role of macrphage in metabolic syndrome

Upload: anand-shankar

Post on 05-Apr-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    1/10

    Biochem. J. (2012) 442, 253262 (Printed in Great Britain) doi:10.1042/BJ20111708 253

    REVIEW ARTICLE

    Role and function of macrophages in the metabolic syndrome

    Prerna BHARGAVA and Chih-Hao LEE1

    Department of Genetics and Complex Diseases, Department of Nutrition, Division of Biological Sciences, Harvard School of Public Health, 665 Huntington Ave, Boston,

    MA 02115, U.S.A.

    Macrophages are key innate immune effector cells best knownfor their role as professional phagocytes, which also includeneutrophils and dendritic cells. Recent evidence indicates thatmacrophages are also key players in metabolic homoeostasis.Macrophages can be found in many tissues, where theyrespond to metabolic cues and produce pro- and/or anti-inflammatory mediators to modulate metabolite programmes.Certain metabolites, such as fatty acids, ceramides and cholesterolcrystals, elicit inflammatory responses through pathogen-sensing signalling pathways, implicating a maladaptation ofmacrophages and the innate immune system to elevated metabolic

    stress associated with overnutrition in modern societies. Theoutcome of this maladaptation is a feedforward inflammatory

    response leading to a state of unresolved inflammation and acollection of metabolic pathologies, including insulin resistance,fatty liver, atherosclerosis and dyslipidaemia. The presentreview summarizes what is known about the contributions ofmacrophages to metabolic diseases and the signalling pathwaysthat are involved in metabolic stress-induced macrophageactivation. Understanding the role of macrophages in theseprocesses will help us to develop therapies against detrimentaleffects of the metabolic syndrome.

    Key words: inflammatory signalling, insulin resistance, macro-

    phage activation, metabolic disease, metabolic syndrome, whiteadipose tissue inflammation.

    INTRODUCTION

    The innate immune system, which consists of macrophages,dendritic cells and a variety of other effector cells, has evolvedas a formidable defence against external threats to the human

    host. When first described by Elie Metchnikoff in the late 1800s,macrophages were thought to primarily function as phagocytes.In the next several decades, intense effort went into understandingthe mechanisms that led to macrophage activation. It was not untilabout 100 years later that researchers first started to document the

    innumerable roles that macrophages play [1]. We now know thatmacrophages are found throughout the body and are involved intissue homoeostasis, wound healing, general surveillance of hostthreats and, as was first observed, killing of foreign pathogens.

    Abundant sources of food that have high energy contentand are enriched in saturated fats, coupled with a lack ofphysical activity, are responsible for the sharp increase in acollection of pathologies, such as obesity, hepatosteatosis, insulinresistance and atherosclerosis, known as the metabolic syndrome.All of these pathologies increase the chance of developingcardiovascular disease, diabetes mellitus and premature death[2]. The observation that anti-inflammatory salicylates couldimprove insulin sensitivity and glucose responsiveness gave thefirst indication that metabolic syndrome is associated with low-grade chronic inflammation [3]. This was followed by reports thatinflammatory cytokines, such as TNF (tumour necrosis factor), inhibit insulin responsiveness in adipocytes, providing a linkbetween inflammation and metabolic pathways [46]. Subsequentstudies have suggested that lipid overload triggers the immune

    system, especially macrophages, to respond in a deleteriousmanner, in which a resolution state could not be achieved [7,8].Ataround the same time as these studies were occurring, researchersobserved that insulin resistance and hyperglycaemia led to dys-regulated immune responses and increased incidences of infectionin patients with the metabolic syndrome [916]. Together, it hasbecome evident that metabolic pathways are tightly intertwinedwith inflammatory signalling and immune responses.

    As environmental sentries, it is not surprising that macrophagessense and regulate metabolism. This link between macrophages

    and metabolic regulation was strengthened further with the dis-covery of resident macrophage populationsin metabolically activetissues, such as WAT (white adipose tissue) and liver [17,18]. Theinterplay between resident macrophages and pathologies associ-ated with the metabolic syndrome has been studied extensivelysince. In the present review, we highlight major contributionsof macrophages to the metabolic milieu and describe how thefunctions of macrophages are altered during states of metabolicdysfunction. We begin with a brief overview of the recruitmentof macrophages in various metabolic tissues and the diseasesassociated with their activation. We then discuss some proposedmechanisms for the inflammatory pathways that lead to patholo-gies, with an emphasis on insulin resistance/Type 2 diabetes.

    AN ESSENTIAL ROLE FOR MACROPHAGES IN METABOLICPATHOLOGIES

    Resident macrophages in different tissues adapt to theirlocal microenvironment and exhibit diverse functional and

    Abbreviations used: ASC, apoptosis-associated speck-like protein containing a caspase-recruitment domain; ATM, adipose-tissue-resident

    macrophage; BAT, brown adipose tissue; CCR2, CC chemokine receptor 2; CHOP, C/EBP (CCAAT/enhancer-binding protein)-homologous protein;

    DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; ER, endoplasmic reticulum; GPCR, G-protein-coupled receptor; HIF, hypoxia-inducible factor;

    IFN, interferon ; IKK, inhibitor of nuclear factor B kinase; IL, interleukin; IRS-1, insulin receptor substrate-1; JNK, c-Jun N-terminal kinase; LDL,

    low-density lipoprotein; Ldlr, LDL receptor; LXR, liver X receptor; MCP-1, monocyte chemoattractant protein 1; miRNA, microRNA; mTOR, mammalian

    target of rapamycin; NAFLD, non-alcoholic fatty liver disease; NF-B, nuclear factor B; NLRP3, NLR (nucleotide-binding-domain- and leucine-rich-repeat-

    containing) family, pyrin-domain-containing 3; oxLDL, oxidized LDL; PKR, double-stranded RNA-dependent protein kinase; PPAR, peroxisome-proliferator-

    activated receptor; STAT6, signal transducer and activator of transcription 6; SVF, stromal vascular fraction; TLR, Toll-like receptor; TNF, tumour necrosis

    factor ; UPR, unfolded protein response; WAT, white adipose tissue.1

    To whom correspondence should be addressed (email [email protected]).

    c The Authors Journal compilation c 2012 Biochemical Society

    www.biochemj.org

    http://getutopia.com/documents
  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    2/10

    254 P. Bhargava and C.-H. Lee

    Figure 1 Metabolic dysregulation leads to a feedforward cycle of chronicinflammation

    Activation of inflammatory pathways by metabolic stress leads to macrophage recruitmentto sites of affected tissues, such as vasculature, WAT, muscle and liver. There, they pick upexcess lipids and cell debris in an attempt to restore homoeostasis. The presence of persistentpro-inflammatory stimulants (e.g. fatty acids and cholesterol) causes macrophage dysfunction,including defective efferocytosis and unresolved inflammation, resulting in recruitment andactivation of more macrophages. In WAT, infiltrated macrophages are concentrated around thedying fat cells, known as the crown-like structure. In the vasculature, macrophages are enrichedin areas of fatty streaks. Inflammatory cytokines produced by activated macrophages induceinsulin resistance in major metabolic tissues, promote hepatosteatosis and facilitate foam celland plaque formation in the vessel wall.

    morphological phenotypes. Resident populations, such as Kupffercells in the liver, resident macrophages in WAT and alveolarmacrophages in the lung, show expression profiles distinctfrom those of recruited macrophages responding to systemic

    cues [1]. For example, studies have demonstrated that ATMs(adipose-tissue-resident macrophages) are functionally distinctfrom infiltrating macrophages (discussed below). In the processof understanding the aetiology of the metabolic syndrome, wehave begun to understand the general mechanisms underlyingmetabolic diseases, particularly the roles of macrophages inatherosclerosis and in adipose tissue insulin resistance. Owing toa lack of specific markers for resident and recruited macrophagesand the cross-talk between tissues in the progression ofmetabolic diseases, it is often difficult to tease apart the precisecontribution of individual macrophage populations to the onsetand development of systemic metabolic dysfunction. Nonetheless,it is clear that the interaction between macrophages and metaboliccells plays a key role in disease pathogenesis. Macrophages are

    recruited by pro-inflammatory cytokines and chemokines to dealwith lipidoverload-induced tissuedysfunction and are also subjectto the detrimental effect of lipid influx. As such, metabolic stressinitiates a feedforward cycle of inflammatory responses, resultingin a state of unresolved chronic inflammation, with macrophagesas major culprits contributing to metabolic diseases (Figure 1).

    Macrophage infiltration and activation

    Recruitment

    The production of chemokines, the factors that recruit immunecells to sites of trouble, is increased in states of metabolicdysfunction. MCP-1 (monocyte chemoattractant protein 1) [alsoknown as CCL2 (CC chemokine ligand 2)] and its receptor CCR2

    (CC chemokine receptor 2) are the most studied chemokine

    signalling molecules in metabolic diseases. Earlier studieshave demonstrated that MCP-1- and CCR2-knockout mice areprotected from atherosclerosis [1921]. CCR2-knockout micealso display attenuated macrophage accumulation and chronicinflammation in adipose tissue [22]. In addition, mice lackingMCP-1 or expressing a dominant-negative form of MCP-1 showimproved insulin sensitivity [23], whereas transgenic MCP-1

    overexpression in adipose tissue causes insulin resistance andhepatic steatosis [23,24]. Other chemokines, such as osteopontin,angiopoietin-like protein 2 and CXCL14 (CXC chemokine ligand14) have also been shown to play a role in obesity-inducedmacrophage recruitment [2528]. These studies demonstrate thatmacrophage infiltration is an essential step for metabolic diseasepathogenesis.

    Activation

    Macrophages respond to many different types of foreign andhost-derived stimuli and exhibit very precise and co-ordinatedactivation states according to the signals they receive. These stateslie on a spectrum ranging from the pro-inflammatory state to theanti-inflammatory state [29] (Figure 2). Foreign pathogens or Th1cytokines [e.g. TNF, IL (interleukin)-1 and IFN (interferon)] transduce signals that activate macrophages to a pro-inflammatory phenotype, typically referred to as an M1 response,characterized by the expression of inflammatory markers, suchas TNF, IFN and iNOS (inducible nitric oxide synthase) [29].In contrast, M2 macrophages, induced by Th2 cytokines IL-4and IL-13, produce anti-inflammatory mediators, notably IL-10[29]. As discussed below, the M1/M2 paradigm also appliesto metabolic regulation, with M1 inducing and M2 preventingmetabolic diseases respectively (Figure 3). Interestingly, dietaryfatty acids are able to polarize macrophages towards M1 or M2activation states, depending on the signalling molecules withwhich they interact (Figure 2), thus providing a molecular basisfor the cross-talk between metabolic and inflammatory pathways.

    Resolution/deactivation

    The initial pro-inflammatory response is terminated by anti-inflammatory cytokines, such as IL-10, which is induced by bothM1 and M2 cytokines to deactivate macrophages and promoteresolution [3033]. Lack of this termination signal leads topersistent establishment of the inflammatory response [3436].Epidemiological studies have shown that polymorphisms in the

    IL10 gene are associated with obesity and metabolic diseases [3739]. Furthermore, in patients with Type 2 diabetes, circulatingmonocytes express decreased levels of IL-10 [40]. IL-10 andother deactivating signalling molecules are also induced duringefferocytosis, a process by which macrophages engulf and clear

    apoptotic cells in an effort to prevent necrosis and suppressinflammation [41]. This process is thought to help removeoxidized lipids and cholesterol and to activate Akt and NF-B(nuclear factor B) survival pathways [42]. Several studies haveshown that efficient efferocytosis is necessary to inhibit necroticplaqueformation anddefects in efferocytosislead to theformationof unstable lesions and an increase in systemic inflammation [4346]. Faulty efferocytosis has also been observed in diabetic mousemodels and is thought to promote inflammatory signalling [47].Oxidative stress and macrophage insulin resistance are two of thepotential mechanisms that cause defective efferocytosis [41,48].Therefore the combined effects of persistent pro-inflammatorystimulation from increased lipid influx, decreased deactivatingsignals and compromised efferocytosis lead to the unresolved

    inflammation in metabolic dysregulation.

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    3/10

    Role and function of macrophages in the metabolic syndrome 255

    Figure 2 Dietary lipids and inflammatory mediators share common signalling pathways in the control of macrophage activation

    The Th2 (M2) type response, elicited by Th2 cytokines, notably IL-4 and IL-13, and parasitic worm infection, induces an anti-inflammatory phenotype in the macrophage and promotes metabolichomoeostasis, tissue repair, wound healing and angiogenesis. STAT6 is activated by Th2 cytokines to control the expression of PPAR and PPAR. Together, they regulate mitochondrial oxidativemetabolism and macrophage alternative activation. The 3 fatty acids DHA and EPA are known to ligate the GPCR GPR120 to promote anti-inflammatory response. These fatty acids can alsomediate PPAR activation. In contrast, saturated fatty acids (FAs), ceramides (metabolites of fatty acids) and cholesterol crystal can induce pro-inflammatory activation of the macrophage throughpathogen-sensing proteins TLR2/TLR4, which activate the JNK cascade, leading to NF-B relocation to the nucleus and induction of inflammatory cytokines and glycolytic pathways. Excess lipidshave also been shown to induce ER stress and inflammasome activation. Pro-inflammatory cytokines produced by these macrophages, including TNF and IL-1 , cause tissue damage and inhibitinsulin signalling, which contribute to the pathogenesis of metabolic diseases. I B, inhibitor of NF-B; iNOS, inducible nitric oxide synthase.

    Figure 3 Cross-talk between macrophages and adipocytes plays an important role in adipose tissue homoeostasisIn the physiological state, ATMs exhibit an M2 phenotype, mediated by the Th2 cytokines IL-4 and IL-13, and downstream transcription factors STAT6 and PPAR /PPAR. Several cell types withinWAT have been reported to be the sources of Th2 cytokines, including T-lymphocytes, eosinophils and adipocytes. 3 fatty acids (FAs) are also able to induce an anti-inflammatory responsethrough GPR120 and possibly PPARs. IL-10 is one of the well-characterized anti-inflammatory cytokines produced by these macrophages. In obesity, stressed adipocytes produce inflammatorymediators (e.g. MCP-1, TNF and saturated fatty acids) to induce M1 activation through inflammatory transcription factors, such as NF-B. Macrophages respond by up-regulating inflammatorycytokines, which activate JNK and inhibit insulin signalling pathways in the adipocyte.

    Macrophages and metabolic pathologies

    Macrophage infiltration and adipose tissue insulin resistance

    The SVF (stromal vascular fraction) of WAT has beenstudied extensively in the progression of obesity-induced insulinresistance. Adipose tissues from lean animals and humans contain

    a resident macrophage population exhibiting the M2 phenotype

    [49] (Figure 3). In contrast, the obese state is characterized byinfiltration of M1 macrophages that accumulate around apoptoticfat cells (referred to as crown-like structures) and express theCD11c marker [49]. It has been suggested that macrophagesare recruited in response to adipocyte hypertrophy or to signalsfrom dying adipocytes in an attempt to restore homoeostasis

    [5052]. There they can be activated further by non-esterified

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    4/10

    256 P. Bhargava and C.-H. Lee

    fatty acids released by dysfunctional fat cells (discussed below),thus amplifying the inflammatory response. These macrophagescontain lipid droplets derived from direct lipid intake or ingestionof dead fat cells and account for up to 40% of the SVF insevere cases of obesity [22]. They secrete pro-inflammatoryfactors, such as MCP-1, TNF and IL-6, which both amplifyinflammatory responses and inhibit adipocyte insulin signalling

    [17,18]. Similarly, monocytes from Type 2 diabetic patients orobese individuals express more M1 markers [40]. Conditionaldepletion of CD11c-expressing macrophages or inhibition ofmacrophage recruitment (e.g. MCP-1 knockout) in obese miceresulted in a significant reduction in systemic inflammation andan improvement in insulin sensitivity [23,24,53]. It should alsobe noted that adipocytes also produce inflammatory mediators.Because of similarities between adipocytes and macrophages,the source of the pro-inflammatory cytokines/chemokines withinWAT is obscured. In contrast with WAT, macrophage infiltrationinto BAT (brown adipose tissue) seems to be limited and, as such,the role of macrophages in BAT is unclear [54,55].

    Vascular inflammation and atherosclerosis

    The most well-characterized macrophage population in themetabolic syndrome is that of macrophages recruited to sites ofendothelial dysfunction in atherosclerosis [56,57]. Elevated LDL(low-density lipoprotein)/cholesterol levels result in accumulationof LDL particles in the subendothelial matrix. Careful analysesof temporal changes leading to lesion formation revealed thatthis lipid deposition is the crucial initiating event in macrophagerecruitment [57]; the accumulated LDL becomes oxidized(oxLDL) and locally produced cytokines and chemokines, inresponse to oxLDL, recruit monocytes/macrophages into thesubendothelial space.Lipid particles are taken up by macrophagesthrough specialized receptors SR-A (scavenger receptor A) andCD36, among other mechanisms [58]. These processes evoke acharacteristic inflammatory response by releasing inflammatory

    molecules, such as MCP-1, to the ECM (extracellular matrix),which recruit more macrophages. The lipid-laden macrophagesbecome foam cells and undergo necrotic cell death releasingintracellular components leading to a vicious cycle of chronicinflammation [36]. Subsequently, smooth muscle cells migrate tothe lesion in response to inflammatory mediators and contributeto fibrotic plaque formation and rupture [57].

    Hepatosteatosis

    Liver insulin resistance and hepatic steatosis, referred toas NAFLDs (non-alcoholic fatty liver diseases), are majorcomponents of the metabolic syndrome. Lipidomic analyses ofliver tissue during various stages of NAFLD show a strongcorrelation between abnormal fat composition and disease

    pathogenesis [59], with excess fat accumulation leading to therecruitment of inflammatory cells [60]. Together with Kupffercells, these immune cells are responsible for the developmentof hepatic inflammation in both humans and various animalmodels. Studies have shown that depletion of Kupffer cellsor inhibition of pro-inflammatory mediators protect against thedevelopment of NAFLD [6163]. During high-fat-diet treatment,depletion of Kupffer cells by GdCl3 or clodronate-encapsulatedliposomes has been shown to improve insulin sensitivity andglucose tolerance [6466]. A single dose of GdCl3 in mice onnormal chow enhances insulin signalling and reduces glucoseproduction in the liver [65]. In contrast, macrophage-specificPpard/ (peroxisome-proliferator-activated receptor ) mice,whose macrophages/Kupffer cells exhibit a predominantly M1

    phenotype, develop insulin resistance and hepatic steatosis [67].

    Muscle dysfunction

    The resident macrophage population in muscle has not beencharacterized. Macrophages do, however, play a significant rolein muscle repair during exercise and tissue damage [ 68]. Studieshave shown that macrophages infiltrate fat depots formed aroundthe muscle in obesity [17]. Additionally, muscle can be affectedby inflammatory cytokines from other tissues, such as liver and

    adipose tissue, even though it may not be a site of significantproduction of inflammatory mediators [69].

    INFLAMMATORY SIGNALLING AND METABOLIC DISEASES

    Although the role of inflammation in atherogenesis is welldocumented [36,56,57], the underlying mechanisms throughwhich pro-inflammatory signalling pathways cause insulinresistance are still under investigation. One of the better-definedmechanisms is through JNK (c-Jun N-terminal kinase) activation,which phosphorylates IRS-1 (insulin receptor substrate-1) toblock insulin signal transduction [7,70]. Similarly, our knowledgeregarding how anti-inflammatory pathways improve metabolic

    homoeostasis is limited, apart from their ability to antagonizeM1 activation [71]. In this section, we summarize varioussignalling pathways mediating macrophage activation during thepathogenesis of the metabolic syndrome.

    Pro-inflammatory signalling

    JNK and pro-inflammatory cytokines

    JNK is an important regulatory node for inflammatory mediators,including pro-inflammatory cytokines, TLRs (Toll-like receptors)and ER (endoplasmic reticulum) stress (Figure 2). In obesity-induced insulin resistance and hepatic steatosis, deletion ofJNK1 in the haemopoietic compartment by bone marrowtransplantation is beneficial [72]. TNF was one of the firstpro-inflammatory cytokines to be linked to JNK activation[4,70]. TNF-receptor-knockout mice show improved metabolichomoeostasis and insulin sensitivity fed on both normal chowand a high-fat diet [73]. Additionally, Tnf+/+ bone marrowdonated to a Tnf/ mouse induces insulin resistance in anotherwise insulin-sensitive mouse [74]. Recently, PKR (double-stranded RNA-dependent protein kinase), which senses viralinfection, was shown to interact with JNK and IRS-1 [75].Interestingly, PKR is able to phosphorylate IRS-1 directly,suggesting that JNK integrates different inflammatory signalsthrough a multicomponent inflammatory complex.

    TLRs and NF-B

    TLRs are pattern-recognition receptors that respond to pathogenicantigens and propagate inflammatory signalling [76]. In vitro,non-esterified fatty acids (saturated fatty acids, e.g. palmitic acid)or ceramides can signal through TLR2 and TLR4 on macrophagesand induce pro-inflammatory gene expression [7779]. In vivo,whole-body TLR4 deletion improves insulin sensitivity in a lipid-infusion model of transient insulin resistance. Similarly, TLR4loss-of-function mutation or TLR2 deficiency protects againstdiet-induced obesity and insulin resistance [8082]. However,TLR2 and TLR4 are expressed on most tissue types includingadipocytes and hepatocytes, in addition to immune cells. Thecontribution of macrophage TLR4 was examined through haemo-poietic cell-specific TLR4 deletion, which ameliorates high-fat-

    diet-induced hepatic and adipose tissue insulin resistance [83].

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    5/10

    Role and function of macrophages in the metabolic syndrome 257

    TLR4 ligation promotes IKK (inhibitor of NF-B kinase)activation, followed by phosphorylation and nuclear translocationof NF-B to activate inflammatory gene transcription. Systemicdeletions of NF-B or various IKK isoforms (e.g. IKK andIKK) prevent obesity-induced insulin resistance [36,84,85].Myeloid-specific IKK deletion also leads to improved systemicinsulin sensitivity, increased glucose disposal and suppressed

    hepatic glucose production [84]. In line with these observations,salicylate treatment, known to attenuate NF-B activation,increases insulin sensitivity in humans [8688].

    In the setting of atherosclerosis, many studies havedemonstrated that a complete deletion of various TLR familymembers reduces the lesion size in Ldlr/ (low-density-lipoprotein receptor) orApoe/ (apolipoprotein E) mice [89,90].

    Ldlr/ mice transplanted with Tlr2/ bone marrow areprotected from atherosclerosis [89]. Similarly, Ldlr/ micedeficient in myeloid-specific NF-B subunit p50 have decreasedatherosclerotic lesions [91]. Lesions in these animals exhibit nearcomplete loss of foam cells, supporting previous findings thatNF-B regulates genes involved in lipid uptake and foam cellformation [92].

    Inflammasome

    The NLRP3 [NLR (nucleotide-binding-domain- and leucine-rich-repeat-containing) family, pyrin-domain-containing 3]inflammasome senses endogenous danger signals and generatesthe mature secreted forms of IL-1 and IL-18 through caspase 1activation [9396].In vitro, palmitic acid and ceramide are able toinduce IL-1 and caspase 1 processing in the macrophage [97].Mice deficient in NLRP3 or ASC (apoptosis-associated speck-like protein containing a caspase-recruitment domain, an adaptorprotein of the NLRP3 inflammasome) exhibit increased insulinsensitivity in liver, WAT and muscle [9799]. In addition, caspase1 and IL-1 have been shown to mediate inflammasome-inducedinsulin resistance [98]. Along these lines, lack of IL-1R, the

    receptor for IL-1 , confers protection against insulin resistanceinduced by a high-fat diet [100]. Inflammasome activation canalso be detected in atherosclerotic lesions [101,102]. Culturedmacrophages exposed to crystalline cholesterol secrete IL-1 andIL-18 [101,102]. In concert, Ldlr/ mice transplanted with bonemarrow from mice lacking NLRP3 or ASC show a reduction inthe lesion area and IL-18 levels [101].

    ER stress

    The ER is the site of protein folding, vesicle transport andlipid synthesis. When the capacity of ER is overburdened,the UPR (unfolded protein response) is activated characterizedby increased activities of PERK (PKR-like ER kinase),

    IRE1 (inositol-requiring enzyme 1) and ATF6 (activatingtranscription factor 6) [103]. Activation of the UPRthrough these signalling molecules leads to downstreamCHOP [C/EBP (CCAAT/enhancer-binding protein)-homologousprotein] activation and subsequent induction of apoptosis. Theinitiation and propagation of the UPR and ER stress by obesityand metabolic stress have been reviewed in detail [103]. ERstress in adipose tissue or liver leads to insulin resistance, whichis associated with activation of JNK and IKK, suggesting thatER stress is linked to inflammatory pathways [103105]. Inatherosclerosis, chronic activation of the ER stress pathwaycontributes to macrophage death and subsequent plaque necrosis[42]. CHOP expression and macrophage apoptosis have beencorrelated with advanced lesions [106,107], anddeletion of CHOP

    suppresses lesional macrophage death/necrosis [108]. Lastly, it

    has been shown that the ER stress in the macrophage is causedby increased aP2 (adipocyte protein 2) lipid chaperone activity,which promotes atherogenesis [109].

    Hypoxia

    Activation of macrophages to a pro-inflammatory state increases

    glucose influx for glycolysis, which is controlled by HIF(hypoxia-inducible factor)-1 [110113]. Loss of HIF-1 inthe macrophage leads to decreased pro-inflammatory cytokineproduction, whereas deletion of VHL (von HippelLindautumour-suppressor protein), a repressor of HIF-1, results inchronic activation of HIF-1 and uncontrolled inflammation[112,114]. In the context of metabolic dysregulation, adiposetissues in obese mice are hypoxic, which may cause macrophageHIF-1 activation [51,52,115,116]. Systemic hypoxia has beenshown to induce insulin resistance and NAFLD [51,117,118].Furthermore, high levels of hypoxia during adipose expansionmay lead to necrotic death of both adipocytes and macrophages,since HIF-1 activation blocks physiological apoptotic cell death[52,116,119]. Similarly, hypoxia promotes macrophage necrosisin mouse models of atherosclerosis [119121].

    Anti-inflammatory signalling

    Th2 cytokines

    The other end of the macrophage activity spectrum is Th2cytokine-induced alternative activation, characterized by theexpression of M2 markers, such as Arg1, Mgl1 and YM1 [29,71].These macrophages function to repair damage elicited bypro-inflammatory M1 macrophages. As discussed above, in thelean state, ATMs display an M2 phenotype, suggesting that, atthe physiological level, M2 macrophages may play an importantrole in metabolic homoeostasis [49]. In line with this notion, Th2cytokines, notably IL-13, have been detected in WAT and liver

    [122]. Treatment of diet-induced obese mice with IL-4 signif-icantly improves glucose homoeostasis and insulin sensitivity,whereas deletion of STAT6 (signal transducer and activator oftranscription 6), a Th2 effector, worsens insulin resistance [123].In addition, mice infected with helminth worms, which inducean M2 response, exhibit improved insulin sensitivity [124].

    Nuclear receptors

    Nuclear receptors are ligand-activated transcription factors thatcontrol important biological processes [125]. Several lipid-sensing nuclear receptors, such as the PPARs(PPAR, PPAR andPPAR, activated by dietary fatty acids) [126,127] and the LXRs(liver X receptors) (LXR and LXR , activated by cholesterol

    metabolites) aredrug targets to treat metabolic diseases [128,129].In the macrophage, IL-4/IL-13-induced alternative macrophageactivation is associated with increased fatty acid -oxidation andoxidative metabolism [122,130], pathways regulated by PPARs[131]. Accordingly, PPAR and PPAR have been shown to beinduced by Th2 cytokines and control M2 activation [122,132].Mice with myeloid-specific deletion of PPAR or PPAR showincreased M1 and decreased M2 markers in WAT and liver anddevelop systemic insulin resistance [122,132135]. The M1/M2paradigm is also relevant in Kupffer cells in the liver. MyeloidPPAR deletion worsens hepatic steatosis in mice fed on a high-fat diet [122,135]. It has been shown that macrophages fromthese mice produce factors that promote adipocyte and hepatocytedysfunction in vitro [122]. Earlier studies also demonstrated an

    anti-inflammatory role for PPARs and LXRs, which correlates

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    6/10

    258 P. Bhargava and C.-H. Lee

    well with the atheroprotective roles for these nuclear receptors[136138].

    GPCRs (G-protein-coupled receptors)

    Although saturated fatty acids are known to induce inflammatorysignalling pathways, epidemiological studies show that

    Mediterranean diets high in polyunsaturated 3 (n 3) fattyacids actually reduce the incidence of metabolic diseases [139142]. 3 fatty acid derivatives, such as DHA (docosahexaenoicacid), EPA (eicosapentaenoic acid) and resolvins, exhibit anti-inflammatory activities [143]. DHA and EPA have been shown tobind to the GPCR GPR120 on macrophage surfaces. GPR120ligation inhibits the actions of TNF and reverses insulinresistance brought on by a high-fat diet [144]. GPR120 and PPARsshare several fatty acid ligands, suggesting potential cross-talkbetween these two signalling pathways [145].

    CONCLUDING REMARKS

    The host immune systemis essential for defending against foreignpathogens. Macrophages rely on the rapid influx of extracellularglucose for ATP production via anaerobic respiration [146148].As such, the ability of pathogen-stimulated macrophages to inhibitglucose uptake and insulin sensitivity in other metabolicallydemanding tissues is thought to be a mechanism by which immunecells sequester energy stores for defence [149151]. However, instates of gluttony and energy storage, these evolutionary pathwayshave become detrimental. As the metabolic syndrome is a rapidlyrising global epidemic affecting 8% of the U.S. populationalone and over 170 million people worldwide, efforts toidentify effective therapies for treatment of associated pathologiesare paramount [ADA (American Diabetes Association), WHO(World Health Organization)]. Exercise and healthy diets are mosteffective in improving the outcome of the metabolic syndrome

    [152,153]. However, lifestyle changes are often an unachievableideal for many patients, and therefore development of medicationsis crucial. Growing evidence indicates that macrophage activationplays a key role in metabolic dysregulation. Understanding thecontribution of macrophages to disease pathogenesis can helpin the development of immunometabolic therapeutics to treatmetabolic diseases.

    As described above, macrophages have evolved to sensedietary fats. The role of cholesterol in macrophage pro-inflammatory responses and atherosclerosis is well established.Studies have demonstrated further that fatty acids have a broadspectrum of activity on macrophage activation [7779,144].This is particularly relevant in obesity, in which the interactionof adipocyte and macrophage through fatty-acid-dependent

    mechanisms is critical to metabolic homoeostasis (Figure 3).Dietary 3 fatty acids and their derivatives, including EPA,DHA and resolvins, can inhibit inflammatory responses throughGPCRs or PPARs [143,144] and, as such, represent an attractivemeans for dietary intervention to reduce metabolic inflammation.In addition to dietary fats, studies have suggested that some aminoacids, such as arginine and glutamine, promote insulin sensitivityand insulin secretion. However, epidemiological data also suggestthat levels of branched-chain amino acids are associated withdiabetes as early as 12 years before the onset of frank insulinresistance [154,155]. The role of amino-acid-sensing pathways inmacrophage function and metabolic inflammation is currentlyunclear. In cultured macrophages, glutamine and arginineare critical for cytokine secretion, eicosanoid production and

    phagocytic uptake during macrophage activation [156]. mTOR

    (mammalian target of rapamycin), a major amino-acid-sensingmolecule, is anti-inflammatory in the macrophage. However,mTOR inhibitionby rapamycin blocksMCP-1 production, despiteits pro-inflammatory phenotype [157]. Additional work is neededto clarify the roles of amino acids in metabolic syndrome andwhether these effects are mediated, in part, through macrophageactivation.

    Although it is evident that metabolic stress (e.g. fattyacids, ceramides and oxLDL/cholesterol) induces macrophageinflammation, the detailed mechanisms through which pro-inflammatory signalling pathways cause metabolic diseases,notably in the setting of insulin resistance, remain unclear.For example, contradictory results have challenged the role ofmyeloid JNK1 in insulin resistance [158,159]. It appears thatthe initiation of a chronic inflammatory cycle may be due toloss of anti-inflammatory and/or regulatory signalling. However,the beneficial effects associated with alternative activation alsorequire further investigation, as Th2 cytokines are known tomediate allergic responses. Recent studies have implicated severalother immune cell types, such as T-lymphocytes, mast cells,natural killer cells and eosinophils, in the modulation of insulinsensitivity [124,160165]. For example, T-cell-deficient Rag/

    (recombination-activating gene)mice are moresusceptible to diet-induced obesity and adoptive transfer of CD4+Foxp3+ (forkheadbox P3) regulatory T-cells restores energy balance in thesemice [160,165,166]. These studies indicate that the interactionsbetween innate and adaptive immune systems also contribute tothe onset of themetabolic syndrome and implicate an autoimmuneresponse in the associated pathologies. What have not beendescribed are the temporal recruitment and activation of variousimmune cells in metabolic tissues and the role of their interactionin metabolic diseases.

    Other anti-inflammatory or immunomodulatory mechanismsmay be relevant in metabolic homoeostasis. Certain miRNAs(microRNAs) are up-regulated by TLR signalling and act throughNF-B pathwaysto dampenthe inflammatoryresponse[167,168].

    miRNAs have also been implicated in atherosclerotic plaqueinitiation and expansion [169]. In addition, gut flora has beenshown to modulate energy-utilization efficiency and adiposityand contribute to the onset of metabolic diseases [170,171].Helminth infection is known to deactivate the immune response,and mice infected with parasitic worms exhibit improved insulinsensitivity, suggesting the potential use of helminthic antigens fortherapies. Lastly, exercise reduces blood inflammatory markersand improves metabolic parameters. The so-called myokinesproduced by muscle after physical activity may modulatemacrophage activation [172175]. In fact, exercise-inducedimprovements in the inflammatory profile were found to beindependent of weight loss [176,177]. Through understandingthese different aspects of metabolic-related immune responses,

    it may be possible to design drugs to specifically targetthe feedforward loop of metabolic-stress-induced inflammationwithout unwanted side effects, such as immunosuppression orallergic responses.

    ACKNOWLEDGEMENT

    We thank K. Stanya for valuable comments.

    FUNDING

    P.B. is supported by the National Institute of Diabetes and Digestive and Kidney Disease(NIDDK) [training grant number T90DK070078]. C.H.L. is supported by the AmericanDiabetes Association, the American Heart Association and the National Institutes of Health

    [grant number R01DK075046].

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    7/10

    Role and function of macrophages in the metabolic syndrome 259

    REFERENCES

    1 Gordon, S. (2007) The macrophage: past, present and future. Eur. J. Immunol. 37

    (Suppl. 1), S9S17

    2 Grundy, S. M., Brewer, Jr, H. B., Cleeman, J. I., Smith, Jr, S. C. and Lenfant, C. (2004)

    Definition of metabolic syndrome: report of the National Heart, Lung, and Blood

    Institute/American Heart Association conference on scientific issues related to definition.

    Arterioscler. Thromb. Vasc. Biol. 24, e13e18

    3 Williamson, R. T. (1901) On the treatment of glycosuria and diabetes mellitus withsodium salicylate. Br. Med. J. 1, 760762

    4 Hotamisligil, G. S., Murray, D. L., Choy, L. N. and Spiegelman, B. M. (1994) Tumor

    necrosis factor inhibits signaling from the insulin receptor. Proc. Natl. Acad. Sci.

    U.S.A. 91, 48544858

    5 Hotamisligil, G. S., Shargill, N. S. and Spiegelman, B. M. (1993) Adipose expression of

    tumor necrosis factor-: direct role in obesity-linked insulin resistance. Science 259,

    8791

    6 Wellen, K. E. and Hotamisligil, G. S. (2005) Inflammation, stress, and diabetes. J. Clin.

    Invest. 115, 11111119

    7 Hotamisligil, G. S. and Erbay, E. (2008) Nutrient sensing and inflammation in metabolic

    diseases. Nat. Rev. Immunol. 8, 923934

    8 Karin, M., Lawrence, T. and Nizet, V. (2006) Innate immunity gone awry: linking

    microbial infections to chronic inflammation and cancer. Cell 124, 823835

    9 Hartman, M. E., OConnor, J. C., Godbout, J. P., Minor, K. D., Mazzocco, V. R. and

    Freund, G. G. (2004) Insulin receptor substrate-2-dependent interleukin-4 signaling in

    macrophages is impaired in two models of type 2 diabetes mellitus. J. Biol. Chem. 279,

    2804528050

    10 OConnor, J. C., Sherry, C. L., Guest, C. B. and Freund, G. G. (2007) Type 2 diabetes

    impairs insulin receptor substrate-2-mediated phosphatidylinositol 3-kinase activity in

    primary macrophages to induce a state of cytokine resistance to IL-4 in association with

    overexpression of suppressor of cytokine signaling-3. J. Immunol. 178, 68866893

    11 Rosa, L. F., Cury, Y. and Curi, R. (1992) Effects of insulin, glucocorticoids and thyroid

    hormones on the activities of key enzymes of glycolysis, glutaminolysis, the

    pentose-phosphate pathway and the Krebs cycle in rat macrophages. J. Endocrinol.

    135, 213219

    12 Bala, M., Kopp, A., Wurm, S., Buchler, C., Scholmerich, J. and Schaffler, A. (2011) Type

    2 diabetes and lipoprotein metabolism affect LPS-induced cytokine and chemokine

    release in primary human monocytes. Exp. Clin. Endocrinol. Diabetes 119, 370376

    13 Geerlings, S. E., Brouwer, E. C., van Kessel, K. P., Gaastra, W. and Hoepelman, A. M.

    (2000) Cytokine secretion is impaired in women with diabetes mellitus. Adv. Exp. Med.

    Biol. 485, 25526214 Vejlsgaard, R. (1966) Studies on urinary infection in diabetics. I. Bacteriuria in patients

    with diabetes mellitus and in control subjects. Acta Med. Scand. 179, 173182

    15 Geerlings, S. E. and Hoepelman, A. I. (1999) Immune dysfunction in patients with

    diabetes mellitus (DM). FEMS Immunol. Med. Microbiol. 26, 259265

    16 Chelvarajan, R. L., Liu, Y., Popa, D., Getchell, M. L., Getchell, T. V., Stromberg, A. J. and

    Bondada, S. (2006) Molecular basis of age-associated cytokine dysregulation in

    LPS-stimulated macrophages. J. Leukocyte Biol. 79, 13141327

    17 Weisberg, S. P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R. L. and Ferrante, Jr,

    A. W. (2003) Obesity is associated with macrophage accumulation in adipose tissue.

    J. Clin. Invest. 112, 17961808

    18 Xu, H., Barnes, G. T., Yang, Q., Tan, G., Yang, D., Chou, C. J., Sole, J., Nichols, A., Ross,

    J. S., Tartaglia, L. A. and Chen, H. (2003) Chronic inflammation in fat plays a crucial role

    in the development of obesity-related insulin resistance. J. Clin. Invest. 112, 18211830

    19 Dawson, T. C., Kuziel, W. A., Osahar, T. A. and Maeda, N. (1999) Absence of CC

    chemokine receptor-2 reduces atherosclerosis in apolipoprotein E-deficient mice.Atherosclerosis 143, 205211

    20 Gosling, J., Slaymaker, S., Gu, L., Tseng, S., Zlot, C. H., Young, S. G., Rollins, B. J. and

    Charo, I. F. (1999) MCP-1 deficiency reduces susceptibility to atherosclerosis in mice

    that overexpress human apolipoprotein B. J. Clin. Invest. 103, 773778

    21 Boring, L., Gosling, J., Cleary, M. and Charo, I. F. (1998) Decreased lesion formation in

    CCR2 / mice reveals a role for chemokines in the initiation of atherosclerosis. Nature

    394, 894897

    22 Weisberg, S. P., Hunter, D., Huber, R., Lemieux, J., Slaymaker, S., Vaddi, K., Charo, I.,

    Leibel, R. L. and Ferrante, Jr, A. W. (2006) CCR2 modulates inflammatory and metabolic

    effects of high-fat feeding. J. Clin. Invest. 116, 115124

    23 Kanda, H. (2006) MCP-1 contributes to macrophage infiltration into adipose tissue,

    insulin resistance, and hepatic steatosis in obesity. J. Clin. Invest. 116, 14941505

    24 Kamei, N. (2006) Overexpression of monocyte chemoattractant protein-1 in adipose

    tissues causes macrophage recruitment and insulin resistance. J. Biol. Chem. 281,

    2660226614

    25 Kiefer, F. W., Zeyda, M., Gollinger, K., Pfau, B., Neuhofer, A., Weichhart, T., Saemann,

    M. D., Geyeregger, R., Schlederer, M., Kenner, L. and Stulnig, T. M. (2010) Neutralization

    of osteopontin inhibits obesity-induced inflammation and insulin resistance. Diabetes

    59, 935946

    26 Nomiyama, T., Perez-Tilve, D., Ogawa, D., Gizard, F., Zhao, Y., Heywood, E. B., Jones,

    K. L., Kawamori, R., Cassis, L. A., Tschop, M. H. and Bruemmer, D. (2007) Osteopontin

    mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance

    in mice. J. Clin. Invest. 117, 28772888

    27 Nara, N., Nakayama, Y., Okamoto, S., Tamura, H., Kiyono, M., Muraoka, M., Tanaka, K.,

    Taya, C., Shitara, H., Ishii, R. et al. (2007) Disruption of CXC motif chemokine ligand-14

    in mice ameliorates obesity-induced insulin resistance. J. Biol. Chem. 282,

    3079430803

    28 Tabata, M., Kadomatsu, T., Fukuhara, S., Miyata, K., Ito, Y., Endo, M., Urano, T., Zhu,

    H. J., Tsukano, H., Tazume, H. et al. (2009) Angiopoietin-like protein 2 promotes chronic

    adipose tissue inflammation and obesity-related systemic insulin resistance. Cell Metab.

    10, 178188

    29 Mosser, D. M. and Edwards, J. P. (2008) Exploring the full spectrum of macrophage

    activation. Nat. Rev. Immunol. 8, 958969

    30 Del Prete, G., De Carli, M., Almerigogna, F., Giudizi, M. G., Biagiotti, R. and Romagnani,

    S. (1993) Human IL-10 is produced by both type 1 helper (Th1) and type 2 helper (Th2)

    T cell clones and inhibits their antigen-specific proliferation and cytokine production.

    J. Immunol. 150, 353360

    31 Fiorentino, D. F., Zlotnik, A., Vieira, P., Mosmann, T. R., Howard, M., Moore, K. W. and

    OGarra, A. (1991) IL-10 acts on the antigen-presenting cell to inhibit cytokine

    production by Th1 cells. J. Immunol. 146, 3444345132 Fiorentino, D. F., Zlotnik, A., Mosmann, T. R., Howard, M. and OGarra, A. (1991) IL-10

    inhibits cytokine production by activated macrophages. J. Immunol. 147, 38153822

    33 de Waal Malefyt, R., Abrams, J., Bennett, B., Figdor, C. G. and de Vries, J. E. (1991)

    Interleukin 10 (IL-10) inhibits cytokine synthesis by human monocytes: an

    autoregulatory role of IL-10 produced by monocytes. J. Exp. Med. 174, 12091220

    34 Baumgartl, J., Baudler, S., Scherner, M., Babaev, V., Makowski, L., Suttles, J., McDuffie,

    M., Tobe, K., Kadowaki, T., Fazio, S. et al. (2006) Myeloid lineage cell-restricted insulin

    resistance protects apolipoprotein E-deficient mice against atherosclerosis. Cell Metab.

    3, 247256

    35 Kanters, E., Pasparakis, M., Gijbels, M. J., Vergouwe, M. N., Partouns-Hendriks, I.,

    Fijneman, R. J., Clausen, B. E., Forster, I., Kockx, M. M., Rajewsky, K. et al. (2003)

    Inhibition of NF-B activation in macrophages increases atherosclerosis in LDL

    receptor-deficient mice. J. Clin. Invest. 112, 11761185

    36 Baker, R. G., Hayden, M. S. and Ghosh, S. (2011) NF-B, inflammation, and metabolic

    disease. Cell Metab. 13, 1122

    37 Chang, Y. H., Huang, C. N., Wu, C. Y. and Shiau, M. Y. (2005) Association ofinterleukin-10 A-592C and T-819C polymorphisms with type 2 diabetes mellitus. Hum.

    Immunol. 66, 12581263

    38 Erdogan, M., Cetinkalp, S., Ozgen, A. G., Saygili, F., Berdeli, A. and Yilmaz, C. (2011)

    Interleukin-10 ( 1082G/A) gene polymorphism in patients with type 2 diabetes with

    and without nephropathy. Genet. Test. Mol. Biomarkers, doi:10.1089/gtmb.2011.0075

    39 Scarpelli, D., Cardellini, M., Andreozzi, F., Laratta, E., Hribal, M. L., Marini, M. A.,

    Tassi, V., Lauro, R., Perticone, F. and Sesti, G. (2006) Variants of the interleukin-10

    promoter gene are associated with obesity and insulin resistance but not type 2 diabetes

    in Caucasian Italian subjects. Diabetes 55, 15291533

    40 Satoh, N., Shimatsu, A., Himeno, A., Sasaki, Y., Yamakage, H., Yamada, K., Suganami, T.

    and Ogawa, Y. (2010) Unbalanced M1/M2 phenotype of peripheral blood monocytes in

    obese diabetic patients: effect of pioglitazone. Diabetes Care 33, e7

    41 Tabas, I., Tall, A. and Accili, D. (2010) The impact of macrophage insulin resistance on

    advanced atherosclerotic plaque progression. Circ. Res. 106, 5867

    42 Tabas, I. (2010) The role of endoplasmic reticulum stress in the progression of

    atherosclerosis. Circ. Res. 107, 83985043 Ball, R. Y., Stowers, E. C., Burton, J. H., Cary, N. R., Skepper, J. N. and Mitchinson, M. J.

    (1995) Evidence that the death of macrophage foam cells contributes to the lipid core of

    atheroma. Atherosclerosis 114, 4554

    44 Tabas, I. (2005) Consequences and therapeutic implications of macrophage apoptosis in

    atherosclerosis: the importance of lesion stage and phagocytic efficiency. Arterioscler.

    Thromb. Vasc. Biol. 25, 22552264

    45 Hegyi, L., Skepper, J. N., Cary, N. R. and Mitchinson, M. J. (1996) Foam cell apoptosis

    and the development of the lipid core of human atherosclerosis. J. Pathol. 180, 423429

    46 Fadok, V. A., Bratton, D. L. and Henson, P. M. (2001) Phagocyte receptors for apoptotic

    cells: recognition, uptake, and consequences. J. Clin. Invest. 108, 957962

    47 Maree, A. F. M., Komba, M., Dyck, C., abecki, M., Finegood, D. T. and

    Edelstein-Keshet, L. (2005) Quantifying macrophage defects in type 1 diabetes. J. Theor.

    Biol. 233, 533551

    48 Yvan-Charvet, L., Pagler, T. A., Seimon, T. A., Thorp, E., Welch, C. L., Witztum, J. L.,

    Tabas, I. and Tall, A. R. (2010) ABCA1 and ABCG1 protect against oxidative

    stress-induced macrophage apoptosis during efferocytosis. Circ Res. 106, 18611869

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    8/10

    260 P. Bhargava and C.-H. Lee

    49 Lumeng, C. N., Bodzin, J. L. and Saltiel, A. R. (2007) Obesity induces a phenotypic

    switch in adipose tissue macrophage polarization. J. Clin. Invest. 117, 175184

    50 Guerre-Millo, M. (2004) Adipose tissue and adipokines: for better or worse. Diabetes

    Metab. 30, 1319

    51 Ye, J. (2009) Emerging role of adipose tissue hypoxia in obesity and insulin resistance.

    Int. J. Obes. Relat. Metab. Disord. 33, 5466

    52 Yin, J., Gao, Z., He, Q., Zhou, D., Guo, Z. and Ye, J. (2009) Role of hypoxia in

    obesity-induced disorders of glucose and lipid metabolism in adipose tissue. Am. J.

    Physiol. Endocrinol. Metab. 296, E333E34253 Patsouris, D., Li, P., Thapar, D., Chapman, J., Olefsky, J. and Neels, J. (2008) Ablation of

    CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals.

    Cell Metab. 8, 301309

    54 Fitzgibbons, T. P., Kogan, S., Aouadi, M., Hendricks, G. M., Straubhaar, J. and Czech, M.

    P. (2011) Similarity of mouse perivascular and brown adipose tissues and their

    resistance to diet-induced inflammation. Am. J. Physiol. Heart Circ. Physiol. 301,

    H1425H1437

    55 Ortega, M. T., Xie, L., Mora, S. and Chapes, S. K. (2011) Evaluation of macrophage

    plasticity in brown and white adipose tissue. Cell. Immunol. 271, 124133

    56 Glass, C. K. and Witztum, J. L. (2001) Atherosclerosis: the road ahead. Cell 104,

    503516

    57 Moore, K. J. and Tabas, I. (2011) Macrophages in the pathogenesis of atherosclerosis.

    Cell 145, 341355

    58 Kunjathoor, V. V., Febbraio, M., Podrez, E. A., Moore, K. J., Andersson, L., Koehn, S.,

    Rhee, J. S., Silverstein, R., Hoff, H. F. and Freeman, M. W. (2002) Scavenger receptors

    class A-I/II and CD36 are the principal receptors responsible for the uptake of modifiedlow density lipoprotein leading to lipid loading in macrophages. J. Biol. Chem. 277,

    4998249988

    59 Elizondo, A., Araya, J., Rodrigo, R., Poniachik, J., Csendes, A., Maluenda, F., Diaz, J. C.,

    Signorini, C., Sgherri, C., Comporti, M. and Videla, L. A. (2007) Polyunsaturated fatty

    acid pattern in liver and erythrocyte phospholipids from obese patients. Obesity 15,

    2431

    60 Wouters, K., van Gorp, P. J., Bieghs, V., Gijbels, M. J., Duimel, H., Lutjohann, D.,

    Kerksiek, A., van Kruchten, R., Maeda, N., Staels, B. et al. (2008) Dietary cholesterol,

    rather than liver steatosis, leads to hepatic inflammation in hyperlipidemic mouse

    models of nonalcoholic steatohepatitis. Hepatology 48, 474486

    61 Shoelson, S. E., Herrero, L. and Naaz, A. (2007) Obesity, inflammation, and insulin

    resistance. Gastroenterology 132, 21692180

    62 Maher, J. J., Leon, P. and Ryan, J. C. (2008) Beyond insulin resistance: innate immunity

    in nonalcoholic steatohepatitis. Hepatology 48, 670678

    63 Rivera, C. A., Adegboyega, P., van Rooijen, N., Tagalicud, A., Allman, M. and Wallace,

    M. (2007) Toll-like receptor-4 signaling and Kupffer cells play pivotal roles in thepathogenesis of non-alcoholic steatohepatitis. J. Hepatol. 47, 571579

    64 Huang, W., Metlakunta, A., Dedousis, N., Zhang, P., Sipula, I., Dube, J. J., Scott, D. K.

    and ODoherty, R. M. (2010) Depletion of liver Kupffer cells prevents the development of

    diet-induced hepatic steatosis and insulin resistance. Diabetes 59, 347357

    65 Neyrinck, A. M., Cani, P. D., Dewulf, E. M., Backer, F. D., Bindels, L. B. and Delzenne,

    N. M. (2009) Critical role of Kupffer cells in the management of diet-induced diabetes

    and obesity. Biochem. Biophys. Res. Commun. 385, 351356

    66 Lanthier, N., Molendi-Coste, O., Horsmans, Y., van Rooijen, N., Cani, P. D. and Leclercq,

    I. A. (2010) Kupffer cell activation is a causal factor for hepatic insulin resistance. Am. J.

    Physiol. Gastrointest. Liver Physiol. 298, G107G116

    67 Kang, K., Hatano, B. and Lee, C. H. (2007) PPAR agonists and metabolic diseases.

    Curr. Atheroscler. Rep. 9, 7277

    68 Chazaud, B., Brigitte, M., Yacoub-Youssef, H., Arnold, L., Gherardi, R., Sonnet, C.,

    Lafuste, P. and Chretien, F. (2009) Dual and beneficial roles of macrophages during

    skeletal muscle regeneration. Exercise Sport Sci. Rev. 37, 1822

    69 Cai, D., Yuan, M., Frantz, D. F., Melendez, P. A., Hansen, L., Lee, J. and Shoelson, S. E.

    (2005) Local and systemic insulin resistance resulting from hepatic activation of IKK-

    and NF-B. Nat. Med. 11, 183190

    70 Hirosumi, J., Tuncman, G., Chang, L., Gorgun, C. Z., Uysal, K. T., Maeda, K., Karin, M.

    and Hotamisligil, G. S. (2002) A central role for JNK in obesity and insulin resistance.

    Nature 420, 333336

    71 Martinez, F. O., Helming, L. and Gordon, S. (2009) Alternative activation of macrophages:

    an immunologic functional perspective. Annu. Rev. Immunol. 27, 451483

    72 Solinas, G., Vilcu, C., Neels, J., Bandyopadhyay, G., Luo, J., Naugler, W., Grivennikov,

    S., Wynshawboris, A., Scadeng, M. and Olefsky, J. (2007) JNK1 in hematopoietically

    derived cells contributes to diet-induced inflammation and insulin resistance without

    affecting obesity. Cell Metab. 6, 386397

    73 Romanatto, T., Roman, E. A., Arruda, A. P., Denis, R. G., Solon, C., Milanski, M.,

    Moraes, J. C., Bonfleur, M. L., Degasperi, G. R., Picardi, P. K. et al. (2009) Deletion of

    tumor necrosis factor- receptor 1 (TNFR1) protects against diet-induced obesity by

    means of increased thermogenesis. J. Biol. Chem. 284, 3621336222

    74 De Taeye, B. M., Novitskaya, T., Mcguinness, O. P., Gleaves, L., Medda, M., Covington,

    J. W. and Vaughan, D. E. (2007) Macrophage TNF- contributes to insulin resistance

    and hepatic steatosis in diet-induced obesity. Am. J. Physiol. Endocrinol. Metab. 293,

    E713E725

    75 Nakamura, T., Furuhashi, M., Li, P., Cao, H., Tuncman, G., Sonenberg, N., Gorgun, C. Z.

    and Hotamisligil, G. S. (2010) Double-stranded RNA-dependent protein kinase links

    pathogen sensing with stress and metabolic homeostasis. Cell 140, 338348

    76 Kumar, H., Kawai, T. and Akira, S. (2009) Pathogen recognition in the innate immune

    response. Biochem. J.420

    , 116

    77 Nguyen, M. T., Favelyukis, S., Nguyen, A.-K., Reichart, D., Scott, P. A., Jenn, A.,

    Liu-Bryan, R., Glass, C. K., Neels, J. G. and Olefsky, J. M. (2007) 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. J. Biol. Chem. 282,

    3527935292

    78 Shi, H., Kokoeva, M. V., Inouye, K., Tzameli, I., Yin, H. and Flier, J. S. (2006) TLR4 links

    innate immunity and fatty acid-induced insulin resistance. J. Clin. Invest. 116,

    30153025

    79 Holland, W. L., Bikman, B. T., Wang, L.-P., Yuguang, G., Sargent, K. M., Bulchand, S.,

    Knotts, T. A., Shui, G., Clegg, D. J., Wenk, M. R. et al. (2011) Lipid-induced insulin

    resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty

    acid-induced ceramide biosynthesis in mice. J. Clin. Invest. 121, 18581870

    80 Poggi, M., Bastelica, D., Gual, P., Iglesias, M. A., Gremeaux, T., Knauf, C., Peiretti, F.,

    Verdier, M., Juhan-Vague, I., Tanti, J. F. et al. (2007) C3H/HeJ mice carrying a Toll-like

    receptor 4 mutation are protected against the development of insulin resistance in white

    adipose tissue in response to a high-fat diet. Diabetologia 50, 1267127681 Tsukumo, D. M., Carvalho-Filho, M. A., Carvalheira, J. B., Prada, P. O., Hirabara, S. M.,

    Schenka, A. A., Araujo, E. P., Vassallo, J., Curi, R., Velloso, L. A. and Saad, M. J. (2007)

    Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and

    insulin resistance. Diabetes 56, 19861998

    82 Davis, J. E., Gabler, N. K., Walker-Daniels, J. and Spurlock, M. E. (2008) Tlr-4 deficiency

    selectively protects against obesity induced by diets high in saturated fat. Obesity 16,

    12481255

    83 Saberi, M., Woods, N.-B., de Luca, C., Schenk, S., Lu, J. C., Bandyopadhyay, G., Verma,

    I. M. and Olefsky, J. M. (2009) Hematopoietic cell-specific deletion of Toll-like receptor

    4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Cell

    Metab. 10, 419429

    84 Arkan, M. C., Hevener, A. L., Greten, F. R., Maeda, S., Li, Z. W., Long, J. M.,

    Wynshaw-Boris, A., Poli, G., Olefsky, J. and Karin, M. (2005) IKK- links inflammation

    to obesity-induced insulin resistance. Nat. Med. 11, 191198

    85 Chiang, S. H., Bazuine, M., Lumeng, C. N., Geletka, L. M., Mowers, J., White, N. M.,

    Ma, J. T., Zhou, J., Qi, N., Westcott, D. et al. (2009) The protein kinase IKK regulatesenergy balance in obese mice. Cell 138, 961975

    86 Yuan, M. (2001) Reversal of obesity- and diet-induced insulin resistance with salicylates

    or targeted disruption of IKK . Science 293, 16731677

    87 Shoelson, S. E., Lee, J. and Yuan, M. (2003) Inflammation and the IKK/IB/NF-B

    axis in obesity- and diet-induced insulin resistance. Int. J. Obes. Relat. Metab. Disord.

    27 (Suppl. 3), S49S52

    88 Hundal, R. S., Petersen, K. F., Mayerson, A. B., Randhawa, P. S., Inzucchi, S., Shoelson,

    S. E. and Shulman, G. I. (2002) Mechanism by which high-dose aspirin improves

    glucose metabolism in type 2 diabetes. J. Clin. Invest. 109, 13211326

    89 Mullick, A. E., Tobias, P. S. and Curtiss, L. K. (2005) Modulation of atherosclerosis in

    mice by Toll-like receptor 2. J. Clin. Invest. 115, 31493156

    90 Michelsen, K. S., Wong, M. H., Shah, P. K., Zhang, W., Yano, J., Doherty, T. M., Akira, S.,

    Rajavashisth, T. B. and Arditi, M. (2004) Lack of Toll-like receptor 4 or myeloid

    differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice

    deficient in apolipoprotein E. Proc. Natl. Acad. Sci. U.S.A. 101, 1067910684

    91 Kanters, E., Gijbels, M. J., van der Made, I., Vergouwe, M. N., Heeringa, P., Kraal, G.,Hofker, M. H. and de Winther, M. P. (2004) Hematopoietic NF-B1 deficiency results in

    small atherosclerotic lesions with an inflammatory phenotype. Blood 103, 934940

    92 Ferreira, V., van Dijk, K. W., Groen, A. K., Vos, R. M., van der Kaa, J., Gijbels, M. J.,

    Havekes, L. M. and Pannekoek, H. (2007) Macrophage-specific inhibition of NF-B

    activation reduces foam-cell formation. Atherosclerosis 192, 283290

    93 Dunne, A. (2011) Inflammasome activation: from inflammatory disease to infection.

    Biochem. Soc. Trans. 39, 669673

    94 Jin, C. and Flavell, R. A. (2010) Molecular mechanism of NLRP3 inflammasome

    activation. J. Clin. Immunol. 30, 628631

    95 Lumeng, C. N. and Saltiel, A. R. (2011) Inflammatory links between obesity and

    metabolic disease. J. Clin. Invest. 121, 21112117

    96 Martinon, F., Mayor, A. and Tschopp, J. (2009) The inflammasomes: guardians of the

    body. Annu. Rev. Immunol. 27, 229265

    97 Wen, H., Gris, D., Lei, Y., Jha, S., Zhang, L., Huang, M. T., Brickey, W. J. and Ting, J. P.

    (2011) Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin

    signaling. Nat. Immunol. 12, 408415

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    9/10

    Role and function of macrophages in the metabolic syndrome 261

    98 Stienstra, R., Joosten, L. A., Koenen, T., van Tits, B., van Diepen, J. A., van den Berg,

    S. A., Rensen, P. C., Voshol, P. J., Fantuzzi, G., Hijmans, A. et al. (2010) The

    inflammasome-mediated caspase-1 activation controls adipocyte differentiation and

    insulin sensitivity. Cell Metab. 12, 593605

    99 Vandanmagsar, B., Youm, Y.-H., Ravussin, A., Galgani, J. E., Stadler, K., Mynatt, R. L.,

    Ravussin, E., Stephens, J. M. and Dixit, V. D. (2011) The NLRP3 inflammasome

    instigates obesity-induced inflammation and insulin resistance. Nat. Med. 17, 179188

    100 McGillicuddy, F. C., Harford, K. A., Reynolds, C. M., Oliver, E., Claessens, M., Mills, K.

    H.G. and Roche, H. M. (2011) Lack of interleukin-1 receptor I (IL-1RI) protects mice

    from high-fat diet-induced adipose tissue inflammation coincident with improved

    glucose homeostasis. Diabetes 60, 16881698

    101 Duewell, P., Kono, H., Rayner, K. J., Sirois, C. M., Vladimer, G., Bauernfeind, F. G.,

    Abela, G. S., Franchi, L., Nunez, G., Schnurr, M. et al. (2010) NLRP3 inflammasomes are

    required for atherogenesis and activated by cholesterol crystals. Nature 464, 13571361

    102 Rajamaki, K., Lappalainen, J., Oorni, K., Valimaki, E., Matikainen, S., Kovanen, P. T. and

    Eklund, K. K. (2010) Cholesterol crystals activate the NLRP3 inflammasome in human

    macrophages: a novel link between cholesterol metabolism and inflammation. PLoS

    ONE 5, e11765

    103 Hotamisligil, G. S. (2005) Role of endoplasmic reticulum stress and c-Jun NH2-terminal

    kinase pathways in inflammation and origin of obesity and diabetes. Diabetes 54 (Suppl.

    2), S73S78

    104 Hotamisligil, G. S. (2006) Inflammation and metabolic disorders. Nature 444, 860867

    105 Ozcan, U., Cao, Q., Yilmaz, E., Lee, A. H., Iwakoshi, N. N., Ozdelen, E., Tuncman, G.,

    Gorgun, C., Glimcher, L. H. and Hotamisligil, G. S. (2004) Endoplasmic reticulum stress

    links obesity, insulin action, and type 2 diabetes. Science 306, 457461106 Han, S., Liang, C. P., DeVries-Seimon, T., Ranalletta, M., Welch, C. L., Collins-Fletcher,

    K., Accili, D., Tabas, I. and Tall, A. R. (2006) Macrophage insulin receptor deficiency

    increases ER stress-induced apoptosis and necrotic core formation in advanced

    atherosclerotic lesions. Cell Metab. 3, 257266

    107 Myoishi, M., Hao, H., Minamino, T., Watanabe, K., Nishihira, K., Hatakeyama, K., Asada,

    Y., Okada, K., Ishibashi-Ueda, H., Gabbiani, G. et al. (2007) Increased endoplasmic

    reticulum stress in atherosclerotic plaques associated with acute coronary syndrome.

    Circulation 116, 12261233

    108 Thorp, E., Li, G., Seimon, T. A., Kuriakose, G., Ron, D. and Tabas, I. (2009) Reduced

    apoptosis and plaque necrosis in advanced atherosclerotic lesions of Apoe / and

    Ldlr/ mice lacking CHOP. Cell Metab. 9, 474481

    109 Erbay, E., Babaev, V. R., Mayers, J. R., Makowski, L., Charles, K. N., Snitow, M. E., Fazio,

    S., Wiest, M. M., Watkins, S. M., Linton, M. F. and Hotamisligil, G. S. (2009) Reducing

    endoplasmic reticulum stress through a macrophage lipid chaperone alleviates

    atherosclerosis. Nat. Med. 15, 13831391

    110 Blouin, C. C., Page, E. L., Soucy, G. M. and Richard, D. E. (2004) Hypoxic geneactivation by lipopolysaccharide in macrophages: implication of hypoxia-inducible

    factor 1. Blood 103, 11241130

    111 Cramer, T. and Johnson, R. S. (2003) A novel role for the hypoxia inducible transcription

    factor HIF-1: critical regulation of inflammatory cell function. Cell Cycle 2, 192193

    112 Cramer, T., Yamanishi, Y., Clausen, B. E., Forster, I., Pawlinski, R., Mackman, N., Haase,

    V. H., Jaenisch, R., Corr, M., Nizet, V. et al. (2003) HIF-1 is essential for myeloid

    cell-mediated inflammation. Cell 112, 645657

    113 Peyssonnaux, C., Datta, V., Cramer, T., Doedens, A., Theodorakis, E. A., Gallo, R. L.,

    Hurtado-Ziola, N., Nizet, V. and Johnson, R. S. (2005) HIF-1 expression regulates the

    bactericidal capacity of phagocytes. J. Clin. Invest. 115, 18061815

    114 Zinkernagel, A. S., Johnson, R. S. and Nizet, V. (2007) Hypoxia inducible factor (HIF)

    function in innate immunity and infection. J. Mol. Med. 85, 13391346

    115 Hosogai, N., Fukuhara, A., Oshima, K., Miyata, Y., Tanaka, S., Segawa, K., Furukawa, S.,

    Tochino, Y., Komuro, R., Matsuda, M. and Shimomura, I. (2007) Adipose tissue hypoxia

    in obesity and its impact on adipocytokine dysregulation. Diabetes 56, 901911

    116 Rausch, M. E., Weisberg, S., Vardhana, P. and Tortoriello, D. V. (2008) Obesity inC57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell

    infiltration. Int. J. Obes. 32, 451463

    117 Iiyori, N., Alonso, L. C., Li, J., Sanders, M. H., Garcia-Ocana, A., ODoherty, R. M.,

    Polotsky, V. Y. and ODonnell, C. P. (2007) Intermittent hypoxia causes insulin resistance

    in lean mice independent of autonomic activity. Am. J. Respir. Crit. Care Med. 175,

    851857

    118 Aron-Wisnewsky, J., Minville, C., Tordjman, J., Levy, P., Bouillot, J. L., Basdevant, A.,

    Bedossa, P., Clement, K. and Pepin, J. L. (2011) Chronic intermittent hypoxia is a major

    trigger for non-alcoholic fatty liver disease in morbid obese. J. Hepatol. 56, 225233

    119 Deguchi, J. O., Yamazaki, H., Aikawa, E. and Aikawa, M. (2009) Chronic hypoxia

    activates the Akt and -catenin pathways in human macrophages. Arterioscler. Thromb.

    Vasc. Biol. 29, 16641670

    120 Parathath, S., Mick, S. L., Feig, J. E., Joaquin, V., Grauer, L., Habiel, D. M., Gassmann,

    M., Gardner, L. B. and Fisher, E. A. (2011) Hypoxia is present in murine atherosclerotic

    plaques and has multiple adverse effects on macrophage lipid metabolism. Circ. Res.

    109, 11411152

    121 Arnaud, C., Poulain, L., Levy, P. and Dematteis, M. (2011) Inflammation contributes to

    the atherogenic role of intermittent hypoxia in apolipoprotein-E knock out mice.

    Atherosclerosis 219, 425431

    122 Kang, K., Reilly, S. M., Karabacak, V., Gangl, M. R., Fitzgerald, K., Hatano, B. and Lee,

    C. H. (2008) Adipocyte-derived Th2 cytokines and myeloid PPAR regulate macrophage

    polarization and insulin sensitivity. Cell Metab. 7, 485495

    123 Ricardo-Gonzalez, R. R., Red Eagle, A., Odegaard, J. I., Jouihan, H., Morel, C. R.,

    Heredia, J. E., Mukundan, L., Wu, D., Locksley, R. M. and Chawla, A. (2010) IL-4/STAT6

    immune axis regulates peripheral nutrient metabolism and insulin sensitivity. Proc. Natl.Acad. Sci. U.S.A. 107, 2261722622

    124 Wu, D., Molofsky, A. B., Liang, H. E., Ricardo-Gonzalez, R. R., Jouihan, H. A., Bando,

    J. K., Chawla, A. and Locksley, R. M. (2011) Eosinophils sustain adipose alternatively

    activated macrophages associated with glucose homeostasis. Science 332, 243247

    125 Chawla, A., Repa, J. J., Evans, R. M. and Mangelsdorf, D. J. (2001) Nuclear receptors

    and lipid physiology: opening the X-files. Science 294, 18661870

    126 Lee, C. H., Olson, P. and Evans, R. M. (2003) Minireview: lipid metabolism, metabolic

    diseases, and peroxisome proliferator-activated receptors. Endocrinology 144,

    22012207

    127 Reilly, S. M. and Lee, C. H. (2008) PPAR as a therapeutic target in metabolic disease.

    FEBS Lett. 582, 2631

    128 Szanto, A. and Roszer, T. (2008) Nuclear receptors in macrophages: a link between

    metabolism and inflammation. FEBS Lett. 582, 106116

    129 Hong, C. and Tontonoz, P. (2008) Coordination of inflammation and metabolism by

    PPAR and LXR nuclear receptors. Curr. Opin. Genet. Dev. 18, 461467

    130 Vats, D., Mukundan, L., Odegaard, J. I., Zhang, L., Smith, K. L., Morel, C. R., Wagner,R. A., Greaves, D. R., Murray, P. J. and Chawla, A. (2006) Oxidative metabolism and

    PGC-1 attenuate macrophage-mediated inflammation. Cell Metab. 4, 1324

    131 Lee, C. H., Kang, K., Mehl, I. R., Nofsinger, R., Alaynick, W. A., Chong, L. W., Rosenfeld,

    J. M. and Evans, R. M. (2006) Peroxisome proliferator-activated receptor promotes

    very low-density lipoprotein-derived fatty acid catabolism in the macrophage. Proc. Natl.

    Acad. Sci. U.S.A. 103, 24342439

    132 Odegaard, J. I., Ricardo-Gonzalez, R. R., Goforth, M. H., Morel, C. R., Subramanian, V.,

    Mukundan, L., Eagle, A. R., Vats, D., Brombacher, F., Ferrante, A. W. and Chawla, A.

    (2007) Macrophage-specific PPAR controls alternative activation and improves insulin

    resistance. Nature 447, 11161120

    133 Bouhlel, M. A., Derudas, B., Rigamonti, E., Dievart, R., Brozek, J., Haulon, S., Zawadzki,

    C., Jude, B., Torpier, G., Marx, N. et al. (2007) PPAR activation primes human

    monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell

    Metab. 6, 137143

    134 Hevener, A. L., Olefsky, J. M., Reichart, D., Nguyen, M. T., Bandyopadyhay, G., Leung,

    H. Y., Watt, M. J., Benner, C., Febbraio, M. A., Nguyen, A. K. et al. (2007) MacrophagePPAR is required for normal skeletal muscle and hepatic insulin sensitivity and full

    antidiabetic effects of thiazolidinediones. J. Clin. Invest. 117, 16581669

    135 Odegaard, J. I., Ricardo-Gonzalez, R. R., Red Eagle, A., Vats, D., Morel, C. R., Goforth,

    M. H., Subramanian, V., Mukundan, L., Ferrante, A. W. and Chawla, A. (2008) Alternative

    M2 activation of Kupffer cells by PPAR ameliorates obesity-induced insulin resistance.

    Cell Metab. 7, 496507

    136 Chawla, A. (2010) Control of macrophage activation and function by PPARs. Circ. Res.

    106, 15591569

    137 Barish, G. D., Narkar, V. A. and Evans, R. M. (2006) PPAR: a dagger in the heart of the

    metabolic syndrome. J. Clin. Invest. 116, 590597

    138 Rigamonti, E., Chinetti-Gbaguidi, G. and Staels, B. (2008) Regulation of macrophage

    functions by PPAR-, PPAR-, and LXRs in mice and men. Arterioscler. Thromb. Vasc.

    Biol. 28, 10501059

    139 de Lorgeril, M., Renaud, S., Mamelle, N., Salen, P., Martin, J. L., Monjaud, I., Guidollet,

    J., Touboul, P. and Delaye, J. (1994) Mediterranean -linolenic acid-rich diet in

    secondary prevention of coronary heart disease. Lancet 343, 14541459

    140 Esposito, K., Marfella, R., Ciotola, M., Di Palo, C., Giugliano, F., Giugliano, G.,

    DArmiento, M., DAndrea, F. and Giugliano, D. (2004) Effect of a Mediterranean-style

    diet on endothelial dysfunction and markers of vascular inflammation in the metabolic

    syndrome: a randomized trial. JAMA, J. Am. Med. Assoc. 292, 14401446

    141 Kabir, M., Skurnik, G., Naour, N., Pechtner, V., Meugnier, E., Rome, S.,

    Quignard-Boulange, A., Vidal, H., Slama, G., Clement, K. et al. (2007) Treatment for 2

    mo with n 3 polyunsaturated fatty acids reduces adiposity and some atherogenic

    factors but does not improve insulin sensitivity in women with type 2 diabetes: a

    randomized controlled study. Am. J. Clin. Nutr. 86, 16701679

    142 Yashodhara, B. M., Umakanth, S., Pappachan, J. M., Bhat, S. K., Kamath, R. and Choo,

    B. H. (2009) 3 fatty acids: a comprehensive review of their role in health and

    disease. Postgrad. Med. J. 85, 8490

    143 Serhan, C. N. (2009) Systems approach to inflammation resolution: identification of

    novel anti-inflammatory and pro-resolving mediators. J. Thromb. Haemostasis 7

    (Suppl. 1), 4448

    c The Authors Journal compilation c 2012 Biochemical Society

  • 7/31/2019 Role of Macrphage in Metabolic Syndrome

    10/10

    262 P. Bhargava and C.-H. Lee

    144 Oh, D. Y., Talukdar, S., Bae, E. J., Imamura, T., Morinaga, H., Fan, W., Li, P., Lu, W. J.,

    Watkins, S. M. and Olefsky, J. M. (2010) GPR120 is an 3 fatty acid receptor

    mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142,

    687698

    145 Suzuki, T., Igari, S., Hirasawa, A., Hata, M., Ishiguro, M., Fujieda, H., Itoh, Y., Hirano, T.,

    Nakagawa, H., Ogura, M. et al. (2008) Identification of G protein-coupled receptor

    120-selective agonists derived from PPAR agonists. J. Med. Chem. 51, 76407644

    146 Newsholme, P., Curi, R., Gordon, S. and Newsholme, E. A. (1986) Metabolism of

    glucose, glutamine, long-chain fatty acids and ketone bodies by murine macrophages.Biochem. J. 239, 121125

    147 Newsholme, P., Costa Rosa, L. F. B. P., Newsholme, E. A. and Curi, R. (1996) The

    importance of fuel metabolism to macrophage function. Cell Biochem. Funct. 14,

    110

    148 Simon, L. M., Axline, S. G., Horn, B. R. and Robin, E. D. (1973) Adaptations in energy

    metabolism in the cultivated macrophage. J. Exp. Med. 138, 14131425

    149 Virkamaki, A. and Yki-Jarvinen, H. (1994) Mechanisms of insulin resistance during

    acute endotoxemia. Endocrinology 134, 20722078

    150 Yki-Jarvinen, H., Sammalkorpi, K., Koivisto, V. A. and Nikkila, E. A. (1989) Severity,

    duration, and mechanisms of insulin resistance during acute infections. J. Clin.

    Endocrinol. Metab. 69, 317323

    151 Agwunobi, A. O., Reid, C., Maycock, P., Little, R. A. and Carlson, G. L. (2000) Insulin

    resistance and substrate utilization in human endotoxemia. J. Clin. Endocrinol. Metab.

    85, 37703778

    152 Haffner, S., Temprosa, M., Crandall, J., Fowler, S., Goldberg, R., Horton, E., Marcovina,

    S., Mather, K., Orchard, T., Ratner, R. and Barrett-Connor, E. (2005) Intensive lifestyleintervention or metformin on inflammation and coagulation in participants with impaired

    glucose tolerance. Diabetes 54, 15661572

    153 Oliver, E., McGillicuddy, F., Phillips, C., Toomey, S. and Roche, H. M. (2010) The role of

    inflammation and macrophage accumulation in the development of obesity-induced

    type 2 diabetes mellitus and the possible therapeutic effects of long-chain n 3 PUFA.

    Proc. Nutr. Soc. 69, 232243

    154 Wang, T. J., Larson, M. G., Vasan, R. S., Cheng, S., Rhee, E. P., McCabe, E., Lewis, G.

    D., Fox, C. S., Jacques, P. F., Fernandez, C. et al. (2011) Metabolite profiles and the risk

    of developing diabetes. Nat. Med. 17, 448453

    155 Newgard, C. B., An, J., Bain, J. R., Muehlbauer, M. J., Stevens, R. D., Lien, L. F., Haqq,

    A. M., Shah, S. H., Arlotto, M., Slentz, C. A. et al. (2009) A branched-chain amino

    acid-related metabolic signature that differentiates obese and lean humans and

    contributes to insulin resistance. Cell Metab. 9, 311326

    156 Newsholme, P. (2001) Why is L-glutamine metabolism important to cells of the immune

    system in health, postinjury, surgery or infection? J. Nutr. 131 (Suppl.), 2515S2522S

    157 Weichhart, T., Costantino, G., Poglitsch, M., Rosner, M., Zeyda, M., Stuhlmeier, K. M.,

    Kolbe, T., Stulnig, T. M., Horl, W. H., Hengstschlager, M. et al. (2008) The TSCmTOR

    signaling pathway regulates the innate inflammatory response. Immunity 29,

    565577

    158 Sabio, G., Das, M., Mora, A., Zhang, Z., Jun, J. Y., Ko, H. J., Barrett, T., Kim, J. K. and

    Davis, R. J. (2008) A stress signaling pathway in adipose tissue regulates hepatic insulin

    resistance. Science 322, 15391543

    159 Vallerie, S. N., Furuhashi, M., Fucho, R. and Hotamisligil, G. S. (2008) A predominant

    role for parenchymal c-Jun amino terminal kinase (JNK) in the regulation of systemic

    insulin sensitivity. PLoS ONE 3, e3151

    160 Feuerer, M., Herrero, L., Cipolletta, D., Naaz, A., Wong, J., Nayer, A., Lee, J., Goldfine,

    A. B., Benoist, C., Shoelson, S. and Mathis, D. (2009) Lean, but not obese, fat is

    enriched for a unique population of regulatory T cells that affect metabolic parameters.

    Nat. Med. 15, 930939

    161 Zhang, J. and Shi, G.-P. (2011) Mast cells and metabolic syndrome. Biochim. Biophys.

    Acta 1822, 1420

    162 ORourke, R. W., Metcalf, M. D., White, A. E., Madala, A., Winters, B. R., Maizlin, I. I.,

    Jobe, B. A., Roberts, Jr, C. T., Slifka, M. K. and Marks, D. L. (2009) Depot-specific

    differences in inflammatory mediators and a role for NK cells and IFN- in inflammationin human adipose tissue. Int. J. Obes. 33, 978990

    163 Ohmura, K., Ishimori, N., Ohmura, Y., Tokuhara, S., Nozawa, A., Horii, S., Andoh, Y.,

    Fujii, S., Iwabuchi, K., Onoe, K. and Tsutsui, H. (2010) Natural killer T cells are involved

    in adipose tissues inflammation and glucose intolerance in diet-induced obese mice.

    Arterioscler. Thromb. Vasc. Biol. 30, 193199

    164 Nishimura, S., Manabe, I., Nagasaki, M., Eto, K., Yamashita, H., Ohsugi, M., Otsu, M.,

    Hara, K., Ueki, K., Sugiura, S. et al. (2009) CD8 + effector T cells contribute to

    macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15,

    914920

    165 Winer, S., Chan, Y., Paltser, G., Truong, D., Tsui, H., Bahrami, J., Dorfman, R., Wang, Y.,

    Zielenski, J., Mastronardi, F. et al. (2009) Normalization of obesity-associated insulin

    resistance through immunotherapy. Nat. Med. 15, 921929

    166 Ilan, Y., Maron, R., Tukpah, A. M., Maioli, T. U., Murugaiyan, G., Yang, K., Wu, H. Y. and

    Weiner, H. L. (2010) Induction of regulatory T cells decreases adipose inflammation and

    alleviates insulin resistance in ob/obmice. Proc. Natl. Acad. Sci. U.S.A. 107,

    97659770167 ONeill, L. A.J., Sheedy, F. J. and McCoy, C. E. (2011) MicroRNAs: the fine-tuners of

    Toll-like receptor signalling. Nat. Rev. Immunol. 11, 163175

    168 Pandey, A. K., Agarwal, P., Kaur, K. and Datta, M. (2009) MicroRNAs in diabetes: tiny

    players in big disease. Cell. Physiol. Biochem. 23, 221232

    169 Vickers, K. C. and Remaley, A. T. (2010) MicroRNAs in atherosclerosis and lipoprotein

    metabolism. Curr. Opin. Endocrinol. Diabetes Obes. 17, 150155

    170 Ley, R. E., Turnbaugh, P. J., Klein, S. and Gordon, J. I. (2006) Microbial ecology: human

    gut microbes associated with obesity. Nature 444, 10221023

    171 Vijay-Kumar, M., Aitken, J. D., Carvalho, F. A., Cullender, T. C., Mwangi, S.,

    Srinivasan, S., Sitaraman, S. V., Knight, R., Ley, R. E. and Gewirtz, A. T. (2010) Metabolic

    syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 328,

    228231

    172 Brandt, C. and Pedersen, B. K. (2010) The role of exercise-induced myokines in muscle

    homeostasis and the defense against chronic diseases. J. Biomed. Biotechnol. 2010,

    520258

    173 Pedersen, B. K. (2011) Muscles and their myokines. J. Exp. Biol. 214, 337346174 Shaw, C. S., Clark, J. and Wagenmakers, A. J. (2010) The effect of exercise and nutrition

    on intramuscular fat metabolism and insulin sensitivity. Annu. Rev. Nutr. 30, 1334

    175 Pedersen, B. K., Akerstrom, T. C., Nielsen, A. R. and Fischer, C. P. (2007) Role of

    myokines in exercise and metabolism. J. Appl. Physiol. 103, 10931098

    176 Beavers, K. M. and Nicklas, B. J. (2011) Effects of lifestyle interventions on inflammatory

    markers in the metabolic syndrome. Front. Biosci. (Schol. Ed.) 3, 168177

    177 Christ, M., Iannello, C., Iannello, P. G. and Grimm, W. (2004) Effects of a weight

    reduction program with and without aerobic exercise in the metabolic syndrome. Int. J.

    Cardiol. 97, 115122

    Received 23 September 2011/15 November 2011; accepted 16 November 2011Published on the Internet 13 February 2012, doi:10.1042/BJ20111708

    c The Authors Journal compilation c 2012 Biochemical Society