Next Article in Journal
Substance P Activates the Wnt Signal Transduction Pathway and Enhances the Differentiation of Mouse Preosteoblastic MC3T3-E1 Cells
Next Article in Special Issue
Evolving Concepts in the Pathogenesis of NASH: Beyond Steatosis and Inflammation
Previous Article in Journal
Differential Signaling by Protease-Activated Receptors: Implications for Therapeutic Targeting
Previous Article in Special Issue
Evaluation of Hepatic Tissue Blood Flow Using Xenon Computed Tomography with Fibrosis Progression in Nonalcoholic Fatty Liver Disease: Comparison with Chronic Hepatitis C
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Obesity and Its Metabolic Complications: The Role of Adipokines and the Relationship between Obesity, Inflammation, Insulin Resistance, Dyslipidemia and Nonalcoholic Fatty Liver Disease

Center for Food and Nutritional Genomics Research, Kyungpook National University, 1370 Sankyuk Dong Puk-ku, Daegu 702-701, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2014, 15(4), 6184-6223; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15046184
Submission received: 4 February 2014 / Revised: 27 March 2014 / Accepted: 1 April 2014 / Published: 11 April 2014
(This article belongs to the Special Issue Non-Alcoholic Fatty Liver Disease Research)

Abstract

:
Accumulating evidence indicates that obesity is closely associated with an increased risk of metabolic diseases such as insulin resistance, type 2 diabetes, dyslipidemia and nonalcoholic fatty liver disease. Obesity results from an imbalance between food intake and energy expenditure, which leads to an excessive accumulation of adipose tissue. Adipose tissue is now recognized not only as a main site of storage of excess energy derived from food intake but also as an endocrine organ. The expansion of adipose tissue produces a number of bioactive substances, known as adipocytokines or adipokines, which trigger chronic low-grade inflammation and interact with a range of processes in many different organs. Although the precise mechanisms are still unclear, dysregulated production or secretion of these adipokines caused by excess adipose tissue and adipose tissue dysfunction can contribute to the development of obesity-related metabolic diseases. In this review, we focus on the role of several adipokines associated with obesity and the potential impact on obesity-related metabolic diseases. Multiple lines evidence provides valuable insights into the roles of adipokines in the development of obesity and its metabolic complications. Further research is still required to fully understand the mechanisms underlying the metabolic actions of a few newly identified adipokines.

1. Introduction

The worldwide prevalence of obesity and its metabolic complications have increased substantially in recent decades. According to the World Health Organization, the global prevalence of obesity has nearly doubled between 1980 and 2008, and more than 10% of the adults aged 20 and over is obese in 2008 [1]. Projections based on the current obesity trends estimate that there will be 65 million more obese adults in the USA and 11 million more obese adults in the UK by 2030, consequently accruing an additional 6–8.5 million cases of diabetes, 5.7–7.3 million cases of heart disease and stroke for USA and UK combined [2]. The increased prevalence in obesity is also associated with increasing prevalence of nonalcoholic fatty liver disease (NAFLD). Among the Americas, the prevalence of NAFLD is highest in the USA, Belize and Barbados and Mexico, which have a high prevalence of obesity [3]. Obesity, especially abdominal obesity, is one of the predominant underlying risk factors for metabolic syndrome [4]. Obesity increases the risk of developing a variety of pathological conditions, including insulin resistance, type 2 diabetes, dyslipidemia, hypertension and NAFLD (Figure 1). Accumulating evidence suggests that chronic inflammation in adipose tissue may play a critical role in the development of obesity-related metabolic dysfunction [57].
Adipose tissue has been recognized as an active endocrine organ and a main energy store of the body [8]. Excess adiposity and adipocyte dysfunction result in dysregulation of a wide range of adipose tissue-derived secretory factors, referred to as adipokines, which may contribute to the development of various metabolic diseases via altered glucose and lipid homeostasis as well as inflammatory responses [9,10]. In addition, excess fat accumulation promotes the release of free fatty acids into the circulation from adipocytes, which may be a critical factor in modulating insulin sensitivity [11,12]. However, plasma free fatty acid levels do not increase in proportion to the amount of body fat, since their basal adipose tissue lipolysis per kilogram of fat is lower in obese subjects than in lean subjects [13]. This finding has been supported by other studies of adipocytes from obese subjects [14,15] and it was associated with down-regulation of hormone sensitive lipase and adipose triglyceride lipase, key enzymes involved in intracellular degradation of triglycerides [14,1618]. Thus, Karpe et al. [19] have recently suggested that the link between circulating free fatty acid levels and insulin sensitivity in vivo is needed to further elucidate this complicated relationship.
In this review, we will first discuss the critical role of adipose tissue for health and as a repository of free fatty acids. We will also review how the dysregulation of free fatty acids and inflammatory factors released by enlarged adipose tissue is associated with the pathogenesis of metabolic syndrome (insulin resistance, dyslipidemia and NAFLD). In particular, we will focus on the imbalance of pro-inflammatory and anti-inflammatory molecules secreted by adipose tissue which contribute to metabolic dysfunction.

2. Function of Adipose Tissue

Adipose tissue is the major site for storage of excess energy in the form of triglycerides, and it contains multiple cell types, including mostly adipocytes, preadipocytes, endothelial cells and immune cells. During positive energy balance, adipose tissue stores excess energy as triglycerides in the lipid droplets of adipocytes through an increase in the number of adipocyte (hyperplasia) or an enlargement in the size of adipocytes (hypertrophy) [20]. The number of adipocytes is mainly determined in childhood and adolescence and remains constant during adulthood in both lean and obese subjects, even after marked weight loss [21]. Hence, an increase in fat mass in adulthood can primarily be attributed to hypertrophy. However, recent study has reported that normal-weight adults can expand lower-body subcutaneous fat, but not upper-body subcutaneous fat, via hyperplasia in response to overfeeding [22], suggesting hyperplasia of adipocytes can also occur in adulthood. Although overall obesity is associated with metabolic diseases, adipose tissue dysfunction caused by hypertrophy has been suggested to play an important role in the development of metabolic diseases such as insulin resistance [2325]. In contrast to positive energy balance states, when energy is needed between meals or during physical exercise, triglycerides stored in adipocytes can be mobilized through lipolysis to release free fatty acids into circulation and the resulting free fatty acids are transported to other tissues to be used as an energy source. It is generally accepted that free fatty acids, a product of lipolysis, play a critical role in the development of obesity-related metabolic disturbances, especially insulin resistance. In obesity, free fatty acids can directly enter the liver via the portal circulation, and increased levels of hepatic free fatty acids induce increased lipid synthesis and gluconeogenesis as well as insulin resistance in the liver [26]. High levels of circulating free fatty acids can also cause peripheral insulin resistance in both animals and humans [26,27]. Moreover, free fatty acids serve as ligands for the toll-like receptor 4 (TLR4) complex [28] and stimulate cytokine production of macrophages [29], thereby modulating inflammation of adipose tissue which contributes to obesity-associated metabolic complications. However, circulating free fatty acid concentrations do not increase in proportion to fat mass and do not predict the development of metabolic syndrome [3033], although many studies suggest a relationship between the release of free fatty acids from adipose tissue and obesity-related metabolic disorders.
Adipose tissue also has a major endocrine function secreting multiple adipokines (including chemokines, cytokines and hormones) (Figure 2). Many of the adipokines are involved in energy homeostasis and inflammation, including chemokines and cytokines. In the obese state, the adipocyte is integral to the development of obesity-induced inflammation by increasing secretion of various pro-inflammatory chemokines and cytokines [34,35]. Many of them, including monocyte chemotactic protein (MCP)-1, tumor necrosis factor (TNF)-α, interlukin (IL)-1, IL-6 and IL-8, have been reported to promote insulin resistance [3639]. Moreover, the macrophage content of adipose tissue is positively correlated with both adipocyte size and body mass, and expression of pro-inflammatory cytokines, such as TNF-α, is mostly derived from macrophages rather than adipocytes [40]. Along with the increased number of macrophages in adipose tissue, obesity induces a phenotypic switch in these cells from an anti-inflammatory M2 polarization state to a pro-inflammatory M1 polarization state [41]. The accumulation of M1 macrophages in adipose tissue has been shown to result in secretion of a variety of pro-inflammatory cytokines and chemokines that potentially contribute to obesity-related insulin resistance [5,42]. In contrast, M2-polarized macrophages participate in remodeling of adipose tissue, including clearance of dead or dying adipocytes and recruitment and differentiation of adipocyte progenitors [43]. Decreased adipose macrophage infiltration or macrophage ablation reduces expression of inflammatory cytokines in adipose tissue and improves insulin sensitivity in diet-induced obese mice [4447]. Furthermore, weight loss decreases macrophage infiltration and pro-inflammatory gene expression in adipose tissue in obese subjects [48,49]. In addition to M1 macrophages, levels of multiple pro-inflammatory immune cells, such as interferon (IFN)-γ+ T helper type 1 cells and CD8+ T cells, are increased in adipose tissue in obesity [50]. In contrast, secretion of insulin-sensitizing adiponectin is reduced in obese subjects [51].

3. Obesity and Insulin Resistance

Insulin resistance is an integral feature of metabolic syndrome and is a major predictor of the development of type 2 diabetes [52]. It has long been recognized that obesity is associated with type 2 diabetes, and the major basis for this link is the ability of obesity to induce insulin resistance. Insulin resistance is defined as the decreased ability of tissues to respond to insulin action. Adipose tissue is one of the insulin-responsive tissues, and insulin stimulates storage of triglycerides in adipose tissue by multiple mechanisms, including promoting the differentiation of preadipocytes to adipocytes, increasing the uptake of glucose and fatty acids derived from circulating lipoproteins and lipogenesis in mature adipocytes, and inhibiting lipolysis [53]. The metabolic effects of insulin are mediated by a complex insulin-signaling network (Figure 3). Insulin signaling is initiated when insulin binds to its receptor on the cell membrane, leading to phosphorylation/activation of insulin receptor substrate (IRS) proteins that are associated with the activation of two main signaling pathways: the phosphatidylinositol 3-kinase (PI3K)-AKT/protein kinase B (PKB) pathway and the Ras-mitogen-activated protein kinase (MAPK) pathway. The PI3K-AKT/PKB pathway is important for most metabolic actions of insulin. IRS-1, which is phosphorylated by the insulin receptor, activates PI3K by binding to its SH2 domain. PI3K generates phosphatidylinositol-(3,4,5)-triphosphate, a lipid second messenger, which activates several phosphatidylinositol-(3,4,5)-triphosphate-dependent serine/threonine kinases, including AKT/PKB. Ultimately, these signalling events result in the translocation of glucose transporter 4 to the plasma membrane, leading to an increase in adipocyte glucose uptake. The MAPK pathways are not implicated in mediating metabolic actions of insulin but rather in stimulating mitogenic and growth effects of insulin. In the adipose tissue, insulin also has an anti-lipolytic effect, whereby the activation of PI3K stimulates phosphodiesterase-3 so that more adenosine 3′,5′-cyclic monophosphate is hydrolyzed in adipocytes, which in turn limits the release of fatty acids from adipocytes. In addition, the transcription factors, including adipocyte determination and differentiation factor 1/sterol regulatory element-binding protein-1c (SREBP1-c), regulate the expression of multiple genes that are responsible for adipocyte differentiation, lipogenesis and fatty acid oxidation.
Evidence has suggested a role for adipose tissue in the development of insulin resistance. As discussed in the preceding text, free fatty acids and various adipokines released from adipose tissue have been involved in abnormal insulin signaling. It has been suggested that fatty acids and their metabolites, such as acyl-coenzyme A, ceramides and diacyglycerol, can impair insulin signaling by promoting protein kinases such as protein kinase C, MAPK, c-Jun N-terminal kinase (JNK), and the inhibitor of nuclear factor κB kinase β [54]. Saturated fatty acids, but not unsaturated fatty acids, induce the synthesis of ceramide, and inhibition of ceramide synthesis ameliorates saturated fatty acids-induced insulin resistance [55]. TNF-α also promotes ceramide accrual by activating sphingomyelinase, an enzyme that catalyzes the hydrolysis of sphingomyelin to ceramide [56], and ceramide mediates TNF-α-induced insulin resistance in adipocytes [57]. Haus et al. [58] reported that plasma ceramide levels are elevated in obese subjects with type 2 diabetes and it contributes to insulin resistance by activating inflammatory mediators, such as TNF-α. Thus, ceramide has been regarded as mediator linking several metabolic stresses (i.e., TNF-α and saturated fatty acids, but not unsaturated fatty acids) to the induction of insulin resistance [55,57], although the role of TNF-α in insulin resistance is somewhat controversial [59]. Obese subjects had greater whole body free fatty acids rates of appearance in plasma compared with lean subjects [60], and a sustained reduction in plasma free fatty acids levels after treatment of lipolysis inhibitor was associated with an improvement of insulin sensitivity in diabetic obese subjects [61]. The anti-lipolysis drug also decreased fasting plasma free fatty acids levels in lean control, obese nondiabetic, obese subjects with impaired glucose tolerance, and the lowering of plasma free fatty acids levels improved insulin resistance and glucose tolerance in obese subjects, regardless of the degree of their preexisting insulin resistance [62]. Recently, Girousse et al. [63] reported that a decrease in adipose tissue lipolysis improved insulin tolerance and glucose metabolism without altering fat mass. Obesity-induced increases in lipolysis not only increases local extracellular lipid concentrations but also derives accumulation of macrophages in adipose tissue [64], which is associated with systemic hyperinsulinemia and insulin resistance in obese subjects [65]. In fact, macrophage recruitment was increased with fat mass [54], and the phenotype of adipose macrophages and recruitment of macrophages and other immune cells to the adipose tissue play important roles in the development of obesity-related insulin resistance [66].
Obesity-induced insulin resistance is also associated with increased secretion of cytokines and other bioactive substances from adipose tissue as well as the number of adipose macrophages. In the adipose tissue of obese humans and animals, there are a large number of macrophages infiltrations, and this recruitment is linked to the pathogenesis of obesity-induced inflammation and insulin resistance [5,40]. The production of most inflammatory factors by adipose tissue is also increased in the obese state and promotes obesity-linked metabolic diseases [67,68]. Adipocytes and immune cells (primarily macrophages) in the adipose tissue are the primary sources of many inflammatory proteins [67,68]. There are two types of inflammatory proteins: pro-inflammatory and anti-inflammatory. A number of pro-inflammatory proteins, including MCP-1, TNF-α, IL-6, IL-18, leptin, resistin, plasminogen activator inhibitor (PAI)-1, visfatin, retinol binding protein 4 (RBP4) and angiopoietin-like protein 2 (ANGPTL2), are described in more detail in the following text. Additionally, we briefly discuss the metabolic properties of two anti-inflammatory adipokines, adiponectin and secreted frizzled-related protein 5 (SFRP5). There are discrepancies between preclinical studies and clinical trials regarding some adipokines, including TNF-α, resistin and SFRP5. Although the cause of discrepancy between preclinical studies and clinical trials is unclear, it may be due to a number of factors including the discrepancies on the species (e.g., difference in the tissue compositions and gene profiles between animals and humans), outcome measures, pre-morbid conditions and treatment methods. In addition, considering the wide spectrum of pro- and anti-inflammatory adipokines, which are altered in obesity, it is likely that crosstalk of many adipokines rather than a single adipokine in adipose tissue and other tissues may be involved in the metabolic dysregulation. Further studies are still required to clarify their roles in various conditions.

4. Role of Adipose Tissue-Produced Adipokines in Insulin Resistance

4.1. CCL2/MCP-1 and Other Chemokines

Chemokines play a major role in selectively recruiting monocytes, neutrophils, and lymphocytes and in inducing chemotaxis, and chemokines and their receptors are highly expressed in human visceral and subcutaneous adipose tissue in obesity [69]. C-C motif chemokine ligand 2/macrophage chemoattractant protein-1 (CCL2/MCP-1) is one of the key chemokines that regulate migration and infiltration of monocytes/macrophages. It initiates adipose inflammation by attracting inflammatory cells from the blood stream into adipose tissue [66,70]. CCL2/MCP-1 is expressed by adipocytes and circulating levels of CCL2/MCP-1 correlate with adiposity. Over-expression of CCL2/MCP-1 in adipose tissue increases macrophage recruitment and worsens the metabolic phenotype [71,72], whereas a deficiency of CCL2/MCP-1 or its receptor CCR2 reduces pro-inflammatory macrophages accumulation in adipose tissue and provides protection from insulin resistance as well as hepatic steatosis [45,71,73]. Recently, Meijer et al. [74] reported that adipocyte-derived CCL2/MCP-1 can stimulate inflammation independently of macrophages/leukocytes in human adipose tissue, although many cells in adipose tissue, including adipocyte and macrophages/leukocytes, produce CCL2/MCP-1. In a large cohort of Caucasians, circulating CCL2/MCP-1 was increased in type 2 diabetes subjects and presence of the MCP-1 G-2518 allele was associated with decreased plasma CCL2/MCP-1 levels as well as prevalence of insulin resistance and type 2 diabetes [75]. Similarly, MCP-1 G-2518 gene variant was decreased the risk of type 2 diabetes in a Chinese and Turkey populations [76,77]. These results support a role for CCL2/MCP-1 in pathologies associated with hyperinsulinaemia, although there are contradictory results [78,79].
Besides CCL2/MCP-1, several other chemokines such as CCL5, C-X-C motif chemokine ligand 5 (CXCL5) and CXCL14 are also involved in adipose tissue macrophage infiltration and obesity-induced insulin resistance [46,80,81]. High levels of multiple chemokine ligands (CCL2, CCL3, CCL5, CCL7, CCL8, CCL11) and receptors (CCR1, CCR2, CCR3, CCR5) have been observed in adipose tissue of obese subjects and are associated with increased inflammation [69]. Similarly, Tourniaire et al. [82] have reported that expression of numerous chemokines (CCL2, CCL5, CCL7, CCL19, CXCL1, CXCL5, CXCL8, CXCL10) is increased in adipose tissue of obese subjects compared to lean subjects. Thus, these studies suggest possibility that loss of one chemokine may be compensated by other chemokines.

