inflammation and lipid signal.pdf

Upload: inyong-budiono

Post on 02-Jun-2018

233 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    1/23

    INFLAMMATION AND LIPID SIGNALING IN THE ETIOLOGY OF

    INSULIN RESISTANCE

    Christopher K. Glass, M.D.and Jerrold M. Olefsky, M.D.

    Abstract

    Inflammation and lipid signaling are intertwined modulators of homeostasis and immunity. In

    addition to the extensively studied eicosanoids and inositol phospholipids, emerging studies

    indicate that many other lipid species act to positively and negatively regulate inflammatory

    responses. Conversely inflammatory signaling can significantly alter lipid metabolism in the liver,

    adipose tissue, skeletal muscle and macrophage in the context of infection, diabetes and

    atherosclerosis. Here, we review recent findings related to this interconnected network from theperspective of immunity and metabolic disease.

    Introduction

    Insulin resistance is a major metabolic abnormality in the great majority of patients with

    Type 2 diabetes and the incidence of this disease has reached epidemic proportions around

    the globe (Hupfeld et al., 2006; Kahn et al., 2006). Obesity is clearly the most common

    cause of insulin resistance, and it is the parallel epidemic of obesity, which is driving the

    rising incidence of Type 2 diabetes (Kahn et al., 2006). The etiology of insulin resistance

    has been intensely studied (Qatanani and Lazar, 2007; Shulman, 2000), and it is now

    recognized that chronic tissue inflammation is an important cause of obesity-induced insulinresistance (Attie and Scherer, 2009; Lumeng and Saltiel, 2011; Olefsky and Glass, 2010;

    Shoelson et al., 2007). One of the initial lines of evidence for this relationship derived from

    the unexpected observations that TNF, a cytokine associated with cachexia in cancer, was

    elevated in obese adipose tissue in rodents and that inhibition of this cytokine improved

    glucose tolerance and insulin sensitivity (Hotamisligil et al., 1993). Subsequently, studies

    demonstrated that a key mechanism underlying obesity-induced inflammation is the

    accumulation of increased numbers of tissue macrophages, particularly in obese adipose

    tissue (Weisberg et al., 2003; Xu et al., 2003). These adipose tissue macrophages (ATMs)

    can span the spectrum from the most pro-inflammatory, M1-like, cells to anti-inflammatory,

    M2-like, macrophages (Lumeng et al., 2007a; Lumeng et al., 2007b; Nguyen et al., 2007).

    The proinflammatory ATMs are often referred to as classically activated macrophages(CAMs) while the anti-inflammatory macrophages are referred to as alternatively activated

    macrophages (AAMs).

    2012 Elsevier Inc. All rights reserved.

    Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our

    customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of

    the resulting proof before it is published in its final citable form. Please note that during the production process errors may be

    discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

    NIH Public AccessAuthor ManuscriptCell Metab. Author manuscript; available in PMC 2014 September 05.

    Published in final edited form as:

    Cell Metab. 2012 May 2; 15(5): 635645. doi:10.1016/j.cmet.2012.04.001.

    NIH-PAAu

    thorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthorM

    anuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    2/23

    In obesity, the balance is tilted towards the M1-like macrophage polarization state (Lumeng

    et al., 2007a), and these cells secrete a number of different cytokines, such as TNFa, IL-1b,

    etc. In turn, these cytokines can act through paracrine mechanisms to directly inhibit insulin

    action in insulin target cells (i.e. hepatocytes, myocytes, and adipocytes) (Feve and Bastard,

    2009; Hotamisligil, 1999). It is also possible that these tissue cytokines leak into the

    systemic circulation to cause insulin resistance through endocrine effects. Cytokine

    signaling activates stress kinases such as S6K, IKK, JNK1, and PKC, whichphosphorylate IRS1 on inhibitory serine residues and results in impaired downstream

    signaling (Hotamisligil, 2006; Schenk et al., 2008). Cytokines can also inhibit insulin action

    through transcriptional mechanisms by decreasing the expression levels of key insulin

    signaling molecules (Stephens and Pekala, 1992). Finally, cytokines can promote ceramide

    biosynthesis, which directly inhibits Akt activation, as discussed below (Holland et al.,

    2011).

    While ATMs are considered to be the ultimate effector cell elaborating cytokines which

    cause insulin resistance, their recruitment to adipose tissue and their activation state are

    influenced by other immune cell types that populate obese adipose tissue, including T cells,

    B cells, eosinophils, and mast cells (Feuerer et al., 2009; Kintscher et al., 2008; Liu et al.,2009; Nishimura et al., 2009; Winer et al., 2011; Wu et al., 2011). However, inflammation is

    not the only mechanism of insulin resistance in obesity. For example, various abnormalities

    of lipid metabolism have been described which can also impair insulin signaling (Itani et al.,

    2002; Savage et al., 2007). These lipotoxic effects include the production of intracellular

    lipid mediators, such as diacylglycerols (Samuel et al., 2010), as well as more direct effects

    of circulating saturated fatty acids. However, chronic tissue inflammation and lipid

    abnormalities are not completely separable processes, and these two systems are closely

    intertwined, and each can amplify the other in the in vivo pathophysiologic setting. In this

    review, we focus on the inter-connections between abnormal lipid signaling and pro-

    inflammatory pathways in insulin resistant states.

    Effects of Fatty Acids on Inflammation

    Saturated Fatty Acids

    Circulating FFAs are typically elevated in insulin resistant states, and dietary fat intake has

    an important influence on the composition of these FFAs (Fielding et al., 1996). The

    association between elevated FFA levels and insulin resistance is well known (Park et al.,

    2007) and numerous studies have demonstrated that infusions of triglyceride emulsions plus

    heparin to rapidly raise FFA levels, will cause a decrease in insulin sensitivity (Boden et al.,

    1991; Roden et al., 1996; Staehr et al., 2003) . In addition to circulating FFAs, macrophages

    and other immune cell types in adipose tissue are exposed to high local concentrations of

    FFAs released from adipocytes by lipolysis. In this way, these cells exist within a lipid-rich

    environment that could amplify the effects of SFAs within adipose tissue.

    SFAs are pro-inflammatory lipid compounds, with many studies demonstrating that

    treatment of various cell types with SFAs broadly activates inflammatory signaling in

    macrophages, adipocytes, myocytes, hepatocytes, etc. (Hwang and Rhee, 1999; Nguyen et

    al., 2007; Nguyen et al., 2005; Yu et al., 2002). This leads to stimulation of IKK/NFB and

    Glass and Olefsky Page 2

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    3/23

    JNK1/AP1, as well as release of cytokines (Fig. 1). Several laboratories have independently

    reported that pro-inflammatory effects of SFAs are mediated, at least in part, through

    activation of Tlr4 and/or Tlr2 (Lee et al., 2001; Nguyen et al., 2007; Schaeffler et al., 2009;

    Senn, 2006; Shi et al., 2006). Tlr2 and Tlr4 are members of the TLR family of receptors that

    recognize pathogen-associated molecular patterns (PAMPs) as well as endogenously derived

    indicators of tissue injury (Kumar et al., 2009). Tl4 ligands include lipopolysaccharide

    (LPS), a component of the cell walls of gram-negative bacteria while Tlr2 ligands includethe lipoteichoic acid component of gram-positive bacteria (Kumar et al., 2009; Poltorak et

    al., 1998), In the absence of Tlr4, SFA-mediated inflammatory signaling is abrogated in

    adipocytes (Shi et al., 2006). Furthermore, Tlr4 KO mice are resistant to the effects of high

    fat diets to cause decreased insulin sensitivity ((Saberi et al., 2009; Shi et al., 2006), Fig. 1).

    As an additional aspect of Tlr4-mediated inflammation, it has been reported that obesity is

    associated with gastrointestinal dysbiosis in which LPS derived from GI tract microflora

    leaks into the systemic circulation (Brun et al., 2007; Creely et al., 2007).

    It has been suggested that the reported effects of SFAs on widely used indicators of TLR4

    activation in macrophages, such as TNF, are due to contamination of the serum or albumin

    used in these experiments with endotoxin (Erridge and Samani, 2009). While this mayaccount for some of the effects reported in cultured macrophages, infusion of heparin/lipid

    emulsions results in Tlr4-dependent inflammatory responses in adipose tissue (Shi et al.,

    2006). Collectively, there is substantial evidence for functionally important relationships

    between SFAs, TLR signaling and insulin resistance, but the molecular mechanisms remain

    unclear. Structural considerations (Park et al., 2009) and direct binding assays (Schaeffler et

    al., 2009) argue against the possibility that SFAs are direct ligands for Tlr4. There are

    several ways in which SFAs could indirectly activate Tlr signaling. One possibility is

    through interaction with SFA binding proteins that function as TLR co-receptors. A

    precedent for this type of mechanism is provided by the ability of CD36, a fatty acid

    transporter and oxidzed LDL receptor, to interact with Tlr2 and induce signaling in response

    to oxidized LDL (Seimon et al., 2010; Stewart et al., 2010). SFAs have also been reported toinduce Tlr4-depedent gene expression by promoting dimerization of Tlr4, a necessary step

    in receptor activation, within specialized lipid domains termed lipid rafts in macrophages

    (Fig. 1) (Wong et al., 2009). This appears to be dependent on the ability of SFAs to

    stimulate NADPH oxidase production of ROS. In independent studies, SFA treatment led to

    membrane redistribution of c-Src so that it is now concentrated in lipid rafts, where it

    becomes activated (Holzer et al., 2011). This then leads to stimulation of JNK1 and its

    proinflammatory target genes (Fig. 1). It is possible that the dimerization of Tlr4, as well as

    the redistribution of c-Src in lipid rafts upon SFA treatment are mechanistically related

    events, providing a pathway for SFA-induced pro-inflammatory signaling. A third

    possibility is that SFAs may stimulate the production or release of a tissue damage signal

    that is sensed by Tlrs. There are a number of endogenously derived molecules that havebeen shown to exert these types of functions when released from necrotic or damaged cells,

    such as high mobility group proteins (Park et al., 2004). In concert, substantial additional

    work will be required to fully establish the molecular mechanisms linking SFAs to Tlr

    signaling.

    Glass and Olefsky Page 3

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    4/23

    SFAs can also stimulate stress and/or inflammatory pathways by TLR-independent

    mechanisms. Treatment of macrophages with SFAs leads to an increase in ROS production

    and this can activate a number of stress kinases. The ability of SFAs to increase ROS levels

    may also drive stimulation of the NLRP3 (nucleotide binding domain, leucine-rich repeats

    containing family, pyrin domain-containing 3) inflammasome (Wen et al., 2011). NLRP3

    forms a complex with the adaptor protein ASC (apoptotic speck protein containing a caspase

    recruitment domain) that regulates caspase 3-dependent cleavage of pro-IL1to releaseactive IL1 (Ting et al., 2008). Recent studies have demonstrated that SFA-induced ROS

    can activate the NLRP3-ASC inflammasome complex, thereby, increasing release of IL1

    (Wen et al., 2011). When applied to insulin target cells, IL-1can decrease insulin signaling,

    providing another mechanism for SFA mediated inflammatory responses causing insulin

    resistance. Indeed, NLRP3, ASC, and caspase 1 knockout mice are all protected from HFD-

    induced inflammation and insulin resistance (Wen et al., 2011).

    Finally, SFAs are precursors to other lipid products, such as DAGs, and intracellular levels

    of DAGs can be increased in insulin resistant states (Kumashiro et al., 2011). This can lead

    to PKC activation with decreased insulin signaling, and this concept has been the subject of

    recent reviews (Samuel et al., 2010). Taken together, these mechanisms would explain animportant component of lipotoxicity in which SFAs promote the chronic tissue

    inflammatory state which can cause insulin resistance.

    In addition to stimulating inflammatory pathways, SFAs can lead to decreased insulin

    sensitivity through other mechanisms. For example, SFAs are precursors for ceramide

    biosynthesis, and ceramide can directly cause decreased insulin signaling, as discussed in

    more detail below (Stratford et al., 2004; Summers, 2010; Ussher et al., 2010). Finally,

    FetuinA is a glycoprotein primarily made in hepatocytes and secreted into the circulation. In

    vitro studies have demonstrated that SFA treatment of hepatocytes leads to increased

    FetuinA mRNA expression and secretion, and other studies have indicated that FetuinA can

    directly inhibit the early steps of insulin signaling (Dasgupta et al., 2010).

    Ceramide Metabolism

    Lipid and inflammatory pathway signaling are closely intertwined processes which can

    result in insulin resistance. One important crossroad between these two pathways, relevant

    to the development of decreased insulin sensitivity involves ceramide biosynthesis. Many

    papers have demonstrated strong associations between intracellular ceramide levels and the

    development of insulin resistance (Holland et al., 2011; Stratford et al., 2004; Ussher et al.,

    2010). For example, the saturated fatty acid palmitate is a preferential substrate leading to

    the biosynthesis of ceramide, providing another potential mechanism for SFA-induced

    insulin resistance (Ussher et al., 2010). Elevated intracellular ceramide levels can stimulate

    the ability of phosphatase 2A to dephosphorylated Akt and this inhibits insulin signaling,providing a mechanism for ceramide-mediated insulin resistance (Stratford et al., 2004).

    Recent studies have revealed a further role for ceramide at the nexus of inflammation and

    lipid signaling. Thus, independent of their roles as precursors for ceramide biosynthesis,

    SFA stimulation of the Tlr4 pathway causes an increase in expression of the enzymes

    involved in ceramide biosynthesis (Holland et al., 2011). Other non-lipid Tlr4 agonists, such

    Glass and Olefsky Page 4

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    5/23

    as LPS, have a similar effect. Interestingly, stimulation with TNFrecapitulated the

    increased ceramide biosynthesis (Dbaibo et al., 2001; Holland et al., 2011), and these

    authors showed that the effect of Tlr4 and TNFsignaling on ceramide biosynthesis was

    IKKdependent, directly demonstrating how inflammatory pathway signaling inputs can

    promote biosynthesis of a lipid moiety, i.e., ceramide, which directly inhibits insulin

    signaling (Fig. 1).

    Omega 3 Fatty Acids

    While SFAs promote inflammation and insulin resistance, other classes of fatty acids can

    have beneficial effects. For example, long chain polyunsaturated omega 3 FAs, such as

    DHA and EPA, can exert anti-inflammatory effects, and this has been known for many years

    (Calder, 2006; Lang et al., 2003; Polozova and Salem, 2007; Proudman et al., 2008; Weldon

    et al., 2007).

    However, the mechanisms for these effects have been poorly understood until recently.

    There is a family of lipid sensing GPCRs, including GPR40, 41, 43, 84, and 120 (Itoh et al.,

    2003; Tazoe et al., 2008; Wang et al., 2006), and recent reports have indicated that omega 3

    FAs can stimulate GPR120 (Hirasawa et al., 2005; Oh et al., 2010).. With respect to

    inflammation, omega-3 FAs are ligands for GPR120 and can exert robust and broad anti-

    inflammatory effects in various macrophage types (Oh et al., 2010). For example, when

    RAW267.4 cells or intraperitoneal macrophages are treated with Tlr4/2 agonists, or TNF,

    potent stimulation of inflammatory pathways occurs. However, when macrophages are

    pretreated with omega 3 FAs, these proinflammatory effects are inhibited (Lee et al., 2003)

    and these anti-inflammatory effects are completely abrogated in the absence of GPR120 (Oh

    et al., 2010). These studies show that omega 3 FAs are ligands for GPR120 and exert anti-

    inflammatory effects by signaling through this receptor. In this sense, GPR120 emerges as a

    receptor/sensor for this anti-inflammatory class of fatty acids.

    The mechanisms for this effect are also of interest. TAK1 is a proximal kinase in theseproinflammatory pathways and in response to inflammatory signals, the adaptor protein

    TAB1 associates with and activates TAK1, stimulating both the IKK/NFB and the

    JNK/AP1 pathways. Activation of GPR120 by omega 3 FAs specifically inhibits TAK

    phosphorylation and activation, providing a mechanism for the broad inhibition of Tlr and

    TNF signaling (Oh et al., 2010). arrestins are a family of molecules which can serve as

    adaptor or scaffold proteins for a number of different GPCRs (Luttrell and Lefkowitz, 2002).

    Interestingly, omega 3 FA stimulation leads to direct association between GPR120 and

    arrestin-2 and the anti-inflammatory effects of GPR120 are completely arrestin-2

    dependent. The apparent mechanism for this is that DHA stimulation leads to GPR120/

    arrestin-2 association with subsequent internalization of the complex. The internalized

    arrestin-2 then associates with TAB1, stealing it from the TAK1 complex, leading toinactivation of TAK1 with inhibition of downstream inflammatory signaling to IKK/NFB

    and JNK/AP1 ((Oh et al., 2010), and Figure 2).

    These cellular mechanisms translate to the in vivo situation. Thus, omega 3 FA treatment of

    WT obese mice led to marked in vivo anti-inflammatory and insulin sensitizing effects, but

    Glass and Olefsky Page 5

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    6/23

    no such effects were observed in obese GPR120 KO animals. Taken together, it appears that

    an important mechanism for the anti-inflammatory effects of omega 3 FAs operates through

    the GPR120 signaling pathway in macrophages.

    It is possible that additional mechanisms also exist which could participate in the anti-

    inflammatory effects of omega 3 FAs. For example, these FAs can be metabolized to small

    lipid mediators termed protectins/resolvins and studies have shown that addition of

    protectins/resolvins to in vitro systems can produce anti-inflammatory effects (Serhan and

    Chiang, 2008; Serhan et al., 2008) (Fig. 1). Furthermore, in vivo studies in mice also

    indicate anti-inflammatory effects through the resolvin/protectin system. Finally, omega 3

    FAs are metabolized through a variety of lipid biosynthetic pathways and can inhibit the

    production of arachidonic acid and other pro-inflammatory eicosanoids.

    Palmatoleate

    Palmatoleate (C16:1N7) is a nutritional component of many food stuffs and can also be

    produced by de novo lipogenesis. Circulating C16:1N7 levels are directly correlated with

    insulin sensitivity in mice and that inhibition of C16:1N7 production can cause insulin

    resistance (Cao et al., 2008). Based on this work, they suggest that C16:1N7 is a lipokine

    secreted from adipocytes that can modulate glucose homeostasis in muscle and liver through

    endocrine effects. When added to cells C16:1N7 can produce insulin-like effects and these

    may relate to GPR120 in adipocytes. C16:1N7 is a GPR120 agonist and, in adipocytes, it

    stimulates glucose transport through a Gq-dependent signaling pathway that leads to

    GLUT4 translocation.

    Effects of inflammation on lipid metabolism

    While the impact of inflammation on gene expression has been studied in a variety of tissues

    and cell types, much less is known regarding the influence of inflammation on the vast

    repertoire of lipids that function as structural components of cell membranes, sources ofenergy, and signaling molecules. Notable exceptions are provided by the eicosanoids and

    inositol phospholipids, which have been subjected to extensive analysis based on their roles

    as essential signaling molecules in diverse aspects of development, homeostasis and

    immunity. For example, prostaglandins represent a class of eiconsanoids that are generated

    from arachidonic acid in membrane phospholipids through the sequential actions of

    phospholipase A and cyclooxygenase enzymes. Both classes of enzymes are under direct

    control by signaling pathways that respond to PAMPs and inflammatory cytokines, enabling

    rapid and robust increases in prostaglandin generation. The resulting prostaglandin

    metabolites have diverse biological roles, including amplification of inflammatory responses

    that contribute to the cardinal signs of inflammation (redness, heat, pain, swelling). In the

    case of the inositol phospholipids, extensive work over the past decade has revealed diverseroles of phosphatidylinositol 3-kinases, particularly PI3Kand PI3K, in responding to

    inflammatory signals to orchestrate specific aspects of innate and adaptive immune

    responses. These topics have been the focus of recent reviews (Dennis et al., 2011; Fung-

    Leung, 2011; Ghigo et al., 2010; Ricciotti and FitzGerald, 2011).

    Glass and Olefsky Page 6

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    7/23

    In contrast, knowledge of effects of inflammation on metabolism of most other classes of

    lipids remains at a relatively rudimentary level. In this section, we discuss the impact of

    inflammation on lipid metabolism from the perspective of the relationship between

    inflammation and metabolic disease. We begin with a brief description of the effects of

    innate immunity-driven inflammation on lipid metabolism at the whole body level, and then

    consider effects at the organ and cellular level in the liver, adipose tissue, skeletal muscle

    and macrophage. We conclude this section with a discussion of the intersection ofinflammatory signaling pathways with transcription factors that control cholesterol and fatty

    acid biosynthesis.

    Inflammation and lipid metabolism at the whole body level

    At the whole body level, inflammation initiated by severe bacterial and viral infection can

    result in marked changes in lipid and lipoprotein metabolism. For example, infection with

    gram-negative organisms has long been known to induce marked hypertriglyceridemia and

    elevated circulating free fatty acid levels (Gallin et al., 1969). Although these changes are

    likely to reflect metabolic adaptations that enable the host to resist infection, there is also

    evidence that high density lipoproteins (HDL) and triglyceride-rich lipoproteins themselves

    function to bind and neutralize bacterial endotoxins (Harris et al., 2000). Experimental

    studies using LPS to mimic infection indicate that increases in very low density lipoproteins

    (VLDL) account for the majority of the rise in serum triglycerides, resulting from both

    increased production and decreased catabolism (Feingold et al., 1992b). The hyperlipidemia

    of sepsis was initially considered to be a consequence of catecholamine release, which

    stimulate release of free fatty acids from adipose tissue that are taken up by the liver,

    metabolized and released in VLDL particles (Nonogaki et al., 1994). In addition, TNF-

    was found to suppress lipoprotein lipase synthesis, which could contribute to

    hypertriglyceridemia by decreasing the rate of triglyceride clearance in the periphery (Figure

    3) (Kawakami et al., 1987). Additional cell-autonomous mechanisms also contribute to

    increased lipolysis, as discussed in further detail below.

    In addition to the potential roles of SFAs as endogenous activators of TLR signaling, low

    levels of endotoxin derived from bowel flora are present in the portal circulation, with levels

    varying dependent on various factors, including diet (Amar et al., 2008; Backhed et al.,

    2004). The concentration of LPS required to increase VLDL secretion is orders of

    magnitude lower than that required to induce sepsis, raising the possibility that endogenous

    endotoxins may contribute to low levels of TLR4 activation (Feingold et al., 1992b). A

    recent study of TLR4/mice indicated a modestly exaggerated hypoglycemic response to

    fasting and elevated levels of circulating and skeletal muscle triglycerides. No effect on

    insulin sensitivity was observed, suggesting a role of TLR4 in glucose and lipid metabolism

    independent of insulin during fasting.(Pang et al., 2010)

    Toll like receptors, including TLR4, are most highly expressed in cells that play direct roles

    in innate immune responses, such as macrophages, that are major sources of cytokines and

    chemokines in the setting of infection and tissue injury. However, TLRs are also expressed

    on a broad range of other cells types, including adipocytes, hepatocytes, and skeletal muscle

    cells (e.g., (Lang et al., 2003; Matsumura et al., 2000; Shi et al., 2006)). The expression of

    Glass and Olefsky Page 7

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    8/23

    TLRs in these cells may enable them to complement the functions of specialized immune

    cells through, for example, the production of chemokines and cytokines. Recent studies

    further suggest that TLR signaling within parenchymal cells of the liver, adipose tissue and

    skeletal muscle serves to modulate diverse aspects of their specific roles in metabolism. As

    noted above, inhibitory effects of LPS and cytokines such as TNFand IL1on insulin

    signaling pathways are well established (reviewed in (Olefsky and Glass, 2010)). Inhibition

    of insulin signaling can itself potentially impact on lipid metabolism, but there is substantialevidence that inflammatory signaling pathways affect many aspects of lipid metabolism

    independently of their effects on insulin. For example, direct infusion of a relatively low

    dose of TNFinto normal human subjects increased whole body lipolysis by 40% and a

    corresponding increase of free fatty acids over a 4h time period (Plomgaard et al., 2008). No

    changes in glucose homeostasis were observed in this acute treatment protocol.

    Because TLR4 signaling stimulates expression of a broad range of cytokines that are linked

    to insulin resistance, such as TNFand IL1, it will be of particular interest to tease apart

    direct and indirect effects on lipid homeostasis in specific cell types in vivo. Bone marrow

    reconstitution experiments, in which wild type bone marrow was replaced with TLR4-

    deficient bone marrow, conferred protection against diet/obesity-induced insulin resistance,consistent with important roles in macrophages and other bone marrow-derived cells (Saberi

    et al., 2009). However, these studies did not address the effects of hematopoietic deletion of

    TLR4 signaling on lipid metabolism.

    In addition to contributing to development of insulin resistance, TLRs have been shown to

    contribute to the development of atherosclerosis (reviewed in (Coban et al., 2009; Curtiss

    and Tobias, 2009; Olefsky and Glass, 2010)). In this context, TLR-driven inflammation is

    thought to contribute to pathology by amplifying recruitment of macrophages and other

    immune cells to atherosclerotic lesions and to influence processes that promote

    modifications of LDL that increase its uptake by macrophages and smooth muscle cells,

    such as oxidation (reviewed in (Rocha and Libby, 2009)). As discussed in further detail

    below, studies in cell culture models suggest important effects of TLR4 signaling in

    hepatocytes, skeletal muscle cells, and adipocytes. Development of a conditional allele of

    TLR4 would be a valuable resource for investigation of direct and indirect effects of TLR4

    engagement in tissues other than the hematopoietic system.

    Inflammation and lipid metabolism in the liver

    Early studies of the effects of LPS on lipoprotein metabolism demonstrated that low doses

    of LPS stimulated hepatic de novo fatty acid synthesis and lipolysis, both of which provided

    a source of fatty acids for the increase in hepatic triglyceride production (Figure 3)

    (Feingold et al., 1992b). The molecular basis for this effect, which is observed within a few

    hours of LPS treatment, has not been clearly established. Becaues LPS injection inducesnumerous cytokines, including TNFand IL1, effects in liver could be primary responses

    to LPS or secondary responses to cytokines, or both. De novo fatty acid biosynthesis is

    primarily regulated by sterol response element binding protein 1c (SREBP1c) (reviewed in

    (Horton, 2002)). Liver X receptors induce expression of SREBP1c, as well as directly

    regulate numerous genes involved in fatty acid biosynthesis and metabolism (Repa et al.,

    Glass and Olefsky Page 8

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    9/23

    2000; Schultz et al., 2000). Increased production of fatty acids would therefore presumably

    require activation of the SREBP pathway either at a transcriptional or post-transcriptional

    level, but this has not been directly shown in an LPS model. A recent study of TNFin the

    livers of fasting mice indicated that TNFinterferes with adaptation to fasting and activates

    the insulin-Insig-SREBP signaling pathway (Fon Tacer et al., 2007). Significant alterations

    were observed in lipid homeostasis, including reduction of HDL-cholesterol, an increase in

    LDL-cholesterol, and elevated expression of cholesterogenic genes. This was accompaniedby an increase of pre-cholesterol metabolites and suppression of cholesterol elimination

    through bile acids. At the transcriptional level, a shift in the expression of genes promoting

    fatty oxidation toward those involved in fatty acid synthesis was observed. Consistent with

    these studies, a different form of inflammatory stress induced by casein injection increased

    hepatic cholesterol accumulation and enhanced sterol regulatory element binding protein 2

    (SREBP2), low-density lipoprotein receptor (LDLr) and HMGCoA-reductase mRNA and

    protein expression in livers of C57BL/6J mice and in HepG2 cells (Ma et al., 2008). In

    addition, treatment of HepG2 cells with IL-6 or IL-1increased nuclear SREBP2, increased

    HMGCoA reductase protein levels and partially inhibited the normal feedback inhibition of

    the cholesterol biosynthetic pathway in response to cholesterol loading (Zhao et al., 2011).

    LPS-induced changes in the liver sinusoidal endothelial cells predicted to reduce access ofhepatocytes to circulating lipoproteins has also been suggested to contribute to

    hypertriglyceridemia (Cheluvappa et al., 2010). Hepatocyte overexpression of IB kinase

    (IKK), the major kinase required for activation of NFB downstream of TLR, TNFand

    IL1signaling was recently shown to result in increased VLDL synthesis (van Diepen et al.,

    2011), indicating that specific activation of IKKwithin hepatocytes is sufficient to induce

    hypertriglyceridemia.

    Many questions remain regarding the impact of inflammation on lipid metabolism in the

    liver. In particular, defining the mechanisms that connect inflammatory signaling pathways

    to altered functions of the SREBPs is of particular interest, as these mechanisms might be

    amenable to therapeutic interventions. In addition, there is a very limited understanding ofthe impact of inflammation on specific lipid species within broad lipid categories (fatty

    acids, sterols, phospholipids, triglycerides, etc). Inflammatory responses have the potential

    to reprogram lipid metabolic pathways so as to change membrane properties, signaling

    events, energy utilization, and other basic cellular processes. The outcome of inflammatory

    stimuli on lipid metabolism may also be influenced by the sterol and fatty acid composition

    of the diet, for example the relative content of -3 and -6 fatty acids.

    Inflammation and lipid metabolism in muscle

    Recent studies in human skeletal muscle, rodent skeletal muscle, and skeletal muscle cell

    cultures using both gain and loss of function approaches provided evidence that TLR4

    signaling reduces fatty acid oxidation (Frisard et al., 2010; Pang et al., 2010) and that actions

    in muscle may contribute to increased circulating triglycerides (Figure 3). Fasting animals

    with a loss of TLR4 function exhibited increased oxidative capacity in skeletal muscle,

    lower levels of triglycerides and non-esterified free fatty acids. Changes in substrate

    metabolism under conditions of low (metabolic) LPS concentrations were suggested to be

    Glass and Olefsky Page 9

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    10/23

    independent of skeletal muscle-derived pro-inflammatory cytokine production (Frisard et al.,

    2010).

    One important consequence of inflammatory signaling in muscle is an increase in

    production of ceramide, which as discussed above, may be a key intermediate that links

    inflammation to insulin resistance by blocking activation of Akt/PKB. Studies in c2c12

    myoblasts indicated that the LPS or saturated fatty acid-induced ceramide production was

    associated with IKK-dependent upregulation of genes driving ceramide biosynthesis.

    Importantly, increased ceramide production was not required for TLR4-dependent induction

    of inflammatory cytokines, but it was essential for TLR4-dependent insulin resistance

    (Holland et al., 2011).

    Interleukin (IL)-6 is an inflammatory cytokine that is chronically elevated in type 2 diabetes

    but also during exercise. Acute (4h) infusion of physiological concentrations of IL4 into

    healthy human subjects had no effects on glucose levels but increased systemic fatty acid

    oxidation approximately twofold after 60 min, which remained elevated by 2h after the

    infusion (Wolsk et al., 2010). The increase in oxidation was followed by an increase in

    systemic lipolysis. Adipose tissue lipolysis and fatty acid kinetics were unchanged, but

    skeletal muscle unidirectional fatty acid and glycerol release was increased, indicative of an

    increase in lipolysis. The increased lipolysis in muscle could account for the systemic

    changes. These effects in muscle were correlated with activation of STAT3, a downstream

    target of IL-6, whereas no changes in phosphorylated AMP-activated protein kinase or

    acetyl-CoA carboxylase levels could be observed. Il-6 could thus contribute to some of the

    observed changes in skeletal muscle lipid metabolism seen in the context of TLR4

    activation, acting as an autocrine factor or as a paracrine/endocrine factor released by

    immune cells such as macrophages.

    As in the case of the liver, inflammation induces substantial changes in fatty acid

    metabolism in skeletal muscle, but the mechanisms remain very poorly understood and the

    impact on classes of lipids beyond fatty acids and ceramides have not been extensively

    evaluated. The differential effects of LPS and IL-6 signaling underscore the complex nature

    of the inflammatory response, which rather than being a single entity evolves as a highly

    variable and context dependent process. IL-6 signaling exerts effects on gene expression by

    controlling phosphorylation of STAT3. Intriguingly, STAT3 has been shown to directly

    regulate the activities of the mitochondrial electron transport chain in liver and heart, which

    could provide a connection between effects of IL-6 signaling and fatty acid oxidation in

    skeletal muscle (Wegrzyn et al., 2009).

    Inflammation and lipid metabolism in adipose tissue

    LPS, TNFand IL1induce lipolysis in primary adipocytes and adipocyte cell lines,indicating a cell-autonomous effect that can act independently of catecholamine- induced

    lipolysis (Figure 3) (Feingold et al., 1992a; Franchini et al., 2010; Kawakami et al., 1987;

    Ryden et al., 2002; Zhang et al., 2002; Zu et al., 2008). Inhibitor studies suggest that TNF

    increases lipolysis in adipocytes by activation of mitogen-activated protein kinase kinase

    (MEK), extracellular signal-related kinase (ERK), Jun N-terminal kinase (JNK) and

    Glass and Olefsky Page 10

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    11/23

    elevation of intracellular cAMP leading to activation of PKA, with some variations in

    relative importance observed dependent on the specific cellular system used (Ryden et al.,

    2002; Zhang et al., 2002). PKA activation directly affects lipolysis by phosphorylating the

    lipid droplet associated protein perilipin (Granneman et al., 2007). This results in release of

    CGI-58/Abhd5, which binds to and co-activates adipose triglyceride lipase (ATGL)

    (Granneman et al., 2009). ATGL has recently been shown to be the major lipase responsible

    for TNF-induced lipolysis in 3T3L1 adipocytes (Yang et al., 2011). These studies alsoprovided evidence that TNFsignaling profoundly suppresses the expression of G0S2, a

    recently identified endogenous inhibitor of ATGL. Thus, TNFsignaling acts

    simultaneously to stimulate activators and suppress inhibitors of ATGL activity. Given the

    central importance of triglyceride hydrolysis in adipose tissue, liver and skeletal muscle in

    overall fatty acid metabolism, it will be of great interest to explore the importance of these

    regulatory pathways in each tissue in vivo. Intriguingly, salicylates block the lipolytic

    response to TNF (Zu et al., 2008), an effect that may contribute to their insulin-sensitizing

    activities.

    The production of TNFand IL1could potentially account for the effects of LPS on

    adipose tissue lipolysis , but the use of adrenergic or interleukin-1 receptor antagonists andTNF-neutralizing antibodies did not abolish increases in serum levels of FFA and

    triglycerides in endotoxemic rats (Feingold et al., 1992b; Nonogaki et al., 1994). Notably,

    neither adipocytokine secretion nor NFB activation is involved in endotoxin-induced

    lipolysis (Zu et al., 2009). In contrast to catecholamines and TNF, endotoxin stimulates

    lipolysis without elevating cAMP production and activating protein kinase A and protein

    kinase C. Instead, endotoxin induces phosphorylation of Raf-1, MEK1/2, and ERK1/2.

    Upon inhibition of ERK1/2 but not JNK and p38 MAPK, endotoxin-stimulated lipolysis

    ceases. Endotoxin causes down-regulation and phosphorylation of perilipin, which would be

    expected to result in activation of ATGL by CGI-58.

    Intriguingly, recent studies have found that deletion of BLT1, a receptor for leukotriene B4

    (LTB4), protects mice from high fat diet-induced insulin resistance (Spite et al., 2011).

    BLT1-deficient mice exhibited a marked reduction in accumulation of classically activated

    adipose tissue macrophages and a reduction in pro-inflammatory cytokines. Although LTB4

    itself was not directly measured in these assays, these findings suggest that the pro-

    inflammatory response of adipose tissue to a high fat diet results in LTB4 production from

    arachidonic acid, which serves to recruit LTB4-positive monocytes into adipose tissue.

    Pharmacological blockade of BLT1 might thus have insulin-sensitizing effects in the context

    of obesity.

    Interferon regulatory factor 4 (IRF4), which plays key roles in regulation of function of

    immune cells, was recently shown to regulate the transcriptional response to nutrient

    availability in adipocytes. Fasting induces IRF4 in an insulin- and FoxO1-dependent

    manner. IRF4 is required for lipolysis, at least in part due to direct effects on the expression

    of ATGL and hormone-sensitive lipase. Conversely, reduction of IRF4 enhances lipid

    synthesis (Eguchi et al., 2011).

    Glass and Olefsky Page 11

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    12/23

    The extent to which inflammation-driven lipolysis in adipose tissue contributes to insulin

    resistance remains to be clearly established. The influence of free fatty acids on

    inflammation, described above, and the proximity of adipose tissue macrophages to

    lipolysis-generated FFAs suggests the possibility of a local positive feedback loop that

    amplifies insulin resistance. It will therefore be of interest to evaluate the consequences of

    inhibiting lipolysis on inflammation and insulin resistance in vivo.

    Inflammation and lipid metabolism in the macrophage

    Based on the key pathogenic roles of macrophages and TLRs in the development of

    atherosclerosis (Moore and Tabas, 2011) and diabetes (Olefsky and Glass, 2010), the LIPID

    MAPS consortium recently reported a comprehensive analysis of the effects of TLR4

    signaling on the lipidome and transcriptome of RAW264.7 macrophages (Dennis et al.,

    2010). These studies demonstrated immediate responses in fatty acid metabolism,

    represented by increases in eicosanoid synthesis, and delayed responses characterized by

    alterations in sphingolipid and sterol biosynthesis. TLR4 activation generated increases in

    almost every category of sphingolipid analyzed, including free sphingoid bases and their

    phosphates (e.g. sphingosine- 1-phosphate), ceramides, sphingomyelins, and

    glycosphingolipids. It would not be surprising for inflammatory signaling to have similar

    effects on the lipidomes of other metabolically important cell types, including adipocytes,

    hepatocytes and skeletal muscle cells. Consistent with this, the impact of TLR4 signaling on

    ceramide production in RAW264.7 cells and C2C12 skeletal muscle cells is similar (Holland

    et al., 2011). Notably, lipid remodeling of glycerolipids, glycerophospholipids, and prenols

    was also observed in activated RAW264.7 cells, indicating that TLR4 signaling leads to

    alterations in a majority of mammalian lipid categories (Dennis et al., 2010). Thus, it is

    likely that extension of these methods to primary cells and tissues would be highly

    informative. Along these lines, a recent lipidomic analysis of adipose tissue macrophages

    from ob/ob mice indicates that macrophage triglycerides accumulate during the transition

    from insulin-sensitive to obese/insulin resistance. Treatment with a PPARagonist

    prevented triglyceride accumulation in these cells (Prieur et al., 2011).

    Signaling pathways that connect inflammation to lipid metabolism

    The TLR, IL1and TNFsignaling pathways are best understood with respect to their roles

    in initiation and amplification of innate immune responses through the activation of latent

    transcription factors that include members of the NFB, AP-1, and IRF gene families. Much

    less is known, however, with respect to the mechanisms by which inflammatory signaling

    pathways affect lipid metabolism. Regulation of post-translational modifications, e.g.,

    perilipin phosphorylation, is likely to play an important role. In addition, significant effects

    of inflammatory signaling pathways on the expression and function of transcription factors

    that regulate lipid homeostasis have been identified. For example, TLR4 activation is apotent suppressor of PPARexpression in the macrophage (Barish et al., 2005), and TLR3

    and TLR4 inhibit LXR function by a mechanism that involves the TRIF adapter protein

    (Castrillo et al., 2003). It will be of interest to explore whether inflammatory signaling

    influences the expression or processing of the SREBP transcription factors that play central

    roles in fatty acid and cholesterol biosynthesis. A large number of other transcription factors

    Glass and Olefsky Page 12

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    13/23

    also influence lipid metabolism, including PPAR, PPAR, Forkhead transcription factors,

    KLFs and others. The emergence of sensitive and quantitative methods to measure major

    and minor lipid species by mass spectroscopy, coupled with genome-wide measurement of

    gene expression, are likely to important new insights into the relationships between

    inflammation, lipid metabolism and disease in the years ahead.

    Conclusions

    Striking progress has been made in the last few years in deconvoluting the complex

    relationships between lipid metabolism and inflammation that contribute to metabolic

    disease. In particular, new molecular mechanisms accounting for pro-inflammatory effects

    of saturated FFAs and anti-inflammatory effects of -3 PUFAs have been discovered. In

    addition, the production of ceramide in response to inflammatory signaling in skeletal

    muscle and macrophages is emerging as an important mechanism contributing to insulin

    resistance. At the level of the impact of inflammation on lipid metabolism, recent studies

    have uncovered likely molecular mechanisms by which inflammatory mediators such as

    LPS and TNFfunction to stimulate lipolysis in adipose tissue, through the indirect

    regulation of ATGL. Studies extending the analysis of these mechanisms in vivo should be

    productive lines of investigation for the future. Many aspects of the relationship between

    inflammation and lipid metabolism remain poorly understood. In particular, mechanisms by

    which inflammatory signaling regulates the functions of SREBPs, LXRs and PPARs remain

    almost completely unknown, and are potential targets for therapeutic intervention. The

    ability to investigate the extent to which inflammation reprograms lipid metabolism to alter

    the production, catabolism and inter-conversion of specific lipid species has until recently

    been significantly limited by technology. The recent development of improved

    instrumentation and methodologies for analysis of diverse lipid species by mass

    spectrometry is now enabling substantial efforts to understand how the lipidome is regulated

    at a more global level. It will be of particular interest to bring the combination of lipidomics

    and transcriptomics to the analysis of the impact of dietary interventions on lipid metabolismin key metabolic organs. Fatty acid composition of the diet can vary widely, from being

    relatively high in saturated (animal-derived) FFAs such as palmitic acid that have direct pro-

    inflammatory effects, to relatively high in -6 (e.g., corn oil-derived) FFAs such as linoleic

    acid that can be further elongated, de-saturated and converted to pro-inflammatory

    prostaglandins, to relatively high in -3 (e.g., fatty fish and certain seed oil-derived) FFAs

    that exert anti-inflammatory effects. As there is now substantial interest in the agricultural

    and food products industries to develop new food oils and food products with increased

    levels of -3 PUFAs, the rationale, specific form and advisability of these efforts should rest

    on a deeper knowledge of the relationship between lipid metabolism, inflammation and

    disease.

    References

    Amar J, Burcelin R, Ruidavets JB, Cani PD, Fauvel J, Alessi MC, Chamontin B, Ferrieres J. Energy

    intake is associated with endotoxemia in apparently healthy men. Am J Clin Nutr. 2008; 87:1219

    1223. [PubMed: 18469242]

    Attie AD, Scherer PE. Adipocyte metabolism and obesity. Journal of lipid research. 2009;

    50(Suppl):S395S399. [PubMed: 19017614]

    Glass and Olefsky Page 13

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    14/23

    Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut

    microbiota as an environmental factor that regulates fat storage. Proceedings of the National

    Academy of Sciences of the United States of America. 2004; 101:1571815723. [PubMed:

    15505215]

    Barish GD, Downes M, Alaynick WA, Yu RT, Ocampo CB, Bookout AL, Mangelsdorf DJ, Evans

    RM. A Nuclear Receptor Atlas: macrophage activation. Molecular endocrinology. 2005; 19:2466

    2477. [PubMed: 16051664]

    Boden G, Jadali F, White J, Liang Y, Mozzoli M, Chen X, Coleman E, Smith C. Effects of fat on

    insulin-stimulated carbohydrate metabolism in normal men. The Journal of clinical investigation.

    1991; 88:960966. [PubMed: 1885781]

    Brun P, Castagliuolo I, Di Leo V, Buda A, Pinzani M, Palu G, Martines D. Increased intestinal

    permeability in obese mice: new evidence in the pathogenesis of nonalcoholic steatohepatitis. Am J

    Physiol Gastrointest Liver Physiol. 2007; 292:G518525. [PubMed: 17023554]

    Calder PC. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr.

    2006; 83:1505S1519S. [PubMed: 16841861]

    Cao H, Gerhold K, Mayers JR, Wiest MM, Watkins SM, Hotamisligil GS. Identification of a lipokine,

    a lipid hormone linking adipose tissue to systemic metabolism. Cell. 2008; 134:933944. [PubMed:

    18805087]

    Castrillo A, Joseph SB, Vaidya SA, Haberland M, Fogelman AM, Cheng G, Tontonoz P. Crosstalk

    between LXR and toll-like receptor signaling mediates bacterial and viral antagonism of cholesterol

    metabolism. Molecular cell. 2003; 12:805816. [PubMed: 14580333]

    Cheluvappa R, Denning GM, Lau GW, Grimm MC, Hilmer SN, Le Couteur DG. Pathogenesis of the

    hyperlipidemia of Gram-negative bacterial sepsis may involve pathomorphological changes in

    liver sinusoidal endothelial cells. International journal of infectious diseases : IJID : official

    publication of the International Society for Infectious Diseases. 2010; 14:e857e867. [PubMed:

    20609608]

    Coban C, Ishii KJ, Akira S. Immune interventions of human diseases through toll-like receptors.

    Advances in experimental medicine and biology. 2009; 655:6380. [PubMed: 20047036]

    Creely SJ, McTernan PG, Kusminski CM, Fisher M, Da Silva NF, Khanolkar M, Evans M, Harte AL,

    Kumar S. Lipopolysaccharide activates an innate immune system response in human adipose

    tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab. 2007; 292:E740E747.

    [PubMed: 17090751]

    Curtiss LK, Tobias PS. Emerging role of Toll-like receptors in atherosclerosis. Journal of lipid

    research. 2009; 50(Suppl):S340S345. [PubMed: 18980945]

    Dasgupta S, Bhattacharya S, Biswas A, Majumdar SS, Mukhopadhyay S, Ray S. NF-kappaB mediates

    lipid-induced fetuin-A expression in hepatocytes that impairs adipocyte function effecting insulin

    resistance. Biochem J. 2010; 429:451462. [PubMed: 20482516]

    Dbaibo GS, El-Assaad W, Krikorian A, Liu B, Diab K, Idriss NZ, El-Sabban M, Driscoll TA, Perry

    DK, Hannun YA. Ceramide generation by two distinct pathways in tumor necrosis factor alpha-

    induced cell death. FEBS Lett. 2001; 503:712. [PubMed: 11513845]

    Dennis EA, Cao J, Hsu YH, Magrioti V, Kokotos G. Phospholipase A2 enzymes: physical structure,

    biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem

    Rev. 2011; 111:61306185. [PubMed: 21910409]

    Dennis EA, Deems RA, Harkewicz R, Quehenberger O, Brown HA, Milne SB, Myers DS, Glass CK,

    Hardiman G, Reichart D, et al. A mouse macrophage lipidome. The Journal of biological

    chemistry. 2010; 285:3997639985. [PubMed: 20923771]

    Eguchi J, Wang X, Yu S, Kershaw EE, Chiu PC, Dushay J, Estall JL, Klein U, Maratos-Flier E, Rosen

    ED. Transcriptional control of adipose lipid handling by IRF4. Cell metabolism. 2011; 13:249259. [PubMed: 21356515]

    Erridge C, Samani NJ. Saturated fatty acids do not directly stimulate Toll-like receptor signaling.

    Arterioscler Thromb Vasc Biol. 2009; 29:19441949. [PubMed: 19661481]

    Feingold KR, Doerrler W, Dinarello CA, Fiers W, Grunfeld C. Stimulation of lipolysis in cultured fat

    cells by tumor necrosis factor, interleukin-1, and the interferons is blocked by inhibition of

    prostaglandin synthesis. Endocrinology. 1992a; 130:1016. [PubMed: 1370149]

    Glass and Olefsky Page 14

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    15/23

    Feingold KR, Staprans I, Memon RA, Moser AH, Shigenaga JK, Doerrler W, Dinarello CA, Grunfeld

    C. Endotoxin rapidly induces changes in lipid metabolism that produce hypertriglyceridemia: low

    doses stimulate hepatic triglyceride production while high doses inhibit clearance. Journal of lipid

    research. 1992b; 33:17651776. [PubMed: 1479286]

    Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C,

    Shoelson S, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells

    that affect metabolic parameters. Nature medicine. 2009; 15:930939.

    Feve B, Bastard JP. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev

    Endocrinol. 2009; 5:305311. [PubMed: 19399017]

    Fielding BA, Callow J, Owen RM, Samra JS, Matthews DR, Frayn KN. Postprandial lipemia: the

    origin of an early peak studied by specific dietary fatty acid intake during sequential meals. Am J

    Clin Nutr. 1996; 63:3641. [PubMed: 8604667]

    Fon Tacer K, Kuzman D, Seliskar M, Pompon D, Rozman D. TNF-alpha interferes with lipid

    homeostasis and activates acute and proatherogenic processes. Physiological genomics. 2007;

    31:216227. [PubMed: 17566076]

    Franchini M, Monnais E, Seboek D, Radimerski T, Zini E, Kaufmann K, Lutz T, Reusch C,

    Ackermann M, Muller B, et al. Insulin resistance and increased lipolysis in bone marrow derived

    adipocytes stimulated with agonists of Toll-like receptors. Horm Metab Res. 2010; 42:703709.

    [PubMed: 20603780]

    Frisard MI, McMillan RP, Marchand J, Wahlberg KA, Wu Y, Voelker KA, Heilbronn L, Haynie K,

    Muoio B, Li L, et al. Toll-like receptor 4 modulates skeletal muscle substrate metabolism.

    American journal of physiology Endocrinology and metabolism. 2010; 298:E988E998. [PubMed:

    20179247]

    Fung-Leung WP. Phosphoinositide 3-kinase delta (PI3Kdelta) in leukocyte signaling and function.

    Cell Signal. 2011; 23:603608. [PubMed: 20940048]

    Gallin JI, Kaye D, OLeary WM. Serum lipids in infection. The New England journal of medicine.

    1969; 281:10811086. [PubMed: 5824173]

    Ghigo A, Damilano F, Braccini L, Hirsch E. PI3K inhibition in inflammation: Toward tailored

    therapies for specific diseases. Bioessays. 2010; 32:185196. [PubMed: 20162662]

    Granneman JG, Moore HP, Granneman RL, Greenberg AS, Obin MS, Zhu Z. Analysis of lipolytic

    protein trafficking and interactions in adipocytes. The Journal of biological chemistry. 2007;

    282:57265735. [PubMed: 17189257]

    Granneman JG, Moore HP, Krishnamoorthy R, Rathod M. Perilipin controls lipolysis by regulating the

    interactions of AB-hydrolase containing 5 (Abhd5) and adipose triglyceride lipase (Atgl). The

    Journal of biological chemistry. 2009; 284:3453834544. [PubMed: 19850935]

    Harris HW, Gosnell JE, Kumwenda ZL. The lipemia of sepsis: triglyceride-rich lipoproteins as agents

    of innate immunity. Journal of endotoxin research. 2000; 6:421430. [PubMed: 11521066]

    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. Nature medicine. 2005; 11:9094.

    Holland WL, Bikman BT, Wang LP, Yuguang G, Sargent KM, Bulchand S, Knotts TA, Shui G, Clegg

    DJ, Wenk MR, et al. Lipid-induced insulin resistance mediated by the proinflammatory receptor

    TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. The Journal of clinical

    investigation. 2011; 121:18581870. [PubMed: 21490391]

    Holzer RG, Park EJ, Li N, Tran H, Chen M, Choi C, Solinas G, Karin M. Saturated fatty acids induce

    c-Src clustering within membrane subdomains, leading to JNK activation. Cell. 2011; 147:173

    184. [PubMed: 21962514]

    Horton JD. Sterol regulatory element-binding proteins: transcriptional activators of lipid synthesis.

    Biochemical Society transactions. 2002; 30:10911095. [PubMed: 12440980]

    Hotamisligil GS. Mechanisms of TNF-alpha-induced insulin resistance. Exp Clin Endocrinol Diabetes.

    1999; 107:119125. [PubMed: 10320052]

    Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006; 444:860867. [PubMed:

    17167474]

    Glass and Olefsky Page 15

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    16/23

    Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha:

    direct role in obesity-linked insulin resistance. Science. 1993; 259:8791. [PubMed: 7678183]

    Hupfeld, CJ.; Courtney, CH.; Olefsky, JM. Chapter 41: Type 2 Diabetes Mellitus: Etiology,

    Pathogenesis, and Natural History. In: DeGroot, aJLJLJ., editor. Endocrinology. Philadelphia:

    Elsevier/Saunders; 2006.

    Hwang D, Rhee SH. Receptor-mediated signaling pathways: potential targets of modulation by dietary

    fatty acids. Am J Clin Nutr. 1999; 70:545556. [PubMed: 10500025]

    Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin resistance in human muscle isassociated with changes in diacylglycerol, protein kinase C, and IkappaB-alpha. Diabetes. 2002;

    51:20052011. [PubMed: 12086926]

    Itoh Y, Kawamata Y, Harada M, Kobayashi M, Fujii R, Fukusumi S, Ogi K, Hosoya M, Tanaka Y,

    Uejima H, et al. Free fatty acids regulate insulin secretion from pancreatic beta cells through

    GPR40. Nature. 2003; 422:173176. [PubMed: 12629551]

    Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2

    diabetes. Nature. 2006; 444:840846. [PubMed: 17167471]

    Kawakami M, Murase T, Ogawa H, Ishibashi S, Mori N, Takaku F, Shibata S. Human recombinant

    TNF suppresses lipoprotein lipase activity and stimulates lipolysis in 3T3-L1 cells. Journal of

    biochemistry. 1987; 101:331338. [PubMed: 3495531]

    Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M, Fischer-Posovszky P,

    Barth TF, Dragun D, Skurk T, et al. T-lymphocyte infiltration in visceral adipose tissue: a primary

    event in adipose tissue inflammation and the development of obesity-mediated insulin resistance.

    Arterioscler Thromb Vasc Biol. 2008; 28:13041310. [PubMed: 18420999]

    Kumar H, Kawai T, Akira S. Toll-like receptors and innate immunity. Biochem Biophys Res

    Commun. 2009; 388:621625. [PubMed: 19686699]

    Kumashiro N, Erion DM, Zhang D, Kahn M, Beddow SA, Chu X, Still CD, Gerhard GS, Han X,

    Dziura J, et al. Cellular mechanism of insulin resistance in nonalcoholic fatty liver disease. Proc

    Natl Acad Sci U S A. 2011; 108:1638116385. [PubMed: 21930939]

    Lang CH, Silvis C, Deshpande N, Nystrom G, Frost RA. Endotoxin stimulates in vivo expression of

    inflammatory cytokines tumor necrosis factor alpha, interleukin-1beta, 6, and high-mobility-

    group protein-1 in skeletal muscle. Shock. 2003; 19:538546. [PubMed: 12785009]

    Lee JY, Plakidas A, Lee WH, Heikkinen A, Chanmugam P, Bray G, Hwang DH. Differential

    modulation of Toll-like receptors by fatty acids: preferential inhibition by n-3 polyunsaturated

    fatty acids. Journal of lipid research. 2003; 44:479486. [PubMed: 12562875]

    Lee JY, Sohn KH, Rhee SH, Hwang D. Saturated fatty acids, but not unsaturated fatty acids, induce

    the expression of cyclooxygenase-2 mediated through Toll-like receptor 4. The Journal of

    biological chemistry. 2001; 276:1668316689. [PubMed: 11278967]

    Liu J, Divoux A, Sun J, Zhang J, Clement K, Glickman JN, Sukhova GK, Wolters PJ, Du J, Gorgun

    CZ, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced

    obesity and diabetes in mice. Nature medicine. 2009; 15:940945.

    Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue

    macrophage polarization. J Clin Invest. 2007a; 117:175184. [PubMed: 17200717]

    Lumeng CN, Deyoung SM, Bodzin JL, Saltiel AR. Increased inflammatory properties of adipose

    tissue macrophages recruited during diet-induced obesity. Diabetes. 2007b; 56:1623. [PubMed:

    17192460]

    Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. The Journal of

    clinical investigation. 2011; 121:21112117. [PubMed: 21633179]

    Luttrell LM, Lefkowitz RJ. The role of beta-arrestins in the termination and transduction of G-protein-

    coupled receptor signals. J Cell Sci. 2002; 115:455465. [PubMed: 11861753]

    Ma KL, Ruan XZ, Powis SH, Chen Y, Moorhead JF, Varghese Z. Inflammatory stress exacerbates

    lipid accumulation in hepatic cells and fatty livers of apolipoprotein E knockout mice. Hepatology.

    2008; 48:770781. [PubMed: 18752326]

    Matsumura T, Ito A, Takii T, Hayashi H, Onozaki K. Endotoxin and cytokine regulation of toll-like

    receptor (TLR) 2 and TLR4 gene expression in murine liver and hepatocytes. Journal of interferon

    Glass and Olefsky Page 16

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    17/23

    & cytokine research : the official journal of the International Society for Interferon and Cytokine

    Research. 2000; 20:915921.

    Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011; 145:341355.

    [PubMed: 21529710]

    Nguyen MT, Favelyukis S, Nguyen AK, Reichart D, Scott PA, Jenn A, Liu-Bryan R, Glass CK, Neels

    JG, Olefsky JM. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is

    activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. The

    Journal of biological chemistry. 2007; 282:3527935292. [PubMed: 17916553]

    Nguyen MT, Satoh H, Favelyukis S, Babendure JL, Imamura T, Sbodio JI, Zalevsky J, Dahiyat BI,

    Chi NW, Olefsky JM. JNK and tumor necrosis factor-alpha mediate free fatty acid-induced insulin

    resistance in 3T3-L1 adipocytes. The Journal of biological chemistry. 2005; 280:3536135371.

    [PubMed: 16085647]

    Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K,

    Sugiura S, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue

    inflammation in obesity. Nature medicine. 2009; 15:914920.

    Nonogaki K, Moser AH, Feingold KR, Grunfeld C. Alpha-adrenergic receptors mediate the

    hypertriglyceridemia induced by endotoxin, but not tumor necrosis factor, in rats. Endocrinology.

    1994; 135:26442650. [PubMed: 7988454]

    Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, Olefsky JM.

    GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-

    sensitizing effects. Cell. 2010; 142:687698. [PubMed: 20813258]

    Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annual review of

    physiology. 2010; 72:219246.

    Pang S, Tang H, Zhuo S, Zang YQ, Le Y. Regulation of fasting fuel metabolism by toll-like receptor

    4. Diabetes. 2010; 59:30413048. [PubMed: 20855545]

    Park BS, Song DH, Kim HM, Choi BS, Lee H, Lee JO. The structural basis of lipopolysaccharide

    recognition by the TLR4-MD-2 complex. Nature. 2009; 458:11911195. [PubMed: 19252480]

    Park E, Wong V, Guan X, Oprescu AI, Giacca A. Salicylate prevents hepatic insulin resistance caused

    by short-term elevation of free fatty acids in vivo. J Endocrinol. 2007; 195:323331. [PubMed:

    17951543]

    Park JS, Svetkauskaite D, He Q, Kim JY, Strassheim D, Ishizaka A, Abraham E. Involvement of toll-

    like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. The Journal of

    biological chemistry. 2004; 279:73707377. [PubMed: 14660645]

    Plomgaard P, Fischer CP, Ibfelt T, Pedersen BK, van Hall G. Tumor necrosis factor-alpha modulates

    human in vivo lipolysis. The Journal of clinical endocrinology and metabolism. 2008; 93:543549.

    [PubMed: 18029463]

    Polozova A, Salem N Jr. Role of liver and plasma lipoproteins in selective transport of n-3 fatty acids

    to tissues: a comparative study of 14C-DHA and 3H-oleic acid tracers. J Mol Neurosci. 2007;

    33:5666. [PubMed: 17901547]

    Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos

    C, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene.

    Science. 1998; 282:20852088. [PubMed: 9851930]

    Prieur X, Mok CY, Velagapudi VR, Nunez V, Fuentes L, Montaner D, Ishikawa K, Camacho A,

    Barbarroja N, ORahilly S, et al. Differential lipid partitioning between adipocytes and tissue

    macrophages modulates macrophage lipotoxicity and M2/M1 polarization in obese mice. Diabetes.

    2011; 60:797809. [PubMed: 21266330]

    Proudman SM, Cleland LG, James MJ. Dietary omega-3 fats for treatment of inflammatory joint

    disease: efficacy and utility. Rheum Dis Clin North Am. 2008; 34:469479. [PubMed: 18638687]

    Qatanani M, Lazar MA. Mechanisms of obesity-associated insulin resistance: many choices on the

    menu. Genes Dev. 2007; 21:14431455. [PubMed: 17575046]

    Repa JJ, Liang G, Ou J, Bashmakov Y, Lobaccaro JM, Shimomura I, Shan B, Brown MS, Goldstein

    JL, Mangelsdorf DJ. Regulation of mouse sterol regulatory element-binding protein-1c gene

    (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes & development. 2000;

    14:28192830. [PubMed: 11090130]

    Glass and Olefsky Page 17

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    18/23

    Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;

    31:9861000. [PubMed: 21508345]

    Rocha VZ, Libby P. Obesity, inflammation, and atherosclerosis. Nature reviews Cardiology. 2009;

    6:399409.

    Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW, Shulman GI. Mechanism of

    free fatty acid-induced insulin resistance in humans. The Journal of clinical investigation. 1996;

    97:28592865. [PubMed: 8675698]

    Ryden M, Dicker A, van Harmelen V, Hauner H, Brunnberg M, Perbeck L, Lonnqvist F, Arner P.Mapping of early signaling events in tumor necrosis factor-alpha - mediated lipolysis in human fat

    cells. The Journal of biological chemistry. 2002; 277:10851091. [PubMed: 11694522]

    Saberi M, Woods NB, de Luca C, Schenk S, Lu JC, Bandyopadhyay G, Verma IM, Olefsky JM.

    Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue

    insulin resistance in high-fat-fed mice. Cell metabolism. 2009; 10:419429. [PubMed: 19883619]

    Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling the mechanism.

    Lancet. 2010; 375:22672277. [PubMed: 20609972]

    Savage DB, Petersen KF, Shulman GI. Disordered lipid metabolism and the pathogenesis of insulin

    resistance. Physiol Rev. 2007; 87:507520. [PubMed: 17429039]

    Schaeffler A, Gross P, Buettner R, Bollheimer C, Buechler C, Neumeier M, Kopp A, Schoelmerich J,

    Falk W. Fatty acid-induced induction of Toll-like receptor-4/nuclear factor-kappaB pathway in

    adipocytes links nutritional signalling with innate immunity. Immunology. 2009; 126:233245.

    [PubMed: 18624726]

    Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutrients and inflammation. The

    Journal of clinical investigation. 2008; 118:29923002. [PubMed: 18769626]

    Schultz JR, Tu H, Luk A, Repa JJ, Medina JC, Li L, Schwendner S, Wang S, Thoolen M, Mangelsdorf

    DJ, et al. Role of LXRs in control of lipogenesis. Genes & development. 2000; 14:28312838.

    [PubMed: 11090131]

    Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R,

    Witztum JL, Tsimikas S, et al. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent

    apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell metabolism. 2010;

    12:467482. [PubMed: 21035758]

    Senn JJ. Toll-like receptor-2 is essential for the development of palmitate-induced insulin resistance in

    myotubes. The Journal of biological chemistry. 2006; 281:2686526875. [PubMed: 16798732]

    Serhan CN, Chiang N. Endogenous pro-resolving and anti-inflammatory lipid mediators: a new

    pharmacologic genus. Br J Pharmacol. 2008; 1(153 Suppl):S200S215. [PubMed: 17965751]

    Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-

    resolution lipid mediators. Nat Rev Immunol. 2008; 8:349361. [PubMed: 18437155]

    Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty

    acid-induced insulin resistance. The Journal of clinical investigation. 2006; 116:30153025.

