Skip to main content
Sports Medicine and Health Science logoLink to Sports Medicine and Health Science
. 2022 Jan 20;4(1):18–28. doi: 10.1016/j.smhs.2022.01.001

Exercise training modulates adipokine dysregulations in metabolic syndrome

Parvin Babaei a,b,c,, Rastegar Hoseini d
PMCID: PMC9219261  PMID: 35782776

Abstract

Metabolic syndrome (MetS) is a cluster of risk factors for various metabolic diseases, and it is characterized by central obesity, dyslipidemia, hypertension, and insulin resistance. The core component for MetS is adipose tissue, which releases adipokines and influences physical health. Adipokines consist of pro and anti-inflammatory cytokines and contribute to various physiological functions. Generally, a sedentary lifestyle promotes fat accumulation and secretion of pro-inflammatory adipokines. However, regular exercise has been known to exert various beneficial effects on metabolic and cognitive disorders. Although the mechanisms underlying exercise beneficial effects in MetS are not fully understood, changes in energy expenditure, fat accumulation, circulatory level of myokines, and adipokines might be involved. This review article focuses on some of the selected adipokines in MetS, and their responses to exercise training considering possible mechanisms.

Keywords: Adipokines, Insulin resistance, Aerobic training, Resistance training, Metabolic syndrome

Graphical abstract

Image 1

Highlights

  • Adipokines are a potential link between visceral fats and complications of MetS.

  • Physical inactivity increases pro-inflammatory adipokines.

  • Moderate aerobic exercise increases anti-inflammatory adipokines partly via reducing adipose tissue mass.

  • Physical exercise-induced myokines might mediate beneficial effects via a cross-talk with adipose tissues.

Abbreviations

AMPK

5′ Adenosine Monophosphate-Activated Protein Kinase

AT

Aerobic Training

BAIBA

β-amino-isobutyric Acid

ERK

Extracellular Signal-Regulated Protein Kinase

FOXO

Forkhead Box O

GLUT4

Glucose Transporter Type 4

JNK

c-Jun N-terminal kinases

HDL

High-Density Lipoprotein

IKK

Inhibitory-κB Kinase

IL

Interleukin

IRS-1/2

Insulin receptor substrate 1/2

LDL

Low-Density Lipoprotein

MAPK

Mitogen-Activated Protein Kinase

MetS

Metabolic Syndrome

NF-κB

Nuclear Factor Kappa-Light-Chain-Enhancer Of Activated B Cells

PGC-1α

PPARγ Co-Activator 1α

PKB

Protein kinase B

PPAR

Peroxisome Proliferator-Activated Receptor

PI3K

Phosphoinositide 3-kinase

RT

Resistance Training

SFRP5

Secreted Frizzled Related Protein 5

SREBP-1c

Sterol Regulatory Element-Binding Protein 1c

TG

Triglyceride

TNFR

Tumor Necrosis Factor receptor

TNF-α

Tumor Necrosis Factor- Alpha

eNOS

Nitric Oxide Synthase

ROS

Reactive Oxygen Species

TLR4

Toll-Like Receptors 4

Wnt5a

Wnt Family Member 5A

p38–MAPK

p38 Mitogen-Activated Protein Kinase

HOMA-IR

Homeostatic Model Assessment for Insulin Resistance

mTOR

Mammalian Target of Rapamycin

TNFR1

Tumor Necrosis Factor Receptor Type 1

HRmax

Maximum Heart Rate

Introduction

Metabolic syndrome

Metabolic syndrome (MetS) is a clustering of symptoms or conditions including; central obesity, high blood pressure, hyperglycemia, insulin resistance, and dyslipidemia.1 Among them, central obesity, has been viewed as a serious problem of the 21st-century,2 and is associated with various metabolic disorders.3 Excess visceral fat, causes the secretion of bioactive peptides; known as adipokines, and thus, links with other parts of the body. Adipokines comprise classical cytokines and chemokines and contribute to different physiological functions such as regulation of appetite, energy expenditure, and metabolism.4,5 Currently their numbers supersede 800, because of heterogeneity of the adipose tissue. To simplify, they are categorized into two distinct classes of pro-inflammatory and anti-inflammatory adipokines.6,7 These peptides fuel the crosstalk feedback loops with other organs8,9 particularly with skeletal muscles and mediate metabolic regulations. Since the production and secretion of adipokines, play a central role in chronic inflammation,10 understanding the scenario behind physiological mechanisms, may provide appropriate strategies for controlling insulin resistance and further related disorders.

Insulin and its signaling cascade normally control cell growth, metabolism, and survival through activation of Mitogen-Activated Protein Kinases (MAPKs) and Phosphoinositide-3-Kinase (PI3K), of which activation of PI-3K-associated with Insulin Receptor Substrate (IRS)-1/2, Protein Kinase B (Akt), Forkhead Box O1 (FOXO1). Inactivation of Akt and activation of FOXO1, through suppression of IRS1 and IRS-2 in different organs following hyperinsulinemia and metabolic inflammation, may provide the underlying mechanisms for MetS in humans.11 Insulin regulates fat metabolism in peripheral tissues by activating complex signaling pathways including PI3K/AKT, and MAPK, by binding to FOXO and proliferator-activated receptor γ (PPARγ) transcription factors. Insulin resistance disrupts these pathways and causes hyperglycemia and dyslipidemia. Dyslipidemia, or fatty disorders, is the result of the accumulation of free fatty acids in the liver along with insulin-enhanced lipogenesis, which increases triglyceride production and secretion. This condition, along with increased hepatic absorption and renal clearance of High-Density Lipoprotein (HDL) cholesterol, causes a disturbance of the fat profile, i.e., decreasing HDL cholesterol and increasing triglycerides (TG), both of which are seen in the MetS.11

MetS prevention and treatment require appropriate behavioral interventions including both dietary and exercise. Considering the important roles of adipokines in MetS, and the molecular link between exercise and other tissues, here we review the effects of aerobic and resistance training (AT and RT) on the selected adipokines in MetS models. Some of the other classical adipokines have been summarized in Table 1 briefly.

Table 1.

Summary of the selected adipokines alteration in response to MetS, RT, and AT.

Adipokines MetS AT Possible Mechanisms RT Possible Mechanisms
Irisin Decrease Increase Decreased body fat percentage and insulin resistance Increase Improved body composition and insulin function
Visfatin Increase Decrease Body fat mass reduction Decrease Improved body composition
Resistin Increase Decrease Improved insulin resistance, inflammatory markers, and glycosylated hemoglobin Decrease Decreased body fat percentage and insulin resistance
Chemerin Increase Decrease Decreased body fat percentage and improved lipid profile Decrease Decreased body fat percentage and insulin resistance
Retinol Binding Protein 4 ​(RBP4) Increase Decrease Decreased body fat percentage Decrease Decreased glucose and improved insulin sensitivity
Interleukin 8 (IL-8) Increase Decrease Decreased body fat mass and inflammatory markers Decrease Decreased body fat mass and increased proinflammatory cytokines
Interleukin 10 (IL-10) Increase Decrease Weight loss and body fat percentage reduction Decrease Body fat mass reduction
Interferon gamma (IFN-γ) Increase Decrease Decreased inflammatory markers Decrease Improved body composition and insulin sensitivity
C1q/TNF-related protein (CTRP4) Increase Decrease Decreased insulin and increased fat oxidation Decrease Decreased body fat percentage and insulin resistance

It is worth mentioning that information provided in the manuscript is the most current knowledge on signaling pathways and these pathways are not fully elucidated.

Effects of exercise on MetS

A sedentary lifestyle results in hypertension, dyslipidemia, high blood glucose, and obesity, while regular physical activity successfully prevents the progression of metabolic diseases.12 Exercise is an important non-pharmacological tool, which exerts remarkable beneficial effects on various functional systems of the human body.12 Although the exact mechanism of the beneficial effect of regular exercise on organs function has not been understood yet, several biological mechanisms may be responsible including reduced visceral adiposity, TG, Low-Density Lipoproteins (LDL),13 blood pressure,14 and systemic inflammation,15 increased HDL16,17 and improved insulin sensitivity.18 Regular exercise, importantly, performed at low/moderate intensities, exerts anti-inflammatory effects19,20 and modulate metabolic homeostasis,21 partly by increasing anti-inflammatory adipokines.22,23

It has been known that skeletal muscles and adipose tissues are among the first target organs for exercise training. They are characterized as an endocrine organ due to the various cytokines production (those are produced by adipose tissues are named adipokines, and those by skeletal muscles are named myokines).24 These molecules demonstrate autocrine, paracrine, and endocrine effects, and have metabolic consequents.25 Exercise alters adipokines levels by modulating genes expression and also activating/inactivating proteins involved in their signaling pathways.26 For example, the lipolytic action of exercise needs phosphorylation of 5′ Adenosine Monophosphate-Activated Protein Kinase (AMPK) to reduce the Sterol Regulatory Element-Binding Protein 1c (SREBP-1c).27,28 AMPK inhibits hepatic TG synthesis suppressing acetyl-CoA carboxylase,29 and promotes fatty acid oxidation and reduction in fat mass.30,31 Other beneficial effects of exercise are related to increased peripheral glucose uptake32 which takes place via activating PI3K, and translocating the Glucose Transporter Type 4 (GLUT4) into the skeletal muscles cell membranes,33 and also increasing AMPK activity, which increases insulin sensitivity.34 Some of these signaling pathways are likely regulated by adipokines secreted by adipose tissues during exercise which provide health benefits. Bellow, we review some of the important adipokines alteration, first in response to MetS and then exercise training.

Adiponectin in MetS

Human adiponectin is a peptide, comprised of 244 amino acids produced by adipose tissues, skeletal muscles, and cardiac cells.35,36 It has been known that low adiponectin level is associated with MetS prevalence,35,36 insulin resistance,37,38 coronary heart disease,39 and inflammation-related diseases.40,41 Adiponectin exerts a multitude of physiological actions against inflammation, insulin resistance, MetS, obesity, cardiac fibrosis,42 fat accumulation in the liver,37,38 and atherosclerosis.43 The increased adiponectin induces insulin sensitivity in skeletal muscles by binding with adiponectin receptor type 1 and 2, leading to the activation of various signaling pathways such as IRS-1/2, AMPK, and p38 Mitogen-Activated Protein Kinase (p38–MAPK).44 Also adiponectin alters nitric oxide level and leads to vasoprotective effects via activating AMPK45 and cyclooxygenase II.46 Moreover, adiponectin alleviates the inflammatory responses by inhibiting Tumor Necrosis Factor-Alpha (TNF-α), Interleukin-6 (IL-6), Interleukin-10 (IL-10), and Interleukin-1 (IL-1),42 via Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells (NF-κB), and Protein Kinase A (PKA).47 In consistency, deficiency in adiponectin receptors or AMPK, leads to significantly reduced glucose intolerance, increased hepatic triglyceride, inflammation, and oxidative stress.48

Effect of AT and RT on adiponectin levels

AT has been shown to cause weight loss and increase lipolysis,49 partly via elevating adiponectin secretion in both human and animal models.50,51 Subsequently, adiponectin modifies HDL and LDL concentration and stimulates glucose uptake in peripheral tissues.52,53 It is assumed that lactate accumulation, elevated adrenalin, glycogen depletion, and acidosis, have stimulatory effects on adiponectin secretion in both ovariectomized rats model of MetS54 and obese humans.55,56 To date, the exact effect of Resistance Training (RT) on adiponectin is not apparent. Ward et al.57 showed a significantly decreased plasma adiponectin in postmenopausal women and elevation after 15 weeks of RT engagement. Also, de Mello et al.58 showed a significant increased in adiponectin level, following both protocols of AT singly, and combined with RT, however, combination of AT and RT showed superiority to AT in obese adolescents with MetS.

Mechanistically, RT has been shown to increase muscle mass, decrease body fat percentage by increasing resting energy consumption in MetS patients,59 increasing blood flow, and shifting adiponectin to plasma.60 Moreover, a combination of AT with RT, has been shown to increase adiponectin levels in obese Type 2 diabetes (T2D) patients and reduce insulin resistance and central adiposity.61 Although other mechanisms such as releasing myokines, or various hormones such as adrenalin and glucocorticoids are warranted.62

Finally, exercise-increased adiponectin level demonstrates an excellent potential strategy for developing novel therapeutic approaches for MetS.

Leptin in MetS

Leptin is a 167 amino acid peptide, which is transcribed from the LEP gene, located on chromosome 7 in adipocytes. It acts on a specific cytokine family receptor in the arcuate and ventromedial nucleus of the hypothalamus,63 and regulates energy balance by suppressing appetite and increasing energy expenditure.64,65 Also leptin acts peripherally, regulating lipid and glucose metabolism, through other hormones such as insulin, glucagon, insulin-like growth factor, growth hormone, and glucocorticoids.64,65 An animal study reported that both leptin muted mice of ob/ob and db/db are markedly hyperglycemic and glucose intolerant, independently of their body weight and that, the hyperglycemia of ob/ob mice can be readily normalized by leptin infusion into the brain.66 Circulating leptin levels are correlated with body fat stores, so that, the decreased leptin levels set off a series of biological reactions to reduce energy expenditure and prevent weight loss.67 The binding of leptin to its receptors leads to activation of Janus Kinase 2 (JAK2)/Signal Transducer And Activator Of Transcription 3 (STAT3), IRS-1, PI3K, and AMPK signaling pathways.68, 69, 70 In particular, the activation of JAK2/STAT3 signaling was found to be critically involved in the modulatory effects of leptin on energy homeostasis.70 When the cytosolic domain of the leptin receptor becomes phosphorylated by JAK proteins, it activates the MAPK/Extracellular Signal-Regulated Protein Kinase (ERK) 1/2 pathway. Pharmacological inhibition of this domain and ERK1/2 in the hypothalamus blocks the anorectic effects of leptin and results in mild obesity.71 While, leptin activates AMPK in hepatocytes and muscle tissue,72 it inhibits AMPK in the hypothalamus, hence reducing food intake and body weight.73,74

Although leptin reduces appetite as a circulating signal, obese individuals exhibit a higher circulating leptin concentration than normal-weight, suggesting a leptin resistance condition in obese individuals.75

Effect of AT and RT on leptin levels

Studies showed that lower physical activity levels, induce leptin resistance and hyperleptinemia,76 but higher levels, decrease circulating leptin by targeting a negative energy balance in obese individuals.77 It has been suggested that AT (in Wistar rats)78,79 and RT (in elderly postmenopausal women),80 exert beneficial effects on leptin sensitivity with or without weight loss. AT has been shown to reverse leptin resistance and reduce serum leptin levels in obese mice by down-regulating the suppressor of cytokines-3 in the JAK/STAT pathway.81 Moreover, leptin is very sensitive to energy-deficient status, so that, two or three days of fasting, lowers plasma leptin levels even before any loss in body fat mass.64,65 There is evidence showing that AT might regulate leptin secretion by increasing energy expenditure, sympathetic activity, and exercise stress metabolites.77

However, some studies suggested relatively unchanged or reduced serum leptin following RT in overweight individuals,82,83 and some studies reported an increased fat-free mass together with decreased leptin concentrations in MetS following RT.67,84 For an instant, Fedewa et al.85 reported that chronic exercise training (≥2 weeks; both AT or RT) leads to reduced leptin levels in elderly postmenopausal women, regardless of age and sex, but dependent on body fat percentage. Besides reducing fat mass, other mechanisms involved during a moderate to severe RT on leptin levels include peripheral glucose uptake, in the presence of lactate and acidosis, sympathetic stimulation of the adrenal gland, and glycogen depletion in elderly postmenopausal women.86

The mechanisms by which exercise training increases leptin levels have not been elucidated yet. It seems that PI3K/AKT, and Mammalian Target of Rapamycin (mTOR),87,88 PPARγ pathways, might be involved.89,90 Exercise training also affects the secretion of leptin, indirectly by altering glucocorticoids, serotonin, and estrogen levels in obese children.91 Insulin and glucocorticoid are known to function synergistically as long-term regulators of leptin expression by transcriptional or post-transcriptional mechanisms.92

In conclusion, long-term AT and RT with moderate to high intensities, modulate leptin serum concentration and thus regulate energy balance, appetite, and lipid/glucose metabolism in MetS.89,90

Omentin in MetS

Omentin is a 313 amino acids adipokine, with two isoforms of omentin-1 and omentin-2. Omentin-1 mRNA is expressed in visceral adipose tissue and stands as a predominant form of plasma.93,94 Clinical studies showed that circulating omentin-1 concentration is decreased in MetS and obesity.95,96 In contrast, an increased level of omentin might reflect the physiological compensatory mechanism in regulating insulin sensitivity97 and glucose homeostasis.95 Omentin-1 stimulates IRS and increases high-density lipoprotein, and finally, stimulates lipolysis.98,99 Also, administration of exogenous omentin-1, significantly decreases blood pressure in normotensive rats probably via enhancing the synthesis of NO and inhibiting Interleukin 6 (IL-6),100 besides energy expenditure regulation in skeletal muscles.101,102 In addition, omentin exerts extensive protective effects via various cell signaling pathways including AMPK.101,102 Then omentin-induced AMPK phosphorylation reduces the Ras/ERK signaling cascade,103 and suppresses TNF-α in macrophages.104 In addition, omentin-1 enhances glucose uptake via activating PKB105,106 and synergistically potentiates the adiponectin functions and alleviates insulin resistance.107

Effect of AT and RT on omentin levels

Studies showed that both AT and RT protocols increase omentin levels, and omentin exerts anti-inflammatory and insulin-sensitizing effects by inserting GLUT4 into the target cells membranes.95,96 Animal studies showed an increase in serum omentin level parallel with alleviating MetS components in ovariectomized obese rats following AT and RT intervention.108 For example, Madsen et al.109 demonstrated increased serum omentin level following strenuous and moderate AT in ovariectomized rats. In contrast, Urbanová et al.110 and Faramarzi et al.111 showed no change in omentin following long-term low intensity AT intervention, despite a significant reduction in body weight, fasting insulin, and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR). Interestingly, Huang L, reported a reduction in serum omentin levels in obese rats following AT (75% V˙O2max).112 Collectively, AT is capable to modulate the production of omentin, as a crosstalk cytokine linking muscle and adipose tissues in T2D Mellitus rat models.99 Finally, increased omentin level by AT and RT improves insulin sensitivity and glucose metabolism113 via stimulating phosphorylation of PKB and AMPK.102,114 Conversely, a negative correlation between omentin-1 and insulin, glucose, and IL-6 99,115 but a positive with adiponectin levels114 have been found. Thus, exercise-induced omentin elevation might regulate glucose homeostasis and stimulate lipolysis. Taken all together, high and moderate-intensity AT induce more favorable effects on the increased omentin production and secretion. RT is also capable to increase omentin serum levels, however, the evidence on different intensities is rare.

Apelin in MetS

Apelin is a 36 amino-acid peptide that contributes to glucose metabolism, lipolysis, blood pressure, cardiovascular functions, fluid homeostasis, food intake, and vasodilation.116 A positive association has been reported between high circulating apelin levels and insulin resistance,117,118 hypertension, heart failure, central obesity,118 high blood glucose, and dyslipidemia,119 although elevated apelin might be a physiological compensatory response to MetS.120 Apelin is expressed by adipose tissue and promotes glucose uptake through GLUT4, and alleviates insulin resistance by activating PI3K/AKT.121 In agreement with these findings, administration of recombinant apelin, increases glucose utilization122 and reduces blood glucose.123, 124, 125 Apelin receptor is a G protein-coupled receptor, which is expressed in vascular smooth muscle and myocardial cells.126 Clinical and experimental studies support a role for apelin in cardiovascular and metabolic disorders127 via AMPK, Endothelial Nitric Oxide Synthase (eNOS), PKB,124 and ERK1/2,128 pathways. Apelin improves glucose tolerance and insulin sensitivity, mainly by improving the skeletal muscles' metabolic functions125 and stimulating glucose transport in an AMPK-dependent manner.124 The ability of apelin to decrease blood glucose together with vasodilation and also reduction in blood pressure may open new therapeutic avenues for MetS.

Effect of AT and RT on apelin levels

The literature regarding exercise effects on apelin concentration in MetS status is inconsistent. For example, Besse-Patin et al.129 reported a twofold increase in apelin mRNA level in muscle, but not in adipose tissue following an 8-week AT program indicating upregulated muscle apelin expression in obese men. Another study in overweight subjects reported a considerable upregulation in apelin following 12 weeks of AT even with no significant weight loss in patients with T2D.130 Jang et al.131 and Nikseresht et al.132 suggested a significant elevation in apelin only following AT, but not RT in obese individuals. However a recent study reported an elevated apelin by RT.133 Considering the mechanisms underlying the effects of exercise on apelin, rodent studies have shown that the AT mediates GLUT-4 translocation to the cell membranes, and then might decrease apelin gene expression.129,132 On the other hand, RT-induced fatty acid oxidation might reduce apelin levels by increasing lipoprotein lipase activity in muscles and decreasing insulin resistance.133

Taken all together, long-term AT and RT, are efficient in reducing apelin levels in favor of AT.

Future studies are needed to clarify the effect of exercise intensity on apelin level in MetS.

Vaspin in MetS

Vaspin is formed of 412–415 amino acids released by adipose tissue that is associated with insulin resistance, obesity, and glucose intolerance in many diseases.134,135 Kloting et al.136 stated in their study that insulin sensitivity is the critical determinant of vaspin gene expression in adipose tissue. Surprisingly, administration of insulin, significantly upregulated vaspin mRNA in subcutaneous adipose tissue but reduced visceral fat.137,138 Conversely, administration of recombinant human vaspin in diabetic mice, significantly improved insulin sensitivity and glucose tolerance, increased GLUT4 and adiponectin in adipose tissues, whereas, suppressed leptin, resistin, and TNF-α.137,138 Overall, vaspin serves as an insulin sensitizer and anti-inflammatory adipokine.139

It has been reported that the effects of vaspin on glycemic control are mediated by inhibiting kallikrein 7; an in vitro protease degradation of human insulin.140 Also, vaspin might protect blood vessels by preventing free fatty acid-induced apoptosis in human vascular endothelial cells via PI3K/AKT/eNOS signaling pathway upregulation.141,142 In addition, vaspin inhibits NF-κB/Protein Kinase C (PKC) in vascular smooth muscle cells,143,144 and suppresses the JAK2/STAT3 signaling pathway activity.145

Effect of AT and RT on vaspin levels

Studies suggested that vaspin levels could be suppressed following different exercise protocols.146,147 It is known that vaspin concentration is lower in well-trained individuals compared to those with low physical fitness.148 Chang et al. and Shahdadi et al.135,149 showed a reduction in serum vaspin levels after AT in obese subjects. In contrast, a higher vaspin level has been found following 4-week AT in obese and T2D subjects.148 Also, Kadoglou et al. and Youn et al.130,148 reported an increase in serum vaspin levels in T2D patients and suggested that vaspin improves insulin sensitivity. Similar to AT, vaspin serum level is changed differently in response to RT. For example, two studies reported a significant reduction in serum vaspin level following RT in obese individuals,135,149 in contrast with Mahdirejei et al.150 who reported no significant alteration. RT might induce decreased vaspin mRNA expression secondary to muscle hypertrophy, increased basal metabolic rate, fat oxidation, and ultimately weight loss.151 It seems that vaspin serum concentration might be differentially regulated in an enzyme (nicotinamide phosphoribosyl transferase enzyme)-dependent manner.135,149 The exact signaling pathways by which AT and RT suppress vaspin production needs more investigation.

TNF-α and IL-6 in MetS

MetS is accompanied by an increase in pro-inflammatory cytokines,152 particularly TNF-α and IL-6.153,154 TNF-α is secreted by abdominal adipose tissue and immune system, and plays significant roles in inflammation and insulin resistance.155 For instance, TNF-α and its receptor genetic deletion, significantly improved insulin signaling in muscle and adipose tissue.156 Although the baseline expression of TNF-α in adipose tissues is relatively low, it is positively correlated with obesity and negatively with weight loss.157 TNF-α elevation has been repo reported to associate with morbidity and mortality in MetS.158,159 A parallel increase in both TNF-α and insulin resistance, together with visceral fat accumulation has been also found in rats model of MetS.160,161 It has been known that TNF-α induces insulin resistance in the skeletal muscles by promoting fatty acids incorporation into triacylglycerol via increasing several kinases.162 Some of these stress-related kinases, perpetuate a positive feedback mechanism for more TNF-α production, and promote chronic insulin resistance.163 In addition, increased TNF-α level induces hepatic fatty acids uptake, reduces fatty acid oxidation and TG export, together with elevation in mitochondrial Reactive Oxygen Species (ROS).164 Besides, higher level of TNF-α causes elevation in IL-6 as the secondary defense response. IL-6 secreted from adipose tissues has both pro-and anti-inflammatory effects and causes a wide range of different effects on lipid and glucose metabolism.165,166 Thus, TNF-α likely stands as a pro-inflammatory adipokine, and representative marker in the MetS pathogenesis and cardiovascular burden. TNF-α neutralization by antibodies or antagonists might be a future candidate therapy in MetS.

Two TNF-α receptors have been identified: Tumor Necrosis Factor Receptor Type 1 (TNFR1) and 2 (TNFR2). It is believed that TNFR1 is responsible for inflammatory actions, and mediates insulin resistance167 via down-regulating the insulin receptor expression, insulin-related substrate-1, stress-related kinases, and GLUT4.168 In other words, increased TNF-α directly inhibits IRS-1 tyrosine phosphorylation, and also activates protein-tyrosine phosphatases,169 and indirectly acts via increasing IL-6, to reduce insulin-dependent glucose uptake.170 IL-6 enhances lipolysis and fat oxidation via activation of AMPK171 and increases the activity of the insulin-degrading enzyme, and thus inhibits downstream signaling of insulin.172 Generally, TNF-α and IL-6 synergistically impair insulin signaling and induce chronic MetS.173

Effect of AT and RT on TNF-α and IL-6 levels

Exercise training has been known as the best non-invasive intervention without serious side effects, which alleviates inflammation and immune function in patients with MetS and diabetes.174,175 Since adipose tissue releases inflammatory markers, long-term weight loss is a useful strategy to reduce the risk of high TNF-α and IL-6 levels in overweight and obese individuals.176 However, existing literature regarding the TNF-α alteration following exercise seems to be dual. For instant, acute, intensive, and unaccustomed exercise training sessions increase the TNF- α and IL-6, while adaptation to long-term exercise protocols might reduce TNF- α and IL-6.177,178 Allen et al.179 reported no change in serum TNF- α following 9-week high-intensity AT in sedentary adults. While Abd El-Kader et al.180 showed significantly decreased TNF-α and IL-6 following 3 months of AT in obese T2D patients (both men and women; aged 40–55 years). They also reported that moderate AT (65%–75% of maximum heart rate [HRmax]) was more effective in reducing TNF-α and IL-6 than mild AT (55%–65% of HRmax). Gerosa-Neto et al.181 investigated the impact of 16-week high-intensity interval (90% HRmax) and moderate-intensity AT (70% HRmax) on subclinical inflammation in overweight or obese adults. They demonstrated that 16 weeks of training decreased blood levels of IL-6, but increased TNF-α in the high-intensity group. Interestingly TNF-α was decreased in the moderate-intensity group, suggesting efficacy for both high and moderate-intensity AT promoting changes in inflammatory profile in overweight or obesity subjects, in favor of moderate-intensity in case of TNF-α responses.

It seems that besides the contracting skeletal muscle cells, the local connective tissue produces more IL-6 as well, in response to a single-bout prolonged exercise in women with MetS.182 It should be noticed that a single exercise session induces an acute robust inflammatory response, while chronic AT induces long-lasting adaptation that might be different from the primary response regardless of fat loss.152,183 Studies showed that AT and RT (to a fewer extent) could be effective in the prevention and delay of chronic inflammatory diseases onset via reducing pro-inflammatory cytokines, particularly TNF-α in patients with MetS.62,184,185

Anti-inflammatory effects of exercise are related not only to adipose tissue but also to the skeletal muscle and peripheral blood mononuclear cells.186 One of the possible exercise-induced mechanisms (in favor of AT) might be the reduced Toll-Like Receptors 4 (TLR4) expression in monocytes,187,188 which are capable to induce TNF-α and IL-6 expression by activating NF-κB.189 Increase in Inhibitory-κB Kinase (IKK) β phosphorylation, but inhibition in nicotinamide adenine dinucleotide phosphate oxidase, are the mechanisms by which RT might lead to reduced TNF-α and IL-6 mRNA expression and secretion in men with MetS.174,190 Furthermore, in an animal study, 15-week moderate AT has been shown to confront metabolic disorders by suppressing TNF-α signaling responses and also promoting muscle energy-sensing network proteins, including AMPK, Sirtuin-1, PPARγ Co-Activator 1α (PGC-1α).191

Overall, regarding anti-inflammatory effects, long-term moderate AT and RT protocols, especially those inducing fat loss and muscle hypertrophy might be a good candidate therapy in MetS.

Wnt5a and SFRP-5 in MetS

Extensive investigations have reported the significance of the Wnt Family Member 5A (WNT5a) pathways in regulating body mass, glucose metabolism, lipogenesis, LDL clearance, vascular smooth muscle plasticity, liver fat, and liver inflammation.192,193 WNT5a is an adipokine contributing to obesity-associated inflammation.194,195 The WNT5a activity is regulated by Secreted Frizzled-Related Protein 5 (SFRP-5), an extracellular Wnt signaling antagonist. SFRP5 is an anti-inflammatory adipokine, secreted by adipocytes acts as a decoy receptor by binding WNT5a and preventing its association with frizzled proteins. Lower SFRP-5 levels are correlated with obesity, impaired glucose tolerance, insulin resistance, and T2D196,197 which results in the activation of WNT5A canonical/non-canonical signaling pathways.198,199 WNT5a exerts both inflammatory and anti-inflammatory effects, in part by modulating the NF-κB pathway.200 Moreover, TNF-α has been known to induce WNT5a secretion from adipocytes and causes an imbalance in WNT5a/SFRP5 signaling.194,201

The Wnt signaling pathway is a ubiquitous signaling cascade that regulates a wide range of physiologic processes. There are three signaling pathways; the canonical pathway (β-catenin dependent), the non-canonical, and the Wnt/calcium signaling pathway.193,202 In the canonical pathway, Wnt ligands bind to the frizzled receptor and low-density lipoprotein receptor-related protein (LRP) 5 or 6, resulting in the internuclear β-catenin accumulation and leading to transcriptional regulation.193 In the non-canonical pathway, the Wnt ligands bind to the frizzled receptor and the receptor-like tyrosine kinase/RTK-like orphan co-receptors and activate the Rho/Rac signaling cascades. However, the non-canonical pathway promotes pro-inflammatory cytokines expression.203 It should be also noticed that Wnt and SFRP-5 proteins undergo several post-translational modifications, including N-glycosylation and acylation, both of which are required for signaling activity rather than secretion.204 Also, lipid modification, which is identified as palmitoleoylation, is essential for Wnt protein secretion.204 In the Wnt/calcium signaling pathway, Wnt ligands bind to the frizzled receptor and activate phospholipase C, and then increase intracellular calcium level.205 Some studies suggested non-canonical Wnt signaling as a metabolic dysregulation biomarker in both rats and humans with MetS.195,206 Inversely, a higher SFRP5 level inhibits the non-canonical WNT5A pathway to improve insulin sensitivity.207 Non-canonical Wnt signaling shifts the lipids storage from adipose tissue to liver and muscle, promoting metabolic complications of obesity such as insulin resistance208,209 via activating c-Jun N-terminal Kinases (JNK) cascade, and IRS-1 serine phosphorylation.210 Therefore, crosstalk of these signaling pathways promotes the pro-inflammatory state and MetS progression.194,211 On the other hand, SFRP5 neutralizes non-canonical JNK activation by WNT5a in macrophages and adipocytes.192 The JNK signaling pathway in adipocytes and macrophages has emerged as an essential mediator of adipose tissue inflammation that affects systemic metabolism.192,212 Thus, the SFRP5/JNK regulatory axis in fat represents a potential target for controlling obesity-linked abnormalities in glucose homeostasis by blocking the WNT5a.213 Generally, impaired canonical Wnt signaling and the activation of non-canonical Wnt signaling constitute the underlying mechanisms for cardio-metabolic abnormalities.

Effect of AT and RT on Wnt and SFRP-5

It has been reported that an increase in non-canonical Wnt signaling in human visceral adipose tissue is positively associated with cardiovascular risk factors and insulin resistance, which can be alleviated by high-intensity AT.160,195 Few studies considered the effect of RT on WNT5A/SFRP5. For example, Leal et al.214 stated that eight-week RT triggers more significant responses in the Wnt pathway, and potently elevates the expression of Wnt pathway genes and β-catenin.215,216 However, Mir et al.217 showed an improvement of T2D as a result of increased SFRP5 serum level and decreased WNT5a serum level after 12 weeks of combined exercise (high intensity AT and RT) possibly due to the reduction in fat mass. They also reported a significant negative relationship between SFRP5 and WNT5A.

It seems that dysregulated Wnt signaling pathway, which underlies the pleiotropy of MetS might be alleviated by both AT and RT protocols. RT might potentiate the Wnt function by influencing various post-translational mediators of this pathway.218 Then, Wnt binds to the cell surface transmembrane receptors which involve direct binding to several intracellular proteins including glycogen synthase kinase-3β, and disheveled.204,219,220

Therefore, combined AT and RT might elevate SFRP5 and reduce Wnt more significantly than a single protocol of either AT or RT.

Conclusion

Considering diverse and complex adipokines functions in MetS, summarizing their role and selecting one core link to the MetS, seems to be an oversimplification. Thus, a panel of adipokines rather than an individual biomarker would be a useful and relatively reliable marker for identifying those who are at risk for developing MetS and related diseases.

Our search strategy for this review focused on those adipokines that have been studied on MetS in various laboratories including ours. Data for this review were identified by searches of science direct advance search, PubMed, and WOS to get any related articles available using the terms” adipokine, adiponectin, omentin, apelin, leptin, Wnt5a, Sfrp5, IL-6, and MetS, AT and RT. We tried to include articles on the English language with preference to publications from the past 15 years.

The strength of this review is summarizing the selected adipokines alteration in MetS and response to AT and RT. However, there are some limitations such as not including myokines and other adipokines due to complexities and varieties.

Reviewing literature revealed that adipokines are categorized into two groups of pro and anti-inflammatory molecules. Pro-inflammatory is related to insulin resistance and inflammation, in contrast to anti-inflammatory which exerts insulin sensitivity and lipid homeostasis. Furthermore, both moderate AT and RT, successfully modulate the adipokines profile toward the health-promoting adipokines. Also, it could be concluded that the constant, long-lasting alterations in adipokines level are more prominent following long-term exercise protocols, in contrast with acute and high-intensity exercise which stimulates pro-inflammatory adipokines.

Although the exact mechanisms underlying the beneficial effects of AT and RT in MetS have not been understood well, modulation of adipokines secreted from adipose tissues, together with weight loss consequences, might be the most important factor linking the molecular signaling pathways with improved glucose homeostasis, and better metabolic state.

Moderate-intensity AT and RT are associated with improved insulin sensitivity, improved circulation, mitochondrial biogenesis, and the release of numerous adipokines. Currently, among various mechanisms, adiponectin/AMPK signaling seems to be the main mediator of metabotropic effects of exercise. These molecular signalings anticipate a better understanding of mechanisms that will enable the development of pharmaceuticals, particularly for sedentary individuals who are at higher risk of developing Mets (Fig. 1, Graphical abstract).

Fig. 1.

Fig. 1

Role of adipokines and AMPK/PPARɣ, mTOR and NF-Kβ, pathways in the development or progression of insulin resistance. in metabolic syndrome.

For future studies, several important points should be considered:

  • -

    To clarify the source of circulating adipokines in response to MetS, AT, and RT

  • -

    To elucidate both up and down streams of adipokines together in response to a specified protocol of exercise to distinguish between cause and effect, compensatory or operative functions.

  • -

    To administrate recombinant adipokines, and also monoclonal antibodies in a rodent model to evaluate their effects on MetS.

Authors' contributions

Parvin Babaei drafted the MetS part based on the studies carried out in her lab and coordinated the contents of the manuscript. Rastegar Hoseini wrote the theoretical parts of the exercise. Both authors have read and approved the final version of the manuscript and agreed with the order of presentation of the authors.

Funding

No external source of funding was used in the preparation of this article.

Submission statement

Hereby we attest that the submitted manuscript has not been published previously, and it is not under consideration for publication elsewhere. We also agree that if accepted, it will not be published elsewhere including electronically in the same form, in English or in any other language.

Conflict of interest

The authors report no biomedical financial interests or potential conflicts of interest.

Contributor Information

Parvin Babaei, Email: p_babaei@gums.ac.ir.

Rastegar Hoseini, Email: r.hoseini@razi.ac.ir.

References

  • 1.Alonso-Gómez A.M., Tojal Sierra L., Fortuny Frau E., et al. Diastolic dysfunction and exercise capacity in patients with metabolic syndrome and overweight/obesity. Int J Cardiol Heart Vasc. 2018;22:67–72. doi: 10.1016/j.ijcha.2018.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Dibaise J.K., Foxx-Orenstein A.E. Role of the gastroenterologist in managing obesity. Expet Rev Gastroenterol Hepatol. 2013;7(5):439–451. doi: 10.1586/17474124.2013.811061. [DOI] [PubMed] [Google Scholar]
  • 3.Abdelaal M., le Roux C.W., Docherty N.G. Morbidity and mortality associated with obesity. Ann Transl Med. 2017;5(7):161. doi: 10.21037/atm.2017.03.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Funcke J.-B., Scherer P.E. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res. 2019;60(10):1648–1684. doi: 10.1194/jlr.r094060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Santilli F., D'Ardes D., Teresa Guagnano M., Davi G. Metabolic syndrome: sex-related cardiovascular risk and therapeutic approach. Curr Med Chem. 2017;24(24):2602–2627. doi: 10.2174/0929867324666170710121145. [DOI] [PubMed] [Google Scholar]
  • 6.Chung H.S., Choi K.M. Adipokines and myokines: a pivotal role in metabolic and cardiovascular disorders. Curr Med Chem. 2018;25(20):2401–2415. doi: 10.2174/0929867325666171205144627. [DOI] [PubMed] [Google Scholar]
  • 7.Peterson J.M., Clark W.A., Marrs J.-A., Alamian A. Serum adipokines and metabolic syndrome risk factors in hispanic children. Faseb J. 2017;31(S1):1037. doi: 10.1096/fasebj.31.1_supplement.1037.5. [DOI] [Google Scholar]
  • 8.Hu X., Guo F. Amino acid sensing in metabolic homeostasis and health. Endocr Rev. 2021;42(1):56–76. doi: 10.1210/endrev/bnaa026. [DOI] [PubMed] [Google Scholar]
  • 9.Sinha R.A., Singh B.K., Yen P.M. Reciprocal crosstalk between autophagic and endocrine signaling in metabolic homeostasis. Endocr Rev. 2017;38(1):69–102. doi: 10.1210/er.2016-1103. [DOI] [PubMed] [Google Scholar]
  • 10.You T., Nicklas B.J., Ding J., et al. The metabolic syndrome is associated with circulating adipokines in older adults across a wide range of adiposity. J Gerontol A Biol Sci Med Sci. 2008;63(4):414–419. doi: 10.1093/gerona/63.4.414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Guo S. Insulin signaling, resistance, and the metabolic syndrome: insights from mouse models to disease mechanisms. J Endocrinol. 2014;220(2):T1–T23. doi: 10.1530/joe-13-0327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nieman D.C., Wentz L.M. The compelling link between physical activity and the body's defense system. J Sport Health Sci. 2019;8(3):201–217. doi: 10.1016/j.jshs.2018.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Smith G.I., Mittendorfer B., Klein S. Metabolically healthy obesity: facts and fantasies. J Clin Invest. 2019;129(10):3978–3989. doi: 10.1172/jci129186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Imazu A.A., Goessler K.F., Casonatto J., Polito M.D. The influence of physical training status on postexercise hypotension in patients with hypertension: a cross-sectional study. Blood Pres Monit. 2017;22(4):196–201. doi: 10.1097/mbp.0000000000000255. [DOI] [PubMed] [Google Scholar]
  • 15.Batista M., Jr., Rosa J., Lopes R., et al. Exercise training changes IL-10/TNF-α ratio in the skeletal muscle of post-MI rats. Cytokine. 2010;49(1):102–110. doi: 10.1016/j.cyto.2009.10.007. [DOI] [PubMed] [Google Scholar]
  • 16.Warburton D.E., Nicol C.W., Bredin S.S. Health benefits of physical activity: the evidence. CMAJ (Can Med Assoc J) 2006;174(6):801–809. doi: 10.1503/cmaj.051351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mitsui R., Fukushima M., Taniguchi A., et al. Insulin secretory capacity and insulin sensitivity in impaired fasting glucose in Japanese. J Diabetes Investig. 2012;3(4):377–383. doi: 10.1111/j.2040-1124.2012.00201.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Tan X., Chapman C.D., Cedernaes J., Benedict C. Association between long sleep duration and increased risk of obesity and type 2 diabetes: a review of possible mechanisms. Sleep Med Rev. 2018;40:127–134. doi: 10.1016/j.smrv.2017.11.001. [DOI] [PubMed] [Google Scholar]
  • 19.Lira F.S., Rosa J.C., Zanchi N.E., et al. Regulation of inflammation in the adipose tissue in cancer cachexia: effect of exercise. Cell Biochem Funct. 2009;27(2):71–75. doi: 10.1002/cbf.1540. [DOI] [PubMed] [Google Scholar]
  • 20.Batista Júnior M.L., Lopes R.D., Seelaender M.C.L., Lopes A.C. Anti-inflammatory effect of physical training in heart failure: role of TNF-α and IL-10. Arq Bras Cardiol [in Portuguese] 2009;93(6):692–700. doi: 10.1590/s0066-782x2009001200021. [DOI] [PubMed] [Google Scholar]
  • 21.Kruk J., Kotarska K., Aboul-Enein B.H. Physical exercise and catecholamines response: benefits and health risk: possible mechanisms. Free Radic Res. 2020;54(2-3):105–125. doi: 10.1080/10715762.2020.1726343. [DOI] [PubMed] [Google Scholar]
  • 22.Bruunsgaard H. Physical activity and modulation of systemic low-level inflammation. J Leukoc Biol. 2005;78(4):819–835. doi: 10.1189/jlb.0505247. [DOI] [PubMed] [Google Scholar]
  • 23.Neves M., Retameiro A.C.B., Tavares A.L.F., et al. Physical exercise and low-level laser therapy on the nociception and leukocyte migration of Wistar rats submitted to a model of rheumatoid arthritis. Laser Med Sci. 2020;35(6):1277–1287. doi: 10.1007/s10103-019-02905-2. [DOI] [PubMed] [Google Scholar]
  • 24.Trayhurn P., Drevon C.A., Eckel J. Secreted proteins from adipose tissue and skeletal muscle–adipokines, myokines and adipose/muscle cross-talk. Arch Physiol Biochem. 2011;117(2):47–56. doi: 10.3109/13813455.2010.535835. [DOI] [PubMed] [Google Scholar]
  • 25.Kershaw E.E., Flier J.S. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89(6):2548–2556. doi: 10.1210/jc.2004-0395. [DOI] [PubMed] [Google Scholar]
  • 26.Sjøberg K.A., Frøsig C., Kjøbsted R., et al. Exercise increases human skeletal muscle insulin sensitivity via coordinated increases in microvascular perfusion and molecular signaling. Diabetes. 2017;66(6):1501–1510. doi: 10.2337/db16-1327. [DOI] [PubMed] [Google Scholar]
  • 27.Yoon K.J., Zhang D., Kim S.J., Lee M.C., Moon H.Y. Exercise-induced AMPK activation is involved in delay of skeletal muscle senescence. Biochem Biophys Res Commun. 2019;512(3):604–610. doi: 10.1016/j.bbrc.2019.03.086. [DOI] [PubMed] [Google Scholar]
  • 28.Huang J., Wang X., Zhu Y., et al. Exercise activates lysosomal function in the brain through AMPK-SIRT1-TFEB pathway. CNS Neurosci Ther. 2019;25(6):796–807. doi: 10.1111/cns.13114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Marwarha G., Claycombe-Larson K., Lund J., Ghribi O. Palmitate-Induced SREBP1 expression and activation underlies the increased BACE 1 activity and amyloid beta genesis. Mol Neurobiol. 2019;56(7):5256–5269. doi: 10.1007/s12035-018-1451-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Olivier S., Foretz M., Viollet B. Promise and challenges for direct small molecule AMPK activators. Biochem Pharmacol. 2018;153:147–158. doi: 10.1016/j.bcp.2018.01.049. [DOI] [PubMed] [Google Scholar]
  • 31.Islam H., Hood D.A., Gurd B.J. Looking beyond PGC-1α: emerging regulators of exercise-induced skeletal muscle mitochondrial biogenesis and their activation by dietary compounds. Appl Physiol Nutr Metabol. 2020;45(1):11–23. doi: 10.1139/apnm-2019-0069. [DOI] [PubMed] [Google Scholar]
  • 32.Ramachandran V., Saravanan R. Glucose uptake through translocation and activation of GLUT4 in PI3K/Akt signaling pathway by asiatic acid in diabetic rats. Hum Exp Toxicol. 2015;34(9):884–893. doi: 10.1177/0960327114561663. [DOI] [PubMed] [Google Scholar]
  • 33.Vega R.B., Konhilas J.P., Kelly D.P., Leinwand L.A. Molecular mechanisms underlying cardiac adaptation to exercise. Cell Metabol. 2017;25(5):1012–1026. doi: 10.1016/j.cmet.2017.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhang X., Xu A., Chung S.K., et al. Selective inactivation of c-Jun NH2-terminal kinase in adipose tissue protects against diet-induced obesity and improves insulin sensitivity in both liver and skeletal muscle in mice. Diabetes. 2011;60(2):486–495. doi: 10.2337/db10-0650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Banu N., Elango K. Adiponectin level in type 2 diabetes and its complication–A review. J Pharmaceut Sci Res. 2019;11(4):1172–1174. [Google Scholar]
  • 36.Zohmangaihi D., Sharma S., Madhu S. Adiponectin, IL-6 and hsCRP: interplay of inflammation with obesity and type 2 diabetes in Indian population. J Diabetes Metabol. 2019;10(3):1–7. doi: 10.35248/2155-6156.19.10.822. [DOI] [Google Scholar]
  • 37.Adiyaman S.C., Ozer M., Saydam B.O., Akinci B. The role of adiponectin in maintaining metabolic homeostasis. Curr Diabetes Rev. 2020;16(2):95–103. doi: 10.2174/1573399815666190702155733. [DOI] [PubMed] [Google Scholar]
  • 38.Bouassida A., Chamari K., Zaouali M., Feki Y., Zbidi A., Tabka Z. Review on leptin and adiponectin responses and adaptations to acute and chronic exercise. Br J Sports Med. 2010;44(9):620–630. doi: 10.1136/bjsm.2008.046151. [DOI] [PubMed] [Google Scholar]
  • 39.Lindberg S., Jensen J.S., Bjerre M., et al. Low adiponectin levels at baseline and decreasing adiponectin levels over 10 years of follow-up predict risk of the metabolic syndrome. Diabetes Metab. 2017;43(2):134–139. doi: 10.1016/j.diabet.2016.07.027. [DOI] [PubMed] [Google Scholar]
  • 40.Muppala S., Konduru S.K., Merchant N., et al. Adiponectin: its role in obesity-associated colon and prostate cancers. Crit Rev Oncol Hematol. 2017;116:125–133. doi: 10.1016/j.critrevonc.2017.06.003. [DOI] [PubMed] [Google Scholar]
  • 41.Wang X., Chen Q., Pu H., et al. Adiponectin improves NF-κB-mediated inflammation and abates atherosclerosis progression in apolipoprotein E-deficient mice. Lipids Health Dis. 2016;15:33. doi: 10.1186/s12944-016-0202-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tore F., Tonchev A., Fiore M., et al. From adipose tissue protein secretion to adipopharmacology of disease. Immunol Endocr Metab Agents Med Chem. 2007;7(2):149–155. doi: 10.2174/187152207780363712. [DOI] [Google Scholar]
  • 43.Okamoto Y., Kihara S., Ouchi N., et al. Adiponectin reduces atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2002;106(22):2767–2770. doi: 10.1161/01.cir.0000042707.50032.19. [DOI] [PubMed] [Google Scholar]
  • 44.Roy B., Palaniyandi S.S. Tissue-specific role and associated downstream signaling pathways of adiponectin. Cell Biosci. 2021;11(1):77. doi: 10.1186/s13578-021-00587-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Miyamoto L., Yamane M., Tomida Y., et al. Nitrite activates 5′ AMP-activated protein kinase-endothelial nitric oxide synthase pathway in human glomerular endothelial cells. Biol Pharm Bull. 2017;40(11):1866–1872. doi: 10.1248/bpb.b17-00316. [DOI] [PubMed] [Google Scholar]
  • 46.Wang Y., Ma X.L., Lau W.B. Cardiovascular adiponectin resistance: the critical role of adiponectin receptor modification. Trends Endocrinol Metabol. 2017;28(7):519–530. doi: 10.1016/j.tem.2017.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen Y., Zheng Y., Liu L., et al. Adiponectin inhibits TNF-α-activated PAI-1 expression via the cAMP-PKA-AMPK-NF-κB axis in human umbilical vein endothelial cells. Cell Physiol Biochem. 2017;42(6):2342–2352. doi: 10.1159/000480006. [DOI] [PubMed] [Google Scholar]
  • 48.Yamauchi T., Nio Y., Maki T., et al. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med. 2007;13(3):332–339. doi: 10.1038/nm1557. [DOI] [PubMed] [Google Scholar]
  • 49.Yamada H., Suzuki D., Kakei M., Kusaka I., Ishikawa S. Close association of hypoadiponectinemia and increased insulin resistance in non-obese Japanese type 2 diabetes with visceral adiposity. J Metab Syndrome. 2016;5(4):215. doi: 10.4172/2167-0943.1000215. [DOI] [Google Scholar]
  • 50.Zeng Q., Isobe K., Fu L., et al. Effects of exercise on adiponectin and adiponectin receptor levels in rats. Life Sci. 2007;80(5):454–459. doi: 10.1016/j.lfs.2006.09.031. [DOI] [PubMed] [Google Scholar]
  • 51.Sun L., Lv Y., Tian J., et al. Regular swimming exercise attenuated neuroma pain in rats: involvement of leptin and adiponectin. J Pain. 2019;20(9):1112–1124. doi: 10.1016/j.jpain.2019.02.097. [DOI] [PubMed] [Google Scholar]
  • 52.Jürimäe J., Purge P., Jürimäe T. Adiponectin and stress hormone responses to maximal sculling after volume-extended training season in elite rowers. Metabolism. 2006;55(1):13–19. doi: 10.1016/j.metabol.2005.06.020. [DOI] [PubMed] [Google Scholar]
  • 53.Magherini F., Fiaschi T., Marzocchini R., et al. Oxidative stress in exercise training: the involvement of inflammation and peripheral signals. Free Radic Res. 2019;53(11-12):1155–1165. doi: 10.1080/10715762.2019.1697438. [DOI] [PubMed] [Google Scholar]
  • 54.Damirchi A., Mehdizade R., Ansar M., Soltani B., Babaei P. Effects of aerobic exercise training on visceral fat and serum adiponectin concentration in ovariectomized rats. Climacteric. 2010;13(2):171–178. doi: 10.3109/13697130903360234. [DOI] [PubMed] [Google Scholar]
  • 55.Lee J.A., Kim J.W., Kim D.Y. Effects of yoga exercise on serum adiponectin and metabolic syndrome factors in obese postmenopausal women. Menopause. 2012;19(3):296–301. doi: 10.1097/gme.0b013e31822d59a2. published correction appears in Menopause. 2012 Apr;19(4):486. [DOI] [PubMed] [Google Scholar]
  • 56.ADv Frankenberg, Reis A.F., Gerchman F. Relationships between adiponectin levels, the metabolic syndrome, and type 2 diabetes: a literature review. Arch Endocrinol Metab. 2017;61(6):614–622. doi: 10.1590/2359-3997000000316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ward L.J., Nilsson S., Hammar M., et al. Resistance training decreases plasma levels of adipokines in postmenopausal women. Sci Rep. 2020;10(1):19837. doi: 10.1038/s41598-020-76901-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.de Mello M.T., de Piano A., Carnier J., et al. Long-term effects of aerobic plus resistance training on the metabolic syndrome and adiponectinemia in obese adolescents. J Clin Hypertens. 2011;13(5):343–350. doi: 10.1111/j.1751-7176.2010.00388.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Strasser B., Siebert U., Schobersberger W. Resistance training in the treatment of the metabolic syndrome : a systematic review and meta-analysis of the effect of resistance training on metabolic clustering in patients with abnormal glucose metabolism. Sports Med. 2010;40(5):397–415. doi: 10.2165/11531380-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 60.Gastebois C., Villars C., Drai J., et al. Effects of training and detraining on adiponectin plasma concentration and muscle sensitivity in lean and overweight men. Eur J Appl Physiol. 2016;116(11-12):2135–2144. doi: 10.1007/s00421-016-3466-z. [DOI] [PubMed] [Google Scholar]
  • 61.Lucotti P., Monti L.D., Setola E., et al. Aerobic and resistance training effects compared to aerobic training alone in obese type 2 diabetic patients on diet treatment. Diabetes Res Clin Pract. 2011;94(3):395–403. doi: 10.1016/j.diabres.2011.08.002. [DOI] [PubMed] [Google Scholar]
  • 62.Ostman C., Smart N., Morcos D., Duller A., Ridley W., Jewiss D. The effect of exercise training on clinical outcomes in patients with the metabolic syndrome: a systematic review and meta-analysis. Cardiovasc Diabetol. 2017;16(1):110. doi: 10.1186/s12933-017-0590-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Choi J.R., Kim J.Y., Huh J.H., Kim S.H., Koh S.B. Contribution of obesity as an effect regulator to an association between serum leptin and incident metabolic syndrome. Clin Chim Acta. 2018;487:275–280. doi: 10.1016/j.cca.2018.09.038. [DOI] [PubMed] [Google Scholar]
  • 64.Ahsan F., Sharif M.K., Butt M.S., Shehzad A., Khan M.I. Pathophysiological role of leptin for human health: a review. Pakistan J Food Sci. 2017;27(1):46–52. [Google Scholar]
  • 65.Perry R.J., Wang Y., Cline G.W., et al. Leptin mediates a glucose-fatty acid cycle to maintain glucose homeostasis in starvation. Cell. 2018;172(1-2):234–248. doi: 10.1016/j.cell.2017.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Considine R.V., Sinha M.K., Heiman M.L., et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292–295. doi: 10.1056/nejm199602013340503. [DOI] [PubMed] [Google Scholar]
  • 67.Ghadge A.A., Khaire A.A. Leptin as a predictive marker for metabolic syndrome. Cytokine. 2019;121:154735. doi: 10.1016/j.cyto.2019.154735. [DOI] [PubMed] [Google Scholar]
  • 68.Barnes T.M., Shah K., Allison M.B., et al. Identification of the leptin receptor sequences crucial for the STAT3-Independent control of metabolism. Mol Metabol. 2020;32:168–175. doi: 10.1016/j.molmet.2019.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ghasemi A., Saeidi J., Azimi-Nejad M., Hashemy S.I. Leptin-induced signaling pathways in cancer cell migration and invasion. Cell Oncol. 2019;42(3):243–260. doi: 10.1007/s13402-019-00428-0. [DOI] [PubMed] [Google Scholar]
  • 70.Barrios-Correa A.A., Estrada J.A., Contreras I. Leptin signaling in the control of metabolism and appetite: lessons from animal models. Mol Neurosci. 2018;66(3):390–402. doi: 10.1007/s12031-018-1185-0. [DOI] [PubMed] [Google Scholar]
  • 71.Zhang E.E., Chapeau E., Hagihara K., Feng G.-S. Neuronal Shp2 tyrosine phosphatase controls energy balance and metabolism. Proc Natl Acad Sci U S A. 2004;101(45):16064–16069. doi: 10.1073/pnas.0405041101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Uotani S., Abe T., Yamaguchi Y. Leptin activates AMP-activated protein kinase in hepatic cells via a JAK2-dependent pathway. Biophys Res Commun. 2006;351(1):171–175. doi: 10.1016/j.bbrc.2006.10.015. [DOI] [PubMed] [Google Scholar]
  • 73.Minokoshi Y., Alquier T., Furukawa N., et al. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 2004;428(6982):569–574. doi: 10.1038/nature02440. [DOI] [PubMed] [Google Scholar]
  • 74.Claret M., Smith M.A., Batterham R.L., et al. AMPK is essential for energy homeostasis regulation and glucose sensing by POMC and AgRP neurons. J Clin Invest. 2007;117(8):2325–2336. doi: 10.1172/jci31516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zimmermann G.S., Bastos M.F., Dias Gonçalves T.E., Chambrone L., Duarte P.M. Local and circulating levels of adipocytokines in obese and normal weight individuals with chronic periodontitis. J Periodontol. 2013;84(5):624–633. doi: 10.1902/jop.2012.120254. [DOI] [PubMed] [Google Scholar]
  • 76.De Git K., Adan R. Leptin resistance in diet-induced obesity: the role of hypothalamic inflammation. Obes Rev. 2015;16(3):207–224. doi: 10.1111/obr.12243. [DOI] [PubMed] [Google Scholar]
  • 77.Gar C., Rottenkolber M., Grallert H., et al. Physical fitness and plasma leptin in women with recent gestational diabetes. PLoS One. 2017;12(6) doi: 10.1371/journal.pone.0179128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Garcia N.F., Silva C.P., Ferreira M., Jr., Oharomari L.K., Rocha T., Moraes Cd. 7-week aerobic exercise training reduces adipocyte area and improves insulin sensitivity in Wistar rats fed a highly palatable diet. Motriz: Revista de Educação Física. 2016;22(1):12–17. doi: 10.1590/s1980-65742016000100002. [DOI] [Google Scholar]
  • 79.Zhao J., Tian Y., Xu J., Liu D., Wang X., Zhao B. Endurance exercise is a leptin signaling mimetic in hypothalamus of Wistar rats. Lipids Health Dis. 2011;10:225. doi: 10.1186/1476-511X-10-225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Botero J.P., Shiguemoto G.E., Prestes J., et al. Effects of long-term periodized resistance training on body composition, leptin, resistin and muscle strength in elderly post-menopausal women. J Sports Med Phys Fit. 2013;53(3):289–294. [PubMed] [Google Scholar]
  • 81.Ramos-Lobo A.M., Donato J., Jr. The role of leptin in health and disease. Temperature (Austin) 2017;4(3):258–291. doi: 10.1080/23328940.2017.1327003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lau P.W., Kong Z., Choi C-r, et al. Effects of short-term resistance training on serum leptin levels in obese adolescents. J Exerc Sci Fit. 2010;8(1):54–60. doi: 10.1016/s1728-869x(10)60008-1. [DOI] [Google Scholar]
  • 83.Ahmadizad S., Ghorbani S., Ghasemikaram M., Bahmanzadeh M. Effects of short-term nonperiodized, linear periodized and daily undulating periodized resistance training on plasma adiponectin, leptin and insulin resistance. Clin Biochem. 2014;47(6):417–422. doi: 10.1016/j.clinbiochem.2013.12.019. [DOI] [PubMed] [Google Scholar]
  • 84.Nappo A., Gonzalez-Gil E., Ahrens W., et al. Analysis of the association of leptin and adiponectin concentrations with metabolic syndrome in children: results from the IDEFICS study. Nutr Metabol Cardiovasc Dis. 2017;27(6):543–551. doi: 10.1016/j.numecd.2017.04.003. [DOI] [PubMed] [Google Scholar]
  • 85.Fedewa M.V., Hathaway E.D., Ward-Ritacco C.L., Williams T.D., Dobbs W.C. The effect of chronic exercise training on leptin: a systematic review and meta-analysis of randomized controlled trials. Sports Med. 2018;48(6):1437–1450. doi: 10.1007/s40279-018-0897-1. [DOI] [PubMed] [Google Scholar]
  • 86.Prestes J., da Cunha Nascimento D., de Sousa Neto I.V., et al. The effects of muscle strength responsiveness to periodized resistance training on resistin, leptin, and cytokine in elderly postmenopausal women. J Strength Condit Res. 2018;32(1):113–120. doi: 10.1519/jsc.0000000000001718. [DOI] [PubMed] [Google Scholar]
  • 87.Marques-Oliveira G.H., Silva T.M., Lima W.G., Valadares H.M.S., Chaves V.E. Insulin as a hormone regulator of the synthesis and release of leptin by white adipose tissue. Peptides. 2018;106:49–58. doi: 10.1016/j.peptides.2018.06.007. [DOI] [PubMed] [Google Scholar]
  • 88.Dantas W.S., Roschel H., Murai I.H., et al. Exercise-induced increases in insulin sensitivity after bariatric surgery are mediated by muscle extracellular matrix remodeling. Diabetes. 2020;69(8):1675–1691. doi: 10.2337/db19-1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Petridou A., Tsalouhidou S., Tsalis G., Schulz T., Michna H., Mougios V. Long-term exercise increases the DNA binding activity of peroxisome proliferator–activated receptor γ in rat adipose tissue. Metabolism. 2007;56(8):1029–1036. doi: 10.1016/j.metabol.2007.03.011. [DOI] [PubMed] [Google Scholar]
  • 90.Tsai M., Asakawa A., Amitani H., Inui A. Stimulation of leptin secretion by insulin. Indian J Endocrinol Metab. 2012;16(Suppl 3):S543–S548. doi: 10.4103/2230-8210.105570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Sirico F., Bianco A., D'Alicandro G., et al. Effects of physical exercise on adiponectin, leptin, and inflammatory markers in childhood obesity: systematic review and meta-analysis. Child Obes. 2018;14(4):207–217. doi: 10.1089/chi.2017.0269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.McGee S.L., Hargreaves M. Exercise adaptations: molecular mechanisms and potential targets for therapeutic benefit. Nat Rev Endocrinol. 2020;16(9):495–505. doi: 10.1038/s41574-020-0377-1. [DOI] [PubMed] [Google Scholar]
  • 93.Shibata R., Ouchi N., Ohashi K., Murohara T. The role of adipokines in cardiovascular disease. J Cardiol. 2017;70(4):329–334. doi: 10.1016/j.jjcc.2017.02.006. [DOI] [PubMed] [Google Scholar]
  • 94.Yang R.-Z., Lee M.-J., Hu H., et al. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 2006;290(6):E1253–E1261. doi: 10.1152/ajpendo.00572.2004. [DOI] [PubMed] [Google Scholar]
  • 95.Buyukinan M., Atar M., Can U., Pirgon O., Guzelant A., Deniz I. The association between serum vaspin and omentin-1 levels in obese children with metabolic syndrome. Metab Syndr Relat Disord. 2018;16(2):76–81. doi: 10.1089/met.2017.0133. [DOI] [PubMed] [Google Scholar]
  • 96.Zhang M., Tan X., Yin C., Wang L., Tie Y., Xiao Y. Serum levels of omentin-1 are increased after weight loss and are particularly associated with increases in obese children with metabolic syndrome. Acta Paediatr. 2017;106(11):1851–1856. doi: 10.1111/apa.14026. [DOI] [PubMed] [Google Scholar]
  • 97.Sitticharoon C., Nway N.C., Chatree S., Churintaraphan M., Boonpuan P., Maikaew P. Interactions between adiponectin, visfatin, and omentin in subcutaneous and visceral adipose tissues and serum, and correlations with clinical and peripheral metabolic factors. Peptides. 2014;62:164–175. doi: 10.1016/j.peptides.2014.10.006. [DOI] [PubMed] [Google Scholar]
  • 98.Pan X., Kaminga A.C., Wen S.W., Acheampong K., Liu A. Omentin-1 in diabetes mellitus: a systematic review and meta-analysis. PLoS One. 2019;14(12) doi: 10.1371/journal.pone.0226292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Castro CAd, Silva KAd, Rocha M.C., et al. Exercise and omentin: their role in the crosstalk between muscle and adipose tissues in type 2 diabetes mellitus rat models. Front Physiol. 2019;9:1881. doi: 10.3389/fphys.2018.01881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Brunetti L., Leone S., Orlando G., et al. Hypotensive effects of omentin-1 related to increased adiponectin and decreased interleukin-6 in intra-thoracic pericardial adipose tissue. Pharmacol Rep. 2014;66(6):991–995. doi: 10.1016/j.pharep.2014.06.014. [DOI] [PubMed] [Google Scholar]
  • 101.Stejskal D., Vaclavik J., Smekal A., Svobodova G., Richterova R., Svestak M. Omentin-1 levels in patients with premature coronary artery disease, metabolic syndrome and healthy controls. Short communication. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2016;160(2):219–221. doi: 10.5507/bp.2016.019. [DOI] [PubMed] [Google Scholar]
  • 102.Liu F., Fang S., Liu X., et al. Omentin-1 protects against high glucose-induced endothelial dysfunction via the AMPK/PPARδ signaling pathway. Biochem Pharmacol. 2020;174 doi: 10.1016/j.bcp.2020.113830. [DOI] [PubMed] [Google Scholar]
  • 103.Watanabe K., Watanabe R., Konii H., et al. Counteractive effects of omentin-1 against atherogenesis. Cardiovasc Res. 2016;110(1):118–128. doi: 10.1093/cvr/cvw016. [DOI] [PubMed] [Google Scholar]
  • 104.Hiramatsu-Ito M., Shibata R., Ohashi K., et al. Omentin attenuates atherosclerotic lesion formation in apolipoprotein E-deficient mice. Cardiovasc Res. 2016;110(1):107–117. doi: 10.1093/cvr/cvv282. [DOI] [PubMed] [Google Scholar]
  • 105.Zhou Y., Hao C., Li C., et al. Omentin-1 protects against bleomycin-induced acute lung injury. Mol Immunol. 2018;103:96–105. doi: 10.1016/j.molimm.2018.09.007. [DOI] [PubMed] [Google Scholar]
  • 106.Jiang R., Lönnerdal B. Cloning and characterization of the human lactoferrin receptor gene promoter. Biometals. 2018;31(3):357–368. doi: 10.1007/s10534-018-0080-z. [DOI] [PubMed] [Google Scholar]
  • 107.Rashid R., Maqbool M., Jan A., Geer M.I. Role of adipokines and free fatty acids in insulin resistance-a review. Int J Adv Res Sci Eng. 2018;7(4):2115–2123. [Google Scholar]
  • 108.Babaei P., Pourrahim Ghouroghchi A., Damirchi A., Soltani Tehrani B. The interactive effect of aerobic-resistance training and estrogen therapy on metabolic syndrome indices and omentin-1. Physiol Pharmacol. 2015;19(3):200–207. [Google Scholar]
  • 109.Madsen S.M., Thorup A.C., Bjerre M., Jeppesen P.B. Does 8 weeks of strenuous bicycle exercise improve diabetes-related inflammatory cytokines and free fatty acids in type 2 diabetes patients and individuals at high-risk of metabolic syndrome? Arch Physiol Biochem. 2015;121(4):129–138. doi: 10.3109/13813455.2015.1082600. [DOI] [PubMed] [Google Scholar]
  • 110.Urbanová M., Dostálová I., Trachta P., et al. Serum concentrations and subcutaneous adipose tissue mRNA expression of omentin in morbid obesity and type 2 diabetes mellitus: the effect of very-low-calorie diet, physical activity and laparoscopic sleeve gastrectomy. Physiol Res. 2014;63(2):207–218. doi: 10.33549/physiolres.932530. [DOI] [PubMed] [Google Scholar]
  • 111.Faramarzi M., Banitalebi E., Nori S., Farzin S., Taghavian Z. Effects of rhythmic aerobic exercise plus core stability training on serum omentin, chemerin and vaspin levels and insulin resistance of overweight women. J Sports Med Phys Fit. 2016;56(4):476–482. [PubMed] [Google Scholar]
  • 112.Ge H., Huang L., Pourbahrami T., Li C. Generation of soluble leptin receptor by ectodomain shedding of membrane-spanning receptors in vitro and in vivo. J Biol Chem. 2002;277(48):45898–45903. doi: 10.1074/jbc.m205825200. [DOI] [PubMed] [Google Scholar]
  • 113.Yan P., Liu D., Long M., Ren Y., Pang J., Li R. Changes of serum omentin levels and relationship between omentin and adiponectin concentrations in type 2 diabetes mellitus. Exp Clin Endocrinol Diabetes. 2011;119(4):257–263. doi: 10.1055/s-0030-1269912. [DOI] [PubMed] [Google Scholar]
  • 114.Kataoka Y., Shibata R., Ohashi K., et al. Omentin prevents myocardial ischemic injury through AMP-activated protein kinase-and Akt-dependent mechanisms. J Am Coll Cardiol. 2014;63(24):2722–2733. doi: 10.1016/j.jacc.2014.03.032. [DOI] [PubMed] [Google Scholar]
  • 115.de Souza Batista C.M., Yang R.-Z., Lee M.-J., et al. Omentin plasma levels and gene expression are decreased in obesity. Diabetes. 2007;56(6):1655–1661. doi: 10.2337/db06-1506. [DOI] [PubMed] [Google Scholar]
  • 116.Boucher J., Masri B., Daviaud D., et al. Apelin, a newly identified adipokine up-regulated by insulin and obesity. Endocrinology. 2005;146(4):1764–1771. doi: 10.1210/en.2004-1427. [DOI] [PubMed] [Google Scholar]
  • 117.Kleinz M.J., Davenport A.P. Emerging roles of apelin in biology and medicine. Pharmacol Ther. 2005;107(2):198–211. doi: 10.1016/j.pharmthera.2005.04.001. [DOI] [PubMed] [Google Scholar]
  • 118.Li L., Yang G., Li Q., et al. Changes and relations of circulating visfatin, apelin, and resistin levels in normal, impaired glucose tolerance, and type 2 diabetic subjects. Exp Clin Endocrinol Diabetes. 2006;114(10):544–548. doi: 10.1055/s-2006-948309. [DOI] [PubMed] [Google Scholar]
  • 119.Babaei P., Dastras A., Tehrani B.S., Pourali Roudbaneh S. The effect of estrogen replacement therapy on visceral fat, serum glucose, lipid profiles and apelin level in ovariectomized rats. J Menopausal Med. 2017;23(3):182–189. doi: 10.6118/jmm.2017.23.3.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Choi Y.S., Yang H.I., Cho S., et al. Serum asymmetric dimethylarginine, apelin, and tumor necrosis factor-α levels in non-obese women with polycystic ovary syndrome. Steroids. 2012;77(13):1352–1358. doi: 10.1016/j.steroids.2012.08.005. [DOI] [PubMed] [Google Scholar]
  • 121.Zhu S., Sun F., Li W., et al. Apelin stimulates glucose uptake through the PI3K/Akt pathway and improves insulin resistance in 3T3-L1 adipocytes. Mol Cell Biochem. 2011;353(1-2):305–313. doi: 10.1007/s11010-011-0799-0. [DOI] [PubMed] [Google Scholar]
  • 122.Dray C., Knauf C., Daviaud D., et al. Apelin stimulates glucose utilization in normal and obese insulin-resistant mice. Cell Metabol. 2008;8(5):437–445. doi: 10.1016/j.cmet.2008.10.003. [DOI] [PubMed] [Google Scholar]
  • 123.Castan-Laurell I., Masri B., Valet P. The apelin/APJ system as a therapeutic target in metabolic diseases. Expert Opin Ther Targets. 2019;23(3):215–225. doi: 10.1080/14728222.2019.1561871. [DOI] [PubMed] [Google Scholar]
  • 124.Bertrand C., Valet P., Castan-Laurell I. Apelin and energy metabolism. Front Physiol. 2015;6:115. doi: 10.3389/fphys.2015.00115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Castan-Laurell I., Dray C., Attané C., Duparc T., Knauf C., Valet P. Apelin, diabetes, and obesity. Endocrine. 2011;40(1):1–9. doi: 10.1007/s12020-011-9507-9. [DOI] [PubMed] [Google Scholar]
  • 126.Maguire J.J., Kleinz M.J., Pitkin S.L., Davenport A.P. [Pyr1] apelin-13 identified as the predominant apelin isoform in the human heart: vasoactive mechanisms and inotropic action in disease. Hypertension. 2009;54(3):598–604. doi: 10.1161/hypertensionaha.109.134619. [DOI] [PubMed] [Google Scholar]
  • 127.Yu X.-H., Tang Z.-B., Liu L.-J., et al. Apelin and its receptor APJ in cardiovascular diseases. Clin Chim Acta. 2014;428:1–8. doi: 10.1016/j.cca.2013.09.001. [DOI] [PubMed] [Google Scholar]
  • 128.Masri B., Morin N., Pedebernade L., Knibiehler B., Audigier Y. The apelin receptor is coupled to Gi1 or Gi2 protein and is differentially desensitized by apelin fragments. J Biol Chem. 2006;281(27):18317–18326. doi: 10.1074/jbc.m600606200. [DOI] [PubMed] [Google Scholar]
  • 129.Besse-Patin A., Montastier E., Vinel C., et al. Effect of endurance training on skeletal muscle myokine expression in obese men: identification of apelin as a novel myokine. Int J Obes. 2014;38(5):707–713. doi: 10.1038/ijo.2013.158. [DOI] [PubMed] [Google Scholar]
  • 130.Kadoglou N.P., Vrabas I.S., Kapelouzou A., et al. The impact of aerobic exercise training on novel adipokines, apelin and ghrelin, in patients with type 2 diabetes. Med Sci Mon Int Med J Exp Clin Res. 2012;18(5):CR290–CR295. doi: 10.12659/msm.882734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Jang S.-H., Paik I.-Y., Ryu J.-H., Lee T.-H., Kim D.-E. Effects of aerobic and resistance exercises on circulating apelin-12 and apelin-36 concentrations in obese middle-aged women: a randomized controlled trial. BMC Wom Health. 2019;19(1):23. doi: 10.1186/s12905-019-0722-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Nikseresht M., Rajabi H., Nikseresht A. The effects of nonlinear resistance and aerobic interval training on serum levels of apelin and insulin resistance in middle-aged obese men. Tehran Univ Med J. 2015;73(5):375–383. [Google Scholar]
  • 133.Ghanbari-Niaki A., Saeidi A., Gharahcholo L., et al. Influence of resistance training and herbal supplementation on plasma apelin and metabolic syndrome risk factors in postmenopausal women. Sci Sports. 2020;35(2):109.e1–109.e5. doi: 10.1016/j.scispo.2019.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Green J.S., Lowe R.C., Pronk N., Jacobsen D., Rohack J.J., Crouse S.F. Low and high intensity endurance exercise training does not significantly alter the apolipoprotein-b/apoliporotienal ratio in hypercholesterolemic men. Med Sci Sports Exerc. 2005;37:S470. doi: 10.1097/00005768-200505001-02459. [DOI] [Google Scholar]
  • 135.Chang H.M., Lee H.J., Park H.S., et al. Effects of weight reduction on serum vaspin concentrations in obese subjects: modification by insulin resistance. Obesity. 2010;18(11):2105–2110. doi: 10.1038/oby.2010.60. [DOI] [PubMed] [Google Scholar]
  • 136.Klöting N., Kovacs P., Kern M., et al. Central vaspin administration acutely reduces food intake and has sustained blood glucose-lowering effects. Diabetologia. 2011;54(7):1819–1823. doi: 10.1007/s00125-011-2137-1. [DOI] [PubMed] [Google Scholar]
  • 137.Klöting N., Berndt J., Kralisch S., et al. Vaspin gene expression in human adipose tissue: association with obesity and type 2 diabetes. Biochem Biophys Res Commun. 2006;339(1):430–436. doi: 10.1016/j.bbrc.2005.11.039. [DOI] [PubMed] [Google Scholar]
  • 138.Mansour S.W., Tawfiq M.S., Khalefa A.A., Hadhoud S.E., El-Shorbgy E.A.A. Effect of diet regimen on serum vaspin level in obese diabetic female patients. Zagazig University Medical Journal. 2019;25(5):699–707. doi: 10.21608/zumj.2019.10713.11170. [DOI] [Google Scholar]
  • 139.El-Lebedy D.H., Ibrahim A.A., Ashmawy I.O. Novel adipokines vaspin and irisin as risk biomarkers for cardiovascular diseases in type 2 diabetes mellitus. Diabetes Metab Syndr. 2018;12(5):643–648. doi: 10.1016/j.dsx.2018.04.025. [DOI] [PubMed] [Google Scholar]
  • 140.Heiker J.T., Klöting N., Kovacs P., et al. Vaspin inhibits kallikrein 7 by serpin mechanism. Cell Mol Life Sci. 2013;70(14):2569–2583. doi: 10.1007/s00018-013-1258-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Jung C.H., Lee W.J., Hwang J.Y., et al. Vaspin protects vascular endothelial cells against free fatty acid-induced apoptosis through a phosphatidylinositol 3-kinase/Akt pathway. Biochem Biophys Res Commun. 2011;413(2):264–269. doi: 10.1016/j.bbrc.2011.08.083. [DOI] [PubMed] [Google Scholar]
  • 142.Zieger K., Weiner J., Krause K., et al. Vaspin suppresses cytokine-induced inflammation in 3T3-L1 adipocytes via inhibition of NFκB pathway. Mol Cell Endocrinol. 2018;460:181–188. doi: 10.1016/j.mce.2017.07.022. [DOI] [PubMed] [Google Scholar]
  • 143.Phalitakul S., Okada M., Hara Y., Yamawaki H. Vaspin prevents TNF-α-induced intracellular adhesion molecule-1 via inhibiting reactive oxygen species-dependent NF-κB and PKCθ activation in cultured rat vascular smooth muscle cells. Pharmacol Res. 2011;64(5):493–500. doi: 10.1016/j.phrs.2011.06.001. [DOI] [PubMed] [Google Scholar]
  • 144.Liu S., Dong Y., Wang T., et al. Vaspin inhibited proinflammatory cytokine induced activation of nuclear factor-kappa B and its downstream molecules in human endothelial EA. hy926 cells. Diabetes Res Clin Pract. 2014;103(3):482–488. doi: 10.1016/j.diabres.2013.12.002. [DOI] [PubMed] [Google Scholar]
  • 145.Tantiwong P., Shanmugasundaram K., Monroy A., et al. NF-κB activity in muscle from obese and type 2 diabetic subjects under basal and exercise-stimulated conditions. Am J Physiol Endocrinol Metab. 2010;299(5):E794–E801. doi: 10.1152/ajpendo.00776.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Choe S.S., Huh J.Y., Hwang I.J., Kim J.I., Kim J.B. Adipose tissue remodeling: its role in energy metabolism and metabolic disorders. Front Endocrinol. 2016;7:30. doi: 10.3389/fendo.2016.00030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Fabre O., Ingerslev L.R., Garde C., Donkin I., Simar D., Barres R. Exercise training alters the genomic response to acute exercise in human adipose tissue. Epigenomics. 2018;10(8):1033–1050. doi: 10.2217/epi-2018-0039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Youn B.-S., Klöting N., Kratzsch J., et al. Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes. 2008;57(2):372–377. doi: 10.2337/db07-1045. [DOI] [PubMed] [Google Scholar]
  • 149.Shahdadi A., Molaei K. The effect of 8 Weeks rhythmic aerobic exercise on vaspin levels and lipid profile in overweight and obese women. Mediterr J Soc Sci. 2016;7(4):163–168. doi: 10.5901/mjss.2016.v7n4s2p163. [DOI] [Google Scholar]
  • 150.Amouzad Mahdirejei H., Fadaei Reyhan Abadei S., Abbaspour Seidi A., et al. Effects of an eight-week resistance training on plasma vaspin concentrations, metabolic parameters levels and physical fitness in patients with type 2 diabetes. Cell J. 2014;16(3):367–374. [PMC free article] [PubMed] [Google Scholar]
  • 151.Oberbach A., Kirsch K., Lehmann S., et al. Serum vaspin concentrations are decreased after exercise-induced oxidative stress. Obes Facts. 2010;3(5):328–331. doi: 10.1159/000321637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Briken S., Rosenkranz S.C., Keminer O., et al. Effects of exercise on Irisin, BDNF and IL-6 serum levels in patients with progressive multiple sclerosis. J Neuroimmunol. 2016;299:53–58. doi: 10.1016/j.jneuroim.2016.08.007. [DOI] [PubMed] [Google Scholar]
  • 153.Keller C., Steensberg A., Hansen A.K., Fischer C.P., Plomgaard P., Pedersen B.K. Effect of exercise, training, and glycogen availability on IL-6 receptor expression in human skeletal muscle. J Appl Physiol (1985) 2005;99(6):2075–2079. doi: 10.1152/japplphysiol.00590.2005. [DOI] [PubMed] [Google Scholar]
  • 154.Nieman D.C., Zwetsloot K.A., Lomiwes D.D., Meaney M.P., Hurst R.D. Muscle glycogen depletion following 75-km of cycling is not linked to increased muscle IL-6, IL-8, and MCP-1 mRNA expression and protein content. Front Physiol. 2016;7:431. doi: 10.3389/fphys.2016.00431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Samarghandian S., Azimi-Nezhad M., Farkhondeh T. Crocin attenuate Tumor Necrosis Factor-alpha (TNF-α) and interleukin-6 (IL-6) in streptozotocin-induced diabetic rat aorta. Cytokine. 2016;88:20–28. doi: 10.1016/j.cyto.2016.08.002. [DOI] [PubMed] [Google Scholar]
  • 156.da Costa R.M., Neves K.B., Mestriner F.L., Louzada-Junior P., Bruder-Nascimento T., Tostes R.C. TNF-α induces vascular insulin resistance via positive modulation of PTEN and decreased Akt/eNOS/NO signaling in high fat diet-fed mice. Cardiovasc Diabetol. 2016;15(1):119. doi: 10.1186/s12933-016-0443-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Dunmore S.J., Brown J. The role of adipokines in b-cell failure of type 2 diabetes. J Endocrinol. 2013;216(1):T37–T45. doi: 10.1530/joe-12-0278. [DOI] [PubMed] [Google Scholar]
  • 158.Hagstrom A.D., Marshall P.W., Lonsdale C., et al. The effect of resistance training on markers of immune function and inflammation in previously sedentary women recovering from breast cancer: a randomized controlled trial. Breast Cancer Res Treat. 2016;155(3):471–482. doi: 10.1007/s10549-016-3688-0. [DOI] [PubMed] [Google Scholar]
  • 159.Sardeli A.V., Tomeleri C.M., Cyrino E.S., Fernhall B., Cavaglieri C.R., Chacon-Mikahil M.P.T. Effect of resistance training on inflammatory markers of older adults: a meta-analysis. Exp Gerontol. 2018;111:188–196. doi: 10.1016/j.exger.2018.07.021. [DOI] [PubMed] [Google Scholar]
  • 160.Fayaz E., Damirchi A., Zebardast N., Babaei P. Cinnamon extract combined with high-intensity endurance training alleviates metabolic syndrome via non-canonical WNT signaling. Nutrition. 2019;65:173–178. doi: 10.1016/j.nut.2019.03.009. [DOI] [PubMed] [Google Scholar]
  • 161.Kouhestani S., Zare S., Babaei P. Flavonoids fraction of mespilus germanica alleviates insulin resistance in metabolic syndrome model of ovariectomized rats via reduction in tumor necrosis factor-α. J Menopausal Med. 2018;24(3):169–175. doi: 10.6118/jmm.2018.24.3.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Wang J., Leung K.-S., Chow S.K.-H., Cheung W.-H. Inflammation and age-associated skeletal muscle deterioration (sarcopaenia) J orthopaedic translat. 2017;10:94–101. doi: 10.1016/j.jot.2017.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Diehl A.M. Tumor necrosis factor and its potential role in insulin resistance and nonalcoholic fatty liver disease. Clin Liver Dis. 2004;8(3):619–x. doi: 10.1016/j.cld.2004.04.012. [DOI] [PubMed] [Google Scholar]
  • 164.Joshi-Barve S., Barve S.S., Butt W., Klein J., McClain C.J. Inhibition of proteasome function leads to NF-κB-independent IL-8 expression in human hepatocytes. Hepatology. 2003;38(5):1178–1187. doi: 10.1053/jhep.2003.50470. [DOI] [PubMed] [Google Scholar]
  • 165.Vozarova B., Weyer C., Hanson K., Tataranni P.A., Bogardus C., Pratley R.E. Circulating interleukin-6 in relation to adiposity, insulin action, and insulin secretion. Obes Res. 2001;9(7):414–417. doi: 10.1038/oby.2001.54. [DOI] [PubMed] [Google Scholar]
  • 166.Oberhauser F., Schulte D., Faust M., et al. Weight loss due to a very low calorie diet differentially affects insulin sensitivity and interleukin-6 serum levels in nondiabetic obese human subjects. Horm Metab Res. 2012;44(6):465–470. doi: 10.1055/s-0032-1306341. [DOI] [PubMed] [Google Scholar]
  • 167.Akash M.S.H., Rehman K., Liaqat A. Tumor necrosis factor-alpha: role in development of insulin resistance and pathogenesis of type 2 diabetes mellitus. J Cell Biochem. 2018;119(1):105–110. doi: 10.1002/jcb.26174. [DOI] [PubMed] [Google Scholar]
  • 168.Zand H., Morshedzadeh N., Naghashian F. Signaling pathways linking inflammation to insulin resistance. Diabetes Metab Syndr. 2017;11(Suppl 1):S307–S309. doi: 10.1016/j.dsx.2017.03.006. [DOI] [PubMed] [Google Scholar]
  • 169.Bode J.G., Schweigart J., Kehrmann J., et al. TNF-α induces tyrosine phosphorylation and recruitment of the Src homology protein-tyrosine phosphatase 2 to the gp130 signal-transducing subunit of the IL-6 receptor complex. J Immunol. 2003;171(1):257–266. doi: 10.4049/jimmunol.171.1.257. [DOI] [PubMed] [Google Scholar]
  • 170.Rotter V., Nagaev I., Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-α, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem. 2003;278(46):45777–45784. doi: 10.1074/jbc.m301977200. [DOI] [PubMed] [Google Scholar]
  • 171.Grosicki G.J., Barrett B., Englund D., et al. Circulating interleukin-6 is associated with skeletal muscle strength, quality, and functional adaptation with exercise training in mobility-limited older adults. J Frailty Aging. 2020;9(1):57–63. doi: 10.14283/jfa.2019.30. [DOI] [PubMed] [Google Scholar]
  • 172.Kurauti M.A., Costa-Júnior J.M., Ferreira S.M., et al. Interleukin-6 increases the expression and activity of insulin-degrading enzyme. Sci Rep. 2017;7 doi: 10.1038/srep46750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Alipourfard I., Datukishvili N., Mikeladze D. TNF-α downregulation modifies Insulin Receptor Substrate 1 (IRS-1) in metabolic signaling of diabetic insulin-resistant hepatocytes. Mediat Inflamm. 2019 doi: 10.1155/2019/3560819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Kränkel N., Bahls M., Van Craenenbroeck E.M., et al. Exercise training to reduce cardiovascular risk in patients with metabolic syndrome and type 2 diabetes mellitus: how does it work? Eur J Prev Cardiol. 2019;26(7):701–708. doi: 10.1177/2047487318805158. [DOI] [PubMed] [Google Scholar]
  • 175.Martínez P.Y.O., López J.A.H., Diaz D.P., Trujillo D.A.Z., Teixeira A.M. Effects of three months of water-based exercise training on metabolic syndrome components in older women. Retos: nuevas tendencias en educación física, deporte y recreación. 2019;35:181–184. doi: 10.47197/retos.v0i35.62041. [DOI] [Google Scholar]
  • 176.Ho T., Zhao X., Courville A., et al. Effects of a 12-month moderate weight loss intervention on insulin sensitivity and inflammation status in nondiabetic overweight and obese subjects. Horm Metab Res. 2015;47(4):289–296. doi: 10.1055/s-0034-1382011. [DOI] [PubMed] [Google Scholar]
  • 177.Scott J.P., Sale C., Greeves J.P., Casey A., Dutton J., Fraser W.D. Effect of exercise intensity on the cytokine response to an acute bout of running. Med Sci Sports Exerc. 2011;43(12):2297–2306. doi: 10.1249/mss.0b013e31822113a9. [DOI] [PubMed] [Google Scholar]
  • 178.Kohut M., McCann D., Russell D., et al. Aerobic exercise, but not flexibility/resistance exercise, reduces serum IL-18, CRP, and IL-6 independent of β-blockers, BMI, and psychosocial factors in older adults. Brain Behav Immun. 2006;20(3):201–209. doi: 10.1016/j.bbi.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 179.Allen N.G., Higham S.M., Mendham A.E., Kastelein T.E., Larsen P.S., Duffield R. The effect of high-intensity aerobic interval training on markers of systemic inflammation in sedentary populations. Eur J Appl Physiol. 2017;117(6):1249–1256. doi: 10.1007/s00421-017-3613-1. [DOI] [PubMed] [Google Scholar]
  • 180.Abd El-Kader S., Gari A., El-Den A.S. Impact of moderate versus mild aerobic exercise training on inflammatory cytokines in obese type 2 diabetic patients: a randomized clinical trial. Afr Health Sci. 2013;13(4):857–863. doi: 10.4314/ahs.v13i4.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gerosa-Neto J., Antunes B.M., Campos E.Z., et al. Impact of long-term high-intensity interval and moderate-intensity continuous training on subclinical inflammation in overweight/obese adults. J Exerc Rehabil. 2016;12(6):575–580. doi: 10.12965/jer.1632770.385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Farinha J.B., Steckling F.M., Stefanello S.T., et al. Response of oxidative stress and inflammatory biomarkers to a 12-week aerobic exercise training in women with metabolic syndrome. Sports Med Open. 2015;1(1):19. doi: 10.1186/s40798-015-0011-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Beavers K.M., Hsu F.-C., Isom S., et al. Long-term physical activity and inflammatory biomarkers in older adults. Med Sci Sports Exerc. 2010;42(12):2189–2196. doi: 10.1249/mss.0b013e3181e3ac80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Cavalcante P.A.M., Gregnani M.F., Henrique J.S., Ornellas F.H., Araújo R.C. Aerobic but not resistance exercise can induce inflammatory pathways via toll-like 2 and 4: a systematic review. Sports Med Open. 2017;3(1):42. doi: 10.1186/s40798-017-0111-2. published correction appears in Sports Med Open. 2018 Jan 31;4(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Monteiro-Junior R.S., de Tarso Maciel-Pinheiro P., Portugal EdMM., et al. Effect of exercise on inflammatory profile of older persons: systematic review and meta-analyses. J Phys Activ Health. 2018;15(1):64–71. doi: 10.1123/jpah.2016-0735. [DOI] [PubMed] [Google Scholar]
  • 186.Kasapis C., Thompson P.D. The effects of physical activity on serum C-reactive protein and inflammatory markers: a systematic review. J Am Coll Cardiol. 2005;45(10):1563–1569. doi: 10.1016/j.jacc.2004.12.077. [DOI] [PubMed] [Google Scholar]
  • 187.Cavalcante P.A.M., Gregnani M.F., Henrique J.S., Ornellas F.H., Araújo R.C. Aerobic but not resistance exercise can induce inflammatory pathways via toll-like 2 and 4: a systematic review. Sports Med Open. 2017;3(1):42. doi: 10.1186/s40798-017-0111-2. published correction appears in Sports Med Open. 2018 Jan 31;4(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Flynn M.G., McFarlin B.K., Phillips M.D., Stewart L.K., Timmerman K.L. Toll-like receptor 4 and CD14 mRNA expression are lower in resistive exercise-trained elderly women. J Appl Physiol (1985) 2003;95(5):1833–1842. doi: 10.1152/japplphysiol.00359.2003. [DOI] [PubMed] [Google Scholar]
  • 189.Kanayama A., Seth R.B., Sun L., et al. TAB2 and TAB3 activate the NF-κB pathway through binding to polyubiquitin chains. Mol Cell. 2004;15(4):535–548. doi: 10.1016/j.molcel.2004.08.008. [DOI] [PubMed] [Google Scholar]
  • 190.Silveira Martins M., Farinha J.B., Basso Benetti C., et al. Positive effects OF resistance training ON inflammatory parameters IN men with metabolic syndrome risk factors. Nutr Hosp. 2015;32(2):792–798. doi: 10.3305/nh.2015.32.2.8696. [DOI] [PubMed] [Google Scholar]
  • 191.Asokan S.M., Wang T., Wang M.-F., Lin W.-T. A novel dipeptide from potato protein hydrolysate augments the effects of exercise training against high-fat diet-induced damages in senescence-accelerated mouse-prone 8 by boosting pAMPK/SIRT1/PGC-1α/pFOXO3 pathway. Aging (N Y) 2020;12(8):7334–7349. doi: 10.18632/aging.103081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Ouchi N., Higuchi A., Ohashi K., et al. Sfrp5 is an anti-inflammatory adipokine that modulates metabolic dysfunction in obesity. Science. 2010;329(5990):454–457. doi: 10.1126/science.1188280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Abou Ziki M.D., Mani A. The interplay of canonical and noncanonical Wnt signaling in metabolic syndrome. Nutr Res. 2019;70:18–25. doi: 10.1016/j.nutres.2018.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Catalán V., Gómez-Ambrosi J., Rodríguez A., et al. Activation of noncanonical Wnt signaling through WNT5A in visceral adipose tissue of obese subjects is related to inflammation. Clin Endocrinol Metabol. 2014;99(8):E1407–E1417. doi: 10.1210/jc.2014-1191. [DOI] [PubMed] [Google Scholar]
  • 195.Zuriaga M.A., Fuster J.J., Farb M.G., et al. Activation of non-canonical WNT signaling in human visceral adipose tissue contributes to local and systemic inflammation. Sci Rep. 2017;7(1):17326. doi: 10.1038/s41598-017-17509-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Hu Z., Deng H., Qu H. Plasma SFRP5 levels are decreased in Chinese subjects with obesity and type 2 diabetes and negatively correlated with parameters of insulin resistance. Diabetes Res Clin Pract. 2013;99(3):391–395. doi: 10.1016/j.diabres.2012.11.026. [DOI] [PubMed] [Google Scholar]
  • 197.Carstensen M., Herder C., Kempf K., et al. Sfrp5 correlates with insulin resistance and oxidative stress. Eur J Clin Invest. 2013;43(4):350–357. doi: 10.1111/eci.12052. [DOI] [PubMed] [Google Scholar]
  • 198.El Asmar Z., Terrand J., Jenty M., et al. Convergent signaling pathways controlled by LRP1 (receptor-related protein 1) cytoplasmic and extracellular domains limit cellular cholesterol accumulation. J Biol Chem. 2016;291(10):5116–5127. doi: 10.1074/jbc.m116.714485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Fujino T., Asaba H., Kang M.-J., et al. Low-density lipoprotein receptor-related protein 5 (LRP5) is essential for normal cholesterol metabolism and glucose-induced insulin secretion. Proc Natl Acad Sci U S A. 2003;100(1):229–234. doi: 10.1073/pnas.0133792100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Makarov S.S. NF-κB as a therapeutic target in chronic inflammation: recent advances. Mol Med Today. 2000;6(11):441–448. doi: 10.1016/s1357-4310(00)01814-1. [DOI] [PubMed] [Google Scholar]
  • 201.Pessin J.E., Kwon H. Adipokines mediate inflammation and insulin resistance. Front Endocrinol. 2013;4:71. doi: 10.3389/fendo.2013.00071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.De A. Wnt/Ca2+ signaling pathway: a brief overview. Acta Biochim Biophys Sin. 2011;43(10):745–756. doi: 10.1093/abbs/gmr079. [DOI] [PubMed] [Google Scholar]
  • 203.Komiya Y., Habas R. Wnt signal transduction pathways. Organogenesis. 2008;4(2):68–75. doi: 10.4161/org.4.2.5851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Baarsma H.A., Königshoff M., Gosens R. The WNT signaling pathway from ligand secretion to gene transcription: molecular mechanisms and pharmacological targets. Pharmacol Ther. 2013;138(1):66–83. doi: 10.1016/j.pharmthera.2013.01.002. [DOI] [PubMed] [Google Scholar]
  • 205.Kohn A.D., Moon R.T. Wnt and calcium signaling: β-catenin-independent pathways. Cell Calcium. 2005;38(3-4):439–446. doi: 10.1016/j.ceca.2005.06.022. [DOI] [PubMed] [Google Scholar]
  • 206.Ackers I., Malgor R. Interrelationship of canonical and non-canonical Wnt signalling pathways in chronic metabolic diseases. Diabetes Vasc Dis Res. 2018;15(1):3–13. doi: 10.1177/1479164117738442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Lu Y.C., Wang C.P., Hsu C.C., et al. Circulating secreted frizzled-related protein 5 (Sfrp5) and wingless-type MMTV integration site family member 5a (Wnt5a) levels in patients with type 2 diabetes mellitus. Diabetes Metab Res Rev. 2013;29(7):551–556. doi: 10.1002/dmrr.2426. [DOI] [PubMed] [Google Scholar]
  • 208.Gustafson B., Hammarstedt A., Hedjazifar S., Smith U. Restricted adipogenesis in hypertrophic obesity: the role of WISP2, WNT, and BMP4. Diabetes. 2013;62(9):2997–3004. doi: 10.2337/db13-0473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Laudes M. Role of WNT signalling in the determination of human mesenchymal stem cells into preadipocytes. J Mol Endocrinol. 2011;46(2):R65–R72. doi: 10.1530/jme-10-0169. [DOI] [PubMed] [Google Scholar]
  • 210.Weir G.C. Glucolipotoxicity, β-cells, and diabetes: the emperor has no clothes. Diabetes. 2020;69(3):273–278. doi: 10.2337/db19-0138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Fuster J.J., Zuriaga M.A., Ngo D.T.-M., et al. Noncanonical Wnt signaling promotes obesity-induced adipose tissue inflammation and metabolic dysfunction independent of adipose tissue expansion. Diabetes. 2015;64(4):1235–1248. doi: 10.2337/db14-1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Tilg H., Moschen A.R. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 2006;6(10):772–783. doi: 10.1038/nri1937. [DOI] [PubMed] [Google Scholar]
  • 213.Shi Z., Xu M., Chen X., Wang J., Zhao T., Zha D. The regulatory role of SFRP5/WNT5A axis in allergic rhinitis through inhibiting JNK pathway activation and lowering mucin generation in human nasal epithelial cells. Exp Mol Pathol. 2021;118:104591. doi: 10.1016/j.yexmp.2020.104591. [DOI] [PubMed] [Google Scholar]
  • 214.Leal M.L., Lamas L., Aoki M.S., et al. Effect of different resistance-training regimens on the WNT-signaling pathway. Eur J Appl Physiol. 2011;111(10):2535–2545. doi: 10.1007/s00421-011-1874-7. [DOI] [PubMed] [Google Scholar]
  • 215.Wagenmakers A.J., Pedersen B.K. The anti-inflammatory effect of exercise: its role in diabetes and cardiovascular disease control. Essays Biochem. 2006;42:105–117. doi: 10.1042/bse0420105. [DOI] [PubMed] [Google Scholar]
  • 216.Karki S., Ngo D.T., Farb M.G., et al. WNT5A regulates adipose tissue angiogenesis via antiangiogenic VEGF-A165b in obese humans. Am J Physiol Heart Circ Physiol. 2017;313(1):H200–H206. doi: 10.1152/ajpheart.00776.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Mir E., Moazzami M., Bijeh N., Dokht E.H., Rahimi N. Changes in SFRP5, WNT5A, HbA1c, BMI, PBF, and insulin resistance in men with type 2 diabetes after 12 weeks of combined exercise (HIIT and resistance) Int J Diabetes Dev Ctries. 2020;40(2):248–254. doi: 10.1007/s13410-019-00790-7. [DOI] [Google Scholar]
  • 218.Newmire D., Willoughby D.S. Wnt and β-catenin signaling and skeletal muscle myogenesis in response to muscle damage and resistance exercise and training. Int J Kinesiol Sports Sci. 2015;3(4):40–49. doi: 10.7575/aiac.ijkss.v.3n.4p.40. [DOI] [Google Scholar]
  • 219.McCubrey J.A., Steelman L., Bertrand F.E., et al. Multifaceted roles of GSK-3 and Wnt/β-catenin in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention. Leukemia. 2014;28(1):15–33. doi: 10.1038/leu.2013.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Aschenbach W.G., Ho R.C., Sakamoto K., et al. Regulation of Dishevelled and β-catenin in rat skeletal muscle: an alternative exercise-induced GSK-3β signaling pathway. Am J Physiol Endocrinol Metab. 2006;291(1):E152–E158. doi: 10.1152/ajpendo.00180.2005. [DOI] [PubMed] [Google Scholar]

Articles from Sports Medicine and Health Science are provided here courtesy of Chengdu Sport University

RESOURCES