Hostname: page-component-7c8c6479df-ph5wq Total loading time: 0 Render date: 2024-03-26T20:27:14.150Z Has data issue: false hasContentIssue false

Insights into the role of macrophage migration inhibitory factor in obesity and insulin resistance

Published online by Cambridge University Press:  23 August 2012

Orla M. Finucane
Affiliation:
Insitiute of Molecular Medicine, School of Medicine, Trinity Centre for Health Sciences, St James Hospital, Dublin 8, Republic of Ireland Nutrigenomics Research Group, UCD Conway Institute, School of Public Health and Population Science, University College Dublin, Dublin 4, Republic of Ireland
Clare M. Reynolds
Affiliation:
Nutrigenomics Research Group, UCD Conway Institute, School of Public Health and Population Science, University College Dublin, Dublin 4, Republic of Ireland
Fiona C. McGillicuddy
Affiliation:
Nutrigenomics Research Group, UCD Conway Institute, School of Public Health and Population Science, University College Dublin, Dublin 4, Republic of Ireland
Helen M. Roche*
Affiliation:
Nutrigenomics Research Group, UCD Conway Institute, School of Public Health and Population Science, University College Dublin, Dublin 4, Republic of Ireland
*
*Corresponding author: Professor Helen M. Roche, fax+353 1 716 7601, email Helen.roche@ucd.ie
Rights & Permissions [Opens in a new window]

Abstract

High-fat diet (HFD)-induced obesity has emerged as a state of chronic low-grade inflammation characterised by a progressive infiltration of immune cells, particularly macrophages, into obese adipose tissue. Adipose tissue macrophages (ATM) present immense plasticity. In early obesity, M2 anti-inflammatory macrophages acquire an M1 pro-inflammatory phenotype. Pro-inflammatory cytokines including TNF-α, IL-6 and IL-1β produced by M1 ATM exacerbate local inflammation promoting insulin resistance (IR), which consequently, can lead to type-2 diabetes mellitus (T2DM). However, the triggers responsible for ATM recruitment and activation are not fully understood. Adipose tissue-derived chemokines are significant players in driving ATM recruitment during obesity. Macrophage migration inhibitory factor (MIF), a chemokine-like inflammatory regulator, is enhanced during obesity and is directly associated with the degree of peripheral IR. This review focuses on the functional role of macrophages in obesity-induced IR and highlights the importance of the unique inflammatory cytokine MIF in propagating obesity-induced inflammation and IR. Given MIF chemotactic properties, MIF may be a primary candidate promoting ATM recruitment during obesity. Manipulating MIF inflammatory activities in obesity, using pharmacological agents or functional foods, may be therapeutically beneficial for the treatment and prevention of obesity-related metabolic diseases.

Type
Irish Section Postgraduate Symposium
Copyright
Copyright © The Authors 2012

Abbreviations:
ATM

adipose tissue macrophage

CCL

CC-chemokine ligand

CCR

CC-chemokine receptor

DIO

diet-induced obese

HFD

High-fat diet

IR

insulin resistance

JNK

c-Jun N-terminal kinases

MCP-1

moncyte chemoattractant protein-1

MIF

migration inhibitory factor

T2DM

type-2 diabetes mellitus

TLR

toll-like receptor

The incidence of obesity has escalated to epidemic proportions. In 2008, an estimated 1·45 billion adults were overweight worldwide (BMI > 25 kg/m2), 500 million of whom were obese (BMI > 30 kg/m2)( Reference Flegal, Graubard and Williamson 1 ). Alarmingly, in 2010 approximately 43 million children under 5 years of age were classed as overweight( 2 ). This global surge in obesity can be largely attributed to the increased availability and consumption of high-fat foods in combination with sedentary lifestyle( Reference Finucane, Stevens and Cowan 3 , Reference Swinburn, Sacks and Hall 4 ). Consequently, there has been a substantial rise in obesity-related co-morbidities; dyslipidemia, hypertriglycemia, insulin resistance (IR), type-2 diabetes mellitus (T2DM), CVD and non-alcoholic fatty liver disease, collectively known as the metabolic syndrome( Reference Flegal, Graubard and Williamson 1 , Reference Berrington de Gonzalez 5 ). As such, obesity presents an astronomical financial cost and burden on health care systems( Reference Zimmet, Alberti and Shaw 6 ). Accumulating evidence indicates that chronic low-grade inflammation is the critical link in the pathogenesis of obesity-related disorders. Disproportionate production of pro- and anti-inflammatory adipose tissue-derived cytokines from hypertrophied adipocytes is a critical determinant of obesity-induced IR( Reference Hotamisligil, Shargill and Spiegelman 7 , Reference Weisberg, McCann and Desai 8 ). The increased presence of macrophages within obese adipose tissue has emerged as the predominant source of inflammatory mediators, which contribute to adipose tissue dysfunction and systemic IR( Reference Weisberg, McCann and Desai 8 , Reference Xu, Barnes and Yang 9 ). Unravelling the mechanisms responsible for adipose tissue macrophage (ATM) infiltration, activation and ensuing adipocyte dysregulation is imperative to our understanding of the pathophysiological progression to IR and T2DM. Macrophage migration inhibitory factor (MIF) is a principal regulator of innate immune responses and has been implicated in numerous inflammatory and autoimmune diseases such as septic shock and rheumatoid arthritis( Reference Morand, Leech and Bernhagen 10 ). Several studies have documented an association between MIF and metabolic deregulation and a relationship between MIF and obesity has transpired( Reference Serre-Beinier, Toso and Morel 11 , Reference Koska, Stefan and Dubois 12 ). This review summarises the importance of macrophages in high-fat diet (HFD)-induced adipose tissue inflammation and IR and outlines the known role of MIF in inflammation and metabolic complications.

Obesity and insulin resistance

IR is the hallmark of obesity-induced metabolic complications and is defined by the inability of metabolic tissues including adipose tissue, skeletal muscle and liver to appropriately respond to insulin action( Reference Kahn and Flier 13 ). Insulin is a pleiotropic hormone secreted by pancreatic β-cells. It signals via the tyrosine phosphorylation of the insulin receptor substrate molecule, which subsequently activates a cascade of intracellular signalling events. Insulin activates two main signalling pathways one of which is phosphatidylinositol-3-kinase–AKT pathway primarily responsible for mediating glucose uptake( Reference de Luca and Olefsky 14 , Reference Taniguchi, Kondo and Sajan 15 ). Upon insulin stimulation, the GLUT-4 promptly translocates from intracellular storage sites to the plasma membrane, facilitating glucose uptake( Reference Abel, Peroni and Kim 16 , Reference Schenk, Saberi and Olefsky 17 ). Insulin also exhibits anti-lipolytic activities in adipose tissue; inhibiting hormone-sensitive lipase activity and reducing NEFA release( Reference Duncan, Ahmadian and Jaworski 18 ). Adipose tissue IR materialises as impaired insulin-mediated glucose uptake and elevated lipolysis inducing hyperlipidaemia, hyperglycaemia and hyperinsulinaemia. Insulin-resistant skeletal muscle displays reduced glucose disposal( Reference Shulman 19 ). On the other hand, the liver exhibits selective IR, whereby the inhibitory effect of insulin on hepatic gluconeogenesis is interrupted while its action on lipogenesis is enhanced, resulting in chronic hyperglycaemia and hypertriglyceridemia( Reference Schwarz, Linfoot and Dare 20 , Reference Brown and Goldstein 21 ). Considerable evidence places emphasis on the importance of inflammation in pathogenesis of obesity-induced IR. Macrophage-derived inflammatory mediators including TNF-α and IL-6, impede insulin signalling( Reference Xu, Barnes and Yang 9 ). Thus, we focus on the role of macrophage-mediated inflammation in the development of IR.

Obese adipose tissue and inflammation

Obesity instigates a typical host inflammatory response( Reference Wellen and Hotamisligil 22 ). Circulating levels of inflammatory mediators including acute phase proteins: plasminogen activator inhibitor and C-reactive protein and inflammatory cytokines TNF-α, IL-6 and IL-8 are systemically elevated( Reference Schmidt, Duncan and Sharrett 23 Reference Fantuzzi 25 ). Adipose tissue is considered a critical interface in the pathogenesis of obesity-induced IR. Once perceived as a mere inert storage depot, adipose tissue is now known to be a vital endocrine organ paramount in maintaining systemic glucose and lipid homoeostasis( Reference Suganami and Ogawa 26 ). This metabolically active organ secretes copious amounts of adipokines, most notably leptin and adiponectin( Reference Zhang, Proenca and Maffei 27 , Reference Maeda, Okubo and Shimomura 28 ) and regulates NEFA flux to and from adipocytes maintaining energy homoeostasis( Reference Duncan, Ahmadian and Jaworski 18 , Reference Thompson, Lobo and Bernlohr 29 ). HFD-induced adipose tissue expansion leads to an increase in pro-inflammatory cytokine secretion and a decrease in anti-inflammatory cytokine production( Reference Suganami and Ogawa 26 , Reference Suganami and Ogawa 30 ). Indeed Hotamisligil and co-workers were the first to demonstrate that TNF-α expression was significantly increased in the adipose tissue of obese mice( Reference Hotamisligil, Shargill and Spiegelman 7 ). Moreover, chronic treatment with recombinant TNF-α was shown to disrupt insulin signalling in 3T3-L1 adipocytes. TNF-α activated intracellular inhibitor of NF-κB kinase-β, which in turn, leads to reduced insulin receptor substrate-1 and GLUT-4 mRNA expression in 3T3-L1 adipocytes( Reference Hotamisligil, Shargill and Spiegelman 7 , Reference Hotamisligil, Murray and Choy 31 ). Conversely, whole body deletion of TNF-α or its corresponding receptor TNF receptor 1 (TNFR1) gene partially protected mice from HFD-induced IR( Reference Uysal, Wiesbrock and Marino 32 ). Subsequently, a multitude of adipose tissue derived cytokines including IL-6, IL-1β and CC-chemokine ligand 2 (CCL2)/moncyte chemoattractant protein (MCP)-1 were identified to be overproduced during obesity in human subjects and rodents( Reference Wellen and Hotamisligil 22 , Reference Hotamisligil, Arner and Caro 33 ).

