Review Open Access
Copyright ©2012 Baishideng. All rights reserved.
World J Hypertens. Feb 23, 2012; 2(1): 13-21
Published online Feb 23, 2012. doi: 10.5494/wjh.v2.i1.13
Adipose tissue in the pathophysiology of cardiovascular disease: Who is guilty?
Plinio Cirillo, Fabio Maresca, Vito Di Palma, Francesca Ziviello, Michele Bevilacqua, Department of Internal Medicine, Cardiovascular and Immunological Sciences (Division of Cardiology), University of Naples “Federico II”, via Sergio Pansini, 5, 80131 Naples, Italy
Author contributions: Maresca F and Di Palma V wrote the first draft; Ziviello F and Bevilacqua M checked the appropriate references; Cirillo P revised the final version and gave the final approval.
Correspondence to: Plinio Cirillo, MD, PhD, Assistant Professor, Department of Internal Medicine, Cardiovascular and Immunological Sciences (Division of Cardiology), University of Naples “Federico II”, via Sergio Pansini, 5, 80131 Naples, Italy. pcirillo@unina.it
Telephone: +39-81-7462235 Fax: +39-81-7462235
Received: July 28, 2011
Revised: December 16, 2011
Accepted: January 12, 2012
Published online: February 23, 2012

Abstract

Epidemiological evidence has shown how abdominal obesity is closely associated with the development of cardiovascular disease. It has been demonstrated that patients with extensive adipose tissue usually have other concomitant cardiovascular risk factors, such insulin resistance, hypertension and dyslipidemia. Moreover, obese patients have a significantly higher risk of developing thrombophilic events compared with the non-obese. Thus, obesity is actually considered an independent cardiovascular risk factor. The pathophysiological mechanisms responsible for the association between obesity and cardiovascular disease remain largely unknown. However, it has been postulated that obese patients have an “inflammatory milieu” responsible for their metabolic disorders and vascular disease. In this context, adipocyte-derived molecules with inflammatory activity might play a pivotal role in the development of these mechanisms. In the present report, we provide an updated overview on the molecules produced by adipose tissue that are potentially involved in cardiovascular pathophysiology.

Key Words: Adipokines, Atherosclerosis, Cardiovascular disease, Inflammation, Obesity, Thrombosis



INTRODUCTION

Nowadays, obesity is considered an emerging and rapidly expanding disease, mainly in industrialized countries. In fact, it can be considered a typical “disease of the affluent”[1]. The World Health Organization estimates that more than 1 billion people are overweight and 300 million are obese in the world[2]. For many years, epidemiological evidence has shown that abdominal obesity is closely associated with the development of cardiovascular disease[3,4]. Specifically, patients with extensive adipose tissue have a higher incidence of other cardiovascular risk factors, such as insulin resistance, hypertension and dyslipidemia[5]. Interestingly, obese patients have a thrombophilic risk 1.5 to 2.5 times higher than the non-obese. Taken together, these observations have suggested that obesity might be considered as an independent cardiovascular risk factor[6]. Despite these clinical observations, the pathophysiological mechanisms responsible for the association between obesity and cardiovascular disease remain largely unknown. However, it has been recently postulated that obese patients have an “inflammatory milieu” responsible for their metabolic disorders and vascular disease[7]. In this context, adipocyte-derived molecules with inflammatory activity might play a pivotal role in the development of these inflammatory mechanisms. In the present report, we provide an updated overview of the molecules produced by adipose tissue potentially involved in cardiovascular pathophysiology.

ADIPO(CYTO)KINES

The cells of adipose tissue, adipocytes, are no longer considered only as fat storage cells. They are considered as cells able to produce and secrete several substances with biological activity, known as “adipokines”[8,9]. The biological functions of adipokines are still partially unknown; however, they seem involved in the regulation of many physiological processes, such as appetite regulation and energy balance, lipid metabolism, blood pressure, insulin sensitivity, inflammation, haemostasis and angiogenesis[10]. It is known that, in the plasma of patients with obesity or metabolic syndrome, increased levels of some adipokines can be measured, suggesting that these adipocyte-derived substances might be considered as novel biomarkers and regulators of the metabolic syndrome[11].

It has been observed that adipocytes belonging to the adipose tissue of obese patients can synthesize and secrete several adipokines. Moreover, this tissue appears to be infiltrated by inflammatory cells[7]. Interestingly, adipokines secreted by visceral fat have a more remarkable biological activity than adipokines released by subcutaneous fat[7]. It was also shown that weight loss and exercise could ameliorate the inflammatory milieu of patients with metabolic syndrome by modulating their adipokine profile[11,12]. To date, the adipokines actually discovered can be divided into four groups: (1) adipokines with metabolic functions; (2) adipokines with pro-inflammatory functions; (3) adipokines components of the extracellular matrix; and (4) adipokines with pro- angiogenic and pro-mitogenic action[11]. However, some of them fall out of this schematization and can be placed transversely across multiple categories. In addition, only some of these molecules appear to play an active role in cardiovascular pathophysiology.

ADIPONECTIN

Adiponectin is a 247 amino acid protein with a globular carboxyl-terminal domain and an amino-terminal collagen-like domain[13]. This adipocytokine and complement factor 1q has a similar structure[14]. In humans, the adiponectin gene is located on chromosome 3q27[15]. This adipokine seems to exert protective effects on the cardiovascular system[16]. In fact, patients with a high atherosclerotic burden have low plasma levels of adiponectin[17]. Moreover, it has been demonstrated that low plasma levels of this adipokine are closely related to the progression of coronary atherosclerosis in patients with angina pectoris[18]. Finally, it has been observed that women with low plasma levels of adiponectin have impairment of the coronary flow reserve[19]. Although adipocytes are the main source of adiponectin, patients in which adipose tissue are largely represented, such as the obese and those affected by metabolic syndrome or diabetes mellitus, have low measurable plasma levels of this adipocytokine[20]. In the plasma, three different oligomers of adiponectin have been isolated, each one with a specific biological function[21]. We can identify: (1) low molecular weight (LMW) oligomers, constructed by three molecules of adiponectin; (2) middle molecular weight (MMW) oligomers, formed by six adiponectin fractions; and (3) high molecular weight (HMW) oligomers constituted by 12 -18 molecules of adiponectin[22]. Then, another oligomer has been recently isolated in which three molecules of adiponectin are bound to albumin (Alb-LMW)[23]. In humans, MMW and LMW adiponectin represent 25% while HMW adiponectin represents 50% of whole circulating adiponectin[23]. Since plasma levels of HMW appear to be closely related to insulin sensitivity, it has been suggested that HMW is biologically active[24,25]. Two specific adiponectin receptors have been identified: AdipoR1 and AdipoR2. Binding of adiponectin to the AdipoR2 receptor increases energy consumption and improves fatty acid oxidation. Moreover, when adiponectin binds the AdipoR2 receptor, pro-atherosclerotic processes such as oxidative stress and inflammation are significantly inhibited[26]. In particular, in the atherosclerotic plaques, adiponectin should modulate the inflammatory response by down-regulating the expression of pro-inflammatory mediators, such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and interferon (IFN)-c, and by up-regulating anti-inflammatory molecules, such as the antagonist receptor for IL-1[27]. As reported above, in vivo, adiponectin increases energy consumption and oxidation of fatty acids in the liver and muscles. These phenomena contribute to reducing triglycerides levels in these tissues and improving insulin sensitivity[28]. In endothelial cells, adiponectin induces the activation of endothelial nitric oxide synthase (eNOS) and stimulates nitric oxide production[29]. Moreover, adiponectin inhibits the oxygen free radical production and ameliorates the endothelial function in mice genetically modified to develop hyperlipidemia and atherosclerosis[30].

