Topic Highlight Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Cardiol. Jan 26, 2011; 3(1): 18-24
Published online Jan 26, 2011. doi: 10.4330/wjc.v3.i1.18
Effects of interventions on oxidative stress and inflammation of cardiovascular diseases
Sewon Lee, Yoonjung Park, Mozow Yusof Zuidema, Cuihua Zhang, Department of Internal Medicine, Medical Pharmacology and Physiology and Nutritional Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States
Mark Hannink, Department of Biochemistry, Life Sciences Fellowship Program, University of Missouri, Columbia, MO 65211, United States
Author contributions: Lee S, Park Y and Zuidema MY wrote the manuscript; Hannink M and Zhang C supervised this study, edited and revised the manuscript.
Correspondence to: Cuihua Zhang, MD, PhD, FAHA, Associate Professor, Department of Internal Medicine, Medical Pharmacology and Physiology and Nutrition and Exercise Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States. zhangcu@missouri.edu
Telephone: +1-573-8822427 Fax: +1-573-8844232
Received: August 3, 2010
Revised: December 2, 2010
Accepted: December 9, 2010
Published online: January 26, 2011

Abstract

Excessive oxidative stress and low-grade chronic inflammation are major pathophysiological factors contributing to the development of cardiovascular diseases (CVD) such as hypertension, diabetes and atherosclerosis. Accumulating evidence suggests that a compromised anti-oxidant system can lead to excessive oxidative stress in cardiovascular related organs, resulting in cell damage and death. In addition, increased circulating levels of pro-inflammatory cytokines, such as tumor necrosis factor α, interleukin-6 and C-reactive protein, are closely related to morbidity and mortality of cardiovascular complications. Emerging evidence suggests that interventions including nutrition, pharmacology and exercise may activate expression of cellular anti-oxidant systems via the nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 signaling pathway and play a role in preventing inflammatory processes in CVD. The focus of the present review is to summarize recent evidence showing the role of these anti-oxidant and anti-inflammatory interventions in cardiovascular disease. We believe that these findings may prompt new effective pathogenesis-oriented interventions, based on the exercise-induced protection from disease in the cardiovascular system, aimed at targeting oxidant stress and inflammation.

Key Words: Anti-oxidant, Exercise, Nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 signaling, Pro-inflammatory cytokines



INTRODUCTION

Cell damage that occurs by insults such as oxidative stress and toxicants may contribute to atherosclerosis, coronary artery disease, stroke, myocardial infarction, Alzheimer’s disease, Parkinson’s disease and cancer[1-5]. Of these diseases, excessive oxidative stress and chronic inflammation are both major characteristics of the pathology seen in type 2 diabetes (T2D), cardiovascular diseases (CVD) and the aging process[1,6]. Specifically, T2D and CVD are associated with increased production of reactive oxygen species (ROS) and compromised endogenous anti-oxidant defense. Oxidative stress is tightly regulated by a balance between production and removal of ROS. ROS are natural by-products of metabolism and these molecules play important roles in cell signaling. However, excessive levels of ROS can be toxic to cells, i.e. whenever the expression of anti-oxidant enzymes, including superoxide dismutases (SODs), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO-1), catalase and thioredoxin are not sufficient to control ROS and minimize ROS-induced damage[3]. A compromised anti-oxidant defense system can lead to excessive oxidative stress and ultimately result in cell damage[7-9].

Recent work has indicated that chronic inflammation is an important pathophysiological factor in the development of T2D and CVD, with increased circulating levels of pro-inflammatory cytokines, such as circulating C-reactive protein (CRP), tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β[10-14]. Opposing the pro-inflammatory cytokines, anti-inflammatory cytokines, such as IL-10 and adiponectin, are inversely correlated with the incidence of these diseases. These anti-inflammatory cytokines play a role in inhibiting the action of TNF-α on endothelial cell adhesion, reducing nuclear factor (NF)-κB activation, and delaying macrophage foam-cell development[15-18]. T2D and CVD are associated with aging and a sedentary lifestyle; however, emerging evidence suggests that the anti-inflammatory effects of exercise and/or physical activity can reduce mortality and morbidity of these patients[19-22]. However, the mechanism(s) that confer anti-inflammatory effects following an exercise training regimen have not been clearly identified.

This review addresses the effects of interventions, such as nutrition, pharmacology, genetics and exercise on anti-oxidant systems and on inflammation.

