Crosstalk between fibroblasts and inflammatory cells (original) (raw)
,
1
Berlin-Brandenburg Center for Regenerative Therapies
,
Charité, University Medicine Berlin, Campus Virchow Clinic
,
Berlin
,
Germany
Search for other works by this author on:
,
1
Berlin-Brandenburg Center for Regenerative Therapies
,
Charité, University Medicine Berlin, Campus Virchow Clinic
,
Berlin
,
Germany
Search for other works by this author on:
1
Berlin-Brandenburg Center for Regenerative Therapies
,
Charité, University Medicine Berlin, Campus Virchow Clinic
,
Berlin
,
Germany
2
Department of Cardiology and Pneumology
,
Charité, University Medicine Berlin, Campus Benjamin Franklin
,
Berlin
,
Germany
*Corresponding author. Department of Cardiology and Pneumology, Charité, University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany. Tel: +49 30 84454780; fax: +49 30 84454509, Email: carsten.tschoepe@charite.de
Search for other works by this author on:
This article is part of the Spotlight Issue on: Heterocellular signalling and crosstalk in the heart in ischaemia and heart failure.
Received:
03 January 2014
Revision received:
27 February 2014
Accepted:
27 February 2014
Cite
Sophie Van Linthout, Kapka Miteva, Carsten Tschöpe, Crosstalk between fibroblasts and inflammatory cells, Cardiovascular Research, Volume 102, Issue 2, 1 May 2014, Pages 258–269, https://doi.org/10.1093/cvr/cvu062
Close
Navbar Search Filter Mobile Enter search term Search
Abstract
Fibroblasts, which are traditionally recognized as a quiescent cell responsible for extracellular matrix production, are more and more appreciated as an active key player of the immune system. This review describes how fibroblasts and immune cells reciprocally influence the pathogenesis of fibrosis. An overview is given how fibroblasts are triggered by components of the innate and adaptive immunity on the one hand and how fibroblasts modulate immune cell behaviour via conditioning the cellular and cytokine microenvironment on the other hand. Finally, latest insights into the role of cardiac fibroblasts in the orchestration of inflammatory cell infiltration in the heart, and their impact on heart failure, are outlined.
1. Introduction
Fibrosis is a scarring process, which is characterized by excess deposition of collageneous and non-collagenous extracellular matrix (ECM) due to the accumulation, proliferation, and activation of fibroblasts and myofibroblasts. Consequently, fibrosis leads to dysregulated organ architecture and function.1 Inflammatory and immunological reactions underlie the fibrosis process, by which both components of the innate and adaptive immune system are involved (Figure 1),2 as well as the renin–angiotensin–aldosterone system and metabolic derangements.
Figure 1
Impact of components of the innate and adaptive immunity on the activation of fibroblasts. Cytokines, growth factors, and enzymes released by immune cells directly (black arrows) promote fibroblast activation and indirectly (red arrows) lead to myofibroblast activation via further induction of pro-inflammatory, pro-fibrotic factors in other immune cells. IFN: interferon; IL: interleukin; iNOS: inducible nitric oxide synthase; MBP: major basic protein; MMP: matrix metalloproteinase; PDGF: platelet-derived growth factor; ROS: reactive oxygen species; TGF: transforming growth factor; TIMP: tissue inhibitor of matrix metalloproteinase; TNF: tumour necrosis factor.
Resolution of the inflammatory response requires the elimination of the major part of immune cells, which were recruited and actively proliferated during the acute phase of inflammation. Though, the presence of aberrant activated fibroblasts, myofibroblasts, lead to chronic inflammation via induction of a dysregulated homeostatic balance between leucocyte recruitment, proliferation, emigration, and death.3 This review gives an overview of the multiple interactions of the components of the innate and adaptive immunity on the fibroblast leading to fibroblast activation, and how fibroblasts upon activation can lead to chronic inflammation. Finally, the significance of cardiac inflammation and fibrosis and their mutual interaction in the development of heart failure is briefly discussed.
2. Fibroblasts
2.1 Definition, function, and diversity
Fibroblasts are a heterogeneous population of stromal cells characterized by a spindle-shaped morphology and flat, oval nuclei, the lack of epithelial, vascular, and leucocyte lineage markers, as well as the ability to adhere to plastic. So far, no universal fibroblast marker has been identified. Fibroblasts are traditionally recognized for their structural role in synthesizing and remodelling the ECM in tissues. Though, beyond their role in structural support, fibroblasts are able to secrete and respond to cytokines, chemokines, and growth factors. They maintain the homeostasis of adjacent cells and orchestrate the maintenance of inflammatory infiltrates, indicating their importance in tissue development, differentiation, remodelling, and repair. In the heart, where fibroblasts account for 60–70% of the cells,4,5 compared with cardiomyocytes which only constitute 30–40%, the crosstalk between cardiac fibroblasts and cardiomyocytes is important for both cardiac development and remodelling in response to tissue injury. Cardiac fibroblasts provide contractile co-ordination and electrical coupling between cardiomyocytes, allow for mechanical force distribution throughout the myocardium, and contribute to angiogenesis, all of which are extensively reviewed elsewhere.6,7
Fibroblasts differ from the anatomical site,8,9 the disease status, and even within the same tissue. Consistent with the varying biophysical requirements of different tissues, fibroblasts from distinct tissues differ in proliferation, collagen and matrix metalloproteinase (MMP) production,9 contractility, and immunomodulatory function.8 Importantly, these differences in characteristic phenotypes among fibroblasts from distinct tissues are maintained after extended in vitro culture, supporting the concept that fibroblasts possess positional identity. This concept is corroborated by comparative transcriptome analysis, which revealed that the transcriptional profiles of fibroblasts can be clustered into groups according to the anatomical site.10,11
The diversity of fibroblasts (within the same tissue) can be explained by their distinct cellular origins. Fibroblasts mainly originate from primary mesenchymal cells, but can also arise through epithelial–mesenchymal transition (as seen in the liver12 and kidney13), or endothelial–mesenchymal transition (as seen in the lung,14 heart,15 and cancer16) and can be derived from circulating cells, including mesenchymal stromal cells (MSCs) and fibrocytes. Fibrocytes are defined as circulating monocyte-derived cells that are capable of expressing a fibroblastic phenotype.17 Although they only comprise a small percentage of circulating leucocytes under non-pathological conditions in humans, their number increases by chronic inflammatory and fibrotic derangements including autoimmune18 and cardiovascular disorders,19,20 and contribute to the fibrotic process.
3. Innate and adaptive immune mechanisms rule the development of fibrosis
Substantial evidence postulates that fibrosis develops as a complication of inflammatory processes, in which both innate and adaptive immune mechanisms play a considerable role. Tissue damage, infections with bacteria, viruses, fungi, and parasites, foreign body implants, autoimmune disease, or tumours could progress to an adverse chronic inflammation, which subsequently leads to fibrotic disease. Moreover, even a low-grade, but persistent inflammation promotes fibrosis in cardiovascular diseases and hypertension.21 Therefore, the elimination of an inflammatory trigger and the resolution of inflammation are of critical importance for the prevention of fibroblasts activation, excessive accumulation of ECM, and tissue fibrosis.
3.1 Innate immunity triggers of fibrosis
The first-line defence of the innate immune system largely depends on a sophisticated array of pattern recognition receptors, which recognize conserved pathogen-associated molecular patterns.22 Interestingly, fibroblasts have been shown to express a variety of pattern recognition receptors, including Toll-like receptors (TLRs), and the subsequent ligand activation of those receptors can directly activate fibroblasts and promote their differentiation into collagen-producing myofibroblasts.23,24
3.1.1 Platelets
Also platelets express a range of TLRs,25,26 contributing to their immune cell function in the state of infection and inflammation, besides their substantial role in the coagulation response via fibrin clot formation. Furthermore, a substantial body of evidence has indicated that platelets promote systemic and cardiac inflammatory responses, and ventricular remodelling.27 Activated platelets release several growth factors promoting healing including platelet-derived growth factor, a potent chemotactic agent, and transforming growth factor (TGF)-β, which stimulates the deposition of ECM.28 Moreover, dysregulation in the coagulation signalling cascade may contribute to tissue fibrosis.29 The pro-fibrotic effects of coagulation factor X have been shown in a model of acute lung injury,30 while overexpression of human blood coagulation factor IX is associated with myocardial fibrosis.31 However, deficiency of coagulation factor VII could result in spontaneous cardiac fibrosis32 and coagulation insufficiency is typical for cirrhosis patients.33,34 Therefore, a balanced coagulation response seems to be of critical importance for the prevention of tissue fibrosis.
3.1.2 Neutrophils
Upon tissue injury, damaged epithelial and endothelial cells not only release inflammatory factors triggering the coagulation cascade, but also a cocktail of growth factors and chemokines, which subsequently initiate an influx of neutrophils and monocytes to the site of the damaged tissue. Neutrophils are the first cells attracted to the injured site followed by monocytes, and finally lymphocytes and mast cells.35 Neutrophils act as a first-line defence and initiate an acute inflammatory response to engulf dead cells and tissue debris in order to facilitate tissue repair. However, excessive and persistent neutrophil infiltration or their delayed elimination exacerbates the tissue injury via the release of inflammatory mediators and proteinases.36 Neutrophils count has been utilized as a prognostic biomarker of chronic remodelling of the left ventricle (LV).37
Neutrophils release large amounts of reactive oxygen species during the respiratory burst38 via the multicomponent enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Reactive oxygen species-releasing neutrophils are a proven pro-fibrotic mediator in systemic sclerosis,39 pulmonary fibrosis,40 and hepatic fibrosis.41 Inhibition of NADPH oxidase attenuates cardiac fibrosis post-myocardial infarction.42,43 Besides numerous pro-inflammatory cytokines, neutrophils secrete granules containing enzymes such as MMPs, elastase, and cathepsins capable of cleaving collagenous and non-collagenous connective tissue components, involved in tissue remodelling during the fibrotic process.44 In addition, neutrophils-derived elastase has been shown to play a pivotal role in the pathogenesis of pulmonary fibrosis.45 During the acute phase of inflammation, neutrophils play an indirect pro-fibrotic role by activating further cellular components of the innate immune system such as macrophages promoting fibrogenesis.46
3.1.3 Macrophages
Macrophages are indispensable effector cells involved in tissue remodelling and fibrosis. They are the main source of several types of MMPs (MMP-1, -7, -8, -9, and -12) as well as their endogenous suppressors, tissue inhibitors of MMPs (TIMPs).47 The proper balance of MMPs and TIMPs is crucial for the normal deposition and degradation of the ECM. The MMPs released by macrophages mediate not only signalling through proteolysis of both ECM and non-matrix substrates, they also amplify the inflammatory response and influence the progression of tissue remodelling. MMP-9 is the most relevant pro-fibrotic MMP,48 which inhibition or deletion has been shown to reduce fibrosis in models of dilated cardiomyopathy49 and myocardial infarction.50 MMP-2 is not only responsible for the degradation of matrix proteins. It also cleaves monocyte chemotactic protein (MCP)-3, lowering its chemotactic activity and diminishing the invasion of inflammatory cells and subsequently the inflammatory response and fibrosis in viral-induced myocarditis.51
In addition to MMPs, macrophages are a leading producer of TGF-β, considered the most significant pro-fibrotic agent involved in the progression of chronic fibrotic diseases affecting skin, liver, lung, kidney, and heart.52 Inhibition of alternatively activated macrophages (M2 macrophages) has been shown to abrogate TGF-β-driven lung fibrosis.53 TGF-β has several isoforms (TGF-β1, TGF-β2, and TGF-β3), which are synthesized as latent precursors bound to latent TGF-β-binding proteins (LTBP-1, -3, and -4). LTBPs are removed extracellularly via proteolytic cleavage releasing active TGF-β.54 Activated TGF-β binds to TGF-β receptors and the signalling within the cell is mediated via the SMAD family of transcriptional activators.55 TGF-β induces the expression of ECM genes and suppresses the activity of genes encoding MMPs, which are capable of degrading ECM.56,57 TGF-β induces the expression of pro-fibrotic genes such as Type I collagen and connective tissue growth factor (CTGF) in a SMAD3-dependent manner.58,59 Moreover, TGF-β can promote collagen synthesis in SMAD-independent pathways via mitogen-activated protein kinase cascades, including p38 MAPK, Jun N-terminal kinase, and extracellular signal-regulated kinase.60 TGF-β1 and -β3 as well as LTBPs are up-regulated in patients with cardiac fibrosis, highlighting the implication of active TGF-β in human cardiac fibrogenesis.61 Heart failure patients with preserved ejection fraction exhibit high TGF-β1 expression accompanied by reduced MMP-1 and elevated TIMP-1 favouring the collagen synthesis in the heart of those patients.62 Furthermore, numerous studies have demonstrated the direct effect of TGF-β on fibroblast differentiation to myofibroblasts.63,64 Inhibition of TGF-β via neutralizing antibodies has been shown to be well tolerated and to successfully reduce the development of fibrosis in different experimental models.65–67 For example, TGF-β neutralization prevented cardiac fibrosis and improved diastolic dysfunction in pressure-overloaded rats.68 In contrast, TGF-β-deficient mice exhibit profoundly diminished collagen deposition, though they suffer from a severe wasting syndrome in addition to extensive inflammatory, tissue necrosis, resulting in organ failure, and death.69,70 Early inhibition of TGF-β is associated with increased mortality, leucocyte infiltration, and chemokine expression,71 while the later elimination of TGF-β results in improved survival and reduced tissue fibrosis.72 In addition, TGF-β1-producing regulatory T cells (Tregs) have been demonstrated to reduce bleomycin-induced fibrosis in an interleukin (IL)-10-dependent manner.73 These findings indicate that TGF-β-mediated effects in fibrosis are complex, and timing of action as well as cell environment are of critical importance. In fact, a miscalculated inhibition has led to adverse side effects in human.55,74 Therefore, the development of anti-fibrotic therapies targeting TGF-β and its signalling should be carefully considered and properly evaluated in suitable animal models.
The complex role of macrophages in fibrosis is evident from a study, demonstrating that deletion of the macrophage population either during injury or during repair and resolution has dramatically different effects on the overall fibrotic outcome. During a progressive inflammatory injury, macrophage depletion results in amelioration of fibrosis, while depletion during recovery results in a failure of resolution with persistence of pro-fibrotic cellular and matrix components.75 Furthermore, macrophage depletion in the early phase post-myocardial injury markedly impairs the wound healing and increases remodelling and mortality, indicating that macrophages are a key player in myocardial wound healing.76
Macrophages stimulated by TLR ligands and interferon (IFN)-γ undergo classical M1 activation, while those stimulated by IL-4 and -13 become M2 macrophages.77,78 M2 macrophages are involved in wound healing, tissue remodelling, fibrosis, and inflammatory responses.79,80 They contribute to cardiac fibrosis81 and are shown to directly promote collagen I expression in cardiac fibroblasts.82 M2 macrophages release arginase 1 capable of controlling l-proline production essential for the collagen synthesis of activated myofibroblasts.83 For example, inflammatory Gr1+ monocytes, recruited to the injured liver in a CCR2-dependent manner, give rise to CD11b+F4/80+ macrophages producing TGF-β and inducible nitric oxide synthase directly promoting the progression of liver fibrosis.84 Furthermore, suppression of cardiac monocyte/macrophage infiltration in deoxycorticosterone acetate/salt hypertensive rats subsides myocardial fibrosis.85 Interestingly, in Coxsackievirus-induced myocarditis, two distinct M2 macrophage populations have a completely different role in the development of cardiac fibrosis. TLR4+casp-1+IL-1β+ M2 macrophages exacerbate inflammation and fibrosis, whereas Tim-3+ M2 macrophages decrease the inflammatory and fibrotic response.86
Activated macrophages produce, in addition to TGF-β, cytokines such as IL-4, IL-10, IL-13,87 tumour necrosis factor (TNF)-α, and IL-1, which have been shown to activate fibroblasts, overproducing proteins of the ECM. Furthermore, TNF-α has been found to activate the extracellular regulated kinase-specific pathway in fibroblasts resulting in increased expression of TGF-β88 and support the excessive production of pro-inflammatory cytokines via the nuclear factor-kappa B (NF-κB) pathway.89 Once activated, macrophages not only secrete pro-fibrotic factors, but also recruit myofibroblasts and exacerbate inflammatory cell infiltration to sites of tissue injury, leading to profound production of a variety of chemokines, cytokines, and growth factors, which endpoint of repair turns to excessive and poorly ordered matrix deposition and fibrosis.23,90–93
3.1.4 Eosinophils
Eosinophils secrete TGF-β, granule proteins, major basic protein (MBP), lysosomal hydrolytic enzymes, and eosinophilic peroxidase, factors implicated in tissue remodelling and fibrosis.94 Eosinophil-derived TGF-β induces fibroblast activation and transdifferentiation to myofibroblasts overexpressing ECM proteins.94 MBP-1 has been shown to exert pro-fibrotic effects in muscular dystrophy,95 while granule proteins directly stimulate fibroblast proliferation. Importantly, TGF-β, MBP, and eosinophilic peroxidase induce epithelial–mesenchymal transition further contributing to myofibroblast generation and fibrosis.94 Moreover, toxic eosinophil granular proteins have been involved in the development of endomyocardial fibrosis in the hypereosinophilic heart syndrome.96,97
3.1.5 Mast cells
Mast cells produce a variety of proteases, cytokines, growth factors, vasoactive agents, and other biologically active mediators such as tryptase, chymase, and TGF-β, which are known to activate fibroblasts and subsequently support the development of cardiac fibrosis.98,99 Volume overload is associated with increased mast cell density in the LV, paralleled with activation of MMPs, subsequent collagen degradation and LV dilatation, and cardiac fibrosis.100 Furthermore, a recent study demonstrated that IL-4, most likely produced by mast cells in the heart during pressure overload, is also a significant contributor to cardiac fibrosis.101 In contrast, mast cell deficiency was shown to be protective in pulmonary102 and cardiac fibrosis.103
3.2 Adaptive immunity triggers of fibrosis
3.2.1 Th1 and Th2 cells
While a number of studies suggest a pro-fibrotic role of T cells in fibrosis, it became also evident that T cells are indispensable, proved by the finding that T-cell-deficient mice develop prominent fibrosis.104 The pro-fibrotic effect of T lymphocytes seems to be context-dependent and the factors present in the environment trigger specific T-cell populations, which subsequently determine the fibrotic outcome.
A substantial pile of evidence links T-helper type 2 (Th2) cells, characterized by the secretion of the cytokines IL-4, IL-5, and IL-13, with wound healing and fibrosis.105,106 IL-13 is one of the most noticeable mediators of fibrosis107,108 and in combination with IL-4 is capable of inducing the phenotypic transition of human fibroblasts to myofibroblasts in a c-Jun NH2-terminal kinase-dependent manner.109 Furthermore, IL-13 has been found to inhibit fibroblast MMP synthesis and subsequently down-regulates the matrix degradation, which results in excessive collagen deposition.110 IL-13 has been shown to induce TGF-β1 in macrophages via the IL-13Rα2 receptor, which elimination in vivo reduced the production of TGF-β1 and collagen deposition in bleomycin-induced lung fibrosis.111 Moreover, a very recent study has demonstrated that disturbance of IL-13/TGF-β1 interaction by IL-13Rα2 siRNA prevents cardiac allograft fibrosis.112 Therefore, therapeutic targeting of IL-13Rα2 receptor in the course of extended inflammation could prevent TGF-β1-associated fibrosis. Moreover, IL-3 influences Th17-mediated inflammation and Th2-driven fibrosis113 and facilitates its direct pro-fibrotic activity114 via IL-13Rα1 and IL-13Rα2 expressed directly on the reactive myofibroblasts.
The Th1 cytokine IFN-γ plays a controversial role in inflammation and fibrosis, with numerous reports showing pro-fibrotic and anti-fibrotic effects. The pro-fibrotic effects of IFN-γ follow from IFN-γ-deficient mice having attenuated lung inflammation and fibrosis after intratracheal bleomycin administration.115 In addition, a recent study by Marko et al.116 demonstrated that angiotensin II-treated IFN-γR knockout mice exhibited reduced cardiac hypertrophy, decreased infiltration of cardiac macrophages and T cells, and less cardiac fibrosis. IFN-γ mediates its pro-fibrotic activities via intensifying the production of pro-inflammatory and pro-fibrotic mediators such as TNF-α.117,118 IFN-γ-producing T cells control the differentiation, migration, and activation of macrophages as well as their MCP-1 expression, which subsequently leads to inflammation and fibrosis.106,119 A role of IFN-γ signalling in the differentiation of resident fibroblasts to myofibroblasts has not been elucidated so far.
The anti-fibrotic effects of IFN-γ have been discovered long time ago. IFN-α and -γ potently inhibit the collagen production of human fibroblasts, regulating the normal and pathological fibrogenesis.120 In a bleomycin-induced mouse model of lung fibrosis, IFN-γ initially down-regulated the bleomycin-induced overexpression of TGF-β and subsequently the procollagen expression, resulting in reduced collagen content.121 The exact mechanism of the IFN-γ-mediated suppression of TGF-β has been revealed. IFN-γ signalling via the Jak/STAT1 pathway induces an instant expression of SMAD7, which prevents the TGF-β-mediated phosphorylation of SMAD3 and consequently abrogates the TGF-β signalling to the nucleus.122 Transcriptional modification via STAT1 is another mechanism via which IFN-γ antagonizes TGF-β signalling and collagen deposition in lung fibrosis.123 In addition, IFN-γ inhibits the IL-4- and IL-13-promoted differentiation from fibrocytes into myofibroblasts.124 In a model of a chronic viral myocarditis, IFN-γ reduced TGF-β1, IL-1β, and IL-4-associated inflammation and fibrosis.125
3.2.2 Th17 cells
Th17 cells expressing IL-17A and IL-22 are another essential player in the development and progression of fibrotic disease in lung,126 cardiac,127 and hepatic fibrosis.128 IL-17A is characterized by its ability to induce the expression of a variety of pro-inflammatory mediators, such as IL-1, IL-6, TNF-α, CXCL8, granulocyte colony-stimulating factor, and granulocyte–macrophage colony-stimulating factor by endothelial and epithelial as well as stromal cells, which ultimately results in the recruitment and activation of neutrophils.129 Moreover, Th17 cells can directly chemoattract neutrophils, known for their pro-fibrotic effect, through the production and release of CXCL8.130 Furthermore, IL-17 promotes MMP-1 expression in cardiac fibroblasts via NF-κB, activating protein-1, and CCAAT-enhancer-binding protein (C/EBP)-β activation131 and induces cardiac fibrosis via activation of the protein kinase C (PKC)β/Erk1/2/NF-κB pathway.132 During the course of viral myocarditis, IL-17 causes the proliferation of cardiac fibroblasts and, in parallel, induces the degradation of collagen Type I and III via up-regulation of MMP-2.133 The pro-fibrotic role of IL-22, which is up-regulated in patients with chronic hepatitis B virus infection and liver fibrosis, follows from the finding that IL-22 blockade in hepatitis B virus transgenic mice with T-cell-mediated liver fibrosis restricted the progression of liver fibrosis.134
However, IL-17 and -22 have besides pro-fibrotic also anti-fibrotic features. Nakashima et al.135 have shown that IL-17A down-regulated the expression of CTGF and collagen I in fibroblasts of healthy patients. Th17 cells elicited MCP-1, IL-8, and a MMP-1 response, while simultaneously inhibited Type I collagen production in dermal fibroblasts of healthy and systemic sclerosis patients.136 In models of hypersensitive pneumonitis, IL-17 and -22 even exhibit a dual role in fibrosis, where their pro- or anti-inflammatory/fibrotic effects depend on the particular antigen.137 Taking the above-discussed studies into account, it seems that Th17 cells play a dual role in the fibrosis process. Probably, the presence of regulatory mediators, such as chemokines, transcription factors, and receptors in the particular inflammatory environment, can guide the Th17 response in a pro-fibrotic or anti-fibrotic direction.
3.2.3 Regulatory T cells
Th17 cells are not the only dual cell population. Tregs can also either suppress or promote fibrosis. Tregs release important immunosuppressive cytokines including IL-10 and TGF-β that have control over the inflammatory response and contribute to the maintenance of self-tolerance and host immune defence. Tregs play a considerable role during the inflammatory process and the subsequent progression of fibrosis contributing138 or suppressing139 the development of fibrosis. Adoptive transfer of Tregs in hypertensive hearts has been shown to attenuate cardiac fibrosis and inflammation, to reduce the interstitial myofibroblast numbers, and to decrease the activity of the TGF-β1 system.139 Moreover, IL-10 produced by Tregs in vivo and in vitro significantly inhibits the collagen synthesis by cardiac fibroblasts.140 This is in line with other reports showing pronounced anti-fibrotic features of IL-10 in models of wound healing and Crohn's disease.141,142 IL-10 exerts its anti-fibrotic effects via reduction of STAT3 activity143 and via inhibition of the NF-κB pathway.144 In contrast, long-term overexpression of IL-10 promotes lung fibrosis via fibrocyte recruitment and M2 macrophage activation.145 Moreover, Tregs releasing TGF-β1 have been shown to affect CD4+ T-cell homeostasis in an HIV model by inducing collagen deposition in lymphatic tissues.146 In contrast, depletion of Tregs attenuated the progress of silica-induced lung fibrosis and enhanced the Th1 response.138 Therefore, before considering Tregs application as a suitable anti-fibrotic strategy, efforts should be focused on understanding the exact mechanisms and the particular tissue environment factors modulating whether Tregs would exhibit an anti-fibrotic or pro-fibrotic effect.
3.2.4 B cells
B cells are involved in antigen presentation, produce autoantibodies and various cytokines, and are essential players in immune-mediated disorders.147,148 In addition, B cells have been shown to play a role in hepatic fibrosis in an antibody- and T-cell-independent manner.149 B cells release the pro-fibrotic cytokine IL-6 and trigger liver fibrosis by inducing differentiation of hepatic stellate cells into myofibroblasts, promoting fibroblast proliferation, and augmenting the collagen and TIMP synthesis.150 In addition, Zhou et al.151 have demonstrated that anti-fibrillin-1 autoantibodies from systemic sclerosis patients have a potent pro-fibrotic effect in normal dermal fibroblasts, indicated by the increased expression of ECM components, the phosphorylation and nuclear translocation of SMAD3, as well as the induction of TGF-β1. A recent study illustrated that B cells releasing B-cell-activating factor are prominent inducers of excessive collagen, TIMP1, MMP-9, α-SMA expression in human dermal fibroblasts, as well as of pro-inflammatory and pro-fibrotic cytokines IL-6, CCL2, and TGF-β.152 Furthermore, TNF-α-secreting B cells have been shown to contribute to myocardial fibrosis and fibrosis-related cardiac dysfunction in patients with dilated cardiomyopathy.153 In conclusion, the role of B cells in (cardiac) fibrosis has so far been neglected and requires further investigation.
4. Fibroblasts are key players in the control of tissue damage
Fibroblasts modify the quantity, quality, and duration of the inflammatory infiltrate and play a critical role in the switch of acute resolving to chronic persistent inflammation154 by several means. An overview of their impact on immune cell chemotaxis, infiltration, transendothelial migration, retention, and apoptosis, and underlying mechanisms, is outlined below and illustrated in Figure 2.
Figure 2
Role of (activated) fibroblasts in the inflammatory process. (Activated) fibroblasts from the stromal microenvironment drive homing of circulating leucocytes via the release of chemokines and promote the recruitment of circulating leucocytes on endothelial cells via the induction of adhesion molecules on the endothelium. Furthermore, myofibroblasts activate leucocytes to produce the gelatinase MMP-9, allowing degradation of the basal membrane and subsequent transendothelial migration. Activated fibroblasts enhance local T-cell persistence via (i) up-regulation of CXCR4 on T cells via TGF-β and induced expression of its ligand stromal-derived factor-1 (SDF-1), promoting the SDF-1/CXCR4 axis and via (ii) the reduction of T-cell apoptosis involving the release of IFN-β.
