Inflammation and the Pathogenesis of Atrial Fibrillation

14
NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 1 Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, National Yang‑Ming University, Number 201, Section 2, Shipai Road, Beitou District, Taipei 11217, Taiwan, Republic of China (Y.‑F.H., Y.‑J.L., S.‑A.C.). Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Number 111, Section 3, Hsing‑Long Road, Taipei 11696, Taiwan, Republic of China (Y.‑J.C.) Correspondence to: S.‑A.C. epsachen@ ms41.hinet.net Inflammation and the pathogenesis of atrial fibrillation Yu-Feng Hu, Yi-Jen Chen, Yenn-Jiang Lin and Shih-Ann Chen Abstract | Atrial fibrillation (AF) is the most common cardiac arrhythmia. However, the development of preventative therapies for AF has been disappointing. The infiltration of immune cells and proteins that mediate the inflammatory response in cardiac tissue and circulatory processes is associated with AF. Furthermore, the presence of inflammation in the heart or systemic circulation can predict the onset of AF and recurrence in the general population, as well as in patients after cardiac surgery, cardioversion, and catheter ablation. Mediators of the inflammatory response can alter atrial electrophysiology and structural substrates, thereby leading to increased vulnerability to AF. Inflammation also modulates calcium homeostasis and connexins, which are associated with triggers of AF and heterogeneous atrial conduction. Myolysis, cardiomyocyte apoptosis, and the activation of fibrotic pathways via fibroblasts, transforming growth factor‑ β and matrix metalloproteases are also mediated by inflammatory pathways, which can all contribute to structural remodelling of the atria. The development of thromboembolism, a detrimental complication of AF, is also associated with inflammatory activity. Understanding the complex pathophysiological processes and dynamic changes of AF‑associated inflammation might help to identify specific anti‑inflammatory strategies for the prevention of AF. Hu, Y.‑F. et al. Nat. Rev. Cardiol. advance online publication 27 January 2015; doi:10.1038/nrcardio.2015.2 Introduction Atrial fibrillation (AF) is the most common cardiac arrhythmia and is associated with detrimental conse- quences. In addition to worsening patient quality of life, AF is associated with stroke, heart failure, and increased mortality. 1 Worldwide, AF has affected >30 million indi- viduals since 2010, and the incidence of AF continues to increase. 2,3 Current treatments for AF include pre- venting its recurrence (via rhythm control) and conse- quences (by rate control and antithrombosis). 4,5 In many patients, heart-rate control is sufficient to control a rapid rhythm and its associated symptoms, and the preven- tion of AF recurrence relies primarily on antiarrhythmic drugs. Catheter ablation is considered as an alternative to antiarrhythmic drugs because of its superiority to medical therapy for the maintenance of sinus rhythm. 1,5,6 Although major progress has been made in the treatment of AF, its recurrence and subsequent treatment after medication or catheter ablation, including any associ- ated complications, problems remain. 1,4,5,7 An improved understanding of the pathophysiology underlying AF and subsequent remodelling is necessary for the devel- opment of novel therapeutic approaches. Increasing evidence supports the role of inflammation in the pathophysiology of AF, which suggests that the inflam- matory process is a potential therapeutic target. 8,9 The main pathophysiological mechanisms contributing to AF development and progression include both electrical and structural remodelling of the atria. Moreover, AF itself can induce inflammation during atrial remodel- ling, which perpetuates the arrhythmia—the so-called ‘AF begets AF’ phenomenon. Instead of emphasizing the clinical correlations of inflammation in different scenarios of AF, 8,10 in this Review we discuss the patho- physiological role of inflammation and its interaction with the established mechanisms of AF. We also highlight potential inflammation-based therapeutic options. Sources of inflammation in AF Inflammation in patients with AF can arise from dif- ferent sources, which might have underlying inflam- matory mechanisms and temporal changes (Figure 1). Many systemic diseases (such as coronary artery disease, hypertension, and obesity) are associated with low-grade inflammation and increased levels of proinflammatory cytokines. 11–13 Obesity Obesity is associated with new-onset AF in the general population or in patients after cardiac surgery. 14,15 Obesity-induced immune cell infiltration into the adipose tissue, particularly by M1 macrophages (a pro- inflammatory phenotype), 16,17 as well as inflammation of adipose tissue and secreted proinflammatory cytokines occurs in patients with obesity. 17–19 High levels of inflam- matory activity in the pericardial adipose tissue has been described in patients with AF. 20 For example, epicardial inflammatory activity was 35% higher in 21 patients with Competing interests The authors declare no competing interests. REVIEWS © 2015 Macmillan Publishers Limited. All rights reserved

description

Good

Transcript of Inflammation and the Pathogenesis of Atrial Fibrillation

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 1

    Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, National YangMing University, Number 201, Section2, Shipai Road, Beitou District, Taipei11217, Taiwan, Republic of China (Y.F.H., Y.J.L., S.A.C.). Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Number111, Section3, HsingLong Road, Taipei11696, Taiwan, Republic ofChina (Y.J.C.)

    Correspondence to: S.A.C. epsachen@ ms41.hinet.net

    Inflammation and the pathogenesis ofatrialfibrillationYu-Feng Hu, Yi-Jen Chen, Yenn-Jiang Lin and Shih-Ann Chen

    Abstract | Atrial fibrillation (AF) is the most common cardiac arrhythmia. However, the development of preventative therapies for AF has been disappointing. The infiltration of immune cells and proteins that mediate the inflammatory response in cardiac tissue and circulatory processes is associated with AF. Furthermore, the presence of inflammation in the heart or systemic circulation can predict the onset of AFand recurrence in the general population, as well as in patients after cardiac surgery, cardioversion, and catheter ablation. Mediators of the inflammatory response can alter atrial electrophysiology and structural substrates, thereby leading to increased vulnerability to AF. Inflammation also modulates calcium homeostasis and connexins, which are associated with triggers of AF and heterogeneous atrial conduction. Myolysis, cardiomyocyte apoptosis, and the activation of fibrotic pathways via fibroblasts, transforming growth factor and matrix metalloproteases are also mediated by inflammatory pathways, which can all contribute to structural remodelling of the atria. The development of thromboembolism, a detrimental complication of AF, is also associated with inflammatory activity. Understanding the complex pathophysiological processes and dynamic changes of AFassociated inflammation might help to identify specific antiinflammatory strategies forthe prevention of AF.

    Hu, Y.F. etal. Nat. Rev. Cardiol. advance online publication 27 January 2015; doi:10.1038/nrcardio.2015.2

    IntroductionAtrial fibrillation (AF) is the most common cardiac arrhythmia and is associated with detrimental conse-quences. In addition to worsening patient quality of life, AF is associated with stroke, heart failure, and increased mortality.1 Worldwide, AF has affected >30million indi-viduals since 2010, and the incidence of AF continues to increase.2,3 Current treatments for AF include pre-venting its recurrence (via rhythm control) and conse-quences (by rate control and antithrombosis).4,5 In many patients, heart-rate control is sufficient to control a rapid rhythm and its associated symptoms, and the preven-tion of AF recurrence relies primarily on antiarrhythmic drugs. Catheter ablation is considered as an alternative to antiarrhythmic drugs because of its superiority to medical therapy for the maintenance of sinus rhythm.1,5,6 Although major progress has been made in the treatment of AF, its recurrence and subsequent treatment after medication or catheter ablation, including any associ-ated complications, problems remain.1,4,5,7 An improved understanding of the pathophysiology underlying AF and subsequent remodelling is necessary for the devel-opment of novel therapeutic approaches. Increasing evidence supports the role of inflammation in the pathophysiology of AF, which suggests that the inflam-matory process is a potential therapeutic target.8,9 The main pathophysiological mechanisms contributing to AF development and progression include both electrical

    and structural remodelling of the atria. Moreover, AF itself can induce inflammation during atrial remodel-ling, which perpetuates the arrhythmiathe so-called AF begets AF phenomenon. Instead of emphasizing the clinical correlations of inflammation in different scenarios of AF,8,10 in this Review we discuss the patho-physiological role of inflammation and its interaction with the established mechanisms of AF. We also highlight p otential inflammation-based therapeutic options.

    Sources of inflammation in AFInflammation in patients with AF can arise from dif-ferent sources, which might have underlying inflam-matory mechanisms and temporal changes (Figure1). Many systemic diseases (such as coronary artery disease, hypertension, and obesity) are associated with low-grade in flammationand increased levels of proinflammatory cytokines.1113

    ObesityObesity is associated with new-onset AF in the general population or in patients after cardiac surgery.14,15 Obesity-induced immune cell infiltration into the adipose tissue, particularly by M1 macrophages (a pro-inflammatory phenotype),16,17 as well as inflammation of adipose tissue and secreted proinflammatory cytokines occurs in patients with obesity.1719 High levels of inflam-matory activity in the pericardial adipose tissue has been described in patients with AF.20 For example, epicardial inflammatory activity was 35% higher in 21 patients with

    Competing interestsThe authors declare no competing interests.

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

    mailto:[email protected]:[email protected]://www.nature.com/doifinder/10.1038/nrcardio.2015.2

  • 2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    AF than in 21 matched controls without AF, as meas-ured by 18F-fluorodeoxyglucose PET.20 Proinflammatory cytokines from adipose tissue might reach the atrium via the circulation or paracrine factors. In a study of 34 patients, high-dominant frequencies or complex atrial fractionated electrogram sites were located adjacent to epicardial fat areas, which suggest that epicardial fat might maintain AF by releasing paracrine inflammatory mediators.21,22 Free fatty acid overload in patients with obesity induces lipid accumulation within cardiomyo-cytes and apoptosis, which might also trigger regional inflammation.23 These factors suggest that inflammation is an important pathophysiological mechanism of AF in patients with obesity.