ROLE OF INTERVENTIONS IN ENDOGENOUS ANTIOXIDANT SIGNALING

The anti-oxidant defense system is regulated, in large part, by a transcription factor termed nuclear factor erythroid 2-related factor 2 (Nrf2), which is a member of the cap ‘n’ collar subfamily of the basic leucine zipper transcription factors[5]. Under normal physiological conditions, Nrf2 is bound to a cytoplasmic repressor, termed Kelch-like ECH-associated protein 1 (Keap1)[23]. Keap1 functions as a substrate adaptor for a Cullin3-dependent ubiquitin ligase and targets Nrf2 for degradation by the proteasome[24-26]. The substrate adaptor function of Keap1 is inactivated in response to a range of oxidative and electrophilic stimuli such as ROS, diethyl malonate and certain disease processes, resulting in the accumulation of Nrf2, which enters the nucleus and activates expression of anti-oxidant genes[5,9]. Although most investigators believe that Keap1-mediated repression occurs in the cytoplasm, several studies have shown that Nrf2 and Keap1 can shuttle between the nucleus and the cytoplasm[27-29]. In the nucleus, Nrf2 forms a heterodimer with members of the small musculo-aponeurotic fibrosarcoma (Maf) transcription factor family. These Nrf2/Maf heterodimers bind to antioxidant response elements present in the promoters of numerous anti-oxidant genes, including NQO-1, glutathione S-transferase, glutathione peroxidase (GPx), catalase and HO-1[5,9,30-32] (Figure 1). Nrf2 is widely expressed and it has been studied in many different tissues[7,33,34]. In the cardiovascular system, it has been shown that ischemia/reperfusion (I/R) down-regulates Nrf2 protein expression in rat heart and that aging decreases glutathione synthesis via diminished Nrf2 signaling in rat vascular endothelial and smooth muscle cells, suggesting that Nrf2 may play a critical role in the development of CVD in the aged population[6,35]. He et al[30] have shown a functionally decreased contractility when Nrf2 is genetically deleted from cardiomyocytes due to a marked increase in high-glucose oxidative stress and apoptosis.

Figure 1
Figure 1 The role of interventions in nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 signaling pathway. Nuclear factor erythroid 2-related factor 2 (Nrf2) can be activated by interventions such as nutrition (phytochemical, phenolic acids), pharmacology (MG132, H2S) and oxidative and electrophilic stimuli. Under basal conditions, Nrf2 is sequestered in the cytosol by binding with Kelch-like ECH-associated protein 1 (Keap1). On activation, Nrf2 can be released from Keap1 and translocated into the nucleus. Nrf2 forms a heterodimer with musculo-aponeurotic fibrosarcoma (Maf) and antioxidant response element (ARE) and regulates phase II anti-oxidant enzymes. NQO-1: NAD(P)H quinone oxidoreductase-1; GST: Glutathione S-transferase; GPx: Glutathione peroxidase; HO-1: Heme oxygenase-1.
Role of nutrition in antioxidant signaling

Numerous studies have indicated that increased oxidative stress may be involved in the pathogenesis of CVD. Several animal models suggest that when endogenous anti-oxidant systems are overwhelmed, exogenous anti-oxidant supplementation can be used for preventive and/or therapeutic intervention of oxidative cardiovascular disorders[35,36]. Phenolic acids are a group of compounds that are widely distributed in natural plant foods including fruits, vegetables and whole grains[36]. Yeh et al[36] have shown that 14 d of oral gavage (100 mg/kg) of phenolic acids in male rats increased anti-oxidant capacity via SOD-1, GPx and catalase, while HO-1 mRNA increased via Nrf2 signaling in the heart. Other phytochemicals, such as those found in broccoli sprouts may confer protection against cancer, although little is known about these effects on the cardiovascular system[37,38]. Recently, Mukherjee et al[35] have tested if daily consumption of broccoli, which contains sulforaphane and selenium for 1 mo could be beneficial to the heart. They have found that broccoli induced cardio-protection in I/R through the induction of HO-1[35].

Role of pharmacology and genetics in antioxidant signaling

The proteasome system uses a ubiquitin tag to activate the major intracellular protein degradation in eukaryotic cells[39]. The ubiquitin-proteasome system is critical for degradation of proteins related to the cell cycle and apoptosis[40,41]. In this sense, proteasome inhibition has been highlighted as a promising therapeutic target for treatment of human diseases. For instance, proteasome inhibitors have been proposed as an anti-inflammatory treatment via inhibition of NF-κB[42]. As steady-state levels of Nrf2 increase following proteasome inhibition, Dreger et al[39] have suggested that non-toxic inhibition of the ubiquitin-proteasome system by MG132 (0.5 μmol/L for 48 h) may contribute to protection of rat cardiomyocytes against oxidative stress via Nrf2-mediated transcriptional activation of anti-oxidants. Calvert et al[43] showed that hydrogen sulfide (H2S) may be an attractive pharmacological agent for the treatment of CVD by up-regulating anti-oxidants and anti-apoptogens. They showed that 100 μg/kg precondition by H2S in the form of sodium sulfide resulted in protection against myocardial I/R injury in a mouse model by increasing endogenous anti-oxidant defenses via an Nrf2-dependent manner. In this study, Nrf2 deficient mice showed an exacerbated injury in response to I/R, suggesting that Nrf2 may play an important cardio-protective role in heart disease[43]. On the other hand, Sussan et al[44] have shown that disruption of Nrf2 in apolipoprotein E (ApoE) knockout mice significantly decreased atherosclerotic plaque after 20 wk of high-fat diet. However, Nrf2 knock-out mice showed increased susceptibility to pulmonary emphysema, asthma and sepsis due to increased oxidative stress and inflammation[44]. This study suggested that Nrf2 might promote atherosclerotic plaque development through a mechanism separate from oxidative stress. More studies are required to fully understand the contribution of Nrf2 signaling in regards to atherosclerosis.

Role of exercise and physical activity in antioxidant processes

A sedentary lifestyle is a risk factor for T2D and CVD with several clinical studies illustrating a reduction of mortality and morbidity in physically active individuals compared to sedentary individuals[45-47]. Exercise or physical activity may contribute to improvement of insulin resistance via improved insulin action, improved vascular function via increase of nitric oxide (NO) bioavailability, and by increasing ROS-detoxification and decreasing ROS generation[48-53]. Since generation of ROS is a normal result of aerobic metabolism, it is efficiently removed by cellular anti-oxidant systems under physiological conditions. Several studies have shown that chronic exercise training increases SOD gene expression in vascular systems. Exercise training increased SOD-3 gene expression in mice aorta in NO-dependent manner and up-regulated SOD-1 in Yucatan miniature pig aortas[50,51]. Recently, Moien-Afshari et al[54] have suggested that low intensity exercise training increased SOD-1 protein expression, whereas moderate intensity increased SOD-2 gene expression in diabetic mice aorta with improved NO availability. Even though many studies have shown that exercise and physical activity up-regulated anti-oxidants such as SODs in cardiovascular systems, little is known about how exercise and physical activity may increase phase II anti-oxidant systems via the Nrf2-Keap1 signaling pathway[50,51,54,55]. Even though there are no clear studies to determine if exercise training may alter Nrf2 signaling, Niess et al[56] have shown that leukocytes from endurance trained athletes down-regulate the baseline expression of HO-1, presumably due to the adaptation mechanism of exercise training. Since HO-1 is an anti-oxidant protein that is mainly induced through the Nrf2-Keap1 signaling pathway, exercise training may down-regulate Nrf-2 signaling in humans. However, more studies are needed to further elucidate the effect of exercise on Nrf2 mechanisms in the cardiovascular system.

ROLE OF EXERCISE IN INFLAMMATION
Effect of acute exercise on inflammation

The effect of acute exercise on pro-inflammatory cytokines release has been a matter of considerable debate, since although a majority of studies have reported that acute exercise simulates release of inflammatory cytokine[57-61], some studies have also shown that acute exercise did not change levels of the pro-inflammatory cytokines TNF-α and IL-1β[58,61-63]. These discrepant findings suggest that the level of pro-inflammatory cytokines during and following exercise is dependent on several factors including the pathological condition, intensity and duration of exercise, and timing of sampling[64]. For example, plasma concentration and muscle mRNA expression of TNF-α are elevated in chronic obstructive pulmonary disease patients during continuous moderate-intensity exercise (for 11 min at 40% VO2max) whereas no change occurs in normal individuals[64]. Although the circulating level of TNF-α is not altered during low intensity and long duration of two-leg knee extensor exercise, short duration and high intensity of cycling exercise, approximately 80% VO2max, increases the circulating level of pro-inflammatory cytokines, IL-4, IL-6, TNF-α, interferon (IFN)-γ and anti-inflammatory cytokine such as IL-1β and IL-10[59,64]. Ostrowski et al[57] found that IL-6 and IL-1 receptor antagonist (IL-1ra) levels were enhanced during 2 h of continuous exercise (measured at every 30 min for 2 h) and following exercise, despite no change in the TNF-α level. Of these multiple pro-inflammatory cytokines, IL-6 is the most responsive cytokine that is increased during and following exercise and it is related to exercise intensity, duration, and muscle mass recruited[65,66]. Contracting skeletal muscle is one of the major sources of IL-6 produced during exercise. For example, during even moderate intensity of exercise (50% of maximal power output), 3 h of dynamic two-legged knee-extensor, muscle IL-6 mRNA expression and plasma concentration of IL-6 is increased 16-fold and 20-fold, respectively[67]. An even greater amount of IL-6 is produced in higher intensity and longer duration of exercise[66]. More interestingly, Petersen et al[66] suggest that IL-6 produced from working muscle can play a hormone-like role that stimulates lipolysis and fat oxidation in adipose tissue and induces gluconeogenesis in liver that may enhance exercise capacity. Moreover, IL-6 has been suggested as an anti-inflammatory cytokine because some studies have shown that an infusion of IL-6 decreases TNF-α production in healthy humans and stimulates the release of anti-inflammatory cytokines, IL-1ra and IL-10[68,69]. However, IL-6 is a well-established pro-inflammatory cytokine that is closely linked to various CVD and morbidity and mortality of several diseases. One possible explanation of a paradoxical role of IL-6 as an inflammatory cytokines and as a mediator of beneficial adaptation to exercise is the location of IL-6 production. Muscle contracting-induced local production of IL-6 may play a positive role in lipid and carbohydrate metabolism during exercise whereas systemic IL-6 may result in a negative consequence of tissue injury, chronic infection and diseases.

