Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through activation of inflammatory and fibrogenic cascades that ultimately result in formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of DAMPs that initiate both myocardial and systemic inflammatory responses. TLRs and NLRs recognize DAMPs and transduce downstream pro-inflammatory signals, leading to upregulation of cytokines (such as Interleukin-1, TNF-α and Interleukin-6) and chemokines (such as CCL2) and recruitment of neutrophils, monocytes and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart plays a major role in clearance of the infarct from dead cells and in subsequent stimulation of reparative pathways. Efferocytosis triggers induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and to inhibit arrhythmia generation in patients surviving an myocardial infarction.
Keywords: Basic Science Research, Cardiovascular Disease, Cell Biology/Structural Biology
1. Introduction
Advances in acute care of patients with myocardial infarction (MI) and the introduction of early reperfusion strategies resulted in a marked reduction in short-term mortality (in-hospital and 30-day mortality) from >30% in the 1950s to ~5-8% in the current era1,2,3. In contrast to the marked improvement in early mortality and event rates, effects on long-term mortality have been much less impressive4. In fact, the improved survival of patients suffering acute MI resulted in an expansion of the pool of patients that develop adverse remodeling and heart failure. Progression of heart failure after acute infarction is dependent not only on the size of the acute infarct, but also on the quality of the reparative response. Thus, heart failure after infarction results, at least in part, from faulty healing.
In patients suffering a large MI, sudden loss of up to a billion cardiomyocytes overwhelms the extremely limited regenerative capacity of the adult human heart. Thus, repair of the infarcted heart requires timely activation and suppression of an acute inflammatory reaction that clears the infarct from dead cells and matrix debris and ultimately results in formation of a collagen-based scar5. Immune cells, fibroblasts and vascular cells co-operate to promote healing of the infarcted heart, protecting the ventricle from catastrophic rupture. Perturbations in inflammatory, fibrogenic and angiogenic responses after infarction play an important role in the pathogenesis of adverse post-infarction remodeling and heart failure.
Dissection of the cellular and molecular mechanisms involved in repair of the infarcted heart is important to understand the pathogenesis of post-infarction remodeling and heart failure. This review manuscript discusses our current knowledge on the cellular effectors of cardiac repair, and the molecular signals involved in the reparative cardiac response. We also identify promising therapeutic approaches that may protect the infarcted heart from adverse remodeling by targeting inflammatory and fibrogenic signals involved in cardiac repair.
2. The phases of the reparative response after MI
MI is defined as cardiomyocyte death triggered by an ischemic insult. In most cases, rupture or erosion of a coronary plaque triggers thrombosis leading to acute cessation of flow in the myocardium subserved by the occluded coronary (type 1 infarction). In other cases, ischemia is triggered by an imbalance between myocardial supply and demand caused by an extracardiac stressor, such as hypotension, tachycardia, anemia, or hypertension, in the presence of coronary disease (type 2 infarction). Complete cessation of coronary flow results in almost immediate functional depression (within seconds after the ischemic insult), followed by early ultrastructural changes in ischemic cardiomyocytes (within minutes of coronary occlusion). Ischemic insults longer that 15-20 minutes are sufficient to induce irreversible changes in some subendocardial cardiomyocytes that ultimately culminate in cell death6. The longer the ischemic interval, the larger the extent of the infarct, which has been suggested to progress from the subendocardium to the less vulnerable subepicardium as a “wavefront of necrosis” as the duration of the ischemia is prolonged7.
Massive death of cardiomyocytes in the infarct zone initiates a well-orchestrated reparative response that can be divided into 3 distinct but overlapping phases: the inflammatory phase, the proliferative phase and the maturation/remodeling phase. Dying cardiomyocytes and degraded extracellular matrix proteins release danger signals that activate inflammatory cascades, resulting in recruitment of neutrophils and monocytes in the infarct. Professional phagocytes clear the infarct from death cells and matrix debris, thus activating endogenous pathways that suppress pro-inflammatory signaling and stimulate fibrogenic and angiogenic macrophage phenotypes. Thus, suppression of inflammation is coupled with the transition to the proliferative phase of cardiac repair, during which fibroblasts expand, convert into myofibroblasts and undergo activation8, generating an extracellular matrix network that supports the structural integrity of the ventricle. Formation of neovessels ensures supply of the infarct with blood to support the metabolically active wound. As a matrix network comprised of structural collagens is formed, fibroblasts and myofibroblasts undergo phenotypic transitions to matrifibrocytes, a population of fibroblast-like cells that serve a scar-preserving role, neovessels acquire a coat comprised of vascular mural cells (vascular smooth muscle cells and pericytes) and a mature scar containing large amounts of cross-linked collagen is formed. In large infarcts associated with substantial loss of contractile muscle, replacement of dead cardiomyocytes with collagenous tissue in the infarct zone is accompanied by remodeling of the non-infarcted myocardium, which exhibits macrophage activation and interstitial fibrosis in response to pressure and volume loads. The cellular responses to MI are qualitatively similar in reperfused and non-reperfused infarcts. However, reperfusion attenuates cardiomyocyte loss in the area at risk, while accelerating the myocardial inflammatory response (and its resolution) and promoting earlier activation of fibroblast-driven repair.
3. The inflammatory phase of cardiac repair
3.1. MI triggers both local and systemic inflammatory responses.
MI triggers both systemic activation of inflammation and an intense myocardial inflammatory reaction. Long range signals transfer information from the infarct to the bone marrow and the spleen, thus promoting leukocyte production and mobilization9,10,11. The molecular signals that signal to the immune system the need for leukocyte supply to repair the ventricle remain poorly understood. Angiotensin II activation mobilizes monocytes from the splenic reservoir within hours after MI10, and cytokines (such as IL-1β) released by the infarct stimulate bone marrow hematopoietic stem cell proliferation and monocyte production12. Moreover, in the bone marrow, sympathetic nerves may activate β3 adrenergic receptors in mesenchymal stromal cells13 thus providing local cues that release hematopoietic stem and progenitor cells and ultimately increase the output of neutrophils and monocytes14. On the other hand, myocardial signals derived from dying cells localize the inflammatory response to the infarcted heart.
3.2. Molecular pathways that initiate the myocardial post-infarction inflammatory response: the Damage-Associated Molecular Patterns (DAMPs),
Cell death and protease-mediated degradation of the extracellular matrix generate a broad range of damage-associated substances that act as “danger signals” and initiate the post-infarction inflammatory response. These mediators, called DAMPs, bind to pattern recognition receptors (PRRs) located on the surface of surviving myocardial parenchymal cells or and infiltrating leukocytes and stimulate downstream inflammatory cascades that culminate in secretion of pro-inflammatory cytokines and chemokines, and in induction of cell adhesion molecules15,16,17. In addition to their passive release from dead cells and damaged extracellular matrix, DAMPs can also be expressed and secreted by activated immune cells, cardiomyocytes, and interstitial cells under conditions of stress. Over the last 20 years several different DAMPs have been suggested to contribute to the initiation of the post-infarction inflammatory response, including the nuclear protein high mobility group box-1 (HMGB1), heat shock proteins (HSPs), fibronectin extra domain A (fibronectin-EDA), low molecular weight hyaluronic acid, S100 proteins, interleukin (IL)-1α, cardiac myosin, ATP, mitochondrial DNA, dsRNA, extracellular RNA (eRNA) and others18-22 (Figure 1). The relative significance of each DAMP in activating selected aspects of the inflammatory response remains poorly understood. DAMPs act by binding to PRRs, primarily the toll-like receptor/IL-1 receptor (TLR/IL-1R) cell surface proteins, members of the cytosolic NOD-like receptor (NLR) family, or the membrane receptor for advanced glycation end-products (RAGE). Upon activation, PRRs stimulate downstream mitogen-activated protein kinases (MAPKs) and nuclear factor (NF)-κB cascades21, 23-25, driving induction of pro-inflammatory cytokines and chemokines and upregulation of cell adhesion molecules and facilitating leukocyte trafficking in the infarct. Infiltrating leukocytes serve an important reparative role, clearing the infarct from dead cells and matrix debris and stimulating activation of matrix-producing interstitial cells and angiogenesis. However, excessive, prolonged or dysregulated leukocyte activation may accentuate injury, increasing death of cardiomyocytes, promoting fibrosis and stimulating protease-induced matrix degradation that can lead to left ventricular rupture. Considering that DAMPs may stimulate both reparative and maladaptive inflammatory responses, it is not surprising that various studies inhibiting DAMP-PRR interactions in the infarcted heart have reported both protective and detrimental effects.
Figure 1: Damage-Associated Molecular Patterns (DAMPs) Initiate the post-infarction inflammatory response.
In the infarcted heart, dying cells, damaged extracellular matrix and infiltrating immune cells release a broad range of DAMPs, including the nuclear protein high mobility group box-1 (HMGB1), heat shock proteins (HSP), cardiac myosin, extracellular RNA (eRNA), the S100 proteins S100A8/A9, fibronectin extra domain A (EDA-Fn), low molecular weight hyaluronan (LMWH), and interleukin (IL)-1α. DAMPs bind to pattern recognition receptors expressed on the cell surface of immune cells, vascular cells, fibroblasts and cardiomyocytes, transducing downstream pro-inflammatory cascades and stimulating leukocyte recruitment.
3.2.1. HMGB1.
The non-histone chromatin protein HMGB1 is expressed in mammalian cells and is involved in DNA stabilization and transcription regulation. Upon cell stress and injury, HMGB1 is passively released by necrotic cells26, but also actively secreted by stimulated immune cells27, or by ischemic cardiomyocytes28. HMGB1 release has been documented in both experimental models of myocardial ischemia and in human patients with MI. In animal models of reperfused29, 30 and non-reperfused31 MI, early myocardial induction of HMGB1 expression is accompanied by a marked increase in serum levels. In acute MI patients, increased HMGB1 levels predict adverse outcome31-33.
The pro-inflammatory effects of HMGB1 involve binding to RAGE or members of the TLR family (predominantly TLR2, TLR4 and TLR922, 29, 34, 35, 36) and downstream activation of NF-κB signaling. Moreover, HMGB1 has also been reported to stimulate leukocyte recruitment through direct binding to the CXC chemokine CXCL12/stromal cell-derived factor 1 (SDF-1). HMGB1-CXCL12 heterocomplexes can synergistically enhance leukocyte CXCR4 signaling 37 enhancing inflammation through effects independent of PRR binding.
Studies examining the role of HMGB1 in MI have suggested both injurious and protective actions. In a reperfused MI model, HMGB1 was found to augment injury and increase infarct size, through effects mediated partly via RAGE signaling29, 30. In contrast, in non-reperfused MI models, cardiomyocyte-specific HMGB1 overexpression38, 39, or treatment with exogenous HMGB1 40-42 were found to attenuate cardiac remodeling. Protective effects of HMGB1 in non-reperfused infarcts were attributed to improved repair, enhanced angiogenesis and reduced fibrosis. Consistent with these reparative actions, HMGB1 inhibition using a neutralizing antibody perturbed repair in non-reperfused MI, inducing scar thinning, infarct expansion and adverse remodeling31. These findings underscore the context-dependent role of DAMP-mediated inflammation in injury and repair of the infarcted heart. Reperfusion salvages many cardiomyocytes from death; however, many of these cells may be vulnerable to the injurious effects of inflammation. On the other hand, in non-reperfused infarcts virtually all cardiomyocytes in the area at risk die; thus, HMGB1 signaling may play a central role in activation of inflammatory and reparative cascades that preserve the structure and geometry of the ventricle protecting from rupture and adverse remodeling.
3.2.2. S100 proteins.
The S100 family of calcium-binding cytosolic is comprised of 25 members with a broad range of intracellular and extracellular functions43. Several members of the S100 family were reported to act as DAMPs, which are released following injury and activate pro-inflammatory signaling by binding to TLRs or RAGE. S100A8 (also known as myeloid-related protein 8 (MRP8)] or calgranulin A) and S100A9 (MRP14) are the best characterized S100 family members in MI. Predominantly expressed and secreted by neutrophils and macrophages, S100A8 and A9 associate to form the S100A8/A9 heterodimer, which functions as a DAMP, signaling through activation of RAGE and TLR444, 45. Release of S100A8/A9 has been documented in both experimental models of myocardial ischemia and in human patients with acute MI46, 47, 48. Both pharmacologic and genetic interventions have suggested that S100A8/A9 may accentuate injury during the early stages of the post-MI inflammatory response by promoting granulopoiesis in the bone marrow49 and by accentuating leukocyte infiltration50, while promoting reparative actions at later timepoints through recruitment of reparative macrophages51.
The role of other S100 proteins in the infarcted myocardium remains poorly understood. S100A12 is released in the circulation following MI52; however, its potential role as a DAMP had not been documented. S100A1, the most abundant S100 family member in cardiomyocytes, is also released following MI53, but does not act as a pro-inflammatory stimulus, but is taken up through endocytosis by cardiac fibroblasts to transiently activate TLR4-endolysosomal signaling, promoting an anti-fibrotic phenotype that protects the infarcted heart from adverse remodeling53. S100A4 has also been suggested to exert protective actions, mediated through stimulation of an anti-apoptotic program in cardiomyocytes54.
3.2.3. Other DAMPs
Several additional mediators were found to be released in the infarcted myocardium and were suggested to serve as danger signals that activate the post-infarction inflammatory response. IL-1α is constitutively expressed by many different cell types and is upregulated in response to a broad range of pro-inflammatory signals. Cell necrosis results in rapid release of large amounts of IL-1α that can directly stimulate pro-inflammatory signaling cascades by binding to the type 1 IL-1 receptor (IL-1R1). In contrast, apoptosis sequesters IL-1α in the nucleus preventing release of the DAMP and protecting from inflammatory activation55. In addition to this mechanism, IL-1α can also be localized on the cell surface, where it may serve as an alarmin that activates neighboring cells in response to injury56. Following MI, IL-1α derived from necrotic cardiomyocytes was found to serve as a danger signal that simulates pro-inflammatory MAPK and NF-κB signaling in cardiac interstitial cells through activation of the IL-1R1 pathway, thus promoting adverse remodeling57,58. Moreover, IL-1α was detected on the cell surface of monocytes from patients with MI and was suggested to play a role in mediating the adhesive interactions between leukocytes and endothelial cells that trigger inflammatory cell infiltration59.
Components of the extracellular matrix network have also been suggested to play a role as DAMPs that, when released following injury, activate the inflammatory response60. Fibronectin domains, low molecular-weight hyaluronan, and tenascin-C have been suggested to act as alarmins in various pathologic conditions. Data in MI are limited. Fibronectin is secreted by fibroblasts, vascular cells and macrophages infiltrating the infarct and includes an alternatively spliced exon coding type III repeat extra domain A (EDA) that can bind to TLR4 and transduce downstream inflammatory signaling61, 62. Expression of fibronectin-EDA by parenchymal myocardial cells was found to mediate inflammation and dysfunction in the infarcted heart63. In addition to its pro-inflammatory actions, fibronectin-EDA may also play an important role in activation of reparative myofibroblasts64, in part through recruitment of active TGF-β in the pericellular matrix65.
3.3. PRRs and RAGE mediate the effects of DAMPs in the infarcted myocardium:
There are several major subfamilies of PRRs: the transmembrane TLRs and C-type lectin receptors (CLRs), and the cytoplasmic NLRs, retinoid acid-inducible gene (RIG)-1 like receptors (RLRs) and absent-in-melanoma 2 (AIM2) like receptors (ALR)66,67. Recently, cyclic GMP-AMP synthase (c-GAS)68 and a family of c-GAS-like receptors (cGLRs)69 have been identified and found to recognize mis-localized cytosolic double stranded DNA (dsDNA), thus triggering NF-κB- and interferon-mediated inflammatory responses. Following MI, DAMPs released by injured cells and damaged matrix act predominantly through binding to members of the TLR and NLR subfamilies. Moreover, RAGE, an immunoglobulin-like receptor that acts as a PRR transducing DAMP-mediated signals, has also been implicated in activation of post-infarction inflammatory cascades.
3.3.1. TLRs:
The TLRs are expressed in both immune cells and in myocardial parenchymal cells (including cardiomyocytes, fibroblasts, and vascular cells), and serve as the main transducers of DAMP-mediated inflammation following cardiac injury. Out of the 10 TLR family members described in humans, TLR1, 2, 4, 5 and 6 serve as cell surface receptors, whereas TLR3, 7, 8, 9 and 10 are expressed in endolysosomes21, 23. TLR2 and TLR4 are the best-studied members of the family in myocardial disease. Myocardial TLR4 expression is increased after acute MI70 and in chronic heart failure71, 72. Moreover, in patients with MI, increased expression of TLR4 and downstream induction of inflammatory cytokines was documented in circulating leukocytes73-75 and correlated with heart failure progression74. Both genetic studies and pharmacologic interventions have suggested that TLR4 plays an important role in extending ischemic injury after infarction by accentuating cardiomyocyte loss and by stimulating inflammatory leukocyte activation76, 77,78. The cellular mechanisms responsible for the injurious TLR4 actions in the infarcted myocardium have not been systematically examined but may involve effects on both cardiomyocytes and immune cells. TLR2 exhibits similar patterns of regulation and effects to TLR4. In patients with MI, high expression of TLR2 was found in circulating monocytes79. Moreover, studies in genetic models demonstrated that TLR2 signaling in leukocytes stimulates pro-inflammatory cascades and increases infarct size in reperfused infarcts 80, 81. Pharmacologic inhibition of TLR2 signaling using a neutralizing antibody had protective effects in both rodent and pig models of MI80,82. In addition to their role in extension of ischemic injury in models of reperfused infarction, TLR2 and TLR4 also contribute to adverse remodeling in non-reperfused infarction83 through actions on inflammatory and fibrotic responses, which are independent of changes in infarct size84.
In addition, endolysosomal TLRs (such as TLR3 and TLR7) have also been implicated in extension of injury after MI. Extracellular RNA-mediated activation of TLR3 signaling was found to extend injury in models of reperfused infarction, increasing the size of the infarct and accentuating inflammatory leukocyte infiltration 85,86. Moreover, TLR7 was upregulated in infarcted human and mouse hearts. Genetic loss-of-function experiments suggested that single stranded RNA (ssRNA) released by dying cardiomyocytes is taken up by leukocytes and activate endosomal TLR7, thus stimulating pro-inflammatory signaling, and promoting cardiac rupture and adverse post-infarction remodeling87.
In addition to their involvement in initiation of the post-infarction inflammatory response, several members of the TLR family (including TLR2 and TLR4) have been implicated in protective preconditioning. Early short-term TLR2 and TLR4 activation prior to ischemia and reperfusion was found to protect cardiomyocytes from ischemic death and may be implicated in ischemic preconditioning88, 89,23, 90, 91. These cardioprotective effects are not specific to TLR activation, but have been observed upon early stimulation with several other pro-inflammatory mediators 92,93, 94. The mechanistic basis for the contrasting cytoprotective and pro-inflammatory actions of innate immune pathways in the myocardium remains enigmatic but may reflect cell-specific actions of TLR signaling in cardiomyocytes vs leukocytes, or concentration-dependent actions.
3.3.2. NLRs.
The members of the NLR family are structurally related PRRs that recognize pathogens or danger signals and activate pro-inflammatory cascades95. Depending on the composition of their N-terminus, NLR proteins can be classified into subfamilies that contain acid transactivation, pyrin, CARD (caspase activation and recruitment) or BID domains. Several of the pyrin domain-containing NLRs (NLRPs) are implicated in assembly of the inflammasomes, multi-protein complexes comprised of an activated NLR protein, the adaptor protein ASC (apoptosis speck-like protein containing a caspase-recruitment domain) and procaspase-1. This complex facilitates proteolytic maturation of caspase-1 and subsequent cleavage and activation of pro-inflammatory mediators, such as IL-1β and IL-18. Caspase-1 also cleaves gasdermin-D, generating an active N-terminal product that translocates to the plasma membrane, oligomerizes and forms a pore, mediating not only secretion of active IL-1 and IL-18, but also osmotic swelling of the cell that leads to a form of death called pyroptosis96. NLRP3 is the best studied of the inflammasome sensors in myocardial diseases and is critically involved in stimulation of inflammatory cascades after MI. All the major components of the NLRP3 inflammasome (ASC, NLRP3 and caspase-1) are upregulated and activated within hours after MI predominantly in infiltrating leukocytes, but also in several other cell types, including fibroblasts vascular cells and border zone cardiomyocytes97-99. Extensive evidence suggests an important role for the NLRP3 inflammasome in activation of the inflammatory response and extension of injury after MI100,. Mice with global loss of NLRP3, ASC or caspase-197,101,102 had reduced infarct size and attenuated ventricular dysfunction in a ischemia/reperfusion model. On the other hand, in models of non-reperfused MI, activation of the NLRP3 inflammasome was found to accentuate inflammation and matrix degradation, thus promising adverse remodeling and dysfunction. Caspase-1 activation accentuated chronic apoptosis of cardiomyocytes in the infarct border zone and enhanced MMP activity causing increased ventricular dilation 103. Moreover, activation of the NLRP3 inflammasome in leukocytes was found to promote chronic adverse remodeling by increasing levels of pro-inflammatory cytokines104. It should be emphasized that protective actions of the NLRP3 inflammasome have also been also reported in models of reperfused infarction and may predominantly involve activation of cytoprotective pathways in cardiomyocytes105. These effects are similar to the protective actions of TLR-mediated innate immune signals discussed in the previous section.
In contrast to the abundance of evidence on the role of the NLRP3 inflammasome, very little is known regarding the role of other inflammasomes in infarcted hearts. Evidence derived from chronic heart failure models (both ischemic and non-ischemic) suggested that the NLRC4 and AIM2 inflammasomes are activated in failing hearts and may be localized in macrophages and in cardiomyocytes106.107. However, the relative contribution of these inflammasomes in activation and regulation of post-infarction inflammation remains unknown.
3.3.3. The c-GAS-STING axis:
c-GAS is an enzyme that acts as a sensor for pathogen-derived or host-derived dsDNA that has entered the cytosol. Upon recognition of cytosolic dsDNA, cGAS synthesizes the nucleotide second messenger cGAMP that directly binds and activates the receptor protein STING (stimulator of Interferon Genes). STING then recruits and activates TANK-binding kinase (TBK)1, subsequently phosphorylating the transcription factor IRF3, which enters the nucleus and induces type 1 interferons. A growing body of evidence has implicated the c-GAS-STING-IRF3 axis in mediating the post-infarction inflammatory response. Uptake of cellular debris by a subpopulation of cardiac macrophages was found to contribute to inflammatory cell infiltration, cytokine upregulation and ventricular dysfunction after MI via activation of a c-GAS/STING/IRF3 cascade108,109. Moreover, pharmacologic inhibition of STING in a model of non-reperfused infarction attenuated adverse remodeling and dysfunction110. Although macrophages are considered major cellular targets of the c-GAS-STING cascade108,109, the potential role of the pathway in other infarct cell types has not been investigated.
3.3.4. RAGE
RAGE is a PRR expressed as a transmembrane receptor in a broad range of cell types, including immune cells, vascular cells, interstitial cells and cardiomyocytes, but also exists in a soluble form (sRAGE) that can be either cleaved through the actions of proteases on the cell surface, or through alternative splicing of the RAGE gene, resulting in generation of endogenous secreted RAGE (esRAGE). Several DAMPs, including HMGB1 and S100A8/9 can activate cell surface RAGE, stimulating downstream NF-κB- and MAPK-dependent pro-inflammatory cascades25. The central role of RAGE in activation of the post-inflammatory response has been demonstrated by experiments in global RAGE knockout mice, which exhibit reduced infarct size, associated with attenuated leukocyte infiltration and inflammatory cytokine expression after reperfused infarction. These protective actions of RAGE loss decrease adverse remodeling and ameliorate dysfunction29, 48. Although some investigations suggested that the pro-inflammatory actions of RAGE may involve actions on endothelial cells111, experiments in bone marrow chimeric mice suggested that leukocytes may be the predominant effectors of RAGE-dependent inflammation after MI48.
3.3.5. Downstream pathways activated by PRRs: a system of adaptor molecules fine tunes the inflammatory response.
Cell surface TLRs respond to binding with various DAMPs in a similar manner: ligand recognition results in formation of dimers and triggers recruitment of the adaptor proteins and downstream activation of pro-inflammatory NF-κB and MAPK cascades. Moreover, a wide range of accessory molecules provide essential signals in regulation of TLR cascades, acting as co-factors for ligand recognition and delivery, or mediating crosstalk with other pathways. Except for TLR3, all other TLRs associate with and signal through MyD88 (myeloid differentiation factor 88), an adaptor protein that also transduces IL-1R signaling. Upon ligand binding, MyD88 undergoes dimerization and recruits the IL-1R-associated kinases (IRAKs), thus forming a molecular platform that recruits TRAF6 (TNF receptor associated factor 6) and ultimately activates downstream pro-inflammatory cascades112,113. Studies using global KO mice demonstrated a central role for MyD88 in activation of the post-infarction inflammatory response114,115. MyD88 loss reduced infarct size in a model of reperfused infarction, through effects attributed to attenuation of inflammatory injury. In non-reperfused infarction, pharmacologic inhibition of MyD88 decreased post-infarction remodeling, despite the absence of effects on the size of the infarct116. The effects of MyD88 in the post-infarction inflammatory response may involve actions on several different cell types, including bone marrow-derived leukocytes117, resident cardiac macrophages118, cardiomyocytes115 and fibroblasts57. A recent study in a model of left ventricular pressure overload suggested important regulatory effects of MyD88 on T lymphocyte phenotype119; however, the significance of these actions in MI has not been investigated. Endosomal TLR4 or TLR3 can also transduce pro-inflammatory signaling through MyD88-independent pathways, by recruiting another cytoplasmic adaptor protein called TRIF (TIR domain-containing adaptor inducing interferon [IFN]-β)86,120,121. This pathway activates both NF-κB and IRF3-mekdiated pro-inflammatory cascades.
3.3.6. Activation of the NF-κB system
Several PRR-mediated pathways converge on the NF-κB system, the central transcriptional effector of pro-inflammatory signaling after MI92. Activation and nuclear translocation of NF-κB is responsible for upregulation of a broad range of pro-inflammatory cytokines, chemokines, and adhesion molecules in the infarcted heart, thus mediating inflammatory leukocyte recruitment and activation. The mammalian NF-κB family is comprised of 5 members (RelA /p65, RelB, c-Rel, p50, and p52) that form homo- or heterodimers to modulate gene transcription. p50/p65 is the predominant NF-κB complex in the heart, where it exists as an inactive cytoplasmic dimer bound to one of the inhibitor of κB family members, IκBα. NF-κB activation requires activation of IκB kinase (IKK), which induces IκBα phosphorylation and subsequent ubiquitination and proteasomal degradation. IκBα loss unmasks the nuclear localization sequence of the NF-kB sequence, resulting in nuclear translocation of the complex and transcriptional activation. In addition to this canonical pathway of NF-κB activation, a non-canonical pathway has been described that involves slow and persistent activation of NF-κB-inducing kinase (NIK), NIK-mediated p100 phosphorylation and subsequent p100 processing and nuclear translocation of non-canonical NF-κB members, such as p52/RelB122. Several TNFR superfamily members (such as the lymphotoxin-b receptor (LTβR) and CD40) are the best-studied activators of non-canonical NF-κB signaling. Dysregulated non-canonical NF-κB signaling in various cell types has been implicated in the pathogenesis of inflammatory and autoimmune conditions122.
In animal models of MI, NF-κB is rapidly activated in the infarct zone within the first 1-3 hours after ischemia123-125. In contrast, NF-κB activation in the remote remodeling myocardium is noted several weeks after infarction, likely reflecting the consequences of pressure and volume loads in animals with heart failure due to large non-reperfused infarcts126. Studies using genetic and pharmacologic interventions to target components of the NF-κB system have produced conflicting results. In mouse models, cardiomyocyte-restricted overexpression of mutant phosphorylation-resistant IκBα127, cardiomyocyte-specific p65 deletion128, IκBα overexpression via gene transfer129, NF-κB double-stranded decoy DNA transfection130, and pharmacological blockade of IκBα131, or IKKβ132 have supported the notion that NF-κB activation extends the size of the infarct, accentuates leukocyte infiltration and increases ventricular dysfunction after reperfused infarction. These effects may predominantly reflect actions on cardiomyocytes that may involve p65. In non-reperfused infarcts, several investigations using mice with cardiomyocyte-restricted overexpression of mutant phosphorylation-resistant IκBα126, the NF-κB signaling inhibitor A20133, and IκBα gene transfer134 also suggested detrimental effects of NF-κB, associated with accentuated adverse remodeling and increased inflammation. Studies using p50 KO mice also suggested a role for p50 in post-infarction remodeling in non-reperfused infarcts135, 136.
On the other hand, several other studies have suggested protective actions of the NF-κB system in the infarcted myocardium that may involve pro-survival effects on cardiomyocytes, but also reparative actions on leukocytes. In models of ischemic preconditioning, acute NF-κB activation has been suggested to play an essential role in late cardioprotection induced by ischemic preconditioning137, 138. In the ischemic and reperfused heart, RelB was found to transduce cytoprotective signaling in cardiomyocytes139. Moreover, endogenous leukocyte p50 was found to protect the remodeling heart after non-reperfused MI, by attenuating inflammation, reducing matrix metalloproteinase activity and improving repair140. The conflicting observations between various in vivo studies likely reflect the distinct characteristics of each intervention on specific components and functions of the NF-κB system. Subunit- and cell-specific actions of the NF-κB system, as well as the efficiency and context of the intervention (reperfused vs. non-reperfused MI, timing, etc) need to be carefully considered to interpret the findings of investigations targeting the NF-κB system in MI.
3.4. Pro-inflammatory cytokines and chemokines
Stimulation of innate immune pathways and downstream activation of NF-κB and MAPK cascades results in secretion of a broad range of pro-inflammatory cytokines and chemokines in the infarcted heart. TNF-α, IL-1 and IL-6 are the best-studied pro-inflammatory cytokines in MI and were found to exert a broad range of actions on all cell types involved in cardiac injury, repair and remodeling. Several other cytokines have been suggested to play roles in modulation of immune cell phenotype, thus contributing to the post-infarction inflammatory response. Cytokines are notoriously pleiotropic and multifunctional. For this reason, cytokine functions on the post-MI inflammatory response include both cytoprotective-reparative and injurious actions. Thus, the consequences of cytokine targeting in the infarcted heart are dependent on the context, timing and on the magnitude duration of cytokine inhibition.
3.4.1. TNF-α
TNF-α is rapidly induced141 and secreted142 in the infarcted myocardium. Experimental studies have suggested both protective and detrimental actions of TNF-α signaling in the infarcted myocardium, reflecting the pleiotropic actions of the cytokine. Global loss of TNF-α or treatment with anti-TNF-α antibody attenuated leukocyte infiltration and reduced infarct size in models of reperfused MI143. In contrast, other studies have demonstrated cardioprotective actions of TNF-α signaling144 that may involve activation of NF-κB-dependent anti-apoptotic pathways in cardiomyocytes145. The conflicting findings may reflect divergent actions of the TNFR receptors, TNFR1 and TNFR2 in the cell types involved in ischemic injury, inflammation and cardiac repair126, 146, 147 and may explain, at least in part, the negative clinical trials of TNF inhibition strategies in heart failure patients.
3.4.2. IL-1
Induction and secretion of IL-1 has been consistently documented in MI patients and in experimental models of coronary ischemia. Extensive experimental evidence from both genetic and pharmacologic inhibition studies suggests a central role for IL-1-mediated signaling in the pathogenesis of cardiac dysfunction and adverse remodeling after MI. Mice with genetic disruption of the type 1 IL-1 receptor (IL-1R1), the only signaling receptor for IL-1α and IL-1β attenuated adverse remodeling after MI, markedly suppressing leukocyte infiltration, but without affecting the size of the infarct148. Pharmacologic targeting of IL-1 cascades also showed protective effects in experimental models of MI, attenuating adverse remodeling149, reducing cardiomyocyte apoptosis150 and suppressing arrhythmogenesis151. IL-1-mediated accentuation of post-MI remodeling and dysfunction has been attributed stimulation of pro-apoptotic signaling and functional depression in cardiomyocytes152,153,154, to pro-inflammatory leukocyte activation12, 155 to induction of endothelial expression of adhesion molecules that promote leukocyte recruiment156, and to stimulation of a protease-expressing, matrix-degrading fibroblast phenotype155. Another member of the IL-1 family, IL-18 has been also suggested to extend inflammatory injury after MI, thus contributing to the pathogenesis of adverse remodeling123,103.
3.4.3. IL-6
IL-6, the prototypical member of the gp130 family of cytokines, is expressed by several different cell types, including cardiomyocytes157, leukocytes142 and fibroblasts158, and plays a central role in regulation of inflammatory and reparative responses after MI. The bulk of the evidence suggests that the effects of IL-6 signaling in the infarcted heart accentuate post-MI inflammation and promote dysfunction. Persistent gp130/STAT3 signaling enhanced inflammation in remodeling infarcted hearts159. Moreover, pharmacologic blockade of IL-6 through administration of an anti-IL6R antibody attenuated dilation and improved function in a model of post-infarction heart failure160. Understanding of the role of IL-6 in healing infarction is hampered by the notoriously pleiotropic and cell-specific actions of the cytokine and by the distinct effects of trans vs cis IL-6 signaling. Classic IL-6 pathways involve binding to the IL-6R on the cell surface, and subsequent association of the IL-6/IL-6R complex with gp130, which dimerizes and initiates signaling. However, it has been suggested that IL-6 can also act on cells that do not express IL-6R on their surface. ADAM proteases can cleave IL-6R from receptor-expressing cells, generating soluble IL-6R (sIL-6R), which can then associate with IL-6, stimulating gp130 signaling on IL-6R-negative cells. This pathway is called “trans-signaling”, and has been suggested to play a major role in inflammatory responses161. In MI models, inhibition of IL-6 trans signaling was found to be more effective in attenuating inflammation and post-MI dysfunction than global IL-6 blockade162. The cellular targets of IL-6 in the infarcted heart have not been systematically investigated. IL-6 exerts important modulatory effects on several different cell types, suppressing function163 and promoting hypertrophy in cardiomyocytes164,165,166 through the gp130-STAT3 pathway, stimulating fibroblast proliferation and activation 167,168, 169 and mediating both pro- and anti-inflammatory effects on macrophages and lymphocytes170,171,172, 173,174.
3.4.4. The chemokines:
Chemokines are a family of small chemotactic cytokines that regulate cell migration and positioning in development, homeostasis and inflammatory injury175. Based on their structure, chemokines can be classified into 2 major subfamilies (CC and CXC chemokines) and 2 much smaller subfamilies (CX3C and C chemokines). This classification has functional implications: most CC chemokines are potent mononuclear cell chemoattractants, whereas the CXC chemokines that contain the glutamic acid-leucine-arginine (ELR) motif in their aminoterminal region (ELR+ CXC chemokines) serve to recruit neutrophils in sites of inflammation. Several members of the chemokine family are upregulated in healing infarcts and are potent regulators of the inflammatory response176. In comparison to the much more pleiotropic cytokines, most chemokines exhibit higher cellular specificity, acting predominantly on trafficking of specific leukocyte subpopulations that express the corresponding chemokine receptors.
3.4.5. CC chemokines
Several CC chemokines have been implicated in chemotactic recruitment of proinflammatory monocytes in the infarcted region177. CCL2/Monocyte Chemoattractant Protein (MCP)-1, the best-studied CC chemokine in myocardial pathology, is rapidly upregulated in the infarcted heart and mediates recruitment of CCR2+ monocytes177. Pharmacologic or genetic disruption of the CCL2/CCR2 axis in reperfused MI was found to reduce infarct size in some 178-181, but not in all studies177, and consistently attenuated post-MI remodeling177, 182. In addition to its protective actions against adverse remodeling, complete CCL2 loss also delayed phagocytic removal of dead cardiomyocytes, perturbing the reparative response177, highlight the dual role of inflammatory pathways in repair and adverse remodeling. Several other CC chemokines have been implicated in activation of the post-MI inflammatory response, including CCL5183, 184, CCL21 and the CCR9 and CCR1 systems185-187. CCL17 was also found to exert pro-inflammatory actions in the infarcted heart, not by recruiting inflammatory leukocytes, but through inhibition of regulatory T cell (Treg) infiltration. CCL17-mediated inhibition of Treg chemotaxis was attributed to an interaction between CCL17 and the chemokine receptor CCR4 that inhibits CCL22-mediated chemotaxis188. It is important to emphasize that CC chemokines are not uniformly pro-inflammatory. Some members of the subfamily may suppress inflammation by recruiting anti-inflammatory subpopulations of monocytes. For example, CCR5 signaling was found to play a role in recruitment of regulatory T cells (Tregs) in the infarct, mediating suppression of the post-infarction inflammatory response189.
3.4.6. CXC chemokines
Several ELR+ CXC chemokines were found to be upregulated in the infarcted myocardium and may play a role in chemotactic recruitment of neutrophils. The absence of a mouse homolog for CXCL8/IL-8, the prototypical human ELR+ CXC chemokine, hampers understanding of the role of specific members of the family in the post-MI inflammatory response. CXCL8/IL-8 is markedly upregulated in large animal models of MI190, 191 and has been implicated in neutrophil recruitment in the infarct in some, but not all studies190,192. On the other hand, mouse studies suggested that CXCR2, the chemokine receptor for most ELR+ CXC chemokines, not only mediates inflammatory leukocyte recruitment, but may also exert direct protective effects on cardiomyocytes193. In addition, IL-8 and other CXCR2 ligands have been suggested to be potent angiogenic mediators194,195. The concept of compartmentalized CCR2-mediated actions that may exert protective effects on cardiomyocytes and vascular cells, but accentuate leukocyte-mediated inflammatory injury has also been supported by experiments investigating the role of another (non-canonical) CXCR2 ligand, the cytokine macrophage migration-inhibitory factor (MIF)196.
In contrast to the effects of the ELR+ subfamily, CXC chemokines lacking ELR cannot attract neutrophils, but have been implicated in lymphocytes chemotaxis197-199. Moreover, a robust body of literature suggests that several ELR-negative CXC chemokines may also modulate fibroblast and vascular cell phenotype, exerting angiostatic and anti-fibrotic actions200-202. CXCL10/ Interferon-γ-inducible protein (IP)-10, the best studied member of this subfamily, is markedly upregulated in both canine and mouse myocardial infarcts203, 204. Studies using a global loss of function model suggested that CXCL10 protects the infarcted heart from excessive fibrotic remodeling, limiting infiltration of the infarct with activated myofibroblast by inhibiting growth factor-induced fibroblast migration205. The anti-fibrotic effects of CXCL10 did not involve its canonical receptor CXCR3 but were mediated through interactions with cell surface proteoglycans206.
CXCL12/Stromal cell-derived factor (SDF)-1 stands out as a homeostatic chemokine with a central role in embryonic development207 and in mobilization, survival and differentiation of progenitor cells. CXCL12 is upregulated in the infarcted heart208-214 and has been suggested to regulate inflammatory, reparative and cell death pathways through effects involving the CXCR4 and ACKR3/CXCR7 receptors. In addition to its role in inflammatory activation215, CXCL12 was also found to exert angiogenic actions216-227 and to inhibit cardiomyocyte apoptosis222, 225, 228. The broad range of cellular effects of CXCL12 may explain the conflicting findings from studies testing the effects of CXCL12/CXCR4 inhibition after MI229,230, 231.
3.5. The cellular effectors of the inflammatory response
A broad range of cell types, both resident myocardial cells and recruited cells from the bone marrow or the spleen (Figure 2), are implicated in the post-infarction inflammatory response. These cells co-operate to clear the infarct from dead cells and matrix debris and to set the stage for stimulation of a reparative program.
Figure 2: Temporal course of immune cell accumulation in myocardial infarction (MI) is linked to phase-specific cell functions.
ROS, reactive oxygen species. NET, neutrophils extracellular traps. LCN2, lipocalin 2. IL, interleukin. VEGF, vascular endothelial growth factor. SPP1, secreted phosphoprotein 1. TGF, transforming growth factor.
3.5.1. Cardiomyocytes as pro-inflammatory cells
Dying cardiomyocytes contribute the central stimulus that activates post-MI inflammation by releasing DAMPs that transduce innate immune signals16. Moreover, surviving cardiomyocytes in the infarct border zone are subjected to mechanical stress and are exposed to a cytokine-rich environment. These unique microenvironmental conditions may promote synthesis and release of inflammatory mediators that may contribute to the intense infiltration of the infarct border zone with leukocytes. Studies in large animal models have demonstrated that cardiomyocyte in the viable border zone may be a significant source of cytokines, such as IL-6157. Considering that several other cell types can produce large amounts of cytokines the relative contribution of border zone cardiomyocytes as pro-inflammatory cells is unknown.
3.5.2. Resident and recruited macrophages
Macrophages are the most prevalent immune cell type residing in the normal mammalian heart. Primitive macrophages infiltrate the developing heart early during embryogenesis and assist in coronary vessel maturation. Their progeny can persist into adulthood through self-renewal in situ. In addition, and over time, monocytes infiltrate the healthy heart and contribute to the resident cardiac macrophage population. Cardiac macrophages in mice and humans have been classified into at least two major subpopulations according to their differential expression of the chemokine receptor CCR2. While monocytes give rise to CCR2+ macrophages, the CCR2− population harbors embryonically derived macrophages with divergent reports regarding the extent of substitution by monocyte-derived cells according to lineage tracing experiments in mice and analysis of sex-mismatched heart transplants or mutation variant allele frequencies in humans232,233,234,235. Differential gene expression profiles of CCR2+ and CCR2− cardiac macrophages share great similarities between mice and humans, supporting the translational value of the mouse model.
Cardiac macrophages preserve tissue homeostasis through efferocytosis of cardiomyocyte exosomes containing damaged cell organelles, electrical coupling to cardiomyocytes, and defense against pathogens236,237. With aging and continuous monocyte recruitment, however, release of the anti-inflammatory cytokine IL-10 by cardiac macrophages can induce a profibrotic phenotype which promotes diastolic dysfunction in mice238. IL-10 is considered a prototypical M2-like macrophage cytokine; however, the extensive use of single cell technologies for characterization of cell profiles has demonstrated that the dichotomous M1/M2 macrophage nomenclature does not adequately reflect primary cardiac macrophage heterogeneity.
Single cell RNA-seq cluster analysis identifies at least three major macrophage subpopulations in the healthy adult murine heart. Macrophages expressing high levels of TIMD4, FOLR2 and/or LYVE1, but low levels of MHC II and CCR2, are commonly referred to as the resident population. This marker profile is relatively preserved across different tissues and organs, the respective population shows little or slow replacement by recruited monocytes, and transcriptional activation of endocytosis, angiogenesis and regeneration related pathways234,239,240. Two LYVE1low MHC IIhigh populations can be distinguished based on CCR2 expression with more pronounced or complete monocyte dependence. In line with high MHC II expression, their transcriptional profiles are associated with antigen processing and presentation capacity and inflammatory pathways which are particularly enriched within the MHC IIhigh CCR2+ subset 241. Another minor inflammatory cell cluster features many type I interferon response genes and is termed according to its signature gene Isg15 (interferon-stimulated gene 15) or IFNIC (interferon-inducible cells). These heterogeneous cardiac macrophage populations react differentially in response to MI. In murine models of non-reperfused infarction, most tissue resident macrophages in the infarct area die and are replaced by recruited monocytes and their progeny. In the remote myocardium or with timely reperfusion of the ischemic area, many of the original macrophages survive and expand through local proliferation supplemented by newly recruited cells242,243. Notably, MHC IIhigh CCR2+ macrophages residing in the tissue prior to MI are activated via TLR signaling and attract monocytes and neutrophils to the infarcted heart within few hours following MI118,244,245. The pericardial cavity has been proposed as an additional, monocyte-independent source of infarct infiltrating macrophages246; however, the relevance and magnitude of this process has been challenged247.
Depleting all monocytes and cardiac macrophages by clodronate liposomes impairs proper healing of experimental MI248,249, underscoring the critical role of cardiac macrophages in clearing cell debris and orchestrating granulation tissue formation. Depleting CCR2+ resident cardiac macrophages selectively while preserving the CCR2− resident population prior to MI, or limiting the recruitment of monocytes into the infarct in the inflammatory phase improves subsequent cardiac remodeling, but imposes an increased risk of acute post-MI ventricular arrhythmia118,250. Depleting resident cardiac macrophages while allowing for monocyte influx and replacement is detrimental to MI healing, both in terms of structural remodeling and arrhythmogenesis118, 250. Mechanistically, macrophages phagocytosing apoptotic cells (efferocytosis) in the infarct during the inflammatory phase is essential for preventing arrhythmia and secondary necrosis which counteracts the resolution of tissue inflammation and transition into the proliferative phase of MI healing, during which efferocytosis receptor expression peaks251, 250. In addition, amphiregulin released by cardiac macrophages controls connexin 43 phosphorylation and translocation in cardiomyocytes and suppresses arrhythmia252. Dying cardiomyocytes overexpress the “don’t eat me signal protein” CD47 on their surface which binds to Signal-regulatory protein alpha (SIRPa) on cardiac macrophages hindering efferocytosis. Blockade of CD47 at the onset of MI improves clearance of apoptotic cardiomyocytes and cardiac remodelling253. However, uptake of apoptotic cells can lead to intracellular sensing of nucleic acids, activation of interferon regulatory factor 3 (IRF3) pathways and induction of type I interferons (IFN) which amplify the inflammatory response108. Phagolysosomal destabilization and oxidative stress-related uncoupling of the thioredoxin-interacting protein within macrophages, and P2X7-receptor binding of extracellular ATP released in the infarct, facilitate potassium efflux and subsequent activation of the NLRP3 inflammasome. Its expression peaks within the first three days post MI. NLRP3 activation and oligomerization leads to the release of mature IL-1β, the hallmark pro-inflammatory cytokine, via Gasdermin pores in the cell membrane254. Gasdermin D deficiency reduces IL-1β and IL-18 release in the infarct255. Other gene signatures of infiltrating monocytes and early or transient cardiac macrophage populations that dominate the inflammatory phase are related to hypoxia, leukocyte migration, T cell activation, interferon-response, glycolysis, proliferation and extracellular matrix degradation 256,257,258,259.
3.5.3. Mast cells
In addition to macrophages the adult mammalian heart also contains smaller populations of resident mast cells260,261 and dendritic cells262,263. Mast cells are strategically located around vessels and degranulate in response to activating signals, such as DAMPs, adenosine, or components of the complement cascade, thus releasing their pre-formed pro-inflammatory mediators and rapidly stimulating the immune response. TNF-α, histamine and tryptase released by degranulating mast cells may activate macrophages and endothelial cells, thus promoting inflammation142,264.
3.5.4. Fibroblasts
The normal heart contains large numbers of interstitial and perivascular fibroblasts 265 that regulate homeostasis of the extracellular matrix network. During the inflammatory phase of cardiac repair, stimulation with TLR ligands, or with cytokines delays myofibroblast conversion and induces a pro-inflammatory and matrix-degrading fibroblast phenotype, associated with secretion of cytokines and chemokines, and with release of matrix metalloproteinases (MMPs)155. Considering that several other cell types (including cardiomyocytes, macrophages and endothelial cells) can also produce the same pro-inflammatory mediators, the relative significance of fibroblast-derived inflammatory cytokines is unclear. Studies investigating the role of fibroblast-specific inflammatory activation are lacking and the evidence supporting the pro-inflammatory roles of fibroblasts in the infarcted heart is for the most part associative. In experimental models of MI, infarct fibroblasts activate the NRLP3 inflammasome97,99, and could serve as an important source of active IL-1β. Moreover, infarct fibroblasts secrete large amounts of granulocyte/macrophage colony-stimulating factor (GM-CSF) 266, a mediator involved in monocyte recruitment, and are a major source of IL-6158.
3.5.5. Endothelial cells.
The heart is a highly vascular organ; thus, endothelial cells are the most abundant non-cardiomyocytes in the adult mammalian myocardium265. Endothelial cells play a central role in the post-infarction inflammatory response by secreting cytokines and chemokines and by interacting with leukocytes, thus promoting their transmigration to the infarct. DAMPs released by dying cells inflammatory cytokines (such as IL-1 and TNF) and autacoids (such as histamine) stimulate endothelial cells in the infarct zone, inducing upregulation of endothelial adhesion molecules, thus triggering adhesive interactions with activated leukocytes. Histamine rapidly mobilizes preformed P-selectin from Weibel-Palade bodies to the endothelial cell surface through effects involving H1 receptors267,268. E-selectin is also upregulated in the infarct endothelium in response to the cytokine-rich environment. Upon expression on the endothelial cell surface, selectins engage in weak transient interaction with their leukocyte ligands, capturing leukocytes and mediating rolling along the endothelium 269,270. Subsequently, rolling leukocytes expressing specific chemokine and cytokine receptors interact with the corresponding ligands, which are bound to glycosaminoglycans on the endothelial cell surface. These interactions induce conformational changes of leukocyte integrins (such as LFA-1, Mac-1 and VLA-4) that strengthen the adhesive interaction 271,272, resulting in arrest and firm adhesion of the leukocyte to the endothelial surface. The integrin-mediated adhesive interactions involve binding to endothelial ICAM-1 or VCAM-1, which are upregulated on the endothelial cell surface in response to cytokine stimulation59. Leukocyte transmigration follows. Leukocytes actively crawl towards “transmigration hotspots”, regions of the endothelial and pericyte layers that serve as exit points for extravasating cells273,274,275.
3.5.6. Pericytes
The heart also contains a large population of pericytes 276,277. During the inflammatory phase, cytokine-mediated actions may perturb the capillary barrier function of pericytes increasing microvascular permeability278. Moreover, infarct pericytes were found to exhibit a migratory phenotype279 and may dissociate from their close relation with endothelial cells. The cytokine pro-nerve growth factor (pro-NGF), is upregulated in ischemic hearts and may promote microvascular dysfunction through effects on pericyte p75 NGF receptor signaling280. Although pericytes can express cytokines and chemokines in response to DAMPs, their relative role as inflammatory cells in MI has not been studied. Regardless of their significance as cellular sources of inflammatory mediators, pericytes are likely to serve as key regulators of leukocyte trafficking274. Perhaps, the best-documented function of pericytes in the inflammatory phase of MI is their role in mediating the “no-reflow phenomenon”, the failure of subendocardial microvascular reperfusion of the ischemic myocardium despite full restoration of coronary blood flow281. In a rat model of myocardial ischemia/reperfusion, sites of microvascular occlusion were spatially associated with pericytes, suggesting that pericyte constriction may contribute to leukocyte entrapment within the microvessels. Adenosine administration to induce pericyte relaxation supported the proposed role of pericytes in microvascular occlusion282. It should be emphasized that other cell types, including leukocytes and endothelial cells, likely also contribute to the pathogenesis of no-reflow.
3.5.7. Neutrophils
Neutrophils dominate the tissue infiltrate within the first 24h after MI5, 283, Neutrophils are recruited in the infarct in response to a broad range of chemoattractants, including cytokines and chemokines, endogenous lipid mediators (such as prostaglandin E2 and leukotrienes), histamine, and components of the complement cascade284-286. Extravasation of neutrophils in the infarct requires activation of adhesive interactions with endothelial cells and pericytes. Neutrophils are major contributors to post-infarction inflammation and act by releasing their granular contents (that include cytokines and proteases), by generating reactive oxygen species (ROS), by forming extracellular traps (NET), and by phagocytosis cells and matrix debris. Neutrophils releasing S100A8/9 alarmins prime NLRP3 activation in an autocrine feedback loop leading to IL-1b production, which in turn stimulates granulopoiesis in the bone marrow by reverse-migrating activated neutrophils49,287. Neutrophil derived lipocalin-2 (Lcn2) stimulates ROS production in MI tissue and facilitates ventricular arrhythmia250. Inhibition of myeloperoxidase (MPO) released by neutrophils in the infarct improves cardiac remodeling particularly when initiated within the first 24 hours post-MI288,289. In contrast, neutrophil NADPH oxidase-deficiency did not influence the infarct size290. In analogy to the M1/M2 nomenclature in macrophage polarization, a biphasic response of pro-inflammatory N1 dominating the inflammatory phase followed by anti-inflammatory CD206+ IL-10 producing N2 neutrophils in the proliferative phase has been proposed291. However, recent scRNAseq analysis has painted a more nuanced and diversified picture distinguishing up to 6 clusters of post MI neutrophils in a time-dependent manner292. During the acute phase post MI infiltrating Siglecflow neutrophils express high levels of inflammatory Lcn2, Oncostatin-M and Cxcl3. From day 3 post MI onward, a large fraction of neutrophils in the MI tissue expresses high levels of Siglecf, a surface marker shared with eosinophils, which they acquire in the tissue alongside ICAM1 expression292. Neutrophils have a short lifespan and become apoptotic, requiring clearance by macrophages. Neutrophils may undergo a specific form of cell death termed NETosis while releasing extracellular DNA and cell lytic histones and enzymes. Inhibition of NETosis limits MI inflammation and injury293. Studies depleting neutrophils after MI have produced conflicting results that may reflect, at least in part, difference in timing and effectiveness of depletion. Neutrophil depletion restricted to the time of experimentally induced MI improves infarct healing by limiting early inflammation289,294. However, prolonged neutrophil depletion during the inflammatory and into the proliferative phase post MI has detrimental effects295. Mechanistically, lack of neutrophil-derived LCN2 inhibits the development of reparative macrophages with high efferocytosis capacity in the late infarct. Similarly, neutrophil-derived Annexin A1 induces a pro-angiogenic cardiac macrophage phenotype post MI and limits inflammatory cell recruitment296. Interestingly, MMP-12 expression by cardiac macrophages increases during the late inflammatory phase, with MMP-12 degrading neutrophil chemoattractants. Thus Ly6Clow macrophages arising from infiltrating Ly6Chigh monocytes242 contain neutrophil influx and facilitate the transition from the inflammatory to the proliferative phase of MI healing297.
3.5.8. Monocytes
The peak of monocyte recruitment in the infarcted heart follows the wave of neutrophil infiltration. In mice, two monocyte subsets are distinguished by differential Ly6C and CCR2 expression. Ly6Chigh monocytes infiltrate the injured heart and give rise to cardiac macrophages. Ly6CLow monocytes derive from Ly6Chigh monocytes and patrol the vasculature rather than differentiating into MI macrophages242. Murine Ly6Chigh/CCR2+ monocytes correspond to the CD14high/CD16low classical and CD14high/CD16high intermediate monocyte subsets in humans, whereas murine Ly6Clow/CCR2− monocytes correspond to CD14low/CD16high non-classical monocytes in humans298, although scRNAseq and mass cytometry studies have identified additional monocyte subtypes in human blood 299,300. In human populations, high numbers of classical or intermediate monocytes associate with future cardiovascular events predict adverse outcomes after MI301,302,303.
In-vivo time-lapse imaging in mice suggests that monocytes rolling in coronary venules are the first to infiltrate the infarct within 30 minutes post MI, preceding neutrophil invasion which, however, dominates cell infiltration within the first 24 hours post MI quantitatively244. Subsequently, monocytes are mobilized from the splenic reservoir in an angiotensin II dependent manner10,304. CC chemokines, such as CCL2 and CCL7, released from endothelial cells, CCR2+ cardiac macrophages and stromal cells recruit Ly6Chigh monocytes to the activated endothelium and facilitate transmigration into the injured myocardium176,177,118,257,243. Monocytes and macrophages infiltrating the infarct are a major source of pro-inflammatory cytokines, such as IL-1β, and in turn IL-1β stimulates both medullary and extramedullary monocytopoiesis and cell recruitment to the infarct305,12,148. During emergency myelopoiesis, monocytes acquire granulocyte marker transcripts306. In the chronic phase post MI, endogenous beta-adrenergic stimulation of the bone marrow niche suppresses hematopoietic quiescence, inhibits the retention factors CXCL12 and Angiopoietin-1, and stimulates extramedullary hematopoiesis243. Nevertheless, the precise pathological significance of monocytes in the healing process of MI remains unclear, primarily due to the absence of experimental tools that allow for the selective study of their role without interfering with their differentiation into cardiac macrophages.
3.5.9. Lymphocytes
Lymphocytes are recruited in the infarcted heart along with monocytes283,307. Most of the studies on the role of lymphocytes in cardiac repair have focused on their anti-inflammatory actions and their modulatory effects on macrophage phenotype during the proliferative phase of cardiac repair. Data on potential pro-inflammatory actions of lymphocytes are limited and predominantly suggest effects of B lymphocytes. B-cells infiltrate the infarcted heart during the first 24h after MI and persist during the proliferative phase283. ScRNA-seq showed accumulation of diverse polyclonal B-cell subsets in infarcted mouse hearts without evidence of antigen specificity, paralleled by mild clonal expansion of germinal center B cells in the mediastinal lymph nodes308. The role of B cells in regulation of the post-infarction inflammatory response remains understudied. However, Zouggari et al 309 showed that in the infarcted myocardium mature B-cells produce the chemokine CCL7, mobilizing pro-inflammatory monocytes that infiltrate the myocardium. Inhibition of B-cells through CD20 neutralization improved left ventricular remodeling after MI in mice309. In addition to production of monocyte chemoattractant chemokines, B cells have also been suggested to serve as an important source of TGF-β1308.
Moreover, associative data on the kinetics of lymphocytes in human STEMI patients suggested that CX3CL1/fractalkine may mediate recruitment of a pro-inflammatory subset of effector T cells in the infarct, promoting microvascular occlusion and accentuating injury310. The role of T cells in MI healing is discussed in detail in section 4.2.4.
4. The proliferative phase
The transition to the proliferative phase of cardiac repair is marked by suppression of the inflammatory response and subsequent activation of fibrogenic and angiogenic cascades. Macrophages, lymphocytes, fibroblasts and vascular cells co-operate to form a highly cellular granulation tissue, ultimately leading to deposition of a matrix network that protects the infarcted heart from catastrophic rupture.
4.1. Mediators involved in suppression and resolution of inflammation.
Timely suppression of the post-infarction inflammatory response is not a passive process that simply reflects the transient nature of pro-inflammatory cytokine and chemokine induction but is dependent on de novo synthesis and release of anti-inflammatory mediators and on activation of intracellular STOP signals that inhibit the innate immune response. Anti-inflammatory cytokines (such as IL-10) and certain members of the TGF-b superfamily are the best studied mediators that have been implicated in suppression of post-infarction inflammatory response 311. Defective suppression, resolution and containment of inflammation after infarction may underlie adverse remodeling in patients surviving MI.
IL-10 is a potent anti-inflammatory cytokine that inhibits expression of pro-inflammatory cytokines and chemokines in macrophages312 through activation of the STAT3 cascade313. IL-10 is markedly upregulated in healing infarcts307,204, and has been localized in T lymphocytes307 (with particularly high levels in regulatory T cells/Tregs189), in subsets of macrophages307 and in regulatory B cells314. Although studies in IL-10 KO mice support the role of endogenous IL-10 in negative regulation of pro-inflammatory cytokines in healing infarcts315, 316 the evidence on the functional impact of IL-10 actions in the infarcted heart is conflicting. In a mouse model of myocardial ischemia/reperfusion, IL-10 loss was associated with markedly increased early mortality315, whereas in another study using a closed-chest model of reperfused infarction and a much higher sample size showed no effects of IL-10 absence on mortality or dysfunction, despite increased myocardial levels of TNF and CCL2316. Several investigations suggested that administration of exogenous IL-10 can improve remodeling after MI, at least in part by attenuating the inflammatory response317,318.
The anti-inflammatory cytokines IL-4 and IL-13 have also been implicated in negative regulation of post-infarction inflammation and repair after MI and can be secreted, not only by macrophages and lymphocytes, but also by eosinophils319,320 and basophils321 that infiltrate the infarct. Myeloid cell-specific IL-4Rα signaling was found to protect the infarcted heart from adverse remodeling, at least in part by suppressing inflammation and by regulating matrix remodeling322. Exogenous administration of IL-4 and of IL-13 were suggested to exert protective actions after infarction by promoting an anti-inflammatory and reparative macrophage phenotypes323,324
Several members of the TGF-β superfamily (including TGF-βs and Growth Differentiation Factor (GDF)-15) exert potent anti-inflammatory actions and have been implicated in negative regulation of the post-infarction inflammatory reaction. The anti-inflammatory actions of TGF-βs may involve effects on both macrophages and lymphocytes. Although TGF-βs are potent monocyte chemoattractants, their effects on macrophages are anti-inflammatory promoting an M2-like phenotype325. TGF-β-mediated inhibition of macrophage expression of pro-inflammatory cytokines and chemokines326,327,328,329 has been attributed to MyD88 degradation, resulting in attenuated NF-κB activity330. Moreover, TGF-βs have profound effects on T cells, inhibiting Th1 helper and cytotoxic T cell responses331,332, while enhancing differentiation of anti-inflammatory Tregs333. The in vivo significance of the anti-inflammatory actions of TGF-β in MI are supported by neutralization experiments using gene therapy with the extracellular domain of the type II TGF-β receptor (TβRII). Early TGF-β inhibition worsened dysfunction accentuating the inflammatory response, whereas late disruption was protective, reflecting the role of TGF-β in activation of fibroblasts334, thus supporting the notion that TGF-β may act as a “master switch” in the transition from inflammation to fibrosis. The anti-inflammatory effects of TGF–β on infarct macrophages involve activation of Smad3, but not Smad2335,336.
Another member of the TGF-β superfamily, GDF-15 exerts potent anti-inflammatory actions by inhibiting chemokine-mediated leukocyte integrin activation. In mice, GDF-15 loss is associated with fatal cardiac rupture after infarction, caused by accentuated post-infarction inflammation337. In STEMI patients, elevated plasma GDF-15 levels were found to predict mortality338, possibly reflecting activation of an anti-inflammatory pathway in patients with an accentuated post-infarction inflammatory reaction.
A broad range of pro-resolving lipid mediators, such as the resolvins, lipoxins, protectins and maresins339 potently inhibit inflammation and contribute to resolution of the inflammatory infiltrate340. Whether release of endogenous pre-resolving lipids plays a role in suppression of inflammation after MI is not known. However, several studies have suggested that administration of exogenous mediators, such as resolvins E1 and D1 and 15-epi-lipoxin protect the infarcted heart, attenuating post-infarction inflammation341, 342,343.
4.2. The cellular basis of cardiac repair
4.2.1. The role of macrophages in suppression of inflammation and in repair.
As the influx of monocytes subsides, cardiac macrophages become proliferative and acquire reparative properties, secreting growth factors and matricellular proteins and stimulating fibroblast activation and angiogenesis. In fact, efferocytosis can induce non-inflammatory macrophage proliferation344, and contribute to the re-emergence of steady state macrophage progenies in the infarct258,241. Efferocytosis also reprograms macrophages to express inflammation-resolving and reparative mediators345,346. The majority of accumulating infarct macrophages during the proliferative phase of repair are monocyte-derived but form heterogenous subpopulations. One-third of them re-acquire resident cell-like profiles. Other macrophages retain an inflammatory signature whereas many exhibit a lipid-associated macrophage gene signature similar to atherosclerotic plaque-associated foam cells and characterized by triggering receptor expressed on myeloid cells 2 (TREM2) expression. TREM2 can be released by these macrophages and suppresses inflammation in an autocrine and paracrine fashion258,347.
Efferocytosing macrophages also secrete reparative matricellular proteins and growth factors, such as Spp1 (encoding osteopontin) and TGF-βs258,347. Osteopontin induction is a prominent characteristic of reparative infarct macrophages and is mediated by cytokines (such as IL-10)348 and chemokines (such as CXCL4)349. Macrophage-derived TGF-β on the other hand plays a central reparative role by activating fibroblasts and by exerting autocrine actions on macrophages that trigger a phagocytic profile (through activation of Smad3 signaling) and stimulate expression of anti-inflammatory mediators335. In addition to their actions in regulation of inflammation and in fibroblast activation, infarct macrophages also support the formation of new blood and lymphatic vessels (Figure 3). In the infarcted heart, neovascularization arises mainly from preexisting endothelial cells starting from the border zone and extending into the necrotic core350,351. Infarct macrophages serve as a major source of the potent angiogenic growth factor vascular endothelial growth factor (VEGF) A352. Several mechanisms have been implicated in VEGF upregulation in infarct macrophages. First, efferocytosis induces VEGFs in cardiac macrophages, stimulating formation of both blood and lymphatic vessels352,353. Second, binding of components of the provisional matrix network, such as fibronectin, to macrophage integrin α5 on the macrophage surface was found to induce VEGF synthesis and release, enhancing infarct angiogenesis and improving function354. Third, neutrophils stimulate an angiogenic VEGF-expressing macrophage phenotype by secreting Annexin A1 that binds to formyl peptide receptor 2 on cardiac macrophages296.
Figure 3: The temporal dynamics of angiogenesis in healing myocardial infarction.
During the inflammatory phase, pericytes in the infarct zone disassociate from endothelial cells (EC), increasing microvascular permeability. During the proliferative phase, efferocytosis and components of the provisional extracellular matrix (such as fibronectin) stimulate an angiogenic phenotype in macrophages, promoting expression of Vascular Endothelial Growth Factor (VEGF) and deposition of angiogenic matricellular proteins. Macrophages and fibroblasts contribute to an angiogenic environment, resulting in formation of abundant microvessels that lack a mural cell coat. As the scar matures, pericytes are recruited through pathways involving PDGF-BB-PDGFRβ and TGF-β–TGFBR2 interactions, thus forming coated mature microvessels that stabilize the scar. PDGF, Platelet=Derived Growth Factor; TGF, Transforming Growth Factor.
4.2.2. Dendritic cells:
Using cardiac lymphatics, immune cells traffic to draining mediastinal lymph nodes355, where dendritic cells (DC) act as antigen presenting cells linking innate to adaptive immune responses356,357. Like macrophages, myocardial DCs are a heterogenous cell population, albeit much less abundant: Two conventional DC (cDC) subsets, CD103+ cDC1 cells and CD11b+ cDC2 cells, a PDCA1+ plasmacytoid (p)DC population of non-monocyte origin, and a population of CD64+ monocyte-derived cells (moDC) are found in the heart by differential marker expression358,357. During the proliferative phase of cardiac repair, cDC and moDC numbers peak in the heart and in draining mediastinal lymph nodes (mLN), with moDC dominating the infarct infiltrate and cDC2 dominating the mLN infiltrate. Post-MI matured and activated cDC2 are most efficient but not exclusive in presenting cardiomyocyte self-antigens to auto-reactive T cells in the mLN357. Controversies exist regarding the role of cDC in MI, likely arising from the use and interpretation of different experimental models. Diphtheria toxin-mediated depletion in CD11c-DTR mice resulted in enhanced and prolonged inflammation in the infarct and adverse cardiac remodeling, suggesting a central role for DCs in negative regulation of inflammation359. However, non-selective DC depletion and dependence of the model on lethal irradiation and reconstitution may represent potential confounders. In contrast, selective cDC depletion using Zbtb46-DTR mice suppressed inflammation in the infarct and improved cardiac function358. Clec9a-deficiency impairs cross-presentation of cDC1 to cytotoxic CD8+ T cells and improves cardiac remodeling. Depletion of pDC in mice does not impact MI healing significantly358. On the other hand, subcutaneous administration of tolerogenic DC generated ex vivo by exposure to cardiac lysate from MI mice can induce MI-specific regulatory T cells which suppress inflammation and improve cardiac remodeling360. Whether the divergent effects of DC in the heart depend on T cell intermediaries or direct interactions with stromal cells is unknown.
4.2.3. The potential role of basophils and eosinophils
In addition to the rapid and intense infiltration with neutrophilic granulocytes (discussed in section 3.5.7), infarcts also recruit much smaller numbers of basophilic and eosinophilic granulocytes that have been suggested to play important cells in regulating inflammation and repair. Similar to neutrophilia361, elevated eosinophil counts after infarction are associated with adverse outcome362, whereas high basophil counts were found to predict better outcomes in acute coronary syndromes321 and worse outcomes in stable coronary artery disease363. Recruitment of basophils and eosinophils in the infarct lags that of neutrophils, and experimental evidence suggests that both these cell types may exert protective reparative actions after myocardial infarction. Basophils are a major source of IL-4 and IL-13 in the infarct tissue and instruct a reparative phenotype in macrophages321. A similar protective mechanism of paracrine macrophage polarization was proposed for eosinophils infiltrating the infarcted heart320. Eosinophil-derived IL-4 and eosinophil-associated RNase 1 were found to mitigate cardiomyocyte apoptosis and fibroblast activation319. These eosinophil-mediated protective effects can be exploited therapeutically by injecting IL-5 in mice undergoing myocardial infarction364.
4.2.4. T-lymphocytes in cardiac repair
Several studies have suggested important roles for T lymphocytes in repair of the infarcted heart. CD4+ T cells infiltrate the infarcted myocardium during the proliferative phase and play a central role in infarct healing. The role of CD4+ T-cells was studied using CD4 KOs and mice that exhibit defective T cell antigen recognition (OT-II mice). Both mouse strains exhibited perturbed repair after MI, associated with left ventricular rupture and adverse remodeling365. T-cells can be activated through antigens binding to its specific T-cell receptor. The above-mentioned almost identical phenotype in CD4-KO and OTII mice suggests antigen-dependent mechanisms in myocardial healing after MI. By screening over 150 class-II-restricted epitopes, myosin heavy chain (MHC) was identified as a dominant cardiac antigen. To address the role of T-cell dependent MHC-autoimmunity T cells specific to a MYHCA614-629 (myosin heavy alpha chain-derived peptide), termed TCR-M cells, were transferred in wildtype mice before MI. Those cells accumulated in the myocardium and in mediastinal lymph nodes, which was associated with improved cardiac healing366. Characterization on a single cell level revealed that TCRM cells rapidly differentiated in myosin-specific regulatory CD4+ T cells in the infarcted myocardium. This suppressed in situ inflammation and improved healing while supporting stromal cell function associated to tissue repair367. This is especially interesting before the background that TCRM mice, i.e. mice that have a lifelong exposure to TCRM, develop autoimmune myocarditis and finally a phenotype of lethal dilated cardiomyopathy368 This indicates that short episodes of autoimmunity may be beneficial for healing, in contrast prolonged autoimmunity may result in a maladaptive phenotype.
In addition to CD4+ T cells, several other lymphocyte subsets were found to contribute to cardiac repair by regulating inflammation and by modulating macrophage and fibroblast phenotype. Regulatory T cells (Tregs) infiltrate the infarcted myocardium through chemokine-dependent mechanisms189. Studies using both genetic and antibody-based approaches to deplete Tregs after MI demonstrated that CD4+/Foxp3+ Tregs promote repair, attenuating post-infarction inflammation.369,370. The effects of Tregs were attributed to modulation of macrophage phenotype towards an anti-inflammatory profile369 and to inhibitory effects on fibroblast-mediated protease expression370. Other studies have suggested important reparative roles for invariant Natural Killer T cells (iNKT cells) in reperfused371 and in non-reperfused MI372, mediated in part through release of anti-inflammatory cytokines, such as IL-10. The role of CD8+ T cells in the infarcted heart remains poorly defined. Absence of functional CD8+ cells was associated with severe healing defects and increased inflammation after MI, resulting in left ventricular rupture373.. However, surviving mice with deficient CD8+ cells had improved post-MI remodeling. The basis for these conflicting observations is unclear.
Thus, several T cell populations (including CD4+ T cells, Tregs, iNKT cells and possibly also CD8+ T cells) are involved in negative regulation of post-infarction inflammation. In contrast, CD4−γδT-cells were found to promote neutrophil and macrophage infiltration and exert detrimental effects in cardiac remodeling374. Maladaptive phenotypic changes in T cells have also been implicated in progression of chronic post-infarction heart failure375,376
4.2.5. Activated fibroblasts mediate repair of the infarcted heart.
Expansion of the fibroblast population is the hallmark of the proliferative phase of cardiac repair. In the healing infarct, activated fibroblasts are the main source of structural collagens 377,378, and mediate reparative fibrosis, forming an organized scar that protects the ventricle from catastrophic rupture8. Infarct fibroblasts exhibit remarkable heterogeneity. Emergence of fibroblast-like cells that express contractile proteins (such as α-smooth muscle actin (α-SMA)), termed myofibroblasts, is a common characteristic of healing wounds in many different organs, including the heart379,380. Although infarct myofibroblasts exhibit increased fibrogenic acitivity381,377,382, the basis for their accentuated matrix-synthetic capacity is unclear, and is not caused by increased α-SMA expression383. Over the last 5 years, single cell transcriptomic studies have contributed additional insights into the diversity of infarct fibroblasts384, and defined clusters with distinct transcriptional profiles257. High expression of Cthrc1 (encoding the glycoprotein collagen triple helix repeat containing 1) identified a subpopulation of reparative infarct fibroblasts that protect from rupture385. Expression of Cilp (the gene encoding the matricellular protein Cartilage Intermediate Layer Protein 1 (CILP1)) may identify another activated fibroblast subpopulation386 with proliferative properties and a role in maladaptive fibrotic remodeling after infarction387.
The origin of the expanding population of infarct fibroblasts has been hotly debated. Although early investigations suggested that endothelial cells388, or myeloid cells (including hematopoietic myeloid progenitors and macrophages)389,390 may undergo fibroblast conversion after infarction, accounting for significant fractions of infarct fibroblasts, more recent investigations using robust lineage tracing strategies showed that the majority of infarct fibroblasts and myofibroblasts originate from resident fibroblast populations that proliferate in the growth factor-rich environment if the infarct391,392. Lineage tracing studies coupled with single cell transcriptomics demonstrated that a small fraction (~4-5%) of infarct fibroblasts are derived from pericytes 279. Although myeloid cells and vascular cells exhibit very limited conversion to fibroblasts, their contribution to fibroblast activation is significant, due to their high expression of fibrogenic growth factors and matricellular proteins279,278,348.
4.2.6. Molecular signals that activate infarct fibroblasts.
Fibroblast activation in healing infarcts involves the co-operation of a broad range of mediators, including neurohumoral mediators, cytokines, growth factors, and matricellular proteins (Figure 4). Angiotensin II potently activates infarct fibroblasts, stimulating fibroblast proliferation promoting myofibroblast conversion, and inducing synthesis of extracellular matrix proteins. These fibroblast-activating effects of angiotensin II are mediated through the angiotensin II type 1 receptor (AT1R) 393,394,395,396; in contrast the AT2 receptor has been suggested to play an anti-fibrotic role, suppressing AT1R-mediated fibroblast activation397,398. Although angiotensin II is a potent activator of fibroblasts, AT1R blockade in both patients with MI and in experimental models of infarction not only does not perturb repair but exerts protective actions399. To what extent the protective effects of AT1R blockade are mediated through inhibition of maladaptive fibrosis is unclear. Angiotensin II has a broad range of effects on many different cell types, including cardiomyocytes, macrophages, vascular cells and fibroblasts that may be involved in adverse post-infarction remodeling.
Figure 4: Fibroblast activation in cardiac repair.
During the proliferative phase of infarct healing, pericytes, macrophages and lymphocytes secrete a broad range of growth factors (such as Platelet-Derived Growth Factors (PDGF), Fibroblast Growth Factors (FGF) and Transforming Growth Factor (TGF)-β) and fibrogenic cytokines (such as Interleukin (IL)-4, IL-6, IL-10, IL-13) that stimulate fibroblasts promoting myofibroblast conversion and activation of a matrix-synthetic, matrix-preserving phenotype. Neurohumoral mediators (such as angiotensin II) and matricellular proteins that enrich the extracellular matrix (ECM) network also contribute to fibroblast activation. Scar maturation is associated with loss of myofibroblast features and acquisition of a matrifibrocyte phenotype by infarct fibroblasts. Fibroblast apoptosis may also contribute to de-activation of the fibrogenic response.
4.2.7. Growth factors: the role of TGF-βs, Fibroblast Growth Factors (FGF)s and PDGFs.
Several fibrogenic growth factors have been implicated in activation of fibroblasts in the infarcted heart. TGF-β is upregulated and activated in healing infarcts and plays a central role in activation of reparative fibroblasts400. Matricellular proteins401, proteases402 and αv integrin-mediated actions403,404 generate bioactive TGF–β in pericellular areas, thus contributing to a spatially localized fibrogenic response. Active TGF-β transduces signaling through members of the Smad family of transcription factors, or via non-Smad cascades405. In infarct fibroblasts, the Smad3 cascade plays a central role in activation of a reparative fibroblast phenotype and in formation of well-aligned arrays of activated myofibroblasts that protect the infarcted heart from rupture and from adverse remodeling406,407. Smad3-dependent fibroblast activation triggers an integrin-dependent oxidative response that plays a central role in formation of a well-organized matrix network that protects the infarcted heart407. In contrast to the central involvement of fibroblast-specific Smad3 signaling in cardiac repair after infarction, the Smad2 pathway does not play an important role in reparative fibrosis of the infarcted heart408.
Several additional growth factors are critical regulators of fibroblast phenotype in vitro; however, their in vivo role remains poorly defined. Expansion of the mast cell population during the proliferative phase of infarct healing409 increases local expression of fibrogenic tryptase410,264 and of chymase, a protease involved in local generation of angiotensin II411. Members of the fibroblast growth factor (FGF) family (such as FGF2) potently activate fibroblast proliferation and migration412,413; however studies investigating the role of their effects on infarct fibroblasts in vivo are lacking. Moreover, FGF signaling also exert angiogenic effects414 and protective actions on cardiomyocytes415,416 that may outweigh its fibrogenic actions. The Platelet-derived growth factor (PDGF) family has also been implicated in cardiac fibroblast activation417, promoting their survival and proliferation418,419,420. It has been suggested that PDGF signaling may stimulate acquisition of a myofibroblast phenotype in mesenchymal stem cells (MSCs), thus contributing to the expansion of fibroblasts after infarction421. In vivo experiments using antibody neutralization strategies suggested that PDGFRα signaling may promote fibroblast activation, whereas PDGFRβ may be involved in infarct angiogenesis417. The isoform-specific effects of PDGFs on the infarcted heart may have important therapeutic implications. Administration of PDGF-AB in a porcine model of MI was found to exert protective actions, improving the mechanical properties of the scar, and promoting angiogenesis422.
Fibroblast activation during the proliferative phase of infarct healing involves not only the effects of secreted growth factors and neurohumoral mediators, but also stimulation of extracellular matrix-mediated pathways. Macrophages and interstitial cells produce large amounts of matricellular proteins, specialized matrix proteins that do not play a major structural role, but transform the matrix into a signaling hub, interacting with cytokines, growth factors and proteases and modulating signaling through a broad range of integrin and non-integrin cell surface receptors423. Tenascin-C, the thrombospondins, osteopontin, osteoglycin, periostin, SPARC (secreted protein acidic and rich in cysteine), cartilage intermediate layer protein 1 (CILP)1, and ED-A fibronectin enrich the healing infarct and mediate fibroblast activation and myofibroblast conversion424,425,63,426,427,428,429,430,387. The matricellular proteins do not act exclusively on fibroblasts, but also modulate macrophage and vascular cell phenotype and function.
4.2.8. Fibroblasts and arrhythmogenesis after infarction
In addition to their reparative functions and their role in maladaptive fibrosis after MI, infarct fibroblasts may also be implicated in arrhythmia generation431. Several studies have suggested the presence of heterocellular electrical coupling between fibroblasts and cardiomyocytes in the infarct border zone that involves formation of gap junctions432,433,434,435. A recent study used a model of exogenous optically-stimulated depolarization to demonstrate that fibroblast-cardiomyocyte coupling can generate arrhythmias after MI431. Heterocellular coupling of infarct fibroblasts and border zone cardiomyocytes was suggested to result from synergistic actions between gap junctional proteins and a poorly understood non-gap junctional mechanism (termed ephaptic), involving generation of electrical fields in the interstitium between the coupled cells431. Given the close spatial association between fibroblasts and cardiomyocytes in the infarct border zone, this is a plausible mechanism; however, whether it plays a significant role in the arrhythmic events that occur in infarcted and remodeling hearts is not known.
5. The maturation phase.
5.1. Endogenous inhibitory pathways suppress fibroblast activation
Although fibroblast-mediated deposition of structural matrix proteins plays a central role in repair of the infarcted heart, excessive, prolonged or expansive fibroblast activation can have catastrophic consequences, resulting in adverse fibrotic remodeling and causing dysfunction and heart failure progression. Thus, the inhibitory signals leading to formation of a mature scar are of central significance in protection from heart failure progression. During the infarct maturation phase, myofibroblasts reduce their matrix-synthetic activity and ultimately disassemble the α-SMA+ stress fibers382. Although a fraction of infarct myofibroblasts may undergo apoptosis436, the majority of the activated infarct fibroblasts convert to matrifibrocytes, cells with low levels of a-SMA and high expression of cartilage and tendon ECM proteins437. The signals responsible for de-activation of infarct myofibroblasts remain enigmatic. However, several negative regulators of fibroblast activation have been identified and found to play an important role in protecting the heart from adverse remodeling.
TGF-β-mediated fibroblast activation stimulates expression of the inhibitory Smads, Smad6 and Smad7, important negative regulators of TGF-β superfamily signaling438. Smad7 is markedly upregulated in infarct myofibroblasts and protects from heart failure, attenuating adverse remodeling and fibrosis, by restraining collagen accumulation and myofibroblast conversion. The anti-fibrotic effects of Smad7 are only partially mediated through inhibition of TGF-β -induced Smad-dependent and non-Smad-dependent cascades, but also involve direct inhibitory actions of Smad7 on the receptor tyrosine kinase Erbb2439. Associative evidence links several other anti-fibrotic signals with suppression of fibroblast activity in the infarcted heart. Induction of the nuclear TGF-β repressor c-Ski during the maturation phase of infarct healing may be involved in fibroblast de-activation440,441. Activation of the intracellular kinase AMP-activated protein kinase (AMPK)α1 may restrain fibroblast activation and proliferation following MI through effects that were attributed to connexin 43 upregulation442. Increased HIF-1a expression in infarct fibroblasts was found to restrain their activation and proliferation, thus preventing excessive fibrotic remodeling after MI443. Moreover, changes in the composition of the extracellular matrix may also transduce de-activating signals. Collagen V was found to restrain integrin expression in infarct fibroblasts, thus attenuating their activity and reducing scar expansion444. It is plausible to hypothesize that suppression of fibroblast activity may involve the co-operation of several different pathways that may induce fibroblast quiescence, while promoting a matrix-preserving fibroblast phenotype involve in scar maintenance.
5.2. Coating of infarct neovessels with mural cells mediates vascular maturation.
Scar maturation is accompanied by dynamic changes in the infarct neovasculature (Figure 3). The robust VEGF-driven angiogenic response that characterizes the proliferative phase417, 445, is followed by suppression of angiogenesis and recruitment of mural cells (pericytes and vascular smooth muscle cells) that coat and stabilize the infarct neovessels. Investment of the neovessels with a mural coat involves activation of PDGF-PDGF receptor-β (PDGFR-β)417 and TGF-β279 signaling pathways Defective mural cell recruitment results in persistence of pro-inflammatory neovessels in the infarct, accentuating post-infarction inflammation and promoting dysfunction.
6. Targeting the inflammatory and reparative response in MI.
Considering their diverse roles in injury, repair and fibrosis, inflammatory and fibrogenic mediators are challenging therapeutic targets in the context of MI. Early attempts to inhibit post-MI inflammation for therapeutic gain were based on the concept of cytotoxic inflammatory injury. A large body of evidence using pharmacologic interventions suggested that blockade of mediators involved in post-MI inflammation (such as the leukocyte integrins) markedly reduced the size of the infarct after infarction311. However, 3 small clinical trials using anti-integrin approaches failed to reduce the size of the infarct in MI patients446,447,448. Administration of pexelizumab, an antibody targeting the complement system, an upstream activator of the innate immune response, produced equally disappointing results in a large clinical trial in STEMI patients449. Moreover, treatment of acute coronary syndrome (ACS) patients with the P-selectin inhibitor inclacumab reduced the release of enzymes associated with cardiac injury, but was associated with trends towards worse clinical outcome450,451. Despite these early disappointments, interest in myocardial inflammation continued to increase, fueled by exciting developments in the field of immunology and new insights into the molecular signals underlying the inflammatory response. The recent experience with anti-cytokine approaches offered a glimmer of hope regarding the future of anti-inflammatory therapies in MI patients.
In the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) trial, treatment of patients with prior MI and elevated hsCRP (suggestive of active inflammation) with the anti-IL1β monoclonal antibody canakinumab reduced the risk of the composite endpoint (death, non-fatal MI, or non-fatal stroke) by 15% in comparison to standard treatment452. Although the study was designed to test the hypothesis that IL-1β-mediated inflammation may be involved in atherothrombotic complications, a pre-specified exploratory analysis showed that IL-1β inhibition may reduce heart failure hospitalizations or heart failure-related death453. The CANTOS data along with early phase trials suggesting protection of STEMI patients from adverse remodeling upon IL-1 blockade with anakinra454,455,456 support the role of IL-1 as a therapeutic target in MI. Other pro-inflammatory cytokines, such as IL-6, may also be involved in adverse remodeling after infarction and may be promising therapeutic targets. In the ASSAIL-MI trial, IL-6 receptor inhibition with tocilizumab increased myocardial salvage in STEMI patients457. In addition to approaches selectively targeting specific cytokines, repurposing anti-inflammatory agents with broader mechanisms of action has also been attempted. In the COLCOT trial, low-dose colchicine administration within 30 days after MI reduced major adverse cardiovascular events by 22%458. Thus, colchicine has become the first guideline-recommended anti-inflammatory medication in cardiovascular secondary prevention. Whether the beneficial effects of colchicine involve improved repair and attenuated remodeling, in addition to a reduction in atherothrombotic events is not known.
Strategies targeting specific cell types may also hold promise for patients with MI. Building on promising results of B-cell inhibition in experimental MI models309, safety and feasibility of B cell depletion using the monoclonal anti-CD20 antibody rituximab has been demonstrated459, paving the way for outcome trials. Chimeric antigen receptor (CAR)+ T cells engineered to target activated fibroblasts have been suggested to improve function in models of interstitial fibrosis induced through neurohumoral activation460,461, and may also hold promise in MI patients with overactive fibrogenic responses. Moreover, amplification of Tregs using a superagonistic anti-CD28 antibody may protect the infarcted heart by suppressing excessive inflammation462,463.
In addition to approaches inhibiting pro-inflammatory mediators and cells to reduce their deleterious effects on the remodeling heart, interventions aimed at enhancing the reparative actions of growth factors have also been suggested. For example, PDGF-AB administration has been suggested to improve repair in both rodent and large animal models of infarction. Although the risks of growth factor supplementation approaches may include accentuation of fibrosis, cautious and selective administration in patients with reparative perturbations may hold promise. CXCL12 treatment may also promote recruitment of reparative cells, increasing infarct angiogenesis, and improving left ventricular mechanics216, 219, 220, 228.
Although several different strategies targeting the inflammatory and reparative responses after MI hold promise, therapeutic translation remains challenging. The duality of the effects of inflammatory mediators after infarction and their involvement in both injurious and reparative responses complicate therapeutic implementation. Rational design of therapeutic interventions targeting inflammatory pathways after MI should take into account important temporal and spatial considerations. Our understanding of the temporal sequence of events during the three phases of cardiac repair suggests that there is a narrow window of therapeutic opportunity for safe and effective targeting of specific inflammatory signals.
The remarkable pathophysiologic heterogeneity of MI patients poses another major challenge for therapeutic translation464,311. Heart failure progression and adverse remodeling after MI are only in part dependent on the extent of initial injury and the size of the infarct. The inflammatory, reparative and fibrogenic responses after MI are greatly affected by many other factors, including age, sex, genetic substrate, the presence or absence of concomitant conditions (such as diabetes, obesity, hypertension etc), the pattern of atherosclerotic disease, and the use of other medications that may interfere with inflammatory or fibrogenic signaling. Some patient subpopulations may have defects in negative regulation and resolution of inflammation and perturbations in fibroblast activation, thus exhibiting prolonged or expanded inflammatory responses and perturbed scar formation. Others may have overactive fibrotic reactions, resulting in increased stiffness and diastolic dysfunction. This pathophysiologic heterogeneity cannot be recapitulated by our animal models, which are better suited for dissection of underlying cellular mechanisms and not as predictors of therapeutic efficacy464. What is needed is a pathophysiologic classification of patients on the basis of clinical features, biochemical profile, and/or functional responses to identify individuals with accentuated post-infarction inflammatory responses that may benefit from targeted anti-inflammatory strategies, subjects with overactive fibrogenic pathways who may require TGF–β inhibition and patients with impaired reparative fibrosis who may be treated with a fibrogenic growth factor, such as PDGF-AB. For example, patients with diabetes have a high incidence of diastolic dysfunction following MI, despite a smaller infarct size and comparable systolic dysfunction465. On the other hand, aging is associated with a prolonged inflammatory phase and defective reparative growth factor signaling, resulting in formation of scars with low levels of collagenous matrix466. In addition to clinical criteria, biomarkers reflecting inflammatory and fibrogenic activation467,468 and imaging approaches informing on the extent of fibrotic remodeling469,470,471 may be used to design personalized therapeutic approaches.
7. Conclusions:
Over the last 30 years, both experimental and clinical studies contributed to our understanding of the role of inflammatory and reparative pathways in infarct healing and post-infarction remodeling. The field survived early skepticism and was strengthened by the failures of initial approaches to target inflammation in MI patients. As several promising candidates for intervention have emerged, there is enough evidence to support cautious optimism on the future of therapeutic translation,
SOURCES OF FUNDING:
Dr. Hilgendorf’s laboratory is supported by German Research Foundation grants HI 1573/10-1, HI 1573/9-1, and SFB1425 grant 422691945. Dr. Frantz is supported by the Collaborative Research Center (CRC) 1525 “Cardio-immune Interfaces” and received funding from the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) — project number: 453989101. Dr Frangogiannis’ laboratory is supported by NIH grants R01 HL76246, R01 HL85440, and R01 HL149407, and by U.S. Department of Defense grant PR211352.
Footnotes
DISCLOSURES: None.
REFERENCES:
- 1.Laforgia PL, Auguadro C, Bronzato S, Durante A. The Reduction of Mortality in Acute Myocardial Infarction: From Bed Rest to Future Directions. Int J Prev Med. 2022; 13:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Rosamond WD, Chambless LE, Heiss G, Mosley TH, Coresh J, Whitsel E, Wagenknecht L, Ni H, Folsom AR. Twenty-two-year trends in incidence of myocardial infarction, coronary heart disease mortality, and case fatality in 4 US communities, 1987-2008. Circulation. 2012; 125:1848–1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.McManus DD, Gore J, Yarzebski J, Spencer F, Lessard D, Goldberg RJ. Recent trends in the incidence, treatment, and outcomes of patients with STEMI and NSTEMI. Am J Med. 2011; 124:40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Thrane PG, Olesen KKW, Thim T, Gyldenkerne C, Mortensen MB, Kristensen SD, Maeng M. Mortality Trends After Primary Percutaneous Coronary Intervention for ST-Segment Elevation Myocardial Infarction. J Am Coll Cardiol. 2023; 82:999–1010. [DOI] [PubMed] [Google Scholar]
- 5.Frangogiannis NG. Regulation of the inflammatory response in cardiac repair. Circ Res. 2012; 110:159–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jennings RB, Murry CE, Steenbergen C Jr., Reimer KA. Development of cell injury in sustained acute ischemia. Circulation. 1990; 82:II2–12. [PubMed] [Google Scholar]
- 7.Reimer KA, Lowe JE, Rasmussen MM, Jennings RB. The wavefront phenomenon of ischemic cell death. 1. Myocardial infarct size vs duration of coronary occlusion in dogs. Circulation. 1977; 56:786–794. [DOI] [PubMed] [Google Scholar]
- 8.Venugopal H, Hanna A, Humeres C, Frangogiannis NG. Properties and Functions of Fibroblasts and Myofibroblasts in Myocardial Infarction. Cells. 2022; 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Courties G, Moskowitz MA, Nahrendorf M. The innate immune system after ischemic injury: lessons to be learned from the heart and brain. JAMA Neurol. 2014; 71:233–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo JL, Kohler RH, Chudnovskiy A, Waterman P, Aikawa E, Mempel TR, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009; 325:612–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science. 2013; 339:161–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sager HB, Heidt T, Hulsmans M, Dutta P, Courties G, Sebas M, Wojtkiewicz GR, Tricot B, Iwamoto Y, Sun Y, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Targeting Interleukin-1beta Reduces Leukocyte Production After Acute Myocardial Infarction. Circulation. 2015; 132:1880–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, Scadden DT, Ma'ayan A, Enikolopov GN, Frenette PS. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010; 466:829–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Courties G, Herisson F, Sager HB, Heidt T, Ye Y, Wei Y, Sun Y, Severe N, Dutta P, Scharff J, Scadden DT, Weissleder R, et al. Ischemic stroke activates hematopoietic bone marrow stem cells. Circ Res. 2015; 116:407–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Silvis MJM, Kaffka Genaamd Dengler SE, Odille CA, Mishra M, van der Kaaij NP, Doevendans PA, Sluijter JPG, de Kleijn DPV, de Jager SCA, Bosch L, van Hout GPJ. Damage-Associated Molecular Patterns in Myocardial Infarction and Heart Transplantation: The Road to Translational Success. Front Immunol. 2020; 11:599511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Shah M, Yellon DM, Davidson SM. The Role of Extracellular DNA and Histones in Ischaemia-Reperfusion Injury of the Myocardium. Cardiovasc Drugs Ther. 2020; 34:123–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Arslan F, de Kleijn DP, Pasterkamp G. Innate immune signaling in cardiac ischemia. Nat Rev Cardiol. 2011; 8:292–300. [DOI] [PubMed] [Google Scholar]
- 18.de Haan JJ, Smeets MB, Pasterkamp G, Arslan F. Danger signals in the initiation of the inflammatory response after myocardial infarction. Mediators Inflamm. 2013; 2013:206039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Eltzschig HK, Eckle T. Ischemia and reperfusion--from mechanism to translation. Nat Med. 2011; 17:1391–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ghigo A, Franco I, Morello F, Hirsch E. Myocyte signalling in leucocyte recruitment to the heart. Cardiovascular research. 2014; 102:270–280. [DOI] [PubMed] [Google Scholar]
- 21.Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harb Perspect Biol. 2012; 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Timmers L, Pasterkamp G, de Hoog VC, Arslan F, Appelman Y, de Kleijn DP. The innate immune response in reperfused myocardium. Cardiovasc Res. 2012; 94:276–283. [DOI] [PubMed] [Google Scholar]
- 23.Mann DL. The emerging role of innate immunity in the heart and vascular system: for whom the cell tolls. Circulation research. 2011; 108:1133–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140:821–832. [DOI] [PubMed] [Google Scholar]
- 25.Xie J, Mendez JD, Mendez-Valenzuela V, Aguilar-Hernandez MM. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cellular signalling. 2013; 25:2185–2197. [DOI] [PubMed] [Google Scholar]
- 26.Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002; 418:191–195. [DOI] [PubMed] [Google Scholar]
- 27.Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Yang H, Ivanova S, Borovikova L, Manogue KR, Faist E, et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science. 1999; 285:248–251. [DOI] [PubMed] [Google Scholar]
- 28.Loukili N, Rosenblatt-Velin N, Li J, Clerc S, Pacher P, Feihl F, Waeber B, Liaudet L. Peroxynitrite induces HMGB1 release by cardiac cells in vitro and HMGB1 upregulation in the infarcted myocardium in vivo. Cardiovasc Res. 2011; 89:586–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Andrassy M, Volz HC, Igwe JC, Funke B, Eichberger SN, Kaya Z, Buss S, Autschbach F, Pleger ST, Lukic IK, Bea F, Hardt SE, et al. High-mobility group box-1 in ischemia-reperfusion injury of the heart. Circulation. 2008; 117:3216–3226. [DOI] [PubMed] [Google Scholar]
- 30.Xu H, Yao Y, Su Z, Yang Y, Kao R, Martin CM, Rui T. Endogenous HMGB1 contributes to ischemia-reperfusion-induced myocardial apoptosis by potentiating the effect of TNF-α/JNK. American journal of physiology Heart and circulatory physiology. 2011; 300:H913–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kohno T, Anzai T, Naito K, Miyasho T, Okamoto M, Yokota H, Yamada S, Maekawa Y, Takahashi T, Yoshikawa T, Ishizaka A, Ogawa S. Role of high-mobility group box 1 protein in post-infarction healing process and left ventricular remodelling. Cardiovasc Res. 2009; 81:565–573. [DOI] [PubMed] [Google Scholar]
- 32.Cirillo P, Giallauria F, Pacileo M, Petrillo G, D'Agostino M, Vigorito C, Chiariello M. Increased high mobility group box-1 protein levels are associated with impaired cardiopulmonary and echocardiographic findings after acute myocardial infarction. Journal of cardiac failure. 2009; 15:362–367. [DOI] [PubMed] [Google Scholar]
- 33.Sorensen MV, Pedersen S, Mogelvang R, Skov-Jensen J, Flyvbjerg A. Plasma high-mobility group box 1 levels predict mortality after ST-segment elevation myocardial infarction. JACC Cardiovascular interventions. 2011; 4:281–286. [DOI] [PubMed] [Google Scholar]
- 34.Park JS, Svetkauskaite D, He Q, Kim JY, Strassheim D, Ishizaka A, Abraham E. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem. 2004; 279:7370–7377. [DOI] [PubMed] [Google Scholar]
- 35.Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, Parroche P, Drabic S, Golenbock D, Sirois C, Hua J, An LL, et al. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nature immunology. 2007; 8:487–496. [DOI] [PubMed] [Google Scholar]
- 36.Liu FY, Fan D, Yang Z, Tang N, Guo Z, Ma SQ, Ma ZG, Wu HM, Deng W, Tang QZ. TLR9 is essential for HMGB1-mediated post-myocardial infarction tissue repair through affecting apoptosis, cardiac healing, and angiogenesis. Cell Death Dis. 2019; 10:480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Schiraldi M, Raucci A, Munoz LM, Livoti E, Celona B, Venereau E, Apuzzo T, De Marchis F, Pedotti M, Bachi A, Thelen M, Varani L, et al. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4. The Journal of experimental medicine. 2012; 209:551–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kitahara T, Takeishi Y, Harada M, Niizeki T, Suzuki S, Sasaki T, Ishino M, Bilim O, Nakajima O, Kubota I. High-mobility group box 1 restores cardiac function after myocardial infarction in transgenic mice. Cardiovasc Res. 2008; 80:40–46. [DOI] [PubMed] [Google Scholar]
- 39.Nakamura Y, Suzuki S, Shimizu T, Miyata M, Shishido T, Ikeda K, Saitoh S, Kubota I, Takeishi Y. High Mobility Group Box 1 Promotes Angiogenesis from Bone Marrow-derived Endothelial Progenitor Cells after Myocardial Infarction. J Atheroscler Thromb. 2015; 22:570–581. [DOI] [PubMed] [Google Scholar]
- 40.Limana F, Germani A, Zacheo A, Kajstura J, Di Carlo A, Borsellino G, Leoni O, Palumbo R, Battistini L, Rastaldo R, Muller S, Pompilio G, et al. Exogenous high-mobility group box 1 protein induces myocardial regeneration after infarction via enhanced cardiac C-kit+ cell proliferation and differentiation. Circ Res. 2005; 97:e73–83. [DOI] [PubMed] [Google Scholar]
- 41.Takahashi K, Fukushima S, Yamahara K, Yashiro K, Shintani Y, Coppen SR, Salem HK, Brouilette SW, Yacoub MH, Suzuki K. Modulated inflammation by injection of high-mobility group box 1 recovers post-infarction chronically failing heart. Circulation. 2008; 118:S106–114. [DOI] [PubMed] [Google Scholar]
- 42.Limana F, Esposito G, D'Arcangelo D, Di Carlo A, Romani S, Melillo G, Mangoni A, Bertolami C, Pompilio G, Germani A, Capogrossi MC. HMGB1 attenuates cardiac remodelling in the failing heart via enhanced cardiac regeneration and miR-206-mediated inhibition of TIMP-3. PloS one. 2011; 6:e19845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Xia C, Braunstein Z, Toomey AC, Zhong J, Rao X. S100 Proteins As an Important Regulator of Macrophage Inflammation. Front Immunol. 2017; 8:1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA, Nacken W, Foell D, van der Poll T, Sorg C, Roth J. Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med. 2007; 13:1042–1049. [DOI] [PubMed] [Google Scholar]
- 45.Hofmann MA, Drury S, Fu C, Qu W, Taguchi A, Lu Y, Avila C, Kambham N, Bierhaus A, Nawroth P, Neurath MF, Slattery T, et al. RAGE mediates a novel proinflammatory axis: a central cell surface receptor for S100/calgranulin polypeptides. Cell. 1999; 97:889–901. [DOI] [PubMed] [Google Scholar]
- 46.Katashima T, Naruko T, Terasaki F, Fujita M, Otsuka K, Murakami S, Sato A, Hiroe M, Ikura Y, Ueda M, Ikemoto M, Kitaura Y. Enhanced expression of the S100A8/A9 complex in acute myocardial infarction patients. Circ J. 2010; 74:741–748. [DOI] [PubMed] [Google Scholar]
- 47.Morrow DA, Wang Y, Croce K, Sakuma M, Sabatine MS, Gao H, Pradhan AD, Healy AM, Buros J, McCabe CH, Libby P, Cannon CP, Braunwald E, Simon DI. Myeloid-related protein 8/14 and the risk of cardiovascular death or myocardial infarction after an acute coronary syndrome in the Pravastatin or Atorvastatin Evaluation and Infection Therapy: Thrombolysis in Myocardial Infarction (PROVE IT-TIMI 22) trial. American heart journal. 2008; 155:49–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Volz HC, Laohachewin D, Seidel C, Lasitschka F, Keilbach K, Wienbrandt AR, Andrassy J, Bierhaus A, Kaya Z, Katus HA, Andrassy M. S100A8/A9 aggravates post-ischemic heart failure through activation of RAGE-dependent NF-kappaB signaling. Basic Res Cardiol. 2012; 107:250. [DOI] [PubMed] [Google Scholar]
- 49.Sreejit G, Abdel-Latif A, Athmanathan B, Annabathula R, Dhyani A, Noothi SK, Quaife-Ryan GA, Al-Sharea A, Pernes G, Dragoljevic D, Lal H, Schroder K, et al. Neutrophil-Derived S100A8/A9 Amplify Granulopoiesis After Myocardial Infarction. Circulation. 2020; 141:1080–1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Marinkovic G, Grauen Larsen H, Yndigegn T, Szabo IA, Mares RG, de Camp L, Weiland M, Tomas L, Goncalves I, Nilsson J, Jovinge S, Schiopu A. Inhibition of pro-inflammatory myeloid cell responses by short-term S100A9 blockade improves cardiac function after myocardial infarction. Eur Heart J. 2019; 40:2713–2723. [DOI] [PubMed] [Google Scholar]
- 51.Marinkovic G, Koenis DS, de Camp L, Jablonowski R, Graber N, de Waard V, de Vries CJ, Goncalves I, Nilsson J, Jovinge S, Schiopu A. S100A9 Links Inflammation and Repair in Myocardial Infarction. Circ Res. 2020; 127:664–676. [DOI] [PubMed] [Google Scholar]
- 52.Zhang X, Cheng M, Gao N, Li Y, Yan C, Tian X, Liu D, Qiu M, Wang X, Luan B, Deng J, Wang S, et al. Utility of S100A12 as an Early Biomarker in Patients With ST-Segment Elevation Myocardial Infarction. Front Cardiovasc Med. 2021; 8:747511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rohde D, Schon C, Boerries M, Didrihsone I, Ritterhoff J, Kubatzky KF, Volkers M, Herzog N, Mahler M, Tsoporis JN, Parker TG, Linke B, et al. S100A1 is released from ischemic cardiomyocytes and signals myocardial damage via Toll-like receptor 4. EMBO Mol Med. 2014; 6:778–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Doroudgar S, Quijada P, Konstandin M, Ilves K, Broughton K, Khalafalla FG, Casillas A, Nguyen K, Gude N, Toko H, Ornelas L, Thuerauf DJ, et al. S100A4 protects the myocardium against ischemic stress. J Mol Cell Cardiol. 2016; 100:54–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cohen I, Rider P, Carmi Y, Braiman A, Dotan S, White MR, Voronov E, Martin MU, Dinarello CA, Apte RN. Differential release of chromatin-bound IL-1alpha discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation. Proc Natl Acad Sci U S A. 2010; 107:2574–2579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Di Paolo NC, Shayakhmetov DM. Interleukin 1alpha and the inflammatory process. Nat Immunol. 2016; 17:906–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lugrin J, Parapanov R, Rosenblatt-Velin N, Rignault-Clerc S, Feihl F, Waeber B, Muller O, Vergely C, Zeller M, Tardivel A, Schneider P, Pacher P, Liaudet L. Cutting Edge: IL-1alpha Is a Crucial Danger Signal Triggering Acute Myocardial Inflammation during Myocardial Infarction. J Immunol. 2015; 194:499–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lugrin J, Parapanov R, Milano G, Cavin S, Debonneville A, Krueger T, Liaudet L. The systemic deletion of interleukin-1alpha reduces myocardial inflammation and attenuates ventricular remodeling in murine myocardial infarction. Sci Rep. 2023; 13:4006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schunk SJ, Triem S, Schmit D, Zewinger S, Sarakpi T, Becker E, Hutter G, Wrublewsky S, Kuting F, Hohl M, Alansary D, Prates Roma L, et al. Interleukin-1alpha Is a Central Regulator of Leukocyte-Endothelial Adhesion in Myocardial Infarction and in Chronic Kidney Disease. Circulation. 2021; 144:893–908. [DOI] [PubMed] [Google Scholar]
- 60.Frevert CW, Felgenhauer J, Wygrecka M, Nastase MV, Schaefer L. Danger-Associated Molecular Patterns Derived From the Extracellular Matrix Provide Temporal Control of Innate Immunity. J Histochem Cytochem. 2018; 66:213–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gondokaryono SP, Ushio H, Niyonsaba F, Hara M, Takenaka H, Jayawardana ST, Ikeda S, Okumura K, Ogawa H. The extra domain A of fibronectin stimulates murine mast cells via toll-like receptor 4. Journal of leukocyte biology. 2007; 82:657–665. [DOI] [PubMed] [Google Scholar]
- 62.Schoneveld AH, Hoefer I, Sluijter JP, Laman JD, de Kleijn DP, Pasterkamp G. Atherosclerotic lesion development and Toll like receptor 2 and 4 responsiveness. Atherosclerosis. 2008; 197:95–104. [DOI] [PubMed] [Google Scholar]
- 63.Arslan F, Smeets MB, Riem Vis PW, Karper JC, Quax PH, Bongartz LG, Peters JH, Hoefer IE, Doevendans PA, Pasterkamp G, de Kleijn DP. Lack of fibronectin-EDA promotes survival and prevents adverse remodeling and heart function deterioration after myocardial infarction. Circ Res. 2011; 108:582–592. [DOI] [PubMed] [Google Scholar]
- 64.Serini G, Bochaton-Piallat ML, Ropraz P, Geinoz A, Borsi L, Zardi L, Gabbiani G. The fibronectin domain ED-A is crucial for myofibroblastic phenotype induction by transforming growth factor-beta1. J Cell Biol. 1998; 142:873–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Klingberg F, Chau G, Walraven M, Boo S, Koehler A, Chow ML, Olsen AL, Im M, Lodyga M, Wells RG, White ES, 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]
- 66.Amarante-Mendes GP, Adjemian S, Branco LM, Zanetti LC, Weinlich R, Bortoluci KR. Pattern Recognition Receptors and the Host Cell Death Molecular Machinery. Front Immunol. 2018; 9:2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bezhaeva T, Karper J, Quax PHA, de Vries MR. The Intriguing Role of TLR Accessory Molecules in Cardiovascular Health and Disease. Front Cardiovasc Med. 2022; 9:820962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kumar V. A STING to inflammation and autoimmunity. J Leukoc Biol. 2019; 106:171–185. [DOI] [PubMed] [Google Scholar]
- 69.Li Y, Slavik KM, Toyoda HC, Morehouse BR, de Oliveira Mann CC, Elek A, Levy S, Wang Z, Mears KS, Liu J, Kashin D, Guo X, et al. cGLRs are a diverse family of pattern recognition receptors in innate immunity. Cell. 2023; 186:3261–3276 e3220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Timmers L, Sluijter JP, van Keulen JK, Hoefer IE, Nederhoff MG, Goumans MJ, Doevendans PA, van Echteld CJ, Joles JA, Quax PH, Piek JJ, Pasterkamp G, de Kleijn DP. Toll-like receptor 4 mediates maladaptive left ventricular remodeling and impairs cardiac function after myocardial infarction. Circ Res. 2008; 102:257–264. [DOI] [PubMed] [Google Scholar]
- 71.Frantz S, Kobzik L, Kim YD, Fukazawa R, Medzhitov R, Lee RT, Kelly RA. Toll4 (TLR4) expression in cardiac myocytes in normal and failing myocardium. J Clin Invest. 1999; 104:271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Liu L, Wang Y, Cao ZY, Wang MM, Liu XM, Gao T, Hu QK, Yuan WJ, Lin L. Up-regulated TLR4 in cardiomyocytes exacerbates heart failure after long-term myocardial infarction. J Cell Mol Med. 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Methe H, Kim JO, Kofler S, Weis M, Nabauer M, Koglin J. Expansion of circulating Toll-like receptor 4-positive monocytes in patients with acute coronary syndrome. Circulation. 2005; 111:2654–2661. [DOI] [PubMed] [Google Scholar]
- 74.Satoh M, Shimoda Y, Maesawa C, Akatsu T, Ishikawa Y, Minami Y, Hiramori K, Nakamura M. Activated toll-like receptor 4 in monocytes is associated with heart failure after acute myocardial infarction. Int J Cardiol. 2006; 109:226–234. [DOI] [PubMed] [Google Scholar]
- 75.van der Pouw Kraan TC, Bernink FJ, Yildirim C, Koolwijk P, Baggen JM, Timmers L, Beek AM, Diamant M, Chen WJ, van Rossum AC, van Royen N, Horrevoets AJ, Appelman YE. Systemic toll-like receptor and interleukin-18 pathway activation in patients with acute ST elevation myocardial infarction. Journal of molecular and cellular cardiology. 2014; 67:94–102. [DOI] [PubMed] [Google Scholar]
- 76.Oyama J, Blais C Jr., Liu X, Pu M, Kobzik L, Kelly RA, Bourcier T. Reduced myocardial ischemia-reperfusion injury in toll-like receptor 4-deficient mice. Circulation. 2004; 109:784–789. [DOI] [PubMed] [Google Scholar]
- 77.Kim SC, Ghanem A, Stapel H, Tiemann K, Knuefermann P, Hoeft A, Meyer R, Grohe C, Knowlton AA, Baumgarten G. Toll-like receptor 4 deficiency: smaller infarcts, but no gain in function. BMC physiology. 2007; 7:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Shimamoto A, Chong AJ, Yada M, Shomura S, Takayama H, Fleisig AJ, Agnew ML, Hampton CR, Rothnie CL, Spring DJ, Pohlman TH, Shimpo H, Verrier ED. Inhibition of Toll-like receptor 4 with eritoran attenuates myocardial ischemia-reperfusion injury. Circulation. 2006; 114:I270–274. [DOI] [PubMed] [Google Scholar]
- 79.Selejan S, Poss J, Walter F, Hohl M, Kaiser R, Kazakov A, Bohm M, Link A. Ischaemia-induced up-regulation of Toll-like receptor 2 in circulating monocytes in cardiogenic shock. European heart journal. 2012; 33:1085–1094. [DOI] [PubMed] [Google Scholar]
- 80.Arslan F, Smeets MB, O'Neill LA, Keogh B, McGuirk P, Timmers L, Tersteeg C, Hoefer IE, Doevendans PA, Pasterkamp G, de Kleijn DP. Myocardial ischemia/reperfusion injury is mediated by leukocytic toll-like receptor-2 and reduced by systemic administration of a novel anti-toll-like receptor-2 antibody. Circulation. 2010; 121:80–90. [DOI] [PubMed] [Google Scholar]
- 81.Favre J, Musette P, Douin-Echinard V, Laude K, Henry JP, Arnal JF, Thuillez C, Richard V. Toll-like receptors 2-deficient mice are protected against postischemic coronary endothelial dysfunction. Arterioscler Thromb Vasc Biol. 2007; 27:1064–1071. [DOI] [PubMed] [Google Scholar]
- 82.Arslan F, Houtgraaf JH, Keogh B, Kazemi K, de Jong R, McCormack WJ, O'Neill LA, McGuirk P, Timmers L, Smeets MB, Akeroyd L, Reilly M, Pasterkamp G, de Kleijn DP. Treatment with OPN-305, a humanized anti-Toll-Like receptor-2 antibody, reduces myocardial ischemia/reperfusion injury in pigs. Circ Cardiovasc Interv. 2012; 5:279–287. [DOI] [PubMed] [Google Scholar]
- 83.Riad A, Jager S, Sobirey M, Escher F, Yaulema-Riss A, Westermann D, Karatas A, Heimesaat MM, Bereswill S, Dragun D, Pauschinger M, Schultheiss HP, Tschope C. Toll-like receptor-4 modulates survival by induction of left ventricular remodeling after myocardial infarction in mice. J Immunol. 2008; 180:6954–6961. [DOI] [PubMed] [Google Scholar]
- 84.Shishido T, Nozaki N, Yamaguchi S, Shibata Y, Nitobe J, Miyamoto T, Takahashi H, Arimoto T, Maeda K, Yamakawa M, Takeuchi O, Akira S, Takeishi Y, Kubota I. Toll-like receptor-2 modulates ventricular remodeling after myocardial infarction. Circulation. 2003; 108:2905–2910. [DOI] [PubMed] [Google Scholar]
- 85.Lu C, Ren D, Wang X, Ha T, Liu L, Lee EJ, Hu J, Kalbfleisch J, Gao X, Kao R, Williams D, Li C. Toll-like receptor 3 plays a role in myocardial infarction and ischemia/reperfusion injury. Biochim Biophys Acta. 2014; 1842:22–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chen C, Feng Y, Zou L, Wang L, Chen HH, Cai JY, Xu JM, Sosnovik DE, Chao W. Role of extracellular RNA and TLR3-Trif signaling in myocardial ischemia-reperfusion injury. J Am Heart Assoc. 2014; 3:e000683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.de Kleijn DPV, Chong SY, Wang X, Yatim S, Fairhurst AM, Vernooij F, Zharkova O, Chan MY, Foo RSY, Timmers L, Lam CSP, Wang JW. Toll-like receptor 7 deficiency promotes survival and reduces adverse left ventricular remodelling after myocardial infarction. Cardiovasc Res. 2019; 115:1791–1803. [DOI] [PubMed] [Google Scholar]
- 88.Ha T, Hu Y, Liu L, Lu C, McMullen JR, Kelley J, Kao RL, Williams DL, Gao X, Li C. TLR2 ligands induce cardioprotection against ischaemia/reperfusion injury through a PI3K/Akt-dependent mechanism. Cardiovascular research. 2010; 87:694–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Dong JW, Vallejo JG, Tzeng HP, Thomas JA, Mann DL. Innate immunity mediates myocardial preconditioning through Toll-like receptor 2 and TIRAP-dependent signaling pathways. American journal of physiology Heart and circulatory physiology. 2010; 298:H1079–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Chao W. Toll-like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart. American journal of physiology Heart and circulatory physiology. 2009; 296:H1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ha T, Hua F, Liu X, Ma J, McMullen JR, Shioi T, Izumo S, Kelley J, Gao X, Browder W, Williams DL, Kao RL, Li C. Lipopolysaccharide-induced myocardial protection against ischaemia/reperfusion injury is mediated through a PI3K/Akt-dependent mechanism. Cardiovascular research. 2008; 78:546–553. [DOI] [PubMed] [Google Scholar]
- 92.Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-kappaB in the heart: to be or not to NF-kappaB. Circ Res. 2011; 108:1122–1132. [DOI] [PubMed] [Google Scholar]
- 93.Davidson SM, Selvaraj P, He D, Boi-Doku C, Yellon RL, Vicencio JM, Yellon DM. Remote ischaemic preconditioning involves signalling through the SDF-1alpha/CXCR4 signalling axis. Basic research in cardiology. 2013; 108:377. [DOI] [PubMed] [Google Scholar]
- 94.Morimoto H, Hirose M, Takahashi M, Kawaguchi M, Ise H, Kolattukudy PE, Yamada M, Ikeda U. MCP-1 induces cardioprotection against ischaemia/reperfusion injury: role of reactive oxygen species. Cardiovasc Res. 2008; 78:554–562. [DOI] [PubMed] [Google Scholar]
- 95.Chou WC, Jha S, Linhoff MW, Ting JP. The NLR gene family: from discovery to present day. Nat Rev Immunol. 2023; 23:635–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, Shao F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015; 526:660–665. [DOI] [PubMed] [Google Scholar]
- 97.Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, Hongo M, Noda T, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011; 123:594–604. [DOI] [PubMed] [Google Scholar]
- 98.Mezzaroma E, Toldo S, Farkas D, Seropian IM, Van Tassell BW, Salloum FN, Kannan HR, Menna AC, Voelkel NF, Abbate A. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc Natl Acad Sci U S A. 2011; 108:19725–19730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sandanger O, Ranheim T, Vinge LE, Bliksoen M, Alfsnes K, Finsen AV, Dahl CP, Askevold ET, Florholmen G, Christensen G, Fitzgerald KA, Lien E, et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovascular research. 2013; 99:164–174. [DOI] [PubMed] [Google Scholar]
- 100.Olsen MB, Gregersen I, Sandanger O, Yang K, Sokolova M, Halvorsen BE, Gullestad L, Broch K, Aukrust P, Louwe MC. Targeting the Inflammasome in Cardiovascular Disease. JACC Basic Transl Sci. 2022; 7:84–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Zhang KZ, Shen XY, Wang M, Wang L, Sun HX, Li XZ, Huang JJ, Li XQ, Wu C, Zhao C, Liu JL, Lu X, Gao W. Retinol-Binding Protein 4 Promotes Cardiac Injury After Myocardial Infarction Via Inducing Cardiomyocyte Pyroptosis Through an Interaction With NLRP3. J Am Heart Assoc. 2021; 10:e022011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Merkle S, Frantz S, Schon MP, Bauersachs J, Buitrago M, Frost RJ, Schmitteckert EM, Lohse MJ, Engelhardt S. A role for caspase-1 in heart failure. Circ Res. 2007; 100:645–653. [DOI] [PubMed] [Google Scholar]
- 103.Frantz S, Ducharme A, Sawyer D, Rohde LE, Kobzik L, Fukazawa R, Tracey D, Allen H, Lee RT, Kelly RA. Targeted deletion of caspase-1 reduces early mortality and left ventricular dilatation following myocardial infarction. J Mol Cell Cardiol. 2003; 35:685–694. [DOI] [PubMed] [Google Scholar]
- 104.Louwe MC, Olsen MB, Kaasboll OJ, Yang K, Fosshaug LE, Alfsnes K, Ogaard JDS, Rashidi A, Skulberg VM, Yang M, de Miranda Fonseca D, Sharma A, et al. Absence of NLRP3 Inflammasome in Hematopoietic Cells Reduces Adverse Remodeling After Experimental Myocardial Infarction. JACC Basic Transl Sci. 2020; 5:1210–1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Sandanger O, Gao E, Ranheim T, Bliksoen M, Kaasboll OJ, Alfsnes K, Nymo SH, Rashidi A, Ohm IK, Attramadal H, Aukrust P, Vinge LE, Yndestad A. NLRP3 inflammasome activation during myocardial ischemia reperfusion is cardioprotective. Biochem Biophys Res Commun. 2016; 469:1012–1020. [DOI] [PubMed] [Google Scholar]
- 106.Durga Devi T, Babu M, Makinen P, Kaikkonen MU, Heinaniemi M, Laakso H, Yla-Herttuala E, Rieppo L, Liimatainen T, Naumenko N, Tavi P, Yla-Herttuala S. Aggravated Postinfarct Heart Failure in Type 2 Diabetes Is Associated with Impaired Mitophagy and Exaggerated Inflammasome Activation. Am J Pathol. 2017; 187:2659–2673. [DOI] [PubMed] [Google Scholar]
- 107.Onodi Z, Ruppert M, Kucsera D, Sayour AA, Toth VE, Koncsos G, Novak J, Brenner GB, Makkos A, Baranyai T, Giricz Z, Gorbe A, et al. AIM2-driven inflammasome activation in heart failure. Cardiovasc Res. 2021; 117:2639–2651. [DOI] [PubMed] [Google Scholar]
- 108.King KR, Aguirre AD, Ye YX, Sun Y, Roh JD, Ng RP Jr., Kohler RH, Arlauckas SP, Iwamoto Y, Savol A, Sadreyev RI, Kelly M, et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. 2017; 23:1481–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Cao DJ, Schiattarella GG, Villalobos E, Jiang N, May HI, Li T, Chen ZJ, Gillette TG, Hill JA. Cytosolic DNA Sensing Promotes Macrophage Transformation and Governs Myocardial Ischemic Injury. Circulation. 2018; 137:2613–2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Rech L, Abdellatif M, Pottler M, Stangl V, Mabotuwana N, Hardy S, Rainer PP. Small molecule STING inhibition improves myocardial infarction remodeling. Life Sci. 2022; 291:120263. [DOI] [PubMed] [Google Scholar]
- 111.Ziegler T, Horstkotte M, Lange P, Ng J, Bongiovanni D, Hinkel R, Laugwitz KL, Sperandio M, Horstkotte J, Kupatt C. Endothelial RAGE exacerbates acute postischaemic cardiac inflammation. Thromb Haemost. 2016; 116:300–308. [DOI] [PubMed] [Google Scholar]
- 112.Bayer AL, Alcaide P. MyD88: At the heart of inflammatory signaling and cardiovascular disease. J Mol Cell Cardiol. 2021; 161:75–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Balka KR, De Nardo D. Understanding early TLR signaling through the Myddosome. J Leukoc Biol. 2019; 105:339–351. [DOI] [PubMed] [Google Scholar]
- 114.Feng Y, Zhao H, Xu X, Buys ES, Raher MJ, Bopassa JC, Thibault H, Scherrer-Crosbie M, Schmidt U, Chao W. Innate immune adaptor MyD88 mediates neutrophil recruitment and myocardial injury after ischemia-reperfusion in mice. Am J Physiol Heart Circ Physiol. 2008; 295:H1311–H1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Singh MV, Swaminathan PD, Luczak ED, Kutschke W, Weiss RM, Anderson ME. MyD88 mediated inflammatory signaling leads to CaMKII oxidation, cardiac hypertrophy and death after myocardial infarction. J Mol Cell Cardiol. 2012; 52:1135–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Van Tassell BW, Seropian IM, Toldo S, Salloum FN, Smithson L, Varma A, Hoke NN, Gelwix C, Chau V, Abbate A. Pharmacologic inhibition of myeloid differentiation factor 88 (MyD88) prevents left ventricular dilation and hypertrophy after experimental acute myocardial infarction in the mouse. J Cardiovasc Pharmacol. 2010; 55:385–390. [DOI] [PubMed] [Google Scholar]
- 117.Feng Y, Zou L, Si R, Nagasaka Y, Chao W. Bone marrow MyD88 signaling modulates neutrophil function and ischemic myocardial injury. Am J Physiol Cell Physiol. 2010; 299:C760–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bajpai G, Bredemeyer A, Li W, Zaitsev K, Koenig AL, Lokshina I, Mohan J, Ivey B, Hsiao HM, Weinheimer C, Kovacs A, Epelman S, et al. Tissue Resident CCR2− and CCR2+ Cardiac Macrophages Differentially Orchestrate Monocyte Recruitment and Fate Specification Following Myocardial Injury. Circ Res. 2019; 124:263–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Bayer AL, Smolgovsky S, Ngwenyama N, Hernandez-Martinez A, Kaur K, Sulka K, Amrute J, Aronovitz M, Lavine K, Sharma S, Alcaide P. T-Cell MyD88 Is a Novel Regulator of Cardiac Fibrosis Through Modulation of T-Cell Activation. Circ Res. 2023; 133:412–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Li W, Feng G, Gauthier JM, Lokshina I, Higashikubo R, Evans S, Liu X, Hassan A, Tanaka S, Cicka M, Hsiao HM, Ruiz-Perez D, et al. Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J Clin Invest. 2019; 129:2293–2304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Singh MV, Cicha MZ, Nunez S, Meyerholz DK, Chapleau MW, Abboud FM. Angiotensin II-induced hypertension and cardiac hypertrophy are differentially mediated by TLR3- and TLR4-dependent pathways. Am J Physiol Heart Circ Physiol. 2019; 316:H1027–H1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sun SC. The non-canonical NF-kappaB pathway in immunity and inflammation. Nat Rev Immunol. 2017; 17:545–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Venkatachalam K, Prabhu SD, Reddy VS, Boylston WH, Valente AJ, Chandrasekar B. Neutralization of interleukin-18 ameliorates ischemia/reperfusion-induced myocardial injury. J Biol Chem. 2009; 284:7853–7865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Chandrasekar B, Freeman GL. Induction of nuclear factor kappaB and activation protein 1 in postischemic myocardium. FEBS Lett. 1997; 401:30–34. [DOI] [PubMed] [Google Scholar]
- 125.Chandrasekar B, Smith JB, Freeman GL. Ischemia-reperfusion of rat myocardium activates nuclear factor-KappaB and induces neutrophil infiltration via lipopolysaccharide-induced CXC chemokine. Circulation. 2001; 103:2296–2302. [DOI] [PubMed] [Google Scholar]
- 126.Hamid T, Guo SZ, Kingery JR, Xiang X, Dawn B, Prabhu SD. Cardiomyocyte NF-kappaB p65 promotes adverse remodelling, apoptosis, and endoplasmic reticulum stress in heart failure. Cardiovascular research. 2011; 89:129–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Brown M, McGuinness M, Wright T, Ren X, Wang Y, Boivin GP, Hahn H, Feldman AM, Jones WK. Cardiac-specific blockade of NF-kappaB in cardiac pathophysiology: differences between acute and chronic stimuli in vivo. American journal of physiology Heart and circulatory physiology. 2005; 289:H466–476. [DOI] [PubMed] [Google Scholar]
- 128.Zhang XQ, Tang R, Li L, Szucsik A, Javan H, Saegusa N, Spitzer KW, Selzman CH. Cardiomyocyte-specific p65 NF-kappaB deletion protects the injured heart by preservation of calcium handling. Am J Physiol Heart Circ Physiol. 2013; 305:H1089–1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Squadrito F, Deodato B, Squadrito G, Seminara P, Passaniti M, Venuti FS, Giacca M, Minutoli L, Adamo EB, Bellomo M, Marini R, Galeano M, Marini H, Altavilla D. Gene transfer of IkappaBalpha limits infarct size in a mouse model of myocardial ischemia-reperfusion injury. Lab Invest. 2003; 83:1097–1104. [DOI] [PubMed] [Google Scholar]
- 130.Morishita R, Sugimoto T, Aoki M, Kida I, Tomita N, Moriguchi A, Maeda K, Sawa Y, Kaneda Y, Higaki J, Ogihara T. In vivo transfection of cis element "decoy" against nuclear factor-kappaB binding site prevents myocardial infarction. Nat Med. 1997; 3:894–899. [DOI] [PubMed] [Google Scholar]
- 131.Onai Y, Suzuki J, Kakuta T, Maejima Y, Haraguchi G, Fukasawa H, Muto S, Itai A, Isobe M. Inhibition of IkappaB phosphorylation in cardiomyocytes attenuates myocardial ischemia/reperfusion injury. Cardiovascular research. 2004; 63:51–59. [DOI] [PubMed] [Google Scholar]
- 132.Moss NC, Stansfield WE, Willis MS, Tang RH, Selzman CH. IKKbeta inhibition attenuates myocardial injury and dysfunction following acute ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2007; 293:H2248–2253. [DOI] [PubMed] [Google Scholar]
- 133.Li HL, Zhuo ML, Wang D, Wang AB, Cai H, Sun LH, Yang Q, Huang Y, Wei YS, Liu PP, Liu DP, Liang CC. Targeted cardiac overexpression of A20 improves left ventricular performance and reduces compensatory hypertrophy after myocardial infarction. Circulation. 2007; 115:1885–1894. [DOI] [PubMed] [Google Scholar]
- 134.Trescher K, Bernecker O, Fellner B, Gyongyosi M, Schafer R, Aharinejad S, DeMartin R, Wolner E, Podesser BK. Inflammation and postinfarct remodeling: overexpression of IkappaB prevents ventricular dilation via increasing TIMP levels. Cardiovascular research. 2006; 69:746–754. [DOI] [PubMed] [Google Scholar]
- 135.Frantz S, Tillmanns J, Kuhlencordt PJ, Schmidt I, Adamek A, Dienesch C, Thum T, Gerondakis S, Ertl G, Bauersachs J. Tissue-specific effects of the nuclear factor kappaB subunit p50 on myocardial ischemia-reperfusion injury. The American journal of pathology. 2007; 171:507–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Frantz S, Hu K, Bayer B, Gerondakis S, Strotmann J, Adamek A, Ertl G, Bauersachs J. Absence of NF-kappaB subunit p50 improves heart failure after myocardial infarction. Faseb J. 2006; 20:1918–1920. [DOI] [PubMed] [Google Scholar]
- 137.Xuan YT, Tang XL, Banerjee S, Takano H, Li RC, Han H, Qiu Y, Li JJ, Bolli R. Nuclear factor-kappaB plays an essential role in the late phase of ischemic preconditioning in conscious rabbits. Circulation research. 1999; 84:1095–1109. [DOI] [PubMed] [Google Scholar]
- 138.Tranter M, Ren X, Forde T, Wilhide ME, Chen J, Sartor MA, Medvedovic M, Jones WK. NF-kappaB driven cardioprotective gene programs; Hsp70.3 and cardioprotection after late ischemic preconditioning. Journal of molecular and cellular cardiology. 2010; 49:664–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Evans S, Tzeng HP, Veis DJ, Matkovich S, Weinheimer C, Kovacs A, Barger PM, Mann DL. TNF receptor-activated factor 2 mediates cardiac protection through noncanonical NF-kappaB signaling. JCI Insight. 2018; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Timmers L, van Keulen JK, Hoefer IE, Meijs MF, van Middelaar B, den Ouden K, van Echteld CJ, Pasterkamp G, de Kleijn DP. Targeted deletion of nuclear factor kappaB p50 enhances cardiac remodeling and dysfunction following myocardial infarction. Circulation research. 2009; 104:699–706. [DOI] [PubMed] [Google Scholar]
- 141.Herskowitz A, Choi S, Ansari AA, Wesselingh S. Cytokine mRNA expression in postischemic/reperfused myocardium. Am J Pathol. 1995; 146:419–428. [PMC free article] [PubMed] [Google Scholar]
- 142.Frangogiannis NG, Lindsey ML, Michael LH, Youker KA, Bressler RB, Mendoza LH, Spengler RN, Smith CW, Entman ML. Resident cardiac mast cells degranulate and release preformed TNF-alpha, initiating the cytokine cascade in experimental canine myocardial ischemia/reperfusion. Circulation. 1998; 98:699–710. [DOI] [PubMed] [Google Scholar]
- 143.Maekawa N, Wada H, Kanda T, Niwa T, Yamada Y, Saito K, Fujiwara H, Sekikawa K, Seishima M. Improved myocardial ischemia/reperfusion injury in mice lacking tumor necrosis factor-alpha. J Am Coll Cardiol. 2002; 39:1229–1235. [DOI] [PubMed] [Google Scholar]
- 144.Kurrelmeyer KM, Michael LH, Baumgarten G, Taffet GE, Peschon JJ, Sivasubramanian N, Entman ML, Mann DL. Endogenous tumor necrosis factor protects the adult cardiac myocyte against ischemic-induced apoptosis in a murine model of acute myocardial infarction. Proc Natl Acad Sci U S A. 2000; 97:5456–5461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Misra A, Haudek SB, Knuefermann P, Vallejo JG, Chen ZJ, Michael LH, Sivasubramanian N, Olson EN, Entman ML, Mann DL. Nuclear factor-kappaB protects the adult cardiac myocyte against ischemia-induced apoptosis in a murine model of acute myocardial infarction. Circulation. 2003; 108:3075–3078. [DOI] [PubMed] [Google Scholar]
- 146.Garlie JB, Hamid T, Gu Y, Ismahil MA, Chandrasekar B, Prabhu SD. Tumor necrosis factor receptor 2 signaling limits beta-adrenergic receptor-mediated cardiac hypertrophy in vivo. Basic research in cardiology. 2011; 106:1193–1205. [DOI] [PubMed] [Google Scholar]
- 147.Hamid T, Gu Y, Ortines RV, Bhattacharya C, Wang G, Xuan YT, Prabhu SD. Divergent tumor necrosis factor receptor-related remodeling responses in heart failure: role of nuclear factor-kappaB and inflammatory activation. Circulation. 2009; 119:1386–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bujak M, Dobaczewski M, Chatila K, Mendoza LH, Li N, Reddy A, Frangogiannis NG. Interleukin-1 receptor type I signaling critically regulates infarct healing and cardiac remodeling. Am J Pathol. 2008; 173:57–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Toldo S, Mezzaroma E, Van Tassell BW, Farkas D, Marchetti C, Voelkel NF, Abbate A. Interleukin-1beta blockade improves cardiac remodelling after myocardial infarction without interrupting the inflammasome in the mouse. Exp Physiol. 2013; 98:734–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Abbate A, Salloum FN, Vecile E, Das A, Hoke NN, Straino S, Biondi-Zoccai GG, Houser JE, Qureshi IZ, Ownby ED, Gustini E, Biasucci LM, et al. Anakinra, a recombinant human interleukin-1 receptor antagonist, inhibits apoptosis in experimental acute myocardial infarction. Circulation. 2008; 117:2670–2683. [DOI] [PubMed] [Google Scholar]
- 151.De Jesus NM, Wang L, Lai J, Rigor RR, Francis Stuart SD, Bers DM, Lindsey ML, Ripplinger CM. Antiarrhythmic effects of interleukin 1 inhibition after myocardial infarction. Heart Rhythm. 2017; 14:727–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Kumar A, Thota V, Dee L, Olson J, Uretz E, Parrillo JE. Tumor necrosis factor alpha and interleukin 1beta are responsible for in vitro myocardial cell depression induced by human septic shock serum. J Exp Med. 1996; 183:949–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Gulick T, Chung MK, Pieper SJ, Lange LG, Schreiner GF. Interleukin 1 and tumor necrosis factor inhibit cardiac myocyte beta-adrenergic responsiveness. Proc Natl Acad Sci U S A. 1989; 86:6753–6757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Ing DJ, Zang J, Dzau VJ, Webster KA, Bishopric NH. Modulation of cytokine-induced cardiac myocyte apoptosis by nitric oxide, Bak, and Bcl-x. Circ Res. 1999; 84:21–33. [DOI] [PubMed] [Google Scholar]
- 155.Saxena A, Chen W, Su Y, Rai V, Uche OU, Li N, Frangogiannis NG. IL-1 Induces Proinflammatory Leukocyte Infiltration and Regulates Fibroblast Phenotype in the Infarcted Myocardium. J Immunol. 2013; 191:4838–4848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Leszczynski D, Josephs MD, Foegh ML. IL-1 beta-stimulated leucocyte-endothelial adhesion is regulated, in part, by the cyclic-GMP-dependent signal transduction pathway. Scand J Immunol. 1994; 39:551–556. [DOI] [PubMed] [Google Scholar]
- 157.Gwechenberger M, Mendoza LH, Youker KA, Frangogiannis NG, Smith CW, Michael LH, Entman ML. Cardiac myocytes produce interleukin-6 in culture and in viable border zone of reperfused infarctions. Circulation. 1999; 99:546–551. [DOI] [PubMed] [Google Scholar]
- 158.Alter C, Henseler AS, Owenier C, Hesse J, Ding Z, Lautwein T, Bahr J, Hayat S, Kramann R, Kostenis E, Scheller J, Schrader J. IL-6 in the infarcted heart is preferentially formed by fibroblasts and modulated by purinergic signaling. J Clin Invest. 2023; 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Hilfiker-Kleiner D, Shukla P, Klein G, Schaefer A, Stapel B, Hoch M, Muller W, Scherr M, Theilmeier G, Ernst M, Hilfiker A, Drexler H. Continuous glycoprotein-130-mediated signal transducer and activator of transcription-3 activation promotes inflammation, left ventricular rupture, and adverse outcome in subacute myocardial infarction. Circulation. 2010; 122:145–155. [DOI] [PubMed] [Google Scholar]
- 160.Kobara M, Noda K, Kitamura M, Okamoto A, Shiraishi T, Toba H, Matsubara H, Nakata T. Antibody against interleukin-6 receptor attenuates left ventricular remodelling after myocardial infarction in mice. Cardiovasc Res. 2010; 87:424–430. [DOI] [PubMed] [Google Scholar]
- 161.Rose-John S. Interleukin-6 Family Cytokines. Cold Spring Harb Perspect Biol. 2018; 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.George MJ, Jasmin NH, Cummings VT, Richard-Loendt A, Launchbury F, Woollard K, Turner-Stokes T, Garcia Diaz AI, Lythgoe M, Stuckey DJ, Hingorani AD, Gilroy DW. Selective Interleukin-6 Trans-Signaling Blockade Is More Effective Than Panantagonism in Reperfused Myocardial Infarction. JACC Basic Transl Sci. 2021; 6:431–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Kinugawa K, Takahashi T, Kohmoto O, Yao A, Aoyagi T, Momomura S, Hirata Y, Serizawa T. Nitric oxide-mediated effects of interleukin-6 on [Ca2+]i and cell contraction in cultured chick ventricular myocytes. Circ Res. 1994; 75:285–295. [DOI] [PubMed] [Google Scholar]
- 164.Melendez GC, McLarty JL, Levick SP, Du Y, Janicki JS, Brower GL. Interleukin 6 mediates myocardial fibrosis, concentric hypertrophy, and diastolic dysfunction in rats. Hypertension. 2010; 56:225–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Sano M, Fukuda K, Kodama H, Pan J, Saito M, Matsuzaki J, Takahashi T, Makino S, Kato T, Ogawa S. Interleukin-6 family of cytokines mediate angiotensin II-induced cardiac hypertrophy in rodent cardiomyocytes. J Biol Chem. 2000; 275:29717–29723. [DOI] [PubMed] [Google Scholar]
- 166.Kunisada K, Tone E, Fujio Y, Matsui H, Yamauchi-Takihara K, Kishimoto T. Activation of gp130 transduces hypertrophic signals via STAT3 in cardiac myocytes. Circulation. 1998; 98:346–352. [DOI] [PubMed] [Google Scholar]
- 167.Chou CH, Hung CS, Liao CW, Wei LH, Chen CW, Shun CT, Wen WF, Wan CH, Wu XM, Chang YY, Wu VC, Wu KD, Lin YH, Group TS. IL-6 trans-signalling contributes to aldosterone-induced cardiac fibrosis. Cardiovasc Res. 2018; 114:690–702. [DOI] [PubMed] [Google Scholar]
- 168.Leicht M, Briest W, Zimmer HG. Regulation of norepinephrine-induced proliferation in cardiac fibroblasts by interleukin-6 and p42/p44 mitogen activated protein kinase. Mol Cell Biochem. 2003; 243:65–72. [DOI] [PubMed] [Google Scholar]
- 169.Mir SA, Chatterjee A, Mitra A, Pathak K, Mahata SK, Sarkar S. Inhibition of signal transducer and activator of transcription 3 (STAT3) attenuates interleukin-6 (IL-6)-induced collagen synthesis and resultant hypertrophy in rat heart. J Biol Chem. 2012; 287:2666–2677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mitani H, Katayama N, Araki H, Ohishi K, Kobayashi K, Suzuki H, Nishii K, Masuya M, Yasukawa K, Minami N, Shiku H. Activity of interleukin 6 in the differentiation of monocytes to macrophages and dendritic cells. Br J Haematol. 2000; 109:288–295. [DOI] [PubMed] [Google Scholar]
- 171.Mauer J, Chaurasia B, Goldau J, Vogt MC, Ruud J, Nguyen KD, Theurich S, Hausen AC, Schmitz J, Bronneke HS, Estevez E, Allen TL, et al. Signaling by IL-6 promotes alternative activation of macrophages to limit endotoxemia and obesity-associated resistance to insulin. Nat Immunol. 2014; 15:423–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Sackett SD, Otto T, Mohs A, Sander LE, Strauch S, Streetz KL, Kroy DC, Trautwein C. Myeloid cells require gp130 signaling for protective anti-inflammatory functions during sepsis. FASEB J. 2019; 33:6035–6044. [DOI] [PubMed] [Google Scholar]
- 173.Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011; 1813:878–888. [DOI] [PubMed] [Google Scholar]
- 174.Kraakman MJ, Kammoun HL, Allen TL, Deswaerte V, Henstridge DC, Estevez E, Matthews VB, Neill B, White DA, Murphy AJ, Peijs L, Yang C, et al. Blocking IL-6 trans-signaling prevents high-fat diet-induced adipose tissue macrophage recruitment but does not improve insulin resistance. Cell Metab. 2015; 21:403–416. [DOI] [PubMed] [Google Scholar]
- 175.Sokol CL, Luster AD. The chemokine system in innate immunity. Cold Spring Harb Perspect Biol. 2015; 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Chen B, Frangogiannis NG. Chemokines in Myocardial Infarction. J Cardiovasc Transl Res. 2021; 14:35–52. [DOI] [PubMed] [Google Scholar]
- 177.Dewald O, Zymek P, Winkelmann K, Koerting A, Ren G, Abou-Khamis T, Michael LH, Rollins BJ, Entman ML, Frangogiannis NG. CCL2/Monocyte Chemoattractant Protein-1 regulates inflammatory responses critical to healing myocardial infarcts. Circ Res. 2005; 96:881–889. [DOI] [PubMed] [Google Scholar]
- 178.Grisanti LA, Traynham CJ, Repas AA, Gao E, Koch WJ, Tilley DG. beta2-Adrenergic receptor-dependent chemokine receptor 2 expression regulates leukocyte recruitment to the heart following acute injury. Proc Natl Acad Sci U S A. 2016; 113:15126–15131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wang J, Seo MJ, Deci MB, Weil BR, Canty JM, Nguyen J. Effect of CCR2 inhibitor-loaded lipid micelles on inflammatory cell migration and cardiac function after myocardial infarction. Int J Nanomedicine. 2018; 13:6441–6451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Leuschner F, Dutta P, Gorbatov R, Novobrantseva TI, Donahoe JS, Courties G, Lee KM, Kim JI, Markmann JF, Marinelli B, Panizzi P, Lee WW, et al. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat Biotechnol. 2011; 29:1005–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Lu W, Xie Z, Tang Y, Bai L, Yao Y, Fu C, Ma G. Photoluminescent Mesoporous Silicon Nanoparticles with siCCR2 Improve the Effects of Mesenchymal Stromal Cell Transplantation after Acute Myocardial Infarction. Theranostics. 2015; 5:1068–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Kaikita K, Hayasaki T, Okuma T, Kuziel WA, Ogawa H, Takeya M. Targeted deletion of CC chemokine receptor 2 attenuates left ventricular remodeling after experimental myocardial infarction. Am J Pathol. 2004; 165:439–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Montecucco F, Braunersreuther V, Lenglet S, Delattre BM, Pelli G, Buatois V, Guilhot F, Galan K, Vuilleumier N, Ferlin W, Fischer N, Vallee JP, Kosco-Vilbois M, Mach F. CC chemokine CCL5 plays a central role impacting infarct size and post-infarction heart failure in mice. Eur Heart J. 2012; 33:1964–1974. [DOI] [PubMed] [Google Scholar]
- 184.Braunersreuther V, Montecucco F, Pelli G, Galan K, Proudfoot AE, Belin A, Vuilleumier N, Burger F, Lenglet S, Caffa I, Soncini D, Nencioni A, Vallee JP, Mach F. Treatment with the CC chemokine-binding protein Evasin-4 improves post-infarction myocardial injury and survival in mice. Thromb Haemost. 2013; 110:807–825. [DOI] [PubMed] [Google Scholar]
- 185.Huang Y, Wang D, Wang X, Zhang Y, Liu T, Chen Y, Tang Y, Wang T, Hu D, Huang C. Abrogation of CC chemokine receptor 9 ameliorates ventricular remodeling in mice after myocardial infarction. Sci Rep. 2016; 6:32660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Jiang Y, Bai J, Tang L, Zhang P, Pu J. Anti-CCL21 antibody attenuates infarct size and improves cardiac remodeling after myocardial infarction. Cell Physiol Biochem. 2015; 37:979–990. [DOI] [PubMed] [Google Scholar]
- 187.Liehn EA, Merx MW, Postea O, Becher S, Djalali-Talab Y, Shagdarsuren E, Kelm M, Zernecke A, Weber C. Ccr1 deficiency reduces inflammatory remodelling and preserves left ventricular function after myocardial infarction. J Cell Mol Med. 2008; 12:496–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Feng G, Bajpai G, Ma P, Koenig A, Bredemeyer A, Lokshina I, Lai L, Forster I, Leuschner F, Kreisel D, Lavine KJ. CCL17 Aggravates Myocardial Injury by Suppressing Recruitment of Regulatory T Cells. Circulation. 2022; 145:765–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Dobaczewski M, Xia Y, Bujak M, Gonzalez-Quesada C, Frangogiannis NG. CCR5 signaling suppresses inflammation and reduces adverse remodeling of the infarcted heart, mediating recruitment of regulatory T cells. Am J Pathol. 2010; 176:2177–2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Kukielka GL, Smith CW, LaRosa GJ, Manning AM, Mendoza LH, Daly TJ, Hughes BJ, Youker KA, Hawkins HK, Michael LH, et al. Interleukin-8 gene induction in the myocardium after ischemia and reperfusion in vivo. J Clin Invest. 1995; 95:89–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Ivey CL, Williams FM, Collins PD, Jose PJ, Williams TJ. Neutrophil chemoattractants generated in two phases during reperfusion of ischemic myocardium in the rabbit. Evidence for a role for C5a and interleukin-8. J Clin Invest. 1995; 95:2720–2728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Boyle EM Jr., Kovacich JC, Hebert CA, Canty TG Jr., Chi E, Morgan EN, Pohlman TH, Verrier ED. Inhibition of interleukin-8 blocks myocardial ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 1998; 116:114–121. [DOI] [PubMed] [Google Scholar]
- 193.Tarzami ST, Miao W, Mani K, Lopez L, Factor SM, Berman JW, Kitsis RN. Opposing effects mediated by the chemokine receptor CXCR2 on myocardial ischemia-reperfusion injury: recruitment of potentially damaging neutrophils and direct myocardial protection. Circulation. 2003; 108:2387–2392. [DOI] [PubMed] [Google Scholar]
- 194.Oral H, Kanzler I, Tuchscheerer N, Curaj A, Simsekyilmaz S, Sonmez TT, Radu E, Postea O, Weber C, Schuh A, Liehn EA. CXC chemokine KC fails to induce neutrophil infiltration and neoangiogenesis in a mouse model of myocardial infarction. J Mol Cell Cardiol. 2013; 60:1–7. [DOI] [PubMed] [Google Scholar]
- 195.Xie Q, Sun Z, Chen M, Zhong Q, Yang T, Yi J. IL-8 up-regulates proliferative angiogenesis in ischemic myocardium in rabbits through phosphorylation of Akt/GSK-3beta(ser9) dependent pathways. Int J Clin Exp Med. 2015; 8:12498–12508. [PMC free article] [PubMed] [Google Scholar]
- 196.Liehn EA, Kanzler I, Konschalla S, Kroh A, Simsekyilmaz S, Sonmez TT, Bucala R, Bernhagen J, Weber C. Compartmentalized protective and detrimental effects of endogenous macrophage migration-inhibitory factor mediated by CXCR2 in a mouse model of myocardial ischemia/reperfusion. Arterioscler Thromb Vasc Biol. 2013; 33:2180–2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Moser B, Loetscher P. Lymphocyte traffic control by chemokines. Nat Immunol. 2001; 2:123–128. [DOI] [PubMed] [Google Scholar]
- 198.Fiorina P, Ansari MJ, Jurewicz M, Barry M, Ricchiuti V, Smith RN, Shea S, Means TK, Auchincloss H Jr., Luster AD, Sayegh MH, Abdi R. Role of CXC chemokine receptor 3 pathway in renal ischemic injury. J Am Soc Nephrol. 2006; 17:716–723. [DOI] [PubMed] [Google Scholar]
- 199.Khan IA, MacLean JA, Lee FS, Casciotti L, DeHaan E, Schwartzman JD, Luster AD. IP-10 is critical for effector T cell trafficking and host survival in Toxoplasma gondii infection. Immunity. 2000; 12:483–494. [DOI] [PubMed] [Google Scholar]
- 200.Shiraha H, Glading A, Gupta K, Wells A. IP-10 inhibits epidermal growth factor-induced motility by decreasing epidermal growth factor receptor-mediated calpain activity. J Cell Biol. 1999; 146:243–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Strieter RM, Polverini PJ, Arenberg DA, Walz A, Opdenakker G, Van Damme J, Kunkel SL. Role of C-X-C chemokines as regulators of angiogenesis in lung cancer. J Leukoc Biol. 1995; 57:752–762. [DOI] [PubMed] [Google Scholar]
- 202.Strieter RM, Polverini PJ, Kunkel SL, Arenberg DA, Burdick MD, Kasper J, Dzuiba J, Van Damme J, Walz A, Marriott D, et al. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J Biol Chem. 1995; 270:27348–27357. [DOI] [PubMed] [Google Scholar]
- 203.Frangogiannis NG, Mendoza LH, Lewallen M, Michael LH, Smith CW, Entman ML. Induction and suppression of interferon-inducible protein 10 in reperfused myocardial infarcts may regulate angiogenesis. FASEB J. 2001; 15:1428–1430. [DOI] [PubMed] [Google Scholar]
- 204.Dewald O, Ren G, Duerr GD, Zoerlein M, Klemm C, Gersch C, Tincey S, Michael LH, Entman ML, Frangogiannis NG. Of mice and dogs: species-specific differences in the inflammatory response following myocardial infarction. Am J Pathol. 2004; 164:665–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Bujak M, Dobaczewski M, Gonzalez-Quesada C, Xia Y, Leucker T, Zymek P, Veeranna V, Tager AM, Luster AD, Frangogiannis NG. Induction of the CXC chemokine interferon-gamma-inducible protein 10 regulates the reparative response following myocardial infarction. Circ Res. 2009; 105:973–983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Saxena A, Bujak M, Frunza O, Dobaczewski M, Gonzalez-Quesada C, Lu B, Gerard C, Frangogiannis NG. CXCR3-independent actions of the CXC chemokine CXCL10 in the infarcted myocardium and in isolated cardiac fibroblasts are mediated through proteoglycans. Cardiovasc Res. 2014; 103:217–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Nagasawa T, Hirota S, Tachibana K, Takakura N, Nishikawa S, Kitamura Y, Yoshida N, Kikutani H, Kishimoto T. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996; 382:635–638. [DOI] [PubMed] [Google Scholar]
- 208.Pillarisetti K, Gupta SK. Cloning and relative expression analysis of rat stromal cell derived factor-1 (SDF-1)1: SDF-1 alpha mRNA is selectively induced in rat model of myocardial infarction. Inflammation. 2001; 25:293–300. [DOI] [PubMed] [Google Scholar]
- 209.Askari AT, Unzek S, Popovic ZB, Goldman CK, Forudi F, Kiedrowski M, Rovner A, Ellis SG, Thomas JD, DiCorleto PE, Topol EJ, Penn MS. Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy. Lancet. 2003; 362:697–703. [DOI] [PubMed] [Google Scholar]
- 210.Frangogiannis NG. The stromal cell-derived factor-1/CXCR4 axis in cardiac injury and repair. J Am Coll Cardiol. 2011; 58:2424–2426. [DOI] [PubMed] [Google Scholar]
- 211.Uematsu M, Yoshizaki T, Shimizu T, Obata JE, Nakamura T, Fujioka D, Watanabe K, Watanabe Y, Kugiyama K. Sustained myocardial production of stromal cell-derived factor-1alpha was associated with left ventricular adverse remodeling in patients with myocardial infarction. Am J Physiol Heart Circ Physiol. 2015; 309:H1764–1771. [DOI] [PubMed] [Google Scholar]
- 212.Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004; 10:858–864. [DOI] [PubMed] [Google Scholar]
- 213.Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, Giordano FJ. Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation. 2004; 110:3300–3305. [DOI] [PubMed] [Google Scholar]
- 214.Ma J, Ge J, Zhang S, Sun A, Shen J, Chen L, Wang K, Zou Y. Time course of myocardial stromal cell-derived factor 1 expression and beneficial effects of intravenously administered bone marrow stem cells in rats with experimental myocardial infarction. Basic Res Cardiol. 2005; 100:217–223. [DOI] [PubMed] [Google Scholar]
- 215.Chen J, Chemaly E, Liang L, Kho C, Lee A, Park J, Altman P, Schecter AD, Hajjar RJ, Tarzami ST. Effects of CXCR4 gene transfer on cardiac function after ischemia-reperfusion injury. Am J Pathol. 2010; 176:1705–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Saxena A, Fish JE, White MD, Yu S, Smyth JW, Shaw RM, DiMaio JM, Srivastava D. Stromal cell-derived factor-1alpha is cardioprotective after myocardial infarction. Circulation. 2008; 117:2224–2231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Zhang M, Mal N, Kiedrowski M, Chacko M, Askari AT, Popovic ZB, Koc ON, Penn MS. SDF-1 expression by mesenchymal stem cells results in trophic support of cardiac myocytes after myocardial infarction. Faseb J. 2007; 21:3197–3207. [DOI] [PubMed] [Google Scholar]
- 218.Sasaki T, Fukazawa R, Ogawa S, Kanno S, Nitta T, Ochi M, Shimizu K. Stromal cell-derived factor-1alpha improves infarcted heart function through angiogenesis in mice. Pediatr Int. 2007; 49:966–971. [DOI] [PubMed] [Google Scholar]
- 219.MacArthur JW Jr., Purcell BP, Shudo Y, Cohen JE, Fairman A, Trubelja A, Patel J, Hsiao P, Yang E, Lloyd K, Hiesinger W, Atluri P, Burdick JA, Woo YJ. Sustained release of engineered stromal cell-derived factor 1-alpha from injectable hydrogels effectively recruits endothelial progenitor cells and preserves ventricular function after myocardial infarction. Circulation. 2013; 128:S79–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Macarthur JW Jr., Cohen JE, McGarvey JR, Shudo Y, Patel JB, Trubelja A, Fairman AS, Edwards BB, Hung G, Hiesinger W, Goldstone AB, Atluri P, et al. Preclinical evaluation of the engineered stem cell chemokine stromal cell-derived factor 1alpha analog in a translational ovine myocardial infarction model. Circ Res. 2014; 114:650–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Elmadbouh I, Haider H, Jiang S, Idris NM, Lu G, Ashraf M. Ex vivo delivered stromal cell-derived factor-1alpha promotes stem cell homing and induces angiomyogenesis in the infarcted myocardium. J Mol Cell Cardiol. 2007; 42:792–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Mayorga ME, Kiedrowski M, McCallinhart P, Forudi F, Ockunzzi J, Weber K, Chilian W, Penn MS, Dong F. Role of SDF-1:CXCR4 in Impaired Post-Myocardial Infarction Cardiac Repair in Diabetes. Stem Cells Transl Med. 2018; 7:115–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Cheng M, Huang K, Zhou J, Yan D, Tang YL, Zhao TC, Miller RJ, Kishore R, Losordo DW, Qin G. A critical role of Src family kinase in SDF-1/CXCR4-mediated bone-marrow progenitor cell recruitment to the ischemic heart. J Mol Cell Cardiol. 2015; 81:49–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Wang M, Hu R, Yang Y, Xiang L, Mu Y. In Vivo Ultrasound Molecular Imaging of SDF-1 Expression in a Swine Model of Acute Myocardial Infarction. Front Pharmacol. 2019; 10:899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Huang FY, Xia TL, Li JL, Li CM, Zhao ZG, Lei WH, Chen L, Liao YB, Xiao D, Peng Y, Wang YB, Liu XJ, Chen M. The bifunctional SDF-1-AnxA5 fusion protein protects cardiac function after myocardial infarction. J Cell Mol Med. 2019; 23:7673–7684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Goldstone AB, Burnett CE, Cohen JE, Paulsen MJ, Eskandari A, Edwards BE, Ingason AB, Steele AN, Patel JB, MacArthur JW, Shizuru JA, Woo YJ. SDF 1-alpha Attenuates Myocardial Injury Without Altering the Direct Contribution of Circulating Cells. J Cardiovasc Transl Res. 2018; 11:274–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Liu Y, Gao S, Wang Z, Yang Y, Huo H, Tian X. Effect of stromal cell-derived factor-1 on myocardial apoptosis and cardiac function recovery in rats with acute myocardial infarction. Exp Ther Med. 2016; 12:3282–3286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Hu X, Dai S, Wu WJ, Tan W, Zhu X, Mu J, Guo Y, Bolli R, Rokosh G. Stromal cell derived factor-1 alpha confers protection against myocardial ischemia/reperfusion injury: role of the cardiac stromal cell derived factor-1 alpha CXCR4 axis. Circulation. 2007; 116:654–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Dai S, Yuan F, Mu J, Li C, Chen N, Guo S, Kingery J, Prabhu SD, Bolli R, Rokosh G. Chronic AMD3100 antagonism of SDF-1alpha-CXCR4 exacerbates cardiac dysfunction and remodeling after myocardial infarction. J Mol Cell Cardiol. 2010; 49:587–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Proulx C, El-Helou V, Gosselin H, Clement R, Gillis MA, Villeneuve L, Calderone A. Antagonism of stromal cell-derived factor-1alpha reduces infarct size and improves ventricular function after myocardial infarction. Pflugers Arch. 2007; 455:241–250. [DOI] [PubMed] [Google Scholar]
- 231.Jujo K, Hamada H, Iwakura A, Thorne T, Sekiguchi H, Clarke T, Ito A, Misener S, Tanaka T, Klyachko E, Kobayashi K, Tongers J, et al. CXCR4 blockade augments bone marrow progenitor cell recruitment to the neovasculature and reduces mortality after myocardial infarction. Proc Natl Acad Sci U S A. 2010; 107:11008–11013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, Brija T, Gautier EL, Ivanov S, Satpathy AT, Schilling JD, Schwendener R, et al. Embryonic and Adult-Derived Resident Cardiac Macrophages Are Maintained through Distinct Mechanisms at Steady State and during Inflammation. Immunity. 2014; 40:91–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Bajpai G, Schneider C, Wong N, Bredemeyer A, Hulsmans M, Nahrendorf M, Epelman S, Kreisel D, Liu Y, Itoh A, Shankar TS, Selzman CH, Drakos SG, Lavine KJ. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat Med. 2018; 24:1234–1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Chakarov S, Lim HY, Tan L, Lim SY, See P, Lum J, Zhang XM, Foo S, Nakamizo S, Duan K, Kong WT, Gentek R, et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science. 2019; 363. [DOI] [PubMed] [Google Scholar]
- 235.Dederichs TS, Yerdenova A, Horstmann H, Vico TA, Nubling S, Peyronnet R, Pfeifer D, von Zur Muehlen C, Heidt T, Wolf D, Czerny M, Westermann D, Hilgendorf I. Nonpreferential but Detrimental Accumulation of Macrophages With Clonal Hematopoiesis-Driver Mutations in Cardiovascular Tissues-Brief Report. Arterioscler Thromb Vasc Biol. 2024; 44:690–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Nicolas-Avila JA, Lechuga-Vieco AV, Esteban-Martinez L, Sanchez-Diaz M, Diaz-Garcia E, Santiago DJ, Rubio-Ponce A, Li JL, Balachander A, Quintana JA, Martinez-de-Mena R, Castejon-Vega B, et al. A Network of Macrophages Supports Mitochondrial Homeostasis in the Heart. Cell. 2020; 183:94–109 e123. [DOI] [PubMed] [Google Scholar]
- 237.Hulsmans M, Clauss S, Xiao L, Aguirre AD, King KR, Hanley A, Hucker WJ, Wulfers EM, Seemann G, Courties G, Iwamoto Y, Sun Y, et al. Macrophages Facilitate Electrical Conduction in the Heart. Cell. 2017; 169:510–522 e520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Hulsmans M, Sager HB, Roh JD, Valero-Munoz M, Houstis NE, Iwamoto Y, Sun Y, Wilson RM, Wojtkiewicz G, Tricot B, Osborne MT, Hung J, et al. Cardiac macrophages promote diastolic dysfunction. J Exp Med. 2018; 215:423–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Dick SA, Wong A, Hamidzada H, Nejat S, Nechanitzky R, Vohra S, Mueller B, Zaman R, Kantores C, Aronoff L, Momen A, Nechanitzky D, et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Sci Immunol. 2022; 7:eabf7777. [DOI] [PubMed] [Google Scholar]
- 240.Zaman R, Hamidzada H, Kantores C, Wong A, Dick SA, Wang Y, Momen A, Aronoff L, Lin J, Razani B, Mital S, Billia F, et al. Selective loss of resident macrophage-derived insulin-like growth factor-1 abolishes adaptive cardiac growth to stress. Immunity. 2021; 54:2057–2071 e2056. [DOI] [PubMed] [Google Scholar]
- 241.Dick SA, Macklin JA, Nejat S, Momen A, Clemente-Casares X, Althagafi MG, Chen J, Kantores C, Hosseinzadeh S, Aronoff L, Wong A, Zaman R, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. 2019; 20:29–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Hilgendorf I, Gerhardt LM, Tan TC, Winter C, Holderried TA, Chousterman BG, Iwamoto Y, Liao R, Zirlik A, Scherer-Crosbie M, Hedrick CC, Libby P, et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res. 2014; 114:1611–1622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Sager HB, Hulsmans M, Lavine KJ, Moreira MB, Heidt T, Courties G, Sun Y, Iwamoto Y, Tricot B, Khan OF, Dahlman JE, Borodovsky A, et al. Proliferation and Recruitment Contribute to Myocardial Macrophage Expansion in Chronic Heart Failure. Circ Res. 2016; 119:853–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Jung K, Kim P, Leuschner F, Gorbatov R, Kim JK, Ueno T, Nahrendorf M, Yun SH. Endoscopic time-lapse imaging of immune cells in infarcted mouse hearts. Circ Res. 2013; 112:891–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Li W, Hsiao HM, Higashikubo R, Saunders BT, Bharat A, Goldstein DR, Krupnick AS, Gelman AE, Lavine KJ, Kreisel D. Heart-resident CCR2(+) macrophages promote neutrophil extravasation through TLR9/MyD88/CXCL5 signaling. JCI Insight. 2016; 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Deniset JF, Belke D, Lee WY, Jorch SK, Deppermann C, Hassanabad AF, Turnbull JD, Teng G, Rozich I, Hudspeth K, Kanno Y, Brooks SR, et al. Gata6(+) Pericardial Cavity Macrophages Relocate to the Injured Heart and Prevent Cardiac Fibrosis. Immunity. 2019; 51:131–140 e135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Jin H, Liu K, Huang X, Huo H, Mou J, Qiao Z, He B, Zhou B. Genetic Lineage Tracing of Pericardial Cavity Macrophages in the Injured Heart. Circ Res. 2022; 130:1682–1697. [DOI] [PubMed] [Google Scholar]
- 248.Frantz S, Hofmann U, Fraccarollo D, Schafer A, Kranepuhl S, Hagedorn I, Nieswandt B, Nahrendorf M, Wagner H, Bayer B, Pachel C, Schon MP, et al. Monocytes/macrophages prevent healing defects and left ventricular thrombus formation after myocardial infarction. FASEB J. 2013; 27:871–881. [DOI] [PubMed] [Google Scholar]
- 249.van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH, van Luyn MJ. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. Am J Pathol. 2007; 170:818–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Grune J, Lewis AJM, Yamazoe M, Hulsmans M, Rohde D, Xiao L, Zhang S, Ott C, Calcagno DM, Zhou Y, Timm K, Shanmuganathan M, et al. Neutrophils incite and macrophages avert electrical storm after myocardial infarction. Nat Cardiovasc Res. 2022; 1:649–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Wan E, Yeap XY, Dehn S, Terry R, Novak M, Zhang S, Iwata S, Han X, Homma S, Drosatos K, Lomasney J, Engman DM, et al. Enhanced efferocytosis of apoptotic cardiomyocytes through myeloid-epithelial-reproductive tyrosine kinase links acute inflammation resolution to cardiac repair after infarction. Circ Res. 2013; 113:1004–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Sugita J, Fujiu K, Nakayama Y, Matsubara T, Matsuda J, Oshima T, Liu Y, Maru Y, Hasumi E, Kojima T, Seno H, Asano K, et al. Cardiac macrophages prevent sudden death during heart stress. Nature communications. 2021; 12:1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Zhang S, Yeap XY, DeBerge M, Naresh NK, Wang K, Jiang Z, Wilcox JE, White SM, Morrow JP, Burridge PW, Procissi D, Scott EA, Frazier W, Thorp EB. Acute CD47 Blockade During Ischemic Myocardial Reperfusion Enhances Phagocytosis-Associated Cardiac Repair. JACC Basic Transl Sci. 2017; 2:386–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018; 15:203–214. [DOI] [PubMed] [Google Scholar]
- 255.Ye X, Zhang P, Zhang Y, Luan J, Xu C, Wu Z, Ju D, Hu W. GSDMD contributes to myocardial reperfusion injury by regulating pyroptosis. Front Immunol. 2022; 13:893914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Mouton AJ, DeLeon-Pennell KY, Rivera Gonzalez OJ, Flynn ER, Freeman TC, Saucerman JJ, Garrett MR, Ma Y, Harmancey R, Lindsey ML. Mapping macrophage polarization over the myocardial infarction time continuum. Basic Res Cardiol. 2018; 113:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Farbehi N, Patrick R, Dorison A, Xaymardan M, Janbandhu V, Wystub-Lis K, Ho JW, Nordon RE, Harvey RP. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. Elife. 2019; 8:e43882. doi: 10.7554/eLife.43882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Jung SH, Hwang BH, Shin S, Park EH, Park SH, Kim CW, Kim E, Choo E, Choi IJ, Swirski FK, Chang K, Chung YJ. Spatiotemporal dynamics of macrophage heterogeneity and a potential function of Trem2(hi) macrophages in infarcted hearts. Nature communications. 2022; 13:4580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Kaminski AR, Moore ET, Daseke MJ 2nd, Valerio FM, Flynn ER, Lindsey ML. The compendium of matrix metalloproteinase expression in the left ventricle of mice following myocardial infarction. Am J Physiol Heart Circ Physiol. 2020; 318:H706–H714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Varricchi G, Marone G, Kovanen PT. Cardiac Mast Cells: Underappreciated Immune Cells in Cardiovascular Homeostasis and Disease. Trends Immunol. 2020; 41:734–746. [DOI] [PubMed] [Google Scholar]
- 261.Frangogiannis NG, Burns AR, Michael LH, Entman ML. Histochemical and morphological characteristics of canine cardiac mast cells. Histochem J. 1999; 31:221–229. [DOI] [PubMed] [Google Scholar]
- 262.Hart DN, Fabre JW. Demonstration and characterization of Ia-positive dendritic cells in the interstitial connective tissues of rat heart and other tissues, but not brain. J Exp Med. 1981; 154:347–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Forte E, Perkins B, Sintou A, Kalkat HS, Papanikolaou A, Jenkins C, Alsubaie M, Chowdhury RA, Duffy TM, Skelly DA, Branca J, Bellahcene M, et al. Cross-Priming Dendritic Cells Exacerbate Immunopathology After Ischemic Tissue Damage in the Heart. Circulation. 2021; 143:821–836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Somasundaram P, Ren G, Nagar H, Kraemer D, Mendoza L, Michael LH, Caughey GH, Entman ML, Frangogiannis NG. Mast cell tryptase may modulate endothelial cell phenotype in healing myocardial infarcts. J Pathol. 2005; 205:102–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D'Antoni ML, Debuque R, Chandran A, Wang L, Arora K, Rosenthal NA, Tallquist MD. Revisiting Cardiac Cellular Composition. Circ Res. 2016; 118:400–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Anzai A, Choi JL, He S, Fenn AM, Nairz M, Rattik S, McAlpine CS, Mindur JE, Chan CT, Iwamoto Y, Tricot B, Wojtkiewicz GR, et al. The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes. J Exp Med. 2017; 214:3293–3310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Weyrich AS, Ma XY, Lefer DJ, Albertine KH, Lefer AM. In vivo neutralization of P-selectin protects feline heart and endothelium in myocardial ischemia and reperfusion injury. J Clin Invest. 1993; 91:2620–2629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Asako H, Kurose I, Wolf R, DeFrees S, Zheng ZL, Phillips ML, Paulson JC, Granger DN. Role of H1 receptors and P-selectin in histamine-induced leukocyte rolling and adhesion in postcapillary venules. J Clin Invest. 1994; 93:1508–1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013; 13:159–175. [DOI] [PubMed] [Google Scholar]
- 270.McEver RP. Selectins: initiators of leucocyte adhesion and signalling at the vascular wall. Cardiovasc Res. 2015; 107:331–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Detmers PA, Lo SK, Olsen-Egbert E, Walz A, Baggiolini M, Cohn ZA. Neutrophil-activating protein 1/interleukin 8 stimulates the binding activity of the leukocyte adhesion receptor CD11b/CD18 on human neutrophils. J Exp Med. 1990; 171:1155–1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Herter J, Zarbock A. Integrin Regulation during Leukocyte Recruitment. J Immunol. 2013; 190:4451–4457. [DOI] [PubMed] [Google Scholar]
- 273.Wang S, Voisin MB, Larbi KY, Dangerfield J, Scheiermann C, Tran M, Maxwell PH, Sorokin L, Nourshargh S. Venular basement membranes contain specific matrix protein low expression regions that act as exit points for emigrating neutrophils. J Exp Med. 2006; 203:1519–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Proebstl D, Voisin MB, Woodfin A, Whiteford J, D'Acquisto F, Jones GE, Rowe D, Nourshargh S. Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J Exp Med. 2012; 209:1219–1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Gronloh MLB, Arts JJG, van Buul JD. Neutrophil transendothelial migration hotspots - mechanisms and implications. J Cell Sci. 2021; 134. [DOI] [PubMed] [Google Scholar]
- 276.Nees S, Weiss DR, Senftl A, Knott M, Forch S, Schnurr M, Weyrich P, Juchem G. Isolation, bulk cultivation, and characterization of coronary microvascular pericytes: the second most frequent myocardial cell type in vitro. Am J Physiol Heart Circ Physiol. 2012; 302:H69–84. [DOI] [PubMed] [Google Scholar]
- 277.Alex L, Tuleta I, Harikrishnan V, Frangogiannis NG. Validation of Specific and Reliable Genetic Tools to Identify, Label, and Target Cardiac Pericytes in Mice. J Am Heart Assoc. 2022; 11:e023171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Quijada P, Park S, Zhao P, Kolluri KS, Wong D, Shih KD, Fang K, Pezhouman A, Wang L, Daraei A, Tran MD, Rathbun EM, et al. Cardiac pericytes mediate the remodeling response to myocardial infarction. J Clin Invest. 2023; 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Alex L, Tuleta I, Hernandez SC, Hanna A, Venugopal H, Astorkia M, Humeres C, Kubota A, Su K, Zheng D, Frangogiannis NG. Cardiac Pericytes Acquire a Fibrogenic Phenotype and Contribute to Vascular Maturation After Myocardial Infarction. Circulation. 2023; 148:882–898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Siao CJ, Lorentz CU, Kermani P, Marinic T, Carter J, McGrath K, Padow VA, Mark W, Falcone DJ, Cohen-Gould L, Parrish DC, Habecker BA, et al. ProNGF, a cytokine induced after myocardial infarction in humans, targets pericytes to promote microvascular damage and activation. J Exp Med. 2012; 209:2291–2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Allencherril J, Jneid H, Atar D, Alam M, Levine G, Kloner RA, Birnbaum Y. Pathophysiology, Diagnosis, and Management of the No-Reflow Phenomenon. Cardiovasc Drugs Ther. 2019; 33:589–597. [DOI] [PubMed] [Google Scholar]
- 282.O'Farrell FM, Mastitskaya S, Hammond-Haley M, Freitas F, Wah WR, Attwell D. Capillary pericytes mediate coronary no-reflow after myocardial ischaemia. Elife. 2017; 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Yan X, Anzai A, Katsumata Y, Matsuhashi T, Ito K, Endo J, Yamamoto T, Takeshima A, Shinmura K, Shen W, Fukuda K, Sano M. Temporal dynamics of cardiac immune cell accumulation following acute myocardial infarction. J Mol Cell Cardiol. 2013; 62:24–35. [DOI] [PubMed] [Google Scholar]
- 284.Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011; 11:519–531. [DOI] [PubMed] [Google Scholar]
- 285.Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nature reviews Immunology. 2008; 8:349–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Soehnlein O, Lindbom L. Phagocyte partnership during the onset and resolution of inflammation. Nat Rev Immunol. 2010; 10:427–439. [DOI] [PubMed] [Google Scholar]
- 287.Sreejit G, Nooti SK, Jaggers RM, Athmanathan B, Ho Park K, Al-Sharea A, Johnson J, Dahdah A, Lee MKS, Ma J, Murphy AJ, Nagareddy PR. Retention of the NLRP3 Inflammasome-Primed Neutrophils in the Bone Marrow Is Essential for Myocardial Infarction-Induced Granulopoiesis. Circulation. 2022; 145:31–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Ali M, Pulli B, Courties G, Tricot B, Sebas M, Iwamoto Y, Hilgendorf I, Schob S, Dong A, Zheng W, Skoura A, Kalgukar A, et al. Myeloperoxidase Inhibition Improves Ventricular Function and Remodeling After Experimental Myocardial Infarction. JACC Basic Transl Sci. 2016; 1:633–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Guthoff H, Hof A, Klinke A, Maass M, Konradi J, Mehrkens D, Geissen S, Nettersheim FS, Braumann S, Michaelsson E, Nies RJ, Lee S, et al. Protective Effects of Therapeutic Neutrophil Depletion and Myeloperoxidase Inhibition on Left Ventricular Function and Remodeling in Myocardial Infarction. Antioxidants (Basel). 2022; 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Hoffmeyer MR, Jones SP, Ross CR, Sharp B, Grisham MB, Laroux FS, Stalker TJ, Scalia R, Lefer DJ. Myocardial ischemia/reperfusion injury in NADPH oxidase-deficient mice. Circ Res. 2000; 87:812–817. [DOI] [PubMed] [Google Scholar]
- 291.Ma Y, Yabluchanskiy A, Iyer RP, Cannon PL, Flynn ER, Jung M, Henry J, Cates CA, Deleon-Pennell KY, Lindsey ML. Temporal neutrophil polarization following myocardial infarction. Cardiovasc Res. 2016; 110:51–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Vafadarnejad E, Rizzo G, Krampert L, Arampatzi P, Arias-Loza AP, Nazzal Y, Rizakou A, Knochenhauer T, Bandi SR, Nugroho VA, Schulz DJJ, Roesch M, et al. Dynamics of Cardiac Neutrophil Diversity in Murine Myocardial Infarction. Circ Res. 2020; 127:e232–e249. [DOI] [PubMed] [Google Scholar]
- 293.Savchenko AS, Borissoff JI, Martinod K, De Meyer SF, Gallant M, Erpenbeck L, Brill A, Wang Y, Wagner DD. VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice. Blood. 2014; 123:141–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Schloss MJ, Horckmans M, Nitz K, Duchene J, Drechsler M, Bidzhekov K, Scheiermann C, Weber C, Soehnlein O, Steffens S. The time-of-day of myocardial infarction onset affects healing through oscillations in cardiac neutrophil recruitment. EMBO Mol Med. 2016; 8:937–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, Weber C, Soehnlein O, Steffens S. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J. 2017; 38:187–197. [DOI] [PubMed] [Google Scholar]
- 296.Ferraro B, Leoni G, Hinkel R, Ormanns S, Paulin N, Ortega-Gomez A, Viola JR, de Jong R, Bongiovanni D, Bozoglu T, Maas SL, D'Amico M, et al. Pro-Angiogenic Macrophage Phenotype to Promote Myocardial Repair. J Am Coll Cardiol. 2019; 73:2990–3002. [DOI] [PubMed] [Google Scholar]
- 297.Kubota A, Suto A, Suzuki K, Kobayashi Y, Nakajima H. Matrix metalloproteinase-12 produced by Ly6C(low) macrophages prolongs the survival after myocardial infarction by preventing neutrophil influx. J Mol Cell Cardiol. 2019; 131:41–52. [DOI] [PubMed] [Google Scholar]
- 298.Hilgendorf I, Swirski FK, Robbins CS. Monocyte fate in atherosclerosis. Arterioscler Thromb Vasc Biol. 2015; 35:272–279. [DOI] [PubMed] [Google Scholar]
- 299.Villani AC, Satija R, Reynolds G, Sarkizova S, Shekhar K, Fletcher J, Griesbeck M, Butler A, Zheng S, Lazo S, Jardine L, Dixon D, et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. 2017; 356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Hamers AAJ, Dinh HQ, Thomas GD, Marcovecchio P, Blatchley A, Nakao CS, Kim C, McSkimming C, Taylor AM, Nguyen AT, McNamara CA, Hedrick CC. Human Monocyte Heterogeneity as Revealed by High-Dimensional Mass Cytometry. Arterioscler Thromb Vasc Biol. 2019; 39:25–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Berg KE, Ljungcrantz I, Andersson L, Bryngelsson C, Hedblad B, Fredrikson GN, Nilsson J, Bjorkbacka H. Elevated CD14++CD16− monocytes predict cardiovascular events. Circ Cardiovasc Genet. 2012; 5:122–131. [DOI] [PubMed] [Google Scholar]
- 302.Askari N, Lipps C, Voss S, Staubach N, Grun D, Klingenberg R, von Jeinsen B, Wolter JS, Kriechbaum S, Dorr O, Nef H, Liebetrau C, Hamm CW, Keller T. Circulating Monocyte Subsets Are Associated With Extent of Myocardial Injury but Not With Type of Myocardial Infarction. Front Cardiovasc Med. 2021; 8:741890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Boidin M, Lip GYH, Shantsila A, Thijssen D, Shantsila E. Dynamic changes of monocytes subsets predict major adverse cardiovascular events and left ventricular function after STEMI. Sci Rep. 2023; 13:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Leuschner F, Panizzi P, Chico-Calero I, Lee WW, Ueno T, Cortez-Retamozo V, Waterman P, Gorbatov R, Marinelli B, Iwamoto Y, Chudnovskiy A, Figueiredo JL, et al. Angiotensin-converting enzyme inhibition prevents the release of monocytes from their splenic reservoir in mice with myocardial infarction. Circ Res. 2010; 107:1364–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Leuschner F, Rauch PJ, Ueno T, Gorbatov R, Marinelli B, Lee WW, Dutta P, Wei Y, Robbins C, Iwamoto Y, Sena B, Chudnovskiy A, et al. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J Exp Med. 2012; 209:123–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Rizzo G, Gropper J, Piollet M, Vafadarnejad E, Rizakou A, Bandi SR, Arampatzi P, Krammer T, DiFabion N, Dietrich O, Arias-Loza AP, Prinz M, et al. Dynamics of monocyte-derived macrophage diversity in experimental myocardial infarction. Cardiovasc Res. 2023; 119:772–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Frangogiannis NG, Mendoza LH, Lindsey ML, Ballantyne CM, Michael LH, Smith CW, Entman ML. IL-10 is induced in the reperfused myocardium and may modulate the reaction to injury. J Immunol. 2000; 165:2798–2808. [DOI] [PubMed] [Google Scholar]
- 308.Heinrichs M, Ashour D, Siegel J, Buchner L, Wedekind G, Heinze KG, Arampatzi P, Saliba AE, Cochain C, Hofmann U, Frantz S, Campos Ramos G. The healing myocardium mobilizes a distinct B-cell subset through a CXCL13-CXCR5-dependent mechanism. Cardiovasc Res. 2021; 117:2664–2676. [DOI] [PubMed] [Google Scholar]
- 309.Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, Sage AP, Guerin C, Vilar J, Caligiuri G, Tsiantoulas D, Laurans L, Dumeau E, Kotti S, et al. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med. 2013; 19:1273–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Boag SE, Das R, Shmeleva EV, Bagnall A, Egred M, Howard N, Bennaceur K, Zaman A, Keavney B, Spyridopoulos I. T lymphocytes and fractalkine contribute to myocardial ischemia/reperfusion injury in patients. J Clin Invest. 2015; 125:3063–3076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. 2014; 11:255–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O'Garra A. IL-10 inhibits cytokine production by activated macrophages. J Immunol. 1991; 147:3815–3822. [PubMed] [Google Scholar]
- 313.Matsukawa A, Kudo S, Maeda T, Numata K, Watanabe H, Takeda K, Akira S, Ito T. Stat3 in resident macrophages as a repressor protein of inflammatory response. J Immunol. 2005; 175:3354–3359. [DOI] [PubMed] [Google Scholar]
- 314.Jiao J, He S, Wang Y, Lu Y, Gu M, Li D, Tang T, Nie S, Zhang M, Lv B, Li J, Xia N, Cheng X. Regulatory B cells improve ventricular remodeling after myocardial infarction by modulating monocyte migration. Basic Res Cardiol. 2021; 116:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Yang Z, Zingarelli B, Szabo C. Crucial role of endogenous interleukin-10 production in myocardial ischemia/reperfusion injury. Circulation. 2000; 101:1019–1026. [DOI] [PubMed] [Google Scholar]
- 316.Zymek P, Nah DY, Bujak M, Ren G, Koerting A, Leucker T, Huebener P, Taffet G, Entman M, Frangogiannis NG. Interleukin-10 is not a critical regulator of infarct healing and left ventricular remodeling. Cardiovasc Res. 2007; 74:313–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Gupta R, Liu L, Zhang X, Fan X, Krishnamurthy P, Verma S, Tongers J, Misener S, Ashcherkin N, Sun H, Tian J, Kishore R. IL-10 provides cardioprotection in diabetic myocardial infarction via upregulation of Heme clearance pathways. JCI Insight. 2020; 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.Jung M, Ma Y, Iyer RP, DeLeon-Pennell KY, Yabluchanskiy A, Garrett MR, 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]
- 319.Liu J, Yang C, Liu T, Deng Z, Fang W, Zhang X, Li J, Huang Q, Liu C, Wang Y, Yang D, Sukhova GK, et al. Eosinophils improve cardiac function after myocardial infarction. Nature communications. 2020; 11:6396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Toor IS, Ruckerl D, Mair I, Ainsworth R, Meloni M, Spiroski AM, Benezech C, Felton JM, Thomson A, Caporali A, Keeble T, Tang KH, et al. Eosinophil Deficiency Promotes Aberrant Repair and Adverse Remodeling Following Acute Myocardial Infarction. JACC Basic Transl Sci. 2020; 5:665–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Sicklinger F, Meyer IS, Li X, Radtke D, Dicks S, Kornadt MP, Mertens C, Meier JK, Lavine KJ, Zhang Y, Kuhn TC, Terzer T, et al. Basophils balance healing after myocardial infarction via IL-4/IL-13. J Clin Invest. 2021; 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.Song J, Frieler RA, Whitesall SE, Chung Y, Vigil TM, Muir LA, Ma J, Brombacher F, Goonewardena SN, Lumeng CN, Goldstein DR, Mortensen RM. Myeloid interleukin-4 receptor alpha is essential in postmyocardial infarction healing by regulating inflammation and fibrotic remodeling. Am J Physiol Heart Circ Physiol. 2021; 320:H323–H337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Shiraishi M, Shintani Y, Shintani Y, Ishida H, Saba R, Yamaguchi A, Adachi H, Yashiro K, Suzuki K. Alternatively activated macrophages determine repair of the infarcted adult murine heart. J Clin Invest. 2016; 126:2151–2166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Alvarez-Argote S, Paddock SJ, Flinn MA, Moreno CW, Knas MC, Almeida VA, Buday SL, Bakhshian Nik A, Patterson M, Chen YG, Lin CW, O'Meara CC. IL-13 promotes functional recovery after myocardial infarction via direct signaling to macrophages. JCI Insight. 2024; 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Zhang F, Wang H, Wang X, Jiang G, Liu H, Zhang G, Wang H, Fang R, Bu X, Cai S, Du J. TGF-beta induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Werner F, Jain MK, Feinberg MW, Sibinga NE, Pellacani A, Wiesel P, Chin MT, Topper JN, Perrella MA, Lee ME. Transforming growth factor-beta 1 inhibition of macrophage activation is mediated via Smad3. J Biol Chem. 2000; 275:36653–36658. [DOI] [PubMed] [Google Scholar]
- 327.Feinberg MW, Shimizu K, Lebedeva M, Haspel R, Takayama K, Chen Z, Frederick JP, Wang XF, Simon DI, Libby P, Mitchell RN, Jain MK. Essential role for Smad3 in regulating MCP-1 expression and vascular inflammation. Circ Res. 2004; 94:601–608. [DOI] [PubMed] [Google Scholar]
- 328.Kitamura M, Suto T, Yokoo T, Shimizu F, Fine LG. Transforming growth factor-beta 1 is the predominant paracrine inhibitor of macrophage cytokine synthesis produced by glomerular mesangial cells. J Immunol. 1996; 156:2964–2971. [PubMed] [Google Scholar]
- 329.Xiao YQ, Freire-de-Lima CG, Janssen WJ, Morimoto K, Lyu D, Bratton DL, Henson PM. Oxidants selectively reverse TGF-beta suppression of proinflammatory mediator production. J Immunol. 2006; 176:1209–1217. [DOI] [PubMed] [Google Scholar]
- 330.Naiki Y, Michelsen KS, Zhang W, Chen S, Doherty TM, Arditi M. Transforming growth factor-beta differentially inhibits MyD88-dependent, but not TRAM- and TRIF-dependent, lipopolysaccharide-induced TLR4 signaling. J Biol Chem. 2005; 280:5491–5495. [DOI] [PubMed] [Google Scholar]
- 331.Batlle E, Massague J. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity. 2019; 50:924–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332.Gorelik L, Flavell RA. Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity. 2000; 12:171–181. [DOI] [PubMed] [Google Scholar]
- 333.Strainic MG, Shevach EM, An F, Lin F, Medof ME. Absence of signaling into CD4(+) cells via C3aR and C5aR enables autoinductive TGF-beta1 signaling and induction of Foxp3(+) regulatory T cells. Nat Immunol. 2013; 14:162–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334.Ikeuchi M, Tsutsui H, Shiomi T, Matsusaka H, Matsushima S, Wen J, Kubota T, Takeshita A. Inhibition of TGF-beta signaling exacerbates early cardiac dysfunction but prevents late remodeling after infarction. Cardiovasc Res. 2004; 64:526–535. [DOI] [PubMed] [Google Scholar]
- 335.Chen B, Huang S, Su Y, Wu YJ, Hanna A, Brickshawana A, Graff J, Frangogiannis NG. Macrophage Smad3 Protects the Infarcted Heart, Stimulating Phagocytosis and Regulating Inflammation. Circ Res. 2019; 125:55–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.Chen B, Li R, Hernandez SC, Hanna A, Su K, Shinde AV, Frangogiannis NG. Differential effects of Smad2 and Smad3 in regulation of macrophage phenotype and function in the infarcted myocardium. J Mol Cell Cardiol. 2022; 171:1–15. [DOI] [PubMed] [Google Scholar]
- 337.Kempf T, Zarbock A, Widera C, Butz S, Stadtmann A, Rossaint J, Bolomini-Vittori M, Korf-Klingebiel M, Napp LC, Hansen B, Kanwischer A, Bavendiek U, et al. GDF-15 is an inhibitor of leukocyte integrin activation required for survival after myocardial infarction in mice. Nat Med. 2011; 17:581–588. [DOI] [PubMed] [Google Scholar]
- 338.Kempf T, Bjorklund E, Olofsson S, Lindahl B, Allhoff T, Peter T, Tongers J, Wollert KC, Wallentin L. Growth-differentiation factor-15 improves risk stratification in ST-segment elevation myocardial infarction. Eur Heart J. 2007; 28:2858–2865. [DOI] [PubMed] [Google Scholar]
- 339.Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014; 510:92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340.Fredman G, Serhan CN. Specialized pro-resolving mediators in vascular inflammation and atherosclerotic cardiovascular disease. Nat Rev Cardiol. 2024. [DOI] [PubMed] [Google Scholar]
- 341.Keyes KT, Ye Y, Lin Y, Zhang C, Perez-Polo JR, Gjorstrup P, Birnbaum Y. Resolvin E1 protects the rat heart against reperfusion injury. Am J Physiol Heart Circ Physiol. 2010; 299:H153–164. [DOI] [PubMed] [Google Scholar]
- 342.Kain V, Ingle KA, Colas RA, Dalli J, Prabhu SD, Serhan CN, Joshi M, Halade GV. Resolvin D1 activates the inflammation resolving response at splenic and ventricular site following myocardial infarction leading to improved ventricular function. Journal of molecular and cellular cardiology. 2015; 84:24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343.Kain V, Liu F, Kozlovskaya V, Ingle KA, Bolisetty S, Agarwal A, Khedkar S, Prabhu SD, Kharlampieva E, Halade GV. Resolution Agonist 15-epi-Lipoxin A(4) Programs Early Activation of Resolving Phase in Post-Myocardial Infarction Healing. Sci Rep. 2017; 7:9999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344.Gerlach BD, Ampomah PB, Yurdagul A Jr., Liu C, Lauring MC, Wang X, Kasikara C, Kong N, Shi J, Tao W, Tabas I. Efferocytosis induces macrophage proliferation to help resolve tissue injury. Cell Metab. 2021; 33:2445–2463 e2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345.Cai B, Thorp EB, Doran AC, Subramanian M, Sansbury BE, Lin CS, Spite M, Fredman G, Tabas I. MerTK cleavage limits proresolving mediator biosynthesis and exacerbates tissue inflammation. Proc Natl Acad Sci U S A. 2016; 113:6526–6531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346.DeBerge M, Yeap XY, Dehn S, Zhang S, Grigoryeva L, Misener S, Procissi D, Zhou X, Lee DC, Muller WA, Luo X, Rothlin C, Tabas I, Thorp EB. MerTK Cleavage on Resident Cardiac Macrophages Compromises Repair After Myocardial Ischemia Reperfusion Injury. Circ Res. 2017; 121:930–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347.Rizzo G, Gropper J, Piollet M, Vafadarnejad E, Rizakou A, Bandi SR, Arampatzi P, Krammer T, DiFabion N, Dietrich O, Arias-Loza AP, Prinz M, et al. Dynamics of monocyte-derived macrophage diversity in experimental myocardial infarction. Cardiovasc Res. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348.Shirakawa K, Endo J, Kataoka M, Katsumata Y, Yoshida N, Yamamoto T, Isobe S, Moriyama H, Goto S, Kitakata H, Hiraide T, Fukuda K, Sano M. IL (Interleukin)-10-STAT3-Galectin-3 Axis Is Essential for Osteopontin-Producing Reparative Macrophage Polarization After Myocardial Infarction. Circulation. 2018; 138:2021–2035. [DOI] [PubMed] [Google Scholar]
- 349.Hoeft K, Schaefer GJL, Kim H, Schumacher D, Bleckwehl T, Long Q, Klinkhammer BM, Peisker F, Koch L, Nagai J, Halder M, Ziegler S, et al. Platelet-instructed SPP1(+) macrophages drive myofibroblast activation in fibrosis in a CXCL4-dependent manner. Cell Rep. 2023; 42:112131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 350.He L, Huang X, Kanisicak O, Li Y, Wang Y, Li Y, Pu W, Liu Q, Zhang H, Tian X, Zhao H, Liu X, et al. Preexisting endothelial cells mediate cardiac neovascularization after injury. J Clin Invest. 2017; 127:2968–2981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351.Wu X, Reboll MR, Korf-Klingebiel M, Wollert KC. Angiogenesis after acute myocardial infarction. Cardiovasc Res. 2021; 117:1257–1273. [DOI] [PubMed] [Google Scholar]
- 352.Howangyin KY, Zlatanova I, Pinto C, Ngkelo A, Cochain C, Rouanet M, Vilar J, Lemitre M, Stockmann C, Fleischmann BK, Mallat Z, Silvestre JS. Myeloid-Epithelial-Reproductive Receptor Tyrosine Kinase and Milk Fat Globule Epidermal Growth Factor 8 Coordinately Improve Remodeling After Myocardial Infarction via Local Delivery of Vascular Endothelial Growth Factor. Circulation. 2016; 133:826–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353.Glinton KE, Ma W, Lantz C, Grigoryeva LS, DeBerge M, Liu X, Febbraio M, Kahn M, Oliver G, Thorp EB. Macrophage-produced VEGFC is induced by efferocytosis to ameliorate cardiac injury and inflammation. J Clin Invest. 2022; 132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354.Li R, Chen B, Kubota A, Hanna A, Humeres C, Hernandez SC, Liu Y, Ma R, Tuleta I, Huang S, Venugopal H, Zhu F, et al. Protective effects of macrophage-specific integrin alpha5 in myocardial infarction are associated with accentuated angiogenesis. Nature communications. 2023; 14:7555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355.Vieira JM, Norman S, Villa Del Campo C, Cahill TJ, Barnette DN, Gunadasa-Rohling M, Johnson LA, Greaves DR, Carr CA, Jackson DG, Riley PR. The cardiac lymphatic system stimulates resolution of inflammation following myocardial infarction. J Clin Invest. 2018; 128:3402–3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356.DeBerge M, Yu S, Dehn S, Ifergan I, Yeap XY, Filipp M, Becker A, Luo X, Miller S, Thorp EB. Monocytes prime autoreactive T cells after myocardial infarction. Am J Physiol Heart Circ Physiol. 2020; 318:H116–H123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 357.Van der Borght K, Scott CL, Nindl V, Bouche A, Martens L, Sichien D, Van Moorleghem J, Vanheerswynghels M, De Prijck S, Saeys Y, Ludewig B, Gillebert T, et al. Myocardial Infarction Primes Autoreactive T Cells through Activation of Dendritic Cells. Cell Rep. 2017; 18:3005–3017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358.Lee JS, Jeong SJ, Kim S, Chalifour L, Yun TJ, Miah MA, Li B, Majdoubi A, Sabourin A, Keler T, Guimond JV, Haddad E, et al. Conventional Dendritic Cells Impair Recovery after Myocardial Infarction. J Immunol. 2018; 201:1784–1798. [DOI] [PubMed] [Google Scholar]
- 359.Anzai A, Anzai T, Nagai S, Maekawa Y, Naito K, Kaneko H, Sugano Y, Takahashi T, Abe H, Mochizuki S, Sano M, Yoshikawa T, et al. Regulatory role of dendritic cells in postinfarction healing and left ventricular remodeling. Circulation. 2012; 125:1234–1245. [DOI] [PubMed] [Google Scholar]
- 360.Choo EH, Lee JH, Park EH, Park HE, Jung NC, Kim TH, Koh YS, Kim E, Seung KB, Park C, Hong KS, Kang K, et al. Infarcted Myocardium-Primed Dendritic Cells Improve Remodeling and Cardiac Function After Myocardial Infarction by Modulating the Regulatory T Cell and Macrophage Polarization. Circulation. 2017; 135:1444–1457. [DOI] [PubMed] [Google Scholar]
- 361.Arruda-Olson AM, Reeder GS, Bell MR, Weston SA, Roger VL. Neutrophilia predicts death and heart failure after myocardial infarction: a community-based study. Circ Cardiovasc Qual Outcomes. 2009; 2:656–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362.Golino M, Corna G, De Ponti R, Abbate A, Van Tassell B. One-year outcomes of patients with ST-segment elevation myocardial infarction according to eosinophil blood count. Eur Heart J Acute Cardiovasc Care. 2024. [DOI] [PubMed] [Google Scholar]
- 363.Pizzolo F, Castagna A, Olivieri O, Girelli D, Friso S, Stefanoni F, Udali S, Munerotto V, Baroni M, Cetera V, Luciani GB, Faggian G, Bernardi F, Martinelli N. Basophil Blood Cell Count Is Associated With Enhanced Factor II Plasma Coagulant Activity and Increased Risk of Mortality in Patients With Stable Coronary Artery Disease: Not Only Neutrophils as Prognostic Marker in Ischemic Heart Disease. J Am Heart Assoc. 2021; 10:e018243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364.Xu JY, Xiong YY, Tang RJ, Jiang WY, Ning Y, Gong ZT, Huang PS, Chen GH, Xu J, Wu CX, Hu MJ, Xu J, et al. Interleukin-5-induced eosinophil population improves cardiac function after myocardial infarction. Cardiovasc Res. 2022; 118:2165–2178. [DOI] [PubMed] [Google Scholar]
- 365.Hofmann U, Beyersdorf N, Weirather J, Podolskaya A, Bauersachs J, Ertl G, Kerkau T, Frantz S. Activation of CD4+ T lymphocytes improves wound healing and survival after experimental myocardial infarction in mice. Circulation. 2012; 125:1652–1663. [DOI] [PubMed] [Google Scholar]
- 366.Rieckmann M, Delgobo M, Gaal C, Buchner L, Steinau P, Reshef D, Gil-Cruz C, Horst ENT, Kircher M, Reiter T, Heinze KG, Niessen HW, et al. Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses. J Clin Invest. 2019; 129:4922–4936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367.Delgobo M, Weiss E, Ashour D, Richter L, Popiolkowski L, Arampatzi P, Stangl V, Arias-Loza P, Mariotti-Ferrandiz E, Rainer PP, Saliba AE, Ludewig B, et al. Myocardial Milieu Favors Local Differentiation of Regulatory T Cells. Circ Res. 2023; 132:565–582. [DOI] [PubMed] [Google Scholar]
- 368.Nindl V, Maier R, Ratering D, De Giuli R, Zust R, Thiel V, Scandella E, Di Padova F, Kopf M, Rudin M, Rulicke T, Ludewig B. Cooperation of Th1 and Th17 cells determines transition from autoimmune myocarditis to dilated cardiomyopathy. Eur J Immunol. 2012; 42:2311–2321. [DOI] [PubMed] [Google Scholar]
- 369.Weirather J, Hofmann UD, Beyersdorf N, Ramos GC, Vogel B, Frey A, Ertl G, Kerkau T, Frantz S. Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation. Circ Res. 2014; 115:55–67. [DOI] [PubMed] [Google Scholar]
- 370.Saxena A, Dobaczewski M, Rai V, Haque Z, Chen W, Li N, Frangogiannis NG. Regulatory T cells are recruited in the infarcted mouse myocardium and may modulate fibroblast phenotype and function. Am J Physiol Heart Circ Physiol. 2014; 307:H1233–1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 371.Homma T, Kinugawa S, Takahashi M, Sobirin MA, Saito A, Fukushima A, Suga T, Takada S, Kadoguchi T, Masaki Y, Furihata T, Taniguchi M, et al. Activation of invariant natural killer T cells by alpha-galactosylceramide ameliorates myocardial ischemia/reperfusion injury in mice. J Mol Cell Cardiol. 2013; 62:179–188. [DOI] [PubMed] [Google Scholar]
- 372.Sobirin MA, Kinugawa S, Takahashi M, Fukushima A, Homma T, Ono T, Hirabayashi K, Suga T, Azalia P, Takada S, Taniguchi M, Nakayama T, et al. Activation of natural killer T cells ameliorates postinfarct cardiac remodeling and failure in mice. Circ Res. 2012; 111:1037–1047. [DOI] [PubMed] [Google Scholar]
- 373.Ilatovskaya DV, Pitts C, Clayton J, Domondon M, Troncoso M, Pippin S, DeLeon-Pennell KY. CD8(+) T-cells negatively regulate inflammation post-myocardial infarction. Am J Physiol Heart Circ Physiol. 2019; 317:H581–H596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 374.Yan X, Shichita T, Katsumata Y, Matsuhashi T, Ito H, Ito K, Anzai A, Endo J, Tamura Y, Kimura K, Fujita J, Shinmura K, et al. Deleterious effect of the IL-23/IL-17A axis and gammadeltaT cells on left ventricular remodeling after myocardial infarction. J Am Heart Assoc. 2012; 1:e004408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375.Bansal SS, Ismahil MA, Goel M, Zhou G, Rokosh G, Hamid T, Prabhu SD. Dysfunctional and Proinflammatory Regulatory T-Lymphocytes Are Essential for Adverse Cardiac Remodeling in Ischemic Cardiomyopathy. Circulation. 2019; 139:206–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376.Bansal SS, Ismahil MA, Goel M, Patel B, Hamid T, Rokosh G, Prabhu SD. Activated T Lymphocytes are Essential Drivers of Pathological Remodeling in Ischemic Heart Failure. Circ Heart Fail. 2017; 10:e003688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377.Cleutjens JP, Verluyten MJ, Smiths JF, Daemen MJ. Collagen remodeling after myocardial infarction in the rat heart. Am J Pathol. 1995; 147:325–338. [PMC free article] [PubMed] [Google Scholar]
- 378.Khalil H, Kanisicak O, Vagnozzi RJ, Johansen AK, Maliken BD, Prasad V, Boyer JG, Brody MJ, Schips T, Kilian KK, Correll RN, Kawasaki K, Nagata K, Molkentin JD. Cell-specific ablation of Hsp47 defines the collagen-producing cells in the injured heart. JCI Insight. 2019; 4:e128722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 379.Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J Cell Biol. 1993; 122:103–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 380.Darby I, Skalli O, Gabbiani G. Alpha-smooth muscle actin is transiently expressed by myofibroblasts during experimental wound healing. Lab Invest. 1990; 63:21–29. [PubMed] [Google Scholar]
- 381.Willems IE, Havenith MG, De Mey JG, Daemen MJ. The alpha-smooth muscle actin-positive cells in healing human myocardial scars. Am J Pathol. 1994; 145:868–875. [PMC free article] [PubMed] [Google Scholar]
- 382.Frangogiannis NG, Michael LH, Entman ML. Myofibroblasts in reperfused myocardial infarcts express the embryonic form of smooth muscle myosin heavy chain (SMemb). Cardiovasc Res. 2000; 48:89–100. [DOI] [PubMed] [Google Scholar]
- 383.Li Y, Li C, Liu Q, Wang L, Bao AX, Jung JP, Dodlapati S, Sun J, Gao P, Zhang X, Francis J, Molkentin JD, Fu X. Loss of Acta2 in cardiac fibroblasts does not prevent the myofibroblast differentiation or affect the cardiac repair after myocardial infarction. J Mol Cell Cardiol. 2022; 171:117–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 384.Krstevski C, Cohen CD, Dona MSI, Pinto AR. New perspectives of the cardiac cellular landscape: mapping cellular mediators of cardiac fibrosis using single-cell transcriptomics. Biochem Soc Trans. 2020; 48:2483–2493. [DOI] [PubMed] [Google Scholar]
- 385.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, Palacio M, Gavira JJ, Bastarrika G, et al. Single-Cell RNA Sequencing Analysis Reveals a Crucial Role for CTHRC1 (Collagen Triple Helix Repeat Containing 1) Cardiac Fibroblasts After Myocardial Infarction. Circulation. 2020; 142:1831–1847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 386.McLellan MA, Skelly DA, Dona MSI, Squiers GT, Farrugia GE, Gaynor TL, Cohen CD, Pandey R, Diep H, Vinh A, Rosenthal NA, Pinto AR. High-Resolution Transcriptomic Profiling of the Heart During Chronic Stress Reveals Cellular Drivers of Cardiac Fibrosis and Hypertrophy. Circulation. 2020; 142:1448–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387.Zhang QJ, He Y, Li Y, Shen H, Lin L, Zhu M, Wang Z, Luo X, Hill JA, Cao D, Luo RL, Zou R, et al. Matricellular Protein Cilp1 Promotes Myocardial Fibrosis in Response to Myocardial Infarction. Circ Res. 2021; 129:1021–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 388.Aisagbonhi O, Rai M, Ryzhov S, Atria N, Feoktistov I, Hatzopoulos AK. Experimental myocardial infarction triggers canonical Wnt signaling and endothelial-to-mesenchymal transition. Dis Model Mech. 2011; 4:469–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389.Haider N, Bosca L, Zandbergen HR, Kovacic JC, Narula N, Gonzalez-Ramos S, Fernandez-Velasco M, Agrawal S, Paz-Garcia M, Gupta S, DeLeon-Pennell K, Fuster V, Ibanez B, Narula J. Transition of Macrophages to Fibroblast-Like Cells in Healing Myocardial Infarction. J Am Coll Cardiol. 2019; 74:3124–3135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390.Mollmann H, Nef HM, Kostin S, von Kalle C, Pilz I, Weber M, Schaper J, Hamm CW, Elsasser A. Bone marrow-derived cells contribute to infarct remodelling. Cardiovasc Res. 2006; 71:661–671. [DOI] [PubMed] [Google Scholar]
- 391.Kanisicak O, Khalil H, Ivey MJ, Karch J, Maliken BD, Correll RN, Brody MJ, SC JL, Aronow BJ, Tallquist MD, Molkentin JD. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nature communications. 2016; 7:12260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392.Moore-Morris T, Cattaneo P, Guimaraes-Camboa N, Bogomolovas J, Cedenilla M, Banerjee I, Ricote M, Kisseleva T, Zhang L, Gu Y, Dalton ND, Peterson KL, et al. Infarct Fibroblasts Do Not Derive From Bone Marrow Lineages. Circ Res. 2018; 122:583–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 393.Schorb W, Booz GW, Dostal DE, Conrad KM, Chang KC, Baker KM. Angiotensin II is mitogenic in neonatal rat cardiac fibroblasts. Circ Res. 1993; 72:1245–1254. [DOI] [PubMed] [Google Scholar]
- 394.Sadoshima J, Izumo S. Molecular characterization of angiotensin II--induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts. Critical role of the AT1 receptor subtype. Circ Res. 1993; 73:413–423. [DOI] [PubMed] [Google Scholar]
- 395.Crabos M, Roth M, Hahn AW, Erne P. Characterization of angiotensin II receptors in cultured adult rat cardiac fibroblasts. Coupling to signaling systems and gene expression. J Clin Invest. 1994; 93:2372–2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396.Thibault G, Lacombe MJ, Schnapp LM, Lacasse A, Bouzeghrane F, Lapalme G. Upregulation of alpha(8)beta(1)-integrin in cardiac fibroblast by angiotensin II and transforming growth factor-beta1. Am J Physiol Cell Physiol. 2001; 281:C1457–1467. [DOI] [PubMed] [Google Scholar]
- 397.Ohkubo N, Matsubara H, Nozawa Y, Mori Y, Murasawa S, Kijima K, Maruyama K, Masaki H, Tsutumi Y, Shibazaki Y, Iwasaka T, Inada M. Angiotensin type 2 receptors are reexpressed by cardiac fibroblasts from failing myopathic hamster hearts and inhibit cell growth and fibrillar collagen metabolism. Circulation. 1997; 96:3954–3962. [DOI] [PubMed] [Google Scholar]
- 398.Kurisu S, Ozono R, Oshima T, Kambe M, Ishida T, Sugino H, Matsuura H, Chayama K, Teranishi Y, Iba O, Amano K, Matsubara H. Cardiac angiotensin II type 2 receptor activates the kinin/NO system and inhibits fibrosis. Hypertension. 2003; 41:99–107. [DOI] [PubMed] [Google Scholar]
- 399.Schieffer B, Wirger A, Meybrunn M, Seitz S, Holtz J, Riede UN, Drexler H. Comparative effects of chronic angiotensin-converting enzyme inhibition and angiotensin II type 1 receptor blockade on cardiac remodeling after myocardial infarction in the rat. Circulation. 1994; 89:2273–2282. [DOI] [PubMed] [Google Scholar]
- 400.Frangogiannis NG. Transforming growth factor-beta in myocardial disease. Nat Rev Cardiol. 2022; 19:435–455. [DOI] [PubMed] [Google Scholar]
- 401.Frangogiannis NG, Ren G, Dewald O, Zymek P, Haudek S, Koerting A, Winkelmann K, Michael LH, Lawler J, Entman ML. The critical role of endogenous Thrombospondin (TSP)-1 in preventing expansion of healing myocardial infarcts. Circulation. 2005; 111:2935–2942. [DOI] [PubMed] [Google Scholar]
- 402.Yu Q, Stamenkovic I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Genes Dev. 2000; 14:163–176. [PMC free article] [PubMed] [Google Scholar]
- 403.Sarrazy V, Koehler A, Chow ML, Zimina E, Li CX, Kato H, Caldarone CA, Hinz B. Integrins alphavbeta5 and alphavbeta3 promote latent TGF-beta1 activation by human cardiac fibroblast contraction. Cardiovasc Res. 2014; 102:407–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404.Chen YW, Cheng PP, Yin YF, Cai H, Chen JZ, Feng MH, Guo W, Zhao P, Zhang C, Shan XL, Chen HH, Guo S, Lu Y, Xu M. Integrin alphaV mediated activation of myofibroblast via mechanoparacrine of transforming growth factor beta1 in promoting fibrous scar formation after myocardial infarction. Biochem Biophys Res Commun. 2024; 692:149360. [DOI] [PubMed] [Google Scholar]
- 405.Frangogiannis NG. Transforming Growth Factor (TGF)-beta in tissue fibrosis. J Exp Med. 2020; 217:e20190103. https://doi.org/201901 10.20191084/jem.20190103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406.Dobaczewski M, Bujak M, Li N, Gonzalez-Quesada C, Mendoza LH, Wang XF, Frangogiannis NG. Smad3 signaling critically regulates fibroblast phenotype and function in healing myocardial infarction. Circ Res. 2010; 107:418–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 407.Kong P, Shinde AV, Su Y, Russo I, Chen B, Saxena A, Conway SJ, Graff JM, Frangogiannis NG. Opposing Actions of Fibroblast and Cardiomyocyte Smad3 Signaling in the Infarcted Myocardium. Circulation. 2018; 137:707–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 408.Huang S, Chen B, Su Y, Alex L, Humeres C, Shinde AV, Conway SJ, Frangogiannis NG. Distinct roles of myofibroblast-specific Smad2 and Smad3 signaling in repair and remodeling of the infarcted heart. J Mol Cell Cardiol. 2019; 132:84–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 409.Frangogiannis NG, Perrard JL, Mendoza LH, Burns AR, Lindsey ML, Ballantyne CM, Michael LH, Smith CW, Entman ML. Stem cell factor induction is associated with mast cell accumulation after canine myocardial ischemia and reperfusion. Circulation. 1998; 98:687–698. [DOI] [PubMed] [Google Scholar]
- 410.Ruoss SJ, Hartmann T, Caughey GH. Mast cell tryptase is a mitogen for cultured fibroblasts. J Clin Invest. 1991; 88:493–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 411.Wei CC, Hase N, Inoue Y, Bradley EW, Yahiro E, Li M, Naqvi N, Powell PC, Shi K, Takahashi Y, Saku K, Urata H, Dell'italia LJ, Husain A. Mast cell chymase limits the cardiac efficacy of Ang I-converting enzyme inhibitor therapy in rodents. J Clin Invest. 2010; 120:1229–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 412.House SL, Branch K, Newman G, Doetschman T, Schultz JlJ. Cardioprotection induced by cardiac-specific overexpression of fibroblast growth factor-2 is mediated by the MAPK cascade. Am J Physiol Heart Circ Physiol. 2005; 289:H2167–2175. [DOI] [PubMed] [Google Scholar]
- 413.Virag JA, Rolle ML, Reece J, Hardouin S, Feigl EO, Murry CE. Fibroblast growth factor-2 regulates myocardial infarct repair: effects on cell proliferation, scar contraction, and ventricular function. Am J Pathol. 2007; 171:1431–1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 414.Zhao T, Zhao W, Chen Y, Ahokas RA, Sun Y. Acidic and basic fibroblast growth factors involved in cardiac angiogenesis following infarction. Int J Cardiol. 2011; 152:307–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415.Matsiukevich D, House SL, Weinheimer C, Kovacs A, Ornitz DM. Fibroblast growth factor receptor signaling in cardiomyocytes is protective in the acute phase following ischemia-reperfusion injury. Front Cardiovasc Med. 2022; 9:1011167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 416.Kardami E, Detillieux K, Ma X, Jiang Z, Santiago JJ, Jimenez SK, Cattini PA. Fibroblast growth factor-2 and cardioprotection. Heart Fail Rev. 2007; 12:267–277. [DOI] [PubMed] [Google Scholar]
- 417.Zymek P, Bujak M, Chatila K, Cieslak A, Thakker G, Entman ML, Frangogiannis NG. The role of platelet-derived growth factor signaling in healing myocardial infarcts. J Am Coll Cardiol. 2006; 48:2315–2323. [DOI] [PubMed] [Google Scholar]
- 418.Simm A, Nestler M, Hoppe V. Mitogenic effect of PDGF-AA on cardiac fibroblasts. Basic Res Cardiol. 1998; 93 Suppl 3:40–43. [DOI] [PubMed] [Google Scholar]
- 419.Saraswati S, Lietman CD, Li B, Mathew S, Zent R, Young PP. Small proline-rich repeat 3 is a novel coordinator of PDGFRbeta and integrin beta1 crosstalk to augment proliferation and matrix synthesis by cardiac fibroblasts. FASEB J. 2020; 34:7885–7904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420.Ivey MJ, Kuwabara JT, Riggsbee KL, Tallquist MD. Platelet-derived growth factor receptor-alpha is essential for cardiac fibroblast survival. Am J Physiol Heart Circ Physiol. 2019; 317:H330–H344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 421.Hamid T, Xu Y, Ismahil MA, Rokosh G, Jinno M, Zhou G, Wang Q, Prabhu SD. Cardiac Mesenchymal Stem Cells Promote Fibrosis and Remodeling in Heart Failure: Role of PDGF Signaling. JACC Basic Transl Sci. 2022; 7:465–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 422.Thavapalachandran S, Grieve SM, Hume RD, Le TYL, Raguram K, Hudson JE, Pouliopoulos J, Figtree GA, Dye RP, Barry AM, Brown P, Lu J, et al. Platelet-derived growth factor-AB improves scar mechanics and vascularity after myocardial infarction. Sci Transl Med. 2020; 12. [DOI] [PubMed] [Google Scholar]
- 423.Frangogiannis NG. The extracellular matrix in myocardial injury, repair, and remodeling. J Clin Invest. 2017; 127:1600–1612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 424.Dugina V, Fontao L, Chaponnier C, Vasiliev J, Gabbiani G. Focal adhesion features during myofibroblastic differentiation are controlled by intracellular and extracellular factors. J Cell Sci. 2001; 114:3285–3296. [DOI] [PubMed] [Google Scholar]
- 425.Kohan M, Muro AF, White ES, Berkman N. EDA-containing cellular fibronectin induces fibroblast differentiation through binding to alpha4beta7 integrin receptor and MAPK/Erk 1/2-dependent signaling. FASEB J. 2010; 24:4503–4512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 426.Lenga Y, Koh A, Perera AS, McCulloch CA, Sodek J, Zohar R. Osteopontin expression is required for myofibroblast differentiation. Circ Res. 2008; 102:319–327. [DOI] [PubMed] [Google Scholar]
- 427.Tamaoki M, Imanaka-Yoshida K, Yokoyama K, Nishioka T, Inada H, Hiroe M, Sakakura T, Yoshida T. Tenascin-C regulates recruitment of myofibroblasts during tissue repair after myocardial injury. Am J Pathol. 2005; 167:71–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 428.Schellings MW, Vanhoutte D, Swinnen M, Cleutjens JP, Debets J, van Leeuwen RE, d'Hooge J, Van de Werf F, Carmeliet P, Pinto YM, Sage EH, Heymans S. Absence of SPARC results in increased cardiac rupture and dysfunction after acute myocardial infarction. J Exp Med. 2009; 206:113–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 429.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, Molkentin JD. Genetic manipulation of periostin expression reveals a role in cardiac hypertrophy and ventricular remodeling. Circ Res. 2007; 101:313–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 430.Van Aelst LN, Voss S, Carai P, Van Leeuwen R, Vanhoutte D, Sanders-van Wijk S, Eurlings L, Swinnen M, Verheyen FK, Verbeken E, Nef H, Troidl C, et al. Osteoglycin prevents cardiac dilatation and dysfunction after myocardial infarction through infarct collagen strengthening. Circ Res. 2015; 116:425–436. [DOI] [PubMed] [Google Scholar]
- 431.Wang Y, Li Q, Tao B, Angelini M, Ramadoss S, Sun B, Wang P, Krokhaleva Y, Ma F, Gu Y, Espinoza A, Yamauchi K, et al. Fibroblasts in heart scar tissue directly regulate cardiac excitability and arrhythmogenesis. Science. 2023; 381:1480–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 432.Nguyen TP, Xie Y, Garfinkel A, Qu Z, Weiss JN. Arrhythmogenic consequences of myofibroblast-myocyte coupling. Cardiovasc Res. 2012; 93:242–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 433.Gaudesius G, Miragoli M, Thomas SP, Rohr S. Coupling of cardiac electrical activity over extended distances by fibroblasts of cardiac origin. Circ Res. 2003; 93:421–428. [DOI] [PubMed] [Google Scholar]
- 434.Quinn TA, Camelliti P, Rog-Zielinska EA, Siedlecka U, Poggioli T, O'Toole ET, Knopfel T, 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]
- 435.Rubart M, Tao W, Lu XL, Conway SJ, Reuter SP, Lin SF, Soonpaa MH. Electrical coupling between ventricular myocytes and myofibroblasts in the infarcted mouse heart. Cardiovasc Res. 2018; 114:389–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 436.Hayakawa K, Takemura G, Kanoh M, Li Y, Koda M, Kawase Y, Maruyama R, Okada H, Minatoguchi S, Fujiwara T, Fujiwara H. Inhibition of granulation tissue cell apoptosis during the subacute stage of myocardial infarction improves cardiac remodeling and dysfunction at the chronic stage. Circulation. 2003; 108:104–109. [DOI] [PubMed] [Google Scholar]
- 437.Fu X, Khalil H, Kanisicak O, Boyer JG, Vagnozzi RJ, Maliken BD, Sargent MA, Prasad V, Valiente-Alandi I, Blaxall BC, 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]
- 438.Miyazawa K, Miyazono K. Regulation of TGF-beta Family Signaling by Inhibitory Smads. Cold Spring Harb Perspect Biol. 2017; 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 439.Humeres C, Shinde AV, Hanna A, Alex L, Hernandez SC, Li R, Chen B, Conway SJ, Frangogiannis NG. Smad7 effects on TGF-beta and ErbB2 restrain myofibroblast activation and protect from postinfarction heart failure. J Clin Invest. 2022; 132:e146926. doi: 1469 10.141172/JCI146926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 440.Cunnington RH, Wang B, Ghavami S, Bathe KL, Rattan SG, Dixon IM. Antifibrotic properties of c-Ski and its regulation of cardiac myofibroblast phenotype and contractility. Am J Physiol Cell Physiol. 2011; 300:C176–186. [DOI] [PubMed] [Google Scholar]
- 441.Landry NM, Rattan SG, Filomeno KL, Meier TW, Meier SC, Foran SJ, Meier CF, Koleini N, Fandrich RR, Kardami E, Duhamel TA, Dixon IMC. SKI activates the Hippo pathway via LIMD1 to inhibit cardiac fibroblast activation. Basic Res Cardiol. 2021; 116:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 442.Dufeys C, Daskalopoulos EP, Castanares-Zapatero D, Conway SJ, Ginion A, Bouzin C, Ambroise J, Bearzatto B, Gala JL, Heymans S, Papageorgiou AP, Vinckier S, et al. AMPKalpha1 deletion in myofibroblasts exacerbates post-myocardial infarction fibrosis by a connexin 43 mechanism. Basic Res Cardiol. 2021; 116:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 443.Janbandhu V, Tallapragada V, Patrick R, Li Y, Abeygunawardena D, Humphreys DT, Martin E, Ward AO, Contreras O, Farbehi N, Yao E, Du J, et al. Hif-1a suppresses ROS-induced proliferation of cardiac fibroblasts following myocardial infarction. Cell Stem Cell. 2022; 29:281–297 e212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 444.Yokota T, McCourt J, Ma F, Ren S, Li S, Kim TH, Kurmangaliyev YZ, Nasiri R, Ahadian S, Nguyen T, Tan XHM, Zhou Y, et al. Type V Collagen in Scar Tissue Regulates the Size of Scar after Heart Injury. Cell. 2020; 182:545–562 e523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 445.Ren G, Michael LH, Entman ML, Frangogiannis NG. Morphological characteristics of the microvasculature in healing myocardial infarcts. J Histochem Cytochem. 2002; 50:71–79. [DOI] [PubMed] [Google Scholar]
- 446.Baran KW, Nguyen M, McKendall GR, Lambrew CT, Dykstra G, Palmeri ST, Gibbons RJ, Borzak S, Sobel BE, Gourlay SG, Rundle AC, Gibson CM, Barron HV. Double-blind, randomized trial of an anti-CD18 antibody in conjunction with recombinant tissue plasminogen activator for acute myocardial infarction: limitation of myocardial infarction following thrombolysis in acute myocardial infarction (LIMIT AMI) study. Circulation. 2001; 104:2778–2783. [DOI] [PubMed] [Google Scholar]
- 447.Faxon DP, Gibbons RJ, Chronos NA, Gurbel PA, Sheehan F. The effect of blockade of the CD11/CD18 integrin receptor on infarct size in patients with acute myocardial infarction treated with direct angioplasty: the results of the HALT-MI study. J Am Coll Cardiol. 2002; 40:1199–1204. [DOI] [PubMed] [Google Scholar]
- 448.Rusnak JM, Kopecky SL, Clements IP, Gibbons RJ, Holland AE, Peterman HS, Martin JS, Saoud JB, Feldman RL, Breisblatt WM, Simons M, Gessler CJ Jr., Yu AS. An anti-CD11/CD18 monoclonal antibody in patients with acute myocardial infarction having percutaneous transluminal coronary angioplasty (the FESTIVAL study). Am J Cardiol. 2001; 88:482–487. [DOI] [PubMed] [Google Scholar]
- 449.Armstrong PW, Granger CB, Adams PX, Hamm C, Holmes D Jr., O'Neill WW, Todaro TG, Vahanian A, Van de Werf F. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA. 2007; 297:43–51. [DOI] [PubMed] [Google Scholar]
- 450.Tardif JC, Tanguay JF, Wright SS, Duchatelle V, Petroni T, Gregoire JC, Ibrahim R, Heinonen TM, Robb S, Bertrand OF, Cournoyer D, Johnson D, et al. Effects of the P-selectin antagonist inclacumab on myocardial damage after percutaneous coronary intervention for non-ST-segment elevation myocardial infarction: results of the SELECT-ACS trial. J Am Coll Cardiol. 2013; 61:2048–2055. [DOI] [PubMed] [Google Scholar]
- 451.Seropian IM, Toldo S, Van Tassell BW, Abbate A. Anti-inflammatory strategies for ventricular remodeling following ST-segment elevation acute myocardial infarction. J Am Coll Cardiol. 2014; 63:1593–1603. [DOI] [PubMed] [Google Scholar]
- 452.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017; 377:1119–1131. [DOI] [PubMed] [Google Scholar]
- 453.Everett BM, Cornel JH, Lainscak M, Anker SD, Abbate A, Thuren T, Libby P, Glynn RJ, Ridker PM. Anti-Inflammatory Therapy With Canakinumab for the Prevention of Hospitalization for Heart Failure. Circulation. 2019; 139:1289–1299. [DOI] [PubMed] [Google Scholar]
- 454.Abbate A, Van Tassell BW, Biondi-Zoccai G, Kontos MC, Grizzard JD, Spillman DW, Oddi C, Roberts CS, Melchior RD, Mueller GH, Abouzaki NA, Rengel LR, et al. Effects of interleukin-1 blockade with anakinra on adverse cardiac remodeling and heart failure after acute myocardial infarction [from the Virginia Commonwealth University-Anakinra Remodeling Trial (2) (VCU-ART2) pilot study]. Am J Cardiol. 2013; 111:1394–1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 455.Abbate A, Kontos MC, Grizzard JD, Biondi-Zoccai GG, Van Tassell BW, Robati R, Roach LM, Arena RA, Roberts CS, Varma A, Gelwix CC, Salloum FN, et al. Interleukin-1 blockade with anakinra to prevent adverse cardiac remodeling after acute myocardial infarction (Virginia Commonwealth University Anakinra Remodeling Trial [VCU-ART] Pilot study). Am J Cardiol. 2010; 105:1371–1377 e1371. [DOI] [PubMed] [Google Scholar]
- 456.Abbate A, Wohlford GF, Del Buono MG, Chiabrando JG, Markley R, Turlington J, Kadariya D, Trankle CR, Biondi-Zoccai G, Lipinski MJ, Van Tassell BW. Interleukin-1 blockade with anakinra and heart failure following ST-segment elevation myocardial infarction: results from a pooled analysis of the VCUART clinical trials. Eur Heart J Cardiovasc Pharmacother. 2022; 8:503–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 457.Broch K, Anstensrud AK, Woxholt S, Sharma K, Tollefsen IM, Bendz B, Aakhus S, Ueland T, Amundsen BH, Damas JK, Berg ES, Bjorkelund E, et al. Randomized Trial of Interleukin-6 Receptor Inhibition in Patients With Acute ST-Segment Elevation Myocardial Infarction. J Am Coll Cardiol. 2021; 77:1845–1855. [DOI] [PubMed] [Google Scholar]
- 458.Tardif JC, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, Pinto FJ, Ibrahim R, Gamra H, Kiwan GS, Berry C, Lopez-Sendon J, et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N Engl J Med. 2019; 381:2497–2505. [DOI] [PubMed] [Google Scholar]
- 459.Zhao TX, Aetesam-Ur-Rahman M, Sage AP, Victor S, Kurian R, Fielding S, Ait-Oufella H, Chiu YD, Binder CJ, McKie M, Hoole SP, Mallat Z. Rituximab in patients with acute ST-elevation myocardial infarction: an experimental medicine safety study. Cardiovasc Res. 2022; 118:872–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 460.Aghajanian H, Kimura T, Rurik JG, Hancock AS, Leibowitz MS, Li L, Scholler J, Monslow J, Lo A, Han W, Wang T, Bedi K, et al. Targeting cardiac fibrosis with engineered T cells. Nature. 2019; 573:430–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 461.Rurik JG, Tombacz I, Yadegari A, Mendez Fernandez PO, Shewale SV, Li L, Kimura T, Soliman OY, Papp TE, Tam YK, Mui BL, Albelda SM, et al. CAR T cells produced in vivo to treat cardiac injury. Science. 2022; 375:91–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 462.Tang TT, Yuan J, Zhu ZF, Zhang WC, Xiao H, Xia N, Yan XX, Nie SF, Liu J, Zhou SF, Li JJ, Yao R, et al. Regulatory T cells ameliorate cardiac remodeling after myocardial infarction. Basic Res Cardiol. 2012; 107:232. [DOI] [PubMed] [Google Scholar]
- 463.Gladow N, Hollmann C, Ramos G, Frantz S, Kerkau T, Beyersdorf N, Hofmann U. Treatment of mice with a ligand binding blocking anti-CD28 monoclonal antibody improves healing after myocardial infarction. PLoS One. 2020; 15:e0227734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 464.Frangogiannis NG. Why animal model studies are lost in translation. J Cardiovasc Aging. 2022; 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 465.Stone PH, Muller JE, Hartwell T, York BJ, Rutherford JD, Parker CB, Turi ZG, Strauss HW, Willerson JT, Robertson T, et al. The effect of diabetes mellitus on prognosis and serial left ventricular function after acute myocardial infarction: contribution of both coronary disease and diastolic left ventricular dysfunction to the adverse prognosis. The MILIS Study Group. J Am Coll Cardiol. 1989; 14:49–57. [DOI] [PubMed] [Google Scholar]
- 466.Bujak M, Kweon HJ, Chatila K, Li N, Taffet G, Frangogiannis NG. Aging-related defects are associated with adverse cardiac remodeling in a mouse model of reperfused myocardial infarction. J Am Coll Cardiol. 2008; 51:1384–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 467.Seropian IM, Sonnino C, Van Tassell BW, Biasucci LM, Abbate A. Inflammatory markers in ST-elevation acute myocardial infarction. Eur Heart J Acute Cardiovasc Care. 2016; 5:382–395. [DOI] [PubMed] [Google Scholar]
- 468.Westman PC, Lipinski MJ, Luger D, Waksman R, Bonow RO, Wu E, Epstein SE. Inflammation as a Driver of Adverse Left Ventricular Remodeling After Acute Myocardial Infarction. J Am Coll Cardiol. 2016; 67:2050–2060. [DOI] [PubMed] [Google Scholar]
- 469.Thackeray JT, Derlin T, Haghikia A, Napp LC, Wang Y, Ross TL, Schafer A, Tillmanns J, Wester HJ, Wollert KC, Bauersachs J, Bengel FM. Molecular Imaging of the Chemokine Receptor CXCR4 After Acute Myocardial Infarction. JACC Cardiovasc Imaging. 2015; 8:1417–1426. [DOI] [PubMed] [Google Scholar]
- 470.Wollenweber T, Roentgen P, Schafer A, Schatka I, Zwadlo C, Brunkhorst T, Berding G, Bauersachs J, Bengel FM. Characterizing the inflammatory tissue response to acute myocardial infarction by clinical multimodality noninvasive imaging. Circ Cardiovasc Imaging. 2014; 7:811–818. [DOI] [PubMed] [Google Scholar]
- 471.Nahrendorf M, Frantz S, Swirski FK, Mulder WJ, Randolph G, Ertl G, Ntziachristos V, Piek JJ, Stroes ES, Schwaiger M, Mann DL, Fayad ZA. Imaging systemic inflammatory networks in ischemic heart disease. J Am Coll Cardiol. 2015; 65:1583–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]