Skip to content
Publicly Available Published by De Gruyter April 10, 2014

Roles of oxidative stress, adiponectin, and nuclear hormone receptors in obesity-associated insulin resistance and cardiovascular risk

  • Morihiro Matsuda EMAIL logo and Iichiro Shimomura

Abstract

Obesity leads to the development of type 2 diabetes mellitus, which is a strong risk factor for cardiovascular disease. A better understanding of the molecular basis of obesity will lead to the establishment of effective prevention strategies for cardiovascular diseases. Adipocytes have been shown to generate a variety of endocrine factors termed adipokines/adipocytokines. Obesity-associated changes to these adipocytokines contribute to the development of cardiovascular diseases. Adiponectin, which is one of the most well-characterized adipocytokines, is produced exclusively by adipocytes and exerts insulin-sensitizing and anti-atherogenic effects. Obese subjects have lower levels of circulating adiponectin, and this is recognized as one of the factors involved in obesity-induced insulin resistance and atherosclerosis. Another pathophysiological feature of obesity may involve the low-grade chronic inflammation in adipose tissue. This inflammatory process increases oxidative stress in adipose tissue, which may affect remote organs, leading to the development of diabetes, hypertension, and atherosclerosis. Nuclear hormone receptors (NRs) regulate the transcription of the target genes in response to binding with their ligands, which include metabolic and nutritional substrates. Among the various NRs, peroxisome proliferator-activated receptor γ promotes the transcription of adiponectin and antioxidative enzymes, whereas mineralocorticoid receptor mediates the effects of aldosterone and glucocorticoid to induce oxidative stress in adipocytes. It is hypothesized that both play crucial roles in the pathophysiology of obesity-associated insulin resistance and cardiovascular diseases. Thus, reduced adiponectin and increased oxidative stress play pathological roles in obesity-associated insulin resistance to increase the cardiovascular disease risk, and various NRs may be involved in this pathogenesis.

Introduction

Over the past few decades, obesity has been a growing threat to the health of people in an increasing number of countries [1]. Obesity, especially visceral fat obesity, causes insulin resistance and leads to the development of type 2 diabetes mellitus (T2DM), which is a strong risk factor for cardiovascular disease and cancers that are associated with a high mortality rate [2–4]. Furthermore, visceral fat obesity leads to the risk-clustering status known as metabolic syndrome, which is characterized by high plasma triglycerides (TG), low plasma high-density lipoprotein (HDL) cholesterol, high fasting plasma glucose, and high blood pressure, which is also a risk factor for cardiovascular events [5, 6]. A better understanding of the molecular basis of obesity will lead to the development of effective prevention strategies for obesity-associated cardiovascular diseases.

A series of studies have revealed that adipocytes generate and secrete a variety of endocrine factors known as adipokines/adipocytokines and that obesity-associated changes in adipocytokines contribute to the development of cardiovascular diseases [7]. Adiponectin, one of the most well-characterized adipocytokines, is produced exclusively by adipocytes and exerts insulin-sensitizing and anti-atherogenic effects [8–10]. Obese subjects have lower levels of circulating adiponectin [11, 12], which is recognized as a molecular factor contributing to obesity-induced insulin resistance and atherosclerosis.

Another important contributing factor for obesity-associated insulin resistance may involve low-grade chronic inflammation in adipose tissue [13, 14]. This inflammatory process includes an increase in pro-inflammatory adipocytokines [15–17] and an increase in oxidative stress [18]. In obese humans and rodents, the levels of oxidative stress-associated markers have been found to be elevated in plasma and urine [18]. In obese mice, oxidative stress was especially increased in adipose tissue [18]. This oxidative stress may remotely affect the oxidative stress levels in β cells [19, 20], vascular endothelium [21, 22], and the brain [23, 24], leading to the development of diabetes, hypertension, and atherosclerosis.

Nuclear hormone receptors (NRs) regulate the transcription of target genes in response to binding to their specific ligands, such as steroid hormones, fatty acids, oxysterol, and bile acids. NRs are expressed in tissues involved in lipid, carbohydrate, and energy homeostasis, translating hormonal, metabolic, and nutritional signals into alterations in gene expression [25]. In particular, peroxisome proliferator-activated receptor (PPAR) γ is an essential NR in adipocytes, playing an important role in the differentiation of mature adipocytes, and can promote the transcription of adiponectin and antioxidative enzymes [26–31]. Recently, mineralocorticoid receptor (MR) has been demonstrated to mediate the effects of aldosterone and glucocorticoid to induce oxidative stress in adipocytes [32–34]. Thus, many studies have shown that NRs play various roles in the pathophysiology of obesity-associated insulin resistance and cardiovascular diseases.

In this review, we describe the roles of reduced adiponectin and increased oxidative stress in obesity-associated insulin resistance as well as cardiovascular risk and the contribution of various NRs to the pathophysiology of obesity.

The roles of oxidative stress in obesity

Obesity-associated increase in oxidative stress in adipose tissue

The Framingham study revealed that urinary levels of 8-epi-prostaglandin F2α (8-epi-PGF2α), a systemic oxidative stress marker, were significantly associated with body mass index [35]. We have shown that urinary 8-epi-PGF2α levels are associated more closely with the visceral fat area than with the subcutaneous fat area measured by abdominal computed tomography [36]. In obese mice, the oxidative stress levels in plasma were elevated in comparison to those in control mice [18]. Moreover, lipid peroxide levels and hydrogen peroxide generation were elevated in adipose tissue, but not in the liver, skeletal muscle, or aorta [18]. These data suggest that the adipose tissue in obese individuals may represent a major source of reactive oxygen species (ROS). The question is why oxidative stress increases in adipose tissue with obesity. Several possible explanations have been proposed.

Increased expression of nicotinamide adenine dinucleotide phosphate oxidase subunits

In adipose tissue, increased expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a plasma membrane enzyme that converts molecular oxygen to superoxide radicals, may be associated with increased oxidative stress levels. In obese mice, messenger ribonucleic acid (mRNA) expression of the NADPH oxidase subunits was increased in adipose tissue but not in the liver or muscles [18]. Moreover, treatment of obese mice with the NADPH oxidase inhibitor apocynin improved hyperglycemia, hyperinsulinemia, hypertriglyceridemia, and hepatic steatosis [18]. These results suggest that ROS generation via NADPH oxidase could play an important role in the pathogenesis of obesity-associated metabolic disorder.

Decreased expression or activity of antioxidative enzymes

Adipose tissue expresses relatively high levels of antioxidant defensive enzymes. However, the expression and activity of antioxidant enzymes such as catalase, superoxide dismutase (SOD) 1, and glutathione peroxidase (GPx) were reduced in the adipose tissues of obese individuals [18, 28, 29, 37]. ROS dose-dependently suppress the expression of PPARγ [18], an important transcriptional factor that induces the expression of catalase [28]. Interestingly, these changes in antioxidant levels were observed in adipose tissue but not in the liver or skeletal muscle. In addition to increased NADPH oxidase levels, decreased antioxidant levels may contribute to increased oxidative stress in the adipose tissue [18].

Involvement of adipose tissue macrophages

Macrophage infiltration into adipose tissue has been considered an important factor in the pathogenesis of insulin resistance in obese individuals [13, 14]. Macrophages are known to produce ROS; thus, adipose tissue macrophages could be involved in increased ROS generation. ROS have been shown to increase monocyte chemotactic protein 1 (MCP-1) expression in adipocytes [18]. Furthermore, the by-products of ROS-associated lipid peroxidation are potent chemoattractants [38]. ROS also augment the mRNA expression of NADPH oxidase subunits in adipocytes [18]; increased ROS generation could thus lead to increased macrophage infiltration and inflammatory changes. Therefore, in obesity, oxidative stress may contribute to the establishment of a vicious cycle that promotes increased inflammation in adipose tissues.

Overnutrition and ROS generation in mitochondria

Glucose is oxidized during the tricarboxylic acid cycle, which generates electron donors such as nicotinamide adenine dinucleotide dehydrogenase (NADH) and flavin adenine dinucleotide (FADH2). Excess glucose leads to the overproduction of electron donors in the mitochondrial electron transport chain, resulting in the generation of superoxide radicals [39]. Similarly, excess free fatty acids (FFAs) lead to increased FFA oxidation by mitochondria, which in turn generate excess NADH and FADH2, leading to the mitochondrial overproduction of ROS [39]. Furthermore, ROS generation is augmented in FFA-loaded adipocytes but can be blocked by treatment with an NADPH oxidase inhibitor, indicating the involvement of NADPH oxidase in ROS generation by fatty acids [18]. FFAs, especially palmitate, can stimulate diacylglycerol synthesis and activate protein kinase C (PKC), which leads to the activation of NADPH oxidase [40]. Thus, excess glucose and FFAs cause oxidative stress in mitochondria and the plasma membrane.

Taken together, various mechanisms, including increased NADPH oxidase expression and decreased antioxidant activities, may be involved in the increased ROS generation by adipose tissue in obesity.

Increased oxidative stress in obesity and insulin resistance

Many studies have reported an association between ROS and insulin resistance [37, 41–45]. In 3T3-L1 adipocytes, insulin resistance can be induced by treatment with either tumor necrosis factor (TNF-α) or glucocorticoid, via increased ROS generation, whereas treatment with an antioxidative agent, either SOD or catalase, can improve insulin resistance [41]. Oxidative stress has also been reported to induce insulin resistance in myocytes [45]. This ROS-induced insulin resistance can be attributed to the activation of stress signals such as c-Jun N-terminal kinase, p38 mitogen-activated protein kinase, nuclear factor κB (NF-κB), and certain isoforms of PKC [39, 41–43]. Meanwhile, hydrogen peroxide is produced transiently in response to insulin in a NADPH oxidase-mediated manner and acts as a second messenger to augment insulin signals in adipocytes [44]. These data indicate that, in adipocytes, transient increases in cellular ROS may play an important role in insulin signaling, but excessive and prolonged exposure to ROS suppresses insulin action. Glutathione (GSH) has been shown to overaccumulate in hypertrophied adipose tissues [37]. Although GSH is an antioxidant, excess GSH suppresses insulin action in adipocytes [37]. Meanwhile, insulin suppresses GPX activity, which leads to the accumulation of GSH in adipocytes [37], indicating a complex interaction between insulin and ROS in adipocytes. In β-cells and isolated islets, oxidative stress suppresses insulin production [19, 46]. This impaired insulin production can be improved with antioxidant treatment in obese mice [20]. Increased oxidative stress impairs insulin production as well as insulin action.