    HypertensionThe association between inflammation and AF in patients with hypertension is not yet established. In spon-taneously hypertensive rats and hypertensive sheep (who received unilateral nephrectomy followed by clamping of the remaining renal artery to 60%), leucocyte infiltration into the atria and inflammation was observed, which was

    Key points

    Inflammation and its associated immune response are involved in the initiation and maintenance of atrial fibrillation (AF)

    AF can further promote inflammation, which contributes to the clinical phenomenon of AF begets AF

    Inflammatory pathways contribute to both electrical and structural atrial remodelling and thrombogenesis in patients with AF

    The mechanisms and dynamic changes that underlie the inflammatory responses in different clinical scenarios of AF should be determined to enable the development of specific, individualized antiinflammatory strategies

    Therapies that target specific inflammatory cascades might be potential therapeutic strategies for the prevention of AF

    followed by atrial fibrosis.24,25 In these models, vulner-ability to the development of AF increased by 62% after the development of atrial fibrosis, but not inflamma-tion alone. However, in another sheep model, in which hypertension was induced by prenatal steroids, increased vulner ability to AF was noted, but atrial inflammation not observed.26 In this model, plasma renin concentra-tion and vascular reactivity to angiotensinII were not altered, whereas in spontaneously hypertensive rats and sheep with unilateral nephrectomy-induced hyperten-sion, the reninangiotensinaldosterone system (RAAS) was activated.2426 The discrepancy between these studies might imply a pathophysiological role of RAAS in AF.

    The atria of angiotensinII-treated mice are character-ized by increased neutrophil infiltration, which is depend-ent on CD11b and CD18 integrins.27 AngiotensinII increases inflammation by stimulating the production of proinflammatory cytokines (such as IL-6, IL-8, and tumour necrosis factor [TNF]) and directly activating immune cells.28 Furthermore, angiotensinII can induce the expression of adhesion molecules such as vascular cell adhesion protein1, intercellular adhesion molecule1, selectins, or CC motif chemokine2 (also known as monocyte chemotactic protein1), which promote the recruitment of immune cells.28 Blocking angiotensin-induced inflammatory cascades (such as TNF and IL-1) might prevent cardiac damage in response to angio-tensinII in a mouse model of AF.29 Several mechanistic hypotheses in addition to the role of RAAS have also been proposed. Atrial stretch, owing to elevated left ventricular diastolic pressure in patients with hypertension, might activate regional RAAS, cardiac apoptosis, and oxidative stress, which can subsequently induce regional inflamma-tion in the heart.30 Cellular stress from reactive oxidative species might be induced by hypertension in patients in AF,31 and reactive oxygen species can further stimulate signal trans duction thereby increasing production of proinflammatory cytokines, such as IL-1, IL-6, and TNF.32

    Coronary artery diseaseAtrial myocardial infarction, or ischaemia that is sec-ondary to an occluded coronary artery, is expected to induce myocardial damage and atrial inflammation during the healing process, and might consequently induce AF.33 The pathophysiology of ischaemic heart disease is more complex than that of occluded arteries, in which low-grade inflammation is an important factor.13 For example, the increased expression of platelet-bound and plasma stromal cell-derived factor1 was observed in patients with AF and ischaemic heart disease com-pared with those in sinus rhythm.34 The increased level of stromal cell-derived factor1 is a risk factor for devel-oping coronary artery disease and is associated with i nflammatory cell recruitment.35,36

    Surgery and ablationInflammation can also be induced by cardiac surgery or catheter ablation. In the ARMYDA-3 study,37 high postoperative C-reactive protein (CRP) levels were associated with an increased risk of AF. Surgery-induced

    Systemic diseaseObesity, hypertension, coronary artery diseases

    Atrial myocardial injuryAtrial ischaemia or infarction, surgical or catheter ablation

    Immune diseases with autoantibodyValvular heart diseases

    Modulating factorsChronic viral infection,

    epicardial fat,genetic predisposition

    Mediator moleculesROS, Ang II, TGF-, MPO, PDGF, HSPs, proinammatory cytokines

    Inammation

    Electrical remodelling Structural remodelling

    Atrial brillation

    Nature Reviews | CardiologyFigure 1 | Sources of inflammation in patients with atrial fibrillation. Activatedinflammatory pathways alter the electrophysiology, structure, and autonomic remodelling of the atria. Inflammation induced by atrial fibrillation canestablish an inflammatory cycle that leads to increased severity of the arrhythmia. Abbreviations: AngII, angiotensinII; HSP, heat shock protein; MPO, myeloperoxidase; PDGF, plateletderived growth factor; ROS, reactive oxygen species; TGF, transforming growth factor.

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 3

    inflammation has been modelled in canine sterile peri-carditis, an experimental model of postoperative AF in humans. AF, induced by burst atrial pacing after open chest operation in these dogs, was reduced by 60% after treatment with anti-inflammatory drugs.38,39 Radiofrequency ablation induces an inflammatory response that develops after thermal injury and is likely to contribute to the maturation of ablation lesions after the procedure.5,40 Patients undergoing radiofrequency ablation for AF have high CRP levels within 3days of the procedure, and the extent of CRP elevation predicts early AF recurrence within this 3-day period, but not late AF recurrence at 3 or 6months.41,42

    Autoimmune reactions in AFWhether the inflammation in AF is associated with an autoimmune response remains unclear. Certain auto antibodies, such as those against the muscarinic acetylcholine receptorM2 and heat shock proteins (HSPs), have also been associated with AF.4345 However, whether these autoantibodies cause or are only released in response to AF is unknown. Valvular heart diseases are associated with volume or pressure overload in the atrium and also increase atrial stretch, which activates RAAS, reactive oxygen species, matrix metalloprotein-ases (MMPs), cardiac myolysis, and apoptosis, and might subsequently increase inflammation and vulnerability toAF.30

    Polymorphisms in the genes encoding IL-1, IL-6, and IL-10, which are responsible for modulating expression levels of inflammatory cytokines, are independently associated with AF in humans.4649 For example, the 174G>C IL-6 polymorphism is associated with the new onset of AF after surgery.50 In two large cohort studies of patients with viral infection, such as HIV or herpes simplex virus, both latent and chronic viral infection were independently associated with the devel-opment of AF, possibly through inflammatory pro-cesses.51,52 However, the underlying mechanisms of AF in patients with a viral infection remain unclear. In indi-vidualswithHIV, several mechanisms were proposed to explain the HIV-induced dilated cardiomyopathy. For example, theendothelium in the heart can act as a reservoir of HIV particles and cytokines, such as TNF and IL-6, and reactive oxygen species, which all increase inflammation.53 Moreover, HIV-associated proteins, such as immunodeficiency virus transactivating regulatory protein (Tat), can lead to destruction of mitochondria, which results in myocardial damage.54

    Inflammation leads to AFIn patients with lone AF, atrial pathology reveals infil-tration of lymphomononuclear cells and necrosis of the adjacent myocytes, which is not present in patients who are in sinus rhythm.55 In a number of case-controlled studies, higher levels of inflammatory markers (such as CRP, HSP 1 [commonly known as HSP27], IL-6, IL-8, and TNF) as well as elevated neutrophil and lymphocyte ratios have been reported in patients with AF compared with those in sinus rhythm.8,10,5659 Increased CRP levels

    have been reported to predict the development of new-onset AF in several large, prospective cohorts.6062 In the Cardiovascular Health Study60 (5,806 patients followed up for a mean period of 6.9years), higher CRP levels (>3.41 mg/l) were associated with the presence of AF compared with lower levels (

  • 4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    Graded increases in the levels of CRP, HSP27, and TNF are also observed in patients with persistent AF compared with those with paroxysmal AF or without arrhythmias.57,68,69 The maintenance of sinus rhythm after cardioversion or catheter ablation of persistent or long-lasting AF leads to a gradual decrease in CRP levels relative to levels before the procedure (from 0.29 0.13 mg/dl to 0.10 0.06 mg/dl; P 50% (P 30% (P 200units) could be used to predict AF recur-rence after catheter ablation (HR4.2, 95%CI 1.214.6, P = 0.02).90 A higher percentage of activated Tlympho-cytes (CD3+ and HLA-DR+) was observed in the periph-eral blood of patients with paroxysmal or persistent AF compared with healthy control individuals (36% versus 27%; P

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 5

    lymphocytes (3.0 in mice with AF versus 2.4 in mice without AF; P = 0.001) in the peripheral circulation has been associated with an increased incidence of new-onset AF (OR1.10 per unit increase, P = 0.04).95 An elevated neutrophil-to-lymphocyte ratio before or after catheter ablation is associated with increased AF r ecurrence after the procudure.96,97

    The contributions of acute and chronic inflammation in AF, which might mediate distinct inflammatory cascades and signals, remain poorly understood. Atrial neutrophil infiltration is mediated by CD11bintegrin in patients with AF.27 Myeloperoxidase is most abundantly expressed in neutrophil granulocytes.98 In patients undergoing off-pump CABG surgery, levels of IL-6 and IL-8 (but not TNF) are elevated immediately after surgery (IL-6 from 0 to 435 pg/ml; IL-8 from 10 to 50 pg/ml).99 The increase in the level of IL-6 after surgery is associated with postopera-tive AF (OR7.63 if IL-6 >401 pg/ml, P = 0.04).99 Among patients with AF who receive catheter ablation, the levels of IL-6 and CRP both significantly increase after ablation (IL-6 from 1.1 2.5 to 12.4 15.3 pg/ml; P = 0.007; CRP from 2.4 2.9 to 20.1 9.2 mg/l, P = 0.001); however, levels of IL-8, IL-10, IL-12, stromal cell-derived factor1, and TNF remain unchanged.100

    In addition to their role in allergic and immune responses, mast cells also participate in cardiovascu-lar disease-related inflammation.101 Mast cells might actively induce inflammation and atrial fibrosis in patients with AF and secrete platelet-derived growth factorA (PDGF-A) and promote cell proliferation and collagen expression in cardiac fibroblasts (Figure2).94 Cardiomyocytes, fibroblasts, and endothelial cells can also induce inflammatory responses;85 however, whether innate and adaptive immune responses lead to different infiltration patterns in AF requires additional studies.

    Acute and chronic inflammation might also interact and contribute to the pathogenesis of AF. For example, preoperative and postoperative CRP levels are both pre-dictive of the development of AF after cardiac surgery.58 Cardiac injury after myocardial infarction is associated with early stimulation of inflammatory signalling.85 The timely increase of anti-inflammatory mediators can stop excessive inflammatory injury and repair cardiac tissue.85 However, the dynamic changes in inflammatory responses during different stresses before the onset or maintenance of AF have yet to be defined. Understanding the temporal changes in these inflammatory responses might be important for the selection of appropriate inflammatory pathway targets to treat AF.

