [CrossRef] [Green Version]
  181. MacDonald, M.L.; van Eck, M.; Hildebrand, R.B.; Wong, B.W.; Bissada, N.; Ruddle, P.; Kontush, A.; Hussein, H.; Pouladi, M.A.; Chapman, M.J.; et al. Despite antiatherogenic metabolic characteristics, SCD1-deficient mice have increased inflammation and atherosclerosis. Arter. Thromb. Vasc. Biol. 2009, 29, 341–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Mukherjee, P.; Hough, G.; Chattopadhyay, A.; Grijalva, V.; O’Connor, E.I.; Meriwether, D.; Wagner, A.; Ntambi, J.M.; Navab, M.; Reddy, S.T.; et al. Role of enterocyte stearoyl-Co-A desaturase-1 in LDLR-null mice. J. Lipid Res. 2018, 59, 1818–1840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Ducheix, S.; Peres, C.; Hardfeldt, J.; Frau, C.; Mocciaro, G.; Piccinin, E.; Lobaccaro, J.M.; De Santis, S.; Chieppa, M.; Bertrand-Michel, J.; et al. Deletion of Stearoyl-CoA Desaturase-1 From the Intestinal Epithelium Promotes Inflammation and Tumorigenesis, Reversed by Dietary Oleate. Gastroenterology 2018, 155, 1524–1538.e1529. [Google Scholar] [CrossRef] [PubMed]
  184. Chen, C.; Shah, Y.M.; Morimura, K.; Krausz, K.W.; Miyazaki, M.; Richardson, T.A.; Morgan, E.T.; Ntambi, J.M.; Idle, J.R.; Gonzalez, F.J. Metabolomics reveals that hepatic stearoyl-CoA desaturase 1 downregulation exacerbates inflammation and acute colitis. Cell Metab. 2008, 7, 135–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Ralston, J.C.; Metherel, A.H.; Stark, K.D.; Mutch, D.M. SCD1 mediates the influence of exogenous saturated and monounsaturated fatty acids in adipocytes: Effects on cellular stress, inflammatory markers and fatty acid elongation. J. Nutr. Biochem. 2016, 27, 241–248. [Google Scholar] [CrossRef] [PubMed]
  186. Malodobra-Mazur, M.; Dziewulska, A.; Kozinski, K.; Dobrzyn, P.; Kolczynska, K.; Janikiewicz, J.; Dobrzyn, A. Stearoyl-CoA desaturase regulates inflammatory gene expression by changing DNA methylation level in 3T3 adipocytes. Int. J. Biochem. Cell Biol. 2014, 55, 40–50. [Google Scholar] [CrossRef]
  187. Oshima, M.; Pechberty, S.; Bellini, L.; Gopel, S.O.; Campana, M.; Rouch, C.; Dairou, J.; Cosentino, C.; Fantuzzi, F.; Toivonen, S.; et al. Stearoyl CoA desaturase is a gatekeeper that protects human beta cells against lipotoxicity and maintains their identity. Diabetologia 2020, 63, 395–409. [Google Scholar] [CrossRef] [Green Version]
  188. Liu, X.; Miyazaki, M.; Flowers, M.T.; Sampath, H.; Zhao, M.; Chu, K.; Paton, C.M.; Joo, D.S.; Ntambi, J.M. Loss of Stearoyl-CoA desaturase-1 attenuates adipocyte inflammation: Effects of adipocyte-derived oleate. Arter. Thromb. Vasc. Biol. 2010, 30, 31–38. [Google Scholar] [CrossRef]
  189. Brown, J.M.; Chung, S.; Sawyer, J.K.; Degirolamo, C.; Alger, H.M.; Nguyen, T.; Zhu, X.; Duong, M.N.; Wibley, A.L.; Shah, R.; et al. Inhibition of stearoyl-coenzyme A desaturase 1 dissociates insulin resistance and obesity from atherosclerosis. Circulation 2008, 118, 1467–1475. [Google Scholar] [CrossRef]
  190. Peter, A.; Weigert, C.; Staiger, H.; Machicao, F.; Schick, F.; Machann, J.; Stefan, N.; Thamer, C.; Haring, H.U.; Schleicher, E. Individual stearoyl-coa desaturase 1 expression modulates endoplasmic reticulum stress and inflammation in human myotubes and is associated with skeletal muscle lipid storage and insulin sensitivity in vivo. Diabetes 2009, 58, 1757–1765. [Google Scholar] [CrossRef] [Green Version]
  191. Dalla Valle, A.; Vertongen, P.; Spruyt, D.; Lechanteur, J.; Suain, V.; Gaspard, N.; Brion, J.P.; Gangji, V.; Rasschaert, J. Induction of Stearoyl-CoA 9-Desaturase 1 Protects Human Mesenchymal Stromal Cells against Palmitic Acid-Induced Lipotoxicity and Inflammation. Front. Endocrinol. (Lausanne) 2019, 10, 726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Hirasawa, A.; Tsumaya, K.; Awaji, T.; Katsuma, S.; Adachi, T.; Yamada, M.; Sugimoto, Y.; Miyazaki, S.; Tsujimoto, G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat. Med. 2005, 11, 90–94. [Google Scholar] [CrossRef] [PubMed]
  193. Ichimura, A.; Hirasawa, A.; Poulain-Godefroy, O.; Bonnefond, A.; Hara, T.; Yengo, L.; Kimura, I.; Leloire, A.; Liu, N.; Iida, K.; et al. Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and human. Nature 2012, 483, 350–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Crosstalk between adipocyte and macrophage leading to enhanced inflammation. FFAs (free fatty acids) produced as a consequence of SFA (saturated fatty acid) overload activate the TLR4 pathway, leading to MCP-1 (Monocyte chemoattracting protein-1), IL-6 (Interleukin-6) and TNF-α (Tumor necrosis factor alpha) secretion by adipocytes via NF-κB (Nuclear factor-kappa B) nuclear translocation. TNF-α activates TNFR (Tumor necrosis factor receptor) on recruited macrophages which, in combination with the TLR4 pathway, triggers NF-κB nuclear import and production of NLRP3 (NOD-like receptor family, pyrin domain containing 3), pro-IL-1β and pro-IL-18. Lysosomal disruption, as a consequence of ATP (adenosine triphosphate) and ROS (reactive oxygen species) accumulation, triggers NLRP3 activation and results in IL-1β/IL-18 maturation and secretion. This figure was generated with BioRender.
Figure 1. Crosstalk between adipocyte and macrophage leading to enhanced inflammation. FFAs (free fatty acids) produced as a consequence of SFA (saturated fatty acid) overload activate the TLR4 pathway, leading to MCP-1 (Monocyte chemoattracting protein-1), IL-6 (Interleukin-6) and TNF-α (Tumor necrosis factor alpha) secretion by adipocytes via NF-κB (Nuclear factor-kappa B) nuclear translocation. TNF-α activates TNFR (Tumor necrosis factor receptor) on recruited macrophages which, in combination with the TLR4 pathway, triggers NF-κB nuclear import and production of NLRP3 (NOD-like receptor family, pyrin domain containing 3), pro-IL-1β and pro-IL-18. Lysosomal disruption, as a consequence of ATP (adenosine triphosphate) and ROS (reactive oxygen species) accumulation, triggers NLRP3 activation and results in IL-1β/IL-18 maturation and secretion. This figure was generated with BioRender.
Ijms 22 00330 g001
Figure 2. Triglyceride production in the liver. The chylomicrons bring fatty acids (mostly palmitate and oleate) to the liver, where they are used by GPAT (Glycerol-3-phosphate acyltransferase), AGPAT (1-Acylglycerol-3-phosphate-O-acyltransferase) and DGAT (Diacylglycerol-O-acyltransferase) enzymes to produce triglycerides. Alternatively, fatty acids can be synthesized de novo Figure 1. (Stearoyl-CoA desaturase-1) and ELOVL6 (Fatty acid elongase 6). Triglycerides are assembled into LDs (lipid droplets) and/or associated with apoB-100 (Apolipoprotein B-100) for secretion as VLDL (very low-density lipoproteins). This figure was generated with Servier Medical ART.
Figure 2. Triglyceride production in the liver. The chylomicrons bring fatty acids (mostly palmitate and oleate) to the liver, where they are used by GPAT (Glycerol-3-phosphate acyltransferase), AGPAT (1-Acylglycerol-3-phosphate-O-acyltransferase) and DGAT (Diacylglycerol-O-acyltransferase) enzymes to produce triglycerides. Alternatively, fatty acids can be synthesized de novo Figure 1. (Stearoyl-CoA desaturase-1) and ELOVL6 (Fatty acid elongase 6). Triglycerides are assembled into LDs (lipid droplets) and/or associated with apoB-100 (Apolipoprotein B-100) for secretion as VLDL (very low-density lipoproteins). This figure was generated with Servier Medical ART.
Ijms 22 00330 g002