Effect of chronic exercise on inflammation

Exercise training and/or a high level of physical activity has a beneficial effect on inflammation through a reduction of pro-inflammatory cytokines and an increase in anti-inflammatory cytokines. Cross-sectional studies show lower plasma levels of IL-6, TNF-α and CRP while higher plasma levels of IL-10 and adiponectin occur in physically active individuals compared to physically inactive groups[16,70-72]. Exercise decreases pro-inflammatory cytokines and indicators of systemic inflammation. For example, long-term exercise (for 6 mo) significantly attenuates the production of pro-inflammatory cytokines, TNF-α and IFN-γ, and enhances the anti-inflammatory cytokine IL-10 in individuals at risk of developing ischemic heart disease[73]. Participation in an exercise training program for 6 mo in patients with stable chronic heart failure (CHF) significantly decreases the mRNA expression of TNF-α, IL-6, IL-1β in skeletal muscle, compared to the healthy individuals[74]. On the other hand, some studies demonstrate that the levels of pro-inflammatory cytokines are not significantly altered after exercise training in the obese individuals and healthy elderly[75-77]. This discrepancy may be derived from differences in experimental design and disease status of the subjects. The studies showing the effectiveness of exercise training on pro-inflammatory cytokines investigated the patients with severe disease conditions such as CHF and ischemic heart disease where basal levels of cytokines were already elevated compared to the healthy individuals before the exercise training[73,74]. In contrast, no apparent change in pro-inflammatory cytokines is shown in relatively less severe conditions, such moderate obesity (approximately 40% of % body fat) and aging (approximately 66 years old)[75-77]. Moreover, local change in inflammation after exercise training is an important factor to be considered. For example, mRNA expression of pro-inflammatory cytokines, TNF-α, IL-6, IL-1β in skeletal muscle are reduced after exercise training although the circulating levels of those cytokines are not changed[74]. This finding suggests that exercise training does not play a role in reducing systemic inflammation and is not effective enough to reduce the circulating levels of cytokines. However, regional expression of cytokines in skeletal muscle are affected. This regional reduction of pro-inflammatory cytokines in skeletal muscle may have a beneficial effect in skeletal muscles homeostasis despite the lack of effect on systemic inflammation.

Mechanisms of anti-inflammatory effect of exercise

As previously described, acute exercise stimulates production of pro-inflammatory cytokines and superoxide (O2-•) that can cause the tissue injury. Interestingly, exercise induced pro-inflammatory cytokines are triggers to generate the anti-inflammatory cytokines such as IL-10, IL-1ra and transforming growth factor β and the anti-oxidant, SOD-2 that have protective functions[58,60,63]. The major role of these cytokines is to recruit neutrophils and monocytes into injured tissue for repair[78]. During this process, anti-inflammatory cytokines and anti-oxidant mechanisms can be initiated and limit the inflammatory reaction in response to exercise. It is suggested that this stimulated anti-inflammatory mechanism, in turn, may down-regulate production of pro-inflammatory cytokines during and following exercise.

CONCLUSION

Oxidative stress plays a critical role in the pathology of CVD. Exogenous anti-oxidant supplementations such as broccoli, curcumin and phenolic acids as well as stimulators of endogenous pathways such as MG132, H2S and exercise seemed to be effective in providing cellular protection. However, large discrepancies are noted among several studies. For example, Sussan et al[44] have shown that double deletions of ApoE and Nrf2 genes in mice aortas showed a decrease in plaque area compared with ApoE knock-out mice in spite of the anti-oxidant effect of Nrf2. This suggests that upregulation of Nrf2 may play a detrimental role in generation of atherosclerosis. On the other hand, several studies have suggested beneficial roles of Nrf2 in the cardiovascular systems. These contradictory findings based on narrowly focused studies indicate that a broader understanding of Nrf2 is needed to understand the role of the oxidant/anti-oxidant system in cardiovascular disease. Physical activity and/or exercise training provides an ideal experimental context for further study of Nrf2 and other cytokines because acute exercise induces an increase in pro-inflammatory cytokine production that eventually stimulates anti-inflammatory responses to achieve an overall beneficial anti-inflammatory effect. The elucidation of the mechanisms governing exercise-induced protection from disease in the cardiovascular system is needed to devise more effective therapies.

Footnotes

Peer reviewers: Yuri V Bobryshev, PhD, Associate Professor, School of Medical Sciences, Faculty of Medicine University of New South Wales, Kensington NSW 2052, Australia; Gianluca Di Bella, MD, PhD, Clinical and Experimental Department of Medicine and Pharmacology, University of Messina, via Consolare Valeria N 1, CAP 98100, Messina, Italy

S- Editor Cheng JX L- Editor Negro F E- Editor Zheng XM

References
1.  Kojda G, Harrison D. Interactions between NO and reactive oxygen species: pathophysiological importance in atherosclerosis, hypertension, diabetes and heart failure. Cardiovasc Res. 1999;43:562-571.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Adams V, Linke A, Kränkel N, Erbs S, Gielen S, Möbius-Winkler S, Gummert JF, Mohr FW, Schuler G, Hambrecht R. Impact of regular physical activity on the NAD(P)H oxidase and angiotensin receptor system in patients with coronary artery disease. Circulation. 2005;111:555-562.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Gao L, Mann GE. Vascular NAD(P)H oxidase activation in diabetes: a double-edged sword in redox signalling. Cardiovasc Res. 2009;82:9-20.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Zhang Y, Gordon GB. A strategy for cancer prevention: stimulation of the Nrf2-ARE signaling pathway. Mol Cancer Ther. 2004;3:885-893.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Surh YJ, Kundu JK, Na HK. Nrf2 as a master redox switch in turning on the cellular signaling involved in the induction of cytoprotective genes by some chemopreventive phytochemicals. Planta Med. 2008;74:1526-1539.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ungvari Z, Parrado-Fernandez C, Csiszar A, de Cabo R. Mechanisms underlying caloric restriction and lifespan regulation: implications for vascular aging. Circ Res. 2008;102:519-528.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Lee JM, Li J, Johnson DA, Stein TD, Kraft AD, Calkins MJ, Jakel RJ, Johnson JA. Nrf2, a multi-organ protector? FASEB J. 2005;19:1061-1066.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Mann GE, Niehueser-Saran J, Watson A, Gao L, Ishii T, de Winter P, Siow RC. Nrf2/ARE regulated antioxidant gene expression in endothelial and smooth muscle cells in oxidative stress: implications for atherosclerosis and preeclampsia. Shengli Xuebao. 2007;59:117-127.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Motohashi H, Yamamoto M. Nrf2-Keap1 defines a physiologically important stress response mechanism. Trends Mol Med. 2004;10:549-557.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Picchi A, Gao X, Belmadani S, Potter BJ, Focardi M, Chilian WM, Zhang C. Tumor necrosis factor-alpha induces endothelial dysfunction in the prediabetic metabolic syndrome. Circ Res. 2006;99:69-77.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Gao X, Belmadani S, Picchi A, Xu X, Potter BJ, Tewari-Singh N, Capobianco S, Chilian WM, Zhang C. Tumor necrosis factor-alpha induces endothelial dysfunction in Lepr(db) mice. Circulation. 2007;115:245-254.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Hallenbeck JM. The many faces of tumor necrosis factor in stroke. Nat Med. 2002;8:1363-1368.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005;352:1685-1695.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Park Y, Capobianco S, Gao X, Falck JR, Dellsperger KC, Zhang C. Role of EDHF in type 2 diabetes-induced endothelial dysfunction. Am J Physiol Heart Circ Physiol. 2008;295:H1982-H1988.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Pajkrt D, van der Poll T, Levi M, Cutler DL, Affrime MB, van den Ende A, ten Cate JW, van Deventer SJ. Interleukin-10 inhibits activation of coagulation and fibrinolysis during human endotoxemia. Blood. 1997;89:2701-2705.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Jankord R, Jemiolo B. Influence of physical activity on serum IL-6 and IL-10 levels in healthy older men. Med Sci Sports Exerc. 2004;36:960-964.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  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: ]
18.  Ouchi N, Kihara S, Arita Y, Okamoto Y, Maeda K, Kuriyama H, Hotta K, Nishida M, Takahashi M, Muraguchi M. Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation. 2000;102:1296-1301.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Albert MA, Glynn RJ, Ridker PM. Effect of physical activity on serum C-reactive protein. Am J Cardiol. 2004;93:221-225.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Bruun JM, Helge JW, Richelsen B, Stallknecht B. Diet and exercise reduce low-grade inflammation and macrophage infiltration in adipose tissue but not in skeletal muscle in severely obese subjects. Am J Physiol Endocrinol Metab. 2006;290:E961-E967.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Flynn MG, McFarlin BK, Markofski MM. State of the art reviews: The anti-inflammatory actions of exercise training. Am J Lifestyle Med. 2007;1:220-235.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Flynn MG, McFarlin BK, Phillips MD, Stewart LK, Timmerman KL. Toll-like receptor 4 and CD14 mRNA expression are lower in resistive exercise-trained elderly women. J Appl Physiol. 2003;95:1833-1842.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, Yamamoto M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. G. enes Dev. 1999;13:76-86.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Zhang DD, Lo SC, Cross JV, Templeton DJ, Hannink M. Keap1 is a redox-regulated substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex. Mol Cell Biol. 2004;24:10941-10953.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Cullinan SB, Gordan JD, Jin J, Harper JW, Diehl JA. The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: oxidative stress sensing by a Cul3-Keap1 ligase. Mol Cell Biol. 2004;24:8477-8486.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, Igarashi K, Yamamoto M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol. 2004;24:7130-7139.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Nguyen T, Sherratt PJ, Nioi P, Yang CS, Pickett CB. Nrf2 controls constitutive and inducible expression of ARE-driven genes through a dynamic pathway involving nucleocytoplasmic shuttling by Keap1. J Biol Chem. 2005;280:32485-32492.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Velichkova M, Hasson T. Keap1 regulates the oxidation-sensitive shuttling of Nrf2 into and out of the nucleus via a Crm1-dependent nuclear export mechanism. Mol Cell Biol. 2005;25:4501-4513.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Sun Z, Zhang S, Chan JY, Zhang DD. Keap1 controls postinduction repression of the Nrf2-mediated antioxidant response by escorting nuclear export of Nrf2. Mol Cell Biol. 2007;27:6334-6349.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  He X, Kan H, Cai L, Ma Q. Nrf2 is critical in defense against high glucose-induced oxidative damage in cardiomyocytes. J Mol Cell Cardiol. 2009;46:47-58.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Kobayashi M, Yamamoto M. Nrf2-Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species. Adv Enzyme Regul. 2006;46:113-140.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Zhang DD. Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab Rev. 2006;38:769-789.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Xue M, Qian Q, Adaikalakoteswari A, Rabbani N, Babaei-Jadidi R, Thornalley PJ. Activation of NF-E2-related factor-2 reverses biochemical dysfunction of endothelial cells induced by hyperglycemia linked to vascular disease. Diabetes. 2008;57:2809-2817.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Balogun E, Hoque M, Gong P, Killeen E, Green CJ, Foresti R, Alam J, Motterlini R. Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Biochem J. 2003;371:887-895.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Mukherjee S, Gangopadhyay H, Das DK. Broccoli: a unique vegetable that protects mammalian hearts through the redox cycling of the thioredoxin superfamily. J Agric Food Chem. 2008;56:609-617.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Yeh CT, Ching LC, Yen GC. Inducing gene expression of cardiac antioxidant enzymes by dietary phenolic acids in rats. J Nutr Biochem. 2009;20:163-171.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Matusheski NV, Swarup R, Juvik JA, Mithen R, Bennett M, Jeffery EH. Epithiospecifier protein from broccoli (Brassica oleracea L. ssp. italica) inhibits formation of the anticancer agent sulforaphane. J Agric Food Chem. 2006;54:2069-2076.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Finley JW. Reduction of cancer risk by consumption of selenium-enriched plants: enrichment of broccoli with selenium increases the anticarcinogenic properties of broccoli. J Med Food. 2003;6:19-26.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Dreger H, Westphal K, Weller A, Baumann G, Stangl V, Meiners S, Stangl K. Nrf2-dependent upregulation of antioxidative enzymes: a novel pathway for proteasome inhibitor-mediated cardioprotection. Cardiovasc Res. 2009;83:354-361.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Naujokat C, Hoffmann S. Role and function of the 26S proteasome in proliferation and apoptosis. Lab Invest. 2002;82:965-980.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Jesenberger V, Jentsch S. Deadly encounter: ubiquitin meets apoptosis. Nat Rev Mol Cell Biol. 2002;3:112-121.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Elliott PJ, Zollner TM, Boehncke WH. Proteasome inhibition: a new anti-inflammatory strategy. J Mol Med. 2003;81:235-245.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Calvert JW, Jha S, Gundewar S, Elrod JW, Ramachandran A, Pattillo CB, Kevil CG, Lefer DJ. Hydrogen sulfide mediates cardioprotection through Nrf2 signaling. Circ Res. 2009;105:365-374.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Sussan TE, Jun J, Thimmulappa R, Bedja D, Antero M, Gabrielson KL, Polotsky VY, Biswal S. Disruption of Nrf2, a key inducer of antioxidant defenses, attenuates ApoE-mediated atherosclerosis in mice. PLoS One. 2008;3:e3791.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Manson JE, Greenland P, LaCroix AZ, Stefanick ML, Mouton CP, Oberman A, Perri MG, Sheps DS, Pettinger MB, Siscovick DS. Walking compared with vigorous exercise for the prevention of cardiovascular events in women. N Engl J Med. 2002;347:716-725.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Niebauer J, Hambrecht R, Velich T, Hauer K, Marburger C, Kälberer B, Weiss C, von Hodenberg E, Schlierf G, Schuler G. Attenuated progression of coronary artery disease after 6 years of multifactorial risk intervention: role of physical exercise. Circulation. 1997;96:2534-2541.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Lindström J, Louheranta A, Mannelin M, Rastas M, Salminen V, Eriksson J, Uusitupa M, Tuomilehto J. The Finnish Diabetes Prevention Study (DPS): Lifestyle intervention and 3-year results on diet and physical activity. Diabetes Care. 2003;26:3230-3236.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Green DJ, Maiorana A, O'Driscoll G, Taylor R. Effect of exercise training on endothelium-derived nitric oxide function in humans. J Physiol. 2004;561:1-25.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Wang J, Wolin MS, Hintze TH. Chronic exercise enhances endothelium-mediated dilation of epicardial coronary artery in conscious dogs. Circ Res. 1993;73:829-838.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Fukai T, Siegfried MR, Ushio-Fukai M, Cheng Y, Kojda G, Harrison DG. Regulation of the vascular extracellular superoxide dismutase by nitric oxide and exercise training. J Clin Invest. 2000;105:1631-1639.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Rush JW, Turk JR, Laughlin MH. Exercise training regulates SOD-1 and oxidative stress in porcine aortic endothelium. Am J Physiol Heart Circ Physiol. 2003;284:H1378-H1387.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Laughlin MH, McAllister RM. Exercise training-induced coronary vascular adaptation. J Appl Physiol. 1992;73:2209-2225.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Trott DW, Gunduz F, Laughlin MH, Woodman CR. Exercise training reverses age-related decrements in endothelium-dependent dilation in skeletal muscle feed arteries. J Appl Physiol. 2009;106:1925-1934.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Moien-Afshari F, Ghosh S, Elmi S, Rahman MM, Sallam N, Khazaei M, Kieffer TJ, Brownsey RW, Laher I. Exercise restores endothelial function independently of weight loss or hyperglycaemic status in db/db mice. Diabetologia. 2008;51:1327-1337.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Moien-Afshari F, Ghosh S, Elmi S, Khazaei M, Rahman MM, Sallam N, Laher I. Exercise restores coronary vascular function independent of myogenic tone or hyperglycemic status in db/db mice. Am J Physiol Heart Circ Physiol. 2008;295:H1470-H1480.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Niess AM, Passek F, Lorenz I, Schneider EM, Dickhuth HH, Northoff H, Fehrenbach E. Expression of the antioxidant stress protein heme oxygenase-1 (HO-1) in human leukocytes. Free Radic Biol Med. 1999;26:184-192.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Ostrowski K, Hermann C, Bangash A, Schjerling P, Nielsen JN, Pedersen BK. A trauma-like elevation of plasma cytokines in humans in response to treadmill running. J Physiol. 1998;513:889-894.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Ostrowski K, Rohde T, Asp S, Schjerling P, Pedersen BK. Pro- and anti-inflammatory cytokine balance in strenuous exercise in humans. J Physiol. 1999;515:287-291.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Zaldivar F, Wang-Rodriguez J, Nemet D, Schwindt C, Galassetti P, Mills PJ, Wilson LD, Cooper DM. Constitutive pro- and anti-inflammatory cytokine and growth factor response to exercise in leukocytes. J Appl Physiol. 2006;100:1124-1133.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Nieman DC, Henson DA, Smith LL, Utter AC, Vinci DM, Davis JM, Kaminsky DE, Shute M. Cytokine changes after a marathon race. J Appl Physiol. 2001;91:109-114.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Steensberg A, Keller C, Starkie RL, Osada T, Febbraio MA, Pedersen BK. IL-6 and TNF-alpha expression in, and release from, contracting human skeletal muscle. Am J Physiol Endocrinol Metab. 2002;283:E1272-E1278.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Febbraio MA, Steensberg A, Starkie RL, McConell GK, Kingwell BA. Skeletal muscle interleukin-6 and tumor necrosis factor-alpha release in healthy subjects and patients with type 2 diabetes at rest and during exercise. Metabolism. 2003;52:939-944.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Drenth JP, Van Uum SH, Van Deuren M, Pesman GJ, Van der Ven-Jongekrijg J, Van der Meer JW. Endurance run increases circulating IL-6 and IL-1ra but downregulates ex vivo TNF-alpha and IL-1 beta production. J Appl Physiol. 1995;79:1497-1503.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Rabinovich RA, Figueras M, Ardite E, Carbó N, Troosters T, Filella X, Barberà JA, Fernandez-Checa JC, Argilés JM, Roca J. Increased tumour necrosis factor-alpha plasma levels during moderate-intensity exercise in COPD patients. Eur Respir J. 2003;21:789-794.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Febbraio MA, Pedersen BK. Muscle-derived interleukin-6: mechanisms for activation and possible biological roles. FASEB J. 2002;16:1335-1347.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Petersen AM, Pedersen BK. The anti-inflammatory effect of exercise. J Appl Physiol. 2005;98:1154-1162.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Fischer CP, Hiscock NJ, Penkowa M, Basu S, Vessby B, Kallner A, Sjöberg LB, Pedersen BK. Supplementation with vitamins C and E inhibits the release of interleukin-6 from contracting human skeletal muscle. J Physiol. 2004;558:633-645.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans. FASEB J. 2003;17:884-886.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Steensberg A, Fischer CP, Keller C, Møller K, Pedersen BK. IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. Am J Physiol Endocrinol Metab. 2003;285:E433-E437.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Pischon T, Hankinson SE, Hotamisligil GS, Rifai N, Rimm EB. Leisure-time physical activity and reduced plasma levels of obesity-related inflammatory markers. Obes Res. 2003;11:1055-1064.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Panagiotakos DB, Kokkinos P, Manios Y, Pitsavos C. Physical activity and markers of inflammation and thrombosis related to coronary heart disease. Prev Cardiol. 2004;7:190-194.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Colbert LH, Visser M, Simonsick EM, Tracy RP, Newman AB, Kritchevsky SB, Pahor M, Taaffe DR, Brach J, Rubin S. Physical activity, exercise, and inflammatory markers in older adults: findings from the Health, Aging and Body Composition Study. J Am Geriatr Soc. 2004;52:1098-1104.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Smith JK, Dykes R, Douglas JE, Krishnaswamy G, Berk S. Long-term exercise and atherogenic activity of blood mononuclear cells in persons at risk of developing ischemic heart disease. JAMA. 1999;281:1722-1727.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Gielen S, Adams V, Möbius-Winkler S, Linke A, Erbs S, Yu J, Kempf W, Schubert A, Schuler G, Hambrecht R. Anti-inflammatory effects of exercise training in the skeletal muscle of patients with chronic heart failure. J Am Coll Cardiol. 2003;42:861-868.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Marcell TJ, McAuley KA, Traustadóttir T, Reaven PD. Exercise training is not associated with improved levels of C-reactive protein or adiponectin. Metabolism. 2005;54:533-541.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Nassis GP, Papantakou K, Skenderi K, Triandafillopoulou M, Kavouras SA, Yannakoulia M, Chrousos GP, Sidossis LS. Aerobic exercise training improves insulin sensitivity without changes in body weight, body fat, adiponectin, and inflammatory markers in overweight and obese girls. Metabolism. 2005;54:1472-1479.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Hammett CJ, Oxenham HC, Baldi JC, Doughty RN, Ameratunga R, French JK, White HD, Stewart RA. Effect of six months' exercise training on C-reactive protein levels in healthy elderly subjects. J Am Coll Cardiol. 2004;44:2411-2413.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Nieman DC. Immune response to heavy exertion. J Appl Physiol. 1997;82:1385-1394.  [PubMed]  [DOI]  [Cited in This Article: ]