You are on page 1of 8

European Journal of Pharmacology 785 (2016) 207–214

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Fatty acids and chronic low grade inflammation associated with


obesity and the metabolic syndrome
Aoife A. Cooke, Ruth M. Connaughton, Claire L. Lyons, Aoibheann M. McMorrow,
Helen M. Roche n
Nutrigenomics Group, UCD Conway Institute for Biomolecular and Biomedical Research & UCD Institute of Food & Health, School of Public Health, Phy-
siotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland

art ic l e i nf o a b s t r a c t

Article history: The metabolic syndrome is a group of obesity associated metabolic conditions that result in increased
Received 3 December 2015 risk of cardiovascular disease and type 2 diabetes. Global increases in obesity rates have led to an in-
Received in revised form crease in metabolic syndrome resulting in a demand for increased understanding of the mechanisms
2 April 2016
involved. This review examines the relationship between adipose tissue biology, lipid metabolism and
Accepted 11 April 2016
Available online 12 April 2016
chronic low grade inflammation relating to obesity and insulin resistance.
& 2016 Published by Elsevier B.V.
Keywords:
Metabolic syndrome
Obesity
Fatty acids
Inflammation

1. Introduction often associated with metabolic abnormalities including type


2 diabetes (T2D) and cardiovascular disease (CVD) (Vague, 1947).
First described in the 1920s as a combination of metabolic In 1988 Gerald Reaven used the term Syndrome X to describe a
disturbances, the scientific community has somewhat struggled to collection of symptoms including resistance to insulin-stimulated
define the Metabolic Syndrome (MetS) (Kylin, 1923). In the 1940s glucose uptake, glucose intolerance, hyperinsulinemia, increased
it was recognised that upper body adiposity was the type most very-low-density lipoprotein triglyceride or triacylglycerol (TAG),
decreased high-density lipoprotein (HDL) cholesterol and hy-
pertension. However, obesity was notably absent from this list
Abbreviations: AMPK, 5′ Adenosine Monophosphate-Activated Protein Kinase; (Reaven, 1988). The most recent formal definition for MetS avail-
ATGL, Adipose Triacylglycerol Lipase; BAT, Brown Adipose Tissue; BMI, Body Mass able identifies raised blood pressure, elevated TAG, lowered HDL
Index; CVD, Cardiovascular Disease; DAG, Diacylglycerol; DHA, Docosahexaenoic cholesterol, raised fasting glucose, and central obesity as its cri-
Acid; DIO, Diet Induced Obesity; EPA, Eicosapentaenoic Acid; ERK, Extra-Cellular
teria (Alberti et al., 2009). Individuals with MetS experience at
Signal Related Kinase; GLUT-4, Glucose Transporter Type  4; HDL, High-Density
Lipoprotein; HFD, High Fat Diet; HSL, Hormone Sensitive Lipase; Ig, Im- least a fivefold increased risk of developing T2D and a twofold
munoglobulin; IKK-β, Inhibitor of Nuclear Factor Kappa-B Kinase Subunit –Beta; IL, increased risk of CVD, with profound implications for morbidity
Interleukin; IL-1R-1, Interleukin-1 Receptor-1; IRS, Insulin Receptor Substrate; and quality of life (Eckel et al., 2010, 2005).
IκBα, Nuclear Factor Kappa Light Polypeptide Chain Gene Enhancer in B-Cells In- The global increase in obesity rates is of grave concern. As of
hibitor, Alpha; JAK, Janus Kinase; JNK, c-Jun-N-Terminal Protein Kinase; LPL, Lipo-
protein Lipase; MAPK, Mitogen Activated Protein Kinase; MCP-1, Monocyte Che-
2011, the number of individuals classified as overweight is greater
moattractant Protein  1; MetS, Metabolic Syndrome; MUFA, Monounsaturated than those classified as undernourished, with more than 1.7 billion
Fatty Acid; (n  3 LCPUFA), n  3 Long Chain Polyunsaturated Fatty Acid; NAFLD, overweight individuals worldwide (Chawla et al., 2011). Increasing
Non-Alcoholic Fatty Liver Disease; NFκB, Nuclear Factor Kappa Light Chain En- obesity rates are closely linked to greater prevalence of the MetS,
hancer of β-Cells; PKC, Protein Kinase C; PKC-θ, Protein Kinase C-θ; PKR, Protein
non-alcoholic fatty liver disease (NAFLD), T2D and CVD (Hong
Kinase R; PPAR-γ, Peroxisome Proliferator Activated Receptor γ; PUFA, Poly-
unsaturated Fatty Acid; SAT, Subcutaneous Adipose Tissue; SFA, Saturated Fatty et al., 2015; Sung et al., 2012; Targher and Arcaro, 2007; Targher,
Acid; SOCS-3, Suppressor of Cytokine Signalling-3; STAT-3, Signal Transducer and 2007; Targher et al., 2006). These statistics represent an enormous
Activator of Transcription-3; SVF, Stromal Vascular Fraction; T2D, Type 2 Diabetes; stress on global health services. It is imperative then that the
TAG, Triacylglycerol; Th-1, T Helper  1; TLR-4, Toll like Receptor-4; TNFR-1, Tu- mechanisms behind obesity and MetS are understood, in order to
mour Necrosis Factor Receptor – 1; TNF-α, Tumour Necrosis Factor – α; Treg, T-
Regulatory; VAT, Vascular Adipose Tissue; WAT, White Adipose Tissue
improve both preventative measures and outcomes. This review
n
Corresponding author. will focus on the inter-relationship between fatty acids and
E-mail address: helen.roche@ucd.ie (H.M. Roche). chronic low-grade inflammation in adipose tissue, which in turn

http://dx.doi.org/10.1016/j.ejphar.2016.04.021
0014-2999/& 2016 Published by Elsevier B.V.
208 A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214

Fig. 1. The inter-relationship between fatty acids, inflammation and insulin signalling - Cellular cross talk between macrophages and adipocytes. Fatty acids and their
derivatives have been shown to modulate inflammatory pathways in immune cells. This results in increased pro-inflammatory cytokine secretion from immune cells such as
macrophages and T cells which in turn act on adipocytes to disrupt insulin signalling. (This figure was prepared using the Servier medical art website http:// www.servier.fr/
servier-medical-art.).

modulates insulin signalling (Fig. 1). Given the importance of lipid (HSL) to release free fatty acids for β-oxidation (Lafontan and
metabolism in this paradigm, our perspective will focus on whe- Langin, 2009).
ther different fatty acids may mediate differential effects on adi- During persistent positive energy balance, WAT is forced to
pose tissue inflammation and insulin resistance, within the expand to facilitate increased TAG storage (Virtue and Vidal-Puig,
broader context of the MetS and T2D risk. 2010). Depending on its location, increased adipose tissue expan-
sion can result in considerable adipose tissue dysfunction, which is
characterised by decreased insulin sensitivity (Hotamisligil et al.,
2. Adipose tissue 1993), increased immune cell infiltration (Hotamisligil, 2006), fi-
brosis (Sun et al., 2013), hypoxia (Sun et al., 2013) and increased
Obesity is characterised by excess adipose tissue, which itself intracellular and systemic free fatty acid flux (Yu and Ginsberg,
can be categorised into two types: brown adipose tissue (BAT) and 2005). Expansion of WAT is peroxisome proliferator- activated
white adipose tissue (WAT) (Rosenwald and Wolfrum, 2014). BAT receptor-γ (PPAR-γ) dependent and occurs via an increase in adi-
is known to play a role in thermogenic processes (Nedergaard pocyte cell size (hypertrophy), cell number (hyperplasia) or both
et al., 2007). Until recently, BAT was thought to be present only in (Jo et al., 2009). It is now known that increased hyperplasia is
new-born humans. However, recent work has suggested a role for linked to a more favourable phenotype, which likely reflects less
BAT in thermoregulatory processes in adults (Nedergaard et al., adipose tissue inflammation (Lundgren et al., 2007).
2007). In contrast, WAT plays a significant role in regulating en- In addition to cell morphology, distribution of excess adipose
ergy metabolism and storing excess energy as TAG (Schweiger and tissue has a significant impact on pathology, wherein increased
Schreiber, 2006). visceral adipose tissue (VAT) to a greater extent than subcutaneous
Adipocytes represent the primary cellular component of WAT. adipose tissue (SAT) is associated with MetS development
The lipid stored within adipocytes is in constant flux and readily (Després and Lemieux, 2006). Additionally, SAT is rich in pre-
adapts to fluctuating nutritional demand (Lafontan and Langin, adipocytes prone to rapid replication, which increases its buffering
2009). In the postprandial state, lipoprotein lipase (LPL), located capacity (Tchkonia et al., 2005). Vidal-Puig and colleagues hy-
within the endothelial lining of blood vessels hydrolyses circulat- pothesised that individuals reach a limit of SAT expansion, after
ing TAG. This process releases free fatty acids which are taken up which excess TAG can no longer be facilitated within the sub-
by adipocytes and re-esterified to generate TAG for storage. In cutaneous region (Virtue and Vidal-Puig, 2008). This inability of
contrast, in times of energy deficit or increased energy require- SAT to buffer macronutrient surplus promotes lipotoxicity,
ments, TAG undergoes lipolysis, wherein it is hydrolysed by adi- wherein excess energy is directed towards the VAT depot as well
pose triacylglycerol lipase (ATGL) and hormone sensitive lipase as to non-adipose locations, such as skeletal muscle, the liver and
A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214 209

the pancreas (Virtue and Vidal-Puig, 2008). This concept is known T cells are necessary for the activation, differentiation and mi-
as overspill and is detrimental to organ signalling and function- gration of macrophages (Nishimura et al., 2009). Adipose tissue
ality owing to the fact that increased lipid uptake is not matched macrophage content increases from 10% to 15% in lean adipose to
by increased β-oxidation (Tumova et al., 2015). 45–60% within obese adipose (Weisberg et al., 2003). In a micro-
Furthermore, impaired insulin signalling reduces postprandial array analysis of adipose tissue, a third of the transcripts that were
inhibition of lipolysis in response to insulin (Basu et al., 2001). positively correlated with body mass belonged to macrophages.
Thus, insulin resistance is associated with increased and persistent Adipocyte area in multiple adipose depots is a strong positive
free fatty acid flux from the adipose tissue into the circulation. predictor of F4/80 þ macrophage number, a result also observed
Visceral adipocytes in particular have been shown to be the pri- with increasing body mass index (BMI) in human SAT (Weisberg
mary contributors of free fatty acids to circulation, attributable to et al., 2003). For example, CD45.2 mice transplanted with CD45.1
higher basal lipolytic activity in VAT relative to SAT (Arner, 1995). bone marrow, displayed 80% of F4/80 þ macrophages from the
Furthermore, obese subjects have demonstrated reduced ability to CD45.1 donor within adipose tissue following 6 weeks of HFD,
clear free fatty acids following a high-fat meal (Lewis and O’Meara, demonstrating the influx of immune cells from the periphery
1990). Thus, adipose tissue dysfunction gives rise to increased (Weisberg et al., 2003). Chemokines, such as monocyte chemoat-
systemic free fatty acids owing to limited SAT buffering capacity, tractant protein 1 (MCP-1) play a role in the recruitment of mac-
increased endogenous free fatty acid release and decreased ability rophages to the adipose tissue in both HFD and genetic models of
to clear exogenous free fatty acids. obesity (Kamei et al., 2006). Once the immune cells populate the
adipose tissue, their secreted cytokines attract more immune cells
into the local area. Resident macrophages undergo a phenotypic
3. Adipose tissue immune cell infiltration switch from the anti-inflammatory M2 to the pro-inflammatory
M1 adipose tissue macrophages, through cytokine and immune
While adipocytes are the largest component of adipose tissue, cell action (Lumeng et al., 2008). Eosinophils maintain macro-
the stromal vascular fraction (SVF), although smaller, plays a key phages in an M2 state in an IL-4/IL-13 dependent manner but their
role in adipose function. The SVF of the adipose is where immune numbers decrease with increasing adiposity (Wu et al., 2011),
cells, such as macrophages (10–15%), CD3 þ T cells (14%; 94% of therefore an M1 phenotype is the dominant immunophenotype in
which are CD4 þ or CD8 þ ), mast cells, eosinophils (4–5%) and B obesity. Eosinophil-deficient mice display obesity with glucose
cells reside (Nishimura et al., 2009; Weisberg et al., 2003; Winer intolerance, indicating their role in metabolic homeostasis. The
et al., 2011; Wu et al., 2011). In the lean insulin sensitive state, innate immune mast cell has been implicated in DIO as their
anti-inflammatory T regulatory (Treg) and M2 macrophages are numbers increase with obesity, inflammation and insulin re-
the prominent cell types. Resident immune cells have house- sistance (Winer et al., 2011). B cells increase inflammatory med-
keeping roles, including, but not limited to, clearance of apoptotic iators, such as secretion of tumor necrosis factor-α (TNF-α); in-
adipocytes, extracellular remodeling, angiogenesis and adipogen- fluence CD8 þ and T helper-1 (Th1) T cell activation; and produce
esis (Schipper et al., 2012). immunoglobulin (Ig) antibodies, which are involved in macro-
As the VAT expands, immune cells infiltrate the tissue to form phage pro-inflammatory polarization. B cell knockout models are
crown-like structures around necrotizing adipocytes, in order to protected against disrupted glucose homeostasis despite equiva-
clear cellular debris and perform an adaptive function (Nishimura lent levels of obesity compared to their control counterparts
et al., 2009; Xu et al., 2003). In addition the immune cell compo- (Winer et al., 2011). This demonstrates that both the innate and
sition and phenotype is altered in the obese setting contributing to adaptive immune systems play a pivotal role in the development
adipose tissue dysfunction and insulin resistance. Obesity is as- of insulin resistance and MetS, mainly through the release of pro-
sociated with a state of chronic low-grade inflammation and this inflammatory cytokines.
provides signals that drive a pro-inflammatory and persistent
immune response (which is discussed in the next section). In ob-
ese adipose, immune cells are under increased demand associated 4. Localised inflammatory mediators
with adipose expansion. Adipose tissue macrophages and mast cell
localization around microvessels provide angiogenic support for Free fatty acids arising from inflamed adipose tissue activate a
adipose tissue development and leukocyte infiltration into the host of intracellular signalling cascades that sense and commu-
expanding depot. An increase in CD31 þ endothelial cells is also nicate adipocyte nutrient overload. This occurs through the acti-
observed in dietary induced obesity (DIO) to facilitate increased vation of the c-Jun N-terminal protein kinases (JNK), inhibitor of
angiogenesis (Nishimura et al., 2009). Extracellular remodeling is nuclear factor kappa-B kinase subunit beta (IKK-β), and protein
undertaken by the mast cells to facilitate growth (Liu et al., 2009). kinase R (PKR) signalling pathways, all of which are involved in the
The influx of immune cells begins with CD8 þ T cells as early as inflammatory response (Hirosumi et al., 2002; Nakamura et al.,
4 weeks on a high-fat diet (HFD), followed by macrophage re- 2010; Yuan et al., 2001). Activation of these signalling pathways
cruitment, with a decrease in CD4 þhelper and Treg cells occurring results in inhibition of the insulin signalling pathway by serine
at later time points (Nishimura et al., 2009). Treatment with a CD8 phosphorylation of insulin receptor substrate (IRS) proteins
specific antibody, in conjunction with a HFD, results in reduced M1 (Aguirre et al., 2002; Hotamisligil et al., 1996). Furthermore, JNK
polarization with no change to M2 or CD4 þ T cell numbers; de- and IKK-β activation initiates a paracrine inflammatory response
monstrating the interplay between the different immune cell involving cytokines, such as TNF-α and IL-1β. These can broadcast
types. Amelioration of the inflammatory response and insulin re- intracellular conditions to other adipocytes and importantly to
sistant state is observed following inhibition of CD8 þ T cell action, resident leukocytes in the immediate microenvironment (Ode-
even in a pre-established inflammatory state, indicating T cell in- gaard and Chawla, 2013).
volvement in both the propagation and maintenance of adipose TNF-α, the first pro-inflammatory cytokine linked to obesity-
tissue inflammation (Nishimura et al., 2009). When reduction of associated insulin resistance activates a number of serine kinases,
Treg cells is induced, insulin levels increase (Feuerer et al., 2009). such as JNK and IKKβ, leading to serine phosphorylation of IRS-1
Treg cells produce large amounts of the anti-inflammatory cyto- (Gao et al., 2002; Hirosumi et al., 2002; Hotamisligil et al., 1993).
kine interleukin (IL)  10, which itself has insulin sensitizing effects This is in contrast to tyrosine phosphorylation of IRS-1 in an in-
(Lumeng et al., 2008). sulin sensitive state. Moreover, TNF-α, in conjunction with IL-6,
210 A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214

