Open Access

Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review)

  • Authors:
    • Fani-Niki Varra
    • Michail Varras
    • Viktoria-Konstantina Varra
    • Panagiotis Theodosis-Nobelos
  • View Affiliations

  • Published online on: April 9, 2024     https://doi.org/10.3892/mmr.2024.13219
  • Article Number: 95
  • Copyright: © Varra et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Obesity reaches up to epidemic proportions globally and increases the risk for a wide spectrum of co‑morbidities, including type‑2 diabetes mellitus (T2DM), hypertension, dyslipidemia, cardiovascular diseases, non‑alcoholic fatty liver disease, kidney diseases, respiratory disorders, sleep apnea, musculoskeletal disorders and osteoarthritis, subfertility, psychosocial problems and certain types of cancers. The underlying inflammatory mechanisms interconnecting obesity with metabolic dysfunction are not completely understood. Increased adiposity promotes pro‑inflammatory polarization of macrophages toward the M1 phenotype, in adipose tissue (AT), with subsequent increased production of pro‑inflammatory cytokines and adipokines, inducing therefore an overall, systemic, low‑grade inflammation, which contributes to metabolic syndrome (MetS), insulin resistance (IR) and T2DM. Targeting inflammatory mediators could be alternative therapies to treat obesity, but their safety and efficacy remains to be studied further and confirmed in future clinical trials. The present review highlights the molecular and pathophysiological mechanisms by which the chronic low‑grade inflammation in AT and the production of reactive oxygen species lead to MetS, IR and T2DM. In addition, focus is given on the role of anti‑inflammatory agents, in the resolution of chronic inflammation, through the blockade of chemotactic factors, such as monocytes chemotractant protein‑1, and/or the blockade of pro‑inflammatory mediators, such as IL‑1β, ΤΝF‑α, visfatin, and plasminogen activator inhibitor‑1, and/or the increased synthesis of adipokines, such as adiponectin and apelin, in obesity‑associated metabolic dysfunction. 

Introduction

Adipose tissue (AT) is the main energy store derived from food intake in the form of triglycerides (TGs) and controls lipid mobilization (1,2). Furthermore, AT is an active endocrine organ, since it synthesizes and secretes several hormones, cytokines and other bioactive factors, signaling to other metabolic organs, such as the liver, pancreas and brain modulating systemic metabolism, whilst also maintaining body temperature (1,2). AT contains multiple cell types, including adipocytes, adipocytes progenitors, endothelial cells, macrophages, fibroblasts and leucocytes (13). Adipocytes, also called adipose cells or fat cells, are the predominant cell type in AT. There are three types of adipocytes including white, brown and beige (brite). They differ in structure, location, abundance of mitochondria, thermogenic gene expression and function (2,4). White adipocytes are unilocular with a low number of mitochondria and low oxidative rate (2). The cells have a high capacity of energy storage in the form of TGs. In addition, white adipocytes can prevent ectopic lipid deposition and therefore protect organs such as skeletal muscle and the liver from lipotoxicity (2). Brown adipocytes are specialized cells with multilocular lipid droplets, high numbers of mitochondria and enrichment of uncoupling protein 1 (UCP1), a high oxidative capacity and actively participate in energy consumption via thermogenesis (2). In newborn humans, brown AT plays an important role in thermogenesis mediated by the expression of UCP1 (5). In adult humans, it has been found that the amount of brown AT is inversely associated to body mass index (BMI), especially in older individuals indicating the importance of brown AT in energy metabolism (5). Beige adipocytes are a distinct type of adipocyte with multilocular morphology within white AT (WAT) and extremely low UCP1 expression and are capable of thermogenesis (2). Beige adipocytes exist mainly in subcutaneous white fat, but a small portion in visceral fat can be found as well. Acute cold exposure markedly triggers the recruitment and activation of beige adipocytes (2). Based on its location in the body, WAT can be further divided into two types of specific regional depots, the subcutaneous depots and the visceral depots (6). Subcutaneous fat is located under the skin in areas such as the abdomen, thighs, hips and buttocks, however, visceral fat surrounds the intrathoraci organs, the intraperitoneal organs, such as the greater and lesser omentum, mesentery, mesocolon and peritoneum, and the retroperitoneal organs, such as the pancreas, duodenum, ascending and descending colon and kidneys (7). AT responds to stimulation by extra nutrients via hyperplasia (proliferation) and hypertrophy of adipocytes (8). Excessive calories are efficiently stored in the form of neutral TGs in AT, which results in adipose hypertrophy and subsequent obesity (2,9). When adipocytes cannot uptake the excess of TGs, the body synthesizes new adipocytes (hyperplasia), which creates space for fat storage through the lipogenic pathway (9,10). In circumstances of reduction of food intake or an increase in energy expenditure, TGs from adipocytes are broken down into glycerol and fatty acids through the lipolytic pathway to provide energy. Subsequently, fatty acids and glycerol can be transported with the blood to other organs (11). Then, the lipids infiltrate into multiple ectopic organs such as the skeletal muscle, heart and liver and into the visceral adipose depots, leading to systemic low-grade chronic inflammation (12). Moreover, with progressive adipocyte expansion and obesity, the blood supply to adipocytes may be reduced, leading to hypoxia, adipocyte necrosis and macrophage infiltration into AT (13,14). During this process, AT produces and releases a variety of pro-inflammatory and anti-inflammatory factors as well, including adipokines such as leptin, adiponectin and resistin, as well as cytokines and chemokines, such as TNF-α, IL-6 and monocyte chemoattractant protein (MCP) −1 (11,1416). The biological action of adipokine is mainly mediated by binding to their cell surface receptors on the cell membrane of target cells activating appropriate intracellular signaling pathways (2).

Obesity is defined by the National Institute of Health based on the BMI, calculated as the weight of a patient in kilograms divided by the square of height in meters, with BMI values >30 causing concern (17). Subcutaneous AT depots seem to be negatively associated with cardiovascular risk factors, while higher levels of visceral AT have been highly associated with cardio-metabolic disease (18,19). Unhealthy expansion of adipocytes is associated with abdominal obesity, promotion of the obesity-associated metabolic complications, recruitment of macrophages and other immune cells, promotion of systemic inflammation and accumulation of visceral fat (20,21). Indeed, fat accumulation intra-abdominally in men is associated with higher risk for cardiometabolic diseases, independent of BMI (2224). In addition, abdominal visceral fat is also a strong predictor of mortality in obese women (25). A number of human studies have shown that omental adipocyte size positively associates with insulin resistance (IR) (26,27). Notably, individuals of certain ethnic backgrounds, regardless of the present country of residence and citizenship, show predisposition to central obesity and significant obesity-related medical complications (2830). Indeed, several studies demonstrated that South Asian, Japanese and Chinese obese populations have a greater risk for IR, type 2 diabetes mellitus (T2DM) and cardiovascular diseases (CVDs) than Caucasians (31,32). Depending on the degree and duration of weight gain, obesity can progressively cause and/or exacerbate a wide spectrum of co-morbidities, including T2DM, hypertension, dyslipidemia, CVDs, non-alcoholic fatty liver disease (NAFLD), kidney diseases, respiratory disorders, sleep apnea, musculoskeletal disorders, osteoarthritis, sub-fertility, psychosocial problems and certain types of cancers (33,34) (Table I).

Table I.

Comorbidities caused or aggravated by obesity.

Table I.

Comorbidities caused or aggravated by obesity.

T2DM
Hypertension
Dyslipidemia
NAFLD
Cancer
Reproductive problems
CVDs
Mental disorders
Myosceletal problems-osteoarthritis
Psoriasis
Respiratory problems-sleep apnea
Neurogenerative problems

[i] T2DM, type-2 diabetes mellitus; CVDs, cardiovascular diseases; NAFLD, non-alcoholic fatty liver disease.

Obesity-related inflammation and oxidative stress (OS)

Obesity is associated with chronic low-grade inflammation in AT (35). Obesity-related inflammation is associated with the increased release of chemotactic factors, anti-inflammatory adipokines, pro-inflammatory adipokines and pro-inflammatory cytokines (Fig. 1). Inflammation of the AT in obesity is linked to a shift of the anti-inflammatory M2 macrophages in adipocytes from lean individuals to the pro-inflammatory M1 macrophages (35). In AT of lean individuals, most resident macrophages are the anti-inflammatory M2 macrophages that contribute to insulin sensitivity by secreting anti-inflammatory cytokines, such as IL-10, IL-4, IL-11, IL-13, IL-1 receptor antagonist (IL-1Ra), arginase-1 and transforming growth factor-β and anti-inflammatory adipokines, such as adiponectin and apelin (Fig. 2).

It has become evident that the presence of excessive AT enhances lipogenesis and activates the innate immune system (17,34,3650). Compiling evidence suggests that AT, during the course of excessive fat accumulation, in obese patients, and the expansion of the fat mass, produces several chemotactic factors, such as MCP-1, −2, −3 and −4, RANTES [or chemokine CC motif ligand 5 (CCL5)], eotaxin [chemokine CC motif ligand 11 (CCL11)] and interferon γ-induced protein 10 [chemokine CXC motif ligand (CXCL10)] (17,34,3650). In response to such stimuli, monocytes are recruited from the blood, transmigrate and infiltrate into AT depots, through adhesion processes to endothelial cells, increasing the number of activated pro-inflammatory M1 macrophages. In turn, the growing population of pro-inflammatory M1 macrophages enhances the inflammatory changes and secretes pro-inflammatory cytokines (such as TNF-α, IL-1β, IL-6, IL-8, IL-12 and IL-23), pro-inflammatory adipokines [such as leptin, plasminogen activator inhibitor type 1 (PAI-1), visfatin and resistin] and inducible nitric oxide synthase (iNOS) (17,34,3650). The initiation of a low-grade inflammation in AT of obese individuals contributes to an increase of leptin, visfatin, resistin and PAI-1 and to a decrease of adiponectin (17,34,3650). This status leads to IR in adipocytes, which generates free fatty acids (FFAs) in serum, impairs glucose metabolism and favors hepatic, muscular and AT accumulation of fats and glucose (17,34,3650). These events promote higher mitochondrial and peroxisomal oxidation, which results in the production of free radicals (FRs), OS, mitochondrial DNA injury, depletion of adenosine triphosphate (ATP) and finally, lipotoxicity (17,34,3650). Cellular damage leads to high production of pro-inflammatory cytokines, such as TNF-α, which generates further reactive oxygen species (ROS) in tissues and increases the lipid peroxidation rate. An imbalance between the antioxidant capacity and the production of FR induces OS and promotes a systemic low-grade inflammation (17,34,3650) (Fig. 3).

Chemotactic factor, MCP-1

MCP-1 or CCL2 is a 13-kDa pro-inflammatory chemokine. MCP-1 is a member of the MCP family consisting of at least four members (MCP-1, −2, −3, −4) and it exerts its action by binding to its chemokine receptor, C-C motif chemokine receptor 2 (CCR2), which is a CC motif receptor (51). The CCR2A isoform is expressed by mononuclear cells and vascular smooth muscle cells (VSMCs), while CCR2B is expressed by monocytes and natural killer cells (52). MCP-1 plays a role in the recruitment, migration and infiltration of monocytes, microglia and memory T lymphocytes to sites of infection and injury (5356). MCP-1 is secreted predominately by macrophages and endothelial cells (52). Also, MCP-1 is produced from adipocytes and its expression is higher in visceral AT (VAT) than in subcutaneous AT (STAT). The release of MCP-1 is inhibited by adiponectin (57). There is a close relationship between MCP-1 and the number of resident macrophages in adipocytes (5759). Plasma levels of MCP-1 are markedly elevated in obesity and T2DM (52,58,6063). In obesity, the production of MCP-1 by adipocytes results in recruitment of monocytes and activation of macrophages, which causes AT inflammation (56,64). It has been suggested that obesity-associated inflammation in WAT is the causal factor of systemic IR (65). In addition, serum MCP-1 levels are higher in patients with atherosclerosis and the expression of mRNA MPC-1 is increased in atherosclerotic lesions as well (6668). Also, inhibition of MPC-1 expression or its receptor (CCR2) reduces the extent of atheroma formation in hypercholesterolemic mice (55,66,6971).

Anti-inflammatory adipokines-adiponectin as a biomarker of AT

Adiponectin belongs to adipokines and is a 30-kDa peptide secreted only by AT and in particular in large amounts by adipocytes of white AT (2,7274). It acts via two receptors (ADIPOR1 and ADIPOR2) that elicit AMPK signaling and may be modulated by T-cadherin (75). Adiponectin has been described as a main anti-inflammatory adipokine (76). Its anti-inflammatory actions are partly due to its ability to reduce TNF-α activity, via suppression of adiponectin-induced NFκB (77). Also, adiponectin was shown to directly decrease production of pro-inflammatory cytokines TNF-α and IL-6 by macrophages (78). Additionally, adiponectin inhibits generation of ROS induced by high glucose and oxidized low-density lipoprotein (LDL) via a cAMP/PKA-dependent pathway (48,79,80). High ROS levels in adipocytes suppress adiponectin expression and secretion (81,82). Accordingly, there is an inverse association between human serum adiponectin levels and systemic OS (83). Adiponectin increases insulin sensitivity in multiple tissues via up-regulation in insulin signaling. This insulin-sensitizing effect of adiponectin seems to be mediated by increased fatty acid oxidation through AMP-activated protein kinase and peroxisome proliferator-activating receptor-α (PPARα) activation (84,85). In addition, adiponectin reduces glucose content in tissues, by trasfering cytoplasmic glucose trasporter type 4 (GLUT4) toward the surface of the cytoplasmic membrane (73). Also, adiponectin inhibits gluconeogenesis within the liver (86). Adiponectin exhibits cardiovascular protection by suppressing inflammatory processes occurring in the early phases of atherosclerosis and microangiopathy through inhibition of the adhesion of monocytes to blood vessel endothelial cells, as a result of down-regulated expression of adhesion proteins, decreasing of the transformation of macrophages into foam cells and down-regulating intimal smooth muscle cell proliferation (2,48,77,8791). It also enhances nitrogen oxide (NO) synthesis in endothelial cells and stimulates angiogenesis (73,80,92,93).

Adiponectin synthesis is regulated by insulin and insulin-like growth factor-1, which leads to increased concentration of this adipokine (73,94). However, its synthesis is inhibited by pro-inflammatory cytokines, such as TNF-α and IL-6. This suggests that obesity and IR are important factors contributing to low levels of serum adiponectin (95). Also, low serum levels of adiponectin are found in obese, insulin-resistant individuals with related pathologies including T2DM, dyslipidemia and CVDs (89,93,9699). On the other hand, weight loss increases adiponectin concentrations (73). Hence, elevated levels of adiponectin may lead to the decrease in the risk of T2DM (100).

Apelin as a biomarker of AT

Apelin is a short peptide hormone, which belongs to adipokines and is produced by adipocytes in proportion to the present amount of fat; it plays an important role in energy metabolism and is considered to be linked with obesity and diabetes (39,101,102). Apelin exerts its effects by binding with angiotensin II protein J (APJ) receptor (101,103). Apelin promotes brown adipocyte development through the phosphatidylinositol 3-kinase (PI3K)/Akt and AMPK signaling pathways (102). Also, apelin is able to increase the browning in white adipocytes (102,104). In addition, it has been found that apelin relieves the TNF-α suppression on brown adipogenesis (102). Apelin stimulates glucose uptake, increases insulin sensitivity and regulates lipolysis and fatty acid oxidation (101). It has been found that serum apelin levels are increased in obesity. This may be due either to potential resistance to apelin or to its attempt to delay or reduce tissue IR (105). Nevertheless, current literature suggests that apelin administration protects diabetic and/or obese mice (101) by lowering glucose levels and hence, it may have a therapeutic role against obesity and related metabolic diseases (104). Apelin with its activity on enthothelial APJ receptors may additionally improve nitric oxide (NO) release and endothelium-dependent vasodilation (39,106). Apelin has antioxidant effects because it suppresses ROS production and release in AT and improves the antioxidant state in OS-related conditions (107). Apelin promotes the synthesis of antioxidant enzymes via Ras/Raf/mitogen-activated protein kinase (MAPK)/ERK and AMP-activated protein kinase (AMPK) pathways (104), suppresses the expression of pro-oxidant enzymes via the same pathway, and increases mitochondrial oxidative capacity (104,108111).

Pro-inflammatory adipokines and their relationship with obesity
Leptin

Leptin belongs to adipokines and is a 16-KDa peptide, encoded by the obese (ob) gene. It is mainly secreted by WAT and its secretion increases according to the volume of AT and to the TGs stored in adipocytes (2,48,112116). When body energy stores are adequate, leptin suppresses food intake, regulating appetite, energy balance and causing satiety (2,48,112114). The sensation of satiety of leptin is achieved by crossing the blood-brain barrier and targeting the hypothalamus (2,112,117119). Hypothalamic leptin signaling is mediated by leptin receptor and downstream processes, including JAK2/STAT3 pathway (120). However, obesity is associated with increased leptin protein and mRNA levels compared with lean controls. The failure of the elevated leptin levels to correct the metabolic complications seen in obesity, is mainly related to leptin resistance, in tissues with decreased sensitivity to leptin (3,35,39,121123). The influence of leptin on IR is not yet fully understood (35). However, it has been found that IR is associated with elevated serum leptin levels (35). A possible explanation is that leptin resistance causes reduction of lipid oxidation, which leads to lipid accumulation and IR (124,125). Also, leptin in obese humans causes an increase in blood pressure, through sympathetic activation at vascular and/or renal levels (126). Leptin has pro-inflammatory actions, which are related to structural and functional similarities with the cytokine IL-6 (115). Also, leptin promotes OS and endothelial cell dysfunction and activation, increases phagocytic activity by macrophages and induces the production of pro-inflammatory cytokines such as TNF-α, IL-6, IL-2 and IL-1 (2,127,128). In addition, it has been found that leptin administration increases c-reactive protein (CRP) levels, which confirms further its inflammatory effects (123).

Visfatin

Visfatin also known as nicotinamide phosphoribosyltranferase and pre-B-cell colony enhancing factor (129,130) is a 52 kDa adipokine, which is predominantly expressed in human VAT (131), an area of fat tissue, whose accumulation is strongly associated with an enhanced cardiovascular (CV) risk (48,129). Visfatin is also secreted by macrophages, bone marrow, skeletal muscles and various organs including the liver, lungs, brain, heart and pancreas (49,129). However, a specific receptor for visfatin has not been identified yet (93). Several authors have suggested that visfatin levels increase with obesity, T2DM, MetS or CVDs (132134). However, other studies have shown conflicting results regarding the relationship of visfatin with MetS (135,136). It has been found that weight loss decreases visfatin levels in obese patients (134). Moreover, leucocytes from obese patients produce higher amounts of visfatin compared with lean patients (137). Visfatin levels beyond a threshold appear to be associated with IR and obesity-related vascular disorders (130,138). Specifically, visfatin appears to contribute to the release of pro-inflammatory cytokines IL-1β, IL-6, IL-8 and TNF-α, through a regulation of the JAK2/STAT3 and IKN/NF-kB signaling pathways, promoting inflammation (129,131,139144). Moreover, in experimental studies, it has been found that visfatin induces endothelial dysfunction, via the NF-κB pathway, in the vascular endothelium and promotes the proliferation of human VSMCs (129,138). Additionally, visfatin induces NF-κB pathway dependent OS, and blockade of this pathway, via selective IκΒ Kinase (IKK-2) inhibition, leads to a partial reduction in OS, which it is independent of the MAPK/ERK signaling pathway (138,145).

Resistin

Resistin is a 12.5 kDa adipokine, which in human AT is secreted predominantly in macrophages (93). Resistin is also known as adipocyte-secreted factor or Found in Inflammatory Zone 3 (93). The resistin receptor remains unknown, but resistin binding to the Toll-like receptor 4, adenylyl cyclase-associated protein 1 receptor and G protein-coupled receptors was proposed to mediate resistin inflammatory responses in human cells (93,146,147). Resistin has also been associated with the inflammatory response, by promoting activation of the pro-inflammatory cytokines IL-6, IL-1β and TNF-α (16,131,148150). Moroeover, resistin upregulates several adhesion molecules, through NF-κB, in vascular endothelial cells (148,151). In animal models, resistin promotes IR, but in humans there are conflicting reports about the potency of resistin in metabolic diseases (93,152,153). Several studies indicated that increased serum resistin levels are associated with increased obesity, visceral fat, IR and T2DM (154157), while other studies failed to reach to such conclusions (158,159). Also, resistin generates OS, which activates MAPK signaling and inhibits endothelial nitric oxide synthase (eNOS) gene expression (160). Moreover, resistin reduces NO production, by inducing the proliferation of VSMCs, and causes endothelial dysfunction (160). In turn, reduction of NO availability results in impaired vasodilation, increased vascular permeability, endothelial cell adhesion and damage leading to CVDs (160,161).

Plasminogen activator inhibitor-1 (PAI-1)

PAI-1, also known as SERPINE1 is a physiological inhibitor of tissue plasminogen activators (tPAs) (162). Increased PAI-1 activity is associated with reduced fibrinolytic activity and thus, increased risk for thrombus formation and CVDs (163). PAI-1 is synthesized in AT, especially in visceral fat, as well as in preadipocytes, fibroblasts, vascular endothelial cells and in immune cells (2,163165). Increased PAI-1 plasma levels have been found in obese patients and reduced levels were achieved with weight loss (129,166). In experimental studies, using obese mouse models with MetS, it has been found that the deletion of PAI-1 inhibited carotid artery atherosclerosis (167), while pharmacological PAI-1 inhibition attenuated atherosclerosis by inhibiting macrophage accumulation and eliminating senescent cells in the atherosclerotic plaques (168). In human studies, PAI-1 was found to be associated with IR, MetS and atherosclerosis in obesity (163,169). Also, in animal studies using mice fed with a high-fat diet, a deficiency of PAI-1 led to a decrease of body weight gain and improvement of IR (170). In addition, adipocyte hypertrophy in obesity may create local hypoxic areas, which activates hypoxia-inducible factor-1α (HIF-1α). The increase in HIF-1α causes an increased expression of several pro-inflammatory cytokines, such as TNF-α, IL-6, IL-1β and ROS, leading to higher PAI-1 expression in adipocytes (171).

Pro-inflammatory cytokines and their relationship with obesity
TNF-α

TNF-α is a 26-kDa pro-inflammatory cytokine produced by macrophages, adipocytes and vascular endothelial cells, in response to chronic inflammatory activity (37,85,172,173). TNF-α exists in two forms, membrane-bound (mTNF-α) and free soluble (fTNF-α). TNF-α is synthesized as a transmembrane monomer, which afterwards can be cleaved by TNF-α converting enzyme, to yield the 17-kDa soluble form (52). Both forms exist as trimers that have biological activity. TNF-α has two distinct TNF-α receptors, TNF-R1 and TNF-R2, that are similar in their extracellular ligand-binding domains but differ markedly in their intracellular signaling domains (52,85). The majority of signaling in AT is downstream of the TNFR1 (52). TNF-α is a pro-inflammatory cytokine characterized by various biological effects including metabolic, inflammatory, proliferative and necrotic (173). TNF-α directly impairs peripheral glucose uptake, by increasing serine phosphorylation of insulin receptor substrate 1 (IRS-1), inhibits GLUT 4 translocation to the plasma membrane and results in peripheral IR (173,174). TNF-α also potentially increases lipolysis in human adipocytes by regulating hormone-sensitive lipase in adipocytes, resulting in increased circulating FFA levels and peripheral IR in obesity (173). Expression of TNF-α is increased in obesity and IR in humans (37), whilst treatment with TNF-α induces IR in AT (48). Serum TNF-α levels are decreased during weight loss (175). TNF-α is a part of a complex inflammatory network and is capable of initiating cytokine cascade activation that involves both synergistic and inhibitory reactions, which control the synthesis and expression of other cytokines, hormones and their receptors (176). ROS production can also be induced by TNF-α binding to its TNF-R1 receptor promoting NF-κB, ERK, p38MAPK and FADD/pro-caspase-8 signaling pathways (177179). TNF-α causes systemic acute-phase response via the release of other pro-inflammatory cytokines, such as IL-6 and the reduction of anti-inflammatory adiponectins (48). TNF-α also increases the induction of OS and the production of superoxide anions (180).

IL-1β

IL-1β is a 31-kDa pro-inflammatory cytokine, secreted by M1 macrophages (181). It is produced as pro-IL-1β, a pro-inflammatory cytokine, which becomes biologically active by cleavage, requiring the protease caspase-1 (182), which is activated through the NLRP3 inflammasome complex (183). Obesity appears to be directly related to the deregulation of IL-1β expression and the increase in its levels. The increased IL-1β levels contribute to the induction of chronic inflammatory diseases (184). High levels of IL-1β promote ectopic fat accumulation, recruitment of immune cells to AT and liver, fibrosis, IR, T2DM and atheromatous plaque formation in obese patients (181,184). In particular, the increase in IL-1β levels, in STAT, contributes to the suppression of PPARγ expression and the inhibition of the differentiation of preadipocytes to mature adipocytes. However, there is a discrepancy about the effects of IL-1β on lipogenesis and lipolysis (185188). High levels of IL-1β are observed in the VAT of obese individuals (189,190), leading to reduced TG storage in WAT and hepatic steatosis (184). In parallel, high levels of IL-1β in AT are associated with extracellular matrix (ECM) disorganization, diminishing AT hyperplasia (191) and provoking fibrosis and inflammation (192). Elevated IL-1β levels contribute to high immune cell recruitment, promote local inflammation via increase in expression of VCAM-1, ICAM-1, MCP-1 and suppression of PPARα (193). Moreover, the increased recruitment of immune cells is responsible for the hepatic cirrhosis promotion (184). IL-1β contributes to an increase in PAI-1 secretion and promotes fibrosis and chronic liver disease (194). IL-1β appears to lead to activation of the serine/threonine kinase, janus kinase (JAK) 1, which inactivates IRS1 and IRS2 (184), downregulates the expression of PI3K, p85, pAkt and GLUT4, leading to suppression of insulin signal transduction, with a subsequent increase in blood glucose levels (195). Elevated FFAs bind to receptors in the liver, activating the IKK/NFκB pathway and stimulating increased IL-1β expression (196). Respectively, high glucose levels stimulate NADPH oxidase, ROS production and thioredoxin-interacting protein expression, that binds to NIPR3 (196) and activates IL-1β production. As a result, IL-1β binds to the IL-R1 receptor on pancreatic β-cells and leads to increased expression of cytokines and chemokines (197), facilitating the accumulation of hematopoietic cells, such as macrophages, which secrete pro-inflammatory cytokines that further exacerbate the inflammation cascade causing impairment of insulin secretion (184). High IL-1β concentration is further capable of leading to pancreatic β-cell apoptosis by the NF-κB pathway regulation and FAs overexpression (198), or through the depletion of calcium ions (Ca2+), in the c-Jun N-terminal kinase (JNK)-mediated endoplasmic reticulum (ER) with the mediation of JNK (199). Finally, IL-1β enhances the proliferation and migration of VSMCs promoting the atherosclerotic plaque development (184).

IL-6

IL-6 is produced by numerous different cell types, including adipocytes, endothelial cells, pancreatic β-cells, macrophages and monocytes (14,200202). There are two signaling pathways for IL-6 including its classical signaling mechanism involving the binding of IL-6 to its receptor complex (IL6-Ra) that subsequently interacts with an IL-6ST signaling protein (also known as glycoprotein 130, gp130) at the plasma membrane, and the non-classical signaling mechanism which is related to the interaction of the IL-6ST protein with a soluble form of the IL-6-binding receptor (203). Both IL-6 signaling pathways lead to activation of the JAK1-STAT3 pathway (203). Its actions can be beneficial or harmful to the organism, depending on the site of action, the magnitude of production and the duration of the response (204,205). In summary, its beneficial actions are related to its production by skeletal muscles, in response to physical exercise, contributing to lipid metabolism and enhancement of insulin sensitivity in muscles (204,205), as well as appetite suppression (206). Other actions of IL-6 include immune response and hematopoiesis (14,200202). It participates in the regulation of neural differentiation, maturation and function and in energy homeostasis (14,200202). In obesity, serum IL-6 levels are elevated and 10–35% of IL-6 produced is attributed to WAT (207). In obesity, high caloric intake in combination with reduced energy expenditure is directly related to changes in the physiology and morphology of WAT (207). In particular, WAT expansion is observed through an increase in the number or size of adipocytes (208,209). Therefore, in the WAT of obese patients there is an infiltration of immune cells, including T cells and macrophages (19). Both adipocytes, as well as immune cells in WAT, are the main sources of increased circulating IL-6 levels. The VAT secretes a number of substances that further promote IL-6 expression and releases ~2-3 times more IL-6 concentrations than STAT (19). Chronic exposure to high serum IL-6 levels has been associated with an elevated likelihood of impaired glucose tolerance, T2DM, high blood pressure and obesity (48), while is also positively associated with IR in obesity (210,211). Furthermore, exogenous IL-6 administration causes IR in humans, while weight loss results in IL-6 decrease and bariatric surgery improves IR (212215). Thus, if the elevated plasma IL-6 levels in obesity are considered, it could be suggested that chronic low-grade inflammation in obesity links IL-6 as causal factor for IR through the progressive tissue-infiltration by immune cells (216).

Obesity, OS and insulin resistance (IR)

Insulin is an anabolic, peptide hormone, secreted by the pancreatic β-cells of the islets of Langerhans, in response to high blood glucose levels and controls the metabolism of carbohydrates, proteins and fats by stimulating the absorption of glucose from the blood into lipid cells, skeletal muscle cells and the liver, for ATP production or storage as glycogen and TGs (217). More specifically, in fed states, the exogenous glucose uptake increases the circulating glucose levels and stimulates insulin secretion (123,217,218220). Also, other nutrients from food such as FAs and amino acids increase insulin secretion, which stimulates lipogenesis and protein synthesis (218,221). Under fasting conditions, lipolysis is induced from stored TGs in AT and supplies i) glycerol for hepatic glucose production (gluconeogenesis) and ii) FAs for β-oxidation (2,220). In the liver, insulin suppresses hepatic gluconeogenesis (220). Also, insulin reduces the rate of breakdown of glycogen in muscles and liver (glycogenolysis), retaining normal glucose levels (222). Regarding the effect of insulin on lipid metabolism, insulin inhibits lipolysis (antilipolytic action), increases hepatic lipid synthesis for subsequent TGs storage in AT (223) and stimulates glucose uptake into the skeletal muscles, heart and AT (220). In order to exert its effects, insulin binds to its receptor (IF), a tyrosine kinase receptor. A reduction in insulin signaling triggers IR that could affect the metabolic actions of insulin (224). If a decrease of the blood glucose levels is not achieved by insulin, then pancreatic β-cells increase insulin release resulting in hyperinsulinemia, which is the key for IR (225). Therefore, hyperinsulinemia often precedes the development of marked IR and fat mass gain (226).

Insulin signaling is initiated by the phosphorylation/activation of the cytoplasmatic insulin tyrosine kinase receptor that is associated with the activation of two main signaling pathways: i) PI3K/AKT [also known as protein kinase B (PKB)] pathway; and ii) the MAPK pathway (2,227). In healthy subjects, by simultaneously stimulating these distinct pathways (PI3K and MAPK), insulin couples metabolic and hemodynamic homeostasis.

PI3K/AKT2 signaling pathway (also known as protein kinase B or PKB)

In the PI3K-AKT/PKB pathway the binding of insulin to its cell surface receptor activates the lipid kinase, PI3K, binding to its Src homology 2 domain, which activates several phosphatidylinositol-(3,4,5)-triphosphate-dependent serine/threonine kinases, including AKT/PKB (2). Ultimately, these signaling events result in the translocation of the insulin-dependent GLUT4 from its cytoplasmic storage vesicle to the plasma membrane, leading to an increase in glucose uptake (2). The PI3K-AKT signaling pathway regulates the metabolic insulin actions by promoting glucose utilization, protein synthesis and lipogenesis (228).

MAPK pathway

MAPK activation triggers a cascade that regulates the effects of insulin on mitogenesis, growth and differentiation and is not implicated in the metabolic actions of insulin (2). Also, MAPK-dependent insulin signaling pathway controls secretion of the vasoconstrictor, endothelin-1, from endothelium (63).

IR

When a decrease of blood glucose levels is not achieved by insulin, then pancreatic β-cells increase the release of insulin, resulting in hyperinsulinemia, which is the key for IR (224,225). Therefore, hyperinsulinemia often precedes the development of marked IR and fat mass gain (226,229) (Fig. 4). The IR in AT, skeletal muscles and liver is commonly linked with obesity, which is a pathophysiological factor of T2DM (230233). When IR develops in fat tissues, insulin-mediated inhibition of lipolysis is impaired leading to increased lipolysis (123,234) (Fig. 4). The resulting increase in circulating FAs in turn worsen IR, causing alterations in the insulin signaling cascade in different organs, thus creating a vicious cycle (235,236) (Fig. 4). Moreover, in IR, there is a reduced insulin ability to suppress glycogenolysis in hepatocytes and myocytes (234). In IR, FFAs, in muscles, affect IRS-1-associated PI3K activity, leading to decreased GLUT4 translocation to the surface and reduced glucose uptake (235). In parallel, in IR the FFAs act on the liver to promote gluconeogenesis. Therefore, insulin-resistant individuals fail to inhibit hepatic glucose production and hyperglycemia. This results in a hyperinsulinemic state to maintain normal glucose levels (123). However, this compensation eventually fails, leading to decreased insulin levels, which is further exacerbated by the lipotoxic effect of FFAs on pancreatic β-cells (123,236,237). Additionally, in IR, the FFAs act on the liver and promote lipogenesis (Fig. 4). It is essential to note that visceral lipolysis increases the supply of FFAs, directly to the liver, via the splanchnic circulation, thus making visceral fat deposits more important contributors to IR than subcutaneous fat (123). Together with this, there is increased de novo hepatic TG synthesis and a disruption of β-oxidation in hepatic mitochondria (238241). This net leads to increased hepatic very low-density lipoproteins (VLDLs) secretion and hypertriglyceridemia (238241). The hepatic accumulation of TGs and the toxic levels of FFAs results in hepatic lipotoxicity (238241). This mechanism contributes to the production of ROS and the development of NAFLD, which is associated with the development of MetS, which may progress to the more serious non-alcoholic steatohepatitis (NASH) with subsequent hepatic fibrosis, cirrhosis and cancer (225,242). In fact, there is a by-directional relationship between obesity-related IR and NAFLD, since obesity-related IR causes fatty liver and excessive hepatic fat accumulation, promotes IR and weight gain (243). In addition, high concentrations of FFAs increase cholesterol esters and triglyceride (TG) synthesis and subsequently the production of VLDLs rich in TGs (244,245). These in turn, activate cholesterol ester transfer protein, promote TG transfer from VLDL to high-density lipoprotein (HDL), increase HDL clearance and decrease its concentrations (244,245). Moreover, triglyceride-rich LDL, formed after exchange with LDL cholesterol ester, becomes hydrolyzed by lipoprotein lipase or hepatic lipase, leading to cholesterol-depleted small dense LDL particles (244,245). All these alterations in lipoprotein concentrations constitute the hallmark of atherogenic dyslipidemia, caused by IR, in MetS (244,245). Another contribution of IR to MetS is the development of hypertension caused partly by the loss of the vasodilatory effect of insulin and by FFA-induced vasoconstriction due to ROS production and the subsequent scavenging of NO (246). Other mechanisms involve the increased sympathetic stimulation and the renin-induced sodium reabsorption in the kidneys (247). Finally, the contribution of IR, to the promotion of atherogenic processes and the increase of CVD risk, is the development of a higher serum viscosity and a pro-thrombotic state, caused by the increased levels of fibrinogen and PAI-1 (34,165,169,248253). IR and IL-6 produced during the acute phase reaction contribute to elevated fibrinogen concentrations (34,254). Fibrinogen is synthesized by hepatocytes and holds a pivotal role in the coagulation cascade, being a major determinant of plasma viscosity and platelet aggregation, whilst potentially plays a pro-inflammatory role in vascular wall disease (34,254,255). In parallel, PAI-1 is elevated in IR, obesity and MetS (34,256). PAI-1 regulates the endogenous fibrinolytic system and constitutes the main inhibitor of fibrinolysis by binding and inactivating the tPA (34,248,257260). Therefore, elevated PAI-1 levels lead to decreased clearance of clots (34,248,257260). Enhanced AT expression of PAI-1 has been reported in obesity, particularly in VAT (34,261), whilst there is an inverse relationship between PAI-1 activity and adiponectin in overweight and obese women (34,259,260).

Figure 4.

Schematic illustration of the pathways associated with the development of T2DM in obese individuals. The cellular mechanisms involved in the pathogenesis of obesity-associated T2DM include: i) Alterations in the insulin signaling in metabolic tissues, such as liver, adipose tissue and skeletal muscles; ii) pancreatic β-cell dysfunction; and iii) chronic low-grade inflammation and increased oxidative stress. Obesity causes IR and hyperglycemia. Hyperglycemia causes glucotoxicity and an increase in AGEs, resulting in the up-regulation of NF-κΒ, which is a mediator of inflammation and immunity. Increased AGEs affect vascular endothelium and block the activity of NO. The inhibition of NO enhances the oxidative stress. Additionally, glycotoxicity induces the glycolysis and the Krebs cycle resulting in an increased flow of NADPH and FADH2, that act as electron donors to the mitochondrial chain, resulting in an excess of electrons in coenzyme Q and the production of mitochondrial superoxide, resulting in an increase in ROS. Chronic exposure to high intracellular ROS levels in adipocytes ultimately causes mitochondrial dysfunction and perpetuates adipose tissue inflammation, resulting in IR and T2DM. In addition, peroxide-induced OS, causes impairment of the AKT, which results in IR and T2DM. Also, chronic hyperglycemia causes alterations in IR in the liver, adipose tissue and skeletal muscles. Furthermore, chronic hyperglycemia creates chronic low-grade inflammation, which increases OS. The increased FAs cause lipotoxicity and increase the secretion of IL-1β, which is responsible for the deficiency of insulin secretion from pancreatic β-cells resulting in IR and T2DM. Moreover, infiltration of adipocytes by macrophages causes an increase in TNF-α and IL-6. The increase in TNF-α is responsible for tissue inflammation, generation of ROS and IR propagation in peripheral tissues and adipocytes, which is important for the onset of T2DM. In addition, the increase in IL-6 levels contributes to indirect prevention of insulin binding to its receptor, as well as the induction of CRP, which like TNF-α, is associated with IR and T2DM. T2DM, type 2 diabetes mellitus; IR, insulin resistance; AGEs, advanced glycation end products; NO, nitric oxide; FADH2, flavin adenine dinucleotide (reduced form); ROS, reactive oxygen species; FAs, fatty acids; CRP, c-reactive protein.

Obesity, OS and metabolic syndrome (MetS)

MetS is a complex disorder defined by a cluster of clinical and metabolic conditions that occur together and increase the risk for IR, T2DM, dyslipidemia, CVDs, prothrombotic state and stroke (262264). According to the International Diabetes Federation (IDF), metabolic syndrome (MetS) is characterized as the presence of three or more of the following features: i) obesity; ii) hyperglycemia; iii) hypertension; iv) low HDL cholesterol levels; and/or v) hypertriglyceridemia (263). Obesity is the most frequently observed component of MetS (265). It has been established that patients with MetS are five times more likely to develop T2DM and have a 2–3 times higher risk of CVDs (stroke and myocardial infarction), compared with healthy subjects (264,266,267). In addition, MetS has been associated with other clinical conditions, such as hepatic steatosis and NAFLD, hypogonadism, polycystic ovarian syndrome, obstructive sleep apnea, vascular dementia, Alzheimer's disease and carcinomas, especially breast, pancreatic and colorectal cancers (218,268,269). Obesity is the most frequently observed component of MetS (265). The IDF estimates that MetS affects almost a quarter of the adult general population in Western societies (269271). The prevalence of MetS in men does not differ before and after 50 years of age, but women >50 years, show a sharp increase in prevalence (272). It is associated with higher mortality risk in younger adults than in elders (273). Accumulating evidence indicates that dysregulation of the production a wide range of adipocytokines and cytokines, due to excessive accumulation of body fat, participates in the pathogenesis of obesity associated MetS (128,163,169). The link between PAI-1 and MetS with obesity is well established. Increased PAI-1 serum levels are associated with the development of IR, MetS, atherosclerosis and thrombosis in obese patients (128,163,169). Treatment with TNF-α contributes to the development of IR in AT (175). In patients with MetS, chronic exposure to increased IL-6 levels is related to the development of IR by depletion of GLUT4 and disruption of insulin signaling (210,211). Leptin is also involved in the pathophysiology of MetS (274). High plasma levels of leptin are directly associated with IR, MetS and lipid accumulation due to leptin resistance (35,39,124,125). Moreover, visfatin plays a central role in MetS. Elevated visfatin serum levels are associated to IR, T2DM and decreased function of pancreatic β-cells (129,275). Conversely, a protective role of adiponectin against MetS has been reported, since it directly attenuates production of IL-6 and TNF-α, by macrophages, through its ability to suppress NF-κB activation (78,276). In addition, apelin reduces MetS risk. It has been demonstrated that apelin stimulates glucose uptake, increases insulin sensitivity and regulates lipolysis in patients with MetS (101). OS is also critically involved in the pathogenesis of MetS and in the progression of its complications (277280). In patients with MetS there are higher levels of oxidative markers, as well as reduced antioxidant defenses (277). In obesity, the chronic low-grade inflammation, produced by adipocytes exacerbates OS (35) (Fig. 4). Visceral fat accumulation induces an increase in mitochondrial and peroxisomal oxidation of FAs, and the production of ROS (35,281,282). Furthermore, visceral fat accumulation causes over-consumption of oxygen, which generates FRs in the mitochondrial respiratory chain (35,281,282). In addition, a lipid-rich diet can alter oxygen metabolism and generate ROS (35,281,282). Moreover, high ROS production and a decrease in antioxidant capacity leads to a reduction in the bioavailability of vasodilators, particularly NO, and an increase in endothelium-derived contractile factors, favoring atherosclerotic disease (35,281,282) (Fig. 4). With regards to hypertension, elevated OS, in vascular wall leads to vasoconstriction, vascular remodeling, inflammation and fibrosis which results in hypertension and atherosclerosis (268,277,283). Regarding NAFLD, it has been documented that elevated OS appears to be a key mechanism in promoting liver injury and liver inflammation in NAFLD (284). Finally, it has been found that dyslipidemia is associated with higher ROS release and lower eNOS synthesis (277).

Obesity, OS and T2DM

T2DM is a heterogeneous, chronic metabolic disorder, characterized by elevated blood glucose levels with a high prevalence, up to 90%, of all diagnosed diabetic cases in adults (285,286). IR leads to hyperglycemia and over time to T2DM (286). It has been found that the relative risk of T2DM, in adult men and women, increases for a BMI, >24 kg/m2 in men, and 22 kg/m2 in women (34). Women with T2DM are 3–4 times more prone to CVDs compared with 2–3 times in men with T2DM (287). Obesity is an important independent risk factor for IR and T2DM (288291). IR is responsible for the development of hyperglycemia and over time may evolve to T2DM. IR alone is not capable of causing an increase in blood sugar (292), since the pancreas has mechanisms to adapt, by increasing the mass of β-cells and the ability to produce insulin (292,293). Thus, despite reduced peripheral insulin sensitivity, blood sugar levels could retain stable (292). The cellular mechanisms involved in the pathogenesis of obesity-associated T2DM include: i) alterations in the insulin signaling; ii) pancreatic β-cell dysfunction and failure; and iii) chronic low-grade inflammation and increased OS (294). The mechanisms interrelating obesity to the pathogenesis of T2DM are depicted in Fig. 4. Obesity causes generalized IR in AT, liver and skeletal muscles and is associated with increased insulin secretion and chronic hyperinsulinemia, which promotes further weight gain (292,295,296). Therefore, there is a bi-directional relationship between obesity and hyperinsulinemia. Insulin resistant conditions in T2DM could be caused by signaling defects at a number of levels of the insulin-signaling cascade in metabolic tissues, such as liver, AT and skeletal muscles (292,243,297). In addition, in IR and T2DM, the liver fails to suppress glycogenolysis and gluconeogenesis, despite compensatory hyperinsulinemia, and is associated with accelerated glucose synthesis and fasting hyperglycemia (292,243,297). In T2DM, the IR in skeletal muscles is associated with postprandial hyperglycemia, since in these patients, skeletal muscles exhibit decreased insulin sensitivity, which results in impaired insulin-stimulated glucose uptake (298). As aforementioned, IR does not necessarily imply T2DM. A harmful mechanism for the functionality of β-cells is ‘glucotoxicity’. Glucotoxicity is dependent on the duration and degree of hyperglycemia, in which the elevated glucose levels, characteristic of T2DM, contribute to desensitization of pancreatic β-cells in insulin secretion (292,294). Another mechanism that contributes to further loss of β-cells and pancreatic dysfunction is ‘lipotoxicity’. It is directly related to fat occuring obesity and is accompanied by an underlying predisposition to T2DM and increased serum FFA levels (292,299). FFAs are elevated in the plasma of patients with T2DM, due to uncontrolled lipolysis, by insulin-sensitive lipase, in adipocytes (300). The high levels of FFAs impair the function of pancreatic β-cells and the glucose-induced insulin secretion (300) (Fig. 4). Another mechanism, related to the disruption of pancreatic β-cells, is their low antioxidant defense, since they do not express, in high ratio, antioxidant enzymes, which make them susceptible to oxidative damage (301). Finally, due to the accumulated fat during obesity, increased levels of cytokines from macrophages are observed, such as IL-1β (302), which is also responsible for the deficiency of insulin secretion from β-cells (34) (Fig. 4). In addition to BMI, there is a strong association between abdominal obesity (central obesity) and the incidence of T2DM (24,303308). Abdominal obesity is associated with the following conditions that may lead to systemic inflammation and IR: i) increased levels of glucose and non-esterified fatty acids; ii) hormonal imbalance with increased leptin levels and decreased adiponectin levels; and iii) increased secretion of cytokines and pro-inflammatory substances from fat cells (309,310). In more detail, in terms of the secretion of cytokines and pro-inflammatory substances from adipocytes, the driving force is the excess visceral fat that triggers the cascade of inflammation (309,310). In response to the secretion of these substances, mononuclear cells migrate from blood circulation to the AT, and they differentiate into macrophages (309,310). Macrophages secrete cytokines, thus in obesity an increased secretion of TNF-α and IL-6 is observed (309,310) (Fig. 4). Secretion of TNF-α is responsible for tissue inflammation, due to its role in the generation of ROS, and activation of transcription-induced pathways, and for IR, in peripheral tissues and adipocytes, which is important for the onset of T2DM (309,310). The increase in IL-6 levels contributes to the prevention of the binding of insulin to its receptor, through the induction of proteins associated with it (310312), as well as the induction of CRP, which like TNF-α, is associated with IR (309,310) (Fig. 4). Additionally, hyperglycemia, characteristic in T2DM, is associated with the generation of advanced glycation end products (AGEs), binding to their receptors. AGEs are responsible for the up-regulation of the transcription factor NF-κB, which is a mediator of inflammation and immunity (313), while AGEs also block the activity of NO in the vascular endothelium and promote the production of ROS (313) (Fig. 4). Finally, a key role for chronic inflammation and OS in obesity related T2DM is mitochondrial dysfunction (314316). Specifically, due to the increased concentration of sugars, glycolysis and the Krebs cycle are induced and cause an increased flow of NADH and FADH2 (flavin adenine dinucleotide; reduced form) (314316). They act as electron donors to the mitochondrial chain, resulting in the accumulation of electron donors in coenzyme Q (314316). This results in the production of mitochondrial superoxide radical (FR), an important source of ROS from adipocytes (314316). Chronic exposure to high intracellular ROS levels in adipocytes ultimately causes mitochondrial dysfunction and perpetuates AT inflammation, together with impairment of the AKT signaling pathway that induces IR (314316) (Fig. 4).

Therapeutic interventions on inflammatory mediators for the therapy of obesity-associated metabolic diseases

Treating inflammation by blocking IL-1R

In patients with T2DM, the high blood glucose levels induce cytokine IL-1β production and secretion, in the β-cells of the pancreatic islets of Langerhans, leading to pro-inflammatory immune responses, β-cell dysfunction, decreased β-cell proliferation and increased β-cell apoptosis (198,317,318). Therefore, short-term IL-1 receptor (IL-1R) blockage could lead to improvements in both metabolic and inflammatory parameters in patients with MetS and T2DM and may represent a potential targeted therapeutic approach for these patients. For instance, Larsen et al tested the effects of anakinra therapy in two studies (319,320). In their first study, Larsen et al (319) examined the effects of anakinra treatment, in a double-blind, parallel-group trial with 70 patients with T2DM, that were randomly assigned to receive a placebo, or 100 mg of anakinra, subcutaneously, once daily. Anakinra is a human recombinant IL-1Ra, which prevents signal dunsduction of IL-1α and IL-1β (321,322). Anakinra is approved by the Food and Drug Administration (FDA) for the treatment of rheumatoid arthritis in adults and neonatal onset multisystem inflammatory disease (323). During a 13-week treatment period, anakinra administration improved glycemia and pancreatic β-cell secretory function, compared with the placebo group. This occurred by reducing the glycated hemoglobin levels (HbA1c), the ratio of proinsulin to insulin (marker of pancreatic β-cell dysfunction and reduced insulin secretory capacity), the serum levels of systemic inflammatory markers (IL-6 and CRP), and enhanced C-peptide and insulin secretion (319) (Table II). Furthermore, Larsen et al (320) in a 39-week follow-up study examined the durability of anakinra administration on the management of T2DM and found maintenance of increased insulin secretion and reduction of insulin requirements (320). These findings suggest that IL-1R blockade with anakinra may improve glucose control and β-cell secretory function for a long period (319,321). Van Asseldonk et al (324) in a randomized, double-blind, crossover study examined the effects of anakinra in nondiabetic, obese individuals, with MetS, at a dose of 150 mg, subcutaneously, once daily for a 4-week treatment period. The authors found that anakinra administration compared with the placebo group, led to a significantly lower degree of inflammation by reducing the circulating CRP levels and the number of leukocytes accompanied by a significant increase in the disposition index and improvement in pancreatic β-cell function (324) (Table II). Nevertheless, anakinra did not significantly improve insulin sensitivity (324) (Table II). Also, van Poppel et al (325) assessed the effects of anakinra therapy in another double-blind, randomized, placebo-controlled crossover study, involving 16 subjects, with impaired glucose tolerance, assigned to receive 150 mg anakinra daily, for 8 weeks. A significant improvement in the first-phase insulin secretion and pancreatic β-cell function was found (325) (Table II). In line with these findings, Cucak et al (326) evaluated in female non-obese diabetic (NOD) mice, the effects of SER140, which is a 10-amino-acid peptide antagonist of IL-1β receptors (IL-1Ra), on the progression of diabetes and pancreatic β-cell changes. The study consisted of an 8-week treatment period. The results of this study showed a reduction in the incidence of diabetes, by >50%, compared with the control group, a decrease in non-fasting plasma glucose concentrations and an increase in plasma insulin levels. Additionally, SER140 administration changed the immune-endocrine dynamics in the NOD mouse pancreas. The authors suggested that the SER140 treatment can postpone the onset of diabetes in female NOD mice by competing with IL-1β for IL-1β receptors (IL-1R) (326).

Table II.

Summary of representative experimental studies for anti-inflammatory therapies in the management of human metabolic dysfunction.

Table II.

Summary of representative experimental studies for anti-inflammatory therapies in the management of human metabolic dysfunction.

First author/s yearDisordersNo. of participants and type of clinical TrialExperimental drugMechanismDosesRouteFollow-upMain effects on metabolic dysfunction(Refs.)
Paquot et al 2000ObesitySingle-center, single blind,Ro 45-2081:Recombinant50 mgIV inj.6 daysIneffective in IR reduction(347)
with IRsequential treatmentSolubleTNF-α
(placebo, followed byTNF-receptorantagonist
active drug) clinical trial;p55 linked to
n=7the fc portion
of human IgG1
Dominguez et alObesityRandomized, parallel-EtanerceptAnti-ΤΝF-α25 mg twice/Sub-Q inj.4 weeksIneffective in IR reduction(348)
2005with T2DMgroup, open-label clinical week
trial; n=20
Kiortsis et alIR and RAn=28 with RA; n=17 withInfliximabAnti-ΤΝF-α3 mg/kg at 0, 2,IV inj.6 monthsImprovement in insulin(338)
2005or ASAS; clinical prospective 6 weeks and resistance
study thereafter every
8 weeks
Bernstein et alObesityRandomized double-blindEtanerceptAnti-ΤΝF-α50 mg/weekSub-Q inj.4 weeksReduction in inflam-(343)
2006with MetScontrolled clinical trial; mation markers (CRP,
n=28 drug; n=28 placebo; fibronectin); increase in
2 withdrew in each group. total adiponectin levels;
no effects on IR
Gonzalez-GayIR and RAn=27; cohort studyInfliximabAnti-ΤΝF-α3 or 5 mg/kgIV inj.1 monthImprovement in IR and(339)
et al 2006 without placebo group every 6 or insulin sensitivity
due the severity of RA. 8 weeks
according to the
disease severity
Larsen et alT2DMn=70; randomizedAnakinraRecombinant10 mg/daySub-Q inj.13 weeksReduction in HbA1c(319)
2007 parallel-group, double- IL-1Ra values; reduction in
blind clinical trial. pro-insulin/insulin ratio;
reduction in systemic
inflammatory markers
(CRP, IL-6); rise in
C-peptide secretion; rise
in insulin secretion
Lo et al 2007Obesityn=56; randomized double-EtanerceptAnti-ΤΝF-α50 mg/weekSub-Q inj.4 weeksIncrease in total adipo-(344)
with MetSblind placebo-controlled nectin levels; reduction
clinical trial in blood HMWA/total
adiponectin ratio
Larsen et al 2009T2DMRandomized parallel-AnakinraRecombinant10 mg/daySub-Q inj.39 weeksIncreased insulin(320)
group, double-blind trial; IL-1Ra secretion; reduction in
n=34 drug group; n=36 insulin requirements
placebo group
Ramos-ZavalaT2DMRandomized double-blind,DiacereinAnti-IL-1β in50 mg once orper os2 monthsIncrease in insulin(352)
et al 2011 placebo-controlled clinical combinationtwice/day secretion; decrease in
trial; n=20 drug group; with TNF-α fasting glucose levels
n=20 placebo group antagonism
Stanley et alObesityRandomized placebo-EtanerceptAnti-ΤΝF-α50 mg twice/Sub-Q inj.6 monthsReduction in fasting blood(346)
2011with MetScontrolled double blind week for the glucose levels; increase in
clinical trial; n=16 drug first 3 months blood HMWA/total
group; n=24 placebo and 50 mg/week adiponectin ratio
group for the final
3 months
van AsseldonkObesityN=9; randomized, double-AnakinraRecombinant150 mg/daySub-Q inj.4 weeksCRP reduction values;(324)
et al 2011with MetSblind, placebo-controlled, IL-1Ra WBC number reduction;
withouttwo period crossover trial improvement in pancreatic
T2DM β-cell function; no effects
on insulin sensitivity
Cavelti-WederT2DMRandomized placebo-GevokimumabAnti-IL-1βIncreasing dosesIV inj. or2 monthsReduction in HbA1c(327)
et al 2012 controlled trial; n=81 (0.01, 0.03, 0.1,Sub-Q inj.(US arm);values; reduction in
drug group (n=56 in the 0.3 and 1 mg/kg) 3 monthsCRP; rise in C-peptide
US arm) and (n=25 in (US and Swiss (Swisssecretion; rise in insulin
the Swiss arm); n=17 arm) and arm)secretion; improvement
placebo group 3 mg/kg in insulin sensitivity
Ridker et al 2012T2DM withn=556; randomizedCanakinumabAnti-IL-1β5, 15, 50 orSub-Q inj.5 monthsIneffective in HbA1c,(330)
high CV placebo-controlled 150 mg/month fasting glucose and
riskmultinational phase IIb insulin values; reduction
clinical trial in systemic inflammatory
markers (CRP, IL-6 and
fibrinogen)
Rissanen et alIGT orn=190, randomizedCanakinumabAnti-IL-1β150 mg singleSub-Q inj.4 weeksTrend towards increased(328)
2012T2DMparallel-group, placebo- dose insulin secretion
undercontrolled clinical trial
insulin +
metformin
treatment
Faghihimani et alT2DMRandomized, double-SalsalateNF-κΒ3 g/dayper os12 weeksReduction in HbA1c(361)
2013 blind, placebo-controlled inhibition values and fasting glucose
clinical trial; n=30 drug levels
group; n=30 placebo group
Godfine et al 2013T2DMRandomized placebo-SalsalateNF-κΒ3.5 g/dayper os48 weeksReduction in HbA1c(362)
controlled parallel trial; inhibition values; improvement in
n=146 drug group; glycemia
n=140 placebo group
Hensen et al 2013T2DMRandomized, parallel-CanakinumabAnti-IL-1β5, 15, 50 orSub-Q inj.4 monthsReduction in HbA1c(329)
undergroup, double-blind, 150 mg/month values; improvement in
Metforminplacebo-controlled trial; pancreatic β-cell function
treatmentn=372 drug group; n=179
placebo group
Sloan-LancasterT2DMn=106; randomizedLY2189102Anti-IL-1β0.6, 18 orSub-Q inj.12 weeksReduction in postprandial(333)
et al 2013 phase II, parallel-group, 180 mg/week glycemic levels; reduction
double-blind, placebo- in inflammatory markers
controlled clinical trial (CRP, IL-6)
Di Prospero et alT2DMRandomized double-JNJ-41443532CCR2250 mg vsper os4 weeksReduction in HbA1c(357)
2014 blind, placebo-controlled antagonist1,000 mg values
study; n=41 drug group
and completed trial;
n=20 placebo group;
n=20 pioglitazone group
Noe et al 2014T2DMRandomized multicenter,CanakinumabAnti-IL-1β0.03, 0.1, 0.3,IV inj.12 weeksSuppression of high(332)
underdouble-blind, placebo- 1.5 or 10 mg/kg sensitivity CRP (hsCRP)
Metfomincontrolled, dose-escalation (single dose)
treatmentclinical trial; cohort 1:
n=10 drug group and n=5
placebo; cohort 2: n=45
drug group; n=45 received
placebo; cohort 3: n=72
drug group with different
doses; n=34 placebo
group; cohort 4: n=20
drug group and n=34
placebo group
van Poppel et alIGTn=16; randomized doubleAnakinraRecombinant150 mg/daySub-Q inj.4 weeksImprovement in the first(325)
2014 blind, placebo-controlled IL-1Ra phase insulin secretion
cross-over trial
Choudhury et alASCVDRandomized phase II,CanakinumabAnti-IL-1β150 mg/monthSub-Q inj.12 monthsReduction in inflammation(331)
2016and T2DMdouble-blind, placebo- markers (hsCRP, IL-6)
or IGTcontrolled clinical trial;
n=95 drug group; n=94
placebo group
Cardoso et alT2DMRandomized double-DiacereinAnti-IL-1β in100 mg/dayper os48 weeksReduction in HbA1c with(353)
2017 blind, placebo-controlled, combination(single dose) peak of effect at 24 weeks
parallel, clinical trial; with TNF-α of treatment
n=43 drug group; n=41 antagonism
placebo group
Piovesan et alT2DM andRandomized placebo-DiacereinAnti-IL-1β in50 mg twice/dayper os90 daysImprovement in metabolic(356)
2017chroniccontrolled, parallel-group combination control of T2DM;
kidneytrial; n=36 drug group; with TNF-α reduction in nighttime
diseasen=36 placebo group antagonism blood pressure; no effects
in GFR and ACR
Everett et al 2018Prior MIn=10,061; randomizedCanakinumabAnti-IL-1β50 or 150 orSub-Q inj.3.7 yearsNo long-term benefits in(334)
with ordouble-blind, placebo- 300 mg every HbA1c values; No effects
without pre-controlled trial 12 weeks on the reduction of new-
diabetes or onset T2DM
T2DM
Tres et al 2018T2DMRandomized double-blind,DiacereinAnti-IL-1β in50 mg twice/dayper os12 weeksReduction in HbA1c(354)
parallel, placebo- combination in patients with long
controlled clinical trial; with TNF-α established T2DM under
n=36 drug group; n=36 antagonism long-term anti-diabetic
placebo group treatment.
Tuttle et al 2018T2DM andn=129; randomizedBaricitinibAnti- JAK1/0.75, 1.5 orper os24 weeksReduction in HbA1c(360)
diabeticphase II, parallel-group, JAK24 mg/day values, inflammation and
kidneydouble blind, placebo- albuminuria
diseasecontrolled clinical trial
Genovese et alRA withPost hoc analysis of threeSarilumab vs.IL-6RaSarilumab: 150Sub-Q inj.Up toSarilumab was associated(366)
2020or withoutrandomized, controlledadalimumabvs TNF-αor 200 mg every 24 weekswith more reduction in
T2DMclinical trials; n=184 inhibitor2 weeks vs. adali- HbA1c values compared
patients with T2DM; mumab: 40 mg to adalimumab
n=1,928 without diabetes. every 2 weeks
Ruscitti et al 2021RA andRandomized open-label,AnakinraRecombinantAnakinra:Anakinra:6 andAnakinra was associated(351)
T2DMprospective, controlled,versus anti-IL-1Ra100 mg/day;Sub-Q inj.;18 monthswith more reduction in
parallel-group clinicalTNF agentsversus anti-Anti-TNFanti-TNF HbA1c values and
trial; n=17 received(Etanercept,TNF-αagents: dosesagents: antidiabetic drugs
anakinra; n=15 receivedInfliximab, according to theadmin- compared to TNF-α
anti-TNFα agent for theadalimumab, internationalistration inhibitors at 6 months
6-month follow-up; forgolimumab, guidelinesroute follow-up; anakinra had
the last follow-up n=15certolizumab according no effects in HbA1c values
received anakinra; n=14pegol) to the compared to TNF-α
anti-TNFα agent guidelines inhibitors at 18 months
follow-up but continued to
reduce antidiabetic drugs
compared to TNF-α
inhibitors
van den OeverRA or OAn=36 with RA and n=33Adalimumabanti-TNF-α40mg/2weeksSub-Q inj.6 monthsImprovement in IR and(337)
et al 2021and IRwith OA (control group) β-cell function in RA
patients with active disease
JangsripornpakornT2DMRandomized, double blind,DiacereinAnti-IL-1β in50mg/dayper os12 weeksReduction in HbA1c(355)
et al 2022 placebo-controlled trial; combination(single dose) values
n=18 drug group; n=17 with TNF-α
placebo group antagonism

[i] IL-1Ra, IL-1 receptor antagonist; IL-6Ra, IL-6 receptor antagonist; SQ inj., subcutaneous injection; IV inj., intravenous injection; MI, myocardiac infraction; CCR2, C-C chemokine receptor-2 antagonist; ACR, urinary albumin/creatinine ratio; GFR, glomerular filtration rate; T2DM, type 2 diabetes mellitus; IR, insulin resistance; RA, rheumatoid arthritis; OA, osteoarthritis; ASCVD, atherosclerotic cardiovascular disease; IGT, impaired glucose tolerance; hsCRP, high sensitivity CRP; AS, ankylosing spondylitis.

Treating inflammation with anti-IL-1β therapies

Other promising IL-1β blocking therapies have demonstrated antidiabetic potential as well. Cavelti-Weder et al (327) evaluated the safety and biological activity of gevokizumab in patients with T2DM. Gevokizumab is a recombinant human monoclonal anti-IL-1β antibody. In this study a total of 98 patients with T2DM participated, 17 patients to the control group and 81 patients to the gevokizumab treatment group, at increasing doses. It was found that gevokizumab treatment was safe and led to significant reduction in HbA1c values (−0.85%) after 3 months, accompanied by augmented C-peptide secretion, increased insulin sensitivity and decreased CRP levels (327) (Table II). Rissanen et al (328) evaluated the effects of a single dose of canakinumab, a recombinant human monoclonal antibody targeting circulating IL-1β in patients with impaired glucose tolerance or T2DM treated with insulin and metformin. The authors found a trend towards increased insulin secretion (Table II). In another study conducted with 551 metformin-treated patients, with T2DM, Hensen et al (329) assessed the safety, tolerability and effects of different monthly doses of canakinumab (5, 15, 50 or 150 mg). The authors found that canakinumab treatment was safe and well tolerated. In addition, canakinumab (50 mg) led to a reduction in HBA1c values compared with the placebo group (329) (Table II). These findings suggest that monthly adjuvant treatment, with 50 mg canakinumab, on metformin-treated patients, with T2DM, could potentially improve pancreatic β-cell function (329). However, Ridker et al (330) did not find alterations in HbA1c, glucose and insulin levels after canakinumab treatment in patients with T2DM with high cardiovascular risk (Table II). By contrast, cankinumab significantly reduced inflammation markers such as CRP, IL-6 and fibrinogen (330) (Table II). Choudhury et al (331) examined the effects of cankinumab in patients with atherosclerotic cardiovascular disease and T2DM or impaired glucose tolerance for 12 months and found reduction in inflammation markers (hsCRP, IL-6) compared with the control group (Table II). Furthermore, Noe et al (332) found similar results (Table II). In addition, Sloan-Lancaster et al (333) examined the effects of LY2189102 in the treatment of patients with T2DM. LY2189102 is a recombinant human monoclonal antibody (IgG4) that binds to IL-1β with high affinity and neutralizes its activity by forming a complex with circulating IL-1β. The authors demonstrated that the weekly subcutaneous administration of LY2189102 for 12 weeks can reduce postprandial glycemic levels and improve anti-inflammatory effects in patients with T2DM (Table II). In a canakinumab anti-inflammatory thrombosis outcomes study, IL-1β inhibition by canakinumab did not show long-term (over a median period of 3.7 years) benefits in the reduction of HbA1c values in patients prior to myocardial infarction with or without pre-diabetes or T2DM (334) (Table II). In addition, canakinumab administration was ineffective in reducing the occurrence of new onset T2DM (334) (Table II). Also, the development of vaccines against IL-1β represents a treatment option for IL-1β-dependent diseases such as T2DM (327).

Treating inflammation with anti-TNF-α therapies

Since obese humans have increased circulating levels of TNF-α and TNF-α levels, in human AT, is positively associated with BMI and hyperinsulinemia (317,335), their levels have been proposed to play a role in the development and pathogenesis of IR and T2DM (317,336). Indeed, van den Oever et al (337) in their study found that adalimumab administration in patients with RA or OA and IR improves IR and pancreatic β-cell function (337) (Table II). Adalimumab binds with specificity to TNF-α and inhibits its interaction with the p55 and p75 cell surface TNF receptors (337). Also, infliximab is a chimeric monoclonal antibody that binds TNF-α with high affinity and neutralizes TNF-α. Kiortsis et al (338) found that the administration of infliximab in patients with RA or ankylosing spondylitis and IR improved insulin sensitivity (Table II). Similar results were also found by Gonzalez-Gay et al (339) in insulin resistant patients, with RA (Table II). In addition, Haida et al (340) demonstrated that infliximab treatment prevents hyperglycemia and liver gluconeogenesis, in high fat diet-fed mice. Moreover, infliximab seems to ameliorate TNF-α-induced IR, in 3T3-L1 adipocytes, in vitro, by improving the insulin signaling pathway, via inhibition of protein tyrosine phosphatase 1B (341). Additionally, Abdelhamid et al (342) showed that infliximab administration in rats reduces TGs, increases HDL-c levels and reverses fructose-induced adiponectin resistance. Therefore, the authors suggested that infliximab may affect the manifestation of MetS. However, infliximab failed to affect MetS-mediated hyperglycemia, hypertension and the elevated peroxidation levels, as the levels of malondialdehyde dictate (342). Bernstein et al (343) investigated the effects of the inhibition of TNF-α with entanercept (a TNF-α blocker), in patients with MetS, for a 4-week treatment period. The authors concluded that etanercept reduced CRP levels (Table II). Also, Lo et al (344) randomized obese patients with MetS on etanercept and demonstrated increased circulating levels of total adiponectin but the ratio of high molecular weight adiponectin (HMWA) to total adiponectin was reduced (Table II); HMWA is the most biologically active form of the adipokine and is thought to mediate insulin sensitivity (345). Conversely, Stanley et al (346) found that the administration of etanercept, on obese individuals, with MetS, increased the ratio of HMWA to total adiponectin (Table II). Paquot et al (347) in their clinical trial failed to show an improvement in insulin sensitivity after TNF-α neutralization, following a single intravenous administration of a TNF receptor antagonist (Ro45-2081; a soluble TNF-receptor-IgG fusion protein) in obese insulin resistant patients (Table II). Moreover, Dominguez et al (348) failed to reverse vascular and metabolic IR, after short-term etanercept treatment, in obese patients with T2DM (Table II). This evidence may support the hypothesis that AT TNF-α, which is not secreted in the systemic circulation may act in an autocrine or paracrine manner. Therefore, the anti-TNF-α agents may not reach the AT microcirculation, which is markedly impaired in T2DM (347,349,350). Thus, anti-TNF-α therapy may fail to improve insulin sensitivity in such cases. In summary, treatment with anti-TNF-α agents in patients with T2DM did not yield consistent results for glucose and HbA1c reduction. Ruscitti et al (351) investigated the effects of anti-IL-1 treatment with anakinra compared with TNF-α inhibitors, such as etanercept, adalimumad, infliximad, certolizumab pegol or golimumab, in patients with RA and T2DM in an open label, prospective, controlled, parallel-group trial. The authors found that anakinra reduced HbA1c values compared with TNF-α inhibitors after a 6 month treatment period and also reduced antidiabetic drugs defined as the reduction of administered dosages, change from combination therapy to monotherapy or discontinuation of anti-diabetic drugs (351). However, after the mean follow-up of 18 months anakrinra had no effects in HbA1c values compared with TNF-α inhibitors but continued to reduce the use of antidiabetic drugs. On the contrary, an increase of anti-diabetic therapies was needed in participants treated with TNF-α inhibitors to reduce HbA1c levels (351) (Table II).

Treating inflammation with synergic anti-IL-1β and anti-TNF-α therapies

Diacerein is both an IL-1βR blocker and a TNF-α antagonist by its active metabolite, rhein (352). Ramos-Zavala et al (352) found that diacerein administration in patients with T2DM increased insulin secretion and decreased fasting glucose levels (Table II). In addition, Cardoso et al (353) found that diacerein administration reduced HbA1c values in patients with T2DM (Table II). These findings are in agreement with the studies reported from Tres et al (354) (Table II) and Jangsiripornpakorn et al (355) (Table II). In patients with T2DM and chronic kidney disease, intervention with diacerein improves the metabolic control of T2DM and reduces nighttime blood pressure but has no effects in glomerular filtration rate and urinary albumin/creatinine ratio (356) (Table II).

Treating inflammation with CCR2 antagonists

Drugs targeting immune cell infiltration have been tested for anti-inflammatory and anti-obesity therapy as well. Di Prospero et al (357) evaluated the safety, tolerability, pharmacokinetics and pharmacodynamics of JNJ-41443532, a CCR2 antagonist, in a small sample size, double-blind, placebo-controlled, randomized, multicenter study for 4-weeks, in patients with T2DM. The authors found that JNJ-41443532 treatment was well tolerated in patients with T2DM and showed modestly improved glycemic parameters compared with the placebo group (357) (Table II). Also, Mulder et al (358) examined in male mice whether propagermanium, an inhibitor of CCR2, could attenuate tissue inflammation and NASH development. The results of this study showed that early propagermanium intervention was more effective than late intervention in attenuating IR, WAT inflammation and NASH development (358). In addition, Huh et al (359) investigated the effects of PF4178903, an antagonist for dual CCRs, CCR2 and CCR5, on obesity and IR, in high fat diet fed mice. The authors demonstrated that the dual CCR2 and CCR5 antagonist, PF4178903, attenuated metabolic dysfunction, induced by a high-fat diet. There was a decrease in body weight gain, blood glucose levels, lipid levels, adipocyte size and systemic inflammation, and an improvement in glucose tolerance and insulin sensitivity (359). Particularly, PF4178903 significantly shifted the M1 macrophage phenotype towards the M2 phenotype, in high fat diet-induced obesity, suggesting that the dual CCR2 and CCR5 blockade regulates macrophage polarization in AT macrophages (359).

Treating inflammation with NF-κΒ inhibition

Tuttle et al (360) found that baricitinib, an oral, reversible, selective inhibitor of JAK1 and JAK2 decreases inflammation, HBA1c and albuminuria in patients with T2DM and diabetic kidney disease. Salsalate is a product of salicylate showing anti-inflammatory effects by inhibiting the IKKb/NF-κΒ and JNK singnaling pathways. Faghihimani et al (361) found that the adiminstration of salsalate in T2DM reduced HbA1c values and fasting glucose levels (Table II). Similar results were shown in the study by Godfine et al (362) (Table II). Greater improvement of glycemic control of salsalate might be seen with newly diagnoseed patients with T2DM or with longer duration of antidiabetic treatment (363).

Treating inflammation with IL-6R inhibitors

Sarilumab is a human anti-IL-6 receptor (IL-6R) monoclonal IgG1 antibody that targets both the membrane-bound and soluble IL-6 receptor forms (364,365). Therefore, sarilumab blocks both the cis- and trans-inflammatory signaling cascades of IL-6 and reduces the activity of pro-inflammatory cytokines and inflammation (364,365). In particular, sarilumab has been shown to reduce HbA1c values after a 24 week treatment period compared with adalimumab in patients with rheumatoid arthritis with or without T2DM (366) (Table II).

Side effects of anticytokine therapies

However, anticytokine therapy is not devoid of unwanted side effects (367369). Serious side effects derived from the use of anticytokines include infections, reactivation of latent tuberculosis and hepatitis B virus infection, hepatotoxicity, demyelinating disorders of the central nervous system and adverse cardiac events (369). Therefore, potential benefits should be carefully weighed against potential side effects related to the use of these medications (369).

Anti-inflammatory effects of thiazolidinediones (TZDs) in the treatment of obesity-associated T2DM

Pharmacological elevation of plasma levels of adiponectin could become a promising therapeutic strategy in countering-balacing obesity-associated T2DM (370). The thiazolidinediones (TZDs) are agonists of peroxisome proliferation activating receptor-γ (PPARγ), with TZDs such as troglitazone, rosiglitazone, glitazone and pioglitazone having been shown to increase the activation of PPARγ, elevate serum adiponectin concentrations, restore lipogenic function and decrease inflammation (371373). TZDs also block the ability of TNF-α to inhibit insulin signaling through increased serine phosphorylation of IRS-1 (374). Wolf et al (375) demonstrated in vitro that adiponectin displays potent immunosuppressive effects inducing the production of anti-inflammatory cytokines IL-10 and IL-1Ra in myeloid cell types. In addition, IL-10 can inhibit the production of pro-inflammatory mediators by macrophages, including IL-1, IL-2, IL-6, IL-12, interferon gamma (INFγ) and TNF-α (375,376). Furthermore, adiponectin rapidly up-regulates IL-10 and subsequently increases the levels of tissue inhibitor metalloproteinase-1 (an inhibitor of matrix metalloproteinases) in human macrophages preventing the degradation of the ECM (78). Although, TZDs are effective in controlling glycemia and IR, and are not associated with hypoglycemia, when used as monotherapy (377,378), there are some serious safety concerns that must be considered when selecting TZDs for the treatment of metabolic disorders (370). For example, troglitazone was removed from the market after the FDA received reports of 94 cases of troglitazone-induced liver failure (379). Also, pioglitazone usage increases the risk of bladder cancer (380) and edema in T2DM (381,382). Another important safety issue of TZDs, is their risk for heart failure due to fluid retention (383). Moreover, glitazone has been associated with macular edema of the retina that leads to vision loss (384,385). Additionally, TZDs decrease bone density and therefore increase the bone fracture risk (386). Apart from the aforementioned side effects of TZDs, treatment with TZDs is associated with a rise in body weight due to increased fat mass and fluid retention in patients with T2DM (387,388).

Conclusion and future perspectives

Obesity has evolved to an epidemic condition that causes health impairment by increasing the risk of developing other relevant conditions, such as MetS, IR, T2DM, hypertension, atherosclerosis, dyslipidemia, CVDs, respiratory disorders and several types of cancer. The molecular and pathophysiological mechanisms linking visceral obesity and MetS are mediated by chronic low-grade inflammation and OS, but they are not fully understood. Particularly, obesity results in a pro-inflammatory state in the adipocytes characterized by increased recruitment, accumulation and AT infiltration of M1 macrophages with a consequent release of highly pro-inflammatory cytokines, such as IL-1β, IL-6 and TNF-α, and pro-inflammatory adipokines, such as PAI-1, visfatin, resistin and leptin. The polarization of macrophages toward the M1 pro-inflammatory state results in reduced adiponectin levels. Accumulating evidence indicates that obesity related factors, such as a high-calorie diet, sedentary lifestyle, AT micro-environment and gut microbiota deregulation, exacerbate chronic tissue inflammation. To date, clinical studies, which tested the safety, tolerability and efficacy of molecular therapies targeting obesity-associated inflammation, have shown hopeful results by enhancing insulin sensitivity and improving metabolic function and IR, but they still remain unsatisfactory with poor treatment outcomes and in numerous cases are accompanied with serious side effects. Therefore, new efficacious and safe molecular targeted agents need to be discovered.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

FNV and PTN conceptualized the study; FNV created all the figures; FNV, MNV, VKV and PTN wrote and edited the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AGEs

advanced glycation end products

AMPK

AMP-activated protein kinase

APJ

angiotensin II protein J

AT

adipose tissue

ATP

adenosine triphosphate

BMI

body mass index

CCR

C-C motif chemokine receptor

CRP

c-reactive protein

CVDs

cardiovascular diseases

eNOS

endothelial nitric oxide synthase

FADH2

flavin adenine dinucleotide (reduced form)

FFAs

free fatty acids

FRs

free radicals

HDL

high-density lipoprotein

HIF-1α

hypoxia-inducible factor-1α

HMWA

high molecular weight adiponectin

IL-1Ra

interleukin-1 receptor antagonist

iNOS

inducible nitric oxide synthase

IR

insulin resistance

IRS-1

insulin receptor substrate 1

JAK

janus kinase

JNK

c-Jun N-terminal kinase

MAPK

Ras/Raf/mitogen-activated protein kinase

MetS

metabolic syndrome

MCP

monocytes chemoattractant protein

NAFLD

non-alcoholic fatty liver disease

NASH

non-alcoholic steatohepatitis

NO

nitrogen oxide

NOD

non-obese diabetic

OS

oxidative stress

PAI-1

plasminogen activator inhibitor-1

PI3K

phosphatidylinositol 3-kinase

PKB

protein kinase B

PPAR

peroxisome proliferation activating receptor

ROS

reactive oxygen species

STAT

subcutaneous adipose tissue

T2DM

type-2 diabetes mellitus

TGs

triglycerides

TZDs

thiazolidinediones

UCP1

uncoupling protein 1

VAT

visceral adipose tissue

VLDLs

very low density lipoproteins

VSMC

vascular smooth muscle cells

WAT

white adipose tissue

References

1 

Sethi JK and Vidal-Puig AJ: Thematic review series: Adipocyte biology. Adipose tissue function and plasticity orchestrate nutritional adaptation. J Lipid Res. 48:1253–1262. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Luo L and Liu M: Adipose tissue in control of metabolism. J Endocrinol. 231:R77–R99. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Jung UJ and Choi MS: Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci. 15:6184–6223. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Curat CA, Miranville A, Sengenè C, Diehl M, Tonus C, Busse R and Bouloumié A: From blood monocytes to adipose tissue-resident macrophages: Induction of diapedesis by human mature adipocytes. Diabetes. 53:1285–1292. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC, Palmer EL, Tseng YH, Doria A, et al: Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 360:1509–1517. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Nauli AM and Matin S: Why do men accumulate abdominal visceral fat? Front Physiol. 10:14862019. View Article : Google Scholar : PubMed/NCBI

7 

Kahn CR, Wang G and Lee KY: Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 129:3990–4000. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Blüher M: Adipose tissue dysfunction in obesity. Exp Clin Endocrinol Diabetes. 117:241–250. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Tan CY and Vidal-Puig A: Adipose tissue expandability: The metabolic problems of obesity may arise from the inability to become more obese. Biochem Soc Trans. 36:935–940. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Sebo ZL and Rodeheffer MS: Assembling the adipose organ: Adipocyte lineage segragation and adipogenesis in vivo. Development. 146:dev1720982019. View Article : Google Scholar : PubMed/NCBI

11 

Lafontan M and Langin D: Lipolysis and lipid modilization in human adipose tissue. Prog Lipid Res. 48:275–297. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Frayn KN: Adipose tissue as a buffer for daily lipid flux. Diabetologia. 45:1201–1210. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS and Obin MS: Adipocyte death defines macrophage location and function in adipose tissue of obese mice and humans. J Lipid Res. 46:2347–2355. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Ellulu MS, Patimah I, Khazaai H, Rahmat A and Abed Y: Obesity and inflammation: The linking mechanism and the complications. Arch Med Sci. 13:851–863. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Hotamisligil GS: Inflammation and metabolic disorders. Nature. 444:860–867. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Fantuzzi G: Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol. 115:911–919. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Weir CB and Jan A: BMI classification percentile and cut off points. StatPearls Treasure Island, FL: StatPearls Publishing; 2020

18 

Marcadenti A and de Abreu-Silva EO: Different adipose tissue depots: Metabolic implications and effects of surgical removal. Endocrinol Nutr. 62:458–464. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Fuster JJ, Ouchi N, Gokce N and Walsh K: Obesity-induced changes in adipose tissue microenvironment and their impact on cardiovascular disease. Circ Res. 118:1786–1807. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Osborn O and Olefsky JM: The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med. 18:363–374. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Gustafson B, Hedjazifar S, Gogg S, Hammarstedt A and Smith U: Insulin resistance and impaired adipogenesis. Trends Endocrinol Metab. 26:193–200. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Kuk JL, Katzmarzyk PT, Nichaman MZ, Church TS, Blair SN and Ross R: Visceral fat in an independent predictor of all-cause mortality in men. Obesity (Silver Spring). 14:336–341. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Klein J, Permana PA, Owecki M, Chaldakov GN, Böhm M, Hausman G, Lapière CM, Atanassova P, Sowiński J, Fasshauer M, et al: What are subcutaneous adipocytes really good for? Exp Dermatol. 16:45–70. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Cameron AJ, Magliano DJ and Soderberg S: A systemic review of the impact of including both waist and hip circumference in risk models for cardiovascular diseases, diabetes and mortality. Obes Rev. 14:86–94. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Koster A, Murphy RA, Eiriksdottir G, Aspelund T, Sigurdsson S, Lang TF, Gudnason V, Launer LJ and Harris TB: Fat distribution and mortality: The AGES-Reykjavik study. Obesity (Silver Spring). 23:893–897. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Arner P, Andersson DP, Thörne A, Wirén M, Hoffstedt J, Näskybd E, Thorell A and Rydén M: Variations in the size of the major omentum are primarily determined by fat cell number. J Clin Endocrinol Metab. 98:E897–E901. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Chait A and den Hartigh LJ: Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med. 25:222020. View Article : Google Scholar : PubMed/NCBI

28 

James WP: Assessing obesity: Are ethnic differences in body mass index and waist classification criteria justified? Obes Rev. 6:179–181. 2005. View Article : Google Scholar : PubMed/NCBI

29 

James WP, Rigby N and Leach R: Obesity and the metabolic syndrome: The stress on society. Ann N Y Acad Sci. 1083:1–10. 2006. View Article : Google Scholar : PubMed/NCBI

30 

El-Sayed AM, Scarborough P and Galea S: Ethnic inequalities in obesity among children and adults in the UK: A systematic review of the literature. Obes Rev. 12:e516–e534. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Barnett AH, Dixon AN, Bellary S, Hanif MW, O'Hare JP, Raymond NT and Kumar S: Type 2 diabetes and cardiovascular risk in the UK south Asian community. Diabetologia. 49:2234–2246. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Misra A and Khurana L: Obesity-related non-communicable diseases: South Asians vs White Caucasians. Int J Obes (Lond). 35:167–187. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Pi-Sunyer X: The medical risks of obesity. Postgrad Med. 121:21–33. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Kyrou I, Randeva HS, Tsigos C, Kaltsas G, Weickert MO, Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, et al: Clinical problems caused by obesity. Endotext [Internet] South Dartmouth (MA): MDText.com, Inc; 2018

35 

Khanna D, Khanna S, Khanna P, Kahar P and Patel BM: Obesity: A chronic low-grade inflammation and its markers. Cureus. 14:e227112022.PubMed/NCBI

36 

Bobbert T, Rochlitz H, Wegewitz U, Akpulat S, Mai K, Weickert MO, Möhlig M, Pfeiffer AFH and Spranger J: Changes of adiponectin oligomer composition by moderate weight reduction. Diabetes. 54:2712–2719. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Hotamisligil GS, Shargill NS and Spiegelman BM: Adipose expression of tumor necrosis factor-α: Direct role in obesity-linked insulin resistance. Science. 259:87–91. 1993. View Article : Google Scholar : PubMed/NCBI

38 

Mohlig M, Weickert MO, Ghadamgadai E, Machlitt A, Pfüller B, Arafat AM, Pfeiffer AFH and Schöfl C: Adipocyte fatty acid-binding protein is associated with marker of obesity, but is an unlikely link between obesity, insulin resistance and hyperandrogenism in polycystic ovary syndrome women. Eur J Endocrinol. 157:195–200. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Fonseca-Alaniz MH, Takada J, Alonso-Vale MI and Lima FB: Adipose tissue as an endocrine organ: From theory to practice. J Pediatr. 83:192–203. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Maury E and Brichard SM: Adipokine dysregulation, adipose tissue inflammation and metabolic syndrome. Mol Cell Endocrinol. 314:1–16. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Gregor MF and Hotamilsigil GS: Inflammatory mechanisms in obesity. Annu Rev Immunol. 29:415–445. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Weickert MO, Hodges P, Tan BK and Randeva HS: Neuroendocrine and endocrine dysfunction in the hyperinsulinemic PCOS patient: The role of metformin. Minerva Endocrinol. 37:25–40. 2012.PubMed/NCBI

43 

Randeva HS, Tan BK, Weickert MO, Lois K, Nestler JE, Sattar N and Lehnert H: Cardiometabolic aspects of the polycystic ovary syndrome. Endocr Rev. 33:812–841. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Makki K, Froguel P and Wolowczuk I: Adipose tissue in obesity-related inflammation and insulin resistance. Cells, cytokines and chemokines. ISRN Inflamm. 2013:1392392013. View Article : Google Scholar : PubMed/NCBI

45 

Sivakumar K, Bari MF, Adaikalakoteswari A, Guller S, Weickert MO, Randeva HS, Grammatopoulos DK, Bastie CC and Vatish M: Elevated fetal adispin/acylation-stimulating protein (ASP) in obese pregnancy: Novel placental secretion via Hofbauer cells. J Clin Endocrinol Metab. 98:4113–4122. 2013. View Article : Google Scholar : PubMed/NCBI

46 

von Loeffelholz C, Mohlig M, Arafat AM, Isken F, Spranger J, Mai K, Randeva HS, Pfeiffer AFH and Weickert MO: Circulation vaspin is unrelated to insulin sensitivity in a cohort of nondiabetic humans. Eur J Endocrinol. 162:507–513. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Elmasry SA, Al-Azzawi MA, Ghoneim AH, Nasr MY and AboZaid MMN: Role of oxidant-antioxidant imbalance in the pathogenesis of chronic obstructive pulmonary disease. Egypt J Chest Dis Tuberc. 64:813–820. 2015. View Article : Google Scholar

48 

Marseglia L, Manti S, D'Angelo G, Nicotera A, Parisi E, Di Rosa G, Gitto E and Arrigo T: Oxidative stress in obesity: A critical component in human diseases. Int J Mol Sci. 16:378–400. 2015. View Article : Google Scholar

49 

Reilly SM and Saltiel AR: Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol. 13:633–643. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Rheinheimer J, de Souza BM, Cardoso NS, Bauer AC and Crispim D: Current role of the NLRP3 inflammasome on obesity and insulin resistance: A systematic review. Metabolism. 74:1–9. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Deshmane SL, Kremley S, Amini S and Sawaya BE: Monocyte chemoattractant protein-1 (MCP-1): An overview. J Interferon Cytokine Res. 29:313–326. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Taylor EB: The complex role of adipokines in obesity, inflammation and autoimmunity. Clin Sci (Lond). 135:731–752. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Matsushima K, Larsen CG, DuBois GC and Oppenheim JJ: Purification and characterization of a novel monocyte chemotactic and activating factor produced by a human myolomonocytic cell line. J Exp Med. 169:1485–1490. 1989. View Article : Google Scholar : PubMed/NCBI

54 

Rollins BJ: Chemokines. Blood. 90:909–928. 1997. View Article : Google Scholar : PubMed/NCBI

55 

Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, Kitazawa S, Miyachi H, Maeda S, Egashira K and Kasuga M: MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 116:1494–1505. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Singh S, Anshita D and Ravichandiran V: MCP-1: Function, regulation, and involvement in disease. Int Immunopharmacol. 101((PtB)): 1075982021. View Article : Google Scholar : PubMed/NCBI

57 

Dietze-Schroeder D, Sell H, Uhlig M, Koener M and Eckel J: Autocrine action of adiponectin on human fat cells prevents the release of insulin resistance-inducing factors. Diabetes. 54:2003–2011. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Christiansen T, Richelsen B and Bruun JM: Monocyte chemoattractant protein-1 is produced in isolated adipocytes, associated with adiposity and reduced after weight loss in morbid obese subjects. Int J Obes (Lond). 29:146–150. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Cancello R, Henegar C, Viguerie N, Taleb S, Poitou C, Rouault C, Coupaye M, Pelloux V, Hugol D, Bouillot JL, et al: Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes. 54:2277–2286. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Piemonti L, Calori G, Mercalli A, Lattuada G, Monti P, Garancini MP, Constantio F, Ruotolo G, Luzi L and Perseglin G: Fasting plasma leptin, tumor necrosis factor-alpha receptor 2, and monocyte chemoattracting protein 1 concentration in a population of glucose-tolerant and glucose-intolerant women: Impact on cardiovascular mortality. Diabetes Care. 26:2883–2889. 2003. View Article : Google Scholar : PubMed/NCBI

61 

Simeoni E, Hoffman MM, Winkelman BR, Ruiz J, Fleury S, Boehm BO, März W and Vassalli G: Association between the A-2518G polymorphism in the monocyte chemoattractant protein-1 gene and insulin resistance and type 2 diabetes mellitus. Diabetologia. 47:1574–1580. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Herder C, Baumert J, Thorand B, Koenig W, de Jager W, Meisinger C, Illig T, Martin S and Kolb H: Chemokines as risk factors for type 2 diabetes: Results from the MONICA/KORA Augsburg study, 1984–2002. Diabetologia. 49:921–929. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Kim CS, Park HS, Kawada T, Kim JH, Lim D, Hubbard NE, Kwon BS, Rrickson KL and Yu R: Circulating levels of MCP-1 and IL-8 are elevated in human obese subjects and associated with obesity-related parameters. Int J Obes (Lond). 30:1347–1355. 2006. View Article : Google Scholar : PubMed/NCBI

64 

Shimobayashi M, Albert V, Woelnerhanssen B, Frei IC, Weissenberger D, Meyer-Gerpach AC, Clement N, Moes S, Colombi M, Meier JA, et al: Insulin resistance causes inflammation in adipose tissue. J Clin Invest. 128:1538–1550. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Zhu B, Guo X, Xu H, Jiang B, Li H, Wang Y, Yin O, Zhou T, Cai JJ, Glaser S, et al: Adipose tissue inflammation and systemic insulin resistance in mice with diet-induced obesity is possibly associated with disruption of PFKFB3 in hematopoietic cells. Lab Invest. 101:328–340. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Ylä-Herttuala S, Lipton A, Rosenfeld ME, Särkioja T, Yoshimura T, Leonard EJ, Witztum JL and Steinberg D: Expression of monocyte chemoattractant protein 1 in macrophage-rich areas of human and rabbit atherosclerotic lesions. Proc Natl Acad Sci USA. 88:5252–5256. 1991. View Article : Google Scholar : PubMed/NCBI

67 

Arakelyan A, Petrkova J, Hermanova Z, Boyajyan A, Lukl J and Petrek M: Serum levels of the MCP-1 chemokine in patients with ischemic stroke and myocardiac infarction. Mediators Inflamm. 14:175–179. 2005. View Article : Google Scholar : PubMed/NCBI

68 

Sukumar D, Partridge C, Wang X and Shapses SA: The high serum monocyte chemoattractant protein-1 in obesity is influenced by high parathyroid hormone and not adiposity. J Clin Endocrinol Metab. 296:1852–1858. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Takeya M, Yoshimura T, Leonard EJ and Takahashi K: Detection of monocyte chemoattractant protein-1 in human atherosclerotic lesions by an anti-monocyte chemoattractant protein-1 monoclonal antibody. Hum Pathol. 24:534–539. 1993. View Article : Google Scholar : PubMed/NCBI

70 

Gu L, Οkada Y, Clinton SK, Gerard C, Sukhova GK, Libby P and Rollins BJ: Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-defined mice. Mol Cell. 2:275–281. 1998. View Article : Google Scholar : PubMed/NCBI

71 

Boring M, Gosling J, Cleary M and Charo IF: Decreased lesion in CCR2-/- mice reveals a role for chemokines in the initation of atherosclerosis. Nature. 394:894–897. 1998. View Article : Google Scholar : PubMed/NCBI

72 

Deng Y and Scherer PE: Adipokines as novel biomarkers and regulators of the metabolic syndrome. Ann N Y Acad Sci. 1212:E1–E19. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Dutheil F, Gordon BA, Naughton G, Crendal E, Courteix D, Chaplais E, Thivel D, Lac G and Benson AC: Cardiovascular risk of adipokines: A review. J Inter Med Res. 46:2082–2095. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Szumilas K, Szumilas P, Słuczanowska-Głąbowsk S, Zgutka K and Pawlik A: Role of adiponectin in the pathogenesis of Rheumatoid arthritis. Int J Mol Sci. 21:82652020. View Article : Google Scholar : PubMed/NCBI

75 

Adolph TE, Grander C, Grabherr F and Tilg H: Adipokines and non-alcoholic fatty liver disease: Multiple interactions. Int J Mol Sci. 18:16492017. View Article : Google Scholar : PubMed/NCBI

76 

Neumann UH, Chen S, Tam YY, Baker RK, Covey SD, Dullis PP and Kieffer TJ: IGFBP2 is neither sufficient nor necessary for the physiological actions of leptin on glucose homeostasis in male ob/ob mice. Endocrinology. 155:716–725. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Zorena K, Jachimowicz-Duda O, Ślęzak D, Robakowska M and Mrugacz M: Adipokines in obesity. Potential lind to metabolic disorders and chronic complications. Int J Mol Sci. 21:35702020. View Article : Google Scholar : PubMed/NCBI

78 

Kumada M, Kihara S, Ouchi N, Kobayashi H, Okamoto Y, Ohashi K, Maeda K, Nagaretani H, Kishida K, Maeda N, et al: Adiponectin specifically increased tissue inhibitor of metalloproteinase-1 through interleukin-10 expression in human macrophages. Circulation. 109:2046–2049. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Quedraogo R, Wu X, Xu SQ, Fuchsel L, Motoshima H, Mahadev K, Hough K, Scalia R and Goldstein BJ: Adiponectin suppression of high-glucose-induced reactive oxygen species in vascular endothelial cells: Evidence for involvement of a cAMP signaling pathway. Diabetes. 55:1840–1846. 2006. View Article : Google Scholar : PubMed/NCBI

80 

Yuan F, Li YN, Liu YH, Yi B, Tian JW and Liu FY: Adiponectin inhibits the generation of reactive oxygen species induced by high glucose and promotes endothelial NO synthase formation in human mesangial cells. Mol Med Rep. 6:449–453. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M and Shimomura I: Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 114:1752–1761. 2004. View Article : Google Scholar : PubMed/NCBI

82 

Castro JR, Grune T and Speckmann B: The two faces of reactive oxygen species (ROS) in adipocyte function and dysfunction. Biol Chem. 397:709–724. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Fujita K, Nishizawa H, Funahashi T, Shimomura I and Shimabukuro M: Systemic oxidative stress is associated with visceral fat accumulation and the metabolic syndrome. Circulation. 70:1437–1442. 2006. View Article : Google Scholar : PubMed/NCBI

84 

Kanazawa I, Yamaguchi T, Yano S, Yamauchi M, Yamamoto M and Sugimoto T: Adiponectin and AMP kinase activator stimulate proliferation, differentiation, and mineralization of osteoblastic MC3T3-E1 cells. BMC Cell Biol. 8:512007. View Article : Google Scholar : PubMed/NCBI

85 

Xie C and Chen Q: Adipokines: New therapeutic target for osteoarthritis? Curr Reumatol Rep. 21:712020.

86 

Gamberi Τ, Μagherini F, Modesti A and Fiaschi T: Adiponectin signaling pathways in liver diseases. Biomedicines. 6:522018. View Article : Google Scholar : PubMed/NCBI

87 

Ouchi N, Kihara S, Arita Y, Nishida M, Matsuyama A, Okamoto Y, Ishigami M, Kuriyama H, Kishida K, Nishizawa H, et al: Adipocyte-derived plasma protein, adiponectin, suppresses lipid accumulation and class A scavenger receptor expression in human monocyte-derived macrophages. Circulation. 103:1057–1063. 2001. View Article : Google Scholar : PubMed/NCBI

88 

Matsuda M, Shimomura I, Sata M, Arita Y, Nishida M, Maeda N, Kumada M, Okamoto Y, Nagaretani H, Nishizawa H, et al: Role of adiponectin in preventing vascular stenosis. The missing link of adipo-vascular axis. J Biol Chem. 277:37487–37491. 2002. View Article : Google Scholar : PubMed/NCBI

89 

Kadowaki T and Yamauchi T: Adiponectin and adiponectin receptors. Endocr Rev. 26:439–451. 2005. View Article : Google Scholar : PubMed/NCBI

90 

Zha N, Wu X and Gao P: Adiponectin and its receptors in diabetic kidney disease: Molecular mechanisms and clinical potential. Endocrinol. 158:2022–2034. 2017. View Article : Google Scholar

91 

Alnaggar ARLR, Sayed M, El-Deena KE, Gomma M and Hamed Y: Evaluation of serum adiponectin levels in diabetic nephropathy. Diabetes Metab Syndr. 13:128–131. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Ouedraogo R, Gong Y, Berzins B, Wu X, Mahadev K, Hough K, Chan L, Goldstein BJ and Scalia R: Adiponectin deficiency increases leukocyte-endothelium interactions via up-regulation of endothelial cell adhesion molecules in vivo. J Clin Invest. 117:1718–1761. 2007. View Article : Google Scholar : PubMed/NCBI

93 

Abella V, Scotece M, Conde J, López V, Lazzaro V, Pino J, Gómez-Rein O and Gualillo O: Adipokines, metabolic syndrome and rheumatic diseases. J Immunol Res. 2014:3437462014. View Article : Google Scholar : PubMed/NCBI

94 

Stefan N and Stumvoll M: Adiponectin-its role in metabolism and beyond. Horm Metab Res. 34:469–474. 2002. View Article : Google Scholar : PubMed/NCBI

95 

Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE and Tataranni PA: Hypoadiponectinemia in obesity and type 2 diabetes: Close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab. 86:1930–1935. 2001. View Article : Google Scholar : PubMed/NCBI

96 

Arita Y, Kihara S, Ouchi N, Takahashi M, Maed K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, et al: Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun. 257:79–83. 1999. View Article : Google Scholar : PubMed/NCBI

97 

Scherer PE: Adipose tissue. From lipid storage compartment to endocrine organ. Diabetes. 55:1537–1545. 2006. View Article : Google Scholar : PubMed/NCBI

98 

Trujillo ME and Scherer PE: Adipose tissue-derived factors: Impact on health and disease. Endocr Rev. 27:762–778. 2006. View Article : Google Scholar : PubMed/NCBI

99 

Oh DK, Ciaraldi T and Henry RR: Adiponectin in health and disease. Diabetes Obes Metab. 9:282–289. 2007. View Article : Google Scholar : PubMed/NCBI

100 

Li S, Shin HJ, Ding EL and van Dam RM: Adiponectin levels and risk of type 2 diabetes: A systematic review and meta-analysis. JAMA. 302:179–188. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Li C, Cheng H, Adhikari BK, Wang S, Yang N, Liu W, Sun J and Wang Y: The role of apelin-APJ system in diabetes and obesity. Front Endocrinol (Lausanne). 2022:132022.

102 

Al-Mansoori L, Al-Jaber H, Price MS and Elrayess MA: Role of inflammatory cytokines, growth factors and adipokines in adipogenesis and insulin resistance. Inflammation. 45:31–44. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Vykoukal D and Davies MG: Vascular biology of metabolic syndrome. J Vasc Surg. 54:819–831. 2011. View Article : Google Scholar : PubMed/NCBI

104 

Than A, He HL, Chua SH, Xu D, Sun L, Leow MKS and Chen P: Apelin enhances brown adipogenesis and browning of white adipocytes. J Biol Chem. 290:1469–14691. 2015. View Article : Google Scholar

105 

Yamamoto T, Habata Y, Matsumoto Y, Yasuhara Y, Hashimoto T, Hamajyo H, Anayama H, Fujii R, Fuse H, Shintani Y and Mori M: Apelin-transgenic mice exhibit a resistance against diet-induced obesity by increasing vascular mass and mitochondrial biogenesis in skeletal muscle. Biochim Biophys Acta. 1810:853–862. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Mughal A and O'Rourke ST: Vascular effects on apelin: Mechanisms and therapeutic potential. Pharmacol Ther. 190:139–147. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Yamazaki S, Sekiguchi A, Uchiyama A, Fujiwara C, Inoue Y, Yokoyama Y, Ogino S, Torii R, Hosoi M, Akai R, et al: Apelin/APJ signaling suppresses the pressure ulcer formation in cutaneous ischemia-perfusion injury mouse model. Sci Rep. 10:13492020. View Article : Google Scholar : PubMed/NCBI

108 

Attané C, Foussal C, Gonidec SL, Benani A, Daviaud D, Wanecq E, Guzmán-Ruiz R, Dray C, Bezaire V, Rancoule C, et al: Apelin treatment increases complete fatty acid oxidation, mitochondrial oxidative capacity and biogenesis in muscle of insulin-resistant mice. Diabetes. 61:310–320. 2012. View Article : Google Scholar : PubMed/NCBI

109 

Lay SL, Simard G, Martinez MC and Andriantsitohaina R: Oxidative stress and metabolic pathologies: From an adipocentric point of view. Oxid Med Cell Longev. 2014:9085392014.PubMed/NCBI

110 

Kim S, Kim S, Hwang AR, Choi HC, Lee JY and Woo CH: Apelin-13 inhibits methylglyoxal-induced unfolded protein responses and endothelial dysfuction via regulating AMPK pathway. Int J Mol Sci. 21:40692020. View Article : Google Scholar : PubMed/NCBI

111 

Fibbi B, Marroncini G, Naldi L and Peri A: The Yin and Yang effects of the apelinergic system in oxidative stress. Int J Mol Sci. 24:47452023. View Article : Google Scholar : PubMed/NCBI

112 

Zhang Y, Proenca R, Maffei M, Barone M, Leopold L and Friedman JM: Positional cloning of the mouse obes gene and its human homologue. Nature. 372:425–432. 1994. View Article : Google Scholar : PubMed/NCBI

113 

Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK and Friedman JM: Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 269:543–546. 1995. View Article : Google Scholar : PubMed/NCBI

114 

Caro JF, Sinha MK, Kolaczynski JW, Zhang PL and Considine RV: Leptin: The tale of an obesity gene. Diabetes. 45:1455–1462. 1996. View Article : Google Scholar : PubMed/NCBI

115 

Fantuzzi G and Faggioni R: Leptin in the regulation of immunity, inflammation and haematopoiesis. J Leukoc Biol. 68:437–446. 2000. View Article : Google Scholar : PubMed/NCBI

116 

Münzberg H and Morrison CD: Structure, production and signaling of leptin. Metabolism. 64:13–23. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Trayhurn P, Duncan JS, Hoggard N and Rayner DV: Regulation of leptin production: A dominant role for the sympathetic nervous system? Proc Nutr Soc. 57:413–419. 1998. View Article : Google Scholar : PubMed/NCBI

118 

Dieguez C, Vazquez MJ, Romero A, Lopez M and Nogueiras R: Hypothalamic control of lipid metabolism: Focus on leptin, ghrelin and melanocortins. Neuroendocrinology. 94:1–11. 2011. View Article : Google Scholar : PubMed/NCBI

119 

Morton GJ and Schwartz MW: Leptin and the central nervous system control of glucose metabolism. Physiol Rev. 91:389–411. 2011. View Article : Google Scholar : PubMed/NCBI

120 

Sahu A: Leptin signaling in the hypothalamus: Emphasis on energy homeostasis and leptin resistance. Front Neuroendocrinol. 24:225–253. 2003. View Article : Google Scholar : PubMed/NCBI

121 

Blaszczak AM, Jalilvand A and Hsueh WA: Adipocytes, innate immunity and obesity: A mini-review. Front Immunol. 12:6507682021. View Article : Google Scholar : PubMed/NCBI

122 

Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, Gojobori T and Isenovic ER: Leptin and obesity: Role and clinical implication. Front Endocrinol (Lausanne). 12:5858872021. View Article : Google Scholar : PubMed/NCBI

123 

Fahed G, Aoun L, Zerdan MB, Allam S, Zerdan MB, Bouferraa Y and Assi HI: Metabolic syndrome: Updates on pathophysiology and management in 2021. Int J Mol Sci. 23:7862022. View Article : Google Scholar : PubMed/NCBI

124 

van den Hoek AM, Teusink B, Voshol PJ, Havekes LM, Romijn JA and Piji H: Leptin deficiency per se dictates body composition and insulin action in ob/ob mice. J Neuroendocrinol. 20:120–127. 2008. View Article : Google Scholar : PubMed/NCBI

125 

Zhang H, Xie H, Zhao Q, Xie GQ, Wu XP, Liao EY and Luo XH: Relationships between serum adiponectin, apelin, leptin, resistin, visfatin levels and bone mineral density, and bone biochemical markers in post-menopausal Chinese women. J Endocrinol Invest. 33:707–711. 2010. View Article : Google Scholar : PubMed/NCBI

126 

Aizawa-Abe M, Ogawa Y, Masuzaki H, Ebihara K, Satoh N, Iwai H, Matsuoka N, Hayashi T, Hosoda K, Inoue G, et al: Pathophysiological role of leptin in obesity-related hypertension. J Clin Investig. 105:1243–1252. 2000. View Article : Google Scholar : PubMed/NCBI

127 

Ferri C, Desideri G, Valenti M, Bellini C, Pasin M, Santucci A and De Mattia G: Early up-regulation of endothelial adhesion molecules in obese hypertensive men. Hypertension. 34:568–573. 1999. View Article : Google Scholar : PubMed/NCBI

128 

Hukshorn CJ, Lindeman JH, Toet KH, Saris WH, Eilers PH, Westerterp-Plantenga MS and Kooistra T: Leptin and the proinflammatory state associated with human obesity. J Clin Endocrinol Metab. 89:1773–1778. 2004. View Article : Google Scholar : PubMed/NCBI

129 

Kim JE, Kim JS, Jo MJ, Cho E, Ahn SY, Kwon YJ and Ko GJ: The roles and associated mechanisms of adipokines in development of metabolic syndrome. Molecules. 27:3342022. View Article : Google Scholar : PubMed/NCBI

130 

Romacho T, Valencia I, Ramos-González MR, Vallejo S, López-Esteban M, Lorenzo O, Cannata P, Romero A, Hipólito-Luengo AS, Gómez-Cerezo JF, et al: Visfatin/eNampt induces endothelial dysfunction in vivo: A role for toll-like receptor 4 and NLRP3 inflammasome. Sci Rep. 10:53862020. View Article : Google Scholar : PubMed/NCBI

131 

Toussirot E: Mini review: The contribution of adipokines to joint inflammation in inflammatory rheumatic diseases. Front Endocrinol (Lausanne). 11:6065602020. View Article : Google Scholar : PubMed/NCBI

132 

Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Silva C, Rotellar F, Cienfuegos JA, Salvador J and Frühbeck G: Association of increased visfatin/PBEF/NAMPT circulating concentrations and gene expression levels in peripheral blood cells with lipid metabolism and fatty liver in human morbid obesity. Nutr Metab Cardiovasc Dis. 21:245–253. 2011.PubMed/NCBI

133 

Chang YH, Chang DM, Lin KC, Shin SJ and Lee YJ: Visfatin in overweight/obesity, type 2 diabetes mellitus, insulin resistance, metabolic syndrome and cardiovascular diseases: A meta-analysis and systemic review. Diabetes Metab Res Rev. 27:515–527. 2011. View Article : Google Scholar : PubMed/NCBI

134 

Martos-Moreno GA, Kratzch J, Korner A, Barrios V, Hawkins F, Kiess W and Argente J: Serum visfatin and vispin levels in prepubertal childres: Effect of obesity and weitht loss after beharior modifications on their secretion and relationship with glucose metabolism. Int J Obes (Lond). 35:1355–1362. 2011. View Article : Google Scholar : PubMed/NCBI

135 

Olszanecka-Glinianowicz M, Kocełak P, Nylec M, Chudek J and Zahorska-Markiewicz B: Circulating visfatin level and visfatin/insulin ration in obese women with metabolic syndrome. Arch Med Sci. 8:214–218. 2012. View Article : Google Scholar : PubMed/NCBI

136 

de Luis DA, Aller R, Sagrado MG, Conde R, Izaola O and de la Fuente B: Serum visfatin levels and metabolic syndrome criteria in obese female subjects. Diabetes Metab Res Rev. 29:576–581. 2013. View Article : Google Scholar : PubMed/NCBI

137 

Friebe D, Neef M, Kratzch J, Erbs S, Dittrich K, Garten A, Petzold-Qunque S, Blüher S, Reinehr T, Stumvoll M, et al: Leucocytes are a major source of circulating nicotinamide phorsphoribosyltransferase (NAMPT)/pre-B cell colony (PBEF)/visfatin linking obesity and inflammation in humans. Diabetologia. 54:1200–1211. 2011. View Article : Google Scholar : PubMed/NCBI

138 

Kim SR, Bae YH, Bae SK, Choi KS, Yoon KH, Koo TH, Jang HO, Yun Il, Kim KW, Kwon YG, et al: Visfatin enhances ICAM-1 and VCAM-1 expression through ROS-dependent NF-κΒ activation in endothelial cells. Biochim Biophys Acta. 1783:886–895. 2008. View Article : Google Scholar : PubMed/NCBI

139 

Patel SD, Rajala MW, Rossetti L, Scherer PE and Shapiro L: Disulfide-dependent multimeric assembly of resistin family hormones. Science. 304:1154–1158. 2004. View Article : Google Scholar : PubMed/NCBI

140 

Oki K, Yamane K, Kamei N, Nojima H and Kohno N: Circulatin visfatin level is correlated with inflammation, but not with insulin resistance. Clin Endocrinol (Oxf). 67:796–800. 2007. View Article : Google Scholar : PubMed/NCBI

141 

Moschen AR, Kaser A, Enrich B, Mosheimer B, Theurl M, Niederegger H and Tigl H: Visfatin an adipocytokine with proinflammatory and immunomodulating properties. J Immunol. 178:1748–1758. 2007. View Article : Google Scholar : PubMed/NCBI

142 

Krysiak R, Handzlik-Orlik G and Okopien B: The role of adipokines in connective tissue diseases. Eur J Nutr. 51:513–528. 2012. View Article : Google Scholar : PubMed/NCBI

143 

Heo YJ, Choi SE, Jeon JY, Han SJ, Kim DJ, Kang Y, Lee KW and Kim HJ: Visfatin induces inflammation and insulin resistance via the NF-κΒ and STAT3 signaling pathways in hepatocytes. J Diabet Res. 2019:40216232019. View Article : Google Scholar : PubMed/NCBI

144 

Francisco V, Sanz MJ, Real JT, Marques P, Capuozzo M, Eldjoudi DA and Gualillo O: Adipokines in non-alcoholic fatty liver disease: Are we on the road toward new biomarkers and therapeutic targets? Biology (Basel). 11:12372022.PubMed/NCBI

145 

Oita RC, Ferdinando D, Wilson S, Bunce C and Mazzatti DJ: Visfatin induces oxidative stress in differentiated C2C12 myotubes in an Akt- and MAPK-independent, NFκΒ-dependent manner. Pflugers Arch. 459:619–630. 2010. View Article : Google Scholar : PubMed/NCBI

146 

Lee S, Lee HC, Kwon YW, Lee SE, Cho Y, Kim J, Lee S, Kim JY, Lee J, Yang HM, et al: Adenylyl cyclase-associated protein 1 (CAP1) is a receptor for human resistin and mediated inflammatory actions of human monocytes. Cell Metab. 19:484–497. 2014. View Article : Google Scholar : PubMed/NCBI

147 

Li Y, Yang Q, Cai D, Guo H, Fang J, Cui H, Gou L, Deng J, Wang Z and Zuo Z: Resistin, a novel host defence peptide of innate immunity. Front Immunol. 12:6998072021. View Article : Google Scholar : PubMed/NCBI

148 

Kawanami D, Maemura K, Takeda N, Harada T, Nojiri T, Imai Y, Manabe I, Utsunomiya K and Nagai R: Direct reciprocal effects of resistin and adiponectin on vascular endothelial cells: A new insight into adipocytokine-endothelial cell interactions. Biochem Biophys Res Commun. 314:415–419. 2004. View Article : Google Scholar : PubMed/NCBI

149 

Bokarewa M, Nagaev I, Dahlberg L, Smith U and Tarkowski A: Resistin, an adipokine with potent proimflammatory properties. J Immunol. 174:5789–5795. 2005. View Article : Google Scholar : PubMed/NCBI

150 

Agaev I, Bokarewa M, Tarkowski A and Smith U: Human resistin is a systemic immune-derived proinflammatory cytokine targeting both leukocytes and adipocytes. PLoS One. 1:e312006. View Article : Google Scholar : PubMed/NCBI

151 

Ebihara T, Matsumoto H, Matsubara T, Matsuura H, Hirose T, Shimizu K, Ogura H, Kang S, Tanaka T and Shimazu T: Adipocytokine profile reveals resistin forming a prognostic-related cytokine network in the acute phase of sepsis. Shock. 56:718–726. 2021. View Article : Google Scholar : PubMed/NCBI

152 

Heilbronn LK, Rood J, Janderova L, Albu JB, Kelley DE, Ravussin E and Smith SR: Relationship between serum resistin concentrations and insulin resistance in nonobese, obese, and obese diabetic subjects. J Clin Endocrinol Metabol. 89:1844–1848. 2004. View Article : Google Scholar : PubMed/NCBI

153 

Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS and Lazar MA: The hormone resistin links obesity to diabetes. Nature. 409:307–312. 2001. View Article : Google Scholar : PubMed/NCBI

154 

Vidal-Puig A and O'Rahilly S: Resistin: A new link between obesity and insulin resistance? Clin Endocrinol (Oxf). 55:437–438. 2001. View Article : Google Scholar : PubMed/NCBI

155 

McTernan CL, McTernan PG, Harte AL, Levick PL, Barnet AH and Kumar S: Resistin, central obesity, and type 2 diabetes. Lancet. 359:46–47. 2002. View Article : Google Scholar : PubMed/NCBI

156 

Wang H, Chu WS, Hemphill C and Elbein SC: Hunan resistin gene: Molecular scanning and evaluation of association with insulin sensitivity and type 2 diabetes in Caucasians. J Clin Endocrinol Metabol. 87:2520–2524. 2002. View Article : Google Scholar : PubMed/NCBI

157 

Osawa H, Yamada K, Onuma H, Murakami A, Ochi M, Kawata H, Nishimiya T, Niiya T, Shimizu I, Nishida W, et al: The G/G genotype of resistin single-nucleotide polymorphism at −420 increases type 2 diabetes mellitus susceptibility by inducing promoter activity through specific binding of Sp1/3. Am J Hum Genet. 75:678–686. 2004. View Article : Google Scholar : PubMed/NCBI

158 

Kielstein JT, Becker B, Graf S, Brabant G, Haller H and Fliser D: Increased resistin blood levels are not associated with insulin resistance in patients with renal disease. Am J Kidney Dis. 42:62–66. 2003. View Article : Google Scholar : PubMed/NCBI

159 

Patel L, Buckels AC, Kinghorn IJ, Mourdock PR, Holbrook JD, Plumpton C, Macphee CH and Smith SA: Resistin is expressed in human macrophages and directly regulated by PPAR gamma activators. Biochem Biophys Res Commun. 300:472–476. 2003. View Article : Google Scholar : PubMed/NCBI

160 

Chen C, Jiang J, Lü JM, Chai H, Wang X, Lin PH and Yao Q: Resistin descreases expression of endothelial nitric oxide synthase through oxidative stress in human coronary artery endothelial cells. Am J Physiol Heart Circ Physiol. 299:H193–H201. 2010. View Article : Google Scholar : PubMed/NCBI

161 

Acquarone E, Monacelli F, Borghi R, Nencioni A and Odetti P: Resistin: A reappraisal. Mech Ageing Dev. 178:46–63. 2019. View Article : Google Scholar : PubMed/NCBI

162 

Iwaki T, Urano T and Umemura K: PAI-1, progress in understanding the clinical problem and its aetiology. Br J Heamatol. 157:291–298. 2012. View Article : Google Scholar

163 

Para I, Albu A and Porojan MD: Adipokines and arterial stiffness in obesity. Medicina (Kaunas). 57:6532021. View Article : Google Scholar : PubMed/NCBI

164 

Mertens I and Van Gaal LF: Obesity, haemostasis and the fibrinolytic system. Obes Rev. 3:85–101. 2002. View Article : Google Scholar : PubMed/NCBI

165 

Juhan-Vague I, Alessi MC, Mavri A and Morange PE: Plasminogen activator inhibitor-1, inflammation, obesity, insulin resistance and vascular risk. J Thromb Haemost. 1:1575–1579. 2003. View Article : Google Scholar : PubMed/NCBI

166 

Tschoner A, Sturm W, Engl J, Kaser S, Laimer M, Laimer E, Klaus A, Patsch JR and Ebenbichler CF: Plasminogen activator inhibitor 1 and visceral obesity during pronounced weight loss after bariatric surgery. Nutr Metab Cardiovasc Dis. 22:340–346. 2012. View Article : Google Scholar : PubMed/NCBI

167 

Khoukaz HB, Ji Y, Braet DJ, Vadali M, Abdelhamid AA, Emal CD, Lawrence DA and Fay WP: Drug targeting of plasminogen activator inhibitor-1 inhibits metabolic dysfunction and atherosclerosis in murine model of metabolic syndrome. Arterioscler Thromb Vasc Biol. 40:1479–1490. 2020. View Article : Google Scholar : PubMed/NCBI

168 

Eitzman DT, Westrick RJ, Xu Z, Tyson J and Grinsburg D: Plasminogen activator inhibitor-1 deficiency protects against atherosclerosis progression in the mouse carotid artery. Blood. 96:4212–4215. 2000. View Article : Google Scholar : PubMed/NCBI

169 

Alessi MC and Juhan-Vague I: PAI-1 and the metabolic syndrome: Links, causes and consequences. Arterioscler Thromb Vasc Biol. 16:2200–2207. 2006. View Article : Google Scholar : PubMed/NCBI

170 

Ma LJ, Mao SL, Taylor KL, Kanjanabuch T, Guan Y, Zhang Y, Brown NJ, Swift LL, McGuinness OP, Wasserman DH, et al: Prevention of obesity and insulin resistance in mice lacking plasminogen activator inhibitor 1. Diabetes. 53:336–346. 2004. View Article : Google Scholar : PubMed/NCBI

171 

Gleeson LE, Sheedy FJ, Palsson-McDermott EM, Triglia D, O'Leary SM, O'Sullivan MP, O'Neill LA and Keane J: Cytting edge: Mycobacterium tuberculosis induces aerobic glycolysis in human alveolar macrophages that is required for control of intracellular bacillary replication. J Immunol. 196:2444–2449. 2016. View Article : Google Scholar : PubMed/NCBI

172 

Shoelson SE, Herrero L and Naaz A: Obesity, inflammation and insulin resistance. Gastroenterology. 132:2169–2180. 2007. View Article : Google Scholar : PubMed/NCBI

173 

Stojsavljević S, Palčić MG, Jukić LV, Duvnjak LS and Duvnjak M: Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World J Gastroenterol. 20:18070–18091. 2014. View Article : Google Scholar : PubMed/NCBI

174 

Plomgaard P, Bouzakri K, Krogh-Madsen R, Mittendorfer B, Zierath JR and Pedersen BK: Tumor necrosis factor-alpha induces skeletal muscle insulin resistance in healthy human subjects via inhibition of Akt substrate 160 phosphorylation. Diabetes. 54:2936–2945. 2005. View Article : Google Scholar

175 

Ruan H and Lodish HF: Insulin resistance in adipose tissue: Direct and indirect effects of tumor necrosis factor-α. Cytokine Growth Factor Rev. 14:447–455. 2003. View Article : Google Scholar : PubMed/NCBI

176 

Illei GG and Lipsky PE: Novel, antigen-specific therapeutic approaches to autoimmuneinflammatory diseses. Curr Opin Immunol. 12:712–718. 2000. View Article : Google Scholar : PubMed/NCBI

177 

Chandel NS, Schumacker PT and Arch RH: Reactive oxygen species are downstream products of TRAF-mediated signal trasduction. J Biol Chem. 276:42728–42736. 2001. View Article : Google Scholar : PubMed/NCBI

178 

Micheau O and Tschopp J: Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 114:181–190. 2003. View Article : Google Scholar : PubMed/NCBI

179 

Campbell J, Ciesielski CJ, Hunt AE, Horwood NJ, Beech JT, Hayes LA, Denys A, Feldmann M, Brennan FM and Foxwell BMJ: A novel mechanism for TNF-alpha regulation by p38 MAPK: Involvement of NF-kappa B with implications for therapy in rheumatoid arthritis. J Immunol. 173:6928–6937. 2004. View Article : Google Scholar : PubMed/NCBI

180 

Wang B and Trayhurn P: Acute and prolonged effects of TNF-alpha on the expression and secretion of inflammation-related adipokines by human adipocytes differentiated in culture. Pflugers Arch. 452:418–427. 2006. View Article : Google Scholar : PubMed/NCBI

181 

Rider P, Carmi Y, Guttman O, Braiman A, Cohen I, Voronov E, White MR, Dinarello CA and Apte RN: IL-1α and IL-1β recruit different myeloid cells and promote different stages of sterile inflammation. J Immunol. 187:4835–4843. 2011. View Article : Google Scholar : PubMed/NCBI

182 

Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR and Aunins J: A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 356:768–774. 1992. View Article : Google Scholar : PubMed/NCBI

183 

Akdis M, Arab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, Duan S, Eiwegger T, Eljaszewicz A, Ferstl R, et al: Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J Allergy Clin Immunol. 138:984–1010. 2016. View Article : Google Scholar : PubMed/NCBI

184 

Ghabari M, Maragheh SM, Aghazadeh A, Mehrjuyan SR, Hussen BM, Shadbad MA, Dastmalchi N and Safaralizadeh R: Interleukin-1 in obesity-related low-grade inflammation: From molecular mechanisms to therapeutic strategies. Int Immunopharmacol. 96:1077652021. View Article : Google Scholar : PubMed/NCBI

185 

Speaker KJ and Fleshner M: Interleukin-1 beta: A potential link between stress and the development of visceral obesity. BMC Physiol. 12:1–15. 2012. View Article : Google Scholar : PubMed/NCBI

186 

Bruun JM, Pedersen SB, Kristensen K and Richelsen B: Effects of pro-inflammatory cytokines and chemokines on leptin production in human adipose tissue in vitro. Mol Cell Endocrinol. 190:91–99. 2002. View Article : Google Scholar : PubMed/NCBI

187 

Gonzalez RR and Leavis P: Leptin upregulates β3-integrin expression and interleukin-1β upregulates leptin and leptin receptor expression in human endometrial epithelial cell cultures. Endocrine. 16:21–28. 2021. View Article : Google Scholar

188 

Müller G, Ertl J, Gerl M and Preidisch G: Leptin impairs metabolic actions of insulin in isolated rat adipocytes. J Biol Chem. 272:10585–10593. 1997. View Article : Google Scholar : PubMed/NCBI

189 

Moschen AR, Molnar C, Enrich B, Geiger S, Ebenbichler CF and Tilg H: Adipose and liver expression of interleukin (IL)-1 family members in morbid obesity and effects of weight loss. Mol Med. 17:840–845. 2011. View Article : Google Scholar : PubMed/NCBI

190 

Shaul ME, Bennett G, Strissel KJ, Greenberg AS and Obin MS: Dynamic, M2-like remodeling phenotypes of CD11c+ adipose tissue macrophages during high-fat diet-induced obesity in mice. Diabetes. 59:1171–1181. 2010. View Article : Google Scholar : PubMed/NCBI

191 

Schoettl T, Fischer IP and Ussar S: Heterogeneity of adipose tissue in development and metabolic function. J Exp Biol. 221 (Pt Suppl 1):jeb1629582018. View Article : Google Scholar : PubMed/NCBI

192 

Buechler C, Krautbauer S and Eisinger K: Adipose tissue fibrosis. World J Diabetes. 6:548–553. 2015. View Article : Google Scholar : PubMed/NCBI

193 

Shikama Y, Aki N, Hata A, Nishimura M, Oyadomari S and Funaki M: Palmitate-stimulated monocytes induce adhesion molecule expression in endothelial cells via IL-1 signaling pathway. J Cell Physiol. 230:732–742. 2015. View Article : Google Scholar : PubMed/NCBI

194 

Miura K, Kodama Y, Inokuchi S, Scnabl B, Aoyama T, Ohnishi H, Olefsky JM, Brenner DA and Seki E: Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology. 139:323–334. 2010. View Article : Google Scholar : PubMed/NCBI

195 

Gao D, Madi M, Ding C, Fok M, Steele T, Ford C, Hunter L and Bing C: Interleukin-1β mediates macrophage-induced impairemnet of insulin signalin in human primary adipocytes. Am J Physiol Endocrinol Metab. 307:E289–E304. 2014. View Article : Google Scholar : PubMed/NCBI

196 

Zhou R, Tardivel A, Thorens B, Choi I and Tschopp J: Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 11:136–140. 2010. View Article : Google Scholar : PubMed/NCBI

197 

Calabrese L, Fiocco Z, Satoh TK, Peris K and French LE: Therapeutic potential of targeting interleukin-1 family cytokines in chronic inflammatory skin diseases. Br J Dermatol. 186:925–941. 2022. View Article : Google Scholar : PubMed/NCBI

198 

Maedler K, Sergeev P, Ris F, Oberholzer J, Holler-Jemelka H, Spinas GA, Kaiser N, Halban PA and Donath MY: Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest. 110:851–860. 2002. View Article : Google Scholar : PubMed/NCBI

199 

Wang Q, Zhang H, Zhao B and Fei H: IL-1beta caused pancreatic beta-cells apoptosis is mediated in part by endoplasmic reticulum stress via the induction of endoplasmic reticulum Ca2+ release through the c-Jun N-terminal kinase pathway. Mol Cell Biochem. 324:183–190. 2009. View Article : Google Scholar : PubMed/NCBI

200 

Brichory FM, Misek DE, Yim AM, Krause MC, Giordano TJ, Beer DG and Hanash SM: An immune response manifested by the common occurrence of annexins I and II aytoantibodies and high circulating levels of IL-6 in lung cancer. Proc Natl Acad Sci USA. 98:9824–9829. 2001. View Article : Google Scholar : PubMed/NCBI

201 

Scheller J, Chalaris A, Schmidt-Arras D and Rose-John S: The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 1813:878–888. 2011. View Article : Google Scholar : PubMed/NCBI

202 

Tanaka T, Narazaki M and Kishimoto T: IL-6 in inflammation, immunity and disease. Cold Spring Harb Perspect Biol. 6:a0162952014. View Article : Google Scholar : PubMed/NCBI

203 

IL6R Genetics Consortium Emerging Risk Factors and Collaboration, . Sarwar N, Butterworth AS, Freitag DF, Gregson J, Willeit P, Gorman DN, Gao P, Saleheen D, Rendon A, et al: Interleukin-6 receptor pathways in coronary heart disease: A collaborative meta-analysis of 82 studies. Lancet. 379:1205–1213. 2012. View Article : Google Scholar : PubMed/NCBI

204 

Elhage R, Clamens S, Besnard S, Mallat Z, Tedgui A, Arnal J, Maret A and Bayard F: Involvement of interleukin-6 in atherosclerosis but not in the prevention of fatty streak formation by 17beta-estradiol in apolipoprotein E-deficient mice. Atherosclerosis. 156:315–320. 2001. View Article : Google Scholar : PubMed/NCBI

205 

Schieffer B, Selle T, Hilfiker A, Hilfiker-Kleiner D, Grote K, Tietge UJF, Trautwein C, Luchtefeld M, Schmittkamp C, Heeneman S, et al: Impact of interleukin-6 on plaque development and morphology in experimental atherosclerosis. Circulation. 110:3493–3500. 2004. View Article : Google Scholar : PubMed/NCBI

206 

Mishra D, Richard JE, Maric I, Porteiro B, Häring M, Kooijman S, Musovic S, Eerola K, López-Ferreras L, Peris E, et al: Parabrachial interleukin-6 reduces body weight and food intake and increases thermogenesis to regulate energy metabolism. Cell Rep. 26:3011–3026. 2019. View Article : Google Scholar : PubMed/NCBI

207 

Rosen ED and Spiegelman BM: What we talk about when we talk about fat. Cell. 156:20–44. 2014. View Article : Google Scholar : PubMed/NCBI

208 

Mohamed-Ali V, Goodrick S, Rawesh A, Katz DR, Miles JM, Yudkin JS, Klein S and Coppack SW: Subcutaneous adipose tissue related interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin Endocrinol Metab. 82:4196–4200. 1997. View Article : Google Scholar : PubMed/NCBI

209 

Sopasakis VR, Sandqvist M, Gustafson B, Hammerstedt A, Schmelz M, Yang X, Jansson PA and Smith U: High local concentrations and effects on differentiation implicate interleukin-6 as a paracrine regulator. Obes Res. 12:454–460. 2004. View Article : Google Scholar : PubMed/NCBI

210 

Fernandez-Real JM and Ricart W: Insulin resistance and chronic cardiovascular inflammatory sundrome. Endocr Rev. 24:278–301. 2003. View Article : Google Scholar : PubMed/NCBI

211 

Charles BA, Doumatey A, Huang H, Zhou J, Chen G, Shriner D, Adeyemo A and Rotimi CN: The roles of IL-6, IL-10 and IL-1RA in obesity and insulin resistance in African-Americans. J Clin Endocrinol Metab. 96:E2018–E2022. 2011. View Article : Google Scholar : PubMed/NCBI

212 

Tsigos C, Papanicolaou DA, Kyrou I, Defensor R, Mitsiadis CS and Chrousos GP: Dose-dependent effects of recombinant human interleukin-6 on glucose regulation. J Clin Endocrinol Metab. 82:4167–4170. 1997. View Article : Google Scholar : PubMed/NCBI

213 

Pradhan AD, Manson JE, Rifai N, Buring JE and Ridker PM: C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 286:327–334. 2001. View Article : Google Scholar : PubMed/NCBI

214 

Bastard JP, Maachi M, van Nhieu JT, Jardel C, Bruckert E, Grimaldi A, Robert JJ, Capeau J and Hainque B: Adipose tissue IL-6 content correlates with resistance to insulin activation of glucose uptake both in vivo and in vitro. J Clin Endocrinol Metab. 87:2084–2089. 2002. View Article : Google Scholar : PubMed/NCBI

215 

Kopp HP, Kopp CW, Festa A, Krzyzanowaska K, Kriwanek S, Minar E, Roka R and Schernthaner G: Impact of weight loss on inflammatory proteins and their association with the insulin resistance syndrome in morbidly obese patients. Arterioscler Throm Vasc Biol. 23:1042–1047. 2003. View Article : Google Scholar : PubMed/NCBI

216 

Xu E, Pereira MMA, Karakasilioti I, Theurich S, Al-Maarri M, Rappl G, Waisman A, Wenderlich FT and Brüning JC: Temporal and tissue-specific requirements for T-lympocyte IL-6 signaling in obesity-associated inflammation and insulin resistance. Nat Commun. 8:148032017. View Article : Google Scholar : PubMed/NCBI

217 

Wondmkum YT: Obesity, insulin resistance, and type 2 diabetes: Associations and therapeutic implications. Diabetes Metab Syndr Obes. 13:3611–3616. 2020. View Article : Google Scholar : PubMed/NCBI

218 

Fu Z, Gilbert ER and Liu D: Regulation of insulin synthesis and secretion and pancreatic Beta-cell dysfunction in diabetes. Curr Diabetes Rev. 9:25–53. 2013. View Article : Google Scholar : PubMed/NCBI

219 

Röder PV, Wu B, Liu Y and Han W: Pancreatic regulation of glucose homeostasis. Exp Mol Med. 48:e2192016. View Article : Google Scholar : PubMed/NCBI

220 

Fazakerley DJ, Krycer JR, Kearney AL, Hocking SL and James DE: Muscle and adipose tissue insulin resistance: Malady without mechanism? J Lipid Res. 60:1720–1732. 2019. View Article : Google Scholar : PubMed/NCBI

221 

Newsholme P and Krause M: Nutritional regulation of insulin secretion: Implications for diabetes. Clin Biochem Rev. 33:35–47. 2012.PubMed/NCBI

222 

Dashty M: A quick look at biochemistry: Carbohydrate metabolism. Clin Biochem. 46:1339–1352. 2013. View Article : Google Scholar : PubMed/NCBI

223 

Samuel VT and Shulman GI: The pathogenesis of insulin resistance: Intergrating signaling pathways and substrate flux. J Clin Invest. 126:12–22. 2016. View Article : Google Scholar : PubMed/NCBI

224 

Taniguchi CM, Emanuelli B and Kahn CR: Critical nodes in signaling pathways: Insights into insulin action. Nat Rev Mol Cell Biol. 7:85–96. 2006. View Article : Google Scholar : PubMed/NCBI

225 

Merry TL, Hedges CP, Masson SW, Laube B, Pöhlmann D, Wueest S, Walsh ME, Arnold M, Langhans W, Konrad D, et al: Partial impairment of insulin receptor expression mimics fasting to prevent diet-induced fatty liver diseasee. Nat Commun. 11:20802020. View Article : Google Scholar : PubMed/NCBI

226 

Shanik MH, Xu Y, Skrha J, Dankner R, Zick Y and Roth J: Insulin resistance and hyperinsulinemia: Is hyperinsulinemia the cart or the horse? Diabetes Car. 31 (Suppl 2):S262–S268. 2008. View Article : Google Scholar : PubMed/NCBI

227 

Braccini L, Ciraolo E, Campa CC, Perino A, Longo DL, Tibolla G, Pregnolato M, Cao Y, Tassone B, Damilano F, et al: PI3K-C2γ is a Rab5 effector selectively controlling endosomal Akt2 activation downstream of insulin signalling. Nat Commun. 6:74002015. View Article : Google Scholar : PubMed/NCBI

228 

Huang X, Liu G, Guo J and Su Z: The PI3K/AKT pathway in obesity and type 2 diabetes. Int J Biol Sci. 14:1483–1496. 2018. View Article : Google Scholar : PubMed/NCBI

229 

Gray SL, Donald C, Jetha A, Covey SD and Kieffer TJ: Hyperinsulinemia precedes insulin resistance in mice lacking pancreatic beta-cell leptin signaling. Endocrinology. 151:4178–4186. 2010. View Article : Google Scholar : PubMed/NCBI

230 

Wild S, Roglic G, Green A, Sicree R and King H: Global prevalence of diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care. 27:1047–1053. 2004. View Article : Google Scholar : PubMed/NCBI

231 

Smith BW and Adams LA: Nonalcoholic faty liver disease and diabetes mellitus: Pathogenesis and treatment. Nat Rev Endocrinol. 7:456–465. 2011. View Article : Google Scholar : PubMed/NCBI

232 

Willians AJK and Long AE: Following the fate of the failing β-cell: New insights from first-phase insulin responses. Diabetes. 62:3990–3992. 2013. View Article : Google Scholar

233 

Gariani K, Philippe J and Jornayvaz FR: Non-alcoholic fatty liver disease and insulin resistance: From bench to bedside. Diabetes Metab. 39:16–26. 2013. View Article : Google Scholar : PubMed/NCBI

234 

Zhao J, Wu Y, Rong X, Zheng C and Guo J: Anti-lipolysis induced by insulin in diverse pathophysiologic conditions of adipose tissue. Diabetes Metab Syndr Obes. 13:1575–1585. 2020. View Article : Google Scholar : PubMed/NCBI

235 

Griffin ME, Marcucci MJ, Cline GW, Bell K, Barucci N, Lee D, Goodyear LJ, Kraegen EW, White MF and Shulman GI: Free fatty acid-induced insulin resistance is associated with activation of protein kinase C theta and alterations in the insulin signaling cascade. Diabetes. 48:1270–1274. 1999. View Article : Google Scholar : PubMed/NCBI

236 

Boden G and Shulman GI: Free fatty acids in obesity and type 2 diabetes: Defining their role in the development of insulin resistance and beta-cell dysfunction. Eur J Clin Invest. 32:14–23. 2002. View Article : Google Scholar : PubMed/NCBI

237 

Unger RH and Zhou YT: Lipotoxicity of beta-cells in obesity and in other causes of fatty acid spillover. Diabetes. 50 (Suppl 1):S118–S121. 2001. View Article : Google Scholar : PubMed/NCBI

238 

Bugianesi E, Gastaldelli A, Vanni E, Gambino R, Cassader M, Baldi S, Ponti V, Pagano G, Ferranini E and Rizzetto M: Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: Sites and mechanisms. Diabeltologia. 48:634–642. 2005. View Article : Google Scholar

239 

Perry RJ, Samuel VT, Petersen KF and Shulman GI: The role of hepatic insulin resistance and type 2 diabetes. Nature. 510:84–91. 2014. View Article : Google Scholar : PubMed/NCBI

240 

Brown MS and Goldstein JL: Selective versus total insulin resistance: A pathogenic paradox. Cell Metab. 7:95–96. 2008. View Article : Google Scholar : PubMed/NCBI

241 

Kim GT, Kim SJ, Park SH, Lee D and Park TS: Hepatic expression of the serine palmitoyltansferase subunit Sptlc2 reduces lipid droplets in the liver by activating VLDL secretion. J Lipid Atheroscler. 9:291–303. 2020. View Article : Google Scholar : PubMed/NCBI

242 

Hall JE, da Silva AA, do Carmo JM, Dubinion J, Hamza S, Munusamy S, Smith G and Stec DE: Obesity-induced hypertension: Role of sympathetic nervous system, leptin, and melanocortins. J Biol Chem. 285:17271–17276. 2010. View Article : Google Scholar : PubMed/NCBI

243 

Weickert MO and Pfeiffer AFH: Signalling mechanisms linking hepatic glucose and lipid metabolism. Diabetologia. 4:1732–1741. 2006. View Article : Google Scholar : PubMed/NCBI

244 

Murakami T, Michelagnoli S, Longhi R, Gianfranceschi G, Pazzucconi F, Calabresi L, Sirtori CR and Franceschini G: Triglycerides are major determinants of cholesterol esterification/transfer and HDL remodeling in human plasma. Arterioscler Thromb Vasc Biol. 15:1819–1828. 1995. View Article : Google Scholar : PubMed/NCBI

245 

Eisenberg S, Gavish D, Oschry Y, Fainaru M and Deckelbaum RJ: Abnormalities in very low, low and high density lipoproteins in hypertriglyceridemia. Reversal toward normal with bezafibrate treatment. J Clin Invest. 74:470–482. 1984. View Article : Google Scholar : PubMed/NCBI

246 

Tripathy D, Mohanty P, Dhindsa S, Syed T, Ghanim H, Aljada A and Dandona P: Elevation of free fatty acids induces inflammation and impairs vascular reactivity in healthy subjects. Diabetes. 52:2882–2887. 2003. View Article : Google Scholar : PubMed/NCBI

247 

Esler M, Rumantir M, Wiesner G, Kaye D, Hastings J and Lambert G: Sympathetic nervous system and insulin resistance from obesity to diabetes. Am J Hypertens. 14((11 Pt 2)): 304S–309S. 2001. View Article : Google Scholar : PubMed/NCBI

248 

Samad F and Ruf W: Inflammation, obesity and thrombosis. Blood. 122:3415–3422. 2013. View Article : Google Scholar : PubMed/NCBI

249 

Ernst E and Resch KL: Fibrinogen as a cardiovascular risk factor: A meta-analysis and review of the literature. Ann Intern Med. 118:956–963. 1993. View Article : Google Scholar : PubMed/NCBI

250 

Kannel WB, Wolf PA, Castelli WP and D'Agostino RB: Fibrionogen and risk of cardiovascular disease. The Framingham study. JAMA. 258:1183–1186. 1987. View Article : Google Scholar : PubMed/NCBI

251 

Nieuwdorp M, Stroes ES, Meijers JC and Buller H: Hypercoagulability in the metabolic syndrome. Curr Opin Pharmacol. 5:155–159. 2005. View Article : Google Scholar : PubMed/NCBI

252 

Raynaud E, Perez-Martin A, Brun JF, Aïssa-Benhaddad A, Fédou C and Mercier J: Atherosclerosis. 150:365–370. 2000. View Article : Google Scholar : PubMed/NCBI

253 

Tabrez S, Jabir NR, Shakil S and Alama MN: Association of plasma fibrinogen level with insulin resistance in angiographically confirmed coronary artery disease patients. Crit Rev Eukaryot Gene Expr. 29:277–285. 2019. View Article : Google Scholar : PubMed/NCBI

254 

Bryk-Wiązania AH and Undas A: Hypofribrinolysis in type 2 diabetes and its clinical implications: From mechanisms to pharmacological modulation. Cardiovasc Diabetol. 20:1912021. View Article : Google Scholar : PubMed/NCBI

255 

Davalos D and Akassoglou K: Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol. 34:43–62. 2012. View Article : Google Scholar : PubMed/NCBI

256 

Nawaz SS and Siddiqui K: Plasminogen activator inhibitor-1 mediated downregulation of adiponectin in type 2 diabetic patients with metabolic syndrome. Cytokine X. 4:1000642002. View Article : Google Scholar : PubMed/NCBI

257 

Chen R, Yan J, Liu P, Wang Z and Wang C: Plasminogen activator inhibitor links obesity and thrombotic cerebrovascular diseases: The roles of PAI-1 and obesity on stroke. Metab Brain Dis. 32:667–673. 2017. View Article : Google Scholar : PubMed/NCBI

258 

Matsuzawa Y: The metabolic syndrome and adipocytokines. FEBS Lett. 580:2917–2921. 2006. View Article : Google Scholar : PubMed/NCBI

259 

Mertens I, Ballaux D, Funahashi T, Matsuzawa Y, Van der Planken M, Verrijken A, Ruge JB and Gaal LFV: Inverse relationship between plasminogen activator inhibitor-I activity and adiponectin in overweight and obese women. Interrelationship with visceral adipose tissue, insulin resistance, HDL-chol and inflammation. Thromb Haemost. 94:1190–1195. 2005. View Article : Google Scholar : PubMed/NCBI

260 

Shimomura I, Funahashi T, Takahashi M, Maeda K, Kotani K, Nakamura T, Yamashita S, Miura N, Fukuda Y, Takemura K, et al: Enhanced expression of PAI-1 in visceral fat: Possible contributor to vascular disease in obesity. Nat Med. 2:800–803. 1996. View Article : Google Scholar : PubMed/NCBI

261 

Kaji H: Adipose tissue-derived plasminogen activator inhibitor-1 function and regulation. Compr Physiol. 6:1873–1896. 2016. View Article : Google Scholar : PubMed/NCBI

262 

Reaven GM: Banting lecture 1988. Role of insulin resistance in human disease. Diabetes. 37:1595–1607. 1988. View Article : Google Scholar : PubMed/NCBI

263 

Alberti KG, Zimmet P and Shaw J; IDF Edipemiology Task Force Consensus Group, : The metabolic syndrome-a new world-wide definition. Lancet. 366:1059–1062. 2005. View Article : Google Scholar : PubMed/NCBI

264 

Zafar U, Khaliq S, Ahmad HU, Manzoor S and Lone KP: Metabolic syndrome: An update on diagnostic criteria, pathogenesis, and genetic links. Hormones (Athens). 17:299–313. 2018. View Article : Google Scholar : PubMed/NCBI

265 

Spiegelman BM and Flier JS: Obesity and the regulation of energy balance. Cell. 104:531–543. 2001. View Article : Google Scholar : PubMed/NCBI

266 

Mottilo S, Filion KB, Genest J, Joseph L, Pilote L, Poirier P, Rinfret S, Schiffrin EL and Eisenberg MJ: The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J Am Coll Cardiol. 56:1113–1132. 2010. View Article : Google Scholar

267 

Weiss R, Bremer AA and Lusting RH: What is metabolic syndrome, and why are children getting it? Ann N Y Acad Sci. 1281:123–140. 2013. View Article : Google Scholar : PubMed/NCBI

268 

Grundy SM: Metabolic syndrome pandemic. Arteroscler Thromb Vasc Biol. 28:629–636. 2008. View Article : Google Scholar : PubMed/NCBI

269 

Ford ES, Li C and Zhao G: Prevalence and correlates of metabolic syndrome based on a harmonious definition among adults in the US. J Diabetes. 2:180–193. 2010. View Article : Google Scholar : PubMed/NCBI

270 

Kahn R, Buse J, Ferrannini E and Stern M: The metabolic syndrome: Time for a critical appraisal. Joint statement from the Americal diabetes association and the European association for the study of diabetes. Diabetologia. 48:1684–1699. 2005. View Article : Google Scholar : PubMed/NCBI

271 

Pi-Sunyer X: The metabolic syndrome: How to approach differing definitions. Med Clin North Am. 91:1025–1040. 2007. View Article : Google Scholar : PubMed/NCBI

272 

Chung G, Jung HS and Kim HJ: Sociodemographic and health characteristics associated with metabolic syndrome in men and women aged ≥50 Years. Metab Sundr Relat Disord. 19:159–166. 2021. View Article : Google Scholar : PubMed/NCBI

273 

Hydrie MZ, Shera AS, Fawwad A, Basit A and Hussain A: Prevalence of metabolic syndrome in urban Pakistan (Karachi): Comparison of newly proposed international diabetes federation and modified adult treatment panel III criteria. Metab Syndr Relat Disord. 7:119–124. 2009. View Article : Google Scholar : PubMed/NCBI

274 

Rosenbaum M, Sy M, Pavlovich K, Leibel RL and Hirsch J: Leptin reverses weight loss-induced changes in regional neural activity responses to visual food stimuli. J Clin Invest. 118:2583–2591. 2008.PubMed/NCBI

275 

Imai SI: Nicotinamide phosphoribosyltrasferase (Nampt): A link between NED biology, metabolism, and disease. Curr Pharm Des. 15:20–28. 2009. View Article : Google Scholar : PubMed/NCBI

276 

Lago F, Dieguez C, Gomez-Reino G and Gulillo O: Adipokines as emerging mediators of immune response and inflammation. Nat Clin Pract Rheumatol. 3:716–724. 2007. View Article : Google Scholar : PubMed/NCBI

277 

Hopps E, Noto D, Caimi G and Averna MR: A novel comoponent of the metabolic syndrome: The oxidative stress. Nutr Metab Cardiovasc Dis. 20:72–77. 2010. View Article : Google Scholar : PubMed/NCBI

278 

Vona R, Gambardella L, Cittadini C, Straface E and Pietraforte D: Biomarkers of oxidative stress in metabolic syndrome and associated dieseases. Oxid Med Cell Longev. 2019:82672342019. View Article : Google Scholar : PubMed/NCBI

279 

Schieber M and Chandel NS: ROS function in redox signaling and oxidative stress. Curr Biol. 24:R453–R462. 2014. View Article : Google Scholar : PubMed/NCBI

280 

Juan CA, de la Lastra JM, Plou FJ and Pérez-Lebeña EP: The chemistry of reactive oxygen species (ROS) revisited: Outlining their role in biological macromolecules (DNA, lipids and proteins) and induced pathologies. Int J Mol Sci. 22:46422021. View Article : Google Scholar : PubMed/NCBI

281 

Grattagliano I, Palmieri VO, Portincasa P, Moschetta A and Palasciano G: Oxidative stress-induced risk factors associated with the metabolic syndrome: A unifying hypothesis. J Nutr Biochem. 19:491–504. 2008. View Article : Google Scholar : PubMed/NCBI

282 

Fernández-Sánchez A, Madrigal-Santillán E, Bautista M, Esquivel-Soto J, Morales-González A, Esquivel-Chirino C, Durante-Montiel I, Sánchez-Rivera G, Valadez-Vega C and Morales-González JA: Inflammation, oxidative stress, and obesity. Int J Mol Sci. 12:3117–3132. 2001. View Article : Google Scholar : PubMed/NCBI

283 

Sena CM, Leadro A, Azul L, Seiça R and Perry G: Vascular oxidative stress: Impact and therapeutic approaches. Front Physiol. 9:16682018. View Article : Google Scholar : PubMed/NCBI

284 

Smirne C, Croce E, Di Benedetoo D, Cantaluppi V, Comi C, Sainaghi PP, Minisini R, Grossini E and Pirisi M: Oxidative stress in non-alchoholic fatty liver disease. Livers. 2:30–76. 2022. View Article : Google Scholar

285 

Field AE, Coakley EH, Must A, Spadano JL, Laird N, Dietz WH, Rimm E and Golditz GA: Imact of overweight on the risk of developing common chronic diseases during a 10-year period. Arch Intern Med. 161:1581–1586. 2001. View Article : Google Scholar : PubMed/NCBI

286 

Rother KI: Diabetes treatment-Bridging the devide. N Engl J Med. 356:1499–1501. 2007. View Article : Google Scholar : PubMed/NCBI

287 

Norhammar A and Schenck-Gustafsson K: Type 2 diabetes and cardiovascular disease in women. Diabetologia. 56:1–9. 2013. View Article : Google Scholar : PubMed/NCBI

288 

Chan JM, Rimm EB, Colditz GA, Stampfer MJ and Willett WC: Obesity, fat distribution, and weight gain as risk factors for clinical diabetes in men. Diabetes Care. 17:961–969. 1994. View Article : Google Scholar : PubMed/NCBI

289 

Colditz GA, Willett WC, Rotnitzky A and Manson JE: Weight gain as a risk factor for clinical diabetes mellitus in women. Ann Intern Med. 122:481–486. 1995. View Article : Google Scholar : PubMed/NCBI

290 

Wannamethee SG and Shaper AG: Weight change and duration of overweight and obesity in the incidence of type 2 diabetes. Diabetes Care. 22:1266–1272. 1999. View Article : Google Scholar : PubMed/NCBI

291 

Schienkiewitz A, Schulz MB, Hoffmann K, Kroke A and Boeing H: Body mass index history and risk of type 2 diabetes: Results from the European Prospective Investigation into cancer nutrition (EPIC)-Potsdam study. Am J Clin Nutr. 84:427–433. 2006. View Article : Google Scholar : PubMed/NCBI

292 

DeFronzo RA: Pathogenesis of type 2 diabetes mellitus. Med Clin North Am. 88:787–835. 2004. View Article : Google Scholar : PubMed/NCBI

293 

Ferrannini E, Gastaldelli A, Miyazaki Y, Matsuda M, Mari A and DeFronzo RA: Beta-cell function in subjects spanning the range from normal glucose tolerance to overt diabetes: A new analysis. J Clin Endocrinol Metab. 90:493–500. 2005. View Article : Google Scholar : PubMed/NCBI

294 

Panenin F, Castantino S and Cosentino F: Insulin resistance, diabetes, and cardiovascular risk. Curr Atheroscler Rep. 16:4192014. View Article : Google Scholar : PubMed/NCBI

295 

Abdul-Ghami MA and DeFronzo RA: Phathophysiology of prediabetes. Curr Diab Rep. 9:193–199. 2009. View Article : Google Scholar : PubMed/NCBI

296 

Reaven GM: Insulin resistance: The link between obesity and cardiovascular disease. Med Clin North Am. 95:875–892. 2011. View Article : Google Scholar : PubMed/NCBI

297 

DeFronzo RA, Ferrannini E and Simonson DC: Fasting hyperglycemia in non-insulin-dependent diabetes mellitus: contributions of excessive hepatic glucose production and impaired tissue glucose uptake. Metabolism. 38:387–395. 1989. View Article : Google Scholar : PubMed/NCBI

298 

Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA and Shulman RG: Quatitation of muscle glycogen synthesis in normal subjects and subjects with non-insulin-dependent diabetes by 13C nuclear magnetic resonance spectroscopy. N Engl J Med. 322:223–228. 1990. View Article : Google Scholar : PubMed/NCBI

299 

McGarry JD: Banting lecture 2001: Dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes. 51:7–18. 2002. View Article : Google Scholar : PubMed/NCBI

300 

Kashyap S, Belfort R, Gastaldelli A, Pratipanawatr T, Berria R, Pratipanawatr W, Bajaj M, Madarino L, DeFronzo R and Cusi K: A substained increase in plasma free fatty acids impairs insulin secretion in nondiabetic sujects genetically predisposed to develop type 2 diabetes. Diabetes. 52:2461–2474. 2003. View Article : Google Scholar : PubMed/NCBI

301 

Lei XG and Vatamaniuk MZ: Two tales of antioxidant enzymes on β cells and diabetes. Antioxid Redox Signal. 14:489–503. 2011. View Article : Google Scholar : PubMed/NCBI

302 

Krebs M, Krssaak M, Bernroider E, Anderwald C, Brehm A, Meyerspeer M, Nowotny P, Roth E, Waldhäusl W and Roden M: Mechanism of amino acid-induced skeletal muscle insulin resistance in humans. Diabetes. 51:599–605. 2002. View Article : Google Scholar : PubMed/NCBI

303 

Pi-Sunyer FX: The epidemiology of central fat distribution in relation to disease. Nutr Rev. 62((7 Pt2)): S120–S126. 2004. View Article : Google Scholar : PubMed/NCBI

304 

Despres JP: Intra-abdominal obesity: An untreated risk factor for type 2 diabetes and cardiovascular disease. J Endocrinol Invest. 29 (3 Suppl):S77–S82. 2006.

305 

Klein S, Allison DB, Heymsfield SB, Kelley DE, Leibel RL, Nomas C and Kahn R; Association for Weight Management and Obesity Prevention; NASSO, the Obesity Society; American Society for Nutrition, : American Diabetes Association: Waist circumference and cardiometabolic risk: A consensus statement from shaping America's health: Association for weight management and obesity pevention; NAASO, the obesity society; the American society for nutrition; and the American diabetes association. Diabetes Care. 30:1647–1652. 2007. View Article : Google Scholar : PubMed/NCBI

306 

Ashwell M, Gumn P and Gibson S: Waist-to-height is a better screening tool than waist cincumference and BMI for adult cardiometabolic risk factors: Systemic review and meta-analysis. Obes Rev. 13:275–286. 2012. View Article : Google Scholar : PubMed/NCBI

307 

Kouli GM, Panagiotakos DB, Kyrou I, Georgousopoulou EN, Chryoshoou C, Tsigos C, Tousoulis D and Pitsavos C: Visceral adiposity index and 10-year cardiovascular disease incidence. The ATTICA study. Nutr Metab Cardiovasc Dis. 27:881–889. 2017. View Article : Google Scholar : PubMed/NCBI

308 

Weiss R: Fat distribution and storage: How much, where, and how? Eur J Endocrinol. 157 (Suppl 1):S39–S45. 2007. View Article : Google Scholar : PubMed/NCBI

309 

Montague CT and O'Rahilly S: The perils of portliness: Causes and consequences of viscelar adiposity. Diabetes. 49:883–888. 2000. View Article : Google Scholar : PubMed/NCBI

310 

Yang X and Smith U: Adipose tissue distribution and risk of metabolic disease: Does thiazolidinedione-induced adipose tissue redistribution provide a clue to the anwer? Diagetologia. 50:1127–1139. 2007. View Article : Google Scholar : PubMed/NCBI

311 

Peraldi P and Spiegelman B: TNF-α and insulin resistance: Summary and future prospects. Mol Cell Biochem. 182:169–175. 1998. View Article : Google Scholar : PubMed/NCBI

312 

Pickup JC: Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care. 27:813–823. 2004. View Article : Google Scholar : PubMed/NCBI

313 

Han CY: Roles of reactive oxygen species on insulin resistance in adipose tissue. Diabetes Metab J. 40:272–279. 2016. View Article : Google Scholar : PubMed/NCBI

314 

Yan LJ: Redox imbalance stress in diabetes mellitus: Role of the polyol pathway. Animal Model Exp Med. 1:7–13. 2018. View Article : Google Scholar : PubMed/NCBI

315 

Dutta BJ, Singh S, Seksaria S, Gupta GD and Singh A: Inside the diabetic brain: Insulin resistance and molecular mechanism associated with cognitive impairment and its possible therapeutic strategies. Pharmacol Res. 182:1063582022. View Article : Google Scholar : PubMed/NCBI

316 

Li H, Ren J, Li Y, Wu Q and Wei J: Oxidative stress: The nexus of obesity and cognitive dysfunction in diabetes. Front Endocrinol (Lausanne). 14:11340252023. View Article : Google Scholar : PubMed/NCBI

317 

Emanuela F, Grazia M, Marco DR, Paola LM, Giorgio F and Marco B: Inflammation as a link between obesity and metabolic syndrome. J Nutr Metab. 2012:4763802012. View Article : Google Scholar : PubMed/NCBI

318 

Böni-Schnetzler M and Meier DT: Islet inflammation in type 2 diabetes. Semin Immunopathol. 41:501–513. 2019. View Article : Google Scholar : PubMed/NCBI

319 

Larsen CN, Faulenbach A, Vaag Α, Vølund A, Ehses JA, Seifert B, Mandrup-Poulsen T and Donath MY: Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 356:1517–1526. 2007. View Article : Google Scholar : PubMed/NCBI

320 

Larsen CM, Faulenbach M, Vaag A, Ehses JA, Donath MY and Mandrup-Poulsen T: Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes. Diabetes Care. 32:1663–1668. 2009. View Article : Google Scholar : PubMed/NCBI

321 

Infante M, Padilla N, Alejandro R, Caprio M, Della-Morte D, Fabbri A and Ricordi C: Diabetes-modifying antirheumatic drugs: The roles of DMARDs as glucose-lowering agents. Medicina (Kaunas). 58:5712022. View Article : Google Scholar : PubMed/NCBI

322 

Powers NE, Swartzwelter B, Marchetti C, de Graaf DM, Lerchner A, Schlapschy M, Datar R, Binder U, Edwards CK III, Skerra A and Dinarell CA: PASylation of IL-1 receptor antagonist (IL-1Ra) retains IL-1 blockade and extends its duration in mouse urate crystal-induced peritonitis. J Biol Chem. 295:868–882. 2020. View Article : Google Scholar : PubMed/NCBI

323 

Tegtmeyer K, Atassi G, Zhao J, Maloney NJ and Lio PA: Off-Label studies on anakinra in dermatology: A review. J Dermatolog Treat. 33:73–86. 2022. View Article : Google Scholar : PubMed/NCBI

324 

van Asseldonk EJ, Stienstra R, Koenen TB, Joosten LA, Netea MG and Tack CJ: Treatment with Anakinra improves disposition index but not insulin sensitivity in nondiabetic subjects with the metabolic syndrome: A randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab. 96:2119–2126. 2011. View Article : Google Scholar : PubMed/NCBI

325 

van Poppel PCM, van Asseldonk EJP, Holst JJ, Vilsbøll T, Netea MG and Tack CJ: The interleukin-1 receptor antagonist anakinra improves first-phase insulin secretion and insulinogenic index in subjects with impaired glucose tolerance. Diabetes Obes Metab. 16:1269–1273. 2014. View Article : Google Scholar : PubMed/NCBI

326 

Cucak H, Hansen G, Vrang N, Skarsfeldt T, Steiness E and Jelsing J: The IL-1β receptor antagonist SER140 postpones the onset of diabetes in female nonobese diabetic mice. J Diabetes Res. 2016:74846012016. View Article : Google Scholar : PubMed/NCBI

327 

Cavelti-Weder C, Babians-Brunner A, Keller C, Stahel MA, Kurz-Levin M, Zayed H, Solinger AM, Mandrup-Poulsen T, Dinarello CA and Donath MY: Effects of gevokizumab on glycemia and inflammatory markers in type 2 diabetes. Diabetes Care. 35:1654–1662. 2012. View Article : Google Scholar : PubMed/NCBI

328 

Rissanen A, Howard CP, Botha J and Thuren T; Global Investigators, : Effects of anti-IL-1β antibody (canakinumab) on insulin secretion rates in impaired glucose tolerance or type 2 diabetes: Results of a randomized placebo-controlled trial. Diabetes Obes Metab. 14:1088–1096. 2012. View Article : Google Scholar : PubMed/NCBI

329 

Hensen J, Howard CP, Walter V and Thuren T: Impact of interleukin-1β antibody (canakinumab) on glycemic indicators in patients with type 2 diabetes mellitus: Results of secondary endpoints from a randomized, placebo-controlled trial. Diabetes Metab. 39:524–531. 2013. View Article : Google Scholar : PubMed/NCBI

330 

Ridker PM, Howard CP, Walter V, Everett B, Libby P, Hensen J and Thuren T; on the behalf of CANTOS Pilot Investigative Group, : Effects of Interleukin-1β Inhibition with Canakinumab on Hemoglobin A1c, Lipids, C-Reactive Protein, Interleukin-6, and Fibrinogen: A Phase IIb Randomized, Placebo-Controlled Trial. Circulation. 126:2739–2748. 2012. View Article : Google Scholar : PubMed/NCBI

331 

Choudhury RP, Birks JS, Manii V, Biasiolli L, Robson MD, L'Allier PL, Gingras MA, Alie N, McLaughlin MA, Basson CT, et al: Artherial effects of canakinumab in patients with atherosclerosis and type 2 diabetes or glucose intolerance. J Am Coll Cardiol. 68:1769–1780. 2016. View Article : Google Scholar : PubMed/NCBI

332 

Noe A, Howard C, Thuren T, Taylor A and Skerjanec A: Pharmacokinetic and pharmacodynamics characteristics of single-dose canakinumab in patients with type 2 diabetes mellitus. Clin Ther. 36:1625–1637. 2014. View Article : Google Scholar : PubMed/NCBI

333 

Sloan-Lancaster J, Abu-Raddad E, Polzer J, Miller JW, Schere JC, De Gaetano A, Berg JK and Landschulz WH: Double blind, randomized study evaluating the glycemic and anti-inflammatory effects of subcutaneous LY2189102, a neutralizing IL-1 antibody, in patients with type 2 diabetes. Diabetes Care. 36:2239–2246. 2013. View Article : Google Scholar : PubMed/NCBI

334 

Everett BM, Donath MY, Pradhan AD, Thuren T, Pais P, Nicolau JC, Glynn RJ, Libby P and Ridker PM: Anti-inflimmatory therapy with canakinumad for the prevention and management of diabetes. J Am Coll Cardiol. 71:2392–2401. 2018. View Article : Google Scholar : PubMed/NCBI

335 

Olson NC, Callas PW, Hanley AJG, Festa A, Haffner SM, Wagenknecht LE and Tracy RP: Circulating levels of TNF-α are associated with impaired glucose tolerance, increased insulin resistance, and ethnicity: The insulin resistance atherosclerosis study. J Clin Endocrinol Metab. 97:1032–1040. 2012. View Article : Google Scholar : PubMed/NCBI

336 

Wascher TC, Lindeman JHN, Sourij H, Kooistra T, Pacini G and Roden M: Chronic TNF-α neutralization does not improve insulin resistance or endothelial function in ‘healthy’ men with metabolic syndrome. Mol Med. 17:189–193. 2011. View Article : Google Scholar : PubMed/NCBI

337 

van den Oever IAM, Baniaamam M, Simsek S, Raterman HG, van Denderen JC, van Eijk IC, Peters MJL, van der Horst-Bruinsma IE, Smulders YM and Nurmohamed MT: The effect of anti-TNF treatment on body composition and insulin resistance in patients with rheumatoid arthritis. Rheumatol Int. 41:319–328. 2021. View Article : Google Scholar : PubMed/NCBI

338 

Kiortsis DN, Mavridis AK, Vasakos S, Nikas SN and Drosos AA: Effects of infliximab treatment on insulin resistance in patients with rheumatoid arthritis and ankylosing spondylitis. Ann Rheum Dis. 64:765–766. 2005. View Article : Google Scholar : PubMed/NCBI

339 

Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua G, Sanchez-Andrade A, Martin J and Llorca J: Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 24:83–86. 2006.PubMed/NCBI

340 

Haida KS, Bertachini G, Tavoni T, Guilhermetti M, Loures MR and Bazotte RB: Infliximab treatment prevents hyperglycemia and the intensification of hepatic gluconeogenesis in an animal model of high fat diet-induced liver glucose overproduction. Braz Arch Biol Technol. 55:389–393. 2012. View Article : Google Scholar

341 

Méndez-García LA, Trejo-Millán F, Martínez-Reyes CP, Majarrez-Reyna AN, Esquivel-Velázquez M, Melendez-Mier G, Islas-Andrade S, Rojas-Bernbé A, Kzhyshkowska J and Escobedo G: Infliximab ameriorates tumor necrosis factor-alpha-induced insulin resistance by attenuating PTP1B activation in 3T3L1 adipocytes in vitro. Scan J Immunol. 88:e127162018. View Article : Google Scholar : PubMed/NCBI

342 

Abdelhamid YA, Elyamany MF, Al-Shorbagy MY and Badary OA: Effects of TNF-α antagonist infliximad on fructose-induced metabolic syndrome in rats. Hum Exp Toxicol. 40:801–811. 2021. View Article : Google Scholar : PubMed/NCBI

343 

Bernstein LE, Berry J, Kim S, Canavan B and Grinspoon SK: Effects of etanercept in patients with the metabolic syndrome. Arch Intern Med. 166:902–908. 2006. View Article : Google Scholar : PubMed/NCBI

344 

Lo J, Bernstein LE, Canavan B, Torriani M, Jackson MB, Ahima RS and Grinspoon SK: Effects of TNF-alpha neutralization on adipocytokines and skeletal muscle adiposity in the metabolic syndrome. Am J Physiol Endocrinol Metabol. 293:E102–E109. 2007. View Article : Google Scholar : PubMed/NCBI

345 

Bravo C, Cataldo LR, Galgani J, Parada J and Santos JL: Leptin/Adiponectin ratios using either total or high molecular weight adiponectin as biomarkers of systemic insulin sensitivity in normoglycemic women. Diabetes Res. 2017:90310792017.PubMed/NCBI

346 

Stanley TL, Zanni MV, Johnsen S, Rasheed S, Makimura H, Lee H, Khor VK, Ahima RS and Grinspoon SK: TNF-α antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J Clin Endocrinol Metab. 96:E146–E150. 2011. View Article : Google Scholar : PubMed/NCBI

347 

Paquot N, Castillo MJ, Lefèbvre PJ and Scheen AJ: No increased insulin sensitivity after a single intravenous administration of a recombinant human tumor necrosis factor receptor: Fc fusion protein in obese insulin-resistant patients. J Clin Endocrinol Metab. 85:1316–1319. 2000. View Article : Google Scholar : PubMed/NCBI

348 

Dominguez H, Storgaard H, Rask-Madsen C, Hermann TS, Ihlemann N, Nielsen DB, Spohr C, Kober L, Vaag A and Torp-Pedersen C: Metabolic and vascular effects of tumor necrosis factor-α blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res. 42:517–525. 2005. View Article : Google Scholar : PubMed/NCBI

349 

Ronti T, Lupattelli G and Mannarino E: The endocrine function of adipose tissue: An update. Clin Endocrinol (Oxf). 64:355–365. 2006. View Article : Google Scholar : PubMed/NCBI

350 

Hu D, Russell RD, Remash D, Greenaway T, Rattigan S, Squibb KA, Jones G, Ross RM, Roberts CK, Premilovac D, et al: Are the metabolic benefits of resistance in type 2 diabetes linked to improvement in adipose tissue microvascular blood flow? Am J Physiol Endocrinol Metab. 315:E1242–E1250. 2018. View Article : Google Scholar : PubMed/NCBI

351 

Ruscitti P, Berardicurti O, Cipriani P and Giacomelli R; TRACK Study Group, : Benefits of anakinra versus TNF inhibitors in rheumatoid arthritis and type 2 diabetes: Long-term findings from participants furtherly followed-up in the TRACK study, a multicentre, open-label, randomized, controlled trial. Clin Exp Rheumatol. 39:403–406. 2021. View Article : Google Scholar : PubMed/NCBI

352 

Ramos-Zavala MG, Gonzalez-Ortiz M, Martinez-Abundis E, Robles-Cervantes JA, Gonzalez-Lopez R and Santiago-Hernandez NJ: Effect of diacerein on insulin secretion and metabolic control in drug-naïve patients with type 2 diabetes. Diabetes Care. 34:1591–1594. 2011. View Article : Google Scholar : PubMed/NCBI

353 

Cardoso CRL, Leite NC, Carlos FO, Loureiro AA, Viegas BB and Salles GF: Efficacy and safety of diacerein in patients with inadequately controlled type 2 diabetes: A randomized controlled trial. Diabetes Care. 40:1356–1363. 2017. View Article : Google Scholar : PubMed/NCBI

354 

Tres GS, Fuchs SC, Piovesan F, Koehler-Santos P, Pereira FD, Camey S, Lisboa HK and Moreira LB: Effect of diacerein on metabolic control and inflammatory markers in patients with type 2 diabetes using antidiabetic agents: A randomized controlled trial. J Diabetes Res. 2018:42465212018. View Article : Google Scholar : PubMed/NCBI

355 

Jangsiripornpakorn J, Srisuk S, Chailurkit L, Nimitphong H, Saetung S and Ongphiphadhanakul B: The glucose-lowering effect of low-dose diacerein and its responsiveness metabolic markers in uncontrolled diabetes. BMC Res Notes. 15:912022. View Article : Google Scholar : PubMed/NCBI

356 

Piovesan F, Tres GS, Moreira LB, Andrades ME, Lisboa HK and Fucks SC: Effects of diacerein on renal function and inflammatory cytokines in participants with type 2 diabetes mellitus and chronic kidney disease: A randomized controlled trial. PLoS One. 12:e01865542017. View Article : Google Scholar : PubMed/NCBI

357 

Di Prospero NA, Artis E, Andrade-Gordon P, Johnson DL, Vaccaro N, Xi L and Rothenberg P: CCR2 antagonism in patients with type 2 diabetes mellitus: A randomized, placebo-controlled study. Diabetes Obes Metab. 16:1055–1064. 2014. View Article : Google Scholar : PubMed/NCBI

358 

Mulder P, van den Hoek AM and Kleemann R: The CCR2 inhibitor propagermanium attenuates diet-induced insulin resistance, adipose tissue inflammation and non-alcoholic steatohepatitis. PLoS One. 12:e01697402017. View Article : Google Scholar : PubMed/NCBI

359 

Huh JH, Kim HM, Lee ES, Kwon MH, Lee BR, Ko HJ and Chung CH: Dual CCR2/5 antagonist attenuates obesity-induced insulin resistance by regulating macrophage recruitment and M1/M2 status. Obesity (Silver Spring). 26:378–386. 2018. View Article : Google Scholar : PubMed/NCBI

360 

Tuttle KR, Brosius FC III, Adler SG, Kretzler M, Mehta RL, Tumlin JA, Tanaka Y, Haneda M, Liu J, Silk ME, et al: JAK1/JAK2 inhibition by baricitinib in diabetic kidney disease: Results from a phase 2 randomized controlled clinical trial. Nephrol Dial Transplant. 33:1950–1959. 2018. View Article : Google Scholar : PubMed/NCBI

361 

Faghihimani E, Amnorroaya A, Rezvanian H, Adibi P, Ismail-Beigi F and Amini M: Salsalate improves glycemic control in patients with newly diagnosed type 2 diabetes. Acta Diabetol. 50:537–543. 2013. View Article : Google Scholar : PubMed/NCBI

362 

Goldfine AB, Fonseca V, Jablonski KA, Chen YD, Tipton L, Staten MA and Steven E; Targeting Inflammation Using Salsalate in Type 2 Diabetes Study Team, : Salicylate (Salsalate) in patients with type 2 diabetes: A randomized trial. Ann Intern Med. 159:1–12. 2013. View Article : Google Scholar : PubMed/NCBI

363 

Li D, Zhong J, Zhang Q and Zhang J: Effects of anti-inflammatory therapies on glycemic control in type 2 diabetes mellitus. Front Immunol. 14:11251162023. View Article : Google Scholar : PubMed/NCBI

364 

Raimondo MG, Biggioggero M, Crotti C, Becciolini A and Favalli EG: Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Design Dev Ther. 11:1593–1603. 2017. View Article : Google Scholar : PubMed/NCBI

365 

Klinder A, Waletzko-Hellwig J, Sellin ML, Seyfarth-Sehlke A, Wolfien M, Prehn F, Bader R and Jonitz-Heincke A: Effects of the interleukin-6 receptor blocker sarilumab on metabolic activity and differentiation capacity of primary human osteoblasts. Pharmaceutics. 14:13902022. View Article : Google Scholar : PubMed/NCBI

366 

Genovese MC, Burmester GR, Hagino O, Thangavelu K, Iglesias-Rodriguez M, John GT, González-Gay MA, Mandrup-Poulsen T and Fleischmann R: Interleukin-6 receptor blockade or TNFα inhibition for reducing glycaemia in patients with RA and diabetes: Post hoc analyses of three randomised, controlled trials. Arthritis Res Ther. 22:2062020. View Article : Google Scholar : PubMed/NCBI

367 

Drutskaya MS, Efimou GA, Kruglou AA and Nedospasou SA: Can we design a better anti-cytokine therapy? Semin Arthritis and Rhematism. 49:S39–S42. 2019.PubMed/NCBI

368 

Nosenko MA, Atretkhany KSN, Mokhonov VV, Vasilenko EA, Kruglov AA, Tillib SV, Drutskaya MS and Nedospasov SA: Moduatation of bioavailability of proinflammatory cytokines produced by myeloid cells. Semin Arthritis Rheum. 49:S39–S42. 2019. View Article : Google Scholar : PubMed/NCBI

369 

Velikova TV, Kabakchieva PP, Assyov YS and Georgiev TA: Targeting inflammatory cytokines to improve type 2 diabetes control. Biomed Res Int. 2021:72974192021. View Article : Google Scholar : PubMed/NCBI

370 

Achari A and Jain SK: Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci. 18:13212017. View Article : Google Scholar : PubMed/NCBI

371 

Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka T, et al: The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med. 7:941–946. 2001. View Article : Google Scholar : PubMed/NCBI

372 

Combs TP, Wagner JA, Berger J, Doebber T, Wang WJ, Zhang BB, Tanen M, Berg AH, O'Rahilly S, Savage DB, et al: Induction of adipocyte complement-related protein of 30 kilodaltons by PPRgamma agonists: A potential mechanism of insulin sensitization. Endocrinology. 143:998–1007. 2002. View Article : Google Scholar : PubMed/NCBI

373 

Kolak M, Yki-Järvinen H, Kannisto K, Tiikkainen M, Hamsten A, Eriksson P and Fisher RM: Effects of chronic rosiglitazone therapy on gene expression in human adipose tissue in vivo in patients with type 2 diabetes. J Clin Endocrinol Metab. 92:720–724. 2007. View Article : Google Scholar : PubMed/NCBI

374 

Peraldi P, Xu M and Spiegelman BM: Thiazolidinediones block tumor necrosis factor-alpha-incuded inhibition of insulin signaling. J Clin Invest. 100:1863–1869. 1997. View Article : Google Scholar : PubMed/NCBI

375 

Wolf AM, Wolf D, Rumpold H, Enrich B and Tilg H: Adiponectin induces the anti-inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem Biophys Res Commun. 323:630–635. 2004. View Article : Google Scholar : PubMed/NCBI

376 

Mosser DM and Zhang X: Interleukin-10: New perspectives on an old cytokine. Immunol Rev. 226:205–218. 2008. View Article : Google Scholar : PubMed/NCBI

377 

Maclsaac RJ and Jerum G: Clinical indications for thiazolidinediones. Aust Prescr. 27:70–74. 2004. View Article : Google Scholar

378 

Quinn CE, Hamilton PK, Lockhart CJ and McVeigh GE: Thiazolidinediones: Effects on insulin resistance and the cardiovascular system. Br J Pharmacol. 153:636–645. 2008. View Article : Google Scholar : PubMed/NCBI

379 

Graham DJ, Green L, Senior JR and Nourjah P: Troglitazone-induced liver failure: A case study. Am J Med. 114:299–306. 2003. View Article : Google Scholar : PubMed/NCBI

380 

Tuccori M, Filion KB, Yin H, Yu OH, Platt RW and Azoulay L: Pioglitazone use and risk of bladder cancer: Population based cohort study. BMJ. 352:i15412016. View Article : Google Scholar : PubMed/NCBI

381 

Aronoff S, Rosenblatt S, Braithwaite S, Egan JW, Mathisen AL and Schneider RL: Pioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: A 6-month randomized placebo-controlled dose-response study. The pioglitazone 001 study group. Diabetes Care. 23:1605–1611. 2000. View Article : Google Scholar : PubMed/NCBI

382 

Mudaliar S, Chang AR and Henry RR: Thiazolidinediones, peripheral edema, and type 2 diabetes: Incidence, pathophysiology, and clinical implications. Endocr Pract. 9:406–416. 2003. View Article : Google Scholar : PubMed/NCBI

383 

Arnold SV, Inzucchi SE, Echouffo-Tcheugui JB, Tang F, Lam CSP, Sperling LS and Kosiborod M: Understanding contemporary use of thiazolidinediones. Cir Heart Fail. 12:e0058552019. View Article : Google Scholar : PubMed/NCBI

384 

Ferris FL III and Patz A: Macular edema. A complication of diabetic retinopathy. Surv Ophthalmol. 28:452–461. 1984. View Article : Google Scholar : PubMed/NCBI

385 

Ryan EH Jr, Han DP, Ramsay RC, Cantrill HL, Bennett SR, Dev S and Williams DF: Diabetic macular edema associated with glitazone use. Retina. 26:562–570. 2006. View Article : Google Scholar : PubMed/NCBI

386 

Vestergaard P: Discrepacies in bone mineral density and fracture risk in patients with type 1 and type 2 diabetes-a meta-analysis. Osteoporos Int. 18:427–444. 2007. View Article : Google Scholar : PubMed/NCBI

387 

Fonseca V: Effect of thiazolidinediones on body weight in patients with diabetes mellitus. Am J Med. 115:42–48. 2003. View Article : Google Scholar

388 

Ko KD, Kim KK and Lee KR: Does weight gain associated with thiazolidinedione use negatively affect cardiometabolic health? J Obes Metab Syndr. 26:102–106. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 29 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Varra F, Varras M, Varra V and Theodosis-Nobelos P: Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review). Mol Med Rep 29: 95, 2024
APA
Varra, F., Varras, M., Varra, V., & Theodosis-Nobelos, P. (2024). Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review). Molecular Medicine Reports, 29, 95. https://doi.org/10.3892/mmr.2024.13219
MLA
Varra, F., Varras, M., Varra, V., Theodosis-Nobelos, P."Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review)". Molecular Medicine Reports 29.6 (2024): 95.
Chicago
Varra, F., Varras, M., Varra, V., Theodosis-Nobelos, P."Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review)". Molecular Medicine Reports 29, no. 6 (2024): 95. https://doi.org/10.3892/mmr.2024.13219