4.2. TNF-α

TNF-α is a pro-inflammatory cytokine that may contribute to the pathogenesis of obesity and insulin resistance [36]. Expression of TNF-α is increased in obesity and insulin resistance in humans and is positively correlated with insulin resistance [36]. Treatment with TNF-α induces insulin resistance in adipose tissue [83], whereas deletion of TNF-α or its receptors improves insulin sensitivity in obese animals [84]. However, the correlation between plasma TNF-α levels and insulin resistance is relatively weak [36,85], and chronic neutralization of TNF-α does not improve insulin resistance in healthy overweight subjects with metabolic syndrome and insulin resistance, despite improvements in inflammatory status [86]. Bernstein et al. [87] have also reported that administration of TNF-α antagonist does not improve insulin sensitivity in humans. The absence of an effect on insulin sensitivity may be due to a compensatory role of other cytokines in the absence of TNF-α, since metabolic dysregulation has been attributed to numerous pro-inflammatory cytokines secreted by adipose tissue, including TNF-α, IL-1, and IL-6, all of which have been involved in disrupting insulin signaling [88]. TNF-α is a part of complex inflammation network and is capable of initiating cytokine cascades involving both synergistic and inhibitory reactions, which control the synthesis and expression of other cytokines, hormones, and their receptors [89]. For example, In TNF-α null mice, serum IL-12 levels were increased [90]. Because IL-12 and TNF-α are co-stimulators for IFN-α, one of the essential cytokines for regulation of the inflammation and insulin resistance in obesity [91], the up-regulation of IL-12 in the absence of TNF-α could act in a compensatory manner to induce and maintain appropriate IFN-α levels. In addition, TNF-α does not induce insulin resistance when IL-6 is down-regulated in adipose tissue [92].

4.3. IL-6 and IL-18

IL-6 is another cytokine that plays an important role in the development of insulin resistance in obesity [93]. Adipose tissue contributes to 10%–35% of circulating IL-6 levels in humans [94], and hypertrophic enlargement of adipocytes is accompanied by increased production of IL-6 by adipose tissue [95]. Expression of adipose IL-6 positively correlates with insulin resistance both in vivo and in vitro [96]. Hyperglycemia results in increased IL-6 levels [97], and treatment with IL-6 induces hyperglycemia and insulin resistance in humans [98]. However, the correlation between IL-6 and obesity or insulin resistance is controversial. A lack of IL-6 has been shown to cause obesity and insulin resistance in mice [99], but Di Gregorio et al. [100] did not observe any obvious phenotype related to obesity and diabetes in IL-6-deficient mice compared with wild-type mice. IL-6 appears to have different actions depending on the tissue (i.e., skeletal muscle vs. adipose tissue). Treatment of IL-6 enhances insulin-stimulated glucose disposal in humans in vivo, and it increases glucose uptake and fatty acid oxidation in cultured L6 myotubes via activation of adenosine monophosphate-activated protein kinase (AMPK), as well as having an anti-inflammatory effect [101,102], whereas IL-6 induces insulin resistance in adipocytes [39]. Thus, the different tissue-specific functions of IL-6 may account for the controversial findings regarding the correlation between IL-6 and insulin resistance.
IL-18 is also a pro-inflammatory cytokine and has been suggested to be produced by adipose tissue [103]. Circulating IL-18 levels have been shown to be increased in obese subjects and reduced with weight loss [104]. Moreover, overexpression of IL-18 aggravated insulin resistance in a rat model of metabolic syndrome [105]. However, a lack of IL-18 or its receptor in mice induces hyperphagia, obesity and insulin resistance [106]. Thus, further studies are needed to evaluate the role of IL-6 and IL-18 in the pathogenesis of obesity and insulin resistance.

4.4. Leptin

Leptin is abundantly expressed in adipose tissue, specifically adipocytes and is involved in the regulation of energy homeostasis [107]. It inhibits appetite and food intake and stimulates energy expenditure [107]. However, circulating leptin levels [108] and its mRNA expression in adipose tissue [109] are increased in obese subjects, probably due to the existence of leptin resistance [107]. Leptin also plays an important role in the regulation of glucose homeostasis, independent of actions on food intake, energy expenditure or body weight. Leptin improves insulin sensitivity in the liver and skeletal muscle and regulates pancreatic β-cell function [110], whereas it impairs insulin signaling in murine adipocytes [111,112]. In addition, leptin is suggested to have pro-inflammatory effects; Leptin has a cytokine-like structure, and its receptor is member of the class I cytokine receptor (gp130) superfamily [113]. It not only promotes the production of the pro-inflammatory cytokines, IL-2 and IFN-γ, but also inhibits the production of the anti-inflammatory cytokine IL-4 by T cells or mononuclear cells [114]. Concomitantly, circulating leptin levels and its expression in adipose tissues are increased in response to pro-inflammatory cytokines (TNF, IL-1) and endotoxin (lipopolysaccharide, LPS) [115]. Accordingly, the interactions between leptin and inflammation are bidirectional: Pro-inflammatory cytokines increase the synthesis and release of leptin, which in turn contribute to maintain a chronic inflammatory state in obesity [113].

4.5. Resistin

Resistin is also an adipocyte-specific secreted adipokine, and it promotes both inflammation and insulin resistance in murine models. Levels of circulating resistin are increased in obese mice and correlated with insulin resistance [116,117], whereas a lack of resistin protects mice from diet-induced hyperglycemia by increasing the activity of AMPK and decreasing the expression of gluconeogenic enzymes in the liver [118]. Moreover, resistin inhibits multiple steps involved in insulin signaling in 3T3-L1 adipocytes and induces the expression of suppressor of cytokine signaling-3 (SOCS-3), a known inhibitor of insulin signaling, in both 3T3-L1 adipocytes and murine adipose tissues [119]. However, there are conflicting reports of the potency of resistin in metabolic diseases in humans. Several studies have consistently reported a close relationship between resistin levels and obesity, insulin resistance, or type 2 diabetes [120124]. However, other studies have shown that circulating resistin levels and adipocyte expression are not associated with insulin resistance in humans [125,126]. Unlike mouse resistin, human resistin is exclusively expressed in mononuclear cells including macrophages [126], and macrophage-derived human resistin exacerbates adipose tissue inflammation and insulin resistance in mice [127].

4.6. PAI-1

PAI-1, a primary inhibitor of fibrinolysis, is also synthesized by adipocytes as well as stromal vascular cells, such as preadipocytes, fibroblasts, vascular endothelial cells, and a variety of immune cells, in adipose tissue, and its levels in plasma are increased in obesity and insulin resistance [128,129]. A deficiency of PAI-1 decreases body weight gain, increases total energy expenditure, and improves insulin resistance in mice fed a high-fat diet [130]. Moreover, mice lacking PAI-1 have promoted adipocyte differentiation and enhanced basal glucose uptake as well as insulin-stimulated glucose uptake [131]. PAI-1 regulates expression of inflammatory factors, such as IL-8 and leukotriene B4, and monocyte migration, and its expression is regulated by various cytokine inducers such as cigarette smoke extraction and LPS [132].

4.7. Visfatin

Visfatin, which was previously identified as a modulator of β-cell differentiation that is expressed in a variety of tissues and cell types, including lymphocytes, bone marrow, muscle and liver [133], has been reported to be secreted by adipose tissue, especially visceral adipose tissue, and exhibit insulin-like activities in mice [134,135]. Visfatin was highly expressed in the visceral adipose tissue of mice as well as humans, and treatment with visfatin enhances glucose uptake in adipocytes and myocytes [134]. However, several studies have failed to confirm that visfatin is expressed predominantly in visceral white adipose tissue [136139] and that expression of visfatin in adipose tissue is related to obesity [136,139,140]. Moreover, several studies have reported that circulating visfatin levels are high in subjects with obesity and type 2 diabetes and are positively associated with insulin resistance [139141]. However, serum visfatin levels and expression of visfatin in adipose tissue are not correlated with glucose metabolism or insulin resistance [137,142]. A recent study has demonstrated that central visfatin improves hypothalamic insulin signaling and increases glucose-stimulated insulin secretion and β-cell mass without changing serum visfatin levels in diabetic rats [143]. Oki et al. [144] reported that, although not related to insulin resistance in humans, serum visfatin levels are positively correlated with serum levels of IL-6 and C-reactive protein, which are known to be pro-inflammatory markers. Therefore, further studies are needed to clarify the role of visfatin in the pathogenesis of obesity induced-insulin resistance.

4.8. RBP4

RBP4 is a hepatocyte-synthesized protein that is involved in the transport of vitamin A (retinol) in the body [145]. Recently, it has been suggested that RBP4 is also secreted by adipocytes and affects insulin sensitivity [146]. In states of obesity and insulin resistance, RBP4 is preferentially produced by visceral adipose tissue compared with subcutaneous adipose tissue, and thus it is linked to intra-abdominal adipose tissue expansion [147]. Expression of RBP4 is increased in adipose tissue in insulin-resistant mice, and adipose tissue RBP4 mRNA expression is correlated with changes in serum RBP4 levels [147]. In primary human adipocytes, RBP4 inhibits insulin-induced phosphorylation of IRS-1 and ERK1/2, which may be involved in integrating nutrient sensing with insulin signaling [148]. Clinical studies have also reported that circulating RBP4 levels are associated with insulin resistance in subjects with obesity, impaired glucose tolerance, or type 2 diabetes as well as in nonobese subjects [149,150]. Along with markers of obesity and insulin resistance, RBP4 is correlated with inflammatory factors [151]. Several RBP4 gene variants are associated with adiposity and insulin resistance [152154]. However, in several clinical studies, circulating RBP4 levels were not associated with obesity and insulin resistance [155,156]. Furthermore, some studies showed no correlation between serum RBP4 levels and expression of RBP4 in adipose tissue [155,157]. Thus, the relationship between adipose RBP4 expression, circulating levels of RBP4, obesity and insulin resistance in humans needs to be evaluated in future studies.

4.9. ANGPTL2

ANGPTL2 was recently identified as an adipocyte-derived inflammatory mediator that promotes inflammation and insulin resistance [158]. Expression of ANGPTL2 in adipose tissue and circulating levels of ANGPTL2 are higher in diet-induced obese mice than in control mice, and circulating levels of ANGPTL2 are closely related to adiposity, insulin resistance, and inflammation in mice [158]. A deficiency of ANGPTL2 improves adipose tissue inflammation and insulin resistance in diet-induced obese mice, whereas its overexpression in adipose tissue promotes inflammation as well as insulin resistance in mice [158]. ANGPTL2 is also closely associated with adiposity and inflammation in humans [158]. Recently, Doi et al. [159] reported that circulating ANGPTL2 levels are positively correlated with the development of type 2 diabetes in humans, and this relationship is independent of other risk factors for type 2 diabetes, including high-sensitivity C-reactive protein levels. Further studies are needed to identify the association of human adipose ANGPTL2 expression with the development of type 2 diabetes.

4.10. Adiponectin

Adiponectin is a well-known adipose-specific adipokine that produces insulin-sensitizing effects. Levels of adiponectin are low in obese subjects, and treatment with adiponectin increases insulin sensitivity in animal models [160,161]. Expression of adiponectin in adipose tissue is lower in subjects with obesity and insulin resistance than in lean subjects and is associated with higher degrees of insulin sensitivity and lower adipose TNF-α expression [162]. A deficiency of adiponectin in mice induces insulin resistance, whereas over-expression of adiponectin in mice improves insulin sensitivity and glucose tolerance [163]. It has been reported that the adiponectin receptors, adiponectin receptor (AdipoR)1 and AdipoR2 are reduced in obesity-related insulin resistance and mediate the anti-metabolic actions of adiponectin [164].

4.11. SFRP5

SFRP5 is a new adipokine with insulin sensitizing and anti-inflammatory properties that exhibits beneficial effects on metabolic dysfunction [165]. The SFRP5 gene and protein are expressed at higher levels in adipose tissue than in other tissues and, in particular, gene expression is confined to adipocytes rather than stromal vascular cells [165]. A deficiency of SFRP5 in mice induced impaired insulin sensitivity, increased risk of developing NAFLD and aggravated adipose inflammation compared with control mice when fed a high-calorie diet, although SFRP5-deficient mice did not show detectable phenotype changes on a regular diet [165]. Conversely, administration of SFRP5 improves metabolic function and reduces adipose inflammation in obese and diabetic mice. The metabolic dysfunction observed in SFRP5-deficient mice is associated with increased accumulation of macrophages and enhanced production of pro-inflammatory cytokines in adipose tissue [165]. Furthermore, in SFRP5-deficient mice, JNK1 loss reverses the impaired insulin sensitivity and increased adipose inflammation [165], suggesting that a deficiency of SFRP5 promotes obesity-induced inflammation and metabolic dysfunction via activation of JNK1 in adipose tissue. Clinical study also demonstrated that plasma levels of SFRP5 were lower in adult subjects with impaired glucose intolerance and type 2 diabetes than normal glucose tolerance subjects, and its levels were negatively correlated with body mass index (BMI), waist-to-hip ratio and HOMA-IR [166,167]. In addition, circulating SFRP5 was associated with obesity and metabolic syndrome in obese children, and its levels were increased after weight loss [168]. However, Carstensen et al. [169] reported a positive correlation between serum SFRP5 levels and parameters of glucose homeostasis or insulin resistance in healthy and obese subjects. Future clinical studies are required to determine the role of adipose SFRP5 in the control of obesity-related abnormalities in glucose homeostasis and insulin sensitivity.

5. Obesity and Dyslipidemia

Obesity is also linked to an increased prevalence of dyslipidemia. Dyslipidemia is an abnormal amount of lipids, such as cholesterol and triglyceride, in the blood and is a widely accepted risk factor for cardiovascular disease. Obesity-related dyslipidemia is primarily characterized by increased levels of plasma free fatty acids and triglycerides, decreased levels of high-density lipoprotein (HDL), and abnormal low-density lipoprotein (LDL) composition (Figure 4). The most significant contributing factor for obesity-related dyslipidemia is likely uncontrolled fatty acid release from adipose tissue, especially visceral adipose tissue, through lipolysis, which causes increased delivery of fatty acids to the liver and synthesis of very-low-density lipoprotein (VLDL). Increased levels of free fatty acids can decrease mRNA expression or activity of lipoprotein lipase (LPL) in adipose tissue and skeletal muscle, and increased synthesis of VLDL in the liver can inhibit lipolysis of chylomicrons, which promotes hypertriglyceridemia [170172]. Hypertriglyceridemia further triggers a cholesterylester transfer protein-mediated exchange of triglycerides for cholesterol esters between triglyceride-rich lipoproteins (VLDL, immediate-density lipoprotein) and lipoproteins, which are relatively richer in cholesterol esters (LDL, HDL), which leads to a decreased HDL-cholesterol concentration and a reduction in triglyceride content in LDL [173]. The increased triglyceride content in LDL is hydrolyzed by hepatic lipase (HL) [173], leading to the formation of small, dense LDL particles that are associated with a higher risk of cardiovascular disease [174]. For decades, in clinical practice, LDL cholesterol has been the cornerstone measurement for assessing cardiovascular risk and is typically estimated using Friedewald formula [175]. As the formula is calculated based on measurements of total cholesterol, triglyceride, HDL cholesterol, the accuracy of Friedewald formula depends on the accuracy of these values. Therefore, recently, the limitation and errors of the Friedewald equation are not well appreciated by clinicians although well-documented. Currently, there are several homogeneous assays for LDL cholesterol based on selective detergents or other elimination methods to separate chylomicrons, VLDL, and HDL from LDL [176]. A homogeneous assay for measurement of small, dense LDL cholesterol has also been developed [177]. The selective measurement of the small, dense LDL cholesterol concentration is crucial for evaluating the actual atherogenic risk of individuals, since a high concentration of small, dense LDL cholesterol is closely related to a high prevalence of cardiovascular disease [178]. Rizzo et al. [179] suggested the predictive role of small, dense LDL beyond traditional cardiovascular risk factors in subjects with metabolic syndrome events, and National Cholesterol Education Program-Adult Treatment Panel III has accepted the sd-LDL as a novel cardiovascular risk factor.
Adipocyte size is suggested to be an important factor for determining the degree to which adipose tissue contributes to dyslipidemia. Enlargement of adipocytes is associated with an increase in lipolysis [180], which leads to further increases in levels of circulating free fatty acids and their delivery to the liver to increase triglyceride synthesis. Along with triglyceride synthesis in the liver, the increased delivery of free fatty acids to the liver exacerbates insulin resistance, which promotes dyslipidemia. Obese subjects have higher whole body fatty acid release compared with lean subjects because of their greater fat mass, although their basal adipose tissue lipolysis per kilogram of fat is lower [13]. A recent study reported the association between enlargement of visceral adipocytes, but not subcutaneous adipocytes, and dyslipidemia independent of body composition and fat distribution in obese subjects [181]. A relationship between visceral adipose tissue and dyslipidemia was also found in patients with type 2 diabetes [182]. The content of visceral adipose tissue is positively correlated with the number of VLDLs and LDLs, even when controlling for BMI and distribution of subcutaneous adipose tissue [182]. Expansion of visceral adipose tissue has also been associated with larger VLDL particles as well as smaller LDL and HDL particles, which have a lower capacity to transfer cholesteryl esters in reverse cholesterol transport and predict atherosclerosis [182]. Visceral adipose tissue has higher lipolytic rates than subcutaneous adipose tissue, and free fatty acids are directly delivered to the liver through the portal vein [183]. Independent of total body fat, the expanded visceral adipose tissue is positively correlated with high hepatic triglyceride lipase activity [184]. A high amount of visceral adipose tissue is also positively correlated with increased HL activity [185] which is associated with increased cardiovascular risk [174].
Many adipose-produced inflammatory molecules, including TNF-α, IL-6, IL-1, serum amyloid A (SAA) and adiponectin, and the number of adipose macrophages also play an important role in the development of dyslipidemia. As noted in the preceding text, obese subjects have higher levels of macrophage infiltration into adipose tissue compared with lean controls, leading to the increased levels of pro-inflammatory cytokines and circulating free fatty acids that are involved in the pathogenesis of dyslipidemia. Macrophage infiltration into visceral adipose tissue is positively correlated with circulating triglyceride levels in obese patients, and a negative relationship has been found with plasma HDL cholesterol levels [186]. Moreover, in subcutaneous adipose tissue, a macrophage-specific marker (CD68) is positively correlated with levels of plasma free fatty acid as well as LDL and negatively correlated with HDL levels [69]. In addition, inflammation can modify the size, composition and function of HDLs, which leads to the impairment of reverse cholesterol transport and parallel changes in apolipoproteins, cholesterol metabolism-related enzymes, anti-oxidant capacity, and adenosine triphosphate binding cassette A1-dependent efflux [187]. Several adipokines also stimulate lipolysis in adipocytes [188,189] and reduce the clearance of triglyceride-rich particles [190192]. For example, IL-6 and TNF-α enhanced lipolysis and suppressed activity of LPL, a key regulatory enzyme in the catabolism and clearance of triglyceride-rich lipoproteins, in adipocytes [188,189,191,192].

6. Role of Adipose Tissue-Produced Adipokines in Dyslipidemia

6.1. Cytokines

TNF-α was originally identified as a factor that induces hypertriglyceridemia in bacteria infected-animals [193]. Levels of plasma TNF-α are higher in hyperlipidemic patients compared with healthy controls and are positively correlated with concentrations of VLDL triglyceride [194]. These effects are related to the promotion of hepatic triglyceride synthesis and secretion [195] as well as inhibition of LPL [196]. In addition, TNF-α directly promotes the overproduction of hepatic apolipoprotein (apo) B100-containing VLDL through impairment of hepatic insulin signaling in animals [197]. Like TNF-α, IL-6 is also associated with hypertriglyceridemia. Subjects with hypertriglyceridemia have a higher production capacity of IL-6 as well as TNF-α [198,199], and increased levels of serum triglycerides are associated with increased levels of IL-6 [200]. Conversely, when anti-inflammatory cytokine (IL-10) levels are increased, plasma triglyceride levels are also increased [201]. Along with TNF-α, pro-inflammatory cytokines, such as IL-6, IL-1, IFN-α and IFN-γ, stimulate triglyceride synthesis in HepG2 cells [202] and/or promote lipolysis in adipocytes [188,189]. In addition, IL-1, IL-6 and IFN-α as well as TNF-α reduce LPL activity in vivo and in vitro [190192].
Levels of serum pro-inflammatory cytokines, including TNF-α and IL-6, are negatively correlated with serum HDL-cholesterol levels in healthy subjects and patients with cardiovascular disease [203205], whereas a positive correlation exists between the anti-inflammatory cytokine (IL-10) concentration and plasma HDL-cholesterol levels [201]. The administration of pro-inflammatory cytokines, such as TNF-α, IL-6 and IL-1, also reduces expression of apo A1 in hepatic cells and plasma in animals [190,206]. Apo A1 is the major protein component of HDL in plasma, and low concentrations of apoA1 are independent predictors for presence and severity of cardiovascular disease [207].
In contrast to HDL cholesterol, pro-inflammatory cytokines, including TNF-α, IL-6 and IL-1, increase circulating total cholesterol and LDL-cholesterol levels in animals by activating cholesterol synthesis [190,208210], whereas increased IL-10 levels are negatively associated with high levels of total cholesterol and LDL cholesterol [201]. Pro-inflammatory cytokines (TNF-α, transforming growth factor-β or IL-1) not only promote lipoprotein uptake by scavenger receptor and LDL receptor but also inhibit adenosine triphosphate binding cassette transporter A1 (ABCA1)-mediated cholesterol efflux to HDL, which may contribute to lipid deposition and foam cell formation [211213]. In addition, TNF-α increases secretion of apo B in rat hepatocyte cultures in the absence of extracellular fatty acids [214]. The level of circulating apo B has been reported to be a strong predictor of coronary artery disease compared with circulating HDL-cholesterol levels [215]. Moreover, in HepG2 cells, the treatment of cytokines (TNF-α, IL-6 and IL-1) stimulates the hepatic production and secretion of phospholipase A2 [216] which accelerate the development of atherosclerosis [217].

6.2. SAA

SAA is an apolipoprotein that can replace apo A1 as the major apolipoprotein of HDL [218]. SAA is found in the adipose tissue and the liver in humans [34,219], and mice mainly express it in adipocytes [220]. Expression of SAA in adipose tissue and circulating levels of SAA are higher in obese subjects than in lean subjects and are decreased by caloric restriction [219]. A number of studies have suggested a role of SAA in the inflammatory process [221,222]. Treatment with SAA increases expression of the pro-inflammatory cytokines IL-6 and TNF-α in preadipocytes and adipocytes in vitro [221,222]. In addition, SAA affects the metabolism of HDL cholesterol through its inhibitory effects on HDL binding and selective lipid uptake mediated by scavenger receptor SR-BI [223], an HDL receptor that mediates the cellular uptake of cholesteryl esters from HDL, thereby promoting the process of reverse cholesterol transport from the periphery to the liver [224]. Lewis et al. [225] suggested that SAA might be a potential contributor to atherosclerosis directly by mediating retention of SAA-enriched HDL to vascular proteoglycans, independent of an adverse effect on plasma lipoproteins. Thus, increased expression of SAA can promote dyslipidemia by affecting the HDL structure and function as well as inflammation.

6.3. Adiponectin

Adiponectin has beneficial effects on lipid metabolism and also plays a role as a vasoprotective adipokine [226]. Levels of plasma adiponectin have been negatively correlated with triglycerides and positively correlated with HDL cholesterol [71]. Decreased adiponectin levels are associated with dyslipidemia and cardiovascular disease compared with matched controls [227,228]. Adiponectin stimulates fatty acid oxidation and glucose utilization through activation of AMPK in the liver and skeletal muscle, which has been associated with many of the positive effects of adiponectin on lipoprotein metabolism as well as insulin sensitivity [229]. Adiponectin also induces activation of LPL, thereby enhancing VLDL clearance and reducing plasma triglyceride levels [230]. In addition, in subjects with type 2 diabetes and normal controls, low levels of adiponectin have been related to increased HL activity, which may be responsible for the decreased levels of HDL cholesterol [231]. More recently, Matsuura et al. [232] reported that adiponectin increases the mRNA expression and secretion of apo A1 as well as ABCA1 mRNA and protein expression in HepG2 cells, suggesting that adiponectin might increase HDL assembly in the liver. In adiponectin-knockout mice, plasma and hepatic apo A1 protein levels and hepatic ABCA1 gene and protein expression were shown to be decreased compared with wild-type-mice [233]. Recently, Chang et al. [234] suggested that hypoadiponectinemia may be a useful marker of dyslipidemia in subjects with polycystic ovarian syndrome, who have an increased risk of dyslipidemia.

7. Obesity and NAFLD

NAFLD is currently the most common form of chronic liver disease [235], and its incidence has increased in parallel to the rise in the incidence of obesity [3,236]. More than two-thirds of patients with NAFLD are obese [237]. NAFLD is characterized by two steps of liver injury: (1) accumulation of triglycerides in the liver (hepatic steatosis) and (2) inflammation and subsequent fibrosis (nonalcoholic steatohepatitis, NASH) [238]. The “two-hit” hypothesis is widely accepted to explain the development of NAFLD and the progression from simple steatosis to NASH [239]. The “first hit” is the accumulation of hepatic lipids, and the “second hit” promotes hepatocyte injury, inflammation and fibrosis. A number of factors, including proinflammatory cytokines, adipokines, mitochondrial dysfunction, oxidative stress and subsequent lipid peroxidation, initiate the second hit [239]. The classical “two-hit” hypothesis has now been modified by “multi-hit” hypothesis due to involvement of complex factors and interactions leading from lipid dysregulation, adipokine imbalance, adipose inflammation, oxidative stress, insulin resistance to NAFLD [240,241] (Figure 5). In the “multi-hit” hypothesis, imbalanced lipid metabolism and insulin resistance is considered as the “first hit”. Hyperinsulinemia, caused by insulin resistance, results in steatosis via increased de novo hepatic lipogenesis, decreased free fatty acid oxidation, decreased hepatic VLDL secretion and increased efflux of free fatty acids due to increased lipolysis from adipose tissue. After the development of steatosis, liver becomes more vulnerable to “multi-hit” including the gut-derived bacterial toxins, adipokine/cytokine imbalance, mitochondrial dysfunction, oxidative damage, dysregulated hepatocyte apoptosis, release of pro-fibrogenic factors and pro-inflammatory mediators from impaired organelles and activation of hepatic stellate cell and Kupffer cell. Such multiple factors may collectively stimulate inflammation, apoptosis and fibrosis that ultimately leading to progressive liver disease.
Excessive lipid accumulation in the liver generally occurs when the influx of lipids, via increased fatty acid import or de novo fatty acid synthesis, exceeds the ability of hepatic lipid clearance by fatty acid oxidation or triglyceride export [242,243]. Recent study has confirmed that increased de novo lipogenesis is a distinct characteristic of subjects with NAFLD [244]. As described in the preceding text, adipose tissue is suggested to be a source of free fatty acids and other factors entering the portal circulation [183,245]. Expanded adipose tissue promotes macrophage infiltration and secretion of many pro-inflammatory chemokines, cytokines and adipokines that are closely related to insulin resistance [5,6,40]. A failure to suppress lipolysis by insulin then results in increased release of free fatty acids from adipose tissue [246,247]. The increased lipolysis in adipose tissue, especially visceral adipose tissue [13,180,248], increases free fatty acid influx directly into the liver by the portal vein [246]. The free fatty acids from enlarged adipose tissue are then taken up by the hepatocytes, which lead to reduced hepatic insulin clearance with a further increase in circulating insulin levels [53]. In the liver, free fatty acids promote increased glucose production and triglyceride synthesis and impair insulin suppression of hepatic glucose output [53]. In addition, free fatty acids are ligands of the membrane-bound TLR4 and can promote inflammation [24,28]. However, it is still unclear to what extent the portally drained adipose tissue influences hepatic steatosis. The contribution of visceral adipose tissue lipolysis to the delivery of hepatic free fatty acids has been shown to be only 5%–10% in normal-weight subjects and up to 25% in intra-abdominally obese subjects [245]; however, in the fasting state, hepatic fatty acids originate predominantly from circulating free fatty acids [249] and delivery of the portal free fatty acids to the liver is increased postprandially [250]. Nevertheless, increased free fatty acid influx is a key contributor to promoting accumulation of lipids in the liver, irrespective of the origin of the free fatty acids [247,251].
In the obese state, pro-inflammatory and anti-inflammatory factors secreted by inflamed adipose tissue are also associated with NAFLD [247]. Among them, adiponectin is suggested to protect the liver from steatosis and inflammation. In the liver, adiponectin increases the ability of insulin to suppress glucose production and glucose output [160]. Moreover, it inhibits hepatic lipogenesis by down-regulating the lipogenic transcription factor, SREBP1-c [252] and promotes glucose utilization and fatty-acid oxidation in the liver by activating AMPK [229]. These findings are supported by studies in which recombinant adiponectin given to obese mice not only ameliorated hepatomegaly, hepatic steatosis and inflammation but also normalized levels of alanine aminotransferase (ALT) [253] which is a sensitive indicator of liver injury and often used as a surrogate marker for NAFLD [254]. In addition to its metabolic effects, adiponectin has anti-inflammatory activities that might protect the progression of hepatic steatosis to fibrosis. In KK-Ay obese mice, adiponectin attenuated LPS-induced liver injury by decreasing TNF-α levels and activating peroxisome proliferator-activated receptor (PPAR)α in the liver [255]. Moreover, liver fibrosis induced by the administration of carbon tetrachloride was enhanced in adiponectin-deficient mice, whereas injection of adiponectin attenuated liver fibrosis in wild-type mice treated with carbon tetrachloride [256]. In accordance with animal studies, a number of clinical studies have suggested a protective role of adiponectin in NAFLD. Circulating adiponectin levels are lower in subjects with NAFLD than in healthy controls [257] and negatively correlated with liver function markers in healthy subjects [258]. Similarly, low adiponectin levels predict hepatic steatosis and increased liver injury enzyme levels in obese subjects [259]. In addition, expression of adiponectin and its receptor (AdipoR2) is significantly reduced in the liver of patients with NASH compared with those with simple steatosis [260]. Polymorphisms in the gene encoding AdipoR1 are also associated with hepatic steatosis in human [261].
Leptin is regarded as another key regulator of NAFLD. It directly stimulates AMPK which is involved in activation of lipid oxidation, such as β-oxidation and glycolysis, as well as inhibition of lipogenesis [262]. Expression of SREBP1-c is increased in the liver of leptin-unresponsive fa/fa Zucker diabetic fatty rats [263], and infusion of adenovirus-leptin not only decreases hepatic triglyceride synthesis but also increases β-oxidation through down-regulation of SREBP1-c and up-regulation of PPARα [264]. Moreover, a negative correlation between serum leptin levels and hepatic injury has been observed in humans [265]. Conversely, several clinical studies have reported that the concentration of circulating leptin is positively correlated with high serum ALT or hepatic steatosis, independent of BMI and body fat mass [266,267]. Leptin also increases hepatic fibrosis, whereas a deficiency of leptin is related to the decreased hepatic injury in animal models [268]. Leptin enhances the expression of pro-fibrogenic cytokine (transforming growth factor-β1) in Kupffer cells [269,270] and has a direct action on hepatic fibrogenesis by activating hepatic stellate cells and stimulating production of α-smooth muscle actin, collagen and tissue inhibitor of metalloproteinase 1 [267,269]. Leptin has been reported to be a potent hepatic stellate cell mitogen and inhibit hepatic stellate cell apoptosis, which promotes the pathogenesis of liver fibrosis [270].
In animal models, resistin also regulates glucose and lipid metabolism in the liver and acts as a mediator of hepatic insulin resistance. Circulating levels of resistin are increased in patients with NAFLD [271,272]. When NAFLD patients is divided according to liver histology (pure fatty liver vs. NASH), serum resistin levels are higher in patients with NASH than in those with pure fatty liver and positively correlated with the NASH score, an index that takes into account necrosis, inflammation, and fibrosis in liver biopsies and reflects the severity of the disease. However, the role of resistin in humans remains uncertain. In addition, TNF-α mediates not only the early stages of NAFLD but also the transition to more advanced stages of liver damage in animals and human, suggesting TNF-α has been proposed to play a key role in the development of NASH/NAFLD [273275]. Moreover, IL-6 and TNF-α increase expression of SOCS in the liver which is involved in increased hepatic SREBP-1c expression and insulin resistance [276]. Acylation-stimulating protein and angiotensinogen have also been observed in adipose tissue, and angiotensinogen levels are increased in obese subjects [277279]. The levels of acylation-stimulating protein correlate with insulin resistance in NAFLD [277], and angiotensin II antagonists have been shown to improve liver function test results in patients with NAFLD and attenuated fibrosis in animal models [280].

8. Conclusions

Obesity, especially visceral obesity, is associated with metabolic disturbances, such as insulin resistance, dyslipidemia and NAFLD. Enlarged adipose tissue results in the infiltration of macrophages and unbalance of pro-inflammatory and anti-inflammatory factors secreted by adipose tissue, which lead to the promotion of inflammation, impairment of insulin sensitivity and dysregulation of lipid metabolism. Excess free fatty acids also contribute to the initiation and progression of obesity-induced metabolic complications. The adipose tissue can affect many other tissues, including the liver, skeletal muscle and heart, via the production of free fatty acids and many pro-inflammatory and anti-inflammatory factors, and therefore has a critical role in the pathogenesis of insulin resistance, dyslipidemia and NAFLD. Although the cause-and-effect association has not been definitively established, available evidence have provided great insight into the critical role of adipose tissue in metabolic syndrome. Thus, further elucidation of the functions and mechanisms of adipose tissue-released bioactive substances will lead to a better understanding of the development of obesity-related metabolic syndrome, and it may provide novel therapeutic approaches to prevent or treat obesity and its metabolic complications. In addition, it will be also worthwhile to focus on how each adipokine signaling pathway integrates with multiple intracellular signaling cascades activated by other factors in the adipose tissue and other tissues.

Acknowledgments

This research was supported by the SRC program (Center for Food & Nutritional Genomics: No. 2012–0000644) through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology. This work was also supported by the Bio-Synergy Research Project (NRF-2012M3A9C4048818) of the Ministry of Science, ICT and Future Planning through the National Research Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Obesity and Overweight. Available online: http://www.who.int/mediacentre/factsheets/fs311/en/ (accessed on 5 May 2013).
  2. Wang, Y.C.; McPherson, K.; Marsh, T.; Gortmaker, S.L.; Brown, M. Health and economic burden of the projected obesity trends in the USA and the UK. Lancet 2011, 378, 815–825. [Google Scholar]
  3. López-Velázquez, J.A.; Silva-Vidal, K.V.; Ponciano-Rodríguez, G.; Chávez-Tapia, N.C.; Arrese, M.; Uribe, M.; Méndez-Sánchez, N. The prevalence of nonalcoholic fatty liver disease in the Americas. Ann. Hepatol 2014, 13, 166–178. [Google Scholar]
  4. Carr, D.B.; Utzschneider, K.M.; Hull, R.L.; Kodama, K.; Retzlaff, B.M.; Brunzell, J.D.; Shofer, J.B.; Fish, B.E.; Knopp, R.H.; Kahn, S.E. Intra-abdominal fat is a major determinant of the National Cholesterol Education Program Adult Treatment Panel III criteria for the metabolic syndrome. Diabetes 2004, 53, 2087–2094. [Google Scholar]
  5. Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Investig 2003, 112, 1821–1830. [Google Scholar]
  6. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar]
  7. Lumeng, C.N.; Saltiel, A.R. Inflammatory links between obesity and metabolic disease. J. Clin. Investig 2011, 121, 2111–2117. [Google Scholar]
  8. Kershaw, E.E.; Flier, J.S. Adipose tissue as an endocrine organ. J. Clin. Endocrinol. Metab 2004, 89, 2548–2556. [Google Scholar]
  9. Hauner, H. Secretory factors from human adipose tissue and their functional role. Proc. Nutr. Soc 2005, 64, 163–169. [Google Scholar]
  10. Halberg, N.; Wernstedt-Asterholm, I.; Scherer, P.E. The adipocyte as an endocrine cell. Endocrinol. Metab. Clin. N. Am 2008, 37, 753–768. [Google Scholar]
  11. Perseghin, G.; Ghosh, S.; Gerow, K.; Shulman, G.I. Metabolic defects in lean nondiabetic offspring of NIDDM parents: A cross-sectional study. Diabetes 1997, 46, 1001–1009. [Google Scholar]
  12. Boden, G. Obesity and free fatty acids. Endocrinol. Metab. Clin. N. Am 2008, 37, 635–646. [Google Scholar]
  13. Horowitz, J.F.; Coppack, S.W.; Paramore, D.; Cryer, P.E.; Zhao, G.; Klein, S. Effect of short-term fasting on lipid kinetics in lean and obese women. Am. J. Physiol 1999, 276, E278–E284. [Google Scholar]
  14. Large, V.; Reynisdottir, S.; Langin, D.; Fredby, K.; Klannemark, M.; Holm, C.; Arner, P. Decreased expression and function of adipocyte hormone-sensitive lipase in subcutaneous fat cells of obese subjects. J. Lipid Res 1999, 40, 2059–2066. [Google Scholar]
  15. Hellström, L.; Reynisdottir, S. Influence of heredity for obesity on adipocyte lipolysis in lean and obese subjects. Int. J. Obes. Relat. Metab. Disord 2000, 24, 340–344. [Google Scholar]
  16. McQuaid, S.E.; Hodson, L.; Neville, M.J.; Dennis, A.L.; Cheeseman, J.; Humphreys, S.M.; Ruge, T.; Gilbert, M.; Fielding, B.A.; Frayn, K.N.; et al. Downregulation of adipose tissue fatty acid trafficking in obesity: A driver for ectopic fat deposition? Diabetes 2011, 60, 47–55. [Google Scholar]
  17. Langin, D.; Dicker, A.; Tavernier, G.; Hoffstedt, J.; Mairal, A.; Rydén, M.; Arner, E.; Sicard, A.; Jenkins, C.M.; Viguerie, N.; et al. Adipocyte lipases and defect of lipolysis in human obesity. Diabetes 2005, 54, 3190–3197. [Google Scholar]
  18. Jocken, J.W.; Langin, D.; Smit, E.; Saris, W.H.; Valle, C.; Hul, G.B.; Holm, C.; Arner, P.; Blaak, E.E. Adipose triglyceride lipase and hormone-sensitive lipase protein expression is decreased in the obese insulin-resistant state. J. Clin. Endocrinol. Metab 2007, 92, 2292–2299. [Google Scholar]
  19. Karpe, F.; Dickmann, J.R.; Frayn, K.N. Fatty acids, obesity, and insulin resistance: Time for a reevaluation. Diabetes 2011, 60, 2441–2449. [Google Scholar]
  20. Hausman, D.B.; DiGirolamo, M.; Bartness, T.J.; Hausman, G.J.; Martin, R.J. The biology of white adipocyte proliferation. Obes. Rev 2001, 2, 239–254. [Google Scholar]
  21. Spalding, K.L.; Arner, E.; Westermark, P.O.; Bernard, S.; Buchholz, B.A.; Bergmann, O.; Blomqvist, L.; Hoffstedt, J.; Naslund, E.; Britton, T.; et al. Dynamics of fat cell turnover in humans. Nature 2008, 453, 783–787. [Google Scholar]
  22. Tchoukalova, Y.D.; Votruba, S.B.; Tchkonia, T.; Giorgadze, N.; Kirkland, J.L.; Jensen, M.D. Regional differences in cellular mechanisms of adipose tissue gain with overfeeding. Proc. Natl. Acad. Sci. USA 2010, 107, 18226–18231. [Google Scholar]
  23. Wang, Y.; Rimm, E.B.; Stampfer, M.J.; Willett, W.C.; Hu, F.B. Comparison of abdominal adiposity and overall obesity in predicting risk of type 2 diabetes among men. Am. J. Clin. Nutr 2005, 81, 555–563. [Google Scholar]
  24. Kim, J.Y.; van de Wall, E.; Laplante, M.; Azzara, A.; Trujillo, M.E.; Hofmann, S.M.; Schraw, T.; Durand, J.L.; Li, H.; Li, G.; et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J. Clin. Investig 2007, 117, 2621–2637. [Google Scholar]
  25. Weyer, C.; Foley, J.E.; Bogardus, C.; Tataranni, P.A.; Pratley, R.E. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia 2000, 43, 1498–1506. [Google Scholar]
  26. Boden, G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 1997, 46, 3–10. [Google Scholar]
  27. Kelley, D.E.; Mokan, M.; Simoneau, J.A.; Mandarino, L.J. Interaction between glucose and free fatty acid metabolism in human skeletal muscle. J. Clin. Investig 1993, 92, 91–98. [Google Scholar]
  28. Shi, H.; Kokoeva, M.V.; Inouye, K.; Tzameli, I.; Yin, H.; Flier, J.S. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Investig 2006, 116, 3015–3025. [Google Scholar]
  29. Suganami, T.; Nishida, J.; Ogawa, Y. A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: Role of free fatty acids and tumor necrosis factor alpha. Arterioscler. Thromb. Vasc. Biol 2005, 25, 2062–2068. [Google Scholar]
  30. Byrne, C.D.; Maison, P.; Halsall, D.; Martensz, N.; Hales, C.N.; Wareham, N.J. Cross-sectional but not longitudinal associations between non-esterified fatty acid levels and glucose intolerance and other features of the metabolic syndrome. Diabet. Med 1999, 16, 1007–1015. [Google Scholar]
  31. Cho, S.J.; Jung, U.J.; Choi, M.S. Differential effects of low-dose resveratrol on adiposity and hepatic steatosis in diet-induced obese mice. Br. J. Nutr 2012, 108, 2166–2175. [Google Scholar]
  32. Charles, M.A.; Fontbonne, A.; Thibult, N.; Claude, J.R.; Warnet, J.M.; Rosselin, G.; Ducimetière, P.; Eschwège, E. High plasma nonesterified fatty acids are predictive of cancer mortality but not of coronary heart disease mortality: Results from the paris prospective study. Am. J. Epidemiol 2001, 153, 292–298. [Google Scholar]
  33. Reeds, D.N.; Stuart, C.A.; Perez, O.; Klein, S. Adipose tissue, hepatic, and skeletal muscle insulin sensitivity in extremely obese subjects with acanthosis nigricans. Metabolism 2006, 55, 1658–1663. [Google Scholar]
  34. Jernas, M.; Palming, J.; Sjoholm, K.; Jennische, E.; Svensson, P.A.; Gabrielsson, B.G.; Levin, M.; Sjogren, A.; Rudemo, M.; Lystig, T.C.; et al. Separation of human adipocytes by size: Hypertrophic fat cells display distinct gene expression. FASEB J 2006, 20, 1540–1542. [Google Scholar]
  35. Skurk, T.; Alberti-Huber, C.; Herder, C.; Hauner, H. Relationship between adipocyte size and adipokine expression and secretion. J. Clin. Endocrinol. Metab 2007, 92, 1023–1033. [Google Scholar]
  36. Hotamisligil, G.S.; Shargill, N.S.; Spiegelman, B.M. Adipose expression of tumor necrosis factor-α: Direct role in obesity-linked insulin resistance. Science 1993, 259, 87–91. [Google Scholar]
  37. Amrani, A.; Jafarian-Tehrani, M.; Mormède, P.; Durant, S.; Pleau, J.M.; Haour, F.; Dardenne, M.; Homo-Delarche, F. Interleukin-1 effect on glycemia in the non-obese diabetic mouse at the pre-diabetic stage. J. Endocrinol 1996, 148, 139–148. [Google Scholar]
  38. Sartipy, P.; Loskutoff, D.J. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc. Natl. Acad. Sci. USA 2003, 100, 7265–7270. [Google Scholar]
  39. Rotter, V.; Nagaev, I.; Smith, U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J. Biol. Chem 2003, 278, 45777–45784. [Google Scholar]
  40. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig 2003, 112, 1796–1808. [Google Scholar]
  41. Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Investig 2007, 117, 175–184. [Google Scholar]
  42. Jiao, P.; Chen, Q.; Shah, S.; Du, J.; Tao, B.; Tzameli, I.; Yan, W.; Xu, H. Obesity-related upregulation of monocyte chemotactic factors in adipocytes: Involvement of nuclear factor-kappaB and c-Jun NH2-terminal kinase pathways. Diabetes 2009, 58, 104–115. [Google Scholar]
  43. Lee, Y.H.; Thacker, R.I.; Hall, B.E.; Kong, R.; Granneman, J.G. Exploring the activated adipogenic niche: Interactions of macrophages and adipocyte progenitors. Cell Cycle 2014, 13, 184–190. [Google Scholar]
  44. Nomiyama, T.; Perez-Tilve, D.; Ogawa, D.; Gizard, F.; Zhao, Y.; Heywood, E.B.; Jones, K.L.; Kawamori, R.; Cassis, L.A.; Tschöp, M.H.; et al. Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J. Clin. Investig 2007, 117, 2877–2888. [Google Scholar]
  45. Weisberg, S.P.; Hunter, D.; Huber, R.; Lemieux, J.; Slaymaker, S.; Vaddi, K.; Charo, I.; Leibel, R.L.; Ferrante, A.W., Jr. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Investig 2006, 116, 115–124. [Google Scholar]
  46. Nara, N.; Nakayama, Y.; Okamoto, S.; Tamura, H.; Kiyono, M.; Muraoka, M.; Tanaka, K.; Taya, C.; Shitara, H.; Ishii, R.; et al. Disruption of CXC motif chemokine ligand-14 in mice ameliorates obesity-induced insulin resistance. J. Biol. Chem 2007, 282, 30794–30803. [Google Scholar]
  47. Feng, B.; Jiao, P.; Nie, Y.; Kim, T.; Jun, D.; van Rooijen, N.; Yang, Z.; Xu, H. Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS One 2011, 6, e24358. [Google Scholar]
  48. Clément, K.; Viguerie, N.; Poitou, C.; Carette, C.; Pelloux, V.; Curat, C.A.; Sicard, A.; Rome, S.; Benis, A.; Zucker, J.D.; et al. Weight loss regulates inflammation-related genes in white adipose tissue of obese subjects. FASEB J 2004, 18, 1657–1669. [Google Scholar]
  49. Cancello, R.; Henegar, C.; Viguerie, N.; Taleb, S.; Poitou, C.; Rouault, C.; Coupaye, M.; Pelloux, V.; Hugol, D.; Bouillot, J.L.; 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 2005, 54, 2277–2286. [Google Scholar]
  50. Schipper, H.S.; Prakken, B.; Kalkhoven, E.; Boes, M. Adipose tissue-resident immune cells: Key players in immunometabolism. Trends. Endocrinol. Metab 2012, 23, 407–415. [Google Scholar]
  51. Arita, Y.; Kihara, S.; Ouchi, N.; Takahashi, M.; Maeda, 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 1999, 257, 79–83. [Google Scholar]
  52. Lillioja, S.; Mott, D.M.; Spraul, M.; Ferraro, R.; Foley, J.E.; Ravussin, E.; Knowler, W.C.; Bennett, P.H.; Bogardus, C. Insulin resistance and insulin secretory dysfunction as precursors of non-insulin-dependent diabetes mellitus. N. Engl. J. Med 1993, 329, 1988–1992. [Google Scholar]
  53. Kahn, B.B.; Flier, J.S. Obesity and insulin resistance. J. Clin. Investig 2000, 106, 473–481. [Google Scholar]
  54. Schenk, S.; Saberi, M.; Olefsky, J.M. Insulin sensitivity: Modulation by nutrients and inflammation. J. Clin. Investig 2008, 118, 2992–3002. [Google Scholar]
  55. Holland, W.L.; Brozinick, J.T.; Wang, L.P.; Hawkins, E.D.; Sargent, K.M.; Liu, Y.; Narra, K.; Hoehn, K.L.; Knotts, T.A.; Siesky, A.; et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metab 2007, 5, 167–179. [Google Scholar]
  56. Dressler, K.A.; Mathias, S.; Kolesnick, R.N. Tumor necrosis factor-alpha activates the sphingomyelin signal transduction pathway in a cell-free system. Science 1992, 255, 1715–1718. [Google Scholar]
  57. Teruel, T.; Hernandez, R.; Lorenzo, M. Ceramide mediates insulin resistance by tumor necrosis factor-alpha in brown adipocytes by maintaining Akt in an inactive dephosphorylated state. Diabetes 2001, 50, 2563–2571. [Google Scholar]
  58. Haus, J.M.; Kashyap, S.R.; Kasumov, T.; Zhang, R.; Kelly, K.R.; Defronzo, R.A.; Kirwan, J.P. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 2009, 58, 337–343. [Google Scholar]
  59. Holland, W.L.; Bikman, B.T.; Wang, L.P.; Yuguang, G.; Sargent, K.M.; Bulchand, S.; Knotts, T.A.; Shui, G.; Clegg, D.J.; Wenk, M.R.; et al. Lipid-induced insulin resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. J. Clin. Investig 2011, 121, 1858–1870. [Google Scholar]
  60. Horowitz, J.F.; Klein, S. Whole body and abdominal lipolytic sensitivity to epinephrine is suppressed in upper body obese women. Am. J. Physiol. Endocrinol. Metab 2000, 278, E1144–E1152. [Google Scholar]
  61. Bajaj, M.; Suraamornkul, S.; Romanelli, A.; Cline, G.W.; Mandarino, L.J.; Shulman, G.I.; DeFronzo, R.A. Effect of a sustained reduction in plasma free fatty acid concentration on intramuscular long-chain fatty Acyl-CoAs and insulin action in type 2 diabetic patients. Diabetes 2005, 54, 3148–3153. [Google Scholar]
  62. Santomauro, A.T.; Boden, G.; Silva, M.E.; Rocha, D.M.; Santos, R.F.; Ursich, M.J.; Strassmann, P.G.; Wajchenberg, B.L. Overnight lowering of free fatty acids with Acipimox improves insulin resistance and glucose tolerance in obese diabetic and nondiabetic subjects. Diabetes 1999, 48, 1836–1841. [Google Scholar]
  63. Girousse, A.; Tavernier, G.; Valle, C.; Moro, C.; Mejhert, N.; Dinel, A.L.; Houssier, M.; Roussel, B.; Besse-Patin, A.; Combes, M.; et al. Partial inhibition of adipose tissue lipolysis improves glucose metabolism and insulin sensitivity without alteration of fat mass. PLoS Biol 2013, 11, e1001485. [Google Scholar]
  64. Kosteli, A.; Sugaru, E.; Haemmerle, G.; Martin, J.F.; Lei, J.; Zechner, R.; Ferrante, A.W., Jr. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. J. Clin. Investig 2010, 120, 3466–3479. [Google Scholar]
  65. Apovian, C.M.; Bigornia, S.; Mott, M.; Meyers, M.R.; Ulloor, J.; Gagua, M.; McDonnell, M.; Hess, D.; Joseph, L.; Gokce, N. Adipose macrophage infiltration is associated with insulin resistance and vascular endothelial dysfunction in obese subjects. Arterioscler. Thromb. Vasc. Biol 2008, 28, 1654–1659. [Google Scholar]
  66. Anderson, E.K.; Gutierrez, D.A.; Hasty, A.H. Adipose tissue recruitment of leukocytes. Curr. Opin. Lipidol 2010, 21, 172–177. [Google Scholar]
  67. Fain, J.N. Release of inflammatory mediators by human adipose tissue is enhanced in obesity and primarily by the nonfat cells: A review. Mediat. Inflamm 2010, 2010, 513948. [Google Scholar]
  68. Ouchi, N.; Parker, J.L.; Lugus, J.J.; Walsh, K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol 2011, 11, 85–97. [Google Scholar]
  69. Huber, J.; Kiefer, F.W.; Zeyda, M.; Ludvik, B.; Silberhumer, G.R.; Prager, G.; Zlabinger, G.J.; Stulnig, T.M. CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. J. Clin. Endocrinol. Metab 2008, 93, 3215–3221. [Google Scholar]
