Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Oct 1.
Published in final edited form as: Clin Chim Acta. 2007 Feb 2;380(1-2):24–30. doi: 10.1016/j.cca.2007.01.026

Adiponectin as an anti-inflammatory factor

Noriyuki Ouchi 1, Kenneth Walsh 1
PMCID: PMC2755046  NIHMSID: NIHMS23075  PMID: 17343838

Abstract

Obesity is characterized by low-grade systemic inflammation. Adiponectin is an adipose tissue-derived hormone, which is downregulated in obesity. Adiponectin displays protective actions on the development of various obesity-linked diseases. Several clinical studies demonstrate the inverse relationship between plasma adiponectin levels and several inflammatory markers including C-reactive protein. Adiponectin attenuates inflammatory responses to multiple stimuli by modulating signaling pathways in a variety of cell types. The anti-inflammatory properties of adiponectin may be a major component of its beneficial effects on cardiovascular and metabolic disorders including atherosclerosis and insulin resistance. In this reviews, we focus on the role of adiponectin in regulation of inflammatory response and discuss its potential as an antiinflammatory marker.

Keywords: adiponectin, anti-inflammatory, cardioprotection, biomarker

1. Introduction

Increasing evidence indicates that chronic mild inflammation linked to obesity is closely associated with the development of insulin resistance and cardiovascular disorders [1]. A number of bioactive substances secreted from fat tissue, referred to as adipokines, could contribute to the complications of obesity through the regulation of inflammatory and immune responses [2, 3]. Adipokines include pro-inflammatory cytokines/chemokines such as leptin, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) [2, 3]. In contrast, the adipokine adiponectin exerts anti-inflammatory actions on a number of cell types.

Adiponectin, also referred to as ACRP30, AdipoQ and gelatin-binding protein-28 [4-6], is expressed almost exclusively in adipose tissue [5, 6]. Adiponectin is abundantly present in blood stream, and the average levels of plasma adiponectin in human range 3 to 30 μg/ml [2, 7]. Adiponectin is a 244-amino acid protein that contains a putative signal sequence and a collagen-like domain followed by a globular domain similar to collagens VIII and X and compliment factor C1q. Of importance, plasma adiponectin levels are paradoxically decreased in obese subjects [7]. Low plasma adiponectin levels, known as hypoadiponectinemia, are closely associated with obesity-linked complications including type 2 diabetes, coronary heart disease and hypertension. A number of experimental studies suggest that adiponectin can act as a protective factor against insulin resistance and cardiovascular disease. In this review, we focus on the possible role of adiponectin in obesity-associated inflammatory states.

2. Experimental data

2.1. Vascular protection

2.1.1. Atherosclerosis

Atherosclerosis is characterized by chronic systemic inflammation. Endothelial cell activation by pro-inflammatory stimuli and subsequent monocyte adherent to injured endothelium is a precipitating event in atherogenesis [8]. Adiponectin treatment reduces TNF-α-stimulated expression of vascular cell adhesion molecule-1 (VCAM-1), E-selectin, intracellular adhesion molecule-1 and IL-8 in human aortic endothelial cells as well as monocyte attachment to TNF-α-stimulated endothelial cells (Figure) [9, 10]. Adiponectin also inhibits TNF-α-induced nuclear factor-κB (NF-κB) activation in endothelial cells (Figure) [10, 11]. The inhibitory effect of adiponectin on NF-κB pathway is mediated, at least in part, by its ability to promote signaling through cyclicAMP (cAMP)/protein kinase A (PKA)(Figure). A recent study has also demonstrated that adiponectin attenuates high glucose-induced production of reactive oxygen species in endothelial cells through a cAMP-PKA-dependent mechanism (Figure) [12]. Thus, activation of cAMP-PKA signaling is an important mechanism by which adiponectin protects endothelial cells from activation.

Figure.

Figure

Adiponectin-mediated regulation of inflammatory responses in different cell types. Adiponectin negatively regulates CRP and TNF-α expression in adipose tissue, whereas adiponectin expression is inhibited by TNF-α and IL-6. Adiponectin inhibits IL-8, VCAM-1 and ROS production in endothelial cells through cAMP-PKA-dependent signaling. Adiponectin also stimulates AMPK activation in endothelial cells, leading to activation of eNOS. Adiponectin suppresses TNF-α production in cardiac cells through its ability to stimulate the COX-2-PGE2 pathway. In macrophages, adiponectin attenuates TNF-α and IL-6 production through its ability to suppress NF-κB activation, and enhances IL-10 expression resulting in increase in TIMP-1 production. Adiponectin inhibits foam cell formation by reducing SR-A expression.

The accumulation of lipid-laden foam cells and macrophage-related inflammation are key features of atherosclerotic lesion progression. Adiponectin inhibits macrophage-to-foam cell transformation and reduces intracellular cholesteryl ester content in human macrophages by suppressing expression of class A scavenger receptor (SR-A) [13]. Pretreatment with adiponectin reduces lipopolysaccharide (LPS)-stimulated TNF-α production in human and porcine macrophages (Figure) [14, 15]. Adiponectin treatment also inhibits Toll-like receptor-mediated NF-κB activation in mouse macrophages [16]. Adiponectin stimulates production of IL-10, an anti-inflammatory cytokine in porcine macrophages [15], and it increases the production of tissue inhibitor of metalloproteinase-1 in human macrophages through its ability to stimulate IL-10 expression [17]. Furthermore, a recent clinical study showed a positive correlation between plasma adiponectin and IL-10 concentrations (Figure) [18].

Consistent with these in vitro findings, adiponectin exhibit anti-atherogenic actions in a mouse model [19, 20]. Adenovirus-mediated supplementation of adiponectin inhibits the formation of atherosclerotic lesions and decreases mRNA levels of SR-A, TNF-α and VCAM-1 in the vascular wall [19]. Importantly, adiponectin does not affect glucose and lipid parameters in this model. Thus, adiponectin appears to attenuate atherogenesis through multiple anti-inflammatory actions on macrophages and vascular endothelial cells.

