Skip to main content
  • Review Article
  • Open access
  • Published:

Adipokines and Insulin Resistance

Abstract

Obesity is associated with an array of health problems in adult and pediatric populations. Understanding the pathogenesis of obesity and its metabolic sequelae has advanced rapidly over the past decades. Adipose tissue represents an active endocrine organ that, in addition to regulating fat mass and nutrient homeostasis, releases a large number of bioactive mediators (adipokines) that signal to organs of metabolic importance including brain, liver, skeletal muscle, and the immune system— thereby modulating hemostasis, blood pressure, lipid and glucose metabolism, inflammation, and atherosclerosis. In the present review, we summarize current data on the effect of the adipose tissue-derived hormones adiponectin, chemerin, leptin, omentin, resistin, retinol binding protein 4, tumor necrosis factor-α and interleukin-6, vaspin, and visfatin on insulin resistance.

Introduction

Adipose tissue is composed of adipocytes embedded in a loose connective tissue meshwork containing adipocyte precursors, fibroblasts, immune cells, and various other cell types. Adipose tissue was traditionally considered an energy storage depot with few interesting attributes. Due to the dramatic rise in obesity and its metabolic sequelae during the past decades, adipose tissue gained tremendous scientific interest. It is now regarded as an active endocrine organ that, in addition to regulating fat mass and nutrient homeostasis, releases a large number of bioactive mediators (adipokines) modulating hemostasis, blood pressure, lipid and glucose metabolism, inflammation, and atherosclerosis. Table 1 summarizes the adipokines discussed in this review article, the interplay between adipokines, and their effects on glucose homeostasis.

Table 1 Adipokines, adipokine interplay and the effects on glucose homeostasisa

Adiponectin

Adiponectin expression occurs from an intermediate stage of adipogenesis onwards (1,2), and represents the most abundant protein secreted by adipose tissue. Unlike most other adipokines, plasma adiponectin levels were reduced in animal models of obesity and insulin resistance (2,3). Administration of recombinant adiponectin to rodents resulted in increased glucose uptake and fat oxidation in muscle, reduced hepatic glucose production, and improved whole-body insulin sensitivity (46). Adiponectin transgenic mice showed partial amelioration of insulin resistance and diabetes (7) and suppression of endogenous glucose production (8). In contrast, adiponectin-deficient mice exhibited insulin resistance and glucose intolerance (911). In addition to its insulin-sensitizing effects, adiponectin may alter glucose metabolism through stimulation of pancreatic insulin secretion in vivo (12). Apart from its peripheral actions, adiponectin was shown to modulate food intake and energy expenditure during fasting (increased food intake and reduced energy expenditure) and refeeding (opposite effects) through its effects in the central nervous system (13).

In humans, plasma adiponectin levels were correlated negatively with adiposity (1417), insulin resistance (16,18,19), type 2 diabetes (16,20), and metabolic syndrome (2123), yet positively correlated with markers of insulin sensitivity in frequently sampled intravenous glucose tolerance testing (17,24,25) and clamp studies (15,19). Prospective and longitudinal studies indicated that lower adiponectin levels were associated with a higher incidence of type 2 diabetes (2633). Adiponectin single nucleotide polymorphisms (SNPs) have been associated variably with increased body mass index (BMI), insulin resistance-related traits, and type 2 diabetes (34). However, in a systematic meta-analysis of all published data on adiponectin SNPs, only the +276G→T variant was a strong determinant of insulin resistance with minor allele homozygotes having a lower homeostasis model assessment of insulin resistance index than carriers of other genotypes. No consistent effect on BMI or risk of type 2 diabetes was observed (34).

Adiponectin circulates in plasma as a low-molecular weight trimer, a middle-molecular weight hexamer, and high-molecular weight (HMW) 12- to 18-mer, and these forms were postulated to differ in biologic activity (35,36). HMW adiponectin was proposed to be the biologically active form of the hormone (37), and, although not unchallenged (38), was shown to be superior to total adiponectin in predicting insulin resistance and the metabolic syndrome trait cluster (3941). Adiponectin expression and secretion was demonstrated to be upregulated by thiazolidinediones (TZDs) (4244), and HMW adiponectin is the predominant form of adiponectin increased by TZDs (37).

Adiponectin’s effects on glucose metabolism are mediated through two distinct receptors termed adiponectin receptor 1 (AdipoR1) and adiponectin receptor 2 (AdipoR2). AdipoR1 is expressed ubiquitously, whereas AdipoR2 is expressed most abundantly in the liver (45). Similar to adiponectin, expression of both receptors was decreased in mouse models of obesity and insulin resistance (46,47). Yamauchi et al. (47) reported that liver-specific adenoviral expression of AdipoR1 in leptin-receptor deficient db/db mice resulted in activation of 5′AMP-activated protein kinase (AMPK), leading to reduced expression of genes encoding hepatic gluconeogenic enzymes such as glucose-6-phosphatase and phosphoenolpyruvate carboxykinase 1, and of genes encoding molecules involved in lipogenesis, such as sterol regulatory element-binding protein 1c. Hepatic expression of AdipoR2 increased expression of genes involved in hepatic glucose uptake such as glucokinase, and of peroxisome proliferator-activated receptor-α (PPAR-α) and its target genes such as acyl-CoA oxidase and uncoupling protein 2. Activation of AMPK reduced endogenous hepatic glucose production, while expression of both receptors increased fatty acid oxidation, decreased hepatic triglyceride content and improved insulin resistance. Conversely, targeted disruption of AdipoR1 resulted in the abrogation of adiponectin-induced AMPK activation, and in increased endogenous glucose production and insulin resistance. Knockout of AdipoR2 caused decreased activity of PPAR-α signaling pathways and insulin resistance. Simultaneous disruption of both AdipoR1 and AdipoR2 abolished adiponectin binding and actions, resulting in increased glucose intolerance and insulin resistance compared with the single knockout models (47). The role of T-cadherin, another putative adiponectin receptor (48), in adiponectin signaling appeared to be minor since, in contrast to control mice, administration of adiponectin to AdipoR1/R2 double knockout mice did not improve plasma glucose levels (47). The impact of AdipoR1 and AipoR2 on glucose metabolism in rodents has been examined by two more studies with, in part, conflicting results. Bjursell et al. (49) reported that AdipoR1 knockout mice showed increased adiposity associated with decreased glucose tolerance, reduced spontaneous locomotor activity, and decreased energy expenditure. Unexpectedly, however, AdipoR2 deficient mice were lean and resistant to high fat diet-induced obesity associated with improved glucose tolerance and increased spontaneous locomotor activity and energy expenditure. Consistent with these data, Liu et al. (50) demonstrated that disruption of AdipoR2 diminished high fat-induced insulin resistance and reduced plasma glucose levels in leptin-deficient ob/ob mice. However, glucose homeostasis in these animals on long-term high fat diet deteriorated because of failure of pancreatic β-cells to compensate for the moderate insulin resistance.

In humans, data regarding a possible association of adiponectin receptor expression in adipose tissue or skeletal muscle and obesity or insulin resistance were highly divergent and dependent on the population studied (5159). Furthermore, although polymorphisms in both adiponectin receptor genes have been found to be associated with insulin resistance and type 2 diabetes (34,60), these associations have not been replicated widely across populations. Thus, the number of studies available to date is still too small to draw firm conclusions on the role of variability in AdipoR1 and/or AdipoR2 expression in predicting insulin resistance and related disorders.

In summary, adiponectin is an abundantly expressed adipokine that exerts a potent insulin-sensitizing effect through binding to its receptors AdipoR1 and AdipoR2, leading to activation of AMPK, PPAR-α, and presumably other yet-unknown signaling pathways. In obesity-linked insulin resistance, both adiponectin and adiponectin receptors are downregulated. Upregulation of adiponectin/adiponectin receptors or enhancing adiponectin receptor function may represent an interesting therapeutic strategy for obesity-linked insulin resistance.

Chemerin

Chemerin (RARRES2 or TIG2) is a recently discovered chemokine (61) highly expressed in liver and white adipose tissue (62,63). It exerts potent antiinflammatory effects on activated macrophages expressing the chemerin receptor CMKLR1 (chemokine-like receptor-1) in a cysteine protease-dependent manner (64). Furthermore, chemerin is crucial for normal adipocyte differentiation and modulates the expression of adipocyte genes involved in glucose and lipid homeostasis, such as glucose transporter-4, fatty acid synthase, and adiponectin via its own receptor (62,63,65). In 3T3-L1 adipocytes, chemerin was reported to enhance insulin-stimulated glucose uptake and insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation, suggesting that chemerin may increase insulin sensitivity in adipose tissue (66). Conflicting data exist regarding the association of chemerin with obesity and diabetes in rodents. Chemerin expression was shown to be decreased in adipose tissue of db/db mice compared with controls (66). In contrast, chemerin expression was significantly higher in adipose tissue of impaired glucose tolerant and diabetic Psammomys obesus compared with normal glucose tolerant sand rats (62). In humans, chemerin levels did not differ significantly between subjects with type 2 diabetes and normal controls. However, in normal glucose tolerant subjects, chemerin levels were associated significantly with BMI, triglycerides, and blood pressure (62). Further studies are needed to determine the physiological role of chemerin in glucose metabolism, and to identify chemerin’s target tissues as well as relevant signal transduction pathways.

Leptin

Since its identification in 1994, leptin has attracted much attention as one of the most important signals for the regulation of food intake and energy homeostasis (6769). Hypothalamic as well as brain stem nuclei play a critical role in integrating the information on absorbed food, on the amount of energy stored in the form of fat and on blood glucose levels to regulate feeding, energy storage, or expenditure. Leptin receptor activation at these sites leads to repression of orexigenic pathways (for example, those involving neuropeptide Y [NPY] and agouti-related peptide [AgRP]) and induction of anorexigenic pathways (such as those involving pro-opiomelanocortin [POMC] and cocaine and amphetamine-regulated transcript [CART]) via Janus kinase (JAK)-signal transducers and activators of transcription (STAT) and IRS/phosphoinositide-3 kinase (PI3K) signaling (70). Although changes in food intake and total body fat clearly can affect insulin sensitivity in peripheral tissues, several observations suggested that leptin regulation of glucose homeostasis occurs independently of its effects on food intake through central and peripheral mechanisms. Hypothalamic arcuate nucleus (ARC)-specific expression of leptin receptor in leptin receptor-deficient mice resulted in a modest reduction of food intake and body fat mass, yet normalized blood glucose and insulin levels (71). ARC-specific restoration of leptin receptor in obese, leptin receptor-deficient Koletsky rats markedly improved insulin sensitivity via a mechanism that was not dependent on reduced food intake, but was attenuated by intraventricular infusion of a PI3K inhibitor (72). ARC-directed expression of a constitutively active mutant of protein kinase B, an enzyme activated by PI3K, mimicked the insulin-sensitizing effect of restored hypothalamic leptin signaling in these animals. In contrast, mice with a mutant leptin receptor that cannot signal via the JAK-STAT pathway, yet otherwise functions normally, developed severe hyperphagia and obesity, but unlike leptin receptor-deficient mice, exhibited only mild disturbances of glucose homeostasis that can be prevented by caloric restriction (73). These results suggested that although leptin receptor-mediated JAK-STAT signaling is essential for regulation of food intake and body weight, leptin-stimulated PI3K signaling appears to be important for regulation of glucose metabolism. Leptin also limits accumulation of triglycerides in liver and skeletal muscle through a combination of direct activation of AMPK and indirect actions mediated through central neural pathways, thereby improving insulin sensitivity (74,75). Furthermore, leptin modulates pancreatic β-cells function through direct actions (76,77) and indirectly through central neural pathways (78,79). Leptin was shown to inhibit insulin secretion in lean animals. As body weight increased, leptin signaling protected the β-cell from adverse effects of overnutrition such as lipid accumulation, thus improving β-cell function (77). Insulin stimulates both leptin biosynthesis and secretion from adipose tissue establishing a classic endocrine adipo-insular feedback loop; the so-called “adipo-insular axis” (80).

The concept of “leptin resistance” was introduced when increased adipose leptin production was observed in the majority of obese individuals without adequate leptin-mediated end-organ response (81). Leptin improves glucose homeostasis in humans with lipodystrophy or congenital leptin deficiency (82,83). However, results in humans with ‘typical’ obesity were disappointing in this regard (84). Studies in obese rodents suggested that leptin resistance is associated with impairment of leptin transport across the blood-brain-barrier (BBB), reduction of leptin-mediated JAK-STAT signaling, and induction of suppressor of cytokine signaling-3 (SOCS-3) (81,85). Attenuation of leptin sensitivity in the brain leads to excess triglyceride accumulation in adipose tissue, as well as muscle, liver, and pancreas, resulting in impaired insulin sensitivity and secretion (86). The concept of “leptin resistance” has been challenged recently by an alternate concept of “hypothalamic leptin insufficiency.” The major tenet of this postulation is that BBB restricts the blood-to-brain entry of leptin in response to hyperleptinemia resulting in leptin insufficiency at multiple target sites in the brain (87).

In summary, leptin serves as a major ‘adipostat’ by repressing food intake and promoting energy expenditure. Independent of these effects, leptin improves peripheral (hepatic and skeletal muscle) insulin sensitivity and modulates pancreatic β-cell function. In the majority of cases of obesity, despite both an intact leptin receptor and high circulating leptin levels, leptin fails to induce weight loss. This diminished response to the anorexigenic and insulin-sensitizing effects of leptin is called “leptin resistance.”

Omentin

Omentin is a fat depot-specific secretory protein synthesized by visceral stromal vascular cells, but not adipocytes. Omentin enhanced insulin-stimulated glucose transport and Akt phosphorylation in human subcutaneous and visceral adipocytes, suggesting that omentin may improve insulin sensitivity (88). Plasma omentin-1 levels, the major circulating isoform in human plasma, were correlated inversely with obesity and insulin resistance as determined by homeostasis model assessment yet correlated positively with adiponectin and HDL levels (89). Administration of glucose and insulin to human omental adipose tissue explants resulted in a dose-dependent reduction of omentin-1 expression. Furthermore, prolonged insulin-glucose infusion in healthy individuals resulted in significantly decreased plasma omentin-1 levels (90). The physiological role of omentin in glucose metabolism, omentin’s target tissues, a receptor, or relevant signal transduction pathways still need to be determined.

Resistin

Resistin, a member of the resistin-like molecule (RELM) family of cysteine-rich proteins, has a controversial history regarding its role in the pathogenesis of obesity-mediated insulin resistance and type 2 diabetes. Resistin was discovered in 2001 as a TZD-downregulated gene in mouse adipocytes (91). In rodents, circulating levels of resistin were increased in obesity (92), and both gain- (91,9396) and loss-of-function studies (91,9799) demonstrated a role for resistin in mediating hepatic or skeletal muscle (depending on the animal model) insulin resistance (94,95,97,98). There is considerable controversy about the role of resistin in humans. Human resistin is produced and secreted mainly by peripheral-blood mononuclear cells (100). Human studies over the past years reported contradictory findings with regards to a physiological role for resistin in glucose metabolism. Several groups suggested resistin levels and SNPs to be associated with obesity, insulin resistance, and type 2 diabetes (19,101106). However, other groups failed to identify changes in resistin levels or SNPs in these conditions (107114). Although a clear function for resistin in glucose metabolism in humans is still lacking, data indicate that resistin has a role in inflammatory processes. The expression of resistin in human peripheral-blood mononuclear cells is upregulated by the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) (115). Conversely, resistin induced the expression of TNF-α and IL-6 in white adipose tissue and in peripheral-blood mononuclear cells (116118). Plasma resistin levels were reported to be associated with many inflammatory markers including C-reactive protein (119), soluble TNF-α receptor-2, IL-6, and lipoprotein-associated phospholipase A2 (120) in several pathophysiological conditions. Resistin was shown to be associated with disease activity in patients with inflammatory bowel disease (121), to correlate with severity of disease in severe sepsis and septic shock (122), and to be associated with coronary artery disease (120). Furthermore, resistin may be involved in the pathogenesis of rheumatoid arthritis (117). Considering the crosstalk between inflammatory pathways and the insulin signaling cascade (see below “Tumor necrosis factor-α and interleukin-6”), resistin may represent a link between inflammation and metabolic signals (123).