Figure 3. Monounsaturated fatty acids have anti-inflammatory effects. SFAs (saturated fatty acids) activate TLR4 (Toll-Like receptor 4) to induce NF-κB (Nuclear factor-kappaB) nuclear translocatable 3. (NOD-like receptor family, pyrin domain containing 3) and pro-IL-1β (pro-Interleukin-1beta) expression, leading to IL-1β secretion and macrophage M1 polarization. MUFAs (monounsaturated fatty acids) can inhibit NF-κB and NLRP3 activation, respectively, through direct binding to GPR120 (G-protein coupled receptor 120) or PPARs (Peroxysome proliferator activated receptors), and through AMPK (AMP-activated protein kinase) phosphorylation. By inhibiting macrophage M1 polarization, MUFAs potentiate M2 polarization. This figure was generated with Servier Medical ART.
Figure 3. Monounsaturated fatty acids have anti-inflammatory effects. SFAs (saturated fatty acids) activate TLR4 (Toll-Like receptor 4) to induce NF-κB (Nuclear factor-kappaB) nuclear translocatable 3. (NOD-like receptor family, pyrin domain containing 3) and pro-IL-1β (pro-Interleukin-1beta) expression, leading to IL-1β secretion and macrophage M1 polarization. MUFAs (monounsaturated fatty acids) can inhibit NF-κB and NLRP3 activation, respectively, through direct binding to GPR120 (G-protein coupled receptor 120) or PPARs (Peroxysome proliferator activated receptors), and through AMPK (AMP-activated protein kinase) phosphorylation. By inhibiting macrophage M1 polarization, MUFAs potentiate M2 polarization. This figure was generated with Servier Medical ART.
Ijms 22 00330 g003
Table 1. Main outcome of human clinical trials involving MUFA-rich diets.
Table 1. Main outcome of human clinical trials involving MUFA-rich diets.
StudiesParticipantsDurationPopulationDietMain Outcome
Martin-Pelaez 2017 [120]12 volunteers, aged 46–67 years3 weeksHypercholesterolemic volunteers25 mg/day of virgin olive oilImproved cholesterolemia profile
De Filippis 2016 [126]153 individuals1 weekHealthy people, omnivore/vegetarian or veganMediterranean dietIncrease of Prevotella and Firmicutes within gut microbiota, improvement of atherosclerosis profile
Cesari 2018 [128]421 subjects, aged > 90 years12 monthsNAMediterranean dietDecreased risk factors for cardiovascular disease development
Schwingshackl 2015 [131]3106 participants>4 weeksNAOlive oil interventionsImprovement of inflammatory profile, CRP plasma level decreased
Paniagua 2007 [133]11 volunteers4 weeksDiabetic subjectMediterranean dietImproved insulin sensitivity and adiponectin secretion
Konstandinidou 2013 [136]90 participants, aged 20–50 years1 yearHealthy volunteersMediterranean dietProtective gene expression associated with inflammation improvement
Esposito 2004 [138]90 patients3 yearsDiabetic patientMediterranean dietDecreased insulin resistance, reduced plasma CRP and IL-6 level
Toscano 2020 [139]6 volunteers, aged 25–35 yearsNAHealthy volunteersOne oral emulsion of olive oilEnhanced M2 macrophage polarization, reduced proinflammatory profile
Abbreviations: NA (not applicable), CRP (C-reactive protein), IL-6 (Interleukin-6).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ravaut, G.; Légiot, A.; Bergeron, K.-F.; Mounier, C. Monounsaturated Fatty Acids in Obesity-Related Inflammation. Int. J. Mol. Sci. 2021, 22, 330. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010330

AMA Style

Ravaut G, Légiot A, Bergeron K-F, Mounier C. Monounsaturated Fatty Acids in Obesity-Related Inflammation. International Journal of Molecular Sciences. 2021; 22(1):330. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010330

Chicago/Turabian Style

Ravaut, Gaetan, Alexandre Légiot, Karl-F. Bergeron, and Catherine Mounier. 2021. "Monounsaturated Fatty Acids in Obesity-Related Inflammation" International Journal of Molecular Sciences 22, no. 1: 330. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010330

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop