Nagai Fibro Mac Rev

  • Upload
    hn

  • View
    242

  • Download
    0

Embed Size (px)

DESCRIPTION

Nagai Fibro Mac Rev

Citation preview

  • INVITED REVIEW

    Contributions of cardiomyocytecardiac fibroblastimmunecell interactions in heart failure development

    Katsuhito Fujiu Ryozo Nagai

    Received: 22 February 2013 / Revised: 13 March 2013 / Accepted: 21 March 2013 / Published online: 6 June 2013

    Springer-Verlag Berlin Heidelberg 2013

    Abstract The heart contains various types of cells,

    including cardiomyocytes, cardiac fibroblasts, many kinds of

    immune cells and vascular cells. Initial studies mainly

    focused on cardiomyocytes, which directly reflect the con-

    tractile function of the heart. Recently, pivotal functions of

    cardiac fibroblasts have been revealed in the maintenance of

    cardiac function, physiological cardiac remodeling after heart

    stress and pathological remodeling using genetically engi-

    neered mouse models, like the fibroblast-specific gene

    knockout mouse, bone marrow transplantation and immune

    cell-specific gene knockout. Moreover, chronic inflammation

    is considered to be a basic pathological mechanism that

    underlies various diseases, including heart failure. In the

    development of heart failure, the contributions of immune

    cells like T lymphocytes and monocyte/macrophage lineage

    cells have been also reported. Immune cells have diverse and

    multiple functions in regulating both pro-inflammatory

    effects and the resolution of heart failure. On the one hand,

    immune cells have protective effects to compensate for and

    overcome heart stresses. On the other hand, they also con-

    tribute to sustained inflammation and result in the develop-

    ment of heart failure. These observations prompted a shift in

    the heart-related studies to include the complex communi-

    cations between cardiomyocytes and other kinds of cardiac

    cells, including inflammatory cells residing in or recruited to

    the heart. This review will summarize the current knowledge

    regarding cellcell interactions during cardiac remodeling

    and the development of heart failure. We will especially

    focus on the interactions among cardiomyocytes, cardiac

    fibroblasts and immune cells.

    Keywords Cardiac fibroblast Cardiomyocyte Cellcellinteraction Heart failure Immune cell

    Introduction

    In the heart, cardiac fibroblasts, immune cells and vascular

    cells, including vascular endothelial cells and vascular

    smooth muscle cells, are all present. Under steady-state

    conditions, non-myocytes may exhibit a quiescent pheno-

    type, whereas cardiomyocytes are always beating. After the

    exposure to heart stress, such as myocardial infarction,

    pressure overload, myocarditis, volume overload and so on,

    the cardiac fibroblasts change their phenotype to become

    activated fibroblasts, which produce growth factors, cyto-

    kines, chemokines and extracellular matrix [46]. Simulta-

    neously, a variety of immune cells, such as T lymphocytes

    and monocyte/macrophage lineage cells infiltrate into the

    heart. These T lymphocytes and monocytes/macrophages

    have diverse functions, which are regulated in a spatial and

    This article is part of the Topical Collection Novel Perspectives on

    Heart Failure.

    K. Fujiu (&) R. Nagai (&)Department of Cardiovascular Medicine, University of Tokyo,

    7-3-1, Hongo, Bunkyo, Tokyo 113-8655, Japan

    e-mail: [email protected]

    R. Nagai

    e-mail: [email protected]

    K. Fujiu

    Translational Systems Biology and Medicine Initiative

    (TSBMI), the University of Tokyo Graduate School of Medicine,

    Tokyo, Japan

    R. Nagai

    Funding Program for World-Leading Innovative R&D on

    Science and Technology (FIRST Program), Tokyo, Japan

    R. Nagai

    Jichi Medical University, Tochigi, Japan

    123

    Basic Res Cardiol (2013) 108:357

    DOI 10.1007/s00395-013-0357-x

  • temporal manner [93, 106]. The cellcell interactions

    among cardiomyocytes and non-myocytes within the

    interstitium of the heart have become an increasingly

    popular focus of research on heart failure. These interac-

    tions promote adaptive responses against heart stress, and

    compensate or overcome these stresses. On the other hand,

    these interactions can also provoke pathological remodel-

    ing associated with heart diseases, and can thereby result in

    heart failure and cardiac death. In this review, we will

    present the current knowledge regarding cellcell interac-

    tions in the heart after heart stress, such as myocardial

    infarction and pressure overload. We will especially focus

    on the contribution of cellcell interactions among

    cardiomyocytes, cardiac fibroblasts and immune cells to

    heart failure.

    Cellular components of the heart

    Cardiomyocytes, cardiac fibroblasts, endothelial cells,

    vascular smooth muscle cells and immune cells are all

    major cellular components of the heart. In the 1970s,

    efforts to establish the cellular populations of the adult

    heart in rats were reported, based on a morphological

    analysis using electron microscopy or gradient centrifuga-

    tion. Recently, several common cardiac fibroblast markers

    have been reported. Discoidin domain receptor 2 (DDR2)

    [8] and CD90/thymus cell antigen-1 (Thy1) [42] are cell-

    surface markers that can be used for fluorescence-activated

    cell sorting (FACS). These discoveries showed that the

    adult murine myocardium is composed of 56 % myocytes,

    27 % fibroblasts, 7 % endothelial cells and 10 % vascular

    smooth muscle cells [8]. Moreover, in the adult rat heart,

    the number of cardiomyocytes was 30 %, that of fibroblasts

    was 64 % and non-myocyte and non-fibroblast cell popu-

    lations made up the remaining 6 %, which includes

    immune cells and vascular cells [8]. The approaches using

    FACS enabled the analysis of the relative percentages of

    distinct, definable cell types present in the developing and

    adult heart, as well as the elucidation of the cellcell

    communications among various cells within the heart

    during the homeostatic state, adaptive response state and

    during the development of heart disease.

    Cardiomyocytecardiac fibroblast interaction

    The important role of cellcell communication between

    cardiomyocytes and cardiac fibroblasts in both develop-

    ment and/or cardiovascular diseases was implied by an

    initial in vitro study using conditioned media or a simpli-

    fied co-culture system [25]. For example, murine cardio-

    myocytes developed a hypertrophied phenotype when they

    were co-cultured with cardiac fibroblasts or their condi-

    tioned media supplemented with interleukin (IL)-6. In that

    report, the IL-6 signaling in cardiomyocytes in co-culture

    with cardiac fibroblasts was suggested to be key in pro-

    moting cardiomyocyte hypertrophy [31]. In addition to

    IL-6, the tumor necrosis factor (TNF)a production was alsoupregulated by cardiomyocytes when they were co-cul-

    tured with cardiac fibroblasts [9]. These results from co-

    culture systems suggested that paracrine factors are one of

    the method by which cardiomyocytes and cardiac fibro-

    blasts communicate. However, forced physical disconnec-

    tion between cardiomyocytes and cardiac fibroblast using

    an antibody for a cardiac fibroblast plasma membrane

    protein or connexin 43 inhibited cell adhesion and

    decreased the IL-6 production, but did not decrease the

    TNFa production [9]. These results highlighted theimportance of direct cellcell interactions between

    cardiomyocytes and cardiac fibroblasts, in addition to the

    existence of multiple non-physical communication path-

    ways mediated by cytokine production, during the devel-

    opment of cardiac hypertrophy (Fig. 1). In addition to

    cardiomyocyte hypertrophy, a recent in vitro study sug-

    gested that cardiac fibroblasts also affected the contractile

    activity [59] and electrophysiological conditions of the

    cardiomyocytes [77]. These results suggested that cardiac

    fibroblasts may have pivotal pathological roles via their

    interactions with cardiomyocytes in vivo. Many studies

    have shown that cardiac fibroblasts were activated by heart

    stresses and that they became proliferative and secreted

    cytokines and growth factors and eventually differentiated

    into cardiac myofibroblasts, which expressed smooth

    muscle a-actin. There are a wide variety of heart stresses,including mechanical stress, hypoxia [76], immune cell-

    derived inflammatory cytokines [107] and augmented

    neurohormonal stimulation [64].

    Among these stresses, the reninangiotensin system and

    b-adrenergic stimulation have been extensively investi-gated. Cardiac fibroblasts were found to express the

    angiotensin II receptor [99] and b-adrenergic receptors[100] and these receptors were involved in the pathological

    process of heart diseases due to enhanced reninangioten-

    sinaldosterone signaling [35, 82, 84] and chronic

    b-adrenergic overstimulation [73]. These two signalingpathways have been established as standard targets for the

    treatment of heart failure. Recently, the interdependence

    between angiotensin II receptor 1 and the serotonin

    receptor [5-HT(2B)] in cardiac fibroblasts was reported

    [44]. These two receptors in cardiac fibroblasts affected the

    sympathetic overstimulation-dependent heart failure, thus

    suggesting that 5-HT(2B) might be a novel therapeutic

    target for adrenergic overstimulation-dependent heart fail-

    ure [44]. In addition to these signaling pathways, nuclear

    factor kappa-light-chain-enhancer of activated B cells

    Page 2 of 15 Basic Res Cardiol (2013) 108:357

    123

  • (NF-jB) [5, 53], the Smad signaling pathway[38], mitogenactivated protein kinase [72] and phosphoinositide 3-kinase

    (PI3K) [15] were also suggested to be involved in cardiac

    fibroblast activation during the development of heart fail-

    ure, and might also be candidate therapeutic targets.

    Cyclic nucleotide phosphodiesterase 1A (PDE1A) is a

    key regulator of cardiac fibroblast activation induced by

    angiotensin II and TGF-b through the Ca2?/calmodulinpathway both in vitro and in vivo [71].

    The extracellular heterodimeric protein S100A8/A9 is

    produced in fibroblasts and macrophages in the heart fol-

    lowing myocardial ischemia [104]. The production of

    S100A8/A9 leads to increases in the activity of nuclear

    factor (NF)-jB and the expression of proinflammatorycytokines in cardiac fibroblasts and macrophages [104].

    S100A8/A9 recruits macrophages into the heart by acti-

    vating the receptor of advanced glycation end-products

    (RAGE) on macrophage [104]. Secreted S100A8/A9

    affects cardiomyocytes by activating MAP kinases JNK,

    ERK1/2 and NF-jB, which mediates signals downstreamof RAGE following ischemic heart failure and results in a

    reduced cardiac function [104].

    The TNF receptor superfamily member fibroblast

    growth factor-inducible molecule 14 (Fn14) is produced

    from cardiac fibroblasts in response to endothelin-1 stim-

    ulated by right ventricular pressure overload [74]. Fn14

    activates and leads to the proliferation of cardiac fibroblast

    cells autonomously, results in collagen synthesis via RhoA-

    dependent nuclear translocation of myocardin-related

    transcription factor-A (MRTF-A)/MAL [74].

    In just the past decade, more details about the cellcell

    interactions between cardiomyocytes and cardiac fibro-

    blasts have been revealed by genetically manipulated mice

    using the Cre-loxP system and estrogen receptor-inducible

    system. This technique enabled the generation of cardio-

    myocyte-specific [4, 39] and fibroblast-specific [45]

    IL-6 family IL-6 family

    GP130 GP130

    cardiomyocyte cardiac fibroblast

    JAK MAPK PI3K

    hypertrophy cell death

    iNOS

    SERCAPLBRYR

    contraction

    extracellularmatrix

    production

    migrationproliferation

    XONADPH

    superoxideNO

    peroxynitrite

    Connexin43

    IL-6 production

    Stressinduced AngIIproduction

    AngIIAT1

    Fibroblast activation

    AngIIIL-6 family

    Fig. 1 Interleukin-6 family regulates the heart remodeling bymodulating cardiomyocytes and cardiac fibroblasts. Current knowl-

    edge of cellular molecular mechanisms is shown. In cardiomyocytes,

    IL-6 induces cardiac hypertrophy via JAK signaling, cardiomyocyte

    death via MAPK and inducible nitric oxide synthase, xanthine

    oxidase and NADPH oxidase expression through their receptor

    GP130. Induced superoxide and nitric oxide produce peroxynitrite.

    Peroxynitrite decreases contractility through its effects on SERCA2a,

    ryanodine receptor and phospholamban. In cardiac fibroblasts, IL-6

    induces activation of extracellular matrix production and migration

    and proliferation of cardiac fibroblasts. Cardiac stresses induce

    angiotensin II production from cardiac fibroblasts and secreted

    angiotensin II induces IL-6 production of cardiomyocytes. IL

    interleukin, JAK Janus Kinase, MAPK mitogen activated protein

    kinase, PI3K Phosphoinositide 3 kinase, SERCA Sarcoplasmic

    reticulum calcium ATPase, XO xanthine oxidase, PLB phospholam-

    ban, iNOS inducible nitric oxide synthase, RYR ryanodine receptor,

    AngII angiotensin II, AT1 angiotensin II receptor type 1, IL-6 family

    IL-6 leukemia inhibitory factor and cardiotropin-1

    Basic Res Cardiol (2013) 108:357 Page 3 of 15

    123

  • knockout or overexpression mice, and allowed the analysis

    of cell type-specific functions in vivo. Haploinsufficiency

    of a transcription factor, Klf5, inhibited cardiac hypertro-

    phy and cardiac fibrosis after angiotensin II infusion or

    pressure overload [86, 94]. However, cardiomyocyte-spe-

    cific Klf5 knockout mice did not show any reduction of the

    cardiac hypertrophy and fibrosis after left ventricular

    pressure overload. On the other hand, fibroblast-specific

    Klf5 knockout mice showed significantly suppressed car-

    diac hypertrophy and fibrosis [94]. However, the fibroblast-

    specific Klf5 knockout mice subsequently developed severe

    heart failure, because of a truncation of the adaptive car-

    diac response against pressure overload, including cardiac

    hypertrophy and fibrosis, which resulted in heart failure

    and cardiac death (Fig. 2).

    Placental growth factor (Pgf) is another key regulator

    of the interaction between cardiomyocytes and cardiac

    fibroblasts. Pgf knockout mice died of severe heart failure

    within a week after pressure overload [1]. Reductions of

    fibroblast activation and angiogenesis after pressure

    overload were observed. In contrast, the overexpression

    PGF in cardiomyocytes augmented the cardiac hypertro-

    phy in response to pressure overload [1]. Because the PGF

    receptor is expressed in cardiac fibroblasts, but not in

    cardiomyocytes, the PGF secreted from cardiomyocytes

    only affects non-cardiomyocytes (mainly cardiac fibro-

    blasts) and promoted cardiac adaptive responses after

    pressure overload. Ras-associated domain family 1 iso-

    form A (Rassf1a), a tumor suppressor gene, was reported

    to enhance the secretion of TNFa by cardiac fibroblasts,and this secreted TNFa facilitates the increases of thecardiac hypertrophy and cardiac fibroblast proliferation

    [20]. These reports also suggested that cardiac hypertro-

    phy and cardiac fibrosis are part of an appropriate adap-

    tive response, and that the loss of these adaptive responses

    clearly resulted in a poor prognosis. Although cardiac

    hypertrophy and cardiac fibrosis can be both a positive

    and negative response to the physiological conditions, the

    differences in the survival rates after heart stress in

    genetically manipulated mice strongly support the

    importance of these responses in the protection against

    heart stress.

    KLF5

    IGF-1PDGF-A

    KLF5 KLF5

    Wild type cardiac fibroblast specific Klf5knockout mouse

    a

    b

    pressure overloadangiotensin II infusion

    cardiac hypertrophycardiac fibrosis

    adaptive responsetruncation of adaptive responseby Klf5 deficiency in cardiac fibroblasts

    htaeddnaeruliaftraehevila

    pressure overload

    Fig. 2 Cardiac fibroblasts facilitate adaptive responses againstcardiac pressure overload. a In cardiac fibroblasts, transcriptionfactor Kruppel-like factor 5 induces insulin-like growth factor 1 and

    platelet-derived growth factor A chain after pressure overload. IGF1

    is required for induced cardiac remodeling including cardiac hyper-

    trophy and cardiac fibrosis after pressure overload and angiotensin II

    infusion. b Although in wild-type mice, cardiac hypertrophy and

    cardiac fibrosis are observed after pressure overload, this stress is

    compensated by these appropriate responses and all mice can survive.

    Cardiac fibroblast-specific Kruppel-like factor 5 knock mice develop

    heart failure and show high mortality by lack of these adaptive

    responses. KLF5 Kruppel-like factor 5, IGF1 insulin-like growth

    factor 1, PDGF-A platelet-derived growth factor A chain

    Page 4 of 15 Basic Res Cardiol (2013) 108:357

    123

  • Cardiomyocytes and neuronal/vascular cell interactions

    The sympathetic nervous system controls cardiac contrac-

    tility, heart rate and cardiac hypertrophy through interac-

    tion of neurons and cardiomyocytes by norepinephrine

    secretion [109]. In neurons, angiotensin II induces and

    nitric oxide inhibits sympathetic activation and norepi-

    nephrine release [119]. In experimental models, ablation of

    sympathetic nerves results in reduced cardiomyocytes size

    [113]. These effects are mediated by the ubiquitin protea-

    some followed by activation of autophagy. This atrophic

    remodeling is caused by the reduction of basal stimulation

    of adrenergic b2-receptors [113] and can be mimicked byneuron-specific angiotensin converting enzyme 2 overex-

    pression in mice which have lower norepinephrine levels.

    The reduction of sympathetic activity protects against

    cardiac hypertrophy induced by angiotensin II infusion

    [27]. These results suggest that sympathetic neuroncar-

    diomyocyte interactions induced by norepinephrine pro-

    duction are required for the regulation of the

    cardiomyocyte volume in maintenance and pathological

    remodeling.

    In terms of inflammation, vascular endothelial cells also

    contribute to heart failure development by expressing cell

    adhesion molecules. Cell adhesion molecules, including

    selectins, integrins and the immunoglobulin gene super-

    family of adhesion receptors, provoke leukocyte migration

    from blood vessels into the cardiac interstitium [34].

    Endothelial cell dysfunction via reduction of nitric oxide

    production induces the expression of intercellular adhesion

    molecule-1 (ICAM-1) and vascular cell adhesion mole-

    cules-1 (VCAM-1). These cell adhesion molecules induce

    the interaction of leukocytes with endothelial cells and

    result in the accumulation of leukocytes; infiltrating leu-

    kocytes provoke the interaction of immune cells, cardio-

    myocytes and fibroblast via the actions of proinflammatory

    cytokines.

    Blood vessels supply oxygen for cardiac contraction. In

    response to pressure overload, cardiomyocytes produce

    VEGF and induce angiogenesis, thus adapting supply to the

    increased oxygen demand of hypertrophic cardiomyocytes.

    In later stages, p53 inhibits VEGF production from

    cardiomyocytes and reduced oxygen supply contributes to

    reduced contractile function and heart failure [83].

    Contribution of inflammation in heart failure

    development

    The levels of many cytokines, chemokines and growth

    factors were observed to be augmented during the adaptive

    responses against heart stress and the development of heart

    failure. These factors induce phenotypic changes in steady-

    state cardiomyocytes and quiescent cardiac fibroblasts to

    generate the active form of these cells, i.e. abundant

    cytokine-producing cardiomyocytes and cardiac fibro-

    blasts. Consequently, the increased paracrine factors from

    these two kinds of cells can recruit immune cells and

    provoke acute inflammatory changes. Recruited inflam-

    matory cells from the bone marrow and spleen interact with

    cardiomyocytes and cardiac fibroblasts, and make more

    complex inflammatory statuses [46]. An acute response

    that includes the infiltration of immune cells, like granu-

    locytes and inflammatory monocytes/macrophages, into the

    heart and the increased vascular permeability mainly exerts

    favorable effects on the heart suffering from stresses and

    injury, and enhances the cardiac repair, wound healing,

    adaptive cardiac hypertrophy, etc. Supporting this idea, the

    inhibition of acute inflammatory cell infiltration into the

    heart by depletion of CC chemokine receptor 5 or intra-

    venous injections of clodronate-containing liposomes

    worsened the cardiac remodeling and healing after myo-

    cardial infarction in model mice [102, 114]. However,

    chronic inflammation has recently been reported to be a

    common mechanism underlying many kinds of lifestyle-

    related diseases, including coronary heart disease, diabetes

    mellitus and cancer. Serum markers of inflammation, such

    as the sustained elevation of inflammatory cytokines and

    growth factors, in heart failure patients also augmented and

    reflected the severity of their heart failure and clinical

    outcomes [81, 97].

    In chronic inflammation, macrophages undergo polari-

    zation and contribute to tissue injury. In myocardial

    infarction, classically activated macrophages produce pro-

    inflammatory cytokines (M1 macrophages), and alterna-

    tively activated macrophages (M2) anti-inflammatory

    cytokines; both, are increased in the heart. Class A scav-

    enger receptor (SRA) knockout mice have a reduction of

    infiltrated M2 macrophages and increased M1 macro-

    phages that result in exacerbation of the cardiac dysfunc-

    tion and fibrosis following myocardial infarction [41]. A

    particular importance of SRA for the M2 phenotype

    polarization has been reported [41].

    Bone marrow cells-derived mesenchymal stromal cells

    (MSCs) also contribute to repair through their anti-

    inflammatory effects. MSCs infiltrate into the heart fol-

    lowing myocardial infarction and secrete interleukin-10,

    which mediates a switch from an inflammatory monocyte/

    macrophage lineage to anti-inflammatory lineages [41].

    Regulatory T-cells are also suggested to contribute to

    M2 polarization following myocardial infarction via IL-10

    production [95].

    In this context, the communication among immune cells,

    cardiomyocytes and cardiac fibroblasts in the interstitium

    of the heart leads to the formation of an immunoregulatory

    network, and chronic inflammation might occur if these

    Basic Res Cardiol (2013) 108:357 Page 5 of 15

    123

  • cells fail to switch off their inflammatory programs, which

    might lead to the inappropriate survival and retention of

    leukocytes [11].

    Intercellular mediators control cellcell interactions

    Intercellular mediators produced by immune cells, cardio-

    myocytes and cardiac fibroblasts are key factors in the

    complex mechanism of connection among the different

    types of cells, and have provided new therapeutic targets

    for heart failure. In fact, during the development of heart

    failure, large numbers of pro-inflammatory mediators are

    secreted and contribute to the cellular dysfunction, modu-

    late cellcell interactions, affect numerous signaling path-

    ways and the cross-talk among signaling pathways [65].

    Below, we describe the best-characterized mediators by

    which immune cells, cardiomyocytes and cardiac fibro-

    blasts interact during the development of heart failure and/

    or the progression of other heart diseases.