    Many subpopulations of Tlymphocytes and monocyte or macrophages have different proinflammatory or anti-inflammatory responses. For example, M1 macrophages are proinflammatory and recruited early during tissue damage to clear debris and dead cells.102 By contrast, M2 macrophages, which are recruited after M1 macrophages, have reparative functions and secrete proangiogenic or fibrotic mediators to promote wound healing.102 CD4+ (Thelper1 or Thelper2 cells), CD8+, natural killer, and Tregulatory cells all have different roles during chronic inflammation.103 However, little is known about

    how these subpopulations of immune cells and their temporary changes affect the pathogenesis of AF.

    Cytokines, chemokines, and mediatorsThe involvement of different inflammation-associated cytokines and chemokines has been proposed in the pathogenesis of AF (Figure2, Table1). Clinical data indicate an important association between CRP levels and AF. However, in the prospective Copenhagen City Heart Study,61 increases in CRP levels owing to genetic polymorphisms (CRP polymorphism: rs1205, rs1130864, rs3091244, and rs3093077) did not increase the incidence of AF, suggesting that CRP indicates the systemic or regional inflammatory state, but does not have a pathophysiological role. Substantial differences exist between the studies to investigate cytokines and chemokines in the pathogenesis of AF, including study design, patient number, enrolled populations, sample collection, treatment, and follow-up strategies. The mechanisms that underlie postoperative or postabla-tion AF recurrence might differ from those underlying AF onset in the general population and could lead to activation of different cytokines. Consequently, incon-sistent results among these studies are not unexpected. Furthermore, only CRP has been linked to new-onset AF in the generalpopulation.104

    In mice, cardiac-specific expression of TNF or TGF-1 can increase the vulnerability to AF and atrial remod-elling, including fibrosis and heterogeneous conduc-tion.105108 In clinical studies, the serum or atrial tissue levels of TNF or TGF-1 increase in patients with AF, compared with individuals in sinus rhythm, which further supports a detrimental role for these proteins in AF.88,92,109111 The TGF- inhibitor tranilast can prevent atrial remodelling and development of AF in a canine model,112 but no reports exist of studies investigat-ing anti-TNF strategies in the treatment or prevention of AF in humans. However, in the ATTACH trial,113 the use of anti-TNF antibodies in the treatment of patients with heart failure was associated with a higher combined risk of death from any cause or hospitaliza-tion for heart failure than placebo (HR2.84, 95%CI 1.017.97, P = 0.043). Because heart failure is a common comorbidity in patients with AF, an anti-TNF antibody as a cytokine therapy must be used with caution. The levels of myeloperoxidase in atrial tissue are higher in patients with AF than in individuals in sinus rhythm, and increased myeloperoxidase levels in the blood have been associated with early AF recurrence after catheter ablation (HR2.12, 95%CI 1.713.27, P = 0.032).42,114,115 Increased atrial myeloperoxidase levels are also associated with AF vulnerability in canine models.39 In mice pre-treated with angiotensinII, myeloperoxidase deficiency decreases atrial fibrosis and protects mice from AF, which was reversed after restoring myeloper oxidase.114 These data suggest that TNF, TGF-1, and myeloperoxidase might be therapeutic targets for the treatment of AF.

    HSPs have multifunctional cardioprotective roles.116 HSPs are a family of proteins that prevent toxic protein aggregation by binding to unfolded proteins,117 and can

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • 6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    prevent AF by reversing atrial structural remodelling (by mediating apoptosis, fibrosis, and myolysis) and abnormal calcium homeostasis.118 Most studies of HSPs in patients with AF focus on HSP27, HSP60, and HSP70 (Figure3).117118 HSP27 levels progressively decrease from 7.2 0.5 ng/ml in control individuals to 6.1 0.5 ng/ml in patients with paroxysmal AF and 4.7 0.5 ng/ml in those with persistent AF (P = 0.02 for the trend).57 Moreover, HSP27 levels are correlated with increased left atrial size (left atrial diameter 4 cm versus

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 7

    mitochondria and cytosol to the plasma membrane and is released to the extracellular space.129 HSP60 induces cardiomyocyte apoptosis partially via TLR4129,130 and cytokine production through the TLR4MYD88p38/NF-B pathway.131 In addition, HSP60 activates mono-cytes, macrophages, and Tlymphocytes through TLR2 or TLR4(Figure2).132,133

    In a prospective study of 329 patients undergoing elective CABG surgery, increased postoperative anti-HSP65 was independently associated with post operative AF (OR1.4, P = 0.04),43 and higher anti-HSP70 levels were recorded in patients with persistent AF than in those with paroxysmal AF (median 53 g/ml versus 43 g/ml; P = 0.035).45 These findings indicate a patho-genic role of humoral immune responses in patients with AF.43,45 However, ongoing studies to address the mechanisms of a humoral immune response in AF have not yet been reported. Whether an anti-HSP60 or anti-HSP70 autoantibody functions as a trigger or inhibitor for AF-related inflammation remains unclear. HSPs that function as autoantigens might induce an adaptive immune response and activate Blymphocytes to produce antibodies that neutral-ize and promote the clearance of autoantigens.134 The HSPautoantibody complex might also induce a com-plement pathway that leads to macrophage activation and consequently promote inflammation. For example, anti-HSP60 autoantibodies can induce atherosclerosis via endothelial damage.135,136 Autoantibodies against HSP60 canmediate endothelial injury by activating complement-mediated or antibody-dependent cellular cytotoxicity by peripheral blood mononuclear cells.135,136 Conversely, the binding of autoantibodies to HSPs enables the clearance of extracellular HSPs from the tissue and prevents the continuous activation of immune systems.135,137 High serum anti-HSP70 levels (>119.6 g/ml) were also associated with a 61% reduced risk of cardio vascular complications compared with low HSP70

    levels in patients with diabetes mellitus (95%CI 0.170.87).137 Ifautoantibodies against HSPs are a trigger of inflammation, induction of immune tolerance to HSP60 might decrease immune responses to DAMPs via Treg-ulatory cells and inhibit the release of proinflammatory immune cells and cytokines.138 However, if these autoan-tibodies are an inhibitor of inflammation, they might be used as a new DAMP-specific anti-inflammatory therapy to treat AF,139 in an approach similar to that of IgM antibodies against oxidised LDL, which can prevent inflammation and atherosclerosis.140

    Inflammation and electrical remodellingThe detailed pathophysiological mechanisms of electri-cal and structural remodelling in AF have been exten-sively reviewed previously.141 Different inflammatory cytokines modulate the function of ion channels and calcium homeostasis (Figure4). TNF induces abnor-mal Ca2+ handling and arrhythmogenicity in pulmo-nary vein cardiomyocytes.142 TNF can also decrease the expression of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase2a (SERCA2a) by enhancing methylation in the promoter region.143 Mice that selectively overexpress TNF in myocardial tissue have prolonged action poten-tial and Ca2+ transient durations, and higher diastolic and lower systolic Ca2+ currents than those with normal TNF levels.105,106 Furthermore, mice with an elevated TNF level have increased vulnerability to AF and also develop spontaneous episodes of AF.105,106 These findings suggest that TNF can directly alter Ca2+ handling in cardiomyo-cytes, which is crucial for the initiation of AF and atrial el ectrical remodelling.105,106,142,143

    PDGF from myofibroblasts can reduce the dura-tion of action potentials and Ca2+ transients when directly applied to cardiomyocytes, which supports a role for PDGF in electrical remodelling.144 IL-2 is pre-dominantly secreted by activated Tlymphocytes,145 and changes the amplitude of electrically-stimulated

    Increase cardiac apoptosis

    Innate and adaptiveimmune cells

    Inhibit TLR-4 expressionAnti-inammatory cytokines

    Increase proinammatorycytokines

    HSP60

    HSP60HSP70

    HSP27

    Restore L-type calcium currentPrevent action potential duration shorteningPrevent myolysisPrevent F-actin stress bre

    TLR-2,TLR-4

    Cardiomyocytes

    Intracellular cardiomyocyte(HSP27, HSP70)

    HSPs

    Extracellular(HSP27, HSP60, HSP70)

    Secrete anti-HSP autoantibodies

    Nature Reviews | CardiologyFigure 3 | HSPs in atrial fibrillation. HSPs prevent protein aggregation and stabilize protein folding. However, HSPs can function as damageassociated molecular patterns and induce an immune response. Intracellular HSP27 and HSP70 restore abnormal calcium currents and prevent shortening of the action potential duration, myolysis, and Factin stress fibre formation, which can reverse AFrelated electrical and structural remodelling in cardiomyocytes. Extracellular HSP70 and HSP60 activate TLR2 and TLR4 on immune cells and cardiomyocytes. HSP60 can induce cardiomyocyte apoptosis. HSP60 and HSP70 also activate immune cells that secrete proinflammatory cytokines and induce humoral responses to produce antiHSP autoantibodies. HSP27 might inhibit TLR4 expression and its associated NFB pathway, and increase secretion ofIL10; these responses are considered antiinflammatory. Abbreviations: HSP, heat shock protein; TLR, Tolllike receptor.

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • 8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    andcaffeine-induced Ca2+ transients in ventricular myocytes, which has a similar effect to, and might be explained by, a suppression of SERCA2a function and increase of Na+/Ca2+ exchanger activity without changing L-type calcium channels.146 However, this effect has not yet been demonstrated in atrial myocytes.

    Inflammation also alters the conduction properties of the atria. Acute atrial inflammation after right atriotomy in dogs increases the heterogeneity of conduction and AF duration, which can be prevented by the systemic administration of methylprednisolone.39 The increased heterogeneity of conduction also correlates with higher atrial myeloperoxidase activity.39 Heterogeneous con-duction can be created by the local application of ara-chidonic acid (a mediator of inflammation) in the left atria of dogs and be prevented by the topical application of methylprednisolone.147 Furthermore, heterogeneous conduction might be the result of the altered expres-sion or distribution of gap junction-5 protein (com-monly known as connexin-40 [Cx40]), gap junction-1 protein (commonly known as connexin-43 [Cx43]), or atrial fibrosis.108,148 Reduced expression and transmural gradient of Cx40 and Cx43 (both of which are absent in the epicardium, decreased in the mid-myocardium, and normal in the endocardium) in the canine sterile pericarditis model is associated with markedly abnormal

    atrial conduction and vulnerability to the induction and maintenance of AF.148 TNF also downregulates Cx40 andchanges the intracellular distribution of Cx43 in cardio myocytes (which is dispersed from the intercalated discs) in mice.106,149

    In clinical studies, mediators of the inflammatory response are associated with atrial electrical proper-ties.57,90,150 CD36 levels are positively correlated with atrial voltage (rcoefficient=0.63, P = 0.001).90 Low levels of HSP27 (

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 9

    which high CRP or IL-6 levels independently predicted stroke in patients with AF.156,157 These mechanisms might be associated with hypercoagulation, platelet activation, and endothelial dysfunction (Figure5). The vonWillebrand factor (vWF) and asymmetric dimethyl-arginine (ADMA), biomarkers of endothelial dysfunc-tion were both predictors of stroke in patients with AF in a prospective cohort.158160 In 994 patients in the SPAFIII trial,158 levels of vWF independently predicted the occurrence of stroke (relative risk1.2 per 20 IU/dl increase of vWF, 95% CI 1.01.5, P = 0.06) and vascu-lar events (RR1.2 per 20 IU/dl increase of vWF, 95% CI 1.01.4, P = 0.02). ADMA levels were also used, in a single hospital cohort, to predict adverse cardiovascu-lar events including cardiovascular death and ischae-mic stroke (HR1.36 per 0.1 mol/l increase of ADMA, 95%CI 1.071.74, P = 0.01).160 In patients with AF, atrial lymphomononuclear infiltration was concomitant with increased expression of vWF and tissue factor.161,162 High endocardial levels of vWF are also associated with addi-tional platelet adhesion and thrombus formation on the atrial endocardium.163 A lack of endothelial cells and endothelial nitric oxide synthase in the cellular region with platelet adhesion and thrombus formation sug-gests that endothelial dysfunction also contributes to t hrombogenesis in AF.163

    TLR4 might also be an underlying immune mecha-nism to induce atrial endothelial dysfunction. TLR4 knock-out mice have a lower incidence of atrial throm-bosis after thoracic transverse aortic constriction compared with wild-type mice.83 TLR4-related NF-B signal pathways can activate mitogen-activated protein kinase p38, decrease phosphorylation of endothelial nitric oxide synthase, and increase vascular cell adhe-sion protein1 and plasminogen activator inhibitor1

    expression in mice atria.83 Prothrombin and throm-bin receptor (protease-activated receptor1) were also highly expressed in left atrial endocardium concomitant with monocyte infiltration and tissue fibrosis.164 Tissue factor and thrombin activate intrinsic coagulation pathways and platelet aggregation, further contributing tothrombogenesis.164

    Proinflammatory cytokines from immune cells and leucocyteplatelet interactions might also mediate pro-thrombotic states (Figure5).8 Proinflammatory cytokines such as IL-6 can induce platelet activation165 and are associated with spontaneous echo contrast and adverse cardiovascular outcomes in patients with AF.157,166 Acute onset of AF will induce plateletleucocyte interactions.167 Platelets can interact with neutrophils and monocytes and be activated via CD40, P-selectin, and CD36 in AF. However, in clinical studies, inconsistent results linking the levels of soluble CD40 or P-selectin to thrombo-embolism in AF have been reported.156,168,169 Whether monocyte CD36 is associated with thromboembolism is currently unclear.

    Current anti-inflammatory therapiesTo date, no drug has been designed to target the inflam-matory pathway specifically in patients with AF, but most drugs used to prevent AF are arbitrarily consid-ered anti-inflammatory as part of their pleiotropic effects. Angiotensin-converting-enzyme inhibitors, a ngiotensin-receptor blockers, statins, and n-3 poly-unsaturated fatty acids have been studied in large, pro-spective, randomized trials and meta-analyses for both primary and secondary prevention of AF.4,5,170 Owing to the heterogeneity between studies and disappointing results in prospective trials, only angiotensin-converting-enzyme inhibitors and angiotensin-receptor blockers are considered reasonable approaches for the primary pre-vention of new-onset AF in patients with heart failure and reduced left ventricular ejection fraction (classIIa indication, level of evidenceB).4

    These findings should not discourage the develop-ment of anti-inflammatory therapies in the prevention of AF, because most of these studies did not demonstrate a downregulation of inflammation after drug applica-tion.171 Even among positive results, the prevention of AF does not seem to be the result of reduced inflammation. For example, in the ARMYDA-3 trial,37 treatment with atorvastatin significantly reduced the incidence of post-operative AF after elective cardiac surgery with cardio-pulmonary bypass surgery (OR0.39, 95%CI 0.180.85, P = 0.017); however, CRP levels did not significantly decrease after statin use. Colchicine might prevent AF by treating pericarditis after surgery or ablation. For example, colchicine seems to reduce postoperative AF (from 22.0% to 12.0%; P = 0.02) and decreases the com-plication rate and length of hospital stay.172,173 Colchicine also prevents early AF recurrences in patients at 3months after pulmonary vein isolation (from 33.5% to 16.0%; P = 0.01).174 This effect is associated with a signifi-cant decrease in inflammatory mediators, including CRP and IL-6 (CRP: 0.46 mg/l; interquartile range: 0.78

    Platelet

    P-selectinCD36

    CD40Thrombus

    Neutrophil

    Monocyte

    Proinammatorycytokines: IL-6

    von Willebrand factor

    Damagedendothelium

    Thrombin

    Tissue factor

    PAR-1

    eNOS

    Atrium

    Immunecell

    inltration

    Plateletleucocyteinteraction Coagulation

    factor

    Nature Reviews | CardiologyFigure 5 | The pathophysiological link between inflammation and thrombogenesis. Immune cell infiltration induces endothelial dysfunction, which decreases eNOSexpression, but increases the expression of vonWillebrand factor, thrombin, tissue factor, and PAR1 on the atrial endocardium. Leucocytes (neutrophils andmonocytes) that are partially activated by proinflammatory cytokines might interact with and activate platelets via CD36, CD40, and Pselectin. The adhesion and activation of platelets and coagulation factors also contributes to a thromboticclot. Abbreviations: eNOS, endothelial nitric oxide synthase; PAR1, proteinaseactivated receptor1.

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • 10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    to 0.08 mg/l, P

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 11

    1. Camm, A.J. etal. 2012 focused update of the ESC guidelines for the management of atrial fibrillation: an update of the 2010 ESC guidelines for the management of atrial fibrillationdeveloped with the special contribution of the European Heart Rhythm Association. Europace 14, 13851413 (2012).

    2. Chugh, S.S. etal. Worldwide epidemiology of atrial fibrillation: a Global Burden of Disease 2010 Study. Circulation 129, 837847 (2014).

    3. Naccarelli, G.V., Varker, H., Lin, J. &Schulman,K.L. Increasing prevalence of atrialfibrillation and flutter in the United States. Am. J. Cardiol. 104, 15341539 (2009).

    4. January, C.T. etal. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. J. Am. Coll. Cardiol. 64, e1e76 (2014).

    5. Calkins, H. etal. 2012HRS/EHRA/ECAS expert consensus statement on catheter and surgical ablation of atrial fibrillation: recommendations for patient selection, procedural techniques, patient management and followup, definitions, endpoints, and research trial design: a report ofthe Heart Rhythm Society (HRS) Task Force onCatheter and Surgical Ablation of Atrial Fibrillation. Developed in partnership with the European Heart Rhythm Association (EHRA), aregistered branch of the European Society ofCardiology (ESC) and the European Cardiac Arrhythmia Society (ECAS); and in collaboration with the American College of Cardiology (ACC), American Heart Association (AHA), the Asia Pacific Heart Rhythm Society (APHRS), and the Society of Thoracic Surgeons (STS). Endorsed bythe governing bodies of the American College of Cardiology Foundation, the American Heart Association, the European Cardiac Arrhythmia Society, the European Heart Rhythm Association, the Society of Thoracic Surgeons, the Asia Pacific Heart Rhythm Society, and the Heart Rhythm Society. Heart Rhythm 9, 632696.e21 (2012).

    6. Morillo, C.A. etal. Radiofrequency ablation vsantiarrhythmic drugs as firstline treatment ofparoxysmal atrial fibrillation (RAAFT2): arandomized trial. JAMA 311, 692700 (2014).

    7. Calkins, H. etal. Treatment of atrial fibrillation with antiarrhythmic drugs or radiofrequency ablation: two systematic literature reviews and metaanalyses. Circ. Arrhythm. Electrophysiol. 2, 349361 (2009).

    8. Guo, Y., Lip, G.Y. & Apostolakis, S. Inflammationin atrial fibrillation. J. Am. Coll. Cardiol. 60, 22632270 (2012).

    9. Savelieva, I., Kakouros, N., Kourliouros, A. &Camm, A.J. Upstream therapies for management of atrial fibrillation: review of clinical evidence and implications for European Society of Cardiology guidelines. PartI: primary prevention. Europace 13, 308328 (2011).

    10. Patel, P., Dokainish, H., Tsai, P. & Lakkis, N. Update on the association of inflammation and atrial fibrillation. J. Cardiovasc. Electrophysiol. 21, 10641070 (2010).

    11. Gregor, M.F. & Hotamisligil, G.S. Inflammatory mechanisms in obesity. Annu. Rev. Immunol. 29, 415445 (2011).

    12. Harrison, D.G. etal. Inflammation, immunity, and hypertension. Hypertension 57, 132140 (2011).

    13. Marzilli, M. etal. Obstructive coronary atherosclerosis and ischemic heart disease: anelusive link! J. Am. Coll. Cardiol. 60, 951956 (2012).

    14. Wang, T.J. etal. Obesity and the risk of newonset atrial fibrillation. JAMA 292, 24712477 (2004).

    15. Zacharias, A. etal. Obesity and risk of newonset atrial fibrillation after cardiac surgery. Circulation 112, 32473255 (2005).

    16. Lumeng, C.N., DelProposto, J.B., Westcott, D.J. & Saltiel, A.R. Phenotypic switching of adipose tissue macrophages with obesity is generated byspatiotemporal differences in macrophage subtypes. Diabetes 57, 32393246 (2008).