Increased oxidative stress in obesity and cardiovascular disease

Oxidative stress is strongly involved in the development of atherosclerosis [47]. Excess ROS attenuates nitric oxide bioavailability, and superoxide easily reacts with nitric oxide, leading to the generation of harmful peroxynitrite and, subsequently, to endothelial dysfunction [22]. Increased ROS facilitates the oxidation of low-density lipoprotein (LDL) in atherosclerotic lesions [48], thus facilitating immune reactions in endothelial cells, including the increased expression of adhesion molecules, which results in macrophage migration, and the formation of lipid-laden macrophages [47]. These processes aggravate vascular endothelial damage.

Several mechanisms that increase oxidative stress locally in the vascular wall have been postulated as atherosclerosis pathogeneses. Increased NADPH oxidase expression has been observed in pre-atherosclerotic vascular endothelium [49, 50], whereas angiotensin II acts through the angiotensin type I receptor to trigger a powerful stimulus for ROS generation from NADPH oxidase [21, 51].

Several molecules may be involved in the modulation of hyperglycemia-induced oxidative stress, including those in the polyol pathway, advanced glycation end products (AGEs), and PKC [52–55]. The polyol pathway is facilitated by hyperglycemic states. Aldose reductase, a polyol pathway enzyme, utilizes and depletes NADPH to convert excess glucose to sorbitol. In particular, AGE-receptor interactions activate NADPH oxidase, leading to ROS generation [56]. Mitochondrial ROS (described in a previous section) are also involved in cellular AGE accumulation, activation of the polyol pathway, and PKC [54]. These oxidative stresses are aggravated in obesity and diabetes.

The prolonged presence of increased TG-rich lipoproteins in the circulation induces oxidative stress in the endothelium [57, 58]. Our recent clinical study suggested that in high-risk DM patients treated with statins, the circulating levels of malondialdehyde-modified LDL (MDA-LDL), a surrogate marker of oxidized LDL, were significantly correlated with the TG and HDL cholesterol levels [59]. Adiponectin levels were also significantly correlated with MDA-LDL levels, although not independently of TG and HDL cholesterol [59]. The serum MDA-LDL level was significantly associated with serum remnant lipoprotein cholesterol levels [59]. Activation of lectin-like oxidized LDL receptor 1 by remnant lipoprotein particles induced NADPH oxidase-dependent production of superoxide in endothelial cells [60], which may explain the significant association between LDL oxidation and remnant lipoproteins rich in TG. Moreover, HDL protects against the oxidation of LDL [61]. Collectively, the management of dyslipidemic metabolic syndrome components is important for reducing the oxidization of LDL, and ultimately, to the development of atherosclerosis (Figure 1).

Figure 1 Schematic diagram of the effects of adiponectin and oxidative stress on circulating lipoproteins and atherosclerosis under healthy conditions and in metabolic syndrome.In metabolic syndrome, the reduction of adiponectin leads to the increase of TG-rich remnant lipoproteins via decreased insulin sensitivity, which contributes to generation of ROS, resulting in the oxidation of LDL that accelerates the development of atherosclerosis. Moreover, the anti-atherosclerosis force is ameliorated directly by the reduction of adiponectin and indirectly through the decrease of HDL that contributes to reverse cholesterol transport and suppresses the oxidation of LDL.
Figure 1

Schematic diagram of the effects of adiponectin and oxidative stress on circulating lipoproteins and atherosclerosis under healthy conditions and in metabolic syndrome.

In metabolic syndrome, the reduction of adiponectin leads to the increase of TG-rich remnant lipoproteins via decreased insulin sensitivity, which contributes to generation of ROS, resulting in the oxidation of LDL that accelerates the development of atherosclerosis. Moreover, the anti-atherosclerosis force is ameliorated directly by the reduction of adiponectin and indirectly through the decrease of HDL that contributes to reverse cholesterol transport and suppresses the oxidation of LDL.

Roles of adiponectin in obesity-associated insulin resistance and cardiovascular risk

Clinical implications of adiponectin in obesity-associated diseases

Circulating levels of adiponectin are low in patients with visceral fat obesity [11] or T2DM [12], and the levels are correlated with the indices of insulin sensitivity [62, 63]. People with high levels of circulating adiponectin are less likely than those with low concentrations to develop T2DM [64]. Circulating adiponectin levels are also decreased with hypertension in humans, irrespective of insulin resistance [65]. Furthermore, adiponectin concentrations correlate positively with HDL cholesterol concentrations and negatively with TG concentrations [66]. Patients with missense mutations in the adiponectin gene present with low adiponectin concentrations and have been reported to exhibit T2DM and metabolic syndrome phenotypes [67]. Thus, low circulating adiponectin levels are associated with various coronary risk factors. A case-control study demonstrated that low adiponectin levels are associated with a high risk of coronary artery disease [68], whereas a clinical prospective study demonstrated that men with high circulating adiponectin levels had a significantly lower prevalence of myocardial infarction than those with low adiponectin levels [69]. Recent clinical studies have demonstrated that low levels of circulating adiponectin are significantly associated with lipid-rich plaques in coronary arteries, as assessed by intravascular ultrasound [70]. Low adiponectin levels were significantly associated with multivessel coronary atherosclerosis assessed on computed tomography angiography independently of conventional risk factors in patients with suspected coronary artery disease and were predictive of multivessel coronary atherosclerosis in combination with age, sex, hypertension, and diabetes [71]. Taken together with the basic researches described in detail in the following sections, these clinical studies indicate that circulating adiponectin protein should directly protect coronary artery walls from pro-atherogenic stresses induced by conventional risk factors as well as enhance insulin sensitivity, and hence, the vascular walls with reduced adiponectin protein should be susceptible to pro-atherogenic stresses, facilitating the development of coronary atherosclerosis.

Insulin-sensitizing effects of adiponectin

The insulin-sensitizing or anti-diabetes effects of adiponectin have been demonstrated in vivo in animal studies. Adiponectin-deficient mice showed marked elevations in plasma glucose and insulin levels, as well as insulin resistance, relative to wild-type mice, when fed a high-fat and high-sucrose diet, although they did not present this phenotype on a normal diet [9]. Adiponectin supplementation via transfection with an adiponectin-generating adenovirus reduced the development of insulin resistance in adiponectin-deficient mice that consumed a high-fat and high-sucrose diet [9].

A study by Kadowaki and his colleagues has elucidated the precise molecular mechanism involved in the adiponectin-mediated modulation of insulin sensitivity. Adiponectin exerts insulin-sensitizing effects via adenosine monophosphate-activated protein kinase (AMPK) activation and facilitates fatty acid oxidization via PPARα activation [72]. These effects are mediated by the membrane receptor proteins AdipoR1 and AdipoR2, which specifically bind to adiponectin [73]. Analysis of mice deficient in both AdipoR1 and AdipoR2 revealed that these proteins play essential roles in mediating the effects of adiponectin with regard to insulin sensitization and the suppression of inflammation or oxidative stress [74].

Anti-inflammatory and anti-atherogenic effects of adiponectin

Anti-atherosclerotic effects have been demonstrated in vivo in animal studies of adiponectin-deficient mice. When compared with wild-type mice, adiponectin-deficient mice developed more severe intimal thickening, with more active smooth muscle cell proliferation after receiving an experimental vascular injury [10]. Treatment with adiponectin-producing adenovirus suppressed this intimal thickening [10]. Adiponectin overexpression attenuated plaque formation in apolipoprotein E-deficient mice [75, 76]. The various biological properties of adiponectin that suppress pro-inflammatory or pro-atherosclerotic processes have been elucidated. Adiponectin can suppress the expression of adhesion molecules, such as intracellular adhesion molecule 1, by inhibiting the TNF-α-mediated activation of NF-κB in endothelial cells, leading to the suppression of monocyte adhesion, which is an initial step in atherosclerosis [77]. Adiponectin predominantly inhibits the proliferation of myelomonocytic lineage cells and suppresses mature macrophage functions, including phagocytic activity and lipopolysaccharide-induced TNF-α production [78]. In macrophages, adiponectin can suppress foam cell transformation by inhibiting scavenger receptor class A expression [79]. Adiponectin overexpression significantly reduces the vascular wall expression of scavenger receptor class A, TNF-α, and intracellular adhesion molecule 1 in apolipoprotein E-deficient mice and suppresses atherosclerosis [75, 76]. It also suppresses growth factor-induced vascular smooth muscle cell proliferation by inhibiting mitogen-activated protein kinase [80]. Furthermore, adiponectin increases the expression of a tissue inhibitor of metalloproteinase in macrophages, which contributes to coronary artery plaque stabilization by inhibiting matrix metalloproteinases [81]. The adiponectin protein exists in the aortic endothelium under steady state conditions and may protect vasculature from the initiation of atherosclerosis [82]. Adiponectin increases HDL assembly by enhancing the ATP-binding cassette transporter A1 (ABCA1) pathway and apolipoprotein A-1 synthesis in the liver [83, 84], leading to an enhancement of reverse cholesterol transport. Collectively, these properties contribute to the anti-atherosclerotic and anti-inflammatory functions of adiponectin.

The oxidative stress and adiponectin antagonism

Adiponectin suppresses the harmful effects of oxidative stress

Besides its suppressive effects on atherosclerosis, adiponectin inhibits pressure overload-induced myocardial hypertrophy, decreases angiotensin II-induced cardiac fibrosis, and protects the heart from ischemia-reperfusion injury [85–87]. In a myocardial infarction/reperfusion model, adiponectin played a protective role against oxidative stress-induced myocardial damage. It is possible that adiponectin decreases oxidative/nitrative stress by inhibiting inducible nitric oxide synthase and suppressing the expression of gp91phox, a NADPH oxidase subunit [88], in an AMPK-independent manner [89]. Similar effects of adiponectin have also been observed in the endothelium. Adiponectin can suppress oxidative/nitrative stress in the arteries of hyperlipidemic rats [90]. In addition, adiponectin exerts cardioprotective effects against the oxidative stress-induced remodeling processes in cardiomyocytes by activating AMPK and inhibiting extracellular signal-regulated kinases and NF-κB [91]. Interestingly, this cardioprotective effect of adiponectin was reduced in mice in which diabetes was induced through a high fat diet [92], which may be associated with reduced AdipoR1 and AdipoR2 expression in response to insufficient insulin activity [93].