Several experimental studies have clearly demonstrated that adiponectin plays a protective role for the cardiovascular system since it is able to interfere with the early steps of atherosclerotic disease. In particular, it has been shown that adiponectin deficiency increases leukocyte-endothelium interactions via up-regulation of endothelial cell adhesion molecules (CAMs) in vivo[31]. Conversely, the expression of adhesion molecules is reduced when increased levels of adiponectin are measurable[32]. Moreover, adiponectin suppresses smooth muscle cells proliferation[33] and inhibits lipopolysaccharide-induced adventitial fibroblast migration and transition to myofibroblasts via the AdipoR1-AMPK-iNOS pathway[34]. Again, adiponectin suppresses lipid accumulation and class A scavenger receptor expression in human monocyte-derived macrophages[35]. Finally, this adipocytokine reduces lipid accumulation in macrophage foam cells[36] and is able to reduce atherosclerosis in apolipoprotein E-deficient mice[37]. Of note, adiponectin seems to play an important role in modulating the inflammatory network involved in the pathophysiology of cardiovascular disease because it regulates the expression of several important chemokines, such as IP-10, Mig and I-TAC, which bind to the chemokine receptor CXCR3, an important regulator of chemotaxis of lymphocytes within the atherosclerotic plaque[38]. The protective role played by adiponectin has also been confirmed by recent studies showing that the adiponectin receptor is detectable on platelets, thus suggesting that this adipocytokine acts as an endogenous antithrombotic factor[39].

LEPTIN

Leptin is a polypeptide consisting of 167 amino acids, encoded by the “ob” gene and implicated in the regulation of body weight and energy balance[40]. Experimental and clinical evidence has shown that this peptide might be involved in the pathophysiology of metabolic syndrome[41]. In fact, elevated leptin plasma levels are usually detectable in the plasma of obese patients[42]. In this context, the relationship between obesity, elevated plasma levels of leptin and cardiovascular disease appears of particular interest. Several clinical studies have shown that patients with increased plasma concentrations of leptin are at high risk of developing myocardial infarction[43] and stroke[44]. In addition, elevated serum levels of leptin were measured in patients with myocardial infarction with ST elevation[45]. Finally, a large prospective study on leptin and cardiovascular risk, the West of Scotland Coronary Prevention Study, confirmed that leptin is an independent predictor of coronary events[46]. This adipokine has recently been identified as a good prognostic marker of future cardiovascular events in patients with angiographically proven atherosclerosis[47]. Elevated leptin baseline levels are associated with increased risk of cardiac death, new myocardial infarction, stroke and coronary revascularization, even in patients without diabetes[47]. Hyperleptinemia is closely associated with in-stent restenosis in patients undergoing coronary stenting[48]. Moreover, the ratio of leptin:adiponectin seems to be directly correlated with the magnitude of the intima-media thickness of the common carotid artery, a good index of subclinical atherosclerosis[49]. Finally, it has been suggested that leptin, by stimulating the sympathetic nervous system, might also play an important role in the pathophysiology of hypertension[50]. These clinical observations have been supported by experimental studies which have strongly reinforced the hypothesis that leptin might be involved in the pathophysiology of cardiovascular disease. Leptin seems to be able to modulate platelet aggregation[51] and arterial thrombosis[52,53]. In a recent paper, it has been demonstrated that leptin, at concentrations usually measurable in the plasma of patients with acute coronary syndrome, induces a pro-atherothrombotic phenotype in human coronary endothelial cells through the expression of tissue factor (TF) and CAMs[54]. Moreover, leptin can induce the expression of TF in human peripheral blood mononuclear cells[55].

The progression of atherosclerotic plaques might be modulated by leptin. Treatment of apo-lipoprotein E deficient mice with leptin causes faster progression of vessel atherosclerosis and increases the amount of calcium in the vessel wall[56]. In addition, it has been shown that treatment of hyper-lipidemic mice with recombinant leptin increases the atherosclerotic burden and promotes a faster thrombus formation[53]. Interestingly, leptin deficiency suppresses progression of atherosclerosis in apoE-deficient mice[57]. Recently, it has been demonstrated that leptin stimulates the production of C-reactive protein (CRP) in human coronary endothelial cells[58]. Since it has been previously evidenced that CRP induces vascular thrombosis[59], taken together, these observations suggest that leptin might promote the development of the atherosclerotic disease and that it might be involved in the pathophysiology of acute coronary syndromes.

RESISTIN

Resistin is a cysteine-rich protein of 12.5 kDa, consisting in humans of 108 amino acids: 17 amino acids form the N-terminal signal sequence, 37 in the variable portion and the remaining in the constant area at the C-terminal. Its gene is located on chromosome 19. Recently, a family of resistin-like molecules (RELMs) have been described. These polypeptides consist of 105-114 amino acids and are composed of three domains: a signal sequence at the N-terminal, a variable central portion and a highly conserved C-terminal. RELM α is secreted mainly by adipose tissue, while RELM-β is expressed only in the gastrointestinal tract and in neoplastic cells, suggesting a possible role in cell proliferation. RELM-γ, recently discovered, was found in hematopoietic tissue, where it is supposed to have a cytokine-like activity. In rodents, adipocytes are the main source of resistin, while in humans, macrophages have this function[60]. Initially, this adipokine was proposed as a potential link between obesity and diabetes by modulating the mechanisms responsible for insulin resistance[61]. Then, experimental evidence in vivo and in vitro has shown that resistin is able to trigger the mechanisms involved in inflammation[60]. In addition, plasma levels of resistin appear to be closely correlated with other markers of inflammation, such as TNF-α, type 2 soluble receptor for TNF-α and IL-6[62-64]. Of note, patients with acute coronary syndrome have resistin plasma levels significantly higher than patients with stable angina and healthy controls[65]. Then, it was also shown that resistin might be an independent predictor of coronary atherosclerosis in humans[62-64]. Finally, since plasma resistin is associated with myocardium injury in patients with acute coronary syndrome, it has been proposed as a marker of ischemic injury[66]. Recent experimental evidence has indicated that resistin promotes endothelial dysfunction. In fact, it induces expression of adhesion molecules and cytokine in human endothelial cells[67]. In addition, it influences the expression of PI3Kp85 and stimulates the release of plasminogen activator inhibitor 1, von Willebrand factor and endothelin[68]. Again, resistin inhibits the expression of eNOS. Finally, recent studies have shown that this molecule is able to promote proliferation and migration of smooth muscle cells through the activation of ERKs and PI3K kinase[69,70]. In a recent in vitro study, it has also been shown that resistin induces the synthesis and expression of active TF in human coronary artery cells[71].

TNF-α

TNF-α is another important chemical mediator involved in the progression of atherosclerotic disease[72]. It has pro-inflammatory activity and is produced mainly by inflammatory cells, such as monocytes and macrophages. This molecule is actively involved in the modulation of inflammatory and autoimmune diseases. Adipose tissue of animals used as experimental models of obesity and type 2 diabetes has increased expression of TNF-α[73]. Similarly, elevated levels of TNF-α have been detected in adipose tissue and plasma of obese patients. Interestingly, TNF-α plasma levels are closely related to the extent of visceral adipose tissue, while weight loss and lifestyle changes reduce these levels[74]. TNF-α has been isolated in atherosclerotic plaques and it seems to be a marker of plaque growth[75-77]. The role played by this pro-inflammatory molecule was also highlighted by elegant experiments in mice genetically modified to be deficient in expression of TNF-α: these animals showed less atherosclerosis compared to the same animals not genetically modified. In addition, these animals showed a poor inflammatory profile with low levels of IL-1β, IFN-γ, ICAM-1, VCAM-1, MCP-1, GM-CSF and nuclear factor (NF)-κB[78]. TNF-α also appears to be able to regulate the expression of lectin-like type 1 receptor for oxidized LDL, an important facilitating atherosclerosis mediator in various cell types, including endothelial cells and macrophages[79].

IL-6

IL-6 is another pro-inflammatory cytokine with plasma levels closely related to the amount of visceral fat[80]. In fact, obese patients have elevated plasma levels of IL-6 which can be reduced by weight loss[74]. It is estimated that the adipose tissue is responsible for the production of about one third of the circulating levels of IL-6[80] and it has been hypothesized that the secretion of IL-6 in obese individuals contributes to metabolic dysfunction. In fact, plasma levels of IL-6 can be considered predictive for the development of type 2 diabetes and, in turn, high levels of this cytokine are detectable in diabetics[81]. IL-6 plays an important role in the pathophysiology of atherothrombosis: it modulates the release of pro-inflammatory cytokines and of other pro-thrombotic mediators, promotes lipoprotein oxidation, activates matrix metalloproteinases and, finally, it stimulates secretion of the acute phase proteins[82]. IL-6 is secreted directly into the portal circulation; thus, it reaches the liver where it stimulates release of CRP[11], an important predictor of future cardiovascular events both in patients with documented cardiovascular damage and in apparently healthy people[83].

VISFATIN

Visfatin, identified in 2004, derives its name from the fact that it is produced mainly in visceral fat. It is composed of 491 amino acids and has a molecular weight of 52 kDa. Visfatin gene corresponds to the gene of the Pre-B cell colony enhancing factor (PBEF). PBEF was described in 1994 as a cytokine produced by lymphocytes, involved in regulation of inflammatory mechanisms.

Interestingly, this adipokine is produced by macrophages resident in adipose tissue and not directly by adipocytes. In this regard, the levels of visfatin are believed to be the expression of macrophages infiltrating the adipose tissue, where they produce it in response to inflammatory signals[84]. Circulating levels of visfatin are closely correlated with white adipose tissue accumulation[9]. However, the relationship between circulating visfatin levels and anthropometric and metabolic parameters of obesity and type 2 diabetes is still not completely understood. The role of visfatin in cardiovascular disease is not clear. Increased expression of visfatin has been demonstrated in macrophages of human unstable carotid and coronary atherosclerotic plaques[85]. Again, endothelial dysfunction has been described in those patients with elevated plasma levels of visfatin[86]. Moreover, it has been observed that visfatin is able to increase the expression of adhesion molecules by activating the transcription factor NF-κB[87]. Taken together, these data suggest that visfatin might play a role in plaque destabilization.

“OTHER” ADIPOKINES

Several “other” adipokines with paracrine/endocrine activities are produced by adipose tissue. Some of these molecules have been partially characterized: apelin is a bioactive peptide produced by adipocytes, stromal vascular cells and cardiac myocytes. In obese patients with hyperinsulinemia, increased plasma levels of apelin are measurable[88]. In animal models of heart failure, cardiac apelin is down-regulated by angiotensin II, while its production is restored after treatment with an angiotensin receptor 1 antagonist[89]. In rats, the cardiac production of apelin is increased by hypoxia[90] and ischemia[91]. In spontaneously hypertensive rats, exercise has also been shown to stimulate the production of apelin[92]. This molecule also has a positive hemodynamic effect, acting with an inotropic mechanism in rats with heart failure, as well as in isolated cardiomyocytes[93]. In humans, low plasma apelin was observed in patients with chronic heart failure[94] and in patients with atrial fibrillation[95]. In these, cardiac resynchronization therapy is accompanied by increases in the concentrations of apelin[96]. Despite the available evidence, it is still unclear whether the form of metabolically active apelin is produced by adipose or by cardiac tissue. Thus, more studies are needed to clarify the role of this interesting peptide in the pathophysiology of cardiovascular disease and its potential therapeutic use.

Omentin is a secretory protein selectively expressed in the visceral adipose tissue, where it is synthesized by the visceral stromal vascular cells[97,98]. It has also been found in the human lung, intestine, ovaries, placenta and heart[98]. Omentin plasma levels are reduced in obese patients[99]. Conversely, elevated levels of omentin are measurable in the plasma of lean subjects with increased levels of adiponectin and high-density lipoproteins[99,100]. Omentin increases insulin-stimulated glucose uptake in both omental and subcutaneous adipocytes and promotes Akt/PKB phosphorylation[101]. Interestingly, it has been shown that low levels of omentin are measurable in patients with severe coronary atherosclerotic disease[102-104]. Finally, the association of circulating omentin level with arterial stiffness and carotid plaque in patients with diabetes has been demonstrated[105]. Recently, it has been proposed that omentin might be an early marker of metabolic dysfunction[106].

Vaspin is a novel adipokine expressed in the visceral and subcutaneous adipose tissue, involved in the development of obesity and insulin resistance[107-110]. The relationship between vaspin and cardiovascular disease is still obscure. It has been demonstrated that patients with unstable angina have reduced plasma and mRNA levels of this adipokine[111]. In addition, vaspin serum concentrations were significantly low in patients with atherosclerosis of carotid arteries[112].

Table 1: Overview of adipocitokines and of their potential cardiovascular functions

Table 1 Sources and potential role in cardiovascular disease of key adipokines.
AdipokinePrimary source(s)Potential role in CVD
AdiponectinAdipocytes↑Insulin sensitivity, energy consumption, fatty acid oxidation
↓Oxidative stress
Anti-inflammatory activity
↓TNF-α, IL-6, interferon-c ↑IL-1R antagonist
Modulation of chemokine expression
Improved endothelial function
eNOS induction ↑NO ↓ROS
regulation of adhesion molecules
Regulation of macrophage function
Antiaggregants effects
Decreased progression of atherosclerotic lesions
LeptinAdipocytesAppetite regulation and modulation of energy expenditure
Independent prognostic marker of ACS
Modulation of blood pressure
Regulation of platelet aggregation and induction of arterial thrombosis
TF, CRP and adhesion molecules induction in endothelial cells
TF induction in peripheral blood mononuclear cells
Maintenance of progression of atherosclerotic disease
Resistin/RELMsRELM-α: Adipose tissue macrophages (human), adipocytes (mice)Induction of insulin resistance
Cell proliferation (RELM-β)
Cytokine-like functionality (RELM-γ)
RELM-β: Tumor and gastro-intestinal cellsPro-inflammatory activity
Independent predictive marker of atherosclerosis and severity of ischemic injury
RELM-γ: Hematopoietic tissueEffects on endothelial cells
↑Adhesion molecules, cytokines, TF, plasminogen activator inhibitor, von Willebrand factor, endothelin
↓e-NOS expression
Smooth muscle cell proliferation and migration
TNF-αInflammatory cells, monocytes, macrophages, adipocytesReduction of insulin signaling
Induction of insulin resistance
Maintaining proinflammatory state and atherosclerosis
IL-6Inflammatory cells, stromal vascular fraction cells, adipocytes, liver, muscleInduction of insulin resistance
Maintaining pro-inflammatory status
Modulation of pro-inflammatory cytokines and pro-thrombotic mediators release
Promotion of lipoproteins oxidation
Activation of matrix metalloproteinase
Induction of CRP production by the liver
VisfatinLymphocytes, macrophages, adipocytes, other cellsMonocyte chemotactic activity
Endothelial dysfunction
Atherosclerotic plaque destabilization
TF induction
Other adipokines
ApelinAdipocytes, stromal vascular cells and cardiac myocytesCardiomyocyte function regulation
OmentinStromal vascular cells of visceral adipose tissue↑Insulin-stimulated glucose uptake in both Low levels in severe coronary atherosclerotic disease and arterial stiffness and carotid plaque in patients with diabetes
Possible marker of metabolic dysfunction
VaspinVisceral and subcutaneous adipose tissueReduced plasma and mRNA levels in patients with unstable angina
Low serum concentrations in patients with carotid atherosclerosis
CONCLUSION

It is well established that obesity and cardiovascular events are closely related. However, although it has been clearly demonstrated that obesity is also associated with other conditions that may accelerate atherosclerosis, the molecular mechanisms underlying these phenomena are not fully defined yet. In this context, adipose tissue, abundantly represented in obese individuals, might play a role by producing substances, called adipokines, with an active role in the pathophysiology of cardiovascular disease. Although some of these molecules, such as adiponectin, leptin and resistin, now have a well defined position in the complex network between inflammation, obesity and cardiovascular diseases, many others are still poorly defined functionally. It seems clear that the adipokines panorama needs new experimental evidence and clinical trials to compose the complex puzzle that link a “modern” and wellness disease like obesity with cardiovascular disease, the leading cause of mortality in industrialized world. As in a complex crime scene, we have several potentially guilty candidates.

Footnotes

Peer reviewer: Po-Shiuan Hsieh, MD, PhD, Professor, Department of Physiology and Biophysics, National Defense Medical Center, Taipei, 114, Taiwan, China

S- Editor Wang JL L- Editor Roemmele A E- Editor Zheng XM

References
1.  Ogden CL, Yanovski SZ, Carroll MD, Flegal KM. The epidemiology of obesity. Gastroenterology. 2007;132:2087-2102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1006]  [Cited by in F6Publishing: 1054]  [Article Influence: 62.0]  [Reference Citation Analysis (0)]
2.  Guilbert JJ. The world health report 2002 - reducing risks, promoting healthy life. Educ Health (Abingdon). 2003;16:230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 1085]  [Article Influence: 51.7]  [Reference Citation Analysis (0)]
3.  Phillips SD, Roberts WC. Comparison of body mass index among patients with versus without angiographic coronary artery disease. Am J Cardiol. 2007;100:18-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
4.  Buettner HJ, Mueller C, Gick M, Ferenc M, Allgeier J, Comberg T, Werner KD, Schindler C, Neumann FJ. The impact of obesity on mortality in UA/non-ST-segment elevation myocardial infarction. Eur Heart J. 2007;28:1694-1701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 89]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
5.  Faber DR, de Groot PG, Visseren FL. Role of adipose tissue in haemostasis, coagulation and fibrinolysis. Obes Rev. 2009;10:554-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 133]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
6.  Sowers JR. Obesity as a cardiovascular risk factor. Am J Med. 2003;115 Suppl 8A:37S-41S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 365]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
7.  Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol. 2011;11:85-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3038]  [Cited by in F6Publishing: 2861]  [Article Influence: 220.1]  [Reference Citation Analysis (0)]
8.  Attie AD, Scherer PE. Adipocyte metabolism and obesity. J Lipid Res. 2009;50 Suppl:S395-S399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 153]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
9.  Wang Z, Nakayama T. Inflammation, a link between obesity and cardiovascular disease. Mediators Inflamm. 2010;2010:535918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 247]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
10.  Trayhurn P, Wood IS. Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br J Nutr. 2004;92:347-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1428]  [Cited by in F6Publishing: 1392]  [Article Influence: 69.6]  [Reference Citation Analysis (0)]
11.  Deng Y, Scherer PE. Adipokines as novel biomarkers and regulators of the metabolic syndrome. Ann N Y Acad Sci. 2010;1212:E1-E19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 338]  [Cited by in F6Publishing: 360]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
12.  Kadoglou NP, Iliadis F, Angelopoulou N, Perrea D, Ampatzidis G, Liapis CD, Alevizos M. The anti-inflammatory effects of exercise training in patients with type 2 diabetes mellitus. Eur J Cardiovasc Prev Rehabil. 2007;14:837-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 203]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
13.  Barseghian A, Gawande D, Bajaj M. Adiponectin and vulnerable atherosclerotic plaques. J Am Coll Cardiol. 2011;57:761-770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
14.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2259]  [Cited by in F6Publishing: 2236]  [Article Influence: 77.1]  [Reference Citation Analysis (0)]
15.  Takahashi M, Arita Y, Yamagata K, Matsukawa Y, Okutomi K, Horie M, Shimomura I, Hotta K, Kuriyama H, Kihara S. Genomic structure and mutations in adipose-specific gene, adiponectin. Int J Obes Relat Metab Disord. 2000;24:861-868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 275]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
16.  Nanasato M, Murohara T. Role of adiponectin in cardiovascular protection. Circ J. 2010;74:432-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
17.  Hasan-Ali H, Abd El-Mottaleb NA, Hamed HB, Abd-Elsayed A. Serum adiponectin and leptin as predictors of the presence and degree of coronary atherosclerosis. Coron Artery Dis. 2011;22:264-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
18.  Liang KW, Sheu WH, Lee WL, Liu TJ, Ting CT, Hsieh YC, Wang KY, Chen YT, Lee WJ. Decreased circulating protective adiponectin level is associated with angiographic coronary disease progression in patients with angina pectoris. Int J Cardiol. 2008;129:76-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
19.  Eroglu S, Sade LE, Bozbas H, Haberal A, Ozbicer S, Demir O, Muderrisoglu H. Association of serum adiponectin levels and coronary flow reserve in women with normal coronary angiography. Eur J Cardiovasc Prev Rehabil. 2009;16:290-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
20.  Matsuzawa Y, Funahashi T, Kihara S, Shimomura I. Adiponectin and metabolic syndrome. Arterioscler Thromb Vasc Biol. 2004;24:29-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 838]  [Cited by in F6Publishing: 812]  [Article Influence: 38.7]  [Reference Citation Analysis (0)]
21.  Bobbert T, Rochlitz H, Wegewitz U, Akpulat S, Mai K, Weickert MO, Möhlig M, Pfeiffer AF, Spranger J. Changes of adiponectin oligomer composition by moderate weight reduction. Diabetes. 2005;54:2712-2719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 212]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
22.  Pajvani UB, Du X, Combs TP, Berg AH, Rajala MW, Schulthess T, Engel J, Brownlee M, Scherer PE. Structure-function studies of the adipocyte-secreted hormone Acrp30/adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem. 2003;278:9073-9085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 798]  [Cited by in F6Publishing: 796]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
23.  Ebinuma H, Miyazaki O, Yago H, Hara K, Yamauchi T, Kadowaki T. A novel ELISA system for selective measurement of human adiponectin multimers by using proteases. Clin Chim Acta. 2006;372:47-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 125]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
24.  Lara-Castro C, Luo N, Wallace P, Klein RL, Garvey WT. Adiponectin multimeric complexes and the metabolic syndrome trait cluster. Diabetes. 2006;55:249-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 357]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
25.  Murdolo G, Nowotny B, Celi F, Donati M, Bini V, Papi F, Gornitzka G, Castellani S, Roden M, Falorni A. Inflammatory adipokines, high molecular weight adiponectin, and insulin resistance: a population-based survey in prepubertal schoolchildren. PLoS One. 2011;6:e17264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
26.  Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, Iwabu M, Okada-Iwabu M, Kawamoto S, Kubota N, Kubota T. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med. 2007;13:332-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 968]  [Cited by in F6Publishing: 986]  [Article Influence: 58.0]  [Reference Citation Analysis (0)]
27.  Tilg H, Hotamisligil GS. Nonalcoholic fatty liver disease: Cytokine-adipokine interplay and regulation of insulin resistance. Gastroenterology. 2006;131:934-945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 252]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
28.  Stofkova A. Leptin and adiponectin: from energy and metabolic dysbalance to inflammation and autoimmunity. Endocr Regul. 2009;43:157-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
29.  Cheng KK, Lam KS, Wang Y, Huang Y, Carling D, Wu D, Wong C, Xu A. Adiponectin-induced endothelial nitric oxide synthase activation and nitric oxide production are mediated by APPL1 in endothelial cells. Diabetes. 2007;56:1387-1394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 240]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
30.  Chen X, Zhang H, McAfee S, Zhang C. The reciprocal relationship between adiponectin and LOX-1 in the regulation of endothelial dysfunction in ApoE knockout mice. Am J Physiol Heart Circ Physiol. 2010;299:H605-H612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
31.  Ouedraogo R, Gong Y, Berzins B, Wu X, Mahadev K, Hough K, Chan L, Goldstein BJ, Scalia R. Adiponectin deficiency increases leukocyte-endothelium interactions via upregulation of endothelial cell adhesion molecules in vivo. J Clin Invest. 2007;117:1718-1726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 207]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
32.  Ouchi N, Kihara S, Arita Y, Maeda K, Kuriyama H, Okamoto Y, Hotta K, Nishida M, Takahashi M, Nakamura T. Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation. 1999;100:2473-2476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1551]  [Cited by in F6Publishing: 1518]  [Article Influence: 60.7]  [Reference Citation Analysis (0)]
33.  Arita Y, Kihara S, Ouchi N, Maeda K, Kuriyama H, Okamoto Y, Kumada M, Hotta K, Nishida M, Takahashi M. 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;105:2893-2898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 511]  [Cited by in F6Publishing: 483]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
34.  Cai XJ, Chen L, Li L, Feng M, Li X, Zhang K, Rong YY, Hu XB, Zhang MX, Zhang Y. Adiponectin inhibits lipopolysaccharide-induced adventitial fibroblast migration and transition to myofibroblasts via AdipoR1-AMPK-iNOS pathway. Mol Endocrinol. 2010;24:218-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
35.  Ouchi N, Kihara S, Arita Y, Nishida M, Matsuyama A, Okamoto Y, Ishigami M, Kuriyama H, Kishida K, Nishizawa H. Adipocyte-derived plasma protein, adiponectin, suppresses lipid accumulation and class A scavenger receptor expression in human monocyte-derived macrophages. Circulation. 2001;103:1057-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 897]  [Cited by in F6Publishing: 881]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
36.  Tian L, Luo N, Klein RL, Chung BH, Garvey WT, Fu Y. Adiponectin reduces lipid accumulation in macrophage foam cells. Atherosclerosis. 2009;202:152-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 77]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
37.  Okamoto Y, Kihara S, Ouchi N, Nishida M, Arita Y, Kumada M, Ohashi K, Sakai N, Shimomura I, Kobayashi H. Adiponectin reduces atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2002;106:2767-2770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 788]  [Cited by in F6Publishing: 752]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
38.  Okamoto Y, Folco EJ, Minami M, Wara AK, Feinberg MW, Sukhova GK, Colvin RA, Kihara S, Funahashi T, Luster AD. Adiponectin inhibits the production of CXC receptor 3 chemokine ligands in macrophages and reduces T-lymphocyte recruitment in atherogenesis. Circ Res. 2008;102:218-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 148]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
39.  Kato H, Kashiwagi H, Shiraga M, Tadokoro S, Kamae T, Ujiie H, Honda S, Miyata S, Ijiri Y, Yamamoto J. Adiponectin acts as an endogenous antithrombotic factor. Arterioscler Thromb Vasc Biol. 2006;26:224-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 136]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
40.  Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395:763-770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3710]  [Cited by in F6Publishing: 3569]  [Article Influence: 137.3]  [Reference Citation Analysis (0)]
41.  Gnacińska M, Małgorzewicz S, Stojek M, Łysiak-Szydłowska W, Sworczak K. Role of adipokines in complications related to obesity: a review. Adv Med Sci. 2009;54:150-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 105]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
42.  Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334:292-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4210]  [Cited by in F6Publishing: 4079]  [Article Influence: 145.7]  [Reference Citation Analysis (0)]
43.  Söderberg S, Ahrén B, Jansson JH, Johnson O, Hallmans G, Asplund K, Olsson T. Leptin is associated with increased risk of myocardial infarction. J Intern Med. 1999;246:409-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 261]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
44.  Söderberg S, Ahrén B, Stegmayr B, Johnson O, Wiklund PG, Weinehall L, Hallmans G, Olsson T. Leptin is a risk marker for first-ever hemorrhagic stroke in a population-based cohort. Stroke. 1999;30:328-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 119]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
45.  Taneli F, Yegane S, Ulman C, Tikiz H, Bilge AR, Ari Z, Uyanik BS. Increased serum leptin concentrations in patients with chronic stable angina pectoris and ST-elevated myocardial infarction. Angiology. 2006;57:267-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
46.  Wallace AM, McMahon AD, Packard CJ, Kelly A, Shepherd J, Gaw A, Sattar N. Plasma leptin and the risk of cardiovascular disease in the west of Scotland coronary prevention study (WOSCOPS). Circulation. 2001;104:3052-3056.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 576]  [Cited by in F6Publishing: 560]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
47.  Wolk R, Berger P, Lennon RJ, Brilakis ES, Johnson BD, Somers VK. Plasma leptin and prognosis in patients with established coronary atherosclerosis. J Am Coll Cardiol. 2004;44:1819-1824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 158]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
48.  Piatti P, Di Mario C, Monti LD, Fragasso G, Sgura F, Caumo A, Setola E, Lucotti P, Galluccio E, Ronchi C. Association of insulin resistance, hyperleptinemia, and impaired nitric oxide release with in-stent restenosis in patients undergoing coronary stenting. Circulation. 2003;108:2074-2081.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 139]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
49.  Norata GD, Raselli S, Grigore L, Garlaschelli K, Dozio E, Magni P, Catapano AL. Leptin: adiponectin ratio is an independent predictor of intima media thickness of the common carotid artery. Stroke. 2007;38:2844-2846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 143]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
50.  Beltowski J. Leptin and atherosclerosis. Atherosclerosis. 2006;189:47-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 324]  [Cited by in F6Publishing: 333]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
51.  Konstantinides S, Schäfer K, Koschnick S, Loskutoff DJ. Leptin-dependent platelet aggregation and arterial thrombosis suggests a mechanism for atherothrombotic disease in obesity. J Clin Invest. 2001;108:1533-1540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 245]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
52.  Bodary PF, Westrick RJ, Wickenheiser KJ, Shen Y, Eitzman DT. Effect of leptin on arterial thrombosis following vascular injury in mice. JAMA. 2002;287:1706-1709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 178]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
53.  Bodary PF, Gu S, Shen Y, Hasty AH, Buckler JM, Eitzman DT. Recombinant leptin promotes atherosclerosis and thrombosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25:e119-e122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 98]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
54.  Cirillo P, Angri V, De Rosa S, Calì G, Petrillo G, Maresca F, D'Ascoli GL, Maietta P, Brevetti L, Chiariello M. Pro-atherothrombotic effects of leptin in human coronary endothelial cells. Thromb Haemost. 2010;103:1065-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
55.  Napoleone E, DI Santo A, Amore C, Baccante G, di Febbo C, Porreca E, de Gaetano G, Donati MB, Lorenzet R. Leptin induces tissue factor expression in human peripheral blood mononuclear cells: a possible link between obesity and cardiovascular risk? J Thromb Haemost. 2007;5:1462-1468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 63]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
56.  Zeadin M, Butcher M, Werstuck G, Khan M, Yee CK, Shaughnessy SG. Effect of leptin on vascular calcification in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2009;29:2069-2075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 52]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
57.  Chiba T, Shinozaki S, Nakazawa T, Kawakami A, Ai M, Kaneko E, Kitagawa M, Kondo K, Chait A, Shimokado K. Leptin deficiency suppresses progression of atherosclerosis in apoE-deficient mice. Atherosclerosis. 2008;196:68-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 61]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  De Rosa S, Cirillo P, Pacileo M, Di Palma V, Paglia A, Chiariello M. Leptin stimulated C-reactive protein production by human coronary artery endothelial cells. J Vasc Res. 2009;46:609-617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
59.  Cirillo P, Golino P, Calabrò P, Calì G, Ragni M, De Rosa S, Cimmino G, Pacileo M, De Palma R, Forte L. C-reactive protein induces tissue factor expression and promotes smooth muscle and endothelial cell proliferation. Cardiovasc Res. 2005;68:47-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 102]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
60.  Filková M, Haluzík M, Gay S, Senolt L. The role of resistin as a regulator of inflammation: Implications for various human pathologies. Clin Immunol. 2009;133:157-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 291]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
61.  Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA. The hormone resistin links obesity to diabetes. Nature. 2001;409:307-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3205]  [Cited by in F6Publishing: 3111]  [Article Influence: 135.3]  [Reference Citation Analysis (1)]
62.  Yaturu S, Daberry RP, Rains J, Jain S. Resistin and adiponectin levels in subjects with coronary artery disease and type 2 diabetes. Cytokine. 2006;34:219-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 62]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
63.  Burnett MS, Lee CW, Kinnaird TD, Stabile E, Durrani S, Dullum MK, Devaney JM, Fishman C, Stamou S, Canos D. The potential role of resistin in atherogenesis. Atherosclerosis. 2005;182:241-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 168]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
64.  Reilly MP, Lehrke M, Wolfe ML, Rohatgi A, Lazar MA, Rader DJ. Resistin is an inflammatory marker of atherosclerosis in humans. Circulation. 2005;111:932-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 627]  [Cited by in F6Publishing: 625]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
65.  Wang H, Chen DY, Cao J, He ZY, Zhu BP, Long M. High serum resistin level may be an indicator of the severity of coronary disease in acute coronary syndrome. Chin Med Sci J. 2009;24:161-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
66.  Chu S, Ding W, Li K, Pang Y, Tang C. Plasma resistin associated with myocardium injury in patients with acute coronary syndrome. Circ J. 2008;72:1249-1253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 50]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
67.  Verma S, Li SH, Wang CH, Fedak PW, Li RK, Weisel RD, Mickle DA. Resistin promotes endothelial cell activation: further evidence of adipokine-endothelial interaction. Circulation. 2003;108:736-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 476]  [Cited by in F6Publishing: 463]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
68.  Li Y, Wang Y, Li Q, Chen Y, Sun SZ, Zhang WD, Jia Q. Effect of resistin on vascular endothelium secretion dysfunction in rats. Endothelium. 2007;14:207-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
69.  Calabro P, Samudio I, Willerson JT, Yeh ET. Resistin promotes smooth muscle cell proliferation through activation of extracellular signal-regulated kinase 1/2 and phosphatidylinositol 3-kinase pathways. Circulation. 2004;110:3335-3340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 225]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
70.  Jung HS, Park KH, Cho YM, Chung SS, Cho HJ, Cho SY, Kim SJ, Kim SY, Lee HK, Park KS. Resistin is secreted from macrophages in atheromas and promotes atherosclerosis. Cardiovasc Res. 2006;69:76-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 183]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
71.  Calabrò P, Cirillo P, Limongelli G, Maddaloni V, Riegler L, Palmieri R, Pacileo G, De Rosa S, Pacileo M, De Palma R. Tissue factor is induced by resistin in human coronary artery endothelial cells by the NF-ĸB-dependent pathway. J Vasc Res. 2011;48:59-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
72.  Zhang L, Peppel K, Sivashanmugam P, Orman ES, Brian L, Exum ST, Freedman NJ. Expression of tumor necrosis factor receptor-1 in arterial wall cells promotes atherosclerosis. Arterioscler Thromb Vasc Biol. 2007;27:1087-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 75]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
73.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5334]  [Cited by in F6Publishing: 5219]  [Article Influence: 168.4]  [Reference Citation Analysis (0)]
74.  Esposito K, Pontillo A, Di Palo C, Giugliano G, Masella M, Marfella R, Giugliano D. Effect of weight loss and lifestyle changes on vascular inflammatory markers in obese women: a randomized trial. JAMA. 2003;289:1799-1804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1081]  [Cited by in F6Publishing: 1010]  [Article Influence: 48.1]  [Reference Citation Analysis (0)]
75.  Barath P, Fishbein MC, Cao J, Berenson J, Helfant RH, Forrester JS. Detection and localization of tumor necrosis factor in human atheroma. Am J Cardiol. 1990;65:297-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 332]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
76.  Rus HG, Niculescu F, Vlaicu R. Tumor necrosis factor-alpha in human arterial wall with atherosclerosis. Atherosclerosis. 1991;89:247-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 164]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
77.  Ohta H, Wada H, Niwa T, Kirii H, Iwamoto N, Fujii H, Saito K, Sekikawa K, Seishima M. Disruption of tumor necrosis factor-alpha gene diminishes the development of atherosclerosis in ApoE-deficient mice. Atherosclerosis. 2005;180:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 252]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
78.  Xiao N, Yin M, Zhang L, Qu X, Du H, Sun X, Mao L, Ren G, Zhang C, Geng Y. Tumor necrosis factor-alpha deficiency retards early fatty-streak lesion by influencing the expression of inflammatory factors in apoE-null mice. Mol Genet Metab. 2009;96:239-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
79.  Zhang H, Cui J, Zhang C. Emerging role of adipokines as mediators in atherosclerosis. World J Cardiol. 2010;2:370-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
80.  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;83:847-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 481]  [Cited by in F6Publishing: 420]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
81.  Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 2001;286:327-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2974]  [Cited by in F6Publishing: 2900]  [Article Influence: 126.1]  [Reference Citation Analysis (1)]
82.  Schuett H, Luchtefeld M, Grothusen C, Grote K, Schieffer B. How much is too much? Interleukin-6 and its signalling in atherosclerosis. Thromb Haemost. 2009;102:215-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 194]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
83.  Ridker PM, Koenig W, Fuster V. C-reactive protein and coronary heart disease. N Engl J Med. 2004;351:295-298; author reply 295-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
84.  Saddi-Rosa P, Oliveira CS, Giuffrida FM, Reis AF. Visfatin, glucose metabolism and vascular disease: a review of evidence. Diabetol Metab Syndr. 2010;2:21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 91]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
85.  Dahl TB, Yndestad A, Skjelland M, Øie E, Dahl A, Michelsen A, Damås JK, Tunheim SH, Ueland T, Smith C. Increased expression of visfatin in macrophages of human unstable carotid and coronary atherosclerosis: possible role in inflammation and plaque destabilization. Circulation. 2007;115:972-980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 343]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
86.  Takebayashi K, Suetsugu M, Wakabayashi S, Aso Y, Inukai T. Association between plasma visfatin and vascular endothelial function in patients with type 2 diabetes mellitus. Metabolism. 2007;56:451-458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 122]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
87.  Kim SR, Bae YH, Bae SK, Choi KS, Yoon KH, Koo TH, Jang HO, Yun I, Kim KW, Kwon YG. Visfatin enhances ICAM-1 and VCAM-1 expression through ROS-dependent NF-kappaB activation in endothelial cells. Biochim Biophys Acta. 2008;1783:886-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 219]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
88.  Lee DK, George SR, O'Dowd BF. Unravelling the roles of the apelin system: prospective therapeutic applications in heart failure and obesity. Trends Pharmacol Sci. 2006;27:190-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 88]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
89.  Iwanaga Y, Kihara Y, Takenaka H, Kita T. Down-regulation of cardiac apelin system in hypertrophied and failing hearts: Possible role of angiotensin II-angiotensin type 1 receptor system. J Mol Cell Cardiol. 2006;41:798-806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 122]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
90.  Ronkainen VP, Ronkainen JJ, Hänninen SL, Leskinen H, Ruas JL, Pereira T, Poellinger L, Vuolteenaho O, Tavi P. Hypoxia inducible factor regulates the cardiac expression and secretion of apelin. FASEB J. 2007;21:1821-1830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 127]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
91.  Atluri P, Morine KJ, Liao GP, Panlilio CM, Berry MF, Hsu VM, Hiesinger W, Cohen JE, Joseph Woo Y. Ischemic heart failure enhances endogenous myocardial apelin and APJ receptor expression. Cell Mol Biol Lett. 2007;12:127-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 71]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
92.  Zhang J, Ren CX, Qi YF, Lou LX, Chen L, Zhang LK, Wang X, Tang C. Exercise training promotes expression of apelin and APJ of cardiovascular tissues in spontaneously hypertensive rats. Life Sci. 2006;79:1153-1159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
93.  Higuchi K, Masaki T, Gotoh K, Chiba S, Katsuragi I, Tanaka K, Kakuma T, Yoshimatsu H. Apelin, an APJ receptor ligand, regulates body adiposity and favors the messenger ribonucleic acid expression of uncoupling proteins in mice. Endocrinology. 2007;148:2690-2697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 151]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
94.  Chong KS, Gardner RS, Morton JJ, Ashley EA, McDonagh TA. Plasma concentrations of the novel peptide apelin are decreased in patients with chronic heart failure. Eur J Heart Fail. 2006;8:355-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 148]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
95.  Ellinor PT, Low AF, Macrae CA. Reduced apelin levels in lone atrial fibrillation. Eur Heart J. 2006;27:222-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
96.  Francia P, Salvati A, Balla C, De Paolis P, Pagannone E, Borro M, Gentile G, Simmaco M, De Biase L, Volpe M. Cardiac resynchronization therapy increases plasma levels of the endogenous inotrope apelin. Eur J Heart Fail. 2007;9:306-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
97.  Schäffler A, Neumeier M, Herfarth H, Fürst A, Schölmerich J, Büchler C. Genomic structure of human omentin, a new adipocytokine expressed in omental adipose tissue. Biochim Biophys Acta. 2005;1732:96-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 238]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
98.  Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, Shuldiner AR, Fried SK, McLenithan JC, Gong DW. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 2006;290:E1253-E1261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 583]  [Article Influence: 32.4]  [Reference Citation Analysis (0)]
99.  Pan HY, Guo L, Li Q. Changes of serum omentin-1 levels in normal subjects and in patients with impaired glucose regulation and with newly diagnosed and untreated type 2 diabetes. Diabetes Res Clin Pract. 2010;88:29-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 220]  [Article Influence: 15.7]  [Reference Citation Analysis (1)]
100.  de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, Ndubuizu K, Patil S, Schwartz A, Kligman M. Omentin plasma levels and gene expression are decreased in obesity. Diabetes. 2007;56:1655-1661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 506]  [Cited by in F6Publishing: 512]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
101.  Maruyama S, Shibata R, Kikuchi R, Izumiya Y, Rokutanda T, Araki S, Kataoka Y, Ohashi K, Daida H, Kihara S. Fat-derived factor omentin stimulates endothelial cell function and ischemia-induced revascularization via endothelial nitric oxide synthase-dependent mechanism. J Biol Chem. 2012;287:408-417.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Shang FJ, Wang JP, Liu XT, Zheng QS, Xue YS, Wang B, Zhao LY. Serum omentin-1 levels are inversely associated with the presence and severity of coronary artery disease in patients with metabolic syndrome. Biomarkers. 2011;16:657-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 64]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
103.  Shibata R, Ouchi N, Kikuchi R, Takahashi R, Takeshita K, Kataoka Y, Ohashi K, Ikeda N, Kihara S, Murohara T. Circulating omentin is associated with coronary artery disease in men. Atherosclerosis. 2011;219:811-814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
104.  Zhong X, Zhang HY, Tan H, Zhou Y, Liu FL, Chen FQ, Shang DY. Association of serum omentin-1 levels with coronary artery disease. Acta Pharmacol Sin. 2011;32:873-878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
105.  Yoo HJ, Hwang SY, Hong HC, Choi HY, Yang SJ, Seo JA, Kim SG, Kim NH, Choi KM, Choi DS. Association of circulating omentin-1 level with arterial stiffness and carotid plaque in type 2 diabetes. Cardiovasc Diabetol. 2011;10:103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 94]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
106.  Prats-Puig A, Bassols J, Bargalló E, Mas-Parareda M, Ribot R, Soriano-Rodríguez P, Berengüí A, Díaz M, de Zegher F, Ibánez L. Toward an early marker of metabolic dysfunction: omentin-1 in prepubertal children. Obesity (Silver Spring). 2011;19:1905-1907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
107.  Klöting N, Berndt J, Kralisch S, Kovacs P, Fasshauer M, Schön MR, Stumvoll M, Blüher M. Vaspin gene expression in human adipose tissue: association with obesity and type 2 diabetes. Biochem Biophys Res Commun. 2006;339:430-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 230]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
108.  Cho JK, Han TK, Kang HS. Combined effects of body mass index and cardio/respiratory fitness on serum vaspin concentrations in Korean young men. Eur J Appl Physiol. 2010;108:347-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 27]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
109.  Youn BS, Klöting N, Kratzsch J, Lee N, Park JW, Song ES, Ruschke K, Oberbach A, Fasshauer M, Stumvoll M. Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes. 2008;57:372-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 284]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
110.  Ye Y, Hou XH, Pan XP, Lu JX, Jia WP. Serum vaspin level in relation to postprandial plasma glucose concentration in subjects with diabetes. Chin Med J (Engl). 2009;122:2530-2533.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Li HL, Peng WH, Cui ST, Lei H, Wei YD, Li WM, Xu YW. Vaspin plasma concentrations and mRNA expressions in patients with stable and unstable angina pectoris. Clin Chem Lab Med. 2011;49:1547-1554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
112.  Aust G, Richter O, Rohm S, Kerner C, Hauss J, Klöting N, Ruschke K, Kovacs P, Youn BS, Blüher M. Vaspin serum concentrations in patients with carotid stenosis. Atherosclerosis. 2009;204:262-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]