    Interleukins

    Interleukins are a group of cytokines which are mainly

    produced by immune cells, like T cells and monocyte/

    macrophage lineage cells, as well as cardiomyocytes, car-

    diac fibroblasts and vascular endothelial cells. IL-6 and its

    signal transducer, a 130-kDa glycoprotein (gp130; CD130)

    is expressed in cardiomyocytes and cardiac fibroblasts, and

    is associated with cardiac hypertrophy and cardiac fibrosis

    [30] (Fig. 1). IL-6 is upregulated in the heart after myo-

    cardial infarction [14, 22, 47]. The angiotensin II secreted

    from cardiac fibroblasts also induced IL-6 production from

    the cardiomyocytes, and an AT-1 receptor antagonist

    inhibited cardiomyocyte hypertrophy and fibroblast pro-

    liferation [30]. These results suggested that the IL-6 pro-

    duction in cardiomyocytes contributes significantly to

    cardiomyocyte hypertrophy by an autocrine pathway and to

    cardiac fibroblast proliferation by a paracrine pathway.

    Angiotensin II can induce various members of the IL-6

    family, including IL-6, leukemia inhibitory factor and

    cardiotropin-1, in cardiac fibroblasts [82]. These IL-6

    family members induce significant hypertrophy of cardio-

    myocytes through gp130 [82]. The administration of a

    neutralizing antibody against IL-6 or gp130 (CD130)

    resulted in the reduction of hypertrophic gene expression.

    IL-6 blockade also led to reduced cardiac fibroblast pro-

    liferation. Signal transducer and activator of transcription 3

    (STAT3) is a key signal transducer that acts downstream of

    gp130 in both cardiomyocytes and cardiac fibroblasts [28,

    118]. Gp130 and STAT3 phosphorylation are altered in

    end-stage dilated cardiomyopathy patients [79]. The

    induction of signaling via the gp130STAT3 axis provoked

    unfavorable downstream activation of this critical pathway

    during heart failure [40]. The IL-6 family and gp130 axis is

    the most extensively analyzed signal transduction pathway

    between cardiomyocytes and cardiac fibroblasts in terms of

    the effects on cardiac hypertrophy and cardiac fibrosis.

    IL-1b and its receptor, the type I IL-1 receptor (IL-1RI), are also markedly induced in the infarcted heart,

    and this signaling is essential for the activation of the

    fibrogenic pathways in the healing heart [12]. IL-1bleads to extra cellular matrix remodeling by inducing the

    migration of cardiac fibroblasts [72] and matrix

    metalloproteinase production [87], which promotes car-

    diac dilatation and cardiac rupture. IL-1b also inducescardiac dysfunction through nitric oxide production [85],

    modifying b-adrenergic signaling [37] and via a reduc-tion of phospholamban [70]. On the other hand, IL-1RI

    knockout mice showed decreased infiltration of neutro-

    phils and macrophages into the myocardium after

    ischemia/reperfusion injury [12]. Mice lacking the IL-1

    receptor also showed a reduction of myofibroblast acti-

    vation and collagen deposition, but no reduction of the

    infarct size [12]. These results suggest that IL-1 signal-

    ing regulates pathogenic cardiac remodeling in both

    cardiomyocytes and cardiac fibroblasts.

    IL-17, a recently identified cytokine, is released from

    Th17 cells, which are a subset of CD4 effector T cells. IL-

    17 is also induced in the infarct area after myocardial

    infarction, and depletion of IL-17 by genetic deficiency or

    a neutralizing antibody resulted in a reduction of the infarct

    size and preserved cardiac function [7, 63]. The mecha-

    nisms underlying these effects of IL-17 are suggested to be

    due to the fact that IL-17 provokes the apoptosis of

    cardiomyocytes and induces chemokines that mediate

    neutrophil migration [63]. In addition to Th17 cells, recent

    in vitro studies suggested that cardiac fibroblasts can pro-

    duce IL-17 and provoke collagen and metalloproteinase 1

    production through the IL-17 receptor expressed on the

    cardiac fibroblasts [16, 103]. These results imply that IL-17

    is a key mediator linking Th17 cells, cardiac fibroblasts and

    cardiomyocytes that contributes to pathological

    remodeling.

    IL-10 is a general anti-inflammatory cytokine that is

    also induced after cardiac reperfusion injury or heart

    failure in a canine model [29] and in humans [108].

    Elevated IL-10 is considered to antagonize the effects of

    pro-inflammatory cytokines like TNFa [51]. A studydemonstrated that IL-10 reduced the oxidative stress and

    TNFa-induced apoptosis of cardiomyocytes in vitro, andthe administration of IL-10 after myocardial infarction led

    to a reduction of the inflammatory cell invasion and

    inflammatory cytokine production and resulted in an

    ameliorated infarct area and cardiac remodeling [23].

    These results suggest that IL-10 has important roles in the

    Page 6 of 15 Basic Res Cardiol (2013) 108:357

    123

  • adaptive response during myocardial infarction and heart

    failure by affecting both cardiomyocytes and cardiac

    fibroblasts.

    TNFa

    Tumor necrosis factor is a cytokine involved in inflam-

    mation that is recognized by its receptors, TNF receptor

    type I (TNF-R1) and TNF-R2 [53]. The serum level of

    TNFa is elevated in heart failure patients in a mannercorresponding to the severity of the heart failure [60].

    TNFa provokes various effects on TNF-R expressing tis-sues, including the myocardium [98], through the MAPK

    and/or NF-jB pathways. Pathological conditions likehemodynamic pressure overloading or myocardial infarc-

    tion induce TNFa production in both cardiomyocytes andnon-cardiomyocytes [18, 43, 49]. TNFa induced the pro-liferation of cardiac fibroblasts and collagen deposition,

    increasing the matrix metalloproteinase activity and

    inflammatory cytokine production from cardiac fibroblasts,

    and also induced the apoptosis of cardiomyocytes and

    resulted in cardiac dysfunction and heart failure [91]. As

    expected based on these findings, Tnf knockout mice

    showed a reduction of cardiac death, infarct size, myo-

    cardial apoptosis, inflammatory cell infiltration, inflam-

    matory cytokine production, MMP activity and deposition

    of extracellular matrix after myocardial infarction [91].

    Phosphatase and tensin homolog deleted on chromo-

    some ten (PTEN) inactivates protein kinase Akt and pro-

    motes cell death in the heart [75]. PTEN is also induced in

    the heart following myocardial infarction, while PTEN

    heterozygous knockout mice show a reduction of the

    number of infiltrated immune cells in the heart and a pre-

    served cardiac function following myocardial infarction

    [75]. In PTEN heterozygous knockout mice, the production

    of TNFa and MMP-2 is decreased and the production ofIL-10 is increased in the heart following myocardial

    infarction [75]. In addition, inhibition of IL-10 receptors

    increases TNFa and MMP-2 production following myo-cardial infarction [75]. PTEN is also important in remote

    ischemic preconditioning (RIPC) [13]. PTEN is inactivated

    in limb muscles following lower limb RIPC. The inacti-

    vation of PTEN promotes STAT3 phosphorylation and

    induces IL-10 production in limb muscles. The IL-10

    released from ischemic skeletal muscles activates protec-

    tive signaling pathways in the heart [13]. These results

    suggest that PTEN is critically involved in the post-myo-

    cardial infarction remodeling induced by TNFa via theAkt/IL-10 signaling pathway.

    TNFa directly impairs the contractility of cardiomyo-cytes through sphingosine, a metabolite stimulated by

    TNFa binding, by decreasing the intracellular calciumrelease and inotropic activity [32, 96], inhibiting the

    cardiac L-type calcium channel current and contractile

    calcium transients [55], reducing the SERCA2A expression

    and activity [48] and reducing the b-adrenergic respon-siveness [37]. In addition, TNFa can directly induce car-diomyocyte hypertrophy [110] and apoptosis [56]. These

    findings indicate that TNFa might be a key to the devel-opment of heart failure and could be a therapeutic target for

    heart failure. Indeed, several preclinical studies showed

    promising data that indicated favorable results for a TNFablocking strategy against heart failure. In an animal model

    of myocardial infarction, TNFa also had ambivalentfunctions [88]. Indeed, clinical trials of anti-TNFa therapyusing the soluble TNFa selective antagonist, etanercept, forNew York Heart Association class II to IV chronic heart

    failure patients (ejection fraction B 30 %) were terminated

    prematurely due to a lack of benefit. A subanalysis of these

    studies concluded that the outcome of patients after the

    administration of etanercept was worsened compared to

    patients who did not receive the drug [66]. These results

    implied that the TNFa signaling in cardiomyocytes andnon-cardiomyocytes in patients with heart failure is more

    complex than was previously thought. Moreover, the

    intervention targeting this single molecule associated with

    inflammation and heart failure did not bring about favor-

    able effects, thus suggesting that chronic inflammation

    after heart stress may actually be a favorable adaptive

    response, and that excessive blockade of the inflammatory

    responses during heart failure might lead to decompensa-

    tion. More cell type-specific analyses of this signaling

    pathway under different conditions will be required.

    Transforming growth factor-b

    Transforming growth factor b (TGF-b) is expressed incardiac fibroblasts, cardiomyocytes and vascular cells, and

    is induced in the myocardium by myocardial infarction and

    heart failure [19, 21]. Classical TGF-b signaling occurs viabinding to the TGF-b type 2 receptor and activation of theTGF-b type I receptor. Thereafter, both a Smad-dependentpathway and non-canonical pathway (Smad-independent)

    are activated. In the non-canonical pathway, PDE1A-

    mediated cardiac fibroblast activation and cardiac fibrosis

    in response to TGFb have recently been reported [71]. Inthis report, PDE1A has a pivotal function in the develop-

    ment of fibrosis induced by both TGFb and angiotensin II[71]. Blockade of the classical TGF-b signaling usingsystemic Smad3 knockout in mice led to their development

    of severe hypertrophy and less fibrosis after left ventricular

    pressure overload [24]. On the other hand, cardiomyocyte-

    specific forced non-canonical TGF-b activation by over-expression of TGF-b activated kinase 1 (TAK1) alsoinduced cardiac hypertrophy and heart failure in mice

    [117]. A left ventricular pressure overload mouse model

    Basic Res Cardiol (2013) 108:357 Page 7 of 15

    123

  • treated with a TGF-b signal neutralizing antibody resultedin suppression of the classical pathway (Smad) activation

    in the interstitium (non-cardiomyocytes), but not in

    cardiomyocytes, and the non-canonical activation (TAK1

    activation) was also not affected. Although the cardiac

    fibrosis was markedly suppressed, the cardiac dysfunction

    was not ameliorated in this mouse model [54]. These

    results based on systemic knockout mice and systemic

    protein blockade methods have limitations for analyzing

    the mechanism(s) underlying complex cellcell interac-

    tions, because the same gene may work differentially in

    different kinds of cells. A cell type-specific analysis of

    TGF-b signaling will be necessary to better understand therole of the molecule in heart failure.

    In response to this need, cardiomyocyte-specific TGF-breceptor knockout mice were recently reported [54]. These

    mice showed reduced cardiac hypertrophy and cardiac

    fibrosis as a result of the inhibited cardiomyocyte and

    interstitial Smad and TAK1 activation [54] (Fig. 3). Inter-

    estingly, the non-Smad pathways in cardiomyocytes,

    including the Ras-MEK, Rho GTP-ase, phosphoinositide-3-

    kinase and TAK1 pathways, might be the predominant

    pathways leading to cardiac hypertrophy and cardiac dys-

    function, because cardiomyocyte-specific Smad4 knockout

    mice exhibit hypertrophy and heart failure [105] and Smad3

    knockout worsens the hypertrophic response to pressure

    overload [24]. Among the non-Smad pathways, TAK1 can

    phosphorylate P38-MAPK, which promotes cardiac dys-

    function [62]. In cardiomyocytes, connective tissue growth

    factor and bone morphogenetic protein 7 are key paracrine

    factors connecting these cells to cardiac fibroblasts in

    response to TGF-b signaling [54]. Connecting tissue growthfactor is thought to promote myofibroblast activation and to

    affect cardiomyocyte dysfunction, in addition to Smad

    signaling in the development of cardiac fibrosis, hypertro-

    phy and myocarditis [36, 54, 68, 111]. Bone morphogenetic

    protein 7 was also shown to suppress TGF-b-mediatedcardiac fibrosis and the epithelial mesenchymal transition

    [116]. TGF-b1 stimulation can suppress bone morphoge-netic protein 7 by a TAK1-dependent pathway [54]. The

    non-Smad pathway in cardiomyocytes is becoming gradu-

    ally recognized to have a major role in TGF-b signaling,especially in the communication between cardiomyocytes

    and cardiac fibroblasts, in terms of heart failure.

    Insulin-like growth factor

    Insulin-like growth factor-1 (IGF1) is expressed in cardiac

    fibroblasts and promotes cardiac cardiomyocyte hypertro-

    phy through phosphoinositide 3-kinase signaling [69].

    IGF1 is a downstream target of KLF5 that is activated by

    KLF5 after pressure overload [94] (Fig. 2). The adminis-

    tration of an IGF-1 inhibitor to wild-type mice with

    pressure overload also led to the development of severe

    heart failure [94]. These results clearly show that KLF5-

    IGF-1 signaling in cardiac fibroblasts, but not in cardio-

    myocytes, is required for adaptive responses, like cardiac

    hypertrophy and cardiac fibrosis, during pressure overload.

    Natriuretic peptide

    The serum levels of atrial natriuretic peptide (ANP) and

    brain natriuretic peptide (BNP) are clinically used as

    diagnostic markers which reflect volume overload in heart

    failure and the therapeutic index of heart failure treatment.

    Mice that were genetically deficient in the common

    receptor for these two peptides, guanylyl cyclase-A,

    showed marked cardiac hypertrophy and fibrosis, suggest-

    ing that ANP and BNP have protective effects against

    cardiac hypertrophy and fibrosis [52]. ANP and BNP are

    produced by cardiomyocytes, and BNP directly affects and

    protects cardiomyocytes following myocardial infarction

    by opening ATP-sensitive potassium (KATP) channel [17].

    BNP also directly affects cardiac fibroblasts and limits

    TGF-b-mediated gene expression associated with cardiacfibrosis and cardiac fibroblast proliferation [50]. In addi-

    tion, ANP reduces the endothelin-1 expression, which can

    exert proliferative effects on cardiac fibroblasts in vitro

    [33]. These data suggest that these two peptides from

    cardiomyocytes antagonize the fibrogenic activity of car-

    diac fibroblasts in a paracrine fashion.

    Immune cells and cardiomyocytes/cardiac fibroblasts

    interactions

    Several kinds of immune cells proliferate or infiltrate into

    the heart after cardiac stress, and are considered to promote

    cardiac remodeling. Monocytes (Ly-6Chigh at acute phase

    and Ly-6Clow at late phage) and macrophages are one of

    the major immune cell types that accumulate in the heart

    during stress, such as myocardial infarction or heart failure.

    The origin of cardiac macrophages is believed to be from

    bone marrow-derived monocytes, which differentiate into

    macrophages in the heart, or may be the result of the

    proliferation of resident macrophages within the heart.

    Recently, splenic monocytes were reported to function as a

    reservoir of Ly-6C high-inflammatory monocytes that

    participate in the repair of heart disease in the acute phase

    [92]. In addition, this rapid deployment of inflammatory

    monocytes from the spleen depends on angiotensin sig-

    naling [92].

    Monocyte chemotactic protein-1 (MCP-1) is a major

    chemokine that recruits inflammatory monocytes into var-

    ious tissues. In a pressure overload model, MCP-1 was

    upregulated in the early stage of heart remodeling. Chronic

    Page 8 of 15 Basic Res Cardiol (2013) 108:357

    123

  • treatment using a neutralizing antibody against MCP-1

    inhibited the inflammatory monocyte/macrophage infiltra-

    tion and cardiac fibroblast proliferation. This treatment

    attenuated cardiac fibrosis through the inhibition of TGF-binduction, and ameliorated the left ventricular diastolic

    dysfunction, but not the cardiomyocyte hypertrophy [57].

    These results suggest that MCP-1 mediates the accumula-

    tion of monocyte/macrophage lineage cells into the heart,

    and that this promotes myocardial fibrosis and diastolic

    dysfunction in the pressure overloaded heart through a

    TGF-b-mediated process.The reninangiotensinaldosterone system is associated

    with cardiac remodeling, and a competitive antagonist of

    aldosterone (spironolactone) or a mineral corticoid-specific

    inhibitor (eplerenone) has been indicated for reducing the

    risk of cardiac death in patients with heart failure and

    myocardial infarction. Myeloid cell-specific mineralocor-

    ticoid receptor knockout mice showed a lack of classical

    activation of infiltrated macrophages into the heart after

    angiotensin II infusion, and the infiltrated macrophages

    exhibited alternative activation profiles [101]. These mice

    exhibited reduced cardiac hypertrophy and fibrosis, sug-

    gesting that the mineralocorticoid receptor in cardiac

    macrophages is critical for cardiac macrophage polarity

    (pro-classical activation), cardiac hypertrophy and cardiac

    fibrosis [101].

    On the other hand, monocyte/macrophage depletion by

    clodronate liposome administration, which can damage or

    deplete monocytes and macrophages via their phagocytic

    activity, resulted in exacerbated left ventricular ejection

    function in a hypertensive heart disease model [115].

    Monocyte/macrophage lineage cell depletion in hyperten-

    sive hearts led to the abundant infiltration of inflammatory

    cells within areas of cardiomyocyte loss, predominantly

    CD4? T lymphocytes [115]. These results imply that

    monocytes/macrophages have protective effects on adap-

    tive stress through inhibiting T cell infiltration into the

    heart. Monocytes/macrophages have diverse and bivalent

    TGF-TGF- type I receptor

    cardiomyocyte cardiac fibroblast

    Smad dependentpathway

    non Smad pathway

    hypertrophy contraction

    extracellularmatrix

    production

    myofibroblast activation

    dominant

    Smad3, Smad4

    suppressor

    no effect

    TAK1Ras-MEKRho GTPasePI3K

    P38MAPK

    CTGF

    BMP7

    TGF- type I receptor

    TGF-

    fibrosis

    suppress

    Fig. 3 TGF-b signaling in cardiomyocytes and cardiac fibroblasts.TGF-b signaling is associated with hypertrophy of cardiomyocyte andcardiac fibrosis revealed by recent works are shown. In cardiomyo-

    cytes, TGF-b induces hypertrophy through TGF-b type I receptor.TGF-b type I receptor transduces its signals after cardiac stressesmainly through non-Smad pathways. For example, TGF-beta acti-

    vated kinase 1 (TAK1) induces hypertrophy of cardiomyocytes and

    reduces cardiac contractility via P38MAPK. In addition, TAK1

    activation inhibits bone morphogenetic protein 7 (BMP7) production

    that can inhibit myofibroblast activation. Smad 4 might inhibit cardiac

    hypertrophy under TGF-b signaling. In cardiac fibroblasts, TGF-bsignaling provokes myofibroblast activation through TGF-b type Ireceptor and promotes cardiac fibrosis. TAK1 activation in cardio-

    myocytes also produced connective tissue growth factor (CTGF).

    CTGF reduces contractility of cardiomyocytes and inhibits myofi-

    broblast activation. TGF-b transforming growth factor beta, TAK1TGF-beta activated kinase 1, MEK MAPK/ERK kinase, Rho GTPase

    Rho guanosine triphosphate, PI3K phosphoinositide-3-kinase, BMP7

    bone morphogenetic protein 7, CTGF connective tissue growth factor

    Basic Res Cardiol (2013) 108:357 Page 9 of 15

    123

  • effects on tissue remodeling after heart stress. More sub-

    type analyses of monocytes/macrophages should be per-

    formed in the future to better understand their roles. The

    balances of pro-inflammatory monocytes/macrophages

    versus alternatively activated monocytes/macrophages in

    the heart at every time point after heart stress might have

    complex effects on cardiomyocyte and cardiac fibroblast

    phenotypic modulation and activation.

    Mast cells are also key effector cells during allergic

    reactions, and show immune responses resulting from the

    degranulation of synthesized bioactive agents, like

    growth factors, cytokines and other molecules. Mast cells

    are also increased during cardiac hypertrophy and heart

    failure in humans [90]. These cells secrete platelet-

    derived growth factor A, TGF-b, TNF-a and histamine,and affect the cardiac function. Mast cell stabilizing

    drugs ameliorated heart failure via reduced cardiac

    remodeling in a volume overload model [10]. In addi-

    tion, after pressure overload in a mouse model, the

    infiltrated mast cells induced platelet-derived growth

    factor A chain synthesis and promoted the proliferation

    and collagen synthesis of cardiac fibroblasts. This mast

    cellcardiac fibroblast interaction is required for atrial

    fibrosis and affects the susceptibility to atrial fibrillation,

    which is the most common type of arrhythmia occurring

    in heart failure [61]. These results suggest that cardiac

    mast cells have a key role in the regulation of atrial

    myocardial remodeling and in the communication

    between cardiomyocytes and cardiac fibroblasts. In

    addition to platelet-derived growth factor A chain, it has

    recently been reported that angiotensin II-induced Rac 1

    activation leads to atrial remodeling and atrial fibrillation

    via the CTGF and lysyl oxidase-mediated miR-21

    expression [2].

    To date, several T cell subsets have been reported.

    CD4? T cells are divided into four subtypes, including

    helper T cells (Th1, Th2), Th17 cells and regulatory T

    cells (Tregs). T cells are also suggested to contribute to

    cardiac remodeling via cellcell interactions, as well as

    the production of cytokines and growth factors. In gen-

    eral, Th1 cells secrete Th1 cytokines like interferon-c,and promote an anti-fibrogenic response, while Th2

    cytokines, like IL-4,-5 and -13, promote fibrosis [67]. In

    the early stage of inflammation, Th1 cytokines are

    secreted to eradicate intracellular pathogens. In the late

    phase of inflammation, Th2 cytokines enhance chronic

    inflammation, leading to the elimination of extracellular

    organisms and contributing to the development of

    chronic inflammatory diseases. The administration of

    N(G)-nitro-L-arginine methyl ester (L-NAME: NO syn-

    thesis inhibitor) to mice can induce hypertension. Dif-

    ferent strains of mice, including C57BL/6 SCID mice,

    which lack T and B lymphocytes, C57BL/6 wild-type

    mice and BALB/c mice subjected to L-NAME-induced

    hypertension showed different Th1/Th2 polarity, i.e. null,

    Th1 polarity and Th2 polarity, respectively, whereas all

    of the strains showed the same degree of hypertension.

    The hypertension-induced cardiac fibrosis is decreased in

    SCID mice, unchanged in C57BL/6 wild-type mice and

    significantly increased in BALB/c mice [112]. Another

    hypertension model (an angiotensin II continuous infu-

    sion model) showed severe left ventricular dilatation,

    thinning and fibrosis in BALB/c mice, but not in C57BL/

    6 mice, whereas the same degree of hypertrophied

    cardiomyocytes was observed in both BALB/c and

    C57BL/6 mice [78]. These results suggest that Th1 cells

    and cytokines might initiate fibroblastic activity, and that

    Th2 cells and cytokines might contribute to the late or

    chronic stage of fibrosis.

    IL-17- and IL-22-producing CD4? cells, called Th17

    cells, play an important role in promoting inflammation

    during tissue remodeling. IL-17 can induce or functionally

    augment inflammatory cytokines, including IL-6, IL-1band TNFa, and can promote extracellular matrix remod-eling by producing matrix metalloproteinases or inhibiting

    repair components, like proteoglycans [3]. IL-17 is a

    pleiotropic cytokine that can modulate other cytokine

    functions in multiple ways. Among them, the post-tran-

    scriptional modification of IL-17 can stabilize the mRNA

    of many cytokines through AU-rich elements in the 30UTR,a common feature of inflammatory cytokine genes [80]. IL-

    17 blockade using an anti-IL-17 antibody resulted in

    decreased cardiac fibrosis in an isoproterenol-infusion rat

    heart failure model [26]. In this model, the MMP-1 and

    receptor activator of nuclear factor-rB ligand (RANKL)expression and collagen synthesis were inhibited, and the

    levels of tissue inhibitor of metalloproteinases and osteo-

    protegerin (OPG) were increased in cardiac fibroblasts

    [26]. These results indicate that Th17 cells control cardiac

    fibrosis by the IL-17-RANKL/OPG system and MMP

    production in cardiac fibroblasts, or via the stabilization of

    pro-inflammatory cytokines mRNA in various cardiac

    cells and immune cells.

    Tregs are an anti-inflammatory lineage of CD4? T

    lymphocytes that express FoxP3 and produce anti-

    inflammatory cytokines/growth factors, such as IL-10

    and/or TGF-b. In a heart disease model, CD4?CD25?

    Tregs were increased by angiotensin II infusion and

    myocardial infarction, and adoptive transfer of Tregs led

    to a reduction of cardiac hypertrophy, anti-inflammatory

    and anti-fibrotic effects and resulted in a preserved car-

    diac function via IL-10 production and a direct cellcell

    interaction [58, 95]. These results suggested that there is

    a close relationship among Tregs, cardiomyocytes and

    cardiac fibroblasts, and confirms their importance in

    cardiac remodeling.

    Page 10 of 15 Basic Res Cardiol (2013) 108:357

    123

  • Natural killer T cells (NKT cells)

    NKT cells have been shown to be involved in inflammation

    and tissue remodeling. The infiltration of NKT cells was

    increased in the non-infarct area of the left ventricle after

    myocardial infarction in mice [89]. Moreover, the admin-

    istration of a NKT cell activator, a-galactosylceramide(aGC) in a mouse myocardial infarction model, led toenhanced infiltration of NKT cells in the non-infarcted

    area. The left ventricular dilatation and mortality due to

    heart failure were significantly attenuated in the aGCadministration group [89]. It was suggested that these

    effects are dependent on NKT cells, because NKT deficient

    mice did not show these effects, and because IL-10 is the

    most potent effector cytokine for this process [89]. NKT

    cells play a protective role after myocardial infarction and

    heart failure via IL-10 production.

    Dendritic cells

    Dendritic cells infiltrate in the infarct heart. Depletion of

    bone marrow-derived dendritic cell using CD11c-diphteria

    toxin receptor transgenic mice showed deteriorated left

    ventricular function and remodeling after myocardial

    infarction model. The dendritic cells ablation group

    exhibited long-lasting inflammatory cytokines like IL-1b,IL-18 and TNFa. In addition, anti-inflammatory cellsincluding Ly-6Clow monocytes and alternatively activated

    macrophages were significantly infiltrated in the hearts of

    dendritic cell depleted group [6]. These results suggest that

    cardiac dendritic cells have a potent immunoprotective

    function after myocardial infarction.

    Concluding remarks

    Recent heart failure research has been focused not only on

    cardiomyocytes, but also on non-myocytes, which have

    been recognized as quiescent and structural cells under

    normal conditions. Unveiling the pivotal functions of non-

    myocytes has demonstrated that they have dynamic and

    diverse functions during tissue remodeling as an adaptive

    response and during the development of heart failure. In

    this review, we summarized the recent reports of the cell

    cell interactions in the heart. However, there is room for

    further research into the cellcell interactions under both

    physiological and pathological conditions in the heart.

    Importantly, immune cells are mainly derived from non-

    heart organs, like the bone marrow, spleen, thymus and

    intestinal organs. Moreover, the recruitment of these

    immune cells is at least partly controlled by the brain via

    neurotransmitters or humoral proteins. In the next decade,

    research should focus on further elucidating the mecha-

    nisms of heart failure in terms of cellcell interactions and

    organorgan communication.

    Acknowledgments This study was supported by the FundingProgram for World-Leading Innovative R&D on Science and Tech-

    nology (FIRST Program) (to R.N.), Grants-in-Aid for Scientific

    Research (S) and (B), and Grants-in-Aid for Young Scientists

    (B) from JSPS (23390203, 22229006, 23790835) (to R.N., K.F.); a

    grant for Translational Systems Biology and Medicine Initiative (to

    R.N.) from JST.

    Conflict of interest On behalf of all authors, the correspondingauthor states that there is no conflict of interest.

    References

    1. Accornero F, van Berlo JH, Benard MJ, Lorenz JN, Carmeliet P,

    Molkentin JD (2011) Placental growth factor regulates cardiac

    adaptation and hypertrophy through a paracrine mechanism.

    Circ Res 109:272280. doi:10.1161/CIRCRESAHA.111.240820

    2. Adam O, Lohfelm B, Thum T, Gupta SK, Puhl SL, Schafers HJ,

    Bohm M, Laufs U (2012) Role of miR-21 in the pathogenesis of

    atrial fibrosis. Basic Res Cardiol 107:278. doi:10.1007/s00395-

    012-0278-0

    3. Afzali B, Lombardi G, Lechler RI, Lord GM (2007) The role of

    T helper 17 (Th17) and regulatory T cells (Treg) in human organ

    transplantation and autoimmune disease. Clin Exp Immunol

    148:3246. doi:10.1111/j.1365-2249.2007.03356.x

    4. Agah R, Frenkel PA, French BA, Michael LH, Overbeek PA,

    Schneider MD (1997) Gene recombination in postmitotic cells.

    Targeted expression of Cre recombinase provokes cardiac-

    restricted, site-specific rearrangement in adult ventricular mus-

    cle in vivo. J Clin Invest 100:169179. doi:10.1172/jci119509

    5. Aggarwal BB (2003) Signalling pathways of the TNF super-

    family: a double-edged sword. Nat Rev Immunol 3:745756.

    doi:10.1038/nri1184

    6. Anzai A, Anzai T, Nagai S, Maekawa Y, Naito K, Kaneko H,

    Sugano Y, Takahashi T, Abe H, Mochizuki S, Sano M, Yos-

    hikawa T, Okada Y, Koyasu S, Ogawa S, Fukuda K (2012)

    Regulatory role of dendritic cells in postinfarction healing and

    left ventricular remodeling. Circulation 125:12341245.

    doi:10.1161/circulationaha.111.052126

    7. Avalos AM, Apablaza FA, Quiroz M, Toledo V, Pena JP, Mi-

    chea L, Irarrazabal CE, Carrion FA, Figueroa FE (2012) IL-17A

    levels increase in the infarcted region of the left ventricle in a rat

    model of myocardial infarction. Biol Res 45:193200.

    doi:10.1590/S0716-97602012000200012

    8. Banerjee I, Fuseler JW, Price RL, Borg TK, Baudino TA (2007)

    Determination of cell types and numbers during cardiac devel-

    opment in the neonatal and adult rat and mouse. Am J Physiol

    Heart Circ Physiol 293:H1883H1891. doi:10.1152/ajpheart.

    00514.2007

    9. Bowers SL, Borg TK, Baudino TA (2010) The dynamics of

    fibroblast-myocyte-capillary interactions in the heart. Ann NY

    Acad Sci 1188:143152. doi:10.1111/j.1749-6632.2009.05094.x

    10. Brower GL, Janicki JS (2005) Pharmacologic inhibition of mast

    cell degranulation prevents left ventricular remodeling induced

    by chronic volume overload in rats. J Card Fail 11:548556.

    doi:10.1016/j.cardfail.2005.05.005

    11. Buckley CD, Pilling D, Lord JM, Akbar AN, Scheel-Toellner D,

    Salmon M (2001) Fibroblasts regulate the switch from acute

    Basic Res Cardiol (2013) 108:357 Page 11 of 15

    123

  • resolving to chronic persistent inflammation. Trends Immunol

    22:199204. doi:10.1016/S1471-4906(01)01863-4

    12. Bujak M, Dobaczewski M, Chatila K, Mendoza LH, Li N,

    Reddy A, Frangogiannis NG (2008) Interleukin-1 receptor type I

    signaling critically regulates infarct healing and cardiac

    remodeling. Am J Pathol 173:5767. doi:10.2353/ajpath.

    2008.070974

    13. Cai ZP, Parajuli N, Zheng X, Becker L (2012) Remote ischemic

    preconditioning confers late protection against myocardial

    ischemiareperfusion injury in mice by upregulating interleukin-

    10. Basic Res Cardiol 107:277. doi:10.1007/s00395-012-0277-1

    14. Chandrasekar B, Mitchell DH, Colston JT, Freeman GL (1999)

    Regulation of CCAAT/enhancer binding protein, interleukin-6,

    interleukin-6 receptor, and gp130 expression during myocardial

    ischemia/reperfusion. Circulation 99:427433. doi:10.1161/

    01.CIR.99.3.427

    15. Colombo F, Gosselin H, El-Helou V, Calderone A (2003) Beta-

    adrenergic receptor-mediated DNA synthesis in neonatal rat

    cardiac fibroblasts proceeds via a phosphatidylinositol 3-kinase

    dependent pathway refractory to the antiproliferative action of

    cyclic AMP. J Cell Physiol 195:322330. doi:10.1002/jcp.10251

    16. Cortez DM, Feldman MD, Mummidi S, Valente AJ, Steffensen

    B, Vincenti M, Barnes JL, Chandrasekar B (2007) IL-17 stim-

    ulates MMP-1 expression in primary human cardiac fibroblasts

    via p38 MAPK- and ERK1/2-dependent C/EBP-beta, NF-kap-

    paB, and AP-1 activation. Am J Physiol Heart Circ Physiol

    293:H3356H3365. doi:10.1152/ajpheart.00928.2007

    17. DSouza SP, Yellon DM, Martin C, Schulz R, Heusch G, Onody

    A, Ferdinandy P, Baxter GF (2003) B-type natriuretic peptide

    limits infarct size in rat isolated hearts via KATP channel

    opening. Am J Physiol Heart Circ Physiol 284:H1592H1600.

    doi:10.1152/ajpheart.00902.2002

    18. Dorge H, Schulz R, Belosjorow S, Post H, van de Sand A,

    Konietzka I, Frede S, Hartung T, Vinten-Johansen J, Youker

    KA, Entman ML, Erbel R, Heusch G (2002) Coronary micro-

    embolization: the role of TNF-a in contractile dysfunction.J Mol Cell Cardiol 34:5162. doi:10.1006/jmcc.2001.1489

    19. Dean RG, Balding LC, Candido R, Burns WC, Cao Z, Twigg

    SM, Burrell LM (2005) Connective tissue growth factor and

    cardiac fibrosis after myocardial infarction. J Histochem Cyto-

    chem 53:12451256. doi:10.1369/jhc.4A6560.2005

    20. Del Re DP, Matsuda T, Zhai P, Gao S, Clark GJ, Van Der

    Weyden L, Sadoshima J (2010) Proapoptotic Rassf1A/Mst1

    signaling in cardiac fibroblasts is protective against pressure

    overload in mice. J Clin Invest 120:35553567. doi:10.1172/

    JCI43569

    21. Deten A, Holzl A, Leicht M, Barth W, Zimmer HG (2001)

    Changes in extracellular matrix and in transforming growth

    factor beta isoforms after coronary artery ligation in rats. J Mol

    Cell Cardiol 33:11911207. doi:10.1006/jmcc.2001.1383

    22. Deten A, Volz HC, Briest W, Zimmer HG (2002) Cardiac

    cytokine expression is upregulated in the acute phase after

    myocardial infarction. Experimental studies in rats. Cardiovasc

    Res 55:329340. doi:10.1016/S0008-6363(02)00413-3

    23. Dhingra S, Sharma AK, Arora RC, Slezak J, Singal PK (2009)

    IL-10 attenuates TNF-alpha-induced NF kappaB pathway acti-

    vation and cardiomyocyte apoptosis. Cardiovasc Res 82:5966.

    doi:10.1093/cvr/cvp040

    24. Divakaran V, Adrogue J, Ishiyama M, Entman ML, Haudek S,

    Sivasubramanian N, Mann DL (2009) Adaptive and maladaptive

    effects of SMAD3 signaling in the adult heart after hemody-

    namic pressure overloading. Circ Heart Fail 2:633642.

    doi:10.1161/CIRCHEARTFAILURE.108.823070

    25. Erlich JH, Boyle EM, Labriola J, Kovacich JC, Santucci RA,

    Fearns C, Morgan EN, Yun W, Luther T, Kojikawa O, Martin

    TR, Pohlman TH, Verrier ED, Mackman N (2000) Inhibition of

    the tissue factor-thrombin pathway limits infarct size after

    myocardial ischemiareperfusion injury by reducing inflamma-

    tion. Am J Pathol 157:18491862. doi:10.1016/S0002-9440

    (10)64824-9

    26. Feng W, Li W, Liu W, Wang F, Li Y, Yan W (2009) IL-17

    induces myocardial fibrosis and enhances RANKL/OPG and

    MMP/TIMP signaling in isoproterenol-induced heart failure.

    Exp Mol Pathol 87:212218. doi:10.1016/j.yexmp.2009.06.001

    27. Feng Y, Hans C, McIlwain E, Varner KJ, Lazartigues E (2012)

    Angiotensin-converting enzyme 2 over-expression in the central

    nervous system reduces angiotensin-II-mediated cardiac hyper-

    trophy. PLoS One 7:e48910. doi:10.1371/journal.pone.0048910

    28. Fischer P, Hilfiker-Kleiner D (2007) Survival pathways in

    hypertrophy and heart failure: the gp130STAT axis. Basic Res

    Cardiol 102:393411. doi:10.1007/s00395-007-0674-z

    29. Frangogiannis NG, Mendoza LH, Lindsey ML, Ballantyne CM,

    Michael LH, Smith CW, Entman ML (2000) IL-10 is induced in

    the reperfused myocardium and may modulate the reaction to

    injury. J Immunol 165:27982808

    30. Fredj S, Bescond J, Louault C, Delwail A, Lecron JC, Potreau D

    (2005) Role of interleukin-6 in cardiomyocyte/cardiac fibroblast

    interactions during myocyte hypertrophy and fibroblast prolif-

    eration. J Cell Physiol 204:428436. doi:10.1002/jcp.20307

    31. Fredj S, Bescond J, Louault C, Potreau D (2005) Interactions

    between cardiac cells enhance cardiomyocyte hypertrophy and

    increase fibroblast proliferation. J Cell Physiol 202:891899.

    doi:10.1002/Jcp.20197

    32. Friedrichs GS, Swillo RE, Jow B, Bridal T, Numann R, Warner

    LM, Killar LM, Sidek K (2002) Sphingosine modulates myocyte

    electrophysiology, induces negative inotropy, and decreases

    survival after myocardial ischemia. J Cardiovasc Pharmacol

    39:1828. doi:10.1097/00005344-200201000-00003

    33. Glenn DJ, Rahmutula D, Nishimoto M, Liang F, Gardner DG

    (2009) Atrial natriuretic peptide suppresses endothelin gene

    expression and proliferation in cardiac fibroblasts through a

    GATA4-dependent mechanism. Cardiovasc Res 84:209217.

    doi:10.1093/cvr/cvp208

    34. Golias C, Tsoutsi E, Matziridis A, Makridis P, Batistatou A,

    Charalabopoulos K (2007) Review. Leukocyte and endothelial

    cell adhesion molecules in inflammation focusing on inflam-

    matory heart disease. In Vivo 21:757769

    35. Gray MO, Long CS, Kalinyak JE, Li HT, Karliner JS (1998)

    Angiotensin II stimulates cardiac myocyte hypertrophy via

    paracrine release of TGF-beta 1 and endothelin-1 from fibro-

    blasts. Cardiovasc Res 40:352363. doi:10.1161/01.CIR.101.

    20.2338

    36. Gruhle S, Sauter M, Szalay G, Ettischer N, Kandolf R, Klingel

    K (2012) The prostacyclin agonist iloprost aggravates fibrosis

    and enhances viral replication in enteroviral myocarditis by

    modulation of ERK signaling and increase of iNOS expression.

    Basic Res Cardiol 107:287. doi:10.1007/s00395-012-0287-z

    37. Gulick T, Chung MK, Pieper SJ, Lange LG, Schreiner GF

    (1989) Interleukin 1 and tumor necrosis factor inhibit cardiac

    myocyte beta-adrenergic responsiveness. Proc Natl Acad Sci

    USA 86:67536757. doi:10.1073/pnas.86.17.6753

    38. Hao J, Wang B, Jones SC, Jassal DS, Dixon IMC (2000)

    Interaction between angiotensin II and Smad proteins in fibro-

    blasts in failing heart and in vitro. Am J Physiol Heart Circ

    Physiol 279:H3020H3030

    39. Heine HL, Leong HS, Rossi FM, McManus BM, Podor TJ

    (2005) Strategies of conditional gene expression in myocardium:

    an overview. Methods Mol Med 112:109154. doi:10.1007/978-

    1-59259-879-3_8

    40. Hilfiker-Kleiner D, Kaminski K, Podewski E, Bonda T, Schaefer

    A, Sliwa K, Forster O, Quint A, Landmesser U, Doerries C,

    Luchtefeld M, Poli V, Schneider MD, Balligand JL, Desjardins

    Page 12 of 15 Basic Res Cardiol (2013) 108:357

    123

  • F, Ansari A, Struman I, Nguyen NQ, Zschemisch NH, Klein G,

    Heusch G, Schulz R, Hilfiker A, Drexler H (2007) A cathepsin

    D-cleaved 16 kDa form of prolactin mediates postpartum car-

    diomyopathy. Cell 128:589600. doi:10.1016/j.cell.2006.12.036

    41. Hu Y, Zhang H, Lu Y, Bai H, Xu Y, Zhu X, Zhou R, Ben J, Xu

    Y, Chen Q (2011) Class A scavenger receptor attenuates myo-

    cardial infarction-induced cardiomyocyte necrosis through sup-

    pressing M1 macrophage subset polarization. Basic Res Cardiol

    106:13111328. doi:10.1007/s00395-011-0204-x

    42. Ieda M, Tsuchihashi T, Ivey KN, Ross RS, Hong TT, Shaw RM,

    Srivastava D (2009) Cardiac fibroblasts regulate myocardial

    proliferation through beta1 integrin signaling. Dev Cell

    16:233244. doi:10.1016/j.devcel.2008.12.007

    43. Jacobs M, Staufenberger S, Gergs U, Meuter K, Brandstatter K,

    Hafner M, Ertl G, Schorb W (1999) Tumor necrosis factor-alpha

    at acute myocardial infarction in rats and effects on cardiac

    fibroblasts. J Mol Cell Cardiol 31:19491959. doi:10.1006/

    jmcc.1999.1007

    44. Jaffre F, Bonnin P, Callebert J, Debbabi H, Setola V, Doly S,

    Monassier L, Mettauer B, Blaxall BC, Launay JM, Maroteaux L

    (2009) Serotonin and angiotensin receptors in cardiac fibroblasts

    coregulate adrenergic-dependent cardiac hypertrophy. Circ Res

    104:113123. doi:10.1161/CIRCRESAHA.108.180976

    45. Joseph NM, Mosher JT, Buchstaller J, Snider P, McKeever PE,

    Lim M, Conway SJ, Parada LF, Zhu Y, Morrison SJ (2008) The

    loss of Nf1 transiently promotes self-renewal but not tumori-

    genesis by neural crest stem cells. Cancer Cell 13:129140.

    doi:10.1016/j.ccr.2008.01.003

    46. Kakkar R, Lee RT (2010) Intramyocardial fibroblast myocyte

    communication. Circ Res 106:4757. doi:10.1161/circresaha.

    109.207456

    47. Kaneko K, Kanda T, Yokoyama T, Nakazato Y, Iwasaki T,

    Kobayashi I, Nagai R (1997) Expression of interleukin-6 in the

    ventricles and coronary arteries of patients with myocardial

    infarction. Res Commun Mol Pathol Pharmacol 97:312

    48. Kao YH, Chen YC, Cheng CC, Lee TI, Chen YJ, Chen SA

    (2010) Tumor necrosis factor-alpha decreases sarcoplasmic

    reticulum Ca2?-ATPase expressions via the promoter methyla-

    tion in cardiomyocytes. Crit Care Med 38:217222.

    doi:10.1097/CCM.0b013e3181b4a854

    49. Kapadia SR, Oral H, Lee J, Nakano M, Taffet GE, Mann DL

    (1997) Hemodynamic regulation of tumor necrosis factor-alpha

    gene and protein expression in adult feline myocardium. Circ

    Res 81:187195. doi:10.1161/01.RES.81.2.187

    50. Kapoun AM, Liang F, OYoung G, Damm DL, Quon D, White

    RT, Munson K, Lam A, Schreiner GF, Protter AA (2004) B-type

    natriuretic peptide exerts broad functional opposition to trans-

    forming growth factor-beta in primary human cardiac fibro-

    blasts: fibrosis, myofibroblast conversion, proliferation, and

    inflammation. Circ Res 94:453461. doi:10.1161/01.RES.0000

    117070.86556.9F

    51. Kaur K, Sharma AK, Singal PK (2006) Significance of changes

    in TNF-alpha and IL-10 levels in the progression of heart failure

    subsequent to myocardial infarction. Am J Physiol Heart Circ

    Physiol 291:H106H113. doi:10.1152/ajpheart.01327.2005

    52. Kishimoto I, Rossi K, Garbers DL (2001) A genetic model

    provides evidence that the receptor for atrial natriuretic peptide

    (guanylyl cyclase-A) inhibits cardiac ventricular myocyte hyper-

    trophy. Proc Natl Acad Sci USA 98:27032706. doi:10.1073/

    pnas.051625598

    53. Kleinbongard P, Heusch G, Schulz R (2010) TNFalpha in ath-

    erosclerosis, myocardial ischemia/reperfusion and heart failure.

    Pharmacol Ther 127:295314. doi:10.1016/j.pharmthera.

    2010.05.002

    54. Koitabashi N, Danner T, Zaiman AL, Pinto YM, Rowell J,

    Mankowski J, Zhang D, Nakamura T, Takimoto E, Kass DA

    (2011) Pivotal role of cardiomyocyte TGF-beta signaling in the

    murine pathological response to sustained pressure overload.

    J Clin Invest 121:23012312. doi:10.1172/JCI44824

    55. Krown KA, Yasui K, Brooker MJ, Dubin AE, Nguyen C, Harris

    GL, McDonough PM, Glembotski CC, Palade PT, Sabbadini RA

    (1995) TNF alpha receptor expression in rat cardiac myocytes:

    TNF alpha inhibition of L-type Ca2? current and Ca2? transients.

    FEBS Lett 376:2430. doi:10.1016/0014-5793(95)01238-5

    56. Krown KA, Page MT, Nguyen C, Zechner D, Gutierrez V,

    Comstock KL, Glembotski CC, Quintana PJ, Sabbadini RA

    (1996) Tumor necrosis factor alpha-induced apoptosis in cardiac

    myocytes. Involvement of the sphingolipid signaling cascade in

    cardiac cell death. J Clin Invest 98:28542865. doi:10.1172/

    JCI119114

    57. Kuwahara F, Kai H, Tokuda K, Takeya M, Takeshita A, Egashira

    K, Imaizumi T (2004) Hypertensive myocardial fibrosis and

    diastolic dysfunction: another model of inflammation? Hyper-

    tension 43:739745. doi:10.1161/01.HYP.0000118584.33350.7d

    58. Kvakan H, Kleinewietfeld M, Qadri F, Park JK, Fischer R,

    Schwarz I, Rahn HP, Plehm R, Wellner M, Elitok S, Gratze P,

    Dechend R, Luft FC, Muller DN (2009) Regulatory T cells

    ameliorate angiotensin II-induced cardiac damage. Circulation

    119:29042912. doi:10.1161/CIRCULATIONAHA.108.832782

    59. LaFramboise WA, Scalise D, Stoodley P, Graner SR, Guthrie

    RD, Magovern JA, Becich MJ (2007) Cardiac fibroblasts

    influence cardiomyocyte phenotype in vitro. Am J Physiol Cell

    Physiol 292:C1799C1808. doi:10.1152/ajpcell.00166.2006

    60. Levine B, Kalman J, Mayer L, Fillit HM, Packer M (1990)

    Elevated circulating levels of tumor necrosis factor in severe

    chronic heart failure. N Engl J Med 323:236241. doi:10.1056/

    NEJM199007263230405

    61. Liao CH, Akazawa H, Tamagawa M, Ito K, Yasuda N, Kudo Y,

    Yamamoto R, Ozasa Y, Fujimoto M, Wang P, Nakauchi H,

    Nakaya H, Komuro I (2010) Cardiac mast cells cause atrial

    fibrillation through PDGF-A-mediated fibrosis in pressure-

    overloaded mouse hearts. J Clin Invest 120:242253.

    doi:10.1172/JCI39942

    62. Liao P, Georgakopoulos D, Kovacs A, Zheng M, Lerner D, Pu

    H, Saffitz J, Chien K, Xiao RP, Kass DA, Wang Y (2001) The

    in vivo role of p38 MAP kinases in cardiac remodeling and

    restrictive cardiomyopathy. Proc Natl Acad Sci USA

    98:1228312288. doi:10.1073/pnas.211086598

    63. Liao YH, Xia N, Zhou SF, Tang TT, Yan XX, Lv BJ, Nie SF,

    Wang J, Iwakura Y, Xiao H, Yuan J, Jevallee H, Wei F, Shi GP,

    Cheng X (2012) Interleukin-17A contributes to myocardial

    ischemia/reperfusion injury by regulating cardiomyocyte apop-

    tosis and neutrophil infiltration. J Am Coll Cardiol 59:420429.

    doi:10.1016/j.jacc.2011.10.863

    64. Long CS, Hartogensis WE, Simpson PC (1993) Beta-adrenergic

    stimulation of cardiac non-myocytes augments the growth-pro-

    moting activity of non-myocyte conditioned medium. J Mol Cell

    Cardiol 25:915925. doi:10.1006/jmcc.1993.1104

    65. Mann DL (2002) Inflammatory mediators and the failing heart:

    past, present, and the foreseeable future. Circ Res 91:988998.

    doi:10.1161/01.RES.0000043825.01705.1B

    66. Mann DL, McMurray JJ, Packer M, Swedberg K, Borer JS,

    Colucci WS, Djian J, Drexler H, Feldman A, Kober L, Krum H,

    Liu P, Nieminen M, Tavazzi L, van Veldhuisen DJ, Walden-

    strom A, Warren M, Westheim A, Zannad F, Fleming T (2004)

    Targeted anticytokine therapy in patients with chronic heart

    failure: results of the Randomized Etanercept Worldwide

    Evaluation (RENEWAL). Circulation 109:15941602.

    doi:10.1161/01.CIR.0000124490.27666.B2

    67. Marra F, Aleffi S, Galastri S, Provenzano A (2009) Mononuclear

    cells in liver fibrosis. Semin Immunopathol 31:345358.

    doi:10.1007/s00281-009-0169-0

    Basic Res Cardiol (2013) 108:357 Page 13 of 15

    123

  • 68. Matsui Y, Sadoshima J (2004) Rapid upregulation of CTGF in

    cardiac myocytes by hypertrophic stimuli: implication for car-

    diac fibrosis and hypertrophy. J Mol Cell Cardiol 37:477481.

    doi:10.1016/j.yjmcc.2004.05.012

    69. McMullen JR (2008) Role of insulin-like growth factor 1 and

    phosphoinositide 3-kinase in a setting of heart disease. Clin Exp

    Pharmacol Physiol 35:349354. doi:10.1111/j.1440-

    1681.2007.04873.x

    70. McTiernan CF, Lemster BH, Frye C, Brooks S, Combes A,

    Feldman AM (1997) Interleukin-1 beta inhibits phospholamban

    gene expression in cultured cardiomyocytes. Circ Res

    81:493503. doi:10.1161/01.RES.81.4.493

    71. Miller C, Cai Y, Oikawa M, Thomas T, Dostmann W, Zaccolo

    M, Fujiwara K, Yan C (2011) Cyclic nucleotide phosphodies-

    terase 1A: a key regulator of cardiac fibroblast activation and

    extracellular matrix remodeling in the heart. Basic Res Cardiol

    106:10231039. doi:10.1007/s00395-011-0228-2

    72. Mitchell MD, Laird RE, Brown RD, Long CS (2007) IL-1beta

    stimulates rat cardiac fibroblast migration via MAP kinase

    pathways. Am J Physiol Heart Circ Physiol 292:H1139H1147.

    doi:10.1152/ajpheart.00881.2005

    73. Murray DR, Prabhu SD, Chandrasekar B (2000) Chronic beta-

    adrenergic stimulation induces myocardial proinflammatory

    cytokine expression. Circulation 101:23382341. doi:10.1161/

    01.CIR.101.20.2338

    74. Novoyatleva T, Schymura Y, Janssen W, Strobl F, Swiercz JM,

    Patra C, Posern G, Wietelmann A, Zheng TS, Schermuly RT,

    Engel FB (2013) Deletion of Fn14 receptor protects from right

    heart fibrosis and dysfunction. Basic Res Cardiol 108:325.

    doi:10.1007/s00395-012-0325-x

    75. Parajuli N, Yuan Y, Zheng X, Bedja D, Cai Z (2012) Phos-

    phatase PTEN is critically involved in post-myocardial infarc-

    tion remodeling through the Akt/interleukin-10 signaling

    pathway. Basic Res Cardiol 107:248. doi:10.1007/s00395-012-

    0248-6

    76. Pawlinski R, Tencati M, Hampton CR, Shishido T, Bullard TA,

    Casey LM, Andrade-Gordon P, Kotzsch M, Spring D, Luther T,

    Abe J, Pohlman TH, Verrier ED, Blaxall BC, Mackman N

    (2007) Protease-activated receptor-1 contributes to cardiac

    remodeling and hypertrophy. Circulation 116:22982306.

    doi:10.1161/CIRCULATIONAHA.107.692764

    77. Pedrotty DM, Klinger RY, Kirkton RD, Bursac N (2009) Car-

    diac fibroblast paracrine factors alter impulse conduction and

    ion channel expression of neonatal rat cardiomyocytes. Car-

    diovasc Res 83:688697. doi:10.1093/Cvr/Cvp164

    78. Peng H, Yang X-P, Carretero OA, Nakagawa P, DAmbrosio M,

    Leung P, Xu J, Peterson EL, Gonzalez GE, Harding P, Rhaleb

    N-E (2011) Angiotensin II-induced dilated cardiomyopathy in

    Balb/c but not C57BL/6J mice. Exp Physiol 96:756764.

    doi:10.1113/expphysiol.2011.057612

    79. Podewski EK, Hilfiker-Kleiner D, Hilfiker A, Morawietz H,

    Lichtenberg A, Wollert KC, Drexler H (2003) Alterations in

    Janus kinase (JAK)-signal transducers and activators of tran-

    scription (STAT) signaling in patients with end-stage dilated

    cardiomyopathy. Circulation 107:798802. doi:10.1161/

    01.CIR.0000057545.82749.FF

    80. Qian Y, Liu C, Hartupee J, Altuntas CZ, Gulen MF, Jane-Wit D,

    Xiao J, Lu Y, Giltiay N, Liu J, Kordula T, Zhang QW, Vallance

    B, Swaidani S, Aronica M, Tuohy VK, Hamilton T, Li X (2007)

    The adaptor Act1 is required for interleukin 17-dependent sig-

    naling associated with autoimmune and inflammatory disease.

    Nat Immunol 8:247256. doi:10.1038/ni1439

    81. Rauchhaus M, Doehner W, Francis DP, Davos C, Kemp M,

    Liebenthal C, Niebauer J, Hooper J, Volk HD, Coats AJ, Anker

    SD (2000) Plasma cytokine parameters and mortality in patients

    with chronic heart failure. Circulation 102:30603067.

    doi:10.1161/01.CIR.102.25.3060

    82. Sano M, Fukuda K, Kodama H, Pan J, Saito M, Matsuzaki J,

    Takahashi T, Makino S, Kato T, Ogawa S (2000) Interleukin-6

    family of cytokines mediate angiotensin II-induced cardiac

    hypertrophy in rodent cardiomyocytes. J Biol Chem

    275:2971729723. doi:10.1074/jbc.M003128200

    83. Sano M, Minamino T, Toko H, Miyauchi H, Orimo M, Qin Y,

    Akazawa H, Tateno K, Kayama Y, Harada M, Shimizu I,

    Asahara T, Hamada H, Tomita S, Molkentin JD, Zou Y,

    Komuro I (2007) p53-Induced inhibition of Hif-1 causes cardiac

    dysfunction during pressure overload. Nature 446:444448.

    doi:10.1038/nature05602

    84. Schultz Jel J, Witt SA, Glascock BJ, Nieman ML, Reiser PJ, Nix

    SL, Kimball TR, Doetschman T (2002) TGF-beta1 mediates the

    hypertrophic cardiomyocyte growth induced by angiotensin II.

    J Clin Invest 109:787796. doi:10.1172/JCI14190

    85. Schulz R, Panas DL, Catena R, Moncada S, Olley PM, Lopas-

    chuk GD (1995) The role of nitric oxide in cardiac depression

    induced by interleukin-1 beta and tumour necrosis factor-alpha.

    Br J Pharmacol 114:2734. doi:10.1111/j.1476-

    5381.1995.tb14901.x

    86. Shindo T, Manabe I, Fukushima Y, Tobe K, Aizawa K, Mi-

    yamoto S, Kawai-Kowase K, Moriyama N, Imai Y, Kawakami

    H, Nishimatsu H, Ishikawa T, Suzuki T, Morita H, Maemura K,

    Sata M, Hirata Y, Komukai M, Kagechika H, Kadowaki T,

    Kurabayashi M, Nagai R (2002) Kruppel-like zinc-finger tran-

    scription factor KLF5/BTEB2 is a target for angiotensin II

    signaling and an essential regulator of cardiovascular remodel-

    ing. Nat Med 8:856863. doi:10.1038/nm738

    87. Siwik DA, Chang DL, Colucci WS (2000) Interleukin-1beta and

    tumor necrosis factor-alpha decrease collagen synthesis and

    increase matrix metalloproteinase activity in cardiac fibroblasts

    in vitro. Circ Res 86:12591265. doi:10.1161/01.RES.86.

    12.1259

    88. Skyschally A, Gres P, Hoffmann S, Haude M, Erbel R, Schulz

    R, Heusch G (2007) Bidirectional role of tumor necrosis factor-

    alpha in coronary microembolization: progressive contractile

    dysfunction versus delayed protection against infarction. Circ

    Res 100:140146. doi:10.1161/01.RES.0000255031.15793.86

    89. Sobirin MA, Kinugawa S, Takahashi M, Fukushima A, Homma

    T, Ono T, Hirabayashi K, Suga T, Azalia P, Takada S, Tanig-

    uchi M, Nakayama T, Ishimori N, Iwabuchi K, Tsutsui H (2012)

    Activation of natural killer T cells ameliorates postinfarct car-

    diac remodeling and failure in mice. Circ Res 111:10371047.

    doi:10.1161/CIRCRESAHA.112.270132

    90. Sperr WR, Bankl HC, Mundigler G, Klappacher G, Grosssch-

    midt K, Agis H, Simon P, Laufer P, Imhof M, Radaszkiewicz T,

    Glogar D, Lechner K, Valent P (1994) The human cardiac mast

    cell: localization, isolation, phenotype, and functional charac-

    terization. Blood 84:38763884

    91. Sun M, Dawood F, Wen WH, Chen M, Dixon I, Kirshenbaum LA,

    Liu PP (2004) Excessive tumor necrosis factor activation after

    infarction contributes to susceptibility of myocardial rupture and

    left ventricular dysfunction. Circulation 110:32213228.

    doi:10.1161/01.CIR.0000147233.10318.23

    92. Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-

    Retamozo V, Panizzi P, Figueiredo J-L, Kohler RH, Chudnov-

    skiy A, Waterman P, Aikawa E, Mempel TR, Libby P, Weiss-

    leder R, Pittet MJ (2009) Identification of splenic reservoir

    monocytes and their deployment to inflammatory sites. Science

    325:612616. doi:10.1126/science.1175202

    93. Swirski FK, Nahrendorf M (2013) Leukocyte behavior in ath-

    erosclerosis, myocardial infarction, and heart failure. Science

    339:161166. doi:10.1126/science.1230719

    Page 14 of 15 Basic Res Cardiol (2013) 108:357

    123

  • 94. Takeda N, Manabe I, Uchino Y, Eguchi K, Matsumoto S, Ni-

    shimura S, Shindo T, Sano M, Otsu K, Snider P, Conway SJ,

    Nagai R (2010) Cardiac fibroblasts are essential for the adaptive

    response of the murine heart to pressure overload. J Clin Invest

    120:254265. doi:10.1172/Jci40295

    95. Tang TT, Yuan J, Zhu ZF, Zhang WC, Xiao H, Xia N, Yan XX,

    Nie SF, Liu J, Zhou SF, Li JJ, Yao R, Liao MY, Tu X, Liao YH,

    Cheng X (2012) Regulatory T cells ameliorate cardiac remod-

    eling after myocardial infarction. Basic Res Cardiol 107:232.

    doi:10.1007/s00395-011-0232-6

    96. Thielmann M, Dorge H, Martin C, Belosjorow S, Schwanke U,

    van de Sand A, Konietzka I, Buchert A, Kruger A, Schulz R,

    Heusch G (2002) Myocardial dysfunction with coronary mi-

    croembolization: signal transduction through a sequence of

    nitric oxide, tumor necrosis factor-a, and sphingosine. Circ Res90:807813. doi:10.1161/01.res.0000014451.75415.36

    97. Torre-Amione G, Kapadia S, Benedict C, Oral H, Young JB,

    Mann DL (1996) Proinflammatory cytokine levels in patients

    with depressed left ventricular ejection fraction: a report from

    the Studies of Left Ventricular Dysfunction (SOLVD). J Am

    Coll Cardiol 27:12011206. doi:10.1016/0735-1097(95)00589-7

    98. Torre-Amione G, Kapadia S, Lee J, Durand JB, Bies RD, Young

    JB, Mann DL (1996) Tumor necrosis factor-alpha and tumor

    necrosis factor receptors in the failing human heart. Circulation

    93:704711. doi:10.1161/01.CIR.93.4.704

    99. Tsutsumi Y, Matsubara H, Ohkubo N, Mori Y, Nozawa Y,

    Murasawa S, Kijima K, Maruyama K, Masaki H, Moriguchi Y,

    Shibasaki Y, Kamihata H, Inada M, Iwasaka T (1998) Angio-

    tensin II type 2 receptor is upregulated in human heart with

    interstitial fibrosis, and cardiac fibroblasts are the major cell type

    for its expression. Circ Res 83:10351046. doi:10.1161/

    01.RES.83.10.1035

    100. Turner NA, Porter KE, Smith WH, White HL, Ball SG, Balm-

    forth AJ (2003) Chronic beta2-adrenergic receptor stimulation

    increases proliferation of human cardiac fibroblasts via an

    autocrine mechanism. Cardiovasc Res 57:784792. doi:10.1016/

    S0008-6363(02)00729-0

    101. Usher MG, Duan SZ, Ivaschenko CY, Frieler RA, Berger S,

    Schutz G, Lumeng CN, Mortensen RM (2010) Myeloid miner-

    alocorticoid receptor controls macrophage polarization and

    cardiovascular hypertrophy and remodeling in mice. J Clin

    Invest 120:33503364. doi:10.1172/JCI41080

    102. van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH,

    van Luyn MJ (2007) Macrophage depletion impairs wound

    healing and increases left ventricular remodeling after myocar-

    dial injury in mice. Am J Pathol 170:818829. doi:10.2353/

    ajpath.2007.060547

    103. Venkatachalam K, Mummidi S, Cortez DM, Prabhu SD, Valente

    AJ, Chandrasekar B (2008) Resveratrol inhibits high glucose-

    induced PI3K/Akt/ERK-dependent interleukin-17 expression in

    primary mouse cardiac fibroblasts. Am J Physiol Heart Circ

    Physiol 294:H2078H2087. doi:10.1152/ajpheart.01363.2007

    104. Volz HC, Laohachewin D, Seidel C, Lasitschka F, Keilbach K,

    Wienbrandt AR, Andrassy J, Bierhaus A, Kaya Z, Katus HA,

    Andrassy M (2012) S100A8/A9 aggravates post-ischemic heart

    failure through activation of RAGE-dependent NF-kappaB sig-

    naling. Basic Res Cardiol 107:250. doi:10.1007/s00395-012-

    0250-z

    105. Wang J, Xu N, Feng X, Hou N, Zhang J, Cheng X, Chen Y,

    Zhang Y, Yang X (2005) Targeted disruption of Smad4 in

    cardiomyocytes results in cardiac hypertrophy and heart failure.

    Circ Res 97:821828. doi:10.1161/01.RES.0000185833.42544.06

    106. Wei L (2011) Immunological aspect of cardiac remodeling: T

    lymphocyte subsets in inflammation-mediated cardiac fibrosis.

    Exp Mol Pathol 90:7478. doi:10.1016/j.yexmp.2010.10.004

    107. Westermann D, Lindner D, Kasner M, Zietsch C, Savvatis K,

    Escher F, von Schlippenbach J, Skurk C, Steendijk P, Riad A,

    Poller W, Schultheiss HP, Tschope C (2011) Cardiac inflam-

    mation contributes to changes in the extracellular matrix in

    patients with heart failure and normal ejection fraction. Circ

    Heart Fail 4:4452. doi:10.1161/CIRCHEARTFAILURE.

    109.931451

    108. Yamaoka M, Yamaguchi S, Okuyama M, Tomoike H (1999)

    Anti-inflammatory cytokine profile in human heart failure:

    behavior of interleukin-10 in association with tumor necrosis

    factor-alpha. Jpn Circ J 63:951956. doi:10.1253/jcj.63.951

    109. Yamazaki T, Komuro I, Yazaki Y (1998) Signalling pathways

    for cardiac hypertrophy. Cell Signal 10:693698. doi:10.1016/

    S0898-6568(98)00036-9

    110. Yokoyama T, Nakano M, Bednarczyk JL, McIntyre BW, Ent-

    man M, Mann DL (1997) Tumor necrosis factor-alpha provokes

    a hypertrophic growth response in adult cardiac myocytes.

    Circulation 95:12471252. doi:10.1161/01.CIR.95.5.1247

    111. Yoon PO, Lee MA, Cha H, Jeong MH, Kim J, Jang SP, Choi

    BY, Jeong D, Yang DK, Hajjar RJ, Park WJ (2010) The

    opposing effects of CCN2 and CCN5 on the development of

    cardiac hypertrophy and fibrosis. J Mol Cell Cardiol

    49:294303. doi:10.1016/j.yjmcc.2010.04.010

    112. Yu Q, Horak K, Larson DF (2006) Role of T lymphocytes in

    hypertension-induced cardiac extracellular matrix remodeling.

    Hypertension 48:98104. doi:10.1161/01.HYP.0000227247.

    27111.b2

    113. Zaglia T, Milan G, Franzoso M, Bertaggia E, Pianca N, Pia-

    sentini E, Voltarelli VA, Chiavegato D, Brum PC, Glass DJ,

    Schiaffino S, Sandri M, Mongillo M (2013) Cardiac sympathetic

    neurons provide trophic signal to the heart via beta2-adreno-

    ceptor-dependent regulation of proteolysis. Cardiovasc Res

    97:240250. doi:10.1093/cvr/cvs320

    114. Zamilpa R, Kanakia R, Cigarroa J 4th, Dai Q, Escobar GP,

    Martinez H, Jimenez F, Ahuja SS, Lindsey ML (2011) CC

    chemokine receptor 5 deletion impairs macrophage activation

    and induces adverse remodeling following myocardial infarc-

    tion. Am J Physiol Heart Circ Physiol 300:H1418H1426.

    doi:10.1152/ajpheart.01002.2010

    115. Zandbergen HR, Sharma UC, Gupta S, Verjans JW, van den

    Borne S,