    17. Lumeng, C.N., Bodzin, J.L. & Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 117, 175184 (2007).

    18. Hotamisligil, G.S., Arner, P., Caro, J.F., Atkinson,R.L. & Spiegelman, B.M. Increased adipose tissue expression of tumor necrosis factoralpha in human obesity and insulin resistance. J. Clin. Invest. 95, 24092415 (1995).

    19. MohamedAli, V. etal. Subcutaneous adipose tissue releases interleukin6, but not tumor necrosis factoralpha, invivo. J. Clin. Endocrinol. Metab. 82, 41964200 (1997).

    20. Mazurek, T. etal. Relation of proinflammatory activity of epicardial adipose tissue to the occurrence of atrial fibrillation. Am. J. Cardiol. 113, 15051508 (2014).

    21. Nagashima, K. etal. Does location of epicardial adipose tissue correspond to endocardial high dominant frequency or complex fractionated atrial electrogram sites during atrial fibrillation? Circ. Arrhythm. Electrophysiol. 5, 676683 (2012).

    22. Kanazawa, H. etal. Importance of pericardial fatin the formation of complex fractionated atrial electrogram region in atrial fibrillation. Int.J. Cardiol. 174 557564 (2014).

    23. Poirier, P. etal. Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, PhysicalActivity, and Metabolism. Circulation 113, 898918 (2006).

    24. Lau, D.H. etal. Hypertension and atrial fibrillation: evidence of progressive atrial remodeling with electrostructural correlate in a conscious chronically instrumented ovine model. Heart Rhythm 7, 12821290 (2010).

    25. Lau, D.H. etal. Atrial arrhythmia in ageing spontaneously hypertensive rats: unraveling the substrate in hypertension and ageing. PLoS ONE 8, e72416 (2013).

    26. Kistler, P.M. etal. Atrial electrical and structural abnormalities in an ovine model of chronic blood pressure elevation after prenatal corticosteroid exposure: implications for development of atrial fibrillation. Eur. Heart J. 27, 30453056 (2006).

    27. Friedrichs, K. etal. Induction of atrial fibrillation by neutrophils critically depends on CD11b/CD18 integrins. PLoS ONE 9 e89307 (2014).

    28. Suzuki, Y. etal. Inflammation and angiotensin II. Int. J. Biochem. Cell Biol. 35, 881900 (2003).

    29. Wang, Y. etal. TNF and IL1 neutralization ameliorates angiotensinIIinduced cardiac damage in male mice. Endocrinology 155, 26772687 (2014).

    30. De Jong, A.M. etal. Mechanisms of atrial structural changes caused by stretch occurring before and during early atrial fibrillation. Cardiovasc. Res. 89, 754765 (2011).

    31. Zhang, J. etal. NOX4dependent hydrogen peroxide overproduction in human atrial fibrillation and HL1 atrial cells: relationship to hypertension. Front. Physiol. 3, 140 (2012).

    32. Hori, M. & Nishida, K. Oxidative stress and leftventricular remodelling after myocardial infarction. Cardiovasc. Res. 81, 457464 (2009).

    33. Sinno, H. etal. Atrial ischemia promotes atrial fibrillation in dogs. Circulation 107, 19301936 (2003).

    34. Stellos, K. etal. Expression of plateletbound stromal cellderived factor1 in patients with nonvalvular atrial fibrillation and ischemic heart disease. J. Thromb. Haemost. 10, 4955 (2012).

    35. Mehta, N.N. etal. The novel atherosclerosis locus at 10q11 regulates plasma CXCL12 levels. Eur. Heart J. 32, 963971 (2011).

    36. Kim, D. etal. CXCL12 secreted from adipose tissue recruits macrophages and induces insulin resistance in mice. Diabetologia 57, 14561465 (2014).

    37. Patti, G. etal. Randomized trial of atorvastatin for reduction of postoperative atrial fibrillation inpatients undergoing cardiac surgery: results of the ARMYDA3 (Atorvastatin for Reduction of MYocardial Dysrhythmia After cardiac surgery) study. Circulation 114, 14551461 (2006).

    38. Zhang, Z. etal. n3 polyunsaturated fatty acids prevents atrial fibrillation by inhibiting inflammation in a canine sterile pericarditis model. Int. J. Cardiol. 153, 1420 (2011).

    39. Ishii, Y. etal. Inflammation of atrium after cardiacsurgery is associated with inhomogeneity of atrial conduction and atrial fibrillation. Circulation 111, 28812888 (2005).

    40. Grubman, E. etal. Histopathologic effects of radiofrequency catheter ablation in previously infarcted human myocardium. J. Cardiovasc. Electrophysiol. 10, 336342 (1999).

    41. Lim, H.S. etal. Time course of inflammation, myocardial injury, and prothrombotic response after radiofrequency catheter ablation for atrial fibrillation. Circ. Arrhythm. Electrophysiol. 7, 8389 (2014).

    42. Richter, B. etal. Markers of oxidative stress afterablation of atrial fibrillation are associated with inflammation, delivered radiofrequency energy and early recurrence of atrial fibrillation. Clin. Res. Cardiol. 101, 217225 (2012).

    43. Mandal, K. etal. Association of antiheat shock protein 65 antibodies with development of postoperative atrial fibrillation. Circulation 110, 25882590 (2004).

    44. Li, H. etal. Atrial tachycardia provoked in the presence of activating autoantibodies to 2adrenergic receptor in the rabbit. Heart Rhythm 10, 436441 (2013).

    45. Kornej, J. etal. Response of circulating heat shock protein 70 and antiheat shock protein 70antibodies to catheter ablation of atrial fibrillation. J. Transl. Med. 11, 49 (2013).

    46. Kato, K. etal. Genetic factors for lone atrial fibrillation. Int. J. Mol. Med. 19, 933939 (2007).

    47. Lin, H. etal. Targeted sequencing in candidate genes for atrial fibrillation: the Cohorts for Heartand Aging Research in Genomic Epidemiology (CHARGE) Targeted Sequencing Study. HeartRhythm 11, 452457 (2014).

    48. Marcus, G.M. etal. Interleukin6 and atrial fibrillation in patients with coronary artery disease: data from the Heart and Soul Study. Am. Heart J. 155, 303309 (2008).

    49. Gungor, B. etal. Assessment of interleukin1 gene cluster polymorphisms in lone atrial fibrillation: new insight into the role of inflammation in atrial fibrillation. Pacing Clin. Electrophysiol. 36, 12201227 (2013).

    50. Gaudino, M. etal. The 174G/C interleukin6 polymorphism influences postoperative interleukin6 levels and postoperative atrial fibrillation. Is atrial fibrillation an inflammatory

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • 12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    complication? Circulation 108 (Suppl.1), II195II199 (2003).

    51. Chiang, C.H. etal. Herpes simplex virus infection and risk of atrial fibrillation: anationwide study. Int. J. Cardiol. 164, 201204 (2013).

    52. Hsu, J.C. etal. Atrial fibrillation and atrial flutter in human immunodeficiency virusinfected persons: incidence, risk factors, and association with markers of HIV disease severity. J. Am. Coll. Cardiol. 61, 22882295 (2013).

    53. Yearley, J.H. etal. Antigenic stimulation in the simian model of HIV infection yields dilated cardiomyopathy through effects of TNF. AIDS 22, 585594 (2008).

    54. Raidel, S.M. etal. Targeted myocardial transgenic expression of HIV tat causes cardiomyopathy and mitochondrial damage. Am. J. Physiol. Heart Circ. Physiol. 282, H1672H1678 (2002).

    55. Frustaci, A. etal. Histological substrate of atrial biopsies in patients with lone atrial fibrillation. Circulation 96, 11801184 (1997).

    56. Boos, C.J., Anderson, R.A. & Lip, G.Y. Is atrial fibrillation an inflammatory disorder? Eur. Heart J. 27, 136149 (2006).

    57. Hu, Y.F. etal. Electrophysiological correlation and prognostic impact of heat shock protein 27 in atrial fibrillation. Circ. Arrhythm. Electrophysiol. 5, 334340 (2012).

    58. Jacob, K.A. etal. Inflammation in newonsetatrial fibrillation after cardiac surgery: asystematic review. Eur. J. Clin. Invest. 44, 402428 (2014).

    59. Smit, M.D. etal. Role of inflammation in earlyatrial fibrillation recurrence. Europace 14, 810817 (2012).

    60. Aviles, R.J. etal. Inflammation as a risk factor for atrial fibrillation. Circulation 108, 30063010 (2003).

    61. Marott, S.C. etal. Does elevated Creactive protein increase atrial fibrillation risk? AMendelian randomization of 47,000 individuals from the general population. J. Am. Coll. Cardiol. 56, 789795 (2010).

    62. Conen, D. etal. A multimarker approach toassess the influence of inflammation on theincidence of atrial fibrillation in women. Eur.Heart J. 31, 17301736 (2010).

    63. Dernellis, J. & Panaretou, M. Relationship between Creactive protein concentrations during glucocorticoid therapy and recurrent atrial fibrillation. Eur. Heart J. 25, 11001107 (2004).

    64. Bruins, P. etal. Activation of the complement system during and after cardiopulmonary bypass surgery: postsurgery activation involves Creactive protein and is associated with postoperative arrhythmia. Circulation 96, 35423548 (1997).

    65. Ho, K.M. & Tan, J.A. Benefits and risks of corticosteroid prophylaxis in adult cardiac surgery: a doseresponse metaanalysis. Circulation 119, 18531866 (2009).

    66. Marcus, G.M. etal. Markers of inflammation before and after curative ablation of atrial flutter. Heart Rhythm 5, 215221 (2008).

    67. Marcus, G.M. etal. Intracardiac and extracardiac markers of inflammation during atrial fibrillation. Heart Rhythm 7, 149154 (2010).

    68. Chung, M.K. etal. Creactive protein elevation inpatients with atrial arrhythmias: inflammatory mechanisms and persistence of atrial fibrillation. Circulation 104, 28862891 (2001).

    69. Li, J. etal. Role of inflammation and oxidative stress in atrial fibrillation. Heart Rhythm 7, 438444 (2010).

    70. Kallergis, E.M. etal. The role of the postcardioversion time course of hsCRP levels in clarifying the relationship between inflammation and persistence of atrial fibrillation. Heart 94, 200204 (2008).