Adipose tissue macrophages in obesity

Despite compelling evidence signifying an association between chronic adipose tissue inflammation and IR, the cell types responsible for orchestrating this inflammatory response remained unexplored. In 2003, two separate studies demonstrated that expanding adipose mass is accompanied by a progressive infiltration of macrophages into adipose tissue (Fig. 1)( Reference Weisberg, McCann and Desai 8 , Reference Xu, Barnes and Yang 9 ). Weisberg et al. conducted transcriptional profile analysis on adipose tissue isolated from genetic and diet-induced obese (DIO) mouse models and demonstrated 1304 transcripts correlated to body mass, 30% of which encoded proteins that were characteristic of macrophages. Immunohistochemistry analysis of adipose tissue confirmed that macrophage-specific marker F4/80 expression was positively associated with adipocyte size and body mass. Furthermore, ATM were deemed the predominant source of pro-inflammatory TNF-α( Reference Weisberg, McCann and Desai 8 ). Xu et al. showed macrophage-specific genes to be significantly up-regulated in adipose tissue prior to overt hyperinsulinemia, an effect that was dramatically reduced on treatment with the insulin-sensitising drug rosiglitazone( Reference Xu, Barnes and Yang 9 ). In support of these studies, diet or surgery-induced weight loss coincided with reduced ATM infiltration, CCL2/MCP-1 gene expression and low-grade inflammation in morbidly obese individuals( Reference Bruun, Helge and Richelsen 34 , Reference Cancello, Henegar and Viguerie 35 ). These findings placed macrophage biology at the forefront of investigations in an attempt to decipher how immune and metabolic responses converge in obesity and IR. In vitro analysis demonstrated that paracrine ATM-adipocyte interactions exacerbate local inflammation( Reference Suganami and Ogawa 30 , Reference Permana, Menge and Reaven 36 ) and potentiated the development of IR and T2DM( Reference Antuna-Puente, Feve and Fellahi 37 ). Indeed Suganami et al. showed that 3T3-L1 adipocytes exposed to RAW264 macrophage-derived conditioned media exhibited increased MCP-1, IL-6 and TNF-α mRNA expression, while RAW264 macrophages incubated with adipocyte-conditioned media displayed enhanced IL-6 and TNF-α expression. TNF-α emerged as the dominant inflammatory mediator, anti-TNF-α neutralising antibody inhibited the up-regulation of adipocyte MCP-1 expression. RAW264 macrophages treated with adipocyte-derived NEFA demonstrate increased inflammatory phenotype, suggesting NEFA are an important contributor to macrophage inflammation( Reference Suganami and Ogawa 30 ). Permana et al. and Lumeng et al. extended these findings demonstrating that macrophage-secreted factors decreased adipocyte sensitivity by inducing a marked reduction in GLUT-4 and insulin receptor substrate-1 expression coincident with attenuated AKT phosphorylation and impaired insulin-stimulated glucose uptake( Reference Permana, Menge and Reaven 36 , Reference Lumeng, Deyoung and Saltiel 38 ).

Fig. 1. Obesity-induced adipose tissue macrophage (ATM) recruitment. M2 ATM are characteristic of lean adipose tissue. Adipocyte-derived arginase, adiponectin and M2-derived IL-10 are enhanced. During obesity, adipose tissue expansion is accompanied by adipocyte necrosis, hypoxia and enhanced pro-inflammatory cytokine secretion which attract pro-inflammatory M1 macrophages into adipose tissue. In this obesogenic state pro-inflammatory IL-6, IL-1β, TNF-α, inducible nitric oxide synthase (iNOS), monocyte chemoattractant protein (MCP)-1 and adipokines leptin and resistin are up-regulated, exacerbating the inflammatory milieu, while arginase, adiponectin and IL-10 are decreased. Inflammatory signalling pathways c-Jun N-terminal kinases (JNK) and p38 are activated leading to impaired insulin action and systemic insulin resistance (IR).

A dynamic interplay between multiple intracellular signalling pathways within insulin-responsive tissues is imperative in modulating both local and systemic inflammation and subsequent IR. Macrophage-derived pro-inflammatory cytokines and NEFA have been implicated in the chronic activation of c-Jun N-terminal kinases (JNK) and inhibitor of NF-κB kinase-β protein kinases, critical inflammatory regulators in the progression of obesity-related complications( Reference Solinas and Karin 39 ). Activated JNK impairs insulin action via serine phosphorylation of insulin receptor substrate-1, which disrupts downstream activation of phosphatidylinositol-3-kinase–AKT( Reference Hotamisligil, Murray and Choy 31 ). In genetic and DIO rodent models, lack of JNK-1 but not JNK-2 reduced adiposity coincident with attenuated IR and enhanced insulin signalling in liver tissue( Reference Hirosumi, Tuncman and Chang 40 ). Furthermore, whole body JNK-1 knockout and adipose-specific JNK-1 deficiency ameliorated obesity-induced hepatic inflammation and IR( Reference Tuncman, Hirosumi and Solinas 41 , Reference Sabio, Das and Mora 42 ). On the other hand, deletion of JNK-1 in haematopoietic cells did not alter body weight, but provided protection from HFD-induced IR and inflammation, while abrogation of JNK-1 in non-haematopoietic cells reduced adiposity concomitant with improved insulin sensitivity( Reference Solinas, Vilcu and Neels 43 ). Similarly, myeloid-specific deletion of inhibitor of NF-κB kinase-β alleviated HFD promoted IR( Reference Arkan MC and Greten 44 ). Taken together, these two aforementioned studies provide convincing evidence that obesity associated IR is strongly dependent on myeloid cell activity such as ATM.

Dysregulated fatty acid flux has been implicated in the activation of inflammatory macrophages. Toll-like receptor (TLR)-4 is a member of the pattern-recognition receptor family involved in innate immunity( Reference Medzhitov 45 ). Studies indicate that SFA can stimulate both macrophage and adipocyte inflammation by activating the NF-κB and JNK signalling pathway via TLR-4 in vitro ( Reference Shi, Kokoeva and Inouye 46 ). Acute lipid infusion promoted adipose tissue inflammation via NF-κB and systemic IR in WT but not TLR4−/− mice. Coincident DIO TLR4−/− mice exhibited attenuated IR, reduced ATM recruitment and improved adipose tissue inflammation( Reference Shi, Kokoeva and Inouye 46 , Reference Tsukumo, Carvalho-Filho and Carvalheira 47 ). Mice exhibiting a loss of function mutation in TLR-4 displayed reduced HFD-induced weight gain, increased energy expenditure and enhanced insulin sensitivity in peripheral tissues( Reference Tsukumo, Carvalho-Filho and Carvalheira 47 ). Furthermore, haematopoietic cell specific deletion of TLR ameliorated HFD-induced hyperinsulinemia, hepatic and adipose tissue IR, concomitant with impaired ATM content( Reference Saberi, Woods and de Luca 48 ). It is clear from these studies that TLR-4 plays a critical role in adipose tissue inflammation and IR; moreover, macrophages have transpired as the predominant mediators of TLR-4 responses in adipose tissue( Reference Harford, Reynolds and McGillicuddy 49 ).

Adipose tissue macrophages in mice v. men: limitations of the M1/M2 classification

Macrophages are professional phagocytic cells( Reference Kono and Rock 50 ) derived from peripheral blood mononuclear cells and are present in the majority of tissue types. Macrophages present diverse functionality and are classified into two populations (1) ‘classically activated’ M1 macrophages or (2) ‘alternatively activated’ M2 macrophages( Reference Weisberg, Hunter and Huber 51 ). Helper T-cell 1 derived interferon-γ or lipopolysaccharide can prime macrophages towards a pro-inflammatory phenotype( Reference Osborn and Olefsky 52 ). M1 macrophages secrete copious amounts of pro-inflammatory cytokines; TNF-α, IL-1β and IL-6, produce increased nitric oxide synthase and reactive oxygen species( Reference Mosser and Edwards 53 ). Conversely, M2 macrophages exhibit an anti-inflammatory potential and are divided into sub-populations associated with wound healing and regulatory immunity. IL-4 and IL-13 produced during innate or adaptive immunity promote wound-healing macrophages by increasing arginase activity contributing to extracellular matrix production and tissue repair( Reference Mosser and Edwards 53 ). Macrophages treated with IL-4 and/or IL-13 in vitro display reduced pro-inflammatory cytokine signature( Reference Edwards, Zhang and Frauwirth 54 ). Regulatory macrophages are induced by a transient signal such as PG in combination with a TLR ligand and are characterised by their ability to produce anti-inflammatory IL-10 and down-regulate IL-12 production( Reference Mosser and Edwards 53 ).

In 2007, the ‘phenotypic switch’ model was proposed, which suggested that in an obesogenic environment ATM are polarised from an M2 to an M1 activation state( Reference Lumeng, Bodzin and Saltiel 55 ). In these studies, M1 and M2 macrophages were characterised by differential expression of specific surface markers. M1 macrophages are defined by the presence of general macrophage marker F4/80 and myeloid cell markers CD11c, CD11b (F4/80+CD11c+CD11b+)( Reference Lumeng, Bodzin and Saltiel 55 , Reference Lumeng, Deyoung and Bodzin 56 ). M2 ATM are sparsely scattered in the interstitial spaces between adipocytes in lean adipose tissue( Reference Lumeng, Bodzin and Saltiel 55 ) and express significantly elevated levels of anti-inflammatory IL-10 in response to NEFA( Reference Nguyen, Favelyukis and Nguyen 57 ). Immunoflouresence analysis demonstrated that CD11c+ M1 cells are the predominant cell population involved in crown-like structure formation surrounding necrotic adipocytes in obese mice( Reference Lumeng, Bodzin and Saltiel 55 , Reference Nguyen, Favelyukis and Nguyen 57 ). Nguyen et al. demonstrated bone marrow-derived dendritic cells expressing F4/80+CD11b+CD11c+ are activated via TLR2/4 and JNK signalling pathways in response to NEFA and exhibit enhanced inflammatory signature secreting elevated levels of IL-1β and IL-6. Moreover, exposure of skeletal muscle cells to CD11c + -conditioned media impaired glucose uptake. Coincident adipose tissue derived F4/80+CD11b+CD11c+ cells treated with NEFA ex vivo displayed heightened inflammatory activity; increased TNF-α, CC-chemokine receptor 2 (CCR2) and TLR-4 expression, compared with CD11c cells( Reference Nguyen, Favelyukis and Nguyen 57 ). Diphtheria toxin-induced ablation of CD11c+ cells in vivo normalised insulin sensitivity in obese mice coincident with reduced local and systemic inflammatory gene expression( Reference Patsouris, Li and Thapar 58 ).

The M1/M2 macrophage classification system has been largely defined in vitro and is considered as an over-simplification of macrophage phenotype in vivo. It has become increasingly apparent that macrophages display a combination of M1/M2 properties. A recent DIO mouse model demonstrated mixed M1/M2 ATM profiles, wherein three distinct ATM populations were characterised in obese adipose tissue; MGL1+/CD11c, MGL1/CD11c+ and MGL1med/CD11c+( Reference Shaul, Bennett and Strissel 59 ). Importantly, the previously defined as M2 ATM (MGL1+/CD11c), acquired an alternative transcriptional profile on exposure to an HFD. Classical M2 transcripts including IL-1Ra and IL-10 were up-regulated in addition to the expression of a small number of M1 transcripts including inducible nitric oxide synthase. M1 MGL1/CD11c+ ATM exhibited a characteristic increase in pro-inflammatory genes and gene expression analysis revealed genes involved in matrix remodelling and angiogenesis that were up-regulated. Intriguingly 60–70% of CD11c+ cells co-expressed MGL1 (MGL1med/CD11c+) and this sub-population displayed an intermediate phenotype between MGL1+/CD11c and MGL1/CD11c+ ATM( Reference Shaul, Bennett and Strissel 59 ). These findings corroborate with Westcott et al., which unexpectedly showed that mice lacking in the traditional alternatively activated M2 MGL surface marker displayed significant reduction in CD11c+ ATM infiltration, attenuated inflammation and hepatic steatosis( Reference Westcott, Delproposto and Geletka 60 ). A further study also defined three ATM populations in obese adipose. In accordance with the ‘phenotypic switch’ model, DIO caused a shift from MR+/CD11c (M2) ATM population to MR/CD11c+ (M1) population. However, an increase in a third population was observed; the MR/CD11c double negative population, which simulated an M2-like population( Reference Zeyda, Gollinger and Kriehuber 61 ). Li and colleagues further highlight the complexity of M1/M2 prototype in vivo. In their study, HFD-induced IR mice were fed a normal chow diet and CD11c+cell functionality was assessed. CD11c+ ATM number remained consistent; however, inflammatory phenotype was attenuated( Reference Li, Lu and Nguyen 62 ).

Further adding to the complexity of M1/M2 classification extrapolating the significance of ATM in IR from rodent models to human subjects is confounded by differences in gene expression and cell surface receptors; for instance, human subjects exhibit reduced arginase and CD11c expression in ATM( Reference Schneemann and Schoeden 63 , Reference Zeyda and Stulnig 64 ). Thus, the contribution of ATM to HFD-induced IR in human subjects is less well understood. Human ATM recently demonstrated a mixed M1/M2 phenotype. As such, CD206+ cells increased in human subcutaneous adipose tissue and displayed a remodelling phenotype for instance expressing high levels of matrix metalloproteinase-9; paradoxically these macrophages also expressed elevated levels of pro-inflammatory genes TNF-α, IL-8( Reference Bourlier, Zakaroff-Girard and Miranville 65 ). Furthermore, weight loss significantly altered the expression of M1 and M2 surface markers, promoting a less inflammatory profile( Reference Aron-Wisnewsky, Tordjman and Poitou 66 ). Analysis of ATM in obese women demonstrated functionally distinct sub-populations. Coincident with obese rodent models CD11c expressing cells were predominantly located in a crown-like structure in adipose tissue. A subset of CD11c cells co-expressed M2 marker CD206 and exhibit features of M1 and M2 macrophages. Importantly, CD11c+CD206+/CD11c ratio significantly correlated with levels of IR. Ex vivo analyses demonstrated CD11c+CD206+ATM secreted significantly higher levels of pro-inflammatory IL-6, TNF-α and IL-8 relative to CD11cATM. Furthermore, CD11c+CD206+ATM-conditioned media impaired insulin-stimulated glucose uptake into adipocytes( Reference Wentworth, Naselli and Brown 67 ).

Fatty acids and adipose tissue macrophage polarisation

At present, there is a plethora of research attempting to decipher the molecular mechanisms controlling this ‘phenotypic switch’. One hypothesis is that lipids may regulate macrophage polarisation. The identification that the fatty acid sensor PPARγ displayed anti-inflammatory properties was the first unifying link between macrophage lipid metabolism and inflammation( Reference Bouhlel, Derudas and Rigamonti 68 , Reference Prieur, Roszer and Ricote 69 ). PPARγ is a ligand-activated transcription factor central to lipid homoeostasis. It is highly expressed in adipocytes and macrophages and is essential for adipocyte differentiation and lipid storage( Reference Rosen, Sarraf and Troy 70 , Reference Koutnikova, Cock and Watanabe 71 ). PPARγ activity is markedly enhanced by IL-4( Reference Huang, Welch and Ricote 72 ) and in DIO mice, myeloid cell-specific disruption of PPARγ impaired M2 activation predisposing mice to adipose tissue inflammation and systemic IR( Reference Odegaard, Ricardo-Gonzalez and Goforth 73 ). Furthermore, lack of PPARγ suppressed numerous genes involved in nutrient uptake, fatty acid synthesis and β-oxidation( Reference Odegaard, Ricardo-Gonzalez and Goforth 73 ). Short-term treatment with PPARγ agonist rosiglitazone enhanced M2 ATM number and reduced adipose tissue pro-inflammatory IL-18 gene expression in obese mice( Reference Stienstra, Duval and Muller 74 ). Congruent with this, human monocytes treated with PPARγ agonist GW1929 or rosiglitazone, were primed towards an M2 activation state expressing enhanced surface expression of M2 MR and reduced M1 surface marker CD163 in vitro. Intriguingly, in atherosclerotic lesions treatment with PPARγ agonists GW1929 or rosiglitazone did not induce M2 marker expression in resting or M1 macrophages implying PPARγ solely primes native monocytes to an M2 polarisation state( Reference Bouhlel, Derudas and Rigamonti 68 ). More recently, two distinct ATM subsets were characterised: CCR2+ F4/80Hi and F4/80low in db/db obese mice, mice deficient in leptin receptor activity, based on responsiveness to PPARγ agonist. In the absence of an agonist CCR2+ F4/80Hi ATM were abundantly expressed; however, in response to PPARγ agonist CCR2+ F4/80Hi ATM sub-population were reduced coincident with improved insulin sensitivity and inflammation( Reference Guri, Hontecillas and Ferrer 75 ). Bassaganya-Riera et al. ( Reference Bassaganya-Riera, Misyak and Guri 76 ) extended this work demonstrating F4/80Hi and F4/80low ATM subsets differentially express PPARγ and lack of PPARγ favours an M1 ATM polarisation state.

The adipose tissue ‘expandability hypothesis’ postulates that adipose tissue loses its ability to adequately store excess fat resulting in lipid spill over which promotes ectopic lipid accumulation and inflammation in macrophages( Reference Prieur, Roszer and Ricote 69 , Reference Prieur, Mok and Velagapudi 77 ). Transcriptional profile analysis of adipocytes and ATM from leptin-deficient (ob/ob) mice, demonstrated that increasing adiposity was associated with a reduction in lipogenic, lipid uptake and lipid storage gene expression in adipocytes but up-regulation in ATM. Correspondingly, a large portion of CD11c+ M1 macrophages were present within adipose tissue. Lipidomic analysis revealed ATM selectively accumulated SCFA which coincided with M2 to M1 phenotypic switch. Finally, rosiglitazone treatment improved adipocyte lipid storage capacity, improved insulin sensitivity coincident with increased M2 markers. Correspondingly, mice over-expressing PPARγ target gene diacylglycerol acyltransferase 1 in macrophages and adipocytes exhibited enhanced weight gain but were protected from typical obesity-induced macrophage infiltration and inflammatory activation coincident with attenuated IR. Furthermore in this context, NEFA failed to directly activate macrophages which over-expressed diacylglycerol acyltransferase 1( Reference Koliwad, Streeper and Monetti 78 ). Taken together, these data suggest that obesity reduces adipocyte storage capacity and metabolic capabilities, which induces lipid partitioning from adipocytes into macrophages favouring M1 ATM polarisation( Reference Prieur, Mok and Velagapudi 77 ).

Obesity-induced adipose tissue macrophage recruitment

A vital question that must be addressed regarding ATM is what triggers ATM infiltration during obesity. The precise mechanisms underpinning the initiation of ATM recruitment are unclear and are currently under intense investigation( Reference Zeyda and Stulnig 64 ). Adipocyte death and/or hypoxia are possibly both responsible for recruitment. Adipocyte necrosis increases with obesity due to adipocyte hypertrophy, and thus a plausible explanation for macrophage recruitment during obesity is for the removal of dead and dying cells( Reference Cinti, Mitchell and Barbatelli 79 ). Indeed, in obese mice and human subjects greater than 90% of ATM were shown to be specifically localised in a crown-like structure surrounding necrotic adipocytes( Reference Cinti, Mitchell and Barbatelli 79 ). These activated macrophages exhibit increased IL-6 and TNF-α expression and were shown to ingest residual lipid droplets forming large lipid-laden multinucleate giant cells, a characteristic feature of chronic inflammation( Reference Cinti, Mitchell and Barbatelli 79 ). Similarly, adipocyte apoptosis is markedly increased in obese mice and human subjects( Reference Alkhouri, Gornicka and Berk 80 ). Hypoxia is a pathogenic state whereby the surrounding tissue is devoid of adequate oxygen supply resulting in decreased oxygen tension. Local hypoxia is observed in obese adipose tissue in mice and human subjects( Reference Hosogai, Fukuhara and Oshima 81 , Reference Kabon, Nagele and Reddy 82 ). In DIO mice, adipose tissue hypoxia is correlated with enhanced pro-inflammatory gene expression and reduced adiponectin expression. Conversely, weight loss improved adipose oxygenation and increased adiponectin expression( Reference Ye, Gao and Yin 83 ). Moreover, hypoxic conditions up-regulate additional adipokines and adipose-tissue-derived cytokine expression, including leptin and IL-6( Reference Sun and Scherer 84 ). Hypoxia-inducible factor-1α is a critical regulator in cellular hypoxic response, which is rapidly degraded under normoxic conditions and is increased early in obesity. Over-expression of hypoxia-inducible factor-1α in a transgenic mouse model initiated adipose tissue fibrosis, with an accompanying increase in local inflammation( Reference Halberg, Khan and Trujillo 85 ). In human subjects, phosphorylated levels of p38 are significantly up-regulated in visceral adipose tissue. Disrupting p38 activity reduced hypoxia-induced stromal vascular fraction inflammatory responses, implicating a role for p38 activity in the regulation of hypoxia( Reference O'Rourke, White and Metcalf 86 ). These studies strongly suggest that adipocyte cell death and hypoxia-induced inflammation may be potential underlying causes of ATM infiltration during obesity.

Chemokines have emerged as crucial mediators in obesity-associated ATM recruitment into adipose tissue. They are small pro-inflammatory cytokines which instigate monocyte recruitment from the bone marrow to the site of inflammation( Reference Charo and Ransohoff 87 , Reference Gerard and Rollins 88 ). More than 50 cytokines/chemokines have been described and categorised into four distinct groups in accordance with the location of the conserved cysteine residue; CCL, CXCL, CL and CX3CL( Reference Zlotnik, Yoshie and Nomiyama 89 , Reference Chavey and Fajas 90 ). The pathophysological role of CCL2/MCP-1 and its receptor CCR2 has been most thoroughly investigated. CCL2 is ubiquitously expressed and found in monocytes and adipocytes and thus has been implicated in several biological processes and disease states( Reference Rull, Camps and Alonso-Villaverde 91 ). Circulating concentrations and adipose tissue expression levels of CCL2 positively correlate to adiposity( Reference Xu, Barnes and Yang 9 ). In addition, 3T3-L1 adipocytes treated with CCL2 exhibited a marked reduction in insulin-simulated glucose uptake in vitro ( Reference Sartipy and Loskutoff 92 , Reference Chen, Mumick and Zhang 93 ). Absence of CCR2 in DIO mouse model partially ameliorated HFD-induced obesity, hepatic steatosis coincident with attenuated systemic IR. Immunohistochemistry analysis demonstrated reduced ATM recruitment, concomitant with reduced adipose tissue TNF-α expression. Further expression analysis revealed that CCR2 directly altered genes involved in lipid metabolism and adipocyte differentiation, with CCR2−/− mice exhibiting increased expression of fatty acid binding protein-4 and PPARγ. Correspondingly, short-term treatment with a CCR2 antagonist decreased macrophage infiltration and enhanced systemic insulin sensitivity in DIO mice( Reference Weisberg, Hunter and Huber 51 ). In support of these findings, a subsequent study demonstrated lack of CCL2-attenuated adipose tissue inflammation and IR, while over-expression of CCL2 in adipose tissue caused increased ATM number coincident with exaggerated systemic IR( Reference Kanda, Tateya and Tamori 94 , Reference Kamei, Tobe and Suzuki 95 ). However, CCR2 and CCL2 knockout mice models retain significant number of ATM( Reference Weisberg, Hunter and Huber 51 , Reference Kanda, Tateya and Tamori 94 ). The role of CCL2 in obesity maybe further confounded by the effect of CCL2 on metabolism. Lack of CCR2 reduced HFD-induced obesity, while plasma CCL2 levels decreased exponentially with pharmacological-induced weight reduction in DIO mice( Reference Weisberg, Hunter and Huber 51 , Reference Chen, Mumick and Zhang 93 ). A number of studies have disputed the involvement of the CCR2/MCP-1 axis in obesity and IR( Reference Chen, Mumick and Zhang 93 , Reference Inouye, Shi and Howard 96 ). Abrogating CCR2 in DIO mice did not alter body weight, glucose homoeostasis or ATM infiltration( Reference Chen, Mumick and Zhang 93 ). Coincidently, Inouye and co-workers showed that CCL2-deficient mice exhibited increased HFD-induced weight gain and comparable macrophage recruitment capabilities to control mice. CCL2-deficient mice also displayed impaired glucose tolerance, and in fact presented increased plasma glucose and reduced adiponectin levels compared with control mice( Reference Inouye, Shi and Howard 96 ). These studies suggest that infiltrating macrophages are not solely pro-inflammatory and indicate that factors independent of CCR2/CCL2 are involved in macrophage recruitment.

Emerging data have proposed a pathogenic role for alternative chemokines in obesity. CX3CL1, a human chemokine is expressed in adipocytes and increased with obesity( Reference Digby, McNeill and Dyar 97 , Reference Shah, Hinkle and Ferguson 98 ). Furthermore, genetic variations within CX3CR1 gene correlate to increased weight circumference, IR and reduced adiponectin( Reference Shah, Hinkle and Ferguson 98 ). A causal role for CX3CL1 in adipose tissue inflammation has been displayed whereby CX3CL1 induced monocyte adhesion to human adipocytes ex vivo ( Reference Shah, Hinkle and Ferguson 98 ). A role for keratinocyte-derived chemokine, a mouse orthologue for human IL-8, in obesity-induced ATM recruitment has been proposed. Keratinocyte-derived chemokine and its receptor CXCR2 have been implicated in co-ordinating macrophage infiltration into atherosclerotic lesions( Reference Boisvert, Santiago and Curtiss 99 , Reference Boisvert, Rose and Johnson 100 ). Circulating concentrations and adipose tissue expression levels are heightened during obesity. Furthermore, keratinocyte-derived chemokine induced an inflammatory response in 3T3-L1 adipocytes in vitro, up-regulating IL-6, TNF-α and MCP-1 expression. Another CXCR2 ligand is CXCL5. CXCL5 is abundantly expressed in adipose tissue, predominantly within the stromal vascular fraction. Circulating CXCL5 levels increased with obesity concomitant with enhanced secretion from obese adipose tissue explants compared with lean ex vivo. Furthermore, exogenous CXCL5 treatment reduced muscle insulin sensitivity via activation of suppressor of cytokine signalling pathway( Reference Chavey and Fajas 90 ). Confirming the importance of both keratinocyte-derived chemokine and CXCL5 in obesity-induced inflammation and IR mice deficient in CXCR2 exhibit reduced ATM recruitment and attenuated IR in HFD-induced obese mice models( Reference Chavey and Fajas 90 , Reference Straczkowski, Dzienis-Straczkowska and Stepien 101 , Reference Neels, Badeanlou and Hester 102 ). Another potential chemokine receptor that could mediate ATM recruitment and alter IR is CCR5( Reference Kitade, Sawamoto and Nagashimada 103 ). CCR5 and its ligands are up-regulated in both obese human and rodent models( Reference Kitade, Sawamoto and Nagashimada 103 , Reference Huber, Kiefer and Zeyda 104 ). DIO mice lacking CCR5 are protected from the development of hepatic steatosis and exhibit improved insulin sensitivity. Furthermore, deletion of CCR5 alters both ATM number and the M1/M2 polarisation state with a corresponding reduction in adipose tissue inflammation and improved insulin sensitivity( Reference Kitade, Sawamoto and Nagashimada 103 ). These studies illustrate that chemokines are central to ATM recruitment; however, it is yet to be fully established what enhances their secretion.

Macrophage migration inhibitory factor

MIF is a multifunctional pro-inflammatory cytokine whose actions are responsible for an array of inflammatory processes. Macrophages have been identified as a primary source and target of MIF in vitro and in vivo ( Reference Calandra, Bernhagen and Mitchell 105 ). Uniquely, MIF is rapidly released from preformed intracellular pools in response to inflammatory stimuli: Lipopolysaccharide, TNF-α and interferon-γ and can elicit its effects in a paracrine and autocrine manner( Reference Calandra, Bernhagen and Mitchell 105 Reference Calandra and Roger 107 ). In this regard, MIF can inhibit immunosuppressive glucocorticoids, promote secretion of a variety of cytokines including TNF-α, IL-2, IL-6, IL-8, interferon-γγ, IL-1β and inhibit IL-10 propagating a pro-inflammatory response( Reference Calandra, Bernhagen and Mitchell 105 Reference Lue, Thiele and Franz 108 ). Endogenous MIF regulates innate immunity via up-regulation of TLR-4, IL-1R1 and TNFR expression( Reference Roger, David and Glauser 109 , Reference Toh, Aeberli and Lacey 110 ). Roger and co-workers demonstrated that MIF−/− macrophages were hyporesponsive to lipopolysaccharide and failed to secrete TNF-α and IL-6 due to profound suppression of inhibitor of NF-κB kinase-β/NF-кB activity( Reference Roger, David and Glauser 109 ). In addition, MIF deficiency suppressed IL-1 and TNF-α-induced mitogen-activated protein kinases activity and cell proliferation in mouse fibroblasts. Reconstitution of an upstream mitogen-activated protein kinase or MIF reversed these affects( Reference Toh, Aeberli and Lacey 110 ). Furthermore, MIF impairs p53-dependent apoptosis sustaining activated macrophage lifespan, thus further amplifying the inflammatory response( Reference Mitchell, Liao and Chesney 111 ). Another mechanism identified by which MIF promotes inflammation is through amplification of transmigration, recruitment and activation of leukocytes at the site of inflammation through up-regulation of adhesion molecules such as intracellular adhesion molecule-1 and chemokine MCP-1( Reference Gregory, Morand and McKeown 112 Reference Bernhagen, Krohn and Lue 114 ). MIF can exert its chemotactic properties via CXCR2 and CXCR4 in macrophages and T-cells, respectively( Reference Bernhagen, Krohn and Lue 114 ). Binding of MIF to CXCR4 on the surface of fibroblasts and T-cells induced JNK propagating CXCL8 secretion( Reference Lue, Dewor and Leng 115 ). Intriguingly, an alternative MIF receptor CD74, traditionally involved in the activation of mitogen-activated protein kinases pathway, has recently been demonstrated to also mediate macrophage chemotactic responses( Reference Fan, Hall and Santos 116 ). Given its diverse functionality and prominent role in macrophage biology MIF has been implicated in a multitude of inflammatory diseases( Reference Calandra and Roger 107 ). More recently, a pathogenic role has been ascribed to MIF in obesity-associated inflammation and related metabolic complications (Fig. 2).

Fig. 2. Role of macrophage migration inhibitory factor (MIF) in inflammation and adipose tissue dysfunction. 1. MIF activates macrophages inducing inflammatory cytokine secretion; TNF-α and IL-12. 2. Inflammatory stimuli lipopolysaccaride (LPS), TNF-α and interferon-γ can induce MIF secretion from macrophages. 3. Endogenous MIF can regulate toll-like receptor-4 (TLR-4), IL-1 receptor (IL-1R)1, TNF receptor (TNFR) expression 4. MIF inhibits macrophage-derived anti-inflammatory IL-10 secretion. 5. MIF is involved in monocyte recruitment via transmigration and recruitment of monocytes by up-regulating intracellular adhesion molecule-1 (ICAM) and monocyte chemoattractant protein-1 (MCP)-1 and can also by binding to CXC-chemokine receptor (CXCR)2. 6. MIF can promote TNF-α secretion from adipocytes 0·7. MIF can impair insulin-stimulated glucose uptake into adipose tissue and down-regulate phosphorylated AKT expression.

The role of macrophage migration inhibitory factor in obesity and type-2 diabetes mellitus

Epidemiological studies implicate MIF in obesity and T2DM( Reference Ghanim, Aljada and Hofmeyer 117 Reference Vozarova, Stefan and Hanson 120 ). Obese individuals with a BMI of 37·5 (SD 4·9) kg/m2 have significantly elevated plasma MIF concentrations (2·8 (SD 2·0) ng/ml) compared with lean individuals with a BMI of 22·6 (SD 3·4) kg/m2 (1·2 (SD 0·6) ng/ml). During obesity MIF mRNA expression is up-regulated by 60% in circulating peripheral blood mononuclear cells( Reference Ghanim, Aljada and Hofmeyer 117 , Reference Dandona, Aljada and Ghanim 118 ). Furthermore, a positive association between elevated plasma MIF concentrations and peripheral blood mononuclear cells MIF mRNA with BMI, NEFA concentration and impaired glucose tolerance has been shown( Reference Vozarova, Stefan and Hanson 120 , Reference Skurk, Herder and Kraft 121 ). A polymorphism within the MIF gene promoter (MIF_794[CATT]5–8) was associated with obesity in a Japanese population( Reference Sakaue, Ishimaru and Hizawa 122 ). Treatment with the anti-diabetic drug metformin decreased plasma MIF concentration from 2·3 (SD 1·4) to 1·6 (SD 1·2) ng/ml in obese individuals following a 6-week intervention. Cessation of drug treatment reversed these effects( Reference Dandona, Aljada and Ghanim 118 ). Furthermore, a weight loss programme in morbidly obese individuals that achieved a 14·4 kg reduction in body weight was associated with lower serum MIF concentrations and improved pancreatic β-cell function( Reference Church, Willis and Priest 119 ).

It has been demonstrated that patients with T2DM have higher serum MIF (20·7 (13·3) ng/ml) levels compared with non-diabetic controls (5·2 (SD 3·0) ng/ml)( Reference Yabunaka, Nishihira and Mizue 123 ). The Pima Indians display increased susceptibility to T2DM. Healthy lean Pima Indians exhibit elevated serum MIF concentrations relative to healthy lean Caucasians. This elevated MIF concentration was directly associated with impaired insulin action; supporting a link between MIF and increased susceptibility to IR and T2DM( Reference Vozarova, Stefan and Hanson 120 ). A subsequent case–control study based on the Cooperative Health Research in the region of Augsburg Survey 4 (KORA4: 1999–2000) serum MIF concentration increased exponential from normoglycemia to impaired glucose tolerance and overt T2DM. This study indicated that MIF was a more proficient discriminatory factor between healthy individuals and patients with impaired glucose tolerance or T2DM compared with IL-6 or C-reactive protein 2( Reference Herder, Kolb and Koenig 124 ). Results from the population-based study monitoring of trends and determinants in cardiovascular disease/KORA presented evidence of a triangular relationship between MIF levels, SNP within the MIF gene and the incidence of T2DM in women. The C allele of SNP rs1007888 was associated with increased circulating MIF, while rs1007888CC was correlated to T2DM in female subjects. Moreover, elevated MIF concentrations correlated to the incidence of T2DM to a greater extent in obese women compared with lean( Reference Herder, Klopp and Baumert 125 ). More recently, rs1007888 also showed association with enhanced susceptibility to gestational diabetes mellitus and postpartum metabolic syndrome( Reference Aslani, Hossein-nezhad and Maghbooli 126 ).

The role of macrophage migration inhibitory factor in glucose metabolism and adipose tissue dysfunction

Given that epidemiological data suggest an association between MIF, obesity and glucose homoeostasis, molecular studies have focused on whether MIF is a causal factor in relation to dysregulated glucose metabolism and adipose tissue dysfunction. Waeber et al. ( Reference Waeber, Calandra and Roduit 127 ), first implicated a role for MIF within glucose metabolism when they highlighted the presence of MIF within the secretory granules in rodent pancreatic β-cells. The production of MIF from these cells was demonstrated to be glucose dependent and on release regulate insulin secretion in an autocrine manner. Immunoneutralisation of MIF-impaired glucose-induced insulin secretion during exposure to exogenous recombinant MIF potentiated insulin release( Reference Waeber, Calandra and Roduit 127 ). More recently, MIF inhibition provided significant protection from palmitic-acid induced β-cells apoptosis( Reference Saksida, Stosic-Grujicic and Timotijevic 128 ). Together, these studies suggested a potential pathogenic role for MIF in fatty acids induced β-cell dysfunction and hyperinsulinemia.

MIF has been shown to be secreted from murine and human adipocytes( Reference Koska, Stefan and Dubois 12 , Reference Skurk, Herder and Kraft 121 , Reference Hirokawa, Sakaue and Tagami 129 , Reference Hirokawa, Sakaue and Furuya 130 ). Moreover, glucose and insulin reportedly regulate adipocyte MIF expression in vitro ( Reference Sakaue, Nishihira and Hirokawa 131 ). Treatment of 3T3-L1 adipocytes with TNF-α induced MIF secretion, suggesting that TNF-α may regulate MIF production during obesity( Reference Hirokawa, Sakaue and Tagami 129 , Reference Hirokawa, Sakaue and Furuya 130 ). In vivo MIF-deficient mice were hyporesponsive to TNF-α administration and exhibited normoglycemia compared with control mice, indicating MIF is required for successful TNF-α action. Furthermore, 3T3-L1 adipocytes exposed to exogenous MIF demonstrated impaired insulin-stimulated glucose uptake and insulin receptor signal transduction. Similarly, in response to inflammatory stress, MIF−/− mice exhibit a marked improvement in adipose tissue glucose uptake compared with control mice( Reference Atsumi, Cho and Leng 132 ). This study suggested that MIF affects glucose homoeostasis both directly and indirectly via TNF-α regulation. In obese patients subcutaneous adipose tissue explants were shown to release significant quantities of MIF over 24 h (10 000 pg/ml in 24 h) and these concentrations presented a direct correlation to individual donor BMI( Reference Skurk, Herder and Kraft 121 ). More recently, Koska et al. demonstrated that MIF mRNA expression in subcutaneous adipose tissue increased with adipocyte size and was negatively correlated to peripheral and hepatic IR( Reference Koska, Stefan and Dubois 12 ). Correspondingly, intracellular levels of MIF were found to be heightened during adipogenesis in vitro. Conversely, genetic deletion of MIF in rodent models and siRNA knockdown of MIF in 3T3-L1 adipocytes reduced intracellular lipid accumulation and impaired adipogeneic gene PPARγ expression( Reference Atsumi, Cho and Leng 132 , Reference Ikeda, Sakaue and Kamigaki 133 ). Verschuren and co-workers have shown that lack of MIF in an atherosclerotic mouse model (LDLR−/−MIF−/−) maintained on a standard chow diet exhibit attenuated adipose tissue inflammation and enhanced adipose tissue insulin sensitivity coincident with improved glucose homoeostasis and systemic inflammation( Reference Verschuren, Kooistra and Bernhagen 134 ). In response to an acute inflammatory challenge, IL-1β administration, LDLR−/−MIF−/− mice exhibited reduced circulating serum amyloid A, a marker of systemic inflammation. Subsequent histological analysis revealed that lack of MIF reduced adipocyte size and MAC3+ macrophage population. Gene expression analysis indicated that the ratio of M1/M2 was reduced in LDLR−/−MIF−/− mice. Furthermore, thioglycollate-elicited macrophages isolated from MIF−/− mice exhibited reduced IL-6 secretion when stimulated with lipopolysaccharide compared with control mice. Consequently, PI3-kinase activity, a functional marker of insulin signalling, was greater in LDLR−/−MIF−/− adipose tissue. In addition, deletion of MIF reduced monocyte adhesion, macrophage lesion content and atherosclerotic lesion size( Reference Verschuren, Kooistra and Bernhagen 134 ). On the contrary, mature MIF−/− mice maintained on a standard chow diet for 12 months demonstrated impaired glucose tolerance( Reference Serre-Beinier, Toso and Morel 11 ). These contradictory findings underscore the complexity of MIF inflammatory signals within glucose metabolism depending on age.

Conclusion

Extensive evidence implicates inflammation as a critical factor in the progression of obesity-related metabolic abnormalities. ATM accumulation significantly contributes to adipose tissue inflammation resulting in impaired insulin action, thus contributing to systemic IR and the development of T2DM. However, the triggers responsible for ATM recruitment and activation are not fully understood. Adipose tissue-derived chemokines have emerged as principal mediators in propagating inflammation and IR. Given MIF chemotactic properties, MIF may be a primary candidate promoting ATM recruitment during obesity. Pro-inflammatory MIF activity is enhanced during obesity and is directly associated with the degree of peripheral IR. Current anti-inflammatory therapeutics exhibit a broad range of actions which consequently may induce an immunocompromised phenotype( Reference Harford, Reynolds and McGillicuddy 49 ). Targeting the functionality of specific tissues, for example targeting ATM inflammatory responses, may prove to be more beneficial. Indeed, several population studies have identified a number of SNP within the MIF gene promoter region associated with obesity and T2DM. Thus, manipulating MIF inflammatory activities in obesity, using pharmacological agents or functional foods, may be therapeutically beneficial for the treatment and prevention of obesity-related metabolic diseases.

Acknowledgements

H.M.R. was supported by Science Foundation Ireland PI Programme (06/IM.1/B105). The authors declare no conflict of interest. O.M.F. completed the review. H.M.R. advised in relation to review content. H.M.R., C.M.R. and F.C.M. critically evaluated the manuscript. All authors approved the final review.

References

1. Flegal, KM, Graubard, BI, Williamson, DF et al. (2007) Cause-specific excess deaths associated with underweight, overweight, and obesity. JAMA 298, 20282037.Google Scholar
2. The World Health Organisation (2011) Obesity and overweight. Available at: http://www.who.int./mediacentre/factsheet/fs311/en/.Google Scholar
3. Finucane, MM, Stevens, GA, Cowan, MJ et al. (2011) National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9·1 million participants. Lancet 377, 557567.Google Scholar
4. Swinburn, BA, Sacks, G, Hall, KD et al. (2011) The global obesity pandemic: shaped by global drivers and local environments. Lancet 378, 804814.Google Scholar
5. Berrington de Gonzalez, A (2010) Body-Mass index and mortality among 1·46 million white adults. N Engl J Med 365, 22112219.Google Scholar
6. Zimmet, P, Alberti, KG & Shaw, J (2001) Global and societal implications of the diabetes epidemic. Nature 414, 782787.Google Scholar
7. Hotamisligil, GS, Shargill, NS & Spiegelman, BM (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259, 8791.Google Scholar
8. Weisberg, SP, McCann, D, Desai, M et al. (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112, 17961808.Google Scholar
9. Xu, H, Barnes, GT, Yang, Q et al. (2003) Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112, 18211830.Google Scholar
10. Morand, EF, Leech, M & Bernhagen, J (2006) MIF: a new cytokine link between rheumatoid arthritis and atherosclerosis. Nat Rev Drug Discov 5, 399410.Google Scholar
11. Serre-Beinier, V, Toso, C, Morel, P et al. (2011) Macrophage migration inhibitory factor deficiency leads to age-dependent impairment of glucose homeostasis in mice. J Endocrinol 206, 297306.Google Scholar
12. Koska, J, Stefan, N, Dubois, S et al. (2009) mRNA concentrations of MIF in subcutaneous abdominal adipose cells are associated with adipocyte size and insulin action. Int J Obes (Lond) 33, 842850.Google Scholar
13. Kahn, BB, Flier, JS (2000) Obesity and insulin resistance. J Clin Invest 106, 473481.Google Scholar
14. de Luca, C & Olefsky, JM (2008) Inflammation and insulin resistance. FEBS Lett 582, 97105.Google Scholar
15. Taniguchi, CM, Kondo, T, Sajan, M et al. (2006) Divergent regulation of hepatic glucose and lipid metabolism by phosphoinositide 3-kinase via Akt and PKClambda/zeta. Cell Metab 3, 343353.Google Scholar
16. Abel, ED, Peroni, O, Kim, JK et al. (2001) Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409, 729733.Google Scholar
17. Schenk, S, Saberi, M & Olefsky, JM (2008) Insulin sensitivity: modulation by nutrients and inflammation. J Clin Invest 118, 29923002.Google Scholar
18. Duncan, RE, Ahmadian, M, Jaworski, K et al. (2007) Regulation of lipolysis in adipocytes. Annu Rev Nutr 27, 79101.Google Scholar
19. Shulman, GI (2000) Cellular mechanisms of insulin resistance. J Clin Invest 106, 171176.Google Scholar
20. Schwarz, JM, Linfoot, P, Dare, D et al. (2003) Hepatic de novo lipogenesis in normoinsulinemic and hyperinsulinemic subjects consuming high-fat, low-carbohydrate and low-fat, high-carbohydrate isoenergetic diets. Am J Clin Nutr 77, 4350.Google Scholar
21. Brown, MS & Goldstein, JL (2008) Selective versus total insulin resistance: a pathogenic paradox. Cell Metab 7, 9596.Google Scholar
22. Wellen, KE & Hotamisligil, GS (2005) Inflammation, stress, and diabetes. J Clin Invest 115, 11111119.Google Scholar
23. Schmidt, MI, Duncan, BB, Sharrett, AR et al. (1999) Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. Lancet 353, 16491652.Google Scholar
24. Pradhan, AD, Manson, JE, Rifai, N et al. (2001) C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 286, 327334.Google Scholar
25. Fantuzzi, G (2005) Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol 115, 911919.Google Scholar
26. Suganami, T & Ogawa, Y (2010) Adipose tissue macrophages: their role in adipose tissue remodeling. J Leukoc Biol 88, 3339.Google Scholar
27. Zhang, Y, Proenca, R, Maffei, M et al. (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425432.Google Scholar
28. Maeda, K, Okubo, K, Shimomura, I et al. (1996) cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (AdiPose Most abundant Gene transcript 1). Biochem Biophys Res Commun 221, 286289.Google Scholar
29. Thompson, BR, Lobo, S & Bernlohr, DA (2010) Fatty acid flux in adipocytes: the in's and out's of fat cell lipid trafficking. Mol Cell Endocrinol 318, 2433.Google Scholar
30. Suganami, TNJ & Ogawa, Y (2005) A paracrine loop between adipocytes and macrophages aggravates inflammatory changes; role of free fatty acids and tumor necrosis factor alpha. Arterioscler Thromb Vasc Biol 25, 20622068.Google Scholar
31. Hotamisligil, GS, Murray, DL, Choy, LN et al. (1994) Tumor necrosis factor alpha inhibits signaling from the insulin receptor. Proc Natl Acad Sci USA 91, 48544858.Google Scholar
32. Uysal, KT, Wiesbrock, SM, Marino, MW et al. (1997) Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature 389, 610614.Google Scholar
33. Hotamisligil, GS, Arner, P, Caro, JF et al. (1995) Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 95, 24092415.Google Scholar
34. Bruun, JM, Helge, JW, Richelsen, B et al. (2006) Diet and exercise reduce low-grade inflammation and macrophage infiltration in adipose tissue but not in skeletal muscle in severely obese subjects. Am J Physiol Endocrinol Metab 290, E961E967.Google Scholar
35. Cancello, R, Henegar, C, Viguerie, N et al. (2005) Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes 54, 22772286.Google Scholar
36. Permana, PA, Menge, C & Reaven, PD (2006) Macrophage-secreted factors induce adipocyte inflammation and insulin resistance. Biochem Biophys Res Commun 341, 507514.Google Scholar
37. Antuna-Puente, B, Feve, B, Fellahi, S et al. (2008) Adipokines: the missing link between insulin resistance and obesity. Diabetes Metab 34, 211.Google Scholar
38. Lumeng, C, Deyoung, SM & Saltiel, AR (2006) Macrophages block insulin action in adipocytes by altering expression of signaling and glucose transport proteins. Am J Physiol Endocrinol Metab 292, 166174.10.1152/ajpendo.00284.2006Google Scholar
39. Solinas, G & Karin, M (2010) JNK1 and IKKbeta: molecular links between obesity and metabolic dysfunction. FASEB J 24, 25962611.Google Scholar
40. Hirosumi, J, Tuncman, G, Chang, L et al. (2002) A central role for JNK in obesity and insulin resistance. Nature 420, 333336.Google Scholar
41. Tuncman, G, Hirosumi, J, Solinas, G et al. (2006) Functional in vivo interactions between JNK1 and JNK2 isoforms in obesity and insulin resistance. Proc Natl Acad Sci USA 103, 1074110746.Google Scholar
42. Sabio, G, Das, M, Mora, A et al. (2007) A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science 322, 15391543.Google Scholar
43. Solinas, G, Vilcu, C, Neels, JG et al. (2007) JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab 6, 386397.Google Scholar
44. Arkan MC, HA & Greten, FR (2005) IKK-beta links inflammation to obesity induced insulin resistance. Nat Med 11, 191198.Google Scholar
45. Medzhitov, R (2001) Toll-like receptors and innate immunity. Nat Rev Immunol 1, 135145.Google Scholar
46. Shi, H, Kokoeva, MV, Inouye, K et al. (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116, 30153025.Google Scholar
47. Tsukumo, DM, Carvalho-Filho, MA, Carvalheira, JB et al. (2007) Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 56, 19861998.Google Scholar
48. Saberi, M, Woods, NB, de Luca, C et al. (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.Google Scholar
49. Harford, KA, Reynolds, CM, McGillicuddy, FC et al. (2011) Fats, inflammation and insulin resistance: insights to the role of macrophage and T-cell accumulation in adipose tissue. Proc Nutr Soc 70, 408417.Google Scholar
50. Kono, H & Rock, KL (2008) How dying cells alert the immune system to danger. Nat Rev Immunol 8, 279289.Google Scholar
51. Weisberg, SP, Hunter, D, Huber, R et al. (2006) CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest 116, 115124.10.1172/JCI24335Google Scholar
52. Osborn, O & Olefsky, JM (2012) The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med 18, 363374.10.1038/nm.2627Google Scholar
53. Mosser, DM & Edwards, JP (2008) Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8, 958969.Google Scholar
54. Edwards, JP, Zhang, X, Frauwirth, KA et al. (2006) Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol 80, 12981307.Google Scholar
55. Lumeng, CN, Bodzin, JL & Saltiel, AR (2007) Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117, 175184.Google Scholar
56. Lumeng, CN, Deyoung, SM, Bodzin, JL et al. (2007) Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes 56, 1623.Google Scholar
57. Nguyen, MT, Favelyukis, S, Nguyen, AK et al. (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.Google Scholar
58. Patsouris, D, Li, PP, Thapar, D et al. (2008) Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab 8, 301309.Google Scholar
59. Shaul, ME, Bennett, G, Strissel, KJ et al. (2010) Dynamic, M2-like remodeling phenotypes of CD11c+ adipose tissue macrophages during high-fat diet–induced obesity in mice. Diabetes 59, 11711181.Google Scholar
60. Westcott, DJ, Delproposto, JB, Geletka, LM et al. (2009) MGL1 promotes adipose tissue inflammation and insulin resistance by regulating 7/4hi monocytes in obesity. J Exp Med 206, 31433156.Google Scholar
61. Zeyda, M, Gollinger, K, Kriehuber, E et al. (2010) Newly identified adipose tissue macrophage populations in obesity with distinct chemokine and chemokine receptor expression. Int J Obes (Lond) 34, 16841694.Google Scholar
62. Li, P, Lu, M, Nguyen, MT et al. (2010) Functional heterogeneity of CD11c-positive adipose tissue macrophages in diet-induced obese mice. J Biol Chem 285, 1533315345.Google Scholar
63. Schneemann, M & Schoeden, G (2007) Macrophage biology and immunology: man is not a mouse. J Leukoc Biol 81, 579 -discussion 80.Google Scholar
64. Zeyda, M & Stulnig, TM (2007) Adipose tissue macrophages. Immunol Lett 112, 6167.Google Scholar
65. Bourlier, V, Zakaroff-Girard, A, Miranville, A et al. (2008) Remodeling phenotype of human subcutaneous adipose tissue macrophages. Circulation 117, 806815.Google Scholar
66. Aron-Wisnewsky, J, Tordjman, J, Poitou, C et al. (2009) Human adipose tissue macrophages: m1 and m2 cell surface markers in subcutaneous and omental depots and after weight loss. J Clin Endocrinol Metab 94, 46194623.Google Scholar
67. Wentworth, JM, Naselli, G, Brown, WA et al. (2010) Pro-inflammatory CD11c + CD206+ adipose tissue macrophages are associated with insulin resistance in human obesity. Diabetes 59, 16481656.Google Scholar
68. Bouhlel, MA, Derudas, B, Rigamonti, E et al. (2007) PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab 6, 137143.Google Scholar
69. Prieur, X, Roszer, T & Ricote, M (2010) Lipotoxicity in macrophages: evidence from diseases associated with the metabolic syndrome. Biochim Biophys Acta 1801, 327337.Google Scholar
70. Rosen, ED, Sarraf, P, Troy, AE et al. (1999) PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4, 611617.Google Scholar
71. Koutnikova, H, Cock, TA, Watanabe, M et al. (2003) Compensation by the muscle limits the metabolic consequences of lipodystrophy in PPAR gamma hypomorphic mice. Proc Natl Acad Sci USA 100, 1445714462.Google Scholar
72. Huang, JT, Welch, JS, Ricote, M et al. (1999) Interleukin-4-dependent production of PPAR-gamma ligands in macrophages by 12/15-lipoxygenase. Nature 400, 378382.Google Scholar
73. Odegaard, JI, Ricardo-Gonzalez, RR, Goforth, MH et al. (2007) Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature 447, 11161120.Google Scholar
74. Stienstra, R, Duval, C, Muller, M et al. (2007) PPARs, obesity, and inflammation. PPAR Res 2007, 95974.Google Scholar
75. Guri, AJ, Hontecillas, R, Ferrer, G et al. (2008) Loss of PPAR gamma in immune cells impairs the ability of abscisic acid to improve insulin sensitivity by suppressing monocyte chemoattractant protein-1 expression and macrophage infiltration into white adipose tissue. J Nutr Biochem 19, 216228.Google Scholar
76. Bassaganya-Riera, J, Misyak, S, Guri, AJ et al. (2009) PPAR gamma is highly expressed in F4/80(hi) adipose tissue macrophages and dampens adipose-tissue inflammation. Cell Immunol 258, 138146.Google Scholar
77. Prieur, X, Mok, CY, Velagapudi, VR et al. (2011) Differential lipid partitioning between adipocytes and tissue macrophages modulates macrophage lipotoxicity and M2/M1 polarization in obese mice. Diabetes 60, 797809.Google Scholar
78. Koliwad, SK, Streeper, RS, Monetti, M et al. (2010) DGAT1-dependent triacylglycerol storage by macrophages protects mice from diet-induced insulin resistance and inflammation. J Clin Invest 120, 756767.Google Scholar
79. Cinti, S, Mitchell, G, Barbatelli, G et al. (2005) Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46, 23472355.Google Scholar
80. Alkhouri, N, Gornicka, A, Berk, MP et al. (2010) Adipocyte apoptosis, a link between obesity, insulin resistance, and hepatic steatosis. J Biol Chem 285, 34283438.Google Scholar
81. Hosogai, N, Fukuhara, A, Oshima, K et al. (2007) Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56, 901911.Google Scholar
82. Kabon, B, Nagele, A, Reddy, D et al. (2004) Obesity decreases perioperative tissue oxygenation. Anesthesiology 100, 274280.Google Scholar
83. Ye, J, Gao, Z, Yin, J et al. (2007) Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 293, E1118E1128.Google Scholar
84. Sun, KKC & Scherer, PE (2011) Adipose tissue remodelling and obesity. J Clin Invest 121, 20942100.Google Scholar
85. Halberg, N, Khan, T, Trujillo, ME et al. (2009) Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 29, 44674483.Google Scholar
86. O'Rourke, RW, White, AE, Metcalf, MD et al. (2011) Hypoxia-induced inflammatory cytokine secretion in human adipose tissue stromovascular cells. Diabetologia 54, 14801490.Google Scholar
87. Charo, IF & Ransohoff, RM (2006) The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med 354, 610621.Google Scholar
88. Gerard, C & Rollins, BJ (2001) Chemokines and disease. Nat Immunol 2, 108115.Google Scholar
89. Zlotnik, A, Yoshie, O & Nomiyama, H (2006) The chemokine and chemokine receptor superfamilies and their molecular evolution. Genome Biol 7, 243.Google Scholar
90. Chavey, C & Fajas, L (2009) CXCL5 drives obesity to diabetes, and further. Aging (Albany NY) 1, 674677.Google Scholar
91. Rull, A, Camps, J, Alonso-Villaverde, C et al. (2010) Insulin resistance, inflammation, and obesity: role of monocyte chemoattractant protein-1 (or CCL2) in the regulation of metabolism. Mediators Inflamm 10, 11551166.Google Scholar
92. Sartipy, P & Loskutoff, DJ (2003) Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci USA 100, 72657270.Google Scholar
93. Chen, A, Mumick, S, Zhang, C et al. (2005) Diet induction of monocyte chemoattractant protein-1 and its impact on obesity. Obes Res 13, 13111320.10.1038/oby.2005.159Google Scholar
94. Kanda, H, Tateya, S, Tamori, Y et al. (2006) MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest 116, 14941505.Google Scholar
95. Kamei, N, Tobe, K, Suzuki, R et al. (2006) Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J Biol Chem 281, 2660226614.Google Scholar
96. Inouye, KE, Shi, H, Howard, JK et al. (2007) Absence of CC chemokine ligand 2 does not limit obesity-associated infiltration of macrophages into adipose tissue. Diabetes 56, 22422250.Google Scholar
97. Digby, JE, McNeill, E, Dyar, OJ et al. (2010) Anti-inflammatory effects of nicotinic acid in adipocytes demonstrated by suppression of fractalkine, RANTES, and MCP-1 and upregulation of adiponectin. Atherosclerosis 209, 8995.Google Scholar
98. Shah, R, Hinkle, CC, Ferguson, JF et al. (2011) Fractalkine is a novel human adipochemokine associated with type 2 diabetes. Diabetes 60, 15121518.Google Scholar
99. Boisvert, WA, Santiago, R, Curtiss, LK et al. (1998) A leukocyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J Clin Invest 101, 353363.Google Scholar
100. Boisvert, WA, Rose, DM, Johnson, KA et al. (2006) Up-regulated expression of the CXCR2 ligand KC/GRO-alpha in atherosclerotic lesions plays a central role in macrophage accumulation and lesion progression. Am J Pathol 168, 13851395.Google Scholar
101. Straczkowski, M, Dzienis-Straczkowska, S, Stepien, A et al. (2002) Plasma interleukin-8 concentrations are increased in obese subjects and related to fat mass and tumor necrosis factor-alpha system. J Clin Endocrinol Metab 87, 46024606.Google Scholar
102. Neels, JG, Badeanlou, L, Hester, KD et al. (2009) Keratinocyte-derived chemokine in obesity: expression, regulation, and role in adipose macrophage infiltration and glucose homeostasis. J Biol Chem 284, 2069220698.Google Scholar
103. Kitade, H, Sawamoto, K, Nagashimada, M et al. (2012) CCR5 Plays a Critical Role in Obesity-Induced Adipose Tissue Inflammation and Insulin Resistance by Regulating Both Macrophage Recruitment and M1/M2 Status. DiabetesGoogle Scholar
104. Huber, J, Kiefer, FW, Zeyda, M et al. (2008) CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. J Clin Endocrinol Metab 93, 32153221.Google Scholar
105. Calandra, T, Bernhagen, J, Mitchell, RA et al. (1994) The macrophage is an important and previously unrecognized source of macrophage migration inhibitory factor. J Exp Med 179, 18951902.Google Scholar
106. Baugh, JA & Bucala, R (2002) Macrophage migration inhibitory factor. Crit Care Med 30, 2735.Google Scholar
107. Calandra, T & Roger, T (2003) Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immunol 3, 791800.Google Scholar
108. Lue, H, Thiele, M, Franz, J et al. (2007) Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene 26, 50465059.Google Scholar
109. Roger, T, David, J, Glauser, MP et al. (2001) MIF regulates innate immune responses through modulation of Toll-like receptor 4. Nature 414, 920924.Google Scholar
110. Toh, ML, Aeberli, D, Lacey, D et al. (2006) Regulation of IL-1 and TNF receptor expression and function by endogenous macrophage migration inhibitory factor. J Immunol 177, 48184825.Google Scholar
111. Mitchell, RA, Liao, H, Chesney, J et al. (2002) Macrophage migration inhibitory factor (MIF) sustains macrophage proinflammatory function by inhibiting p53: regulatory role in the innate immune response. Proc Natl Acad Sci USA 99, 345350.Google Scholar
112. Gregory, JL, Morand, EF, McKeown, SJ et al. (2006) Macrophage migration inhibitory factor induces macrophage recruitment via CC chemokine ligand 2. J Immunol 177, 80728079.Google Scholar
113. Toso, C, Emamaullee, JA, Merani, S et al. (2008) The role of macrophage migration inhibitory factor on glucose metabolism and diabetes. Diabetologia 51, 19371946.Google Scholar
114. Bernhagen, J, Krohn, R, Lue, H et al. (2007) MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 13, 587596.Google Scholar
115. Lue, H, Dewor, M, Leng, L et al. (2011) Activation of the JNK signalling pathway by macrophage migration inhibitory factor (MIF) and dependence on CXCR4 and CD74. Cell Signal 23, 135144.Google Scholar
116. Fan, H, Hall, P, Santos, LL et al. (2011) Macrophage migration inhibitory factor and CD74 regulate macrophage chemotactic responses via MAPK and Rho GTPase. J Immunol 186, 49154924.Google Scholar
117. Ghanim, H, Aljada, A, Hofmeyer, D et al. (2004) Circulating mononuclear cells in the obese are in a proinflammatory state. Circulation 110, 15641571.Google Scholar
118. Dandona, P, Aljada, A, Ghanim, H et al. (2004) Increased plasma concentration of macrophage migration inhibitory factor (MIF) and MIF mRNA in mononuclear cells in the obese and the suppressive action of metformin. J Clin Endocrinol Metab 89, 50435047.Google Scholar
119. Church, TS, Willis, MS, Priest, EL et al. (2005) Obesity, macrophage migration inhibitory factor, and weight loss. Int J Obes (Lond) 29, 675681.Google Scholar
120. Vozarova, B, Stefan, N, Hanson, R et al. (2002) Plasma concentrations of macrophage migration inhibitory factor are elevated in Pima Indians compared to Caucasians and are associated with insulin resistance. Diabetologia 45, 17391741.Google Scholar
121. Skurk, T, Herder, C, Kraft, I et al. (2005) Production and release of macrophage migration inhibitory factor from human adipocytes. Endocrinology 146, 10061011.Google Scholar
122. Sakaue, S, Ishimaru, S, Hizawa, N et al. (2006) Promoter polymorphism in the macrophage migration inhibitory factor gene is associated with obesity. Int J Obes (Lond) 30, 238242.Google Scholar
123. Yabunaka, N, Nishihira, J, Mizue, Y et al. (2000) Elevated serum content of macrophage migration inhibitory factor in patients with type 2 diabetes. Diabetes Care 23, 256258.Google Scholar
124. Herder, C, Kolb, H, Koenig, W et al. (2006) Association of systemic concentrations of macrophage migration inhibitory factor with impaired glucose tolerance and type 2 diabetes: results from the Cooperative Health Research in the Region of Augsburg, Survey 4 (KORA S4). Diabetes Care 29, 368371.Google Scholar
125. Herder, C, Klopp, N, Baumert, J et al. (2008) Effect of macrophage migration inhibitory factor (MIF) gene variants and MIF serum concentrations on the risk of type 2 diabetes: results from the MONICA/KORA Augsburg Case-Cohort Study, 1984–2002. Diabetologia 51, 276284.Google Scholar
126. Aslani, S, Hossein-nezhad, A, Maghbooli, Z et al. (2011) Genetic variation in macrophage migration inhibitory factor associated with gestational diabetes mellitus and metabolic syndrome. Horm Metab Res 43, 557561.Google Scholar
127. Waeber, G, Calandra, T, Roduit, R et al. (1997) Insulin secretion is regulated by the glucose-dependent production of islet beta cell macrophage migration inhibitory factor. Proc Natl Acad Sci USA 94, 47824787.Google Scholar
128. Saksida, T, Stosic-Grujicic, S, Timotijevic, G et al. (2011). Macrophage migration inhibitory factor deficiency protects pancreatic islets from palmitic acid-induced apoptosis. Immunol Cell Biol 169, 156163.Google Scholar
129. Hirokawa, J, Sakaue, S, Tagami, S et al. (1997) Identification of macrophage migration inhibitory factor in adipose tissue and its induction by tumor necrosis factor-alpha. Biochem Biophys Res Commun 235, 9498.Google Scholar
130. Hirokawa, J, Sakaue, S, Furuya, Y et al. (1998) Tumor necrosis factor-alpha regulates the gene expression of macrophage migration inhibitory factor through tyrosine kinase-dependent pathway in 3T3-L1 adipocytes. J Biochem 123, 733739.Google Scholar
131. Sakaue, S, Nishihira, J, Hirokawa, J et al. (1999) Regulation of macrophage migration inhibitory factor (MIF) expression by glucose and insulin in adipocytes in vitro. Mol Med 5, 361371.Google Scholar
132. Atsumi, T, Cho, YR, Leng, L et al. (2007) The proinflammatory cytokine macrophage migration inhibitory factor regulates glucose metabolism during systemic inflammation. J Immunol 179, 53995406.Google Scholar
133. Ikeda, D, Sakaue, S, Kamigaki, M et al. (2008) Knockdown of macrophage migration inhibitory factor disrupts adipogenesis in 3T3-L1 cells. Endocrinology 149, 60376042.Google Scholar
134. Verschuren, L, Kooistra, T, Bernhagen, J et al. (2009) MIF deficiency reduces chronic inflammation in white adipose tissue and impairs the development of insulin resistance, glucose intolerance, and associated atherosclerotic disease. Circ Res 105, 99107.Google Scholar
Figure 0

Fig. 1. Obesity-induced adipose tissue macrophage (ATM) recruitment. M2 ATM are characteristic of lean adipose tissue. Adipocyte-derived arginase, adiponectin and M2-derived IL-10 are enhanced. During obesity, adipose tissue expansion is accompanied by adipocyte necrosis, hypoxia and enhanced pro-inflammatory cytokine secretion which attract pro-inflammatory M1 macrophages into adipose tissue. In this obesogenic state pro-inflammatory IL-6, IL-1β, TNF-α, inducible nitric oxide synthase (iNOS), monocyte chemoattractant protein (MCP)-1 and adipokines leptin and resistin are up-regulated, exacerbating the inflammatory milieu, while arginase, adiponectin and IL-10 are decreased. Inflammatory signalling pathways c-Jun N-terminal kinases (JNK) and p38 are activated leading to impaired insulin action and systemic insulin resistance (IR).

Figure 1

Fig. 2. Role of macrophage migration inhibitory factor (MIF) in inflammation and adipose tissue dysfunction. 1. MIF activates macrophages inducing inflammatory cytokine secretion; TNF-α and IL-12. 2. Inflammatory stimuli lipopolysaccaride (LPS), TNF-α and interferon-γ can induce MIF secretion from macrophages. 3. Endogenous MIF can regulate toll-like receptor-4 (TLR-4), IL-1 receptor (IL-1R)1, TNF receptor (TNFR) expression 4. MIF inhibits macrophage-derived anti-inflammatory IL-10 secretion. 5. MIF is involved in monocyte recruitment via transmigration and recruitment of monocytes by up-regulating intracellular adhesion molecule-1 (ICAM) and monocyte chemoattractant protein-1 (MCP)-1 and can also by binding to CXC-chemokine receptor (CXCR)2. 6. MIF can promote TNF-α secretion from adipocytes 0·7. MIF can impair insulin-stimulated glucose uptake into adipose tissue and down-regulate phosphorylated AKT expression.