Retinol Binding Protein 4

A potential link between retinol binding protein 4 (RBP4) and type 2 diabetes was suggested by Yang et al. (124) reporting that RBP4 was elevated in insulin-resistant adipose specific GLUT4 knockout mice and humans with obesity and type 2 diabetes. Transgenic overexpression of human RBP4 in wildtype mice or administration of recombinant RBP4 to wildtype mice was shown to cause insulin resistance through induction of hepatic expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase and impairment of skeletal muscle insulin signaling. In contrast, genetic deletion of RBP4 enhanced insulin sensitivity. The effects of RBP4 may be mediated through retinol-dependent (via retinoic acid receptors [RARs] and retinoic acid-X receptors [RXRs] to regulate gene transcription) or retinol-independent mechanisms (for example, interaction with cell surface receptors such as Megalin/gp320). Serum RBP4 concentrations were elevated in insulin-resistant humans with obesity, impaired glucose tolerance and type 2 diabetes, and even in lean normoglycemic subjects with a strong family history of type 2 diabetes (124,125). A large number of subsequent studies confirmed an association of increased circulating RBP4 levels with various aspects of adiposity (126129), insulin resistance (128,130133), type 2 diabetes (127,134,135) and the metabolic syndrome (136). Improving insulin sensitivity by interventions such as exercise training, lifestyle modification, or gastric banding surgery reduced serum RBP4 levels in humans (125,126,137,138). Genetic studies reported an association of RBP4 SNPs and insulin resistance, impaired insulin secretion, and/or type 2 diabetes (139,140). Other studies, however, failed to establish an association of RBP4 levels with obesity (141143), insulin resistance (141144), type 2 diabetes (144) or components of the metabolic syndrome (145). This discrepancy may be explained in part through methodological differences in the measurement of RBP4 (146) as well as differences in the study populations.

Based on current data, the function of RBP4 as an adipokine exerting metabolic effects in glucose metabolism in humans remains uncertain and might be restricted to rodent models (147).

Tumor Necrosis Factor-α and Interleukin-6

Both TNF-α and IL-6, the most widely studied cytokines produced by adipose tissue, were reported to modulate insulin resistance. Evidence supporting a key role for TNF-α in obesity-related insulin resistance came from studies showing that deletion of TNF-α or TNF-α receptors resulted in significantly improved insulin sensitivity in both diet-induced obese mice and leptin-deficient ob/ob mice (148). In humans, adipose tissue TNF-α expression correlated with BMI, percentage of body fat, and hyperinsulinemia, whereas weight loss decreased TNF-α levels (149). Fasting TNF-α plasma levels were associated with insulin resistance in the Framingham Offspring Study (19). Neutralization of TNF-α improved insulin resistance in obese rats (150). However, infusion of TNF-α-neutralizing antibodies to obese, insulin-resistant subjects, or type 2 diabetic patients, did not improve insulin sensitivity (151,152).

Conflicting data exist regarding the role of IL-6 in insulin resistance (153, 154). IL-6 was reported to reduce insulin-dependent hepatic glycogen synthesis (155,156) and glucose uptake in adipocytes (157), whereas insulin-dependent glycogen synthesis and glucose uptake was enhanced in myotubes (158,159). The effect of IL-6 on hepatic glucose production is still under debate (160,161). Kim et al. (160) reported that IL-6 infusion in mice blunted insulin’s ability to suppress hepatic glucose production. In contrast, Inoue et al. (161) demonstrated that intraventricular insulin infusion resulted in IL-6-mediated suppression of hepatic gluconeogenesis. Overall, circulating IL-6 levels are increased in obese and insulin resistant subjects (162,163). One may speculate that persistent systemic increases of IL-6 in states of chronic inflammation such as obesity and type 2 diabetes may trigger insulin resistance, whereas transient increases may contribute to normal glucose homeostasis. TNF-α and IL-6 modulate insulin resistance through several distinct mechanisms, including c-Jun N-terminal kinase 1 (JNK1)-mediated serine phosphorylation of IRS-1, IκB kinase (IKK)-mediated nuclear factor-κB (NF-κB) activation, and induction of SOCS-3 (164).

Vaspin

Visceral adipose tissue-derived serine protease inhibitor (vaspin) was identified in visceral adipose tissue of Otsuka Long-Evans Tokushima fatty rats at an age when body weight and hyperinsulinemia peaked (165). Vaspin expression was shown to decrease with worsening of diabetes and body weight loss. Administration of recombinant human vaspin to a mouse model of diet-induced obesity improved glucose tolerance and insulin sensitivity, suggesting that vaspin may represent an insulin-sensitizing adipokine. Human vaspin mRNA was reported to be expressed in visceral and subcutaneous adipose tissue. It was shown to be regulated in a fat-depot specific manner, and to be associated with obesity and parameters of insulin resistance (166). Likewise, elevated vaspin serum concentrations were correlated with obesity and impaired insulin sensitivity, whereas type 2 diabetes seemed to abrogate this correlation (167).

Much remains to be learned about the role of vaspin in glucose metabolism. Identification of vaspin’s protease substrate is crucial to understand how vaspin may modulate insulin resistance.

Visfatin/PBEF/Nampt

Visfatin, originally isolated as a presumptive cytokine named pre-B cell colony-enhancing factor (PBEF) that enhances the maturation of B cell precursors (168), and displays nicotinamide phosphoribosyltransferase (Nampt) activity (169), was reported to be highly correlated with the amount of visceral fat in humans and in a mouse model of obesity and insulin resistance, to exert insulinmimetic effects in cultured cells, and to lower plasma glucose levels in mice (170). Although this study was retracted in 2007 due to numerous scientific flaws, the original observation was supported by a number of subsequent studies demonstrating that plasma visfatin levels in humans correlate with obesity, visceral fat mass, type 2 diabetes, and presence of the metabolic syndrome (171173). Furthermore, visfatin promoter SNPs were reported to be associated with fasting glucose and insulin levels, as well as type 2 diabetes (174,175). Other studies, however, did not confirm an association of visfatin and visceral adipose tissue or parameters of insulin sensitivity in humans and rodents (176179). Recent data pointed to an important role of visfatin/PBEF/Nampt in pancreatic β-cell function. In contrast to the results of Fukahara et al. (170), Revello et al. (180) demonstrated that the extracellular form of Nampt (eNampt/Visfatin/PBEF), which is secreted through a non-classical secretory pathway, did not show insulinmimetic effects in vitro or in vivo, but rather exhibited robust nicotinamide adenine dinucleotide (NAD) biosynthetic activity. Haplodeficiency and chemical inhibition of Nampt resulted in significantly decreased NAD biosynthesis and glucose-stimulated insulin secretion in pancreatic islets in vitro and in vivo. Conversely, administration of the Nampt reaction product nicotinamide mononucleotide (NMN) resulted in an amelioration of these defects.

In summary, current data suggest that adipose tissue as a natural source of eNampt/visfatin/PBEF may regulate β-cell function through secretion of eNampt and extracellular biosynthesis of NMN.

Adipokine Interplay

Insulin resistance should be conceptualized in a very broad manner that takes into account the interplay between nutrition, glucose, insulin and adipokines in various tissues of metabolic importance. Interactions between distinct adipokines add additional complexity to the picture (Table 1, Figure 1). Current data on adipokine interplay are rather sparse and, in part, contradictory due to examination of different in vitro (different cell types) and in vivo (different species) models.

Figure 1
figure 1

Obesity, adipokines and insulin resistance. Murine resistin is expressed in white adipose tissue, whereas in humans, resistin is mainly produced by peripheral-blood mononuclear cells. Green arrows depict stimulation, red lines suppression of gene expression.

Adiponectin and TNF-α control each other’s synthesis and activity, thus creating a balanced physiologic situation (164). Overnutrition results in activation of inflammatory pathways causing a critical imbalance leading to decreased expression of adiponectin. TNF-α and IL-6 play a key role in the regulation of many adipokines. TNF-α was reported to downregulate RBP4 production in human adipocytes (181). Expression of leptin (182,183), resistin (115), and visfatin/PBEF/eNampt (184) is increased by TNF-α and IL-6. Conversely, leptin (185), resistin (116118), and visfatin/PBEF/eNampt (186) upregulate the production of TNF-α and IL-6, suggesting that these adipokines could trigger or participate in the inflammatory process through direct paracrine and/or autocrine actions. Leptin, however, also was reported to suppress the expression of resistin and RBP4 (92,187,188), and to increase adiponectin expression in leptin-deficient ob/ob mice (188,189). Chemerin and vaspin, like adiponectin, were shown to have antiinflammatory properties. Chemerin inhibited the production of TNF-α and IL-6 by classically activated macrophages (64). Furthermore, knockdown of chemerin in 3T3-L1 adipocytes reduced adiponectin expression (63). Vaspin suppressed the expression of leptin, resistin, and TNF-α in white adipose tissue, yet increased the expression of adiponectin (165). No data have yet been reported on the interaction between omentin and other adipokines.

Conclusion

Obesity has reached dramatic proportions affecting more than 1 billion adults worldwide (190). The epidemic of obesity also affects children becoming overweight at progressively younger ages. Obesity is associated with an array of health problems including insulin resistance and type 2 diabetes, fatty liver disease, atherosclerosis, airway diseases, degenerative disorders, and various types of cancer. Our understanding of the pathogenesis of obesity and its metabolic sequelae has advanced significantly over the past decades. Environmental factors, such as sedentary lifestyle and increased calorie intake, in combination with an unfavorable genotype, are responsible for the epidemic of obesity. Excess visceral fat accumulation results in altered release of adipokines, leading to CNS-mediated skeletal muscle and hepatic insulin resistance (Figure 1). Understanding of the diverse effects of distinct adipokines and the interactions between these bioactive mediators is still incomplete. Unraveling the pathophysiological roles of adipokines in obesity-induced diseases likely will result in new pharmacotherapeutic approaches.

Disclosure

The authors have nothing to disclose.

References

  1. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. (1995) A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol. Chem. 270:26746–9.

    Article  CAS  PubMed  Google Scholar 

  2. Hu E, Liang P, Spiegelman BM. (1996) AdipoQ is a novel adipose-specific gene dysregulated in obesity. JBiol. Chem. 271:10697–703.

    Article  CAS  Google Scholar 

  3. Hotta K, et al. (2001) Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 50:1126–33.

    Article  CAS  PubMed  Google Scholar 

  4. Fruebis J, et al. (2001) Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc. Natl. Acad. Sci. U. S. A. 98:2005–10.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. (2001) The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat. Med. 7:947–53.

    Article  CAS  PubMed  Google Scholar 

  6. Yamauchi T, et al. (2001) The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 7:941–6.

    Article  CAS  PubMed  Google Scholar 

  7. Yamauchi T, et al. (2003) Globular adiponectin protected ob/ob mice from diabetes and ApoE-deficient mice from atherosclerosis. J. Biol. Chem. 278:2461–8.

    Article  CAS  PubMed  Google Scholar 

  8. Combs TP, et al. (2004) A transgenic mouse with a deletion in the collagenous domain of adiponectin displays elevated circulating adiponectin and improved insulin sensitivity. Endocrinology 145:367–83.

    Article  CAS  PubMed  Google Scholar 

  9. Kubota N, et al. (2002) Disruption of adiponectin causes insulin resistance and neointimal formation. JBiol. Chem. 277:25863–6.

    Article  CAS  Google Scholar 

  10. Maeda N, et al. (2002) Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat. Med. 8:731–7.

    Article  CAS  PubMed  Google Scholar 

  11. Nawrocki AR, et al. (2006) Mice lacking adiponectin show decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome proliferator-activated receptor gamma agonists. J Biol. Chem. 281:2654–60.

    Article  CAS  PubMed  Google Scholar 

  12. Okamoto M, et al. (2008) Adiponectin induces insulin secretion in vitro and in vivo at a low glucose concentration. Diabetologia 51:827–35.

    Article  CAS  PubMed  Google Scholar 

  13. Kubota N, et al. (2007) Adiponectin stimulates AMP-activated protein kinase in the hypothalamus and increases food intake. Cell Metab. 6:55–68.

    Article  CAS  PubMed  Google Scholar 

  14. Cnop M, et al. (2003) Relationship of adiponectin to body fat distribution, insulin sensitivity and plasma lipoproteins: evidence for independent roles of age and sex. Diabetologia 46:459–69.

    Article  CAS  PubMed  Google Scholar 

  15. Tschritter O, et al. (2003) Plasma adiponectin concentrations predict insulin sensitivity of both glucose and lipid metabolism. Diabetes 52:239–43.

    Article  CAS  PubMed  Google Scholar 

  16. Weyer C, et al. (2001) Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J. Clin. Endocrinol. Metab. 86:1930–5.

    Article  CAS  PubMed  Google Scholar 

  17. Hanley AJ, et al. (2007) Associations of adiponectin with body fat distribution and insulin sensitivity in nondiabetic Hispanics and African-Americans. J. Clin. Endocrinol. Metab. 92:2665–71.

    Article  CAS  PubMed  Google Scholar 

  18. Bacha F, Saad R, Gungor N, Arslanian SA. (2004) Adiponectin in youth: relationship to visceral adiposity, insulin sensitivity, and beta-cell function. Diabetes Care 27:547–52.

    Article  CAS  PubMed  Google Scholar 

  19. Hivert MF, et al. (2008) Associations of adiponectin, resistin, and tumor necrosis factor-alpha with insulin resistance. J. Clin. Endocrinol. Metab. 93:3165–72.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Hotta K, et al. (2000) Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler. Thromb. Vasc. Biol. 20:1595–9.

    Article  CAS  PubMed  Google Scholar 

  21. Gilardini L, et al. (2006) Adiponectin is a candidate marker of metabolic syndrome in obese children and adolescents. Atherosclerosis 189:401–7.

    Article  CAS  PubMed  Google Scholar 

  22. Mohan V, et al. (2005) Association of low adiponectin levels with the metabolic syndrome— the Chennai Urban Rural Epidemiology Study (CURES-4). Metabolism 54:476–81.

    Article  CAS  PubMed  Google Scholar 

  23. Wang J, et al. (2008) Adiponectin and metabolic syndrome in middle-aged and elderly Chinese. Obesity (Silver Spring) 16:172–8.

    Article  CAS  Google Scholar 

  24. Pellmé F, et al. (2003) Circulating adiponectin levels are reduced in nonobese but insulin-resistant first-degree relatives of type 2 diabetic patients. Diabetes 52:1182–6.

    Article  PubMed  Google Scholar 

  25. Winzer C, et al. (2004) Plasma adiponectin, insulin sensitivity, and subclinical inflammation in women with prior gestational diabetes mellitus. Diabetes Care 27:1721–7.

    Article  CAS  PubMed  Google Scholar 

  26. Lindsay RS, et al. (2002) Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet 360:57–8.

    Article  CAS  PubMed  Google Scholar 

  27. Daimon M, et al. (2003) Decreased serum levels of adiponectin are a risk factor for the progression to type 2 diabetes in the Japanese Population: the Funagata study. Diabetes Care 26:2015–20.

    Article  CAS  PubMed  Google Scholar 

  28. Snehalatha C, et al. (2003) Plasma adiponectin is an independent predictor of type 2 diabetes in Asian Indians. Diabetes Care 26:3226–9.

    Article  PubMed  Google Scholar 

  29. Spranger J, et al. (2003) Adiponectin and protection against type 2 diabetes mellitus. Lancet 361:226–8.

    Article  CAS  PubMed  Google Scholar 

  30. Duncan BB, et al. (2004) Adiponectin and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes 53:2473–8.

    Article  CAS  PubMed  Google Scholar 

  31. Krakoff J, et al. (2003) Inflammatory markers, adiponectin, and risk of type 2 diabetes in the Pima Indian. Diabetes Care 26:1745–51.

    Article  CAS  PubMed  Google Scholar 

  32. Snijder MB, et al. (2006) Associations of adiponectin levels with incident impaired glucose metabolism and type 2 diabetes in older men and women: The Hoorn Study. Diabetes Care 29:2498–503.

    Article  CAS  PubMed  Google Scholar 

  33. Mather KJ, et al. (2008) Adiponectin, change in adiponectin, and progression to diabetes in the Diabetes Prevention Program. Diabetes 57:980–6.

    Article  CAS  PubMed  Google Scholar 

  34. Menzaghi C, Trischitta V, Doria A. (2007) Genetic influences of adiponectin on insulin resistance, type 2 diabetes, and cardiovascular disease. Diabetes 56:1198–209.

    Article  CAS  PubMed  Google Scholar 

  35. Waki H, et al. (2003) Impaired multimerization of human adiponectin mutants associated with diabetes: molecular structure and multimer formation of adiponectin. JBiol. Chem. 278:40352–63.

    Article  CAS  Google Scholar 

  36. Wang Y, et al. (2006) Posttranslational modifications on the four conserved lysine residues within the collagenous domain of adiponectin are required for the formation of its high-molecular-weight oligomeric complex. J Biol. Chem. 281:16391–400.

    Article  CAS  PubMed  Google Scholar 

  37. Pajvani UB, et al. (2004) Complex distribution, not absolute amount of adiponectin, correlates with thiazolidinedione-mediated improvement in insulin sensitivity. J Biol. Chem. 279:12152–62.

    Article  CAS  PubMed  Google Scholar 

  38. Blüher M, et al. (2007) Total and high-molecular weight adiponectin in relation to metabolic variables at baseline and in response to an exercise treatment program: comparative evaluation of three assays. Diabetes Care 30:280–5.

    Article  CAS  PubMed  Google Scholar 

  39. Lara-Castro C, Luo N, Wallace P, Klein RL, Garvey WT. (2006) Adiponectin multimeric complexes and the metabolic syndrome trait cluster. Diabetes 55:249–59.

    Article  CAS  PubMed  Google Scholar 

  40. Hara K, Horikoshi M, Yamauchi T, et al. (2006) Measurement of the high molecular weight form of adiponectin in plasma is useful for the prediction of insulin resistance and metabolic syndrome. Diabetes Care 29:1357–62.

    Article  CAS  PubMed  Google Scholar 

  41. von Eynatten M, Lepper PM, Humpert PM. (2007) Total and high-molecular weight adiponectin in relation to metabolic variables at baseline and in response to an exercise treatment program: comparative evaluation of three assays: response to Bluher et al. Diabetes Care 30:e67.

    Article  Google Scholar 

  42. Combs TP, et al. (2002) Induction of adipocyte complement-related protein of 30 kilodaltons by PPARgamma agonists: a potential mechanism of insulin sensitization. Endocrinology 143:998–1007.

    Article  CAS  PubMed  Google Scholar 

  43. Yu JG, et al. (2002) The effect of thiazolidine-diones on plasma adiponectin levels in normal, obese, and type 2 diabetic subjects. Diabetes 51:2968–74.

    Article  CAS  PubMed  Google Scholar 

  44. Maeda N, et al. (2001) PPARgamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein. Diabetes 50:2094–9.

    Article  CAS  PubMed  Google Scholar 

  45. Yamauchi T, et al. (2003) Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 423:762–9.

    Article  CAS  PubMed  Google Scholar 

  46. Tsuchida A, et al. (2004) Insulin/Foxo1 pathway regulates expression levels of adiponectin receptors and adiponectin sensitivity. J Biol. Chem. 279:30817–22.

    Article  CAS  PubMed  Google Scholar 

  47. Yamauchi T, et al. (2007) Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat. Med. 13:332–9.

    Article  CAS  PubMed  Google Scholar 

  48. Hug C, et al. (2004) T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc. Natl. Acad. Sci. U. S. A. 101:10308–13.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Bjursell M, et al. (2007) Opposing effects of adiponectin receptors 1 and 2 on energy metabolism. Diabetes 56:583–93.

    Article  CAS  PubMed  Google Scholar 

  50. Liu Y, et al. (2007) Deficiency of adiponectin receptor 2 reduces diet-induced insulin resistance but promotes type 2 diabetes. Endocrinology 148:683–92.

    Article  CAS  PubMed  Google Scholar 

  51. Blüher M, et al. (2007) Gene expression of adiponectin receptors in human visceral and subcutaneous adipose tissue is related to insulin resistance and metabolic parameters and is altered in response to physical training. Diabetes Care 30:3110–5.

    Article  CAS  PubMed  Google Scholar 

  52. Rasmussen MS, et al. (2006) Adiponectin receptors in human adipose tissue: effects of obesity, weight loss, and fat depots. Obesity (Silver Spring) 14:28–35.

    Article  CAS  Google Scholar 

  53. Li W, et al. 2007) Insulin-sensitizing effects of thiazolidinediones are not linked to adiponectin receptor expression in human fat or muscle. Am. J. Physiol. Endocrinol. Metab. 292:E1301–7.

    Article  CAS  PubMed  Google Scholar 

  54. Nannipieri M, et al. (2007) Pattern of expression of adiponectin receptors in human adipose tissue depots and its relation to the metabolic state. Int. J. Obes. (Lond.) 31:1843–8.

    Article  CAS  Google Scholar 

  55. Zhang J, Holt H, Wang C, Hadid OH, Byrne CD. (2005) Expression of AdipoR1 in vivo in skeletal muscle is independently associated with measures of truncal obesity in middle-aged Caucasian men. Diabetes Care 28:2058–60.

    Article  CAS  PubMed  Google Scholar 

  56. Blüher M, et al. (2006) Circulating adiponectin and expression of adiponectin receptors in human skeletal muscle: associations with metabolic parameters and insulin resistance and regulation by physical training. J. Clin. Endocrinol. Metab. 91:2310–6.

    Article  CAS  PubMed  Google Scholar 

  57. Debard C, et al. (2004) Expression of key genes of fatty acid oxidation, including adiponectin receptors, in skeletal muscle of Type 2 diabetic patients. Diabetologia 47:917–25.

    Article  CAS  PubMed  Google Scholar 

  58. Civitarese AE, et al. (2004) Adiponectin receptors gene expression and insulin sensitivity in nondiabetic Mexican Americans with or without a family history of Type 2 diabetes. Diabetologia 47:816–20.

    Article  CAS  PubMed  Google Scholar 

  59. Staiger H, et al. (2004) Expression of adiponectin receptor mRNA in human skeletal muscle cells is related to in vivo parameters of glucose and lipid metabolism. Diabetes 53:2195–201.

    Article  CAS  PubMed  Google Scholar 

  60. Crimmins NA, Martin LJ. (2007) Polymorphisms in adiponectin receptor genes ADIPOR1 and ADIPOR2 and insulin resistance. Obes. Rev. 8:419–23.

    Article  CAS  PubMed  Google Scholar 

  61. Wittamer V, et al. (2003) Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J. Exp. Med. 198:977–985.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. Bozaoglu K, et al. (2007) Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology 148:4687–94.

    Article  CAS  PubMed  Google Scholar 

  63. Goralski KB, et al. (2007) Chemerin: A novel adipokine that regulates adipogenesis and adipocyte metabolism. J Biol. Chem. 282:28175–88.

    Article  CAS  PubMed  Google Scholar 

  64. Cash JL, et al. (2008) Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J. Exp. Med. 205:767–75.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  65. Roh S-G, et al. (2007) Chemerin—A new adipokine that modulates adipogenesis via its own receptor. Biochem. Biophys. Res. Commun. 362:1013–8.

    Article  CAS  PubMed  Google Scholar 

  66. Takahashia M, et al. (2008) Chemerin enhances insulin signaling and potentiates insulin-stimulated glucose uptake in 3T3-L1 adipocytes. FEBS Letters 582:573–8.

    Article  CAS  Google Scholar 

  67. Friedman JM, Halaas JL. (1998) Leptin and the regulation of body weight in mammals. Nature 395:763–770.

    Article  CAS  PubMed  Google Scholar 

  68. Elmquist JK, Elias CF, Saper CB. (1999) From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 22:221–232.

    Article  CAS  PubMed  Google Scholar 

  69. Bates SH, Myers MG Jr. (2003) The role of leptin receptor signaling in feeding and neuroendocrine function. Trends Endocrinol. Metab. 14:447–452.

    Article  CAS  PubMed  Google Scholar 

  70. Prodi E, Obici S. (2006) Minireview: the brain as a molecular target for diabetic therapy. Endocrinology 147:2664–9.

    Article  CAS  PubMed  Google Scholar 

  71. Coppari R, et al. (2005) The hypothalamic arcuate nucleus: a key site for mediating leptin’s effects on glucose homeostasis and locomotor activity. Cell Metab. 1:63–72.

    Article  CAS  PubMed  Google Scholar 

  72. Morton GJ, et al. (2005) Leptin regulates insulin sensitivity via phosphatidylinositol-3-OH kinase signaling in mediobasal hypothalamic neurons. Cell Metab. 2:411–420.

    Article  CAS  PubMed  Google Scholar 

  73. Bates SH, Kulkarni RN, Seifert M, Myers MG Jr. (2005) Roles for leptin receptor/STAT3-dependent and -independent signals in the regulation of glucose homeostasis. Cell Metab. 1:169–178.

    Article  CAS  PubMed  Google Scholar 

  74. Kahn BB, Alquier T, Carling D, Hardie DG. (2005) AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 1:15–25.

    Article  CAS  PubMed  Google Scholar 

  75. Minokoshi Y, et al. (2002) Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature 415:339–43.

    Article  CAS  PubMed  Google Scholar 

  76. Niswender KD, Magnuson MA. (2007) Obesity and the beta cell: lessons from leptin. J. Clin. Invest. 117:2753–6.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  77. Morioka T, et al. (2007) Disruption of leptin receptor expression in the pancreas directly affects beta cell growth and function in mice. J. Clin. Invest. 117:2860–8.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  78. Boghossian S, Dube MG, Torto R, Kalra PS, Kalra SP. (2006) Hypothalamic clamp on insulin release by leptin transgene expression. Peptides 27:3245–54.

    Article  CAS  PubMed  Google Scholar 

  79. Boghossian S, Lecklin AH, Torto R, Kalra PS, Kalra SP. (2005) Suppression of fat deposition for the life time of rodents with gene therapy. Peptides 26:1512–9.

    Article  CAS  PubMed  Google Scholar 

  80. Kieffer TJ, Habener JF. (2000) The adipoinsular axis: effects of leptin on pancreatic beta-cells. Am. J. Physiol. Endocrinol. Metab. 278:E1–E14.

    Article  CAS  PubMed  Google Scholar 

  81. Munzberg H, Myers MG Jr. (2005) Molecular and anatomical determinants of central leptin resistance. Nat. Neurosci. 8:566–570.

    Article  CAS  PubMed  Google Scholar 

  82. Farooqi IS, et al. (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J. Clin. Invest. 110:1093–103.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  83. Oral EA, et al. (2002) Leptin-replacement therapy for lipodystrophy. N. Engl. J. Med. 346:570–8.

    Article  CAS  PubMed  Google Scholar 

  84. Hukshorn CJ, et al. (2000) Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J. Clin. Endocrinol. Metab. 85:4003–9.

    Article  CAS  PubMed  Google Scholar 

  85. El-Haschimi K, Pierroz DD, Hileman SM, Bjørbaek C, Flier JS. (2000) Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J. Clin. Invest. 105:1827–32.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  86. Unger RH. (2002) Lipotoxic diseases. Annu. Rev. Med. 53:319–36.

    Article  CAS  PubMed  Google Scholar 

  87. Kalra SP. (2008) Central leptin insufficiency syndrome: an interactive etiology for obesity, metabolic and neural diseases and for designing new therapeutic interventions. Peptides 29:127–38.

    Article  CAS  PubMed  Google Scholar 

  88. Yang RZ, et al. (2006) Identification of omentinas a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am. J. Physiol. Endocrinol. Metab. 290:E1253–61.

    Article  CAS  PubMed  Google Scholar 

  89. de Souza Batista CM, et al. (2007) Omentin plasma levels and gene expression are decreased in obesity. Diabetes 56:1655–61.

    Article  CAS  PubMed  Google Scholar 

  90. Tan BK, et al. (2008) Omentin-1, a novel adipokine, is decreased in overweight insulinresistant women with polycystic ovary syndrome. Ex vivo and in vivo regulation of omentin-1 by insulin and glucose. Diabetes 57:801–8.

    Article  CAS  PubMed  Google Scholar 

  91. Steppan CM, et al. (2001) The hormone resistin links obesity to diabetes. Nature 409:307–12.

    Article  CAS  PubMed  Google Scholar 

  92. Rajala MW, et al. (2004) Regulation of resistin expression and circulating levels in obesity, diabetes, and fasting. Diabetes 53:1671–9.

    Article  CAS  PubMed  Google Scholar 

  93. Rajala MW, Obici S, Scherer PE, Rossetti L. (2003) Adipose-derived resistin and gut-derived resistin-like molecule-beta selectively impair insulin action on glucose production. J. Clin. Invest. 111:225–30.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  94. Satoh H, et al. (2004) Adenovirus-mediated chronic “hyper-resistinemia” leads to in vivo insulin resistance in normal rats. J. Clin. Invest. 114:224–31.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  95. Pravenec M, et al. (2003) Transgenic and recombinant resistin impair skeletal muscle glucose metabolism in the spontaneously hypertensive rat. J. Biol. Chem. 278:45209–15.

    Article  CAS  PubMed  Google Scholar 

  96. Rangwala SM, et al. (2004) Abnormal glucose homeostasis due to chronic hyperresistinemia. Diabetes 53:1937–41.

    Article  CAS  PubMed  Google Scholar 

  97. Banerjee RR, et al. (2004) Regulation of fasted blood glucose by resistin. Science 303:1195–8.

    Article  CAS  PubMed  Google Scholar 

  98. Muse ED, et al. (2004) Role of resistin in diet-induced hepatic insulin resistance. J. Clin. Invest. 114:232–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  99. Kim KH, Zhao L, Moon Y, Kang C, Sul HS. (2004) Dominant inhibitory adipocyte-specific secretory factor (ADSF)/resistin enhances adipogenesis and improves insulin sensitivity. Proc. Natl. Acad. Sci. U. S. A. 101:6780–5.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  100. Savage DB, et al. (2001) Resistin/Fizz3 expression in relation to obesity and peroxisome proliferator-activated receptor-gamma action in humans. Diabetes 50:2199–2202.

    Article  CAS  PubMed  Google Scholar 

  101. Degawa-Yamauchi M, et al. (2003) Serum resistin (FIZZ3) protein is increased in obese humans. J. Clin. Endocrinol. Metab. 88:5452–5.

    Article  CAS  PubMed  Google Scholar 

  102. Heilbronn LK, et al. (2004) Relationship between serum resistin concentrations and insulin resistance in nonobese, obese, and obese diabetic subjects. J. Clin. Endocrinol. Metab. 89:1844–8.

    Article  CAS  PubMed  Google Scholar 

  103. Youn BS, et al. (2004) Plasma resistin concentrations measured by enzyme-linked immunosorbent assay using a newly developed monoclonal antibody are elevated in individuals with type 2 diabetes mellitus. J. Clin. Endocrinol. Metab. 89:150–6.

    Article  CAS  PubMed  Google Scholar 

  104. Osawa H, et al. (2004) The G/G genotype of a resistin singlenucleotide polymorphism at −420 increases type 2 diabetes mellitus susceptibility by inducing promoter activity through specific binding of Sp1/3. Am. J. Hum. Genet. 75:678–86.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  105. Osawa H, et al. (2007) Plasma resistin, associated with single nucleotide plymorphism −402, is correlated with insulin resistance, lower HDL cholesterol, and high sensitivity C-reactive protein in the Japanese general population. Diabetes Care 30:1501–6.

    Article  CAS  PubMed  Google Scholar 

  106. Ochi M, et al. (2007) Frequency of the G/G genotype of resistin single nucleotide polymorphism at −420 appears to be increased in younger-onset type 2 diabetes. Diabetes 56:2834–8.

    Article  CAS  PubMed  Google Scholar 

  107. Gerber M, et al. (2005) Serum resistin levels of obese and lean children and adolescents: biochemical analysis and clinical relevance. J. Clin. Endocrinol. Metab. 90:4503–9.

    Article  CAS  PubMed  Google Scholar 

  108. Chen CC, et al. (2005) Serum resistin level among healthy subjects: Relationship to anthro-pometric and metabolic parameters. Metabolism 54:471–5.

    Article  CAS  PubMed  Google Scholar 

  109. Pfutzner A, Langenfeld M, Kunt T, Lobig M, Forst T. (2003) Evaluation of human resistin assays with serum from patients with type 2 diabetes and different degrees of insulin resistance. Clin. Lab. 49:571–6.

    PubMed  Google Scholar 

  110. Lee JH, et al. (2003) Circulating resistin levels are not associated with obesity or insulin resistance in humans and are not regulated by fasting or leptin administration: cross-sectional and interventional studies in normal, insulin-resistant, and diabetic subjects. J. Clin. Endocrinol. Metab. 88:4848–56.

    Article  CAS  PubMed  Google Scholar 

  111. Kielstein JT, et al. (2003) Increased resistin blood levels are not associated with insulin resistance in patients with renal disease. Am. J. Kidney Dis. 42:62–6.

    Article  CAS  PubMed  Google Scholar 

  112. Pagano C, et al. (2006) Increased serum resistin in nonalcoholic fatty liver disease is related to liver disease severity and not to insulin resistance. J. Clin. Endocrinol. Metab. 91:1081–6.

    Article  CAS  PubMed  Google Scholar 

  113. Perseghin G, et al. (2006) Increased serum resistin in elite endurance athletes with high insulin sensitivity. Diabetologia 49:1893–900.

    Article  CAS  PubMed  Google Scholar 

  114. Beckers S, et al. (2008) Analysis of genetic variations in the resistin gene shows no associations with obesity in women. Obesity 16:905–7.

    Article  CAS  PubMed  Google Scholar 

  115. Kaser S, et al. (2003) Resistin messenger-RNA expression is increased by proinflammatory cytokines in vitro. Biochem. Biophys. Res. Commun. 309:286–90.

    Article  CAS  PubMed  Google Scholar 

  116. Nagaev I, Bokarewa M, Tarkowski A, Smith U. (2006) Human resistin is a systemic immune-derived proinflammatory cytokine targeting both leukocytes and adipocytes. PLoS ONE 1:e31.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  117. Bokarewa M, Nagaev I, Dahlberg L, Smith U, Tarkowski A. (2005) Resistin, an adipokine with potent proinflammatory properties. J. Immunol. 174:5789–95.

    Article  CAS  PubMed  Google Scholar 

  118. Silswal N, et al. (2005) Human resistin stimulates the pro-inflammatory cytokines TNF-α and IL-12 in macrophages by NF-κB-dependent pathway. Biochem. Biophys. Res. Commun. 334:1092–101.

    Article  CAS  PubMed  Google Scholar 

  119. Shetty GK, Economides PA, Horton ES, Mantzoros CS, Veves A. (2004) Circulating adiponectin and resistin levels in relation to metabolic factors, inflammatory markers, and vascular reactivity in diabetic patients and subjects at risk for diabetes. Diabetes Care 27:2450–7.

    Article  CAS  PubMed  Google Scholar 

  120. Reilly MP, etal. (2005) Resistin is an inflammatory marker of atherosclerosis in humans. Circulation 111:932–9.

    Article  CAS  PubMed  Google Scholar 

  121. Konrad A, et al. (2007) Resistin is an inflammatory marker of inflammatory bowel disease in humans. Eur J. Gastroenterol. Hepatol. 19:1070–4.

    Article  CAS  PubMed  Google Scholar 

  122. Sunden-Cullberg J, et al. (2007) Pronounced elevation of resistin correlates with severity of disease in severe sepsis and septic shock. Crit. Care Med. 35:1536–42.

    Article  CAS  PubMed  Google Scholar 

  123. Lehrke M, et al. (2004) An inflammatory cascade leading to hyperresistinemia in humans. PLoS Med. 1:e45.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  124. Yang Q, et al. (2005) Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 436:356–62.

    Article  CAS  PubMed  Google Scholar 

  125. Graham TE, et al. (2006) Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N. Engl. J. Med. 354:2552–63.

    Article  CAS  PubMed  Google Scholar 

  126. Haider DG, et al. (2007) Serum retinol-binding protein 4 is reduced after weight loss in morbidly obese subjects. J. Clin. Endocrinol. Metab. 92:1168–71.

    Article  CAS  PubMed  Google Scholar 

  127. Jia W, et al. (2007) Association of serum retinol-binding protein 4 and visceral adiposity in Chinese subjects with and without type 2 diabetes. J. Clin. Endocrinol. Metab. 92:3224–9.

    Article  CAS  PubMed  Google Scholar 

  128. Klöting N, et al. (2007) 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. 6:79–87.

    Article  CAS  PubMed  Google Scholar 

  129. Aeberli I, et al. (2007) Serum retinol-binding protein 4 concentration and its ratio to serum retinol are associated with obesity and metabolic syndrome components in children. J. Clin. Endocrinol. Metab. 92:4359–65.

    Article  CAS  PubMed  Google Scholar 

  130. Möhlig M, et al. (2008) Retinol-binding protein 4 is associated with insulin resistance, but appears unsuited for metabolic screening in women with polycystic ovary syndrome. Eur. J. Endocrinol. 158:517–23.

    Article  CAS  PubMed  Google Scholar 

  131. Gavi S, et al. (2007) Retinol-binding protein 4 is associated with insulin resistance and body fat distribution in nonobese subjects without type 2 diabetes. J. Clin. Endocrinol. Metab. 92:1886–90.

    Article  CAS  PubMed  Google Scholar 

  132. Perseghin G, et al. (2007) Serum retinol-binding protein-4, leptin, and adiponectin concentrations are related to ectopic fat accumulation. J. Clin. Endocrinol. Metab. 92:4883–8.

    Article  CAS  PubMed  Google Scholar 

  133. Stefan N, et al. (2007) High circulating retinol-binding protein 4 is associated with elevated liver fat but not with total, subcutaneous, visceral, or intramyocellular fat in humans. Diabetes Care 30:1173–8.

    Article  CAS  PubMed  Google Scholar 

  134. Takebayashi K, Suetsugu M, Wakabayashi S, Aso Y, Inukai T. (2007) Retinol binding protein-4 levels and clinical features of type 2 diabetes patients. J. Clin. Endocrinol. Metab. 92:2712–9.

    Article  CAS  PubMed  Google Scholar 

  135. Cho YM, et al. (2006) Plasma retinol-binding protein-4 concentrations are elevated in human subjects with impaired glucose tolerance and type 2 diabetes. Diabetes Care 29:2457–61.

    Article  CAS  PubMed  Google Scholar 

  136. Qi Q, et al. (2007) Elevated retinol-binding protein 4 levels are associated with metabolic syndrome in Chinese people. J. Clin. Endocrinol. Metab. 92:4827–34.

    Article  CAS  PubMed  Google Scholar 

  137. Lim S, et al. (2008) Insulin-sensitizing effects of exercise on adiponectin and retinol binding protein-4 concentrations in young and middle-aged women. J. Clin. Endocrinol. Metab. 93:2263–8.

    Article  CAS  PubMed  Google Scholar 

  138. Balagopal P, et al. (2007) Reduction of elevated serum retinol binding protein in obese children by lifestyle intervention: association with subclinical inflammation. J. Clin. Endocrinol. Metab. 92:1971–4.

    Article  CAS  PubMed  Google Scholar 

  139. Munkhtulga L, et al. (2007) Identification of a regulatory SNP in the retinol binding protein 4 gene associated with type 2 diabetes in Mongolia. Hum. Genet. 120:879–88.

    Article  CAS  PubMed  Google Scholar 

  140. Craig RL, Chu WS, Elbein SC. (2007) Retinol binding protein 4 as a candidate gene for type 2 diabetes and prediabetic intermediate traits. Mol. Genet. Metab. 90:338–44.

    Article  CAS  PubMed  Google Scholar 

  141. Janke J, et al. (2006) Retinol-binding protein 4 in human obesity. Diabetes 55:2805–10.

    Article  CAS  PubMed  Google Scholar 

  142. Yao-Borengasser A, et al. (2007) Retinol binding protein 4 expression in humans: relationship to insulin resistance, inflammation, and response to pioglitazone. J. Clin. Endocrinol. Metab. 92:2590–7.

    Article  CAS  PubMed  Google Scholar 

  143. Broch M, Vendrell J, Ricart W, Richart C, Fernandez-Real JM. (2007) Circulating retinol-binding protein-4, insulin sensitivity, insulin secretion, and insulin disposition index in obese and nonobese subjects. Diabetes Care 30:1802–6.

    Article  CAS  PubMed  Google Scholar 

  144. von Eynatten M, et al. (2007) Retinol-binding protein 4 is associated with components of the metabolic syndrome, but not with insulin resistance, in men with type 2 diabetes or coronary artery disease. Diabetologia 50:1930–7.

    Article  CAS  Google Scholar 

  145. Silha JV, Nyomba BL, Leslie WD, Murphy LJ. (2007) Ethnicity, insulin resistance, and inflammatory adipokines in women at high and low risk for vascular disease. Diabetes Care 30:286–91.

    Article  CAS  PubMed  Google Scholar 

  146. Graham TE, Wason CJ, Blüher M, Kahn BB. (2007) Shortcomings in methodology complicate measurements of serum retinol binding protein (RBP4) in insulin-resistant human subjects. Diabetologia 50:814–23.

    Article  CAS  PubMed  Google Scholar 

  147. von Eynatten M, Humpert PM. (2008) Retinol-binding protein-4 in experimental and clinical metabolic disease. Expert Rev. Mol. Diagn. 8:289–99

    Article  Google Scholar 

  148. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. (1997) Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature 389:610–4.

    Article  CAS  PubMed  Google Scholar 

  149. Jellema A, Plat J, Mensink RP. (2004) Weight reduction, but not a moderate intake of fish oil, lowers concentrations of inflammatory markers and PAI-1 antigen in obese men during the fasting and postprandial state. Eur. J. Clin. Invest. 34:766–73.

    Article  CAS  PubMed  Google Scholar 

  150. Hotamisligil GS, Shargill NS, Spiegelman BM. (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1:87–91.

    Article  Google Scholar 

  151. Bernstein LE, Berry J, Kim S, Canavan B, Grinspoon SK. (2006) Effects of etanercept in patients with the metabolic syndrome. Arch. Intern. Med. 166:902–8.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  152. Dominguez H, et al. (2005) Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J. Vasc. Res. 42:517–25.

    Article  CAS  PubMed  Google Scholar 

  153. Mooney RA. (2007) Counterpoint: interleukin-6 does not have a beneficial role in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102:816–8.

    Article  CAS  PubMed  Google Scholar 

  154. Pedersen BK, Febbraio MA. (2007) Point: interleukin-6 does have a beneficial role in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102:814–6.

    Article  CAS  PubMed  Google Scholar 

  155. Klover PJ, Zimmers TA, Koniaris LG, Mooney RA. (2003) Chronic exposure to interleukin-6 causes hepatic insulin resistance in mice. Diabetes 52:2784–9.

    Article  CAS  PubMed  Google Scholar 

  156. Senn JJ, Klover PJ, Nowak IA, Mooney RA. (2002) Interleukin-6 induces cellular insulin resistance in hepatocytes. Diabetes 51:3391–9.

    Article  CAS  PubMed  Google Scholar 

  157. Rotter V, Nagaev I, Smith U. (2003) 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. 278:45777–84.

    Article  CAS  PubMed  Google Scholar 

  158. Carey AL, et al. (2006) Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes 55:2688–97.

    Article  CAS  PubMed  Google Scholar 

  159. Al Khalili L, et al. (2006) Signaling specificity of interleukin-6 action on glucose and lipid metabolism in skeletal muscle. Mol. Endocrinol. 20:3364–75.

    Article  CAS  PubMed  Google Scholar 

  160. Kim HJ, et al. (2004). Differential effects of interleukin-6 and -10 on skeletal muscle and liver insulin action in vivo. Diabetes 53:1060–7.

    Article  CAS  PubMed  Google Scholar 

  161. Inoue H, et al. (2006) Role of hepatic STAT3 in brain-insulin action on hepatic glucose production. Cell Metab. 3:267–75.

    Article  CAS  PubMed  Google Scholar 

  162. Pickup JC, Mattock MB, Chusney GD, Burt D. (1997) NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Diabetologia 40:1286–92.

    Article  CAS  PubMed  Google Scholar 

  163. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. (2001) Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am. J. Physiol. Endocrinol. Metab. 280:E745–51.

    Article  CAS  PubMed  Google Scholar 

  164. Tilg H, Hotamisligil, GS. (2006) Nonalcoholic fatty liver disease: Cytokine-adipokine interplay and regulation of insulin resistance. Gastroenterology 131:934–45.

    Article  CAS  PubMed  Google Scholar 

  165. Hida K, et al. (2005) Visceral adipose tissue-derived serine protease inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc. Natl. Acad. Sci. U. S. A. 102:10610–5.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  166. Klöting N, et al. (2006) Vaspin gene expression in human adipose tissue: association with obesity and type 2 diabetes. Biochem. Biophys. Res. Commun. 339:430–6.

    Article  CAS  PubMed  Google Scholar 

  167. Youn BS, et al. (2008) Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes 57:372–7.

    Article  CAS  PubMed  Google Scholar 

  168. Samal B, et al. (1994) Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol. Cell. Biol. 14:1431–7.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  169. Rongvaux A, et al. (2002) Pre-B-cell colony-enhancing factor, whose expression is up-regulated in activated lymphocytes, is a nicotinamide phosphoribosyltransferase, a cytosolic enzyme involved in NAD biosynthesis. Eur. J. Immunol. 32:3225–34.

    Article  CAS  PubMed  Google Scholar 

  170. Fukuhara A, et al. (2005) Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science 307:426–30.

    Article  CAS  PubMed  Google Scholar 

  171. Chen MP, et al. (2006) Elevated plasma level of visfatin/pre-B cell colony-enhancing factor in patients with type 2 diabetes mellitus. J. Clin. Endocrinol. Metab. 91:295–9.

    Article  CAS  PubMed  Google Scholar 

  172. Haider DG, et al. (2006) Increased plasma vis-fatin concentrations in morbidly obese subjects are reduced after gastric banding. J. Clin. Endocrinol. Metab. 91:1578–81.

    Article  CAS  PubMed  Google Scholar 

  173. Filippatos TD, et al. (2008) Increased plasma visfatin levels in subjects with the metabolic syndrome. Eur. J. Clin. Invest. 38:71–2.

    Article  CAS  PubMed  Google Scholar 

  174. Bailey SD, et al. (2006) Common polymorphisms in the promoter of the visfatin gene (PBEF1) influence plasma insulin levels in a French-Canadian population. Diabetes 55:2896–902.

    Article  CAS  PubMed  Google Scholar 

  175. Zhang YY, et al. (2006) A visfatin promoter polymorphism is associated with low-grade inflammation and type 2 diabetes. Obesity (Silver Spring) 14:2119–26.

    Article  CAS  Google Scholar 

  176. Berndt J, et al. (2005) Plasma visfatin concentrations and fat depot-specific mRNA expression in humans. Diabetes 54:2911–6.

    Article  CAS  PubMed  Google Scholar 

  177. Klöting N, et al. (2006) Vaspin gene expression in human adipose tissue: association with obesity and type 2 diabetes. Biochem. Biophys. Res. Commun. 339:430–6.

    Article  CAS  PubMed  Google Scholar 

  178. Pagano C, et al. (2006) Reduced plasma visfatin/pre-B cell colony-enhancing factor in obesity is not related to insulin resistance in humans. J. Clin. Endocrinol. Metab. 91:3165–70.

    Article  CAS  PubMed  Google Scholar 

  179. Oki K, Yamane K, Kamei N, Nojima H, Kohno N. (2007) Circulating visfatin level is correlated with inflammation, but not with insulin resistance. Clin. Endocrinol. (Oxf). 67:796–800.

    Article  CAS  PubMed  Google Scholar 

  180. Revollo JR, et al. (2007) Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab. 6:363–75.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  181. Sell H, Eckel J. (2007) Regulation of retinol binding protein 4 production in primary human adipocytes by adiponectin, troglitazone and TNF-alpha. Diabetologia 50:2221–3.

    Article  CAS  PubMed  Google Scholar 

  182. Faggioni R, Feingold KR, Grunfeld C. (2001) Leptin regulation of the immune response and the immunodeficiency of malnutrition. FASEB J. 15:2565–71.

    Article  CAS  PubMed  Google Scholar 

  183. Simons PJ, van den Pangaart PS, van Roomen CP, Aerts JM, Boon L. (2005) Cytokine-mediated modulation of leptin and adiponectin secretion during in vitro adipogenesis: evidence that tumor necrosis factor-alpha- and interleukin-1beta-treated human preadipocytes are potent leptin producers. Cytokine 32:94–103.

    Article  CAS  PubMed  Google Scholar 

  184. Hector J, et al. (2007) TNF-alpha alters visfatin and adiponectin levels in human fat. Horm. Metab. Res. 39:250–5.

    Article  CAS  PubMed  Google Scholar 

  185. Lago F, Dieguez C, Gómez-Reino J, Gualillo O. (2007) Adipokines as emerging mediators of immune response and inflammation. Nat Clin Pract Rheumatol. 3:716–24

    Article  CAS  PubMed  Google Scholar 

  186. Moschen AR, et al. (2007) Visfatin, an adipocytokine with proinflammatory and immunomod-ulating properties. J. Immunol. 178:1748–58.

    Article  CAS  PubMed  Google Scholar 

  187. Asensio C, Cettour-Rose P, Theander-Carrillo C, Rohner-Jeanrenaud F, Muzzin P. (2004) Changes of glycemia by leptin administration or high fat feeding in rodent models of obesity/type 2 diabetes suggest a link between resistin expression and control of glucose homeostasis. Endocrinology 145:2206–13

    Article  CAS  PubMed  Google Scholar 

  188. Zhang W, Della-Fera MA, Hartzell DL, Hausman D, Baile CA. (2008) Adipose tissue gene expression profiles in ob/ob mice treated with leptin. Life Sci. 83:35–42.

    Article  CAS  PubMed  Google Scholar 

  189. Delporte ML, El Mkadem SA, Quisquater M, Brichard SM. (2004) Leptin treatment markedly increased plasma adiponectin but barely decreased plasma resistin of ob/ob mice. Am. J. Physiol. Endocrinol Metab. 287:E446–53.

    Article  CAS  PubMed  Google Scholar 

  190. Obesity and overweight. c2008. Geneva: World Health Organization; [cited 2008 Oct 15]. Available from: https://doi.org/www.who.int/dietphysicalactivity/publications/facts/obesity/en.

Download references

Acknowledgments

This work was supported by a grant from the Deutsche Forschungsgemeinschaft (BR 2151/4-1 to UCB).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Uli C. Broedl.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Rabe, K., Lehrke, M., Parhofer, K.G. et al. Adipokines and Insulin Resistance. Mol Med 14, 741–751 (2008). https://doi.org/10.2119/2008-00058.Rabe

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/2008-00058.Rabe

Keywords