    71. Rotter, M. etal. Decline in Creactive protein aftersuccessful ablation of longlasting persistent atrial fibrillation. J. Am. Coll. Cardiol. 47, 12311233 (2006).

    72. ShiroshitaTakeshita, A., Brundel, B.J., Lavoie, J. & Nattel, S. Prednisone prevents atrial fibrillation promotion by atrial tachycardia remodeling in dogs. Cardiovasc. Res. 69, 865875 (2006).

    73. AimeSempe, C. etal. Myocardial cell death in fibrillating and dilated human right atria. J. Am. Coll. Cardiol. 34, 15771586 (1999).

    74. Tabas, I. & Glass, C.K. Antiinflammatory therapy in chronic disease: challenges and opportunities. Science 339, 166172 (2013).

    75. Das, M. etal. Induction of hepatitis by JNKmediated expression of TNF. Cell 136, 249260 (2009).

    76. Arslan, F., de Kleijn, D.P. & Pasterkamp, G. Innate immune signaling in cardiac ischemia. Nat. Rev. Cardiol. 8, 292300 (2011).

    77. Tousoulis, D. etal. Oxidative stress and inflammatory process in patients with atrial fibrillation: the role of left atrium distension. Int.J. Cardiol. 136, 258262 (2009).

    78. Steiner, I. & Hajkova, P. Patterns of isolated atrialamyloid: a study of 100 hearts on autopsy. Cardiovasc. Pathol. 15, 287290 (2006).

    79. Afzal, A.R. etal. Association of Met439Thr substitution in heat shock protein 70 gene with postoperative atrial fibrillation and serum HSP70 protein levels. Cardiology 110, 445452 (2008).

    80. Brundel, B.J. etal. Heat shock protein upregulation protects against pacinginduced myolysis in HL1 atrial myocytes and in human atrial fibrillation. J. Mol. Cell. Cardiol. 41, 555562 (2006).

    81. Ichiki, H. etal. The role of infection in the development of nonvalvular atrial fibrillation: upregulation of Tolllike receptor 2 expression levels on monocytes. J. Cardiol. 53, 127135 (2009).

    82. Wang, J. etal. TLR2 was overexpressed independent of IL6 in patients with valvular atrial fibrillation. J. Biomed. Res. 25, 178184 (2011).

    83. Katoh, S. etal. Atrial endothelial impairment through Tolllike receptor 4 signaling causes atrial thrombogenesis. Heart Vessels 29, 263272 (2014).

    84. Boyd, J.H., Mathur, S., Wang, Y., Bateman, R.M. & Walley, K.R. Tolllike receptor stimulation in cardiomyoctes decreases contractility and initiates an NFB dependent inflammatory response. Cardiovasc. Res. 72, 384393 (2006).

    85. Frangogiannis, N.G. The inflammatory response in myocardial injury, repair, and remodelling. Nat.Rev. Cardiol. 11, 255265 (2014).

    86. MonneratCahli, G. etal. Tolllike receptor 4 activation promotes cardiac arrhythmias by decreasing the transient outward potassium current (Ito) through an IRF3dependent and MyD88independent pathway. J. Mol. Cell. Cardiol. 76, 116125 (2014).

    87. Yamashita, T. etal. Recruitment of immune cellsacross atrial endocardium in human atrialfibrillation. Circ. J. 74, 262270 (2010).

    88. Qu, Y.C. etal. Activated nuclear factorkappaBand increased tumor necrosis factoralpha in atrial tissue of atrial fibrillation. Scand.Cardiovasc. J. 43, 292297 (2009).

    89. Goette, A. etal. Angiotensin II receptor blockade reduces tachycardiainduced atrial adhesion molecule expression. Circulation 117, 732742 (2008).

    90. Hu, Y.F. etal. Impact of circulating monocyte CD36 level on atrial fibrillation and subsequent catheter ablation. Heart Rhythm 8, 650656 (2011).

    91. Liu, L. etal. Activation of peripheral blood CD3+ Tlymphocytes in patients with atrial fibrillation. Int. Heart J. 53, 221224 (2012).

    92. Corradi, D. etal. Persistent lone atrial fibrillation: clinicopathologic study of 19 cases. Heart Rhythm 11, 12501258 (2014).

    93. Schuessler, R.B. etal. The effects of inflammation on heart rate and rhythm in a canine model of cardiac surgery. Heart Rhythm 9, 432439 (2012).

    94. Liao, C.H. etal. Cardiac mast cells cause atrial fibrillation through PDGFAmediated fibrosis in pressureoverloaded mouse hearts. J. Clin. Invest. 120, 242253 (2010).

    95. Gibson, P.H. etal. Usefulness of neutrophil/lymphocyte ratio as predictor of newonset atrial fibrillation after coronary artery bypass grafting. Am. J. Cardiol. 105, 186191 (2010).

    96. Canpolat, U. etal. Role of preablation neutrophil/lymphocyte ratio on outcomes of cryoballoonbased atrial fibrillation ablation. Am.J. Cardiol. 112, 513519 (2013).

    97. Guo, X. etal. Postablation neutrophil/lymphocyte ratio correlates with arrhythmia recurrence aftercatheter ablation of lone atrial fibrillation. Chin. Med. J. 127, 10331038 (2014).

    98. Klebanoff, S.J. Myeloperoxidase. Proc. Assoc. Am. Physicians 111, 383389 (1999).

    99. Ishida, K. etal. Relation of inflammatory cytokines to atrial fibrillation after offpump coronary artery bypass grafting. Eur. J. Cardiothorac. Surg. 29, 501505 (2006).

    100. Stein, A. etal. Systemic inflammatory changes after pulmonary vein radiofrequency ablation do not alter stem cell mobilization. Europace 10, 444449 (2008).

    101. Kennedy, S., Wu, J., Wadsworth, R.M., Lawrence, C.E. & Maffia, P. Mast cells and vascular diseases. Pharmacol. Ther. 138, 5365 (2013).

    102. ChinettiGbaguidi, G., Colin, S. & Staels, B. Macrophage subsets in atherosclerosis. Nat.Rev. Cardiol. 12, 1017 (2014).

    103. Kared, H., Camous, X. & Larbi, A. Tcells and their cytokines in persistent stimulation of the immune system. Curr. Opin. Immunol. 29, 7985 (2014).

    104. Schnabel, R.B. etal. Relation of multiple inflammatory biomarkers to incident atrial fibrillation. Am. J. Cardiol. 104, 9296 (2009).

    105. Saba, S. etal. Atrial contractile dysfunction, fibrosis, and arrhythmias in a mouse model of cardiomyopathy secondary to cardiacspecific overexpression of tumor necrosis factor. Am. J. Physiol. Heart Circ. Physiol. 289, H1456H1467 (2005).

    106. Sawaya, S.E. etal. Downregulation of connexin40 and increased prevalence of atrial arrhythmias in transgenic mice with cardiacrestricted overexpression of tumor necrosis factor. Am. J. Physiol. Heart Circ. Physiol. 292, H1561H1567 (2007).

    107. Verheule, S. etal. Increased vulnerability to atrial fibrillation in transgenic mice with selective atrialfibrosis caused by overexpression of TGF1. Circ. Res. 94, 14581465 (2004).

    108. Choi, E.K. etal. Triggered firing and atrial fibrillation in transgenic mice with selective atrialfibrosis induced by overexpression of TGF1. Circ. J. 76, 13541362 (2012).

    109. Wu, N. etal. Association of inflammatory factorswith occurrence and recurrence of atrial fibrillation: a metaanalysis. Int. J. Cardiol. 169, 6272 (2013).

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 13

    110. Wang, Y., Hou, X. & Li, Y. Association between transforming growth factor1 polymorphisms and atrial fibrillation in essential hypertensive subjects. J. Biomed. Sci. 17, 23 (2010).

    111. Zhao, F. etal. Calreticulin overexpression correlates with integrin5 and transforming growth factor1 expression in the atria of patients with rheumatic valvular disease and atrial fibrillation. Int. J. Cardiol. 168, 21772185 (2013).

    112. Nakatani, Y. etal. Tranilast prevents atrial remodeling and development of atrial fibrillation in a canine model of atrial tachycardia and left ventricular dysfunction. J. Am. Coll. Cardiol. 61, 582588 (2013).

    113. Chung, E.S., Packer, M., Lo, K.H., Fasanmade,A.A. & Willerson, J.T. Randomized, doubleblind, placebocontrolled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor, in patients with moderatetosevere heart failure: results of the antiTNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation 107, 31333140 (2003).

    114. Rudolph, V. etal. Myeloperoxidase acts as a profibrotic mediator of atrial fibrillation. Nat. Med. 16, 470474 (2010).

    115. Li, S.B. etal. Myeloperoxidase and risk of recurrence of atrial fibrillation after catheter ablation. J. Investig. Med. 61, 722727 (2013).

    116. GhayourMobarhan, M., Saber, H. & Ferns, G.A. The potential role of heat shock protein 27 in cardiovascular disease. Clin. Chim. Acta 413, 1524 (2012).

    117. Brundel, B.J. etal. Heat shock proteins as molecular targets for intervention in atrial fibrillation. Cardiovasc. Res. 78, 422428 (2008).

    118. HoogstraBerends, F. etal. Heat shock proteininducing compounds as therapeutics to restore proteostasis in atrial fibrillation. Trends Cardiovasc. Med. 22, 6268 (2012).

    119. Yang, M. etal. Expression of heat shock proteins in myocardium of patients with atrial fibrillation. Cell Stress Chaperones 12, 142150 (2007).

    120. Brundel, B.J. etal. Induction of heat shock response protects the heart against atrial fibrillation. Circ. Res. 99, 13941402 (2006).

    121. Ke, L. etal. HSPB1, HSPB6, HSPB7 and HSPB8protect against RhoA GTPaseinduced remodeling in tachypaced atrial myocytes. PLoSONE 6, e20395 (2011).

    122. Chang, S.L. etal. Heat shock protein inducer modifies arrhythmogenic substrate and inhibits atrial fibrillation in the failing heart. Int. J. Cardiol. 168, 40194026 (2013).

    123. Wakisaka, O. etal. Hyperthermia treatment prevents angiotensin IImediated atrial fibrosis and fibrillation via induction of heatshock protein 72. J. Mol. Cell. Cardiol. 43, 616626 (2007).

    124. Schafler, A.E. etal. The expression of heat shock protein 60 in myocardium of patients with chronic atrial fibrillation. Basic Res. Cardiol. 97, 258261 (2002).

    125. De, A.K., Kodys, K.M., Yeh, B.S. &MillerGraziano, C. Exaggerated human monocyte IL10 concomitant to minimal TNF induction by heatshock protein 27 (Hsp27) suggests Hsp27 is primarily an antiinflammatory stimulus. J.Immunol. 165, 39513958 (2000).

    126. Jin, C. etal. Human myocardium releases heat shock protein 27 (HSP27) after global ischemia: the proinflammatory effect of extracellular HSP27 through tolllike receptor (TLR)2 and TLR4. Mol. Med. 20, 280289 (2014).

    127. Asea, A. etal. Novel signal transduction pathway utilized by extracellular HSP70: role of tolllike receptor (TLR) 2 and TLR4. J. Biol. Chem. 277, 1502815034 (2002).

    128. Mathur, S., Walley, K.R., Wang, Y., Indrambarya,T. & Boyd, J.H. Extracellular heat shock protein 70 induces cardiomyocyte inflammation and contractile dysfunction via TLR2. Circ. J. 75, 24452452 (2011).

    129. Lin, L. etal. HSP60 in heart failure: abnormal distribution and role in cardiac myocyte apoptosis. Am. J. Physiol. Heart Circ. Physiol. 293, H2238H2247 (2007).

    130. Kim, S.C. etal. Extracellular heat shock protein 60, cardiac myocytes, and apoptosis. Circ. Res. 105, 11861195 (2009).

    131. Tian, J. etal. Extracellular HSP60 induces inflammation through activating and upregulating TLRs in cardiomyocytes. Cardiovasc. Res. 98, 391401 (2013).

    132. Kol, A., Lichtman, A.H., Finberg, R.W., Libby, P. &KurtJones, E.A. Cutting edge: heat shock protein (HSP) 60 activates the innate immune response: CD14 is an essential receptor forHSP60 activation of mononuclear cells. J.Immunol. 164, 1317 (2000).

    133. ZaninZhorov, A., Nussbaum, G., Franitza, S., Cohen, I.R. & Lider, O. Tcells respond to heat shock protein 60 via TLR2: activation of adhesion and inhibition of chemokine receptors. FASEB J. 17, 15671569 (2003).

    134. Rudolphi, U. etal. The Bcell repertoire of patients with rheumatoid arthritis. II. Increased frequencies of IgG+ and IgA+ Bcells specific for mycobacterial heatshock protein 60 or human typeII collagen in synovial fluid and tissue. Arthritis Rheum. 40, 14091419 (1997).

    135. Schett, G. etal. Autoantibodies against heat shock protein 60 mediate endothelial cytotoxicity. J. Clin. Invest. 96, 25692577 (1995).

    136. Foteinos, G., Afzal, A.R., Mandal, K., Jahangiri,M. & Xu, Q. Antiheat shock protein 60 autoantibodies induce atherosclerosis in apolipoproteinEdeficient mice via endothelial damage. Circulation 112, 12061213 (2005).

    137. Gruden, G. etal. Antihsp60 and Antihsp70 antibody levels and micro/macrovascular complications in type1 diabetes: the EURODIAB Study. J. Intern. Med. 266, 527536 (2009).

    138. van Puijvelde, G.H. etal. Induction of oral tolerance to HSP60 or an HSP60peptide activates Tcell regulation and reduces atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 27, 26772683 (2007).

    139. Binder, C.J. etal. Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae and oxidized LDL. Nat. Med. 9, 736743 (2003).

    140. Cesena, F.H. etal. Immunemodulation by polyclonal IgM treatment reduces atherosclerosis in hypercholesterolemic apoE/ mice. Atherosclerosis 220, 5965 (2012).

    141. Wakili, R., Voigt, N., Kaab, S., Dobrev, D. &Nattel, S. Recent advances in the molecular pathophysiology of atrial fibrillation. J. Clin. Invest. 121, 29552968 (2011).

    142. Lee, S.H. etal. Tumor necrosis factor alterscalcium handling and increases arrhythmogenesis of pulmonary vein cardiomyocytes. Life Sci. 80, 18061815 (2007).

    143. Kao, Y.H. etal. Tumor necrosis factor decreases sarcoplasmic reticulum Ca2+ATPase expressions via the promoter methylation in cardiomyocytes. Crit. Care Med. 38, 217222 (2010).

    144. Musa, H. etal. Inhibition of plateletderived growth factorAB signaling prevents electromechanical remodeling of adult atrial myocytes that contact myofibroblasts. Heart Rhythm 10, 10441051 (2013).

    145. Nelson, B.H. IL2, regulatory Tcells, and tolerance. J. Immunol. 172, 39833988 (2004).

    146. Cao, C.M. etal. Influence of interleukin2 on Ca2+ handling in rat ventricular myocytes. J. Mol. Cell. Cardiol. 35, 14911503 (2003).

    147. Tselentakis, E.V., Woodford, E., Chandy, J., Gaudette, G.R. & Saltman, A.E. Inflammation effects on the electrical properties of atrial tissue and inducibility of postoperative atrial fibrillation. J. Surg. Res. 135, 6875 (2006).

    148. Ryu, K. etal. Effects of sterile pericarditis on connexins 40 and 43 in the atria: correlation with abnormal conduction and atrial arrhythmias. Am.J. Physiol. Heart Circ. Physiol. 293, H1231H1241 (2007).

    149. Liew, R. etal. Role of tumor necrosis factoralpha in the pathogenesis of atrial fibrosis and development of an arrhythmogenic substrate. Circ. J. 77, 11711179 (2013).

    150. Lin, Y.J. etal. Prognostic implications of the highsensitive Creactive protein in the catheter ablation of atrial fibrillation. Am. J. Cardiol. 105, 495501 (2010).

    151. Wilhelm, M. etal. Inflammation and atrial remodeling after a mountain marathon. Scand. J. Med. Sci. Sports 24, 519525 (2014).

    152. Li, Y.Y. etal. Myocardial extracellular matrix remodeling in transgenic mice overexpressing tumor necrosis factor alpha can be modulated by antitumor necrosis factor alpha therapy. Proc. Natl Acad. Sci. USA 97, 1274612751 (2000).

    153. Burstein, B., Libby, E., Calderone, A. & Nattel, S. Differential behaviors of atrial versus ventricular fibroblasts: a potential role for plateletderived growth factor in atrialventricular remodeling differences. Circulation 117, 16301641 (2008).

    154. Psychari, S.N. etal. Relation of elevated Creactive protein and interleukin6 levels to left atrial size and duration of episodes in patients with atrial fibrillation. Am. J. Cardiol. 95, 764767 (2005).

    155. Luan, Y. etal. Interleukin18 among atrial fibrillation patients in the absence of structural heart disease. Europace 12, 17131718 (2010).

    156. Lip, G.Y., Patel, J.V., Hughes, E. & Hart, R.G. Highsensitivity Creactive protein and soluble CD40 ligand as indices of inflammation and platelet activation in 880 patients with nonvalvular atrial fibrillation: relationship to stroke risk factors, stroke risk stratification schema, and prognosis. Stroke 38, 12291237 (2007).

    157. Roldan, V. etal. High sensitivity cardiac troponinT and interleukin6 predict adverse cardiovascular events and mortality in anticoagulated patients with atrial fibrillation. J.Thromb. Haemost. 10, 15001507 (2012).

    158. Conway, D.S., Pearce, L.A., Chin, B.S., Hart,R.G. & Lip, G.Y. Prognostic value of plasma von Willebrand factor and soluble Pselectin as indices of endothelial damage and platelet activation in 994 patients with nonvalvular atrial fibrillation. Circulation 107, 31413145 (2003).

    159. Roldan, V. etal. Plasma von Willebrand factor levels are an independent risk factor for adverse events including mortality and major bleeding in anticoagulated atrial fibrillation patients. J. Am. Coll. Cardiol. 57, 24962504 (2011).

    160. Chao, T.F. etal. Plasma asymmetric dimethylarginine and adverse events in patients with atrial fibrillation referred for coronary angiogram. PLoS ONE 8, e71675 (2013).

    161. Nakamura, Y. etal. Tissue factor expression in atrial endothelia associated with nonvalvular atrial fibrillation: possible involvement in intracardiac thrombogenesis. Thromb. Res. 111, 137142 (2003).

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

  • 14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio

    162. Kaireviciute, D. etal. Intracardiac expression ofmarkers of endothelial damage/dysfunction, inflammation, thrombosis, and tissue remodeling, and the development of postoperative atrial fibrillation. J. Thromb. Haemost. 9, 23452352 (2011).

    163. Fukuchi, M. etal. Increased von Willebrand factor in the endocardium as a local predisposing factor for thrombogenesis in overloaded human atrial appendage. J. Am. Coll. Cardiol. 37, 14361442 (2001).

    164. Ito, K. etal. An immunohistochemical analysis oftissue thrombin expression in the human atria. PLoS ONE 8, e65817 (2013).

    165. Marino, M. etal. Novel path to IL6 transsignaling through thrombininduced soluble IL6 receptor release by platelets. J. Biol. Regul. Homeost. Agents 27, 841852 (2013).

    166. Conway, D.S., Buggins, P., Hughes, E. & Lip, G.Y. Relation of interleukin6, Creactive protein, and the prothrombotic state to transesophageal echocardiographic findings in atrial fibrillation. Am. J. Cardiol. 93, 13681373 (2004).

    167. Hayashi, M. etal. Platelet activation and induction of tissue factor in acute and chronic atrial fibrillation: involvement of mononuclear cellplatelet interaction. Thromb. Res. 128, e113e118 (2011).

    168. Duygu, H. etal. Prognostic value of plasma soluble CD40 ligand in patients with chronic nonvalvular atrial fibrillation. Europace 10, 210214 (2008).

    169. Heeringa, J. etal. A longitudinal populationbased study of prothrombotic factors in elderly subjects with atrial fibrillation: the Rotterdam Study 19901999. J. Thromb. Haemost. 4, 19441949 (2006).

    170. Savelieva, I., Kakouros, N., Kourliouros, A. & Camm, A.J. Upstream therapies for management of atrial fibrillation: review of clinical evidence and implications for European Society of Cardiology guidelines. Part II: secondary prevention. Europace 13, 610625 (2011).

    171. Nigam, A. etal. Fish oil for the reduction of atrialfibrillation recurrence, inflammation, andoxidative stress. J. Am. Coll. Cardiol. 64, 14411448 (2014).

    172. Imazio, M. etal. Colchicine reduces postoperative atrial fibrillation: results of theColchicine for the Prevention of the Postpericardiotomy Syndrome (COPPS) atrial fibrillation substudy. Circulation 124, 22902295 (2011).

    173. Imazio, M. etal. COlchicine for the Prevention ofthe Postpericardiotomy Syndrome (COPPS): amulticentre, randomized, doubleblind, placebocontrolled trial. Eur. Heart J. 31, 27492754 (2010).

    174. Deftereos, S. etal. Colchicine for prevention of early atrial fibrillation recurrence after pulmonary vein isolation: a randomized controlled study. J.Am. Coll. Cardiol. 60, 17901796 (2012).

    175. Deftereos, S. etal. Colchicine for prevention of atrial fibrillation recurrence after pulmonary vein isolation: midterm efficacy and effect on quality of life. Heart Rhythm 11, 620628 (2014).

    176. Weis, F. etal. Stress doses of hydrocortisone inhighrisk patients undergoing cardiac surgery: effects on interleukin6 to interleukin10 ratio and early outcome. Crit. Care Med. 37, 16851690 (2009).

    177. Yared, J.P. etal. Effect of dexamethasone on atrial fibrillation after cardiac surgery: prospective, randomized, doubleblind, placebocontrolled trial. J. Cardiothorac. Vasc. Anesth. 21, 6875 (2007).

    178. Andrade, J.G. etal. Corticosteroid use during pulmonary vein isolation is associated with a higher prevalence of dormant pulmonary vein conduction. Heart Rhythm 10, 15691575 (2013).

    179. Koyama, T. etal. Prevention of atrial fibrillation recurrence with corticosteroids after radiofrequency catheter ablation: a randomized controlled trial. J. Am. Coll. Cardiol. 56, 14631472 (2010).

    180. Cappabianca, G., Rotunno, C., de Luca Tupputi Schinosa, L., Ranieri, V.M. &Paparella, D. Protective effects of steroids in cardiac surgery: a metaanalysis of randomized doubleblind trials. J. Cardiothorac. Vasc. Anesth. 25, 156165 (2011).

    181. Won, H. etal. Effect of a single bolus injectionoflowdose hydrocortisone for preventionofatrial fibrillation recurrence after radiofrequency catheter ablation. Circ. J. 77, 5359 (2013).

    182. Cappato, R. etal. Updated worldwide survey onthe methods, efficacy, and safety of catheterablation for human atrial fibrillation. Circ.Arrhythm. Electrophysiol. 3, 3238 (2010).

    183. Themistoclakis, S. etal. Prospective EuropeanSurvey on atrial fibrillation ablation: clinical characteristics of patients and ablation strategies used in different countries. J.Cardiovasc. Electrophysiol. 25, 10741081 (2014).

    184. Kleinbongard, P., Schulz, R. & Heusch, G. TNFinmyocardial ischemia/reperfusion, remodeling and heart failure. Heart Fail. Rev. 16, 4969 (2011).

    185. Price, J.V. etal. Protein microarray analysis reveals BAFFbinding autoantibodies in systemiclupus erythematosus. J. Clin. Invest. 123, 51355145 (2013).

    186. Marik, P.E. & Fromm, R. The efficacy and dosageeffect of corticosteroids for the prevention of atrial fibrillation after cardiac surgery: a systematic review. J. Crit. Care 24, 458463 (2009).

    187. Allessie, M.A. etal. Pathophysiology and prevention of atrial fibrillation. Circulation 103, 769777 (2001).

    188. Kaireviciute, D. etal. Characterisation and validity of inflammatory biomarkers in the prediction of postoperative atrial fibrillation in coronary arterydisease patients. Thromb.Haemost. 104, 122127 (2010).

    189. Rudd, J.H. etal. Imaging atherosclerotic plaque inflammation by fluorodeoxyglucose with positron emission tomography: ready for prime time? J. Am. Coll. Cardiol. 55, 25272535 (2010).

    190. Wu, C.H. etal. Transforming growth factor1 level and outcome after catheter ablation for nonparoxysmal atrial fibrillation. Heart Rhythm 10, 1015 (2013).

    191. Leftheriotis, D.I. etal. The predictive value of inflammatory and oxidative markers following the successful cardioversion of persistent lone atrial fibrillation. Int. J. Cardiol. 135, 361369 (2009).

    192. Cao, H. etal. Heat shock proteins in stabilization of spontaneously restored sinus rhythm in permanent atrial fibrillation patients after mitral valve surgery. Cell Stress Chaperones 16, 517528 (2011).

    193. Schafler, A.E., Hannekum, A. & Kirmanoglou, K. Unchanged heatshock protein 70 expression inmyocardium of patients with permanent atrial fibrillation. J. Cardiovasc. Surg. 51, 923927 (2010).

    194. Mandal, K. etal. Association of high intracellular, but not serum, heat shock protein 70 with postoperative atrial fibrillation. Ann. Thorac. Surg. 79, 865871 (2005).

    195. St. Rammos, K. etal. Low preoperative HSP70 atrial myocardial levels correlate significantly with high incidence of postoperative atrial fibrillation after cardiac surgery. Cardiovasc. Surg. 10, 228232 (2002).

    196. Parthenakis, F.I. etal. Atrial fibrillation is associated with increased neurohumoral activation and reduced exercise tolerance in patients with nonischemic dilated cardiomyopathy. Int. J. Cardiol. 118, 206214 (2007).

    197. Wang, H. etal. Increased serum levels of microvesicles in nonvalvular atrial fibrillation determinated by ELISA using a specific monoclonal antibody AD1. Clin. Chim. Acta 411, 17001704 (2010).

    198. Hak, L. etal. Interleukin2 as a predictor ofearlypostoperative atrial fibrillation aftercardiopulmonary bypass graft (CABG). J.Interferon Cytokine Res. 29, 327332 (2009).

    199. Rizos, I. etal. Interleukin2 serum levels variations in recent onset atrial fibrillation are related with cardioversion outcome. Cytokine 40, 157164 (2007).

    200. Henningsen, K.M. etal. Prognostic impact of hsCRP and IL6 in patients undergoing radiofrequency catheter ablation for atrial fibrillation. Scand. Cardiovasc. J. 43, 285291 (2009).

    201. Henningsen, K.M. etal. Prognostic impact of hsCRP and IL6 in patients with persistent atrial fibrillation treated with electrical cardioversion. Scand. J. Clin. Lab. Invest. 69, 425432 (2009).

    202. Liuba, I. etal. Source of inflammatory markers inpatients with atrial fibrillation. Europace 10, 848853 (2008).

    203. Wu, Z.K. etal. High postoperative interleukin8 levels related to atrial fibrillation in patients undergoing coronary artery bypass surgery. World J. Surg. 32, 26432649 (2008).

    204. De Gennaro, L. etal. Inflammatory activation and carbohydrate antigen125 levels in subjects with atrial fibrillation. Eur. J. Clin. Invest. 42, 371375 (2012).

    205. Nakano, Y. etal. Matrix metalloproteinase9 contributes to human atrial remodeling during atrial fibrillation. J. Am. Coll. Cardiol. 43, 818825 (2004).

    206. Kim, S.K. etal. Serological predictors for the recurrence of atrial fibrillation after electrical cardioversion. Korean Circ. J. 40, 185190 (2010).

    207. Rosenberg, M.A. etal. Circulating fibrosis biomarkers and risk of atrial fibrillation: the Cardiovascular Health Study (CHS). Am. Heart J. 167, 723728.e2 (2014).

    208. Kimura, T. etal. Serum inflammation markers predicting successful initial catheter ablation for atrial fibrillation. Heart Lung Circ. 23, 636643 (2014).

    AcknowledgementsThis authors were supported in part by the Ministry ofScience and Technology, Taiwan (NSC1022325B010005, NSC1012321B075004, and NSC1022911I008001).

    Author contributionsAll authors substantially contributed to the discussionof content, researched data for thearticle,and wrote, reviewed, and edited the manuscript before submission.

    REVIEWS

    2015 Macmillan Publishers Limited. All rights reserved

    Inflammation and the pathogenesis ofatrialfibrillationYu-Feng Hu, Yi-Jen Chen, Yenn-Jiang Lin and Shih-Ann ChenIntroductionSources of inflammation in AFKey pointsFigure 1 | Sources of inflammation in patients with atrial fibrillation. Activatedinflammatory pathways alter the electrophysiology, structure, and autonomic remodelling of the atria. Inflammation induced by atrial fibrillation canestablish an inflammatAutoimmune reactions in AFInflammation leads to AFAF promotes inflammationFigure 2 | Inflammatory cell regulation in lone and postoperative AF. DAMPs can activate cells via TLR2 and TLR4, including immune cells, cardiomyocytes, fibroblasts, and endothelial cells, which induce inflammatory cascades. Innate and adaptive immune reImmune reactions in AFCytokines, chemokines, and mediatorsInflammation and electrical remodellingFigure 3 | HSPs in atrial fibrillation. HSPs prevent protein aggregation and stabilize protein folding. However, HSPs can function as damage-associated molecular patterns and induce an immune response. Intracellular HSP27 and HSP70 restore abnormal calciuFigure 4 | Inflammatory cells and mediators of inflammation modulate cardiacelectrophysiology and structural properties. Calcium homeostasis in cardiomyocytes is regulated by TNF, PDGF, and IL2, which are associated with increased triggering and shortenInflammation and structural remodellingInflammation and thrombogenicityFigure 5 | The pathophysiological link between inflammation and thrombogenesis. Immune cell infiltration induces endothelial dysfunction, which decreases eNOS expression, but increases the expression of vonWillebrand factor, thrombin, tissue factor, and Current anti-inflammatory therapiesFuture targeting of inflammation in AFConclusionsAcknowledgementsAuthor contributions