Abstract
The heart can respond to increased pathophysiological demand through alterations in tissue structure and function 1. This process, called cardiac remodeling, is particularly evident following myocardial infarction (MI), where the blockage of a coronary artery leads to widespread death of cardiac muscle. Following MI, necrotic tissue is replaced with extracellular matrix (ECM), and the remaining viable cardiomyocytes (CMs) undergo hypertrophic growth. ECM deposition and cardiac hypertrophy are thought to represent an adaptive response to increase structural integrity and prevent cardiac rupture. However, sustained ECM deposition leads to the formation of a fibrotic scar that impedes cardiac compliance and can induce lethal arrhythmias. Resident cardiac fibroblasts (CFs) are considered the primary source of ECM molecules such as collagens and fibronectin, particularly after becoming activated by pathologic signals. CFs contribute to multiple phases of post-MI heart repair and remodeling, including the initial response to CM death, immune cell (IC) recruitment, and fibrotic scar formation. The goal of this review is to describe how resident fibroblasts contribute to the healing and remodeling that occurs after MI, with an emphasis on how fibroblasts communicate with other cell types in the healing infarct scar 1–6.
Keywords: Fibrosis, fibroblast, ischemia, remodeling, heart
Introduction
The heart can respond to increased pathophysiological demand through alterations in tissue structure and function 1. This process, called cardiac remodeling, is particularly evident following myocardial infarction (MI), where the blockage of a coronary artery leads to widespread death of cardiac muscle. Following MI, necrotic tissue is replaced with extracellular matrix (ECM), and the remaining viable cardiomyocytes (CMs) undergo hypertrophic growth. ECM deposition and cardiac hypertrophy are thought to represent an adaptive response to increase structural integrity and prevent cardiac rupture. However, sustained ECM deposition leads to the formation of a fibrotic scar that impedes cardiac compliance and can induce lethal arrhythmias. Resident cardiac fibroblasts (CFs) are considered the primary source of ECM molecules such as collagens and fibronectin, particularly after becoming activated by pathologic signals. CFs contribute to multiple phases of post-MI heart repair and remodeling, including the initial response to CM death, immune cell (IC) recruitment, and fibrotic scar formation. The goal of this review is to describe how resident fibroblasts contribute to the healing and remodeling that occurs after MI, with an emphasis on how fibroblasts communicate with other cell types in the healing infarct scar 1–6.
Pathological and physiological cardiac remodeling
The heart is capable of remarkable phenotypic plasticity, which allows for the accommodation of alterations in pathophysiological demand. Increased demand can arise due to acute and chronic disease states (ischemic or non-ischemic pathological cardiac remodeling), or in response to pregnancy or extreme aerobic exercise (physiological cardiac remodeling) (see Fig. 1).
Figure 1: Stress-induced cardiovascular changes.
Physiological stress via exercise causes global hypertrophy that does not impede cardiovascular function or result in fibrosis. In exercise models, CM length and width is increased proportionally by longitudinal hypertrophic growth. Pathological stress can induce concentric hypertrophy where myocyte width increases greater than myocyte length (PO) or can induce eccentric hypertrophy (dilation) where myocytes are stretched, and their length increases much greater than their width (MI or HF). Healthy or exercised hearts lack fibrosis, but in PO models, interstitial and perivascular fibrosis occur throughout the heart while in MI models, large areas of CM death are repaired by replacement fibrosis. MI, myocardial infarction; PO, pressure overload.
Ischemic cardiac remodeling
Ischemic remodeling is often caused by a MI originating from an atherosclerotic plaque or thrombotic blockage in the coronary vasculature, which leads to localized ischemia and widespread CM death. Through well-classified phases of healing, the necrotic myocardial tissue within the infarct zone is replaced with a fibrotic scar called replacement fibrosis. Replacement fibrosis may provide structural support to the remaining healthy myocardium in the remote zone 7. Due to localized death of CMs, the development of a fibrotic infarct zone is associated with dilative remodeling and a decrease in heart function. ECM deposition results in stiffening of the ventricle and impedes pump function, leading to decreased heart function. Classical features of ischemic remodeling include the increase of CM length at the expense of width, resulting in thinned, elongated ventricles with larger surface area (termed eccentric remodeling) 7, 8. Development of cardiac arrhythmia, which is a common cause of sudden cardiac death, is also a consequence of replacement fibrosis due to loss of conductance of the electrical signals that govern proper heart contraction. The deleterious effects of this natural fibrotic remodeling response (which is strictly required to prevent rupture and maintain the structural integrity of the heart following the development of an infarct) dictate the necessity of developing strategies to ameliorate, but not eliminate, the fibrotic response to MI.
Ventricular pressure overload induced cardiac remodeling
Left ventricle pressure overload (PO) is a form of non-ischemic remodeling characterized by CM hypertrophy, loss in cardiac function, and development of fibrosis. PO is often initiated by chronic hypertension, in which increased systemic blood pressure generates a greater resistance to the flow of blood from the heart. In response to hypertension, left ventricle hypertrophic remodeling occurs to compensate for increased wall stress. Hypertrophy in response to PO causes CMs to disproportionately increase in width relative to length, resulting in a universal thickening of the cardiac wall (termed concentric remodeling) 8, 9. Increased pressure within coronary arteries also adds stress to surrounding myocardial tissue; therefore, fibrosis initially develops in patches in perivascular regions to increase wall support of large and small vessels. As PO increases and the CMs reach a maximum width-to-length ratio, CM apoptosis can occur that precipitates a more widespread replacement of myocardium with interstitial collagenous networks intended to strengthen the cardiac wall (termed interstitial fibrosis) 1, 6, 8, 9. Over time, the thickened, increasingly fibrotic myocardium is unable to relax and expand sufficiently to allow for proper diastolic filling, leading to decreased heart function in a similar fashion as described for MI. In contrast to MI, there is currently less evidence for distinct inflammatory and resolution phases in PO10, 11. Also, in contrast to MI, the development of early fibrosis is not localized, but diffused throughout the myocardium in predominantly vascular areas. Work from our group also showed that CFs from MI hearts and PO hearts have distinct genetic expression signatures not only from sedentary control animals, but also from each other - strongly suggesting that development of anti-fibrotic strategies in PO might take different forms than those most effective in MI 12, 13.
Physiological cardiac remodeling
In contrast to pathological cardiac remodeling, volume overload in physiological remodeling in response to exercise or pregnancy is generally correlated preservation of heart function, or even improved cardiac output 14, 15. To accommodate the increased demand for oxygen and nutrients typical in sustained endurance exercise, CM size is increased by longitudinal hypertrophic growth (increase in width-to-length ratio). While this can lead to concentric hypertrophy, physiological CM hypertrophy is not typically accompanied by fibrosis 8, 12, 16, 17, and the cardioprotective benefits of exercise are reported to ameliorate the progression of pathologic cardiac remodeling 18–22. Multiple studies have shown that chronic exercise induces transcriptional signatures in the myocardium that are consistent with improved cardiovascular function 23–26.
Timeline of ischemic cardiac remodeling
Pathological remodeling occurs as a continuum, which has been described in detail following MI (depicted in Fig. 2). The phases of post-MI cardiac repair are broadly defined by: 1) cell death and the initiation of an inflammatory response; 2) resolution of inflammation and stimulation of fibroblast proliferation; and 3) Scar formation and maturation. These major phases are comprised of various partially overlapping and interconnected processes, which will be the focus of the remainder of this review, with a focus on fibroblast contributions to the timing and extent of inflammation and scar formation in ischemic-injury.
Figure 2: Time-course of healing post-MI in mice.
Immediately following an occlusion of a coronary blood vessel, the Necrotic Phase occurs with downstream CM necrosis in response to lack of nutrients. Next, the Inflammatory Phase occurs with a surplus of invading ICs to clear cellular debris and promote pro-inflammatory signaling. During this phase, angiogenesis also begins. CFs begin to proliferate during the Reparative Phase, which also induces fibroblast activation and a shift from pro-inflammatory to pro-reparative IC interactions. Lastly, the Maturation Phase is identified by large amounts of ECM deposits, collagen cross-linking, and the transition of AFs to a matrifibrocyte state. AF, activated fibroblast; CF, cardiac fibroblast; CM, cardiomyocyte; EC, endothelial cell; ECM, extracellular matrix; IC, immune cell.
Defining the cardiac fibroblast
CFs originate from a population of progenitor cells that reside within the epicardium, a single cell layer of mesothelium that lines the heart 27. Epicardium-derived progenitor cells undergo epithelial-to-mesenchymal transition mid-gestation, and differentiate into various cardiac interstitial lineages, including CFs, which colonize the cardiac interstitium and provide structural support for the growing myocardium 28, 29. It is generally accepted that the primary cellular source of ECM in the heart is the resident cardiac fibroblast. Historically, the fibroblast response to cardiac insult has been described as a phenotypic switch, where quiescent resident fibroblasts “transform” into activated myofibroblasts that become contractile and secrete copious amounts of ECM. However, recent transcriptional analyses of non-CM cells in the interstitium of injured or uninjured hearts has identified various distinct CF subtypes and challenged the binary definition of fibroblasts as quiescent or activated. It is becoming more evident that fibroblasts may exist as a heterogeneous and fluid population of cells that contribute varied roles to post-MI injury repair 30–33. One recent study using single cell RNA-sequencing identified at least 10 CF populations, some of which expressed clearly identifiable gene expression profiles and some that were less distinct suggesting various intermediate phenotypes. This study described a population of collagen triple helix repeat containing 1 (Cthrc1)-expressing AF that emerges after MI, which the authors propose is especially important for the healing process 31. Another study using single cell RNA-sequencing identified 11 subpopulations of CFs; in addition to the standard “myofibroblast” definition, they identified 3 transcriptionally different populations (MYO-1, MYO-2, and MYO-3). MYO-2 and MYO-3 supported pro-fibrotic programs while MYO-1 was anti-fibrotic challenging the notion that all myofibroblasts promote pathological fibrosis 30. Finally, to expand on the heterogeneity of myofibroblasts, Ceccato et al. identified different secretory profiles from myofibroblasts depending on the method of activation (physical or biochemical) 32. In vivo, it’s likely that these signals, in addition to others, converge to result in various additional secretory profiles. For the simplicity of this review, we will identify injury-responsive fibroblasts as activated fibroblasts (AFs).
While no single marker is considered fibroblast-specific, CFs are defined by the expression of various ECM molecules including collagens and fibronectin, and the expression of cell surface markers, including platelet-derived growth factor alpha (See Table 1). Recent advances in genome editing have also enabled the generation of genetic lineage tracing strategies based upon the expression of Cre-recombinase from promoters that drive fibroblast-enriched gene expression. Several of the most efficacious lineage tracing strategies make use of Cre knockin at genes that are enriched in the epicardium that restrict to fibroblasts later in life, including at the Tcf21, Wilms Tumor protein 1 (Wt1), and Tbx18 alleles; additional transgenic strategies include Cre driven by a fragment of the Col1a2 promoter in all CFs, or the labeling of AFs using a tamoxifen inducible Postn-Cre mouse line 34–37. The identification of various CF-specific markers has significantly accelerated the study of CF contributions to fibrotic remodeling, as well as the understanding of the genetic processes that underlie fibroblast activation. We will now explore in more depth how CFs behave and respond to environmental ques throughout phases of healing post-MI.
Table 1:
Genetic markers of CF that have been used to restrict desired expression of lineage tracing reporters or create tissue-specific knockout/overexpression models. For a more comprehensive list of markers that is not limited to CF, the subject is thoroughly reviewed in 37.
| Marker | Name | Role |
|---|---|---|
| Col1a1 | Collagen type 1, alpha 1 | Structural component of type I collagen, major component of connective tissue and ECM 38. |
| Col1a2 | Collagen type 1, alpha 2 | Structural component of type I collagen, major component of connective tissue and ECM 39. |
| FSP-1 (S100A4) | S100 calcium-binding protein A4 | Associated with filaments, no longer considered specific for CF 40, 41. |
| PDGFRα (CD140a) | Platelet-derived growth factor receptor alpha | Receptor tyrosine kinase, transduces survival, proliferation, and migration responses. Expression in heart is specific to CF – high levels in quiescent CF, lower in AF 38. |
| Postn | Periostin | Matricellular molecule (see text). Expression in heart is specific to AF – very low levels or absent in quiescent CF 35. |
| Tbx18 | T-box transcription factor 18 | Transcription factor. Early marker of epicardium, Tbx18-expressing progenitor cells give rise to subset of CF as well as myocardial lineages 42, 43. |
| Tcf21 | Transcription factor 21 | Transcription factor, relevant to development in heart, lung, kidney, and spleen. Expression in heart is specific to CF – high levels in quiescent CF, lower in AF 27, 35, 44. |
| Tie2 (CD202b) | Angiopoietin-1 receptor, TEK | Tyrosine kinase receptor, also expressed on endothelial cells and monocytes. Tie2-expressing endocardial cells yield a specific, small subset of CF during development but do not contribute to remodeling in adult phases or injury 42. |
| Wt1 | Wilms’ tumor protein | Transcription factor. Regulates heart development and epithelial-mesenchymal transition of epicardial cells, some of which contribute to resident CF population 45. |
General cardiac fibroblast contributions to post-MI remodeling
In the adult heart, CFs exhibit considerable phenotypic plasticity, which allows for a regulated response to various pathophysiologic cues34, 36, 46, 47. Following MI, CFs deposit an ECM rich scar as part of the healing process and also express a number of secreted cytokines, chemokines, and growth factors 48, 49. While the canonical role of CFs remains the generation of ECM and subsequent contributions to scar formation and healing, fibroblasts have also been called sentinels of tissue injury due to a unique capacity to communicate with and alter the function of neighboring cells through both direct interactions and paracrine signaling (Fig. 3) 50.
Figure 3: Summary of CF interactions post-MI in mice.
Throughout the healing continuum post-MI, CFs interact with CMs, ECs, and ICs. In each phase of healing, different interactions become prominent that contribute to the microenvironment and signaling from CFs to other cardiac cells. During the necrotic and inflammatory phases, DAMPs that originate from dying CM signal the danger response to surrounding cells. Pro-inflammatory signaling is a major component of CFs and ICs interactions during the early phases of healing, and other pro-angiogenic and cell adhesion proteins are also secreted. During the proliferative and reparative phases, enriched gene expression programs include pro-resolution cytokines and matrix building molecules. Positive feedback from ECM tension and growth factors contributes to the progression of fibroblast activation and pathological fibrosis. Lastly, during the maturation phase, collagen cross-linking and modifying factors and chondrocyte factors are secreted from a specialized fibroblast called the matrifibrocyte. ANGPT1, angiopoietin 1; CCL5, C-C motif chemokine ligand 5; CM, cardiomyocyte; CHAD, chondroadherin; CLIP1, cartilage intermediate layer protein; COMP, cartilage oligomeric matrix protein; DAMPs, damage associated molecular patterns; EC, endothelial cell; ECM, extracellular matrix; FN, fibronectin; GM-CSF, granulocyte-macrophage colony-stimulating factor; IC, immune cell; IFNγ, interferon gamma; IL, interleukin; LOX, lysyl oxidase; MFGE8, milk fat globule epidermal growth factor 8; MIF, macrophage migration inhibitory factor; MMPs, matrix metalloproteinases; POSTN, periostin; TGFβ−1, transforming growth factor β−1; TIMPs, tissue inhibitors of MMPs; TNFα, tumor necrosis factor α; TSP, thrombospondin; VEGF, vascular endothelial growth factor.
Scar formation –
Increased levels of anti-inflammatory/pro-fibrotic factors such as TGF-β1, and changes in the biomechanical properties of the cardiac muscle, promote the activation of CFs into AFs, characterized by the robust expression of genes that encode contractile proteins such as (e.g. Acta2 and Tagln), and ECM components (e.g. Postn, Fn, and Col1a1).
Angiogenesis
Specifically, fibroblasts are reported to stimulate endothelial cells (ECs) to promote angiogenesis and revascularization through their secretions of angiopoietin 1 (ANGPT1) 51 and vascular endothelial growth factor (VEGF) 52. ANGPT1 has been reported as an essential factor in regulating the diameter of developing vessels 53. Mesenchymal-endothelial-transition (MEndoT) has also been described suggesting that CFs are active participants in neoangiogenesis, but their overall contribution may be limited 54, 55.
Immune cell polarization
ICs are recruited and polarized in the early phases of MI healing. Monocytes and macrophages respond to the initial ischemic injury by adopting a pro-inflammatory phenotype, sometimes referred to as M1 56. But as healing occurs, ICs can transition through a phagocytotic, proliferative phenotype and later adopt a pro-reparative profile (also called M2) that induces fibroblast activation via secretion of TGF-β1 56. These transitions can be triggered by cytokines (such as IL-1β) that are secreted by CFs, which are the predominant cell type responsible for activation of the inflammasome 57.
Immune resolution
CFs promote resolution of the immune response by secreting anti-inflammatory prostaglandins in the later phases of remodeling post-MI 58, 59. Additionally, increased secretions of IL-10 from CFs during the pro-resolving phase of MI healing can alter macrophage phenotypes to have a pro-resolution signature 60.
Timeline of CF response following MI
Necrotic Phase – Death Sets the Stage
Following occlusion of a coronary artery, the necrotic phase immediately commences as ischemic myocardium begins to die, and lasts about a day in mouse MI models 7. Loss of cardiomyocyte membrane integrity during cell starvation and death leads to release of cytoplasmic content, such as heat shock proteins, high mobility group box 1 protein (HMGB1), histones, S100 proteins, ATP, and mitochondrial DNA, into the interstitial space. The inappropriate localization of these intracellular molecules, or the secretion of certain ECM proteins such as tenascin C and Fibronectin (FN)-extra domain A (EDA), serve as danger signals that are collectively called damage associated molecular patterns (DAMPs) or alarmins (reviewed in 61). For example, HMGB1 has been shown to be released from the chromatin in cells undergoing necrotic cell death. Once released, extracellular HMGB1 interacts with antigen presenting cells via pattern recognition receptors (PRRs), such as dendritic cells, causing them to mature and promote sterile inflammation 62–64. PRRs are a diverse group recognizing many DAMP signaling molecules and encompassing Toll-like receptors (TLRs), nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain-containing proteins (NLRPs), P2X/P2Y purinoreceptors, and interleukin (IL) receptors, amongst others. These processes couple the necrotic death of CMs in response to infarction to the activation of the inflammasome, which is the multiprotein organization system responsible for the coordinated induction of the innate immune response. One of the major consequences of inflammasome activation is the proteolytic cleavage and secretion of ILs-1α/β and −18, which is associated with the initiation of the pro-inflammatory pyroptosis programmed cell death pathway 65–68. In the context of MI, this implies that DAMP release from damaged CMs propagates death signaling in neighboring cells via paracrine signals. Indeed, gene deletion and pharmacological inhibition studies generally indicate that induction of PRRs and the inflammasome leads to maladaptive remodeling after MI 69–71.
PRRs are also expressed on the surface of CF, which contribute to the pro-inflammatory response and the initiation of fibrosis downstream of DAMP-dependent signaling 72,73. Cleaved and activated IL-1α/β released from membrane pores in dying CMs interacts with and activates the extracellular domains of IL-1 receptors (IL-1Rs) on CF 74. Upon activation, IL-1Rs induce downstream myeloid differentiation primary response 88 (MyD88)-dependent signaling that in turn signal through the IL-1R-associated kinases (IRAKs) and leads to the activation of the transcription factor NF-κB. IL-1Rs are not the only proteins that make use of this signaling module – MyD88 is obligately used by virtually all PRRs in mice, though there is evidence that redundancy exists for MyD88 in the human immune response 75, 76. In addition to IL-1α/β, HMGB1, as mentioned above, is an example of a nuclear DAMP secreted from necrotic CMs that signals through PRRs. Both of these examples of CM DAMP secretion result in induction of chemokine synthesis in the CF via the activation of NF-κB-dependent transcription, shifting the CF from quiescence to active participation in the inflammatory phase, described in more detail below.
Inflammatory Phase – Recruitment and Cleanup
The inflammatory phase begins a day after the initial infarction and lasts until roughly day 4 7. At this point, the chance of survival for tissue experiencing prolonged oxygen and nutrient deprivation is minimal. Neutrophils are the first responders in the necrotic and early inflammatory phase, quickly infiltrating the infarct zone to clear cellular debris and propagate pro-inflammatory signals via degranulation, which releases appropriate chemical cues to promote the coming inflammation phase in a spatiotemporally controlled manner 5, 10, 77. The inflammatory phase is also characterized by the acute infiltration of pro-inflammatory monocytes and subsequent activation of tissue macrophages, which contribute to the clearance of cellular debris, as well as alerting cells in more remote areas, including CFs, to the presence of damage.
Once CFs develop a pro-inflammatory phenotype, they produce and secrete molecules that contribute to the pro-inflammatory microenvironment around the infarct. The post-MI secretome is comprised of cytokines such as interleukins (IL-1β, IL-6, IL-9, IL-11, and IL-12), tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and granulocyte-macrophage colony-stimulating factor (GM-CSF); as well as chemokines including the classically pro-inflammatory CCL2 and CCL5 78–82. All of these factors are reported to be upregulated in CF in the inflammatory response 34. In fact, autocrine feedback of IL-11 dependent signaling in CF is an essential step in the synthesis of matricellular proteins 80. Deletion of the IL-11 receptor (IL-11RA) in mice ameliorated fibrosis in hypertension and PO-induced cardiac remodeling; future studies are required to evaluate the efficacy of targeting IL-11 in ischemic remodeling. Additionally, macrophage migration inhibitory factor (MIF) has also been shown to be preferentially upregulated in and secreted from CFs in the ischemic heart 83. MIF is an inflammatory cytokine that interacts with the membrane-bound antigen-presenting protein CD74, triggering pro-survival and pro-inflammatory pathways that are in part mediated by ERK signaling84, 85. Interestingly, MIF can act via paracrine signaling to amplify MIF secretion rate in adjacent CFs, inducing the expression of genes encoding pro-adrenomedullin and CCL2 - thereby propagating CF-derived inflammation in addition to promoting angiogenesis and vasodilation via interaction with ECs 83.
Another example of a CF-secreted factor that acts preferentially on other CFs is milk fat globule epidermal growth factor 8 (MFGE8 or lactadherin). MFGE8 is a cell adhesion protein that was first identified as a connective factor in arteries between the smooth muscle cells and elastin 86. Known properties of MFGE8 signaling involve a phosphatidylserine-binding domain, which allows it to bind to and opsonize apoptotic cells, as well as an integrin-binding motif that facilitates angiogenesis 87, 88. MFGE8 was found to be induced in certain AF populations between 1 and 3 days post-MI, and was found to have a paracrine effect on surrounding CFs that significantly increased their native ability to perform apoptotic cell engulfment - normally the milieu of macrophages 89; however, considering the abundance of professional phagocytes in the infarcted heart, the relative contributions of cell engulfment by CFs is most likely minimal. This property of MFGE8 signaling between CFs is particularly notable, because it most likely helps the transition from the inflammatory phase into the pro-resolution phase through increasing cell engulfment and reducing inflammatory signals.
During the initial injury response, CFs have been shown to contribute to angiogenesis and neovascularization 54, 82. CFs at 3 days post-MI are reported to adopt a pro-angiogenic gene expression profile, including the significant upregulation of VEGF, which corresponds to the timing of neovascularization 82, 90. In addition to pro-angiogenic secreted factors at early timepoints following MI, CFs may also undergo MEndoT. At 3 days after ischemic injury, vascular identifiers (such as VECAD, eNOS, claudin-5, and occludin) are upregulated in a subset of CFs, and incorporation of Col1a2-lineage traced CFs into vascularization in the border zones was observed 54. However, the relative contribution of CFs into newly formed coronary arteries by MEndoT is debated, and more recent work has shown that the majority of new vessels are constructed from pre-existing endothelial cells 55.
Proliferative phase - Cardiac fibroblasts accumulate
The early phase of a cardiac insult is generally characterized by a robust increase in CF numbers. This proliferative phase overlaps with both the inflammatory and reparative phases of post-MI remodeling (described below) before the cell cycle slows13, 91. Interestingly, a recent study suggests that CF proliferation precedes and is distinct in time from fibroblast activation and subsequent ECM deposition 13, 92. Our recent work complements this finding, indicating that increased CF proliferation observed upon Sprr2b/MDM2 dependent p53 degradation does not drive the AF phenotype 13. Regardless, inhibition of CF proliferation in transgenic mice via cell-specific induction of Cdkn1a/p21 ameliorated scar formation following MI, likely by limiting the number of CFs that secrete ECM 93. A better understanding of the mechanisms that control the timing and extent of CF accumulation, and whether proliferation and fibroblast activation are coordinated through complementary signal transduction pathways, may uncover novel anti-fibrotic strategies.
Immune Resolution / Reparative Phase – Building a Scar
The immune resolution/reparative phase, identified by the presence of pro-resolution monocytes, peaks around 7 days post-MI and continues until about day 15 94. This phase is marked by the subsidence of pro-inflammatory factors and the emergence of anti-inflammatory, pro-reparative, and pro-fibrotic factors - many of the latter being matricellular proteins that are incorporated into the surrounding matrix to promote cardiac structural integrity. During the course of this phase, pro-inflammatory ICs are superceded by the emergence of pro-resolution ICs, while CFs transition into a contractile AF phenotype and begin to synthesize ECM in response to signaling cues from their environment (both directly and indirectly via interaction with other cell types). For instance, macrophages are an abundant source of anti-inflammatory, resolution-promoting cytokines such as IL-10. IL-10 infusion has been shown in vivo to have a beneficial effect on the post-MI remodeling process; however, it was also discovered that the effects IL-10 has on fibrosis are not from direct interactions, but rather from macrophages alternatively polarized by IL-10 that then interact with CFs 60.
CF activation
During the resolution phase, some of the most prominent factors that directly alter CF phenotype belong to the transforming growth factor beta (TGF-β) superfamily. TGF-β superfamily members are secreted from most cardiac cells, but the prominent source post-MI in mouse models has been found to be from macrophages 95. Interestingly, while members of the TGF-β superfamily (including TGF-β1/2/3, bone morphogenetic proteins or BMPs, growth differentiation factors or GDFs, activins, and inhibins) make use of myriad serine/threonine kinase receptors and adaptor signal transduction proteins, their signaling primarily converges downstream to activate SMAD proteins and translocate them to the nucleus. As a canonical example, extracellular TGF-β1 binds to TGFBR-II, which causes phosphorylation of the paired TGFBR-I. A zinc double finger FYVE domain-containing adaptor protein (e.g. SARA) then facilitates recruitment of one of the receptor-regulated SMADs (the specific species is ligand-dependent, and for TGF-β1 are SMAD2/3) to the activated TGFBR-I, which then phosphorylates the SMAD on a serine residue. Finally, this phosphorylated SMAD binds a co-SMAD (for TGF-β1, SMAD4), which moves to the nucleus to interact with DNA and influence the transcription of genes whose promoters contain SMAD-responsive elements. Several key factors in the transition from quiescent CF to AF phenotype are regulated by SMAD signaling: Acta2, Tgfb1, Pak2, Col1a1/1a2, Col3a1, Mmp3/14, Timp1, Rhoa, Serpine1, Itga1/2/5, Itgb3/8, Dsp, Scx (itself a transcriptional factor that interacts with and augments SMAD signaling), and Postn serve as salient examples of the diversity of SMAD signaling targets, which encompass (but are not limited to) intracellular kinases and cell cycle control proteins, secreted circulating factors, contractile and cytoskeletal proteins, cell-cell adhesion mediators, and ECM components and modulators 96–101. Speaking to this high complexity, recent efforts continue to focus on building a comprehensive database of SMAD signaling targets using high-throughput strategies 102.
While TGF-β superfamily/SMAD signaling is a major driver of the contractile and secretory AF phenotype, it is not the only transcriptional pathway governing this process. First, non-canonical TGF-β signaling contributes to the development of fibrosis after ischemic injury in mice, evidenced by the amelioration of scar formation in mice lacking p38 (mitogen-activated kinase 14) in fibroblasts 103. Permanent ligation of mice lacking p38 in fibroblasts led to 100% ventricular rupture, presumably due to insufficient replacement fibrosis. In addition to SMAD and p38 dependent signaling, TGF-β also induces the contractile phenotype via the activation of the serum response factor (SRF) pathway, which acts as a key determinant of differentiation and proliferation responses to stress stimuli 104, 105. A prime example of the cooperativity between TGF-β and biomechanical gene regulation is the Acta2 promoter, which contains both SMAD-binding and SRF-binding elements. Alterations in biomechanical tension that lead to filamentous (F) actin polymerization facilitates the nuclear accumulation of myocardin-related transcription factor (MRTF)-A, which interacts with SRF and promotes transcription of the contractile gene program, including Acta2 49, 106, 107. Therefore, the importance of MRTF-A is revealed by studies that demonstrate an abrogation of AFs and fibrosis following MI or in aging 106, 108, 109.
Collagen and elastin deposition
Accompanying the assumption of the contractile phenotype in AFs is a marked and sustained increase in transcription of genes coding for collagens, other ECM-associated structural proteins, matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs). The synthesis of new matrix materials can be attributed primarily to AFs during the resolution phase, with limited evidence that other cell types are major sources of ECM (covered in more detail later) 7, 42, 101. Collagens, representing at least 42 genes coding for 28 protein products, represent possibly the best-known and most diverse family of ECM protein products synthesized by AFs 110. Broadly, collagens can be classified as fibrillar or non-fibrillar based on their molecular structure. Fibrillar collagens, such as the ubiquitous COL1A1, are associated with integumentary, skeletal, and organ structure, while non-fibrillar collagens tend to provide parallel structural support to fibrillary collagens or (in the case of COL4A) are synthesized in sheet structures to create the foundation for the basal lamina 111. Fibrillar collagens, which make up at least 90% of the total collagen mass in mammals, are initially synthesized as single strands that self-assemble into triple helical structures termed procollagens 112. Procollagens are then hydroxylated on proline and lysine residues in a vitamin C-dependent manner by prolyl-4-hydroxylases (P4HA/B) or lysyl oxidase (LOX), either intracellularly or extracellularly, and subsequent processing steps lead to the formation of the mature collagen fibril 113–115. The measurement of collagen (or its characteristic component hydroxyproline) has long been used as a surrogate for the degree of fibrotic remodeling in the heart, and circulating collagen type I and III propeptides have been shown to be a biomarker of multiple types of cardiac pathology in both mice and human patients 116–119. Elastin (ELN) is a highly conserved ECM protein that is in fact absent from MI scar tissue in either its molecular or compound elastic fiber format, but has been shown to be advantageous to ventricular functional recovery post-MI in a rat model if introduced via gene therapy – likely via increasing the elasticity of the rigid MI scar and improving mechanical properties 120–122.
Proteoglycans
While collagens certainly are the main component of the AF-derived ECM, other proteins and macromolecules synthesized by AFs are equally important to ECM biology and the biology of a scar during post-MI remodeling. Proteoglycans such as heparan sulfate are also crucial to the post-MI scar formation process, as well as ventricular functional recovery 123. Heparan sulfate chains bind angiogenic growth factors present in the ECM such as VEGF-A, and their ability to retain these factors is governed at least in part by a pair of endosulfatases (Sulf1/2) that regulate how much VEGF-A is available in the ECM to promote angiogenesis – a crucial step in the revascularization and recovery of the post-MI myocardium 124. Interestingly, this study showed AFs preferentially expressed Sulf1 – implicating AFs not only in heparan sulfate synthesis, but also in its post-translational regulation. Another chief component of the cardiac ECM synthesized by AFs is the glycosaminoglycan hyaluronic acid, which is the product of three hyaluronan synthases (Has1/2/3) located on the cell membrane 125. Hyaluronic acid is synthesized in a wide variety of molecular sizes, which have been demonstrated to have diverse properties and participate in both inflammation and resolution – as well as aiding in the recovery from MI both natively and when implanted as a biomaterial scaffold 126–130. It is interesting to speculate that strategies to promote or recapitulate the native ability of CFs to synthesize larger amounts of these potentially protective matrix molecules may be cardioprotective in the context of MI.
Other matricellular proteins
CFs and other cell types also interact with the ECM via myriad non-collagenous matricellular proteins - of which fibronectin (FN), periostin (POSTN), and thrombospondins (TSPs) are salient examples in MI biology. FN is a large glycoprotein that interacts specifically with membrane-bound integrins as well as collagen and heparan sulfate 131. FN has widespread contributions within cardiac ECM: integrin binding (with the classical example being α5β1, also termed the FN receptor), collagen binding, heparin binding, and interaction with fibrin (a major initial component of the MI scar) 132, 133. FN is found in one of three isoforms based on alternative splicing, and the extra domain A/B-containing isoforms that predominate during fetal development are re-expressed following MI 134–137. Interestingly, FN extra domain A (EDA) is thought to localize TGF-β by acting with the latent TGF-β-binding protein-1 (Ltbp1) in vitro 138. FN-EDA has been shown to stimulate fibroblast activation and collagen production in cutaneous fibrosis 139, and genetic deletion of FN-EDA or pharmacological inhibition of FN polymerization, reduces ischemia-reperfusion injury 140–142.
Another matrix molecule with similarly complicated biology in the heart is the matricellular protein, POSTN, which has been shown to be highly expressed in the embryonic mouse heart but is largely absent from the uninjured post-natal myocardium (reviewed in depth in 143). POSTN has a number of interacting roles in the heart that recall those played by FN: namely integrin binding (primarily avb3 and avb5, the canonical vitronectin receptors), fibrillary collagen binding, and FN binding – consistent with a role in promotion of cell motility by coupling intracellular integrin signaling to changes in the ECM 144, 145. Postn is perhaps best known in recent literature as a reliable, specific marker of AFs – adult resident quiescent CFs express virtually no Postn, but post-injury the CF to AF transition is associated with a significant and persistent increase in Postn expression, which is recapitulated in a transgenic mouse line expressing tamoxifen-inducible Cre recombinase at the Postn locus 12, 13, 35, 92. In the mouse MI model, gain of function in Postn did not cause noticeable increases in fibrosis, but Postn-knockout mice were more likely to experience cardiac rupture due to inferior scar formation during the resolution period 146–148. Although Postn is a recently well-accepted marker of AFs, literature has suggested that it may only be expressed by a subset of CFs marked by Tcf21 expression 82. Additionally, POSTN could certainly be derived from other cell types after cardiac insult, such as smooth muscle cells or macrophages 56, 82.
The final family of non-collagenous matricellular proteins to be discussed here are the TSPs, a five-member family of matrix-binding proteins, which have been thoroughly reviewed elsewhere 149,9, 143, 150. While TSP-1/2 have been shown in multiple studies to discourage angiogenesis, more recently TSP-4 has been implicated in the promotion of neovascularization 151–154. TSP-4 was also shown to directly protect the heart by promoting the nuclear accumulation of ATF6α, an ER stress response transcription factor 155. Furthermore, TSP-3 (once thought to be absent from the heart) was recently found to destabilize sarcolemmal structure and inhibit integrin attachment complex formation due to a difference in molecular structure from the closely related TSPs-4/5 156. Interestingly, by introducing a mutation to incorporate the integrin-binding domain contained in TSPs-4/5 into the TSP-3 protein, the negative effect of TSP-3 during myocardial remodeling was completely reversed. Based on these studies, the TSP family is a particularly attractive candidate for targeted therapies due to the diverse roles played in the post-MI heart.
Matrix metalloproteinases
Discussion of the remodeling ECM is not limited to the components of its synthesis – the ECM, particularly in the response to cardiac insult, is a dynamic environment that is constantly being synthesized and degraded. To this end, one of the most relevant families of matrix-modifying proteins are the MMPs, a large group of calcium-dependent zinc-incorporating enzymes that were first characterized as degrading agents of ECM molecules but have also been implicated in a number of other cellular functions such as cytokine processing and inactivation as well as receptor cleavage 157, 158. MMPs are initially synthesized in a latent form, but are activated by cleavage due to other proteases (including MMPs), by structural changes in the pro-domain, or by reactive oxygen species altering the cysteine residue that maintains the latent configuration 159, 160. As befits such a diverse family of proteins, roles played by MMPs are context-dependent and are not limited to the extracellular space 161. As reviewed recently, MMPs serve in both their native enzymatic capacity and as serum biomarkers in heart failure patients 162. MMPs are primarily responsible for degradation of the ECM, which in MI has been shown to promote chamber dilation 163, 164. Of note, the well-characterized MMPs-2 and −9 (which degrade both gelatinous and fibrillary collagens) have clinically relevant polymorphisms that are associated with poor cardiovascular outcomes across a variety of ethnic backgrounds – suggesting a central role in cardiovascular health 165–173. These proteins are synthesized by both CFs and macrophages – and in at least one study, constitutive production of macrophage MMP-9 improved outcomes for aged mice after MI, making their role in remodeling potentially even more complex 174. Closely related to these proteins are the four TIMPs, which are produced by both CFs and CMs and act to stabilize MMP pro-forms and limit MMP activity 175. TIMP expression has been shown to be generally decreased in the failing heart, in direct contrast to the increased prevalence of MMPs 176. At least one study has also shown non-MMP-related effects of TIMPs in the rat MI model –TIMPs-1 and −3 were found to promote beneficial remodeling post-MI and also were associated with increased CM resistance to apoptosis both in vivo and in vitro 177. This evidence suggests that balancing MMP and TIMP expression in the post-MI heart might be a key to translational development of therapies that target these ECM-modifying molecules.
Maturation Phase – Maintaining and Modifying a Scar
Once the scar has been synthesized during the pro-reparative phase, it begins a long process of dynamic remodeling known as maturation, which is a matter of weeks in small animal models and months in humans 178–181. This scar is distinct from the early fibrous scar in its complexity – LOX facilitates collagen cross-linking, and collagen-binding factors such as decorin and perlecan reinforce the scar and alter its mechanical properties on both micro and macro scales 2, 182–186. Changes occur in resident cell types as well; the AFs that have synthesized the scar now enter a phase marked by limited apoptosis, increased senescence, and irreversible exit from the cell cycle 92, 187–189. Lineage tracing in a recent study showed that AFs residing in the maturing scar lose Acta2 expression as well as the ability to divide, resulting in a distinct state that the authors termed the ‘matrifibrocyte’ 92. These cells retained a number of lineage-specific fibroblast genes but gained expression of bone/cartilage markers (cartilage oligomeric matrix protein – Comp or Tsp5, chondroadherin – Chad, and Cartilage intermediate layer protein 1 – Cilp1) and apoptosis resistance genes, while silencing the expression of Col1 and Col3, and some matrix remodeling factors (which peak during the resolution/reparative phases). Surviving CMs have been shown to maintain a network of connections that allow for limited electrical conduction, even in the infarcted zone 190, and electrical coupling is also accomplished via fibroblasts in the scar 191–194. Over time, infarct thinning and compaction, in which the infarct responds to mechanical stress by altering its thickness, has been noted in both animal models and in human patients, suggesting dynamic alterations in scar properties long after the initial insult 195–200.
Conclusions:
The CF has recently emerged as one of the most phenotypically plastic cells in the heart, with distinct transcriptional and secretory programs that regulate their essential contributions to pathophysiological cardiac remodeling. It is now evident that CFs are not inactive bystanders, but are dynamic contributors to the divergent responses that characterize cardiac adaptations following MI. The enigmatic and adaptable nature of CFs, once an impediment to translational research studies, may now prove to be a powerful target in the development of therapies aimed at limiting fibrosis and inflammation, which underlie pathologic remodeling in the failing heart.
Highlights.
Ischemic cardiac tissue is replaced with a fibrotic scar
The classical role of resident cardiac fibroblasts is the deposition of scar tissue
Fibroblasts play a wide variety of roles in ischemic cardiac remodeling
Fibroblasts interact with other cardiac cell types in the ischemic heart
Fibroblasts contribute to each of the various phases of the injury response
Acknowledgements
Generous grant funding was provided to EMS (NIHR01-HL133761, NIHR01-HL136179, NIHR01-HL144867, NIHUL1-TR002001, NYSTEM-C32566GG); and RMB (F32HL136066 and T32HL007937-15).
Footnotes
Disclosures
None
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Hill JA and Olson EN. Cardiac plasticity. N Engl J Med. 2008;358:1370–80. [DOI] [PubMed] [Google Scholar]
- 2.Dobaczewski M, Gonzalez-Quesada C and Frangogiannis NG. The extracellular matrix as a modulator of the inflammatory and reparative response following myocardial infarction. J Mol Cell Cardiol. 2010;48:504–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Frangogiannis NG. Regulation of the inflammatory response in cardiac repair. Circ Res. 2012;110:159–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. 2014;11:255–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med. 2019;65:70–99. [DOI] [PubMed] [Google Scholar]
- 6.Frey N and Olson EN. Cardiac hypertrophy: the good, the bad, and the ugly. Annu Rev Physiol. 2003;65:45–79. [DOI] [PubMed] [Google Scholar]
- 7.Frangogiannis NG. Pathophysiology of Myocardial Infarction. Compr Physiol. 2015;5:1841–75. [DOI] [PubMed] [Google Scholar]
- 8.Heineke J and Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600. [DOI] [PubMed] [Google Scholar]
- 9.Frangogiannis NG. The Extracellular Matrix in Ischemic and Nonischemic Heart Failure. Circ Res. 2019;125:117–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Carrillo-Salinas FJ, Ngwenyama N, Anastasiou M, Kaur K and Alcaide P. Heart Inflammation: Immune Cell Roles and Roads to the Heart. Am J Pathol. 2019;189:1482–1494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Blanton RM, Carrillo-Salinas FJ and Alcaide P. T-cell recruitment to the heart: friendly guests or unwelcome visitors? Am J Physiol Heart Circ Physiol. 2019;317:H124–H140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lighthouse JK, Burke RM, Velasquez LS, Dirkx RA Jr., Aiezza A 2nd, Moravec CS, Alexis JD, Rosenberg A and Small EM. Exercise promotes a cardioprotective gene program in resident cardiac fibroblasts. JCI Insight. 2019;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Burke RM, Lighthouse JK, Quijada P, Dirkx RA Jr., Rosenberg A, Moravec CS, Alexis JD and Small EM. Small proline-rich protein 2B drives stress-dependent p53 degradation and fibroblast proliferation in heart failure. Proc Natl Acad Sci U S A. 2018;115:E3436–E3445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chung E and Leinwand LA. Pregnancy as a cardiac stress model. Cardiovasc Res. 2014;101:561–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kaplan ML, Cheslow Y, Vikstrom K, Malhotra A, Geenen DL, Nakouzi A, Leinwand LA and Buttrick PM. Cardiac adaptations to chronic exercise in mice. Am J Physiol. 1994;267:H1167–73. [DOI] [PubMed] [Google Scholar]
- 16.McMullen JR, Amirahmadi F, Woodcock EA, Schinke-Braun M, Bouwman RD, Hewitt KA, Mollica JP, Zhang L, Zhang Y, Shioi T, Buerger A, Izumo S, Jay PY and Jennings GL. Protective effects of exercise and phosphoinositide 3-kinase(p110alpha) signaling in dilated and hypertrophic cardiomyopathy. Proc Natl Acad Sci U S A. 2007;104:612–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tham YK, Bernardo BC, Ooi JY, Weeks KL and McMullen JR. Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets. Arch Toxicol. 2015;89:1401–38. [DOI] [PubMed] [Google Scholar]
- 18.Roh JD, Houstis N, Yu A, Chang B, Yeri A, Li H, Hobson R, Lerchenmuller C, Vujic A, Chaudhari V, Damilano F, Platt C, Zlotoff D, Lee RT, Shah R, Jerosch-Herold M and Rosenzweig A. Exercise training reverses cardiac aging phenotypes associated with heart failure with preserved ejection fraction in male mice. Aging Cell. 2020;19:e13159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bezzerides VJ, Platt C, Lerchenmuller C, Paruchuri K, Oh NL, Xiao C, Cao Y, Mann N, Spiegelman BM and Rosenzweig A. CITED4 induces physiologic hypertrophy and promotes functional recovery after ischemic injury. JCI Insight. 2016;1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Konhilas JP, Watson PA, Maass A, Boucek DM, Horn T, Stauffer BL, Luckey SW, Rosenberg P and Leinwand LA. Exercise can prevent and reverse the severity of hypertrophic cardiomyopathy. Circ Res. 2006;98:540–8. [DOI] [PubMed] [Google Scholar]
- 21.Howden EJ, Sarma S, Lawley JS, Opondo M, Cornwell W, Stoller D, Urey MA, Adams-Huet B and Levine BD. Reversing the Cardiac Effects of Sedentary Aging in Middle Age-A Randomized Controlled Trial: Implications For Heart Failure Prevention. Circulation. 2018;137:1549–1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lavie CJ, Ozemek C, Carbone S, Katzmarzyk PT and Blair SN. Sedentary Behavior, Exercise, and Cardiovascular Health. Circ Res. 2019;124:799–815. [DOI] [PubMed] [Google Scholar]
- 23.Lerchenmuller C, Rabolli CP, Yeri A, Kitchen R, Salvador AM, Liu LX, Ziegler O, Danielson K, Platt C, Shah R, Damilano F, Kundu P, Riechert E, Katus HA, Saffitz JE, Keshishian H, Carr SA, Bezzerides VJ, Das S and Rosenzweig A. CITED4 Protects Against Adverse Remodeling in Response to Physiological and Pathological Stress. Circ Res. 2020;127:631–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Liu X, Xiao J, Zhu H, Wei X, Platt C, Damilano F, Xiao C, Bezzerides V, Bostrom P, Che L, Zhang C, Spiegelman BM and Rosenzweig A. miR-222 is necessary for exercise-induced cardiac growth and protects against pathological cardiac remodeling. Cell Metab. 2015;21:584–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bostrom P, Mann N, Wu J, Quintero PA, Plovie ER, Panakova D, Gupta RK, Xiao C, MacRae CA, Rosenzweig A and Spiegelman BM. C/EBPbeta controls exercise-induced cardiac growth and protects against pathological cardiac remodeling. Cell. 2010;143:1072–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chung E, Heimiller J and Leinwand LA. Distinct cardiac transcriptional profiles defining pregnancy and exercise. PLoS One. 2012;7:e42297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Acharya A, Baek ST, Huang G, Eskiocak B, Goetsch S, Sung CY, Banfi S, Sauer MF, Olsen GS, Duffield JS, Olson EN and Tallquist MD. The bHLH transcription factor Tcf21 is required for lineage-specific EMT of cardiac fibroblast progenitors. Development. 2012;139:2139–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Quijada P, Trembley MA and Small EM. The Role of the Epicardium During Heart Development and Repair. Circ Res. 2020;126:377–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Trembley MA, Velasquez LS, de Mesy Bentley KL and Small EM. Myocardin-related transcription factors control the motility of epicardium-derived cells and the maturation of coronary vessels. Development. 2015;142:21–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Farbehi N, Patrick R, Dorison A, Xaymardan M, Janbandhu V, Wystub-Lis K, Ho JW, Nordon RE and Harvey RP. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. Elife. 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ruiz-Villalba A, Romero JP, Hernandez SC, Vilas-Zornoza A, Fortelny N, Castro-Labrador L, San Martin-Uriz P, Lorenzo-Vivas E, Garcia-Olloqui P, Palacios M, Gavira JJ, Bastarrika G, Janssens S, Wu M, Iglesias E, Abizanda G, Martinez de Morentin X, Lasaga M, Planell N, Bock C, Alignani D, Medal G, Pelacho B, Prudovsky I, Jin YR, Ryzhov S, Yin H, Gomez-Cabrero D, Lindner V, Lara-Astiaso D and Prosper F. Single-Cell RNA-seq Analysis Reveals a Crucial Role for Collagen Triple Helix Repeat Containing 1 (CTHRC1) Cardiac Fibroblasts after Myocardial Infarction. Circulation. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ceccato TL, Starbuck RB, Hall JK, Walker CJ, Brown TE, Killgore JP, Anseth KS and Leinwand LA. Defining the Cardiac Fibroblast Secretome in a Fibrotic Microenvironment. J Am Heart Assoc. 2020;9:e017025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kretzschmar K, Post Y, Bannier-Helaouet M, Mattiotti A, Drost J, Basak O, Li VSW, van den Born M, Gunst QD, Versteeg D, Kooijman L, van der Elst S, van Es JH, van Rooij E, van den Hoff MJB and Clevers H. Profiling proliferative cells and their progeny in damaged murine hearts. Proc Natl Acad Sci U S A. 2018;115:E12245–E12254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tallquist MD. Cardiac Fibroblast Diversity. Annu Rev Physiol. 2020;82:63–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kanisicak O, Khalil H, Ivey MJ, Karch J, Maliken BD, Correll RN, Brody MJ, SC JL, Aronow BJ, Tallquist MD and Molkentin JD. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat Commun. 2016;7:12260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tallquist MD and Molkentin JD. Redefining the identity of cardiac fibroblasts. Nat Rev Cardiol. 2017;14:484–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Swonger JM, Liu JS, Ivey MJ and Tallquist MD. Genetic tools for identifying and manipulating fibroblasts in the mouse. Differentiation. 2016;92:66–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’Antoni ML, Debuque R, Chandran A, Wang L, Arora K, Rosenthal NA and Tallquist MD. Revisiting Cardiac Cellular Composition. Circ Res. 2016;118:400–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Florin L, Alter H, Grone HJ, Szabowski A, Schutz G and Angel P. Cre recombinase-mediated gene targeting of mesenchymal cells. Genesis. 2004;38:139–44. [DOI] [PubMed] [Google Scholar]
- 40.Kong P, Christia P, Saxena A, Su Y and Frangogiannis NG. Lack of specificity of fibroblast-specific protein 1 in cardiac remodeling and fibrosis. Am J Physiol Heart Circ Physiol. 2013;305:H1363–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Sayegh MH, Izumo S and Kalluri R. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007;13:952–61. [DOI] [PubMed] [Google Scholar]
- 42.Moore-Morris T, Guimaraes-Camboa N, Banerjee I, Zambon AC, Kisseleva T, Velayoudon A, Stallcup WB, Gu Y, Dalton ND, Cedenilla M, Gomez-Amaro R, Zhou B, Brenner DA, Peterson KL, Chen J and Evans SM. Resident fibroblast lineages mediate pressure overload-induced cardiac fibrosis. J Clin Invest. 2014;124:2921–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cai CL, Martin JC, Sun Y, Cui L, Wang L, Ouyang K, Yang L, Bu L, Liang X, Zhang X, Stallcup WB, Denton CP, McCulloch A, Chen J and Evans SM. A myocardial lineage derives from Tbx18 epicardial cells. Nature. 2008;454:104–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Acharya A, Baek ST, Banfi S, Eskiocak B and Tallquist MD. Efficient inducible Cre-mediated recombination in Tcf21 cell lineages in the heart and kidney. Genesis. 2011;49:870–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.del Monte G, Casanova JC, Guadix JA, MacGrogan D, Burch JB, Perez-Pomares JM and de la Pompa JL. Differential Notch signaling in the epicardium is required for cardiac inflow development and coronary vessel morphogenesis. Circ Res. 2011;108:824–36. [DOI] [PubMed] [Google Scholar]
- 46.Humeres C and Frangogiannis NG. Fibroblasts in the Infarcted, Remodeling, and Failing Heart. JACC Basic Transl Sci. 2019;4:449–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.DeLeon-Pennell KY, Barker TH and Lindsey ML. Fibroblasts: The arbiters of extracellular matrix remodeling. Matrix Biol. 2020;91–92:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C and Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002;3:349–63. [DOI] [PubMed] [Google Scholar]
- 49.Tomasek JJ, McRae J, Owens GK and Haaksma CJ. Regulation of alpha-smooth muscle actin expression in granulation tissue myofibroblasts is dependent on the intronic CArG element and the transforming growth factor-beta1 control element. Am J Pathol. 2005;166:1343–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Daseke MJ 2nd, Tenkorang MAA, Chalise U, Konfrst SR and Lindsey ML. Cardiac fibroblast activation during myocardial infarction wound healing: Fibroblast polarization after MI. Matrix Biol. 2020;91–92:109–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hurley JR, Balaji S and Narmoneva DA. Complex temporal regulation of capillary morphogenesis by fibroblasts. Am J Physiol Cell Physiol. 2010;299:C444–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Chintalgattu V, Nair DM and Katwa LC. Cardiac myofibroblasts: a novel source of vascular endothelial growth factor (VEGF) and its receptors Flt-1 and KDR. J Mol Cell Cardiol. 2003;35:277–86. [DOI] [PubMed] [Google Scholar]
- 53.Jeansson M, Gawlik A, Anderson G, Li C, Kerjaschki D, Henkelman M and Quaggin SE. Angiopoietin-1 is essential in mouse vasculature during development and in response to injury. J Clin Invest. 2011;121:2278–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ubil E, Duan J, Pillai IC, Rosa-Garrido M, Wu Y, Bargiacchi F, Lu Y, Stanbouly S, Huang J, Rojas M, Vondriska TM, Stefani E and Deb A. Mesenchymal-endothelial transition contributes to cardiac neovascularization. Nature. 2014;514:585–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.He L, Huang X, Kanisicak O, Li Y, Wang Y, Li Y, Pu W, Liu Q, Zhang H, Tian X, Zhao H, Liu X, Zhang S, Nie Y, Hu S, Miao X, Wang QD, Wang F, Chen T, Xu Q, Lui KO, Molkentin JD and Zhou B. Preexisting endothelial cells mediate cardiac neovascularization after injury. J Clin Invest. 2017;127:2968–2981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mouton AJ, DeLeon-Pennell KY, Rivera Gonzalez OJ, Flynn ER, Freeman TC, Saucerman JJ, Garrett MR, Ma Y, Harmancey R and Lindsey ML. Mapping macrophage polarization over the myocardial infarction time continuum. Basic Res Cardiol. 2018;113:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kuppers R, Fischer U, Rajewsky K and Gause A. Immunoglobulin heavy and light chain gene sequences of a human CD5 positive immunocytoma and sequences of four novel VHIII germline genes. Immunol Lett. 1992;34:57–62. [DOI] [PubMed] [Google Scholar]
- 58.Lacy SH, Woeller CF, Thatcher TH, Pollock SJ, Small EM, Sime PJ and Phipps RP. Activated Human Lung Fibroblasts Produce Extracellular Vesicles with Antifibrotic Prostaglandins. Am J Respir Cell Mol Biol. 2019;60:269–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lacy SH, Woeller CF, Thatcher TH, Maddipati KR, Honn KV, Sime PJ and Phipps RP. Human lung fibroblasts produce proresolving peroxisome proliferator-activated receptor-gamma ligands in a cyclooxygenase-2-dependent manner. Am J Physiol Lung Cell Mol Physiol. 2016;311:L855–L867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jung M, Ma Y, Iyer RP, DeLeon-Pennell KY, Yabluchanskiy A, Garrett MR and Lindsey ML. IL-10 improves cardiac remodeling after myocardial infarction by stimulating M2 macrophage polarization and fibroblast activation. Basic Res Cardiol. 2017;112:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Turner NA. Inflammatory and fibrotic responses of cardiac fibroblasts to myocardial damage associated molecular patterns (DAMPs). J Mol Cell Cardiol. 2016;94:189–200. [DOI] [PubMed] [Google Scholar]
- 62.Scaffidi P, Misteli T and Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–5. [DOI] [PubMed] [Google Scholar]
- 63.Rovere-Querini P, Capobianco A, Scaffidi P, Valentinis B, Catalanotti F, Giazzon M, Dumitriu IE, Muller S, Iannacone M, Traversari C, Bianchi ME and Manfredi AA. HMGB1 is an endogenous immune adjuvant released by necrotic cells. EMBO Rep. 2004;5:825–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, Tracey KJ and Chiorazzi N. High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol. 2004;173:307–13. [DOI] [PubMed] [Google Scholar]
- 65.LaRock DL, Sands JS, Ettouati E, Richard M, Bushway PJ, Adler ED, Nizet V and LaRock CN. Inflammasome inhibition blocks cardiac glycoside cell toxicity. J Biol Chem. 2019;294:12846–12854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhou W, Chen C, Chen Z, Liu L, Jiang J, Wu Z, Zhao M and Chen Y. NLRP3: A Novel Mediator in Cardiovascular Disease. J Immunol Res. 2018;2018:5702103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, Hongo M, Noda T, Nakayama J, Sagara J, Taniguchi S and Ikeda U. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123:594–604. [DOI] [PubMed] [Google Scholar]
- 68.Toldo S, Mauro AG, Cutter Z and Abbate A. Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2018;315:H1553–H1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wiratkapun C, Piyapan P, Lertsithichai P and Larbcharoensub N. Fibroadenoma versus phyllodes tumor: distinguishing factors in patients diagnosed with fibroepithelial lesions after a core needle biopsy. Diagn Interv Radiol. 2014;20:27–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mezzaroma E, Toldo S, Farkas D, Seropian IM, Van Tassell BW, Salloum FN, Kannan HR, Menna AC, Voelkel NF and Abbate A. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc Natl Acad Sci U S A. 2011;108:19725–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Epelman S, Liu PP and Mann DL. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat Rev Immunol. 2015;15:117–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhang W, Lavine KJ, Epelman S, Evans SA, Weinheimer CJ, Barger PM and Mann DL. Necrotic myocardial cells release damage-associated molecular patterns that provoke fibroblast activation in vitro and trigger myocardial inflammation and fibrosis in vivo. J Am Heart Assoc. 2015;4:e001993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Saxena A, Chen W, Su Y, Rai V, Uche OU, Li N and Frangogiannis NG. IL-1 induces proinflammatory leukocyte infiltration and regulates fibroblast phenotype in the infarcted myocardium. J Immunol. 2013;191:4838–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bageghni SA, Hemmings KE, Yuldasheva NY, Maqbool A, Gamboa-Esteves FO, Humphreys NE, Jackson MS, Denton CP, Francis S, Porter KE, Ainscough JF, Pinteaux E, Drinkhill MJ and Turner NA. Fibroblast-specific deletion of interleukin-1 receptor-1 reduces adverse cardiac remodeling following myocardial infarction. JCI Insight. 2019;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Arancibia SA, Beltran CJ, Aguirre IM, Silva P, Peralta AL, Malinarich F and Hermoso MA. Toll-like receptors are key participants in innate immune responses. Biol Res. 2007;40:97–112. [DOI] [PubMed] [Google Scholar]
- 76.von Bernuth H, Picard C, Jin Z, Pankla R, Xiao H, Ku CL, Chrabieh M, Mustapha IB, Ghandil P, Camcioglu Y, Vasconcelos J, Sirvent N, Guedes M, Vitor AB, Herrero-Mata MJ, Arostegui JI, Rodrigo C, Alsina L, Ruiz-Ortiz E, Juan M, Fortuny C, Yague J, Anton J, Pascal M, Chang HH, Janniere L, Rose Y, Garty BZ, Chapel H, Issekutz A, Marodi L, Rodriguez-Gallego C, Banchereau J, Abel L, Li X, Chaussabel D, Puel A and Casanova JL. Pyogenic bacterial infections in humans with MyD88 deficiency. Science. 2008;321:691–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Chen B and Frangogiannis NG. Chemokines in Myocardial Infarction. J Cardiovasc Transl Res. 2020. [DOI] [PubMed] [Google Scholar]
- 78.Turner NA, Das A, Warburton P, O’Regan DJ, Ball SG and Porter KE. Interleukin-1alpha stimulates proinflammatory cytokine expression in human cardiac myofibroblasts. Am J Physiol Heart Circ Physiol. 2009;297:H1117–27. [DOI] [PubMed] [Google Scholar]
- 79.Rossini A, Zacheo A, Mocini D, Totta P, Facchiano A, Castoldi R, Sordini P, Pompilio G, Abeni D, Capogrossi MC and Germani A. HMGB1-stimulated human primary cardiac fibroblasts exert a paracrine action on human and murine cardiac stem cells. J Mol Cell Cardiol. 2008;44:683–93. [DOI] [PubMed] [Google Scholar]
- 80.Schafer S, Viswanathan S, Widjaja AA, Lim WW, Moreno-Moral A, DeLaughter DM, Ng B, Patone G, Chow K, Khin E, Tan J, Chothani SP, Ye L, Rackham OJL, Ko NSJ, Sahib NE, Pua CJ, Zhen NTG, Xie C, Wang M, Maatz H, Lim S, Saar K, Blachut S, Petretto E, Schmidt S, Putoczki T, Guimaraes-Camboa N, Wakimoto H, van Heesch S, Sigmundsson K, Lim SL, Soon JL, Chao VTT, Chua YL, Tan TE, Evans SM, Loh YJ, Jamal MH, Ong KK, Chua KC, Ong BH, Chakaramakkil MJ, Seidman JG, Seidman CE, Hubner N, Sin KYK and Cook SA. IL-11 is a crucial determinant of cardiovascular fibrosis. Nature. 2017;552:110–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Humeres C, Vivar R, Boza P, Munoz C, Bolivar S, Anfossi R, Osorio JM, Olivares-Silva F, Garcia L and Diaz-Araya G. Cardiac fibroblast cytokine profiles induced by proinflammatory or profibrotic stimuli promote monocyte recruitment and modulate macrophage M1/M2 balance in vitro. J Mol Cell Cardiol. 2016. [DOI] [PubMed] [Google Scholar]
- 82.Mouton AJ, Ma Y, Rivera Gonzalez OJ, Daseke MJ 2nd, Flynn ER, Freeman TC, Garrett MR, DeLeon-Pennell KY and Lindsey ML. Fibroblast polarization over the myocardial infarction time continuum shifts roles from inflammation to angiogenesis. Basic Res Cardiol. 2019;114:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Voss S, Kruger S, Scherschel K, Warnke S, Schwarzl M, Schrage B, Girdauskas E, Meyer C, Blankenberg S, Westermann D and Lindner D. Macrophage Migration Inhibitory Factor (MIF) Expression Increases during Myocardial Infarction and Supports Pro-Inflammatory Signaling in Cardiac Fibroblasts. Biomolecules. 2019;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Bernhagen J, Calandra T, Mitchell RA, Martin SB, Tracey KJ, Voelter W, Manogue KR, Cerami A and Bucala R. MIF is a pituitary-derived cytokine that potentiates lethal endotoxaemia. Nature. 1993;365:756–9. [DOI] [PubMed] [Google Scholar]
- 85.Shi X, Leng L, Wang T, Wang W, Du X, Li J, McDonald C, Chen Z, Murphy JW, Lolis E, Noble P, Knudson W and Bucala R. CD44 is the signaling component of the macrophage migration inhibitory factor-CD74 receptor complex. Immunity. 2006;25:595–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Larsson A, Peng S, Persson H, Rosenbloom J, Abrams WR, Wassberg E, Thelin S, Sletten K, Gerwins P and Westermark P. Lactadherin binds to elastin--a starting point for medin amyloid formation? Amyloid. 2006;13:78–85. [DOI] [PubMed] [Google Scholar]
- 87.Raymond A, Ensslin MA and Shur BD. SED1/MFG-E8: a bi-motif protein that orchestrates diverse cellular interactions. J Cell Biochem. 2009;106:957–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Silvestre JS, Thery C, Hamard G, Boddaert J, Aguilar B, Delcayre A, Houbron C, Tamarat R, Blanc-Brude O, Heeneman S, Clergue M, Duriez M, Merval R, Levy B, Tedgui A, Amigorena S and Mallat Z. Lactadherin promotes VEGF-dependent neovascularization. Nat Med. 2005;11:499–506. [DOI] [PubMed] [Google Scholar]
- 89.Nakaya M, Watari K, Tajima M, Nakaya T, Matsuda S, Ohara H, Nishihara H, Yamaguchi H, Hashimoto A, Nishida M, Nagasaka A, Horii Y, Ono H, Iribe G, Inoue R, Tsuda M, Inoue K, Tanaka A, Kuroda M, Nagata S and Kurose H. Cardiac myofibroblast engulfment of dead cells facilitates recovery after myocardial infarction. J Clin Invest. 2017;127:383–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kobayashi K, Maeda K, Takefuji M, Kikuchi R, Morishita Y, Hirashima M and Murohara T. Dynamics of angiogenesis in ischemic areas of the infarcted heart. Sci Rep. 2017;7:7156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ivey MJ, Kuwabara JT, Pai JT, Moore RE, Sun Z and Tallquist MD. Resident fibroblast expansion during cardiac growth and remodeling. J Mol Cell Cardiol. 2018;114:161–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Fu X, Khalil H, Kanisicak O, Boyer JG, Vagnozzi RJ, Maliken BD, Sargent MA, Prasad V, Valiente-Alandi I, Blaxall BC and Molkentin JD. Specialized fibroblast differentiated states underlie scar formation in the infarcted mouse heart. J Clin Invest. 2018;128:2127–2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Pu W, Han X, He L, Li Y, Huang X, Zhang M, Lv Z, Yu W, Wang QD, Cai D, Wang J, Sun R, Fei J, Ji Y, Nie Y and Zhou B. A genetic system for tissue-specific inhibition of cell proliferation. Development. 2020;147. [DOI] [PubMed] [Google Scholar]
- 94.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R and Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007;204:3037–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Hanna A and Frangogiannis NG. The Role of the TGF-beta Superfamily in Myocardial Infarction. Front Cardiovasc Med. 2019;6:140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Verrecchia F, Vindevoghel L, Lechleider RJ, Uitto J, Roberts AB and Mauviel A. Smad3/AP-1 interactions control transcriptional responses to TGF-beta in a promoter-specific manner. Oncogene. 2001;20:3332–40. [DOI] [PubMed] [Google Scholar]
- 97.Verrecchia F, Tacheau C, Schorpp-Kistner M, Angel P and Mauviel A. Induction of the AP-1 members c-Jun and JunB by TGF-beta/Smad suppresses early Smad-driven gene activation. Oncogene. 2001;20:2205–11. [DOI] [PubMed] [Google Scholar]
- 98.Barnette DN, Hulin A, Ahmed AS, Colige AC, Azhar M and Lincoln J. Tgfbeta-Smad and MAPK signaling mediate scleraxis and proteoglycan expression in heart valves. J Mol Cell Cardiol. 2013;65:137–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bagchi RA and Czubryt MP. Synergistic roles of scleraxis and Smads in the regulation of collagen 1alpha2 gene expression. Biochim Biophys Acta. 2012;1823:1936–44. [DOI] [PubMed] [Google Scholar]
- 100.Chen G, Nakamura I, Dhanasekaran R, Iguchi E, Tolosa EJ, Romecin PA, Vera RE, Almada LL, Miamen AG, Chaiteerakij R, Zhou M, Asiedu MK, Moser CD, Han S, Hu C, Banini BA, Oseini AM, Chen Y, Fang Y, Yang D, Shaleh HM, Wang S, Wu D, Song T, Lee JS, Thorgeirsson SS, Chevet E, Shah VH, Fernandez-Zapico ME and Roberts LR. Transcriptional Induction of Periostin by a Sulfatase 2-TGFbeta1-SMAD Signaling Axis Mediates Tumor Angiogenesis in Hepatocellular Carcinoma. Cancer Res. 2017;77:632–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, Vagnozzi RJ, Liu R, Huynh T, Lee SJ, Karch J and Molkentin JD. Fibroblast-specific TGF-beta-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest. 2017;127:3770–3783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Martin-Malpartida P, Batet M, Kaczmarska Z, Freier R, Gomes T, Aragon E, Zou Y, Wang Q, Xi Q, Ruiz L, Vea A, Marquez JA, Massague J and Macias MJ. Structural basis for genome wide recognition of 5-bp GC motifs by SMAD transcription factors. Nat Commun. 2017;8:2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Molkentin JD, Bugg D, Ghearing N, Dorn LE, Kim P, Sargent MA, Gunaje J, Otsu K and Davis J. Fibroblast-Specific Genetic Manipulation of p38 Mitogen-Activated Protein Kinase In Vivo Reveals Its Central Regulatory Role in Fibrosis. Circulation. 2017;136:549–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Pipes GC, Creemers EE and Olson EN. The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis. Genes Dev. 2006;20:1545–56. [DOI] [PubMed] [Google Scholar]
- 105.Sandbo N, Kregel S, Taurin S, Bhorade S and Dulin NO. Critical role of serum response factor in pulmonary myofibroblast differentiation induced by TGF-beta. Am J Respir Cell Mol Biol. 2009;41:332–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Velasquez LS, Sutherland LB, Liu Z, Grinnell F, Kamm KE, Schneider JW, Olson EN and Small EM. Activation of MRTF-A-dependent gene expression with a small molecule promotes myofibroblast differentiation and wound healing. Proc Natl Acad Sci U S A. 2013;110:16850–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Small EM. The actin-MRTF-SRF gene regulatory axis and myofibroblast differentiation. J Cardiovasc Transl Res. 2012;5:794–804. [DOI] [PubMed] [Google Scholar]
- 108.Small EM, Thatcher JE, Sutherland LB, Kinoshita H, Gerard RD, Richardson JA, Dimaio JM, Sadek H, Kuwahara K and Olson EN. Myocardin-related transcription factor-a controls myofibroblast activation and fibrosis in response to myocardial infarction. Circ Res. 2010;107:294–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Quijada P, Misra A, Velasquez LS, Burke RM, Lighthouse JK, Mickelsen DM, Dirkx RA Jr. and Small EM. Pre-existing fibroblasts of epicardial origin are the primary source of pathological fibrosis in cardiac ischemia and aging. J Mol Cell Cardiol. 2019;129:92–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Karsdal MA, Nielsen SH, Leeming DJ, Langholm LL, Nielsen MJ, Manon-Jensen T, Siebuhr A, Gudmann NS, Ronnow S, Sand JM, Daniels SJ, Mortensen JH and Schuppan D. The good and the bad collagens of fibrosis - Their role in signaling and organ function. Adv Drug Deliv Rev. 2017;121:43–56. [DOI] [PubMed] [Google Scholar]
- 111.Franzke CW, Bruckner P and Bruckner-Tuderman L. Collagenous transmembrane proteins: recent insights into biology and pathology. J Biol Chem. 2005;280:4005–8. [DOI] [PubMed] [Google Scholar]
- 112.Di Lullo GA, Sweeney SM, Korkko J, Ala-Kokko L and San Antonio JD. Mapping the ligand-binding sites and disease-associated mutations on the most abundant protein in the human, type I collagen. J Biol Chem. 2002;277:4223–31. [DOI] [PubMed] [Google Scholar]
- 113.Peterkofsky B Ascorbate requirement for hydroxylation and secretion of procollagen: relationship to inhibition of collagen synthesis in scurvy. Am J Clin Nutr. 1991;54:1135S–1140S. [DOI] [PubMed] [Google Scholar]
- 114.Gorres KL and Raines RT. Prolyl 4-hydroxylase. Crit Rev Biochem Mol Biol. 2010;45:106–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Myllyla R, Majamaa K, Gunzler V, Hanauske-Abel HM and Kivirikko KI. Ascorbate is consumed stoichiometrically in the uncoupled reactions catalyzed by prolyl 4-hydroxylase and lysyl hydroxylase. J Biol Chem. 1984;259:5403–5. [PubMed] [Google Scholar]
- 116.Ravassa S, Ballesteros G, Lopez B, Ramos P, Bragard J, Gonzalez A, Moreno MU, Querejeta R, Vives E, Garcia-Bolao I and Diez J. Combination of Circulating Type I Collagen-Related Biomarkers Is Associated With Atrial Fibrillation. J Am Coll Cardiol. 2019;73:1398–1410. [DOI] [PubMed] [Google Scholar]
- 117.Zelniker TA, Jarolim P, Scirica BM, Braunwald E, Park JG, Das S, Sabatine MS and Morrow DA. Biomarker of Collagen Turnover (C-Terminal Telopeptide) and Prognosis in Patients With Non- ST -Elevation Acute Coronary Syndromes. J Am Heart Assoc. 2019;8:e011444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Johnson R, Nxele X, Cour M, Sangweni N, Jooste T, Hadebe N, Samodien E, Benjeddou M, Mazino M, Louw J and Lecour S. Identification of potential biomarkers for predicting the early onset of diabetic cardiomyopathy in a mouse model. Sci Rep. 2020;10:12352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Cunningham JW, Claggett BL, O’Meara E, Prescott MF, Pfeffer MA, Shah SJ, Redfield MM, Zannad F, Chiang LM, Rizkala AR, Shi VC, Lefkowitz MP, Rouleau J, McMurray JJV, Solomon SD and Zile MR. Effect of Sacubitril/Valsartan on Biomarkers of Extracellular Matrix Regulation in Patients With HFpEF. J Am Coll Cardiol. 2020;76:503–514. [DOI] [PubMed] [Google Scholar]
- 120.Forrest L and Jackson DS. Intermolecular cross-linking of collagen in human and guinea pig scar tissue. Biochim Biophys Acta. 1971;229:681–9. [DOI] [PubMed] [Google Scholar]
- 121.Parmley WW, Chuck L, Kivowitz C, Matloff JM and Swan HJ. In vitro length-tension relations of human ventricular aneurysms. Relation of stiffness to mechanical disadvantage. Am J Cardiol. 1973;32:889–94. [DOI] [PubMed] [Google Scholar]
- 122.Mizuno T, Yau TM, Weisel RD, Kiani CG and Li RK. Elastin stabilizes an infarct and preserves ventricular function. Circulation. 2005;112:I81–8. [DOI] [PubMed] [Google Scholar]
- 123.Rienks M, Papageorgiou AP, Frangogiannis NG and Heymans S. Myocardial extracellular matrix: an ever-changing and diverse entity. Circ Res. 2014;114:872–88. [DOI] [PubMed] [Google Scholar]
- 124.Korf-Klingebiel M, Reboll MR, Grote K, Schleiner H, Wang Y, Wu X, Klede S, Mikhed Y, Bauersachs J, Klintschar M, Rudat C, Kispert A, Niessen HW, Lubke T, Dierks T and Wollert KC. Heparan Sulfate-Editing Extracellular Sulfatases Enhance VEGF Bioavailability for Ischemic Heart Repair. Circ Res. 2019;125:787–801. [DOI] [PubMed] [Google Scholar]
- 125.Schulz T, Schumacher U and Prehm P. Hyaluronan export by the ABC transporter MRP5 and its modulation by intracellular cGMP. J Biol Chem. 2007;282:20999–1004. [DOI] [PubMed] [Google Scholar]
- 126.Chen CH, Wang SS, Wei EI, Chu TY and Hsieh PC. Hyaluronan enhances bone marrow cell therapy for myocardial repair after infarction. Mol Ther. 2013;21:670–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Le LV, Mohindra P, Fang Q, Sievers RE, Mkrtschjan MA, Solis C, Safranek CW, Russell B, Lee RJ and Desai TA. Injectable hyaluronic acid based microrods provide local micromechanical and biochemical cues to attenuate cardiac fibrosis after myocardial infarction. Biomaterials. 2018;169:11–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Missinato MA, Tobita K, Romano N, Carroll JA and Tsang M. Extracellular component hyaluronic acid and its receptor Hmmr are required for epicardial EMT during heart regeneration. Cardiovasc Res. 2015;107:487–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Gaetani R, Feyen DA, Verhage V, Slaats R, Messina E, Christman KL, Giacomello A, Doevendans PA and Sluijter JP. Epicardial application of cardiac progenitor cells in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac function after myocardial infarction. Biomaterials. 2015;61:339–48. [DOI] [PubMed] [Google Scholar]
- 130.Jiang D, Liang J and Noble PW. Hyaluronan in tissue injury and repair. Annu Rev Cell Dev Biol. 2007;23:435–61. [DOI] [PubMed] [Google Scholar]
- 131.Pankov R and Yamada KM. Fibronectin at a glance. J Cell Sci. 2002;115:3861–3. [DOI] [PubMed] [Google Scholar]
- 132.Clark K, Pankov R, Travis MA, Askari JA, Mould AP, Craig SE, Newham P, Yamada KM and Humphries MJ. A specific alpha5beta1-integrin conformation promotes directional integrin translocation and fibronectin matrix formation. J Cell Sci. 2005;118:291–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Talman V and Ruskoaho H. Cardiac fibrosis in myocardial infarction-from repair and remodeling to regeneration. Cell Tissue Res. 2016;365:563–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Samuel JL, Barrieux A, Dufour S, Dubus I, Contard F, Koteliansky V, Farhadian F, Marotte F, Thiery JP and Rappaport L. Accumulation of fetal fibronectin mRNAs during the development of rat cardiac hypertrophy induced by pressure overload. J Clin Invest. 1991;88:1737–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Contard F, Koteliansky V, Marotte F, Dubus I, Rappaport L and Samuel JL. Specific alterations in the distribution of extracellular matrix components within rat myocardium during the development of pressure overload. Lab Invest. 1991;64:65–75. [PubMed] [Google Scholar]
- 136.Gabler U, Berndt A, Kosmehl H, Mandel U, Zardi L, Muller S, Stelzner A and Katenkamp D. Matrix remodelling in dilated cardiomyopathy entails the occurrence of oncofetal fibronectin molecular variants. Heart. 1996;75:358–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kosmehl H, Berndt A and Katenkamp D. Molecular variants of fibronectin and laminin: structure, physiological occurrence and histopathological aspects. Virchows Arch. 1996;429:311–22. [DOI] [PubMed] [Google Scholar]
- 138.Klingberg F, Chau G, Walraven M, Boo S, Koehler A, Chow ML, Olsen AL, Im M, Lodyga M, Wells RG, White ES and Hinz B. The fibronectin ED-A domain enhances recruitment of latent TGF-beta-binding protein-1 to the fibroblast matrix. J Cell Sci. 2018;131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Bhattacharyya S, Tamaki Z, Wang W, Hinchcliff M, Hoover P, Getsios S, White ES and Varga J. FibronectinEDA promotes chronic cutaneous fibrosis through Toll-like receptor signaling. Sci Transl Med. 2014;6:232ra50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Chorawala MR, Prakash P, Doddapattar P, Jain M, Dhanesha N and Chauhan AK. Deletion of Extra Domain A of Fibronectin Reduces Acute Myocardial Ischaemia/Reperfusion Injury in Hyperlipidaemic Mice by Limiting Thrombo-Inflammation. Thromb Haemost. 2018;118:1450–1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Arslan F, Smeets MB, Riem Vis PW, Karper JC, Quax PH, Bongartz LG, Peters JH, Hoefer IE, Doevendans PA, Pasterkamp G and de Kleijn DP. Lack of fibronectin-EDA promotes survival and prevents adverse remodeling and heart function deterioration after myocardial infarction. Circ Res. 2011;108:582–92. [DOI] [PubMed] [Google Scholar]
- 142.Valiente-Alandi I, Potter SJ, Salvador AM, Schafer AE, Schips T, Carrillo-Salinas F, Gibson AM, Nieman ML, Perkins C, Sargent MA, Huo J, Lorenz JN, DeFalco T, Molkentin JD, Alcaide P and Blaxall BC. Inhibiting Fibronectin Attenuates Fibrosis and Improves Cardiac Function in a Model of Heart Failure. Circulation. 2018;138:1236–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Frangogiannis NG. Matricellular proteins in cardiac adaptation and disease. Physiol Rev. 2012;92:635–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Li G, Jin R, Norris RA, Zhang L, Yu S, Wu F, Markwald RR, Nanda A, Conway SJ, Smyth SS and Granger DN. Periostin mediates vascular smooth muscle cell migration through the integrins alphavbeta3 and alphavbeta5 and focal adhesion kinase (FAK) pathway. Atherosclerosis. 2010;208:358–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Takayama G, Arima K, Kanaji T, Toda S, Tanaka H, Shoji S, McKenzie AN, Nagai H, Hotokebuchi T and Izuhara K. Periostin: a novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J Allergy Clin Immunol. 2006;118:98–104. [DOI] [PubMed] [Google Scholar]
- 146.Oka T, Xu J, Kaiser RA, Melendez J, Hambleton M, Sargent MA, Lorts A, Brunskill EW, Dorn GW 2nd, Conway SJ, Aronow BJ, Robbins J and Molkentin JD. Genetic manipulation of periostin expression reveals a role in cardiac hypertrophy and ventricular remodeling. Circ Res. 2007;101:313–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Norris RA, Damon B, Mironov V, Kasyanov V, Ramamurthi A, Moreno-Rodriguez R, Trusk T, Potts JD, Goodwin RL, Davis J, Hoffman S, Wen X, Sugi Y, Kern CB, Mjaatvedt CH, Turner DK, Oka T, Conway SJ, Molkentin JD, Forgacs G and Markwald RR. Periostin regulates collagen fibrillogenesis and the biomechanical properties of connective tissues. J Cell Biochem. 2007;101:695–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Shimazaki M, Nakamura K, Kii I, Kashima T, Amizuka N, Li M, Saito M, Fukuda K, Nishiyama T, Kitajima S, Saga Y, Fukayama M, Sata M and Kudo A. Periostin is essential for cardiac healing after acute myocardial infarction. J Exp Med. 2008;205:295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Adams JC and Lawler J. The thrombospondins. Int J Biochem Cell Biol. 2004;36:961–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kirk JA and Cingolani OH. Thrombospondins in the transition from myocardial infarction to heart failure. J Mol Cell Cardiol. 2016;90:102–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Muppala S, Frolova E, Xiao R, Krukovets I, Yoon S, Hoppe G, Vasanji A, Plow E and Stenina-Adognravi O. Proangiogenic Properties of Thrombospondin-4. Arterioscler Thromb Vasc Biol. 2015;35:1975–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Frolova EG, Pluskota E, Krukovets I, Burke T, Drumm C, Smith JD, Blech L, Febbraio M, Bornstein P, Plow EF and Stenina OI. Thrombospondin-4 regulates vascular inflammation and atherogenesis. Circ Res. 2010;107:1313–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Bonnefoy A and Hoylaerts MF. Thrombospondin-1 in von Willebrand factor function. Curr Drug Targets. 2008;9:822–32. [DOI] [PubMed] [Google Scholar]
- 154.Bonnefoy A, Moura R and Hoylaerts MF. The evolving role of thrombospondin-1 in hemostasis and vascular biology. Cell Mol Life Sci. 2008;65:713–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Lynch JM, Maillet M, Vanhoutte D, Schloemer A, Sargent MA, Blair NS, Lynch KA, Okada T, Aronow BJ, Osinska H, Prywes R, Lorenz JN, Mori K, Lawler J, Robbins J and Molkentin JD. A thrombospondin-dependent pathway for a protective ER stress response. Cell. 2012;149:1257–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Schips TG, Vanhoutte D, Vo A, Correll RN, Brody MJ, Khalil H, Karch J, Tjondrokoesoemo A, Sargent MA, Maillet M, Ross RS and Molkentin JD. Thrombospondin-3 augments injury-induced cardiomyopathy by intracellular integrin inhibition and sarcolemmal instability. Nat Commun. 2019;10:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Verma RP and Hansch C. Matrix metalloproteinases (MMPs): chemical-biological functions and (Q)SARs. Bioorg Med Chem. 2007;15:2223–68. [DOI] [PubMed] [Google Scholar]
- 158.Van Lint P and Libert C. Chemokine and cytokine processing by matrix metalloproteinases and its effect on leukocyte migration and inflammation. J Leukoc Biol. 2007;82:1375–81. [DOI] [PubMed] [Google Scholar]
- 159.Van Wart HE and Birkedal-Hansen H. The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc Natl Acad Sci U S A. 1990;87:5578–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Ra HJ and Parks WC. Control of matrix metalloproteinase catalytic activity. Matrix Biol. 2007;26:587–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Vanhoutte D, Schellings M, Pinto Y and Heymans S. Relevance of matrix metalloproteinases and their inhibitors after myocardial infarction: a temporal and spatial window. Cardiovasc Res. 2006;69:604–13. [DOI] [PubMed] [Google Scholar]
- 162.DeLeon-Pennell KY, Meschiari CA, Jung M and Lindsey ML. Matrix Metalloproteinases in Myocardial Infarction and Heart Failure. Prog Mol Biol Transl Sci. 2017;147:75–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Heymans S, Luttun A, Nuyens D, Theilmeier G, Creemers E, Moons L, Dyspersin GD, Cleutjens JP, Shipley M, Angellilo A, Levi M, Nube O, Baker A, Keshet E, Lupu F, Herbert JM, Smits JF, Shapiro SD, Baes M, Borgers M, Collen D, Daemen MJ and Carmeliet P. Inhibition of plasminogen activators or matrix metalloproteinases prevents cardiac rupture but impairs therapeutic angiogenesis and causes cardiac failure. Nat Med. 1999;5:1135–42. [DOI] [PubMed] [Google Scholar]
- 164.Heymans S, Pauschinger M, De Palma A, Kallwellis-Opara A, Rutschow S, Swinnen M, Vanhoutte D, Gao F, Torpai R, Baker AH, Padalko E, Neyts J, Schultheiss HP, Van de Werf F, Carmeliet P and Pinto YM. Inhibition of urokinase-type plasminogen activator or matrix metalloproteinases prevents cardiac injury and dysfunction during viral myocarditis. Circulation. 2006;114:565–73. [DOI] [PubMed] [Google Scholar]
- 165.Perez-Hernandez N, Vargas-Alarcon G, Martinez-Rodriguez N, Martinez-Rios MA, Pena-Duque MA, Pena-Diaz Ade L, Valente-Acosta B, Posadas-Romero C, Medina A and Rodriguez-Perez JM. The matrix metalloproteinase 2–1575 gene polymorphism is associated with the risk of developing myocardial infarction in Mexican patients. J Atheroscler Thromb. 2012;19:718–27. [DOI] [PubMed] [Google Scholar]
- 166.Vasku A, Goldbergova M, Holla LI, Spinarova L, Spinar J, Vitovec J and Vacha J. Two MMP-2 promoter polymorphisms (−790T/G and −735C/T) in chronic heart failure. Clin Chem Lab Med. 2003;41:1299–303. [DOI] [PubMed] [Google Scholar]
- 167.Beber AR, Polina ER, Biolo A, Santos BL, Gomes DC, La Porta VL, Olsen V, Clausell N, Rohde LE and Santos KG. Matrix Metalloproteinase-2 Polymorphisms in Chronic Heart Failure: Relationship with Susceptibility and Long-Term Survival. PLoS One. 2016;11:e0161666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Delgado-Enciso I, Gonzalez-Hernandez NA, Baltazar-Rodriguez LM, Millan-Guerrero RO, Newton-Sanchez O, Bayardo-Noriega A, Aleman-Mireles A, Enriquez-Maldonado IG, Anaya-Carrillo MJ, Rojas-Martinez A and Ortiz-Lopez R. Association of matrix metalloproteinase-2 gene promoter polymorphism with myocardial infarction susceptibility in a Mexican population. J Genet. 2009;88:249–52. [DOI] [PubMed] [Google Scholar]
- 169.Mizon-Gerard F, de Groote P, Lamblin N, Hermant X, Dallongeville J, Amouyel P, Bauters C and Helbecque N. Prognostic impact of matrix metalloproteinase gene polymorphisms in patients with heart failure according to the aetiology of left ventricular systolic dysfunction. Eur Heart J. 2004;25:688–93. [DOI] [PubMed] [Google Scholar]
- 170.Demacq C, Vasconcellos VB, Marcaccini AM, Gerlach RF, Silva WA Jr. and Tanus-Santos JE. Functional polymorphisms in the promoter of the matrix metalloproteinase-9 (MMP-9) gene are not linked with significant plasma MMP-9 variations in healthy subjects. Clin Chem Lab Med. 2008;46:57–63. [DOI] [PubMed] [Google Scholar]
- 171.Metzger IF, Luizon MR, Lacchini R and Tanus-Santos JE. Genetic variants in matrix metalloproteinase-9 gene modify metalloproteinase-9 levels in black subjects. DNA Cell Biol. 2012;31:504–10. [DOI] [PubMed] [Google Scholar]
- 172.Lacchini R, Metzger IF, Luizon M, Ishizawa M and Tanus-Santos JE. Interethnic differences in the distribution of matrix metalloproteinases genetic polymorphisms are consistent with interethnic differences in disease prevalence. DNA Cell Biol. 2010;29:649–55. [DOI] [PubMed] [Google Scholar]
- 173.Wang X and Shi LZ. Association of matrix metalloproteinase-9 C1562T polymorphism and coronary artery disease: a meta-analysis. J Zhejiang Univ Sci B. 2014;15:256–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Meschiari CA, Jung M, Iyer RP, Yabluchanskiy A, Toba H, Garrett MR and Lindsey ML. Macrophage overexpression of matrix metalloproteinase-9 in aged mice improves diastolic physiology and cardiac wound healing after myocardial infarction. Am J Physiol Heart Circ Physiol. 2018;314:H224–H235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Lovelock JD, Baker AH, Gao F, Dong JF, Bergeron AL, McPheat W, Sivasubramanian N and Mann DL. Heterogeneous effects of tissue inhibitors of matrix metalloproteinases on cardiac fibroblasts. Am J Physiol Heart Circ Physiol. 2005;288:H461–8. [DOI] [PubMed] [Google Scholar]
- 176.Brew K, Dinakarpandian D and Nagase H. Tissue inhibitors of metalloproteinases: evolution, structure and function. Biochim Biophys Acta. 2000;1477:267–83. [DOI] [PubMed] [Google Scholar]
- 177.Uchinaka A, Kawaguchi N, Mori S, Hamada Y, Miyagawa S, Saito A, Sawa Y and Matsuura N. Tissue inhibitor of metalloproteinase-1 and −3 improves cardiac function in an ischemic cardiomyopathy model rat. Tissue Eng Part A. 2014;20:3073–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Dewald O, Ren G, Duerr GD, Zoerlein M, Klemm C, Gersch C, Tincey S, Michael LH, Entman ML and Frangogiannis NG. Of mice and dogs: species-specific differences in the inflammatory response following myocardial infarction. Am J Pathol. 2004;164:665–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Fishbein MC, Maclean D and Maroko PR. The histopathologic evolution of myocardial infarction. Chest. 1978;73:843–9. [DOI] [PubMed] [Google Scholar]
- 180.Fishbein MC, Maclean D and Maroko PR. Experimental myocardial infarction in the rat: qualitative and quantitative changes during pathologic evolution. Am J Pathol. 1978;90:57–70. [PMC free article] [PubMed] [Google Scholar]
- 181.Yang F, Liu YH, Yang XP, Xu J, Kapke A and Carretero OA. Myocardial infarction and cardiac remodelling in mice. Exp Physiol. 2002;87:547–55. [DOI] [PubMed] [Google Scholar]
- 182.Nakahama M, Murakami T, Kusachi S, Naito I, Takeda K, Ohnishi H, Komatsubara I, Oka T, Ninomiya Y and Tsuji T. Expression of perlecan proteoglycan in the infarct zone of mouse myocardial infarction. J Mol Cell Cardiol. 2000;32:1087–100. [DOI] [PubMed] [Google Scholar]
- 183.Doi M, Kusachi S, Murakami T, Ninomiya Y, Murakami M, Nakahama M, Takeda K, Komatsubara I, Naito I and Tsuji T. Time-dependent changes of decorin in the infarct zone after experimentally induced myocardial infarction in rats: comparison with biglycan. Pathol Res Pract. 2000;196:23–33. [DOI] [PubMed] [Google Scholar]
- 184.Fomovsky GM and Holmes JW. Evolution of scar structure, mechanics, and ventricular function after myocardial infarction in the rat. Am J Physiol Heart Circ Physiol. 2010;298:H221–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Ma Y, de Castro Bras LE, Toba H, Iyer RP, Hall ME, Winniford MD, Lange RA, Tyagi SC and Lindsey ML. Myofibroblasts and the extracellular matrix network in post-myocardial infarction cardiac remodeling. Pflugers Arch. 2014;466:1113–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zimmerman SD, Thomas DP, Velleman SG, Li X, Hansen TR and McCormick RJ. Time course of collagen and decorin changes in rat cardiac and skeletal muscle post-MI. Am J Physiol Heart Circ Physiol. 2001;281:H1816–22. [DOI] [PubMed] [Google Scholar]
- 187.Richardson WJ, Clarke SA, Quinn TA and Holmes JW. Physiological Implications of Myocardial Scar Structure. Compr Physiol. 2015;5:1877–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.van den Borne SW, van de Schans VA, Strzelecka AE, Vervoort-Peters HT, Lijnen PM, Cleutjens JP, Smits JF, Daemen MJ, Janssen BJ and Blankesteijn WM. Mouse strain determines the outcome of wound healing after myocardial infarction. Cardiovasc Res. 2009;84:273–82. [DOI] [PubMed] [Google Scholar]
- 189.Sun Y and Weber KT. Infarct scar: a dynamic tissue. Cardiovasc Res. 2000;46:250–6. [DOI] [PubMed] [Google Scholar]
- 190.Rutherford SL, Trew ML, Sands GB, LeGrice IJ and Smaill BH. High-resolution 3-dimensional reconstruction of the infarct border zone: impact of structural remodeling on electrical activation. Circ Res. 2012;111:301–11. [DOI] [PubMed] [Google Scholar]
- 191.Mahoney VM, Mezzano V, Mirams GR, Maass K, Li Z, Cerrone M, Vasquez C, Bapat A, Delmar M and Morley GE. Connexin43 contributes to electrotonic conduction across scar tissue in the intact heart. Sci Rep. 2016;6:26744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Rotenberg MY, Yamamoto N, Schaumann EN, Matino L, Santoro F and Tian B. Living myofibroblast-silicon composites for probing electrical coupling in cardiac systems. Proc Natl Acad Sci U S A. 2019;116:22531–22539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Schultz F, Swiatlowska P, Alvarez-Laviada A, Sanchez-Alonso JL, Song Q, de Vries AAF, Pijnappels DA, Ongstad E, Braga VMM, Entcheva E, Gourdie RG, Miragoli M and Gorelik J. Cardiomyocyte-myofibroblast contact dynamism is modulated by connexin-43. FASEB J. 2019;33:10453–10468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Quinn TA, Camelliti P, Rog-Zielinska EA, Siedlecka U, Poggioli T, O’Toole ET, Knopfel T and Kohl P. Electrotonic coupling of excitable and nonexcitable cells in the heart revealed by optogenetics. Proc Natl Acad Sci U S A. 2016;113:14852–14857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Eaton LW, Weiss JL, Bulkley BH, Garrison JB and Weisfeldt ML. Regional cardiac dilatation after acute myocardial infarction: recognition by two-dimensional echocardiography. N Engl J Med. 1979;300:57–62. [DOI] [PubMed] [Google Scholar]
- 196.Fieno DS, Hillenbrand HB, Rehwald WG, Harris KR, Decker RS, Parker MA, Klocke FJ, Kim RJ and Judd RM. Infarct resorption, compensatory hypertrophy, and differing patterns of ventricular remodeling following myocardial infarctions of varying size. J Am Coll Cardiol. 2004;43:2124–31. [DOI] [PubMed] [Google Scholar]
- 197.Hillenbrand HB, Sandstede J, Stork S, Ramsayer B, Hahn D, Ertl G, Koestler H, Bauer W and Ritter C. Remodeling of the infarct territory in the time course of infarct healing in humans. MAGMA. 2011;24:277–84. [DOI] [PubMed] [Google Scholar]
- 198.Jugdutt BI and Amy RW. Healing after myocardial infarction in the dog: changes in infarct hydroxyproline and topography. J Am Coll Cardiol. 1986;7:91–102. [DOI] [PubMed] [Google Scholar]
- 199.Waller C, Hiller KH, Kahler E, Hu K, Nahrendorf M, Voll S, Haase A, Ertl G and Bauer WR. Serial magnetic resonance imaging of microvascular remodeling in the infarcted rat heart. Circulation. 2001;103:1564–9. [DOI] [PubMed] [Google Scholar]
- 200.Roberts CS, Maclean D, Maroko P and Kloner RA. Early and late remodeling of the left ventricle after acute myocardial infarction. Am J Cardiol. 1984;54:407–10. [DOI] [PubMed] [Google Scholar]