2.1.2. Endothelial function and blood vessel growth

Functional abnormalities of endothelium including endothelial dysfunction, microvascular rarefaction and reduced collateralization occur with many obesity-linked pathological conditions including diabetes and hypertension [21-27]. Several studies have shown that adiponectin exerts beneficial actions on endothelial homeostasis. Adiponectin acts as an important regulator of endothelial nitric oxide synthase (eNOS), a key determinant of endothelial function and angiogenesis. Adiponectin promotes eNOS phosphorylation in endothelial cells through AMP-activated protein kinase (AMPK)-dependent signaling mechanisms (Figure) [28, 29]. Adiponectin enhances eNOS expression/activity in endothelial cells and restores the suppression of eNOS activity by OxLDL [30-32]. Production of endothelial NO suppresses vascular inflammatory responses [33].

In a recent in vivo study, adiponectin-deficient (APN-KO) mice exhibit hypertension on a high salt diet, which is associated with reduced eNOS mRNA and protein levels in aorta [34]. Delivery of adenovirus expressing adiponectin reduces the elevated blood pressure and reverses the decreased eNOS mRNA levels in aorta in APN-KO mice that have been fed high salt diets. Importantly, inhibition of eNOS activity with L-NAME blocked the adiponectin-induced decrease in blood pressure in APN-KO mice on a high salt diet. APN-KO mice also show hypertension and impaired endothelium-dependent vasodilation when fed an atherogenic diet [35]. These studies suggest that adiponectin plays a crucial role in retaining vascular tone and function by induction of eNOS expression and activity.

APN-KO mice exhibit impaired neovascularization in response to hindlimb ischemia, whereas adenovirus-mediated overexpression of adiponectin promotes angiogenic repair which is dependent on the activation of AMPK signaling [36]. In accordance with its angiogenic actions in vivo, adiponectin stimulates endothelial cell migration and differentiation into capillary-like structures in vitro through its ability to promote cross-talk between AMPK and Akt signaling [28, 32]. Adiponectin also prevents apoptosis in endothelial cells through activation of AMPK signaling [37, 38]. Taken together, these favorable actions of adiponectin on endothelial function and vessel growth could contribute to vascular protection.

2.2. Cardioprotection

Recent findings have shown that adiponectin exerts beneficial actions on the heart under pathological conditions. It was shown that myocardial ischemia-reperfusion injury results in larger infarct sizes in APN-KO mice than in wild-type mice [39]. APN-KO mice also exhibit increased myocardial cell apoptosis and TNF-α production in response to ischemia-reperfusion compared with wild-type mice. Adenovirus-mediated overexpression of adiponectin reduces infarct size, myocardial apoptosis and TNF-α production in both APN-KO and wild-type mice. A clinical study has found that there is a significant decrease in circulating adiponectin levels immediately after acute myocardial infarction [40]. The reduction of adiponectin levels following acute myocardial infarction inversely correlates with plasma CRP concentrations, suggesting that hypoadiponectinemia participates in an increased inflammatory response to acute myocardial ischemia.

Cell culture experiments show that adiponectin has anti-inflammatory and anti-apoptotic effects on cardiac myocytes and fibroblasts. Pretreatment with adiponectin reduces LPS-induced TNF-α production in cardiac cells [39]. Cyclooxygenase-2 (COX-2) and its metabolites have been shown to play important protective roles in myocardial ischemia-reperfusion damage [41-44]. Adiponectin treatment stimulates COX-2 expression and prostaglandin E2 (PGE2) synthesis in cardiac cells. Inhibition of the COX-2-PGE2 pathway blocks the inhibitory effects of adiponectin on LPS-induced secretion of TNF-α from cardiac cells. Furthermore, the inhibitory effects of adiponectin on infarct size and TNF-α production in vivo are partially reversed by COX-2 inhibition. Adiponectin also possesses an anti-apoptotic property in cardiac cells in vitro as is found in endothelial cells. Adiponectin inhibits apoptosis in cardiac cells under conditions of serum deprivation and hypoxia-reoxygenation [39]. Adiponectin activates AMPK signaling in cardiac cells, and inhibition of AMPK activation reverses the pro-survival actions of adiponectin. Collectively, adiponectin protects against myocardial ischemia-reperfusion injury through COX-2-mediated anti-inflammatory and AMPK-mediated anti-apoptotic mechanisms (Figure).

Pathological cardiac remodeling is associated with obesity-related diseases [45, 46]. Several experimental studies have demonstrated the impact of adiponectin on pathological cardiac hypertrophy. APN-KO mice have enhanced concentric cardiac hypertrophy and increased mortality in response to pressure overload [47, 48]. Conversely, adenovirus-mediated delivery of adiponectin attenuates cardiac hypertrophy following pressure overload in APN-KO, wild-type and diabetic db/db mice [47]. Adiponectin supplementation also attenuates angiotensin II-stimulated cardiac hypertrophy in wild-type mice. In cultured cardiac myocytes, adiponectin suppresses agonist-stimulated ERK activation and hypertrophy [47, 49]. The suppressive effects of adiponectin on ERK and hypertrophy are reversed by inhibition of AMPK. AMPK activity increases in hearts with pressure overload hypertrophy [50], and AMPK phosphorylation is attenuated in the hearts of APN-KO mice following pressure overload [47]. Taken together, these findings suggest that adiponectin functions as a suppressor of pathological hypertrophic response in the heart by its ability to activate AMPK signaling.

2.3. Metabolic actions

Accumulating evidence from experimental models indicates that adiponectin plays a protective role in the development of insulin resistance and diabetes. APN-KO mice develop severe diet-induced insulin resistance when fed high fat/sucrose [51] or high fat diets [52]. In another study, APN-KO mice exhibit moderate insulin resistance when fed a normal chow diet [53]. In contrast, another strain of APN-KO mice does not display a detectable insulin-resistant phenotype [54]. Adiponectin protein treatment has been shown to reduce hyperglycemia in diabetic mice without affecting insulin levels [55]. Administration of adiponectin increases fatty acid oxidation in muscle and reduces plasma levels of glucose, free fatty acids and triglycerides [56]. These data suggest that adiponectin acts as an insulin-sensitizing factor.

The beneficial actions of adiponectin on insulin resistance appear to be mediated in part by its ability to activate AMPK in skeletal muscle [57, 58], liver [58] and adipocytes [59]. Adenovirus-mediated delivery of adiponectin is reported to enhance AMPK activation in skeletal muscle and increase insulin sensitivity in rats [60]. Adiponectin transgenic mice also show improved insulin sensitivity and increased AMPK activation in liver [61]. AMPK activation by adiponectin is believed to be mediated through its cell surface receptors AdipoR1 and AdipoR2 [62]. Of interest, APN-KO mice show elevated levels of TNF-α mRNA in adipose tissues and high serum TNF-α concentrations [51]. Adenovirus-mediated supplementation of adiponectin reduces the increased TNF-α levels in APN-KO mice, and this effect is accompanied by improvement of insulin resistance. Therefore, the protective actions of adiponectin on insulin resistance are also the result of its ability to suppress inflammatory cytokine production.

