GET THE APP

Polarization of Macrophages in Metabolic Diseases
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2015) Volume 0, Issue 0

Polarization of Macrophages in Metabolic Diseases

Lise Høj Thomsen1 and Alexander Rosendahl1,2*
1Diabetes Complications Biology and Pharmacology, Novo Nordisk A/S, Måløv, Denmark
2Biopharmaceuticals New Haemophilia, Novo Nordisk A/S, Gentofte, Denmark
*Corresponding Author: Alexander Rosendahl, Novo Nordisk A/S, Biopharmaceuticals New Haemophilia, Brogårdsvej 66, DK-2820 Gentofte, Denmark, Tel: +45-30750637 Email:

Abstract

Monocytes originate from progenitor cells in the bone marrow and traffic via the bloodstream to peripheral tissues. During both homeostasis and in responses to clear pathogens, circulating monocytes leave the bloodstream and migrate into tissues where they, following exposure to local growth factors, pro-inflammatory cytokines and microbial products, differentiate into macrophage or dendritic cell sub-populations. Type 1 diabetes (T1D) is a disease characterised by the elimination of the insulin producing β-cells in the pancreatic tissue through activation mainly of the adaptive immune system. In metabolic diseases, i.e. obesity, Type 2 diabetes (T2D) and diabetic complications, inflammation is mostly driven by macrophages and has been shown pivotal for disease progression even when blood glucose levels are well controlled. This review describes some current ideas and trends regarding the monocyte and macrophage involvement and their polarization in metabolic diseases that might open up novel therapeutic areas for the growing diabetic population.

Keywords: Macrophages, Polarization, Metabolic disease, Diabetes, Obesity

Abbreviations

T1D: Type 1 Diabetes; T2D: Type 2 Diabetes; DC: Dendritic Cell; SFA: Saturated Fatty Acids; TLRs: Toll-Like Receptors; ECM: Extracellular Matrix

Origin of Monocytes and Macrophages and Tissue Migration

Monocytes are a cell population that belongs to the innate immune defence which is capable to differentiate into tissue macrophages and dendritic cells (DCs) depending on tissue, local cytokine and growth factor signature and pathogen [1]. Monocytes are developed from myeloid progenitor cells in the bone marrow where they after initial maturation in well-defined steps enter the peripheral blood stream in a controlled manner and move into peripheral tissues as part of homeostasis or inflammation [2]. Monocytes express a unique combination of sensing molecules e.g. pattern recognition receptors (PRRs) including scavenger receptors, Toll-like receptors (TLRs) and cytokine receptors that allow them to monitor the tissue microenvironments and to act appropriately on local infections and tissue damage [3,4]. The ability of monocytes to mobilize and traffic rapidly and specifically is essential to mount the appropriate inflammatory response in response to infections and tissue injury [3,5]. When inappropriately recruited to tissues, activated and polarized into effector monocytes and macrophages, they contribute to and drive initiation and progression of many inflammatory diseases. Once migrated into the tissue, the monocytes can either remain monocytelike or undergo maturation into tissue macrophages [6]. Monocytes are guided towards the site of interest by specific tissue and insult specific induced chemokine gradients and adherence molecules [7]. CCL2 (MCP-1) is the chemokine most well described to participate in metabolic diseases like obesity and diabetes upon interaction with CCR2b expressed mainly by monocytes, macrophages and DCs [8,9]. Other chemokines like MIP-1β (shown important in atherosclerosis), CCL3 (shown to promote microvascular leakage) and MCP3 (shown to promote renal tubulointerstitial fibrosis) has been suggested to contribute to monocyte migration during metabolic diseases [10-13]. Tissue migration is regulated by specific interaction between several integrins and other adhesion molecules expressed on the monocyte and on the vasculature [14]. As example, monocytes express among others Lselectin (CD62L), P-selectin glycoprotein ligand 1 (PSGL1), LFA1 (αLβ2 integrin), MAC1 (αMβ2 integrin), PECAM1 and VLA4 (α4β1 integrin). In metabolic diseases, monocytes interacts with Eselectin and Pselectin expressed on the venule endothelium through PSGL1, with PNAd on endothelial cells and, lymph node high endothelial venules (HEV) through Lselectin and through and with Eselectin and Pselectin expressed on inflamed aortic endothelium in atherosclerosis through VCAM1 interaction [15-17].

Tissue Monocyte Maturation and Polarization in Metabolic Disease

Maturation and polarization of monocytes and macrophages is tissue dependent and regulated by the local cytokine and growth factor signature [18]. Mononuclear phagocytes migrate from the yolk sac during the first stages of embryogenesis to the CNS where that later differentiate to permanently residing microglia cells [19]. In addition to CNS migration, these mononuclear phagocytes also populate the kidney where they mature into kidney resident monocytes [19]. These kidney resident monocytes expand during chronic kidney disease and produce a unique cytokine signature, compared to the inflammation attracted monocytes and function mainly to provide signals regulating organ growth and repair during physiological stress [20,21]. However, aberrant activity of these kidney resident cells has been considered to participate in the extensive fibrotic activity present in chronic metabolic disorders. This hypothesis was supported in mice depleted of these kidney resident monocytes showing significantly decreased collagen matrix deposition [20]. The kidney resident macrophages present during metabolic diseases interact with and activate the kidney fibroblasts in a paracrine manner allowing the fibroblasts to produce enhanced levels of matrix molecules showing a novel signature [22]. In obesity, excessive recruitment of monocytes and macrophages into the adipose tissue and local activation through the increased levels of saturated fatty acids (SFAs) promotes adipose tissue inflammation which is hypothesized to contribute to insulin resistance through a TLR4 dependent interaction (Figure 1) [23-25].

clinical-cellular-immunology-adipose-tissue

Figure 1: Classically activated M1-like macrophages in adipose tissue. Pro-inflammatory macrophages contribute to insulin resistance by feed forward mechanisms of inflammatory cytokines such as TNF and IL-1β produced by the NLRP3 inflammasome. TLRs are activated by ligands like LPS and SFA and initiate intracellular signalling pathways leading to further induction of pro-inflammatory genes and cytokines. Futhermore, signalling through the mineralcorticoid receptor (MR) contributes to M1-polarization by production of pro-inflammatory cytokine production and inhibition of M2-polarization.

The now TLR4-activated adipose tissue macrophage produces significant levels of cytokines which fuels the low-grade inflammation present in metabolic diseases [26]. SFA activation of the TLR4 on monocytes induce an inflammatory signalling cascades mediated by IRF3, JNK and NF-κB which among other cytokines leads to production of pro-inflammatory cytokines such as TNF-α [27,28]. IL1β is produced in adipose macrophages upon activation of the NOD, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome which has a ligand-recognition domain for other receptors, e.g. TLRs [29]. In addition to inducing inflammation, the inflammasome activates caspase-1 thereby inducing cell pyroptosis, a process of programmed cell death distinct from apoptosis [30]. In vivo , adipose tissue macrophages from obese and insulin resistant mice and humans express high levels of TNF-α, IL-6, and IL-1β which reduces the insulin sensitivity in adipocytes in vivo [31,32]. During obesity, these pro-inflammatory cytokines create feedback loops between the inflamed adipocytes and these M1-like macrophages. This include CD5like antigen (CD5L) release by the macrophages through CD36mediated endocytosis and lipolysis upregulated production of CCL2, osteopontin (OPN) and inflammasome dependent cytokines which leads to further recruitment and activation of macrophages [33-35]. However pre-clinical observations in MyD88 deficient mice show a more severe metabolic disease e.g. insulin resistance in response to a high-fat diet (HFD) than wild-type control mice and TLR4 deficient mice on the C57BL/10 genetic background exacerbated diet-induced obesity, steatosis and insulin resistance suggesting that additional signalling pathways to the TLR4 participates in the disease development [36,37]. Indeed, in addition to the inflammasome, engagement of the mineralocorticoid receptor (MR) signalling contributes to M1-like polarization / activation and pro-inflammatory cytokine production of adipose tissue macrophages primarily by inhibiting the M2 polarization in macrophages [38]. Regardless initial activation mechanisms in the adipose tissue, these adipose tissue macrophage derived cytokines leak into the circulation leading to low- grade systemic inflammation and contributing to insulin resistance in other peripheral tissues.

Macrophage Polarization

Macrophages are classically divided into three major sub-groups based on their polarization status: 1) the classically activated / proinflammatory M1 macrophages or 2) the alternatively activated M2 macrophages and 3) the un-polarized M0 macrophages. The macrophage is placed into a specific sub-population based on a combination of the expression of chemokine receptors, specific adhesion / activation molecule expression and production of specific cytokines [39,40]. In vitro studies has provided instrumental information about the molecular patterns contributing to the polarization of the macrophages [41]. Although helpful, caution should be taken when evaluating the macrophages in vivo and in humans according to this in vitro categorization. The data available in vivo and in humans of the macrophage M1/M2 signatures is still emerging and far from clear. Furthermore, an array of evidence exists that macrophages, particularly in vivo , show polarization plasticity and hence are able to continuously re-polarize depending on the local milieu [42,43]. Therefore, we suggest that when evaluating in vivo and human macrophages the term M1-like and M2-like macrophages should be used indicating that these cells display several features associated with the M1 and M2 macrophages, but emphasize the caution that should be taken.

M1 polarized macrophages

Classical activation and polarization of monocytes and macrophages into M1-like macrophages requires an initial priming from IFN-γ through the IFN-γ receptor which is followed by the second signal in form of TNF-α or an inducer of TNF-α [44]. TNF-α can act alone but often occurs in concert with exposure to microbial product such as LPS [45]. This generates a M1 macrophage with a high antigen-presenting capacity, high potency to produce proinflammatory cytokines (e.g. IL-1, TNF-α, IL-6), perform extracellular matrix (ECM) destruction by production of tissue degrading proteolytic enzymes such as lysozymes, neutral proteases (e.g. collagenase, gelatinase, elastase, matrix metalloproteinases MMP2 and MMP9 and plasminogen activator) and acid hydrolases (e.g. lipases, cathepsins, proteases, phosphatases, glycosidases and ribonucleases) (Table 1) [46]. These M1 macrophages secrete an array of chemokines including IL-8/CXCL8, CXCL10, CCL3, CCL4 and CCL5. These create a gradient towards which neutrophils, natural killer cells and activated T-cells are recruited to the local tissue [47,48]. In addition, the M1 macrophages release pro-inflammatory cytokines such as IL-1β, IL-12, IL-6, and TNF-α which participate directly in several metabolic processes like induction of β-cell death, insulin resistance, liver steatosis and vascular smooth muscle cell (VSMC) apoptosis [49-51]. Further, the M1-like macrophage derived proteolytic enzymes MMP-1, -2, -7, -9, and -12 participate in the degradation of collagens, elastin, fibronectins [52,53]. Together, this may lead to hyperglycaemia, diabetes progression, plaque rupture causing myocardial infarction and diabetic nephropathy [54].

  M1 M2a M2b M2c
Stimulation/activation IFN-γ IL4/IL13 ICs IL-10
TNF-α   LPS TGF-β
LPS   TLRinteractions GCs
Expression CD86 CD163 CD86 CD163
CD80 SR    
IL-1R 1 CD206    
TLR2 IL-1R ii    
TLR4 Mouse only:    
iNOS Ym1    
MR Fizz1    
  Arg-1    
Cytokines TNF-α IL-10 IL-1 IL-10
TNFSF1A TGF-β IL-10 TGF-β
IL-1β IL-1ra    
IL-6      
IL-8      
IL-12      
IL-23      
Chemokines when activated CCL10 CCL18    
CCL9 CCL22    
CCL5 CCL24    
CCL4      
CCL3      
CCL2      
CXCL 10      
Enzymes Tissue degrading collagenase Tissue Remodelling and repair    
Matrix Proteinases (MMP2, MMP9, Plasminogen activator) TIMP1    
  PDGF    

Table 1: M1 and M2 subgroups. Macrophages are subdivided into the M1 classically activated and the M2 alternative activated subsets. M1 macrophages are stimulated by INF-γ, TNF-α and LPS which leads to cytokine production of TNF-α, IL-1β, IL-6, IL-8, IL-12, IL-23, CCL2, CCL3, CCL4, CCL5, CCL9 and CCL10. The M1 markers expressed include CD86, CD80, TLR2 and 4 along with iNOS and stimulate tissue degradation by secretion of enzymes such as collagenases and matrixproteinases [135]. In contrast, M2 macrophages express the scavenger receptors (SR), mannose receptor CD206 and CD163 and produce IL-10, IL1ra, CCL18, CCL22 and CCL24 and are involved in tissue remodelling and repair by secretion of TIMP1 and PDGF. The M1 subset is inhibited by IL-4 and IL-10 while the M2 subset is inhibited by IFN- γ [135].

M2 polarized macrophages

The term alternative activated or M2 macrophage is a broad definition of the macrophages that contain all the polarized macrophages not classically M1 polarized. Polarization towards M2 macrophages are dependent on interaction with the classically Th2 associated cytokines such as IL-4, IL-13, IL-10, TGF-β and is also influenced of by interaction with galectin-3 but inhibited by IFN-γ [55]. Upon polarization, the M2-like macrophage can perform pinocytosis of soluble antigen [56-58]. Once processed, these soluble antigens are loaded onto MHC class II molecules in concert with the upregulation of CD80 and CD86 on the M2-like macrophage which subsequently result in M2-macrophage interactions with the adjacent T-cells [59]. The major cytokines and growth factors released by the activated M2 macrophages are IL-10 and TGF-β, both which are associated with resolution of inflammation or often termed antiinflammatory functions to distinguish them from the highly proinflammatory responses obtained with M1-like macrophages. The major biological functions associated with this subset include promoting differentiation of T-cells towards Th2 cells, immuneregulation that limits several classical acute responses, tissue remodelling and repair by promoting ECM deposit by producing MMPs including TIMP1 and TGF-β, PDGF (Table 1) [60].

The M2 macrophages can be further sub-divided into three major subgroups that share some similar features but are distinct in other. These include: 1) the M2a macrophages which are generated and activated by IL-4 and IL-13, 2) the M2b macrophages which are generated and activated by immune complexes or TLR interactions and 3) the M2c macrophages which are generated and activated by IL-10 [61]. These participate specifically in various pathological conditions such as cardiovascular and metabolic disorders such as atherosclerosis and obesity [62]. Monocytes found in adipose tissue from healthy subjects show an M2-like macrophage signature whereas obesity leads to a shift to an M1-like pro-inflammatory state contributing to insulin resistance [63].

Major Macrophage M1 and M2 Populations in the Mouse and in Humans

Macrophages express several general markers, such as CD14, CD16, CD64, F4/80, CD68, CD11b and Ly6C in a heterogeneous fashion which only partially overlap between man and mouse [62,64,65]. Further, the combination of these markers to various other markers like chemokine receptors allow a sub-categorization of macrophages that is classically used to identify M1 and M2 like cells.

Murine M1-like monocytes

In mice, monocytes are identified and divided into three major subpopulations based on the combination of the surface expression of Ly6C, CD43, CD11b and CCR2 [66]. The Ly6Chigh monocytes represent approximately 60% of circulating monocytes in a mouse, often co-express CD11b and are rapidly recruited to the site of infection / inflammation (Table 2). These cells has been shown to primarily produce and secrete pro-inflammatory cytokines and hence could be considered M1-like monocytes [67]. The Ly6ChighCD11b monocyte expresses high levels of CCR2 and low levels of CX3CR1. In CCR2 deficient mice a markedly reduced Ly6Chigh monocyte trafficking to the site of inflammation occurs suggesting that the CCR2b-CCL2 axis is essential for M1-like Ly6Chigh monocyte migration during metabolic conditions [68]. These Ly6Chigh monocytes accumulate in the diabetic kidney where they participate in progression of disease [69].

    Human Mouse
  Name Classically activated macrophages Ly6chigh
Classically Activated Function Phagocytic and microbial activity Produce pro-inflammatory cytokines such as TNF-α
Low pro-inflammatory cytokine production (IL-10, TNF-α, IL-12 etc.)  
Main IL-10 Producer when stimulated with S. aureus  
Production of ROS  
Frequency 90-95% of circulating monocytes 60% of circulating monocytes
Markers CD14high Ly6chigh
CD16neg CD11b+
Low levels of MHC-II, ICAM-1  
CD86, CD64  
Intermediate levels of CD11b  
Cytokine Receptors CCR2high, CX3CR1low CCR2high, CX3CR1low
Intermediate Name Intermediate  
Function Pro-inflammatory: Actively produce TNF-α (in response to LPS), IL-1β, IL-6 and IL-10  
Phagocytic  
Frequency Minor sub-population  
Markers CD14high, CD16int  
High levels of MHC-II, ICAM-1, CD86, CD11b  
Intermediate levels of CD163, CD64 and CD11c  
Cytokine Receptors CCR2low, CX3CR1high  
Alternatively activated Name Alternatively activated Alternatively activated, Ly6Clow
Function Anti-inflammatory: Constitutively produce IL-1RA  
Frequency 5-10% of circulating monocytes 40% of circulating monocytes
Markers CD14low, CD16high Ly6Clow
High levels of CD86 and CD11c  
Intermediate levels of MHC-II  
ICAM-1  
Low levels of CD11b, CD64 and CD163  
Cytokine Receptors CCR2low, CX3CR1high CCR2low, CX3CR1high

Table 2: Main monocyte populations in human and mouse. Mouse monocytes are divided into classically activated M1 monocytes and alternatively activated M2 monocytes. M1 monocytes express high levels of Ly6C and low levels of CX3CR1 and are known for producing proinflammatory cytokines such as TNF-α. M2 monocytes express low levels of Ly6C and high levels of CX3CR. Human monocytes are divided into three subgroups, where the classical group is called CD14highCD16neg. They express high levels of CCR2 but lack CX3CR1 surface expression. Using the mouse homology they are M1-like but functionally they resemble M0-like monocytes. The intermediate CD14highCD16int monocytes

Murine M2-like monocytes

In absence of inflammation, the Ly6Chigh monocytes return to the bone marrow where they may re-differentiate into Ly6Clow monocytes These Ly6Clow monocytes, are characterized by high surface expression levels of CX3CR1 and low levels of CCR2 (Table 2) [40].

They are less prevalent than the Ly6Chigh monocytes and account for approximately 40% of the circulating monocytes in a healthy mouse. They adhere to and migrate along the luminal surface of endothelial cells particularly in small blood vessels independent of the blood flow direction performing immune surveillance of endothelial cells and surrounding tissues [70].

Human M1/2-like CD14highCD16 neg monocytes

Classical monocytes (CD14highCD16neg) are the most prevalent monocyte subset and constitute ~90% of the monocytes in human peripheral blood (Table 2). Similarly to the Ly6Chigh monocytes in mice, the CD14highCD16neg monocytes express high levels of CCR2 but lack CX3CR1 surface expression [40,71]. These cells express low levels of MHC-II, ICAM-1, CD86, CD64 and intermediate levels of CD11b [72]. Functionally, these cells produce low levels of IL-10 and TNF-α in response to LPS, IFN-γ and IL-12 in co-culture with T cells stimulated with purified protein derivative of tuberculin, but are the main monocyte population producing IL-10 in response to Staphylococcus aureus challenge [72]. The CD14highCD16neg cells are shown to be highly phagocytic of both S. aureus and E. coli bacteria that result in strong production of reactive oxygen species (ROS) [72]. Hence, these monocytes based on chemokine expression are M1-like using the mouse homology, but on functional level more resemble M0-like monocytes. CD14highCD16neg numbers are positively correlated with cardiovascular events independent of diabetes, but do not correlate to atherosclerosis [73]. Although contradictory, the CD14highCD16neg monocytes are considered to participate in the inflammatory response that weakens the fibrotic cap while other monocyte populations might play a more determinative role in the build-up of the plaque size and might in fact provide protection against rupture [73].

Human M1/M2 CD14high/lowCD16int/high monocytes

The remaining 10% of the monocytes in the peripheral blood are further divided into two subsets, the CD14highCD16int intermediate and the CD14lowCD16high non-classical monocytes (Table 2) (66). The intermediate CD14highCD16int monocytes express low levels of CCR2 and high levels of CX3CR1 receptors. These monocytes express very high levels of MHC-II, ICAM-1, CD86, CD11b and intermediate levels of CD163, CD64 and CD11c [72]. Functionally, they are highly responsive to LPS measured as significant production of proinflammatory cytokines such as TNF-α, IL-1β and IL-6, but also produce pronounced levels of IL-10. CD14highCD16int monocytes are phagocytic but do not produce high levels of ROS [72,74]. Thus, these resemble the mouse M2-like monocytes in mice on chemokine receptor expression level while functionally behaves as classically proinflammatory M1-like monocytes. CD14highCD16int monocytes are positively strongly associated with cardiovascular events in chronic kidney subjects and with obesity in diabetic subjects [75,76].

The non-classical CD14lowCD16high monocytes express low level of CCR2 and high level of CX3CR1 chemokine receptors. These cells express high levels of CD86 and CD11c, intermediate levels of MHCII, ICAM-1 while low levels of CD11b, CD64 and CD163 [72]. They do not produce IL-1β, IL-8, IL-6 but produce low-intermediate levels of TNF-α and are the main producer of the regulatory cytokine IL-1RA associated with the recovery phase [72]. Hence, both on the chemokine receptor expression signature as well as by the functional responses these monocytes resemble the mouse M2-like monocytes. The CD14lowCD16high monocytes are considered to be the patrolling monocyte population classically observed to perform endothelium crawling in a LFA-1 dependent manner [77]. CD14lowCD16high monocytes has been positively correlated to BMI and atherogenic lipoproteins while negatively correlated with high-density lipoprotein cholesterol implicating a role in atherogenesis as well as with obesity in diabetic subjects [75,76].

In conclusion, similar features but also several discrepancies exists between the murine and human monocyte/macrophage populations with respect to expression of chemokine receptors linked to biological function [78,79]. As several markers used to define the M1 and M2 monocytes are differently expressed between the mouse and humans there is an urgent need to identify strong and reliable human diagnostic and functionally associated human M1 and M2 markers to properly address the pathogenesis and resolution of diseases to better adjust future therapeutic approaches [80].

Diabetes and Obesity

Globally 1.45 billion of the total adult population was estimated to be overweight (body mass index, BMI>25 kg/m2), of which ~500 million were considered obese (BMI>30 kg/m2) in 2008 [81]. Of importance, the number of overweight subjects currently is demonstrated to be ~525 million higher than the number of malnourished subjects on a global level [82]. As a consequence of the rise in worldwide obesity a paralleled epidemic increase of obesityrelated health problems, such as insulin resistance, T2D, coronary artery disease, fatty liver disease, specific cancers and degenerative diseases has been recorded [83-85]. Overweight and obesity result in decreased insulin sensitivity which reduces the capacity in somatic cells to take up glucose from the blood that finally leads to hyperglycaemia. The β-cells in the islets of Langerhans sense the hyperglycaemia as insufficient amounts of insulin have been produced and respond by sending signals to increase the insulin production and secretion. Eventually, this over-production will result in β-cell exhaustion followed by production failure and finally to apoptosis of the β-cell. The consequence will thereafter be significantly reduced production that will lead to development of diabetes [77].

More than 380 million people are estimated to have diabetes (T1D and T2D) on a global level and the number newly diagnosed patients show a steady increase [86]. T1D is an autoimmune disease where a lymphocyte driven destruction the β-cells leads to the reduced capacity to produce insulin [77]. Clinically this leads to a requirement of exogenous treatment with insulin to properly metabolise glucose to ensure energy balance in the cells [87]. Of interest, etiological investigations has suggested that absence of certain bacterial infections and hence the appropriate activation of the corresponding monocyte populations in modern society during early life and hence improper activation of the monocytes later in life may play a biological role as a novel risk factor leaving the immune system to improper alert [87].

More than 90% of all clinically diagnosed diabetic cases are T2D patients who also show a heritable linkage but here to obesity [88]. Recent evidence has strongly suggested that inflammation is involved in both obesity and diabetes.

Inflammation in Diabetes and Obesity

T2D has long been considered purely a metabolic disease, but the recently described low-grade inflammation and activation of the inflammasome has gained support for inflammation in the disease initiation and progression. Low grade chronic inflammation is primarily mediated by the interplay between innate and adaptive immune cells, and is seen as a key pathogenic link between obesity the metabolic diseases arising with obesity [26,89]. Low-grade inflammation is triggered by a nutrient and metabolic surplus, and shares many similarities to classical M1 inflammation with respect to the molecular and signalling signatures involved [26]. These include among others activation of PRRs, activation of the endothelium, upregulation of acute reaction proteins such as C-reactive protein, cytokines, infiltration of leukocytes which classically include M1-like macrophages and induction of TNF-α and IL-1β in these monocytes/ macrophages [90]. A strong positive correlation exists between the degree of obesity and the number of M1-like macrophages immigrating into the adipose tissue in overweight and obese subjects while it not as clear in T2D subjects if the corresponding monocyte/ macrophage population is M1 or M2 polarized or if that is dependent on other so far unknown patient specific risk factors [91,92]. In advanced T2D, the β-cell function is significantly reduced measured as diminished release of insulin and with time, the β-cell mass is reduced [93,94]. Hence, the remaining few β-cells are now functionally unable to compensate for decreased insulin sensitivity. Thus, there are common features in obesity and T2D, such as insulin tolerance, but also clear differences, such as β-cell dysfunction, cell death, and hyperglycaemia. The immunological signature in early diabetes is characterized by an early infiltration of M1 pro-inflammatory macrophages secreting pro-inflammatory cytokines such as TNF-α, IL-β and IL-6 [95]. However, chronic T2D show TGF-β produced from the alternatively activated macrophages for tissue remodelling in combination with a systemic low grade inflammation with an M1/M2 macrophage population contributing to further tissue remodelling by either secreting degrading proteolytic enzymes or by stimulating matrix deposit by secreting inhibitors of the proteolytic enzymes [95].

Involvement of Macrophages in Type 1 Diabetes

Exposure of β-cells to pro-inflammatory cytokines such as IFN-γ, IL-2 and TNF-α during the insulitis (when the mononuclear leukocytes accumulates within and around the islets) leads to upregulation of chemokines such as IL-8 and CCL5 by the islet cells which further enhanced inflammation by further recruitment of mononuclear cells to the islets (Figure 2) [96,97].

clinical-cellular-immunology-inflammatory-cells

Figure 2: Macrophage infiltration in T1D islets. Macrophages and other inflammatory cells are recruited to the islets of Langerhans in the pancreas by secreted cytokines such as CCL2. Within the islets, the macrophages secrete pro-inflammatory cytokines such as IL-1β and TNF- α. These secreted cytokines together with IFN-γ results in β-cell apoptosis. Together with islet associated DCs, macrophages activate islets auto-reactive T-cells and influence their polarization. These activated T-cells cells further enhance recruitment of additional macrophages through a positive feed-back loop which ultimately leads to development of T1D.

IL-12 secreted from islet DCs and macrophages activates the T-cell polarization which leads to IFN-γ production that contributes to the polarization of the islet macrophages towards M1-like phenotype that enhance the production of pro-inflammatory cytokines including TNF and IL-1β [96,97]. The crosstalk between T-cells and macrophages mediate stress on the β-cells and the secreted cytokines, IFN-γ, IL-1β and TNF also induce the expression of ROS including nitric oxide by β-cell. ROS mediates apoptosis [49]. The importance of proinflammatory macrophages in T1D was demonstrated by clodronate depletion of macrophages in diabetogenic CD4+ T-cells in NOD mice that inhibit activation and differentiation of monocytes into effector macrophage by reducing the macrophage islet β-cell lytic capacity and prevents severe diabetes development [98]. Entry of the macrophages into the T1D islets is dependent on complement receptor 3 interactions as complement 3 antibody inhibition markedly delay disease development [99]. Direct in vivo support for macrophage involvement in the initiation of T1D was obtained in RIP-CCL2 transgenic mice bred onto a Rag-1/background which result in mice with CCL2 producing β-cells which lead to a potent recruitment of monocytes to the pancreatic islets resulting in a macrophage dependent destruction of β-cells in absence of mature T- and B-cells [97]. Islet derived monocytes from T1D diabetics show an inflammatory phenotype and secrete high levels of IL-6 and IL-1β known to promote Th17 polarization [100,101]. With the positive correlation between HbA1c and IL-17 secretion in T1D the relevance of macrophages to promote T1D might in addition to provide direct elimination of the β-cells also be to support development of the diabetogenic effector Th17 cells [102].

Macrophages in Obesity and Insulin Resistance

Close to twenty years ago the first firm link between inflammation, obesity and insulin resistance was demonstrated when adipose tissue of obese animals was found to express TNF-α which was suggested to promot insulin resistance via serine phosphorylation of insulin receptor substrate 1 (IRS1) [103]. The origin of the adipose TNF-α was subsequently demonstrated to be due to obesity dependent infiltrating macrophages [91,104]. In lean mice, 10-15% of the adipose tissue cells are F4/80+ macrophages, while 45-60% F4/80+ macrophages are present in obese mice [91]. Hence, obesity substantially alters the ratio of macrophages to adipocytes in the adipose tissue [91]. In addition to the increased numbers, the adipose tissue macrophages in obese animals exhibit distinct cellular localization and inflammatory activity compared to lean mice [105]. Lean adipose tissue macrophages phenotypically resemble the alternatively activated M2-like macrophages with elevated expression of the mannose receptor (CD206), arginase-1 and IL-10 and show a uniform distribution within the tissue micro-environment [105-107]. In contrast, adipose tissue macrophages in obese mice show a pro-inflammatory, classical M1-like phenotype with elevated expression of NOS2, TNF-α and CD11c [45,107]. This shift in the ratio between M1-like and M2-like adipose macrophages is strongly associated with the development of insulin resistance [108]. The M2-like macrophages found in lean adipose tissue provides essential signals to maintaining the insulin sensitivity of adipocytes via the secretion of interleukin-10 which potentiates insulin signalling in the adipocytes [63,105]. In contrast, M1-like macrophages in obese adipose tissue secrete proinflammatory cytokines such as IL-1β, TNF-α which induce insulin resistance in the adipocytes via an IKKβ- and JNK- mediated inhibitory serine phosphorylation of IRS proteins [109,110]. Further, the M1-like adipose macrophages in obesity express CD11c and form so-called crown-like structures by encircle necrotic adipocytes [111]. These M1-like macrophages phagocytize lipids that are released by the necrotic adipocytes and differentiate into lipid-engorged, multinucleated giant macrophage cells similarly as the proinflammatory M1-like macrophages found in atherosclerotic plaques [112,113].

On a mechanistical level, four major paths of evidence support that activated M1-like macrophages contribute to the pathogenesis of obesity-induced insulin resistance (Figure 1): 1) Infiltration of M1-like macrophages in adipose tissue is significantly reduced in CCR2 deficient mice which are protected from obesity-induced inflammation and insulin resistance [23], 2) the classical inflammation is significantly reduced upon CD11c+ M1-like depletion in the CD11cDTR mouse which enhances insulin action without significantly impacting diet-induced obesity [114], 3) M1-like macrophage cell mediated inflammation in adipose tissue is reduced in upon ablation of IKKβ in myeloid cells or reconstitution of mice with JNK-deficient bone marrow which protects against development of obesity-induced insulin resistance [115,116] and 4) M2-like inflammation is reduced and M1-like adipose tissue inflammation enhanced upon GPR120 deficiency, a G protein-coupled receptor mediating the anti-inflammatory actions of omega-3 USFA which significantly enhance insulin resistance [117].

Macrophage Involvement in Type 2 Diabetes

Based on the evidence for macrophage involvement in insulin resistance, recent focus has been to strengthen the linkage of macrophages locally within the diabetic islets in T2D (Figure 3). Initial histological observations of the diabetic islets from pre-clinical and human subjects with T2D demonstrated enhanced presence of macrophages but could not clearly identify the macrophage subpopulations [89,118]. In analogy to the adipose tissue in obesity, elevated levels of M1-like cytokines e.g. IL-6 is present in diabetic islets compared with healthy controls [119]. As the M1-like cytokines IL-6 and TNF-α induce ER stress in the β-cells and upon chronic exposure resulting in β-cell death the identification of the cytokine origin and detailed analysis of the islet macrophages is of high relevance [119].

clinical-cellular-immunology-subset-secretes

Figure 3: Macrophage infiltration in T2D. In early stage of T2D a predominately M1-like macrophage accumulation occurs in the islets of Langerhans. This M1-like subset secretes inflammatory cytokines such as IL-6, TNF-α and IL-1β leading to a low-grade inflammation within the islets. During disease development, the macrophage population switch phenotype towards an M2-like subset producing significantly enhanced levels of TGF-β and galectin-3 which is thought to participate in the β-cell apoptosis and tissue remodeling.

Islet associated macrophages in T2D diabetes

T2D diabetic islets display a 5-fold accumulation of proinflammatory, M1-like macrophages at the time when β-cells start to decline in number and lose their insulin-producing function i.e. “early T2D” (Figure 3) [95]. This coincides with an increased systemic M1- like macrophage population in the spleen fuelling the systemic lowgrade inflammation by enhancing the production of IL-6, TNF-α and IL-1β. In chronic T2D disease, when the β-cell mass is significantly reduced and the insulin production capacity per remaining β-cell is markedly reduced, the splenic macrophage population undergoes a repolarization toward an M2-like profile associated with high production of TGF-β [95]. On a detailed level, the islet-immigrating macrophage sub-population was a novel CD68+F4/80- sub-population not present in healthy non-diabetic islets whereas the islet resident macrophage population was CD68+F4/80+ [95]. Egushi et al., elegantly demonstrated that during palmitate challenge, the islet immigrating macrophages to be M1-like CD11b+Ly6Chigh cells producing significant levels of IL-1β and TNF-α [90]. Within the diabetic islets the vast majority of the resident CD68+F4/80+ macrophages expressed CD11b, while only 40% of the newly immigrant CD68+F4/80- macrophages were CD11b positive [95]. A similar frequency of Ly6Chigh macrophages were demonstrated in the CD68+ macrophages regardless expression of F4/80. This results in a 2-fold higher frequency of M1-like CD11b+Ly6C+ cells, as described by Egushi et al., in the CD68+F4/80- macrophages compared to the CD68+F4/80+ macrophages [90,95]. Thus, by this detailed analysis of the islet macrophages a highly plastic intra-islet migratory signature with recruitment of M1-like CD68+F4/80-CD11b+/- Ly6Chigh/low and expansion of CD68+F4/80+CD11b+ Ly6Chigh/low macrophages in response to diabetic stress was uncovered (90, 95).

Both healthy- and diabetic islets derived macrophages predominately express the M1-associated CD11chigh phenotype [120]. Detailed evaluation has demonstrated that the total number of CD68+F4/80- M1-like macrophages increase almost 10-fold in the diabetic islets [95]. In contrast, the resident CD68+F4/80+ macrophages in the diabetic islets lost their classical CD11chigh M1- like phenotype and did undergo a re-differentiation to become a mixed M1/M2 CD11chighCD206+ macrophage population.

In established T2D, many β-cells undergo apoptosis and galectin-3 is considered to participate in this process [121]. Interestingly, the diabetic islet macrophages show a markedly elevated surface expression of galectin-3 compared to healthy islets macrophages hence suggesting that in addition to produce islet apoptosis cytokines, apoptotic signals to the β-cells through galectin-3 interactions by the macrophages may contributes to disease progression.

Systemic mouse spleen macrophages in T2D

Onset of T2D show a strong M1-like systemic cytokine signature characterized by high levels of TNF-α, IL-6 and IL-1 while chronic T2D show a blunted M1-like cytokines production (Figure 3) [95]. Independent of age and T2D, the vast majority of the splenic macrophages are CD206+ M2-like and the total number of these M2- like macrophages increase as T2D progress rendering the splenic milieu more M2-like polarized in chronic disease compared to during the initiation [95]. Galectin-3 expressing macrophages are negatively associated with the capacity to produce M1-like cytokines and positively associated with the polarization of M2 in vitro [122,123]. A progressive systemic increase during T2D of CD68+F4/80- M2-like macrophages occurs which is considered to contribute to the reduced M1-like cytokine production and the induction of the M1/M2 mixed low grade cytokine signature [95]. Onset of diabetes show a clear M1- like macrophage phenotype and systemic low grade pro-inflammation signature while chronic diabetes resemble a complex M1/M2 towards M2-like disease with alternatively activated M2-like macrophages with a modulated low-grade pro-inflammatory signature and enhanced remodelling capacity.

Diabetic Complications

Chronic hyperglycaemia or frequent fluctuation of glucose levels result in tissue modification which ultimately leads to altered biological function termed diabetic complications e.g. diabetic nephropathy (DN), diabetic retinopathy, atherosclerosis, heart attack and coronary disease [124,125]. DN is characterised by morphological renal changes such as glomerular basement membrane thickening, mesangial expansion, nodular increase in mesangial matrix, glomerulosclerosis and interstitial fibrosis that leads to kidney failure and is the primary cause of end-stage-kidney disease leading to dialysis and transplantation [126]. Recently, several studies have suggested inflammation to play an essential role in DN (Figure 4) [127,128]. An accumulation of infiltrating macrophages has been demonstrated in diabetic kidneys from pre-clinical animal DN models and in human DN biopsies [128,129]. Resident kidney macrophages are present already at sub-clinical stages when the patients show none or only minimal symptoms [128,130]. In this early stage 1 of DN, the kidney show an infiltration of primarily M1-like CD11chigh macrophages that produce high levels of M1-like cytokines i.e. IFN-γ, IL-6 and TNF-α which contribute to the renal injury [131]. During the disease progression, the macrophages re-polarize locally in the kidney from an early CD11b+F4/80low phenotype that potently contribute to apoptosis and tissue destruction to a CD11b+F4/80high macrophage subtype that primarily enhance tissue remodelling and fibrosis present in late stage 3-4 DN [132]. We recently demonstrated in two pre-clinical models of advanced DN that macrophages indeed had re-polarized locally in the tissue towards a phenotype more resembling a mixed M2 to M1/M2- like phenotype with significantly reduced CD11c in combination with enhanced galectin-3 expression [69]. Macrophage galectin-3 strengthen TGFβR signalling by retaining cell surface expression of TGF-β receptors and thus the newly polarized diabetic kidney macrophage might provide novel insight to understand the tissue remodelling processes occurring in DN [133,134].

clinical-cellular-immunology-diabetic-kidney

Figure 4: Macrophage infiltration in the diabetic kidney. In early stages of DN the macrophage subset within the kidneys is primarily M1-like CD11chigh macrophages. These M1-like macrophages secrete IFN-γ, IL-6 and TNF-α which contribute to the renal injury. Established chronic DN is characterized by a re-polarization of the macrophages towards a mix between M1/M2 macrophages showing reduced expression of CD11c and enhanced TGF-β and galectin-3 expression.

Conclusion

Macrophages are a heterogeneous population that can polarize and re-polarize by specific interactions between PPARs, cytokines and growth factors modulated in the local milieu by the disease and then reciprocally modulate the disease. Substantial support is available that suggests that adipose tissue M1-like macrophages contribute to low grade inflammatory signatures in obesity and early diabetes. Growing evidence suggests that islets from both T1D and T2D are populated with macrophages that participate in the disease progression particularly during the initiation of the disease. The involvement of macrophages in the complications of metabolic disease like diabetic nephropathy is still emerging, but it appears that the macrophages undergoes a re-polarization process locally in the diabetic tissue from a M1-like towards a M2-like phenotype with disease progression potentially contributing to the fibrotic events in the disease. In summary, the complex nature of macrophages should be considered when evaluating macrophage biology during metabolic diseases but also raises the possibilities if used correctly to identify novel therapeutic angles.

References

  1. Auffray C, Sieweke MH, Geissmann F (2009) Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol 27: 669-692.
  2. van Furth R, Cohn ZA (1968) The origin and kinetics of mononuclear phagocytes. J Exp Med 128: 415-435.
  3. Iwasaki A, Medzhitov R (2004) Toll-like receptor control of the adaptive immune responses. Nat Immunol 5: 987-995.
  4. Conti L, Cardone M, Varano B, Puddu P, Belardelli F, et al. (2008) Role of the cytokine environment and cytokine receptor expression on the generation of functionally distinct dendritic cells from human monocytes. Eur J Immunol 38: 750-762.
  5. Biswas SK, Chittezhath M, Shalova IN, Lim JY (2012) Macrophage polarization and plasticity in health and disease. Immunol Res 53: 11-24.
  6. Auger MJ (1992) The biology of the macrophage. In: Lewis CE, McGee JO (eds.) The macrophage. New York.
  7. Shi C, Pamer EG (2011) Monocyte recruitment during infection and inflammation. Nat Rev Immunol 11: 762-774.
  8. Boddeke EW, Meigel I, Frentzel S, Gourmala NG, Harrison JK, et al. (1999) Cultured rat microglia express functional beta-chemokine receptors. J Neuroimmunol 98: 176-184.
  9. Vanbervliet B, Homey B, Durand I, Massacrier C, Aït-Yahia S, et al. (2002) Sequential involvement of CCR2 and CCR6 ligands for immature dendritic cell recruitment: possible role at inflamed epithelial surfaces. Eur J Immunol 32: 231-242.
  10. Loughrey BV, McGinty A, Young IS, McCance DR, Powell LA (2013) Increased circulating CC chemokine levels in the metabolic syndrome are reduced by low-dose atorvastatin treatment: evidence from a randomized controlled trial. ClinEndocrinol (Oxf) 79: 800-806.
  11. Heinrichs D, Berres ML, Nellen A, Fischer P, Scholten D, et al. (2013) The chemokine CCL3 promotes experimental liver fibrosis in mice. PLoS One 8: e66106.
  12. Ben-Baruch A, Xu L, Young PR, Bengali K, Oppenheim JJ, et al. (1995) Monocyte chemotactic protein-3 (MCP3) interacts with multiple leukocyte receptors. C-C CKR, a receptor for macrophage inflammatory protein-1 alpha/Rantes, is also a functional receptor for MCP3. J biolChem 270: 22123-22128.
  13. Gonzalez J, Mouttalib S, Delage C, Calise D, Maoret JJ, et al. (2013) Dual effect of chemokine CCL7/MCP-3 in the development of renal tubulointerstitial fibrosis. BiochemBiophys Res Commun 438: 257-263.
  14. Ley K, Laudanna C, Cybulsky MI, Nourshargh S (2007) Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 7: 678-689.
  15. Dole VS, Bergmeier W, Patten IS, Hirahashi J, Mayadas TN, et al. (2007) PSGL-1 regulates platelet P-selectin-mediated endothelial activation and shedding of P-selectin from activated platelets. ThrombHaemost 98: 806-812.
  16. León B, Ardavín C (2008) Monocyte migration to inflamed skin and lymph nodes is differentially controlled by L-selectin and PSGL-1. Blood 111: 3126-3130.
  17. Galkina E, Ley K (2009) Immune and inflammatory mechanisms of atherosclerosis (*). Annu Rev Immunol 27: 165-197.
  18. Harris RA (2014) Spatial, Temporal, and Functional Aspects of Macrophages during "The Good, the Bad, and the Ugly" Phases of Inflammation. Front Immunol 5: 612.
  19. Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, et al. (2012) A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336: 86-90.
  20. Lin SL, Castano AP, Nowlin BT, Lupher ML Jr., Duffield JS (2009) Bone marrow Ly6Chigh monocytes are selectively recruited to injured kidney and differentiate into functionally distinct populations. J Immunol 183: 6733-6743.
  21. Alikhan MA, Jones CV, Williams TM, Beckhouse AG, Fletcher AL, et al. (2011) Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses. Am J Pathol 179: 1243-1256.
  22. Lin SL, Kisseleva T, Brenner DA, Duffield JS (2008) Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol 173: 1617-1627.
  23. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, et al. (2006) CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest 116: 115-124.
  24. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, et al. (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116: 3015-3025.
  25. Erridge C, Samani NJ (2009) Saturated fatty acids do not directly stimulate Toll-like receptor signaling. ArteriosclerThrombVascBiol 29: 1944-1949.
  26. Hotamisligil GS (2006) Inflammation and metabolic disorders. Nature 444: 860-867.
  27. Dobrovolskaia MA, Medvedev AE, Thomas KE, Cuesta N, Toshchakov V, et al. (2003) Induction of in vitro reprogramming by Toll-like receptor (TLR)2 and TLR4 agonists in murine macrophages: effects of TLR "homotolerance" versus "heterotolerance" on NF-kappa B signaling pathway components. J Immunol 170: 508-519.
  28. Verstrepen L, Bekaert T, Chau TL, Tavernier J, Chariot A, et al. (2008) TLR-4, IL-1R and TNF-R signaling to NF-kappaB: variations on a common theme. Cell Mol Life Sci 65: 2964-2978.
  29. Martinon F, Burns K, Tschopp J (2002) The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 10: 417-426.
  30. Fink SL, Cookson BT (2005) Apoptosis, pyroptosis, and necrosis: mechanistic description of dead and dying eukaryotic cells. Infect Immun 73: 1907-1916.
  31. Hotamisligil GS, Shargill NS, Spiegelman BM (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259: 87-91.
  32. Feinstein R, Kanety H, Papa MZ, Lunenfeld B, Karasik A (1993) Tumor necrosis factor-alpha suppresses insulin-induced tyrosine phosphorylation of insulin receptor and its substrates. J BiolChem 268: 26055-26058.
  33. Varol C, Landsman L, Fogg DK, Greenshtein L, Gildor B, et al. (2007) Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. J Exp Med 204: 171-180.
  34. Nomiyama T, Perez-Tilve D, Ogawa D, Gizard F, Zhao Y, et al. (2007) Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J Clin Invest 117: 2877-2888.
  35. Kurokawa J, Arai S, Nakashima K, Nagano H, Nishijima A, et al. (2010) Macrophage-derived AIM is endocytosed into adipocytes and decreases lipid droplets via inhibition of fatty acid synthase activity. Cell Metab 11: 479-492.
  36. Hosoi T, Yokoyama S, Matsuo S, Akira S, Ozawa K (2010) Myeloid differentiation factor 88 (MyD88)-deficiency increases risk of diabetes in mice. PLoS One 5.
  37. Vijay-Kumar M, Aitken JD, Carvalho FA, Ziegler TR, Gewirtz AT, et al. (2011) Loss of function mutation in toll-like receptor-4 does not offer protection against obesity and insulin resistance induced by a diet high in trans fat in mice. J Inflamm (Lond) 8: 2.
  38. Usher MG, Duan SZ, Ivaschenko CY, Frieler RA, Berger S, et al. (2010) Myeloid mineralocorticoid receptor controls macrophage polarization and cardiovascular hypertrophy and remodeling in mice. J clin invest 120: 3350-3364.
  39. Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, et al. (2001) Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med 194: 1361-1373.
  40. Geissmann F, Jung S, Littman DR (2003) Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19: 71-82.
  41. Martinez FO, Gordon S, Locati M, Mantovani A (2006) Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol 177: 7303-7311.
  42. Gratchev A, Kzhyshkowska J, Köthe K, Muller-Molinet I, Kannookadan S, et al. (2006) Mphi1 and Mphi2 can be re-polarized by Th2 or Th1 cytokines, respectively, and respond to exogenous danger signals. Immunobiology 211: 473-486.
  43. Stout RD, Jiang C, Matta B, Tietzel I, Watkins SK, et al. (2005) Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J Immunol 175: 342-349.
  44. Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, et al. (1993) Immune response in mice that lack the interferon-gamma receptor. Science 259: 1742-1745.
  45. Nathan C (1991) Mechanisms and modulation of macrophage activation. Behring Inst Mitt : 200-207.
  46. Laskin DL, Sunil VR, Gardner CR, Laskin JD (2011) Macrophages and tissue injury: agents of defense or destruction? Annu Rev PharmacolToxicol 51: 267-288.
  47. Luster AD (2002) The role of chemokines in linking innate and adaptive immunity. CurrOpinImmunol 14: 129-135.
  48. Sallusto F, Lenig D, Mackay CR, Lanzavecchia A (1998) Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J Exp Med 187: 875-883.
  49. Lehuen A, Diana J, Zaccone P, Cooke A (2010) Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol 10: 501-513.
  50. Mosser DM (2003) The many faces of macrophage activation. J LeukocBiol 73: 209-212.
  51. Geng YJ, Wu Q, Muszynski M, Hansson GK, Libby P (1996) Apoptosis of vascular smooth muscle cells induced by in vitro stimulation with interferon-gamma, tumor necrosis factor-alpha, and interleukin-1 beta. ArteriosclerThrombVascBiol 16: 19-27.
  52. Chizzolini C, Parel Y, Scheja A, Dayer JM (2006) Polarized subsets of human T-helper cells induce distinct patterns of chemokine production by normal and systemic sclerosis dermal fibroblasts. Arthritis Res Ther 8: R10.
  53. Gibbs DF, Warner RL, Weiss SJ, Johnson KJ, Varani J (1999) Characterization of matrix metalloproteinases produced by rat alveolar macrophages. Am J Respir Cell MolBiol 20: 1136-1144.
  54. Boyle JJ, Weissberg PL, Bennett MR (2003) Tumor necrosis factor-alpha promotes macrophage-induced vascular smooth muscle cell apoptosis by direct and autocrine mechanisms. ArteriosclerThrombVascBiol 23: 1553-1558.
  55. Gordon S, Taylor PR (2005) Monocyte and macrophage heterogeneity. Nat Rev Immunol 5: 953-964.
  56. Brombacher F (2000) The role of interleukin-13 in infectious diseases and allergy. Bioessays 22: 646-656.
  57. Montaner LJ, da Silva RP, Sun J, Sutterwala S, Hollinshead M, et al. (1999) Type 1 and type 2 cytokine regulation of macrophage endocytosis: differential activation by IL-4/IL-13 as opposed to IFN-gamma or IL-10. J Immunol 162: 4606-4613.
  58. Conner SD, Schmid SL (2003) Regulated portals of entry into the cell. Nature 422: 37-44.
  59. Harding CV, Ramachandra L, Wick MJ (2003) Interaction of bacteria with antigen presenting cells: influences on antigen presentation and antibacterial immunity. CurrOpinImmunol 15: 112-119.
  60. Murray PJ, Wynn TA (2011) Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol 11: 723-737.
  61. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, et al. (2004) The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25: 677-686.
  62. Balbo P, Silvestri M, Rossi GA, Crimi E, Burastero SE (2001) Differential role of CD80 and CD86 on alveolar macrophages in the presentation of allergen to T lymphocytes in asthma. ClinExp Allergy 31: 625-636.
  63. Lumeng CN, Bodzin JL, Saltiel AR (2007) Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175-184.
  64. Pillai MM, Hayes B, Torok-Storb B (2009) Inducible transgenes under the control of the hCD68 promoter identifies mouse macrophages with a distribution that differs from the F4/80 - and CSF-1R-expressing populations. Exphematol 37: 1387-1392.
  65. Gordon S, Mantovani A (2011) Diversity and plasticity of mononuclear phagocytes. Eur J Immunol 41: 2470-2472.
  66. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, et al. (2010) Nomenclature of monocytes and dendritic cells in blood. Blood 116: e74-80.
  67. Serbina NV, Jia T, Hohl TM, Pamer EG (2008) Monocyte-mediated defense against microbial pathogens. Annu Rev Immunol 26: 421-452.
  68. Kurihara T, Warr G, Loy J, Bravo R (1997) Defects in macrophage recruitment and host defense in mice lacking the CCR2 chemokine receptor. J Exp Med 186: 1757-1762.
  69. Cucak H, Nielsen Fink L, Hojgaard Pedersen M, Rosendahl A (2015) Enalapril treatment increases T cell number and promotes polarization towards M1-like macrophages locally in diabetic nephropathy. IntImmunopharmacol 25: 30-42.
  70. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, et al. (2007) Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 317: 666-670.
  71. Ziegler-Heitbrock HW, Fingerle G, Ströbel M, Schraut W, Stelter F, et al. (1993) The novel subset of CD14+/CD16+ blood monocytes exhibits features of tissue macrophages. Eur J Immunol 23: 2053-2058.
  72. Skrzeczynska-Moncznik J, Bzowska M, Loseke S, Grage-Griebenow E, Zembala M, et al. (2008) Peripheral blood CD14high CD16+ monocytes are main producers of IL-10. Scand J Immunol 67: 152-159.
  73. Berg KE, Ljungcrantz I, Andersson L, Bryngelsson C, Hedblad B, et al. (2012) Elevated CD14++CD16- monocytes predict cardiovascular events. CircCardiovasc Genet 5: 122-131.
  74. Cros J, Cagnard N, Woollard K, Patey N, Zhang SY, et al. (2010) Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity 33: 375-386.
  75. Poitou C, Dalmas E, Renovato M, Benhamo V, Hajduch F, et al. (2011) CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss: relationships with fat mass and subclinical atherosclerosis. ArteriosclerThrombvascbiol 31: 2322-2330.
  76. Rogacev KS, Seiler S, Zawada AM, Reichart B, Herath E, et al. (2011) CD14++CD16+ monocytes and cardiovascular outcome in patients with chronic kidney disease. Eur Heart J 32: 84-92.
  77. Cnop M, Welsh N, Jonas JC, Jörns A, Lenzen S, et al. (2005) Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 54 Suppl 2: S97-107.
  78. Grage-Griebenow E, Flad HD, Ernst M (2001) Heterogeneity of human peripheral blood monocyte subsets. J LeukocBiol 69: 11-20.
  79. Ingersoll MA, Spanbroek R, Lottaz C, Gautier EL, Frankenberger M, et al. (2010) Comparison of gene expression profiles between human and mouse monocyte subsets. Blood 115: e10-19.
  80. Martinez FO, Helming L, Milde R, Varin A, Melgert BN, et al. (2013) Genetic programs expressed in resting and IL-4 alternatively activated mouse and human macrophages: similarities and differences. Blood 121: e57-69.
  81. Flegal KM, Carroll MD, Ogden CL, Curtin LR (2010) Prevalence and trends in obesity among US adults, 1999-2008. JAMA 303: 235-241.
  82. Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, 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: 557-567.
  83. Berrington de Gonzalez A, Hartge P, Cerhan JR, Flint AJ, Hannan L, et al. (2010) Body-mass index and mortality among 1.46 million white adults. N Engl J Med 363: 2211-2219.
  84. Flegal KM, Graubard BI, Williamson DF, Gail MH (2007) Cause-specific excess deaths associated with underweight, overweight, and obesity. JAMA 298: 2028-2037.
  85. Zheng W, McLerran DF, Rolland B, Zhang X, Inoue M, et al. (2011) Association between body-mass index and risk of death in more than 1 million Asians. N Engl J Med 364: 719-729.
  86. American Diabetes Association (2009) Diagnosis and classification of diabetes mellitus. Diabetes Care 32 Suppl 1: S62-67.
  87. Al-Goblan AS, Al-Alfi MA, Khan MZ (2014) Mechanism linking diabetes mellitus and obesity. Diabetes MetabSyndrObes 7: 587-591.
  88. Guariguata L, Whiting D, Weil C, Unwin N (2011) The International Diabetes Federation diabetes atlas methodology for estimating global and national prevalence of diabetes in adults. Diabetes Res ClinPract 94: 322-332.
  89. Shoelson SE, Lee J, Goldfine AB (2006) Inflammation and insulin resistance. J Clin Invest 116: 1793-1801.
  90. Eguchi K, Manabe I, Oishi-Tanaka Y, Ohsugi M, Kono N, et al. (2012) Saturated fatty acid and TLR signaling link β cell dysfunction and islet inflammation. Cell Metab 15: 518-533.
  91. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, et al. (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112: 1796-1808.
  92. Cucak H, Vistisen D, Witte D, Philipsen A, Rosendahl A (2014) Reduction of specific circulating lymphocyte populations with metabolic risk factors in patients at risk to develop type 2 diabetes. PLoS One 9: e107140.
  93. Kahn SE, Hull RL, Utzschneider KM (2006) Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444: 840-846.
  94. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, et al. (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52: 102-110.
  95. Cucak H, Grunnet LG, Rosendahl A (2014) Accumulation of M1-like macrophages in type 2 diabetic islets is followed by a systemic shift in macrophage polarization. J LeukocBiol 95: 149-160.
  96. Eizirik DL, Colli ML, Ortis F (2009) The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nat Rev Endocrinol 5: 219-226.
  97. Martin AP, Grisotto MG, Canasto-Chibuque C, Kunkel SL, Bromberg JS, et al. (2008) Islet expression of M3 uncovers a key role for chemokines in the development and recruitment of diabetogenic cells in NOD mice. Diabetes 57: 387-394.
  98. Calderon B, Suri A, Unanue ER (2006) In CD4+ T-cell-induced diabetes, macrophages are the final effector cells that mediate islet beta-cell killing: studies from an acute model. Am J Pathol 169: 2137-2147.
  99. Hutchings P, Rosen H, O'Reilly L, Simpson E, Gordon S, et al. (1990) Transfer of diabetes in mice prevented by blockade of adhesion-promoting receptor on macrophages. Nature 348: 639-642.
  100. Devaraj S, Glaser N, Griffen S, Wang-Polagruto J, Miguelino E, et al. (2006) Increased monocytic activity and biomarkers of inflammation in patients with type 1 diabetes. Diabetes 55: 774-779.
  101. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F (2007) Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol 8: 942-949.
  102. Jagannathan-Bogdan M, McDonnell ME, Shin H, Rehman Q, Hasturk H, et al. (2011) Elevated proinflammatory cytokine production by a skewed T cell compartment requires monocytes and promotes inflammation in type 2 diabetes. J Immunol 186: 1162-1172.
  103. Rui L, Aguirre V, Kim JK, Shulman GI, Lee A, et al. (2001) Insulin/IGF-1 and TNF-alpha stimulate phosphorylation of IRS-1 at inhibitory Ser307 via distinct pathways. J Clin Invest 107: 181-189.
  104. Neels JG, Olefsky JM (2006) Inflamed fat: what starts the fire? J Clin Invest 116: 33-35.
  105. Lumeng CN, DelProposto JB, Westcott DJ, Saltiel AR (2008) Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 57: 3239-3246.
  106. Dalmas E, Clément K, Guerre-Millo M (2011) Defining macrophage phenotype and function in adipose tissue. Trends Immunol 32: 307-314.
  107. Zeyda M, Stulnig TM (2007) Adipose tissue macrophages. ImmunolLett 112: 61-67.
  108. Fujisaka S, Usui I, Bukhari A, Ikutani M, Oya T, et al. (2009) Regulatory mechanisms for adipose tissue M1 and M2 macrophages in diet-induced obese mice. Diabetes 58: 2574-2582.
  109. Olefsky JM, Glass CK (2010) Macrophages, inflammation, and insulin resistance. Annu Rev Physiol 72: 219-246.
  110. Ferrante AW Jr (2007) Obesity-induced inflammation: a metabolic dialogue in the language of inflammation. J Intern Med 262: 408-414.
  111. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, et al. (2005) Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46: 2347-2355.
  112. Cho HJ, Shashkin P, Gleissner CA, Dunson D, Jain N, et al. (2007) Induction of dendritic cell-like phenotype in macrophages during foam cell formation. Physiol Genomics 29: 149-160.
  113. Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW 2nd, DeFuria J, et al. (2007) Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes 56: 2910-2918.
  114. Patsouris D, Li PP, Thapar D, Chapman J, Olefsky JM, et al. (2008) Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab 8: 301-309.
  115. Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, et al. (2005) IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med 11: 191-198.
  116. Solinas G, Vilcu C, Neels JG, Bandyopadhyay GK, Luo JL, et al. (2007) JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab 6: 386-397.
  117. Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, et al. (2010) GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142: 687-698.
  118. Richardson SJ, Willcox A, Bone AJ, Foulis AK, Morgan NG (2009) Islet-associated macrophages in type 2 diabetes. Diabetologia 52: 1686-1688.
  119. Kristiansen OP, Mandrup-Poulsen T (2005) Interleukin-6 and diabetes: the good, the bad, or the indifferent? Diabetes 54 Suppl 2: S114-124.
  120. Mosser DM, Edwards JP (2008) Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8: 958-969.
  121. Saksida T, Nikolic I, Vujicic M, Nilsson UJ, Leffler H, et al. (2013) Galectin-3 deficiency protects pancreatic islet cells from cytokine-triggered apoptosis in vitro. J Cell Physiol 228: 1568-1576.
  122. Hsu DK, Yang RY, Pan Z, Yu L, Salomon DR, et al. (2000) Targeted disruption of the galectin-3 gene results in attenuated peritoneal inflammatory responses. Am J Pathol 156: 1073-1083.
  123. MacKinnon AC, Farnworth SL, Hodkinson PS, Henderson NC, Atkinson KM, et al. (2008) Regulation of alternative macrophage activation by galectin-3. J Immunol 180: 2650-2658.
  124. Packham DK, Alves TP, Dwyer JP, Atkins R, de Zeeuw D, et al. (2012) Relative incidence of ESRD versus cardiovascular mortality in proteinuric type 2 diabetes and nephropathy: results from the DIAMETRIC (Diabetes Mellitus Treatment for Renal Insufficiency Consortium) database. Am J Kidney Dis 59: 75-83.
  125. Forbes JM, Cooper ME (2013) Mechanisms of diabetic complications. Physiol Rev 93: 137-188.
  126. Tervaert TW, Mooyaart AL, Amann K, Cohen AH, Cook HT, et al. (2010) Pathologic classification of diabetic nephropathy. J Am SocNephrol 21: 556-563.
  127. Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, García-Pérez J (2011) Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol 7: 327-340.
  128. Nguyen D, Ping F, Mu W, Hill P, Atkins RC, et al. (2006) Macrophage accumulation in human progressive diabetic nephropathy. Nephrology (Carlton) 11: 226-231.
  129. Sassy-Prigent C, Heudes D, Mandet C, Bélair MF, Michel O, et al. (2000) Early glomerular macrophage recruitment in streptozotocin-induced diabetic rats. Diabetes 49: 466-475.
  130. Galkina E, Ley K (2006) Leukocyte recruitment and vascular injury in diabetic nephropathy. J Am SocNephrol 17: 368-377.
  131. Sean Eardley K, Cockwell P (2005) Macrophages and progressive tubulointerstitial disease. Kidney Int 68: 437-455.
  132. Fujiu K, Manabe I, Nagai R (2011) Renal collecting duct epithelial cells regulate inflammation in tubulointerstitial damage in mice. J Clin Invest 121: 3425-3441.
  133. Iacobini C, Amadio L, Oddi G, Ricci C, Barsotti P, et al. (2003) Role of galectin-3 in diabetic nephropathy. J Am SocNephrol 14: S264-270.
  134. Rabinovich GA, Toscano MA (2009) Turning 'sweet' on immunity: galectin-glycan interactions in immune tolerance and inflammation. Nat Rev Immunol 9: 338-352.
  135. Shen Z, Lu M, Duan S, Duan S (2011) Macrophage Polarization and Inflammation at the Interface of Cardiovascular Disease and Metabolism. NAJ Med Sci 4: 191-195.
Citation: Thomsen LH, Rosendahl A (2015) Polarization of Macrophages in Metabolic Diseases. J Clin Cell Immunol 6:313.

Copyright: © 2015 Thomsen LH. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top