increases the secretion of suppressor of cytokine signalling-3 chemokines such as TNF-α and IL-6; and in turn their detrimental
(SOCS-3) proteins, which bind to insulin receptors and attenuate biological effect on target cells, such as adipocytes (Hong et al.,
phosphorylation of IRS proteins thus impairing insulin signalling 2009). Other intracellular events affected by IL-10 involve nuclear
(Emanuelli et al., 2000; Ueki et al., 2004). Demonstrating the im- translocation of signal transducer and activator of transcription 3
portant role TNF-α plays in obesity-induced insulin resistance, (STAT3) and suppressor of cytokine signalling 3 (SOCS3). Induction
obese rodents which had been administered a TNF-α neutralising of SOCS3 by IL-10 inhibits signalling that is initiated by IL-6 via the
antibody exhibited reduced hyperinsulinemia (Hotamisligil et al., IL-6 signal transducer (gp130) (Niemand et al., 2003). In adipocyte
1993). Additionally, whole body deletion of TNF-α or its corre- cell culture models, pre-treatment with IL-10 attenuated the in-
sponding receptor tumour necrosis factor receptor-1 (TNFR1) sulin desensitizing effect of macrophage conditioned media, thus
partially protects mice from obesity induced insulin resistance by modulating or attenuating inflammation, IL-10 may potentiate
(Uysal et al., 1997). insulin sensitivity (Oliver et al., 2012).
With respect to IL-6, there is conflicting data regarding its role
in insulin resistance (Pedersen and Febbraio, 2007). IL-6 is se-
creted by adipose tissue, as well as skeletal muscle and liver, sig- 5. Dysregulated fatty acid metabolism and metabolic
nalling via the janus kinase (JAK)/signal transducer and activator homeostasis
of transcription (STAT) and mitogen activated protein kinase
(MAPK) pathways. In adipose tissue, IL-6 reduces glucose uptake The relationship between obesity induced inflammation and
in adipocytes, by reducing insulin-stimulated tyrosine phosphor- lipid dysregulation is bidirectional. It is important to acknowledge
ylation of IRS-1 while concurrently promoting serine phosphor- that several components of dysregulated lipid metabolism and
ylation of IRS-1 (Fasshauer et al., 2003; Rotter et al., 2003). How- chronic low-grade inflammation play a causal role in the patho-
ever in skeletal muscle the role played by IL-6 can vary. IL-6 en- genesis and progression of insulin resistance. However, several
hances fat oxidation in skeletal muscle via activation of 5′ ade- abnormalities of lipid metabolism associated with insulin re-
nosine monophosphate activated protein kinase (AMPK) (Carey sistance may simply reflect the insulin resistant state. On the
et al., 2006). Furthermore, Wolsk et al. have demonstrated that IL- causality side, Schulman's team have demonstrated very elegantly
6 enhances fat metabolism, specifically lipolysis, in skeletal muscle the concept of lipotoxicity, wherein overspill of free fatty acids
but not in adipose (Wolsk et al., 2010). In contrast, other studies leading to ectopic lipid accumulation in skeletal muscle and the
have associated skeletal muscle wastage with chronic systemic IL- liver, can lead to insulin resistance (Kim et al., 2000; Nagle et al.,
6 levels (Fearon et al., 2012). Thus any positive effects of IL-6 in 2007; Samuel and Shulman, 2012). As reviewed extensively, there
skeletal muscle are associated with transient production, rather are two principle routes wherein fatty acids/lipids directly impede
than chronic exposure (Munoz-Canoves et al., 2013). insulin signalling (Johnson and Olefsky, 2013). Firstly, circulating
IL-1 are a pro-inflammatory family of cytokines that play a key free fatty acids can activate cell-signalling pathways, to impede
role in insulin resistance (Osborn and Olefsky, 2012). Consisting of insulin action (Glass and Olefsky, 2012). Secondly, imbalanced lipid
two major proteins, IL-1α and IL-1β, both display similar biological metabolism and/or metabolic derivatives thereof, lead to accu-
properties but minimal sequence homology (Dinarello, 2009). mulation of intracellular lipid products, including diacylglycerol
Recently, studies have established that IL-1β requires the activa- (DAG), in both hepatic and skeletal tissues wherein lipotoxicity
tion of the NLRP3 inflammasome, a protein complex, to produce causes insulin resistance (Jornayvaz and Shulman, 2012). DAG in
its biologically active mature form (Tschopp and Schroder, 2010). turn activates protein kinase Cε (PKC), which then impedes tyr-
Activation of the NLRP3 inflammasome leads to the cleavage of osine phosphorylation of IRS 1 and  2 to precipitate the hepatic
pro-caspase to caspase-1, which in turn cleaves the inactive pro-IL- insulin resistance that underlies NAFLD associated with obesity,
1β to the mature, biologically potent IL-1β (Koenen et al., 2011). MetS and T2D (Jornayvaz and Shulman, 2012). Taken together,
This is distinct to IL-1α, which is produced in its biologically active these routes demonstrate that lipotoxicity plays a causal role in
form (Dinarello, 2009). IL-1β is considerably the more potent in- the MetS.
flammatory cytokine and is involved in many inflammatory dis- Ceramides are another critical lipid intermediate derived from
eases by initiating and potentiating immune and inflammatory saturated fatty acid (SFA), which impede AKT, the activity of which
responses (Dinarello, 2009). In 3T3L1 adipocytes, IL-1β decreases is crucial to insulin signalling (Holland et al., 2011; Powell et al.,
glucose transport via inhibition of IRS-1 expression, by activating 2004). There is also strong evidence that an excessive fatty acid
the inflammatory kinases MAPK and extra-cellular signal related insult upon the mitochondria overwhelm their capacity for β-
kinase (ERK) (Jager et al., 2007). Furthermore, treatment of adi- oxidation, which generates partially oxidised acylcarnitines that
pocytes with IL-1β reduced insulin-stimulated glucose uptake by also have adverse effects on insulin signalling (Muoio and Neufer,
decreasing expression and translocation of glucose transporter 2012). Hepatocellular DAG and acylcarnitine content probably re-
type-4 (GLUT-4) to the plasma membrane (He et al., 2006). HFD flects the balance between lipogenesis and mitochondrial fatty
feeding induces NLRP3 inflammasome activation, via cleavage and acid oxidation, the combination of which affects the degree of
activation of caspase-1, resulting in secretion of IL-1β (Martinon hepatic insulin resistance. Furthermore, Holland and colleagues
et al., 2002; Reynolds et al., 2012). Correspondingly, mice lacking showed the interconnections between ceramide biology and in-
the IL-1 receptor 1(IL-1R1) are protected from HFD-induced glu- flammation. Activation of toll like receptor-4 (TLR4)-dependent
cose intolerance (De Roos et al., 2009; McGillicuddy et al., 2011). inflammatory pathways by SFA augments ceramide biosynthesis
Conversely anti-inflammatory cytokines, such as IL-10, are also (Holland et al., 2011). This interplay between fatty acids and in-
secreted by immune cells and may have insulin sensitising po- flammatory processes occurs at a number of other metabolic-in-
tential. IL-10, produced by monocytes, M2 adipose tissue macro- flammatory hubs, as discussed in the next section.
phages, dendritic cells, T cells and B cells, signals via the IL-10
receptor activating the JAK/STAT pathway (Saraiva and O’Garra,
2010). Protein levels of NFκB inhibitor-α (IκBα), an inhibitor of 6. Different fatty acids ¼different metabolic effects?
nuclear factor kappa light chain enhancer of B cells (NFκB), are
preserved following treatment with IL-10, attenuating NFκB A core theme of our recent research has addressed whether
binding as well as TNF-α levels (Shames et al., 1998). Furthermore different dietary fatty acids may have a variable impact on meta-
IL-10 is a potent inhibitor of pro-inflammatory cytokines and bolic homeostasis. We have some interesting insights; however
A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214 211

