Abstract
Angiotensin II (Ang II) is a primary mediator of profibrotic signaling in the heart and more specifically, the cardiac fibroblast. Ang II-mediated cardiomyocyte hypertrophy in combination with cardiac fibroblast proliferation, activation, and extracellular matrix production compromise cardiac function and increase mortality in humans. Profibrotic actions of Ang II are mediated by increasing production of fibrogenic mediators (e.g. transforming growth factor beta, scleraxis, osteopontin, and periostin), recruitment of immune cells, and via increased reactive oxygen species generation. As a result, drugs that inhibit Ang II production or action, collectively referred to as renin angiotensin system (RAS) inhibitors, are first line therapeutics for heart failure. Moreover, transient RAS inhibition has been found to persistently alter hypertensive cardiac fibroblast responses to injury providing a useful tool to identify novel therapeutic targets. This review summarizes the profibrotic actions of Ang II and the known impact of RAS inhibition on cardiac fibroblast phenotype and cardiac remodeling.
Keywords: fibroblast, angiotensin II, fibrosis, angiotensin converting enzyme inhibitor, angiotensin receptor antagonist
1. Introduction
Cardiac pathology as a product of acute or chronic injury typically manifests in cardiac fibrosis mediated by cardiac fibroblasts. The classical renin angiotensin system (RAS) positively regulates the progression of fibrosis by signaling via angiotensin receptors in the heart. Angiotensin II (Ang II) is a vasoactive peptide with systemic and local effects on the heart via angiotensin type 1 (AT1) and type 2 (AT2) receptors. Angiotensinogen is cleaved by the enzyme renin, to form angiotensin I, which in the presence of angiotensin converting enzyme (ACE) or chymase, is further cleaved to produce angiotensin II. ACE inhibitors and AT1 receptor antagonists are known to both reduce blood pressure and minimize cardiac fibrosis following injury. Cardiac tissues have been shown to uptake extracellular angiotensinogen and renin1,2, and increase expression of both following injury3,4. The presence of ACE has been well-established in the normal5 and remodeling heart6,7 allowing for the generation of Ang II. Thus, Ang II elicits cardiac effects by signaling from the circulation8 as well as by local production of Ang II9. In the heart, Ang II induces cardiomyocyte hypertrophy and cardiac fibroblast activation which together potentiate cardiac fibrosis and dysfunction. Angiotensin-(1–7) (Ang (1–7)) is an alternative product of Ang I by the action of neprilysin or of Ang II by ACE2. Ang (1–7) antagonizes the effects of Ang II via actions at the Mas receptor10 and as a systemic vasodilator11.
The RAS plays a primary role in the long-term control of arterial pressure. Dysregulation of the RAS systemically can result in hypertension. Excess local activation of the RAS can result in tissue remodeling, including fibrosis in the heart12 and other tissues13. All the necessary components for the generation of Ang II can be found in the left ventricle (LV)14,15. Moreover, cardiac fibroblasts also express genes for renin, ACE, angiotensinogen, as well as the AT1 and AT2 receptors4,16–21. Angiotensin II also stimulates aldosterone release from the adrenal cortex, which promotes fibrosis in the heart via the mineralocorticoid receptor. As such, aldosterone antagonists have also shown to be effective in attenuating cardiac fibrosis13. The cardioprotective effects of RAS inhibition are therefore linked to both cardiomyocyte and cardiac fibroblast specific signaling. Fibroblast activation is directly reduced while diminished cardiomyocyte hypertrophy removes additional stimuli. ACE inhibitors and angiotensin receptor blockers (ARBs) are standard of care to slow the development of heart failure induced by chronic hypertension and acute MI and have been associated with reduction in cardiac fibrosis and mortality. More recent combination therapies are proving to be more effective than these monotherapies. Thus, a better understanding of downstream targets in the RAS will help elucidate therapeutic targets that may be more proficient in preventing heart failure.
The pathological impact of Ang II is primarily mediated by AT1 receptor agonism, while AT2 receptor activation can attenuate the fibrogenic signaling22. The relative contribution of AT1 vs. AT2 receptors in mediating fibrosis has not been fully elucidated. AT2 receptor knockout mice have been shown to have lower cardiac collagen content and reduced collagen 1a1 gene expression23, and in response to Ang II infusion, AT2 receptor knockout mice exhibited attenuated collagen deposition24. In contrast, AT1aR deletion alone25 or in combination with overexpression of AT2R, resulted in a greater reduction in fibrosis26. Given that each of these studies were performed in global knockout mice, the relative contribution of blood pressure changes or cardiomyocyte-fibroblast crosstalk is not known. The development of the TCF21-cre and POSTN-cre lines for fibroblast or activated fibroblast-specific knockouts would be important for future studies to identify the importance of fibroblast-specific RAS signaling in the development of fibrosis. Crosstalk between fibroblasts and cardiomyocytes includes pro-inflammatory, pro-death, and activating signals. Whether the injury is slow and progressive or sudden, fibroblasts respond to apoptosis and necrosis of cardiomyocytes which in turn triggers fibroblast activation. Both fibroblasts and cardiomyocytes contribute to the inflammatory milieu, communicate with immune cells, and exacerbate fibrotic remodeling. The details of cardiac cell crosstalk during cardiac remodeling has been reviewed in depth elsewhere27,28.
Cardiac fibroblast activation and transdifferentiation into extracellular matrix (ECM)-producing cells, often referred to as myofibroblasts, is the cornerstone to fibrotic remodeling of the heart29. The structural ECM includes collagen, fibronectin, proteoglycans, glycoproteins and integrins. Activated fibroblasts also secrete modulatory matricellular proteins that transduce inflammatory, growth, and fibrogenic signals. The roles of key matricellular proteins periostin, osteopontin, thrombospondins, tenascin-C, and CCN1-CCN6 are reviewed here and elsewhere30. Markers of cardiac fibroblast activation continue to be refined and remain difficult to determine given low fibroblast specificity and high fibroblast heterogeneity. Thus, the activated fibroblast is currently best determined by a collection of markers which include ɑ-smooth muscle actin (ɑ-SMA), periostin, fibronectin, and collagens I and III among others31. Some effector pathways downstream of AT1 receptor are mitogen-activated protein kinases (MAPK) including extracellular signal-regulated 1/2 (ERK1/2), p38 MAPK, and AKT, which modulate proliferation. Additionally, Ang II induces reactive oxygen species (ROS) production and upregulation of ECM components mainly via transforming growth factor beta (TGF-β) signaling. This review focuses on cardiac-specific RAS signaling and direct effects of Ang II and RAS inhibition on cardiac fibroblast responses.
2. Ang II and cardiac fibroblast activation
Ang II infusion in rodent models produces a phenotype of hypertension, cardiac hypertrophy and cardiac fibrosis32–37. In addition, production of local Ang II has been shown to increase in response to myocardial infarction and play a major role in the wound healing response38,39. Thus, chronic elevations of Ang II and acute increase in local Ang II following an ischemic event each result in fibroblast activation and expansion. Studies that investigate the direct impact of Ang II on cardiac fibroblasts reveal increased measures of activation including proliferation and fibrogenic gene and protein expression. Ang II-induced fibroblast activation and associated ECM production is mediated by fibrogenic signaling molecules, inflammation, and oxidative stress (Figure 1) as outlined in this section. Additionally, epigenetic modulation of gene expression is influenced by the RAS and therefore increases the complexity of Ang II actions on cardiac fibroblast activation. Current and future investigations utilizing single cell RNA-sequencing are working to elucidate novel targets for therapy that range outside of general RAS inhibition.
Figure 1. RAS-mediated mechanisms of cardiac fibroblast activation.

Angiotensin II (Ang II) binds angiotensin type 1 receptors (AT1R) on the fibroblast to initiate three key processes that result in fibroblast activation: fibrogenic gene upregulation, immune cell recruitment, and oxidative stress. Crosstalk between components of each pathway supports redundant feed forward mechanisms that together with cell death resistance perpetuate activated fibroblasts with cardiac injury. Transforming growth factor-beta (TGF-β), scleraxis (SCX), signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinase 1/2 (ERK1/2), cellular communication network factor 2 (CCN2), osteopontin (OPN), periostin (POSTN), alpha-smooth muscle actin (α-SMA), toll-like receptor 2 (TLR2), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), NADPH oxidase (NOX4), reactive oxygen species (ROS), B-cell lymphoma 2 (Bcl-2). Created with BioRender.com.
2.1. Pro-Fibrogenic actions of Ang II-mediated signaling molecules
Ang II has been widely shown to increase TGF-β signaling in the cardiac fibroblast via AT1 receptor stimulation. The pro-fibrotic effects of Ang II pay be mediated in large part by TGF-β signaling. Whole animal Ang II infusion and in vitro stimulation of cardiac fibroblasts with Ang II induces increases in TGF-β mRNA and protein40–42. Activated fibroblasts produce TGF-β, which then can act in an autocrine capacity to bind TGF-β receptors on the cell surface43 and signal through ERK1/2 and Smad proteins41,44. TGF-β-mediated ERK1/2 activation plays a large role in cardiac fibroblast proliferation for expansion of ECM-producing cells in the remodeling heart. Binding of TGF-β also leads to phosphorylation of Smad2 and Smad3, which transfer to the nucleus to increase transcription of both pro-inflammatory and ECM proteins via a process that involves several accessory and modulatory proteins.
TGF-β/Smad3 signaling has been linked to several signaling partners that work to maintain the fibrogenic capacity of fibroblasts. Some more recent investigations have highlighted a role of signal transducer and activator of transcription 3 (STAT3) in maintaining Smad signaling that is initiated in a TGF-β independent manner. JAK/STAT signaling occurs downstream of AT1 receptor45 and toll-like receptor 4 (TLR4)46 in the heart. Ang II infusion upregulates STAT3 activity and is linked with cardiac fibrosis via thrombospondin 147. In the heart, loss of STAT3 prevents Ang II-induced upregulation of fibrogenic TGF-β and collagen46 demonstrating the critical role of TGF-β in potentiating fibrotic remodeling. Upstream inhibition of STAT3 activity by global TLR4 knockout46 or direct pharmacological inhibition48 protects mice from fibrosis induced by Ang II. The STAT/Smad3 association has also been shown to be maintained by the transmembrane ligand, EphrinB2. This ligand is elevated co-incident with fibrosis after 4 weeks of Ang II infusion in mice and in vitro treatment with EphrinB2 stimulates cardiac fibroblast activation49. Further, STAT3-mediated protection of mitochondria prolongs fibroblast activation50. Other agents that inhibit the actions of TGF-β also suppress proliferation and fibrotic signals in models of Ang II infusion in vivo51 and in vitro in cardiac fibroblasts52–56.
TGF-β mediated Smad signaling was shown to be modulated by microRNAs (miRNA). Ang II-mediated pressure overload in mice was shown to be linked to repression of miRNA-221/222 resulting in unrestrained TGF-β signaling57. Protective actions of Klotho include suppression of pro-fibrotic miRNA-132 downstream of Ang II-induced Smad activation58. Four weeks of Ang II suppresses miRNA-1297-mediated inhibition of ULK1 with potential reduction in autophagy, which may contribute to cardiac fibroblast activation59. Cardiac fibroblast proliferation mediated by cyclin D2 and cyclin-dependent kinase 6 (CDK6) is blunted by miRNA-1, which was shown to be downregulated in cardiac fibroblasts from Ang II-infused rats60. On the other hand, miRNA-96 promotes Ang II-induced proliferation and ECM production in murine cardiac fibroblasts61.
TGF-β stimulates expression of the profibrotic transcription factor scleraxis via both Smad-dependent and Smad-independent mechanisms62,63. In turn, scleraxis is critically required for various aspects of TGF-β/Smad signaling in the transdifferentiation of cardiac fibroblasts, including the TGF-β-dependent up-regulation of collagen Iɑ2 and ɑ-SMA, as well as myofibroblast contraction63,64. Further, the production of fibronectin65 and MMP266 is prevented in cardiac fibroblasts isolated from scleraxis null mouse hearts. Scleraxis null cardiac fibroblasts fail to undergo transdifferentiation to myofibroblasts in response to TGF-β or mechanical stretch64,67. We and others have reported that Ang II increases scleraxis expression in cardiac fibroblasts68,69. Our preliminary data indicates that Ang II-mediated up-regulation of ColIα1, ColIα2 and ColIIIα1 is attenuated by scleraxis knockdown, implicating scleraxis as an effector of Ang II fibrotic signaling (M. Czubryt, personal observation).
Periostin and osteopontin are two downstream products of Ang II-mediated fibroblast activation. Periostin is increased after MI and following Ang II infusion41,47,70,71. Genetic lineage tracing of cardiac fibroblasts has confirmed that activated fibroblasts express periostin and arise from resident cardiac fibroblasts of the Tcf21 lineage and that these cells modulate adaptive healing and fibrosis72. Periostin depletion can cause cardiac rupture with MI73, but limiting the persistence of periostin-expressing activated fibroblasts during 4 weeks of Ang II infusion or 3 weeks of MI attenuates fibrosis and improves cardiac function70. Both Tcf21 and periostin promoter-driven therapeutic interventions have been used during Ang II infusion74,75 to attenuate collagen deposition – suggesting a causal role for periostin in Ang II-induced fibrosis.
Osteopontin is associated with fibrogenic signaling but is also involved in the pro-inflammatory status of activated fibroblasts. The Ang II-induced increase in osteopontin, IL-6, and monocyte chemoattractant protein 1 (MCP-1) gene expression in mouse cardiac fibroblasts is prevented in cells from osteopontin knockout mice76. These reductions in gene expression correlate with an attenuation in cardiac fibrosis linking the fibrogenic effects of osteopontin induction with pro-inflammatory signaling. In another study, osteopontin knockout mice exhibit similar increases in ECM gene expression, but reduced adhesion and proliferation which attenuates interstitial cardiac fibrosis in response to Ang II compared to wildtype animals77.
2.2. Role of inflammation and immune cell infiltration in Ang II-dependent fibrosis
In addition to their role in promoting ECM production, activated fibroblasts initiate and perpetuate inflammation, which in turn exacerbates fibrosis in hypertensive hearts78. Cardiac fibrosis that occurs with Ang II infusion is dependent on bone marrow-derived inflammatory cells. Macrophages are recruited to the heart mainly via MCP-1. Macrophage infiltration into the heart peaks after 3 days of Ang II infusion in rodents and prevention of this influx limits fibrosis79. Direct effects of macrophages on fibroblast phenotype have been demonstrated using co-culture experiments. Specifically, macrophages stimulate fibroblasts to produce IL-6 which induces TGF-β-mediated Smad activation in the cardiac fibroblast80 as well as increased expression of collagen and ɑ-SMA79. Suppression of IL-6 in vitro also attenuates Ang II-induced TGF-β expression and downstream production of collagen and ɑ-SMA by fibroblasts81. Furthermore, using the in vivo model of Ang II infusion, TGF-β-mediated ECM and inflammatory signals are attenuated in MCP-134 and IL-680 knockout mice. However, hypertension and cardiac hypertrophy are not impacted by loss of MCP-1, differentiating the systemic hemodynamic response and hypertrophy from the fibrotic response34. Loss of tumor necrosis factor alpha (TNFɑ) receptor results in a reduction of cardiac fibrosis after 1 week of Ang II infusion in mice82. The relationship between Ang II and TNFɑ signaling is tightly linked to ROS production and reviewed here83. IL-6 and TNFɑ are increased by nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB)-mediated transcription downstream of toll-like receptor (TLR) stimulation. TLR2 knockout mice exhibit a marked attenuation in macrophage infiltration and the associated fibrosis upon Ang II infusion84. Given that Ang II mainly upregulated TLR2 expression in innate immune cells, this finding supports a main role for macrophages and neutrophils in the fibrotic response to Ang II.
2.3. Oxidative stress promotes fibrosis downstream of AT1 receptor activation
Oxidative stress is a major inducer of hypertension-related fibrosis and is linked to TGF-β signaling85. Ang II induces ROS generation, which stimulates fibroblast activation. NADPH oxidase (NOX) 1, 2, and 4 are enzymes that produce superoxide and hydrogen peroxide in times of cell stress and are expressed in both cardiomyocytes and cardiac fibroblasts86 which exacerbates cardiac fibrosis87. Of all NOX isoforms, cardiac fibroblasts primarily express NOX488. Ang II promotes increased NOX4 expression and cardiac overexpression of NOX4 worsens Ang II-induced cardiac hypertrophy and fibrosis87. The impact of NOX4 modulation likely depends on the duration of Ang II infusion. The absence of NOX4 does not alter cardiac fibrosis after 2 weeks of Ang II infusion89, but suppression of NOX4 protein and activity does attenuate fibrosis when assessed after 4 weeks of Ang II infusion in rodents86. Knockdown or inhibition of hydrogen peroxide-producing NOX4 attenuates proliferation, migration, and collagen secretion induced by Ang II in adult mouse cardiac fibroblasts90. Further, physical interaction of NOX4 with the AT1 receptor is enhanced by Ang II90. NOX4-dependent cardiac fibroblast migration and collagen production was shown to be mediated by Ang II-induced downregulation of regulator reversion-inducing-cysteine-rich protein with Kazal motifs (RECK) and subsequent unrestrained MMP expression91.
AT1 receptor/NOX mediated ROS production and fibrogenic gene expression (collagen I, collagen III, ɑ-SMA, TGF-β) is inhibited by a cardioprotective gasotransmitter, hydrogen sulfide (H2S) in isolated neonatal cardiac fibroblasts92. In addition, H2S has been shown to reduce fibrosis in hypertensive rats in vivo92, as well as to blunt Ang II-induced upregulation of NOX493. To combat the elevated ROS, antioxidants are initially increased in cardiac tissue after 24 hours of Ang II infusion, but this was not evident after 2 weeks - suggesting a reduced capacity to neutralize free radicals with prolonged RAS activation94. Longer term Ang II infusion with antioxidant co-treatment attenuates fibroblast activation and fibrosis85. Thus, agents that neutralize ROS reduce Ang II-induced fibroblast proliferation, migration, and activation. Various ROS scavengers and antioxidants including N-acetyl cysteine95, tempol90, propofol/alpha-tocopherol96, and combined antioxidant therapy97 prevent Ang II-induced collagen production and ɑ-SMA expression in cardiac fibroblasts, TGF-β activity, and fibroblast proliferation. Ang (1–7) also attenuates Ang II-induced ROS and fibrosis98.
Ultimately, ROS production downstream of AT1 receptor binding is mediated by NOX4-dependent ERK1/2 signaling. Thus, modulation of Ang II, ROS, or other endogenous regulators of the ERK1/2 pathway have an impact on fibrogenic signaling of cardiac fibroblasts. Targets of interest for ERK1/2 range from simple reduction in direct ROS scavenging to more interconnected alterations in ECM components and regulators. Importantly, there is a feed forward mechanism of Ang II and ROS production, which sustains expression and activity of enzymes like NOX4. In addition to the non-canonical responses to ROS, fibroblasts are known to be resistant to apoptosis due to constitutive expression of anti-apoptotic Bcl-299. Recently, Ang II activation of ERK1/2/RSK1 downstream of AT1 receptor stimulation led to inhibition of caspase-3 cleavage in rat and human cardiac myofibroblasts, adding to the anti-apoptotic properties of activated fibroblasts100. This combination of ROS-induced activation and resistance to cell death is key to the persistence of ECM-producing fibroblasts in the hypertensive and injured heart.
2.4. Epigenetic modulation by the RAS
In additional to direct modulation of signaling cascades, Ang II has been shown to induce epigenetic alterations of DNA and histones by methylation and acetylation to influence expression of pro-fibrotic genes101,102. Moreover, fibroblast transdifferentiation into myofibroblasts may also be regulated epigentically103. Histone acetylation promotes relaxation of DNA and increased transcription of genes. Thus, histone deacetylases (HDAC) condense DNA resulting in decreased gene transcription. Ang II104 and hypertension105 increase HDAC activity, while HDAC inhibitors attenuate hypertrophy, fibrosis and cardiac dysfunction in Ang II-infused mice106,107. Inhibition of Ang II-induced HDAC activity attenuates fibrogenic signaling via MMP9/RECK and inhibits the production of ECM proteins fibronectin and collagen in murine cardiac fibroblasts104.
Histone methylation is associated with both repression and activation of gene transcription, depending on the specific location and number of methyl groups on the various histones. However, Ang II108 has been shown to increase levels of histone demethylases in the heart in association with fibrogenic gene activation. Further, the expression of the ACE gene is increased in spontaneously hypertensive rats (SHR) and the ACE promoters are mostly unmethylated109. It is known that pressure overload and in vitro stimulation of cardiac fibroblasts with Ang II induces activation of the pro-fibrotic Wnt/β-catenin pathway110. Increased expression of the demethylase JMJD3 was recently linked with Ang II-mediated β-catenin and ECM production in cardiac fibroblasts111. Demethylation of histones was also recently shown to be controlled by metabolic shifts that occur during fibroblast transdifferentiation,112 further implicating histone methylation in cardiac fibroblast activation. Methylation of DNA within the promoter region of genes by DNA methyltransferases (DNMT) leads to inhibition of specific fibrogenic genes. For example, TGF-β induced ɑ-SMA occurs via inhibition of DNMT1 in cardiac fibroblasts and is correlated with reduced DNMT1 expression in the infarcted heart113. DNA hypomethylation is associated with hypertension114 and heart failure115. The epigenetic regulation of cardiac fibrosis has been reviewed in depth elsewhere116,117.
2.5. Single cell RNA-sequencing: Identifying the impact of Ang II on cardiac fibroblasts and novel therapeutic targets
Technological advances in single cell transcriptomics have identified multiple cardiac fibroblast populations and fibroblast-specific changes in gene expression in the remodeling heart. Cardiac fibroblasts treated in vitro with Ang II exhibit upregulation of 126 genes compared to control with IL-1β and MMP3 being identified as potential therapeutic targets118. In a study of cardiac cells from control vs. Ang II infused mice, single cell RNA sequencing analysis identified nine subpopulations of cardiac fibroblasts. Two of these subpopulations were primarily composed of fibroblasts from Ang II-infused hearts119. These two pathological subpopulations were defined by the expression of either Cilp or Thbs4 and were characterized by increased cell adhesion, wound healing, and ECM production compared to other fibroblast subpopulations. Further, the abundance of Thbs4 expressing fibroblasts was almost doubled in females compared to males among other gene changes that may underlie altered ECM quality and remodeling between the sexes. Another study showed that genetic deletion of Fstl1, an Ang II-responsive gene120, in cardiac fibroblasts increased cardiac rupture and mortality121. Proliferative and fibrogenic gene expression in periostin-traced cardiac fibroblasts after combination Ang II/phenylephrine infusion are reversed after a 2-week washout of the treatment demonstrating the capacity to downregulate these processes after injury resolution72. Several single cell assessments of cardiac cells, including fibroblasts, have been performed following MI. A comprehensive study evaluated a time course of MI in mice from diverse genetic backgrounds and found that mice prone to hypertension (mouse strain 129) exhibited an increased predisposition to cardiac rupture in the proliferative phase of wound healing122. The number of fibroblast subpopulations remained consistent, but the proportion of cells within the myofibroblast subpopulations was increased in mouse strain 129. Ultimately, the altered ratio of subpopulation composition resulted in increased activation and decreased fibrinolysis gene expression, which increased the rate of cardiac rupture122. As Ang II signaling is implicated in the pathology of MI, markers of activated fibroblasts in normotensive hearts may reveal novel targets for RAS inhibition as well. Gladka et al. identified Ckap4 as a novel marker of activated fibroblasts following ischemia/reperfusion injury, a marker which was modulated by TGF-β in vitro123. Farbehi et al. identified three myofibroblast subsets that emerge from activated fibroblasts 7 days post-MI124. Two myofibroblast populations expressed genes associated with fibrogenic pathways with markers of Scx and Thbs4124. The genetic makeup and relative proportion of cardiac fibroblast subpopulations holds valuable information for exacerbated cardiac fibrosis in hypertension and cardiac injury.
3. RAS inhibition alters cardiac fibroblast phenotype and limits fibrosis
As described above, Ang II stimulates fibrogenic and pro-inflammatory cascades in the cardiac fibroblast that fuel fibrotic remodeling. Thus, the inhibition of the production or actions of Ang II are known to attenuate adverse cardiac remodeling. Though the impact of RAS inhibition encompasses hemodynamic alterations and other tissue-specific benefits13, this section focuses on the known effects of RAS inhibition on cardiac fibrosis.
3.1. RAS inhibition and fibrosis in hypertensive heart disease
ACE inhibitors125–133, AT1 receptor antagonists134–137, and aldosterone antagonists134 have been shown to attenuate or reverse cardiac fibrosis in experimental models. Given the direct effects of Ang II on cardiac fibroblasts, it is perhaps not surprising that drugs that inhibit the production or action of this peptide would be effective in reducing fibrosis. Associated with RAS inhibition is an increase in collagenase activity, decreased collagen production and inhibition of TGF-β signaling138–140. In cardiac fibroblasts, activation of ERK1/2, p38 MAPK, and JAK/STAT pathways by Ang II are reduced by ACE inhibition141. Downstream of similar signaling pathways, periostin71 is upregulated in cardiac fibroblasts stimulated with Ang II, which is also shown to increase CCN2 (previously known as CTGF)142, both of which are attenuated by AT1 receptor blockers (ARBs). In addition to suppressing Ang II production, ACE inhibitors also inhibit hydrolysis of N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP)129 thus increasing plasma Ac-SDKP levels which reduces inflammatory cell infiltration and fibrotic remodeling143. Ac-SDKP has been shown to inhibit TGFβ-induced fibroblast activation, fibroblast proliferation, and collagen production - independently of Ang II144,145. Since fibroblasts have been shown to express the requisite components for Ang II production as well as the angiotensin receptors18,146, targeting this local RAS has been a proposed reason for the consistent efficacy of these drugs to attenuate or reverse cardiac fibrosis. Recently, FDA-approved angiotensin receptor-neprilysin inhibitors (ARNI), a combination of ARB/neprilysin inhibitors, not only antagonize AT1 receptors but also prevent the breakdown of vasodilatory natriuretic peptides147. Treatment with ARNIs during pressure overload produces superior cardioprotection than that achieved with either agent alone and is linked with PKG-dependent inhibition of RhoA activation in cardiac fibroblasts148.
3.2. Antihypertensive agents and cardiac fibroblast apoptosis
Hypertension is associated with fibroblast hyperplasia in the hypertrophied LV.149,150 ACE inhibitors, AT1 receptor antagonists, and calcium channel blockers (CCBs), when administered to adult male SHR, each induce a burst of apoptosis within the first 2 weeks of treatment that results in a 30% loss of LV fibroblasts149–154. In addition, AT1 receptor blockade has been shown to induce cardiac fibroblast apoptosis associated with reduced collagen deposition in a model of nitric oxide synthase (NOS) inhibitor-induced cardiac injury155. Similar treatment does not induce apoptosis of fibroblasts in the right ventricle, nor in the LV of normotensive WKY rats,154 suggesting that the excess fibroblasts that are present in the hypertrophied LV may be particularly susceptible to RAS inhibitor and CCB-induced cell death. Moreover, these drugs also protect against cardiomyocyte156–160 and endothelial cell159,161,162 death, suggesting LV fibroblast-specific effects.
The mechanisms by which RAS inhibitors and CCBs each induce cardiac fibroblast apoptosis are unknown, however the extrinsic (i.e. death receptor) pathway is frequently implicated in other cell types163–166. For example, increased levels of TNFα-related apoptosis inducing ligand (TRAIL) accompanied ACE inhibitor-induced apoptosis in intimal cells.164 Fas associated death domain (FADD) and tumor necrosis factor receptor type 1-associated death domain (TRADD) were both upregulated in response to ACE inhibition in apoptotic erythroid precursor cells.165 In addition, caspase-8 was upregulated during aortic vascular smooth muscle cell (VSMC) apoptosis in response to CCB treatment167. The molecular differences that render these subsets of cells uniquely susceptible to apoptosis by these antihypertensive drugs with distinct mechanisms of action have yet to be identified.
The stimulus that leads RAS inhibitors and CCBs to induce cell death also remains elusive. Fibroblast apoptosis appears to be independent of blood pressure lowering effects of these drugs, as beta blockers and hydralazine were unable to produce cell death despite effective antihypertensive response154. It may be that the fibroblasts of the hypertrophied LV are particularly dependent upon Ang II and calcium, and a loss of these survival factors triggers cell death. Given that fibroblast apoptosis precedes regression of LV hypertrophy, it is unlikely that this cell loss is merely secondary to a reduction in mass153. Interestingly, the spatiotemporal association of fibroblast apoptosis and regression of cardiomyocyte hypertrophy suggests that the fibroblasts may play a role in supporting the increased cardiomyocyte size153. This is similar to more recent studies in which a 50% loss of cardiac fibroblasts following germline deletion of scleraxis was accompanied by a ~30% decrease in cardiomyocyte size64.
During angiotensin receptor blockade, selective antagonism of the AT1 receptor leaves the AT2 receptor unopposed. While AT2 receptor stimulation has been shown to have growth inhibitory and pro-apoptotic effects, combined AT1 and AT2 blockade was unable to prevent valsartan-induced apoptosis of interstitial left ventricular fibroblasts152. During ACE inhibition, there is a prolongation of the half-life for bradykinin. Activation of bradykinin B1 and B2 receptors has also been proposed as a potential mechanism for ACE inhibitor-induced fibroblast apoptosis. Although this has not been tested in the LV, evidence from aortic VSMC suggests that while bradykinin B2 receptor activity did not block ACE inhibitor-induced apoptosis, there may be a role for the bradykinin B1 receptor168. In addition, the ACE inhibitor enalaprilat has been shown to prevent Ang II-induced proliferation as well as ROS/p38 activation in cardiac fibroblasts, in vitro169. Future studies aimed at identifying the mechanisms by which these antihypertensive agents induce apoptosis of LV fibroblasts may reveal a novel target for antifibrotic therapies.
3.3. Long-term consequences of transient RAS inhibition on cardiac fibroblasts
Inhibitors of the RAS produce a reduction of arterial pressure and LV hypertrophy that persists even after stopping treatment in adult male SHR14,170–174. Moreover, RAS inhibitors have been shown to be effective in producing long-term resistance to aging-induced cardiac fibrosis - an effect that persisted up to 15 months after stopping treatment134–137,175. In addition, a 2-week treatment of adult male SHR with the ACE inhibitor enalapril conferred protection against a future insult induced by NOS inhibition14,170,175,176. Specifically, rats that were previously treated with an ACE inhibitor showed a reduction in fibrosis, inflammation, and cellular proliferation in response to NOS inhibition14,176. This protection does not appear to be secondary to a reduction in cardiomyocyte loss. In fact, for a given area of infarction there were fewer macrophages infiltrating the hearts of previously-treated rats.14 This protection against NOS inhibitor-induced pathological remodeling was found to be related to a phenotypic difference in the cardiac fibroblasts following transient ACE inhibition. Cardiac fibroblasts isolated from SHR transiently treated with an ACE inhibitor had a lower proliferation rate, collagen I gene expression, and secretion of chemokines when compared to fibroblasts isolated from naive SHR that were treated with a NOS inhibitor177. Taken together, inhibition of the RAS produces persistent effects in the cardiac fibroblasts leading to a less fibrogenic phenotype. Whether this is a result of a loss of a subset of fibrogenic fibroblasts during treatment, or epigenetic changes in the fibroblasts during treatment, is unknown.
3.4. RAS inhibition and protection from myocardial infarction
With MI, the ischemic area of the heart undergoes massive tissue necrosis and the fibrotic remodeling that occurs is necessary for the repair of the heart. Regions remote to the injury also accumulate interstitial fibrosis that contributes to myocardial stiffness and ultimately heart failure. Activation of the RAS occurs with MI178–181 and therefore inhibitors of the RAS have been shown to slow cardiac remodeling and improve cardiac function. Despite no change in infarct size, ACE inhibition improved survival and decreased LV hypertrophy and remote fibrotic remodeling in normotensive182–187 and hypertensive hearts188. The protection in SHRs was associated with a reduced capacity to induce ventricular arrhythmias188. Interestingly, treatment of SHRs with either ramipril or losartan reduces LV fibrosis, but not collagen mRNA suggesting that the protection occurs post-translationally189. Reduction of oxidative stress186 and improved antioxidant activity190 have been associated with ACE inhibitor-mediated protection. A longer-term study found that 25 weeks after MI, collagen and CCN2 protein were elevated, but only collagen was reduced by captopril implying that CCN2 is an important regulator of late cardiac remodeling independent of RAS inhibition184. The combination of an ACE inhibitor with resveratrol190, bradykinin191, soluble guanylyl cyclase activator192, or mineralocorticoid receptor antagonist193 further suppresses collagen deposition.
Like ACE inhibitors, ARBs do not reduce the size of the infarct and therefore also alter remote cardiac remodeling and hypertrophy to improve outcomes194. Mice with MI that were treated with valsartan exhibited reduced hypertrophy, fibroblast proliferation and fibrosis195. Olmesartan administration to mice with MI prevented cardiac rupture due to attenuated periostin expression196. The ARB, telmisartan, is also a peroxisome proliferator-activated receptor-gamma (PPAR-γ) activator, which further reduces hypertrophy, fibrosis, and macrophage infiltration to improve hemodynamic function compared to losartan. Moreover, isolated cardiac fibroblasts pretreated with telmisartan produce less MMP2, MMP9, CCN2, and osteopontin, but more TIMP1 then losartan treated fibroblasts in hypoxic conditions197. Elevated CCN2 in failing hearts after MI is attenuated with losartan142.
Most studies find equivalent protection afforded by either ACE inhibitors or ARBs, suggesting that the reduction in Ang II is the primary mediator of the beneficial effects of these interventions. However, one study found that post-MI rats had greater reductions in fibroblast proliferation and LV collagen content with enalapril compared to losartan with similar reductions in hypertrophy198, while another reported treatment post-MI with valsartan reduced collagen, immune cell infiltration, and TGF-β in the LV to a greater extent than fosinopril199. Both studies evaluated 2 weeks post-MI in normotensive rats that received therapeutic intervention 24 hours after surgery, therefore the differences in findings may be due to drug or dose, rather than class-specific findings. Similar to findings in pressure overload, ARNIs produce greater reductions in fibrotic remodeling compared to an ARB200 or ACE inhibitor201 alone after MI likely due to added reduction in cardiac hypertrophy200.
Cardioprotection can also be achieved by activation of the alternative RAS pathway. Activation of ACE2, which converts Ang II to Ang (1–7), was recently shown to attenuate pathological remodeling in the normotensive rat heart when measured 4 weeks post-MI202. The mechanism of action for Ang (1–7) may be linked to the proinflammatory signaling that occurs acutely following the insult. Treatment of rats with Ang (1–7) promoted resolution of inflammation and reduced mitochondrial dysfunction related to downregulation of C-X-C chemokine receptor 4 (CXCR4) during MI203. The mechanisms of protection discussed here underlie the benefits of RAS inhibition in humans as well. ACE inhibitors204–208, and ARBs209,210 reduce post-MI mortality and secondary events. Collectively, there is consensus on improved cardiac remodeling with RAS inhibition post-MI. Given that the size of the infarct is often unaffected, limiting the fibrotic remodeling of the viable myocardium is of great importance to prevent the development of cardiac dysfunction and heart failure. Clinical use of RAS inhibitors is common, but alternative strategies and novel targets are necessary for development of anti-fibrotic therapies. The known actions of Ang II in the cardiac fibroblast can be used to identify these targets and potentially determine more direct methods of intervention for heart failure.
4. Conclusions and Future Directions: Cardiac fibroblasts as therapeutic targets
Mounting evidence supports that limiting the extent and duration of cardiac fibroblast activation is necessary for cardioprotection after injury and slowed progression toward heart failure. It is important to allow for the appropriate initiation of wound healing for cardiac repair and elasticity for contraction without excessive fibrotic remodeling. These collective data highlight multiple players that contribute to fibroblast activity and ECM production downstream of Ang II, including various fibrogenic signaling pathways, inflammation, and oxidative stress. Thus, protection is afforded by RAS inhibitors as summarized in Figure 2, but the timing of treatment dictates the extent of that protection. During pressure overload and cardiac injury, cardioprotection is achieved with ACE inhibitors, ARBs, and ARNIs when treatment occurs prior to or during the insult. Given the lack of protection after the establishment of fibrotic remodeling, novel targets and approaches are needed. Future directions for identification of mechanisms of cardiac fibroblast activation are summarized in Figure 3.
Figure 2. RAS inhibition reduces cardiac fibrosis and improves outcomes after chronic or acute injury.

Hypertension and cardiac injury (e.g. myocardial infarction) induce changes in the heart that result in cardiac fibrosis, impaired function, and increased mortality. These insults impact several cell types resulting in cardiomyocyte hypertrophy, fibroblast proliferation and activation, and immune cell recruitment to the heart. Together, the adaptive responses of cardiac cells cause heart wall thickening and extracellular matrix production that reduce cardiac contractility and output that are attenuated by RAS inhibition. Created with BioRender.com.
Figure 3. Current knowledge gaps and future areas of study.

Important foci for future investigation are highlighted.
Research in the area of cardiac RAS signaling and modulation of adverse cardiac remodeling has produced the most widely used therapeutics for patients with heart failure secondary to hypertension and MI. The growing knowledge base in fibrogenic proteins, epigenetics, and cardiac fibroblast subpopulations are elucidating novel targets for intervention that may increase the anti-fibrotic efficacy of existing approaches. Furthermore, transient ACE inhibition has been shown to produce persistently altered cardiac fibroblasts with reduced proliferative, pro-inflammatory, and fibrogenic profiles14,170,176,177. Understanding the ways in which cardiac fibroblasts are reprogrammed by transient ACE inhibition, and therefore targeting Ang II-mediated pathways, to manifest suppressed responses to injury may hold new insights for limiting fibrotic remodeling. As single cell investigations of cardiac fibroblasts become more numerous, genetic candidates for excessive cardiac fibroblast activation are being described and providing more avenues for future work. Thus far, TGF-β has been described as the main fibrogenic mediator downstream of AT1 receptor stimulation. However, it is possible that some fibrogenic signals may work independently of TGF-β and warrants investigation. Additionally, the regulation of AT1 receptor expression and the incidence of Ang II-induced cell priming, or feed-forward mechanisms are impact Ang II-mediated signaling. It is also possible that different cardiac cell types have different Ang II receptor expression bias to regulate cell-specific responses. The roles for multiple cell types including cardiac fibroblasts, cardiomyocytes, and immune cells during fibrotic remodeling, cell-specific responses to ROS and the timing and extent of oxidative stress are important areas of further investigation of cell-cell communication. Lastly, most of the reviewed studies were performed in males. The known differences in cardiac remodeling due to sex were recently reviewed211 and the role of estrogen in various fibrogenic signaling cascades was discussed. However, sex-specific differences in fibrotic remodeling remains an important and understudied area of investigation. Research efforts in these areas will increase our mechanistic understanding of how to prevent, limit and reverse fibrosis.
Highlights.
Angiotensin induces fibroblast activation, proliferation, and inflammation
Renin angiotensin system inhibitors attenuate fibrotic remodeling
Targeting angiotensin-mediated pathways may produce lasting effects in fibroblasts
Funding Sources
This work was supported by the National Institutes of Health NHLBI HL141165 (TMH); and the American Heart Association 19AIREA34460000 (TMH) and 19POST34410055 (AMG); and Project Grant PJT-162422 from the Canadian Institutes of Health Research (MPC).
Abbreviations:
- RAS
renin angiotensin system
- ACE
angiotensin converting enzyme
- Ang II
angiotensin II
- AT1
angiotensin receptor type 1
- AT2
angiotensin receptor type 2
- Ang (1–7)
angiotensin-(1–7)
- ARB
angiotensin receptor blocker
- MI
myocardial infarction
- ECM
extracellular matrix
- ɑ-SMA
ɑ-smooth muscle actin
- MAPK
mitogen-activated protein kinases
- ERK1/2
extracellular signal-regulated kinase 1/2
- ROS
reactive oxygen species
- TGF-β
transforming growth factor beta
- STAT3
signal transducer and activator of transcription 3
- TLR4
toll-like receptor 4
- miRNA
microRNA
- CDK6
cyclin-dependent kinase 6
- MCP-1
monocyte chemoattractant protein 1
- AP-1
activator protein-1
- TNFɑ
tumor necrosis factor alpha
- NFκB
nuclear factor kappa-light-chain-enhancer of activated B cells
- NOX
NADPH oxidase
- RECK
reversion-inducing-cysteine-rich protein with Kazal motifs
- DNMT
DNA methyltransferase
- HDAC
histone deacetylases
- Ac-SDKP
N-acetyl-seryl-aspartyl-lysyl-proline
- ARNI
angiotensin receptor-neprilysin inhibitors
- PPAR-γ
peroxisome proliferator-activated receptor-gamma
- CCB
calcium channel blocker
- SHR
spontaneously hypertensive rat
- NOS
nitric oxide synthase
- LV
left ventricle
- VSMC
vascular smooth muscle cell
- TRAIL
TNFα-related apoptosis inducing ligand
- FADD
Fas associated death domain
- TRADD
tumor necrosis factor receptor type 1-associated death domain
- CXCR4
C-X-C chemokine receptor 4
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Müller DN, Fischli W, Clozel J-P, Hilgers KF, Bohlender J, Ménard J, Busjahn A, Ganten D, Luft FC. Local Angiotensin II Generation in the Rat Heart. Circulation research. 1998;82(1):13–20. [DOI] [PubMed] [Google Scholar]
- 2.Heller LJ, Opsahl JA, Wernsing SE, Saxena R, Katz SA. Myocardial and plasma renin-angiotensinogen dynamics during pressure-induced cardiac hypertrophy. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 1998;274(3):R849–R856. [DOI] [PubMed] [Google Scholar]
- 3.Passier RC, Smits JF, Verluyten MJ, Daemen MJ. Expression and localization of renin and angiotensinogen in rat heart after myocardial infarction. Am J Physiol. 1996;271(3 Pt 2):H1040–1048. [DOI] [PubMed] [Google Scholar]
- 4.Sun Y, Zhang J, Zhang JQ, Weber KT. Renin expression at sites of repair in the infarcted rat heart. Journal of molecular and cellular cardiology. 2001;33(5):995–1003. [DOI] [PubMed] [Google Scholar]
- 5.Yamada H, Fabris B, Allen A, Jackson B, Johnston CI, Mendelsohn A. Localization of angiotensin converting enzyme in rat heart. Circulation research. 1991;68(1):141–149. [DOI] [PubMed] [Google Scholar]
- 6.Schunkert H, Dzau VJ, Tang SS, Hirsch AT, Apstein CS, Lorell BH. Increased rat cardiac angiotensin converting enzyme activity and mRNA expression in pressure overload left ventricular hypertrophy. Effects on coronary resistance, contractility, and relaxation. The Journal of clinical investigation. 1990;86(6):1913–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Passier R, Smits J, Verluyten M, Studer R, Drexler H, Daemen M. Activation of angiotensin-converting enzyme expression in infarct zone following myocardial infarction. American Journal of Physiology-Heart and Circulatory Physiology. 1995;269(4):H1268–H1276. [DOI] [PubMed] [Google Scholar]
- 8.van Kats JP, de Lannoy LM, Danser AJ, van Meegen JR, Verdouw PD, Schalekamp MA. Angiotensin II Type 1 (AT1) receptor–mediated accumulation of angiotensin II in tissues and its intracellular half-life in vivo. Hypertension (Dallas, Tex : 1979). 1997;30(1):42–49. [DOI] [PubMed] [Google Scholar]
- 9.van Kats JP, Danser AJ, van Meegen JR, Sassen LM, Verdouw PD, Schalekamp MA. Angiotensin production by the heart: a quantitative study in pigs with the use of radiolabeled angiotensin infusions. Circulation. 1998;98(1):73–81. [DOI] [PubMed] [Google Scholar]
- 10.Tao X, Fan J, Kao G, Zhang X, Su L, Yin Y, Zrenner B. Angiotensin-(1–7) attenuates angiotensin II-induced signalling associated with activation of a tyrosine phosphatase in Sprague-Dawley rats cardiac fibroblasts. Biol Cell. 2014;106(6):182–192. [DOI] [PubMed] [Google Scholar]
- 11.Nakamoto H, Ferrario CM, Fuller SB, Robaczewski DL, Winicov E, Dean RH. Angiotensin-(1–7) and nitric oxide interaction in renovascular hypertension. Hypertension (Dallas, Tex : 1979). 1995;25(4 Pt 2):796–802. [DOI] [PubMed] [Google Scholar]
- 12.Xu J, Carretero OA, Liao TD, Peng H, Shesely EG, Xu J, Liu TS, Yang JJ, Reudelhuber TL, Yang XP. Local angiotensin II aggravates cardiac remodeling in hypertension. American journal of physiology Heart and circulatory physiology. 2010;299(5):H1328–1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.AlQudah M, Hale TM, Czubryt MP. Targeting the renin-angiotensin-aldosterone system in fibrosis. Matrix Biology. 2020;91–92:92–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Biwer LA, D’Souza KM, Abidali A, Tu D, Siniard AL, DeBoth M, Huentelman M, Hale TM. Time course of cardiac inflammation during nitric oxide synthase inhibition in SHR: impact of prior transient ACE inhibition. Hypertension research : official journal of the Japanese Society of Hypertension. 2016;39(1):8–18. [DOI] [PubMed] [Google Scholar]
- 15.Smallegange C, Hale TM, Bushfield TL, Adams MA. Persistent lowering of pressure by transplanting kidneys from adult spontaneously hypertensive rats treated with brief antihypertensive therapy. Hypertension (Dallas, Tex : 1979). 2004;44(1):89–94. [DOI] [PubMed] [Google Scholar]
- 16.Singh VP, Baker KM, Kumar R. Activation of the intracellular renin-angiotensin system in cardiac fibroblasts by high glucose: role in extracellular matrix production. American journal of physiology Heart and circulatory physiology. 2008;294(4):H1675–1684. [DOI] [PubMed] [Google Scholar]
- 17.Sun Y, Cleutjens JP, Diaz-Arias AA, Weber KT. Cardiac angiotensin converting enzyme and myocardial fibrosis in the rat. Cardiovascular research. 1994;28(9):1423–1432. [DOI] [PubMed] [Google Scholar]
- 18.Katwa LC, Campbell SE, Tyagi SC, Lee SJ, Cicila GT, Weber KT. Cultured myofibroblasts generate angiotensin peptides de novo. Journal of molecular and cellular cardiology. 1997;29(5):1375–1386. [DOI] [PubMed] [Google Scholar]
- 19.Sun Y, Weber KT. Cells expressing angiotensin II receptors in fibrous tissue of rat heart. Cardiovascular research. 1996;31(4):518–525. [PubMed] [Google Scholar]
- 20.Anupama V, George M, Dhanesh SB, Chandran A, James J, Shivakumar K. Molecular mechanisms in H2O2-induced increase in AT1 receptor gene expression in cardiac fibroblasts: A role for endogenously generated Angiotensin II. Journal of molecular and cellular cardiology. 2016;97:295–305. [DOI] [PubMed] [Google Scholar]
- 21.Tadevosyan A, Xiao J, Surinkaew S, Naud P, Merlen C, Harada M, Qi X, Chatenet D, Fournier A, Allen BG, Nattel S. Intracellular Angiotensin-II Interacts With Nuclear Angiotensin Receptors in Cardiac Fibroblasts and Regulates RNA Synthesis, Cell Proliferation, and Collagen Secretion. Journal of the American Heart Association. 2017;6(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.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(5):C1457–1467. [DOI] [PubMed] [Google Scholar]
- 23.Walther T, Klostermann K, Heringer-Walther S, Schultheiss HP, Tschöpe C, Stepan H. Fibrosis rather than blood pressure determines cardiac BNP expression in mice. Regul Pept. 2003;116(1–3):95–100. [DOI] [PubMed] [Google Scholar]
- 24.Ichihara S, Senbonmatsu T, Price E Jr., Ichiki T, Gaffney FA, Inagami T. Angiotensin II type 2 receptor is essential for left ventricular hypertrophy and cardiac fibrosis in chronic angiotensin II-induced hypertension. Circulation. 2001;104(3):346–351. [DOI] [PubMed] [Google Scholar]
- 25.Harada K, Sugaya T, Murakami K, Yazaki Y, Komuro I. Angiotensin II type 1A receptor knockout mice display less left ventricular remodeling and improved survival after myocardial infarction. Circulation. 1999;100(20):2093–2099. [DOI] [PubMed] [Google Scholar]
- 26.Voros S, Yang Z, Bove CM, Gilson WD, Epstein FH, French BA, Berr SS, Bishop SP, Conaway MR, Matsubara H, Carey RM, Kramer CM. Interaction between AT1 and AT2 receptors during postinfarction left ventricular remodeling. American journal of physiology Heart and circulatory physiology. 2006;290(3):H1004–1010. [DOI] [PubMed] [Google Scholar]
- 27.Wagner JUG, Dimmeler S. Cellular cross-talks in the diseased and aging heart. Journal of molecular and cellular cardiology. 2020;138:136–146. [DOI] [PubMed] [Google Scholar]
- 28.Zhang P, Su J, Mende U. Cross talk between cardiac myocytes and fibroblasts: from multiscale investigative approaches to mechanisms and functional consequences. American Journal of Physiology-Heart and Circulatory Physiology. 2012;303(12):H1385–H1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.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. The Journal of Clinical Investigation. 2018;128(5):2127–2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Frangogiannis NG. Matricellular proteins in cardiac adaptation and disease. Physiological reviews. 2012;92(2):635–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Tallquist MD, Molkentin JD. Redefining the identity of cardiac fibroblasts. Nature reviews Cardiology. 2017;14(8):484–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Crawford DC, Chobanian AV, Brecher P. Angiotensin II induces fibronectin expression associated with cardiac fibrosis in the rat. Circulation research. 1994;74(4):727–739. [DOI] [PubMed] [Google Scholar]
- 33.Zhang R, Zhang YY, Huang XR, Wu Y, Chung AC, Wu EX, Szalai AJ, Wong BC, Lau CP, Lan HY. C-reactive protein promotes cardiac fibrosis and inflammation in angiotensin II-induced hypertensive cardiac disease. Hypertension (Dallas, Tex : 1979). 2010;55(4):953–960. [DOI] [PubMed] [Google Scholar]
- 34.Haudek SB, Cheng J, Du J, Wang Y, Hermosillo-Rodriguez J, Trial J, Taffet GE, Entman ML. Monocytic fibroblast precursors mediate fibrosis in angiotensin-II-induced cardiac hypertrophy. Journal of molecular and cellular cardiology. 2010;49(3):499–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, Loibner H, Wang XH, Penninger JM, Kassiri Z, Oudit GY. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation. 2010;122(7):717–728, 718 p following 728. [DOI] [PubMed] [Google Scholar]
- 36.Jiang HM, Wang HX, Yang H, Zeng XJ, Tang CS, Du J, Li HH. Role for granulocyte colony stimulating factor in angiotensin II-induced neutrophil recruitment and cardiac fibrosis in mice. American journal of hypertension. 2013;26(10):1224–1233. [DOI] [PubMed] [Google Scholar]
- 37.Bai F, Pang XF, Zhang LH, Wang NP, McKallip RJ, Garner RE, Zhao ZQ. Angiotensin II AT1 receptor alters ACE2 activity, eNOS expression and CD44-hyaluronan interaction in rats with hypertension and myocardial fibrosis. Life Sci. 2016;153:141–152. [DOI] [PubMed] [Google Scholar]
- 38.Sun Y, Weber KT. Angiotensin converting enzyme and myofibroblasts during tissue repair in the rat heart. Journal of molecular and cellular cardiology. 1996;28(5):851–858. [DOI] [PubMed] [Google Scholar]
- 39.Sun Y, Zhang JQ, Zhang J, Ramires FJA. Angiotensin II, Transforming Growth Factor-β1and Repair in the Infarcted Heart. Journal of molecular and cellular cardiology. 1998;30(8):1559–1569. [DOI] [PubMed] [Google Scholar]
- 40.Wong CKS, Falkenham A, Myers T, Légaré J-F. Connective tissue growth factor expression after angiotensin II exposure is dependent on transforming growth factor-β signaling via the canonical Smad-dependent pathway in hypertensive induced myocardial fibrosis. J Renin Angiotensin Aldosterone Syst. 2018;19(1):1470320318759358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Li L, Fan D, Wang C, Wang JY, Cui XB, Wu D, Zhou Y, Wu LL. Angiotensin II increases periostin expression via Ras/p38 MAPK/CREB and ERK1/2/TGF-β1 pathways in cardiac fibroblasts. Cardiovascular research. 2011;91(1):80–89. [DOI] [PubMed] [Google Scholar]
- 42.Campbell SE, Katwa LC. Angiotensin II stimulated expression of transforming growth factor-beta1 in cardiac fibroblasts and myofibroblasts. Journal of molecular and cellular cardiology. 1997;29(7):1947–1958. [DOI] [PubMed] [Google Scholar]
- 43.Lee AA, Dillmann WH, McCulloch AD, Villarreal FJ. Angiotensin II stimulates the autocrine production of transforming growth factor-beta 1 in adult rat cardiac fibroblasts. Journal of molecular and cellular cardiology. 1995;27(10):2347–2357. [DOI] [PubMed] [Google Scholar]
- 44.Hao J, Wang B, Jones SC, Jassal DS, Dixon IM. Interaction between angiotensin II and Smad proteins in fibroblasts in failing heart and in vitro. American journal of physiology Heart and circulatory physiology. 2000;279(6):H3020–3030. [DOI] [PubMed] [Google Scholar]
- 45.McWhinney CD, Hunt RA, Conrad KM, Dostal DE, Baker KM. The type I angiotensin II receptor couples to Stat1 and Stat3 activation through Jak2 kinase in neonatal rat cardiac myocytes. Journal of molecular and cellular cardiology. 1997;29(9):2513–2524. [DOI] [PubMed] [Google Scholar]
- 46.Han J, Ye S, Zou C, Chen T, Wang J, Li J, Jiang L, Xu J, Huang W, Wang Y, Liang G. Angiotensin II Causes Biphasic STAT3 Activation Through TLR4 to Initiate Cardiac Remodeling. Hypertension (Dallas, Tex : 1979). 2018;72(6):1301–1311. [DOI] [PubMed] [Google Scholar]
- 47.Bao Q, Zhang B, Suo Y, Liu C, Yang Q, Zhang K, Yuan M, Yuan M, Zhang Y, Li G. Intermittent hypoxia mediated by TSP1 dependent on STAT3 induces cardiac fibroblast activation and cardiac fibrosis. Elife. 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ye S, Luo W, Khan ZA, Wu G, Xuan L, Shan P, Lin K, Chen T, Wang J, Hu X, Wang S, Huang W, Liang G. Celastrol Attenuates Angiotensin II-Induced Cardiac Remodeling by Targeting STAT3. Circulation research. 2020;126(8):1007–1023. [DOI] [PubMed] [Google Scholar]
- 49.Su SA, Yang D, Wu Y, Xie Y, Zhu W, Cai Z, Shen J, Fu Z, Wang Y, Jia L, Wang Y, Wang JA, Xiang M. EphrinB2 Regulates Cardiac Fibrosis Through Modulating the Interaction of Stat3 and TGF-β/Smad3 Signaling. Circulation research. 2017;121(6):617–627. [DOI] [PubMed] [Google Scholar]
- 50.Zhao J, Gao J-L, Zhu J-X, Zhu H-B, Peng X, Jiang M, Fu Y, Xu J, Mao X-H, Hu N, Ma M-H, Dong D-L. The different response of cardiomyocytes and cardiac fibroblasts to mitochondria inhibition and the underlying role of STAT3. Basic research in cardiology. 2019;114(2):12. [DOI] [PubMed] [Google Scholar]
- 51.Reddy SS, Agarwal H, Barthwal MK. Cilostazol ameliorates heart failure with preserved ejection fraction and diastolic dysfunction in obese and non-obese hypertensive mice. Journal of molecular and cellular cardiology. 2018;123:46–57. [DOI] [PubMed] [Google Scholar]
- 52.Liu M, Yeh J, Huang Y, Redondo A, Ke J, Yao J, Tan G, Tang W, Chen J. EFFECT OF TRIPTOLIDE ON PROLIFERATION AND APOPTOSIS OF ANGIOTENSIN II-INDUCED CARDIAC FIBROBLASTS IN VITRO: A PRELIMINARY STUDY. Afr J Tradit Complement Altern Med. 2017;14(1):145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chung CC, Kao YH, Liou JP, Chen YJ. Curcumin Suppress Cardiac Fibroblasts Activities by Regulating Proliferation, Migration, and the Extracellular Matrix. Acta Cardiol Sin. 2014;30(5):474–482. [PMC free article] [PubMed] [Google Scholar]
- 54.Chen Y, Yang S, Yao W, Zhu H, Xu X, Meng G, Zhang W. Prostacyclin analogue beraprost inhibits cardiac fibroblast proliferation depending on prostacyclin receptor activation through a TGF β-Smad signal pathway. PloS one. 2014;9(5):e98483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Xu X, Pang J, Yin H, Li M, Hao W, Chen C, Cao JM. Hexarelin suppresses cardiac fibroblast proliferation and collagen synthesis in rat. American journal of physiology Heart and circulatory physiology. 2007;293(5):H2952–2958. [DOI] [PubMed] [Google Scholar]
- 56.Samuel CS, Unemori EN, Mookerjee I, Bathgate RA, Layfield SL, Mak J, Tregear GW, Du XJ. Relaxin modulates cardiac fibroblast proliferation, differentiation, and collagen production and reverses cardiac fibrosis in vivo. Endocrinology. 2004;145(9):4125–4133. [DOI] [PubMed] [Google Scholar]
- 57.Verjans R, Peters T, Beaumont FJ, van Leeuwen R, van Herwaarden T, Verhesen W, Munts C, Bijnen M, Henkens M, Diez J, de Windt LJ, van Nieuwenhoven FA, van Bilsen M, Goumans MJ, Heymans S, González A, Schroen B. MicroRNA-221/222 Family Counteracts Myocardial Fibrosis in Pressure Overload-Induced Heart Failure. Hypertension (Dallas, Tex : 1979). 2018;71(2):280–288. [DOI] [PubMed] [Google Scholar]
- 58.Ding J, Tang Q, Luo B, Zhang L, Lin L, Han L, Hao M, Li M, Yu L, Li M. Klotho inhibits angiotensin II-induced cardiac hypertrophy, fibrosis, and dysfunction in mice through suppression of transforming growth factor-β1 signaling pathway. Eur J Pharmacol. 2019;859:172549. [DOI] [PubMed] [Google Scholar]
- 59.Li ML, Li RN, Ma YM, Jiang B, Chen YJ, Hu WX, Qv CL, Zhang YJ, Song YY, Wang Y. MiRNA-1297 inhibits myocardial fibrosis by targeting ULK1. Eur Rev Med Pharmacol Sci. 2020;24(4):2070–2076. [DOI] [PubMed] [Google Scholar]
- 60.Valkov N, King ME, Moeller J, Liu H, Li X, Zhang P. MicroRNA-1-Mediated Inhibition of Cardiac Fibroblast Proliferation Through Targeting Cyclin D2 and CDK6. Front Cardiovasc Med. 2019;6:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Su L, Yao Y, Song W. Downregulation of miR-96 suppresses the profibrogenic functions of cardiac fibroblasts induced by angiotensin II and attenuates atrial fibrosis by upregulating KLF13. Hum Cell. 2020;33(2):337–346. [DOI] [PubMed] [Google Scholar]
- 62.Zeglinski MR, Roche P, Hnatowich M, Jassal DS, Wigle JT, Czubryt MP, Dixon IM. TGFβ1 regulates Scleraxis expression in primary cardiac myofibroblasts by a Smad-independent mechanism. American journal of physiology Heart and circulatory physiology. 2016;310(2):H239–249. [DOI] [PubMed] [Google Scholar]
- 63.Bagchi RA, Czubryt MP. Synergistic roles of scleraxis and Smads in the regulation of collagen 1α2 gene expression. Biochim Biophys Acta. 2012;1823(10):1936–1944. [DOI] [PubMed] [Google Scholar]
- 64.Bagchi RA, Roche P, Aroutiounova N, Espira L, Abrenica B, Schweitzer R, Czubryt MP. The transcription factor scleraxis is a critical regulator of cardiac fibroblast phenotype. BMC Biol. 2016;14:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bagchi RA, Lin J, Wang R, Czubryt MP. Regulation of fibronectin gene expression in cardiac fibroblasts by scleraxis. Cell Tissue Res. 2016;366(2):381–391. [DOI] [PubMed] [Google Scholar]
- 66.Nagalingam RS, Safi HA, Al-Hattab DS, Bagchi RA, Landry NM, Dixon IMC, Wigle JT, Czubryt MP. Regulation of cardiac fibroblast MMP2 gene expression by scleraxis. Journal of molecular and cellular cardiology. 2018;120:64–73. [DOI] [PubMed] [Google Scholar]
- 67.Roche PL, Nagalingam RS, Bagchi RA, Aroutiounova N, Belisle BM, Wigle JT, Czubryt MP. Role of scleraxis in mechanical stretch-mediated regulation of cardiac myofibroblast phenotype. Am J Physiol Cell Physiol. 2016;311(2):C297–307. [DOI] [PubMed] [Google Scholar]
- 68.Czubryt MP, Bagchi RA, Roche PL, Wang R, Devalapurkar S, Schweitzer R. Abstract 192: Scleraxis is a Required Component of the Cardiac Extracellular Matrix Gene Expression Program. Circulation research. 2014;115(suppl_1):A192–A192. [Google Scholar]
- 69.Salameh MM, Katwa LC. Angiotensin II up-regulates the expression of scleraxis in cardiac myofibroblasts isolated from the site of myocardial infarction. The FASEB Journal. 2011;25(S1):1032.1039–1032.1039. [Google Scholar]
- 70.Kaur H, Takefuji M, Ngai CY, Carvalho J, Bayer J, Wietelmann A, Poetsch A, Hoelper S, Conway SJ, Möllmann H, Looso M, Troidl C, Offermanns S, Wettschureck N. Targeted Ablation of Periostin-Expressing Activated Fibroblasts Prevents Adverse Cardiac Remodeling in Mice. Circulation research. 2016;118(12):1906–1917. [DOI] [PubMed] [Google Scholar]
- 71.Iekushi K, Taniyama Y, Azuma J, Katsuragi N, Dosaka N, Sanada F, Koibuchi N, Nagao K, Ogihara T, Morishita R. Novel mechanisms of valsartan on the treatment of acute myocardial infarction through inhibition of the antiadhesion molecule periostin. Hypertension (Dallas, Tex : 1979). 2007;49(6):1409–1414. [DOI] [PubMed] [Google Scholar]
- 72.Kanisicak O, Khalil H, Ivey MJ, Karch J, Maliken BD, Correll RN, Brody MJ, Lin S-CJ, Aronow BJ, Tallquist MD. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nature communications. 2016;7(1):1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.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. Circulation research. 2007;101(3):313–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Dorn LE, Petrosino JM, Wright P, Accornero F. CTGF/CCN2 is an autocrine regulator of cardiac fibrosis. Journal of molecular and cellular cardiology. 2018;121:205–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shimizu T, Narang N, Chen P, Yu B, Knapp M, Janardanan J, Blair J, Liao JK. Fibroblast deletion of ROCK2 attenuates cardiac hypertrophy, fibrosis, and diastolic dysfunction. JCI Insight. 2017;2(13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lorenzen JM, Schauerte C, Hübner A, Kölling M, Martino F, Scherf K, Batkai S, Zimmer K, Foinquinos A, Kaucsar T, Fiedler J, Kumarswamy R, Bang C, Hartmann D, Gupta SK, Kielstein J, Jungmann A, Katus HA, Weidemann F, Müller OJ, Haller H, Thum T. Osteopontin is indispensible for AP1-mediated angiotensin II-related miR-21 transcription during cardiac fibrosis. Eur Heart J. 2015;36(32):2184–2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Collins AR, Schnee J, Wang W, Kim S, Fishbein MC, Bruemmer D, Law RE, Nicholas S, Ross RS, Hsueh WA. Osteopontin modulates angiotensin II-induced fibrosis in the intact murine heart. J Am Coll Cardiol. 2004;43(9):1698–1705. [DOI] [PubMed] [Google Scholar]
- 78.Lindner D, Zietsch C, Tank J, Sossalla S, Fluschnik N, Hinrichs S, Maier L, Poller W, Blankenberg S, Schultheiss HP, Tschöpe C, Westermann D. Cardiac fibroblasts support cardiac inflammation in heart failure. Basic research in cardiology. 2014;109(5):428. [DOI] [PubMed] [Google Scholar]
- 79.Falkenham A, de Antueno R, Rosin N, Betsch D, Lee TD, Duncan R, Légaré JF. Nonclassical resident macrophages are important determinants in the development of myocardial fibrosis. Am J Pathol. 2015;185(4):927–942. [DOI] [PubMed] [Google Scholar]
- 80.Ma F, Li Y, Jia L, Han Y, Cheng J, Li H, Qi Y, Du J. Macrophage-stimulated cardiac fibroblast production of IL-6 is essential for TGF β/Smad activation and cardiac fibrosis induced by angiotensin II. PloS one. 2012;7(5):e35144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zhou J, Jiang K, Ding X, Fu M, Wang S, Zhu L, He T, Wang J, Sun A, Hu K, Chen L, Zou Y, Ge J. Qiliqiangxin inhibits angiotensin II-induced transdifferentiation of rat cardiac fibroblasts through suppressing interleukin-6. J Cell Mol Med. 2015;19(5):1114–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Duerrschmid C, Crawford JR, Reineke E, Taffet GE, Trial J, Entman ML, Haudek SB. TNF receptor 1 signaling is critically involved in mediating angiotensin-II-induced cardiac fibrosis. Journal of molecular and cellular cardiology. 2013;57:59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sriramula S, Francis J. Tumor Necrosis Factor - Alpha Is Essential for Angiotensin II-Induced Ventricular Remodeling: Role for Oxidative Stress. PloS one. 2015;10(9):e0138372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wang L, Li YL, Zhang CC, Cui W, Wang X, Xia Y, Du J, Li HH. Inhibition of Toll-like receptor 2 reduces cardiac fibrosis by attenuating macrophage-mediated inflammation. Cardiovascular research. 2014;101(3):383–392. [DOI] [PubMed] [Google Scholar]
- 85.Zhao W, Zhao T, Chen Y, Ahokas RA, Sun Y. Oxidative stress mediates cardiac fibrosis by enhancing transforming growth factor-beta1 in hypertensive rats. Mol Cell Biochem. 2008;317(1–2):43–50. [DOI] [PubMed] [Google Scholar]
- 86.Zhang Q, Tan Y, Zhang N, Yao F. Polydatin prevents angiotensin II-induced cardiac hypertrophy and myocardial superoxide generation. Exp Biol Med (Maywood). 2015;240(10):1352–1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhao QD, Viswanadhapalli S, Williams P, Shi Q, Tan C, Yi X, Bhandari B, Abboud HE. NADPH oxidase 4 induces cardiac fibrosis and hypertrophy through activating Akt/mTOR and NFκB signaling pathways. Circulation. 2015;131(7):643–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Colston JT, de la Rosa SD, Strader JR, Anderson MA, Freeman GL. H2O2 activates Nox4 through PLA2-dependent arachidonic acid production in adult cardiac fibroblasts. FEBS Lett. 2005;579(11):2533–2540. [DOI] [PubMed] [Google Scholar]
- 89.Bouabout G, Ayme-Dietrich E, Jacob H, Champy MF, Birling MC, Pavlovic G, Madeira L, Fertak LE, Petit-Demoulière B, Sorg T, Herault Y, Mudgett J, Monassier L. Nox4 genetic inhibition in experimental hypertension and metabolic syndrome. Arch Cardiovasc Dis. 2018;111(1):41–52. [DOI] [PubMed] [Google Scholar]
- 90.Somanna NK, Valente AJ, Krenz M, Fay WP, Delafontaine P, Chandrasekar B. The Nox1/4 Dual Inhibitor GKT137831 or Nox4 Knockdown Inhibits Angiotensin-II-Induced Adult Mouse Cardiac Fibroblast Proliferation and Migration. AT1 Physically Associates With Nox4. J Cell Physiol. 2016;231(5):1130–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Siddesha JM, Valente AJ, Sakamuri SS, Yoshida T, Gardner JD, Somanna N, Takahashi C, Noda M, Chandrasekar B. Angiotensin II stimulates cardiac fibroblast migration via the differential regulation of matrixins and RECK. Journal of molecular and cellular cardiology. 2013;65:9–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Meng G, Zhu J, Xiao Y, Huang Z, Zhang Y, Tang X, Xie L, Chen Y, Shao Y, Ferro A, Wang R, Moore PK, Ji Y. Hydrogen Sulfide Donor GYY4137 Protects against Myocardial Fibrosis. Oxid Med Cell Longev. 2015;2015:691070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Pan LL, Liu XH, Shen YQ, Wang NZ, Xu J, Wu D, Xiong QH, Deng HY, Huang GY, Zhu YZ. Inhibition of NADPH oxidase 4-related signaling by sodium hydrosulfide attenuates myocardial fibrotic response. Int J Cardiol. 2013;168(4):3770–3778. [DOI] [PubMed] [Google Scholar]
- 94.Larkin JE, Frank BC, Gaspard RM, Duka I, Gavras H, Quackenbush J. Cardiac transcriptional response to acute and chronic angiotensin II treatments. Physiol Genomics. 2004;18(2):152–166. [DOI] [PubMed] [Google Scholar]
- 95.Bai D, Ge L, Gao Y, Lu X, Wang H, Yang G. Cytoplasmic translocation of HuR contributes to angiotensin II induced cardiac fibrosis. Biochem Biophys Res Commun. 2015;463(4):1273–1277. [DOI] [PubMed] [Google Scholar]
- 96.Cheng TH, Leung YM, Cheung CW, Chen CH, Chen YL, Wong KL. Propofol depresses angiotensin II-induced cell proliferation in rat cardiac fibroblasts. Anesthesiology. 2010;112(1):108–118. [DOI] [PubMed] [Google Scholar]
- 97.Parra-Flores P, Riquelme JA, Valenzuela-Bustamante P, Leiva-Navarrete S, Vivar R, Cayupi-Vivanco J, Castro E, Espinoza-Pérez C, Ruz-Cortés F, Pedrozo Z, Lavandero S, Rodrigo R, Diaz-Araya G. The Association of Ascorbic Acid, Deferoxamine and N-Acetylcysteine Improves Cardiac Fibroblast Viability and Cellular Function Associated with Tissue Repair Damaged by Simulated Ischemia/Reperfusion. Antioxidants (Basel). 2019;8(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chen YL, Fan J, Cao L, Han TL, Zeng M, Xu Y, Ling Z, Yin Y. Unique mechanistic insights into the beneficial effects of angiotensin-(1–7) on the prevention of cardiac fibrosis: A metabolomic analysis of primary cardiac fibroblasts. Exp Cell Res. 2019;378(2):158–170. [DOI] [PubMed] [Google Scholar]
- 99.Mayorga M, Bahi N, Ballester M, Comella JX, Sanchis D. Bcl-2 is a key factor for cardiac fibroblast resistance to programmed cell death. Journal of Biological Chemistry. 2004;279(33):34882–34889. [DOI] [PubMed] [Google Scholar]
- 100.Cai W, Zhong S, Zheng F, Zhang Y, Gao F, Xu H, Cai X, Lan J, Huang D, Shi G. Angiotensin II confers resistance to apoptosis in cardiac myofibroblasts through the AT1/ERK1/2/RSK1 pathway. IUBMB Life. 2019;71(2):261–276. [DOI] [PubMed] [Google Scholar]
- 101.Mudersbach T, Siuda D, Kohlstedt K, Fleming I. Epigenetic control of the angiotensin-converting enzyme in endothelial cells during inflammation. PloS one. 2019;14(5):e0216218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bogdarina I, Welham S, King PJ, Burns SP, Clark AJ. Epigenetic modification of the renin-angiotensin system in the fetal programming of hypertension. Circulation research. 2007;100(4):520–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Duong TE, Hagood JS. Epigenetic Regulation of Myofibroblast Phenotypes in Fibrosis. Curr Pathobiol Rep. 2018;6(1):79–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Somanna NK, Valente AJ, Krenz M, McDonald KS, Higashi Y, Noda M, Chandrasekar B. Histone deacetyltransferase inhibitors Trichostatin A and Mocetinostat differentially regulate MMP9, IL-18 and RECK expression, and attenuate Angiotensin II-induced cardiac fibroblast migration and proliferation. Hypertension research : official journal of the Japanese Society of Hypertension. 2016;39(10):709–716. [DOI] [PubMed] [Google Scholar]
- 105.Lemon DD, Horn TR, Cavasin MA, Jeong MY, Haubold KW, Long CS, Irwin DC, McCune SA, Chung E, Leinwand LA, McKinsey TA. Cardiac HDAC6 catalytic activity is induced in response to chronic hypertension. Journal of molecular and cellular cardiology. 2011;51(1):41–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kee HJ, Sohn IS, Nam KI, Park JE, Qian YR, Yin Z, Ahn Y, Jeong MH, Bang YJ, Kim N, Kim JK, Kim KK, Epstein JA, Kook H. Inhibition of histone deacetylation blocks cardiac hypertrophy induced by angiotensin II infusion and aortic banding. Circulation. 2006;113(1):51–59. [DOI] [PubMed] [Google Scholar]
- 107.Williams SM, Golden-Mason L, Ferguson BS, Schuetze KB, Cavasin MA, Demos-Davies K, Yeager ME, Stenmark KR, McKinsey TA. Class I HDACs regulate angiotensin II-dependent cardiac fibrosis via fibroblasts and circulating fibrocytes. Journal of molecular and cellular cardiology. 2014;67:112–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Rosales W, Carulla J, García J, Vargas D, Lizcano F. Role of Histone Demethylases in Cardiomyocytes Induced to Hypertrophy. Biomed Res Int. 2016;2016:2634976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lee HA, Cho HM, Lee DY, Kim KC, Han HS, Kim IK. Tissue-specific upregulation of angiotensin-converting enzyme 1 in spontaneously hypertensive rats through histone code modifications. Hypertension (Dallas, Tex : 1979). 2012;59(3):621–626. [DOI] [PubMed] [Google Scholar]
- 110.Xiang FL, Fang M, Yutzey KE. Loss of β-catenin in resident cardiac fibroblasts attenuates fibrosis induced by pressure overload in mice. Nat Commun. 2017;8(1):712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Long F, Wang Q, Yang D, Zhu M, Wang J, Zhu Y, Liu X. Targeting JMJD3 histone demethylase mediates cardiac fibrosis and cardiac function following myocardial infarction. Biochemical and Biophysical Research Communications. 2020;528(4):671–677. [DOI] [PubMed] [Google Scholar]
- 112.Lombardi AA, Gibb AA, Arif E, Kolmetzky DW, Tomar D, Luongo TS, Jadiya P, Murray EK, Lorkiewicz PK, Hajnóczky G, Murphy E, Arany ZP, Kelly DP, Margulies KB, Hill BG, Elrod JW. Mitochondrial calcium exchange links metabolism with the epigenome to control cellular differentiation. Nat Commun. 2019;10(1):4509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.He Y, Ling S, Sun Y, Sheng Z, Chen Z, Pan X, Ma G. DNA methylation regulates α-smooth muscle actin expression during cardiac fibroblast differentiation. J Cell Physiol. 2019;234(5):7174–7185. [DOI] [PubMed] [Google Scholar]
- 114.Lin J, Lin S, Wu Y, Wang X, Wu S, Li H. Hypomethylation of the Angiotensin II Type I Receptor (AGTR1) Gene Along with Environmental Factors Increases the Risk for Essential Hypertension. Cardiology. 2017;137(2):126–135. [DOI] [PubMed] [Google Scholar]
- 115.Movassagh M, Choy MK, Goddard M, Bennett MR, Down TA, Foo RS. Differential DNA methylation correlates with differential expression of angiogenic factors in human heart failure. PloS one. 2010;5(1):e8564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Felisbino MB, McKinsey TA. Epigenetics in Cardiac Fibrosis. JACC: Basic to Translational Science. 2018;3(5):704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Russell-Hallinan A, Watson CJ, Baugh JA. Epigenetics of Aberrant Cardiac Wound Healing. Compr Physiol. 2018;8(2):451–491. [DOI] [PubMed] [Google Scholar]
- 118.Shu J, Liu Z, Jin L, Wang H. An RNA-sequencing study identifies candidate genes for angiotensin II-induced cardiac remodeling. Mol Med Rep. 2018;17(1):1954–1962. [DOI] [PubMed] [Google Scholar]
- 119.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.0(0). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Oosterhuis NR, Bongartz LG, Verhaar MC, Cheng C, Xu YJ, van Koppen A, Cramer MJ, Goldschmeding R, Gaillard CA, Doevendans PA, Braam B, Joles JA. Targeting multiple pathways reduces renal and cardiac fibrosis in rats with subtotal nephrectomy followed by coronary ligation. Acta physiologica (Oxford, England). 2017;220(3):382–393. [DOI] [PubMed] [Google Scholar]
- 121.Kretzschmar K, Post Y, Bannier-Hélaouët M, Mattiotti A, Drost J, Basak O, Li VSW, van den Born M, Gunst QD, Versteeg D, Kooijman L, van der Elst S, van Es JH, van Rooij E, van den Hoff MJB, Clevers H. Profiling proliferative cells and their progeny in damaged murine hearts. Proc Natl Acad Sci U S A. 2018;115(52):E12245–e12254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Forte E, Skelly DA, Chen M, Daigle S, Morelli KA, Hon O, Philip VM, Costa MW, Rosenthal NA, Furtado MB. Dynamic Interstitial Cell Response during Myocardial Infarction Predicts Resilience to Rupture in Genetically Diverse Mice. Cell Rep. 2020;30(9):3149–3163.e3146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gladka MM, Molenaar B, De Ruiter H, Van Der Elst S, Tsui H, Versteeg D, Lacraz GP, Huibers MM, Van Oudenaarden A, Van Rooij E. Single-cell sequencing of the healthy and diseased heart reveals cytoskeleton-associated protein 4 as a new modulator of fibroblasts activation. Circulation. 2018;138(2):166–180. [DOI] [PubMed] [Google Scholar]
- 124.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Weidenbach R, Schulz R, Gres P, Behrends M, Post H, Heusch G. Enhanced reduction of myocardial infarct size by combined ACE inhibition and AT(1)-receptor antagonism. British journal of pharmacology. 2000;131(1):138–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ito N, Ohishi M, Yamamoto K, Tatara Y, Shiota A, Hayashi N, Komai N, Yanagitani Y, Rakugi H, Ogihara T. Renin-angiotensin inhibition reverses advanced cardiac remodeling in aging spontaneously hypertensive rats. American journal of hypertension. 2007;20(7):792–799. [DOI] [PubMed] [Google Scholar]
- 127.Katoh M, Egashira K, Mitsui T, Chishima S, Takeshita A, Narita H. Angiotensin-converting enzyme inhibitor prevents plasminogen activator inhibitor-1 expression in a rat model with cardiovascular remodeling induced by chronic inhibition of nitric oxide synthesis. JMolCell Cardiol. 2000;32(1):73–83. [DOI] [PubMed] [Google Scholar]
- 128.Liang B, Leenen FHH. Prevention of salt induced hypertension and fibrosis by angiotensin converting enzyme inhibitors in Dahl S rats. British journal of pharmacology. 2007;152(6):903–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Peng H, Carretero OA, Vuljaj N, Liao TD, Motivala A, Peterson EL, Rhaleb NE. Angiotensin-converting enzyme inhibitors: a new mechanism of action. Circulation. 2005;112(16):2436–2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Stein M, Boulaksil M, Jansen JA, Herold E, Noorman M, Joles JA, van Veen TA, Houtman MJ, Engelen MA, Hauer RN, de Bakker JM, van Rijen HV. Reduction of fibrosis-related arrhythmias by chronic renin-angiotensin-aldosterone system inhibitors in an aged mouse model. AmJPhysiol Heart CircPhysiol. 2010;299(2):H310–H321. [DOI] [PubMed] [Google Scholar]
- 131.Brilla CG. Regression of myocardial fibrosis in hypertensive heart disease: diverse effects of various antihypertensive drugs. Cardiovascular research. 2000;46(2):324–331. [DOI] [PubMed] [Google Scholar]
- 132.Susic D, Varagic J, Frohlich ED. Pharmacologic agents on cardiovascular mass, coronary dynamics and collagen in aged spontaneously hypertensive rats. JHypertens. 1999;17(8):1209–1215. [DOI] [PubMed] [Google Scholar]
- 133.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, Sosnovik DE, Pittet MJ, Swirski FK, Weissleder R, Nahrendorf M. Angiotensin-converting enzyme inhibition prevents the release of monocytes from their splenic reservoir in mice with myocardial infarction. CircRes. 2010;107(11):1364–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Baumann M, Hermans JR, Janssen BJ, Peutz-Kootstra C, Witzke O, Heemann U, Smits JF, Boudier HAS. Transient prehypertensive treatment in spontaneously hypertensive rats: a comparison of spironolactone and losartan regarding long-term blood pressure and target organ damage. Journal of hypertension. 2007;25(12):2504–2511. [DOI] [PubMed] [Google Scholar]
- 135.Baumann M, Janssen BJ, Hermans JJ, Peutz-Kootstra C, Witzke O, Smits JF, Struijker Boudier HA. Transient AT1 receptor-inhibition in prehypertensive spontaneously hypertensive rats results in maintained cardiac protection until advanced age. Journal of hypertension. 2007;25(1):207–215. [DOI] [PubMed] [Google Scholar]
- 136.Peng F, Lin J, Lin L, Tang H. Transient prehypertensive treatment in spontaneously hypertensive rats: A comparison of losartan and amlodipine regarding long-term blood pressure, cardiac and renal protection. International journal of molecular medicine. 2012;30(6):1376–1386. [DOI] [PubMed] [Google Scholar]
- 137.Fornes P, Richer C, Vacher E, Bruneval P, Giudicelli J-F. Losartan’s protective effects in stroke-prone spontaneously hypertensive rats persist durably after treatment withdrawal. Journal of cardiovascular pharmacology. 1993;22(2):305–313. [DOI] [PubMed] [Google Scholar]
- 138.Yamada T, Horiuchi M, Dzau VJ. Angiotensin II type 2 receptor mediates programmed cell death. Proc Natl Acad Sci U S A. 1996;93(1):156–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Guo C, Wang Y, Liang H, Zhang J. ADAMTS-1 contributes to the antifibrotic effect of captopril by accelerating the degradation of type I collagen in chronic viral myocarditis. Eur J Pharmacol. 2010;629(1–3):104–110. [DOI] [PubMed] [Google Scholar]
- 140.Meng G, Wu F, Yang L, Zhu H, Gu J, He M, Xu J. Synergistic attenuation of myocardial fibrosis in spontaneously hypertensive rats by joint treatment with benazepril and candesartan. J CardiovascPharmacol. 2009;54(1):16–24. [DOI] [PubMed] [Google Scholar]
- 141.van Eickels M, Grohé C, Löbbert K, Stimpel M, Vetter H. Angiotensin converting enzyme inhibitors block mitogenic signalling pathways in rat cardiac fibroblasts. Naunyn Schmiedebergs Arch Pharmacol. 1999;359(5):394–399. [DOI] [PubMed] [Google Scholar]
- 142.Ahmed MS, Øie E, Vinge LE, Yndestad A, Øystein Andersen G, Andersson Y, Attramadal T, Attramadal H. Connective tissue growth factor--a novel mediator of angiotensin II-stimulated cardiac fibroblast activation in heart failure in rats. Journal of molecular and cellular cardiology. 2004;36(3):393–404. [DOI] [PubMed] [Google Scholar]
- 143.Rasoul S, Carretero OA, Peng H, Cavasin MA, Zhuo J, Sanchez-Mendoza A, Brigstock DR, Rhaleb NE. Antifibrotic effect of Ac-SDKP and angiotensin-converting enzyme inhibition in hypertension. Journal of hypertension. 2004;22(3):593–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Peng H, Carretero OA, Peterson EL, Rhaleb NE. Ac-SDKP inhibits transforming growth factor-beta1-induced differentiation of human cardiac fibroblasts into myofibroblasts. American journal of physiology Heart and circulatory physiology. 2010;298(5):H1357–1364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Pokharel S, Rasoul S, Roks AJ, van Leeuwen RE, van Luyn MJ, Deelman LE, Smits JF, Carretero O, van Gilst WH, Pinto YM. N-acetyl-Ser-Asp-Lys-Pro inhibits phosphorylation of Smad2 in cardiac fibroblasts. Hypertension (Dallas, Tex : 1979). 2002;40(2):155–161. [DOI] [PubMed] [Google Scholar]
- 146.Weber KT, Sun Y, Campbell SE. Structural remodelling of the heart by fibrous tissue: Role of circulating hormones and locally produced peptides. European Heart Journal. 1995;16(suppl N):12–18. [DOI] [PubMed] [Google Scholar]
- 147.Hubers SA, Brown NJ. Combined Angiotensin Receptor Antagonism and Neprilysin Inhibition. Circulation. 2016;133(11):1115–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Burke RM, Lighthouse JK, Mickelsen DM, Small EM. Sacubitril/Valsartan Decreases Cardiac Fibrosis in Left Ventricle Pressure Overload by Restoring PKG Signaling in Cardiac Fibroblasts. Circ Heart Fail. 2019;12(4):e005565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.deBlois D, Orlov SN, Hamet P. Apoptosis in cardiovascular remodeling--effect of medication. Cardiovasc Drugs Ther. 2001;15(6):539–545. [DOI] [PubMed] [Google Scholar]
- 150.Der Sarkissian S, Marchand EL, Duguay D, Hamet P, deBlois D. Reversal of interstitial fibroblast hyperplasia via apoptosis in hypertensive rat heart with valsartan or enalapril. Cardiovascular research. 2003;57(3):775–783. [DOI] [PubMed] [Google Scholar]
- 151.deBlois D, Tea BS, Beaudry D, Hamet P. Regulation of therapeutic apoptosis: a potential target in controlling hypertensive organ damage. Can J Physiol Pharmacol. 2005;83(1):29–41. [DOI] [PubMed] [Google Scholar]
- 152.Der Sarkissian S, Tea BS, Touyz RM, deBlois D, Hale TM. Role of angiotensin II type 2 receptor during regression of cardiac hypertrophy in spontaneously hypertensive rats. JAmSocHypertens. 2013;7(2):118–127. [DOI] [PubMed] [Google Scholar]
- 153.Duguay D, Pesant S, Deschepper CF, deBlois D. Fibroblast apoptosis precedes cardiomyocyte mass reduction during left ventricular remodeling in hypertensive rats treated with amlodipine. J Hypertens. 2007;25(6):1291–1299. [DOI] [PubMed] [Google Scholar]
- 154.Tea BS, Dam TV, Moreau P, Hamet P, deBlois D. Apoptosis during regression of cardiac hypertrophy in spontaneously hypertensive rats. Temporal regulation and spatial heterogeneity. Hypertension (Dallas, Tex : 1979). 1999;34(2):229–235. [DOI] [PubMed] [Google Scholar]
- 155.Akashiba A, Ono H, Ono Y, Ishimitsu T, Matsuoka H. Valsartan improves L-NAME-exacerbated cardiac fibrosis with TGF-beta inhibition and apoptosis induction in spontaneously hypertensive rats. JCardiol. 2008;52(3):239–246. [DOI] [PubMed] [Google Scholar]
- 156.Inomata K, Tanaka H. Protective effect of benidipine against sodium azide-induced cell death in cultured neonatal rat cardiac myocytes. Journal of pharmacological sciences. 2003;93(2):163–170. [DOI] [PubMed] [Google Scholar]
- 157.Amann K, Tyralla K, Gross ML, Schwarz U, Tornig J, Haas CS, Ritz E, Mall G. Cardiomyocyte loss in experimental renal failure: prevention by ramipril. Kidney international. 2003;63(5):1708–1713. [DOI] [PubMed] [Google Scholar]
- 158.Backlund T, Palojoki E, Saraste A, Gronholm T, Eriksson A, Lakkisto P, Vuolteenaho O, Nieminen MS, Voipio-Pulkki LM, Laine M, Tikkanen I. Effect of vasopeptidase inhibitor omapatrilat on cardiomyocyte apoptosis and ventricular remodeling in rat myocardial infarction. Cardiovascular research. 2003;57(3):727–737. [DOI] [PubMed] [Google Scholar]
- 159.Sun J, Xie J, Kang L, Ferro A, Dong L, Xu B. Amlodipine Ameliorates Ischemia-Induced Neovascularization in Diabetic Rats through Endothelial Progenitor Cell Mobilization. Biomed Res Int. 2016;2016:3182764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Galvez AS, Fiedler JL, Ocaranza MP, Jalil JE, Lavandero S, Diaz-Araya G. Perindopril regulates beta-agonist-induced cardiac apoptosis. J Cardiovasc Pharmacol. 2005;46(3):255–261. [DOI] [PubMed] [Google Scholar]
- 161.Cangiano E, Marchesini J, Campo G, Francolini G, Fortini C, Carra G, Miccoli M, Ceconi C, Tavazzi L, Ferrari R. ACE inhibition modulates endothelial apoptosis and renewal via endothelial progenitor cells in patients with acute coronary syndromes. American journal of cardiovascular drugs : drugs, devices, and other interventions. 2011;11(3):189–198. [DOI] [PubMed] [Google Scholar]
- 162.Monti M, Terzuoli E, Ziche M, Morbidelli L. The sulphydryl containing ACE inhibitor Zofenoprilat protects coronary endothelium from Doxorubicin-induced apoptosis. Pharmacological research. 2013;76:171–181. [DOI] [PubMed] [Google Scholar]
- 163.Deftereos S, Giannopoulos G, Kossyvakis C, Kaoukis A, Raisakis K, Driva M, Panagopoulou V, Lappos S, Rentoukas I, Pyrgakis V, Alpert MA. Effect of quinapril on in-stent restenosis and relation to plasma apoptosis signaling molecules. The American journal of cardiology. 2010;105(1):54–58. [DOI] [PubMed] [Google Scholar]
- 164.Giannopoulos G, Panagopoulou V, Deftereos S, Angelidis C, Davlouros P, Synetos A, Toutouzas K, Alexopoulos D, Cleman MW, Tousoulis D. Effect of Angiotensin converting enzyme inhibitors on soluble tumor-necrosis-factor-related apoptosis-inducing ligand levels - association with neointimal hyperplasia in drug eluting stents. Journal of interventional cardiology. 2014;27(6):582–590. [DOI] [PubMed] [Google Scholar]
- 165.Glezerman I, Patel H, Glicklich D, Croizat H, Devarajan P. Angiotensin-converting enzyme inhibition induces death receptor apoptotic pathways in erythroid precursors following renal transplantation. American journal of nephrology. 2003;23(4):195–201. [DOI] [PubMed] [Google Scholar]
- 166.De la Iglesia Inigo S, Lopez-Jorge CE, Gomez-Casares MT, Lemes Castellano A, Martin Cabrera P, Lopez Brito J, Suarez Cabrera A, Molero Labarta T. Induction of apoptosis in leukemic cell lines treated with captopril, trandolapril and losartan: a new role in the treatment of leukaemia for these agents. Leukemia research. 2009;33(6):810–816. [DOI] [PubMed] [Google Scholar]
- 167.Duguay D, deBlois D. Differential regulation of Akt, caspases and MAP kinases underlies smooth muscle cell apoptosis during aortic remodelling in SHR treated with amlodipine. British journal of pharmacology. 2007;151(8):1315–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Duguay D, Sarkissian SD, Kouz R, Ongali B, Couture R, deBlois D. Kinin B2 receptor is not involved in enalapril-induced apoptosis and regression of hypertrophy in spontaneously hypertensive rat aorta: possible role of B1 receptor. BrJPharmacol. 2004;141(4):728–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Min Y, Yang Z, Hong-Xia S, Du-Juan Y. Inhibitory Effects of Enalaprilat on Rat Cardiac Fibroblast Proliferation via ROS/P38MAPK/TGF-β1 Signaling Pathway. Molecules. 2012;17(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Biwer LA, Broderick TL, Xu H, Carroll C, Hale TM. Protection against L-NAME-induced reduction in cardiac output persists even after cessation of angiotensin-converting enzyme inhibitor treatment. Acta physiologica (Oxford, England). 2013;207(1):156–165. [DOI] [PubMed] [Google Scholar]
- 171.Hale TM, Bushfield TL, Woolard J, Pang JJ, Rees-Milton KJ, Adams MA. Changes critical to persistent lowering of arterial pressure in spontaneously hypertensive rat occur early in antihypertensive treatment. Journal of hypertension. 2011;29(1):113–122. [DOI] [PubMed] [Google Scholar]
- 172.Hale TM, Shoichet MJ, Bushfield TL, Adams MA. Time Course of Vascular Structural Changes During and After Short-Term Antihypertensive Treatment. Hypertension (Dallas, Tex : 1979). 2003;42:171–176. [DOI] [PubMed] [Google Scholar]
- 173.Harrap SB, Mirakian C, Datodi SR, Lever AF. Blood pressure and lifespan following brief ACE inhibitor treatment in young spontaneously hypertensive rats. Clin Exp Pharmacol Physiol. 1994;21(2):125–127. [DOI] [PubMed] [Google Scholar]
- 174.Harrap SB, Van d, Griffin SA, Macpherson F, Lever AF. Brief angiotensin converting enzyme inhibitor treatment in young spontaneously hypertensive rats reduces blood pressure long-term. Hypertension (Dallas, Tex : 1979). 1990;16(6):603–614. [DOI] [PubMed] [Google Scholar]
- 175.Hale TM. Persistent phenotypic shift in cardiac fibroblasts: impact of transient renin angiotensin system inhibition. Journal of molecular and cellular cardiology. 2016;93:125–132. [DOI] [PubMed] [Google Scholar]
- 176.Hale TM, Robertson SJ, Burns KD, deBlois D. Short-term ACE inhibition confers long-term protection against target organ damage. Hypertension research : official journal of the Japanese Society of Hypertension. 2012;35(6):604–610. [DOI] [PubMed] [Google Scholar]
- 177.D’Souza KM, Biwer LA, Madhavpeddi L, Ramaiah P, Shahid W, Hale TM. Persistent change in cardiac fibroblast physiology after transient ACE inhibition. American journal of physiology Heart and circulatory physiology. 2015;309(8):H1346–1353. [DOI] [PubMed] [Google Scholar]
- 178.Hirsch AT, Talsness CE, Schunkert H, Paul M, Dzau VJ. Tissue-specific activation of cardiac angiotensin converting enzyme in experimental heart failure. Circulation research. 1991;69(2):475–482. [DOI] [PubMed] [Google Scholar]
- 179.Reiss K, Capasso JM, Huang HE, Meggs LG, Li P, Anversa P. ANG II receptors, c-myc, and c-jun in myocytes after myocardial infarction and ventricular failure. Am J Physiol. 1993;264(3 Pt 2):H760–769. [DOI] [PubMed] [Google Scholar]
- 180.Nio Y, Matsubara H, Murasawa S, Kanasaki M, Inada M. Regulation of gene transcription of angiotensin II receptor subtypes in myocardial infarction. J Clin Invest. 1995;95(1):46–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Yamagishi H, Kim S, Nishikimi T, Takeuchi K, Takeda T. Contribution of cardiac renin-angiotensin system to ventricular remodelling in myocardial-infarcted rats. Journal of molecular and cellular cardiology. 1993;25(11):1369–1380. [DOI] [PubMed] [Google Scholar]
- 182.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(5):2273–2282. [DOI] [PubMed] [Google Scholar]
- 183.Pfeffer MA, Pfeffer JM, Steinberg C, Finn P. Survival after an experimental myocardial infarction: beneficial effects of long-term therapy with captopril. Circulation. 1985;72(2):406–412. [DOI] [PubMed] [Google Scholar]
- 184.Dean RG, Balding LC, Candido R, Burns WC, Cao Z, Twigg SM, Burrell LM. Connective tissue growth factor and cardiac fibrosis after myocardial infarction. J Histochem Cytochem. 2005;53(10):1245–1256. [DOI] [PubMed] [Google Scholar]
- 185.Gonzalez GE, Wilensky L, Cassaglia P, Morales C, Gelpi RJ. Early administration of Enalapril prevents diastolic dysfunction and ventricular remodeling in rabbits with myocardial infarction. Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology. 2016;25(3):208–213. [DOI] [PubMed] [Google Scholar]
- 186.Inthachai T, Lekawanvijit S, Kumfu S, Apaijai N, Pongkan W, Chattipakorn SC, Chattipakorn N. Dipeptidyl peptidase-4 inhibitor improves cardiac function by attenuating adverse cardiac remodelling in rats with chronic myocardial infarction. Exp Physiol. 2015;100(6):667–679. [DOI] [PubMed] [Google Scholar]
- 187.Kiss K, Fekete V, Pálóczi J, Sárközy M, Murlasits Z, Pipis J, Kheyfets IA, Dugina JL, Sergeeva SA, Epstein OI. Renin-Angiotensin-Aldosterone Signaling Inhibitors-Losartan, Enalapril, and Cardosten-Prevent Infarction-induced Heart Failure Development in Rats. Alternative Therapies in Health & Medicine. 2016;22(2). [PubMed] [Google Scholar]
- 188.Nguyen T, El Salibi E, Rouleau JL. Postinfarction survival and inducibility of ventricular arrhythmias in the spontaneously hypertensive rat : effects of ramipril and hydralazine. Circulation. 1998;98(19):2074–2080. [DOI] [PubMed] [Google Scholar]
- 189.Dixon IM, Ju H, Jassal DS, Peterson DJ. Effect of ramipril and losartan on collagen expression in right and left heart after myocardial infarction. Mol Cell Biochem. 1996;165(1):31–45. [DOI] [PubMed] [Google Scholar]
- 190.Raj P, Aloud BM, Louis XL, Yu L, Zieroth S, Netticadan T. Resveratrol is equipotent to perindopril in attenuating post-infarct cardiac remodeling and contractile dysfunction in rats. J Nutr Biochem. 2016;28:155–163. [DOI] [PubMed] [Google Scholar]
- 191.Qu Z, Xu H, Tian Y. Effects of angiotensin-converting enzyme inhibition and bradykinin peptides in rats with myocardial infarction. Int J Clin Exp Pathol. 2015;8(3):3410–3417. [PMC free article] [PubMed] [Google Scholar]
- 192.Fraccarollo D, Galuppo P, Motschenbacher S, Ruetten H, Schäfer A, Bauersachs J. Soluble guanylyl cyclase activation improves progressive cardiac remodeling and failure after myocardial infarction. Cardioprotection over ACE inhibition. Basic research in cardiology. 2014;109(4):421. [DOI] [PubMed] [Google Scholar]
- 193.Hayashi M, Tsutamoto T, Wada A, Tsutsui T, Ishii C, Ohno K, Fujii M, Taniguchi A, Hamatani T, Nozato Y, Kataoka K, Morigami N, Ohnishi M, Kinoshita M, Horie M. Immediate administration of mineralocorticoid receptor antagonist spironolactone prevents post-infarct left ventricular remodeling associated with suppression of a marker of myocardial collagen synthesis in patients with first anterior acute myocardial infarction. Circulation. 2003;107(20):2559–2565. [DOI] [PubMed] [Google Scholar]
- 194.De Carvalho Frimm C, Sun Y, Weber KT. Angiotensin II receptor blockade and myocardial fibrosis of the infarcted rat heart. J Lab Clin Med. 1997;129(4):439–446. [DOI] [PubMed] [Google Scholar]
- 195.Castiglioni L, Colazzo F, Fontana L, Colombo GI, Piacentini L, Bono E, Milano G, Paleari S, Palermo A, Guerrini U, Tremoli E, Sironi L. Evaluation of Left Ventricle Function by Regional Fractional Area Change (RFAC) in a Mouse Model of Myocardial Infarction Secondary to Valsartan Treatment. PloS one. 2015;10(8):e0135778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Chen B, Lu D, Fu Y, Zhang J, Huang X, Cao S, Xu D, Bin J, Kitakaze M, Huang Q, Liao Y. Olmesartan prevents cardiac rupture in mice with myocardial infarction by modulating growth differentiation factor 15 and p53. British journal of pharmacology. 2014;171(15):3741–3753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Maejima Y, Okada H, Haraguchi G, Onai Y, Kosuge H, Suzuki J, Isobe M. Telmisartan, a unique ARB, improves left ventricular remodeling of infarcted heart by activating PPAR gamma. Lab Invest. 2011;91(6):932–944. [DOI] [PubMed] [Google Scholar]
- 198.Taylor K, Patten RD, Smith JJ, Aronovitz MJ, Wight J, Salomon RN, Konstam MA. Divergent effects of angiotensin-converting enzyme inhibition and angiotensin II-receptor antagonism on myocardial cellular proliferation and collagen deposition after myocardial infarction in rats. J Cardiovasc Pharmacol. 1998;31(5):654–660. [DOI] [PubMed] [Google Scholar]
- 199.Yu CM, Tipoe GL, Wing-Hon Lai K, Lau CP. Effects of combination of angiotensin-converting enzyme inhibitor and angiotensin receptor antagonist on inflammatory cellular infiltration and myocardial interstitial fibrosis after acute myocardial infarction. J Am Coll Cardiol. 2001;38(4):1207–1215. [DOI] [PubMed] [Google Scholar]
- 200.von Lueder TG, Wang BH, Kompa AR, Huang L, Webb R, Jordaan P, Atar D, Krum H. Angiotensin receptor neprilysin inhibitor LCZ696 attenuates cardiac remodeling and dysfunction after myocardial infarction by reducing cardiac fibrosis and hypertrophy. Circ Heart Fail. 2015;8(1):71–78. [DOI] [PubMed] [Google Scholar]
- 201.Kompa AR, Lu J, Weller TJ, Kelly DJ, Krum H, von Lueder TG, Wang BH. Angiotensin receptor neprilysin inhibition provides superior cardioprotection compared to angiotensin converting enzyme inhibition after experimental myocardial infarction. Int J Cardiol. 2018;258:192–198. [DOI] [PubMed] [Google Scholar]
- 202.Badae NM, El Naggar AS, El Sayed SM. Is the cardioprotective effect of the ACE2 activator diminazene aceturate more potent than the ACE inhibitor enalapril on acute myocardial infarction in rats? Can J Physiol Pharmacol. 2019;97(7):638–646. [DOI] [PubMed] [Google Scholar]
- 203.Gómez-Mendoza DP, Marques FD, Melo-Braga MN, Sprenger RR, Sinisterra RD, Kjeldsen F, Santos RA, Verano-Braga T. Angiotensin-(1–7) oral treatment after experimental myocardial infarction leads to downregulation of CXCR4. J Proteomics. 2019;208:103486. [DOI] [PubMed] [Google Scholar]
- 204.Rodrigues EJ, Eisenberg MJ, Pilote L. Effects of early and late administration of angiotensin-converting enzyme inhibitors on mortality after myocardial infarction. Am J Med. 2003;115(6):473–479. [DOI] [PubMed] [Google Scholar]
- 205.Flather MD, Yusuf S, Køber L, Pfeffer M, Hall A, Murray G, Torp-Pedersen C, Ball S, Pogue J, Moyé L, Braunwald E. Long-term ACE-inhibitor therapy in patients with heart failure or left-ventricular dysfunction: a systematic overview of data from individual patients. ACE-Inhibitor Myocardial Infarction Collaborative Group. Lancet. 2000;355(9215):1575–1581. [DOI] [PubMed] [Google Scholar]
- 206.Cleland JG, Erhardt L, Murray G, Hall AS, Ball SG. Effect of ramipril on morbidity and mode of death among survivors of acute myocardial infarction with clinical evidence of heart failure. A report from the AIRE Study Investigators. Eur Heart J. 1997;18(1):41–51. [PubMed] [Google Scholar]
- 207.Hall AS, Murray GD, Ball SG. Follow-up study of patients randomly allocated ramipril or placebo for heart failure after acute myocardial infarction: AIRE Extension (AIREX) Study. Acute Infarction Ramipril Efficacy. Lancet. 1997;349(9064):1493–1497. [DOI] [PubMed] [Google Scholar]
- 208.Pfeffer MA, Braunwald E, Moyé LA, Basta L, Brown EJ Jr., Cuddy TE, Davis BR, Geltman EM, Goldman S, Flaker GC, et al. Effect of captopril on mortality and morbidity in patients with left ventricular dysfunction after myocardial infarction. Results of the survival and ventricular enlargement trial. The SAVE Investigators. N Engl J Med. 1992;327(10):669–677. [DOI] [PubMed] [Google Scholar]
- 209.Dickstein K, Kjekshus J. Effects of losartan and captopril on mortality and morbidity in high-risk patients after acute myocardial infarction: the OPTIMAAL randomised trial. Optimal Trial in Myocardial Infarction with Angiotensin II Antagonist Losartan. Lancet. 2002;360(9335):752–760. [DOI] [PubMed] [Google Scholar]
- 210.McMurray J, Solomon S, Pieper K, Reed S, Rouleau J, Velazquez E, White H, Howlett J, Swedberg K, Maggioni A, Køber L, Van de Werf F, Califf R, Pfeffer M. The effect of valsartan, captopril, or both on atherosclerotic events after acute myocardial infarction: an analysis of the Valsartan in Acute Myocardial Infarction Trial (VALIANT). J Am Coll Cardiol. 2006;47(4):726–733. [DOI] [PubMed] [Google Scholar]
- 211.Kessler EL, Rivaud MR, Vos MA, van Veen TAB. Sex-specific influence on cardiac structural remodeling and therapy in cardiovascular disease. Biol Sex Differ. 2019;10(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
