Skip to main content
American Journal of Lifestyle Medicine logoLink to American Journal of Lifestyle Medicine
. 2014 Oct 27;10(4):268–279. doi: 10.1177/1559827614552986

Immune and Inflammatory Signaling Pathways in Exercise and Obesity

Karsten Krüger 1,2,, Frank C Mooren 1,2, Klaus Eder 1,2, Robert Ringseis 1,2
PMCID: PMC6125063  PMID: 30202282

Abstract

Over the last decades the combination of both a sedentary lifestyle and excessive food availability has led to a significant increase in the prevalence of obesity, which is increasingly recognized as an important risk factor for type 2 diabetes. Several lines of evidence exist demonstrating that expanded visceral adipose tissue produces several pro-inflammatory mediators that activate signaling pathways that contribute to the development of insulin resistance. Exercise training is an important lifestyle factor that is widely used as a tool for preventing and improving lifestyle-related obesity and insulin resistance. In this regard, exercise training is useful to increase energy expenditure thereby counteracting a positive energy balance. Exercise training is also able to attenuate the activation of several obesity-induced pathways of inflammation and oxidative stress. Thus, a better understanding of the molecular mechanisms and immune pathways in exercise, obesity, and diabetes can be extremely useful to exploit optimized lifestyle strategies to combat the increasing incidence of metabolic diseases.

Keywords: adipocytes, physical activity, cytokines, fat tissue, insulin sensitivity


‘It has long been known that exercise training reduces low-grade inflammation and improves insulin sensitivity.’

Obesity is a serious health issue dramatically increasing in an epidemic manner in most countries. Based on estimations from the World Health Organization, 2.3 billion adults will be overweight or obese by the year 2015.1 The main etiologic factors for obesity are excess nutrition and physical inactivity, whereas rare genetic defects as etiologic factors are clearly less frequent, indicating that obesity is dependent on lifestyle and thus preventable in most instances. In recent years, it has become increasingly clear that the pathologic expansion of adipose tissue (AT) typical of obesity leads to the development of insulin resistance (IR)2—a common metabolic complication associated with obesity, which is characterized by an impaired glucose-uptake by insulin sensitive tissues (liver, skeletal muscle, AT). Insulin mediates glucose-uptake through activation of the insulin receptor, which causes phosphorylation of tyrosine residues of the downstream docking protein insulin receptor substrate 1 (IRS-1), subsequently leading to stimulation of a signaling pathway that causes translocation of glucose transporter type 4 (GLUT4) vesicles to the cell surface resulting in cellular glucose uptake.3 IR is critical as it significantly increases the risk of developing several obesity-associated diseases, such as type 2 diabetes mellitus, dyslipidemia, hypertension, coronary heart disease, stroke, metabolic syndrome, nonalcoholic fatty liver disease, and certain cancers.4-7 Development of systemic IR in obesity is explained by the fact that the expanded AT, particularly the visceral adipose tissue (VAT), is not simply a tissue functioning to store triacylglycerols in response to energy excess and to mobilize energy during periods of fasting. Rather, it is a endocrine organ secreting numerous hormone-like mediators, such as adipokines (leptin, resistin, adiponectin), cytokines (interleukin [IL]-6, IL-1β, tumor necrosis factor-α [TNFα]), and chemokines (monocyte chemoattractant protein-1 [MCP-1], macrophage inflammatory protein 1 [MIP1], regulated on activation normal T cell expressed and secreted [RANTES]), reactive oxygen species (ROS), as well as free fatty acids (FFA).8 The circulating levels of leptin, resistin, IL-6, IL-1β, TNFα, MCP-1, ROS, and FFA increase as the adipocytes enlarge and the VAT expands, and which among other negatively affect insulin signaling thereby causing IR.9 In addition, the VAT of obese subjects develops a localized inflammation due to accumulation of immune cells, in particular monocyte-derived macrophages, secreting also pro-inflammatory cytokines and chemotactic factors, which cause recruitment of further immune cells, thereby inducing a vicious cycle of chronic low-grade inflammation.10 Due to secretion of pro-inflammatory cytokines from VAT into the circulation, the inflammation becomes systemic causing activation of diverse cellular inflammatory signaling pathways that converge in the activation of the serine kinases Jun N-terminal kinase (JNK)-1 and inhibitor of kB kinase (IKKβ) in insulin-sensitive tissues.11 Both JNK-1 and IKKβ cause serine phosphorylation of IRS-1, a phosphorylation state associated with impaired insulin signaling and, thereby, systemic IR.12

Increased physical activity as achieved by exercise training is a reasonable approach to support diet-induced weight loss by simultaneously increasing energy expenditure and thereby counteracting a positive energy balance.13 In addition, it has long been known that exercise training reduces low-grade inflammation and improves insulin sensitivity.14-16 In this review, we provide an overview of the most striking obesity-induced changes in immune pathways and their impact on inflammatory signaling pathways mediating IR and the counterregulatory effects of exercise training on these pathways.

Obesity-Induced Changes in Adipose Tissue Physiology and Immune Pathways

The VAT is a multicellular tissue consisting of the adipocyte fraction, which consists of adipocytes and connective tissue with a dense network of capillaries, and the nonadipocyte fraction. The nonadipocyte fraction, also called stromal-vascular fraction, consists of extracellular matrix and different cells including pre-adipocytes, which are mesenchymal stem cells with the ability to differentiate into adipocytes, multipotent stem cells, fibroblasts, endothelial cells, and immune cells including macrophages, T cells, dendritic cells, natural killer cells, and eosinophils.17,18 As described below, the pathologic expansion of the VAT during obesity development is accompanied by prominent morphological changes of the adipocyte and the nonadipocyte fraction, which together contribute to VAT inflammation and systemic IR typical for obesity.

Obesity-Induced Changes in the Adipocyte Fraction

Expansion of VAT occurs by increasing the volume of preexisting adipocytes (adipocyte hypertrophy) and/or by generating new small adipocytes (adipocyte hyperplasia) from pre-adipocytes. However, adipocyte hypertrophy, which is mediated by an increased storage of triacylglycerols by fully differentiated adipocytes leading to an enlargement of adipocytes, contributes most to the increase of AT mass during obesity.19 Adipocyte hypertrophy is critical for developing obesity-related diseases since the adipocyte size is positively correlated with insulin concentrations, IR, and the risk of developing type 2 diabetes.20-22 The crucial role of adipocyte size is further demonstrated by the finding that obese subjects with few large adipocytes are more insulin resistant than those having the same degree of adiposity and small adipocytes.22 One reason for the detrimental effect of enlarged adipocytes on insulin action is that larger adipocytes have a higher basal and stimulated rate of lipolysis leading to chronically elevated circulating levels of FFA, which are known to mediate IR.23 A second important reason is that enlarged adipocytes have an altered secretome, which is characterized by an increased secretion of pro-inflammatory adipokines (eg, leptin, resistin) and cytokines (TNFα, IL-6, IL-1β) and a decreased secretion of anti-inflammatory and insulin-sensitizing adipokines (eg, adiponectin).8 This imbalance between pro- and anti-inflammatory mediators in blood causes IR in insulin-sensitive tissues and promotes a pro-inflammatory environment in VAT leading to immune cell infiltration, which exacerbates the inflammatory process. A further result of adipocyte enlargement during VAT expansion is the development of local hypoxic regions due to hypoperfusion of the expanded VAT and thus insufficient oxygen supply to the enlarged adipocytes.24 The chronic hypoxia found in obese VAT causes hypoxic stress leading to activation of transcription factors that act as sensors of low oxygen tension, such as hypoxia-inducible factor-1α (HIF-1α),25 which stimulates the expression of pro-inflammatory genes and inhibits expression of adiponectin. VAT inflammation is further promoted by the fact that hypertrophic adipocytes degenerate and subsequently die, which explains that adipocyte number remains remarkably stable during weight gain in adults, despite adipocyte hyperplasia from pre-adipocytes. Adipocyte death occurs via a process called pyroptosis involving activation of pro-caspase-1 within the nucleotide-binding oligomerization domain (NOD)-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome leading to the release of active caspase-1, which cleaves the cytokine precursors pro-IL-1β and pro-IL-18 into active IL-1β and IL-18, respectively.26,27 The resulting adipocyte death via inflammasome activation, thus, is inflammation-associated and contributes also to the local VAT inflammation and triggers the migration of monocyte-derived macrophages into VAT.28 The majority of infiltrated macrophages localize around dead or dying adipocytes (identifiable by the lack of perilipin staining) thereby forming crown-like structures (CLS).29 The CLS-associated macrophages phagocytose the cellular debris from dead adipocytes or their residual lipid droplets leading to a foam cell-like appearance. CLS formation in response to adipocyte death in obese subjects is an early event of VAT inflammation and highly correlated with IR.30 Within weeks it can be observed that new adipocytes arising from pre-adipocytes of the stromal-vascular fraction populate the AT, a process called AT remodeling, which causes further AT expansion.31 However, in the case of treatment of obese subjects with the thiazolidinedione group of insulin sensitizers this AT remodeling can be influenced in a beneficial manner, since thiazolidinediones cause the generation of small, insulin-sensitive adipocytes resulting in an improvement of systemic insulin sensitivity despite AT expansion.32

Obesity-Induced Changes in the Stromal-Vascular Fraction

Infiltration and accumulation of monocyte-derived macrophages in VAT triggered by inflammation-associated death of adipocytes is an early event during VAT expansion and weight gain in both humans and rodents.10,28,33,34 Recruitment of monocytes from the circulation in obesity is mediated by chemokines, such as MCP-1 as well as macrophage inflammatory protein (MIP)1α, MIP1β, and regulated on activation normal T cell expressed and secreted (RANTES), released from enlarged adipocytes and resident VAT macrophages.35 The MCP-1 ligand has high affinity for C-C motif receptors (CCR) 2 and 5 expressed on the cell surface of monocytes and binding of MCP-1 to CCR2 and CCR5 markedly stimulates monocyte infiltration into AT in different mouse models of obesity.36-38 The VAT macrophages are phenotypically classified based on their activation/polarization state into “classically activated” pro-inflammatory M1 macrophages, which produce large amounts of pro-inflammatory cytokines (TNFα, IL-1β, IL-6) and generate ROS via inducible nitric oxide synthase, and “alternatively activated” non- or anti-inflammatory M2 macrophages, which have low pro-inflammatory cytokine expression and instead secrete high levels of anti-inflammatory cytokines, such as IL-10 and IL-1 receptor agonist.39,40 Diet-induced obesity was found to induce a phenotypic switch in AT macrophage polarization from an M2-polarized state in lean animals that may protect adipocytes from inflammation to an M1 pro-inflammatory state that contributes to VAT inflammation and IR.41 FFAs that are elevated in obesity exacerbate pro-inflammatory cytokine secretion from M1 macrophages through activation of TLR4 receptor, which induces IL-6, IL-1β, and MCP-1 expression in genetically and diet-induced obese mice.42 Interestingly, lack of CCR5, but not CCR2, in mice causes a shift of the macrophage population toward M2 macrophages indicating that the chemokine CCR5 promotes VAT inflammation not only by recruiting macrophages into VAT but also by causing a shift to the pro-inflammatory M1 phenotype,43 which favors the development of IR. The switch from M2 to M1 polarization of VAT macrophages is not irreversible but can be reverted by administration of thiazolidinediones to obese mice or by switching obese mice from a high-fat diet to a standard diet.44,45

Besides macrophages, several other immune cells in AT have been shown to play a role in obesity and AT inflammation-associated IR. In particular, significant alterations in the number of different subsets of T lymphocytes in VAT during obesity have been reported in different genetic and diet-induced mouse models of obesity. The number of CD8+ T cells was shown to increase in VAT, which, in turn, initiates and propagates an inflammatory cascade involving secretion of pro-inflammatory cytokines and chemokines, thereby promoting accumulation of macrophages in AT of mice in response to a high-fat diet.46 The finding that CD8-specific antibody treatment ameliorates preestablished AT inflammation in these mice indicates that CD8+ T cells are also essential for maintenance of the inflammatory response.46 In contrast, the number of CD4+ regulatory T cells (Tregs) was reported to decrease in the expanding VAT of mice in response to a high-fat diet.46,47 This also exacerbates AT inflammation, because Tregs secrete anti-inflammatory cytokines that inhibit macrophage migration and promote the noninflammatory M2 polarization state of macrophages.47 The number of pro-inflammatory Th1 cells, which differentiate from mature CD4+ T cells and produce mainly IFN-γ, increase in VAT of mice during obesity, which promotes VAT inflammation and systemic IR through activation of macrophages and antagonizing the anti-inflammatory properties of Tregs and Th2 cells.48,49 Th2 cells, which also differentiate from mature CD4+ T cells, are anti-inflammatory due to secretion of anti-inflammatory cytokines (IL-4, IL-13),50 from which IL-4 induces the noninflammatory M2 polarization state of VAT macrophages. In addition, evidence was provided from different animal models of obesity that obesity causes changes in the number of mast cells,51,52 eosinophils,53,54 and natural killer T (NKT) cells.55 However, the biological role of these obesity-induced changes in AT immune cell number and composition, for example, for VAT inflammation and IR, is yet largely undefined. Further outstanding questions originate from the fact that only few data are available with regard to the presence of specific immune cells in human obesity, because human AT, in particular VAT, is not readily available for clinical assessment. Thus, it remains to be demonstrated whether the AT immune cells found in obese mouse models are equivalently found in obese humans and whether they play similar roles.

Obesity-Induced Changes in Inflammatory Signaling Pathways

Despite activation of different intracellular signaling pathways, it is common to the inflammatory mediators secreted from adipocytes and immune cells that insulin sensitivity of target tissues is compromised through negatively interfering with insulin signaling. How insulin signaling in insulin-sensitive tissues (liver, skeletal muscle, AT) is impaired by these mediators in obesity is exemplified below for some of the most important representatives.

Leptin

Leptin is one of several adipokines secreted from adipocytes, and it has long been known that obese subjects have elevated circulating concentrations of leptin compared with normal-weight subjects.56-58 In addition, circulating leptin levels were found to positively correlate with AT mass and adipocyte size.9 Conversely, weight loss decreases circulating concentrations and/or AT expression of leptin in obese subjects.58-60 Leptin exerts its effects in tissues by binding to the leptin receptor at the plasma membrane leading to recruitment and binding of cytoplasmic Janus tyrosin kinases (JAKs), which subsequently become transphosphorylated resulting in activation of signal transducer and activator of transcription (STAT)3 and STAT5.61,62 Through this, acute-phase proteins and pro-inflammatory transcription factors, such as c-fos, c-jun, and activator protein (AP)-1, are induced.61,62 Autophosphorylation of JAKs results also in activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway leading to stimulation of nuclear factor-κB (NF-κB).61,62 Stimulation of NF-κB and other pro-inflammatory transcription factors (AP-1, STATs) is crucial for development of IR because these transcription factors causes upregulation of genes encoding pro-inflammatory mediators, like TNFα and IL-1β, which negatively interfere with insulin signaling.63 In addition, NF-κB can be stimulated by leptin by activating p38 mitogen-activated protein kinase (MAPK) and stress-activated JNK-1.64 Activation of JNK-1 directly impairs insulin signaling via serine phosphorylation of the IRS-1, the phosphorylation state of IRS-1, which is linked to IR and reduced glucose uptake.12 JNK-1 inhibits insulin sensitivity also in a similar manner as NF-κB, AP-1, and STATs through stimulating transcription of pro-inflammatory genes.12

Adiponectin

Unlike leptin, the circulating concentrations of adiponectin, for which 3 major isoforms exist (low molecular weight, middle-molecular weight, and high molecular weight adiponectin), negatively correlate with adipocyte size in human subjects, for example, levels of adiponectin in the circulation are reduced in obese subjects compared with normal-weight subjects.65 Reduced circulating levels of adiponectin, which is best reflected by the high molecular weight form,66 in obesity are explained by inhibition of their secretion from cultured adipocytes by pro-inflammatory cytokines like TNFα and IL-6,67 which are found at elevated levels in obese humans.58,68,69 Conversely, weight loss, which is accompanied by an improvement of insulin sensitivity, induces AT adiponectin expression and/or increases blood levels of adiponectin in obese humans and genetically obese animals, indicating a beneficial role of adiponectin on insulin signaling.70-72 The insulin sensitizing effect of adiponectin is mediated by binding of adiponectin to its receptors (AdipoR1 and AdipoR2) in target tissues, such as liver and skeletal muscle.73 Activation of AdipoR1 and AdipoR2 in these tissues stimulates the peroxisome proliferator-activated receptor α(PPARα) pathway, which leads to increased fatty acid oxidation, and inhibition of the NF-κB pathway, which results in a reduced production of pro-inflammatory cytokines.74 In addition, treatment of different myeloid cells with adiponectin was shown to increase the secretion of anti-inflammatory cytokines, like IL-10 and IL-1 receptor antagonist,75 an effect that is expected to counteract the inhibitory effects of pro-inflammatory cytokines on insulin signaling. Moreover, studies in mice and rats revealed that the insulin sensitizing effect of adiponectin involves activation of the adenosine monophosphate-activated protein kinase (AMPK) pathway.76,77 AMPK is a principal regulator of energy metabolism homeostasis, which stimulates glucose utilization in skeletal muscle and suppresses gluconeogenesis in the liver.76 In addition, AMPK inhibits inflammatory responses induced by NF-κB activation as well as endoplasmic reticulum stress,77 which occurs in different metabolic disorders including obesity and diabetes. In line with the antidiabetic effects of adiponectin, it was recently shown that AdipoR agonists also activate of AMPK and hepatic and muscle PPARα, ameliorate IR and glucose intolerance in mice fed a high-fat diet, but not in AdipoR1 and AdipoR2 double-knockout mice.78

FFA

It has long been known that circulating levels of FFA are chronically elevated in most obese subjects.79 This is due to the fact that the expanded AT releases more FFA due to a stimulation of basal lipolysis in AT,23 and once elevated, FFA inhibit the antilipolytic action of insulin further increasing FFA release from AT into the circulation.80 Elevated FFA levels are considered to be a key factor in developing systemic IR and ultimately type 2 diabetes during obesity. This is based on the observation that pharmacological reduction of circulating FFA levels in obese and type 2 diabetic subjects improves peripheral insulin sensitivity.81,82 Several mechanisms have been proposed to explain the IR-inducing effect of elevated FFA levels in obesity. Circulating FFA, in particular saturated fatty acids, impair insulin sensitivity through binding to the plasma membrane receptor toll-like receptor 4 (TLR4) in tissues of obese animals,83 which leads to activation of signaling proteins, such as IKK, JNK, and MAPK, that negatively interfere with insulin signaling. The critical role of TLR4 for developing IR is shown by the fact that TLR4 knockout mice are protected from IR and weight gain when fed a high-fat diet compared to wild-type controls.84 In addition, acutely increasing plasma FFA levels in vivo was found to activate NF-κB in human skeletal muscle.85 This effect leads to an increased expression of pro-inflammatory cytokines and elevated circulating levels of MCP-1 as shown in rats.86 The latter chemokine acts as a chemotactic signal for immune cells promoting macrophage recruitment into the AT, thereby propagating the inflammatory process. Interestingly, elevated levels of FFA are also known to cause endoplasmic reticulum (ER) stress.87 In line with this, ER stress is also found in the liver of obese mice, which have also elevated blood levels of FFA.88 ER stress describes an imbalance between the folding capacity of the ER and the amount of proteins to be folded leading to accumulation of unfolded or misfolded proteins in the ER lumen.89 As an adaptive response to this ER stress, the cell activates the so-called unfolded protein response (UPR), which is initiated by 3 ER stress sensors: inositol-requiring enzyme 1 (IRE1), RNA-dependent protein kinase (PKR)-like ER kinase (PERK), and activating transcription factor 6 (ATF6).89 ER stress-induced UPR is proposed to mediate IR because IRE1 and PERK cause activation of JNK and IKKβ signaling pathways, which inhibit insulin signaling via serine phosphorylation of IRS-1 and activation of the pro-inflammatory transcription factors NF-κB and AP-1.89 Noteworthy, it was recently shown that TLR4 deficiency protects against high-fat diet-induced ER stress and IR in the main organs for glucose and lipid metabolism (skeletal muscle, liver, AT) in mice, indicating that this receptor, which is activated by elevated FFA levels, is crucial for mediating the inhibitory effect of elevated FFA levels on insulin signaling.90 It also has been proposed that the inhibitory effect of elevated FFA levels on insulin signaling is mediated by intracellular accumulation of toxic lipid derivatives in response to elevations of FFA levels,91,92 for example, increased plasma FFA levels result in the accumulation of intramuscular long-chain fatty (LCFA) acyl-CoAs, diacylglycerol (DAG), triacylglycerol, and ceramide. Both LCFA-CoAs and DAG were shown to impair insulin signaling via activation of protein kinase C (PKC), which increases serine phosphorylation of IRS-1. A similar mechanism was found for ceramide, but ceramide was also shown to interact with Akt signaling.91,92 Activation of Akt inhibits insulin signaling through stimulation of NF-κB.

Pro-Inflammatory Cytokines

A large number of studies have demonstrated that AT expression and/or circulating levels of pro-inflammatory cytokines, such as IL-6 and TNFα, are elevated in insulin-resistant overweight and obese subjects58,68,69,93 and in animal models of obesity and IR.94 In contrast, weight loss in overweight and obese subjects were accompanied by improvements in insulin sensitivity and decreases in circulating levels and/or AT expression of cytokines.58,59,69,95-99 These observations suggest that pro-inflammatory mediators contribute to the obesity-induced impairment of IR. Pro-inflammatory cytokines mediate their actions through specific receptors located at the cell membrane (IL-1R, IL-6R, TNFR). On activation of these receptors different signaling cascades are stimulated that converge at the MAPK and the IKKβ pathway leading to JNK and NF-κB activation100,101) and through serine phosphorylation of IRS-1 to impairment of insulin signaling.12 In addition, activation of NF-κB and other pro-inflammatory transcription factors, such as AP-1 and STAT3, by these cytokines promotes the production of further cytokines in a feed-forward manner, which also induces IR.

ROS

It has been shown that human obesity is positively correlated with systemic oxidative stress on the basis of urinary levels of 8-epi-prostaglandin F2α (8-epi-PGF2α)102 and that systemic oxidative stress is associated with visceral fat accumulation, IR, and metabolic syndrome in humans.103,104 Oxidative stress occurs when the production of ROS exceeds their clearance by the antioxidant defense system. Interestingly, the oxidative stress levels in obese mice were found to be elevated in plasma and AT but not in liver and skeletal muscle compared to control mice, indicating that the AT in obese individuals may represent a major source of ROS.103 Production of ROS in AT is proposed to be the result of an increased expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a plasma membrane enzyme that converts molecular oxygen to superoxide radicals, and decreased antioxidant levels103; for example, the expression and activity of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD) 1, and glutathione peroxidase (GPX) is known to be decreased in the AT of obese individuals.105 ROS are known to activate several signaling pathways, which negatively affect insulin signaling, such as the MAPK, JNK, and NF-κB pathways. In addition, JNK activation potentiates oxidative stress through promoting ROS accumulation.12

Effect of Exercise on Obesity-Induced Changes in Adipose Tissue Physiology and Immune Pathways

Several studies provided evidence that physical activity exhibits multiple effects on obesity and VAT.106 Observational studies demonstrated that there is an inverse relationship between measures of physical activity (total activity as well as exercise training) and measures of fat mass and distribution. Complementary, longitudinal studies revealed that exercise training programs are directly responsible for a reduction of body weight and favorable changes of body composition.107 It is assumed that the exercise-induced increase of total energy expenditure and fat oxidation generates an energy deficit. The loss of body fat is mostly accompanied by the improvement of several obesity-related metabolic and cardiovascular disorders like IR, dyslipidemia, and hypertension.108

In light of the pathologic significance of VAT, it is of considerable interest whether exercise interventions are capable of preferentially targeting VAT. Numerous studies provided evidence for an exercise-induced reduction of VAT. Noteworthy, Ross et al109 demonstrated that an endurance exercise-induced increase of energy expenditure by 700 kcal/day for 14 weeks was followed by a weight loss of 7.5 kg and a decrease in VAT of 52 cm2 corresponding to a decrease of 6.9 cm2 VAT fat per kg of weight loss in premenopausal women with abdominal obesity. Similarly, it was shown that overweight postmenopausal women, who exercised (treadmill walking, stationary bicycling, or strength training) at moderate-intensity 5 days/week for 12 months, lost about 8.5 cm2 of VAT and 1.3 kg of body weight, corresponding to a ratio of 6.5 cm2/kg of weight change.110 In addition, a systematic review of human and clinical trials provided evidence that there is a dose–response relationship between amount of aerobic exercise, such as brisk walking, light jogging, or stationary ergometer usage, and reduction in VAT mass.111 Regarding the physiological mechanisms of exercise-induced VAT reduction, it is known that exercise induces an increase in AT lipolysis, which is attributed to elevated catecholamine concentrations and a decrease in insulin concentrations. It is assumed that VAT is more responsive to adrenergic innervation than other AT depots and therefore benefits more from activity.112

Exercise-Induced Changes in the Adipocyte Fraction

Effects of exercise-induced reduction of VAT mass could be the result of both reduction in adipocyte size and adipocyte number. In this regard, cross-sectional studies revealed that exercise trained subjects exhibit smaller adipoctyes in the abdominal region than aged-matched sedentary controls.113 Interventional studies suggest that exercise training directly reduces adipocyte cell size.114 A reduction of adipocyte size might be a major reason for exercise effects on IR because smaller adipocytes have lower basal rates of lipolysis leading to a reduction of FFA levels in the circulation. Furthermore, smaller adipocytes are known to have a secretome, which is characterized by a reduced expression of pro-inflammatory adipokines (eg, leptin, resistin) and cytokines.115 In contrast, results regarding the effect of exercise training on adipocyte cell number are inconsistent. These inconsistencies are probably partially due to methodological difficulties to determine adipocyte cell number with sufficient accuracy, because most studies estimated adipocyte cell number by calculating the body fat to adipocyte size ratio, which however does not take into account that adipocyte number and size is highly variable.114 A more recent study, in which the adipocyte cell number was assessed from the DNA content of the fat pad, reported that endurance exercise training induced a significant reduction in cell number in VAT of high fat diet–fed rats.116 Thus, while it is beyond question that exercise reduces adipocyte cell size, more studies are necessary to clarify whether exercise-induced reductions in VAT mass can be attributed also to a decrease of adipocyte number.

During exercise, fat oxidation steadily increases resulting in a breakdown of stored triacylglycerols.117 In particular, moderate exercise up to intensities of 60% to 65% of VO2max leads to an increased rate of appearance of FFA in the circulation.118 It has been proposed that subcutaneous AT contributes most to this increase of circulating FFA levels during exercise, whereas the contribution of VAT to this exercise-induced increase of circulating FFA levels is less. Furthermore, intramuscular triacylglycerol stores provide fatty acids as energy substrates during exercise.119 The release of fatty acids from VAT during exercise is influenced by the AT blood flow, the rate of lipolysis, and the rate of fatty acid re-esterification. Important mediators of these effects have been demonstrated to be elevated plasma levels of catecholamines and decreased plasma levels of insulin. In addition, the atrial natriuretic peptide (ANP), which is secreted during exercise,120 has been shown to contribute to lipid mobilization during exercise. The increase of AT blood flow is assumed to attenuate hypoxic stress of adipocytes leading to a reduced translocation of HIF-1α into the nucleus. In this regard, it was demonstrated that endurance exercise training increased expression of AT vascular endothelial growth factor a (VEGFa), an important factor of tissue angiogenesis, and lowered AT lactate levels, an indicator of AT hypoxia, in obese rats. However, these effects are depot-specific and were only demonstrated in intra-abdominal AT.121

Exercise-Induced Changes in the Stromal-Vascular Fraction

Exercise training exhibits several direct and indirect effects on the stromal-vascular fraction in VAT. Regarding macrophages, it has been demonstrated that exercise training (treadmill running at 12-20 m/min, 60 min/day, 16 weeks) attenuated the infiltration and accumulation of inflammatory cells in AT of diet-induced in mice.122 These authors also showed that exercise training affects the macrophage phenotype as demonstrated by a reduced expression of M1 macrophage markers and an increased expression of M2 markers, indicating that exercise training induces phenotypic switching from M1 macrophages to M2 macrophages in AT of obese mice.122 These changes appear to result in a reduced inflammatory state of AT in general, because the reduction of inflammatory macrophages was accompanied by a decrease in the mRNA levels of several inflammatory genes including TNFα, MCP-1, and adhesion molecules in AT of diet-induced obese mice.94 The exercise training–induced amelioration in inflammation status in VAT of diet-induced obese mice has been attributed to a decreased expression and activation of TLR4,122,123 which promotes inflammatory gene expression and IR through activating IKKβ and JNK in diet-induced obesity.124 In contrast to these data from animal studies, only few data are available from human studies. For instance, the combination of exercise training (eg, walking, swimming, aerobics) and hypocaloric diet was reported to reduce macrophage infiltration and inflammatory gene expression in AT of severely obese subjects.16 In addition, it has been shown that exercise training (aerobic and resistance) reduces also expression of pro-inflammatory genes involved in macrophage recruitment, including TLR4, TNFα, and IL-6 in skeletal muscle of obese humans.125 Moreover, Roberts et al126 demonstrated that male diabetic subjects exhibit a reduced expression of pro-inflammatory adhesion molecules, which also play a critical role for macrophage recruitment, after taking part in a daily walking exercise program.

Data about the effects of exercise training on the accumulation of other leukocyte subpopulations in VAT are limited. For both CD4+ and CD8+ lymphocytes, which are known to accumulate during obesity in AT, a reduction of cell number was observed after exercise training in mice.122 However, the underlying mechanisms, the biological significance, and the transferability of data from animal studies to humans have yet to be clarified.

Exercise-Induced Changes in Inflammatory Signaling Pathways

Leptin

Several studies are available about the effects of endurance exercise training on leptin blood levels in humans. Interestingly, the decrease of leptin blood levels were regularly accompanied by a reduction of AT mass.127,128 In line with this, in one study that reported no changes of blood leptin levels there was also no change in body AT mass.129 Besides endurance training, it was also shown that blood leptin levels decreased after 6 months of strength training interventions in overweight inactive elderly persons, with the magnitude of leptin changes increasing with increasing intensity of resistance exercise.130 Regarding the effect of exercise on leptin expression in AT, divergent results were obtained from human and animal studies. While no changes in AT leptin expression after 12 weeks of aerobic exercise training in humans have been reported,131 treadmill running (5 days/week, 6 or 12 week, 40 min/day, 65% to 70% VO2max) reduced the expression of leptin mRNA in VAT of obese mice.132 One might speculate that the exercise-induced reduction of AT leptin expression in obese mice leads to a reduced stimulation of the JAK/STAT and NF-κB signaling pathways, thereby leading to a reduced inflammation.

Adiponectin

Several studies investigated the effects of exercise training on blood levels of adiponectin. While some studies found no changes, most others found increases in serum adiponectin after running training in obese or diabetic subjects.16,130 In human studies, these increases were often accompanied by a reduction of body mass index or body fat. Since adiponectin exerts effects by activation of specific receptors, the effect of exercise was also investigated on AdipoR1 and AdipoR2 receptors. In this regard, it was shown that exercise training of obese mice is followed by an upregulation of adipoR1-2/APPL1 protein levels in liver, AT, and skeletal muscle.133 These increases in receptor expression were accompanied by an improvement of insulin sensitivity. On binding to its receptors, adiponectin induces activation of the PPARα pathway, which is also known to be activated by exercise training.134 PPARα activation is known to increase fatty acid oxidation and to inhibit NF-κB activation. Accordingly, the effect of exercise on adiponectin contributes, at least partially, to the insulin sensitizing and anti-inflammatory effect of exercise.

FFA

Besides serving as important energy substrates in skeletal muscle during exercise, FFA also activate several signaling pathways including MAPK and JNK during exercise.135 Activation of MAPK and JNK signaling by FFA during endurance exercise in skeletal muscle has been shown to be transduced by the transmenbrane proteins TLR2 and TLR4,136,137 which are activated by FFA by a yet unknown mechanism. Although TLR4 activation in response to chronically elevated FFA levels in obese subjects was shown to impair insulin action due to activation of MAPK and JNK,83 it is expected that TLR activation by FFA during acute exercise does not impair peripheral insulin sensitivity. This assumption is based on the fact that TLR2/4 activation by the elevated FFA levels during exercise is an acute effect,136 whereas plasma FFA levels decrease in the long term in response to chronic exercise due to a loss of body weight. In line with the divergent effect of FFA-induced TLR activation on insulin sensitivity between acute exercise and obesity, it was found that the increase of plasma FFA concentration during acute exercise was stronger in TLR2 knockout and TLR4 knockout mice than in wild-type mice, indicating that TLR2 and TLR4 also regulate metabolic functions in tissues or modify repartition of energy substrates during acute exercise.136 However, most data are from animal studies or cell culture experiments and need to be interpreted with caution.

The elevated levels of FFA in obesity are also known to cause ER stress,87 and it was shown that TLR4 is an important mediator of the effect of FFA on obesity-induced ER stress as evidenced from the finding that TLR4 deficiency protects against high fat diet–induced ER stress and IR in liver, muscle, and AT.90 In contrast, da Luz et al138 found that swimming training (1 hour per day, 5 days for week with 5% overload of the body weight for 8 weeks) leads to a reduction in ER stress in AT and liver of diet-induced obese rats, and an improvement of insulin signaling indicating again that FFA-induced TLR activation has different effects on insulin sensitivity between acute exercise and obesity.

To sum up, although TLR activation in response to elevations of plasma FFA levels can be observed during both obesity and acute bouts of exercise, the impact on insulin sensitivity is different between exercise and obesity because FFA levels are only acutely elevated during exercise, while they are chronically elevated in obesity.

Pro-Inflammatory Cytokines

It is well known that both acute bouts of exercise and exercise training influence the secretion of various adipokines, chemokines, and cytokines.139 One of the most responsive cytokines to exercise is IL-6. Several studies presented evidence that IL-6 is expressed in muscle fibers during exercise,140 and activates skeletal muscle AMPK and/or phosphatidylinositol-3-kinase (PI3K) to increase fat oxidation and glucose uptake. Moreover, IL-6 is released from contracting skeletal muscle to the circulation141 and elicits metabolic effects in AT like stimulation of lipolysis and fat oxidation,142 thereby improving the obesity-associated atherogenic lipid profile. Furthermore, IL-6 belongs to several anti-inflammatory cytokines, which are elevated in response to exercise training. In this regard, it has been speculated that the anti-inflammatory cytokine cascade leads to a suppression of TNFα production and thereby offers protection against TNFα-induced IR.142

The results about exercise-induced changes of TNFα expression and blood levels are conflicting. While Straczkowski et al143 demonstrated a significant decrease of serum levels of TNFα and soluble fraction of TNFα receptor 2 in obese woman after 12 weeks of endurance training, Bruun et al16 found no changes in AT expression of TNFα after several weeks of aerobic exercise training. In contrast, in obese mice it was shown that exercise training suppresses TNFα expression in AT.132 Regarding IL-1β, it was reported that intense exercise training (treadmill running) over 7 weeks with weight loss reduces concentrations of IL-1β in AT of rats.144 Similarly, swimming exercise was found to reduce IL-1β serum levels of diet-induced obese mice.145 A recent systemic review has highlighted that acute and chronic exercise may elicit different responses on TNFα expression depending on the health status of subjects, exercise type, and exercise intensity.146 On the one hand, there are some studies that demonstrated that the immune response to exercise differs in subjects with already increased baseline cytokine levels.147,148 On the other hand, more needs to be understood about the nature of exercise that reduce inflammation in patients with chronic inflammatory diseases. In addition, the contribution of an exercise-induced reduction in visceral fat versus other exercise-induced anti-inflammatory mechanisms needs to be better understood.

ROS

Several studies demonstrated that acute exercise is accompanied by an increased generation of ROS. The sources of ROS during exercise include increased mitochondrial oxygen reduction, activation of xanthine oxidase, inflammation, and heme auto-oxidation.149 Although increased ROS formation is widely considered to be detrimental due to causing oxidative damage to cellular lipids, proteins, and DNA, it is becoming increasingly clear that ROS also improve cellular protection mechanisms against ROS through upregulating antioxidant enzymes. In this regard, several studies reported that exercise training induces activities of antioxidant enzymes and the levels of cellular antioxidants in body fluids.150 In muscle and AT, it was demonstrated for instance that both protein levels and activity of SOD, which reduces the superoxide radical to hydrogen peroxide (H2O2), are increased by exercise training. Likewise, in experimental animals it was demonstrated that exercise is able to increase expression of SOD in AT151 and to prevent the increase in the levels of lipid peroxidation and protein oxidation after feeding a high-fat diet. In addition, it was shown that exercise training attenuates protein expression of the NADPH oxidase NOX2, a major source of ROS production in obese AT, in AT of rats.151 Taken together, exercise training causes anti-oxidative effects in tissues by increasing resistance to oxidative stress, thereby decreasing the basal levels of oxidative damage.

Conclusion

An expanded VAT acts as an endocrine organ that secretes various substances that mostly negatively affect insulin signaling. Exercise is an important lifestyle factors that is known to affect fat metabolism, inflammation, and the oxidative capacity of an organism. Considering the data now reviewed, there is a cluster of evidence that exercise training affects several important mediators in these processes that might attenuate pathological consequences of obesity. However, further research is required to better understand the effects of exercise on complete signaling pathways. It would be also helpful to focus more properly on the type, duration, and intensity of exercise training to maximize the benefits in patients.

References

  • 1. World Health Organization. Preventing Chronic Diseases: A Vital Investment. Geneva, Switzerland: World Health Organization; 2005. [Google Scholar]
  • 2. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444:840-846. [DOI] [PubMed] [Google Scholar]
  • 3. González-Sánchez JL, Serrano-Ríos M. Molecular basis of insulin action. Drug News Perspect. 2007;20:527-531. [DOI] [PubMed] [Google Scholar]
  • 4. Rask-Madsen C, Kahn CR. Tissue-specific insulin signaling, metabolic syndrome, and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2012;32:2052-2059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Khandekar MJ, Cohen P, Spiegelman BM. Molecular mechanisms of cancer development in obesity. Nat Rev Cancer. 2011;11:886-895. [DOI] [PubMed] [Google Scholar]
  • 6. Cusi K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 2012;142:711-725. [DOI] [PubMed] [Google Scholar]
  • 7. Klop B, Elte JW, Cabezas MC. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients. 2013;5:1218-1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Galic S, Oakhill JS, Steinberg GR. Adipose tissue as an endocrine organ. Mol Cell Endocrinol. 2009;316:129-139. [DOI] [PubMed] [Google Scholar]
  • 9. Skurk T, Alberti-Huber C, Herder C, Hauner H. Relationship between adipocyte size and adipokine expression and secretion. J Clin Endocrinol Metab. 2007;92:1023-1033. [DOI] [PubMed] [Google Scholar]
  • 10. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796-1808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw. 2006;17:4-12. [PubMed] [Google Scholar]
  • 12. Solinas G, Karin M. JNK and IKKβ: molecular links between obesity and metabolic dysfunction. FASEB J. 2010;24:2596-2611. [DOI] [PubMed] [Google Scholar]
  • 13. Zachwieja JJ. Exercise as treatment for obesity. Endocrinol Metab Clin North Am. 1996;25:965-988. [DOI] [PubMed] [Google Scholar]
  • 14. Cesari M, Penninx BW, Pahor M, et al. Inflammatory markers and physical performance in older persons: the InCHIANTI study. J Gerontol A Biol Sci Med Sci. 2004;59:242-248. [DOI] [PubMed] [Google Scholar]
  • 15. Kohut ML, McCann DA, Russell DW, et al. Aerobic exercise, but not flexibility/resistance exercise, reduces serum IL-18, CRP, and IL-6 independent of beta-blockers, BMI, and psychosocial factors in older adults. Brain Behav Immun. 2006;20:201-209. [DOI] [PubMed] [Google Scholar]
  • 16. Bruun JM, Helge JW, Richelsen B, Stallknecht B. Diet and exercise reduce low-grade inflammation and macrophage infiltration in adipose tissue but not in skeletal muscle in severely obese subjects. Am J Physiol Endocrinol Metab. 2006;290:E961-E967. [DOI] [PubMed] [Google Scholar]
  • 17. Maumus M, Peyrafitte JA, D’Angelo R, et al. Native human adipose stromal cells: localization, morphology and phenotype. Int J Obes (London). 2011;35:1141-1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Mitchell JB, McIntosh K, Zvonic S, et al. The immunophenotype of human adipose derived cells: temporal changes in stromal- and stem cell-associated markers. Stem Cells. 2006;24:376-385. [DOI] [PubMed] [Google Scholar]
  • 19. Spalding KL, Arner E, Westermark PO, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453:783-787. [DOI] [PubMed] [Google Scholar]
  • 20. Lundgren M, Svensson M, Lindmark S, Renström F, Ruge T, Eriksson JW. Fat cell enlargement is an independent marker of insulin resistance and “hyperleptinaemia.” Diabetologia. 2007;50:625-633. [DOI] [PubMed] [Google Scholar]
  • 21. Stern JS, Batchelor BR, Hollander N, Cohn CK, Hirsch J. Adipose-cell size and immunoreactive insulin levels in obese and normal-weight adults. Lancet. 1972;2:948-951. [DOI] [PubMed] [Google Scholar]
  • 22. Weyer C, Foley JE, Bogardus C, Tataranni PA, Pratley RE. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia. 2000;43:1498-1506. [DOI] [PubMed] [Google Scholar]
  • 23. Kosteli A, Sugaru E, Haemmerle G, et al. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. J Clin Invest. 2010;120:3466-3479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Kabon B, Nagele A, Reddy D, et al. Obesity decreases perioperative tissue oxygenation. Anesthesiology. 2004;100:274-280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Jantsch J, Chakravortty D, Turza N, et al. Hypoxia and hypoxia-inducible factor-1 alpha modulate lipopolysaccharide-induced dendritic cell activation and function. J Immunol. 2008;180:4697-4705. [DOI] [PubMed] [Google Scholar]
  • 26. Giordano A, Murano I, Mondini E, et al. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J Lipid Res. 2013;54:2423-2436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Miao EA, Leaf IA, Treuting PM, et al. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat Immunol. 2010;11:1136-1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Cinti S, Mitchell G, Barbatelli G, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res. 2005;46:2347-2355. [DOI] [PubMed] [Google Scholar]
  • 29. Murano I, Barbatelli G, Parisani V, et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res. 2008;49:1562-1568. [DOI] [PubMed] [Google Scholar]
  • 30. Apovian CM, Bigornia S, Mott M, et al. Adipose macrophage infiltration is associated with insulin resistance and vascular endothelial dysfunction in obese subjects. Arterioscler Thromb Vasc Biol. 2008;28:1654-1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Lee MJ, Wu Y, Fried SK. Adipose tissue remodelling in pathophysiology of obesity. Curr Opin Clin Nutr Metab Care. 2010;13:371-376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. de Souza CJ, Eckhardt M, Gagen K, et al. Effects of pioglitazone on adipose tissue remodeling within the setting of obesity and insulin resistance. Diabetes. 2001;50:1863-1871. [DOI] [PubMed] [Google Scholar]
  • 33. Strissel KJ, Stancheva Z, Miyoshi H, et al. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56:2910-2918. [DOI] [PubMed] [Google Scholar]
  • 34. Gornicka A, Fettig J, Eguchi A, et al. Adipocyte hypertrophy is associated with lysosomal permeability both in vivo and in vitro: role in adipose tissue inflammation. Am J Physiol Endocrinol Metab. 2012;303:E597-E606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Ota T. Chemokine systems link obesity to insulin resistance. Diabetes Metab J. 2013;37:165-172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kanda H, Tateya S, Tamori Y, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494-1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Weisberg SP, Hunter D, Huber R, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. 2006;116:115-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Kamei N, Tobe K, Suzuki R, et al. Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J Biol Chem. 2006;281:26602-26614. [DOI] [PubMed] [Google Scholar]
  • 39. Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol. 2006;177:7303-7311. [DOI] [PubMed] [Google Scholar]
  • 40. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677-686. [DOI] [PubMed] [Google Scholar]
  • 41. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue polarization. J Clin Invest. 2007;117:175-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Nguyen MT, Favelyukis S, Nguyen AK, et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J Biol Chem. 2007;282:35279-35292. [DOI] [PubMed] [Google Scholar]
  • 43. Kitade H, Sawamoto K, Nagashimada M, et al. CCR5 plays a critical role in obesity-induced adipose tissue inflammation and insulin resistance by regulating both macrophage recruitment and M1/M2 status. Diabetes. 2012;61:1680-1690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Li P, Lu M, Nguyen MT, et al. Functional heterogeneity of CD11c-positive adipose tissue macrophages in diet-induced obese mice. J Biol Chem. 2010;285:15333-15345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Fujisaka S, Usui I, Bukhari A, et al. Regulatory mechanisms for adipose tissue M1 and M2 macrophages in diet-induced obese mice. Diabetes. 2009;58:2574-2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914-920. [DOI] [PubMed] [Google Scholar]
  • 47. Feuerer M, Herrero L, Cipolletta D, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15:930-939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Strissel KJ, DeFuria J, Shaul ME, Bennett G, Greenberg AS, Obin MS. T-cell recruitment and Th1 polarization in adipose tissue during diet-induced obesity in C57BL/6 mice. Obesity (Silver Spring). 2010;18:1918-1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Winer S, Chan Y, Paltser G, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009;15:921-929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Nishimura S, Manabe I, Takaki S, et al. Adipose natural regulatory B cells negatively control adipose tissue inflammation [published online October 22, 2013]. Cell Metab. doi: 10.1016/j.cmet.2013.09.017. [DOI] [PubMed] [Google Scholar]
  • 51. Altintas MM, Azad A, Nayer B, et al. Mast cells, macrophages, and crown-like structures distinguish subcutaneous from visceral fat in mice. J Lipid Res. 2011;52:480-488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Liu J, Divoux A, Sun J, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med. 2009;15:940-945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Wu D, Molofsky AB, Liang HE, et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science. 2011;332:243-247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Elgazar-Carmon V, Rudich A, Hadad N, Levy R. Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res. 2008;49:1894-1903. [DOI] [PubMed] [Google Scholar]
  • 55. Ji Y, Sun S, Xu A, et al. Activation of natural killer T cells promotes M2 Macrophage polarization in adipose tissue and improves systemic glucose tolerance via interleukin-4 (IL-4)/STAT6 protein signaling axis in obesity. J Biol Chem. 2012;287:13561-13571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Maffei M, Halaas J, Ravussin E, et al. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med. 1995;1:1155-1161. [DOI] [PubMed] [Google Scholar]
  • 57. Considine RV, Sinha MK, Heiman ML, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334:292-295. [DOI] [PubMed] [Google Scholar]
  • 58. Bastard JP, Jardel C, Bruckert E, et al. Elevated levels of interleukin 6 are reduced in serum and subcutaneous adipose tissue of obese women after weight loss. J Clin Endocrinol Metab. 2000;85:3338-3342. [DOI] [PubMed] [Google Scholar]
  • 59. Monzillo LU, Hamdy O, Horton ES, et al. Effect of lifestyle modification on adipokine levels in obese subjects with insulin resistance. Obes Res. 2003;11:1048-1054. [DOI] [PubMed] [Google Scholar]
  • 60. Rosenbaum M, Goldsmith R, Bloomfield D, et al. Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J Clin Invest. 2005;115:3579-3586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ghilardi N, Skoda RC. The leptin receptor activates Janus kinase 2 and signals for proliferation in a factor-dependent cell line. Mol Endocrinol. 1997;11:393-399. [DOI] [PubMed] [Google Scholar]
  • 62. Banks AS, Davis SM, Bates SH, Myers MG. Activation of downstream signals by the long form of the leptin receptor. J Biol Chem. 2000;275:14563-14572. [DOI] [PubMed] [Google Scholar]
  • 63. Cai D, Yuan M, Frantz DF, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med. 2005;11:183-190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Sweeney G. Leptin signalling. Cell Signal. 2002;14:655-663. [DOI] [PubMed] [Google Scholar]
  • 65. Bahceci M, Gokalp D, Bahceci S, Tuzcu A, Atmaca S, Arikan S. The correlation between adiposity and adiponectin, tumor necrosis factor α, interleukin-6 and high sensitivity C-reactive protein levels. Is adipocyte size associated with inflammation in adults? J Endocrinol Invest. 2007;30:210-214. [DOI] [PubMed] [Google Scholar]
  • 66. Almeda-Valdes P, Cuevas-Ramos D, Mehta R, et al. Total and high molecular weight adiponectin have similar utility for the identification of insulin resistance. Cardiovasc Diabetol. 2010;9:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Fasshauer M, Kralisch S, Klier M, et al. Adiponectin gene expression and secretion is inhibited by interleukin-6 in 3T3-L1 adipocytes. Biochem Biophys Res Commun. 2003;301:1045-1050. [DOI] [PubMed] [Google Scholar]
  • 68. Winkler G, Salamon F, Salamon D, Speer G, Simon K, Cseh K. Elevated serum tumour necrosis factor-alpha levels can contribute to the insulin resistance in Type II (non-insulin-dependent) diabetes and in obesity. Diabetologia. 1998;41:860-861. [DOI] [PubMed] [Google Scholar]
  • 69. Ryan AS, Nicklas BJ. Reductions in plasma cytokine levels with weight loss improve insulin sensitivity in overweight and obese postmenopausal women. Diabetes Care. 2004;27:1699-1705. [DOI] [PubMed] [Google Scholar]
  • 70. Faraj M, Havel PJ, Phélis S, Blank D, Sniderman AD, Cianflone K. Plasma acylation-stimulating protein, adiponectin, leptin, and ghrelin before and after weight loss induced by gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metab. 2003;88:1594-1602. [DOI] [PubMed] [Google Scholar]
  • 71. Yang WS, Lee WJ, Funahashi T, et al. Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab. 2001;86:3815-3819. [DOI] [PubMed] [Google Scholar]
  • 72. Milan G, Granzotto M, Scarda A, et al. Resistin and adiponectin expression in visceral fat of obese rats: effect of weight loss. Obes Res. 2002;10:1095-1103. [DOI] [PubMed] [Google Scholar]
  • 73. Yamauchi T, Kadowaki T. Physiological and pathophysiological roles of adiponectin and adiponectin receptors in the integrated regulation of metabolic and cardiovascular diseases. Int J Obes (Lond). 2008;32(suppl 7):S13-S18. [DOI] [PubMed] [Google Scholar]
  • 74. Wang AYH, Hickman IJ, Richards AA, Whitehead JP, Prins JB, MacDonald GA. High molecular weight adiponectin correlates with insulin sensitivity in patients with hepatitis C genotype 3, but not genotype 1 infection. Am J Gastroenterol. 2005;100:2717-2723. [DOI] [PubMed] [Google Scholar]
  • 75. Wolf AM, Wolf D, Rumpold H, Enrich B, Tilg H. Adiponectin induces the anti-inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem Biophys Res Commun. 2004;323:630-635. [DOI] [PubMed] [Google Scholar]
  • 76. Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med. 2002;8:1288-1295. [DOI] [PubMed] [Google Scholar]
  • 77. Guo J, Bian Y, Bai R, Li H, Fu M, Xiao C. Globular adiponectin attenuates myocardial ischemia/reperfusion injury by upregulating endoplasmic reticulum Ca2+-ATPase activity and inhibiting endoplasmic reticulum stress. J Cardiovasc Pharmacol. 2013;62:143-153. [DOI] [PubMed] [Google Scholar]
  • 78. Okada-Iwabu M, Yamauchi T, Iwabu M, et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature. 2013;503:493-499. [DOI] [PubMed] [Google Scholar]
  • 79. Opie LH, Walfish PG. Plasma free fatty acid concentrations in obesity. N Engl J Med. 1963;268:757-760. [DOI] [PubMed] [Google Scholar]
  • 80. Jensen MD, Haymond MW, Rizza RA, Cryer PE, Miles JM. Influence of body fat distribution on free fatty acid metabolism in obesity. J Clin Invest. 1989;83:1168-1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Santomauro AT, Boden G, Silva ME, et al. Overnight lowering of free fatty acids with Acipimox improves insulin resistance and glucose tolerance in obese diabetic and nondiabetic subjects. Diabetes. 1999;48:1836-1841. [DOI] [PubMed] [Google Scholar]
  • 82. Liang H, Tantiwong P, Sriwijitkamol A, et al. Effect of a sustained reduction in plasma free fatty acid concentration on insulin signalling and inflammation in skeletal muscle from human subjects. J Physiol. 2013;591:2897-2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. 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;116:3015-3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Tsukumo DM, Carvalho-Filho MA, Carvalheira JB, et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes. 2007;56:1986-1998. [DOI] [PubMed] [Google Scholar]
  • 85. Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IκB-α. Diabetes. 2002;51:2005-2011. [DOI] [PubMed] [Google Scholar]
  • 86. Boden G, She P, Mozzoli M, et al. Free fatty acids produce insulin resistance and activate the proinflammatory nuclear factor-κB pathway in rat liver. Diabetes. 2005;54:3458-3465. [DOI] [PubMed] [Google Scholar]
  • 87. Kharroubi I, Ladrière L, Cardozo AK, Dogusan Z, Cnop M, Eizirik DL. Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-κB and endoplasmic reticulum stress. Endocrinology. 2004;145:5087-5096. [DOI] [PubMed] [Google Scholar]
  • 88. Kawasaki N, Asada R, Saito A, Kanemoto S, Imaizumi K. Obesity-induced endoplasmic reticulum stress causes chronic inflammation in adipose tissue. Sci Rep. 2012;2:799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Cao SS, Kaufman RJ. Targeting endoplasmic reticulum stress in metabolic disease. Expert Opin Ther Targets. 2013;17:437-448. [DOI] [PubMed] [Google Scholar]
  • 90. Pierre N, Deldicque L, Barbé C, Naslain D, Cani PD, Francaux M. Toll-like receptor 4 knockout mice are protected against endoplasmic reticulum stress induced by a high-fat diet. PLoS One. 2013;8:e65061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Consitt LA, Bell JA, Houmard JA. Intramuscular lipid metabolism, insulin action, and obesity. IUBMB Life. 2009;61:47-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Schmitz-Peiffer C. Targeting ceramide synthesis to reverse insulin resistance. Diabetes. 2010;59:2351-2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. 1995;95:2409-2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Bradley RL, Jeon JY, Liu FF, Maratos-Flier E. Voluntary exercise improves insulin sensitivity and adipose tissue inflammation in diet-induced obese mice. Am J Physiol Endocrinol Metab. 2008;295:E586-E594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Bastard JP, Jardel C, Bruckert E, Vidal H, Hainque B. Variations in plasma soluble tumour necrosis factor receptors after diet-induced weight loss in obesity. Diabetes Obes Metab. 2000;2:323-325. [DOI] [PubMed] [Google Scholar]
  • 96. Manco M, Fernandez-Real JM, Equitani F, et al. Effect of massive weight loss on inflammatory adipocytokines and the innate immune system in morbidly obese women. J Clin Endocrinol Metab. 2007;92:483-490. [DOI] [PubMed] [Google Scholar]
  • 97. Moschen AR, Molnar C, Geiger S, et al. Anti-inflammatory effects of excessive weight loss: potent suppression of adipose interleukin 6 and tumour necrosis factor α expression. Gut. 2010;59:1259-1264. [DOI] [PubMed] [Google Scholar]
  • 98. Roth CL, Kratz M, Ralston MM, Reinehr T. Changes in adipose-derived inflammatory cytokines and chemokines after successful lifestyle intervention in obese children. Metabolism. 2011;60:445-452. [DOI] [PubMed] [Google Scholar]
  • 99. Illán-Gómez F, Gonzálvez-Ortega M, Orea-Soler I, et al. Obesity and inflammation: change in adiponectin, C-reactive protein, tumour necrosis factor-α and interleukin-6 after bariatric surgery. Obes Surg. 2012;22:950-955. [DOI] [PubMed] [Google Scholar]
  • 100. Aggarwal BB. Tumour necrosis factors receptor associated signalling molecules and their role in activation of apoptosis, JNK and NF-κB. Ann Rheum Dis. 2000;59(suppl 1):i6-i16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Kuno K, Matsushima K. The IL-1 receptor signaling pathway. J Leukoc Biol. 1994;56:542-547. [DOI] [PubMed] [Google Scholar]
  • 102. Keaney JF, Jr, Larson MG, Vasan RS, et al. ; Framingham Study. Obesity and systemic oxidative stress: clinical correlates of oxidative stress in the Framingham Study. Arterioscler Thromb Vasc Biol. 2003;23:434-439. [DOI] [PubMed] [Google Scholar]
  • 103. Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114:1752-1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Fujita K, Nishizawa H, Funahashi T, Shimomura I, Shimabukuro M. Systemic oxidative stress is associated with visceral fat accumulation and the metabolic syndrome. Circ J. 2006;70:1437-1442. [DOI] [PubMed] [Google Scholar]
  • 105. Lee YS, Kim AY, Choi JW, et al. Dysregulation of adipose glutathione peroxidase 3 in obesity contributes to local and systemic oxidative stress. Mol Endocrinol. 2008;22:2176-2189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Sakurai T, Ogasawara J, Kizaki T, et al. The effects of exercise training on obesity-induced dysregulated expression of adipokines in white adipose tissue. Int J Endocrinol. 2013;2013:801743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Imayama I, Ulrich CM, Alfano CM, et al. Effects of a caloric restriction weight loss diet and exercise on inflammatory biomarkers in overweight/obese postmenopausal women: a randomized controlled trial. Cancer Res. 2012;72:2314-2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. You T, Arsenis NC, Disanzo BL, Lamonte MJ. Effects of exercise training on chronic inflammation in obesity: current evidence and potential mechanisms. Sports Med. 2013;43:243-256. [DOI] [PubMed] [Google Scholar]
  • 109. Ross R, Janssen I, Dawson J, et al. Exercise-induced reduction in obesity and insulin resistance in women: a randomized controlled trial. Obes Res. 2004;12:789-798. [DOI] [PubMed] [Google Scholar]
  • 110. Irwin ML, Yasui Y, Ulrich CM, et al. Effect of exercise on total and intra-abdominal body fat in postmenopausal women: a randomized controlled trial. JAMA. 2003;289:323-330. [DOI] [PubMed] [Google Scholar]
  • 111. Ohkawara K, Tanaka S, Miyachi M, Ishikawa-Takata K, Tabata I. A dose-response relation between aerobic exercise and visceral fat reduction: systematic review of clinical trials. Int J Obes (Lond). 2007;31:1786-1797. [DOI] [PubMed] [Google Scholar]
  • 112. Arner P, Hellström L, Wahrenberg H, Brönnegård M. Beta-adrenoceptor expression in human fat cells from different regions. J Clin Invest. 1990;86:1595-1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Mauriège P, Prud’Homme D, Marcotte M, Yoshioka M, Tremblay A, Després JP. Regional differences in adipose tissue metabolism between sedentary and endurance-trained women. Am J Physiol. 1997;273(3 pt 1):E497-E506. [DOI] [PubMed] [Google Scholar]
  • 114. Després JP, Bouchard C, Savard R, Tremblay A, Marcotte M, Thériault G. The effect of a 20-week endurance training program on adipose-tissue morphology and lipolysis in men and women. Metabolism. 1984;33:235-239. [DOI] [PubMed] [Google Scholar]
  • 115. El Bouazzaoui F, Henneman P, Thijssen P, et al. Adipocyte telomere length associates negatively with adipocyte size, whereas adipose tissue telomere length associates negatively with the extent of fibrosis in severely obese women. Int J Obes (Lond). 2014;38:746-749. [DOI] [PubMed] [Google Scholar]
  • 116. Gollisch KS, Brandauer J, Jessen N, et al. Effects of exercise training on subcutaneous and visceral adipose tissue in normal- and high-fat diet-fed rats. Am J Physiol Endocrinol Metab. 2009;297:E495-E504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Ahlborg G, Felig P, Hagenfeldt L, Hendler R, Wahren J. Substrate turnover during prolonged exercise in man. Splanchnic and leg metabolism of glucose, free fatty acids, and amino acids. J Clin Invest. 1974;53:1080-1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Spriet LL. New insights into the interaction of carbohydrate and fat metabolism during exercise. Sports Med. 2014;44(suppl 1):87-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Romijn JA, Coyle EF, Sidossis LS, Rosenblatt J, Wolfe RR. Substrate metabolism during different exercise intensities in endurance-trained women. J Appl Physiol. 2000;88:1707-1714. [DOI] [PubMed] [Google Scholar]
  • 120. Moro C, Pillard F, de Glisezinski I, et al. Sex differences in lipolysis-regulating mechanisms in overweight subjects: effect of exercise intensity. Obesity (Silver Spring). 2007;15:2245-2255. [DOI] [PubMed] [Google Scholar]
  • 121. Disanzo BL, You T. Effects of exercise training on indicators of adipose tissue angiogenesis and hypoxia in obese rats. Metabolism. 2014;63:452-455. [DOI] [PubMed] [Google Scholar]
  • 122. Kawanishi N, Yano H, Yokogawa Y, Suzuki K. Exercise training inhibits inflammation in adipose tissue via both suppression of macrophage infiltration and acceleration of phenotypic switching from M1 to M2 macrophages in high-fat-diet-induced obese mice. Exerc Immunol Rev. 2010;16:105-118. [PubMed] [Google Scholar]
  • 123. Oliveira AG, Carvalho BM, Tobar N, et al. Physical exercise reduces circulating lipopolysaccharide and TLR4 activation and improves insulin signaling in tissues of DIO rats. Diabetes. 2011;60:784-796. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 124. Kim F, Pham M, Luttrell I, et al. Toll-like receptor-4 mediates vascular inflammation and insulin resistance in diet-induced obesity. Circ Res. 2007;100:1589-1596. [DOI] [PubMed] [Google Scholar]
  • 125. Lambert CP, Wright NR, Finck BN, Villareal DT. Exercise but not diet-induced weight loss decreases skeletal muscle inflammatory gene expression in frail obese elderly persons. J Appl Physiol (1985). 2008;105:473-478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Roberts CK, Won D, Pruthi S, Lin SS, Barnard RJ. Effect of a diet and exercise intervention on oxidative stress, inflammation and monocyte adhesion in diabetic men. Diabetes Res Clin Pract. 2006;73:249-259. [DOI] [PubMed] [Google Scholar]
  • 127. Halle M, Berg A, Garwers U, Grathwohl D, Knisel W, Keul J. Concurrent reductions of serum leptin and lipids during weight loss in obese men with type II diabetes. Am J Physiol. 1999;277(2 pt 1):E277-E282. [DOI] [PubMed] [Google Scholar]
  • 128. Ishii T, Yamakita T, Yamagami K, et al. Effect of exercise training on serum leptin levels in type 2 diabetic patients. Metabolism. 2001;50:1136-1140. [DOI] [PubMed] [Google Scholar]
  • 129. Houmard JA, Cox JH, MacLean PS, Barakat HA. Effect of short-term exercise training on leptin and insulin action. Metabolism. 2000;49:858-861. [DOI] [PubMed] [Google Scholar]
  • 130. Fatouros IG, Tournis S, Leontsini D, et al. Leptin and adiponectin responses in overweight inactive elderly following resistance training and detraining are intensity related. J Clin Endocrinol Metab. 2005;90:5970-5977. [DOI] [PubMed] [Google Scholar]
  • 131. Christiansen T, Paulsen SK, Bruun JM, Pedersen SB, Richelsen B. Exercise training versus diet-induced weight-loss on metabolic risk factors and inflammatory markers in obese subjects: a 12-week randomized intervention study. Am J Physiol Endocrinol Metab. 2010;298:E824-E831. [DOI] [PubMed] [Google Scholar]
  • 132. Vieira VJ, Valentine RJ, Wilund KR, Antao N, Baynard T, Woods JA. Effects of exercise and low-fat diet on adipose tissue inflammation and metabolic complications in obese mice. Am J Physiol Endocrinol Metab. 2009;296:E1164-E1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Farias JM, Maggi RM, Tromm CB, et al. Exercise training performed simultaneously to a high-fat diet reduces the degree of insulin resistance and improves adipoR1-2/APPL1 protein levels in mice. Lipids Health Dis. 2012;11:134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Ringseis R, Mooren FC, Keller J, et al. Regular endurance exercise improves the diminished hepatic carnitine status in mice fed a high-fat diet. Mol Nutr Food Res. 2011;55(suppl 2):S193-S202. [DOI] [PubMed] [Google Scholar]
  • 135. Zbinden-Foncea H, van Loon LJ, Raymackers JM, Francaux M, Deldicque L. Contribution of nonesterified fatty acids to mitogen-activated protein kinase activation in human skeletal muscle during endurance exercise. Int J Sport Nutr Exerc Metab. 2013;23:201-209. [DOI] [PubMed] [Google Scholar]
  • 136. Zbinden-Foncea H, Raymackers JM, Deldicque L, Renard P, Francaux M. TLR2 and TLR4 activate p38 MAPK and JNK during endurance exercise in skeletal muscle. Med Sci Sports Exerc. 2012;44:1463-1472. [DOI] [PubMed] [Google Scholar]
  • 137. Francaux M. Toll-like receptor signalling induced by endurance exercise. Appl Physiol Nutr Metab. 2009;34:454-458. [DOI] [PubMed] [Google Scholar]
  • 138. da Luz G, Frederico MJ, da Silva S, et al. Endurance exercise training ameliorates insulin resistance and reticulum stress in adipose and hepatic tissue in obese rats. Eur J Appl Physiol. 2011;111:2015-2023. [DOI] [PubMed] [Google Scholar]
  • 139. Petersen AM, Pedersen BK. The role of IL-6 in mediating the anti-inflammatory effects of exercise. J Physiol Pharmacol. 2006;57(suppl 10):43-51. [PubMed] [Google Scholar]
  • 140. Plomgaard P, Penkowa M, Pedersen BK. Fiber type specific expression of TNF-α, IL-6 and IL-18 in human skeletal muscles. Exerc Immunol Rev. 2005;11:53-63. [PubMed] [Google Scholar]
  • 141. Steensberg A, van Hall G, Osada T, Sacchetti M, Saltin B, Klarlund Pedersen B. Production of interleukin-6 in contracting human skeletal muscles can account for the exercise-induced increase in plasma interleukin-6. J Physiol. 2000;529(pt 1):237-242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Andersen K, Pedersen BK. The role of inflammation in vascular insulin resistance with focus on IL-6. Horm Metab Res. 2008;40:635-639. [DOI] [PubMed] [Google Scholar]
  • 143. Straczkowski M, Kowalska I, Dzienis-Straczkowska S, et al. Changes in tumor necrosis factor-α system and insulin sensitivity during an exercise training program in obese women with normal and impaired glucose tolerance. Eur J Endocrinol. 2001;145:273-280. [DOI] [PubMed] [Google Scholar]
  • 144. Gomez-Merino D, Drogou C, Guezennec CY, Chennaoui M. Effects of chronic exercise on cytokine production in white adipose tissue and skeletal muscle of rats. Cytokine. 2007;40:23-29. [DOI] [PubMed] [Google Scholar]
  • 145. Wasinski F, Bacurau RF, Moraes MR, et al. Exercise and caloric restriction alter the immune system of mice submitted to a high-fat diet. Mediators Inflamm. 2013;2013:395672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Ploeger HE, Takken T, de Greef MH, Timmons BW. The effects of acute and chronic exercise on inflammatory markers in children and adults with a chronic inflammatory disease: a systematic review. Exerc Immunol Rev. 2009;15:6-41. [PubMed] [Google Scholar]
  • 147. Tirakitsoontorn P, Nussbaum E, Moser C, Hill M, Cooper DM. Fitness, acute exercise, and anabolic and catabolic mediators in cystic fibrosis. Am J Respir Crit Care Med. 2001;164(8 pt 1):1432-1437. [DOI] [PubMed] [Google Scholar]
  • 148. Rabinovich RA, Figueras M, Ardite E, et al. Increased tumour necrosis factor-alpha plasma levels during moderate-intensity exercise in COPD patients. Eur Respir J. 2003;21:789-794. [DOI] [PubMed] [Google Scholar]
  • 149. Vollaard NB, Shearman JP, Cooper CE. Exercise-induced oxidative stress: myths, realities and physiological relevance. Sports Med. 2005;35:1045-1062. [DOI] [PubMed] [Google Scholar]
  • 150. Finkler M, Lichtenberg D, Pinchuk I. The relationship between oxidative stress and exercise. J Basic Clin Physiol Pharmacol. 2013;17:1-11. [DOI] [PubMed] [Google Scholar]
  • 151. Sakurai T, Izawa T, Kizaki T, et al. Exercise training decreases expression of inflammation-related adipokines through reduction of oxidative stress in rat white adipose tissue. Biochem Biophys Res Commun. 2009;379:605-609. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Lifestyle Medicine are provided here courtesy of SAGE Publications

RESOURCES