the full nature of the inter-relationship between fatty acid, low adverse effects of high SFA diets as reviewed in detail previously
grade chronic inflammation and obesity related insulin resistance (Phillips et al., 2008). However we do not yet fully understand if/
is not fully understood. Fatty acids are ubiquitous in the diet, how different fatty acids mediate their adverse effect. Also it is
distributed throughout the body and play a key role in lipid me- unlikely that this effect can be ascribed only to IL-1β mediated
tabolism. Fatty acids may be present in their free form, as free fatty inflammation and insulin resistance. For example, it has been
acids. However, they are more abundantly represented in several shown that overfeeding SFA and PUFA has different effects on
complex lipid forms, such as TAG, phospholipids or as cholesterol hepatic and VAT lipid accumulation (Rosqvist et al., 2014). Also
esters. Exogenously these fatty acid-enriched complex lipid spe- different dietary lipids may affect skeletal muscle fatty acid
cies are present in lipid containing food. Dietary fat is principally handling (Jans et al., 2012b).
composed of TAG, with lesser amounts of phospholipids and Other recent and exciting developments in this field include
cholesterol esters. Endogenous lipid metabolism is principally the discovery of novel endogenous lipid species which have anti-
determined by the liver, with interactions between adipose tissue diabetic and anti-inflammatory effects. Yore et al. demonstrated
and skeletal muscle, the former a storage site representing an enhanced adipose lipogenesis, greater glucose tolerance and anti-
important endogenous source of fatty acids and the latter being a inflammatory properties, in spite of obesity. This was attributable
catabolic site. Lipids play a fundamental role in intra-organ and to elevated concentrations of a novel class of lipids in GLUT-4
cellular biology as lipid storage molecules (TAG) and cellular transgenic animals. These novel lipids were characterised by
membrane components (phospholipids). Fatty acids can also act as branched ester linkage between a fatty acid and a hydroxy-fatty
potent intracellular second messenger signalling molecules acid and are referred to as FAHFAs. The most dramatically upre-
wherein they may be present in DAG ceramides and other lipid gulated FAHFA in GLUT-4 transgenic mice was palmitic-acid-hy-
derivatives. droxyl-stearic acid (PAHSA). PAHSAs are synthesised in vivo and
There is a large body of evidence that shows that obesity per se, regulated by both fasting and HFD feeding. Importantly, PAHSA
induced by HFD, instigates an inflammatory cascade that impedes levels were lower in serum and adipose tissue of insulin resistant
insulin signalling, as previously reviewed (Murphy et al., 2014). For subjects. Yore et al. demonstrated that PAHSAs had multiple ef-
example, Reynolds et al. demonstrated that this priming and ac- fects that resulted in improved glucose-insulin homeostasis, which
tivation of inflammation via TLR4 and NLRP3 by dietary SFA is suggests that restoration of PAHSA levels in patients with insulin
evident in bone marrow dendritic cells, in a manner that is con- resistance may have potential beneficial effects on metabolism. In
current with that of adipose tissue inflammation activation (Rey- animal studies, oral administration of PAHSA to insulin-resistant
nolds et al., 2012). In the case of the liver, this paradigm is not mice on a HFD improved glucose tolerance (Yore et al., 2014). In-
necessarily correct. Galbo et al. demonstrated that TLR-4 mediated terestingly PAHSA signalled via G protein-coupled receptor 120
hepatic ceramide synthesis signalling is not directly required for (GPR120). GPR120 was also identified as an important signalling
SFA or polyunsaturated fatty acid (PUFA) induced hepatic insulin sensor/receptor that mediated the anti-inflammatory, insulin-
resistance. It is of note that this study investigated the short-term sensitising effects of the two n-3 long chain PUFA ((n  3 LCPUFA))
(3 days) impact of dietary SFA versus PUFA, wherein both SFA and eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) (Oh
PUFA-HFD led to hepatic DAG accumulation, PKCε activation and et al., 2010). However, in this context the dietary doses of (n  3
impaired insulin signaling (Galbo et al., 2013). LCPUFA) were supra-physiological and not necessarily translatable
Our recent work has focused on SFA mediated priming and to humans. Nevertheless, GPR120 is an important control point in
activation of the NLRP3 inflammasome resulting in IL-1β-medi- the integration of anti-inflammatory insulin sensitisation and the
ated adipose tissue inflammation and insulin resistance. Interest- focus of on-going research as a therapeutic target within the
ingly replacement of SFA for monounsaturated fatty acid (MUFA) context of obesity related inflammation and insulin resistance in
in HFD reduced adipose tissue pro-IL-1β priming and attenuated T2D (Oh et al., 2014). There are several other fatty acid sensitive,
insulin resistance in an AMPK mediated fashion (Finucane et al., molecular targets at the inter-section of metabolic-inflammation,
2015). Despite being obese, MUFA-HFD-fed mice displayed im- including ChREBP, NF-κB, PPARγ which for the sake of brevity are
proved insulin sensitivity coincident with reduced pro-IL-1β not discussed here, but are well documented (Benhamed et al.,
priming, attenuated adipose IL-1β secretion and sustained adipose 2012; Herman et al., 2012; Murphy et al., 2014; Osborn and
activation of the nutritional sensor AMPK. Adipose tissue mor- Olefsky, 2012). Whilst we have focused on fatty acids; it is im-
phology was markedly different between diets, which we suspect portant to note that there are additional non-lipid nutrients with
had a very significant physiological impact upon the whole body anti-inflammatory potential, such as lycopene, ECGC, vitamin C
insulin and glucose homeostasis. We speculate that greater acti- and resveratrol, as reviewed in detail elsewhere (Connaughton
vation of IL-1β in the adipose milieu by SFA affected the adipo- et al., 2016; Schrauwen and Timmers, 2014).
genic potential of preadipocytes creating a hypertrophic adipose
morphology. In contrast despite equal adipose tissue weight, the
MUFA-HFD mice displayed hyperplasic adipose tissue with pre- 7. Translational perspectives – efficacy of dietary fatty acid
served insulin sensitivity, which we attributed to attenuated modulation in humans?
NLRP3 mediated IL-1β activation in adipose. Whilst this work is
very interesting in terms of the relative impact of SFA versus In the acute setting, SFA are more potent than MUFA or PUFA in
MUFA, we are as yet unsure the extent to which this paradigm inducing an insulin resistant or glucose intolerant state (Jans et al.,
relates to man. Previous work from our group has demonstrated 2012a; Xiao et al., 2006). Fatty acid challenges rapidly induce an
that increasing SFA but not MUFA induced insulin resistance in insulin resistant state, which is mediated in a number of ways.
T2D subjects suggesting that a HFD rich in MUFA are not as de- These include acylcarnitine accumulation, beta-cell dysfunction,
leterious. Furthermore we examined data from subjects with altered intra-myocellular lipid partitioning and oxidation, reduced
MetS, which demonstrated that when subjects with a habitual non-oxidative glucose disposal, PKCθ activation and DAG enrich-
high MUFA intake underwent a MUFA dietary intervention they ment in skeletal muscle, as well as inducing a postprandial pro-
had improved insulin response to glucose and improved first- inflammatory state (Cruz-Teno et al., 2012; Jans et al., 2012a;
phase insulin response. This was in contrast to high-SFA habitual Nowotny et al., 2013; Stephens et al., 2014; Wuesten et al., 2005).
consumers who experienced no such improvement (Finucane However the effects of chronic dietary fat modifications are more
et al., 2015). The prospective evidence is strong with respect to the complex and the data are mixed, as reviewed extensively
212 A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214

elsewhere (Murphy et al., 2014). Briefly, there is no doubt that Basu, A., Basu, R., Shah, P., Vella, A., Rizza, R.A., Jensen, M.D., 2001. Systemic and
obesity as a result of HFD, particularly if enriched in SFA, promotes regional free fatty acid metabolism in type 2 diabetes. Am. J. Physiol. En-
docrinol. Metab. 280, E1000–E1006.
a pro-inflammatory, insulin resistant state characteristic of MetS, Benhamed, F., Denechaud, P.D., Lemoine, M., Robichon, C., Moldes, M., Bertrand-
with a greater risk of T2D (Minihane et al., 2015). Indeed recent Michel, J., Ratziu, V., Serfaty, L., Housset, C., Capeau, J., Girard, J., Guillou, H.,
mechanistic studies have provided very strong evidence in relation Postic, C., 2012. The lipogenic transcription factor ChREBP dissociates hepatic
steatosis from insulin resistance in mice and humans. J. Clin. Investig. 122,
to molecular processes involved. Nevertheless, even if this para- 2176–2194. http://dx.doi.org/10.1172/JCI41636.
digm is true, simply removing dietary SFA, with or without re- Carey, A.L., Steinberg, G.R., Macaulay, S.L., Thomas, W.G., Holmes, A.G., Ramm, G.,
placement with other fatty acid (e.g. MUFA, (n  3 LCPUFA)), does Prelovsek, O., Hohnen-Behrens, C., Watt, M.J., James, D.E., Kemp, B.E., Pedersen,
B.K., Febbraio, M.A., 2006. Interleukin-6 increases insulin-stimulated glucose
not necessarily resolve the pro-inflammatory, insulin resistant
disposal in humans and glucose uptake and fatty acid oxidation in vitro via
state. A recent review highlighted the challenges arising from the AMP-activated protein kinase. Diabetes 55, 2688–2697. http://dx.doi.org/
inconsistencies within the scientific evidence to date (Minihane 10.2337/db05-1404.
Chawla, A., Nguyen, K.D., Goh, Y.P.S., 2011. Macrophage-mediated inflammation in
et al., 2015). All diet and lifestyle intervention studies are chal-
metabolic disease. Nat. Rev. Immunol. 11, 738–749. http://dx.doi.org/10.1038/
lenged by apparent responders versus non-responders, wherein nri3071.
typically 30–40% of individuals ‘respond’ to the intervention. This Connaughton, R.M., McMorrow, A.M., McGillicuddy, F.C., Lithander, F.E., Roche, H.
apparent difference in treatment efficacy goes beyond simple M., 2016. Impact of anti-inflammatory nutrients on obesity-associated meta-
bolic-inflammation from childhood through to adulthood. Proc. Nutr. Soc.,
compliance and may reflect different genetic or phenotypic char- 1–10. http://dx.doi.org/10.1017/S0029665116000070.
acteristics. For example, Yubero-Serrano et al. demonstrated that Cruz-Teno, C., Pérez-Martínez, P., Delgado-Lista, J., Yubero-Serrano, E.M., García-
MetS subjects with insulin resistance, displaying more extensive Ríos, A., Marín, C., Gómez, P., Jiménez-Gómez, Y., Camargo, A., Rodríguez-Can-
talejo, F., Malagón, M.M., Pérez-Jiménez, F., Roche, H.M., López-Miranda, J.,
metabolic complications, responded more effectively to dietary fat 2012. Dietary fat modifies the postprandial inflammatory state in subjects with
modification, wherein dietary SFA were reduced by replacement metabolic syndrome: the LIPGENE study. Mol. Nutr. Food Res. 56, 854–865.
with MUFA and (n  3 LCPUFA) (Yubero-Serrano et al., 2015). http://dx.doi.org/10.1002/mnfr.201200096.
De Roos, B., Rungapamestry, V., Ross, K., Rucklidge, G., Reid, M., Duncan, G., Horgan,
Conversely, MetS subjects without insulin resistance were more G., Toomey, S., Browne, J., Loscher, C.E., Mills, K.H.G., Roche, H.M., 2009. At-
sensitive to the detrimental effects of dietary SFA intake. Thus the tenuation of inflammation and cellular stress-related pathways maintains in-
metabolic phenotype (or metabotypes) of subjects may modulate sulin sensitivity in obese type I interleukin-1 receptor knockout mice on a high-
fat diet. Proteomics 9, 3244–3256.
response to the quantity and quality of dietary fatty acid on MetS
Després, J.-P., Lemieux, I., 2006. Abdominal obesity and metabolic syndrome. Nat-
risk factors; which suggests that targeted or personalized dietary ure 444, 881–887. http://dx.doi.org/10.1038/nature05488.
therapies based on distinct metabotypes may be more effective Dinarello, C.A., 2009. Immunological and inflammatory functions of the inter-
leukin-1 family. Annu. Rev. Immunol. 27, 519–550. http://dx.doi.org/10.1146/
(Phillips et al., 2012). This variation in responsiveness is the focus
annurev.immunol.021908.132612.
of current research, as we attempt to address the knowledge gap Eckel, R.H., Alberti, K., Grundy, S.M., Zimmet, P.Z., 2010. The metabolic syndrome.
with respect to those genotype-phenotype combinations that de- Lancet 375, 181–183. http://dx.doi.org/10.1016/S0140-6736(09)61794-3.
termine sensitivity to fatty acids; both in terms of identifying risk Eckel, R.H., Grundy, S.M., Zimmet, P.Z., 2005. The metabolic syndrome. Lancet 365,
1415–1428. http://dx.doi.org/10.1016/S0140-6736(05)66378-7.
and enhancing therapeutic efficacy. Furthermore, systemic bio- Emanuelli, B., Peraldi, P., Filloux, C., Sawka-Verhelle, D., Hilton, D., Van Obberghen,
markers of inflammation do not necessarily reflect the potent E., 2000. SOCS-3 is an insulin-induced negative regulator of insulin signaling. J.
biological effects of inflammatory mediators in the metabolic tis- Biol. Chem. 275, 15985–15991.
Fasshauer, M., Klein, J., Lossner, U., Paschke, R., 2003. Interleukin (IL)  6 mRNA
sues, including adipose tissue, liver and skeletal muscle. Clearly, expression is stimulated by insulin, isoproterenol, tumour necrosis factor alpha,
there are several gaps in knowledge relating to the inter-re- growth hormone, and IL-6 in 3T3-L1 adipocytes. Horm. Metab. Res. 35,
lationships between dietary fatty acid, inflammation and meta- 147–152. http://dx.doi.org/10.1055/s-2003-39075.
Fearon, K.C.H., Glass, D.J., Guttridge, D.C., 2012. Cancer cachexia: mediators, sig-
bolism. These need to be resolved given the continued risks arising naling, and metabolic pathways. Cell Metab. 16, 153–166. http://dx.doi.org/
from excessive dietary fat intake and escalating obesity rates with 10.1016/j.cmet.2012.06.011.
concomitant risk of NALFD, MetS, T2D and CVD. Feuerer, M., Herrero, L., Cipolletta, D., Naaz, A., Wong, J., Nayer, A., Lee, J., Goldfine,
A.B., Benoist, C., Shoelson, S., Mathis, D., 2009. Lean, but not obese, fat is en-
riched for a unique population of regulatory T cells that affect metabolic
parameters. Nat. Med. 15, 930–939. http://dx.doi.org/10.1038/nm.2002.
Acknowledgements 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., O’Reilly, M.E., Tierney, A.C.,
Morine, M.J., Alcala-Diaz, J.F., Lopez-Miranda, J., O’Connor, D.P., O’Neill, L.A.,
AAC, RMC, CLL and HMR are supported by Science Foundation McGillicuddy, F.C., Roche, H.M., 2015. Monounsaturated fatty acid–enriched
Ireland Principal Investigator Programme (11/PI/1119). AMM and high-fat diets impede adipose NLRP3 inflammasome–mediated IL-1β secretion
and insulin resistance despite obesity. Diabetes . http://dx.doi.org/10.2337/
HMR were funded by the Irish Department of Agriculture, Food
db14-1098, db141098.
and the Marine, NUTRIMAL Programme (14F 882). RMC and AMM Galbo, T., Perry, R.J., Jurczak, M.J., Camporez, J.-P.G., Alves, T.C., Kahn, M., Guigni, B.
were funded by the National Children's Research Centre, Crumlin, A., Serr, J., Zhang, D., Bhanot, S., Samuel, V.T., Shulman, G.I., 2013. Saturated and
Ireland (Grant number: B/11/1). unsaturated fat induce hepatic insulin resistance independently of TLR-4 sig-
naling and ceramide synthesis in vivo. Proc. Natl. Acad. Sci. USA 110,
12780–12785. http://dx.doi.org/10.1073/pnas.1311176110.
Gao, Z., Hwang, D., Bataille, F., Lefevre, M., York, D., Quon, M.J., Ye, J., 2002. Serine
References phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase
complex. J. Biol. Chem. 277, 48115–48121. http://dx.doi.org/10.1074/jbc.
M209459200.
Aguirre, V., Werner, E.D., Giraud, J., Lee, Y.H., Shoelson, S.E., White, M.F., 2002. Glass, C.K., Olefsky, J.M., 2012. Inflammation and lipid signaling in the etiology of
Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions insulin resistance. Cell Metab. 15, 635–645. http://dx.doi.org/10.1016/j.
with the insulin receptor and inhibits insulin action. J. Biol. Chem. 277, cmet.2012.04.001.
1531–1537. http://dx.doi.org/10.1074/jbc.M101521200. He, J., Usui, I., Ishizuka, K., Kanatani, Y., Hiratani, K., Iwata, M., Bukhari, A., Haruta, T.,
Alberti, K.G.M.M., Eckel, R.H., Grundy, S.M., Zimmet, P.Z., Cleeman, J.I., Donato, K.A., Sasaoka, T., Kobayashi, M., 2006. Interleukin-1alpha inhibits insulin signaling
Fruchart, J.-C., James, W.P.T., Loria, C.M., Smith, S.C., 2009. Harmonizing the with phosphorylating insulin receptor substrate-1 on serine residues in 3T3-L1
metabolic syndrome: a joint interim statement of the International Diabetes adipocytes. Mol. Endocrinol. 20, 114–124. http://dx.doi.org/10.1210/me.2005–
Federation Task Force on Epidemiology and Prevention; National Heart, Lung, 0107.
and Blood Institute; American Heart Association; World Heart Federation; In- Herman, M.A., Peroni, O.D., Villoria, J., Schön, M.R., Abumrad, N.A., Blüher, M., Klein,
ternational Atherosclerosis Society; and International Association for the Study S., Kahn, B.B., 2012. A novel ChREBP isoform in adipose tissue regulates sys-
of Obesity. Circulation 120, 1640–1645. http://dx.doi.org/10.1161/ temic glucose metabolism. Nature 484, 333–338. http://dx.doi.org/10.1038/
CIRCULATIONAHA.109.192644. nature10986.
Arner, P., 1995. Differences in lipolysis between human subcutaneous and omental Hirosumi, J., Tuncman, G., Chang, L., Görgün, C.Z., Uysal, K.T., Maeda, K., Karin, M.,
adipose tissues. Ann. Med. 27, 435–438. http://dx.doi.org/10.3109/ Hotamisligil, G.S., 2002. A central role for JNK in obesity and insulin resistance.
07853899709002451. Nature 420, 333–336. http://dx.doi.org/10.1038/nature01137.
A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214 213

Holland, W.L., Bikman, B.T., Wang, L., Yuguang, G., Sargent, K.M., Bulchand, S., 10.2337/db10-1278.
Knotts, T.A., Shui, G., Clegg, D.J., Wenk, M.R., Pagliassotti, M.J., Scherer, P.E., Minihane, A.M., Vinoy, S., Russell, W.R., Baka, A., Roche, H.M., Tuohy, K.M., Teeling, J.
Summers, S.A., 2011. Lipid-induced insulin resistance mediated by the proin- L., Blaak, E.E., Fenech, M., Vauzour, D., McArdle, H.J., Kremer, B.H.A., Sterkman,
flammatory receptor TLR4 requires saturated fatty acid – induced ceramide L., Vafeiadou, K., Benedetti, M.M., Williams, C.M., Calder, P.C., 2015. Low-grade
biosynthesis in mice. J. Clin. Investig., 121. http://dx.doi.org/10.1172/ inflammation, diet composition and health: current research evidence and its
JCI43378.1858. translation. Br. J. Nutr. 114, 999–1012. http://dx.doi.org/10.1017/
Hong, E.-G., Ko, H.J., Cho, Y.-R., Kim, H.-J., Ma, Z., Yu, T.Y., Friedline, R.H., Kurt-Jones, S0007114515002093.
E., Finberg, R., Fischer, M.A., Granger, E.L., Norbury, C.C., Hauschka, S.D., Phil- Munoz-Canoves, P., Scheele, C., Pedersen, B.K., Serrano, A.L., 2013. Interleukin-6
brick, W.M., Lee, C.-G., Elias, J.A., Kim, J.K., 2009. Interleukin-10 prevents diet- myokine signaling in skeletal muscle: a double-edged sword? FEBS J. 280,
induced insulin resistance by attenuating macrophage and cytokine response in 4131–4148. http://dx.doi.org/10.1111/febs.12338.
skeletal muscle. Diabetes 58, 2525–2535. http://dx.doi.org/10.2337/db08-1261. Muoio, D.M., Neufer, P.D., 2012. Lipid-Induced mitochondrial stress and insulin
Hong, H.C., Hwang, S.Y., Ryu, J.Y., Yoo, H.J., Seo, J.-A., Kim, S.G., Kim, N.H., Baik, S.H., action in muscle. Cell Metab. 15, 595–605. http://dx.doi.org/10.1016/j.
Choi, D.S., Choi, K.M., 2015. The synergistic impact of nonalcoholic fatty liver cmet.2012.04.010.
disease and metabolic syndrome on subclinical atherosclerosis. Clin. En- Murphy, A.M., Lyons, C.L., Finucane, O.M., Roche, H.M., 2014. Interactions between
docrinol. . http://dx.doi.org/10.1111/cen.12940 differential fatty acids and inflammatory stressors-impact on metabolic health.
Hotamisligil, G.S., 2006. Inflammation and metabolic disorders. Nature 444, Prostaglandins Leukot. Essent. Fat. Acids 92, 49–55. http://dx.doi.org/10.1016/j.
860–867. http://dx.doi.org/10.1038/nature05485. plefa.2014.05.003.
Hotamisligil, G.S., Peraldi, P., Budavari, A., Ellis, R., White, M.F., Spiegelman, B.M., Nagle, C.A., An, J., Shiota, M., Torres, T.P., Cline, G.W., Liu, Z.X., Wang, S., Catlin, R.L.,
1996. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in Shulman, G.I., Newgard, C.B., Coleman, R.A., 2007. Hepatic overexpression of
TNF-alpha- and obesity-induced insulin resistance. Science 271, 665–668. glycerol-sn-3-phosphate acyltransferase 1 in rats causes insulin resistance. J.
Hotamisligil, G.S., Shargill, N.S., Spiegelman, B.M., 1993. Adipose expression of tu- Biol. Chem. 282, 14807–14815. http://dx.doi.org/10.1074/jbc.M611550200.
mor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Sci- Nakamura, T., Furuhashi, M., Li, P., Cao, H., Tuncman, G., Sonenberg, N., Gorgun, C.Z.,
ence 259, 87–91 (80). Hotamisligil, G.S., 2010. Double-stranded RNA-dependent protein kinase links
Jager, J., Grémeaux, T., Cormont, M., Le Marchand-Brustel, Y., Tanti, J.-F., 2007. In- pathogen sensing with stress and metabolic homeostasis. Cell 140, 338–348.
terleukin-1beta-induced insulin resistance in adipocytes through down-reg- http://dx.doi.org/10.1016/j.cell.2010.01.001.
ulation of insulin receptor substrate-1 expression. Endocrinology 148, 241–251. Nedergaard, J., Bengtsson, T., Cannon, B., 2007. Unexpected evidence for active
http://dx.doi.org/10.1210/en.2006–0692. brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 293,
Jans, A., Konings, E., Goossens, G.H., Bouwman, F.G., Moors, C.C., Boekschoten, M.V., E444–E452. http://dx.doi.org/10.1152/ajpendo.00691.2006.
Afman, L.A., Müller, M., Mariman, E.C., Blaak, E.E., 2012a. PUFAs acutely affect Niemand, C., Nimmesgern, A., Haan, S., Fischer, P., Schaper, F., Rossaint, R., Heinrich,
triacylglycerol-derived skeletal muscle fatty acid uptake and increase post- P.C., Müller-Newen, G., 2003. Activation of STAT3 by IL-6 and IL-10 in primary
prandial insulin sensitivity. Am. J. Clin. Nutr. 95, 825–836. http://dx.doi.org/ human macrophages is differentially modulated by suppressor of cytokine
10.3945/ajcn.111.028787. signaling 3. J. Immunol. 170, 3263–3272.
Jans, A., Van Hees, A.M.J., Gjelstad, I.M.F., Sparks, L.M., Tierney, A.C., Ris??rus, U., Nishimura, S., Manabe, I., Nagasaki, M., Eto, K., Yamashita, H., Ohsugi, M., Otsu, M.,
Drevon, C.A., Schrauwen, P., Roche, H.M., Blaak, E.E., 2012b. Impact of dietary fat Hara, K., Ueki, K., Sugiura, S., Yoshimura, K., Kadowaki, T., Nagai, R., 2009.
quantity and quality on skeletal muscle fatty acid metabolism in subjects with CD8þ effector T cells contribute to macrophage recruitment and adipose tissue
the metabolic syndrome. Metabolism 61, 1554–1565. http://dx.doi.org/10.1016/ inflammation in obesity. Nat. Med. 15, 914–920. http://dx.doi.org/10.1038/
j.metabol.2012.04.003. nm.1964.
Jo, J., Gavrilova, O., Pack, S., Jou, W., Mullen, S., Sumner, A.E., Cushman, S.W., Periwal, Nowotny, B., Zahiragic, L., Krog, D., Nowotny, P.J., Herder, C., Carstensen, M., Yosh-
V., 2009. Hypertrophy and/or hyperplasia : dynamics of adipose tissue growth. imura, T., Szendroedi, J., Phielix, E., Schadewaldt, P., Schloot, N.C., Shulman, G.I.,
PLoS Comput. Biol., 5. http://dx.doi.org/10.1371/journal.pcbi.1000324. Roden, M., 2013. Mechanisms underlying the onset of oral lipid – induced
Johnson, A.M.F., Olefsky, J.M., 2013. The origins and drivers of insulin resistance. skeletal muscle insulin resistance in humans. Diabetes 62, 2240–2248. http:
Cell 152, 673–684. http://dx.doi.org/10.1016/j.cell.2013.01.041. //dx.doi.org/10.2337/db12-1179.
Jornayvaz, F.R., Shulman, G.I., 2012. Diacylglycerol activation of protein kinase Cε Odegaard, J.I., Chawla, A., 2013. Pleiotropic actions of insulin resistance and in-