    [PubMed: 17053832]

    Shoelson SE, Herrero L, Naaz A. Obesity, inflammation, and insulin resistance. Gastroenterology.

    2007; 132:21692180. [PubMed: 17498510]

    Shulman GI. Cellular mechanisms of insulin resistance. J Clin Invest. 2000; 106:171176. [PubMed:

    10903330]

    Spite M, Hellmann J, Tang Y, Mathis SP, Kosuri M, Bhatnagar A, Jala VR, Haribabu B. Deficiency of

    the Leukotriene B4 Receptor, BLT-1, Protects against Systemic Insulin Resistance in Diet-Induced

    Obesity. Journal of immunology. 2011; 187:19421949.

    Staehr P, Hother-Nielsen O, Landau BR, Chandramouli V, Holst JJ, Beck-Nielsen H. Effects of freefatty acids per se on glucose production, gluconeogenesis, and glycogenolysis. Diabetes. 2003;

    52:260267. [PubMed: 12540595]

    Stephens JM, Pekala PH. Transcriptional repression of the C/EBP-alpha and GLUT4 genes in 3T3-L1

    adipocytes by tumor necrosis factor-alpha. Regulations is coordinate and independent of protein

    synthesis. The Journal of biological chemistry. 1992; 267:1358013584. [PubMed: 1618860]

    Glass and Olefsky Page 18

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    19/23

    Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R,

    Frazier WA, et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like

    receptor 4 and 6 heterodimer. Nat Immunol. 2010; 11:155161. [PubMed: 20037584]

    Stratford S, Hoehn KL, Liu F, Summers SA. Regulation of insulin action by ceramide: dual

    mechanisms linking ceramide accumulation to the inhibition of Akt/protein kinase B. The Journal

    of biological chemistry. 2004; 279:3660836615. [PubMed: 15220355]

    Summers SA. Sphingolipids and insulin resistance: the five Ws. Curr Opin Lipidol. 2010; 21:128135.

    [PubMed: 20216312]

    Tazoe H, Otomo Y, Kaji I, Tanaka R, Karaki SI, Kuwahara A. Roles of short-chain fatty acids

    receptors, GPR41 and GPR43 on colonic functions. J Physiol Pharmacol. 2008; 2(153 Suppl):

    251262. [PubMed: 18812643]

    Ting JP, Willingham SB, Bergstralh DT. NLRs at the intersection of cell death and immunity. Nat Rev

    Immunol. 2008; 8:372379. [PubMed: 18362948]

    Ussher JR, Koves TR, Cadete VJ, Zhang L, Jaswal JS, Swyrd SJ, Lopaschuk DG, Proctor SD, Keung

    W, Muoio DM, et al. Inhibition of de novo ceramide synthesis reverses diet-induced insulin

    resistance and enhances whole-body oxygen consumption. Diabetes. 2010; 59:24532464.

    [PubMed: 20522596]

    van Diepen JA, Wong MC, Guigas B, Bos J, Stienstra R, Hodson L, Shoelson SE, Berbee JF, Rensen

    PC, Romijn JA, et al. Hepatocyte-specific IKK-beta activation enhances VLDL-triglyceride

    production in APOE*3-Leiden mice. Journal of lipid research. 2011; 52:942950. [PubMed:

    21357939]

    Wang J, Wu X, Simonavicius N, Tian H, Ling L. Medium-chain fatty acids as ligands for orphan G

    protein-coupled receptor GPR84. The Journal of biological chemistry. 2006; 281:3445734464.

    [PubMed: 16966319]

    Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, Koeck T, Derecka M, Szczepanek K, Szelag M,

    Gornicka A, et al. Function of mitochondrial Stat3 in cellular respiration. Science. 2009;

    323:793797. [PubMed: 19131594]

    Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated

    with macrophage accumulation in adipose tissue. The Journal of clinical investigation. 2003;

    112:17961808. [PubMed: 14679176]

    Weldon SM, Mullen AC, Loscher CE, Hurley LA, Roche HM. Docosahexaenoic acid induces an anti-

    inflammatory profile in lipopolysaccharide-stimulated human THP-1 macrophages more

    effectively than eicosapentaenoic acid. J Nutr Biochem. 2007; 18:250258. [PubMed: 16781858]

    Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT, Brickey WJ, Ting JP. Fatty acid-induced NLRP3-

    ASC inflammasome activation interferes with insulin signaling. Nat Immunol. 2011; 12:408

    415. [PubMed: 21478880]

    Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, Tsui H, Wu P, Davidson MG, Alonso

    MN, et al. B cells promote insulin resistance through modulation of T cells and production of

    pathogenic IgG antibodies. Nature medicine. 2011; 17:610617.

    Wolsk E, Mygind H, Grondahl TS, Pedersen BK, van Hall G. IL-6 selectively stimulates fat

    metabolism in human skeletal muscle. American journal of physiology Endocrinology and

    metabolism. 2010; 299:E832840. [PubMed: 20823453]

    Wong SW, Kwon MJ, Choi AM, Kim HP, Nakahira K, Hwang DH. 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. The Journal of biological chemistry. 2009;

    284:2738427392. [PubMed: 19648648]

    Wu D, Molofsky AB, Liang HE, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, Chawla A, Locksley

    RM. Eosinophils sustain adipose alternatively activated macrophages associated with glucosehomeostasis. Science. 2011; 332:243247. [PubMed: 21436399]

    Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, et al.

    Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin

    resistance. The Journal of clinical investigation. 2003; 112:18211830. [PubMed: 14679177]

    Glass and Olefsky Page 19

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    20/23

    Yang X, Zhang X, Heckmann BL, Lu X, Liu J. Relative contribution of adipose triglyceride lipase and

    hormone-sensitive lipase to TNF-{alpha}-induced lipolysis in adipocytes. The Journal of

    biological chemistry. 2011

    Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, Bergeron R, Kim JK, Cushman SW, Cooney

    GJ, et al. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1

    (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. The Journal of biological

    chemistry. 2002; 277:5023050236. [PubMed: 12006582]

    Zhang HH, Halbleib M, Ahmad F, Manganiello VC, Greenberg AS. Tumor necrosis factor-alpha

    stimulates lipolysis in differentiated human adipocytes through activation of extracellular signal-

    related kinase and elevation of intracellular cAMP. Diabetes. 2002; 51:29292935. [PubMed:

    12351429]

    Zhao L, Chen Y, Tang R, Li Q, Gong J, Huang A, Varghese Z, Moorhead JF, Ruan XZ. Inflammatory

    stress exacerbates hepatic cholesterol accumulation via increasing cholesterol uptake and de novo

    synthesis. Journal of gastroenterology and hepatology. 2011; 26:875883. [PubMed: 21488946]

    Zu L, He J, Jiang H, Xu C, Pu S, Xu G. Bacterial endotoxin stimulates adipose lipolysis via toll-like

    receptor 4 and extracellular signal-regulated kinase pathway. The Journal of biological

    chemistry. 2009; 284:59155926. [PubMed: 19122198]

    Zu L, Jiang H, He J, Xu C, Pu S, Liu M, Xu G. Salicylate blocks lipolytic actions of tumor necrosis

    factor-alpha in primary rat adipocytes. Molecular pharmacology. 2008; 73:215223. [PubMed:

    17911533]

    Glass and Olefsky Page 20

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    21/23

    Figure 1. The effects of fatty acids on inflammation and insulin resistance

    Saturated fatty acids (SFAs) stimulate inflammatory pathways through Tlr4 dependent and

    independent mechanisms. SFAs are also precursors for ceramide biosynthesis, and

    inflammatory pathway stimulation by SFAs, as well as cytokines, induces the enzymes

    which produce ceramide, providing two mechanisms for increased ceramide content. Omega

    3 FAs produce anti-inflammatory effects by stimulating GPR120 and production of

    resolvins and protectins. Omega 6 FAs can be proinflammatory through their metabolism to

    leukotrienes and prostaglandins.

    Glass and Olefsky Page 21

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    22/23

    Figure 2.

    (A) Tlr4 and TNF receptor signaling stimulates the TAK1/TAB1 complex which then

    activates IKKand MKK4 to stimulate the downstream NFB and JNK1 inflammatorypathways. (B) Omega 3 FAs activate GPR120 causing it to associate with arrestin-2. The

    GPR120arrestin-2 complex then undergoes endocytosis and the internalized arrestin-2

    binds to TAB1, stealing it from the TAK1 complex, causing TAK1 inactivation with

    inhibition of subsequent downstream proinflammatory signaling. Adapted from (Oh et al.,

    2010).

    Glass and Olefsky Page 22

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 8/11/2019 Inflammation and Lipid Signal.pdf

    23/23

    Figure 3.

    Impact of inflammation on lipid metabolism. Inducers and amplifiers of inflammation

    include ligands for pattern recognition receptors (e.g. LPS), cytokines (e.g., Il1) and

    saturated fatty acids (SFAs). Each of these inflammatory stimuli influence lipid metabolism

    in liver, skeletal muscle and adipose tissue by acting on both parenchymal cells and resident

    immune cells. Although some effects are seen in all three organs (e.g., increased TG

    hydrolysis), other effects may be organ-specific (e.g., increased fatty acid biosynthesis in

    liver).

    Glass and Olefsky Page 23

    Cell Metab. Author manuscript; available in PMC 2014 September 05.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript