Open access peer-reviewed chapter

The Pivotal Role of Macrophages in Metabolic Distress

Written By

Joseph Roberts, Padraic G. Fallon and Emily Hams

Submitted: 15 February 2019 Reviewed: 23 April 2019 Published: 09 July 2019

DOI: 10.5772/intechopen.86474

From the Edited Volume

Macrophage Activation - Biology and Disease

Edited by Khalid Hussain Bhat

Chapter metrics overview

1,647 Chapter Downloads

View Full Metrics

Abstract

Obesity is a prevalent condition with several associated co-morbidities including the development of metabolic diseases. In obesity there is immune cell infiltration into the white adipose tissue and this is associated with the generation of inflammation and insulin resistance (IR). A large majority of the infiltrating leukocytes in obese adipose tissue are pro-inflammatory macrophages, which upon activation induce a switch in metabolism from oxidative phosphorylation, as is utilised by macrophages in lean adipose tissue, towards aerobic glycolysis. The signalling pathways evoked in the recruited macrophages induce the release of pro-inflammatory cytokines, in signalling pathways which directly interfere with insulin signalling and thus induce a state of IR. As macrophages appear to play such a pivotal role in the generation of IR and are the largest leukocyte population in the adipose tissue, they provide a promising therapeutic target. Indeed, there are several strategies currently being studied to induce a ‘switch’ in macrophages associated with obese adipose tissue, towards the phenotype of those associated with lean adipose tissue, with arguably the most promising being those strategies designed to target the metabolic pathways within the macrophages. This chapter will discuss the polarisation and activation of macrophages within lean and obese adipose tissue and how these cells can be targeted therapeutically.

Keywords

  • macrophage
  • obesity
  • metabolism
  • inflammation

1. Introduction

Obesity is defined as abnormal or excessive fat accumulation and is linked with increased risk of development of multiple co-morbidities, including cardiovascular disease, type 2 diabetes, musculoskeletal disorders and certain cancers. Obesity and its associated co-morbidities are a significant health concern facing the global population. Worldwide obesity has tripled since 1975, with 39% of adults considered overweight and 13% considered obese [1]. This situation is prominent in childhood, with 41 million of the global under five population overweight or obese [1].

Obesity induces a state of low-grade systemic inflammation, characterized by increased serum levels of pro-inflammatory mediators, including C Reactive Protein (CRP), Tumour Necrosis Factor (TNF)-α, Interleukin (IL)-1β and IL-6, which contributes to metabolic dysfunction and insulin resistance (IR) [2]. Although the mechanisms underlying this inflammatory response are not fully understood, activation of adipose tissue macrophages (ATM) contributes to this inflammatory state, and therefore to the development of insulin resistance (IR) [3, 4]. Conversely, in lean individuals the immune repertoire constitutes a more anti-inflammatory phenotype, with ATM alongside regulatory T cells (Tregs) releasing cytokines such as IL-10 and transforming growth factor (TGF)-β, which increase insulin sensitivity [5]. Therefore, the role of ATM in metabolic function is clearly an area of interest, indeed transcriptional profiling has identified how quickly macrophages can respond and adapt to alterations in their microenvironment [6]. This chapter will focus on the role macrophages play in the pathogenesis of metabolic disorders and explore if re-education of these cells provides a target for therapeutic intervention in obesity and its related co-morbidities.

Advertisement

2. The microenvironment of the adipose tissue in lean and obese individuals

Obesity historically was believed to be due to a combination of genetic predisposition and environmental factors, however, more recently it has been recognised that immunological factors can also contribute to the pathogenesis of obesity. Indeed, while over 30 gene loci combinations have been associated with the development of obesity and metabolic disease, these loci are only associated with 2–3% of the incidence of these conditions [7]. Further the energy-dense modern Western diet combined with a sedentary lifestyle undoubtedly adds to the obesity epidemic. Recent work has identified the links between dysbiosis in the intestinal microbiome and immune cell activation, linked to the ingestion of high-fat, low-fibre diets, and the development of obesity [8].

2.1 The role of the microbiome

The intestinal microbiome is essential for processing dietary polysaccharides and has been identified as a key regulator of systemic inflammation in obesity [9]. Mouse studies are routinely used to study the mechanisms underlying obesity and metabolic disease. Due to the nature of obesity being largely related to diet, diet-induced models are often favoured over genetic models (for example, leptin deficient ob/ob mice). Indeed, studies using a high-fat diet (HFD; equivalent to 60% animal-derived fats in the diet) have been used to study the potential implications of alteration in the microbiome related to diet as well as other obesity-related pathogenesis. It has been shown that the microbiome in obese mice has an increased capacity to harvest energy from the diet compared to the microbiome from lean mice [10]. Microbiome transfer studies, in which intestinal microbiota from mice raised in conventional housing was transferred into germ-free mice, induced a 60% increase in body fat and IR within 14 days, despite a reduction in food consumption [11]. The transfer of microbiota-derived products such as lipopolysaccharides and peptidoglycans, have shown to promote metabolic endotoxemia, which induces pro-inflammation in adipose tissue [12]. In contrast, the microbiota of the gut bacterial fermentation of dietary fibre was shown to have anti-inflammatory effects [12]. Indeed, it was shown that the transfer of intestinal microbiota from lean donors increased the insulin sensitivity in individuals with metabolic syndrome [13].

2.2 Adipose tissue

Adipose tissue (AT) is an important metabolic organ, which helps orchestrates metabolic and endocrine functions as well as immune responses [12]. AT functions to store excess nutrients as triacylglycerides and releases fatty acids in the fasted state, provide cold insulation and protection of vital organs. In the AT of obese individuals, there is significant adipocyte hyperplasia and adipose tissue hypertrophy [14]. AT consists of mature adipocytes, pre-adipocytes, fibroblasts, endothelial cells, histocytes and populations of immune cells including monocytes, macrophages, natural killer (NK) cells, innate lymphoid cells (ILCs) and lymphocytes. AT is classified into three categories, namely white (WAT), beige or ‘brite’ (beige/brite) and brown (BAT). WAT accounts for approximately 50% of body mass and can release free fatty acids (FFA) into circulation when glucose levels are low. Whilst BAT plays an important role in thermogenesis and the production of heat [15].

The AT of obese individuals is in a state of chronic low-grade inflammation with marked infiltration various pro-inflammatory immune cells such as CD8 cells, NK cells, ILC1, Th1 cells, neutrophils and pro-inflammatory macrophages [16]. Conversely, the immune repertoire of AT from lean individuals comprises anti-inflammatory cell populations, including eosinophils, ILC2, Tregs, Th2 cells and anti-inflammatory macrophages [16] (Figure 1). In lean mice, ATM constitutes approximately 5% of cells, conversely in obese mice ATM can account for up to 50% of the cells [3]. Whilst in lean human AT, ATM comprises 4% of cells compared to 12% in excess adiposity [17]. In addition to macrophages, lymphocytes and ILCs also play roles in the regulation of AT inflammation, the roles of which seem to largely involve supporting the polarisation state of the ATM populations. For example, eosinophils provide a source of IL-4 promoting an M2 phenotype. In obese mice adipose eosinophils are decreased, whilst depletion of eosinophils results in increased M1 ATM, weight gain and systemic IR [18]. Furthermore, ILC2 have been identified as a source of IL-5, a key cytokine in eosinophil recruitment. Thus, accumulation of eosinophils and maintenance of M2 ATM relies on ILC2 [19].

Figure 1.

Immune cell composition of adipose tissue in a lean and obese state. In the lean state, eosinophils and type 2 innate lymphoid cells (ILC2s) produce Th2 cytokines (IL-4, IL-5 and IL-13), which promotes eosinophil recruitment and anti-inflammatory polarisation of macrophages towards an M2 phenotype, which is supported in the normoxic state of lean adipose tissue. In turn, M2 macrophages secrete anti-inflammatory cytokines such as IL-10. In the obese state, adipocyte hypertrophy, hyperplasia and hypoxia cause necrotic adipocytes, resulting in pro-inflammatory state and macrophage recruitment, forming crown like structures (CLS) surrounding the adipocytes. These macrophages are polarised towards an M1 phenotype and secrete the pro-inflammatory cytokines IL-6, IL-1β and TNF-α.

2.3 The role of the adaptive immune system in obesity

The adaptive immune system also plays an important role in obesity and metabolic disease. B cells have been shown to be involved in obesity induced inflammation and IR [20]. In obese mice, there was an identified increased in IgG+B cells and IgG production, associated with activation of M1 ATM, increased Th1 cells and conversely, attrition of Treg cells [21]. In addition, transferring B cells into B cell deficient mice induced IR [20]. Furthermore, in obese mice, there is increased CD8+ effector T cell recruitment in epididymal AT. Interestingly, it is reported that CD8+ T cells precede macrophage infiltration and deletion of CD8+ T cells resulted in reduced macrophage infiltration and AT inflammation whilst improving IR [22]. Conversely, both Treg cells and iNKT cells are negatively associated with obesity-induced inflammation and are enriched in lean AT. Indeed, both these immune cells are known to secrete IL-10 which promotes M2 macrophage polarisation [23, 24].

2.4 Adipose tissue macrophages

ATMs appear to play a major role in the regulation of obesity-related inflammation, with different macrophage phenotypes associated with divergent roles in the AT. In lean animals, ATM function to maintain the homeostatic micro-environment in AT by taking up excess lipids and phagocytosing dead adipocytes. Broadly speaking macrophages present in lean AT are of an M2 phenotype, which have been shown to suppress inflammation in AT [25]. Furthermore, M2 macrophages in lean AT have been associated with brown fat activation and ‘beiging’ of WAT in mouse models of obesity, via expression of tyrosine hydroxylase, which induces thermogenesis [26, 27]. However, this process has recently been queried, with IL-4-stimulated macrophages failing to generate sufficient levels of catecholamines to contribute to adipose tissue adaptive thermogenesis [28]. Conversely, excess lipid uptake in obese AT, induces M1 polarisation and along with excess lipid droplets, immune cells and necrotic adipocytes this forms a component called ‘crown-like’ structures (CLS) [29, 30]. Indeed, it has been shown that more than 90% of all macrophages in WAT of obese mice and humans are localized to dead adipocytes [31]. This metabolic activation of M1 macrophages in obese AT is associated with increased pro-inflammatory cytokines in the AT and recruitment and activation of M1 macrophages in the AT [32].

ATM in lean AT is considered a resident macrophage population, which originates from yolk-sac progenitors and self-renews via proliferation under homeostatic conditions. Over time into adulthood resident ATMs are replaced with circulating monocytes derived from bone marrow [33]. Using mouse bone marrow chimera experiments, following transplanting donor CD45.1+ bone marrow into recipient CD45.2+ mice, and maintenance on obesity-inducing HFD, 85% of the ATM were donor-derived compared to 15% that were recipient-derived [3]. Interestingly, the polarization of macrophages in obesity from an M2 to an M1 phenotype has been mainly attributed to the recruitment of monocytes to AT, rather than the conversion of tissue resident M2 macrophages [34]. Murine monocytes can be classified through the expression of Ly6C, with Ly6Chi monocytes considered inflammatory. In the steady state Ly6Chi monocytes differentiate into Ly6Clo monocytes in the circulation, which are believed to differentiate into M2 macrophages in the tissue. However, in obese AT in response to inflammatory stimuli such as the monocyte chemoattractant CCL2, Ly6Chi macrophages are recruited to the AT where they differentiate to M1-like ATM [25]. Indeed, in absence of Ccl2 expression macrophages expressed an M2 gene profile [35].

ATM represents the largest population of leukocytes within the AT and plays many vital homeostatic roles including tissue remodelling and insulin sensitivity. However, with progressive obesity ATM are the key mediators of inflammation, IR and the impairment of adipocyte function.

Advertisement

3. Polarization of ATM and the link to IR

Macrophages are extremely heterogenic in function and phenotype, and have historically been characterized into two phenotypes; M1 and M2. M1 macrophages are often defined as ‘classically activated’ and are generally pro-inflammatory in function, with a vital role in eliminating pathogens and virus-infected cells. Whereas, M2 macrophages and termed ‘alternatively activated’ are anti-inflammatory in function and promote tissue repair and wound healing. This is very simplified and dated model however, evidence now suggests that ATMs include highly plastic cell populations, with their phenotype largely dependent on the microenvironment of the AT. Whilst the exact number and function of ATM in the AT is evolving, it is clear there are distinct populations in the lean and obese AT, with unique tissue distribution, marker expression, transcriptional profiles and functions. Indeed, obese ATM display markers that are largely induced by their metabolic state rather than cytokine stimuli that classically polarise M1 and M2 cells [32].

3.1 M1 and M2 macrophage phenotypes

M1 macrophages are activated by signals associated with infection such as IFN-ɣ as well as bacterial-derived products such lipopolysaccharide (LPS) and free fatty acids (FFA). M1 macrophages are loosely identified by surface expression of F4/80+CD11C+ with high levels of MHC-II, CD68, CD80 and CD86 costimulatory molecules in addition to release of TNF-α and inducible nitric oxide synthase (iNOS) [36]. M2 macrophages are activated via Th2 cytokines, IL-4 and IL-13 as well as by parasitic products. M2 macrophages are loosely identified as F4/80+CD206+CD301+CD11C and express genes encoding anti-inflammatory proteins such as Chil3, Arg1 and Il10 in mice [34]. A crucial transcription factor in M2 macrophage polarisation is peroxisome proliferator-activated receptor (PPAR-γ/δ), which can be driven by adipocyte derived IL-4 and IL-13 [37, 38, 39] (Figure 2). Some markers vary between mice and human macrophages. For example, there are no human homologues of the M2-associated genes Chil3, Arg1 and Fizz1, with human M2 identified based on expression of tranglutaminase-2 (TGM2) and CD68 [40].

Figure 2.

Adipose tissue macrophage (ATM) polarisation in lean and obese state. ATM originate from polarisation of ‘M0’ macrophages. Depending on the stimuli and local environment facilitates the macrophage polarisation. Th1 cytokines, LPS, IFN-ɣ, TNF-α and IL-1β promote a M1 macrophage that is characterised by F4/80+CD11c+. In contrast, Th2 cytokines, IL-4 and IL-13 promote an M2 macrophage that is characterised by F4/80+CD206+CD301+CD11c. Interestingly, in an obese state, the hypoxic environment and metabolic cues such as excess free fatty acids, high insulin and glucose, oxidised phospholipids and low-density lipoprotein, which promotes a metabolic activated (MME) or oxidised (MOX) macrophage. In the obese state, macrophages promote inflammation, IR and tissue damage. In contrast, in the lean state, macrophages promote ant inflammation, insulin sensitivity and tissue repair.

3.2 MMe and Mox macrophage phenotypes

Macrophages with a phenotype associated with obesity are induced by several metabolic stimuli such as FFA, high insulin and glucose, oxidised phospholipids and low-density lipoproteins. These macrophages display surface markers that are neither representative of typical M1 or M2 macrophages and give rise to a population of metabolic activated (MMe) and oxidised (Mox) macrophages [41, 42] (Figure 2). Both these macrophage phenotypes are associated with a state of IR. MMe macrophages cell-surface markers express ABCA1, CD36 and PLIN2 and are involved in the clearance of dead adipocytes through lysosomal exocytosis as well as potentiating inflammation. NADPH-oxidase-2 (NOX2) has been identified as a key driver of the functions of MMe macrophages, with Nox2-deficient mice displaying attenuated ATM inflammation and improved glucose sensitivity in a model of diet-induced obesity, when compared to WT animals [41]. Mox macrophages are driven by oxidised phospholipids derived from oxidised low density lipoproteins (LDL) [43] and express surface markers Srnx-1 and Txnrd-1 [42]. Mox macrophages have been studied primarily in the context of atherosclerosis, where the oxidation of accumulated LDL leads to enrichment of the tissue with oxidised lipids, causing the polarisation of macrophages towards a phenotype dependent on the transcription factor Nrf2 [43]. However, a recent paper identified that ATM with a Mox phenotype (CX3CR1negF4/80lowTxnrd1+HO1+) are the predominant phenotype present the AT of lean mice, driven by individual oxidised phospholipids in the AT [44]. It will be an interesting further area study to appraise the role of these novel macrophage phenotypes within the context of obesity and IR.

3.3 Regulators of macrophage polarization

Macrophage polarization has been well studied over the past decade leading to the discovery of several key regulators which orchestrate macrophage polarization, such as the Signal Transducer and Activator of Transcription (STAT) family, interferons, regulators of lipid metabolism, transcription factor families, microRNAs (miRNAs) and long non-coding RNAs [45] (Figure 3).

Figure 3.

Intracellular signalling of M1 and M2 macrophage polarisation. Respective intracellular key regulators of M1 and M2 macrophage polarisation. Signal transducer and activator of transcription (STAT) family, interferons (IRFs) and microRNA (miRNAs).

3.3.1 STAT family members

The Janus Kinase (JAK)/STAT signalling pathway transmits signals from extracellular cytokines into the nucleus. Indeed, JAK/STATs are arguably the most widely studied pathway within the context of macrophage polarisation, with IFNɣ binding to its receptor triggering activation of JAK1/2-mediated tyrosine phosphorylation and subsequent dimerization of STAT1 one of the first pathways to M1 polarisation identified [46]. In addition, LPS binding to TLR4 induces autocrine production of IFN-ß that activates the type 1 IFN receptor triggering STAT1 and STAT2 phosphorylation and heterodimerisation [47]. STAT3 has a dichotomous role in macrophage polarisation, it is the key transcriptional regulator in the production of the anti-inflammatory cytokine IL-10, which can drive an anti-inflammatory macrophage phenotype, however, STAT3 can also be activated by IL-6 and IFN-ß, inducing a pro-inflammatory phenotype [48]. Conversely, IL-4 and IL-13 induces M2 macrophage polarisation largely through induction of STAT6 and KLF4 via the dual catalytic activities of MCP-1-induced protein and inducing PPAR-ɣ [49, 50, 51].

3.3.2 Interferon regulatory factors

Interferon regulatory factors (IRFs) are intracellular proteins that regulate immune cell maturation and play a pivotal role in macrophage polarization. Two key and opposing IRFs in macrophage polarisation are IRF4 and IRF5, which directly compete for binding to MyD88 and subsequent transcription factors such as NFκB. Interestingly, IRF5 was shown to promote M1 macrophages, while IRF5 expression was upregulated in obese individuals compared to lean individuals at both the mRNA and protein levels and is negatively associated with insulin sensitivity [52, 53, 54]. It was also shown that IRF5 promotes inflammatory macrophage polarization by activating the transcription of IL-12 and repressing IL-10 [55]. Conversely, IRF4 acts as an antagonist of M1 macrophage polarisation, promoting M2 macrophages [53, 54, 56]. In the context of obesity, macrophage-specific knockout of IRF4 resulted in significant IR and an increase in expression of pro-inflammatory genes [57]. Furthermore, IRF6 has also been implicated in macrophage polarisation, promoting M1 macrophages due to suppression of PPARɣ expression, a critical regulator of M2 macrophages. Overexpression of IRF6 reduced M2 activation, whilst IRF6 knockdown enhanced M2 macrophage activation [58]. The impact of IRFs on macrophage polarisation and plasticity is clearly quite complex and further studies will hopefully provide information on how IRFs function in different microenvironments, for example, in lean versus obese AT.

3.3.3 MicroRNAs

miRNA are short inhibitory non-coding RNAs (~22 nucleotides) that degrade specific mRNA targets or block RNA translation, and have also been implicated in driving macrophage polarisation. Indeed, miR-125b expression was shown to be upregulated in murine macrophages following IFN-ɣ stimulation and was identified to promote M1 macrophage polarisation, while suppressing IRF4, an important M2 transcription factor [59]. Additionally, miR-155 was also shown to promote M1 polarisation with expression in murine macrophages increased upon TLR activation or stimulation with pro-inflammatory cytokines (TNF-α, IFN-β or IFN-ɣ) [45]. Whilst in human macrophages, miR-155 was shown to target IL-13Rα1 and inhibit STAT6 activation, thus inhibiting M2 macrophage polarisation [60]. Additionally, miR-9 enhances M1 macrophage polarisation by suppressing PPARδ [61]. Whilst miR-127 suppresses B-Cell lymphoma protein (Bc6), which promotes M1 polarisation [62]. Conversely, miR-124 promotes M2 polarisation via LPS-induced cytokine production by targeting STAT3 to decrease IL-6 production and reduce TNF-α [63]. Furthermore, miR-132, miR-146a and miR-223 induce M2 macrophages by inhibiting NF-κB [64, 65, 66].

3.4 Hypoxia and macrophage polarisation

Of interest in the context of obesity is the potential for hypoxia to influence macrophage polarisation of macrophages (Figure 2). Indeed, hypoxic areas in adipose tissue occur in obese individuals when rapid tissue expansion occurs without sufficient accompanying blood flow to these areas. M1 macrophages display high expression levels of Hypoxia-related genes including Hif1α, which has been shown to induce a pro-inflammatory phenotype in macrophages via TLR4 activation, involving the PI3K/Akt signalling pathway [67, 68]. TLR4 expression in macrophages was shown to increase in a hypoxic environment [69]. Hif1α enhances the transcriptional activity of NF-κB, driving production of pro-inflammatory cytokines and decreasing the induction of immune regulatory mediators [70]. In contrast, M2-like macrophages in obese individuals express Hif2α [71]. Indeed, HIF-2α is also upregulated under low oxygen levels [5]. It was shown that HIF-2α overexpressing macrophages suppressed pro-inflammatory responses and improved IR. Whilst knockdown of HIF-2α in macrophages induced pro-inflammatory gene expression in adipocytes [71]. Thus, it was suggested that HIF-2α counteracts the pro-inflammatory responses to relieve obesity induced IR in AT [71].

3.5 IR and macrophage polarisation

Insulin acts in the adipose tissue to promote uptake and storage of fatty acids, stored as triglycerides, and inhibits the lipolysis of stored triglycerides. IR is a reduced response to insulin in the liver, muscle and AT, due to an impairment in the insulin-signalling pathway, leading to hyperglycaemia [72]. In obesity, the increased recruitment of macrophages to AT positively correlates with IR [4]. Indeed, obese mice show increased macrophage-mediated inflammation that resulted in long-term IR [73], with elevated levels of FFA which activated CD36 expressing macrophages and adipocytes, inducing inflammation and impairing insulin signalling [74]. In AT from lean states, the presence of M2 macrophages maintains insulin sensitivity via anti-inflammatory actions of IL-10 and STAT3 [25]. In obese individuals, there is a change in adipocyte metabolism and gene expression. Consequently, there is increased lipolysis and release of FFA which can lead to TLR4 activation of M1 macrophages [75]. Indeed, TLR4 expression is increased on macrophages in obese individuals. In co-culture studies, it was shown that macrophage activation through TLR4 signalling increased secretion of pro-inflammatory cytokines, blocking the insulin signalling cascade [76, 77]. In obese individuals, the IκB kinase (IKK) complex is activated in macrophages resulting in phosphorylation of IκBα on Ser32 and 36, degrading IκBα and allowing NF-κB to translocate to the nucleus and upregulated target genes such as pro-inflammatory cytokines including TNF-α, IL-1β and IL-6 [78].

Increased expression of pro-inflammatory cytokines negatively affects insulin signalling pathways [79]. The effect of these cytokines on IR are seen locally in AT, but also systemically as they are released into circulation [76]. TNF-α phosphorylates insulin receptor substrates (IRS), consequently preventing downstream signalling via inhibiting IKK, c-Jun n-terminal kinase (JNK) and atypical protein kinase C (aPKC) [80]. Interestingly, males have been shown to have increased TNF-α plasma concentration compared to females, leading to the suggestion that obese males are more susceptible to develop IR [81]. IL-6 is increased in the serum of obese individuals and mice, with weight loss reducing circulating IL-6, which improves insulin sensitivity [82]. Systemic IR has also observed during pregnancy, puberty and during infection such as sepsis driven by TNF-α and IL-6 [83]. It is clear from such studies that the presence of pro-inflammatory cells and the release of pro-inflammatory cytokines lead to a loss of sensitivity to insulin and a state of IR. This is possibly the biggest incentive to therapeutically target the macrophages in cases of metabolic disease.

Advertisement

4. The metabolic signature of macrophages

Metabolism is a series of highly interconnected pathways that generate metabolic products such as energy and macromolecules from nutrients in the microenvironment. Whilst the metabolic pathways are plastic, cells, in particular macrophages, tend to utilise a pathway that suits their immediate energy requirements. M1 macrophages have huge metabolic demands, and rely largely on glycolysis, conversely, M2 macrophages meet their energy requirements using oxidative phosphorylation (OXPHOS) pathways. During glycolysis, extracellular glucose is taken up by the cell and converted to two molecules of pyruvate and ATP; NAD+ is converted to NADH+H+ regenerated through the breakdown of pyruvate to lactate. Glycolysis also provides the first molecule in the pentose-phosphate pathway, glucose-6-phosphate, which provides NADPH to maintain the cellular redox balance and the production of fatty acids. In M1 macrophages, the increased glucose consumption is associated with the capacity for rapid cytokine production and antimicrobial activity through ROS generation [84, 85]. Conversely, in the presence of oxygen cells produce ATP via the electron transport chain (ETC), which is linked to the tricarboxylic acid (TCA) cycle. The TCA cycle uses carbon sources, such as Acetyl CoA, glutamine or fatty acids to fuel a cycle which generates the reducing agents NADH and FADH2 that serve as electron carriers for the ETC for OXPHOS. Glycolysis is a poor producer of energy, with only two molecules of ATP per glucose molecule, compared to 36 molecules produced by OXPHOS. However, the use of each pathway will depend on the environment and functional requirements of the cells, as glycolysis provides energy rapidly.

The TCA cycle is truncated in M1 macrophages, resulting in a reduced production of alpha-ketoglutarate (α-KG) and accumulation of citrate and succinate metabolites [86]. The accumulation of citrate leads to the production of the macrophage specific metabolite itaconic acid, which is a major feature of LPS stimulated macrophages [86]. The build-up of itaconic acid has been identified as a driver for succinate accumulation, through its ability to inhibit succinate dehydrogenase [87, 88]. The excess succinate leads to the induction of IL-1β through the stabilization of HIF-1a further enhancing inflammation in the macrophages [89]. Indeed, blocking glycolysis reduces release of CCL2 from TNF-a or LPS stimulated adipocytes, providing further evidence for a link between metabolism and inflammation [90] (Figure 4).

Figure 4.

Immunometabolic differences in M1 and M2 macrophages. M2 macrophages use OXPHOS and the TCA cycle to produce energy. They have increased ability to uptake free fatty acid (FFA) and fatty acid oxidation (FAO) to facilitate the TCA cycle. In contrast, M1 macrophages increase energy production oxidative glycolysis. M1 macrophages also have a ‘broken’ TCA cycle, resulting in accumulate of citrate and succinate. Increased succinate results in secretion of pro-inflammatory cytokine IL-1β via HIF-1α. TCA, tricarboxylic acid; HIF-1a, hypoxia-inducible factor-1a; a-KG, alpha-ketoglutarate.

Macrophage polarization is also influenced by the metabolism of arginine. M1 macrophages upregulated nitric oxide synthase (iNOS), which catabolize arginine to citrulline and nitric oxide (NO). This NO is important for intracellular killing of pathogens. In addition, M1 macrophages use the pentose phosphate pathway, which generates NADPH for the NADPH oxidase, which produces ROS and NO. Consequently, these metabolic pathways provide M1 macrophages with rapid energy. Conversely, in M2 macrophages, arginase-1 (Arg1) is induced which produces urea, ornithine and polyamines which are key in tissue repair [36, 91].

The metabolic profile of ATM alters dependent on the microenvironment of the AT. In mice, transcriptome and extracellular flux analysis have shown that in lean AT fatty acid oxidation, glycolysis and glutaminolysis all participate in cytokine release by ATM [92]. In obese AT, both glycolysis and OXPHOS are utilised, however glycolysis takes precedence, potentially due to the hypoxic environment in the AT [92].

Advertisement

5. Could targeting macrophages provide a therapeutic strategy in metabolic disorders?

As described above, as macrophages play a significant role in obesity and other metabolic disorders they are an attractive therapeutic target. The therapeutic strategies that target macrophages look to re-educate polarized macrophages, depletion of polarized macrophages or silencing macrophages. Additionally, the link between macrophage polarization and cellular metabolism suggests a potential therapeutic strategy by modulating the macrophage metabolic state. There are several therapeutic strategies commonly used to target macrophages such as depletion, proliferation, inflammation and gene silencing.

5.1 Macrophage depletion

It was shown that macrophages could be depleted in vivo by inducing apoptosis following accumulation of toxic particles [93]. Interesting, it was shown that by depletion of pro-inflammatory macrophages resulted in normalizing insulin sensitivity in IR obese mice [94]. Furthermore, in obese mice, the depletion of visceral adipose tissue macrophages (VATMs) by Intraperitoneal injection of clodronate liposomes, results in improved systemic insulin sensitivity, glucose homeostasis and further blocked high-fat diet-induced weight gain [95, 96]. Consequently, depletion of VATMs also resulted in prevention of CLS in WAT and a low level of blood TNF-α [96]. However, liposomes treatment as a therapy is prone to degradation and significant risks of potential off-target effects. An alternative approach is altering macrophage proliferation, such as using a nanoparticle-based delivery of simvastatin, which may provide therapeutic benefit for atherosclerosis [97]. However, as previously mentioned, in obesity, macrophages are recruited from circulating monocytes, so reducing proliferation may not provide therapeutic benefit for IR [98].

5.2 Biological therapeutics

There are several orally active synthetic ligands for PPARɣ which are used to treat IR in patients with T2D. It has been shown in vivo that pioglitazone, belonging to the chemical class thiazolidinediones, reduces LPS induced TLR2 and TLR4 expression on peritoneal macrophages. Whilst in vitro, pioglitazone reduces the synthesis and gene expression of TLR2, TLR4, IL-1β, TNF-α, IL-6 and MCP-1 in human blood monocytes [99]. However, the use of thiazolidinediones like pioglitazone in clinical studies to treat T2D has resulted in increased cardiovascular events and death [100].

Clinical studies have shown that anti-inflammatories are efficacious in patients with systemic IR. Members of the interferon family have been used to suppress the release of pro-inflammatory cytokines, however, the use of type 1 interferons, as well as other anti-inflammatory strategies, is associated with cell toxicity in long-term use. Recently studies using interferon tau (IFNT), an alternative member of the type 1 interferon family, in mice with diet-induced obesity show enhanced insulin sensitivity when compared to untreated mice. There was also a significant decrease in secretion of pro-inflammatory cytokines and increased M2 macrophages in AT, suggesting IFNT as a novel bio-therapeutic agent for treating obesity-associated disorders [101].

Interestingly, yeast-derived β-glucans (Y-BGs) have been shown to be beneficial in models for obesity. In obese humans, Y-GBs administered orally increased AT expression of anti-inflammatory cytokine IL-10 and serum IL-10 [102]. In addition, macrophages uptake of Y-GBs increased reactive oxygen species (ROS) formation, phagosomal maturation and induction of autophagy [103].

5.3 RNA interference

Another attractive therapeutic approach in targeting macrophage polarisation would be to use RNA interference (RNAi), which reduces gene expression. This approach could target the inflammatory mediators such as TNF-α, IL-6 and IL-1β [98]. Indeed, it was shown that intraperitoneal (i.p.) administration of small interfering RNA (siRNA) selectively silenced genes such as TNF-α in epididymal ATM of obese mice and improved glucose tolerance [7]. Additionally, it was shown that i.p. administration of a rabies virus glycoprotein-derived acetylcholine receptor-binding peptide delivers siRNA into ATM and peritoneal macrophages in HFD mice. This resulted in inhibition of ATM infiltration and reduced pro-inflammatory cytokines, thus improving glucose tolerance and insulin sensitivity [104].

5.4 Metabolic reprogramming

As stated previously, macrophage phenotypes have distinct metabolism pathways. Therefore, altering the metabolic state of macrophages provides a potential therapeutic approach to metabolic disorders. Indeed, the strong link between macrophage polarization and cellular metabolism makes altering the metabolic state of the cells an attractive therapeutic prospect. To prove this principle, inducing oxidative metabolism in M1 macrophages has been shown to shift the phenotype to an M2 profile [105], while blocking oxidative metabolism in macrophages inhibits the M2 phenotype and drives the M1 macrophage phenotype. Furthermore, it was shown that by driving macrophage metabolism with glucose, insulin and fatty acids resulted in an increased pro-inflammatory ATM phenotype in obese mice [32].

Modulation of the metabolic pathways in macrophages has been studied extensively in recent years to assess the extent to which inflammatory status can be influenced by the metabolic profile of the cells. Glucose transporter (GLUT)-1 is upregulated in macrophages localised to the CLS in inflamed obese AT. In vitro studies show that overexpression of GLUT1 increases glucose uptake in the cells and induces release of pro-inflammatory cytokines, linking the metabolic phenotype with the inflammatory function of the cells [106]. Furthermore, knockout of fatty acid transporter protein (FATP)-1 expression, which is elevated in M2 macrophages, is associated with priming of macrophages towards an M1 phenotype, upregulating expression of NOS1 [107]. Further pathways of current interest to therapeutically target include Notch, carbohydrate kinase-like protein (CARKL), mammalian target of rapamycin (mTOR), IL-4 and IL-10, all of which show intricate links between the metabolic and inflammatory pathways in macrophages [108, 109, 110, 111, 112].

Advertisement

6. Conclusions

Obesity has long been considered a low-grade systemic inflammatory condition, which appears to be mediated largely through the prominent populations of ATM. Alterations in environmental cues, including changes in metabolites, the microbiota and inflammatory stimuli act to influence the ATM, coordinating the recruitment of pro-inflammatory monocytes and altering the metabolic state of ATM. While in lean individuals the resident ATM function to clear dead adipocytes and sequester excess lipids from the AT to maintain homeostasis within the AT, the recruited inflammatory ATM release pro-inflammatory cytokines to induce inflammation within the AT and are involved in the pathogenic remodelling of the AT. This state of inflammation within the ATM is largely associated with IR and metabolic dysfunction through interference with insulin signalling pathways. Macrophages are heterogenous and extremely plastic and as such it has historically been difficult to define subsets. With the use of transcriptional and metabolic profiling it is now becoming possible to appraise the full role of ATM in obesity. This knowledge will aid the search for novel therapeutics targeting the metabolic capacity and inflammatory potential of ATM, restoring the homeostatic functions of resident lean ATM, to modulate obesity.

Advertisement

Acknowledgments

EH is an SFI Starting Investigator (15/SIRG/3473) and PF is supported by the National Childrens Research Center and SFI (17/TIDA/5032).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. WHO. WHO—Obesity and Overweight. 2018. Available from: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight [Accessed: 19 April 2019]
  2. 2. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annual Review of Immunology. 2011;29:415-445
  3. 3. Weisberg SP et al. Obesity is associated with macrophage accumulation in adipose tissue. The Journal of Clinical Investigation. 2003;112(12):1796-1808
  4. 4. Xu H et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. The Journal of Clinical Investigation. 2003;112(12):1821-1830
  5. 5. Fang HY et al. Hypoxia-inducible factors 1 and 2 are important transcriptional effectors in primary macrophages experiencing hypoxia. Blood. 2009;114(4):844-859
  6. 6. Xue J et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40(2):274-288
  7. 7. Aouadi M et al. Gene silencing in adipose tissue macrophages regulates whole-body metabolism in obese mice. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(20):8278-8283
  8. 8. Sittipo P et al. Intestinal microbiota and the immune system in metabolic diseases. Journal of Microbiology. 2018;56(3):154-162
  9. 9. Cox LM, Blaser MJ. Pathways in microbe-induced obesity. Cell Metabolism. 2013;17(6):883-894
  10. 10. Turnbaugh PJ et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027-1031
  11. 11. Backhed F et al. The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(44):15718-15723
  12. 12. Mraz M, Haluzik M. The role of adipose tissue immune cells in obesity and low-grade inflammation. The Journal of Endocrinology. 2014;222(3):R113-R127
  13. 13. Vrieze A et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143(4):913-6.e7
  14. 14. Jo J et al. Hypertrophy and/or hyperplasia: Dynamics of adipose tissue growth. PLoS Computational Biology. 2009;5(3):e1000324
  15. 15. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell. 2014;156(1-2):20-44
  16. 16. Guzik TJ et al. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovascular Research. 2017;113(9):1009-1023
  17. 17. Harman-Boehm I et al. Macrophage infiltration into omental versus subcutaneous fat across different populations: Effect of regional adiposity and the comorbidities of obesity. The Journal of Clinical Endocrinology and Metabolism. 2007;92(6):2240-2247
  18. 18. Wu D et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science. 2011;332(6026):243-247
  19. 19. Molofsky AB et al. Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. The Journal of Experimental Medicine. 2013;210(3):535-549
  20. 20. Winer DA et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nature Medicine. 2011;17(5):610-617
  21. 21. DeFuria J et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(13):5133-5138
  22. 22. Nishimura S et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nature Medicine. 2009;15(8):914-920
  23. 23. Exley MA et al. Interplay between the immune system and adipose tissue in obesity. The Journal of Endocrinology. 2014;223(2):R41-R48
  24. 24. Feuerer M et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nature Medicine. 2009;15(8):930-939
  25. 25. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. The Journal of Clinical Investigation. 2007;117(1):175-184
  26. 26. Nguyen KD et al. Alternatively activated macrophages produce catecholamines to sustain adaptive thermogenesis. Nature. 2011;480(7375):104-108
  27. 27. Qiu Y et al. Eosinophils and type 2 cytokine signaling in macrophages orchestrate development of functional beige fat. Cell. 2014;157(6):1292-1308
  28. 28. Fischer K et al. Alternatively activated macrophages do not synthesize catecholamines or contribute to adipose tissue adaptive thermogenesis. Nature Medicine. 2017;23(5):623-630
  29. 29. Prieur X et al. Differential lipid partitioning between adipocytes and tissue macrophages modulates macrophage lipotoxicity and M2/M1 polarization in obese mice. Diabetes. 2011;60(3):797-809
  30. 30. Murano I et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. Journal of Lipid Research. 2008;49(7):1562-1568
  31. 31. Cinti S et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. Journal of Lipid Research. 2005;46(11):2347-2355
  32. 32. Kratz M et al. Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metabolism. 2014;20(4):614-625
  33. 33. Ginhoux F, Guilliams M. Tissue-resident macrophage ontogeny and homeostasis. Immunity. 2016;44(3):439-449
  34. 34. Lumeng CN et al. Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes. 2008;57(12):3239-3246
  35. 35. Oh DY et al. Increased macrophage migration into adipose tissue in obese mice. Diabetes. 2012;61(2):346-354
  36. 36. Galvan-Pena S, O’Neill LAJ. Metabolic reprograming in macrophage polarization. Frontiers in Immunology. 2014;5:420
  37. 37. Van Dyken SJ, Locksley RM. Interleukin-4- and interleukin-13-mediated alternatively activated macrophages: Roles in homeostasis and disease. Annual Review of Immunology. 2013;31:317-343
  38. 38. Ying W et al. MicroRNA-223 is a crucial mediator of PPARgamma-regulated alternative macrophage activation. The Journal of Clinical Investigation. 2015;125(11):4149-4159
  39. 39. Stienstra R et al. Peroxisome proliferator-activated receptor gamma activation promotes infiltration of alternatively activated macrophages into adipose tissue. The Journal of Biological Chemistry. 2008;283(33):22620-22627
  40. 40. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Reports. 2014;6:13
  41. 41. Coats BR et al. Metabolically activated adipose tissue macrophages perform detrimental and beneficial functions during diet-induced obesity. Cell Reports. 2017;20(13):3149-3161
  42. 42. Russo L, Lumeng CN. Properties and functions of adipose tissue macrophages in obesity. Immunology. 2018;155(4):407-417
  43. 43. Kadl A et al. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circulation Research. 2010;107(6):737-746
  44. 44. Serbulea V et al. Macrophage phenotype and bioenergetics are controlled by oxidized phospholipids identified in lean and obese adipose tissue. Proceedings of the National Academy of Sciences of the United States of America. 2018;115(27):E6254-E6263
  45. 45. Li C et al. Macrophage polarization and meta-inflammation. Translational Research. 2018;191:29-44
  46. 46. Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264(5164):1415-1421
  47. 47. Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: Enabling diversity with identity. Nature Reviews. Immunology. 2011;11(11):750-761
  48. 48. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nature Immunology. 2010;11(10):889-896
  49. 49. Kapoor N et al. Transcription factors STAT6 and KLF4 implement macrophage polarization via the dual catalytic powers of MCPIP. Journal of Immunology. 2015;194(12):6011-6023
  50. 50. Malyshev I, Malyshev Y. Current concept and update of the macrophage plasticity concept: Intracellular mechanisms of reprogramming and M3 macrophage “switch” phenotype. BioMed Research International. 2015;2015:341308
  51. 51. Odegaard JI et al. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447(7148):1116-1120
  52. 52. Thomas RS et al. Increased expression of IRF-5 in the adipose tissue in obesity: Implication in metabolic inflammation. The Journal of Immunology. 2016;196(1 Supplement):124.51
  53. 53. Gunthner R, Anders HJ. Interferon-regulatory factors determine macrophage phenotype polarization. Mediators of Inflammation. 2013;2013:731023
  54. 54. Dalmas E et al. Irf5 deficiency in macrophages promotes beneficial adipose tissue expansion and insulin sensitivity during obesity. Nature Medicine. 2015;21(6):610-618
  55. 55. Krausgruber T et al. IRF5 promotes inflammatory macrophage polarization and TH1–TH17 responses. Nature Immunology. 2011;12(3):231-238
  56. 56. El Chartouni C, Schwarzfischer L, Rehli M. Interleukin-4 induced interferon regulatory factor (Irf) 4 participates in the regulation of alternative macrophage priming. Immunobiology. 2010;215(9-10):821-825
  57. 57. Eguchi J et al. Interferon regulatory factor 4 regulates obesity-induced inflammation through regulation of adipose tissue macrophage polarization. Diabetes. 2013;62(10):3394-3403
  58. 58. Li C et al. IRF6 regulates alternative activation by suppressing PPARgamma in male murine macrophages. Endocrinology. 2017;158(9):2837-2847
  59. 59. Chaudhuri AA et al. MicroRNA-125b potentiates macrophage activation. Journal of Immunology. 2011;187(10):5062-5068
  60. 60. Martinez-Nunez RT, Louafi F, Sanchez-Elsner T. The interleukin 13 (IL-13) pathway in human macrophages is modulated by microRNA-155 via direct targeting of interleukin 13 receptor alpha1 (IL13Ralpha1). The Journal of Biological Chemistry. 2011;286(3):1786-1794
  61. 61. Thulin P et al. MicroRNA-9 regulates the expression of peroxisome proliferator-activated receptor delta in human monocytes during the inflammatory response. International Journal of Molecular Medicine. 2013;31(5):1003-1010
  62. 62. Ying H et al. MiR-127 modulates macrophage polarization and promotes lung inflammation and injury by activating the JNK pathway. Journal of Immunology. 2015;194(3):1239-1251
  63. 63. Sun Y et al. MicroRNA-124 mediates the cholinergic anti-inflammatory action through inhibiting the production of pro-inflammatory cytokines. Cell Research. 2013;23(11):1270-1283
  64. 64. Liu F et al. MiR-132 inhibits lipopolysaccharide-induced inflammation in alveolar macrophages by the cholinergic anti-inflammatory pathway. Experimental Lung Research. 2015;41(5):261-269
  65. 65. Taganov KD et al. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(33):12481-12486
  66. 66. Zhuang G et al. A novel regulator of macrophage activation: miR-223 in obesity-associated adipose tissue inflammation. Circulation. 2012;125(23):2892-2903
  67. 67. Fujisaka S et al. Adipose tissue hypoxia induces inflammatory M1 polarity of macrophages in an HIF-1alpha-dependent and HIF-1alpha-independent manner in obese mice. Diabetologia. 2013;56(6):1403-1412
  68. 68. Kim SY et al. PI3K/Akt contributes to increased expression of Toll-like receptor 4 in macrophages exposed to hypoxic stress. Biochemical and Biophysical Research Communications. 2012;419(3):466-471
  69. 69. Kim SY et al. Hypoxic stress up-regulates the expression of Toll-like receptor 4 in macrophages via hypoxia-inducible factor. Immunology. 2010;129(4):516-524
  70. 70. Liu W et al. Targeted genes and interacting proteins of hypoxia inducible factor-1. International Journal of Biochemistry and Molecular Biology. 2012;3(2):165-178
  71. 71. Choe SS et al. Macrophage HIF-2alpha ameliorates adipose tissue inflammation and insulin resistance in obesity. Diabetes. 2014;63(10):3359-3371
  72. 72. Castoldi A et al. The macrophage switch in obesity development. Frontiers in Immunology. 2015;6:637
  73. 73. Lee YS et al. Inflammation is necessary for long-term but not short-term high-fat diet-induced insulin resistance. Diabetes. 2011;60(10):2474-2483
  74. 74. Kennedy DJ et al. A CD36-dependent pathway enhances macrophage and adipose tissue inflammation and impairs insulin signalling. Cardiovascular Research. 2011;89(3):604-613
  75. 75. Nguyen MT et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. The Journal of Biological Chemistry. 2007;282(48):35279-35292
  76. 76. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annual Review of Physiology. 2010;72:219-246
  77. 77. Norseen J et al. Retinol-binding protein 4 inhibits insulin signaling in adipocytes by inducing proinflammatory cytokines in macrophages through a c-Jun N-terminal kinase- and toll-like receptor 4-dependent and retinol-independent mechanism. Molecular and Cellular Biology. 2012;32(10):2010-2019
  78. 78. Chen L et al. Mechanisms linking inflammation to insulin resistance. International Journal of Endocrinology. 2015;2015:508409
  79. 79. Hotamisligil GS. Inflammatory pathways and insulin action. International Journal of Obesity and Related Metabolic Disorders. 2003;27(Suppl 3):S53-S55
  80. 80. Nieto-Vazquez I et al. Insulin resistance associated to obesity: The link TNF-alpha. Archives of Physiology and Biochemistry. 2008;114(3):183-194
  81. 81. El-Haggar SM, Mostafa TM. Adipokines and biochemical changes in Egyptian obese subjects: Possible variation with sex and degree of obesity. Endocrine. 2015;48(3):878-885
  82. 82. Bastard JP et al. Elevated levels of interleukin 6 are reduced in serum and subcutaneous adipose tissue of obese women after weight loss. The Journal of Clinical Endocrinology and Metabolism. 2000;85(9):3338-3342
  83. 83. Lauterbach MA, Wunderlich FT. Macrophage function in obesity-induced inflammation and insulin resistance. Pflügers Archiv. 2017;469(3-4):385-396
  84. 84. West AP et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011;472(7344):476-480
  85. 85. Weichhart T, Hengstschlager M, Linke M. Regulation of innate immune cell function by mTOR. Nature Reviews Immunology. 2015;15(10):599-614
  86. 86. Jha AK et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. 2015;42(3):419-430
  87. 87. Cordes T et al. Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. The Journal of Biological Chemistry. 2016;291(27):14274-14284
  88. 88. Lampropoulou V et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metabolism. 2016;24(1):158-166
  89. 89. Tannahill GM et al. Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha. Nature. 2013;496(7444):238-242
  90. 90. Grant RW, Boudreaux JI, Stephens JM. 2-deoxyglucose inhibits induction of chemokine expression in 3T3-L1 adipocytes and adipose tissue explants. Obesity (Silver Spring). 2017;25(1):76-84
  91. 91. Geelhaar-Karsch A et al. Evaluation of arginine metabolism for the analysis of M1/M2 macrophage activation in human clinical specimens. Inflammation Research. 2013;62(9):865-869
  92. 92. Boutens L et al. Unique metabolic activation of adipose tissue macrophages in obesity promotes inflammatory responses. Diabetologia. 2018;61(4):942-953
  93. 93. van Rooijen N, van Kesteren-Hendrikx E. “In vivo” depletion of macrophages by liposome-mediated “suicide”. Methods in Enzymology. 2003;373:3-16
  94. 94. Patsouris D et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metabolism. 2008;8(4):301-309
  95. 95. Feng B et al. Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS One. 2011;6(9):e24358
  96. 96. Bu L et al. Intraperitoneal injection of clodronate liposomes eliminates visceral adipose macrophages and blocks high-fat diet-induced weight gain and development of insulin resistance. The AAPS Journal. 2013;15(4):1001-1011
  97. 97. Tang J et al. Inhibiting macrophage proliferation suppresses atherosclerotic plaque inflammation. Science Advances. 2015;1(3)
  98. 98. Peterson KR et al. Macrophage-targeted therapeutics for metabolic disease. Trends in Pharmacological Sciences. 2018;39(6):536-546
  99. 99. Dasu MR et al. Pioglitazone inhibits Toll-like receptor expression and activity in human monocytes and db/db mice. Endocrinology. 2009;150(8):3457-3464
  100. 100. Stafylas PC, Sarafidis PA, Lasaridis AN. The controversial effects of thiazolidinediones on cardiovascular morbidity and mortality. International Journal of Cardiology. 2009;131(3):298-304
  101. 101. Ying W et al. Interferon tau alleviates obesity-induced adipose tissue inflammation and insulin resistance by regulating macrophage polarization. PLoS One. 2014;9(6):e98835
  102. 102. Kohl A et al. Increased interleukin-10 but unchanged insulin sensitivity after 4 weeks of (1, 3)(1, 6)-beta-glycan consumption in overweight humans. Nutrition Research. 2009;29(4):248-254
  103. 103. Fatima N et al. Particulate beta-glucan induces early and late phagosomal maturation in murine macrophages. Frontiers in Bioscience (Elite Edition). 2017;9:129-140
  104. 104. Kim J et al. Silencing CCR2 in macrophages alleviates adipose tissue inflammation and the associated metabolic syndrome in dietary obese mice. Molecular Therapy-Nucleic Acids. 2016;5:e280
  105. 105. Rodriguez-Prados JC et al. Substrate fate in activated macrophages: A comparison between innate, classic, and alternative activation. Journal of Immunology. 2010;185(1):605-614
  106. 106. Freemerman AJ et al. Metabolic reprogramming of macrophages: Glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. The Journal of Biological Chemistry. 2014;289(11):7884-7896
  107. 107. Johnson AR et al. Metabolic reprogramming through fatty acid transport protein 1 (FATP1) regulates macrophage inflammatory potential and adipose inflammation. Molecular Metabolism. 2016;5(7):506-526
  108. 108. Xu J et al. NOTCH reprograms mitochondrial metabolism for proinflammatory macrophage activation. The Journal of Clinical Investigation. 2015;125(4):1579-1590
  109. 109. Haschemi A et al. The sedoheptulose kinase CARKL directs macrophage polarization through control of glucose metabolism. Cell Metabolism. 2012;15(6):813-826
  110. 110. Byles V et al. The TSC-mTOR pathway regulates macrophage polarization. Nature Communications. 2013;4:2834
  111. 111. Van den Bossche J et al. Mitochondrial dysfunction prevents repolarization of inflammatory macrophages. Cell Reports. 2016;17(3):684-696
  112. 112. Ip WKE et al. Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science. 2017;356(6337):513-519

Written By

Joseph Roberts, Padraic G. Fallon and Emily Hams

Submitted: 15 February 2019 Reviewed: 23 April 2019 Published: 09 July 2019