and hepatic insulin resistance. Cell Metab. 15, 574–584. http://dx.doi.org/ flammation in metabolic homeostasis. Science 339, 172–177. http://dx.doi.org/
10.1016/j.cmet.2012.03.005. 10.1126/science.1230721.
Kamei, N., Tobe, K., Suzuki, R., Ohsugi, M., Watanabe, T., Kubota, N., Ohtsuka-Ko- Oh, D.Y., Talukdar, S., Bae, E.J., Imamura, T., Morinaga, H., Fan, W., Li, P., Lu, W.J.,
watari, N., Kumagai, K., Sakamoto, K., Kobayashi, M., Yamauchi, T., Ueki, K., Watkins, S.M., Olefsky, J.M., 2010. GPR120 is an omega-3 fatty acid receptor
Oishi, Y., Nishimura, S., Manabe, I., Hashimoto, H., Ohnishi, Y., Ogata, H., To- mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142,
kuyama, K., Tsunoda, M., Ide, T., Murakami, K., Nagai, R., Kadowaki, T., 2006. 687–698. http://dx.doi.org/10.1016/j.cell.2010.07.041.
Overexpression of monocyte chemoattractant protein-1 in adipose tissues Oh, D.Y., Walenta, E., Akiyama, T.E., Lagakos, W.S., Lackey, D., Pessentheiner, A.R.,
causes macrophage recruitment and insulin resistance. J. Biol. Chem. 281, Sasik, R., Hah, N., Chi, T.J., Cox, J.M., Powels, M.A., Di Salvo, J., Sinz, C., Watkins, S.
26602–26614. http://dx.doi.org/10.1074/jbc.M601284200. M., Armando, A.M., Chung, H., Evans, R.M., Quehenberger, O., McNelis, J.,
Kim, J.K., Gavrilova, O., Chen, Y., Reitman, M.L., Shulman, G.I., 2000. Mechanism of Bogner-Strauss, J.G., Olefsky, J.M., 2014. A Gpr120-selective agonist improves
insulin resistance in A-ZIP/F-1 fatless mice. J. Biol. Chem. 275, 8456–8460. http: insulin resistance and chronic inflammation in obese mice. Nat. Med. 20,
//dx.doi.org/10.1074/jbc.275.12.8456. 942–947. http://dx.doi.org/10.1038/nm.3614.
Koenen, T.B., Stienstra, R., Van Tits, L.J., Joosten, L.A.B., Van Velzen, J.F., Hijmans, A., Oliver, E., McGillicuddy, F.C., Harford, K.A., Reynolds, C.M., Phillips, C.M., Ferguson, J.
Pol, J.A., Van Der Vliet, J.A., Netea, M.G., Tack, C.J., Stalenhoef, A.F.H., De Graaf, J., F., Roche, H.M., 2012. Docosahexaenoic acid attenuates macrophage-induced
2011. The inflammasome and caspase-1 activation: a new mechanism under- inflammation and improves insulin sensitivity in adipocytes-specific differ-
lying increased inflammatory activity in human visceral adipose tissue. En- ential effects between LC n  3 PUFA. J. Nutr. Biochem. 23, 1192–1200. http://dx.
docrinology 152, 3769–3778. http://dx.doi.org/10.1210/en.2010–1480. doi.org/10.1016/j.jnutbio.2011.06.014.
Kylin, E., 1923. Studien über das Hypertonie-Hyperglykämie-Hyperurikämie Syn- Osborn, O., Olefsky, J.M., 2012. The cellular and signaling networks linking the
drom. 44. Zentralblatt für Inn. Medizin, pp. 105–112. immune system and metabolism in disease. Nat. Med. 18, 363–374. http://dx.
Lafontan, M., Langin, D., 2009. Lipolysis and lipid mobilization in human adipose doi.org/10.1038/nm.2627.
tissue. Prog. Lipid Res. 48, 275–297. http://dx.doi.org/10.1016/j. Pedersen, B.K., Febbraio, M.A., 2007. Point: Interleukin-6 does have a beneficial role
plipres.2009.05.001. in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102, 814–816.
Lewis, G., O’meara, N., 1990. Postprandial lipoprotein metabolism in normal and http://dx.doi.org/10.1152/japplphysiol.01208.2006.
obese subjects: comparison after the vitamin A fat-loading test*. J. Clin. En- Phillips, C.M., Tierney, A.C., Perez-Martinez, P., Defoort, C., Blaak, E.E., Gjelstad, I.M.
docrinol. Metab., 71. F., Lopez-Miranda, J., Kiec-Klimczak, M., Malczewska-Malec, M., Drevon, C.A.,
Liu, J., Divoux, A., Sun, J., Zhang, J., Clément, K., Glickman, J.N., Sukhova, G.K., Hall, W., Lovegrove, J.A., Karlstrom, B., Risérus, U., Roche, H.M., 2012. Obesity
Wolters, P.J., Du, J., Gorgun, C.Z., Doria, A., Libby, P., Blumberg, R.S., Kahn, B.B., and body fat classification in the metabolic syndrome: impact on cardiometa-
Hotamisligil, G.S., Shi, G.-P., 2009. Genetic deficiency and pharmacological bolic risk metabotype. Obesity 21, 154–161. http://dx.doi.org/10.1038/
stabilization of mast cells reduce diet-induced obesity and diabetes in mice. oby.2012.188.
Nat. Med. 15, 940–945. http://dx.doi.org/10.1038/nm.1994. Phillips, C.M., Tierney, A.C., Roche, H.M., 2008. Gene-nutrient interactions in the
Lumeng, C.N., DelProposto, J.B., Westcott, D.J., Saltiel, A.R., 2008. Phenotypic metabolic syndrome. J. Nutr. Nutr. 1, 136–151. http://dx.doi.org/10.1159/
switching of adipose tissue macrophages with obesity is generated by spatio- 000112461.
temporal differences in macrophage subtypes. Diabetes 57, 3239–3246. http: Powell, D.J., Turban, S., Gray, A., Hajduch, E., Hundal, H.S., 2004. Intracellular cer-
//dx.doi.org/10.2337/db08-0872. amide synthesis and protein kinase Czeta activation play an essential role in
Lundgren, M., Svensson, M., Lindmark, S., 2007. Fat cell enlargement is an in- palmitate-induced insulin resistance in rat L6 skeletal muscle cells. Biochem. J.
dependent marker of insulin resistance and “hyperleptinaemia. Diabetologia. 382, 619–629. http://dx.doi.org/10.1042/BJ20040139.
Martinon, F., Burns, K., Tschopp, J., 2002. The inflammasome: a molecular platform Reaven, G.M., 1988. Role of insulin resistance in human disease. Diabetes 37,
triggering activation of inflammatory caspases and processing of proIL-beta. 1595–1607. http://dx.doi.org/10.2337/diab.37.12.1595.
Mol. Cell 10, 417–426. Reynolds, C.M., Mcgillicuddy, F.C., Harford, K.A., Finucane, O.M., Mills, K.H.G., Roche,
McGillicuddy, F.C., Harford, K.A., Reynolds, C.M., Oliver, E., Claessens, M., Mills, K.H. H.M., 2012. Dietary saturated fatty acids prime the NLRP3 inflammasome via
G., Roche, H.M., 2011. Lack of interleukin-1 receptor I (IL-1RI) protects mice TLR4 in dendritic cells-implications for diet-induced insulin resistance. Mol.
from high-fat diet-induced adipose tissue inflammation coincident with im- Nutr. Food Res. 56, 1212–1222. http://dx.doi.org/10.1002/mnfr.201200058.
proved glucose homeostasis. Diabetes 60, 1688–1698. http://dx.doi.org/ Rosenwald, M., Wolfrum, C., 2014. The origin and definition of brite versus white
214 A.A. Cooke et al. / European Journal of Pharmacology 785 (2016) 207–214