The vascular endothelium is an anatomical defence barrier. Circulating leucocytes from the blood flow have to pass the endothelium to reach the underlying tissue. Traditionally, vascular inflammation has been described as an event whereby, upon endothelial cell activation, leucocytes extravasate and induce an inflammatory response onto the microenvironment via the release of pro-inflammatory stimuli. Though, it is more and more recognized that also the fibroblasts from the stromal microenvironment drive homing of circulating leucocytes155 and promote activation of the endothelium,156–158 indicating a bidirectional activation in response to tissue injury. In detail, fibroblasts are a major source of constitutive and cytokine-induced C–C and C–X–C chemokines including MCP-1, macrophage inflammatory protein (MIP)-1, RANTES, and IP-10 and express chemokine receptors.159 Fibroblasts derived from an environment with cell-mediated inflammatory responses demonstrate a dramatic alteration in their cytokine profile and produce high levels of MCP-1, when compared with normal fibroblasts.160 MCP-1 itself also stimulates collagen expression and endogenous up-regulation of TGF-β expression in fibroblasts, leading to autocrine and/or juxtacrine stimulation of collagen gene expression.161 The induction in chemokine production following activation of fibroblasts with inflammatory stimuli such as TNF-α depends on the transcription factor RelB, which is capable of stabilizing IκB, the endogenous NF-κB inhibitor. In the absence of RelB expression in fibroblasts, but not in macrophages, NF-κB activity is uncontrolled leading to continuous chemokine generation. This has been elegantly demonstrated by experiments with RelB−/− fibroblasts, which exhibited a dramatic persistent induction of chemokine production, including MCP-1, MIP-1α, MIP-1β, MIP-2, IP-10, and RANTES, following lipopolysaccharide. In contrast, upon lipopolysaccharide activation of normal fibroblasts, only a transient production of chemokines, closely followed by induction of RelB expression, was induced.162In vivo, activated RelB−/− fibroblasts dramatically increased the recruitment of granulocytes into tissues,162 further supporting the importance of chemokine production by fibroblasts in tissue inflammation. Interestingly, fibroblasts from fibrotic lesions express higher levels of the MCP-1 (CCL2) receptor, CCR2, compared with those from non-fibrotic lesions,160 further enhancing hereby the inflammatory and fibrotic process. Furthermore, co-culture of macrophages with fibroblasts revealed that a contact-dependent expression of chemokines, especially of the macrophage-derived MIP-1α, is induced in macrophages. Further experiments unravelled that MIP-1 production in macrophages was dependent on the TNF-α-induced expression of intercellular adhesion molecule-1 (ICAM-1) by the fibroblast.163 On the other hand, the CCL2/CCR2 axis enhances vascular cell adhesion molecule-1 (VCAM-1) expression in human synovial fibroblasts promoting monocyte adhesion to human fibroblasts,164 indicating a reciprocal enhancement of monocyte–fibroblast adhesion and chemokine production. Taken together, fibroblasts have the ability to participate in the maintenance of an inflammatory response via the expression of chemokines.160 The interaction between fibroblasts and macrophages may be an important early event in the recruitment of monocytes and may facilitate a cytokine network that maintains the activation of tissue inflammation.
Besides attracting immune cells to the site of injury via the release of chemokines, fibroblasts can promote or inhibit the recruitment of circulating leucocytes on endothelial cells via the induction or decrease of cytokine-induced expression of adhesion molecules on endothelial cells.165 This influence of fibroblasts on the cytokine sensitivity of the vascular endothelium depends on the tissue origin of the fibroblasts and is restricted to normal stromal fibroblasts. Fibroblasts associated with chronic inflammation bypass this and develop a direct inflammatory phenotype.165 Lindner et al.166 further demonstrated that cardiac fibroblasts also activate leucocytes to produce the gelatinase MMP-9, allowing easier transendothelial migration through the basal membrane.
Fibroblasts not only modulate the recruitment of immune cells, but also regulate their behaviour, retention, and survival in damaged tissue. In general, the crosstalk between fibroblasts and leucocytes depends on the interaction between the leucocyte surface antigen CD40 on fibroblasts and its ligand, CD40L, which is expressed on immune cells. This interaction induces, among others, the up-regulation of ICAM-1 and VCAM-1 on fibroblasts,167,168 which in turn are important for the induction of chemokine production (see below) and the reduction in T-cell apoptosis (see above). Furthermore, fibroblasts also express the co-stimulatory molecule B7,169 suggesting that similar to interactions between lymphocytes and antigen-presenting cells, CD40-CD40L and co-stimulatory CTLA4-B7 interactions play an important role in the fibroblast–leucocyte crosstalk.
Activated fibroblasts up-regulate CXCR4 on T cells via TGF-β170 and express its ligand stromal-derived factor-1 (SDF-1),171 suggesting that fibroblasts alter the migratory phenotype of the leucocytes towards a stationary phenotype via SDF-1/CXCR4 interactions, leading to retention of the infiltrated cells. The importance of CXCR4 expression on T cells for the recruitment of activated T cells towards inflammatory sites follows from T-cell-specific CXCR4-deficient mice, which exhibit less T cells into affected joints and a lower incidence of collagen-induced arthritis compared with wild-type littermates.172 Intriguingly, SDF is also an important attractant for fibrocytes. Its significance for fibrocyte recruitment has recently been demonstrated by Garibaldi et al.173 They showed a reduction in acute lung injury by decreasing fibrocyte recruitment and subsequent fibroproliferation involving a down-regulation in SDF expression. On the other hand, also the recruitment of MSC depends on SDF/CXCR4 signalling.174–176 Though, in contrast to fibrocytes and leucocytes, which exert pro-inflammatory and pro-fibrotic effects, MSCs have immunomodulatory177–179 and anti-fibrotic179,180 features. Depending on the local concentration of SDF-1α, this chemokine can act as a chemo-attractant as well as a repellent for leucocytes.3,181 The above-mentioned observations illustrate the delicate and fine-tuned regulation of SDF/CXCR4 interactions and their influence on the retention of leucocytes/fibrocytes as well as of MSC, promoting or reducing the inflammatory/fibrotic process. Besides raising the retention of infiltrated T cells via inducing the SDF/CXCR4 axis, fibroblasts promote local T-cell persistence via reducing T-cell apoptosis. This occurs in an integrin-ligand-dependent manner182 and via the release of IFN-β,183 bringing the cell hereby in a resting G0/G1 state.
5. Link of inflammation and cardiac fibrosis in the development of heart failure
With (i the spleen being a reservoir of monocytes, which are recruited to the inflammatory heart,184 and the findings that (ii) splenectomy improves the outcome of myocardial infarction,185 (iii) the use of an antibody against T cells decreases cardiac damage in myocarditis,186 and (iv) our observation that ex vivo supplementation of splenocytes isolated from mice with virus-induced inflammatory cardiomyopathy to fibroblasts induces more collagen production in fibroblasts compared with splenocytes from control mice,179 we postulated the hypothesis that the cardiosplenic axis is important for cardiac inflammation, fibrosis, and the subsequent development of heart failure.175,187 This hypothesis has recently been elegantly demonstrated and detailed by Ismahil et al.188 Via splenectomy experiments and adoptive transfer of splenocytes from mice with heart failure, but not from sham-operated mice in naive recipients, they showed that activation of mononuclear phagocytes is central to the progression of cardiac remodelling in heart failure and heightened antigen processing in the spleen plays a critical role in this process. Furthermore, the authors illustrated that splenocytes promote immune-mediated fibrosis responses in the failing heart, and retain this memory upon adoptive transfer.188
Via up-regulated chemokine expression, e.g. MCP-1, immune cells from the spleen are attracted to the heart. In a model of suprarenal aortic constriction, increased cardiac expression of MCP-1 was observed preceding TGF-β1 up-regulation and subsequent cardiac fibrosis and diastolic dysfunction.189 The significance of TGF-β1 follows, among others, from experiments in pressure-overloaded rats where administration of an anti-TGF-β1 neutralizing antibody 1 day before operation inhibited fibroblast activation and subsequently prevented collagen mRNA induction and myocardial fibrosis, but not myocyte hypertrophy.68 Westermann et al.62 provided insights into the role of cardiac inflammation as a pro-fibrogenic stimulus in subjects with heart failure with preserved ejection fraction (HFPEF). In endomyocardial biopsy specimens from these patients, they identified increased inflammatory cells and higher TGF-β1 mRNA levels associated with both reduced levels of MMP-1, the major collagenase in the human heart, and elevated TIMP-1 levels. In accordance with these findings indicating TGF-β1-induced collagen synthesis in human subjects with HFPEF, in vitro stimulation of primary human cardiac fibroblasts from HFPEF patients with TGF-β1 resulted in transdifferentiation of fibroblasts to myofibroblasts, which produced more CTGF and more collagen and expressed less MMP-1. Double staining further confirmed the expression of TGF-β1 in immune cells present in endomyocardial biopsies of HFPEF patients, whereas activated human acute monocytic leukemia cell line (THP-1) monocytes were found to express TGF-β1 in a time-dependent manner in vitro. Collectively, these data provide accumulating evidence linking cardiac inflammation with TGF-β1-induced collagen synthesis and diastolic dysfunction.190 The pro-fibrotic factor CTGF, which is induced in cardiac fibroblasts upon TGF-β1 stimulation, also enhances the migration of monocytes.191 The ability of cardiac fibroblasts to induce the chemotaxis of monocytes via their production of CTGF191 and/or chemokines166 and to facilitate transendothelial migration through the basal membrane166 indicate a self-maintaining mechanism supporting the inflammatory and fibrotic process.192 It is suggested that these mechanisms also affect the cardiomyocyte, including the function of titin (Figure 3).193 Further studies still have to prove whether thereby differences occur between HFPEF and heart failure with reduced ejection fraction.
Figure 3
Pivotal role of myofibroblasts in the pathogenesis of heart failure. Risk factors of heart failure (diabetes mellitus, age, smoking, hypertension, etc.) lead to endothelial cell activation and the induction of inflammatory responses, which trigger the transdifferentiation of fibroblasts to myofibroblasts via the release of cytokines and growth factors. Myofibroblasts, in turn, lead to extensive collagen production and the production of chemokines and further activate inflammatory cells to adhere to the endothelium, to release MMP-9 enabling transendothelial migration, and to induce chemokine expression, attracting other inflammatory cells. Besides triggering fibroblasts, inflammatory cells also lead to titin dephosphorylation in cardiomyocytes via the induction of oxidative stress leading to cardiomyocyte stiffness and further contributing to the pathogenesis of heart failure. Figure has been adapted from Tschöpe and Lam193 with kind permission of Springer Science+Business Media.
6. Conclusions and perspectives
There is accumulating evidence showing that fibroblasts are -in contrast to their traditional view of being solely matrix-producing cells -cells with important immunomodulatory properties, playing a pivotal role in the switch to chronic inflammation. The complex interaction among different immune cells triggering the fibroblast on the one hand, and the multifactorial enhancement of tissue inflammation by fibroblasts on the other hand (as summarized in this review) might explain why uni-directed strategies blocking inflammation194 or fibrosis have failed to abrogate the fibrotic process, and indicate the need for therapies with more broaden immunomodulatory effects. In that view, MSC and the recently identified cardiac-derived adherent proliferating cells (CardAPs)195 having both immunomodulatory177–179,196 and anti-fibrotic179,180,197 features are attractive tools to counteract the inflammatory/fibrotic process. They home to the site of injury and their cardioprotective effects are exerted via the cardiosplenic axis.179 CardAPs might further profit from their cardiac niche origin. Especially HFPEF, where the mechanisms triggering inflammation and fibrosis probably represent multifactorial stressors, including metabolic derangements (diabetes mellitus), might profit from a multi-directed strategy like the iv application of MSC. Finally, the differences in functionality among fibroblasts from healthy and diseased tissues, which underlie their diverse intrinsic susceptibility to inflammation, indicate the need to investigate the inflammation/fibrosis status via (endomyocardial) biopsies allowing an optimal stratified strategy.
Funding
This study is supported by the DZHK to S.V.L. and C.T., and by the European 7th Framework Consortia MEDIA and REDDSTAR to C.T.
References
1
Fibrosis and hypertensive heart disease
,
Curr Opin Cardiol
,
2000
, vol.
15
(pg.
264
-
272
)
2
The immunology of fibrosis
,
Ann Rev Immunol
,
2013
, vol.
31
(pg.
107
-
135
)
3
Fibroblasts regulate the switch from acute resolving to chronic persistent inflammation
,
Trends Immunol
,
2001
, vol.
22
(pg.
199
-
204
)
4
Study of non-muscle cells of the adult mammalian heart: a fine structural analysis and distribution
,
Cytobios
,
1980
, vol.
28
(pg.
41
-
61
)
5
Structural and functional characterisation of cardiac fibroblasts
,
Cardiovasc Res
,
2005
, vol.
65
(pg.
40
-
51
)
6
Communication signals between cardiac fibroblasts and cardiac myocytes
,
J Cardiovasc Pharmacol
,
2011
, vol.
57
(pg.
513
-
521
)
7
Cardiac intercellular communication: are myocytes and fibroblasts fair-weather friends?
,
J Cardiovasc Transl Res
,
2012
, vol.
5
(pg.
768
-
782
)
8
Chemokines and cd40 expression in human fibroblasts
,
Eur J Immunol
,
2000
, vol.
30
(pg.
914
-
919
)
9
Differential expression of matrix metalloproteases in human fibroblasts with different origins
,
Biochem Res Int
,
2012
, vol.
2012
pg.
875742
10
Global gene expression profiles in fibroblasts from synovial, skin and lymphoid tissue reveals distinct cytokine and chemokine expression patterns
,
Thromb Haemost
,
2003
, vol.
90
(pg.
688
-
697
)
11
Diversity, topographic differentiation, and positional memory in human fibroblasts
,
Proc Natl Acad Sci USA
,
2002
, vol.
99
(pg.
12877
-
12882
)
12
Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymal transition
,
J Biol Chem
,
2007
, vol.
282
(pg.
23337
-
23347
)
13
Fibroblasts emerge via epithelial-mesenchymal transition in chronic kidney fibrosis
,
Front Biosc
,
2008
, vol.
13
(pg.
6991
-
6998
)
14
Caveolin-1 deficiency induces spontaneous endothelial-to-mesenchymal transition in murine pulmonary endothelial cells in vitro
,
Am J Pathol
,
2013
, vol.
182
(pg.
325
-
331
)
15
Endothelial-to-mesenchymal transition contributes to cardiac fibrosis
,
Nat Med
,
2007
, vol.
13
(pg.
952
-
961
)
16
The role of endothelial-to-mesenchymal transition in cancer progression
,
Br J Cancer
,
2008
, vol.
99
(pg.
1375
-
1379
)
17
Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair
,
Mol Med
,
1994
, vol.
1
(pg.
71
-
81
)
18
Circulating fibrocytes contribute to the pathogenesis of collagen antibody-induced arthritis
,
Arthritis Rheum
,
2012
, vol.
64
(pg.
3583
-
3593
)
19
Elevated circulating fibrocyte levels in patients with hypertensive heart disease
,
J Hypertens
,
2012
, vol.
30
(pg.
1856
-
1861
)
20
Fibrocytes: emerging effector cells in chronic inflammation
,
Nat Rev Immunol
,
2011
, vol.
11
(pg.
427
-
435
)
21
Vascular remodeling in hypertension: roles of apoptosis, inflammation, and fibrosis
,
Hypertension
,
2001
, vol.
38
(pg.
581
-
587
)
22
TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity
,
Trends Immunol
,
2006
, vol.
27
(pg.
352
-
357
)
23
Infectious disease, the innate immune response, and fibrosis
,
J Clin Invest
,
2007
, vol.
117
(pg.
530
-
538
)
24
Intestinal myofibroblasts in innate immune responses of the intestine
,
Gastroenterology
,
2003
, vol.
124
(pg.
1866
-
1878
)
25
Expression of toll-like receptors on human platelets
,
Thromb Res
,
2004
, vol.
113
(pg.
379
-
385
)
26
Platelets express functional toll-like receptor-4
,
Blood
,
2005
, vol.
106
(pg.
2417
-
2423
)
27
Novel role of platelets in mediating inflammatory responses and ventricular rupture or remodeling following myocardial infarction
,
Arterioscler Thromb Vasc Biol
,
2011
, vol.
31
(pg.
834
-
841
)
28
Complements and the wound healing cascade: an updated review
,
Plastic Surgery Int
,
2013
, vol.
2013
pg.
146764
29
Procoagulant signalling mechanisms in lung inflammation and fibrosis: novel opportunities for pharmacological intervention?
,
Br J Pharmacol
,
2008
, vol.
153
Suppl 1
(pg.
S367
-
S378
)
30
Increased local expression of coagulation factor X contributes to the fibrotic response in human and murine lung injury
,
J Clin Invest
,
2009
, vol.
119
(pg.
2550
-
2563
)
31
Myocardial fibrosis in mice with overexpression of human blood coagulation factor ix
,
Blood
,
2003
, vol.
101
(pg.
1871
-
1873
)
32
Severe deficiency of coagulation factor vii results in spontaneous cardiac fibrosis in mice
,
J Pathol
,
2009
, vol.
217
(pg.
362
-
371
)
33
Endothelial dysfunction in cirrhosis and portal hypertension
,
Pharmacol Ther
,
2001
, vol.
89
(pg.
273
-
293
)
34
The platelet and platelet function testing in liver disease
,
Clin Liver Dis
,
2009
, vol.
13
(pg.
11
-
20
)
35
The inflammation-fibrosis link? A Jekyll and Hyde role for blood cells during wound repair
,
J Invest Dermatol
,
2007
, vol.
127
(pg.
1009
-
1017
)
36
Neutrophil clearance: when the party is over, clean-up begins
,
Trends Immunol
,
2011
, vol.
32
(pg.
350
-
357
)
37
Use of neutrophil count in early diagnosis and risk stratification of ami
,
Am J Med
,
2011
, vol.
124
(pg.
534
-
542
)
38
Flavonoids inhibit the respiratory burst of neutrophils in mammals
,
Oxid Med Cell Longev
,
2012
, vol.
2012
pg.
181295
39
Neutrophil-derived reactive oxygen species in SSC
,
Rheumatol (Oxford)
,
2012
, vol.
51
(pg.
1166
-
1169
)
40
Integrating mechanisms of pulmonary fibrosis
,
J Exp Med
,
2011
, vol.
208
(pg.
1339
-
1350
)
41
Involvement of reactive oxygen species and nitric oxide radicals in activation and proliferation of rat hepatic stellate cells
,
Liver
,
2001
, vol.
21
(pg.
1
-
12
)
42
Leonurine (SCM-198) attenuates myocardial fibrotic response via inhibition of NADPH oxidase 4
,
Free Radic Biol Med
,
2013
, vol.
54
(pg.
93
-
104
)
43
Inhibition of NADPH oxidase reduces myocardial oxidative stress and apoptosis and improves cardiac function in heart failure after myocardial infarction
,
Free Radic Biol Med
,
2007
, vol.
43
(pg.
271
-
281
)
44
Neutrophil roles in left ventricular remodeling following myocardial infarction
,
Fibrogenesis Tissue Repair
,
2013
, vol.
6
pg.
11
45
A neutrophil elastase inhibitor prevents bleomycin-induced pulmonary fibrosis in mice
,
Eur Respir J
,
2012
, vol.
40
(pg.
1475
-
1482
)
46
Macrophage-neutrophil interaction: a paradigm for chronic inflammation revisited
,
Immunol Cell Biol
,
2001
, vol.
79
(pg.
502
-
506
)
47
Protective and pathogenic functions of macrophage subsets
,
Nat Rev Immunol
,
2011
, vol.
11
(pg.
723
-
737
)
48
Reduced peribronchial fibrosis in allergen-challenged MMP-9-deficient mice
,
Am J Physiol Lung Cell Mol Physiol
,
2006
, vol.
291
(pg.
L265
-
L271
)
49
Matrix metalloproteinase (MMP)-9, but not MMP-2, is involved in the development and progression of c protein-induced myocarditis and subsequent dilated cardiomyopathy
,
J Immunol
,
2009
, vol.
183
(pg.
4773
-
4781
)
50
Targeted deletion of matrix metalloproteinase-9 attenuates left ventricular enlargement and collagen accumulation after experimental myocardial infarction
,
J Clin Invest
,
2000
, vol.
106
(pg.
55
-
62
)
51
Reduced degradation of the chemokine MCP-3 by matrix metalloproteinase-2 exacerbates myocardial inflammation in experimental viral cardiomyopathy
,
Circulation
,
2011
, vol.
124
(pg.
2082
-
2093
)
52
Transforming growth factor-beta and fibrosis
,
World J Gastroenterol
,
2007
, vol.
13
(pg.
3056
-
3062
)
53
TGF-beta driven lung fibrosis is macrophage dependent and blocked by serum amyloid p
,
Int J Biochem Cell Biol
,
2011
, vol.
43
(pg.
154
-
162
)
54
Making sense of latent tgfbeta activation
,
J Cell Sci
,
2003
, vol.
116
(pg.
217
-
224
)
55
Tgf-beta signaling and the fibrotic response
,
FASEB J
,
2004
, vol.
18
(pg.
816
-
827
)
56
Stimulation of normal human fibroblast collagen production and processing by transforming growth factor-beta
,
Biochem Biophys Res Commun
,
1986
, vol.
138
(pg.
974
-
980
)
57
Independent regulation of collagenase, 72-kDa progelatinase, and metalloendoproteinase inhibitor expression in human fibroblasts by transforming growth factor-beta
,
J Biol Chem
,
1989
, vol.
264
(pg.
1860
-
1869
)
58
Stimulation of Type I collagen transcription in human skin fibroblasts by TGF-beta: involvement of SMAD 3
,
J Invest Dermatol
,
1999
, vol.
112
(pg.
49
-
57
)
59
Identification of novel TGF-beta/SMAD gene targets in dermal fibroblasts using a combined CDNA microarray/promoter transactivation approach
,
J Biol Chem
,
2001
, vol.
276
(pg.
17058
-
17062
)
60
TGF-beta signaling in vascular fibrosis
,
Cardiovasc Res
,
2007
, vol.
74
(pg.
196
-
206
)
61
Involvement of transforming growth factor-beta in the formation of fibrotic lesions in carcinoid heart disease
,
Am J Pathol
,
1993
, vol.
142
(pg.
71
-
78
)
62
Cardiac inflammation contributes to changes in the extracellular matrix in patients with heart failure and normal ejection fraction
,
Circ Heart Fail
,
2011
, vol.
4
(pg.
44
-
52
)
63
IL-1beta suppresses TGF-beta-mediated myofibroblast differentiation in cardiac fibroblasts
,
Growth Fact
,
2013
, vol.
31
(pg.
81
-
89
)
64
P38 inhibitors prevent TGF-beta-induced myofibroblast transdifferentiation in human tenon fibroblasts
,
Invest Ophthalmol Vis Sci
,
2006
, vol.
47
(pg.
1500
-
1509
)
65
Control of scarring in adult wounds by neutralising antibody to transforming growth factor beta
,
Lancet
,
1992
, vol.
339
(pg.
213
-
214
)
66
Neutralization of TGF-beta by anti-TGF-beta antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice
,
Diabetes
,
1996
, vol.
45
(pg.
522
-
530
)
67
Role of transforming growth factor-beta1 in cardiovascular inflammatory changes induced by chronic inhibition of nitric oxide synthesis
,
Hypertension
,
2000
, vol.
35
(pg.
86
-
90
)
68
Transforming growth factor-beta function blocking prevents myocardial fibrosis and diastolic dysfunction in pressure-overloaded rats
,
Circulation
,
2002
, vol.
106
(pg.
130
-
135
)
69
Transforming growth factor-beta 1 knockout mice. A mutation in one cytokine gene causes a dramatic inflammatory disease
,
Am J Pathol
,
1993
, vol.
143
(pg.
3
-
9
)
70
Biology of TGF-beta in knockout and transgenic mouse models
,
Kidney Int
,
1997
, vol.
51
(pg.
1355
-
1360
)
71
Inhibition of TGF-beta signaling exacerbates early cardiac dysfunction but prevents late remodeling after infarction
,
Cardiovasc Res
,
2004
, vol.
64
(pg.
526
-
535
)
72
Postinfarction gene therapy against transforming growth factor-beta signal modulates infarct tissue dynamics and attenuates left ventricular remodeling and heart failure
,
Circulation
,
2005
, vol.
111
(pg.
2430
-
2437
)
73
Transforming growth factor (TGF)-beta1-producing regulatory T cells induce SMAD-mediated interleukin 10 secretion that facilitates coordinated immunoregulatory activity and amelioration of TGF-beta1-mediated fibrosis
,
J Exp Med
,
2003
, vol.
198
(pg.
1179
-
1188
)
74
TGF-beta: a balancing act
,
Int Rev Immunol
,
1998
, vol.
16
(pg.
553
-
580
)
75
Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair
,
J Clin Invest
,
2005
, vol.
115
(pg.
56
-
65
)
76
Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice
,
Am J Pathol
,
2007
, vol.
170
(pg.
818
-
829
)
77
Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm
,
Nat Immunol
,
2010
, vol.
11
(pg.
889
-
896
)
78
Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes
,
Trends Immunol
,
2002
, vol.
23
(pg.
549
-
555
)
79
Macrophage activation and polarization
,
Front Biosci
,
2008
, vol.
13
(pg.
453
-
461
)
80
Alternative activation of macrophages: an immunologic functional perspective
,
Ann Rev Immunol
,
2009
, vol.
27
(pg.
451
-
483
)
81
Cardiovascular effects of arginase inhibition in spontaneously hypertensive rats with fully developed hypertension
,
Cardiovasc Res
,
2010
, vol.
87
(pg.
569
-
577
)
82
Urokinase plasminogen activator induces pro-fibrotic/M2 phenotype in murine cardiac macrophages
,
PLoS ONE
,
2013
, vol.
8
pg.
e57837
83
Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of l-arginine metabolism
,
J Immunol
,
2001
, vol.
167
(pg.
6533
-
6544
)
84
Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes hepatic fibrosis
,
Hepatology
,
2009
, vol.
50
(pg.
261
-
274
)
85
Fasudil attenuates myocardial fibrosis in association with inhibition of monocyte/macrophage infiltration in the heart of DOCA/salt hypertensive rats
,
J Cardiovasc Pharmacol
,
2007
, vol.
50
(pg.
187
-
194
)
86
Alternatively activated macrophages in infection and autoimmunity
,
J Autoimmunity
,
2009
, vol.
33
(pg.
222
-
230
)
87
Macrophage plasticity and polarization: in vivo veritas
,
J Clin Invest
,
2012
, vol.
122
(pg.
787
-
795
)
88
TNF-alpha induces TGF-beta1 expression in lung fibroblasts at the transcriptional level via AP-1 activation
,
J Cell Mol Med
,
2009
, vol.
13
(pg.
1866
-
1876
)
89
NF-κβ in the liver—linking injury, fibrosis and hepatocellular carcinoma
,
Nat Rev Gastrol Hepatol
,
2011
, vol.
8
(pg.
108
-
118
)
90
Cellular and molecular mechanisms of fibrosis
,
J Pathol
,
2008
, vol.
214
(pg.
199
-
210
)
91
Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases
,
J Clin Invest
,
2007
, vol.
117
(pg.
524
-
529
)
92
Innate immune response and hepatic inflammation
,
Semin Liver Dis
,
2007
, vol.
27
(pg.
339
-
350
)
93
TLR4 enhances TGF-beta signaling and hepatic fibrosis
,
Nat Med
,
2007
, vol.
13
(pg.
1324
-
1332
)
94
Relationships between eosinophilic inflammation, tissue remodeling, and fibrosis in eosinophilic esophagitis
,
Immunol Allergy Clin North Am
,
2009
, vol.
29
(pg.
197
-
211
)
xiii-xiv
95
Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy
,
Hum Mol Genet
,
2008
, vol.
17
(pg.
2280
-
2292
)
96
Eosinophilic products lead to myocardial damage
,
Hum Pathol
,
1989
, vol.
20
(pg.
850
-
857
)
97
Deposits of eosinophil granule proteins in cardiac tissues of patients with eosinophilic endomyocardial disease
,
Lancet
,
1987
, vol.
1
(pg.
643
-
647
)
98
Cardiac mast cells mediate left ventricular fibrosis in the hypertensive rat heart
,
Hypertension
,
2009
, vol.
53
(pg.
1041
-
1047
)
99
Chymase induces profibrotic response via transforming growth factor-beta 1/SMAD activation in rat cardiac fibroblasts
,
Mol Cell Biochem
,
2008
, vol.
310
(pg.
159
-
166
)
100
Cardiac mast cell- and chymase-mediated matrix metalloproteinase activity and left ventricular remodeling in mitral regurgitation in the dog
,
J Mol Cell Cardiol
,
2003
, vol.
35
(pg.
311
-
319
)
101
A pro-fibrotic role for interleukin-4 in cardiac pressure overload
,
Cardiovasc Res
,
2012
, vol.
95
(pg.
77
-
85
)
102
Mast cells: a pivotal role in pulmonary fibrosis
,
DNA Cell Biol
,
2013
, vol.
32
(pg.
206
-
218
)
103
Cardiac mast cells: the centrepiece in adverse myocardial remodelling
,
Cardiovasc Res
,
2011
, vol.
89
(pg.
12
-
19
)
104
Ship-deficient mice develop spontaneous intestinal inflammation and arginase-dependent fibrosis
,
Am J Pathol
,
2011
, vol.
179
(pg.
180
-
188
)
105
Liver fibrosis: cellular mechanisms of progression and resolution
,
Clin Sci (Lond)
,
2007
, vol.
112
(pg.
265
-
280
)
106
Fibrotic disease and the T(h)1/T(h)2 paradigm
,
Nat Rev Immunol
,
2004
, vol.
4
(pg.
583
-
594
)
107
Schistosome infection of transgenic mice defines distinct and contrasting pathogenic roles for IL-4 and IL-13: IL-13 is a profibrotic agent
,
J Immunol
,
2000
, vol.
164
(pg.
2585
-
2591
)
108
Anti-IL-13 monoclonal antibody inhibits airway hyperresponsiveness, inflammation and airway remodeling
,
Cytokine
,
2004
, vol.
28
(pg.
224
-
232
)
109
IL-4 and IL-13 induce myofibroblastic phenotype of human lung fibroblasts through c-Jun NH2-terminal kinase-dependent pathway
,
J Allergy Clin Immunol
,
2001
, vol.
107
(pg.
1001
-
1008
)
110
IL-13 promotes collagen accumulation in Crohn's disease fibrosis by down-regulation of fibroblast MMP synthesis: a role for innate lymphoid cells?
,
PLoS ONE
,
2012
, vol.
7
pg.
e52332
111
IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis
,
Nat Med
,
2006
, vol.
12
(pg.
99
-
106
)
112
IL-13 signaling via IL-13ralpha2 triggers TGF-beta1-dependent allograft fibrosis
,
Transplant Res
,
2013
, vol.
2
pg.
16
113
Opposing roles for IL-13 and IL-13 receptor alpha 2 in health and disease
,
Immunol Rev
,
2004
, vol.
202
(pg.
191
-
202
)
114
Unique functions of the Type II interleukin 4 receptor identified in mice lacking the interleukin 13 receptor alpha1 chain
,
Nat Immunol
,
2008
, vol.
9
(pg.
25
-
33
)
115
Attenuation of lung inflammation and fibrosis in interferon-gamma-deficient mice after intratracheal bleomycin
,
Am J Respir Cell Mol Biol
,
2001
, vol.
24
(pg.
545
-
555
)
116
Interferon-gamma signaling inhibition ameliorates angiotensin II-induced cardiac damage
,
Hypertension
,
2012
, vol.
60
(pg.
1430
-
1436
)
117
Activation of human macrophages. Comparison of other cytokines with interferon-gamma
,
J Exp Med
,
1984
, vol.
160
(pg.
600
-
605
)
118
Tumor necrosis factor/cachectin plays a key role in bleomycin-induced pneumopathy and fibrosis
,
J Exp Med
,
1989
, vol.
170
(pg.
655
-
663
)
119
Reciprocal interaction between macrophages and T cells stimulates IFN-gamma and MCP-1 production in Ang II-induced cardiac inflammation and fibrosis
,
PLoS ONE
,
2012
, vol.
7
pg.
e35506
120
Selective inhibition of human diploid fibroblast collagen synthesis by interferons
,
J Clin Invest
,
1984
, vol.
74
(pg.
1112
-
1116
)
121
Molecular mechanisms of antifibrotic effect of interferon gamma in bleomycin-mouse model of lung fibrosis: downregulation of TGF-beta and procollagen I and III gene expression
,
Exp Lung Res
,
1995
, vol.
21
(pg.
791
-
808
)
122
Inhibition of transforming growth factor-beta/SMAD signalling by the interferon-gamma/stat pathway
,
Nature
,
1999
, vol.
397
(pg.
710
-
713
)
123
Molecular mechanisms of TGF-(beta) antagonism by interferon (gamma) and cyclosporine a in lung fibroblasts
,
FASEB J
,
2001
, vol.
15
(pg.
797
-
806
)
124
Pivotal advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation
,
J Leukoc Biol
,
2008
, vol.
83
(pg.
1323
-
1333
)
125
Interferon-gamma protects against chronic viral myocarditis by reducing mast cell degranulation, fibrosis, and the profibrotic cytokines transforming growth factor-beta 1, interleukin-1 beta, and interleukin-4 in the heart
,
Am J Pathol
,
2004
, vol.
165
(pg.
1883
-
1894
)
126
Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17a dependent
,
J Exp Med
,
2010
, vol.
207
(pg.
535
-
552
)
127
IL-17 induces myocardial fibrosis and enhances RANKl/OPG and MMP/TIMP signaling in isoproterenol-induced heart failure
,
Exp Mol Pathol
,
2009
, vol.
87
(pg.
212
-
218
)
128
IL-17 expression is correlated with hepatitis b related liver diseases and fibrosis
,
Int J Mol Med
,
2011
, vol.
27
(pg.
385
-
392
)
129
The biological functions of T helper 17 cell effector cytokines in inflammation
,
Immunity
,
2008
, vol.
28
(pg.
454
-
467
)
130
Evidence for a cross-talk between human neutrophils and Th17 cells
,
Blood
,
2010
, vol.
115
(pg.
335
-
343
)
131
IL-17 stimulates MMP-1 expression in primary human cardiac fibroblasts via p38 MAPK- and ERK1/2-dependent C/EBP-beta, NF-κβ, and AP-1 activation
,
Am J Physiol Heart Circ Physiol
,
2007
, vol.
293
(pg.
H3356
-
H3365
)
132
IL-17 contributes to cardiac fibrosis following experimental autoimmune myocarditis by a PKCbeta/Erk1/2/NF-κβ-dependent signaling pathway
,
Int Immunol
,
2012
, vol.
24
(pg.
605
-
612
)
133
In vivo delivery of adenoviral vector containing interleukin-17 receptor a reduces cardiac remodeling and improves myocardial function in viral myocarditis leading to dilated cardiomyopathy
,
PLoS ONE
,
2013
, vol.
8
pg.
e72158
134
Increased IL-22-producing cells contribute to liver fibrosis through promoting Th17 migration in chronic HBV patients
,
J Immunol
,
2013
, vol.
190
(pg.
202
-
204
)
135
Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts
,
J Immunol
,
2012
, vol.
188
(pg.
3573
-
3583
)
136
Th17 cells favor inflammatory responses while inhibiting type I collagen deposition by dermal fibroblasts: differential effects in healthy and systemic sclerosis fibroblasts
,
Arthritis Res Ther
,
2013
, vol.
15
pg.
R151
137
Gammadelta T cells and Th17 cytokines in hypersensitivity pneumonitis and lung fibrosis
,
Trans Res
,
2009
, vol.
154
(pg.
222
-
227
)
138
CD4+CD25+FOXp3+ regulatory T cells depletion may attenuate the development of silica-induced lung fibrosis in mice
,
PLoS ONE
,
2010
, vol.
5
pg.
e15404
139
CD4(+)CD25(+)FOXp3(+) regulatory t cells suppress cardiac fibrosis in the hypertensive heart
,
J Hypertens
,
2011
, vol.
29
(pg.
1820
-
1828
)
140
Adoptive transfer of regulatory T cells protects against Coxsackievirus B3-induced cardiac fibrosis
,
PLoS ONE
,
2013
, vol.
8
pg.
e74955
141
Protection against TGF-beta1-induced fibrosis effects of IL-10 on dermal fibroblasts and its potential therapeutics for the reduction of skin scarring
,
Arch Dermatol Res
,
2013
, vol.
305
(pg.
341
-
352
)
142
IL-10 treatment is associated with prohibitin expression in the Crohn's disease intestinal fibrosis mouse model
,
Med Inflamm
,
2013
, vol.
2013
pg.
617145
143
IL-10 inhibits inflammation and attenuates left ventricular remodeling after myocardial infarction via activation of STAT3 and suppression of HUR
,
Circ Res
,
2009
, vol.
104
(pg.
e9
-
18
)
144
Interleukin-10 treatment attenuates pressure overload-induced hypertrophic remodeling and improves heart function via signal transducers and activators of transcription 3-dependent inhibition of nuclear factor-κβ
,
Circulation
,
2012
, vol.
126
(pg.
418
-
429
)
145
New concepts of IL-10-induced lung fibrosis: fibrocyte recruitment and M2 activation in a CCL2/CCR2 axis
,
Am J Physiol Lung Cell Mol Physiol
,
2011
, vol.
300
(pg.
L341
-
L353
)
146
Simian immunodeficiency virus-induced lymphatic tissue fibrosis is mediated by transforming growth factor beta 1-positive regulatory T cells and begins in early infection
,
J Infect Dis
,
2007
, vol.
195
(pg.
551
-
561
)
147
B-lymphocyte contributions to human autoimmune disease
,
Immunol Rev
,
2008
, vol.
223
(pg.
284
-
299
)
148
Cytokine-producing b lymphocytes-key regulators of immunity
,
Curr Opin Immunol
,
2008
, vol.
20
(pg.
332
-
338
)
149
Attenuated liver fibrosis in the absence of B cells
,
J Clin Invest
,
2005
, vol.
115
(pg.
3072
-
3082
)
150
Elevated interleukin-6 expression in keloid fibroblasts
,
J Surg Res
,
2000
, vol.
89
(pg.
74
-
77
)
151
Autoantibodies to fibrillin-1 activate normal human fibroblasts in culture through the TGF-beta pathway to recapitulate the ‘scleroderma phenotype’
,
J Immunol
,
2005
, vol.
175
(pg.
4555
-
4560
)
152
B lymphocytes and B-cell activating factor promote collagen and profibrotic markers expression by dermal fibroblasts in systemic sclerosis
,
Arthritis Res Ther
,
2013
, vol.
15
pg.
R168
153
TNF-alpha-secreting B cells contribute to myocardial fibrosis in dilated cardiomyopathy
,
J Clin Immun
,
2013
, vol.
33
(pg.
1002
-
1008
)
154
A stromal address code defined by fibroblasts
,
Trends Immunol
,
2005
, vol.
26
(pg.
150
-
156
)
155
An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice
,
J Clin Invest
,
2009
, vol.
119
(pg.
3637
-
3651
)
156
The local physicochemical environment conditions the proinflammatory response of endothelial cells and thus modulates leukocyte recruitment
,
FEBS Lett
,
2004
, vol.
569
(pg.
13
-
17
)
157
A novel system for investigating the ability of smooth muscle cells and fibroblasts to regulate adhesion of flowing leukocytes to endothelial cells
,
J Immunol Methods
,
2001
, vol.
255
(pg.
73
-
82
)
158
Production of monocyte chemoattractant protein-1 and macrophage inflammatory protein-1 alpha by inflammatory granuloma fibroblasts
,
Am J Pathol
,
1994
, vol.
144
(pg.
711
-
718
)
159
Stimulus and cell-specific expression of C-X-C and C-C chemokines by pulmonary stromal cell populations
,
Am J Physiol
,
1995
, vol.
268
(pg.
L856
-
L861
)
160
Novel roles for chemokines and fibroblasts in interstitial fibrosis
,
Kidney Int
,
1998
, vol.
54
(pg.
2152
-
2159
)
161
Costimulation of fibroblast collagen and transforming growth factor beta1 gene expression by monocyte chemoattractant protein-1 via specific receptors
,
J Biol Chem
,
1996
, vol.
271
(pg.
17779
-
17784
)
162
Relb regulation of chemokine expression modulates local inflammation
,
Am J Pathol
,
1997
, vol.
151
(pg.
375
-
387
)
163
Macrophage/fibroblast coculture induces macrophage inflammatory protein-1alpha production mediated by intercellular adhesion molecule-1 and oxygen radicals
,
J Leukoc Biol
,
1998
, vol.
64
(pg.
636
-
641
)
164
The CCL2/CCR2 axis enhances vascular cell adhesion molecule-1 expression in human synovial fibroblasts
,
PLOS ONE
,
2012
, vol.
7
pg.
e49999
165
Fibroblasts from different sites may promote or inhibit recruitment of flowing lymphocytes by endothelial cells
,
Eur J Immunol
,
2009
, vol.
39
(pg.
113
-
125
)
166
Cardiac fibroblasts as a stimulator for cardiac inflammation
,
Cardiovasc Res
,
2012
, vol.
93
Suppl 1
pg.
S102
167
Fibroblasts as sentinel cells. Synthesis of chemokines and regulation of inflammation
,
Am J Pathol
,
1997
, vol.
151
(pg.
317
-
322
)
168
Ligation of CD40 on fibroblasts induces CD54 (ICAM-1) and CD106 (VCAM-1) up-regulation and IL-6 production and proliferation
,
J Leukoc Biol
,
1995
, vol.
58
(pg.
209
-
216
)
169
IFN-gamma regulates the expression of B7-H1 in dermal fibroblast cells
,
J Derm Sci
,
2005
, vol.
40
(pg.
95
-
103
)
170
Persistent induction of the chemokine receptor CXCR4 by TGF-beta 1 on synovial T cells contributes to their accumulation within the rheumatoid synovium
,
J Immunol
,
2000
, vol.
165
(pg.
3423
-
3429
)
171
Emergence of fibroblasts with a proinflammatory epigenetically altered phenotype in severe hypoxic pulmonary hypertension
,
J Immunol
,
2011
, vol.
187
(pg.
2711
-
2722
)
172
CXC chemokine receptor 4 expressed in T cells plays an important role in the development of collagen-induced arthritis
,
Arthritis Res Ther
,
2010
, vol.
12
pg.
R188
173
Regulatory T cells reduce acute lung injury fibroproliferation by decreasing fibrocyte recruitment
,
Am J Resp Cell Mol Biol
,
2013
, vol.
48
(pg.
35
-
43
)
174
The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities
,
Stem Cells
,
2007
, vol.
25
(pg.
1737
-
1745
)
175
Mesenchymal stem cells and inflammatory cardiomyopathy: cardiac homing and beyond
,
Cardiol Res Pract
,
2011
, vol.
2011
pg.
757154
176
Cardiac migration of mesenchymal stem cells in patients with inflammatory cardiomyopathy
,
Eur Heart J Suppl
,
2010
, vol.
31
(pg.
S464
-
S465
)
177
Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide
,
Cell Stem Cell
,
2008
, vol.
2
(pg.
141
-
150
)
178
Mesenchymal stem cells improve murine acute Coxsackievirus B3-induced myocarditis
,
Eur Heart J
,
2011
, vol.
32
(pg.
2168
-
2178
)
179
Mesenchymal stromal cells but not cardiac fibroblasts exert beneficial systemic immunomodulatory effects in experimental myocarditis
,
PLoS ONE
,
2012
, vol.
7
pg.
e41047
180
Mesenchymal stem cells promote matrix metalloproteinase secretion by cardiac fibroblasts and reduce cardiac ventricular fibrosis after myocardial infarction
,
Stem Cells
,
2009
, vol.
27
(pg.
2734
-
2743
)
181
Active movement of T cells away from a chemokine
,
Nat Med
,
2000
, vol.
6
(pg.
543
-
548
)
182
Inhibition of T cell apoptosis in the rheumatoid synovium
,
J Clin Invest
,
1997
, vol.
99
(pg.
439
-
446
)
183
Interferon-beta mediates stromal cell rescue of T cells from apoptosis
,
Eur J Immunol
,
1999
, vol.
29
(pg.
1041
-
1050
)
184
Coxsackievirus group B type 3 infection upregulates expression of monocyte chemoattractant protein 1 in cardiac myocytes, which leads to enhanced migration of mononuclear cells in viral myocarditis
,
J Virology
,
2004
, vol.
78
(pg.
12548
-
12556
)
185
Angiotensin-converting enzyme inhibition prevents the release of monocytes from their splenic reservoir in mice with myocardial infarction
,
Circ Res
,
2010
, vol.
107
(pg.
1364
-
1373
)
186
Monoclonal antibody therapy for prevention of acute Coxsackievirus B3 myocarditis in mice
,
Circulation
,
1989
, vol.
79
(pg.
1300
-
1308
)
187
Immunomodulatory effects of mesenchymal stromal cells revisited in the context of inflammatory cardiomyopathy
,
Stem Cells Int
,
2013
, vol.
2013
pg.
353097
188
Remodeling of the mononuclear phagocyte network underlies chronic inflammation and disease progression in heart failure: critical importance of the cardiosplenic axis
,
Circ Res
,
2014
, vol.
114
(pg.
266
-
282
)
189
Pressure overload-induced transient oxidative stress mediates perivascular inflammation and cardiac fibrosis through angiotensin ii
,
Hypertens Res
,
2006
, vol.
29
(pg.
711
-
718
)
190
Transforming growth factor-beta: governing the transition from inflammation to fibrosis in heart failure with preserved left ventricular function
,
Circ Heart Fail
,
2011
, vol.
4
(pg.
5
-
7
)
191
CTGF induces monocyte chemoattractant protein-1 expression to enhance monocyte migration in human synovial fibroblasts
,
Biochim Biophys Acta
,
2013
, vol.
1833
(pg.
1114
-
1124
)
192
A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation
,
J Am Coll Cardiol
,
2013
, vol.
62
(pg.
263
-
271
)
193
Diastolic heart failure: what we still don't know. Looking for new concepts, diagnostic approaches, and the role of comorbidities
,
Herz
,
2012
, vol.
37
(pg.
875
-
879
)
194
Therapeutic potential of tumour necrosis factor-alpha antagonists in patients with chronic heart failure
,
Heart Lung Circ
,
2013
, vol.
22
(pg.
323
-
327
)
195
Endomyocardial biopsy derived adherent proliferating cells—a potential cell source for cardiac tissue engineering
,
J Cell Biochem
,
2010
, vol.
109
(pg.
564
-
575
)
196
Immune attributes of cardiac-derived adherent proliferating (CAP) cells in cardiac therapy
,
J Tissue Eng Regen Med
,
2013
, vol.
7
(pg.
362
-
370
)
197
Human cardiac-derived adherent proliferating cells reduce murine acute Coxsackievirus B3-induced myocarditis
,
PLoS ONE
,
2011
, vol.
6
pg.
e28513
Author notes
This article is part of the Spotlight Issue on: Heterocellular signalling and crosstalk in the heart in ischaemia and heart failure.
Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2014. For permissions please email: journals.permissions@oup.com.
Topic:
Advertisement intended for healthcare professionals
Citations
Views
Altmetric
Metrics
Total Views 36,193
29,265 Pageviews
6,928 PDF Downloads
Since 1/1/2017
Month: | Total Views: |
---|---|
January 2017 | 91 |
February 2017 | 274 |
March 2017 | 162 |
April 2017 | 37 |
May 2017 | 45 |
June 2017 | 55 |
July 2017 | 80 |
August 2017 | 99 |
September 2017 | 100 |
October 2017 | 121 |
November 2017 | 150 |
December 2017 | 372 |
January 2018 | 299 |
February 2018 | 354 |
March 2018 | 471 |
April 2018 | 448 |
May 2018 | 377 |
June 2018 | 407 |
July 2018 | 347 |
August 2018 | 394 |
September 2018 | 405 |
October 2018 | 398 |
November 2018 | 461 |
December 2018 | 379 |
January 2019 | 469 |
February 2019 | 436 |
March 2019 | 472 |
April 2019 | 486 |
May 2019 | 478 |
June 2019 | 443 |
July 2019 | 422 |
August 2019 | 386 |
September 2019 | 375 |
October 2019 | 466 |
November 2019 | 448 |
December 2019 | 350 |
January 2020 | 470 |
February 2020 | 449 |
March 2020 | 450 |
April 2020 | 572 |
May 2020 | 351 |
June 2020 | 483 |
July 2020 | 504 |
August 2020 | 457 |
September 2020 | 496 |
October 2020 | 490 |
November 2020 | 490 |
December 2020 | 380 |
January 2021 | 485 |
February 2021 | 453 |
March 2021 | 487 |
April 2021 | 424 |
May 2021 | 628 |
June 2021 | 479 |
July 2021 | 453 |
August 2021 | 379 |
September 2021 | 464 |
October 2021 | 434 |
November 2021 | 439 |
December 2021 | 357 |
January 2022 | 442 |
February 2022 | 491 |
March 2022 | 481 |
April 2022 | 432 |
May 2022 | 459 |
June 2022 | 452 |
July 2022 | 360 |
August 2022 | 437 |
September 2022 | 359 |
October 2022 | 480 |
November 2022 | 408 |
December 2022 | 374 |
January 2023 | 476 |
February 2023 | 340 |
March 2023 | 434 |
April 2023 | 508 |
May 2023 | 473 |
June 2023 | 378 |
July 2023 | 399 |
August 2023 | 458 |
September 2023 | 423 |
October 2023 | 387 |
November 2023 | 399 |
December 2023 | 297 |
January 2024 | 358 |
February 2024 | 392 |
March 2024 | 493 |
April 2024 | 362 |
May 2024 | 372 |
June 2024 | 331 |
July 2024 | 212 |
August 2024 | 217 |
September 2024 | 248 |
October 2024 | 279 |
November 2024 | 151 |
Citations
396 Web of Science
×
Email alerts
Related articles in PubMed
Citing articles via
More from Oxford Academic
Advertisement intended for healthcare professionals