Oxidative stress suppresses adiponectin production

In humans, serum adiponectin levels are inversely correlated with systemic oxidative stress [18, 36]. In adipose cells, ROS exposure suppressed adiponectin mRNA expression and secretion and increased the mRNA expression of pro-inflammatory adipocytokines such as interleukin (IL) 6 and MCP-1 [18]. The antioxidant N-acetylcysteine reversed the effects of ROS and restored gene expression to the basal level [18]. Furthermore, in an in vivo study, treatment of obese mice with apocynin, an NADPH oxidase inhibitor, led to increased adiponectin expression and decreased TNF-α expression, which were accompanied by the suppression of oxidative stress in the adipose tissue [18]. These data suggest that oxidative stress plays a role in reducing adiponectin levels, which in turn contributes to obesity-associated disease pathogenesis. Conversely, oxidative stress was enhanced in AdipoR1- and AdipoR2-deficient mice [74], which provides evidence that the adiponectin-AdipoR pathway contributes to the suppression of oxidative stress.

Roles of NRs in obesity-associated insulin resistance and cardiovascular risk

Peroxisome proliferator-activated receptors

PPARγ is a master regulator that plays a key role in the control of adipocyte-specific gene expression in combination with CCAAT/enhancer binding protein α during adipose differentiation [26]. PPARγ2 is exclusively expressed by adipocytes and plays essential roles in regulating various important genes involved in adipose differentiation as well as in glucose and lipid metabolism in adipocytes. Thiazolidinediones (TZDs), synthetic PPARγ ligands, can improve insulin resistance [94]. TZDs can increase circulating adiponectin levels by activating adiponectin gene transcription through a PPARγ-responsive element in the gene promoter region [27]. Kubota et al. [95] demonstrated that TZDs exert insulin-sensitizing effects in ob/ob mice, mainly by activating AMPK and suppressing gluconeogenesis in the liver. This effect is mediated partly by an adiponectin-dependent pathway.

PPARγ plays an important role in the transcriptional activation of antioxidant enzymes such as SOD1, catalase, and GPX3 [28–31]. The expression of these antioxidant enzymes is decreased in the adipocytes of obese animals [18, 31, 37], whereas TZDs increase the expression, which may contribute to the suppression of oxidative stress [28, 29, 31]. TZDs have been shown to suppress TNF-α-induced oxidative stress [57]. Therefore, the insulin-sensitizing effects of TZDs might be explained partly by their ability to inhibit oxidative stress. Interestingly, PPARγ-mediated regulation of catalase is functionally conserved between mice and humans, although the locations of the PPARγ-responsive sites in the promoter regions are different [28, 29]. Clinically, a randomized controlled trial revealed that pioglitazone, a TZD, reduces the incidence of all-cause mortality, nonfatal myocardial infarction, and stroke in patients with type 2 diabetes, who have a high risk of macrovascular events [96]. PPARγ could prove to be a therapeutic target for insulin resistance to reduce cardiovascular risk, especially in obese subjects.

Although other PPARs are not key regulators of adipogenesis, they can control lipid metabolism. Activation of PPARα results in a reduction of plasma TG levels, through the induction of genes that decrease the availability of TG for hepatic very-low-density lipoprotein secretion, and the induction of genes that promote lipoprotein lipase-mediated lipolysis of TG-rich plasma lipoproteins [97]. Fibrates, synthetic ligands of PPARα, can increase the production of adiponectin via PPARα in adipocytes [98]. Several clinical trials have suggested that fibrates may be effective for the prevention of cardiovascular events in patients with high TG and low HDL-C levels [99, 100]. PPARδ induces the expression of genes required for fatty acid oxidation and energy dissipation in skeletal muscle and contributes to the development of oxidative muscle fiber [101, 102]. Activation or overexpression of PPARδ in mice results in resistance to weight gain and improved insulin sensitivity in high fat diet-induced obesity, as well as genetically predisposed obesity, via enhanced oxidation [103].

Mineralocorticoid receptor

Recent studies have resulted in a better understanding of MR physiology in the heart, vasculature, brain, and adipose tissues [104]. Activation of MR by aldosterone promotes ROS generation through NADPH oxidase in the heart and vasculature [105]. Recent studies have revealed that MR was also involved in oxidative stress in adipose tissue [32–34]. Adipose expression of MR increases in obese mice [33]. Treatment of obese mice with eplerenone, an inhibitory agent of MR, can improve insulin resistance through the suppression of macrophage infiltration, a decrease in inflammatory adipocytokines, and an increase in serum adiponectin levels [32]. Similarly, in 3T3-L1 adipocytes, treatment with aldosterone suppresses the expression of adiponectin, which is blocked by eplerenone [33]. Moreover, aldosterone increases oxidative stress in 3T3-L1 adipocytes, which is blocked by treatment with eplerenone or small interfering RNA of MR, indicating that the effect of aldosterone is mediated by MR [33].

MR can bind not only to aldosterone but also to glucocorticoid, with 10-fold higher affinity than the glucocorticoid receptor (GR) [106]. Glucocorticoids are a potent regulator of adipose differentiation, which is mediated by MR [107]. A selective MR blockade can inhibit adipose differentiation and TG accumulation in 3T3-L1 and 3T3-F442A cells [108]. 11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1), an enzyme that converts cortisone to active cortisol, is expressed in adipose tissue [109]. 11β-HSD1 mRNA in adipose tissue is expressed more highly in obese subjects, suggesting that active cortisol has a role in the pathophysiology of obesity [110, 111]. Fat-specific overexpression of 11β-HSD1 shows a phenotype presenting abdominal obesity, hypertension, and insulin resistance [112]. In 3T3-L1 adipocytes, treatment with glucocorticoid increases the expression of NADPH oxidase subunits, leading to an increase in oxidative stress [34], and decreases the expression of adiponectin and catalase (Figure 2), which is blocked by treatment with eplerenone, suggesting that these effects of glucocorticoid are mediated by MR in adipocytes [34]. The expression and activity of 11β-HSD1 in adipocytes is negatively regulated by PPARγ [113]. Therefore, the reduction of PPARγ leads to an increase in 11β-HSD1 activity, resulting in greater generation of cortisol, leading to increased oxidative stress through MR, which in turn causes a further reduction in PPARγ (Figure 2). This vicious cycle mediated by inappropriate MR activation (by both aldosterone and glucocorticoids) should be associated with the development of insulin resistance and atherosclerosis [114] (Figure 2), which can be blocked by treatment with TZDs or eplerenone.

Figure 2 Working model illustrating the contribution of nuclear receptors to increased oxidative stress and decreased adiponectin expression in adipocytes in obesity.Inappropriate MR activation by both aldosterone and cortisol leads to increased oxidative stress via the activation of NADPH oxidase, which ameliorates PPARγ, which plays an essential role in the expression of adiponectin. The reduction of adiponectin is causally associated with the development of insulin resistance and atherosclerosis. Moreover, the inactivation of PPARγ causes the reduction of catalase, leading to further increase of oxidative stress and increase of 11β-HSD1, which facilitates the conversion of inactive cortisone to active cortisol, which further activates MR activity.
Figure 2

Working model illustrating the contribution of nuclear receptors to increased oxidative stress and decreased adiponectin expression in adipocytes in obesity.

Inappropriate MR activation by both aldosterone and cortisol leads to increased oxidative stress via the activation of NADPH oxidase, which ameliorates PPARγ, which plays an essential role in the expression of adiponectin. The reduction of adiponectin is causally associated with the development of insulin resistance and atherosclerosis. Moreover, the inactivation of PPARγ causes the reduction of catalase, leading to further increase of oxidative stress and increase of 11β-HSD1, which facilitates the conversion of inactive cortisone to active cortisol, which further activates MR activity.

Meanwhile, selective GR stimulation inhibits the expression of pro-inflammatory adipocytokines [115]. A recent research have revealed that cardiomyocyte-specific GR-deficient mice die prematurely from spontaneous cardiovascular disease or display a marked reduction in left ventricular systolic function by 3 months of age [116]. Thus, glucocorticoids via activating GR play a crucial role in protecting target tissues from stresses, whereas they induce oxidative stress via activating MR.

Liver X receptors

There are two liver X receptors (LXRs), termed LXRα and LXRβ. LXRα is expressed in liver, intestine, macrophages, and adipose tissue, whereas LXRβ is ubiquitously expressed [117]. LXR regulates the expression of the ABCA1 [118, 119], which is one of the important transporters for reverse cholesterol transport, the process of cholesterol delivery from the periphery to the liver. A synthetic LXR agonist has been demonstrated to inhibit the development of atherosclerosis in mice [120]. Meanwhile, LXR stimulates lipogenesis through the induction of sterol-regulatory element-binding protein 1c (SREBP1c), a transcriptional factor activating various genes involved in lipogenesis [121]. In addition, LXR activation leads to hypertriglyceridemia via the expression of angiopoietin-like protein 3, suppressing lipoprotein lipase activity [122]. Thus, LXR agonists have a beneficial effect of inhibiting atherosclerosis, but they also have a harmful effect of promoting hepatic steatosis and hypertriglyceridemia.

Both LXRs are highly expressed in white and brown adipose tissues. Although LXRs do not contribute to adipocyte differentiation, they regulate the genes involved in adipocyte growth and glucose homeostasis [123, 124]. LXR activation promotes the expression of glucose transporter 4 (GLUT4) and increases glucose uptake in adipose tissue [125, 126]. Meanwhile, LXR increases fatty acid β-oxidation and decreases glucose oxidation in white adipose tissue [127]. LXR-deficient mice are resistant to diet-induced obesity, which may be mediated by increased energy expenditure in brown adipose tissue [128, 129]. Collectively, these data suggest that LXRs play some role in governing glucose and lipid metabolism in adipose tissue.

Other nuclear hormone receptors

Farnesoid X receptor (FXR), liver receptor homologue 1 (LRH-1), and a small heterodimer partner (SHP) are NRs that have been shown to regulate various genes involved in bile acid metabolism [130–132]. In mice fed a high-fat diet, FXR deletion had a protective effect, reducing weight gain, hyperglycemia, hyperinsulinemia, and glucose intolerance, despite higher plasma TG levels [133]. Meanwhile, FXR activation reduced atherosclerotic lesion formation in LDL receptor null or apolipoprotein E null mice [134]. LRH-1 augments PPARγ-induced transactivation of the adiponectin gene [27]. Treatment with an LRH-1 ligand leads to decreased hepatic TGs and serum glucose [135]. Mutations of the SHP gene are associated with a mildly obese phenotype in Japanese subjects [136]. Meanwhile, SHP knockout mice are resistant to diet-induced obesity [137, 138]. Although the precise mechanism has not yet been fully clarified, SHP has been reported to modulate the activity of PPARγ [139] and PPARα [137]. Other NRs such as chicken ovalbumin upstream promoter transcription factor II [140, 141], RAR-related receptor α (RORα) [142], estrogen-related receptor α (ERRα) [143], ERRγ [144], and REV-ERBα [145] have been shown to be involved in adipogenesis. In the future, the identification of new ligands for NRs may facilitate the development of new therapeutic approaches for the treatment of patients with obesity and insulin resistance to reduce cardiovascular risk, although further studies are required.

Expert opinion

Measuring circulating adiponectin levels and oxidative stress levels is beneficial because they give an indication of obesity-associated cardiovascular risk and are predictive of the occurrence of severe cardiovascular disease, in combination with conventional risk factors. TZDs and eplerenone increase adiponectin levels and decrease oxidative stress levels via the activation of PPARγ and suppression of MR, respectively. Other NRs in adipocytes may be good drug targets for obesity-associated insulin resistance and cardiovascular diseases.

Outlook

The identification of new ligands for NRs will facilitate the development of new therapeutic approaches for the treatment of patients with obesity and insulin resistance to reduce cardiovascular risk.

Highlights

  • Adiponectin is an adipocyte-derived factor that has insulin-sensitizing and anti-atherogenic functions. In obese subjects, plasma adiponectin levels are reduced, which facilitates the development of diabetes and atherosclerosis.

  • In obese subjects, oxidative stress levels are elevated in adipose tissue, which affects the remote organs, contributing to the development of obesity-associated diseases, such as diabetes, hypertension, and atherosclerosis.

  • Adiponectin and oxidative stress are co-associated. Oxidative stress suppresses the production of adiponectin in adipocytes. Adiponectin suppresses oxidative stress-induced damage in the heart and the vascular wall.

  • PPARγ is an essential NR for adipocyte differentiation and regulates the transcription of various genes involved in glucose and lipid metabolism in adipocytes. Oxidative stress suppresses the expression of PPARγ, leading to amelioration of adipocyte function.

  • TZDs, synthetic ligands of PPARγ, can increase the expression of adiponectin and antioxidative enzymes, such as catalase, SOD1, and GPX3.

  • MR is an NR expressed in adipocytes that mediates the effect of aldosterone and glucocorticoid to increase oxidative stress.

  • Treatment of obese mice with eplerenone, an MR antagonist, can decrease oxidative stress and increase adiponectin expression in adipose tissue.

  • There are a number of NRs in adipocytes that may be involved in obesity-associated diseases. Activation or repression of these NRs will be targets for the development of new therapeutic approaches to obesity-associated insulin resistance and atherosclerosis.

Conflict of interest statement: The authors declare no conflict of interest.


Corresponding author: Morihiro Matsuda, Institute for Clinical Research, Division of Preventive Medicine, National Hospital Organization, Kure Medical Center, and Chugoku Cancer Center, 3-1 Aoyama-cho, Kure, Hiroshima 737-0023, Japan, Phone: +81 823 22 3111, Fax: +81 823 21 0478, E-mail:

References

1. Obesity: preventing and managing the global epidemic. Report of a WHO consultation. World Health Organ Tech Rep Ser 2000;894:i–xii, 1–253.Search in Google Scholar

2. Matsuzawa Y, Nakamura T, Shimomura I, Kotani K. Visceral fat accumulation and cardiovascular disease. Obes Res 1995;3:645S–7S.10.1002/j.1550-8528.1995.tb00481.xSearch in Google Scholar PubMed

3. Matsuzawa Y, Shimomura I, Nakamura T, Keno Y, Kotani K, Tokunaga K. Pathophysiology and pathogenesis of visceral fat obesity. Obes Res 1995;3:187S–94S.10.1002/j.1550-8528.1995.tb00462.xSearch in Google Scholar PubMed

4. Vucenik I, Stains JP. Obesity and cancer risk: evidence, mechanisms, and recommendations. Ann NY Acad Sci 2012;1271:37–43.10.1111/j.1749-6632.2012.06750.xSearch in Google Scholar PubMed PubMed Central

5. Isomaa B, Almgren P, Tuomi T, Forsen B, Lahti K, Nissen M, Taskinen MR, Groop L. Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care 2001;24:683–9.10.2337/diacare.24.4.683Search in Google Scholar PubMed

6. Matsuzawa Y, Funahashi T, Nakamura T. The concept of metabolic syndrome: contribution of visceral fat accumulation and its molecular mechanism. J Atheroscler Thromb 2011;18:629–39.10.5551/jat.7922Search in Google Scholar PubMed

7. Matsuzawa Y, Funahashi T, Nakamura T. Molecular mechanism of metabolic syndrome X: contribution of adipocytokines adipocyte-derived bioactive substances. Ann NY Acad Sci 1999;892:146–54.10.1111/j.1749-6632.1999.tb07793.xSearch in Google Scholar PubMed

8. Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Matsubara K. cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (AdiPose Most abundant Gene transcript 1). Biochem Biophys Res Commun 1996;221:286–9.10.1006/bbrc.1996.0587Search in Google Scholar PubMed

9. Maeda N, Shimomura I, Kishida K, Nishizawa H, Matsuda M, Nagaretani H, Furuyama N, Kondo H, Takahashi M, Arita Y, Komuro R, Ouchi N, Kihara S, Tochino Y, Okutomi K, Horie M, Takeda S, Aoyama T, Funahashi T, Matsuzawa Y. Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat Med 2002;8:731–7.10.1038/nm724Search in Google Scholar PubMed

10. Matsuda M, Shimomura I, Sata M, Arita Y, Nishida M, Maeda N, Kumada M, Okamoto Y, Nagaretani H, Nishizawa H, Kishida K, Komuro R, Ouchi N, Kihara S, Nagai R, Funahashi T, Matsuzawa Y. Role of adiponectin in preventing vascular stenosis. The missing link of adipo-vascular axis. J Biol Chem 2002;277:37487–91.10.1074/jbc.M206083200Search in Google Scholar PubMed

11. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 1999;257:79–83.10.1006/bbrc.1999.0255Search in Google Scholar PubMed

12. Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y, Iwahashi H, Kuriyama H, Ouchi N, Maeda K, Nishida M, Kihara S, Sakai N, Nakajima T, Hasegawa K, Muraguchi M, Ohmoto Y, Nakamura T, Yamashita S, Hanafusa T, Matsuzawa Y. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 2000;20:1595–9.10.1161/01.ATV.20.6.1595Search in Google Scholar PubMed

13. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 2003;112:1796–808.10.1172/JCI200319246Search in Google Scholar

14. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 2003;112:1821–30.10.1172/JCI200319451Search in Google Scholar

15. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993;259:87–91.10.1126/science.7678183Search in Google Scholar PubMed

16. Fried SK, Bunkin DA, Greenberg AS. Omental and subcutaneous adipose tissues of obese subjects release interleukin-6: depot difference and regulation by glucocorticoid. J Clin Endocrinol Metab 1998;83:847–50.10.1210/jc.83.3.847Search in Google Scholar

17. Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci USA 2003;100:7265–70.10.1073/pnas.1133870100Search in Google Scholar PubMed PubMed Central

18. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest 2004;114:1752–61.10.1172/JCI21625Search in Google Scholar PubMed PubMed Central

19. Matsuoka T, Kajimoto Y, Watada H, Kaneto H, Kishimoto M, Umayahara Y, Fujitani Y, Kamada T, Kawamori R, Yamasaki Y. Glycation-dependent, reactive oxygen species-mediated suppression of the insulin gene promoter activity in HIT cells. J Clin Invest 1997;99:144–50.10.1172/JCI119126Search in Google Scholar PubMed PubMed Central

20. Kaneto H, Kajimoto Y, Miyagawa J, Matsuoka T, Fujitani Y, Umayahara Y, Hanafusa T, Matsuzawa Y, Yamasaki Y, Hori M. Beneficial effects of antioxidants in diabetes: possible protection of pancreatic beta-cells against glucose toxicity. Diabetes 1999;48:2398–406.10.2337/diabetes.48.12.2398Search in Google Scholar PubMed

21. Rajagopalan S, Kurz S, Munzel T, Tarpey M, Freeman BA, Griendling KK, Harrison DG. Angiotensin II-mediated hypertension in the rat increases vascular superoxide production via membrane NADH/NADPH oxidase activation. Contribution to alterations of vasomotor tone. J Clin Invest 1996;97:1916–23.10.1172/JCI118623Search in Google Scholar PubMed PubMed Central

22. Heitzer T, Schlinzig T, Krohn K, Meinertz T, Munzel T. Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease. Circulation 2001;104:2673–8.10.1161/hc4601.099485Search in Google Scholar PubMed

23. Zhang X, Dong F, Ren J, Driscoll MJ, Culver B. High dietary fat induces NADPH oxidase-associated oxidative stress and inflammation in rat cerebral cortex. Exp Neurol 2005;191: 318–25.10.1016/j.expneurol.2004.10.011Search in Google Scholar PubMed

24. Nagae A, Fujita M, Kawarazaki H, Matsui H, Ando K, Fujita T. Sympathoexcitation by oxidative stress in the brain mediates arterial pressure elevation in obesity-induced hypertension. Circulation 2009;119:978–86.10.1161/CIRCULATIONAHA.108.824730Search in Google Scholar PubMed

25. Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 2006;126:789–99.10.1016/j.cell.2006.06.049Search in Google Scholar PubMed PubMed Central