and classical brown adipocytes. Adipocyte. phosphorylation of insulin receptor substrate proteins by discrete mechanisms.
Rosqvist, F., Iggman, D., Kullberg, J., Cedernaes, J., Johansson, H.E., Larsson, A., Jo- Mol. Cell. Biol. 24, 5434–5446. http://dx.doi.org/10.1128/MCB.24.12.5434.
hansson, L., Ahlström, H., Arner, P., Dahlman, I., Risérus, U., 2014. Overfeeding Uysal, K.T., Wiesbrock, S.M., Marino, M.W., Hotamisligil, G.S., 1997. Protection from
polyunsaturated and saturated fat causes distinct effects on liver and visceral obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature
fat accumulation in humans. Diabetes 63, 2356–2368. http://dx.doi.org/ 389, 610–614. http://dx.doi.org/10.1038/39335.
10.2337/db13-1622. Vague, J., 1947. La différentiation sexuelle, facteur déterinant des formes de l’obé-
Rotter, V., Nagaev, I., Smith, U., 2003. Interleukin-6 (IL-6) induces insulin resistance sité. Presse Méd. 55, 339–340.
in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-, overexpressed Virtue, S., Vidal-Puig, A., 2010. Adipose tissue expandability, lipotoxicity and the
in human fat cells from insulin-resistant subjects. J. Biol. Chem. 278, Metabolic Syndrome–an allostatic perspective. Biochim. Biophys. Acta 1801,
45777–45784. http://dx.doi.org/10.1074/jbc.M301977200. 338–349. http://dx.doi.org/10.1016/j.bbalip.2009.12.006.
Samuel, V.T., Shulman, G.I., 2012. Mechanisms for insulin resistance: common Virtue, S., Vidal-Puig, A., 2008. It's not how fat you are, it's what you do with it that
threads and missing links. Cell 148, 852–871. http://dx.doi.org/10.1016/j. counts. PLoS Biol. 6. http://dx.doi.org/10.1371/journal.pbio.0060237.
cell.2012.02.017. Weisberg, S.P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R.L., Ferrante, A.W.,
Saraiva, M., O’Garra, A., 2010. The regulation of IL-10 production by immune cells. 2003. Obesity is associated with macrophage accumulation in adipose tissue. J.
Nat. Rev. Immunol. 10, 170–181. http://dx.doi.org/10.1038/nri2711. Clin. Investig. 112, 1796–1808. http://dx.doi.org/10.1172/JCI19246.
Schipper, H.S., Prakken, B., Kalkhoven, E., Boes, M., 2012. Adipose tissue-resident Winer, D.A., Winer, S., Shen, L., Wadia, P.P., Yantha, J., Paltser, G., Tsui, H., Wu, P.,
immune cells: key players in immunometabolism. Trends Endocrinol. Metab. Davidson, M.G., Alonso, M.N., Leong, H.X., Glassford, A., Caimol, M., Kenkel, J.A.,
23, 407–415. http://dx.doi.org/10.1016/j.tem.2012.05.011. Tedder, T.F., McLaughlin, T., Miklos, D.B., Dosch, H.-M., Engleman, E.G., 2011. B
Schrauwen, P., Timmers, S., 2014. Can resveratrol help to maintain metabolic cells promote insulin resistance through modulation of T cells and production
health? Proc. Nutr. Soc. 73, 271–277. http://dx.doi.org/10.1017/ of pathogenic IgG antibodies. Nat. Med. 17, 610–617. http://dx.doi.org/10.1038/
S0029665113003856. nm.2353.
Schweiger, M., Schreiber, R., 2006. Adipose triglyceride lipase and hormone-sen- Wolsk, E., Mygind, H., Grøndahl, T.S., Pedersen, B.K., Hall, G.VAN., 2010. IL-6 se-
sitive lipase are the major enzymes in adipose tissue triacylglycerol catabolism. lectively stimulates fat metabolism in human skeletal muscle. Am. J. Physiol.
J. Biol. Endocrinol. Metab. 299, E832–E840. http://dx.doi.org/10.1152/
Shames, B.D., Selzman, C.H., Meldrum, D.R., Pulido, E.J., Barton, H.A., Meng, X., ajpendo.00328.2010.
Harken, A.H., McIntyre, R.C., 1998. Interleukin-10 stabilizes inhibitory kappaB- Wu, D., Molofsky, A.B., Liang, H.-E., Ricardo-Gonzalez, R.R., Jouihan, H.A., Bando, J.K.,
alpha in human monocytes. Shock 10, 389–394.
Chawla, A., Locksley, R.M., 2011. Eosinophils sustain adipose alternatively ac-
Stephens, F., Mendis, B., Shannon, C., Cooper, S., Ortori, C., Barrett, D., Mansell, P.,
tivated macrophages associated with glucose homeostasis. Science 332,
Tsintzas, K., 2014. Fish oil omega-3 fatty acids partially prevent lipid-induced
243–247. http://dx.doi.org/10.1126/science.1201475 (80).
insulin resistance in human skeletal muscle without limiting acylcarnitine ac-
Wuesten, O., Balz, C.H., Bretzel, R.G., Kloer, H.-U., Hardt, P.D., 2005. Effects of oral fat
cumulation. Clin. Sci. 127, 315–322.
load on insulin output and glucose tolerance in healthy control subjects and
Sun, K., Tordjman, J., Clément, K., Scherer, P.E., 2013. Fibrosis and adipose tissue
obese patients without diabetes. Diabetes Care 28, 360–365. http://dx.doi.org/
dysfunction. Cell Metab. 18, 470–477. http://dx.doi.org/10.1016/j.
10.2337/diacare.28.2.360.
cmet.2013.06.016.
Xiao, C., Giacca, A., Carpentier, A., Lewis, G.F., 2006. Differential effects of mono-
Sung, K.-C., Wild, S.H., Kwag, H.J., Byrne, C.D., 2012. Fatty liver, insulin resistance,
unsaturated, polyunsaturated and saturated fat ingestion on glucose-stimu-
and features of metabolic syndrome: relationships with coronary artery cal-
lated insulin secretion, sensitivity and clearance in overweight and obese, non-
cium in 10,153 people. Diabetes Care 35, 2359–2364. http://dx.doi.org/10.2337/
diabetic humans. Diabetologia 49, 1371–1379. http://dx.doi.org/10.1007/
dc12-0515.
Targher, G., 2007. Non-alcoholic fatty liver disease, the metabolic syndrome and the s00125-006-0211-x.
risk of cardiovascular disease: the plot thickens. Diabet. Med. 24, 1–6. http://dx. Xu, H., Barnes, G.T., Yang, Q., Tan, G., Yang, D., Chou, C.J., Sole, J., Nichols, A., Ross, J.S.,
doi.org/10.1111/j.1464–5491.2007.02025.x. Tartaglia, L.A., Chen, H., 2003. Chronic inflammation in fat plays a crucial role in
Targher, G., Arcaro, G., 2007. Non-alcoholic fatty liver disease and increased risk of the development of obesity-related insulin resistance. J. Clin. Investig. 112,
cardiovascular disease. Atherosclerosis 191, 235–240. http://dx.doi.org/10.1016/ 1821–1830. http://dx.doi.org/10.1172/JCI19451.
j.atherosclerosis.2006.08.021. Yore, M.M., Syed, I., Moraes-vieira, P.M., Zhang, T., Herman, M.A., Homan, E.A., Patel,
Targher, G., Bertolini, L., Padovani, R., Poli, F., Scala, L., Tessari, R., Zenari, L., Falezza, R.T., Lee, J., Chen, S., Peroni, O.D., Dhaneshwar, A.S., Hammarstedt, A., Smith, U.,
G., 2006. Increased Prevalence of Cardiovascular Disease in Type 2 Diabetic Mcgraw, T.E., Saghatelian, A., Kahn, B.B., 2014. Discovery of a class of en-
Patients With Non-Alcoholic Fatty Liver Disease, pp. 403–409. doi: 〈http://dx. dogenous mammalian lipids with anti-diabetic and anti-inflammatory effects.
doi.org/10.1111/j.1464-5491.2005.01817.x〉. Cell 159, 318–332. http://dx.doi.org/10.1016/j.cell.2014.09.035.
Tchkonia, T., Tchoukalova, Y.D., Giorgadze, N., Pirtskhalava, T., Karagiannides, I., Yu, Y., Ginsberg, H., 2005. Adipocyte signaling and lipid homeostasis sequelae of
Forse, R.A., Koo, A., Stevenson, M., Chinnappan, D., Cartwright, A., Jensen, M.D., insulin-resistant adipose tissue. Circ. Res.
Kirkland, J.L., Abun-, J.L.K., 2005. Abundance of Two Human Preadipocyte Yuan, M., Konstantopoulos, N., Lee, J., Hansen, L., Li, Z.W., Karin, M., Shoelson, S.E.,
Subtypes With Distinct Capacities for Replication , Adipogenesis , and Apoptosis 2001. Reversal of obesity- and diet-induced insulin resistance with salicylates
Varies Among Fat Depots. Vol. 02118, pp. 267–277. doi: 〈http://dx.doi.org/10. or targeted disruption of Ikkbeta. Science 293, 1673–1677. http://dx.doi.org/
1152/ajpendo.00265.2004〉. 10.1126/science.1061620.
Tschopp, J., Schroder, K., 2010. NLRP3 inflammasome activation: the convergence of Yubero-Serrano, E.M., Delgado-Lista, J., Tierney, A.C., Perez-Martinez, P., Garcia-
multiple signalling pathways on ROS production? Nat. Rev. Immunol. 10, Rios, A., Alcala-Diaz, J.F., Castano, J.P., Tinahones, F.J., Drevon, C.A., Defoort, C.,
210–215. http://dx.doi.org/10.1038/nri2725. Blaak, E.E., Dembinska-Kiec, A., Riserus, U., Lovegrove, J.A., Perez-Jimenez, F.,
Tumova, J., Andel, M., Trnka, J., 2015. Excess of free fatty acids as a cause of me- Roche, H.M., Lopez-Miranda, J., 2015. Insulin resistance determines a differ-
tabolic dysfunction in skeletal muscle. Physiol. Res. ential response to changes in dietary fat modification on metabolic syndrome
Ueki, K., Kondo, T., Kahn, C.R., 2004. Suppressor of cytokine signaling 1 ( SOCS-1 ) risk factors: the LIPGENE study. Am. J. Clin. Nutr., 1509–1517. http://dx.doi.org/
and SOCS-3 cause insulin resistance through inhibition of tyrosine 10.3945/ajcn.115.111286.

You might also like