  70. Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte chemoattractant protein-1 (MCP-1): An overview. J. Interferon Cytokine Res 2009, 29, 313–326. [Google Scholar]
  71. Kanda, H.; Tateya, S.; Tamori, Y.; Kotani, K.; Hiasa, K.; Kitazawa, R.; Kitazawa, S.; Miyachi, H.; Maeda, S.; Egashira, K.; et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Investig 2006, 116, 1494–1505. [Google Scholar]
  72. Kamei, N.; Tobe, K.; Suzuki, R.; Ohsugi, M.; Watanabe, T.; Kubota, N.; Ohtsuka-Kowatari, N.; Kumagai, K.; Sakamoto, K.; Kobayashi, M.; et al. Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J. Biol. Chem 2006, 281, 26602–26614. [Google Scholar]
  73. Lumeng, C.N.; DelProposto, J.B.; Westcott, D.J.; Saltiel, A.R. Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 2008, 57, 3239–3246. [Google Scholar]
  74. Meijer, K.; de Vries, M.; Al-Lahham, S.; Bruinenberg, M.; Weening, D.; Dijkstra, M.; Kloosterhuis, N.; van der Leij, R.J.; van der Want, H.; Kroesen, B.J.; et al. Human primary adipocytes exhibit immune cell function: Adipocytes prime inflammation independent of macrophages. PLoS One 2011, 6, e17154. [Google Scholar]
  75. Simeoni, E.; Hoffmann, M.M.; Winkelmann, B.R.; Ruiz, J.; Fleury, S.; Boehm, B.O.; März, W.; Vassalli, G. Association between the A-2518G polymorphism in the monocyte chemoattractant protein-1 gene and insulin resistance and Type 2 diabetes mellitus. Diabetologia 2004, 47, 1574–1580. [Google Scholar]
  76. Karadeniz, M.; Erdogan, M.; Cetinkalp, S.; Berdeli, A.; Eroglu, Z.; Ozgen, A.G. Monocyte chemoattractant protein-1 (MCP-1) 2518G/A gene polymorphism in Turkish type 2 diabetes patients with nephropathy. Endocrine 2010, 37, 513–517. [Google Scholar]
  77. Jing, Y.; Zhu, D.; Bi, Y.; Yang, D.; Hu, Y.; Shen, S. Monocyte chemoattractant protein 1–2518 A/G polymorphism and susceptibility to type 2 diabetes in a Chinese population. Clin. Chim. Acta 2011, 412, 466–469. [Google Scholar]
  78. Zietz, B.; Büchler, C.; Herfarth, H.; Müller-Ladner, U.; Spiegel, D.; Schölmerich, J.; Schäffler, A. Caucasian patients with type 2 diabetes mellitus have elevated levels of monocyte chemoattractant protein-1 that are not influenced by the −2518 A→G promoter polymorphism. Diabetes Obes. Metab 2005, 7, 570–578. [Google Scholar]
  79. Katakami, N.; Matsuhisa, M.; Kaneto, H.; Matsuoka, T.A.; Imamura, K.; Ishibashi, F.; Kanda, T.; Kawai, K.; Osonoi, T.; Kashiwagi, A.; et al. Monocyte chemoattractant protein-1 (MCP-1) gene polymorphism as a potential risk factor for diabetic retinopathy in Japanese patients with type 2 diabetes. Diabetes Res. Clin. Pract 2010, 89, e9–e12. [Google Scholar]
  80. Keophiphath, M.; Rouault, C.; Divoux, A.; Clement, K.; Lacasa, D. CCL5 promotes macrophage recruitment and survival in human adipose tissue. Arterioscler. Thromb. Vasc. Biol 2009, 30, 39–45. [Google Scholar]
  81. Chavey, C.; Lazennec, G.; Lagarrigue, S.; Clape, C.; Iankova, I.; Teyssier, J.; Annicotte, J.S.; Schmidt, J.; Mataki, C.; Yamamoto, H.; et al. CXC ligand 5 is an adipose-tissue derived factor that links obesity to insulin resistance. Cell Metab 2009, 9, 339–349. [Google Scholar]
  82. Tourniaire, F.; Romier-Crouzet, B.; Lee, J.H.; Marcotorchino, J.; Gouranton, E.; Salles, J.; Malezet, C.; Astier, J.; Darmon, P.; Blouin, E.; et al. Chemokine Expression in Inflamed Adipose Tissue Is Mainly Mediated by NF-κB. PLoS One 2013, 8, e66515. [Google Scholar]
  83. Ruan, H.; Lodish, H.F. Insulin resistance in adipose tissue: Direct and indirect effects of tumor necrosis factor-α. Cytokine Growth Factor Rev 2003, 14, 447–455. [Google Scholar]
  84. Uysal, K.T.; Wiesbrock, S.M.; Marino, M.W.; Hotamisligil, G.S. Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature 1997, 389, 610–614. [Google Scholar]
  85. Miyazaki, Y.; Pipek, R.; Mandarino, L.J.; DeFronzo, R.A. Tumor necrosis factor α and insulin resistance in obese type 2 diabetic patients. Int. J. Obes. Relat. Metab. Disord 2003, 27, 88–94. [Google Scholar]
  86. Wascher, T.C.; Lindeman, J.H.; Sourij, H.; Kooistra, T.; Pacini, G.; Roden, M. Chronic TNF-α neutralization does not improve insulin resistance or endothelial function in “healthy” men with metabolic syndrome. Mol. Med 2011, 17, 189–193. [Google Scholar]
  87. Bernstein, L.E.; Berry, J.; Kim, S.; Canavan, B.; Grinspoon, S.K. Effects of etanercept in patients with the metabolic syndrome. Arch. Intern. Med 2006, 166, 902–908. [Google Scholar]
  88. McArdle, M.A.; Finucane, O.M.; Connaughton, R.M.; McMorrow, A.M.; Roche, H.M. Mechanisms of obesity-induced inflammation and insulin resistance: Insights into the emerging role of nutritional strategies. Front. Endocrinol. (Lausanne) 2013, 4, 52. [Google Scholar]
  89. Illei, G.G.; Lipsky, P.E. Novel, antigen-specific therapeutic approaches to autoimmuneinflammatory diseases. Curr. Opin. Immunol 2000, 12, 712–718. [Google Scholar]
  90. Hodge-Dufour, J.; Marino, M.W.; Horton, M.R.; Jungbluth, A.; Burdick, R.D.; Strieter, R.M.; Noble, P.W.; Hunter, C.A.; Pure, E. Inhibition of interferon gamma induced interleukin 12 production—A potential mechanism for the anti-inflammatory activities of tumor necrosis factor. Proc. Natl. Acad. Sci. USA 1998, 95, 13806–13811. [Google Scholar]
  91. O’Rourke, R.W.; White, A.E.; Metcalf, M.D.; Winters, B.R.; Diggs, B.S.; Zhu, X.; Marks, D.L. Systemic inflammation and insulin sensitivity in obese IFN-γ knockout mice. Metabolism 2012, 61, 1152–1161. [Google Scholar]
  92. Sultan, A.; Strodthoff, D.; Robertson, A.K.; Paulsson-Berne, G.; Fauconnier, J.; Parini, P.; Rydén, M.; Thierry-Mieg, N.; Johansson, M.E.; Chibalin, A.V.; et al. T cell-mediated inflammation in adipose tissue does not cause insulin resistance in hyperlipidemic mice. Circ. Res 2009, 104, 961–968. [Google Scholar]
  93. Fernandez-Real, J.M.; Ricart, W. Insulin resistance and chronic cardiovascular inflammatory syndrome. Endocr. Rev 2003, 24, 278–301. [Google Scholar]
  94. Mohamed-Ali, V.; Goodrick, S.; Rawesh, A.; Katz, D.R.; Miles, J.M.; Yudkin, J.S.; Klein, S.; Coppack, S.W. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha in vivo. J. Clin. Endocrinol. Metab 1997, 82, 4196–4200. [Google Scholar]
  95. Sopasakis, V.R.; Sandqvist, M.; Gustafson, B.; Hammarstedt, A.; Schmelz, M.; Yang, X.; Jansson, P.A.; Smith, U. High local concentrations and effects on differentiation implicate interleukin-6 as a paracrine regulator. Obes. Res 2004, 12, 454–460. [Google Scholar]
  96. Bastard, J.P.; Maachi, M.; van Nhieu, J.T.; Jardel, C.; Bruckert, E.; Grimaldi, A.; Robert, J.J.; Capeau, J.; 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. 2002, 87, 2084–2089. [Google Scholar]
  97. Pradhan, A.D.; Manson, J.E.; Rifai, N.; Buring, J.E.; Ridker, P.M. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 2001, 286, 327–334. [Google Scholar]
  98. Tsigos, C.; Papanicolaou, D.A.; Kyrou, I.; Defensor, R.; Mitsiadis, C.S.; Chrousos, G.P. Dose-dependent effects of recombinant human interleukin-6 on glucose regulation. J. Clin. Endocrinol. Metab 1997, 82, 4167–4170. [Google Scholar]
  99. Wallenius, V.; Wallenius, K.; Ahren, B.; Rudling, M.; Carlsten, H.; Dickson, S.L.; Ohlsson, C.; Jansson, J.O. Interleukin-6-deficient mice develop mature-onset obesity. Nat. Med 2002, 8, 75–79. [Google Scholar]
  100. Di Gregorio, G.B.; Hensley, L.; Lu, T.; Ranganathan, G.; Kern, P.A. Lipid and carbohydrate metabolism in mice with a targeted mutation in the IL-6 gene: Absence of development of age-related obesity. Am. J. Physiol. Endocrinol. Metab 2004, 287, E182–E187. [Google Scholar]
  101. Starkie, R.; Ostrowski, S.R.; Jauffred, S.; Febbraio, M.; Pedersen, B.K. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-α production in humans. FASEB J 2003, 17, 884–886. [Google Scholar]
  102. Carey, A.L.; Steinberg, G.R.; Macaulay, S.L.; Thomas, W.G.; Holmes, A.G.; Ramm, G.; Prelovsek, O.; Hohnen-Behrens, C.; Watt, M.J.; James, D.E.; et al. Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes 2006, 55, 2688–2697. [Google Scholar]
  103. Wood, I.S.; Wang, B.; Jenkins, J.R.; Trayhurn, P. The pro-inflammatory cytokine IL-18 is expressed in human adipose tissue and strongly upregulated by TNFalpha in human adipocytes. Biochem. Biophys. Res. Commun 2005, 337, 422–429. [Google Scholar]
  104. Esposito, K.; Pontillo, A.; Ciotola, M.; di Palo, C.; Grella, E.; Nicoletti, G.; Giugliano, D. Weight loss reduces interleukin-18 levels in obese women. J. Clin. Endocrinol. Metab 2002, 87, 3864–3866. [Google Scholar]
  105. Tan, H.W.; Liu, X.; Bi, X.P.; Xing, S.S.; Li, L.; Gong, H.P.; Zhong, M.; Wang, Z.H.; Zhang, Y.; Zhang, W. IL-18 overexpression promotes vascular inflammation and remodeling in a rat model of metabolic syndrome. Atherosclerosis 2010, 208, 350–357. [Google Scholar]
  106. Netea, M.G.; Joosten, L.A.; Lewis, E.; Jensen, D.R.; Voshol, P.J.; Kullberg, B.J.; Tack, C.J.; van Krieken, H.; Kim, S.H.; Stalenhoef, A.F.; et al. Deficiency of interleukin-18 in mice leads to hyperphagia, obesity and insulin resistance. Nat. Med 2006, 12, 650–656. [Google Scholar]
  107. Friedman, J.M.; Halaas, J.L. Leptin and the regulation of body weight in mammals. Nature 1998, 395, 763–770. [Google Scholar]
  108. Considine, R.V.; Sinha, M.K.; Heiman, M.L.; Kriauciunas, A.; Stephens, T.W.; Nyce, M.R.; Ohannesian, J.P.; Marco, C.C.; McKee, L.J.; Bauer, T.L.; et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med 1996, 334, 292. [Google Scholar]
  109. Kouidhi, S.; Jarboui, S.; Clerget Froidevaux, M.S.; Abid, H.; Demeneix, B.; Zaouche, A.; Benammar Elgaaied, A.; Guissouma, H. Relationship between subcutaneous adipose tissue expression of leptin and obesity in Tunisian patients. Tunis. Med 2010, 88, 569–572. [Google Scholar]
  110. Marroquí, L.; Gonzalez, A.; Ñeco, P.; Caballero-Garrido, E.; Vieira, E.; Ripoll, C.; Nadal, A.; Quesada, I. Role of leptin in the pancreatic β-cell: Effects and signaling pathways. J. Mol. Endocrinol 2012, 49, R9–R17. [Google Scholar]
  111. Müller, G.; Ertl, J.; Gerl, M.; Preibisch, G. Leptin impairs metabolic actions of insulin in isolated rat adipocytes. J. Biol. Chem 1997, 272, 10585–10593. [Google Scholar]
  112. Pérez, C.; Fernández-Galaz, C.; Fernández-Agulló, T.; Arribas, C.; Andrés, A.; Ros, M.; Carrascosa, J.M. Leptin impairs insulin signaling in rat adipocytes. Diabetes 2004, 53, 347–353. [Google Scholar]
  113. Paz-Filho, G.; Mastronardi, C.; Franco, C.B.; Wang, K.B.; Wong, M.L.; Licinio, J. Leptin: Molecular mechanisms, systemic pro-inflammatory effects, and clinical implications. Arq. Bras. Endocrinol. Metabol 2012, 56, 597–607. [Google Scholar]
  114. Lord, G.M.; Matarese, G.; Howard, J.K.; Baker, R.J.; Bloom, S.R.; Lechler, R.I. Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature 1998, 394, 897–901. [Google Scholar]
  115. Grunfeld, C.; Zhao, C.; Fuller, J.; Pollack, A.; Moser, A.; Friedman, J.; Feingold, K.R. Endotoxin and cytokines induce expression of leptin, the ob gene product, in hamsters. J. Clin. Investig 1996, 97, 2152–2157. [Google Scholar]
  116. Satoh, H.; Nguyen, M.T.; Miles, P.D.; Imamura, T.; Usui, I.; Olefsky, J.M. Adenovirus-mediated chronic “hyper-resistinemia” leads to in vivo insulin resistance in normal rats. J. Clin. Investig 2004, 114, 224–231. [Google Scholar]
  117. Qi, Y.; Nie, Z.; Lee, Y.S.; Singhal, N.S.; Scherer, P.E.; Lazar, M.A.; Ahima, R.S. Loss of resistin improves glucose homeostasis in leptin deficiency. Diabetes 2006, 55, 3083–3090. [Google Scholar]
  118. Banerjee, R.R.; Rangwala, S.M.; Shapiro, J.S.; Rich, A.S.; Rhoades, B.; Qi, Y.; Wang, J.; Rajala, M.W.; Pocai, A.; Scherer, P.E.; et al. Regulation of fasted blood glucose by resistin. Science 2004, 303, 1195–1198. [Google Scholar]
  119. Steppan, C.M.; Wang, J.; Whiteman, E.L.; Birnbaum, M.J.; Lazar, M.A. Activation of SOCS-3 by resistin. Mol. Cell. Biol 2005, 25, 1569–1575. [Google Scholar]
  120. Vidal-Puig, A.; O’Rahilly, S. Resistin: A new link between obesity and insulin resistance? Clin. Endocrinol 2001, 55, 437–438. [Google Scholar]
  121. McTernan, C.L.; McTernan, P.G.; Harte, A.L.; Levick, P.L.; Barnett, A.H.; Kumar, S. Resistin, central obesity, and type 2 diabetes. Lancet 2002, 359, 46–47. [Google Scholar]
  122. McTernan, P.G.; McTernan, C.L.; Chetty, R.; Jenner, K.; Fisher, F.M.; Lauer, M.N.; Crocker, J.; Barnett, A.H.; Kumar, S. Increased resistin gene and protein expression in human abdominal adipose tissue. J. Clin. Endocrinol. Metab 2002, 87, 2407–2410. [Google Scholar]
  123. Wang, H.; Chu, W.S.; Hemphill, C.; Elbein, S.C. Human resistin gene: Molecular scanning and evaluation of association with insulin sensitivity and type 2 diabetes in Caucasians. J. Clin. Endocrinol. Metab 2002, 87, 2520–2524. [Google Scholar]
  124. 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 a 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 2004, 75, 678–686. [Google Scholar]
  125. Kielstein, J.T.; Becker, B.; Graf, S.; Brabant, G.; Haller, H.; Fliser, D. Increased resistin blood levels are not associated with insulin resistance in patients with renal disease. Am. J. Kidney Dis 2003, 42, 62–66. [Google Scholar]
  126. Patel, L.; Buckels, A.C.; Kinghorn, I.J.; Murdock, P.R.; Holbrook, J.D.; Plumpton, C.; Macphee, C.H.; Smith, S.A. Resistin is expressed in human macrophages and directly regulated by PPARγ activators. Biochem. Biophys. Res. Commun 2003, 300, 472–476. [Google Scholar]
  127. Qatanani, M.; Szwergold, N.R.; Greaves, D.R.; Ahima, R.S.; Lazar, M.A. Macrophage-derived human resistin exacerbates adipose tissue inflammation and insulin resistance in mice. J. Clin. Investig 2009, 119, 531–539. [Google Scholar]
  128. Mertens, I.; van Gaal, L.F. Obesity, haemostasis and the fibrinolytic system. Obes. Rev 2002, 3, 85–101. [Google Scholar]
  129. Juhan-Vague, I.; Alessi, M.C.; Mavri, A.; Morange, P.E. Plasminogen activator inhibitor-1, inflammation, obesity, insulin resistance and vascular risk. J. Thromb. Haemost 2003, 1, 1575–1579. [Google Scholar]
  130. Ma, L.J.; Mao, S.L.; Taylor, K.L.; Kanjanabuch, T.; Guan, Y.; Zhang, Y.; Brown, N.J.; Swift, L.L.; McGuinness, O.P.; Wasserman, D.H.; et al. Prevention of obesity and insulin resistance in mice lacking plasminogen activator inhibitor 1. Diabetes 2004, 53, 336–346. [Google Scholar]
  131. Liang, X.; Kanjanabuch, T.; Mao, S.L.; Hao, C.M.; Tang, Y.W.; Declerck, P.J.; Hasty, A.H.; Wasserman, D.H.; Fogo, A.B.; Ma, L.J. Plasminogen activator inhibitor-1 modulates adipocyte differentiation. Am. J. Physiol. Endocrinol. Metab 2006, 290, E103–E113. [Google Scholar]
  132. Xu, X.; Wang, H.; Wang, Z.; Xiao, W. Plasminogen activator inhibitor-1 promotes inflammatory process induced by cigarette smoke extraction or lipopolysaccharides in alveolar epithelial cells. Exp. Lung Res 2009, 35, 795–805. [Google Scholar]
  133. Samal, B.; Sun, Y.; Stearns, G.; Xie, C.; Suggs, S.; McNiece, I. Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol. Cell. Biol 1994, 14, 1431–1437. [Google Scholar]
  134. Fukuhara, A.; Matsuda, M.; Nishizawa, M.; Segawa, K.; Tanaka, M.; Kishimoto, K.; Matsuki, Y.; Murakami, M.; Ichisaka, T.; Murakami, H.; et al. Visfatin: A protein secreted by visceral fat that mimics the effects of insulin. Science 2005, 307, 426–430. [Google Scholar]
  135. Revollo, J.R.; Körner, A.; Mills, K.F.; Satoh, A.; Wang, T.; Garten, A.; Dasgupta, B.; Sasaki, Y.; Wolberger, C.; Townsend, R.R.; et al. Nampt/PBEF/Visfatin regulates insulin secretion in β cells as a systemic NAD biosynthetic enzyme. Cell Metab 2007, 6, 363–375. [Google Scholar]
  136. Pagano, C.; Pilon, C.; Olivieri, M.; Mason, P.; Fabris, R.; Serra, R.; Milan, G.; Rossato, M.; Federspil, G.; Vettor, R. Reduced plasma visfatin/pre B-cell colony-enhancing factor in obesity is not related to insulin resistance in humans. J. Clin. Endocrinol. Metab 2006, 91, 3165–3170. [Google Scholar]
  137. Berndt, J.; Klöting, N.; Kralisch, S.; Kovacs, P.; Fasshauer, M.; Schön, M.R.; Stumvollx, M.; Blüher, M. Plasma visfatin concentrations and fat-depot specific mRNA expression in humans. Diabetes 2005, 54, 2911–2916. [Google Scholar]
  138. Fain, J.N.; Sacks, H.S.; Buehrer, B.; Bahouth, S.W.; Garrett, E.; Wolf, R.Y.; Carter, R.A.; Tichansky, D.S.; Madan, A.K. Identification of omentin mRNA in human epicardial adipose tissue: Comparison to omentin in subcutaneous, internal mammary artery periadventitial and visceral abdominal depots. Int. J. Obes 2008, 32, 810–815. [Google Scholar]
  139. Chang, Y.C.; Chang, T.J.; Lee, W.J.; Chuang, L.M. The relationship of visfatin/pre-B-cell colony-enhancing factor/nicotinamide phosphoribosyltransferase in adipose tissue with inflammation, insulin resistance, and plasma lipids. Metabolism 2010, 59, 93–99. [Google Scholar]
  140. Haider, D.G.; Schindler, K.; Schaller, G.; Prager, G.; Wolzt, M.; Ludvik, B. Increased plasma visfatin concentrations in morbidly obese subjects are reduced after gastric banding. J. Clin. Endocrinol. Metab 2006, 91, 1578–1581. [Google Scholar]
  141. El-Mesallamy, H.O.; Kassem, D.H.; El-Demerdash, E.; Amin, A.I. Vaspin and visfatin/Nampt are interesting interrelated adipokines playing a role in the pathogenesis of type 2 diabetes mellitus. Metabolism 2011, 60, 63–70. [Google Scholar]
  142. Varma, V.; Yao-Borengasser, A.; Rasouli, N.; Bodles, A.M.; Phanavanh, B.; Lee, M.J.; Starks, T.; Kern, L.M.; Spencer, H.J., 3rd; McGehee, R.E., Jr.; et al. Human visfatin expression: Relationship to insulin sensitivity, intramyocellular lipids, and inflammation. J. Clin. Endocrinol. Metab 2007, 92, 666–672. [Google Scholar]
  143. Kim da, S.; Kang, S.; Moon, N.R.; Park, S. Central visfatin potentiates glucose-stimulated insulin secretion and β-cell mass without increasing serum visfatin levels in diabetic rats. Cytokine 2014, 65, 159–166. [Google Scholar]
  144. Oki, K.; Yamane, K.; Kamei, N.; Nojima, H.; Kohno, N. Circulating visfatin level is correlated with inflammation, but not with insulin resistance. Clin. Endocrinol 2007, 67, 796–800. [Google Scholar]
  145. Quadro, L.; Blaner, W.S.; Salchow, D.J.; Vogel, S.; Piantedosi, R.; Gouras, P.; Freeman, S.; Cosma, M.P.; Colantuoni, V.; Gottesman, M.E. Impaired retinal function and vitamin A availability in mice lacking retinol-binding protein. EMBO J 1999, 18, 4633–4644. [Google Scholar]
  146. Yang, Q.; Graham, T.E.; Mody, N.; Preitner, F.; Peroni, O.D.; Zabolotny, J.M.; Kotani, K.; Quadro, L.; Kahn, B.B. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 2005, 436, 356–362. [Google Scholar]
  147. Klöting, N.; Graham, T.E.; Berndt, J.; Kralisch, S.; Kovacs, P.; Wason, C.J.; Fasshauer, M.; Schön, M.R.; Stumvoll, M.; Blüher, M.; et al. Serum retinol-binding protein is more highly expressed in visceral than in subcutaneous adipose tissue and is a marker of intra-abdominal fat mass. Cell Metab 2007, 6, 79–87. [Google Scholar]
  148. Ost, A.; Danielsson, A.; Liden, M.; Eriksson, U.; Nystrom, F.H.; Stralfors, P. Retinol-binding protein-4 attenuates insulin-induced phosphorylation of IRS1 and ERK1/2 in primary human adipocytes. FASEB J 2007, 21, 3696–3704. [Google Scholar]
  149. Graham, T.E.; Yang, Q.; Bluher, M.; Hammarstedt, A.; Ciaraldi, T.P.; Henry, R.R.; Wason, C.J.; Oberbach, A.; Jansson, P.A.; Smith, U.; et al. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N. Engl. J. Med 2006, 354, 2552–2563. [Google Scholar]
  150. Gavi, S.; Stuart, L.M.; Kelly, P.; Melendez, M.M.; Mynarcik, D.C.; Gelato, M.C.; McNurlan, M.A. Retinol-binding protein 4 is associated with insulin resistance and body fat distribution in nonobese subjects without type 2 diabetes. J. Clin. Endocrinol. Metab 2007, 92, 1886–1890. [Google Scholar]
  151. Balagopal, P.; Graham, T.E.; Kahn, B.B.; Altomare, A.; Funanage, V.; George, D. Reduction of elevated serum retinol binding protein in obese children by lifestyle intervention: Association with subclinical inflammation. J. Clin. Endocrinol. Metab 2007, 92, 1971–1974. [Google Scholar]
  152. Nair, A.K.; Sugunan, D.; Kumar, H.; Anilkumar, G. Case-control analysis of SNPs in GLUT4, RBP4 and STRA6: Association of SNPs in STRA6 with type 2 diabetes in a South Indian population. PLoS One 2010, 5, e11444. [Google Scholar]
  153. Munkhtulga, L.; Nakayama, K.; Utsumi, N.; Yanagisawa, Y.; Gotoh, T.; Omi, T.; Kumada, M.; Erdenebulgan, B.; Zolzaya, K.; Lkhagvasuren, T.; et al. Identification of a regulatory SNP in the retinol binding protein 4 gene associated with type 2 diabetes in Mongolia. Hum. Genet 2007, 120, 879–888. [Google Scholar]
  154. Munkhtulga, L.; Nagashima, S.; Nakayama, K.; Utsumi, N.; Yanagisawa, Y.; Gotoh, T.; Omi, T.; Kumada, M.; Zolzaya, K.; Lkhagvasuren, T.; et al. Regulatory SNP in the RBP4 gene modified the expression in adipocytes and associated with BMI. Obesity 2010, 18, 1006–1014. [Google Scholar]
  155. Yao-Borengasser, A.; Varma, V.; Bodles, A.M.; Rasouli, N.; Phanavanh, B.; Lee, M.J.; Starks, T.; Kern, L.M.; Spencer, H.J., III; Rashidi, A.A.; et al. Retinol binding protein 4 expression in humans: Relationship to insulin resistance, inflammation, and response to pioglitazone. J. Clin. Endocrinol. Metab 2007, 92, 2590–2597. [Google Scholar]
  156. Ulgen, F.; Herder, C.; Kuhn, M.C.; Willenberg, H.S.; Schott, M.; Scherbaum, W.A.; Schinner, S. Association of serum levels of retinol-binding protein 4 with male sex but not with insulin resistance in obese patients. Arch. Physiol. Biochem 2010, 116, 57–62. [Google Scholar]
  157. Kos, K.; Wong, S.; Tan, B.; Kerrigan, D.; Randeva, H.; Pinkney, J.; Wilding, J. Human RBP4 adipose tissue expression is gender specific and influenced by leptin. Clin. Endocrinol. (Oxf.) 2011, 74, 197–205. [Google Scholar]
  158. Tabata, M.; Kadomatsu, T.; Fukuhara, S.; Miyata, K.; Ito, Y.; Endo, M.; Urano, T.; Zhu, H.J.; Tsukano, H.; Tazume, H.; et al. Angiopoietin-like protein 2 promotes chronic adipose tissue inflammation and obesity-related systemic insulin resistance. Cell Metab 2009, 10, 178–188. [Google Scholar]
  159. Doi, Y.; Ninomiya, T.; Hirakawa, Y.; Takahashi, O.; Mukai, N.; Hata, J.; Iwase, M.; Kitazono, T.; Oike, Y.; Kiyohara, Y. Angiopoietin-like protein 2 and risk of type 2 diabetes in a general Japanese population: The Hisayama study. Diabetes Care 2013, 36, 98–100. [Google Scholar]
  160. Berg, A.H.; Combs, T.P.; Du, X.; Brownlee, M.; Scherer, P.E. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat. Med 2001, 7, 947–953. [Google Scholar]
  161. Diez, J.J.; Iglesias, P. The role of the novel adipocyte-derived hormone adiponectin in human disease. Eur. J. Endocrinol 2003, 148, 293–300. [Google Scholar]
  162. Kern, P.A.; di Gregorio, G.B.; Lu, T.; Rassouli, N.; Ranganathan, G. Adiponectin expression from human adipose tissue: Relation to obesity, insulin resistance, and tumor necrosis factor-alpha expression. Diabetes 2003, 52, 1779–1785. [Google Scholar]
  163. Maeda, N.; Shimomura, I.; Kishida, K.; Nishizawa, H.; Matsuda, M.; Nagaretani, H.; Furuyama, N.; Kondo, H.; Takahashi, M.; Arita, Y.; et al. Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat. Med 2002, 8, 731–737. [Google Scholar]
  164. Kadowaki, T.; Yamauchi, T.; Kubota, N.; Hara, K.; Ueki, K.; Tobe, K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J. Clin. Investig 2006, 116, 1784–1792. [Google Scholar]
  165. Ouchi, N.; Higuchi, A.; Ohashi, K.; Oshima, Y.; Gokce, N.; Shibata, R.; Akasaki, Y.; Shimono, A.; Walsh, K. Sfrp5 is an anti-inflammatory adipokine that modulates metabolic dysfunction in obesity. Science 2010, 329, 454–457. [Google Scholar]
  166. Hu, Z.; Deng, H.; Qu, H. Plasma SFRP5 levels are decreased in Chinese subjects with obesity and type 2 diabetes and negatively correlated with parameters of insulin resistance. Diabetes Res. Clin. Pract 2013, 99, 391–395. [Google Scholar]
  167. Hu, W.; Li, L.; Yang, M.; Luo, X.; Ran, W.; Liu, D.; Xiong, Z.; Liu, H.; Yang, G. Circulating Sfrp5 is a signature of obesity-related metabolic disorders and is regulated by glucose and liraglutide in humans. J. Clin. Endocrinol. Metab 2013, 98, 290–298. [Google Scholar]
  168. Tan, X.; Wang, X.; Chu, H.; Liu, H.; Yi, X.; Xiao, Y. SFRP5 correlates with obesity and metabolic syndrome and increases after weight loss in children. Clin. Endocrinol. (Oxf.) 2013. [Google Scholar] [CrossRef]
  169. Carstensen, M.; Herder, C.; Kempf, K.; Erlund, I.; Martin, S.; Koenig, W.; Sundvall, J.; Bidel, S.; Kuha, S.; Roden, M.; et al. Sfrp5 correlates with insulin resistance and oxidative stress. Eur. J. Clin. Investig 2013, 43, 350–357. [Google Scholar]
  170. Saleh, J.; Sniderman, A.D.; Cianflone, K. Regulation of Plasma fatty acid metabolism. Clin. Chim. Acta 1999, 286, 163–180. [Google Scholar]
  171. Clemente-Postigo, M.; Queipo-Ortuno, M.I.; Fernandez-Garcia, D.; Gomez-Huelgas, R.; Tinahones, F.J.; Cardona, F. Adipose tissue gene expression of factors related to lipid processing in obesity. PLoS One 2011, 6, e24783. [Google Scholar]
  172. Klop, B.; Jukema, J.W.; Rabelink, T.J.; Castro Cabezas, M. A physician’s guide for the management of hypertriglyceridemia: The etiology of hypertriglyceridemia determines treatment strategy. Panminerva Med 2012, 54, 91–103. [Google Scholar]
  173. Klop, B.; Elte, J.W.; Cabezas, M.C. Dyslipidemia in obesity: Mechanisms and potential targets. Nutrients 2013, 5, 1218–1240. [Google Scholar]
  174. Guendouzi, K.; Jaspard, B.; Barbaras, R.; Motta, C.; Vieu, C.; Marcel, Y.; Chap, H.; Perret, B.; Collet, X. Biochemical and physical properties of remnant-HDL2 and of pre beta 1-HDL produced by hepatic lipase. Biochemistry 1999, 38, 2762–2768. [Google Scholar]
  175. Friedewald, W.T.; Levy, R.I.; Fredrickson, D.S. Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge. Clin. Chem 1972, 18, 499–502. [Google Scholar]
  176. Contois, J.H.; Warnick, G.R.; Sniderman, A.D. Reliability of low-density lipoprotein cholesterol, non-high-density lipoprotein cholesterol, and apolipoprotein B measurement. J. Clin. Lipidol 2011, 5, 264–272. [Google Scholar]
  177. Ito, Y.; Fujimura, M.; Ohta, M.; Hirano, T. Development of a homogeneous assay for measurement of small dense LDL cholesterol. Clin. Chem 2011, 57, 57–65. [Google Scholar]
  178. St-Pierre, A.C.; Cantin, B.; Dagenais, G.R.; Mauriege, P.; Bernard, P.-M.; Despres, J.P.; Lamarche, B. Low-density lipoprotein subfractions and the long-term risk of ischemic heart disease in men. Arterioscler. Thromb. Vasc. Biol 2005, 25, 553–559. [Google Scholar]
  179. Rizzo, M.; Pernice, V.; Frasheri, A.; di Lorenzo, G.; Rini, G.B.; Spinas, G.A.; Berneis, K. Small, dense low-density lipoproteins (LDL) are predictors of cardio- and cerebro-vascular events in subjects with the metabolic syndrome. Clin. Endocrinol. (Oxf.) 2009, 70, 870–875. [Google Scholar] [Green Version]
  180. Engfeldt, P.; Arner, P. Lipolysis in human adipocytes, effects of cell size, age and of regional differences. Horm. Metab. Res 1988, 19, 26–29. [Google Scholar]
  181. Veilleux, A.; Caron-Jobin, M.; Noël, S.; Laberge, P.Y.; Tchernof, A. Visceral adipocyte hypertrophy is associated with dyslipidemia independent of body composition and fat distribution in women. Diabetes 2011, 60, 1504–1511. [Google Scholar]
  182. Sam, S.; Haffner, S.; Davidson, M.H.; D’Agostino, R.B., Sr.; Feinstein, S.; Kondos, G.; Perez, A.; Mazzone, T. Relationship of abdominal visceral and subcutaneous adipose tissue with lipoprotein particle number and size in type 2 diabetes. Diabetes 2008, 57, 2022–2027. [Google Scholar]
  183. Johnson, J.A.; Fried, S.K.; Pi-Sunyer, F.X.; Albu, J.B. Impaired insulin action in subcutaneous adipocytes from women with visceral obesity. Am. J. Physiol. Endocrinol. Metab 2001, 280, E40–E49. [Google Scholar]
  184. Despres, J.P.; Ferland, M.; Moorjani, S.; Nadeau, A.; Tremblay, A.; Lupien, P.J.; Theriault, G.; Bouchard, C. Role of hepatic-triglyceride lipase activity in the association between intra-abdominal fat and plasma HDL cholesterol in obese women. Arteriosclerosis 1989, 9, 485–492. [Google Scholar]
  185. Carr, M.C.; Hokanson, J.E.; Zambon, A.; Deeb, S.S.; Barrett, P.H.; Purnell, J.Q.; Brunzell, J.D. The contribution of intraabdominal fat to gender differences in hepatic lipase activity and low/high density lipoprotein heterogeneity. J. Clin. Endocrinol. Metab 2001, 86, 2831–2837. [Google Scholar]
  186. Cancello, R.; Tordjman, J.; Poitou, C.; Guilhem, G.; Bouillot, J.L.; Hugol, D.; Coussieu, C.; Basdevant, A.; Bar Hen, A.; Bedossa, P.; et al. Increased infiltration of macrophages in omental adipose tissue is associated with marked hepatic lesions in morbid human obesity. Diabetes 2006, 55, 1554–1561. [Google Scholar]
  187. Esteve, E.; Ricart, W.; Fernández-Real, J.M. Dyslipidemia and inflammation: An evolutionary conserved mechanism. Clin. Nutr 2005, 24, 16–31. [Google Scholar]
  188. Yang, Y.; Ju, D.; Zhang, M.; Yang, G. Interleukin-6 stimulates lipolysis in porcine adipocytes. Endocrine 2008, 33, 261–269. [Google Scholar]
  189. Hardardóttir, I.; Doerrler, W.; Feingold, K.R.; Grünfeld, C. Cytokines stimulate lipolysis and decrease lipoprotein lipase activity in cultured fat cells by a prostaglandin independent mechanism. Biochem. Biophys. Res. Commun 1992, 186, 237–243. [Google Scholar]
  190. Hardardottir, I.; Moser, A.H.; Memon, R.; Grunfeld, C.; Feingold, K.R. Effects of TNF, IL-1, and the combination of both cytokines on cholesterol metabolism in Syrian hamsters. Lymphokine Cytokine Res 1994, 13, 161–166. [Google Scholar]
  191. Kawakami, M.; Murase, T.; Ogawa, H.; Ishibashi, S.; Mori, N.; Takaku, F.; Shibata, S. Human recombinant TNF suppresses lipoprotein lipase activity and stimulates lipolysis in 3T3-L1 cells. J. Biochem 1987, 10, 331–338. [Google Scholar]
  192. Greenberg, A.S.; Nordan, R.P.; McIntosh, J.; Calvo, J.C.; Scow, R.O.; Jablons, D. Interleukin 6 reduces lipoprotein lipase activity in adipose tissue of mice in vivo and in 3T3-L1 adipocytes (a possible role for interleukin 6 in cancer cachexia). Cancer Res 1992, 52, 4113–4116. [Google Scholar]
  193. Rouzer, C.A.; Cerami, A. Hypertriglyceridemia associated with Trypanosoma brucei brucei infection in rabbits: Role of defective triglyceride removal. Mol. Biochem. Parasitol 1980, 2, 31–38. [Google Scholar]
  194. Jovinge, S.; Hamsten, A.; Tornvall, P.; Proudler, A.; Båvenholm, P.; Ericsson, C.G.; Godsland, I.; de Faire, U.; Nilsson, J. Evidence for a role of tumor necrosis factor alpha in disturbances of triglyceride and glucose metabolism predisposing to coronary heart disease. Metabolism 1998, 47, 113–118. [Google Scholar]
  195. Feingold, K.R.; Serio, M.K.; Adi, S.; Moser, A.H.; Grunfeld, C. Tumor necrosis factor stimulates hepatic lipid synthesis and secretion. Endocrinology 1989, 124, 2336–2342. [Google Scholar]
  196. Kawakami, M.; Cerami, A. Studies of endotoxin-induced decrease in lipoprotein lipase activity. J. Exp. Med 1981, 154, 631–639. [Google Scholar]
  197. Qin, B.; Anderson, R.A.; Adeli, K. Tumor necrosis factor-alpha directly stimulates the overproduction of hepatic apolipoprotein B100-containing VLDL via impairment of hepatic insulin signaling. Am. J. Physiol. Gastrointest. Liver Physiol 2008, 294, G1120–G1129. [Google Scholar]
  198. Mohrschladt, M.F.; Weverling-Rijnsburger, A.W.; de Man, F.H.; Stoeken, D.J.; Sturk, A.; Smelt, A.H.; Westendorp, R.G. Hyperlipoproteinemia affects cytokine production in whole blood samples ex vivo. The influence of lipid-lowering therapy. Atherosclerosis 2000, 148, 413–419. [Google Scholar]
  199. Jonkers, I.J.; Mohrschladt, M.F.; Westendorp, R.G.; van der Laarse, A.; Smelt, A.H. Severe hypertriglyceridemia with insulin resistance is associated with systemic inflammation (reversal with bezafibrate therapy in a randomized controlled trial). Am. J. Med 2002, 112, 275–280. [Google Scholar]
  200. Nappo, F.; Esposito, K.; Cioffi, M.; Giugliano, G.; Molinari, A.M.; Paolisso, G.; Marfella, R.; Giugliano, D. Postprandial endothelial activation in healthy subjects and in type 2 diabetic patients (role of fat and carbohydrate meals). J. Am. Coll. Cardiol 2002, 39, 1145–1150. [Google Scholar]
  201. Van Exel, E.; Gussekloo, J.; de Craen, A.J.; Frolich, M.; Bootsma-van der Wiel, A.; Westendorp, R.G. Leiden 85 Plus Study. Low production capacity of interleukin-10 associates with the metabolic syndrome and type 2 diabetes: The Leiden 85-Plus Study. Diabetes 2002, 51, 1088–1092. [Google Scholar]
  202. Grunfeld, C.; Dinarello, C.A.; Feingold, K.R. Tumor necrosis factor-alpha, interleukin-1, and interferon alpha stimulate triglyceride synthesis in HepG2 cells. Metabolism 1991, 40, 894–898. [Google Scholar]
  203. Fernandez-Real, J.M.; Gutierrez, C.; Ricart, W.; Castineira, M.J.; Vendrell, J.; Richart, C. Plasma levels of the soluble fraction of tumor necrosis factor receptors 1 and 2 are independent determinants of plasma cholesterol and LDL-cholesterol concentrations in healthy subjects. Atherosclerosis 1999, 146, 321–327. [Google Scholar]
  204. Skoog, T.; Dichtl, W.; Boquist, S.; Skoglund-Andersson, C.; Karpe, F.; Tang, R.; Bond, M.G.; de Faire, U.; Nilsson, J.; Eriksson, P.; et al. Plasma tumour necrosis factor-alpha and early carotid atherosclerosis in healthy middle-aged men. Eur. Heart J 2002, 23, 376–383. [Google Scholar]
  205. Mizia-Stec, K.; Zahorska-Markiewicz, B.; Mandecki, T.; Janowska, J.; Szulc, A.; Jastrzekbska-Maj, E.; Gasior, Z. Hyperlipidaemias and serum cytokines in patients with coronary artery disease. Acta Cardiol 2003, 58, 9–15. [Google Scholar]
  206. Saez, Y.; Vacas, M.; Santos, M.; Saez de Lafuente, J.P.; Sagastagoitia, J.D.; Molinero, E.; Iriarte, J.A. Relation of high-density lipoprotein cholesterol and apoprotein A1 levels with presence and severity of coronary obstruction. ISRN Vasc. Med 2012, 2012, 1–5. [Google Scholar]
  207. Ettinger, W.H.; Miller, L.A.; Smith, T.K.; Parks, J.S. Effect of interleukin-1 alpha on lipoprotein lipids in cynomolgus monkeys (comparison to tumor necrosis factor). Biochim. Biophys. Acta 1992, 1128, 186–192. [Google Scholar]
  208. Ettinger, W.H.; Miller, L.D.; Albers, J.J.; Smith, T.K.; Parks, J.S. Lipopolysaccharide and tumor necrosis factor cause a fall in plasma concentration of lecithin (cholesterol acyltransferase in cynomolgus monkeys). J. Lipid Res 1990, 31, 1099–1107. [Google Scholar]
  209. Memon, R.A.; Grunfeld, C.; Moser, A.H.; Feingold, K.R. Tumor necrosis factor mediates the effects of endotoxin on cholesterol and triglyceride metabolism in mice. Endocrinology 1993, 132, 2246–2253. [Google Scholar]
  210. Feingold, K.R.; Pollock, A.S.; Moser, A.H.; Shigenaga, J.K.; Grunfeld, C. Discordant regulation of proteins of cholesterol metabolism during the acute phase response. J. Lipid Res 1995, 36, 1474–1482. [Google Scholar]
  211. Ruan, X.Z.; Varghese, Z.; Fernando, R.; Moorhead, J.F. Cytokine regulation of low-density lipoprotein receptor gene transcription in human mesangial cells. Nephrol. Dial. Transplant 1998, 13, 1391–1397. [Google Scholar]
  212. Ruan, X.Z.; Varghese, Z.; Powis, S.H.; Moorhead, J.F. Dysregulation of LDL receptor under the influence of inflammatory cytokines (a new pathway for foam cell formation). Kidney Int 2001, 60, 1716–1725. [Google Scholar]
  213. Ruan, X.Z.; Moorhead, J.F.; Fernando, R.; Wheeler, D.C.; Powis, S.H.; Varghese, Z. Regulation of lipoprotein trafficking in the kidney (role of inflammatory mediators and transcription factors). Biochem. Soc. Trans 2004, 32, 88–91. [Google Scholar]
  214. Bartolome, N.; Rodriguez, L.; Martinez, M.J.; Ochoa, B.; Chico, Y. Upregulation of apolipoprotein B secretion, but not lipid, by tumor necrosis factor-alpha in rat hepatocyte cultures in the absence of extracellular fatty acids. Ann. N. Y. Acad. Sci 2007, 1096, 55–69. [Google Scholar]
  215. Homma, Y. Predictors of atherosclerosis. J. Atheroscler. Thromb 2004, 11, 265–270. [Google Scholar]
  216. Crowl, R.M.; Stoller, T.J.; Conroy, R.R.; Stoner, C.R. Induction of phospholipase A2 gene expression in human hepatoma cells by mediators of the acute phase response. J. Biol. Chem 1991, 266, 2647–2651. [Google Scholar]
  217. Kugiyama, K.; Ota, Y.; Takazoe, K.; Moriyama, Y.; Kawano, H.; Miyao, Y.; Sakamoto, T.; Soejima, H.; Ogawa, H.; Doi, H.; et al. Circulating levels of secretory type II phospholipase A(2) predict coronary events in patients with coronary artery disease. Circulation 1999, 100, 1280–1284. [Google Scholar]
  218. O’Brien, K.D.; Chait, A. Serum amyloid A: The “other” inflammatory protein. Curr. Atheroscler. Rep 2006, 8, 62–68. [Google Scholar]
  219. Poitou, C.; Viguerie, N.; Cancello, R.; de Matteis, R.; Cinti, S.; Stich, V.; Coussieu, C.; Gauthier, E.; Courtine, M.; Zucker, J.D.; et al. Serum amyloid A: Production by human white adipocyte and regulation by obesity and nutrition. Diabetologia 2005, 48, 519–528. [Google Scholar]
  220. Fasshauer, M.; Klein, J.; Kralisch, S.; Klier, M.; Lossner, U.; Bluher, M.; Paschke, R. Serum amyloid A3 expression is stimulated by dexamethasone and interleukin-6 in 3T3–L1 adipocytes. J. Endocrinol 2004, 183, 561–567. [Google Scholar]
  221. Yang, R.Z.; Lee, M.J.; Hu, H.; Pollin, T.I.; Ryan, A.S.; Nicklas, B.J.; Snitker, S.; Horenstein, R.B.; Hull, K.; Goldberg, N.H.; et al. Acute-phase serum amyloid A: An inflammatory adipokine and potential link between obesity and its metabolic complications. PLoS Med 2006, 3, e287. [Google Scholar]
  222. Chen, C.H.; Wang, P.H.; Liu, B.H.; Hsu, H.H.; Mersmann, H.J.; Ding, S.T. Serum amyloid A protein regulates the expression of porcine genes related to lipid metabolism. J. Nutr 2008, 138, 674–679. [Google Scholar]
  223. Cai, L.; de Beer, M.C.; de Beer, F.C.; van der Westhuyzen, D.R. Serum amyloid A is a ligand for scavenger receptor class B type I and inhibits high density lipoprotein binding and selective lipid uptake. J. Biol. Chem 2005, 280, 2954–2961. [Google Scholar]
  224. Rigotti, A.; Miettinen, H.E.; Krieger, M. The role of the high-density lipoprotein receptor SR-BI in the lipid metabolism of endocrine and other tissues. Endocr. Rev 2003, 24, 357–387. [Google Scholar]
  225. Lewis, K.E.; Kirk, E.A.; McDonald, T.O.; Wang, S.; Wight, T.N.; O’Brien, K.D.; Chait, A. Increase in serum amyloid a evoked by dietary cholesterol is associated with increased atherosclerosis in mice. Circulation 2004, 110, 540–545. [Google Scholar]
  226. Ouchi, N.; Kihara, S.; Arita, Y.; Maeda, K.; Kuriyama, H.; Okamoto, Y.; Hotta, K.; Nishida, M.; Takahashi, M.; Nakamura, T.; et al. Novel modulator for endothelial adhesion molecules: Adipocyte-derived plasma protein adiponectin. Circulation 1999, 100, 2473–2476. [Google Scholar]
  227. Matsubara, M.; Maruoka, S.; Katayose, S. Decreased plasma adiponectin concentrations in women with dyslipidemia. J. Clin. Endocrinol. Metab 2002, 87, 2764–2769. [Google Scholar]
  228. Okamoto, Y.; Kihara, S.; Funahashi, T.; Matsuzawa, Y.; Libby, P. Adiponectin: A key adipocytokine in metabolic syndrome. Clin. Sci. (Lond.) 2006, 110, 267–278. [Google Scholar]
  229. Yamauchi, T.; Kamon, J.; Minokoshi, Y.; Ito, Y.; Waki, H.; Uchida, S.; Yamashita, S.; Noda, M.; Kita, S.; Ueki, K.; et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med 2002, 8, 1288–1295. [Google Scholar]
  230. Berneis, K.K.; Krauss, R.M. Metabolic origins and clinical significance of LDL heterogeneity. J. Lipid Res 2002, 43, 1363–1379. [Google Scholar]
  231. Schneider, J.G.; von Eynatten, M.; Schiekofer, S.; Nawroth, P.P.; Dugi, K.A. Low plasma adiponectin levels are associated with increased hepatic lipase activityin vivo. Diabetes Care 2005, 28, 2181–2186. [Google Scholar]
  232. Matsuura, F.; Oku, H.; Koseki, M.; Sandoval, J.C.; Yuasa-Kawase, M.; Tsubakio-Yamamoto, K.; Masuda, D.; Maeda, N.; Tsujii, K.; Ishigami, M.; et al. Adiponectin accelerates reverse cholesterol transport by increasing high density lipoprotein assembly in the liver. Biochem. Biophys. Res. Commun 2007, 358, 1091–1095. [Google Scholar]
  233. Oku, H.; Matsuura, F.; Koseki, M.; Sandoval, J.C.; Yuasa-Kawase, M.; Tsubakio-Yamamoto, K.; Masuda, D.; Maeda, N.; Ohama, T.; Ishigami, M.; et al. Adiponectin deficiency suppresses ABCA1 expression and ApoA-I synthesis in the liver. FEBS Lett 2007, 581, 5029–5033. [Google Scholar]
  234. Chang, C.Y.; Chen, M.J.; Yang, W.S.; Yeh, C.Y.; Ho, H.N.; Chen, S.U.; Yang, Y.S. Hypoadiponectinemia: A useful marker of dyslipidemia in women with polycystic ovary syndrome. Taiwan J. Obstet. Gynecol 2012, 51, 583–590. [Google Scholar]
  235. Angulo, P. Nonalcoholic fatty liver disease. N. Engl. J. Med 2002, 346, 1221–1231. [Google Scholar]
  236. Starley, B.Q.; Calcagno, C.J.; Harrison, S.A. Nonalcoholic fatty liver disease and hepatocellular carcinoma: A weighty connection. Hepatology 2010, 51, 1820–1832. [Google Scholar]
  237. Lazo, M.; Clark, J.M. The epidemiology of nonalcoholic fatty liver disease: A global perspective. Semin Liver Dis 2008, 28, 339–350. [Google Scholar]
  238. Tarantino, G.; Savastano, S.; Colao, A. Hepatic steatosis, low-grade chronic inflammation and hormone/growth factor/adipokine imbalance. World J. Gastroenterol 2010, 16, 4773–4783. [Google Scholar]
  239. Day, C.P.; James, O.F. Steatohepatitis: A tale of two “hits”? Gastroenterology 1998, 114, 842–845. [Google Scholar]
  240. Tilg, H.; Moschen, A.R. Evolution of inflammation in nonalcoholic fatty liver disease: The multiple parallel hits hypothesis. Hepatology 2010, 52, 1836–1846. [Google Scholar]
  241. Polyzos, S.A.; Kountouras, J.; Zavos, C.; Deretzi, G. Nonalcoholic fatty liver disease: Multimodal treatment options for a pathogenetically multiple-hit disease. J. Clin. Gastroenterol 2012, 46, 272–284. [Google Scholar]
  242. Bradbury, M.W.; Berk, P.D. Lipid metabolism in hepatic steatosis. Clin. Liver Dis 2004, 8, 639–671. [Google Scholar]
  243. Koteish, A.; Diehl, A.M. Animal models of steatosis. Semin. Liver Dis 2001, 21, 89–104. [Google Scholar]
  244. Lambert, J.E.; Ramos-Roman, M.A.; Browning, J.D.; Parks, E.J. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic Fatty liver disease. Gastroenterology 2014, 146, 726–735. [Google Scholar]
  245. Nielsen, S.; Guo, Z.; Johnson, C.M.; Hensrud, D.D.; Jensen, M.D. Splanchnic lipolysis in human obesity. J. Clin. Investig 2004, 113, 1582–1588. [Google Scholar]
  246. Heilbronn, L.; Smith, S.R.; Ravussin, E. Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and type II diabetes mellitus. Int. J. Obes. Relat. Metab. Disord 2004, 28, S12–S21. [Google Scholar]
  247. Roden, M. Mechanisms of disease: Hepatic steatosis in type 2 diabetes—Pathogenesis and clinical relevance. Nat. Clin. Pract. Endocrinol. Metab 2006, 2, 335–348. [Google Scholar]
  248. Montague, C.T.; O’Rahilly, S. The perils of portliness: Causes and consequences of visceral adiposity. Diabetes 2000, 49, 883–888. [Google Scholar]
  249. Donnelly, K.L.; Smith, C.I.; Schwarzenberg, S.J.; Jessurun, J.; Boldt, M.D.; Parks, E.J. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Investig 2005, 115, 1343–1351. [Google Scholar]
  250. Parks, E.J.; Hellerstein, M.K. Thematic review series: Patient-oriented research. Recent advances in liver triacylglycerol and fatty acid metabolism using stable isotope labeling techniques. J. Lipid Res 2006, 47, 1651–1660. [Google Scholar]
  251. Harrison, S.A.; Day, C.P. Benefits of lifestyle modification in NAFLD. Gut 2007, 56, 1760–1769. [Google Scholar]
  252. Shklyaev, S.; Aslanidi, G.; Tennant, M.; Prima, V.; Kohlbrenner, E.; Kroutov, V.; Campbell-Thompson, M.; Crawford, J.; Shek, E.W.; Scarpace, P.J.; et al. Sustained peripheral expression of transgene adiponectin offsets the development of diet-induced obesity in rats. Proc. Natl. Acad. Sci. USA 2003, 100, 14217–14222. [Google Scholar]
  253. Xu, A.; Wang, Y.; Keshaw, H.; Xu, L.Y.; Lam, K.S.; Cooper, G.J. The fat-derived hormone adiponectin alleviates alcoholic and nonalcoholic fatty liver diseases in mice. J. Clin. Investig 2003, 112, 91–100. [Google Scholar] [Green Version]
  254. Schindhelm, R.K.; Diamant, M.; Dekker, J.M.; Tushuizen, M.E.; Teerlink, T.; Heine, R.J. Alanine aminotransferase as a marker of non-alcoholic fatty liver disease in relation to type 2 diabetes mellitus and cardiovascular disease. Diabetes Metab. Res. Rev 2006, 22, 437–443. [Google Scholar]
  255. Masaki, T.; Chiba, S.; Tatsukawa, H.; Yasuda, T.; Noguchi, H.; Seike, M.; Yoshimatsu, H. Adiponectin protects LPS-induced liver injury through modulation of TNF-alpha in KK-Ay obese mice. Hepatology 2004, 40, 177–184. [Google Scholar]
  256. Kamada, Y.; Tamura, S.; Kiso, S.; Matsumoto, H.; Saji, Y.; Yoshida, Y.; Fukui, K.; Maeda, N.; Nishizawa, H.; Nagaretani, H.; et al. Enhanced carbon tetrachloride-induced liver fibrosis in mice lacking adiponectin. Gastroenterology 2003, 125, 1796–1807. [Google Scholar]
  257. Hui, J.M.; Hodge, A.; Farrell, G.C.; Kench, J.G.; Kriketos, A.; George, J. Beyond insulin resistance in NASH: TNF-alpha or adiponectin? Hepatology 2004, 40, 46–54. [Google Scholar]
  258. López-Bermejo, A.; Botas, P.; Funahashi, T.; Delgado, E.; Kihara, S.; Ricart, W.; Fernández-Real, J.M. Adiponectin, hepatocellular dysfunction and insulin sensitivity. Clin. Endocrinol. (Oxf.) 2004, 60, 256–263. [Google Scholar]
  259. Targher, G.; Bertolini, L.; Scala, L.; Poli, F.; Zenari, L.; Falezza, G. Decreased plasma adiponectin concentrations are closely associated with nonalcoholic hepatic steatosis in obese individuals. Clin. Endocrinol. (Oxf.) 2004, 61, 700–703. [Google Scholar]
  260. Kaser, S.; Moschen, A.; Cayon, A.; Kaser, A.; Crespo, J.; Pons-Romero, F.; Ebenbichler, C.F.; Patsch, J.R.; Tilg, H. Adiponectin and its receptors in non-alcoholic steatohepatitis. Gut 2005, 54, 117–121. [Google Scholar]
  261. Stefan, N.; Machicao, F.; Staiger, H.; Machann, J.; Schick, F.; Tschritter, O.; Spieth, C.; Weigert, C.; Fritsche, A.; Stumvoll, M.; et al. Polymorphisms in the gene encoding adiponectin receptor 1 are associated with insulin resistance and high liver fat. Diabetologia 2005, 48, 2282–2291. [Google Scholar]
  262. Minokoshi, Y.; Kim, Y.B.; Peroni, O.D.; Fryer, L.G.; Muller, C.; Carling, D.; Kahn, B.B. Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature 2002, 415, 339–343. [Google Scholar]
  263. Kakuma, T.; Lee, Y.; Higa, M.; Wang, Z.W.; Pan, W.; Shimomura, I.; Unger, R.H. Leptin, troglitazone, and the expression of sterol regulatory element binding proteins in liver and pancreatic islets. Proc. Natl. Acad. Sci. USA 2000, 18, 8536–8441. [Google Scholar]
  264. Lee, Y.; Yu, X.; Gonzales, F.; Mangelsdorf, D.J.; Wang, M.Y.; Richardson, C.; Witters, L.A.; Unger, R.H. PPAR alpha is necessary for the lipogenic action of hyperleptinemia on white adipose and liver tissue. Proc. Natl. Acad. Sci. USA 2002, 99, 11848–11853. [Google Scholar]
  265. Serin, E.; Ozer, B.; Gümürdülü, Y.; Kayaselçuk, F.; Kul, K.; Boyacioğlu, S. Serum leptin level can be a negative marker of hepatocyte damage in nonalcoholic fatty liver. J. Gastroenterol 2003, 38, 471–476. [Google Scholar]
  266. Poordad, F.F. The role of leptin in NAFLD contender or pretender? J. Clin. Gastroenterol 2004, 38, 841–843. [Google Scholar]
  267. Tobe, K.; Ogura, T.; Tsukamoto, C.; Imai, A.; Matsuura, K.; Iwasaki, Y.; Shimomura, H.; Higashi, T.; Tsuji, T. Relationship between serum leptin and fatty liver in Japanese male adolescent university students. Am. J. Gastroenterol 1999, 94, 3328–3335. [Google Scholar]
  268. Marra, F. Leptin and liver fibrosis: A matter of fat. Gastroenterology 2002, 122, 1529–1532. [Google Scholar]
  269. Cao, Q.; Mak, K.M.; Ren, C.; Lieber, C.S. Leptin stimulates tissue inhibitor of metalloproteinase-1 in human hepatic stellate cells: Respective roles of the JAK/STAT and JAK-mediated H2O2-dependent MAPK pathways. J. Biol. Chem 2004, 279, 4292–4304. [Google Scholar]
  270. Saxena, N.K.; Titus, M.A.; Ding, X.; Floyd, J.; Srinivasan, S.; Sitaraman, S.V.; Anania, F.A. Leptin as a novel profibrogenic cytokine in hepatic stellate cells: Mitogenesis and inhibition of apoptosis mediated by extracellular regulated kinase (Erk) and Akt phosphorylation. FASEB J 2004, 18, 1612–1614. [Google Scholar]
  271. Banerjee, R.R.; Lazar, M.A. Resistin: Molecular history and prognosis. J. Mol. Med 2003, 81, 218–226. [Google Scholar]
  272. Pagano, C.; Soardo, G.; Pilon, C.; Milocco, C.; Basan, L.; Milan, G.; Donnini, D.; Faggian, D.; Mussap, M.; Plebani, M.; et al. Increased serum resistin in nonalcoholic fatty liver disease is related to liver disease severity and not to insulin resistance. J. Clin. Endocrinol. Metab 2006, 91, 1081–1086. [Google Scholar]
  273. Crespo, J.; Cayon, A.; Fernandez-Gil, P.; Hernández-Guerra, M.; Mayorga, M.; Domínguez-Díez, A.; Fernández-Escalante, J.C.; Pons-Romero, F. Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patients. Hepatology 2001, 34, 1158–1163. [Google Scholar]
  274. Ding, W.X.; Yin, X.M. Dissection of the multiple mechanisms of TNF-alpha-induced apoptosis in liver injury. J. Cell. Mol. Med 2004, 8, 445–454. [Google Scholar]
  275. Manco, M.; Marcellini, M.; Giannone, G.; Nobili, V. Correlation of serum TNF-alpha levels and histologic liver injury scores in pediatric nonalcoholic fatty liver disease. Am. J. Clin. Pathol 2007, 127, 954–960. [Google Scholar]
  276. Sanal, M.G. The blind men see the elephant-the many faces of fatty liver disease. World J. Gastroenterol 2008, 14, 831–844. [Google Scholar]
  277. Saleh, J.; Christou, N.; Cianflone, K. Regional specificity of ASP binding in human adipose tissue. Am. J. Physiol 1999, 276, E815–E821. [Google Scholar]
  278. Massiera, F.; Bloch-Faure, M.; Ceiler, D.; Murakami, K.; Fukamizu, A.; Gasc, J.M.; Quignard-Boulange, A.; Negrel, R.; Ailhaud, G.; Seydoux, J.; et al. Adipose angiotensinogen is involved in adipose tissue growth and blood pressure regulation. FASEB J 2001, 15, 2727–2729. [Google Scholar]
  279. Umemura, S.; Nyui, N.; Tamura, K.; Hibi, K.; Yamaguchi, S.; Nakamaru, M.; Ishigami, T.; Yabana, M.; Kihara, M.; Inoue, S.; et al. Plasma angiotensinogen concentrations in obese patients. Am. J. Hypertens 1997, 10, 629–633. [Google Scholar]
  280. Dixon, J.B.; Bhathal, P.S.; Jonsson, J.R.; Dixon, A.F.; Powell, E.E.; O’Brien, P.E. Pro-fibrotic polymorphisms predictive of advanced liver fibrosis in the severely obese. J. Hepatol 2003, 39, 967–971. [Google Scholar]
Figure 1. Concept of metabolic syndrome.
Figure 1. Concept of metabolic syndrome.
Ijms 15 06184f1
Figure 2. Secretion of inflammatory adipokines from adipose tissue in obese state. In obese state, the enlarged adipose tissue leads to dysregulated secretion of adipokines and increased release of free fatty acids. The free fatty acids and pro-inflammatory adipokines get to metabolic tissues, including skeletal muscle and liver, and modify inflammatory responses as well as glucose and lipid metabolism, thereby contributing to metabolic syndrome. In addition, obesity induces a phenotypic switch in adipose tissue from anti-inflammatory (M2) to pro-inflammatory (M1) macrophages. On the other hand, the adipose production of insulin-sensitizing adipokines with anti-inflammatory properties, such as adiponectin, is decreased in obese state. The red arrows indicate increased (when pointing upward) or decreased (when pointing downward) responses to obesity. ANGPTL, angiopoietin-like protein; ASP, acylation-stimulating protein; IL, interleukin; MCP-1, monocyte chemotactic protein; NAFLD, nonalcoholic fatty liver disease; PAI-1, plasminogen activator inhibitor-1; RBP4, retinol binding protein 4; SAA, serum amyloid A; SFRP5, secreted frizzled-related protein 5; TGF-β, Transforming growth factor-β; TNF-α, tumor necrosis factor-α.
Figure 2. Secretion of inflammatory adipokines from adipose tissue in obese state. In obese state, the enlarged adipose tissue leads to dysregulated secretion of adipokines and increased release of free fatty acids. The free fatty acids and pro-inflammatory adipokines get to metabolic tissues, including skeletal muscle and liver, and modify inflammatory responses as well as glucose and lipid metabolism, thereby contributing to metabolic syndrome. In addition, obesity induces a phenotypic switch in adipose tissue from anti-inflammatory (M2) to pro-inflammatory (M1) macrophages. On the other hand, the adipose production of insulin-sensitizing adipokines with anti-inflammatory properties, such as adiponectin, is decreased in obese state. The red arrows indicate increased (when pointing upward) or decreased (when pointing downward) responses to obesity. ANGPTL, angiopoietin-like protein; ASP, acylation-stimulating protein; IL, interleukin; MCP-1, monocyte chemotactic protein; NAFLD, nonalcoholic fatty liver disease; PAI-1, plasminogen activator inhibitor-1; RBP4, retinol binding protein 4; SAA, serum amyloid A; SFRP5, secreted frizzled-related protein 5; TGF-β, Transforming growth factor-β; TNF-α, tumor necrosis factor-α.
Ijms 15 06184f2
Figure 3. Schematic view of insulin signaling pathway in adipose tissue. Binding of insulin to its receptor on adipocytes triggers the phosphorylation and activation of insulin receptor substrate, which forms a docking site for phosphatidylinositol 3-kinase (PI3K) at the membrane. When docked, PI3K converts phosphatidylinositol 4,5-bisphosphate to phosphatidylinositol 3,4,5-trisphosphate, a second messenger that activates phosphoinositide-dependent protein kinase 1 and recruits Akt (also known as protein kinase B, PKB) to the cell membrane. Consequently, PI3K-AKT/PKB signaling pathway regulates metabolic processes. The red arrows indicate up-regulation (when pointing upward) or down-regulation (when pointing downward) in response to PI3K-AKT/PKB signaling pathway. The Ras-mitogen-activated protein kinase pathway leads to the activation of genes which are involved in cell growth, thereby promoting inflammation and atherogenesis. IRS-1, insulin receptor substrate; MAPK, mitogen-activated protein kinase; PDK, phosphoinositide-dependent protein kinase 1; PI3K, phosphatidylinositol 3-kinase; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol 3,4,5-trisphosphate; PKB, protein kinase B.
Figure 3. Schematic view of insulin signaling pathway in adipose tissue. Binding of insulin to its receptor on adipocytes triggers the phosphorylation and activation of insulin receptor substrate, which forms a docking site for phosphatidylinositol 3-kinase (PI3K) at the membrane. When docked, PI3K converts phosphatidylinositol 4,5-bisphosphate to phosphatidylinositol 3,4,5-trisphosphate, a second messenger that activates phosphoinositide-dependent protein kinase 1 and recruits Akt (also known as protein kinase B, PKB) to the cell membrane. Consequently, PI3K-AKT/PKB signaling pathway regulates metabolic processes. The red arrows indicate up-regulation (when pointing upward) or down-regulation (when pointing downward) in response to PI3K-AKT/PKB signaling pathway. The Ras-mitogen-activated protein kinase pathway leads to the activation of genes which are involved in cell growth, thereby promoting inflammation and atherogenesis. IRS-1, insulin receptor substrate; MAPK, mitogen-activated protein kinase; PDK, phosphoinositide-dependent protein kinase 1; PI3K, phosphatidylinositol 3-kinase; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol 3,4,5-trisphosphate; PKB, protein kinase B.
Ijms 15 06184f3
Figure 4. Mechanisms of dyslipidemia in obesity. An increased free fatty acids (FFA) release from adipose tissue via lipolysis can result in enhanced delivery of FFA to the liver. The enhanced FFA leads to increased triglyceride (TG) and very-low-density lipoprotein (VLDL) production in the liver as well as inhibition of lipoprotein lipase in adipose tissue and skeletal muscle, thereby promoting hypertriglyceridemia. Moreover, the increased VLDL in the liver can inhibit lipolysis of chylomicrons, which also contributes to hypertriglyceridemia. The TG in VLDL is exchanged for cholesteryl esters from low-density lipoproteins (LDL) and high-density lipoproteins (HDL) by the cholesteryl ester transport protein, producing TG-rich LDL and HDL. The TG in the LDL and HDL is then hydrolyzed by hepatic lipase, producing both small, dense LDL and HDL. The decreased HDL concentration and formation of small, dense LDL particules are linked to a higher risk of cardiovascular disease. The red arrows indicate increased (when pointing upward) or decreased (when pointing downward) responses to obesity. CE, cholesteryl esters; CETP, cholesteryl ester transport protein; FFA, free fatty acids; HDL, high-density lipoproteins; HL, hepatic lipase; LDL, low-density lipoproteins; LPL, lipoprotein lipase; TG, triglyceride; VLDL, very-low-density lipoprotein.
Figure 4. Mechanisms of dyslipidemia in obesity. An increased free fatty acids (FFA) release from adipose tissue via lipolysis can result in enhanced delivery of FFA to the liver. The enhanced FFA leads to increased triglyceride (TG) and very-low-density lipoprotein (VLDL) production in the liver as well as inhibition of lipoprotein lipase in adipose tissue and skeletal muscle, thereby promoting hypertriglyceridemia. Moreover, the increased VLDL in the liver can inhibit lipolysis of chylomicrons, which also contributes to hypertriglyceridemia. The TG in VLDL is exchanged for cholesteryl esters from low-density lipoproteins (LDL) and high-density lipoproteins (HDL) by the cholesteryl ester transport protein, producing TG-rich LDL and HDL. The TG in the LDL and HDL is then hydrolyzed by hepatic lipase, producing both small, dense LDL and HDL. The decreased HDL concentration and formation of small, dense LDL particules are linked to a higher risk of cardiovascular disease. The red arrows indicate increased (when pointing upward) or decreased (when pointing downward) responses to obesity. CE, cholesteryl esters; CETP, cholesteryl ester transport protein; FFA, free fatty acids; HDL, high-density lipoproteins; HL, hepatic lipase; LDL, low-density lipoproteins; LPL, lipoprotein lipase; TG, triglyceride; VLDL, very-low-density lipoprotein.
Ijms 15 06184f4
Figure 5. The Multi-hit hypothesis of NAFLD pathogenesis. The “first hit”, such as insulin resistance and lipid metabolism dysregulation, leads to the development of simple steatosis and renders hepatocytes susceptible to “multi-hit”, which include gut-derived bacterial toxins, adipocytokine imbalance, mitochondrial dysfunction, oxidative damage, dysregulated hepatocyte apoptosis, activation of pro-fibrogenic factors and pro-inflammatory mediators and hepatic stellate cell activation, ultimately leading to NASH and cirrhosis. The red arrows indicate up-regulation (when pointing upward) or down-regulation (when pointing downward) in response to insulin resistance.
Figure 5. The Multi-hit hypothesis of NAFLD pathogenesis. The “first hit”, such as insulin resistance and lipid metabolism dysregulation, leads to the development of simple steatosis and renders hepatocytes susceptible to “multi-hit”, which include gut-derived bacterial toxins, adipocytokine imbalance, mitochondrial dysfunction, oxidative damage, dysregulated hepatocyte apoptosis, activation of pro-fibrogenic factors and pro-inflammatory mediators and hepatic stellate cell activation, ultimately leading to NASH and cirrhosis. The red arrows indicate up-regulation (when pointing upward) or down-regulation (when pointing downward) in response to insulin resistance.
Ijms 15 06184f5

Share and Cite

MDPI and ACS Style

Jung, U.J.; Choi, M.-S. 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. 2014, 15, 6184-6223. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15046184

AMA Style

Jung UJ, Choi M-S. Obesity and Its Metabolic Complications: The Role of Adipokines and the Relationship between Obesity, Inflammation, Insulin Resistance, Dyslipidemia and Nonalcoholic Fatty Liver Disease. International Journal of Molecular Sciences. 2014; 15(4):6184-6223. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15046184

Chicago/Turabian Style

Jung, Un Ju, and Myung-Sook Choi. 2014. "Obesity and Its Metabolic Complications: The Role of Adipokines and the Relationship between Obesity, Inflammation, Insulin Resistance, Dyslipidemia and Nonalcoholic Fatty Liver Disease" International Journal of Molecular Sciences 15, no. 4: 6184-6223. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15046184

Article Metrics

Back to TopTop