3. Clinical features

3.1 Obesity-linked metabolic disorders

Numerous epidemiological studies emphasized the association between adiponectin levels and obesity-linked conditions. Plasma adiponectin levels are significantly lower in obese subjects compared with non-obese subjects [7]. This inverse correlation is observed between plasma adiponectin concentrations and body mass index (BMI) in both genders [7]. In addition, plasma adiponectin levels negatively correlate with visceral fat accumulation in both men and women[63].

Several clinical studies demonstrate an association between hypoadiponectinemia and the development of insulin resistance and type 2 diabetes. A cross-sectional study showed that plasma adiponectin concentrations are lower in type 2 diabetic patients than in age- and BMI-matched nondiabetic men and women [64]. A longitudinal study of Pima Indian demonstrated that high adiponectin levels were associated with a lower risk for developing type 2 diabetes [65]. In support of these findings, other clinical studies show a negative association of circulating adiponectin with the development of insulin resistance and type 2 diabetes [66-69].

Plasma adiponectin levels correlate positively with HDL cholesterol levels and negatively with triglyceride levels [2, 63, 64, 70]. An inverse correlation is observed between adiponectin levels and mean blood pressures in patients with essential hypertension [71], and another study demonstrates that hypoadiponectinemia can be an independent risk factor for hypertension [72]. Taken together, these data suggest that hypoadiponectinemia strongly associates with well-defined conventional risk factors for cardiovascular disease.

3.2 Coronary heart disease

Several studies have investigated the interaction of adiponectin level with coronary artery disease. Hypoadiponectinemia is found in patients with angiographically documented coronary atherosclerosis [9, 73] as well as acute coronary syndrome [74]. The plasma adiponectin levels could significantly predict the extent of coronary atherosclerosis in men [75]. A prospective study of patients with end-stage renal failure demonstrates the inverse relationship between plasma adiponectin levels and cardiovascular events [76]. In addition, two prospective studies show that increased adiponectin levels are associated with a decreased risk of myocardial infarction in healthy men [77] and a moderately decreased coronary artery disease (CAD) risk in diabetic men [78]. On the contrary, two recent prospective studies showed that adiponectin concentrations did not significantly correlate with the risk of coronary heart disease in American Indian and in British women [79, 80]. In a more recent large prospective study in British men with coronary heart disease combined with a meta-analysis of previously published 7 prospective studies, the association between circulating adiponectin levels and the CAD risk was found to be relatively weak [81]. These inconsistent findings might be due to differences in the study populations (e.g. existing disease, ethnicity and gender)(Table). Thus, it remains controversial whether hypoadiponectinemia is a reliable indicator of CAD, and future studies are required to define this relationship.

Table.

Association of adiponectin with risk of CAD in prospective studies.

Study population Gender Association Reference
Renal failure M/F Yes 76
Healthy Americans M Yes 77
Diabetes M Yes 78
American Indians M/F No 79
British W No 80
British M No 81

M: Male, F: Female

3.3 Adiponectin and inflammatory states

Obesity-related disorders including atherosclerosis and insulin resistance are tightly linked to low-grade chronic inflammation. A number of studies have investigated the association between adiponectin levels and pro-inflammatory markers in various populations. The inflammatory marker C-reactive protein (CRP), is considered to be an independent predictor of future risk for cardiovascular outcome and a risk factor of developing the metabolic syndrome [82, 83]. CRP levels are also reported to associate positively with BMI [84, 85], suggesting that CRP is a useful biomarker for obesity-linked chronic inflammatory states.

Plasma CRP levels negatively correlate with plasma adiponectin levels in men [2, 72, 86]. CRP mRNA is expressed in human adipose tissue, indicating that adiponectin may participate in the reduction in plasma CRP levels through its ability to negatively regulate CRP expression in adipose tissue. In another study, adiponectin levels in plasma and adipose tissue were shown to inversely correlate with CRP levels in healthy obese women [87]. Circulating adiponectin levels are negatively correlated with CRP levels in diabetic patients [70, 88, 89]. Furthermore, hypoadiponectinemia is associated with elevated CRP levels in non-diabetic women [90], healthy men [91] and non-diabetic subjects [18]. In contrast, one study did not find a significant correlation between plasma adiponectin levels and CRP levels in patients with CAD [92].

Both IL-6 and TNF-α are important pro-inflammatory adipokines, which contribute to regulation of CRP production in liver [93]. Experimental studies demonstrated that adiponectin expression is negatively regulated by pro-inflammatory cytokines including IL-6 and TNF-α, whereas adiponectin modulates the action and production of TNF-α in various tissues (See section 4). Accordingly, several studies show that hypoadiponectinemia is associated with increased IL-6 levels [18, 87, 89]. However, no evidence suggests an association between plasma adiponectin and TNF-α levels in humans.

4. Regulation of adiponectin

As mentioned above, obesity promotes hypoadiponectinemia. It is believed that chronic inflammation associated with excess adiposity is a key feature of adiponectin downregulation under these conditions. Pro-inflammatory cytokines suppress adiponectin expression in adipocytes. TNF-α treatment suppresses adiponectin expression at the level of transcription in cultured 3T3-L1 adipocytes [94] and reduces the expression and secretion of adiponectin protein in primary human adipocytes [95]. Adiponectin mRNA expression and protein secretion are also inhibited by treatment with IL-6 in 3T3-L1 adipocytes [96]. In epidemiological studies, a positive correlation is found between plasma levels of TNF-α and IL-6 and BMI [85], whereas a negative correlation exists between BMI and adiponectin levels [7]. Conversely, weight reduction correlates with decrease in levels of TNF-α and IL-6 in plasma [97]. Weight loss also results in a significant increase in circulating adiponectin levels [98].

Treatment with insulin-sensitizing agents, thiazolidinediones (TZDs), that activate peroxisome proliferator-activated receptor-gamma (PPAR-gamma) are reported to increase plasma adiponectin levels in humans [94, 99-101]. TZDs treatment increases adiponectin expression and secretion in 3T3-L1 adipocytes as well as circulating adiponectin in obese mice [94]. Recent experimental studies with obese mice lacking adiponectin showed that the beneficial actions of TZDs on insulin resistance are partly dependent on adiponectin [52, 102]. TZDs are believed to exhibit anti-inflammatory and anti-atherogenic properties [103]. TZDs treatment is found to decrease plasma CRP and TNF-α levels in humans [104, 105]. It is conceivable that anti-inflammatory action of TZDs may be mediated by their ability to upregulate adiponectin, but a direct causal link has yet to be established.

Finally, a recent report has shown that adiponectin is expressed in human hearts, and that adiponectin transcripts are downregulated in dilated cardiomyopathy [106]. Thus, dysregulation of adiponectin synthesized in myocardium may contribute to the development of obesity-linked heart diseases.

Conclusion

Adiponectin can function as a modulator of multiple obesity-linked diseases by attenuating excessive inflammatory responses in a variety of tissues. While putative adiponectin receptors are expressed in various cells and tissues where adiponectin exerts anti-inflammatory actions, their involvement in adiponectin-mediated suppression of cellular inflammatory responses has not been definitely established. In contrast, the receptor-mediated signaling events that control metabolic processes downstream from adiponectin are much better understood. Epidemiological studies demonstrate that adiponectin negatively correlates with obesity-related inflammatory disease states, but the relationship of adiponectin to some disease conditions remains controversial. Thus, further clinical and experimental studies are required to clarify the molecular mechanisms that participate in the anti-inflammatory actions of adiponectin.

Acknowledgments

This work was supported by NIH grants HL66957, HL77774, AR40197 and AG15052 to K. Walsh. N. Ouchi was supported by an American Heart Association Scientist Development Grant, Northeast Affiliate.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Wellen KE, Hotamisligil GS. Obesity-induced inflammatory changes in adipose tissue. J Clin Invest. 2003;112:1785–8. doi: 10.1172/JCI20514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ouchi N, Kihara S, Funahashi T, Matsuzawa Y, Walsh K. Obesity, adiponectin and vascular inflammatory disease. Curr Opin Lipidol. 2003;14:561–566. doi: 10.1097/00041433-200312000-00003. [DOI] [PubMed] [Google Scholar]
  • 3.Berg AH, Scherer PE. Adipose tissue, inflammation, and cardiovascular disease. Circ Res. 2005;96:939–949. doi: 10.1161/01.RES.0000163635.62927.34. [DOI] [PubMed] [Google Scholar]
  • 4.Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem. 1996;271:10697–10703. doi: 10.1074/jbc.271.18.10697. [DOI] [PubMed] [Google Scholar]
  • 5.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;221:286–289. doi: 10.1006/bbrc.1996.0587. [DOI] [PubMed] [Google Scholar]
  • 6.Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem. 1995;270:26746–26749. doi: 10.1074/jbc.270.45.26746. [DOI] [PubMed] [Google Scholar]
  • 7.Arita Y, Kihara S, Ouchi N, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun. 1999;257:79–83. doi: 10.1006/bbrc.1999.0255. [DOI] [PubMed] [Google Scholar]
  • 8.Lusis AJ. Atherosclerosis. Nature. 2000;407:233–41. doi: 10.1038/35025203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ouchi N, Kihara S, Arita Y, et al. Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation. 1999;100:2473–2476. doi: 10.1161/01.cir.100.25.2473. [DOI] [PubMed] [Google Scholar]
  • 10.Kobashi C, Urakaze M, Kishida M, et al. Adiponectin inhibits endothelial synthesis of interleukin-8. Circ Res. 2005;97:1245–52. doi: 10.1161/01.RES.0000194328.57164.36. [DOI] [PubMed] [Google Scholar]
  • 11.Ouchi N, Kihara S, Arita Y, et al. Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation. 2000;102:1296–1301. doi: 10.1161/01.cir.102.11.1296. [DOI] [PubMed] [Google Scholar]
  • 12.Ouedraogo R, Wu X, Xu SQ, et al. Adiponectin suppression of high-glucose-induced reactive oxygen species in vascular endothelial cells: evidence for involvement of a cAMP signaling pathway. Diabetes. 2006;55:1840–6. doi: 10.2337/db05-1174. [DOI] [PubMed] [Google Scholar]
  • 13.Ouchi N, Kihara S, Arita Y, et al. Adipocyte-derived plasma protein, adiponectin, suppresses lipid accumulation and class A scavenger receptor expression in human monocyte-derived macrophages. Circulation. 2001;103:1057–1063. doi: 10.1161/01.cir.103.8.1057. [DOI] [PubMed] [Google Scholar]
  • 14.Yokota T, Oritani K, Takahashi I, et al. 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;96:1723–1732. [PubMed] [Google Scholar]
  • 15.Wulster-Radcliffe MC, Ajuwon KM, Wang J, Christian JA, Spurlock ME. Adiponectin differentially regulates cytokines in porcine macrophages. Biochem Biophys Res Commun. 2004;316:924–9. doi: 10.1016/j.bbrc.2004.02.130. [DOI] [PubMed] [Google Scholar]
  • 16.Yamaguchi N, Argueta JG, Masuhiro Y, et al. Adiponectin inhibits Toll-like receptor family-induced signaling. FEBS Lett. 2005;579:6821–6. doi: 10.1016/j.febslet.2005.11.019. [DOI] [PubMed] [Google Scholar]
  • 17.Kumada M, Kihara S, Ouchi N, et al. Adiponectin specifically increased tissue inhibitor of metalloproteinase-1 through interleukin-10 expression in human macrophages. Circulation. 2004;109:2046–9. doi: 10.1161/01.CIR.0000127953.98131.ED. [DOI] [PubMed] [Google Scholar]
  • 18.Choi KM, Ryu OH, Lee KW, et al. Serum adiponectin, interleukin-10 levels and inflammatory markers in the metabolic syndrome. Diabetes Res Clin Pract. 2006 doi: 10.1016/j.diabres.2006.06.019. [DOI] [PubMed] [Google Scholar]
  • 19.Okamoto Y, Kihara S, Ouchi N, et al. Adiponectin reduces atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2002;106:2767–2770. doi: 10.1161/01.cir.0000042707.50032.19. [DOI] [PubMed] [Google Scholar]
  • 20.Yamauchi T, Kamon J, Waki H, et al. Globular adiponectin protected ob/ob mice from diabetes and ApoE-deficient mice from atherosclerosis. J Biol Chem. 2003;278:2461–2468. doi: 10.1074/jbc.M209033200. [DOI] [PubMed] [Google Scholar]
  • 21.Waltenberger J. Impaired collateral vessel development in diabetes: potential cellular mechanisms and therapeutic implications. Cardiovasc Res. 2001;49:554–560. doi: 10.1016/s0008-6363(00)00228-5. [DOI] [PubMed] [Google Scholar]
  • 22.Warley A, Powell JM, Skepper JN. Capillary surface area is reduced and tissue thickness from capillaries to myocytes is increased in the left ventricle of streptozotocin-diabetic rats. Diabetologia. 1995;38:413–421. doi: 10.1007/BF00410278. [DOI] [PubMed] [Google Scholar]
  • 23.Noon JP, Walker BR, Webb DJ, et al. Impaired microvascular dilatation and capillary rarefaction in young adults with a predisposition to high blood pressure. J Clin Invest. 1997;99:1873–1879. doi: 10.1172/JCI119354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yilmaz MB, Biyikoglu SF, Akin Y, Guray U, Kisacik HL, Korkmaz S. Obesity is associated with impaired coronary collateral vessel development. Int J Obes Relat Metab Disord. 2003;27:1541–1545. doi: 10.1038/sj.ijo.0802474. [DOI] [PubMed] [Google Scholar]
  • 25.Lind L, Lithell H. Decreased peripheral blood flow in the pathogenesis of the metabolic syndrome comprising hypertension, hyperlipidemia, and hyperinsulinemia. Am Heart J. 1993;125:1494–1497. doi: 10.1016/0002-8703(93)90446-g. [DOI] [PubMed] [Google Scholar]
  • 26.Steinberg HO, Chaker H, Leaming R, Johnson A, Brechtel G, Baron AD. Obesity/insulin resistance is associated with endothelial dysfunction. Implications for the syndrome of insulin resistance. J Clin Invest. 1996;97:2601–2610. doi: 10.1172/JCI118709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Al Suwaidi J, Higano ST, Holmes DR, Jr, Lennon R, Lerman A. Obesity is independently associated with coronary endothelial dysfunction in patients with normal or mildly diseased coronary arteries. J Am Coll Cardiol. 2001;37:1523–1528. doi: 10.1016/s0735-1097(01)01212-8. [DOI] [PubMed] [Google Scholar]
  • 28.Ouchi N, Kobayashi H, Kihara S, et al. Adiponectin stimulates angiogenesis by promoting cross-talk between AMP-activated protein kinase and Akt signaling in endothelial cells. J Biol Chem. 2004;279:1304–1309. doi: 10.1074/jbc.M310389200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ. Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem. 2003;278:45021–45026. doi: 10.1074/jbc.M307878200. [DOI] [PubMed] [Google Scholar]
  • 30.Hattori Y, Suzuki M, Hattori S, Kasai K. Globular adiponectin upregulates nitric oxide production in vascular endothelial cells. Diabetologia. 2003;46:1543–1549. doi: 10.1007/s00125-003-1224-3. [DOI] [PubMed] [Google Scholar]
  • 31.Motoshima H, Wu X, Mahadev K, Goldstein BJ. Adiponectin suppresses proliferation and superoxide generation and enhances eNOS activity in endothelial cells treated with oxidized LDL. Biochem Biophys Res Commun. 2004;315:264–71. doi: 10.1016/j.bbrc.2004.01.049. [DOI] [PubMed] [Google Scholar]
  • 32.Xi W, Satoh H, Kase H, Suzuki K, Hattori Y. Stimulated HSP90 binding to eNOS and activation of the PI3-Akt pathway contribute to globular adiponectin-induced NO production: vasorelaxation in response to globular adiponectin. Biochem Biophys Res Commun. 2005;332:200–5. doi: 10.1016/j.bbrc.2005.04.111. [DOI] [PubMed] [Google Scholar]
  • 33.Moncada S, Palmer RMJ, Higgs EA. Nitric oxide: physiology, pathophysiology and pharmacology. Pharmacol Rev. 1991;43:109–142. [PubMed] [Google Scholar]
  • 34.Ohashi K, Kihara S, Ouchi N, et al. Adiponectin Replenishment Ameliorates Obesity-Related Hypertension. Hypertension. 2006 doi: 10.1161/01.HYP.0000222368.43759.a1. [DOI] [PubMed] [Google Scholar]
  • 35.Ouchi N, Ohishi M, Kihara S, et al. Association of hypoadiponectinemia with impaired vasoreactivity. Hypertension. 2003;42:231–234. doi: 10.1161/01.HYP.0000083488.67550.B8. [DOI] [PubMed] [Google Scholar]
  • 36.Shibata R, Ouchi N, Kihara S, Sato K, Funahashi T, Walsh K. Adiponectin stimulates angiogenesis in response to tissue ischemia through stimulation of amp-activated protein kinase signaling. J Biol Chem. 2004;279:28670–28674. doi: 10.1074/jbc.M402558200. [DOI] [PubMed] [Google Scholar]
  • 37.Kobayashi H, Ouchi N, Kihara S, et al. Selective suppression of endothelial cell apoptosis by the high molecular weight form of adiponectin. Circ Res. 2004;94:e27–e31. doi: 10.1161/01.RES.0000119921.86460.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lin LY, Lin CY, Su TC, Liau CS. Angiotensin II-induced apoptosis in human endothelial cells is inhibited by adiponectin through restoration of the association between endothelial nitric oxide synthase and heat shock protein 90. FEBS Lett. 2004;574:106–110. doi: 10.1016/j.febslet.2004.08.012. [DOI] [PubMed] [Google Scholar]
  • 39.Shibata R, Sato K, Pimentel DR, et al. Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nat Med. 2005;11:1096–1103. doi: 10.1038/nm1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kojima S, Funahashi T, Sakamoto T, et al. The variation of plasma concentrations of a novel, adipocyte derived protein, adiponectin, in patients with acute myocardial infarction. Heart. 2003;89:667. doi: 10.1136/heart.89.6.667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bolli R, Shinmura K, Tang XL, et al. Discovery of a new function of cyclooxygenase (COX)-2: COX-2 is a cardioprotective protein that alleviates ischemia/reperfusion injury and mediates the late phase of preconditioning. Cardiovasc Res. 2002;55:506–519. doi: 10.1016/s0008-6363(02)00414-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Camitta MG, Gabel SA, Chulada P, et al. Cyclooxygenase-1 and -2 knockout mice demonstrate increased cardiac ischemia/reperfusion injury but are protected by acute preconditioning. Circulation. 2001;104:2453–2458. doi: 10.1161/hc4401.098429. [DOI] [PubMed] [Google Scholar]
  • 43.Xiao CY, Yuhki K, Hara A, et al. Prostaglandin E2 protects the heart from ischemia-reperfusion injury via its receptor subtype EP4. Circulation. 2004;109:2462–2468. doi: 10.1161/01.CIR.0000128046.54681.97. [DOI] [PubMed] [Google Scholar]
  • 44.Hohlfeld T, Meyer-Kirchrath J, Vogel YC, Schror K. Reduction of infarct size by selective stimulation of prostaglandin EP(3)receptors in the reperfused ischemic pig heart. J Mol Cell Cardiol. 2000;32:285–296. doi: 10.1006/jmcc.1999.1072. [DOI] [PubMed] [Google Scholar]
  • 45.Ilercil A, Devereux RB, Roman MJ, et al. Relationship of impaired glucose tolerance to left ventricular structure and function: The Strong Heart Study. Am Heart J. 2001;141:992–998. doi: 10.1067/mhj.2001.115302. [DOI] [PubMed] [Google Scholar]
  • 46.Rutter MK, Parise H, Benjamin EJ, et al. Impact of glucose intolerance and insulin resistance on cardiac structure and function: sex-related differences in the Framingham Heart Study. Circulation. 2003;107:448–454. doi: 10.1161/01.cir.0000045671.62860.98. [DOI] [PubMed] [Google Scholar]
  • 47.Shibata R, Ouchi N, Ito M, et al. Adiponectin-mediated modulation of hypertrophic signals in the heart. Nat Med. 2004;10:1384–1389. doi: 10.1038/nm1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Liao Y, Takashima S, Maeda N, et al. Exacerbation of heart failure in adiponectin-deficient mice due to impaired regulation of AMPK and glucose metabolism. Cardiovasc Res. 2005;67:705–713. doi: 10.1016/j.cardiores.2005.04.018. [DOI] [PubMed] [Google Scholar]
  • 49.Fujioka D, Kawabata K, Saito Y, et al. Role of adiponectin receptors in endothelin-induced cellular hypertrophy in cultured cardiomyocytes and their expression in infarcted heart. Am J Physiol Heart Circ Physiol. 2006;290:H2409–H2416. doi: 10.1152/ajpheart.00987.2005. [DOI] [PubMed] [Google Scholar]
  • 50.Tian R, Musi N, D'Agostino J, Hirshman MF, Goodyear LJ. Increased adenosine monophosphate-activated protein kinase activity in rat hearts with pressure-overload hypertrophy. Circulation. 2001;104:1664–1669. doi: 10.1161/hc4001.097183. [DOI] [PubMed] [Google Scholar]
  • 51.Maeda N, Shimomura I, Kishida K, et al. Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat Med. 2002;8:731–737. doi: 10.1038/nm724. [DOI] [PubMed] [Google Scholar]
  • 52.Nawrocki AR, Rajala MW, Tomas E, et al. Mice lacking adiponectin show decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome proliferator-activated receptor gamma agonists. J Biol Chem. 2006;281:2654–2660. doi: 10.1074/jbc.M505311200. [DOI] [PubMed] [Google Scholar]
  • 53.Kubota N, Terauchi Y, Yamauchi T, et al. Disruption of adiponectin causes insulin resistance and neointimal formation. J Biol Chem. 2002;277:25863–25866. doi: 10.1074/jbc.C200251200. [DOI] [PubMed] [Google Scholar]
  • 54.Ma K, Cabrero A, Saha PK, et al. Increased beta -oxidation but no insulin resistance or glucose intolerance in mice lacking adiponectin. J Biol Chem. 2002;277:34658–61. doi: 10.1074/jbc.C200362200. [DOI] [PubMed] [Google Scholar]
  • 55.Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med. 2001;7:947–953. doi: 10.1038/90992. [DOI] [PubMed] [Google Scholar]
  • 56.Fruebis J, Tsao TS, Javorschi S, et al. 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 USA. 2001;98:2005–2010. doi: 10.1073/pnas.041591798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tomas E, Tsao TS, Saha AK, et al. Enhanced muscle fat oxidation and glucose transport by ACRP30 globular domain: acetyl-CoA carboxylase inhibition and AMP-activated protein kinase activation. Proc Natl Acad Sci USA. 2002;99:16309–16313. doi: 10.1073/pnas.222657499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med. 2002;8:1288–1295. doi: 10.1038/nm788. [DOI] [PubMed] [Google Scholar]
  • 59.Wu X, Motoshima H, Mahadev K, Stalker TJ, Scalia R, Goldstein BJ. Involvement of AMP-activated protein kinase in glucose uptake stimulated by the globular domain of adiponectin in primary rat adipocytes. Diabetes. 2003;52:1355–1363. doi: 10.2337/diabetes.52.6.1355. [DOI] [PubMed] [Google Scholar]
  • 60.Satoh H, Nguyen MT, Trujillo M, et al. Adenovirus-mediated adiponectin expression augments skeletal muscle insulin sensitivity in male Wistar rats. Diabetes. 2005;54:1304–1313. doi: 10.2337/diabetes.54.5.1304. [DOI] [PubMed] [Google Scholar]
  • 61.Combs TP, Pajvani UB, Berg AH, et al. A transgenic mouse with a deletion in the collagenous domain of adiponectin displays elevated circulating adiponectin and improved insulin sensitivity. Endocrinology. 2004;145:367–383. doi: 10.1210/en.2003-1068. [DOI] [PubMed] [Google Scholar]
  • 62.Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 2003;423:762–769. doi: 10.1038/nature01705. [DOI] [PubMed] [Google Scholar]
  • 63.Ryo M, Nakamura T, Kihara S, et al. Adiponectin as a biomarker of the metabolic syndrome. Circ J. 2004;68:975–81. doi: 10.1253/circj.68.975. [DOI] [PubMed] [Google Scholar]
  • 64.Hotta K, Funahashi T, Arita Y, et al. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol. 2000;20:1595–1599. doi: 10.1161/01.atv.20.6.1595. [DOI] [PubMed] [Google Scholar]
  • 65.Lindsay RS, Funahashi T, Hanson RL, et al. Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet. 2002;360:57–8. doi: 10.1016/S0140-6736(02)09335-2. [DOI] [PubMed] [Google Scholar]
  • 66.Spranger J, Kroke A, Mohlig M, et al. Adiponectin and protection against type 2 diabetes mellitus. Lancet. 2003;361:226–228. doi: 10.1016/S0140-6736(03)12255-6. [DOI] [PubMed] [Google Scholar]
  • 67.Snehalatha C, Mukesh B, Simon M, Viswanathan V, Haffner SM, Ramachandran A. Plasma adiponectin is an independent predictor of type 2 diabetes in Asian indians. Diabetes Care. 2003;26:3226–3229. doi: 10.2337/diacare.26.12.3226. [DOI] [PubMed] [Google Scholar]
  • 68.Choi KM, Lee J, Lee KW, et al. Serum adiponectin concentrations predict the developments of type 2 diabetes and the metabolic syndrome in elderly Koreans. Clin Endocrinol (Oxf) 2004;61:75–80. doi: 10.1111/j.1365-2265.2004.02063.x. [DOI] [PubMed] [Google Scholar]
  • 69.Yamamoto Y, Hirose H, Saito I, Nishikai K, Saruta T. Adiponectin, an adipocyte-derived protein, predicts future insulin resistance: two-year follow-up study in Japanese population. J Clin Endocrinol Metab. 2004;89:87–90. doi: 10.1210/jc.2003-031163. [DOI] [PubMed] [Google Scholar]
  • 70.Shetty GK, Economides PA, Horton ES, Mantzoros CS, Veves A. 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. 2004;27:2450–7. doi: 10.2337/diacare.27.10.2450. [DOI] [PubMed] [Google Scholar]
  • 71.Adamczak M, Wiecek A, Funahashi T, Chudek J, Kokot F, Matsuzawa Y. Decreased plasma adiponectin concentration in patients with essential hypertension. Am J Hypertens. 2003;16:72–75. doi: 10.1016/s0895-7061(02)03197-7. [DOI] [PubMed] [Google Scholar]
  • 72.Iwashima Y, Katsuya T, Ishikawa K, et al. Hypoadiponectinemia is an independent risk factor for hypertension. Hypertension. 2004;43:1318–1323. doi: 10.1161/01.HYP.0000129281.03801.4b. [DOI] [PubMed] [Google Scholar]
  • 73.Kumada M, Kihara S, Sumitsuji S, et al. Association of hypoadiponectinemia with coronary artery disease in men. Arterioscler Thromb Vasc Biol. 2003;23:85–89. doi: 10.1161/01.atv.0000048856.22331.50. [DOI] [PubMed] [Google Scholar]
  • 74.Nakamura Y, Shimada K, Fukuda D, et al. Implications of plasma concentrations of adiponectin in patients with coronary artery disease. Heart. 2004;90:528–533. doi: 10.1136/hrt.2003.011114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.von Eynatten M, Schneider JG, Humpert PM, et al. Serum adiponectin levels are an independent predictor of the extent of coronary artery disease in men. J Am Coll Cardiol. 2006;47:2124–6. doi: 10.1016/j.jacc.2006.02.033. [DOI] [PubMed] [Google Scholar]
  • 76.Zoccali C, Mallamaci F, Tripepi G, et al. Adiponectin, metabolic risk factors, and cardiovascular events among patients with end-stage renal disease. J Am Soc Nephrol. 2002;13:134–141. doi: 10.1681/ASN.V131134. [DOI] [PubMed] [Google Scholar]
  • 77.Pischon T, Girman CJ, Hotamisligil GS, Rifai N, Hu FB, Rimm EB. Plasma adiponectin levels and risk of myocardial infarction in men. Jama. 2004;291:1730–1737. doi: 10.1001/jama.291.14.1730. [DOI] [PubMed] [Google Scholar]
  • 78.Schulze MB, Shai I, Rimm EB, Li T, Rifai N, Hu FB. Adiponectin and future coronary heart disease events among men with type 2 diabetes. Diabetes. 2005;54:534–539. doi: 10.2337/diabetes.54.2.534. [DOI] [PubMed] [Google Scholar]
  • 79.Lindsay RS, Resnick HE, Zhu J, et al. Adiponectin and coronary heart disease: the Strong Heart Study. Arterioscler Thromb Vasc Biol. 2005;25:e15–e16. doi: 10.1161/01.ATV.0000153090.21990.8c. [DOI] [PubMed] [Google Scholar]
  • 80.Lawlor DA, Davey Smith G, Ebrahim S, Thompson C, Sattar N. Plasma adiponectin levels are associated with insulin resistance, but do not predict future risk of coronary heart disease in women. J Clin Endocrinol Metab. 2005;90:5677–5683. doi: 10.1210/jc.2005-0825. [DOI] [PubMed] [Google Scholar]
  • 81.Sattar N, Wannamethee G, Sarwar N, et al. Adiponectin and coronary heart disease: a prospective study and meta-analysis. Circulation. 2006 doi: 10.1161/CIRCULATIONAHA.106.618918. In press. [DOI] [PubMed] [Google Scholar]
  • 82.Ridker PM. High-sensitivity C-reactive protein and cardiovascular risk: rationale for screening and primary prevention. Am J Cardiol. 2003;92:17K–22K. doi: 10.1016/s0002-9149(03)00774-4. [DOI] [PubMed] [Google Scholar]
  • 83.Laaksonen DE, Niskanen L, Nyyssonen K, et al. C-reactive protein and the development of the metabolic syndrome and diabetes in middle-aged men. Diabetologia. 2004;47:1403–10. doi: 10.1007/s00125-004-1472-x. [DOI] [PubMed] [Google Scholar]
  • 84.Mendall MA, Patel P, Ballam L, Strachan D, Northfield TC. C reactive protein and its relation to cardiovascular risk factors: a population based cross sectional study. Bmj. 1996;312:1061–5. doi: 10.1136/bmj.312.7038.1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yudkin JS, Stehouwer CD, Emeis JJ, Coppack SW. C-reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose tissue? Arterioscler Thromb Vasc Biol. 1999;19:972–8. doi: 10.1161/01.atv.19.4.972. [DOI] [PubMed] [Google Scholar]
  • 86.Ouchi N, Kihara S, Funahashi T, et al. Reciprocal association of C-reactive protein with adiponectin in blood stream and adipose tissue. Circulation. 2003;107:671–674. doi: 10.1161/01.cir.0000055188.83694.b3. [DOI] [PubMed] [Google Scholar]
  • 87.Engeli S, Feldpausch M, Gorzelniak K, et al. Association between adiponectin and mediators of inflammation in obese women. Diabetes. 2003;52:942–947. doi: 10.2337/diabetes.52.4.942. [DOI] [PubMed] [Google Scholar]
  • 88.Mantzoros CS, Li T, Manson JE, Meigs JB, Hu FB. Circulating adiponectin levels are associated with better glycemic control, more favorable lipid profile, and reduced inflammation in women with type 2 diabetes. J Clin Endocrinol Metab. 2005;90:4542–8. doi: 10.1210/jc.2005-0372. [DOI] [PubMed] [Google Scholar]
  • 89.Krakoff J, Funahashi T, Stehouwer CD, et al. Inflammatory markers, adiponectin, and risk of type 2 diabetes in the Pima Indian. Diabetes Care. 2003;26:1745–1751. doi: 10.2337/diacare.26.6.1745. [DOI] [PubMed] [Google Scholar]
  • 90.Matsubara M, Namioka K, Katayose S. Decreased plasma adiponectin concentrations in women with low-grade C-reactive protein elevation. Eur J Endocrinol. 2003;148:657–662. doi: 10.1530/eje.0.1480657. [DOI] [PubMed] [Google Scholar]
  • 91.Matsushita K, Yatsuya H, Tamakoshi K, et al. Inverse association between adiponectin and C-reactive protein in substantially healthy Japanese men. Atherosclerosis. 2005 doi: 10.1016/j.atherosclerosis.2005.10.031. [DOI] [PubMed] [Google Scholar]
  • 92.von Eynatten M, Hamann A, Twardella D, Nawroth PP, Brenner H, Rothenbacher D. Relationship of adiponectin with markers of systemic inflammation, atherogenic dyslipidemia, and heart failure in patients with coronary heart disease. Clin Chem. 2006;52:853–9. doi: 10.1373/clinchem.2005.060509. [DOI] [PubMed] [Google Scholar]
  • 93.Blake GJ, Ridker PM. Novel clinical markers of vascular wall inflammation. Circ Res. 2001;89:763–771. doi: 10.1161/hh2101.099270. [DOI] [PubMed] [Google Scholar]
  • 94.Maeda N, Takahashi M, Funahashi T, et al. PPARgamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein. Diabetes. 2001;50:2094–9. doi: 10.2337/diabetes.50.9.2094. [DOI] [PubMed] [Google Scholar]
  • 95.Kappes A, Loffler G. Influences of ionomycin, dibutyryl-cycloAMP and tumour necrosis factor-alpha on intracellular amount and secretion of apM1 in differentiating primary human preadipocytes. Horm Metab Res. 2000;32:548–54. doi: 10.1055/s-2007-978684. [DOI] [PubMed] [Google Scholar]
  • 96.Fasshauer M, Kralisch S, Klier M, et al. Adiponectin gene expression and secretion is inhibited by interleukin-6 in 3T3-L1 adipocytes. Biochem Biophys Res Commun. 2003;301:1045–50. doi: 10.1016/s0006-291x(03)00090-1. [DOI] [PubMed] [Google Scholar]
  • 97.Ziccardi P, Nappo F, Giugliano G, et al. Reduction of inflammatory cytokine concentrations and improvement of endothelial functions in obese women after weight loss over one year. Circulation. 2002;105:804–9. doi: 10.1161/hc0702.104279. [DOI] [PubMed] [Google Scholar]
  • 98.Yang WS, Lee WJ, Funahashi T, et al. Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab. 2001;86:3815–3819. doi: 10.1210/jcem.86.8.7741. [DOI] [PubMed] [Google Scholar]
  • 99.Yu JG, Javorschi S, Hevener AL, et al. The effect of thiazolidinediones on plasma adiponectin levels in normal, obese, and type 2 diabetic subjects. Diabetes. 2002;51:2968–74. doi: 10.2337/diabetes.51.10.2968. [DOI] [PubMed] [Google Scholar]
  • 100.Yang WS, Jeng CY, Wu TJ, et al. Synthetic peroxisome proliferator-activated receptor-gamma agonist, rosiglitazone, increases plasma levels of adiponectin in type 2 diabetic patients. Diabetes Care. 2002;25:376–80. doi: 10.2337/diacare.25.2.376. [DOI] [PubMed] [Google Scholar]
  • 101.Combs TP, Wagner JA, Berger J, et al. Induction of adipocyte complement-related protein of 30 kilodaltons by PPARgamma agonists: a potential mechanism of insulin sensitization. Endocrinology. 2002;143:998–1007. doi: 10.1210/endo.143.3.8662. [DOI] [PubMed] [Google Scholar]
  • 102.Kubota N, Terauchi Y, Kubota T, et al. Pioglitazone ameliorates insulin resistance and diabetes by both adiponectin-dependent and -independent pathways. J Biol Chem. 2006;281:8748–55. doi: 10.1074/jbc.M505649200. [DOI] [PubMed] [Google Scholar]
  • 103.Marx N, Imhof A, Froehlich J, et al. Effect of rosiglitazone treatment on soluble CD40L in patients with type 2 diabetes and coronary artery disease. Circulation. 2003;107:1954–7. doi: 10.1161/01.CIR.0000069272.06194.91. [DOI] [PubMed] [Google Scholar]
  • 104.Haffner SM, Greenberg AS, Weston WM, Chen H, Williams K, Freed MI. Effect of rosiglitazone treatment on nontraditional markers of cardiovascular disease in patients with type 2 diabetes mellitus. Circulation. 2002;106:679–84. doi: 10.1161/01.cir.0000025403.20953.23. [DOI] [PubMed] [Google Scholar]
  • 105.Katsuki A, Sumida Y, Murata K, et al. Troglitazone reduces plasma levels of tumour necrosis factor-alpha in obese patients with type 2 diabetes. Diabetes Obes Metab. 2000;2:189–91. doi: 10.1046/j.1463-1326.2000.00072.x. [DOI] [PubMed] [Google Scholar]
  • 106.Wittchen F, Suckau L, Witt H, et al. Genomic expression profiling of human inflammatory cardiomyopathy (DCMi) suggests novel therapeutic targets. J Mol Med. 2006 doi: 10.1007/s00109-006-0122-9. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES