inflammation, advancing age and nutrition || obesity, insulin resistance, and inflammaging

8
Inflammation, Advancing Age and Nutrition. http://dx.doi.org/10.1016/B978-0-12-397803-5.00013-7 Copyright © 2014 Elsevier Inc. All rights reserved. 157 Abbreviations AT adipose tissue ATMs adipose tissue macrophages BMI body mass index CRP C-reactive protein ERK extracellular signal-regulated kinase FFA free fatty acids GDF-15 growth differentiation factor 15 GLP-1 glucagon-like peptide 1 HIF1-α hypoxia inducible factor 1-α IFN-ɣ interferon gamma IKK inhibitor of nuclear factor kappa-B kinase IL interleukin IR insulin resistance IRS insulin receptor substrate JNK c-jun N-terminal kinase LPS lipopolysaccharide MAPK mitogen-activated protein kinase MCP-1 C-C motif chemokine 2/monocyte chemotactic protein 1 MIP-1α C-C motif chemokine 3/macrophage inflammatory protein 1-α NF-κB nuclear factor-kappa-B OPN osteopontin PI-3K phosphatidylinositol 3-kinase PKC protein kinase C SAT subcutaneous adipose tissue SOCS suppressor of cytokine signaling TLR Toll-like receptor VAT visceral adipose tissue. INTRODUCTION Increased population aging in both industrialized and developing countries is associated with an increased susceptibility to chronic noncommunicable diseases, which limit the human life span [1]. The burden of aging-associated chronic conditions is considerable, as they accounted for 42% of the deaths in the European Union in 2007 [2]. Obesity, defined by a body mass index (BMI) 30 kg/m 2 , has spread alarmingly worldwide, especially in elderly individuals, and is independently associated with a greater risk of atherosclerotic cardio- vascular disease, type 2 diabetes mellitus (T2DM), and death [3,4]. If the caloric intake of an adult person, with a normal BMI of 22 kg/m 2 , exceeds energy expenditure by only 50 kcal/day, weight will increase by 2 kg within a year and by 20 kg within 10 years, at which time point that person’s BMI will have reached 30 kg/m 2 . The prev- alence of both generalized obesity and central obesity increases with age [5]. The distribution of adipose tissue (AT) in obesity is important for determining health risks of patients. Visceral obesity due to an increased accumulation of visceral (intra- abdominal) adipose tissue (VAT) is a strong predictor for T2DM and metabolic syndrome [6,7]. Visceral obesity is a major determinant of insulin resistance (IR) [8,9], i.e. a decreased responsiveness to the metabolic actions of insu- lin in AT, muscle, liver, and endothelial and immune cells [10,11]. The degree of IR increases with age in rodents and humans, while compensatory hyperinsulinemia, found in insulin-resistant subjects, may accelerate the aging process [12,13]. IR observed in aging is directly linked to obesity [14]. Physiologically, excess energy is stored as fat, pre- dominantly in subcutaneous adipose tissue (SAT). Once the storage capacity of SAT is exceeded, lipids accumulate in VAT [15]. AT hypertrophy is associated with endoplas- mic reticulum stress, which provokes AT inflammation, particularly in visceral AT. Inflammation triggers an efflux of free fatty acids (FFA) into the circulation [16]. Lipotox- icity due to overload of metabolic intermediates, such as ceramides or diacylglycerol in metabolic tissues includ- ing liver, muscle, and pancreatic islets, further aggravates IR and can lead to diabetes by interfering with pancreatic beta-cell function and survival [17,18]. IR does not occur solely due to lipotoxicity. The expan- sion of the vascular network does not meet the increased CHAPTER 13 Obesity, Insulin Resistance, and Inflammaging Bianca K. Itariu, Thomas M. Stulnig Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy, and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria

Upload: bianca-k

Post on 15-Dec-2016

213 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

C H A P T E R

13Obesity, Insulin Resistance, and Inflammaging

Bianca K. Itariu, Thomas M. StulnigChristian Doppler Laboratory for Cardio-Metabolic Immunotherapy, and Clinical Division of Endocrinology and

Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria

Inflammation, Advancing Age and Nutrition. http://dx.doi.org/10.1016/B978-0-12-397803-5.00013-7

157

AbbreviationsAT adipose tissueATMs adipose tissue macrophagesBMI body mass indexCRP C-reactive proteinERK extracellular signal-regulated kinaseFFA free fatty acidsGDF-15 growth differentiation factor 15GLP-1 glucagon-like peptide 1HIF1-α hypoxia inducible factor 1-αIFN-ɣ interferon gammaIKK inhibitor of nuclear factor kappa-B kinaseIL interleukinIR insulin resistanceIRS insulin receptor substrateJNK c-jun N-terminal kinaseLPS lipopolysaccharideMAPK mitogen-activated protein kinaseMCP-1 C-C motif chemokine 2/monocyte chemotactic protein 1MIP-1α C-C motif chemokine 3/macrophage inflammatory protein 1-αNF-κB nuclear factor-kappa-BOPN osteopontinPI-3K phosphatidylinositol 3-kinasePKC protein kinase CSAT subcutaneous adipose tissueSOCS suppressor of cytokine signalingTLR Toll-like receptorVAT visceral adipose tissue.

INTRODUCTION

Increased population aging in both industrialized and developing countries is associated with an increased susceptibility to chronic noncommunicable diseases, which limit the human life span [1]. The burden of aging-associated chronic conditions is considerable, as they accounted for 42% of the deaths in the European Union in 2007 [2]. Obesity, defined by a body mass index (BMI) ≥ 30 kg/m2, has spread alarmingly worldwide,

Copyright © 2014 Elsevier Inc. All rights reserved.

especially in elderly individuals, and is independently associated with a greater risk of atherosclerotic cardio-vascular disease, type 2 diabetes mellitus (T2DM), and death [3,4]. If the caloric intake of an adult person, with a normal BMI of 22 kg/m2, exceeds energy expenditure by only 50 kcal/day, weight will increase by 2 kg within a year and by 20 kg within 10 years, at which time point that person’s BMI will have reached 30 kg/m2. The prev-alence of both generalized obesity and central obesity increases with age [5].

The distribution of adipose tissue (AT) in obesity is important for determining health risks of patients. Visceral obesity due to an increased accumulation of visceral (intra-abdominal) adipose tissue (VAT) is a strong predictor for T2DM and metabolic syndrome [6,7]. Visceral obesity is a major determinant of insulin resistance (IR) [8,9], i.e. a decreased responsiveness to the metabolic actions of insu-lin in AT, muscle, liver, and endothelial and immune cells [10,11]. The degree of IR increases with age in rodents and humans, while compensatory hyperinsulinemia, found in insulin-resistant subjects, may accelerate the aging process [12,13]. IR observed in aging is directly linked to obesity [14]. Physiologically, excess energy is stored as fat, pre-dominantly in subcutaneous adipose tissue (SAT). Once the storage capacity of SAT is exceeded, lipids accumulate in VAT [15]. AT hypertrophy is associated with endoplas-mic reticulum stress, which provokes AT inflammation, particularly in visceral AT. Inflammation triggers an efflux of free fatty acids (FFA) into the circulation [16]. Lipotox-icity due to overload of metabolic intermediates, such as ceramides or diacylglycerol in metabolic tissues includ-ing liver, muscle, and pancreatic islets, further aggravates IR and can lead to diabetes by interfering with pancreatic beta-cell function and survival [17,18].

IR does not occur solely due to lipotoxicity. The expan-sion of the vascular network does not meet the increased

Page 2: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

13. OBESITY, INSULIN RESISTAN158

oxygen demand of expanded obese AT, resulting in insuf-ficient capillary density, local hypoxia, and upregulation of transcription factors, such as hypoxia inducible factor-1α (HIF-1α) [19]. Under hypoxic conditions, AT secretes proinflammatory mediators that impair insulin signal-ing and potentiate IR [20]. Proinflammatory cytokines are also released from adipocytes and other cell popula-tions from the AT stroma vascular fraction, such as mac-rophages and lymphocytes, upon sensing the increased energy storage levels characteristic of obesity and over-nutrition, partly via pattern recognition receptors of the innate immune system such as Toll-like receptors (TLRs) 2 and 4 [21–23]. Moreover, aging per se increases this inflammatory response to overnutrition [24].

Obesity, IR, and aging are hence associated with chronic systemic low-grade inflammation [25]. This type of inflammation represents a driving force for human aging [26]. Moreover, inflammaging (i.e. inflammation and aging) has been linked to negative health outcomes. Chronic obesity-associated inflammation is character-ized by (1) increased circulating levels of proinflam-matory markers; (2) increased proinflammatory gene expression in insulin target tissues; and (3) immune cell, mainly macrophage and T cell, activation and infiltration in AT [27–29]. Currently, the concept that AT is a highly active endocrine organ, secreting more than a hundred so-called adipokines, e.g. chemokines and cytokines, is well established [30]. In obesity, these molecules orches-trate the inflammatory response locally within AT as well as in the whole body and have predominantly proinflam-matory effects. A link between chronic inflammation and both obesity and IR was first shown for the proinflam-matory cytokine tumor necrosis factor (TNF-α), which directly causes IR; administration of an anti-TNF-α anti-body improves insulin sensitivity, as TNF-α secretion from AT is induced by obesity [31].

Immigrated AT macrophages, which are recruited from the circulation, are the primary sources of pro-inflammatory cytokines and, together with T cells, propagate the inflammatory reactions in AT. Local AT inflammation is thus a key regulator of the chronic sys-temic inflammatory phenotype observed in murine and human obesity [32]. Although the relationship between inflammaging, obesity, and IR is not yet completely understood, the molecular pathophysiology leading to novel therapeutic options is of particular importance for reducing negative health outcomes in aging populations.

INFLAMMATORY MEDIATORS, OBESITY, AND AGING

Acute inflammation is a self-regulated physiological adaptive defense mechanism triggered by infection or harmful stimuli (either self or nonself) that is meant to

CE, AND INFLAMMAGING

protect the host and restore homeostasis [33]. The chronic low-grade systemic inflammation commonly associated with aging and obesity differs from the inflammatory response to infection or autoimmune reactions [34,35]. Although the cardinal signs of inflammation (redness, swelling, heat, pain, and loss of function) are absent in this state, the hallmark of obesity-associated inflamma-tion is the increase in circulating inflammatory factors. Both obesity and aging are characterized by elevated cir-culating levels of:

(1) Acute-phase proteins such as C-reactive protein (CRP) [29] or serum amyloid A [36];

(2) Inflammatory cytokines such as TNF-α, interleukin-6 (IL-6), IL-8 [37], IL-1β [38], and growth differentiation factor 15 (GDF-15) [39];

(3) Chemokines such as monocyte chemotactic protein 1 (MCP-1; CCL2 gene) [37] and macrophage inflammatory protein 1 α (MIP-1α; CCL3 gene) [40];

(4) Soluble adhesion molecules (P-selectin, E-selectin) [41]; and

(5) Multifunctional proteins such as leptin [42] or osteopontin (OPN) [43].

In older persons, serum levels of inflammatory mark-ers are increased 2- to 4-fold [44]. In aging, chronic anti-genic stimulation is thought to be a major driver of the systemic proinflammatory state [45], while AT (mainly VAT) is the starting point of the obesity-associated inflammatory response and a considerable source of pro-inflammatory mediators. Mediators secreted from VAT are directly transported to the liver via portal circulation. Increased IL-6 concentration upregulates CRP secretion in the liver. Both increased IL-6 and CRP concentrations are independent risk factors for T2DM and cardiovas-cular disease [46,47], and positively correlate with BMI.

Serum levels and AT expression of molecules belong-ing to the CC chemokine family and their receptors are increased in obesity, correlate positively with BMI, and are associated with increased systemic inflammation and IR [29,48]. Furthermore, gene sets related to chemo-kine activity and receptor binding are highly upregu-lated in VAT and SAT from insulin-resistant, compared to insulin-sensitive, obese subjects [29,49]. The multi-functional cytokine and extracellular matrix protein OPN is a modulator of IR and inflammation, and may be a therapeutic target for the treatment of IR [50,51]. OPN was found to be massively upregulated in obesity and is critically involved in macrophage activation, cell adhe-sion and migration, inflammation, and fibrosis, as well as an important modulator of IR in AT, liver, and muscle [50,52–54]. OPN knockout mice exhibit improved insu-lin sensitivity and reduced macrophage infiltration into AT, while neutralization of OPN with an anti-OPN anti-body also reduces AT inflammation and IR [50,53]. The exact effects of the inflammatory mediators involved in

Page 3: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

ADIPOSE TISSUE M

obesity-associated inflammation are yet not fully elu-cidated and their characterization represents a prolific field of research, especially when considering their clini-cal utility as predictive biomarkers.

CROSSTALK BETWEEN INSULIN AND INFLAMMATORY SIGNALING

Insulin regulates glucose homeostasis, promotes the storage of lipids and lipogenesis, decreases lipolysis in AT, and promotes protein synthesis in muscle [55].The insulin signaling pathway is fundamental for ensuring nutrient and energy homeostasis, and is highly con-served. Under physiological conditions, insulin binds to the insulin receptor and promotes autophosphoryla-tion of a trio of regulatory loop tyrosine residues, thus disinhibiting tyrosine kinase activity toward the insu-lin receptor substrate (IRS) 1 and 2, which is essential for insulin-mediated metabolic control. The subsequent tyrosine phosphorylation of IRS activates two major pathways: the phosphatidylinositol 3-kinase (PI-3K)/AKT pathway and the mitogen-activated protein kinase (MAPK) pathway [56,57]. The former is involved in insu-lin-stimulated glucose uptake, inhibition of gluconeo-genesis, and regulation of lipid metabolism, whereas the latter primarily controls cell growth and differentiation.

Proinflammatory cytokines impair insulin signaling through activation of downstream inflammatory signal-ing pathways [58], which interfere with insulin signal-ing network transduction downstream of IRS through the induction of serine kinases such as c-jun amino ter-minal kinase (JNK), and activation of nuclear factor-κB (NF-κB) and suppressor of cytokine signaling (SOCS) pathways [58,59]. Obesity- and aging-associated chronic inflammation, but also hyperglycemia and lipotoxicity, drastically impair insulin signaling through activation of MAPK members JNK and extracellular signal-regulated kinase (ERK), inhibitor of nuclear factor kappa-B kinase (IKK), and protein kinase C (PKC) [60,61]. When serine kinases that respond to inflammatory stimuli, such as IKK or JNK, phosphorylate critical serine sites on IRS, downstream insulin signaling is impaired, which results in disinhibited lipolysis in AT and dysregulated glu-cose uptake in muscle [62,63]. The released FFAs further promote the local and systemic inflammatory response [64,65]. In pancreatic beta cells, long-term exposure to FFA inhibits insulin synthesis and secretion by activation of PKC [66]. Insulin itself can activate JNK and other ser-ine kinases as part of a negative feedback mechanism.

Targeting signaling molecules from the inflammatory pathway leads to improved insulin sensitivity: JNK1 knockout mice do not develop high-fat diet-induced obesity and glucose intolerance and are protected from IR [67]. Cytokine signaling (IL-6 and TNF-α) can induce

159ACROPHAGES

in vitro serine phosphorylation via induction of IKK-β and SOCS 1 and 3 expression [68,69], which degrade IRS proteins. IL-1β downregulates IRS expression and IL-6 blocks insulin signaling via IRS and SOCS proteins [68–71].

NF-κB is another transcription factor with a pivotal role in chronic inflammatory disease [72]. NF-κB induces the expression of genes that promote IR. NF-κB signal-ing may be stimulated by (especially saturated) FFA and lipopolysaccharide (LPS) via binding to TLR4, which is expressed on adipocytes and macrophages and upregu-lated in obesity [73]. Lipotoxicity may be an important mediator of IR early in the development of obesity, whereas inflammation becomes more relevant once obe-sity is reached [74]. Elevated circulating FFA concentra-tions found in obesity and insulin-resistant states cause AT macrophages to release TNF-α directly through TLR4 and subsequent NF-κB activation [75]. NF-κB transloca-tion to the nucleus promotes further IL-6 and TNF-α synthesis [76]. Systemic IR continuously worsens with increasing AT inflammation [74]. Enhanced absorption of high-fat diets in the gut increases circulating LPS concentrations. LPS enters the circulation via chylo-microns, elicits low-grade endotoxemia, and potently stimulates TLR4, causing AT inflammation and com-pensatory hyperinsulinemia due to hepatic IR [25,77]. In the absence of functional TLR4 in mice, inflammatory signaling is blunted and the animals are protected from high-fat diet-induced IR [22]. IR is also improved when NF-κB activation is inhibited by salicylates [78]. Thus, FFA- and LPS-sensing in AT may initiate inflammatory and insulin-desensitizing processes. Interestingly, inhi-bition of NF-κB activation in myeloid cells provides the highest protection from systemic IR, highlighting the importance of this cell population in linking metabolic and inflammatory conditions [62,73].

ADIPOSE TISSUE MACROPHAGES

In obesity, AT macrophages (ATMs) play a central role in the crosstalk between inflammation and IR, and may also be key players in inflammaging-related pathological alterations [79–81]. Bone marrow-derived macrophages are recruited from the circulation into obese AT. In order to scavenge cell debris and lipid droplets, they localize mainly as “crown-like structures” around hypertrophic “stressed” adipocytes, which have undergone necrotic-like death [82]. Hence, AT growth by adipocyte hyper-trophy is limited and ongoing stimulated fat deposition leads to a chronic inflammatory response to induce IR.

In obesity, ATM recruitment from the circulation depends on various factors, including endocrine, paracrine, and mechanical ones. ATMs are a main source of inflammatory mediators (such as C-C chemokines, IL-6, and TNF-α), which

Page 4: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

13. OBESITY, INSULIN RESISTAN160

contribute to the recruitment of other macrophages, result-ing in a chronic inflammatory response [83,84]. The MCP-1/C-C motif chemokine receptor 2 (CCR2) axis is involved in macrophage recruitment to sites of inflammation [85,86]. MCP-1 (CCL2 gene) is secreted by AT and its production is stimulated by insulin and increased in obesity [87]. MCP-1 or CCR2 deficiency partially prevents macro-phage infiltration in obese AT and ameliorates IR [88,89]. In animal models, knocking out components of the mac-rophage inflammatory pathway can protect against obe-sity-induced IR [90,91].

Obesity causes a phenotypic switch in macrophage activation and polarization [92]. All ATMs express the overall macrophage marker CD68 [93]. Based on the surface expression of other antigens, ATM can be classi-fied into proinflammatory, classically activated M1 macro-phages [stimulated by interferon gamma (IFN-ɣ) and TLR ligands] and alternatively activated, anti-inflammatory M2 macrophages (stimulated by IL-4) [94]. M1 macrophages express a wide range of proinflammatory cytokines and inflammatory factors involved in IR [92,95] such as: F4/80, CD11c, CCR2 [92,96], and CD40 [92,95,97]. They are the predominant ATM population in obesity, whereas M2 macrophages express mannose and galactose recep-tors (MR/CD206 and MGL-1/CD301), IL-10 and chitinase-3-like protein 3 (Ym1) [98] have immunoregulatory and tissue-remodeling functions and are mainly found in lean subjects and animals. The M1/M2 paradigm is how-ever an oversimplification because in vivo macrophages exhibit great plasticity along the entire continuum from classical to alternative activation, and are characterized by various chemokine receptor expression patterns [99,100]. Mature adipocytes cultured in conditioned medium from activated macrophages exhibit IR and increased NF-κB activation, indicating the functional capability of mac-rophage secretion products to interfere with adipocyte function [101]. Impaired macrophage polarization in the elderly may dysregulate the development of the host response, making them more susceptible to infectious dis-eases; thus, the aging microenvironment may also be a key modulator of these macrophage-elicited responses [102].

ROLE OF OTHER IMMUNE CELL TYPES IN OBESITY-ASSOCIATED

INFLAMMAGING

Macrophages are far from being the only infiltrating immune cells described in AT. Lymphocytes and mast cells have been shown to play a part in obesity-induced inflammation and IR [103,104]. These cells act in an integrated, manner to influence ATM polarization and recruitment. Various T cell subtypes are recruited to AT and modulate inflammation: CD4+ proinflammatory T helper (Th) 1 and CD8+ cytotoxic T cells promote the

CE, AND INFLAMMAGING

attraction and differentiation of ATMs. On the other hand, CD4+ Th2 cells and regulatory T (Treg) cells coun-teract inflammation by inhibiting macrophage migra-tion and inducing alternative ATM activation [105,106]. The number of Th1 cells increases with obesity, while the number of Th2 and Treg cells decreases in mice. A detrimental ratio of Th1 : Th2 cells may also contrib-ute to the inflammatory response associated with obe-sity in humans; however, the obesity-associated loss of Treg cells found in murine AT could not be detected in humans [107]. The function and proportion of T cell sub-sets, and their proliferation and cytokine production is also affected by aging. Thus, aging itself can enhance the inflammatory response associated with obesity [108].

Accumulation of mast cells in AT has been described in murine models of diet-induced obesity and in human obesity, and mast cell-deficient mice have been shown to exhibit enhanced insulin sensitivity [109]. Moreover, mast cells seem to link obesity and T2DM, as the number of mast cells found in the AT of human obese diabetic subjects is increased compared to lean healthy controls. Mast cell numbers in AT correlate with macrophage activation, the degree of inflammation and fibrosis, and most notably with parameters of glucose homeostasis such as fasting glycemia and HbA1c [104].

THERAPEUTIC OPTIONS

The central role of inflammation in obesity should facilitate the identification of drug targets to improve prognosis by interfering with crucial molecular mecha-nisms underlying this inflammatory response. Moreover, a pragmatic approach for assessing the interrelationship between IR and inflammation in humans is analyz-ing the consequences of anti-inflammatory therapies on insulin sensitivity and of antidiabetic therapies on inflammatory status. Dietary factors are either induc-ers or mitigators of obesity-associated inflammation [47,110]. Notably, long-chain n-3 polyunsaturated fatty acids (n-3 PUFAs) are known for their cardioprotective and anti-inflammatory effects, and we have recently shown in a randomized clinical trial that n-3 PUFA suc-ceeded in lessening adipose and systemic inflammation in severely obese nondiabetic subjects after 8 weeks of treatment [111–113]. Adherence to the treatment regi-men of 3.36 g/day of highly purified n-3 PUFA (eicosa-pentaenoic acid and docosahexaenoic acid) was high in this trial and the dose was well tolerated. Long-chain n-3 PUFA decreased AT gene expression of CCL2, CCL3, HIF1A, and TGFB, as well as circulating IL-6 concentra-tions, indicating reduced AT and systemic inflammation. Although improvements in insulin sensitivity were not detected in this setting, n-3 PUFAs have been shown to ameliorate IR and risk profiles in subjects with metabolic

Page 5: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

COnClU

syndrome, indicating that beneficial metabolic effects probably occur with long-term treatment [114].

Antidiabetic and anti-inflammatory medications seem to go hand in hand when it comes to blocking lipolysis. Both metformin and salicylate inhibit TNF-α-induced lipolysis in primary adipocyte cultures in rats [115,116]. Salicylate together with pioglitazone protects pancreatic beta cells against IL-1β-induced damage [117]. Acetyl-salicylic acid inhibits prostaglandin and thromboxane biosynthesis but also favorably affects the formation of new classes of eicosanoids generated from long-chain n-3 PUFA. These so-called resolvins and protectins are anti-inflammatory and can actively resolve inflamma-tory responses even at very low doses [118]. Notably, we detected resolvins E1 and D1, as well as protectin D1, in AT of n-3 PUFA-treated patients. Thus, by increasing substrate availability for the synthesis of anti-inflamma-tory lipid mediators, n-3 PUFAs actively dampen obe-sity-associated inflammation [112].

Salicylates are weak inhibitors if IKKβ and serine phos-phorylation of IRS1, which might explain their favorable effect on glucose tolerance when given in very high doses [119]. High (4.5 g/day) and standard (3 g/day) doses of the anti-inflammatory drug salsalate improved glucose and lipid homeostasis via targeting NF-κB activity, and increased insulin secretion in healthy obese subjects [120,121]. Inter-estingly, methotrexate is an immunosuppressant drug that has in vivo insulin-sensitizing and antilipolytic effects [122]. Patients on methotrexate therapy have a reduced propensity for developing metabolic syndrome [123], and studies are ongoing to assess whether patients at risk of car-diovascular disease can benefit from its anti-inflammatory effects in terms of risk reduction. The anti-inflammatory actions of incretin-based therapies seem to contribute indi-rectly to their insulin-sensitizing effects. Glucagon-like pep-tide 1 (GLP-1) affects immune functions both directly and indirectly, improves insulin secretion [124], and may offer protection against degenerative age-related disorders [125]. Current pharmaceutical investigations are focusing on cytokine and chemokine receptor antagonism, particularly CCR2, as novel options for patients at high cardiometabolic risk [126]. Interference with IL-1 action through admin-istration of an IL-1 receptor (IL-1R) antagonist or specific antibodies against IL-1R lowers blood glucose levels, pre-dominantly improves beta-cell function, and reduces sys-temic inflammation [127,128]. However, investigations into tailored anti-inflammatory strategies for the prevention and treatment of obesity-related complications such as T2DM are still in their initial stages.

CONCLUSION

In the last 20 years, research focus has shifted from the metabolic pathophysiology of IR to the underlying

SIOn 161

chronic low-grade inflammation; this has improved our understanding of aging- and obesity-related complica-tions such as T2DM and cardiovascular disease. Whether increased inflammatory activity causes age-associated pathologies or reflects the sum of ongoing pathologi-cal processes remains uncertain. With respect to obesity, inflamed AT produces proinflammatory adipokines, cytokines, and chemokines, which promote local and sys-temic inflammatory reactions and IR by interfering with insulin signaling. Serum concentrations of adipokines are sensitive markers of subclinical inflammation in the elderly. Anti-inflammatory therapeutic strategies may prove beneficial for preventing obesity- and age-related diseases; however, further studies are necessary for a better understanding of the complex crosstalk between inflammation and metabolism in human disease.

References [1] Suzman RHJ. Chapter 70. World Demography of Aging. In:

Longo DL, Fauci AS, Kasper DL, Hauser SL, Jameson JL, Loscalzo J, editors. Harrison’s Principles of Internal Medicine. 18th ed New York: McGraw-Hill; 2012.

[2] Hasselaar J, Engels Y, Menten J, Jaspers B, Vissers K. The burden of non-acute dying on society: dying of cancer and chronic disease in the European Union. BMJ Supportive & Palliative Care 2012;2(4):334–8.

[3] Bray GA. Medical consequences of obesity. J Clin Endocrinol Metab 2004;89(6):2583–9.

[4] Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML, et al. The global obesity pandemic: shaped by global drivers and local environments. Lancet 2011;378 (9793):804–14.

[5] Pascot A, Lemieux S, Lemieux I, Prud’homme D, Tremblay A, Bouchard C, et al. Age-related increase in visceral adipose tissue and body fat and the metabolic risk profile of premenopausal women. Diabetes Care 1999;22(9):1471–8.

[6] Bjorntorp P. Regional fat distribution—implications for type II diabetes. Int J Obes Relat Metab Disord 1992;16(Suppl. 4):S19–27.

[7] Carr DB, Utzschneider KM, Hull RL, Kodama K, Retzlaff BM, Brunzell JD, et al. Intra-abdominal fat is a major determinant of the National Cholesterol Education Program Adult Treatment Panel III criteria for the metabolic syndrome. Diabetes 2004;53(8):2087–94.

[8] Felber JP, Haesler E, Jequier E. Metabolic origin of insulin resis-tance in obesity with and without type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia 1993;36(12):1221–9.

[9] Albu JB, Kovera AJ, Johnson JA. Fat distribution and health in obesity. Ann N Y Acad Sci 2000;904:491–501.

[10] Lee DE, Kehlenbrink S, Lee H, Hawkins M, Yudkin JS. Getting the message across: mechanisms of physiological cross talk by adipose tissue. Am J Physiol Endocrinol Metab 2009;296(6): E1210–29.

[11] Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modula-tion by nutrients and inflammation. J Clin Invest 2008;118(9): 2992–3002.

[12] Manetta J, Brun JF, Callis A, Mercier J, Prefaut C. Insulin and non-insulin-dependent glucose disposal in middle-aged and young athletes versus sedentary men. Metabolism 2001;50(3):349–54.

[13] Facchini FS, Hua NW, Reaven GM, Stoohs RA. Hyperinsu-linemia: the missing link among oxidative stress and age-related diseases? Free Radic Biol Med 2000;29(12):1302–6.

Page 6: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

13. OBESITY, INSULIN RESISTAN162

[14] Guarner-Lans V, Rubio-Ruiz ME, Perez-Torres I, Banos de MacCarthy G. Relation of aging and sex hormones to metabolic syndrome and cardiovascular disease. Exp Gerontol 2011;46(7): 517–23.

[15] Drolet R, Richard C, Sniderman AD, Mailloux J, Fortier M, Huot C, et al. Hypertrophy and hyperplasia of abdominal adipose tis-sues in women. Int J Obes (Lond) 2008;32(2):283–91.

[16] Chakrabarti P, Kandror KV. Adipose triglyceride lipase: a new target in the regulation of lipolysis by insulin. Curr Diabetes Rev 2011;7(4):270–7.

[17] Cusi K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical im-plications. Gastroenterology 2012;142(4):711–25; e6.

[18] Veilleux A, Caron-Jobin M, Noel S, Laberge PY, Tchernof A. Visceral adipocyte hypertrophy is associated with dyslipidemia independent of body composition and fat distribution in wom-en. Diabetes 2011;60(5):1504–11.

[19] Pasarica M, Sereda OR, Redman LM, Albarado DC, Hymel DT, Roan LE, et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 2009;58(3):718–25.

[20] Hardy OT, Czech MP, Corvera S. What causes the insulin resistance underlying obesity? Curr Opin Endocrinol Diabetes Obes 2012;19(2):81–7.

[21] Senn JJ. Toll-like receptor-2 is essential for the development of palmitate-induced insulin resistance in myotubes. J Biol Chem 2006;281(37):26865–75.

[22] Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 2006;116(11):3015–25.

[23] Fain JN. Release of inflammatory mediators by human adipose tissue is enhanced in obesity and primarily by the nonfat cells: a review. Mediators Inflamm 2010;2010:513948.

[24] Einstein FH, Huffman DM, Fishman S, Jerschow E, Heo HJ, Atzmon G, et al. Aging per se increases the susceptibility to free fatty acid-induced insulin resistance. J Gerontol A Biol Sci Med Sci 2010;65(8):800–8.

[25] Zeyda M, Stulnig TM. Obesity, inflammation, and insulin resistance—a mini-review. Gerontology 2009;55(4):379–86.

[26] Goto M. Inflammaging (inflammation + aging): A driving force for human aging based on an evolutionarily antagonistic pleiot-ropy theory? Biosci Trends 2008;2(6):218–30.

[27] Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol 2011;29:415–45.

[28] Todoric J, Loffler M, Huber J, Bilban M, Reimers M, Kadl A, et al. Adipose tissue inflammation induced by high-fat diet in obese diabetic mice is prevented by n-3 polyunsaturated fatty acids. Diabetologia 2006;49(9):2109–19.

[29] Huber J, Kiefer FW, Zeyda M, Ludvik B, Silberhumer GR, Prager G, et al. CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. J Clin Endocrinol Metab 2008;93(8):3215–21.

[30] Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006;444(7121):860–7.

[31] Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993;259(5091):87–91.

[32] Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. J Clin Invest 2011;121(6):2111–7.

[33] Medzhitov R. Origin and physiological roles of inflammation. Nature 2008;454(7203):428–35.

[34] Medzhitov R. Inflammation 2010: new adventures of an old flame. Cell 2010;140(6):771–6.

[35] Greenberg AS, Obin MS. Obesity and the role of adipose tissue in inflammation and metabolism. Am J Clin Nutr 2006;83(2):461S–5S.

CE, AND INFLAMMAGING

[36] Poitou C, Viguerie N, Cancello R, De Matteis R, Cinti S, Stich V, et al. Serum amyloid A: production by human white adipocyte and regulation by obesity and nutrition. Diabetologia 2005;48(3):519–28.

[37] Kim CS, Park HS, Kawada T, Kim JH, Lim D, Hubbard NE, et al. Circulating levels of MCP-1 and IL-8 are elevated in human obese subjects and associated with obesity-related parameters. Int J Obes (Lond) 2006;30(9):1347–55.

[38] Stienstra R, Tack CJ, Kanneganti TD, Joosten LA, Netea MG. The inflammasome puts obesity in the danger zone. Cell Metab 2012;15(1):10–8.

[39] Vila G, Riedl M, Anderwald C, Resl M, Handisurya A, Clodi M, et al. The relationship between insulin resistance and the cardiovascular biomarker growth differentiation factor-15 in obese patients. Clin Chem 2011;57(2):309–16.

[40] Samaras K, Botelho NK, Chisholm DJ, Lord RV. Subcutaneous and visceral adipose tissue gene expression of serum adipokines that predict type 2 diabetes. Obesity (Silver Spring) 2010;18(5):884–9.

[41] Kvasnicka T, Kvasnicka J, Ceska R, Grauova B, Vrablik M. Increasing plasma levels of soluble cell adhesion molecules (sE-Selectin, sP-Selectin and sICAM-1) in overweight adults with combined hyperlipidemia. Sb Lek 2001;102(4):473–7.

[42] Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998;395(6704):763–70.

[43] Kiefer FW, Zeyda M, Todoric J, Huber J, Geyeregger R, Weichhart T, et al. Osteopontin expression in human and murine obesity: extensive local up-regulation in adipose tissue but minimal systemic alterations. Endocrinology 2008;149(3):1350–7.

[44] Krabbe KS, Pedersen M, Bruunsgaard H. Inflammatory mediators in the elderly. Exp Gerontol 2004;39(5):687–99.

[45] Sansoni P, Vescovini R, Fagnoni F, Biasini C, Zanni F, Zanlari L, et al. The immune system in extreme longevity. Exp Gerontol 2008;43(2):61–5.

[46] Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C- reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 2001;286(3):327–34.

[47] Yusuf S, Hawken S, Ounpuu S, Dans T, Avezum A, Lanas F, et al. Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. Lancet 2004;364(9438):937–52.

[48] Hashimoto I, Wada J, Hida A, Baba M, Miyatake N, Eguchi J, et al. Elevated serum monocyte chemoattractant protein-4 and chronic inflammation in overweight subjects. Obesity (Silver Spring) 2006;14(5):799–811.

[49] Hardy OT, Perugini RA, Nicoloro SM, Gallagher-Dorval K, Puri V, Straubhaar J, et al. Body mass index-independent inflammation in omental adipose tissue associated with insulin resistance in morbid obesity. Surg Obes Relat Dis 2011;7(1):60–7.

[50] Kiefer FW, Zeyda M, Gollinger K, Pfau B, Neuhofer A, Weichhart T, et al. Neutralization of osteopontin inhibits obesity-induced inflammation and insulin resistance. Diabetes 2010;59(4):935–46.

[51] Nomiyama T, Perez-Tilve D, Ogawa D, Gizard F, Zhao Y, Heywood EB, et al. Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J Clin Invest 2007;117(10):2877–88.

[52] Vetrone SA, Montecino-Rodriguez E, Kudryashova E, Kramerova I, Hoffman EP, Liu SD, et al. Osteopontin promotes fibrosis in dystrophic mouse muscle by modulating immune cell subsets and intramuscular TGF-beta. J Clin Invest 2009;119(6):1583–94.

[53] Kiefer FW, Neschen S, Pfau B, Legerer B, Neuhofer A, Kahle M, et al. Osteopontin deficiency protects against obesity-induced hepatic steatosis and attenuates glucose production in mice. Diabetologia 2011;54(8):2132–42.

[54] Chapman J, Miles PD, Ofrecio JM, Neels JG, Yu JG, Resnik JL, et al. Osteopontin is required for the early onset of high fat diet-induced insulin resistance in mice. PLoS One 2010;5(11):e13959.

Page 7: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

COnClU

[55] Nolte Kennedy MS. Chapter 41. Pancreatic Hormones & Antidiabetic Drugs. In: Katzung BG, Masters SB, Trevor AJ, editors. Basic & Clinical Pharmacology. 12e; 2012. http:// www.accessmedicine.com.lp.hscl.ufl.edu/content.aspx?aID=55828662; [accessed 13 04 22].

[56] Kahn CR. Banting Lecture. Insulin action, diabetogenes, and the cause of type II diabetes. Diabetes 1994;43(8):1066–84.

[57] White MF. Insulin signaling in health and disease. Science 2003;302(5651):1710–1.

[58] Nishimura S, Manabe I, Nagai R. Adipose tissue inflammation in obesity and metabolic syndrome. Discov Med 2009;8(41):55–60.

[59] de Luca C, Olefsky JM. Inflammation and insulin resistance. FEBS Lett 2008;582(1):97–105.

[60] Kim JK, Fillmore JJ, Sunshine MJ, Albrecht B, Higashimori T, Kim DW, et al. PKC-theta knockout mice are protected from fat-induced insulin resistance. J Clin Invest 2004;114(6):823–7.

[61] Hotamisligil GS. Role of endoplasmic reticulum stress and c-Jun NH2-terminal kinase pathways in inflammation and origin of obesity and diabetes. Diabetes 2005;54(Suppl. 2):S73–8.

[62] Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, et al. IKK-beta links inflammation to obesity-induced insulin re-sistance. Nat Med 2005;11(2):191–8.

[63] Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, et al. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta. Science 2001;293(5535):1673–7.

[64] Kosteli A, Sugaru E, Haemmerle G, Martin JF, Lei J, Zechner R, et al. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. J Clin Invest 2010;120(10):3466–79.

[65] Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 dia-betes. Nat Rev Mol Cell Biol 2008;9(5):367–77.

[66] Mlinar B, Marc J, Janez A, Pfeifer M. Molecular mechanisms of insulin resistance and associated diseases. Clin Chim Acta 2007;375(1-2):20–35.

[67] Hirosumi J, Tuncman G, Chang L, Gorgun CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Nature 2002;420(6913):333–6.

[68] Emanuelli B, Peraldi P, Filloux C, Sawka-Verhelle D, Hilton D, Van Obberghen E. SOCS-3 is an insulin-induced negative regula-tor of insulin signaling. J Biol Chem 2000;275(21):15985–91.

[69] Ueki K, Kondo T, Tseng YH, Kahn CR. Central role of suppres-sors of cytokine signaling proteins in hepatic steatosis, insulin resistance, and the metabolic syndrome in the mouse. Proc Natl Acad Sci U S A 2004;101(28):10422–7.

[70] Weigert C, Hennige AM, Lehmann R, Brodbeck K, Baumgartner F, Schauble M, et al. Direct cross-talk of interleukin-6 and insulin signal transduction via insulin receptor substrate-1 in skeletal muscle cells. J Biol Chem 2006;281(11):7060–7.

[71] Jager J, Gremeaux T, Cormont M, Le Marchand-Brustel Y, Tanti JF. Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression. Endocrinology 2007;148(1):241–51.

[72] Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcrip-tion factor in chronic inflammatory diseases. N Engl J Med 1997;336(15):1066–71.

[73] Song MJ, Kim KH, Yoon JM, Kim JB. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun 2006;346(3):739–45.

[74] Lee YS, Li P, Huh JY, Hwang IJ, Lu M, Kim JI, et al. Inflammation is necessary for long-term but not short-term high-fat diet-induced insulin resistance. Diabetes 2011;60(10):2474–83.

[75] Lee JY, Ye J, Gao Z, Youn HS, Lee WH, Zhao L, et al. Reciprocal modulation of Toll-like receptor-4 signaling pathways involving MyD88 and phosphatidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids. J Biol Chem 2003;278(39):37041–51.

SIOn 163

[76] Bes-Houtmann S, Roche R, Hoareau L, Gonthier MP, Festy F, Caillens H, et al. Presence of functional TLR2 and TLR4 on human adipocytes. Histochem Cell Biol 2007;127(2):131–7.

[77] Erridge C, Attina T, Spickett CM, Webb DJ. A high-fat meal induces low-grade endotoxemia: evidence of a novel mechanism of postprandial inflammation. Am J Clin Nutr 2007;86(5):1286–92.

[78] Hundal RS, Petersen KF, Mayerson AB, Randhawa PS, Inzucchi S, Shoelson SE, et al. Mechanism by which high-dose aspirin improves glucose metabolism in type 2 diabetes. J Clin Invest 2002;109(10):1321–6.

[79] Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante Jr AW. Obesity is associated with macrophage accumu-lation in adipose tissue. J Clin Invest 2003;112(12):1796–808.

[80] Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obe-sity-related insulin resistance. J Clin Invest 2003;112(12):1821–30.

[81] Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ot-taviani E, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci 2000;908:244–54.

[82] Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 2005;46(11):2347–55.

[83] Fain JN. Release of inflammatory mediators by human adipose tissue is enhanced in obesity and primarily by the nonfat cells: a review. Mediators Inflamm 2006;2010:513948.

[84] Zeyda M, Stulnig TM. Adipose tissue macrophages. Immunol Lett 2007;112(2):61–7.

[85] Boring L, Gosling J, Chensue SW, Kunkel SL, Farese Jr RV, Broxmeyer HE, et al. Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J Clin Invest 1997;100(10):2552–61.

[86] Rollins BJ, Walz A, Baggiolini M. Recombinant human MCP-1/JE induces chemotaxis, calcium flux, and the respiratory burst in human monocytes. Blood 1991;78(4):1112–6.

[87] Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci U S A 2003;100(12):7265–70.

[88] Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest 2006;116(6):1494–505.

[89] Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest 2006;116(1):115–24.

[90] Solinas G, Vilcu C, Neels JG, Bandyopadhyay GK, Luo JL, Naugler W, et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab 2007;6(5):386–97.

[91] Saberi M, Woods NB, de Luca C, Schenk S, Lu JC, Bandyopadhyay G, et al. Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Cell Metab 2009;10(5):419–29.

[92] Lumeng CN, DelProposto JB, Westcott DJ, Saltiel AR. Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 2008;57(12):3239–46.

[93] Holness CL, Simmons DL. Molecular cloning of CD68, a human macrophage marker related to lysosomal glycoproteins. Blood 1993;81(6):1607–13.

[94] Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 2010;11(10):889–96.

[95] Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007;117(1):175–84.

Page 8: Inflammation, Advancing Age and Nutrition || Obesity, Insulin Resistance, and Inflammaging

13. OBESITY, INSULIN RESISTAN164

[96] Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest 2011;121(6):2094–101.

[97] Aron-Wisnewsky J, Tordjman J, Poitou C, Darakhshan F, Hugol D, Basdevant A, et al. Human adipose tissue macrophages: m1 and m2 cell surface markers in subcutaneous and omental depots and after weight loss. J Clin Endocrinol Metab 2009;94(11):4619–23.

[98] Komohara Y, Hirahara J, Horikawa T, Kawamura K, Kiyota E, Sakashita N, et al. AM-3K, an anti-macrophage antibody, recognizes CD163, a molecule associated with an anti-inflammatory macrophage phenotype. J Histochem Cytochem 2006;54(7): 763–71.

[99] Zeyda M, Gollinger K, Kriehuber E, Kiefer FW, Neuhofer A, Stulnig TM. Newly identified adipose tissue macrophage populations in obesity with distinct chemokine and chemokine receptor expression. Int J Obes (Lond) 2011;34(12):1684–94.

[100] Red Eagle A, Chawla A. In obesity and weight loss, all roads lead to the mighty macrophage. J Clin Invest 2010;120(10): 3437–40.

[101] Wentworth JM, Naselli G, Brown WA, Doyle L, Phipson B, Smyth GK, et al. Pro-inflammatory CD11c+CD206+ adipose tissue macrophages are associated with insulin resistance in human obesity. Diabetes 2010;59(7):1648–56.

[102] Mahbub S, Deburghgraeve CR, Kovacs EJ. Advanced age impairs macrophage polarization. J Interferon Cytokine Res 2012;32(1):18–26.

[103] Zeyda M, Huber J, Prager G, Stulnig TM. Inflammation correlates with markers of T-cell subsets including regulatory T cells in adipose tissue from obese patients. Obesity (Silver Spring) 2011;19(4):743–8.

[104] Divoux A, Moutel S, Poitou C, Lacasa D, Veyrie N, Aissat A, et al. Mast cells in human adipose tissue: link with morbid obesity, inflammatory status, and diabetes. J Clin Endocrinol Metab 2012;97(9):E1677–85.

[105] Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 2009;15(8):914–20.

[106] Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 2009;15(8):921–9.

[107] Zeyda M, Huber J, Prager G, Stulnig TM. Inflammation correlates with markers of T-cell subsets including regulatory T cells in adipose tissue from obese patients. Obesity (Silver Spring) 2011;19(4):743–8.

[108] Lee N, Shin MS, Kang I. T-cell biology in aging, with a focus on lung disease. J Gerontol A Biol Sci Med Sci 2012;67(3):254–63.

[109] Liu J, Divoux A, Sun J, Zhang J, Clement K, Glickman JN, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med 2009;15(8):940–5.

[110] Bakker GC, van Erk MJ, Pellis L, Wopereis S, Rubingh CM, Cnubben NH, et al. An antiinflammatory dietary mix modu-lates inflammation and oxidative and metabolic stress in overweight men: a nutrigenomics approach. Am J Clin Nutr 2010;91(4):1044–59.

[111] Zeyda M, Szekeres AB, Saemann MD, Geyeregger R, Stockinger H, Zlabinger GJ, et al. Suppression of T cell signaling by polyunsaturated fatty acids: selectivity in inhibition of mitogen-activated protein kinase and nuclear factor activation. J Immunol 2003;170(12):6033–9.

CE, AND INFLAMMAGING

[112] Itariu BK, Zeyda M, Hochbrugger EE, Neuhofer A, Prager G, Schindler K, et al. Long-chain n-3 PUFAs reduce adipose tissue and systemic inflammation in severely obese nondiabetic patients: a randomized controlled trial. Am J Clin Nutr 2012;96(5):1137–49.

[113] Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: results of the GISSI-Prevenzione trial. Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto miocardico. Lancet 1999;354(9177): 447–55.

[114] Tierney AC, McMonagle J, Shaw DI, Gulseth HL, Helal O, Saris WH, et al. Effects of dietary fat modification on insulin sensitivity and on other risk factors of the metabolic syndrome—LIPGENE: a European randomized dietary intervention study. Int J Obes (Lond) 2011;35(6):800–9.

[115] Ren T, He J, Jiang H, Zu L, Pu S, Guo X, et al. Metformin reduces lipolysis in primary rat adipocytes stimulated by tumor necrosis factor-alpha or isoproterenol. J Mol Endocrinol 2006;37(1):175–83.

[116] Zu L, Jiang H, He J, Xu C, Pu S, Liu M, et al. Salicylate blocks lipolytic actions of tumor necrosis factor-alpha in primary rat adipocytes. Mol Pharmacol 2008;73(1):215–23.

[117] Zeender E, Maedler K, Bosco D, Berney T, Donath MY, Halban PA. Pioglitazone and sodium salicylate protect human beta-cells against apoptosis and impaired function induced by glucose and interleukin-1beta. J Clin Endocrinol Metab 2004;89(10):5059–66.

[118] Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol 2005;6(12):1191–7.

[119] Rumore MM, Kim KS. Potential role of salicylates in type 2 diabetes. Ann Pharmacother 2010;44(7-8):1207–21.

[120] Fernandez-Real JM, Lopez-Bermejo A, Ropero AB, Piquer S, Nadal A, Bassols J, et al. Salicylates increase insulin secretion in healthy obese subjects. J Clin Endocrinol Metab 2008;93(7): 2523–30.

[121] Koska J, Ortega E, Bunt JC, Gasser A, Impson J, Hanson RL, et al. The effect of salsalate on insulin action and glucose tolerance in obese non-diabetic patients: results of a randomised double-blind placebo-controlled study. Diabetologia 2009;52(3):385–93.

[122] DeOliveira CC, Acedo SC, Gotardo EM, Carvalho Pde O, Rocha T, Pedrazzoli Jr J, et al. Effects of methotrexate on inflammatory alterations induced by obesity: an in vivo and in vitro study. Mol Cell Endocrinol 2012;361(1-2):92–8.

[123] Toms TE, Panoulas VF, John H, Douglas KM, Kitas GD. Methotrexate therapy associates with reduced prevalence of the metabolic syndrome in rheumatoid arthritis patients over the age of 60—more than just an anti-inflammatory effect? A cross sectional study. Arthritis Res Ther 2009;11(4):R110.

[124] Drucker DJ, Rosen CF. Glucagon-like peptide-1 (GLP-1) receptor agonists, obesity and psoriasis: diabetes meets dermatology. Diabetologia 2011;54(11):2741–4.

[125] Chen S, Liu AR, An FM, Yao WB, Gao XD. Amelioration of neurodegenerative changes in cellular and rat models of diabetes-related Alzheimer’s disease by exendin-4. Age (Dordr) 2012;34(5):1211–24.

[126] Kang YS, Cha JJ, Hyun YY, Cha DR. Novel C-C chemokine receptor 2 antagonists in metabolic disease: a review of recent developments. Expert Opin Investig Drugs 2011;20(6):745–56.

[127] Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med 2007;356(15):1517–26.

[128] Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol 2011;11(2):98–107.