26. Issemann I, Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 1990;347:645–50.10.1038/347645a0Search in Google Scholar PubMed

27. Iwaki M, Matsuda M, Maeda N, Funahashi T, Matsuzawa Y, Makishima M, Shimomura I. Induction of adiponectin, a fat-derived antidiabetic and antiatherogenic factor, by nuclear receptors. Diabetes 2003;52:1655–63.10.2337/diabetes.52.7.1655Search in Google Scholar PubMed

28. Okuno Y, Matsuda M, Kobayashi H, Morita K, Suzuki E, Fukuhara A, Komuro R, Shimabukuro M, Shimomura I. Adipose expression of catalase is regulated via a novel remote PPARgamma-responsive region. Biochem Biophys Res Commun 2008;366:698–704.10.1016/j.bbrc.2007.12.001Search in Google Scholar PubMed

29. Okuno Y, Matsuda M, Miyata Y, Fukuhara A, Komuro R, Shimabukuro M, Shimomura I. Human catalase gene is regulated by peroxisome proliferator activated receptor-gamma through a response element distinct from that of mouse. Endocr J 2010;57:303–9.10.1507/endocrj.K09E-113Search in Google Scholar PubMed

30. Heo KS, Kim DU, Ryoo S, Nam M, Baek ST, Kim L, Park SK, Myung CS, Hoe KL. PPARgamma activation abolishes LDL-induced proliferation of human aortic smooth muscle cells via SOD-mediated down-regulation of superoxide. Biochem Biophys Res Commun 2007;359:1017–23.10.1016/j.bbrc.2007.06.006Search in Google Scholar PubMed

31. Lee YS, Kim AY, Choi JW, Kim M, Yasue S, Son HJ, Masuzaki H, Park KS, Kim JB. Dysregulation of adipose glutathione peroxidase 3 in obesity contributes to local and systemic oxidative stress. Mol Endocrinol 2008;22:2176–89.10.1210/me.2008-0023Search in Google Scholar PubMed PubMed Central

32. Guo C, Ricchiuti V, Lian BQ, Yao TM, Coutinho P, Romero JR, Li J, Williams GH, Adler GK. Mineralocorticoid receptor blockade reverses obesity-related changes in expression of adiponectin, peroxisome proliferator-activated receptor-gamma, and proinflammatory adipokines. Circulation 2008;117:2253–61.10.1161/CIRCULATIONAHA.107.748640Search in Google Scholar PubMed PubMed Central

33. Hirata A, Maeda N, Hiuge A, Hibuse T, Fujita K, Okada T, Kihara S, Funahashi T, Shimomura I. Blockade of mineralocorticoid receptor reverses adipocyte dysfunction and insulin resistance in obese mice. Cardiovasc Res 2009;84:164–72.10.1093/cvr/cvp191Search in Google Scholar PubMed

34. Hirata A, Maeda N, Nakatsuji H, Hiuge-Shimizu A, Okada T, Funahashi T, Shimomura I. Contribution of glucocorticoid-mineralocorticoid receptor pathway on the obesity-related adipocyte dysfunction. Biochem Biophys Res Commun 2012;419:182–7.10.1016/j.bbrc.2012.01.139Search in Google Scholar PubMed

35. Keaney JF Jr., Larson MG, Vasan RS, Wilson PW, Lipinska I, Corey D, Massaro JM, Sutherland P, Vita JA, Benjamin EJ. Obesity and systemic oxidative stress: clinical correlates of oxidative stress in the Framingham Study. Arterioscler Thromb Vasc Biol 2003;23:434–9.10.1161/01.ATV.0000058402.34138.11Search in Google Scholar PubMed

36. Fujita K, Nishizawa H, Funahashi T, Shimomura I, Shimabukuro M. Systemic oxidative stress is associated with visceral fat accumulation and the metabolic syndrome. Circ J 2006;70:1437–42.10.1253/circj.70.1437Search in Google Scholar PubMed

37. Kobayashi H, Matsuda M, Fukuhara A, Komuro R, Shimomura I. Dysregulated glutathione metabolism links to impaired insulin action in adipocytes. Am J Physiol Endocrinol Metab 2009;296:E1326–34.10.1152/ajpendo.90921.2008Search in Google Scholar PubMed

38. Curzio M, Esterbauer H, Poli G, Biasi F, Cecchini G, Di Mauro C, Cappello N, Dianzani MU. Possible role of aldehydic lipid peroxidation products as chemoattractants. Int J Tissue React 1987;9:295–306.Search in Google Scholar

39. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes 2005;54:1615–25.10.2337/diabetes.54.6.1615Search in Google Scholar PubMed

40. Inoguchi T, Li P, Umeda F, Yu HY, Kakimoto M, Imamura M, Aoki T, Etoh T, Hashimoto T, Naruse M, Sano H, Utsumi H, Nawata H. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C-dependent activation of NAD(P)H oxidase in cultured vascular cells. Diabetes 2000;49:1939–45.10.2337/diabetes.49.11.1939Search in Google Scholar PubMed

41. Houstis N, Rosen ED, Lander ES. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 2006;440:944–8.10.1038/nature04634Search in Google Scholar PubMed

42. Rudich A, Tirosh A, Potashnik R, Hemi R, Kanety H, Bashan N. Prolonged oxidative stress impairs insulin-induced GLUT4 translocation in 3T3-L1 adipocytes. Diabetes 1998;47:1562–9.10.2337/diabetes.47.10.1562Search in Google Scholar PubMed

43. Tirosh A, Potashnik R, Bashan N, Rudich A. Oxidative stress disrupts insulin-induced cellular redistribution of insulin receptor substrate-1 and phosphatidylinositol 3-kinase in 3T3-L1 adipocytes. A putative cellular mechanism for impaired protein kinase B activation and GLUT4 translocation. J Biol Chem 1999;274:10595–602.10.1074/jbc.274.15.10595Search in Google Scholar PubMed

44. Krieger-Brauer HI, Kather H. Human fat cells possess a plasma membrane-bound H2O2-generating system that is activated by insulin via a mechanism bypassing the receptor kinase. J Clin Invest 1992;89:1006–13.10.1172/JCI115641Search in Google Scholar PubMed PubMed Central

45. Maddux BA, See W, Lawrence JC Jr., Goldfine AL, Goldfine ID, Evans JL. Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by mircomolar concentrations of alpha-lipoic acid. Diabetes 2001;50:404–10.10.2337/diabetes.50.2.404Search in Google Scholar PubMed

46. Matsuoka TA, Zhao L, Artner I, Jarrett HW, Friedman D, Means A, Stein R. Members of the large Maf transcription family regulate insulin gene transcription in islet beta cells. Mol Cell Biol 2003;23:6049–62.10.1128/MCB.23.17.6049-6062.2003Search in Google Scholar PubMed PubMed Central

47. Stocker R, Keaney JF Jr. Role of oxidative modifications in atherosclerosis. Physiol Rev 2004;84:1381–478.10.1152/physrev.00047.2003Search in Google Scholar PubMed

48. Azumi H, Inoue N, Ohashi Y, Terashima M, Mori T, Fujita H, Awano K, Kobayashi K, Maeda K, Hata K, Shinke T, Kobayashi S, Hirata K, Kawashima S, Itabe H, Hayashi Y, Imajoh-Ohmi S, Itoh H, Yokoyama M. Superoxide generation in directional coronary atherectomy specimens of patients with angina pectoris: important role of NAD(P)H oxidase. Arterioscler Thromb Vasc Biol 2002;22:1838–44.10.1161/01.ATV.0000037101.40667.62Search in Google Scholar

49. Guzik TJ, Sadowski J, Kapelak B, Jopek A, Rudzinski P, Pillai R, Korbut R, Channon KM. Systemic regulation of vascular NAD(P)H oxidase activity and nox isoform expression in human arteries and veins. Arterioscler Thromb Vasc Biol 2004;24:1614–20.10.1161/01.ATV.0000139011.94634.9dSearch in Google Scholar PubMed

50. Spiekermann S, Landmesser U, Dikalov S, Bredt M, Gamez G, Tatge H, Reepschlager N, Hornig B, Drexler H, Harrison DG. Electron spin resonance characterization of vascular xanthine and NAD(P)H oxidase activity in patients with coronary artery disease: relation to endothelium-dependent vasodilation. Circulation 2003;107:1383–9.10.1161/01.CIR.0000056762.69302.46Search in Google Scholar PubMed

51. Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res 2000;86: 494–501.10.1161/01.RES.86.5.494Search in Google Scholar

52. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001;414:813–20.10.1038/414813aSearch in Google Scholar PubMed

53. Jay D, Hitomi H, Griendling KK. Oxidative stress and diabetic cardiovascular complications. Free Radic Biol Med 2006;40:183–92.10.1016/j.freeradbiomed.2005.06.018Search in Google Scholar

54. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I, Brownlee M. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000;404:787–90.10.1038/35008121Search in Google Scholar

55. Sasaki S, Inoguchi T. The role of oxidative stress in the pathogenesis of diabetic vascular complications. Diabetes Metab J 2012;36:255–61.10.4093/dmj.2012.36.4.255Search in Google Scholar

56. Basta G, Schmidt AM, De Caterina R. Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes. Cardiovasc Res 2004;63:582–92.10.1016/j.cardiores.2004.05.001Search in Google Scholar

57. Ceriello A, Taboga C, Tonutti L, Quagliaro L, Piconi L, Bais B, Da Ros R, Motz E. Evidence for an independent and cumulative effect of postprandial hypertriglyceridemia and hyperglycemia on endothelial dysfunction and oxidative stress generation: effects of short- and long-term simvastatin treatment. Circulation 2002;106:1211–8.10.1161/01.CIR.0000027569.76671.A8Search in Google Scholar

58. Anderson RA, Evans ML, Ellis GR, Graham J, Morris K, Jackson SK, Lewis MJ, Rees A, Frenneaux MP. The relationships between post-prandial lipaemia, endothelial function and oxidative stress in healthy individuals and patients with type 2 diabetes. Atherosclerosis 2001;154:475–83.10.1016/S0021-9150(00)00499-8Search in Google Scholar

59. Matsuda M, Tamura R, Kanno K, Segawa T, Kinoshita H, Nishimoto O, Nishiyama H, Kawamoto T. Impact of dyslipidemic components of metabolic syndrome, adiponectin levels, and anti-diabetes medications on malondialdehyde-modified low-density lipoprotein levels in statin-treated diabetes patients with coronary artery disease. Diabetol Metab Syndr 2013;5:77.10.1186/1758-5996-5-77Search in Google Scholar PubMed PubMed Central

60. Shin HK, Kim YK, Kim KY, Lee JH, Hong KW. Remnant lipoprotein particles induce apoptosis in endothelial cells by NAD(P)H oxidase-mediated production of superoxide and cytokines via lectin-like oxidized low-density lipoprotein receptor-1 activation: prevention by cilostazol. Circulation 2004;109:1022–8.10.1161/01.CIR.0000117403.64398.53Search in Google Scholar PubMed

61. Watson AD, Berliner JA, Hama SY, La Du BN, Faull KF, Fogelman AM, Navab M. Protective effect of high density lipoprotein associated paraoxonase. Inhibition of the biological activity of minimally oxidized low density lipoprotein. J Clin Invest 1995;96:2882–91.10.1172/JCI118359Search in Google Scholar PubMed PubMed Central

62. Stefan N, Vozarova B, Funahashi T, Matsuzawa Y, Weyer C, Lindsay RS, Youngren JF, Havel PJ, Pratley RE, Bogardus C, Tataranni PA. Plasma adiponectin concentration is associated with skeletal muscle insulin receptor tyrosine phosphorylation, and low plasma concentration precedes a decrease in whole-body insulin sensitivity in humans. Diabetes 2002;51:1884–8.10.2337/diabetes.51.6.1884Search in Google Scholar PubMed

63. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA. Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 2001;86:1930–5.10.1210/jcem.86.5.7463Search in Google Scholar

64. Lindsay RS, Funahashi T, Hanson RL, Matsuzawa Y, Tanaka S, Tataranni PA, Knowler WC, Krakoff J. Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet 2002;360:57–8.10.1016/S0140-6736(02)09335-2Search in Google Scholar

65. Iwashima Y, Katsuya T, Ishikawa K, Ouchi N, Ohishi M, Sugimoto K, Fu Y, Motone M, Yamamoto K, Matsuo A, Ohashi K, Kihara S, Funahashi T, Rakugi H, Matsuzawa Y, Ogihara T. Hypoadiponectinemia is an independent risk factor for hypertension. Hypertension 2004;43:1318–23.10.1161/01.HYP.0000129281.03801.4bSearch in Google Scholar PubMed

66. Ryo M, Nakamura T, Kihara S, Kumada M, Shibazaki S, Takahashi M, Nagai M, Matsuzawa Y, Funahashi T. Adiponectin as a biomarker of the metabolic syndrome. Circ J 2004;68:975–81.10.1253/circj.68.975Search in Google Scholar PubMed

67. Kondo H, Shimomura I, Matsukawa Y, Kumada M, Takahashi M, Matsuda M, Ouchi N, Kihara S, Kawamoto T, Sumitsuji S, Funahashi T, Matsuzawa Y. Association of adiponectin mutation with type 2 diabetes: a candidate gene for the insulin resistance syndrome. Diabetes 2002;51:2325–8.10.2337/diabetes.51.7.2325Search in Google Scholar PubMed

68. Kumada M, Kihara S, Sumitsuji S, Kawamoto T, Matsumoto S, Ouchi N, Arita Y, Okamoto Y, Shimomura I, Hiraoka H, Nakamura T, Funahashi T, Matsuzawa Y. Association of hypoadiponectinemia with coronary artery disease in men. Arterioscler Thromb Vasc Biol 2003;23:85–9.10.1161/01.ATV.0000048856.22331.50Search in Google Scholar

69. Pischon T, Girman CJ, Hotamisligil GS, Rifai N, Hu FB, Rimm EB. Plasma adiponectin levels and risk of myocardial infarction in men. J Am Med Assoc 2004;291:1730–7.10.1001/jama.291.14.1730Search in Google Scholar PubMed

70. Marso SP, Mehta SK, Frutkin A, House JA, McCrary JR, Kulkarni KR. Low adiponectin levels are associated with atherogenic dyslipidemia and lipid-rich plaque in nondiabetic coronary arteries. Diabetes Care 2008;31:989–94.10.2337/dc07-2024Search in Google Scholar PubMed

71. Matsuda M, Tamura R, Kishida N, Segawa T, Kanno K, Nishimoto O, Nakamoto K, Nishiyama H, Kawamoto T. Predictive value of adiponectin in patients with multivessel coronary atherosclerosis detected on computed tomography angiography. J Atheroscler Thromb 2013;20:767–76.10.5551/jat.18036Search in Google Scholar PubMed

72. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 2001;7:941–6.10.1038/90984Search in Google Scholar PubMed

73. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, Ohteki T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, Nagai R, Kadowaki T. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003;423:762–9.10.1038/nature01705Search in Google Scholar PubMed

74. Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, Iwabu M, Okada-Iwabu M, Kawamoto S, Kubota N, Kubota T, Ito Y, Kamon J, Tsuchida A, Kumagai K, Kozono H, Hada Y, Ogata H, Tokuyama K, Tsunoda M, Ide T, Murakami K, Awazawa M, Takamoto I, Froguel P, Hara K, Tobe K, Nagai R, Ueki K, Kadowaki T. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med 2007;13:332–9.10.1038/nm1557Search in Google Scholar PubMed

75. Okamoto Y, Kihara S, Ouchi N, Nishida M, Arita Y, Kumada M, Ohashi K, Sakai N, Shimomura I, Kobayashi H, Terasaka N, Inaba T, Funahashi T, Matsuzawa Y. Adiponectin reduces atherosclerosis in apolipoprotein E-deficient mice. Circulation 2002;106:2767–70.10.1161/01.CIR.0000042707.50032.19Search in Google Scholar

76. Yamauchi T, Kamon J, Waki H, Imai Y, Shimozawa N, Hioki K, Uchida S, Ito Y, Takakuwa K, Matsui J, Takata M, Eto K, Terauchi Y, Komeda K, Tsunoda M, Murakami K, Ohnishi Y, Naitoh T, Yamamura K, Ueyama Y, Froguel P, Kimura S, Nagai R, Kadowaki T. Globular adiponectin protected ob/ob mice from diabetes and ApoE-deficient mice from atherosclerosis. J Biol Chem 2003;278:2461–8.10.1074/jbc.M209033200Search in Google Scholar PubMed

77. Ouchi N, Kihara S, Arita Y, Okamoto Y, Maeda K, Kuriyama H, Hotta K, Nishida M, Takahashi M, Muraguchi M, Ohmoto Y, Nakamura T, Yamashita S, Funahashi T, Matsuzawa Y. Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation 2000;102:1296–301.10.1161/01.CIR.102.11.1296Search in Google Scholar

78. Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, Ouchi N, Kihara S, Funahashi T, Tenner AJ, Tomiyama Y, Matsuzawa Y. Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood 2000;96:1723–32.10.1182/blood.V96.5.1723Search in Google Scholar

79. Ouchi N, Kihara S, Arita Y, Nishida M, Matsuyama A, Okamoto Y, Ishigami M, Kuriyama H, Kishida K, Nishizawa H, Hotta K, Muraguchi M, Ohmoto Y, Yamashita S, Funahashi T, Matsuzawa Y. Adipocyte-derived plasma protein, adiponectin, suppresses lipid accumulation and class A scavenger receptor expression in human monocyte-derived macrophages. Circulation 2001;103:1057–63.10.1161/01.CIR.103.8.1057Search in Google Scholar PubMed

80. Arita Y, Kihara S, Ouchi N, Maeda K, Kuriyama H, Okamoto Y, Kumada M, Hotta K, Nishida M, Takahashi M, Nakamura T, Shimomura I, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y. Adipocyte-derived plasma protein adiponectin acts as a platelet-derived growth factor-BB-binding protein and regulates growth factor-induced common postreceptor signal in vascular smooth muscle cell. Circulation 2002;105:2893–8.10.1161/01.CIR.0000018622.84402.FFSearch in Google Scholar

81. Kumada M, Kihara S, Ouchi N, Kobayashi H, Okamoto Y, Ohashi K, Maeda K, Nagaretani H, Kishida K, Maeda N, Nagasawa A, Funahashi T, Matsuzawa Y. Adiponectin specifically increased tissue inhibitor of metalloproteinase-1 through interleukin-10 expression in human macrophages. Circulation 2004;109:2046–9.10.1161/01.CIR.0000127953.98131.EDSearch in Google Scholar PubMed

82. Komura N, Maeda N, Mori T, Kihara S, Nakatsuji H, Hirata A, Tochino Y, Funahashi T, Shimomura I. Adiponectin protein exists in aortic endothelial cells. PLoS One 2013;8:e71271.10.1371/journal.pone.0071271Search in Google Scholar PubMed PubMed Central

83. Matsuura F, Oku H, Koseki M, Sandoval JC, Yuasa-Kawase M, Tsubakio-Yamamoto K, Masuda D, Maeda N, Tsujii K, Ishigami M, Nishida M, Hirano K, Kihara S, Hori M, Shimomura I, Yamashita S. Adiponectin accelerates reverse cholesterol transport by increasing high density lipoprotein assembly in the liver. Biochem Biophys Res Commun 2007;358:1091–5.10.1016/j.bbrc.2007.05.040Search in Google Scholar PubMed

84. Oku H, Matsuura F, Koseki M, Sandoval JC, Yuasa-Kawase M, Tsubakio-Yamamoto K, Masuda D, Maeda N, Ohama T, Ishigami M, Nishida M, Hirano K, Kihara S, Hori M, Shimomura I, Yamashita S. Adiponectin deficiency suppresses ABCA1 expression and ApoA-I synthesis in the liver. FEBS Lett 2007;581:5029–33.10.1016/j.febslet.2007.09.038Search in Google Scholar PubMed

85. Shibata R, Ouchi N, Ito M, Kihara S, Shiojima I, Pimentel DR, Kumada M, Sato K, Schiekofer S, Ohashi K, Funahashi T, Colucci WS, Walsh K. Adiponectin-mediated modulation of hypertrophic signals in the heart. Nat Med 2004;10:1384–9.10.1038/nm1137Search in Google Scholar PubMed PubMed Central

86. Shibata R, Sato K, Pimentel DR, Takemura Y, Kihara S, Ohashi K, Funahashi T, Ouchi N, Walsh K. Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nat Med 2005;11:1096–103.10.1038/nm1295Search in Google Scholar PubMed PubMed Central

87. Fujita K, Maeda N, Sonoda M, Ohashi K, Hibuse T, Nishizawa H, Nishida M, Hiuge A, Kurata A, Kihara S, Shimomura I, Funahashi T. Adiponectin protects against angiotensin II-induced cardiac fibrosis through activation of PPAR-alpha. Arterioscler Thromb Vasc Biol 2008;28:863–70.10.1161/ATVBAHA.107.156687Search in Google Scholar PubMed

88. Tao L, Gao E, Jiao X, Yuan Y, Li S, Christopher TA, Lopez BL, Koch W, Chan L, Goldstein BJ, Ma XL. Adiponectin cardioprotection after myocardial ischemia/reperfusion involves the reduction of oxidative/nitrative stress. Circulation 2007;115:1408–16.10.1161/CIRCULATIONAHA.106.666941Search in Google Scholar PubMed

89. Wang Y, Gao E, Tao L, Lau WB, Yuan Y, Goldstein BJ, Lopez BL, Christopher TA, Tian R, Koch W, Ma XL. AMP-activated protein kinase deficiency enhances myocardial ischemia/reperfusion injury but has minimal effect on the antioxidant/antinitrative protection of adiponectin. Circulation 2009;119:835–44.10.1161/CIRCULATIONAHA.108.815043Search in Google Scholar PubMed PubMed Central

90. Li R, Wang WQ, Zhang H, Yang X, Fan Q, Christopher TA, Lopez BL, Tao L, Goldstein BJ, Gao F, Ma XL. Adiponectin improves endothelial function in hyperlipidemic rats by reducing oxidative/nitrative stress and differential regulation of eNOS/iNOS activity. Am J Physiol Endocrinol Metab 2007;293:E1703–8.10.1152/ajpendo.00462.2007Search in Google Scholar PubMed

91. Essick EE, Ouchi N, Wilson RM, Ohashi K, Ghobrial J, Shibata R, Pimentel DR, Sam F. Adiponectin mediates cardioprotection in oxidative stress-induced cardiac myocyte remodeling. Am J Physiol Heart Circ Physiol 2011;301:H984–93.10.1152/ajpheart.00428.2011Search in Google Scholar PubMed PubMed Central

92. Yi W, Sun Y, Gao E, Wei X, Lau WB, Zheng Q, Wang Y, Yuan Y, Wang X, Tao L, Li R, Koch W, Ma XL. Reduced cardioprotective action of adiponectin in high-fat diet-induced type II diabetic mice and its underlying mechanisms. Antioxid Redox Signal 2011;15:1779–88.10.1089/ars.2010.3722Search in Google Scholar PubMed PubMed Central

93. Tsuchida A, Yamauchi T, Ito Y, Hada Y, Maki T, Takekawa S, Kamon J, Kobayashi M, Suzuki R, Hara K, Kubota N, Terauchi Y, Froguel P, Nakae J, Kasuga M, Accili D, Tobe K, Ueki K, Nagai R, Kadowaki T. Insulin/Foxo1 pathway regulates expression levels of adiponectin receptors and adiponectin sensitivity. J Biol Chem 2004;279:30817–22.10.1074/jbc.M402367200Search in Google Scholar

94. Maeda N, Takahashi M, Funahashi T, Kihara S, Nishizawa H, Kishida K, Nagaretani H, Matsuda M, Komuro R, Ouchi N, Kuriyama H, Hotta K, Nakamura T, Shimomura I, Matsuzawa Y. PPARgamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein. Diabetes 2001;50:2094–9.10.2337/diabetes.50.9.2094Search in Google Scholar

95. Kubota N, Terauchi Y, Kubota T, Kumagai H, Itoh S, Satoh H, Yano W, Ogata H, Tokuyama K, Takamoto I, Mineyama T, Ishikawa M, Moroi M, Sugi K, Yamauchi T, Ueki K, Tobe K, Noda T, Nagai R, Kadowaki T. Pioglitazone ameliorates insulin resistance and diabetes by both adiponectin-dependent and -independent pathways. J Biol Chem 2006;281:8748–55.10.1074/jbc.M505649200Search in Google Scholar

96. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005;366:1279–89.10.1016/S0140-6736(05)67528-9Search in Google Scholar

97. Duval C, Muller M, Kersten S. PPARalpha and dyslipidemia. Biochim Biophys Acta 2007;1771:961–71.10.1016/j.bbalip.2007.05.003Search in Google Scholar

98. Hiuge A, Tenenbaum A, Maeda N, Benderly M, Kumada M, Fisman EZ, Tanne D, Matas Z, Hibuse T, Fujita K, Nishizawa H, Adler Y, Motro M, Kihara S, Shimomura I, Behar S, Funahashi T. Effects of peroxisome proliferator-activated receptor ligands, bezafibrate and fenofibrate, on adiponectin level. Arterioscler Thromb Vasc Biol 2007;27:635–41.10.1161/01.ATV.0000256469.06782.d5Search in Google Scholar

99. The Bezafibrate Infarction Prevention (BIP) Study Group. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation 2000;102:21–7.10.1161/01.CIR.102.1.21Search in Google Scholar

100. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, Forder P, Pillai A, Davis T, Glasziou P, Drury P, Kesaniemi YA, Sullivan D, Hunt D, Colman P, d’Emden M, Whiting M, Ehnholm C, Laakso M. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005;366:1849–61.10.1016/S0140-6736(05)67667-2Search in Google Scholar

101. Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, Muscat GE. The peroxisome proliferator-activated receptor beta/delta agonist, GW501516, regulates the expression of genes involved in lipid catabolism and energy uncoupling in skeletal muscle cells. Mol Endocrinol 2003;17:2477–93.10.1210/me.2003-0151Search in Google Scholar PubMed

102. Luquet S, Lopez-Soriano J, Holst D, Fredenrich A, Melki J, Rassoulzadegan M, Grimaldi PA. Peroxisome proliferator-activated receptor delta controls muscle development and oxidative capability. FASEB J 2003;17:2299–301.10.1096/fj.03-0269fjeSearch in Google Scholar PubMed

103. Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, Evans RM. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 2003;113:159–70.10.1016/S0092-8674(03)00269-1Search in Google Scholar

104. Viengchareun S, Le Menuet D, Martinerie L, Munier M, Pascual-Le Tallec L, Lombes M. The mineralocorticoid receptor: insights into its molecular and (patho)physiological biology. Nucl Recept Signal 2007;5:e012.10.1621/nrs.05012Search in Google Scholar

105. Callera GE, Touyz RM, Tostes RC, Yogi A, He Y, Malkinson S, Schiffrin EL. Aldosterone activates vascular p38MAP kinase and NADPH oxidase via c-Src. Hypertension 2005;45:773–9.10.1161/01.HYP.0000154365.30593.d3Search in Google Scholar

106. Funder JW. Mineralocorticoid receptors: distribution and activation. Heart Fail Rev 2005;10:15–22.10.1007/s10741-005-2344-2Search in Google Scholar

107. Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC. Pivotal role of the mineralocorticoid receptor in corticosteroid-induced adipogenesis. FASEB J 2007;21:2185–94.10.1096/fj.06-7970comSearch in Google Scholar

108. Caprio M, Antelmi A, Chetrite G, Muscat A, Mammi C, Marzolla V, Fabbri A, Zennaro MC, Feve B. Antiadipogenic effects of the mineralocorticoid receptor antagonist drospirenone: potential implications for the treatment of metabolic syndrome. Endocrinology 2011;152:113–25.10.1210/en.2010-0674Search in Google Scholar

109. Bujalska IJ, Kumar S, Stewart PM. Does central obesity reflect “Cushing’s disease of the omentum”? Lancet 1997;349:1210–3.10.1016/S0140-6736(96)11222-8Search in Google Scholar

110. Engeli S, Bohnke J, Feldpausch M, Gorzelniak K, Heintze U, Janke J, Luft FC, Sharma AM. Regulation of 11beta-HSD genes in human adipose tissue: influence of central obesity and weight loss. Obes Res 2004;12:9–17.10.1038/oby.2004.3Search in Google Scholar PubMed

111. Wake DJ, Rask E, Livingstone DE, Soderberg S, Olsson T, Walker BR. Local and systemic impact of transcriptional up-regulation of 11beta-hydroxysteroid dehydrogenase type 1 in adipose tissue in human obesity. J Clin Endocrinol Metab 2003;88:3983–8.10.1210/jc.2003-030286Search in Google Scholar PubMed

112. Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ, Seckl JR, Flier JS. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001;294:2166–70.10.1126/science.1066285Search in Google Scholar PubMed

113. Berger J, Tanen M, Elbrecht A, Hermanowski-Vosatka A, Moller DE, Wright SD, Thieringer R. Peroxisome proliferator-activated receptor-gamma ligands inhibit adipocyte 11beta-hydroxysteroid dehydrogenase type 1 expression and activity. J Biol Chem 2001;276:12629–35.10.1074/jbc.M003592200Search in Google Scholar PubMed

114. Zennaro MC, Caprio M, Feve B. Mineralocorticoid receptors in the metabolic syndrome. Trends Endocrinol Metab 2009;20:444–51.10.1016/j.tem.2009.05.006Search in Google Scholar PubMed

115. Hoppmann J, Perwitz N, Meier B, Fasshauer M, Hadaschik D, Lehnert H, Klein J. The balance between gluco- and mineralo-corticoid action critically determines inflammatory adipocyte responses. J Endocrinol 2010;204:153–64.10.1677/JOE-09-0292Search in Google Scholar PubMed

116. Oakley RH, Ren R, Cruz-Topete D, Bird GS, Myers PH, Boyle MC, Schneider MD, Willis MS, Cidlowski JA. Essential role of stress hormone signaling in cardiomyocytes for the prevention of heart disease. Proc Natl Acad Sci USA 2013;110:17035–40.10.1073/pnas.1302546110Search in Google Scholar PubMed PubMed Central

117. Repa JJ, Mangelsdorf DJ. The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol 2000;16:459–81.10.1146/annurev.cellbio.16.1.459Search in Google Scholar PubMed

118. Venkateswaran A, Laffitte BA, Joseph SB, Mak PA, Wilpitz DC, Edwards PA, Tontonoz P. Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXR alpha. Proc Natl Acad Sci USA 2000;97:12097–102.10.1073/pnas.200367697Search in Google Scholar PubMed PubMed Central

119. Repa JJ, Turley SD, Lobaccaro JA, Medina J, Li L, Lustig K, Shan B, Heyman RA, Dietschy JM, Mangelsdorf DJ. Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science 2000;289:1524–9.10.1126/science.289.5484.1524Search in Google Scholar PubMed

120. Joseph SB, McKilligin E, Pei L, Watson MA, Collins AR, Laffitte BA, Chen M, Noh G, Goodman J, Hagger GN, Tran J, Tippin TK, Wang X, Lusis AJ, Hsueh WA, Law RE, Collins JL, Willson TM, Tontonoz P. Synthetic LXR ligand inhibits the development of atherosclerosis in mice. Proc Natl Acad Sci USA 2002;99:7604–9.10.1073/pnas.112059299Search in Google Scholar PubMed PubMed Central

121. Repa JJ, Liang G, Ou J, Bashmakov Y, Lobaccaro JM, Shimomura I, Shan B, Brown MS, Goldstein JL, Mangelsdorf DJ. Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes Dev 2000;14:2819–30.10.1101/gad.844900Search in Google Scholar PubMed PubMed Central

122. Inaba T, Matsuda M, Shimamura M, Takei N, Terasaka N, Ando Y, Yasumo H, Koishi R, Makishima M, Shimomura I. Angiopoietin-like protein 3 mediates hypertriglyceridemia induced by the liver X receptor. J Biol Chem 2003;278:21344–51.10.1074/jbc.M213202200Search in Google Scholar PubMed

123. Hummasti S, Laffitte BA, Watson MA, Galardi C, Chao LC, Ramamurthy L, Moore JT, Tontonoz P. Liver X receptors are regulators of adipocyte gene expression but not differentiation: identification of apoD as a direct target. J Lipid Res 2004;45:616–25.10.1194/jlr.M300312-JLR200Search in Google Scholar

124. Gerin I, Dolinsky VW, Shackman JG, Kennedy RT, Chiang SH, Burant CF, Steffensen KR, Gustafsson JA, MacDougald OA. LXRbeta is required for adipocyte growth, glucose homeostasis, and beta cell function. J Biol Chem 2005;280:23024–31.10.1074/jbc.M412564200Search in Google Scholar

125. Dalen KT, Ulven SM, Bamberg K, Gustafsson JA, Nebb HI. Expression of the insulin-responsive glucose transporter GLUT4 in adipocytes is dependent on liver X receptor alpha. J Biol Chem 2003;278:48283–91.10.1074/jbc.M302287200Search in Google Scholar

126. Laffitte BA, Chao LC, Li J, Walczak R, Hummasti S, Joseph SB, Castrillo A, Wilpitz DC, Mangelsdorf DJ, Collins JL, Saez E, Tontonoz P. Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue. Proc Natl Acad Sci USA 2003;100:5419–24.10.1073/pnas.0830671100Search in Google Scholar

127. Stenson BM, Ryden M, Steffensen KR, Wahlen K, Pettersson AT, Jocken JW, Arner P, Laurencikiene J. Activation of liver X receptor regulates substrate oxidation in white adipocytes. Endocrinology 2009;150:4104–13.10.1210/en.2009-0676Search in Google Scholar

128. Kalaany NY, Gauthier KC, Zavacki AM, Mammen PP, Kitazume T, Peterson JA, Horton JD, Garry DJ, Bianco AC, Mangelsdorf DJ. LXRs regulate the balance between fat storage and oxidation. Cell Metab 2005;1:231–44.10.1016/j.cmet.2005.03.001Search in Google Scholar

129. Korach-Andre M, Archer A, Barros RP, Parini P, Gustafsson JA. Both liver-X receptor (LXR) isoforms control energy expenditure by regulating brown adipose tissue activity. Proc Natl Acad Sci USA 2011;108:403–8.10.1073/pnas.1017884108Search in Google Scholar

130. Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, Hull MV, Lustig KD, Mangelsdorf DJ, Shan B. Identification of a nuclear receptor for bile acids. Science 1999;284:1362–5.10.1126/science.284.5418.1362Search in Google Scholar

131. Goodwin B, Jones SA, Price RR, Watson MA, McKee DD, Moore LB, Galardi C, Wilson JG, Lewis MC, Roth ME, Maloney PR, Willson TM, Kliewer SA. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell 2000;6:517–26.10.1016/S1097-2765(00)00051-4Search in Google Scholar

132. Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, Mangelsdorf DJ. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell 2000;6:507–15.10.1016/S1097-2765(00)00050-2Search in Google Scholar

133. Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, Velagapudi VR, Dumont J, Bouchaert E, van Dijk TH, Lucas A, Dorchies E, Daoudi M, Lestavel S, Gonzalez FJ, Oresic M, Cariou B, Kuipers F, Caron S, Staels B. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes 2011;60:1861–71.10.2337/db11-0030Search in Google Scholar PubMed PubMed Central

134. Hartman HB, Gardell SJ, Petucci CJ, Wang S, Krueger JA, Evans MJ. Activation of farnesoid X receptor prevents atherosclerotic lesion formation in LDLR–/– and apoE–/– mice. J Lipid Res 2009;50:1090–100.10.1194/jlr.M800619-JLR200Search in Google Scholar PubMed PubMed Central

135. Lee JM, Lee YK, Mamrosh JL, Busby SA, Griffin PR, Pathak MC, Ortlund EA, Moore DD. A nuclear-receptor-dependent phosphatidylcholine pathway with antidiabetic effects. Nature 2011;474:506–10.10.1038/nature10111Search in Google Scholar PubMed PubMed Central

136. Nishigori H, Tomura H, Tonooka N, Kanamori M, Yamada S, Sho K, Inoue I, Kikuchi N, Onigata K, Kojima I, Kohama T, Yamagata K, Yang Q, Matsuzawa Y, Miki T, Seino S, Kim MY, Choi HS, Lee YK, Moore DD, Takeda J. Mutations in the small heterodimer partner gene are associated with mild obesity in Japanese subjects. Proc Natl Acad Sci USA 2001;98:575–80.10.1073/pnas.98.2.575Search in Google Scholar PubMed PubMed Central

137. Park YJ, Kim SC, Kim J, Anakk S, Lee JM, Tseng HT, Yechoor V, Park J, Choi JS, Jang HC, Lee KU, Novak CM, Moore DD, Lee YK. Dissociation of diabetes and obesity in mice lacking orphan nuclear receptor small heterodimer partner. J Lipid Res 2011;52:2234–44.10.1194/jlr.M016048Search in Google Scholar PubMed PubMed Central

138. Wang L, Liu J, Saha P, Huang J, Chan L, Spiegelman B, Moore DD. The orphan nuclear receptor SHP regulates PGC-1alpha expression and energy production in brown adipocytes. Cell Metab 2005;2:227–38.10.1016/j.cmet.2005.08.010Search in Google Scholar PubMed

139. Nishizawa H, Yamagata K, Shimomura I, Takahashi M, Kuriyama H, Kishida K, Hotta K, Nagaretani H, Maeda N, Matsuda M, Kihara S, Nakamura T, Nishigori H, Tomura H, Moore DD, Takeda J, Funahashi T, Matsuzawa Y. Small heterodimer partner, an orphan nuclear receptor, augments peroxisome proliferator-activated receptor gamma transactivation. J Biol Chem 2002;277:1586–92.10.1074/jbc.M104301200Search in Google Scholar PubMed

140. Li L, Xie X, Qin J, Jeha GS, Saha PK, Yan J, Haueter CM, Chan L, Tsai SY, Tsai MJ. The nuclear orphan receptor COUP-TFII plays an essential role in adipogenesis, glucose homeostasis, and energy metabolism. Cell Metab 2009;9:77–87.10.1016/j.cmet.2008.12.002Search in Google Scholar PubMed PubMed Central

141. Xu Z, Yu S, Hsu CH, Eguchi J, Rosen ED. The orphan nuclear receptor chicken ovalbumin upstream promoter-transcription factor II is a critical regulator of adipogenesis. Proc Natl Acad Sci USA 2008;105:2421–6.10.1073/pnas.0707082105Search in Google Scholar PubMed PubMed Central

142. Duez H, Duhem C, Laitinen S, Patole PS, Abdelkarim M, Bois-Joyeux B, Danan JL, Staels B. Inhibition of adipocyte differentiation by RORalpha. FEBS Lett 2009;583:2031–6.10.1016/j.febslet.2009.05.019Search in Google Scholar PubMed

143. Luo J, Sladek R, Carrier J, Bader JA, Richard D, Giguere V. Reduced fat mass in mice lacking orphan nuclear receptor estrogen-related receptor alpha. Mol Cell Biol 2003;23:7947–56.10.1128/MCB.23.22.7947-7956.2003Search in Google Scholar PubMed PubMed Central

144. Kubo M, Ijichi N, Ikeda K, Horie-Inoue K, Takeda S, Inoue S. Modulation of adipogenesis-related gene expression by estrogen-related receptor gamma during adipocytic differentiation. Biochim Biophys Acta 2009;1789:71–7.10.1016/j.bbagrm.2008.08.012Search in Google Scholar PubMed

145. Kumar N, Solt LA, Wang Y, Rogers PM, Bhattacharyya G, Kamenecka TM, Stayrook KR, Crumbley C, Floyd ZE, Gimble JM, Griffin PR, Burris TP. Regulation of adipogenesis by natural and synthetic REV-ERB ligands. Endocrinology 2010;151:3015–25.10.1210/en.2009-0800Search in Google Scholar PubMed PubMed Central

Received: 2014-1-6
Accepted: 2014-3-17
Published Online: 2014-4-10
Published in Print: 2014-8-1

©2014 by De Gruyter

Downloaded on 1.5.2024 from https://www.degruyter.com/document/doi/10.1515/hmbci-2014-0001/html
Scroll to top button