Skip to main content
Lippincott Open Access logoLink to Lippincott Open Access
. 2018 Apr 5;38(7):e108–e116. doi: 10.1161/ATVBAHA.118.311282

Renin-Angiotensin System and Cardiovascular Functions

Chia-Hua Wu 1,2, Shayan Mohammadmoradi 1,2, Jeff Z Chen 1,3, Hisashi Sawada 1, Alan Daugherty 1,2,3, Hong S Lu 1,2,3,
PMCID: PMC6039412  NIHMSID: NIHMS969029  PMID: 29950386

The renin-angiotensin system plays critical roles in maintaining normal cardiovascular functions and contributes to a spectrum of cardiovascular diseases. Classically, the renin-angiotensin system is composed of AGT (angiotensinogen), renin, angiotensin-converting enzyme (ACE), Ang II (angiotensin II), and 2 Ang II receptors (AT1 and AT2 receptors).1,2 AGT, a protein with 452 amino acids, is cleaved by renin to produce Ang I. Ang I is a decapeptide, which is then cleaved by ACE to produce Ang II. Ang II is an octapeptide, acting through binding to its receptors, AT1 and AT2 receptors. AT1 receptor is the major receptor for Ang II to regulate many physiological and pathophysiological functions.36 In mice, AT1 receptor has 2 subtypes, AT1a and AT1b, which have >90% sequence homology, but distinctive distributions and functions.4,712 AT1a receptor is important for blood pressure regulation and contributes to atherosclerosis and aortic aneurysms,5,13,14 whereas AT1b receptor has no evident contribution to these functions15 but is associated with vasculature contractility.16,17 AT2 receptor is abundant during fetal development but becomes low in most tissues after birth.18

In the past 2 decades, many new components in this system have been discovered. These include ACE2, a homologue of ACE, which converts Ang II to Ang(1–7) or converts Ang I to Ang(1–9).19,20 The G protein–coupled receptor Mas1 was identified as the receptor of Ang(1–7).21

This review highlights some recent publications in ATVB that have provided insights into understanding the classic components of the renin-angiotensin system and its alternative components contributing to cardiovascular functions. We will focus on effects of this hormonal system on cardiac dysfunction, hypertension, atherosclerosis, and aortic aneurysms.2229

Angiotensinogen

AGT is the only known substrate of the renin-angiotensin system to produce all downstream angiotensin peptides. AGT regulates blood pressure as demonstrated by multiple mouse models, including global AGT-deficient mouse model and human AGT and renin transgenic mouse model.3033 AGT was also implicated in atherosclerosis using a transgenic mouse model expressing both human angiotensinogen (Agt) and renin genes.34 Two recent studies have provided direct evidence that AGT regulates blood pressure and contributes to atherosclerosis through Ang II–mediated mechanisms.35,36 These studies used multiple genetic manipulations, including AGT hypomorphic mice, bone marrow transplantation, hepatocyte-specific AGT-deficient mouse model, and adeno-associated viral infection to repopulate the manipulated Agt in vivo. These studies demonstrate that hepatocyte-derived AGT is the predominant source to regulate blood pressure and promote atherosclerosis. A pharmacological approach using antisense oligoneucleotides has also opened a door to directly target AGT for preventing high blood pressure and atherosclerosis.36

Renin

Renin is the rate-limiting enzyme of the renin-angiotensin system and the only enzyme known to cleave AGT. These properties make renin a potentially attractive target to inhibit the renin-angiotensin cascade and improve Ang II–mediated cardiovascular dysfunctions.37,38 Inhibition of renin reduces blood pressure and atherosclerosis in animal models.6,36,3943 Unfortunately, renin inhibitors in patients with cardiovascular diseases have not provided superior beneficial effects beyond the well-established ACE inhibitors or AT1 receptor blockers.44

Despite some disappointing findings in human studies of renin inhibition, it has not discouraged research to understand renin-related mechanisms of cardiovascular diseases. The juxtaglomerular cells of the kidney are the major source of renin production and secretion. As an important organ in blood pressure regulation and cardiovascular functions, renal denervation aiming to reduce sympathetic nerve activity has drawn significant attention, although there are conflicting findings that need further research.4548 A recent study using pigs discovered that this approach reduced blood pressure and improved cardiovascular functions through its influence on kidney-brain-heart axis with profound changes of plasma renin activity, implicating the involvement of the renal renin-angiotensin system regulation in the process.49

Angiotensin-Converting Enzymes

In contrast to the rate-limiting and substrate-specific properties of renin, ACE is not sensitive to Ang II concentration changes, and it is an enzyme that cleaves not only Ang I but also many other substrates including bradykinin (a vasodilator) and N-acetyl-Ser-Asp-Lys-Pro (a hemoregulatory peptide).5053 There is a highly consistent literature demonstrating that ACE inhibition reduces blood pressure and atherosclerosis in animal models.6,54,55 ACE inhibitors are one major class for treatment of hypertension, cardiovascular dysfunctions, and diabetic nephropathy in patients.5660 Recent studies have also added new mechanistic insights into guiding the use of ACE inhibitors. It was found that high serum concentration of homocysteine decreased antihypertensive effect of enalapril, an ACE inhibitor, in chronic hypertensive patients.61

ACE is ubiquitously present in many cell types, tissues, and organs.62,63 Leukocyte or smooth muscle cell–derived ACE contributed to atherosclerosis as demonstrated by bone marrow transplantation and cell-specific depletion of ACE, respectively, in mouse models,54,64 although their effects were less potent than pharmacological inhibition of ACE systemically.6 ACE is abundant in endothelial cells.65 However, depletion of ACE in this cell type had no effects on atherosclerosis.64 Global genetic depletion or pharmacological inhibition of ACE reduced blood pressure,6,66 but depletion of ACE in leukocyte, endothelial cells, or smooth muscle cells did not affect blood pressure.54,64 Despite a well-known enzyme discovered half century ago67,68 with impressive success of its inhibitors in clinical patients,69 it is still a long road to define mechanisms by which ACE contributes to multiple cardiovascular functions, including its cellular source that influences blood pressure regulation.

Angiotensin II

As the major bioactive peptide of the renin-angiotensin system, there are broad views of mechanistic insights into understanding how Ang II contributes to multiple cardiovascular physiological and pathophysiological functions. We provide a brief review of the following diseases published recently in ATVB. For most of these studies, the approach used was chronic subcutaneous infusion of Ang II.70,71

Cardiac Dysfunction

Ang II induces several forms of cardiac dysfunction including hypertrophy, arrhythmia, and ventricle function failure.72,73 Basigin is a transmembrane glycoprotein that has multiple functions.74 In a mouse model of transverse aortic constriction, genetic reduction of basigin led to less cardiac hypertrophy, fibrosis, and heart failure.75 Deficiency of smooth muscle stromal interaction molecule 1, an endoplasmic reticulum Ca2+ sensor, also prevented Ang II–induced cardiac hypertrophy.76 These findings are consistent with that renin-angiotensin inhibition is crucial for improving cardiac dysfunction.

Hypertension

There are many factors contributing to hypertension.7779 Salt intake is believed to be a critical factor for high blood pressure.80 Ang II is also a well-recognized contributor to high blood pressure.81,82 However, high salt intake suppresses the renin-angiotensin system, whereas low dietary salt increases Ang II production.83,84 In accord with the paradox between salt intake and the renin-angiotensin regulation, dietary salt intake in blood pressure regulation and its consequent cardiovascular events have also been inconsistent, as reported in both human studies and animal models,8591 implicating complex molecular mechanisms involved in salt versus Ang II–mediated hypertension and related cardiovascular dysfunctions.

Batchu et al78 found that Axl, a receptor tyrosine kinase, in T lymphocytes exerted a significant role in Ang II–mediated blood pressure regulation. This finding is consistent with reports by Guzik et al92 and Norlander et al93 that T-lymphocyte–mediated immune response contributed to Ang II–induced high blood pressure, although this needs to be validated in human studies. In addition to immune cells, smooth muscle cells are a critical cell type in Ang II–mediated blood pressure regulation. Smooth muscle 22α is a cytoskeleton-associated protein in smooth muscle cells. Smooth muscle 22α deficiency in mice reduced Ang II–induced high blood pressure and senescence of vascular smooth muscle cells.93,94 These phenotypes were proposed to be associated with many mediators including p53-dependent pathway.95 Activation of the α7 subtype of nicotinic acetylcholine receptors (α7nAchR) inhibited Ang II–induced senescence in cultured vascular smooth muscle cells and wild-type mice, but not in mice with α7nAchR deficiency. This effect was associated with sirtuin 1 activity because inhibition of sirtuin 1 abrogated this effect.96 microRNA-143 and 145 are abundant in vascular smooth muscle cells and regulate myogenic tone.97 Depletion of these 2 microRNAs did not affect Ang II–induced high blood pressure but caused more severe arterial wall disruption, vascular remodeling, and inflammation.98 Another recent study identified cellular repressor of E1A-stimulated genes as a mediator of Ang II–induced vascular remodeling.99 From these recent studies, we can gather that Ang II–mediated hypertension is a complex process that involves a large spectrum of molecules and many cell types.

Atherosclerosis

Atherosclerosis is a complex disease involving diverse mechanisms including disordered lipoprotein metabolism, inflammation, endothelial dysfunction, reactive oxygen species, and endoplasmic reticulum stress.29,100103 Animal models are a common tool to study these mechanisms and exploring potential therapeutic targets. For example, application of drugs using nanoparticles holds promise to optimize drug delivery and efficacy. In apolipoprotein E–deficient (Apoe−/−) mice fed a high-fat diet and infused with Ang II, nanoparticles containing pioglitazone, an antidiabetic drug that also had peroxisome proliferator–activated receptor-γ agonistic effects, was injected intravenously on a weekly basis for 4 weeks. Although pioglitazone administration did not change atherosclerotic lesion size and macrophage content, it reduced Ly-6C high monocytes, matrix metalloproteinase activity, and cathepsin activity.104

In addition to mouse models, rabbits have been frequently used to study atherosclerosis. In one study, infusion of Ang II to Watanabe heritable hyperlipidemic rabbits led to high death rate (50% for Ang II 100 ng/kg per minute and 92% for Ang II 200 ng/kg per minute) because of acute myocardial infarction with coronary plaque erosion, rupture, and thrombosis.105 Because plaque rupture and thrombosis are high-risk complications in humans,106 this model would be optimal to study mechanisms related to the human disease. In another study, Honda et al107 infused Ang II to Japanese White rabbits when they were fed a high-cholesterol diet and injured using balloon catheter to femoral arteries. This procedure also led to atherothrombotic occlusions. Ezetimibe, a lipid-lowering drug used in patients, profoundly decreased this fatal pathology, providing rationale to determine its extended effects in patients.107

Thoracic Aortic Aneurysms

Thoracic aortic aneurysms (TAA) manifest as profound dilation of the thoracic aorta, accompanied by compromise of aortic wall integrity, dissection, or rupture.108112 Many genetic disorders are involved in this disease process including fibrillin-1,113,114 TGF (transforming growth factor)-β ligands and receptors,115120 smooth muscle cell–specific isoforms of α-actin (encoded by Acta2), and myosin heavy chain (encoded by Myh11).109 In addition to these genetic manipulations, infusion of Ang II also leads to TAA, predominantly localized to the ascending aortic region.121124

The aortic wall is composed of the intima, media, and adventitia. Among the cell types of the aorta, smooth muscle cells are the most abundant cell type and have been the most frequently studied cell type in the development of TAA. Vascular smooth muscle cell phenotypes are associated with aortic aneurysm formation and its pathological process.

Components of TGF-β signaling pathways are important for maintaining aortic wall integrity. However, its effects on TAA and abdominal aortic aneurysm (AAA) formation are controversial. Inhibition of TGF-β by neutralizing antibodies augmented aortic rupture rate and aortic dilation in both abdominal and thoracic aortic regions in Ang II–infused mice125127 but attenuated development of TAA in a Marfan mouse model.114 To explore the conflicting findings in different mouse models and different locations of aortic aneurysms, a recent study determined mechanisms of TGF-β signaling in Ang II–induced TAA and AAA, combined with smooth muscle cell–specific TGF-β receptor 2 deficiency.128 Systemic TGF-β neutralization augmented AAA but had no effects on TAA. In contrast, smooth muscle cell–specific TGF-β receptor 2 deficiency augmented TAA but had no apparent effects on the abdominal aorta.128 This study emphasizes the distinctive mechanisms between TAA and AAA.129

MicroRNA-21 was identified as a critical modulator of proliferation and apoptosis of smooth muscle cells during development of AAA. Overexpression of microRNA-21 reduced AAA, and inhibition of this microRNA augmented AAA in 2 common mouse models.130 A recent study discovered that in mice with Smad3 heterozygous background, aortic miR-21 expression was increased by Ang II infusion, and systemic microRNA-21 deletion exacerbated Ang II–induced TAA formation.131 This study, combined with studies using TGF-β receptor 2 genetically manipulated mice, provides evidence for the importance of TGF-β–mediated mechanisms in the development of TAA.

In addition to components that are important for maintaining the aortic wall structure and integrity, embryonic origins of smooth muscle cells determine their phenotypes and functions. Embryonic origins of smooth muscle cells in the aorta are complex.132 A recent study provided evidence that smooth muscle cells in the ascending aortic region were derived from 2 embryonic origins, with second heart field contributing to the outer layers and cardiac neural crest for the inner medial layers.133 This study adds new insights into understanding mechanisms of TAA from an evolutionary viewpoint.134

Besides critical roles of smooth muscle cells, inflammation is a feature of TAA. Contractile dysfunction in smooth muscle cells is present in aortas of patients with sporadic TAA and dissection and is associated with activation of NLRP3 (nucleotide oligomerization domain–like receptor family, pyrin domain containing 3)-caspase-1 inflammasome.135 A recent study reported that NLRP3 or caspase-1 deficiency in mice significantly reduced Ang II–induced contractile protein degradation and aortic aneurysm formation in both thoracic and abdominal aortic regions.135

Abdominal Aortic Aneurysms

AAA is defined as pathological dilation of the abdominal aorta. Same as individuals afflicted with TAA, aortic rupture is a fatal consequence of AAA.110,112,136,137 There are three commonly used mouse models to study AAA: perfusion of elastase into the infrarenal aorta,138 periaortic application of calcium chloride,139 or subcutaneous infusion of Ang II.70,140 Modifications of these mouse models have also provided mechanistic insights. For example, coadministration of β-aminopropionitrile with Ang II,141,142 coadministration of TGF-β–neutralizing antibody with Ang II125 or administration of TGF-β–neutralizing antibody to mice with elastase-induced AAA,25 or application of calcium chloride with phosphate-buffered saline onto the infrarenal aorta.143

Hypercholesterolemia augments Ang II–induced AAA.144,145 Therefore, Apoe/− mice and low-density lipoprotein receptor–deficient mice are the 2 commonly used mouse models for Ang II–induced AAA studies.70,71,140 Although Ang II–infused mouse model has become a popular model to study AAA, breeding mice to a hypercholesterolemic background has hampered its more broad use.146 A recent study provided a rapid approach for increasing plasma cholesterol and Ang II–induced AAA incidence in C57BL/6 mice by applying a gain-of-function mutation of mouse PCSK9 protein using an adeno-associated viral method,147 which was also frequently used in atherosclerosis studies.148150

Inflammation and extracellular matrix disruption and remodeling are important features of Ang II–induced AAA.112,145,151154 Publications describing Ang II–induced AAA were featured in a recent ATVB Highlights,112 including molecules that promote inflammation involving not only macrophages but also T and B lymphocytes,155164 oxidative stress,165167 and many other factors.112,145,168

In addition to extensive studies to define molecular mechanisms of AAA, some recent studies have emphasized the importance of studying sex differences.29,169171 One study used the 4 core mouse model to generate gonadal male mice with XX or XY chromosomes. This study found that gonadal male mice with an XY chromosome complement exhibited diffuse aortic aneurysms, whereas XX chromosome complement exhibited focal aortic dilation. Orchiectomy attenuated Ang II–induced TAA and AAA in male mice.172

Angiotensin II Receptors

AT1a Receptor

AT1a receptor, a subtype of Ang II receptor, is the major receptor for Ang II–mediated cardiovascular functions in mice. Global deficiency of AT1a receptor ablates atherosclerosis and attenuates Ang II–induced TAA and AAA.5,14,39,173,174 This effect was not attributed to the presence of AT1a receptor on leukocytes39,174 or smooth muscle cells,14,122 whereas endothelial cell–specific depletion of AT1a receptor had modest protective effects on Ang II–induced TAA but not AAA and atherosclerosis.14,122 In agreement with these previous studies, using a well-established Marfan mouse model with genetic disruption of fibrillin-1 expression, Galatioto et al175 found that endothelial cell–specific deletion, but not smooth muscle cell–specific deficiency, of AT1a receptor modestly attenuated TAA development and related aortic rupture.

AT2 Receptor

Although AT2 receptor remains low in most tissues and organs postnatally, many studies have reported increased presence of AT2 receptor under certain pathophysiological conditions as reviewed in a recent article.176 Genetic deletion of AT2 receptor in mice had no effects on general health and development177 but promoted angiogenesis within ischemic muscle.178 A diabetic mouse model with a spontaneous mutation in the insulin 2 gene (Ins2+/C96Y) was bred with AT2 receptor–deficient mouse model. Hindlimb ischemia was induced by ligating femoral artery. Depletion of AT2 receptor augmented blood flow reperfusion and collateral vessel formation that were associated with SH2 domain-containing phosphatase 1 activity and vascular endothelial growth factor action.179

Alternative Pathways

This section introduces an enzyme, a bioactive peptide, and a receptor beyond the classic renin–angiotensin components.

Angiotensin-Converting Enzyme 2

ACE2 prevents atherosclerosis and aortic aneurysms, as demonstrated by deficiency of ACE2 accelerating atherosclerosis and Ang II–induced AAA in hypercholesterolemic mice.180,181 Recently, Moran et al182 reported that ACE2 deficiency in Apoe/− mice augmented incidence of AAA and aortic rupture rate. Of note, deficiency of ACE2 also led to spontaneous AAA formation in the absence of Ang II. Resveratrol, a class of compounds produced by many plants, increased ACE2 and inhibited AAA growth in Ang II–infused mice.

Angiotensin (1–7) and Mas1

Recent studies have implicated that Ang(1–7) has protective effects on multiple cardiovascular functions through its interaction with Mas1.183 Many studies reported that Ang(1–7)/Mas1-mediated actions counteracted actions of Ang II.180,184186 For example, Ang(1–7) had vasodilation effect that was mediated by Mas1, whereas Ang II had potent vasoconstriction effect.187 One study reported that Ang(1–7)-induced NO-mediated vasodilation and increased telomerase activity of endothelial cells.187 In another study, low dose of Ang(1–7) increased angiogenesis and vasodilation through its interaction with Mas1, which had equivalent effects as same low dose of Ang II. Among potential mechanisms, ERK1/2 was essential for Ang(1–7)-induced angiogenesis and vasodilation.186,188

Summary

Although the major renin-angiotensin members were discovered more than a half century ago, this system still attracts a large number of research work in different fields. This implicates the importance of this hormonal system in physiological and pathophysiological functions but also notes that there are many unknowns and conundrums of this system in our knowledge that require more extensive research work.

Sources of Funding

Our research work was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award numbers R01HL133723 and R01HL139748 and the American Heart Association SFRN in Vascular Disease (18SFRN33960001). J.Z. Chen is supported by the National Center for Advancing Translational Sciences (UL1TR001998). H. Sawada is supported by an AHA postdoctoral fellowship (18POST33990468). The content in this article is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Disclosures

None.

Footnotes

*

These authors contributed equally to this article.

References

  • 1.Wu C, Lu H, Cassis LA, Daugherty A. Molecular and pathophysiological features of angiotensinogen: a mini review. N Am J Med Sci (Boston) 2011;4:183–190. doi: 10.7156/v4i4p183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lu H, Cassis LA, Kooi CW, Daugherty A. Structure and functions of angiotensinogen. Hypertens Res. 2016;39:492–500. doi: 10.1038/hr.2016.17. doi: 10.1038/hr.2016.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ito M, Oliverio MI, Mannon PJ, Best CF, Maeda N, Smithies O, Coffman TM. Regulation of blood pressure by the type 1A angiotensin II receptor gene. Proc Natl Acad Sci USA. 1995;92:3521–3525. doi: 10.1073/pnas.92.8.3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Oliverio MI, Best CF, Kim HS, Arendshorst WJ, Smithies O, Coffman TM. Angiotensin II responses in AT1A receptor-deficient mice: a role for AT1B receptors in blood pressure regulation. Am J Physiol. 1997;272(4)(pt 2):F515–F520. doi: 10.1152/ajprenal.1997.272.4.F515. doi: 10.1152/ajprenal.1997.272.4.F515. [DOI] [PubMed] [Google Scholar]
  • 5.Daugherty A, Rateri DL, Lu H, Inagami T, Cassis LA. Hypercholesterolemia stimulates angiotensin peptide synthesis and contributes to atherosclerosis through the AT1A receptor. Circulation. 2004;110:3849–3857. doi: 10.1161/01.CIR.0000150540.54220.C4. doi: 10.1161/01.CIR.0000150540.54220.C4. [DOI] [PubMed] [Google Scholar]
  • 6.Lu H, Balakrishnan A, Howatt DA, Wu C, Charnigo R, Liau G, Cassis LA, Daugherty A. Comparative effects of different modes of renin angiotensin system inhibition on hypercholesterolaemia-induced atherosclerosis. Br J Pharmacol. 2012;165:2000–2008. doi: 10.1111/j.1476-5381.2011.01712.x. doi: 10.1111/j.1476-5381.2011.01712.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Burson JM, Aguilera G, Gross KW, Sigmund CD. Differential expression of angiotensin receptor 1A and 1B in mouse. Am J Physiol. 1994;267(2)(pt 1):E260–E267. doi: 10.1152/ajpendo.1994.267.2.E260. doi: 10.1152/ajpendo.1994.267.2.E260. [DOI] [PubMed] [Google Scholar]
  • 8.Gasc JM, Shanmugam S, Sibony M, Corvol P. Tissue-specific expression of type 1 angiotensin II receptor subtypes. An in situ hybridization study. Hypertension. 1994;24:531–537. doi: 10.1161/01.hyp.24.5.531. [DOI] [PubMed] [Google Scholar]
  • 9.Sugaya T, Nishimatsu S, Tanimoto K, Takimoto E, Yamagishi T, Imamura K, Goto S, Imaizumi K, Hisada Y, Otsuka A. Angiotensin II type 1a receptor-deficient mice with hypotension and hyperreninemia. J Biol Chem. 1995;270:18719–18722. doi: 10.1074/jbc.270.32.18719. [DOI] [PubMed] [Google Scholar]
  • 10.Chen X, Li W, Yoshida H, Tsuchida S, Nishimura H, Takemoto F, Okubo S, Fogo A, Matsusaka T, Ichikawa I. Targeting deletion of angiotensin type 1B receptor gene in the mouse. Am J Physiol. 1997;272(3)(pt 2):F299–F304. doi: 10.1152/ajprenal.1997.272.3.F299. doi: 10.1152/ajprenal.1997.272.3.F299. [DOI] [PubMed] [Google Scholar]
  • 11.Zhu Z, Zhang SH, Wagner C, Kurtz A, Maeda N, Coffman T, Arendshorst WJ. Angiotensin AT1B receptor mediates calcium signaling in vascular smooth muscle cells of AT1A receptor-deficient mice. Hypertension. 1998;31:1171–1177. doi: 10.1161/01.hyp.31.5.1171. [DOI] [PubMed] [Google Scholar]
  • 12.Oliverio MI, Kim HS, Ito M, Le T, Audoly L, Best CF, Hiller S, Kluckman K, Maeda N, Smithies O, Coffman TM. Reduced growth, abnormal kidney structure, and type 2 (AT2) angiotensin receptor-mediated blood pressure regulation in mice lacking both AT1A and AT1B receptors for angiotensin II. Proc Natl Acad Sci USA. 1998;95:15496–15501. doi: 10.1073/pnas.95.26.15496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Crowley SD, Gurley SB, Coffman TM. AT(1) receptors and control of blood pressure: the kidney and more. Trends Cardiovasc Med. 2007;17:30–34. doi: 10.1016/j.tcm.2006.11.002. doi: 10.1016/j.tcm.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 14.Rateri DL, Moorleghen JJ, Balakrishnan A, Owens AP, III, Howatt DA, Subramanian V, Poduri A, Charnigo R, Cassis LA, Daugherty A. Endothelial cell-specific deficiency of Ang II type 1a receptors attenuates Ang II-induced ascending aortic aneurysms in LDL receptor-/- mice. Circ Res. 2011;108:574–581. doi: 10.1161/CIRCRESAHA.110.222844. doi: 10.1161/CIRCRESAHA.110.222844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Poduri A, Owens AP, III, Howatt DA, Moorleghen JJ, Balakrishnan A, Cassis LA, Daugherty A. Regional variation in aortic AT1b receptor mRNA abundance is associated with contractility but unrelated to atherosclerosis and aortic aneurysms. PLoS One. 2012;7:e48462. doi: 10.1371/journal.pone.0048462. doi: 10.1371/journal.pone.0048462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhou Y, Chen Y, Dirksen WP, Morris M, Periasamy M. AT1b receptor predominantly mediates contractions in major mouse blood vessels. Circ Res. 2003;93:1089–1094. doi: 10.1161/01.RES.0000101912.01071.FF. doi: 10.1161/01.RES.0000101912.01071.FF. [DOI] [PubMed] [Google Scholar]
  • 17.Owens AP, III, Subramanian V, Moorleghen JJ, Guo Z, McNamara CA, Cassis LA, Daugherty A. Angiotensin II induces a region-specific hyperplasia of the ascending aorta through regulation of inhibitor of differentiation 3. Circ Res. 2010;106:611–619. doi: 10.1161/CIRCRESAHA.109.212837. doi: 10.1161/CIRCRESAHA.109.212837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mukoyama M, Nakajima M, Horiuchi M, Sasamura H, Pratt RE, Dzau VJ. Expression cloning of type 2 angiotensin II receptor reveals a unique class of seven-transmembrane receptors. J Biol Chem. 1993;268:24539–24542. [PubMed] [Google Scholar]
  • 19.Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, Donovan M, Woolf B, Robison K, Jeyaseelan R, Breitbart RE, Acton S. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ Res. 2000;87:E1–E9. doi: 10.1161/01.res.87.5.e1. [DOI] [PubMed] [Google Scholar]
  • 20.Tipnis SR, Hooper NM, Hyde R, Karran E, Christie G, Turner AJ. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem. 2000;275:33238–33243. doi: 10.1074/jbc.M002615200. doi: 10.1074/jbc.M002615200. [DOI] [PubMed] [Google Scholar]
  • 21.Santos RA, Simoes e Silva AC, Maric C, et al. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci USA. 2003;100:8258–8263. doi: 10.1073/pnas.1432869100. doi: 10.1073/pnas.1432869100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Goldberg IJ. 2017 George Lyman Duff Memorial Lecture: fat in the blood, fat in the artery, fat in the heart: triglyceride in physiology and disease. Arterioscler Thromb Vasc Biol. 2018;38:700–706. doi: 10.1161/ATVBAHA.117.309666. doi: 10.1161/ATVBAHA.117.309666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ley K, Gerdes N, Winkels H. ATVB Distinguished Scientist Award: how costimulatory and coinhibitory pathways shape atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:764–777. doi: 10.1161/ATVBAHA.117.308611. doi: 10.1161/ATVBAHA.117.308611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fisher EA. Regression of atherosclerosis: the journey from the liver to the plaque and back. Arterioscler Thromb Vasc Biol. 2016;36:226–235. doi: 10.1161/ATVBAHA.115.301926. doi: 10.1161/ATVBAHA.115.301926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Raffort J, Lareyre F, Clément M, Hassen-Khodja R, Chinetti G, Mallat Z. Monocytes and macrophages in abdominal aortic aneurysm. Nat Rev Cardiol. 2017;14:457–471. doi: 10.1038/nrcardio.2017.52. doi: 10.1038/nrcardio.2017.52. [DOI] [PubMed] [Google Scholar]
  • 26.Stabley JN, Towler DA. Arterial calcification in diabetes mellitus: preclinical models and translational implications. Arterioscler Thromb Vasc Biol. 2017;37:205–217. doi: 10.1161/ATVBAHA.116.306258. doi: 10.1161/ATVBAHA.116.306258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani R. Pathology of human coronary and carotid artery atherosclerosis and vascular calcification in diabetes mellitus. Arterioscler Thromb Vasc Biol. 2017;37:191–204. doi: 10.1161/ATVBAHA.116.306256. doi: 10.1161/ATVBAHA.116.306256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ho CY, Shanahan CM. Medial arterial calcification: an overlooked player in peripheral arterial disease. Arterioscler Thromb Vasc Biol. 2016;36:1475–1482. doi: 10.1161/ATVBAHA.116.306717. doi: 10.1161/ATVBAHA.116.306717. [DOI] [PubMed] [Google Scholar]
  • 29.Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, III, Rosenfeld ME, Virmani R American Heart Association Council on Arteriosclerosis, Thrombosis and Vascular Biology; and Council on Basic Cardiovascular Sciences. Recommendation on design, execution, and reporting of animal atherosclerosis studies: a scientific statement from the American Heart Association. Arterioscler Thromb Vasc Biol. 2017;37:e131–e157. doi: 10.1161/ATV.0000000000000062. doi: 10.1161/ATV.0000000000000062. [DOI] [PubMed] [Google Scholar]
  • 30.Kim HS, Krege JH, Kluckman KD, Hagaman JR, Hodgin JB, Best CF, Jennette JC, Coffman TM, Maeda N, Smithies O. Genetic control of blood pressure and the angiotensinogen locus. Proc Natl Acad Sci USA. 1995;92:2735–2739. doi: 10.1073/pnas.92.7.2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tanimoto K, Sugiyama F, Goto Y, Ishida J, Takimoto E, Yagami K, Fukamizu A, Murakami K. Angiotensinogen-deficient mice with hypotension. J Biol Chem. 1994;269:31334–31337. [PubMed] [Google Scholar]
  • 32.Fukamizu A, Sugimura K, Takimoto E, Sugiyama F, Seo MS, Takahashi S, Hatae T, Kajiwara N, Yagami K, Murakami K. Chimeric renin-angiotensin system demonstrates sustained increase in blood pressure of transgenic mice carrying both human renin and human angiotensinogen genes. J Biol Chem. 1993;268:11617–11621. [PubMed] [Google Scholar]
  • 33.Merrill DC, Thompson MW, Carney CL, Granwehr BP, Schlager G, Robillard JE, Sigmund CD. Chronic hypertension and altered baroreflex responses in transgenic mice containing the human renin and human angiotensinogen genes. J Clin Invest. 1996;97:1047–1055. doi: 10.1172/JCI118497. doi: 10.1172/JCI118497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sugiyama F, Haraoka S, Watanabe T, Shiota N, Taniguchi K, Ueno Y, Tanimoto K, Murakami K, Fukamizu A, Yagami K. Acceleration of atherosclerotic lesions in transgenic mice with hypertension by the activated renin-angiotensin system. Lab Invest. 1997;76:835–842. [PubMed] [Google Scholar]
  • 35.Wu C, Xu Y, Lu H, Howatt DA, Balakrishnan A, Moorleghen JJ, Vander Kooi CW, Cassis LA, Wang JA, Daugherty A. Cys18-Cys137 disulfide bond in mouse angiotensinogen does not affect AngII-dependent functions in vivo. Hypertension. 2015;65:800–805. doi: 10.1161/HYPERTENSIONAHA.115.05166. doi: 10.1161/HYPERTENSIONAHA.115.05166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lu H, Wu C, Howatt DA, Balakrishnan A, Moorleghen JJ, Chen X, Zhao M, Graham MJ, Mullick AE, Crooke RM, Feldman DL, Cassis LA, Vander Kooi CW, Daugherty A. Angiotensinogen exerts effects independent of angiotensin II. Arterioscler Thromb Vasc Biol. 2016;36:256–265. doi: 10.1161/ATVBAHA.115.306740. doi: 10.1161/ATVBAHA.115.306740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Stanton A. Potential of renin inhibition in cardiovascular disease. J Renin Angiotensin Aldosterone Syst. 2003;4:6–10. doi: 10.3317/jraas.2003.008. doi: 10.3317/jraas.2003.008. [DOI] [PubMed] [Google Scholar]
  • 38.Wood JM, Maibaum J, Rahuel J, et al. Structure-based design of aliskiren, a novel orally effective renin inhibitor. Biochem Biophys Res Commun. 2003;308:698–705. doi: 10.1016/s0006-291x(03)01451-7. [DOI] [PubMed] [Google Scholar]
  • 39.Lu H, Rateri DL, Feldman DL, Charnigo RJ, Jr, Fukamizu A, Ishida J, Oesterling EG, Cassis LA, Daugherty A. Renin inhibition reduces hypercholesterolemia-induced atherosclerosis in mice. J Clin Invest. 2008;118:984–993. doi: 10.1172/JCI32970. doi: 10.1172/JCI32970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nussberger J, Aubert JF, Bouzourene K, Pellegrin M, Hayoz D, Mazzolai L. Renin inhibition by aliskiren prevents atherosclerosis progression: comparison with irbesartan, atenolol, and amlodipine. Hypertension. 2008;51:1306–1311. doi: 10.1161/HYPERTENSIONAHA.108.110932. doi: 10.1161/HYPERTENSIONAHA.108.110932. [DOI] [PubMed] [Google Scholar]
  • 41.Imanishi T, Tsujioka H, Ikejima H, Kuroi A, Takarada S, Kitabata H, Tanimoto T, Muragaki Y, Mochizuki S, Goto M, Yoshida K, Akasaka T. Renin inhibitor aliskiren improves impaired nitric oxide bioavailability and protects against atherosclerotic changes. Hypertension. 2008;52:563–572. doi: 10.1161/HYPERTENSIONAHA.108.111120. doi: 10.1161/HYPERTENSIONAHA.108.111120. [DOI] [PubMed] [Google Scholar]
  • 42.Kühnast S, van der Hoorn JW, van den Hoek AM, Havekes LM, Liau G, Jukema JW, Princen HM. Aliskiren inhibits atherosclerosis development and improves plaque stability in APOE*3Leiden.CETP transgenic mice with or without treatment with atorvastatin. J Hypertens. 2012;30:107–116. doi: 10.1097/HJH.0b013e32834ddd8e. doi: 10.1097/HJH.0b013e32834ddd8e. [DOI] [PubMed] [Google Scholar]
  • 43.Angeli F, Reboldi G, Poltronieri C, Angeli E, De Filippo V, Crocetti A, Bartolini C, D’Ambrosio C, Verdecchia P. Efficacy and safety profile of aliskiren: practical implications for clinicians. Curr Drug Saf. 2014;9:106–117. doi: 10.2174/1389450115666140211113040. [DOI] [PubMed] [Google Scholar]
  • 44.Sen S, Sabirli S, Ozyiğit T, Uresin Y. Aliskiren: review of efficacy and safety data with focus on past and recent clinical trials. Ther Adv Chronic Dis. 2013;4:232–241. doi: 10.1177/2040622313495288. doi: 10.1177/2040622313495288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Bhatt DL, Kandzari DE, O’Neill WW, D’Agostino R, Flack JM, Katzen BT, Leon MB, Liu M, Mauri L, Negoita M, Cohen SA, Oparil S, Rocha-Singh K, Townsend RR, Bakris GL SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med. 2014;370:1393–1401. doi: 10.1056/NEJMoa1402670. doi: 10.1056/NEJMoa1402670. [DOI] [PubMed] [Google Scholar]
  • 46.Böhm M, Mahfoud F, Ukena C, Hoppe UC, Narkiewicz K, Negoita M, Ruilope L, Schlaich MP, Schmieder RE, Whitbourn R, Williams B, Zeymer U, Zirlik A, Mancia G GSR Investigators. First report of the Global SYMPLICITY Registry on the effect of renal artery denervation in patients with uncontrolled hypertension. Hypertension. 2015;65:766–774. doi: 10.1161/HYPERTENSIONAHA.114.05010. doi: 10.1161/HYPERTENSIONAHA.114.05010. [DOI] [PubMed] [Google Scholar]
  • 47.Azizi M. Catheter-based renal denervation for treatment of hypertension. Lancet. 2017;390:2124–2126. doi: 10.1016/S0140-6736(17)32293-6. doi: 10.1016/S0140-6736(17)32293-6. [DOI] [PubMed] [Google Scholar]
  • 48.Mahfoud F, Bakris G, Bhatt DL, Esler M, Ewen S, Fahy M, Kandzari D, Kario K, Mancia G, Weber M, Böhm M. Reduced blood pressure-lowering effect of catheter-based renal denervation in patients with isolated systolic hypertension: data from SYMPLICITY HTN-3 and the global SYMPLICITY Registry. Eur Heart J. 2017;38:93–100. doi: 10.1093/eurheartj/ehw325. doi: 10.1093/eurheartj/ehw325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Uzuka H, Matsumoto Y, Nishimiya K, et al. Renal denervation suppresses coronary hyperconstricting responses after drug-eluting stent implantation in pigs invivothrough the kidney-brain-heart axis. Arterioscler Thromb Vasc Biol. 2017;37:1869–1880. doi: 10.1161/ATVBAHA.117.309777. doi: 10.1161/ATVBAHA.117.309777. [DOI] [PubMed] [Google Scholar]
  • 50.Erdös EG, Skidgel RA. The angiotensin I-converting enzyme. Lab Invest. 1987;56:345–348. [PubMed] [Google Scholar]
  • 51.Fuchs S, Xiao HD, Cole JM, Adams JW, Frenzel K, Michaud A, Zhao H, Keshelava G, Capecchi MR, Corvol P, Bernstein KE. Role of the N-terminal catalytic domain of angiotensin-converting enzyme investigated by targeted inactivation in mice. J Biol Chem. 2004;279:15946–15953. doi: 10.1074/jbc.M400149200. doi: 10.1074/jbc.M400149200. [DOI] [PubMed] [Google Scholar]
  • 52.Bernstein KE, Khan Z, Giani JF, Zhao T, Eriguchi M, Bernstein EA, Gonzalez-Villalobos RA, Shen XZ. Overexpression of angiotensin-converting enzyme in myelomonocytic cells enhances the immune response. F1000Res. 2016;5 doi: 10.12688/f1000research.7508.1. doi: 10.12688/f1000research.7508.1. eCollection 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bernstein KE, Khan Z, Giani JF, Cao DY, Bernstein EA, Shen XZ. Angiotensin-converting enzyme in innate and adaptive immunity. Nat Rev Nephrol. 2018;14:325–336. doi: 10.1038/nrneph.2018.15. doi: 10.1038/nrneph.2018.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chen X, Lu H, Zhao M, Tashiro K, Cassis LA, Daugherty A. Contributions of leukocyte angiotensin-converting enzyme to development of atherosclerosis. Arterioscler Thromb Vasc Biol. 2013;33:2075–2080. doi: 10.1161/ATVBAHA.113.301777. doi: 10.1161/ATVBAHA.113.301777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lu H, Cassis LA, Daugherty A. Atherosclerosis and arterial blood pressure in mice. Curr Drug Targets. 2007;8:1181–1189. doi: 10.2174/138945007782403829. [DOI] [PubMed] [Google Scholar]
  • 56.Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med. 2000;342:145–153. doi: 10.1056/NEJM200001203420301. [DOI] [PubMed] [Google Scholar]
  • 57.Svensson P, de Faire U, Sleight P, Yusuf S, Ostergren J. Comparative effects of ramipril on ambulatory and office blood pressures: a HOPE substudy. Hypertension. 2001;38:E28–E32. doi: 10.1161/hy1101.099502. [DOI] [PubMed] [Google Scholar]
  • 58.Gavish L, Rubinstein C, Bulut A, Berlatzky Y, Beeri R, Gilon D, Gavish L, Harlev M, Reissman P, Gertz SD. Low-level laser irradiation inhibits abdominal aortic aneurysm progression in apolipoprotein E-deficient mice. Cardiovasc Res. 2009;83:785–792. doi: 10.1093/cvr/cvp149. doi: 10.1093/cvr/cvp149. [DOI] [PubMed] [Google Scholar]
  • 59.Mann JF, Anderson C, Gao P, Gerstein HC, Boehm M, Rydén L, Sleight P, Teo KK, Yusuf S ONTARGET investigators. Dual inhibition of the renin-angiotensin system in high-risk diabetes and risk for stroke and other outcomes: results of the ONTARGET trial. J Hypertens. 2013;31:414–421. doi: 10.1097/HJH.0b013e32835bf7b0. doi: 10.1097/HJH.0b013e32835bf7b0. [DOI] [PubMed] [Google Scholar]
  • 60.Jin SM, Han KA, Yu JM, et al. Probucol in albuminuric type 2 diabetes mellitus patients on renin-angiotensin system blockade: a 16-week, randomized, double-blind, placebo-controlled trial. Arterioscler Thromb Vasc Biol. 2016;36:2108–2114. doi: 10.1161/ATVBAHA.116.308034. doi: 10.1161/ATVBAHA.116.308034. [DOI] [PubMed] [Google Scholar]
  • 61.Qin X, Li Y, Sun N, et al. Elevated homocysteine concentrations decrease the antihypertensive effect of angiotensin-converting enzyme inhibitors in hypertensive patients. Arterioscler Thromb Vasc Biol. 2017;37:166–172. doi: 10.1161/ATVBAHA.116.308515. doi: 10.1161/ATVBAHA.116.308515. [DOI] [PubMed] [Google Scholar]
  • 62.Dzau VJ, Bernstein K, Celermajer D, et al. Pathophysiologic and therapeutic importance of tissue ACE: a consensus report. Cardiovasc Drugs Ther. 2002;16:149–160. doi: 10.1023/a:1015709617405. [DOI] [PubMed] [Google Scholar]
  • 63.Shen XZ, Xiao HD, Li P, Billet S, Lin CX, Fuchs S, Bernstein KE. Tissue specific expression of angiotensin converting enzyme: a new way to study an old friend. Int Immunopharmacol. 2008;8:171–176. doi: 10.1016/j.intimp.2007.08.010. doi: 10.1016/j.intimp.2007.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen X, Howatt DA, Balakrishnan A, Moorleghen JJ, Wu C, Cassis LA, Daugherty A, Lu H. Angiotensin-converting enzyme in smooth muscle cells promotes atherosclerosis-brief report. Arterioscler Thromb Vasc Biol. 2016;36:1085–1089. doi: 10.1161/ATVBAHA.115.307038. doi: 10.1161/ATVBAHA.115.307038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ng KK, Vane JR. The conversion of angiotensin I to angiotensin II in vivo. Naunyn Schmiedebergs Arch Exp Pathol Pharmakol. 1968;259:188–189. doi: 10.1007/BF00537778. [DOI] [PubMed] [Google Scholar]
  • 66.Krege JH, John SW, Langenbach LL, Hodgin JB, Hagaman JR, Bachman ES, Jennette JC, O’Brien DA, Smithies O. Male-female differences in fertility and blood pressure in ACE-deficient mice. Nature. 1995;375:146–148. doi: 10.1038/375146a0. doi: 10.1038/375146a0. [DOI] [PubMed] [Google Scholar]
  • 67.Milei J. A cornerstone in the history of hypertension: the seventieth anniversary of the discovery of angiotensin. J Cardiovasc Med (Hagerstown) 2010;11:260–264. doi: 10.2459/JCM.0b013e3283356607. doi: 10.2459/JCM.0b013e3283356607. [DOI] [PubMed] [Google Scholar]
  • 68.Milei J, Provenzano S, Ambrosio G. 75th anniversary of the discovery of angiotensin: a tale of two countries. Eur Heart J. 2015;36:461–463. [PubMed] [Google Scholar]
  • 69.Yusuf S, Lonn E, Bosch J, Gerstein H. Summary of randomized trials of angiotensin converting enzyme inhibitors. Clin Exp Hypertens. 1999;21:835–845. doi: 10.3109/10641969909061013. [DOI] [PubMed] [Google Scholar]
  • 70.Daugherty A, Manning MW, Cassis LA. Angiotensin II promotes atherosclerotic lesions and aneurysms in apolipoprotein E-deficient mice. J Clin Invest. 2000;105:1605–1612. doi: 10.1172/JCI7818. doi: 10.1172/JCI7818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lu H, Howatt DA, Balakrishnan A, Moorleghen JJ, Rateri DL, Cassis LA, Daugherty A. Subcutaneous angiotensin II infusion using osmotic pumps induces aortic aneurysms in mice. JoVE. 2015;103:e53191. doi: 10.3791/53191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schlüter KD, Wenzel S. Angiotensin II: a hormone involved in and contributing to pro-hypertrophic cardiac networks and target of anti-hypertrophic cross-talks. Pharmacol Ther. 2008;119:311–325. doi: 10.1016/j.pharmthera.2008.05.010. doi: 10.1016/j.pharmthera.2008.05.010. [DOI] [PubMed] [Google Scholar]
  • 73.Tham YK, Bernardo BC, Ooi JY, Weeks KL, McMullen JR. Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets. Arch Toxicol. 2015;89:1401–1438. doi: 10.1007/s00204-015-1477-x. doi: 10.1007/s00204-015-1477-x. [DOI] [PubMed] [Google Scholar]
  • 74.Seizer P, Ungern-Sternberg SN, Schönberger T, Borst O, Münzer P, Schmidt EM, Mack AF, Heinzmann D, Chatterjee M, Langer H, Malešević M, Lang F, Gawaz M, Fischer G, May AE. Extracellular cyclophilin A activates platelets via EMMPRIN (CD147) and PI3K/Akt signaling, which promotes platelet adhesion and thrombus formation in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2015;35:655–663. doi: 10.1161/ATVBAHA.114.305112. doi: 10.1161/ATVBAHA.114.305112. [DOI] [PubMed] [Google Scholar]
  • 75.Suzuki K, Satoh K, Ikeda S, et al. Basigin promotes cardiac fibrosis and failure in response to chronic pressure overload in mice. Arterioscler Thromb Vasc Biol. 2016;36:636–646. doi: 10.1161/ATVBAHA.115.306686. doi: 10.1161/ATVBAHA.115.306686. [DOI] [PubMed] [Google Scholar]
  • 76.Kassan M, Ait-Aissa K, Radwan E, Mali V, Haddox S, Gabani M, Zhang W, Belmadani S, Irani K, Trebak M, Matrougui K. Essential role of smooth muscle STIM1 in hypertension and cardiovascular dysfunction. Arterioscler Thromb Vasc Biol. 2016;36:1900–1909. doi: 10.1161/ATVBAHA.116.307869. doi: 10.1161/ATVBAHA.116.307869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sun J, Canton G, Balu N, Hippe DS, Xu D, Liu J, Hatsukami TS, Yuan C. Blood pressure is a major modifiable risk factor implicated in pathogenesis of intraplaque hemorrhage: an invivomagnetic resonance imaging study. Arterioscler Thromb Vasc Biol. 2016;36:743–749. doi: 10.1161/ATVBAHA.115.307043. doi: 10.1161/ATVBAHA.115.307043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Batchu N, Hughson A, Wadosky KM, Morrell CN, Fowell DJ, Korshunov VA. Role of axl in T-lymphocyte survival in salt-dependent hypertension. Arterioscler Thromb Vasc Biol. 2016;36:1638–1646. doi: 10.1161/ATVBAHA.116.307848. doi: 10.1161/ATVBAHA.116.307848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chen TK, Appel LJ, Grams ME, Tin A, Choi MJ, Lipkowitz MS, Winkler CA, Estrella MM. APOL1 risk variants and cardiovascular disease: results from the AASK (African American study of kidney disease and hypertension). Arterioscler Thromb Vasc Biol. 2017;37:1765–1769. doi: 10.1161/ATVBAHA.117.309384. doi: 10.1161/ATVBAHA.117.309384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Meneton P, Jeunemaitre X, de Wardener HE, MacGregor GA. Links between dietary salt intake, renal salt handling, blood pressure, and cardiovascular diseases. Physiol Rev. 2005;85:679–715. doi: 10.1152/physrev.00056.2003. doi: 10.1152/physrev.00056.2003. [DOI] [PubMed] [Google Scholar]
  • 81.Lavoie JL, Sigmund CD. Minireview: overview of the renin-angiotensin system–an endocrine and paracrine system. Endocrinology. 2003;144:2179–2183. doi: 10.1210/en.2003-0150. doi: 10.1210/en.2003-0150. [DOI] [PubMed] [Google Scholar]
  • 82.Gao S, Cui X, Wang X, Burg MB, Dmitrieva NI. Cross-sectional positive association of serum lipids and blood pressure with serum sodium within the normal reference range of 135-145 mmol/L. Arterioscler Thromb Vasc Biol. 2017;37:598–606. doi: 10.1161/ATVBAHA.116.308413. doi: 10.1161/ATVBAHA.116.308413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ingert C, Grima M, Coquard C, Barthelmebs M, Imbs JL. Effects of dietary salt changes on renal renin-angiotensin system in rats. Am J Physiol Renal Physiol. 2002;283:F995–F1002. doi: 10.1152/ajprenal.00321.2001. doi: 10.1152/ajprenal.00321.2001. [DOI] [PubMed] [Google Scholar]
  • 84.Carillo BA, Beutel A, Mirandola DA, Vidonho AF, Jr, Furukawa LN, Casarini D, Campos RR, Dolnikoff MS, Heimann JC, Bergamaschi CT. Differential sympathetic and angiotensinergic responses in rats submitted to low- or high-salt diet. Regul Pept. 2007;140:5–11. doi: 10.1016/j.regpep.2006.11.007. doi: 10.1016/j.regpep.2006.11.007. [DOI] [PubMed] [Google Scholar]
  • 85.Ketonen J, Merasto S, Paakkari I, Mervaala EM. High sodium intake increases vascular superoxide formation and promotes atherosclerosis in apolipoprotein E-deficient mice. Blood Press. 2005;14:373–382. doi: 10.1080/08037050500383687. doi: 10.1080/08037050500383687. [DOI] [PubMed] [Google Scholar]
  • 86.Catanozi S, Rocha JC, Passarelli M, Guzzo ML, Alves C, Furukawa LN, Nunes VS, Nakandakare ER, Heimann JC, Quintão EC. Dietary sodium chloride restriction enhances aortic wall lipid storage and raises plasma lipid concentration in LDL receptor knockout mice. J Lipid Res. 2003;44:727–732. doi: 10.1194/jlr.M200330-JLR200. doi: 10.1194/jlr.M200330-JLR200. [DOI] [PubMed] [Google Scholar]
  • 87.Ivanovski O, Szumilak D, Nguyen-Khoa T, Dechaux M, Massy ZA, Phan O, Mothu N, Lacour B, Drueke TB, Muntzel M. Dietary salt restriction accelerates atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis. 2005;180:271–276. doi: 10.1016/j.atherosclerosis.2004.12.020. doi: 10.1016/j.atherosclerosis.2004.12.020. [DOI] [PubMed] [Google Scholar]
  • 88.Stolarz-Skrzypek K, Kuznetsova T, Thijs L, Tikhonoff V, Seidlerová J, Richart T, Jin Y, Olszanecka A, Malyutina S, Casiglia E, Filipovský J, Kawecka-Jaszcz K, Nikitin Y, Staessen JA European Project on Genes in Hypertension (EPOGH) Investigators. Fatal and nonfatal outcomes, incidence of hypertension, and blood pressure changes in relation to urinary sodium excretion. JAMA. 2011;305:1777–1785. doi: 10.1001/jama.2011.574. doi: 10.1001/jama.2011.574. [DOI] [PubMed] [Google Scholar]
  • 89.Nakandakare ER, Charf AM, Santos FC, Nunes VS, Ortega K, Lottenberg AM, Mion D, Jr, Nakano T, Nakajima K, D’Amico EA, Catanozi S, Passarelli M, Quintão EC. Dietary salt restriction increases plasma lipoprotein and inflammatory marker concentrations in hypertensive patients. Atherosclerosis. 2008;200:410–416. doi: 10.1016/j.atherosclerosis.2007.12.034. doi: 10.1016/j.atherosclerosis.2007.12.034. [DOI] [PubMed] [Google Scholar]
  • 90.Alderman MH, Cohen H, Madhavan S. Dietary sodium intake and mortality: the National health and nutrition examination survey (NHANES I). Lancet. 1998;351:781–785. doi: 10.1016/S0140-6736(97)09092-2. doi: 10.1016/S0140-6736(97)09092-2. [DOI] [PubMed] [Google Scholar]
  • 91.Lu H, Wu C, Howatt DA, Balakrishnan A, Charnigo RJ, Jr, Cassis LA, Daugherty A. Differential effects of dietary sodium intake on blood pressure and atherosclerosis in hypercholesterolemic mice. J Nutr Biochem. 2013;24:49–53. doi: 10.1016/j.jnutbio.2012.03.001. doi: 10.1016/j.jnutbio.2012.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med. 2007;204:2449–2460. doi: 10.1084/jem.20070657. doi: 10.1084/jem.20070657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Norlander AE, Saleh MA, Pandey AK, Itani HA, Wu J, Xiao L, Kang J, Dale BL, Goleva SB, Laroumanie F, Du L, Harrison DG, Madhur MS. A salt-sensing kinase in T lymphocytes, SGK1, drives hypertension and hypertensive end-organ damage. JCI Insight. 2017;2 doi: 10.1172/jci.insight.92801. doi: 10.1172/jci.insight.92801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kunieda T, Minamino T, Nishi J, Tateno K, Oyama T, Katsuno T, Miyauchi H, Orimo M, Okada S, Takamura M, Nagai T, Kaneko S, Komuro I. Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation. 2006;114:953–960. doi: 10.1161/CIRCULATIONAHA.106.626606. doi: 10.1161/CIRCULATIONAHA.106.626606. [DOI] [PubMed] [Google Scholar]
  • 95.Miao SB, Xie XL, Yin YJ, Zhao LL, Zhang F, Shu YN, Chen R, Chen P, Dong LH, Lin YL, Lv P, Zhang DD, Nie X, Xue ZY, Han M. Accumulation of smooth muscle 22α protein accelerates senescence of vascular smooth muscle cells via stabilization of p53 in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2017;37:1849–1859. doi: 10.1161/ATVBAHA.117.309378. doi: 10.1161/ATVBAHA.117.309378. [DOI] [PubMed] [Google Scholar]
  • 96.Li DJ, Huang F, Ni M, Fu H, Zhang LS, Shen FM. α7 Nicotinic acetylcholine receptor relieves angiotensin II-induced senescence in vascular smooth muscle cells by raising nicotinamide adenine dinucleotide-dependent SIRT1 activity. Arterioscler Thromb Vasc Biol. 2016;36:1566–1576. doi: 10.1161/ATVBAHA.116.307157. doi: 10.1161/ATVBAHA.116.307157. [DOI] [PubMed] [Google Scholar]
  • 97.Boettger T, Beetz N, Kostin S, Schneider J, Krüger M, Hein L, Braun T. Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. J Clin Invest. 2009;119:2634–2647. doi: 10.1172/JCI38864. doi: 10.1172/JCI38864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Holmberg J, Bhattachariya A, Alajbegovic A, Rippe C, Ekman M, Dahan D, Hien TT, Boettger T, Braun T, Swärd K, Hellstrand P, Albinsson S. Loss of vascular myogenic tone in miR-143/145 knockout mice is associated with hypertension-induced vascular lesions in small mesenteric arteries. Arterioscler Thromb Vasc Biol. 2018;38:414–424. doi: 10.1161/ATVBAHA.117.310499. doi: 10.1161/ATVBAHA.117.310499. [DOI] [PubMed] [Google Scholar]
  • 99.Li Y, Liu Y, Tian X, Zhang Y, Song H, Liu M, Zhang X, Liu H, Zhang J, Zhang Q, Liu D, Peng C, Yan C, Han Y. Cellular repressor of E1A-stimulated genes is a critical determinant of vascular remodeling in response to angiotensin II. Arterioscler Thromb Vasc Biol. 2017;37:485–494. doi: 10.1161/ATVBAHA.116.308794. doi: 10.1161/ATVBAHA.116.308794. [DOI] [PubMed] [Google Scholar]
  • 100.Tall AR, Yvan-Charvet L, Westerterp M, Murphy AJ. Cholesterol efflux: a novel regulator of myelopoiesis and atherogenesis. Arterioscler Thromb Vasc Biol. 2012;32:2547–2552. doi: 10.1161/ATVBAHA.112.300134. doi: 10.1161/ATVBAHA.112.300134. [DOI] [PubMed] [Google Scholar]
  • 101.Libby P. Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32:2045–2051. doi: 10.1161/ATVBAHA.108.179705. doi: 10.1161/ATVBAHA.108.179705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ait-Oufella H, Taleb S, Mallat Z, Tedgui A. Recent advances on the role of cytokines in atherosclerosis. Arterioscler Thromb Vasc Biol. 2011;31:969–979. doi: 10.1161/ATVBAHA.110.207415. doi: 10.1161/ATVBAHA.110.207415. [DOI] [PubMed] [Google Scholar]
  • 103.Scull CM, Tabas I. Mechanisms of ER stress-induced apoptosis in atherosclerosis. Arterioscler Thromb Vasc Biol. 2011;31:2792–2797. doi: 10.1161/ATVBAHA.111.224881. doi: 10.1161/ATVBAHA.111.224881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Nakashiro S, Matoba T, Umezu R, Koga J, Tokutome M, Katsuki S, Nakano K, Sunagawa K, Egashira K. Pioglitazone-incorporated nanoparticles prevent plaque destabilization and rupture by regulating monocyte/macrophage differentiation in ApoE-/- mice. Arterioscler Thromb Vasc Biol. 2016;36:491–500. doi: 10.1161/ATVBAHA.115.307057. doi: 10.1161/ATVBAHA.115.307057. [DOI] [PubMed] [Google Scholar]
  • 105.Li S, Wang YN, Niimi M, Ning B, Chen Y, Kang D, Wang Z, Yu Q, Waqar AB, Liu E, Zhang J, Shiomi M, Chen YE, Fan J. Angiotensin II destabilizes coronary plaques in watanabe heritable hyperlipidemic rabbits. Arterioscler Thromb Vasc Biol. 2016;36:810–816. doi: 10.1161/ATVBAHA.115.306871. doi: 10.1161/ATVBAHA.115.306871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rader DJ, Daugherty A. Translating molecular discoveries into new therapies for atherosclerosis. Nature. 2008;451:904–913. doi: 10.1038/nature06796. doi: 10.1038/nature06796. [DOI] [PubMed] [Google Scholar]
  • 107.Honda K, Matoba T, Antoku Y, Koga JI, Ichi I, Nakano K, Tsutsui H, Egashira K. Lipid-lowering therapy with ezetimibe decreases spontaneous atherothrombotic occlusions in a rabbit model of plaque erosion: a role of serum oxysterols. Arterioscler Thromb Vasc Biol. 2018;38:757–771. doi: 10.1161/ATVBAHA.117.310244. doi: 10.1161/ATVBAHA.117.310244. [DOI] [PubMed] [Google Scholar]
  • 108.Lu H, Rateri DL, Bruemmer D, Cassis LA, Daugherty A. Involvement of the renin-angiotensin system in abdominal and thoracic aortic aneurysms. Clin Sci (Lond) 2012;123:531–543. doi: 10.1042/CS20120097. doi: 10.1042/CS20120097. [DOI] [PubMed] [Google Scholar]
  • 109.Milewicz DM, Trybus KM, Guo DC, Sweeney HL, Regalado E, Kamm K, Stull JT. Altered smooth muscle cell force generation as a driver of thoracic aortic aneurysms and dissections. Arterioscler Thromb Vasc Biol. 2017;37:26–34. doi: 10.1161/ATVBAHA.116.303229. doi: 10.1161/ATVBAHA.116.303229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mallat Z, Tedgui A, Henrion D. Role of microvascular tone and extracellular matrix contraction in the regulation of interstitial fluid: implications for aortic dissection. Arterioscler Thromb Vasc Biol. 2016;36:1742–1747. doi: 10.1161/ATVBAHA.116.307909. doi: 10.1161/ATVBAHA.116.307909. [DOI] [PubMed] [Google Scholar]
  • 111.Zhang J, Zhao X, Vatner DE, McNulty T, Bishop S, Sun Z, Shen YT, Chen L, Meininger GA, Vatner SF. Extracellular matrix disarray as a mechanism for greater abdominal versus thoracic aortic stiffness with aging in primates. Arterioscler Thromb Vasc Biol. 2016;36:700–706. doi: 10.1161/ATVBAHA.115.306563. doi: 10.1161/ATVBAHA.115.306563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Lu H, Daugherty A. Aortic aneurysms. Arterioscler Thromb Vasc Biol. 2017;37:e59–e65. doi: 10.1161/ATVBAHA.117.309578. doi: 10.1161/ATVBAHA.117.309578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Pereira L, Lee SY, Gayraud B, Andrikopoulos K, Shapiro SD, Bunton T, Biery NJ, Dietz HC, Sakai LY, Ramirez F. Pathogenetic sequence for aneurysm revealed in mice underexpressing fibrillin-1. Proc Natl Acad Sci USA. 1999;96:3819–3823. doi: 10.1073/pnas.96.7.3819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Habashi JP, Judge DP, Holm TM, et al. Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse model of Marfan syndrome. Science. 2006;312:117–121. doi: 10.1126/science.1124287. doi: 10.1126/science.1124287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mizuguchi T, Collod-Beroud G, Akiyama T, et al. Heterozygous TGFBR2 mutations in Marfan syndrome. Nat Genet. 2004;36:855–860. doi: 10.1038/ng1392. doi: 10.1038/ng1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Loeys BL, Schwarze U, Holm T, et al. Aneurysm syndromes caused by mutations in the TGF-beta receptor. N Engl J Med. 2006;355:788–798. doi: 10.1056/NEJMoa055695. doi: 10.1056/NEJMoa055695. [DOI] [PubMed] [Google Scholar]
  • 117.Lindsay ME, Dietz HC. Lessons on the pathogenesis of aneurysm from heritable conditions. Nature. 2011;473:308–316. doi: 10.1038/nature10145. doi: 10.1038/nature10145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Pannu H, Fadulu VT, Chang J, Lafont A, Hasham SN, Sparks E, Giampietro PF, Zaleski C, Estrera AL, Safi HJ, Shete S, Willing MC, Raman CS, Milewicz DM. Mutations in transforming growth factor-beta receptor type II cause familial thoracic aortic aneurysms and dissections. Circulation. 2005;112:513–520. doi: 10.1161/CIRCULATIONAHA.105.537340. doi: 10.1161/CIRCULATIONAHA.105.537340. [DOI] [PubMed] [Google Scholar]
  • 119.Boileau C, Guo DC, Hanna N, et al. National Heart, Lung, and Blood Institute (NHLBI) Go Exome Sequencing Project. TGFB2 mutations cause familial thoracic aortic aneurysms and dissections associated with mild systemic features of Marfan syndrome. Nat Genet. 2012;44:916–921. doi: 10.1038/ng.2348. doi: 10.1038/ng.2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bertoli-Avella AM, Gillis E, Morisaki H, et al. Mutations in a TGF-β ligand, TGFB3, cause syndromic aortic aneurysms and dissections. J Am Coll Cardiol. 2015;65:1324–1336. doi: 10.1016/j.jacc.2015.01.040. doi: 10.1016/j.jacc.2015.01.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Daugherty A, Rateri DL, Charo IF, Owens AP, Howatt DA, Cassis LA. Angiotensin II infusion promotes ascending aortic aneurysms: attenuation by CCR2 deficiency in apoE-/- mice. Clin Sci (Lond) 2010;118:681–689. doi: 10.1042/CS20090372. doi: 10.1042/CS20090372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Rateri DL, Moorleghen JJ, Knight V, Balakrishnan A, Howatt DA, Cassis LA, Daugherty A. Depletion of endothelial or smooth muscle cell-specific angiotensin II type 1a receptors does not influence aortic aneurysms or atherosclerosis in LDL receptor deficient mice. PLoS One. 2012;7:e51483. doi: 10.1371/journal.pone.0051483. doi: 10.1371/journal.pone.0051483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Rateri DL, Davis FM, Balakrishnan A, Howatt DA, Moorleghen JJ, O’Connor WN, Charnigo R, Cassis LA, Daugherty A. Angiotensin II induces region-specific medial disruption during evolution of ascending aortic aneurysms. Am J Pathol. 2014;184:2586–2595. doi: 10.1016/j.ajpath.2014.05.014. doi: 10.1016/j.ajpath.2014.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Trachet B, Piersigilli A, Fraga-Silva RA, Aslanidou L, Sordet-Dessimoz J, Astolfo A, Stampanoni MF, Segers P, Stergiopulos N. Ascending aortic aneurysm in angiotensin II-infused mice: formation, progression, and the role of focal dissections. Arterioscler Thromb Vasc Biol. 2016;36:673–681. doi: 10.1161/ATVBAHA.116.307211. doi: 10.1161/ATVBAHA.116.307211. [DOI] [PubMed] [Google Scholar]
  • 125.Wang Y, Ait-Oufella H, Herbin O, Bonnin P, Ramkhelawon B, Taleb S, Huang J, Offenstadt G, Combadière C, Rénia L, Johnson JL, Tharaux PL, Tedgui A, Mallat Z. TGF-beta activity protects against inflammatory aortic aneurysm progression and complications in angiotensin II-infused mice. J Clin Invest. 2010;120:422–432. doi: 10.1172/JCI38136. doi: 10.1172/JCI38136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Chen X, Rateri DL, Howatt DA, Balakrishnan A, Moorleghen JJ, Cassis LA, Daugherty A. TGF-β neutralization enhances angII-induced aortic rupture and aneurysm in both thoracic and abdominal regions. PLoS One. 2016;11:e0153811. doi: 10.1371/journal.pone.0153811. doi: 10.1371/journal.pone.0153811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chen X, Lu H, Rateri DL, Cassis LA, Daugherty A. Conundrum of angiotensin II and TGF-β interactions in aortic aneurysms. Curr Opin Pharmacol. 2013;13:180–185. doi: 10.1016/j.coph.2013.01.002. doi: 10.1016/j.coph.2013.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Angelov SN, Hu JH, Wei H, Airhart N, Shi M, Dichek DA. TGF-β (Transforming growth factor-β) signaling protects the thoracic and abdominal aorta from angiotensin II-induced pathology by distinct mechanisms. Arterioscler Thromb Vasc Biol. 2017;37:2102–2113. doi: 10.1161/ATVBAHA.117.309401. doi: 10.1161/ATVBAHA.117.309401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Daugherty A, Chen Z, Sawada H, Rateri DL, Sheppard MB. Transforming growth factor-beta in thoracic aortic aneurysms: good, bad, or irrelevant? J Am Heart Assoc. 2017;6:eDD5221. doi: 10.1161/JAHA.116.005221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Maegdefessel L, Azuma J, Toh R, Deng A, Merk DR, Raiesdana A, Leeper NJ, Raaz U, Schoelmerich AM, McConnell MV, Dalman RL, Spin JM, Tsao PS. MicroRNA-21 blocks abdominal aortic aneurysm development and nicotine-augmented expansion. Sci Transl Med. 2012;4:122ra22. doi: 10.1126/scitranslmed.3003441. doi: 10.1126/scitranslmed.3003441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Huang X, Yue Z, Wu J, Chen J, Wang S, Wu J, Ren L, Zhang A, Deng P, Wang K, Wu C, Ding X, Ye P, Xia J. MicroRNA-21 knockout exacerbates angiotensin II-induced thoracic aortic aneurysm and dissection in mice with abnormal transforming growth factor-β-SMAD3 signaling. Arterioscler Thromb Vasc Biol. 2018;38:1086–1101. doi: 10.1161/ATVBAHA.117.310694. doi: 10.1161/ATVBAHA.117.310694. [DOI] [PubMed] [Google Scholar]
  • 132.Majesky MW. Developmental basis of vascular smooth muscle diversity. Arterioscler Thromb Vasc Biol. 2007;27:1248–1258. doi: 10.1161/ATVBAHA.107.141069. doi: 10.1161/ATVBAHA.107.141069. [DOI] [PubMed] [Google Scholar]
  • 133.Sawada H, Rateri DL, Moorleghen JJ, Majesky MW, Daugherty A. Smooth muscle cells derived from second heart field and cardiac neural crest reside in spatially distinct domains in the media of the ascending aorta-brief report. Arterioscler Thromb Vasc Biol. 2017;37:1722–1726. doi: 10.1161/ATVBAHA.117.309599. doi: 10.1161/ATVBAHA.117.309599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sawada H, Chen JZ, Wright BC, Sheppard MB, Lu SH, Daugherty A. Heterogeneity of aortic smooth muscle cells: a determinant for regional characteristics of thoracic aortic aneurysms? J Transl Int Med. doi: 10.2478/jtim-2018-0023. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wu D, Ren P, Zheng Y, Zhang L, Xu G, Xie W, Lloyd EE, Zhang S, Zhang Q, Curci JA, Coselli JS, Milewicz DM, Shen YH, LeMaire SA. NLRP3 (Nucleotide oligomerization domain-like receptor family, pyrin domain containing 3)-caspase-1 inflammasome degrades contractile proteins: implications for aortic biomechanical dysfunction and aneurysm and dissection formation. Arterioscler Thromb Vasc Biol. 2017;37:694–706. doi: 10.1161/ATVBAHA.116.307648. doi: 10.1161/ATVBAHA.116.307648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wei H, Hu JH, Angelov SN, Fox K, Ysan J, Enstrom R, Smith A, Dichek DA. Aortopathy in a mouse model of Marfan syndrome is not mediated by altered transforming growth factor beta signaling. JAHA. 2017;6:eDD4968. doi: 10.1161/JAHA.116.004968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Sénémaud J, Caligiuri G, Etienne H, Delbosc S, Michel JB, Coscas R. Translational relevance and recent advances of animal models of abdominal aortic aneurysm. Arterioscler Thromb Vasc Biol. 2017;37:401–410. doi: 10.1161/ATVBAHA.116.308534. doi: 10.1161/ATVBAHA.116.308534. [DOI] [PubMed] [Google Scholar]
  • 138.Pyo R, Lee JK, Shipley JM, Curci JA, Mao D, Ziporin SJ, Ennis TL, Shapiro SD, Senior RM, Thompson RW. Targeted gene disruption of matrix metalloproteinase-9 (gelatinase B) suppresses development of experimental abdominal aortic aneurysms. J Clin Invest. 2000;105:1641–1649. doi: 10.1172/JCI8931. doi: 10.1172/JCI8931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Longo GM, Xiong W, Greiner TC, Zhao Y, Fiotti N, Baxter BT. Matrix metalloproteinases 2 and 9 work in concert to produce aortic aneurysms. J Clin Invest. 2002;110:625–632. doi: 10.1172/JCI15334. doi: 10.1172/JCI15334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Daugherty A, Cassis L. Chronic angiotensin II infusion promotes atherogenesis in low density lipoprotein receptor -/- mice. Ann N Y Acad Sci. 1999;892:108–118. doi: 10.1111/j.1749-6632.1999.tb07789.x. [DOI] [PubMed] [Google Scholar]
  • 141.Kanematsu Y, Kanematsu M, Kurihara C, Tsou TL, Nuki Y, Liang EI, Makino H, Hashimoto T. Pharmacologically induced thoracic and abdominal aortic aneurysms in mice. Hypertension. 2010;55:1267–1274. doi: 10.1161/HYPERTENSIONAHA.109.140558. doi: 10.1161/HYPERTENSIONAHA.109.140558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Imanishi M, Chiba Y, Tomita N, Matsunaga S, Nakagawa T, Ueno M, Yamamoto K, Tamaki T, Tomita S. Hypoxia-inducible factor-1α in smooth muscle cells protects against aortic aneurysms-brief report. Arterioscler Thromb Vasc Biol. 2016;36:2158–2162. doi: 10.1161/ATVBAHA.116.307784. doi: 10.1161/ATVBAHA.116.307784. [DOI] [PubMed] [Google Scholar]
  • 143.Yamanouchi D, Morgan S, Stair C, Seedial S, Lengfeld J, Kent KC, Liu B. Accelerated aneurysmal dilation associated with apoptosis and inflammation in a newly developed calcium phosphate rodent abdominal aortic aneurysm model. J Vasc Surg. 2012;56:455–461. doi: 10.1016/j.jvs.2012.01.038. doi: 10.1016/j.jvs.2012.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Liu J, Lu H, Howatt DA, Balakrishnan A, Moorleghen JJ, Sorci-Thomas M, Cassis LA, Daugherty A. Associations of ApoAI and ApoB-containing lipoproteins with AngII-induced abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc Biol. 2015;35:1826–1834. doi: 10.1161/ATVBAHA.115.305482. doi: 10.1161/ATVBAHA.115.305482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Liu J, Daugherty A, Lu H. Angiotensin II and abdominal aortic aneurysms: an update. Curr Pharm Des. 2015;21:4035–4048. doi: 10.2174/1381612821666150826093318. [DOI] [PubMed] [Google Scholar]
  • 146.Daugherty A, Tabas I, Rader DJ. Accelerating the pace of atherosclerosis research. Arterioscler Thromb Vasc Biol. 2015;35:11–12. doi: 10.1161/ATVBAHA.114.304833. doi: 10.1161/ATVBAHA.114.304833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lu H, Howatt DA, Balakrishnan A, Graham MJ, Mullick AE, Daugherty A. Hypercholesterolemia induced by a PCSK9 gain-of-function mutation augments angiotensin II-induced abdominal aortic aneurysms in C57BL/6 mice-brief report. Arterioscler Thromb Vasc Biol. 2016;36:1753–1757. doi: 10.1161/ATVBAHA.116.307613. doi: 10.1161/ATVBAHA.116.307613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bjørklund MM, Hollensen AK, Hagensen MK, Dagnaes-Hansen F, Christoffersen C, Mikkelsen JG, Bentzon JF. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ Res. 2014;114:1684–1689. doi: 10.1161/CIRCRESAHA.114.302937. doi: 10.1161/CIRCRESAHA.114.302937. [DOI] [PubMed] [Google Scholar]
  • 149.Roche-Molina M, Sanz-Rosa D, Cruz FM, García-Prieto J, López S, Abia R, Muriana FJ, Fuster V, Ibáñez B, Bernal JA. Induction of sustained hypercholesterolemia by single adeno-associated virus-mediated gene transfer of mutant hPCSK9. Arterioscler Thromb Vasc Biol. 2015;35:50–59. doi: 10.1161/ATVBAHA.114.303617. doi: 10.1161/ATVBAHA.114.303617. [DOI] [PubMed] [Google Scholar]
  • 150.Goettsch C, Hutcheson JD, Hagita S, Rogers MA, Creager MD, Pham T, Choi J, Mlynarchik AK, Pieper B, Kjolby M, Aikawa M, Aikawa E. A single injection of gain-of-function mutant PCSK9 adeno-associated virus vector induces cardiovascular calcification in mice with no genetic modification. Atherosclerosis. 2016;251:109–118. doi: 10.1016/j.atherosclerosis.2016.06.011. doi: 10.1016/j.atherosclerosis.2016.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Daugherty A, Cassis LA, Lu H. Complex pathologies of angiotensin II-induced abdominal aortic aneurysms. J Zhejiang Univ Sci B. 2011;12:624–628. doi: 10.1631/jzus.B1101002. doi: 10.1631/jzus.B1101002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Rateri DL, Howatt DA, Moorleghen JJ, Charnigo R, Cassis LA, Daugherty A. Prolonged infusion of angiotensin II in apoE(-/-) mice promotes macrophage recruitment with continued expansion of abdominal aortic aneurysm. Am J Pathol. 2011;179:1542–1548. doi: 10.1016/j.ajpath.2011.05.049. doi: 10.1016/j.ajpath.2011.05.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Davis FM, Rateri DL, Daugherty A. Mechanisms of aortic aneurysm formation: translating preclinical studies into clinical therapies. Heart. 2014;100:1498–1505. doi: 10.1136/heartjnl-2014-305648. doi: 10.1136/heartjnl-2014-305648. [DOI] [PubMed] [Google Scholar]
  • 154.Davis FM, Rateri DL, Daugherty A. Abdominal aortic aneurysm: novel mechanisms and therapies. Curr Opin Cardiol. 2015;30:566–573. doi: 10.1097/HCO.0000000000000216. doi: 10.1097/HCO.0000000000000216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ijaz T, Sun H, Pinchuk IV, Milewicz DM, Tilton RG, Brasier AR. Deletion of NF-κB/RelA in angiotensin II-sensitive mesenchymal cells blocks aortic vascular inflammation and abdominal aortic aneurysm formation. Arterioscler Thromb Vasc Biol. 2017;37:1881–1890. doi: 10.1161/ATVBAHA.117.309863. doi: 10.1161/ATVBAHA.117.309863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Takei Y, Tanaka T, Kent KC, Yamanouchi D. Osteoclastogenic differentiation of macrophages in the development of abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol. 2016;36:1962–1971. doi: 10.1161/ATVBAHA.116.307715. doi: 10.1161/ATVBAHA.116.307715. [DOI] [PubMed] [Google Scholar]
  • 157.Howatt DA, Balakrishnan A, Moorleghen JJ, Muniappan L, Rateri DL, Uchida HA, Takano J, Saido TC, Chishti AH, Baud L, Subramanian V. Leukocyte calpain deficiency reduces angiotensin II-induced inflammation and atherosclerosis but not abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc Biol. 2016;36:835–845. doi: 10.1161/ATVBAHA.116.307285. doi: 10.1161/ATVBAHA.116.307285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Kusters PJH, Seijkens TTP, Beckers L, Lievens D, Winkels H, de Waard V, Duijvestijn A, Lindquist Liljeqvist M, Roy J, Daugherty A, Newby A, Gerdes N, Lutgens E. CD40L deficiency protects against aneurysm formation. Arterioscler Thromb Vasc Biol. 2018;38:1076–1085. doi: 10.1161/ATVBAHA.117.310640. doi: 10.1161/ATVBAHA.117.310640. [DOI] [PubMed] [Google Scholar]
  • 159.Schaheen B, Downs EA, Serbulea V, Almenara CC, Spinosa M, Su G, Zhao Y, Srikakulapu P, Butts C, McNamara CA, Leitinger N, Upchurch GR, Jr, Meher AK, Ailawadi G. B-Cell depletion promotes aortic infiltration of immunosuppressive cells and is protective of experimental aortic aneurysm. Arterioscler Thromb Vasc Biol. 2016;36:2191–2202. doi: 10.1161/ATVBAHA.116.307559. doi: 10.1161/ATVBAHA.116.307559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Krishna SM, Seto SW, Jose RJ, Li J, Morton SK, Biros E, Wang Y, Nsengiyumva V, Lindeman JH, Loots GG, Rush CM, Craig JM, Golledge J. Wnt signaling pathway inhibitor sclerostin inhibits angiotensin II-induced aortic aneurysm and atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:553–566. doi: 10.1161/ATVBAHA.116.308723. doi: 10.1161/ATVBAHA.116.308723. [DOI] [PubMed] [Google Scholar]
  • 161.Martorell S, Hueso L, Gonzalez-Navarro H, Collado A, Sanz MJ, Piqueras L. Vitamin D receptor activation reduces angiotensin-II-induced dissecting abdominal aortic aneurysm in apolipoprotein E-knockout mice. Arterioscler Thromb Vasc Biol. 2016;36:1587–1597. doi: 10.1161/ATVBAHA.116.307530. doi: 10.1161/ATVBAHA.116.307530. [DOI] [PubMed] [Google Scholar]
  • 162.Nakao T, Horie T, Baba O, et al. Genetic ablation of microRNA-33 attenuates inflammation and abdominal aortic aneurysm formation via several anti-inflammatory pathways. Arterioscler Thromb Vasc Biol. 2017;37:2161–2170. doi: 10.1161/ATVBAHA.117.309768. doi: 10.1161/ATVBAHA.117.309768. [DOI] [PubMed] [Google Scholar]
  • 163.Liu CL, Wang Y, Liao M, et al. Allergic lung inflammation aggravates angiotensin II-induced abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc Biol. 2016;36:69–77. doi: 10.1161/ATVBAHA.115.305911. doi: 10.1161/ATVBAHA.115.305911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Moran CS, Rush CM, Dougan T, Jose RJ, Biros E, Norman PE, Gera L, Golledge J. Modulation of kinin B2 receptor signaling controls aortic dilatation and rupture in the angiotensin II-infused apolipoprotein E-deficient mouse. Arterioscler Thromb Vasc Biol. 2016;36:898–907. doi: 10.1161/ATVBAHA.115.306945. doi: 10.1161/ATVBAHA.115.306945. [DOI] [PubMed] [Google Scholar]
  • 165.Lu WW, Jia LX, Ni XQ, Zhao L, Chang JR, Zhang JS, Hou YL, Zhu Y, Guan YF, Yu YR, Du J, Tang CS, Qi YF. Intermedin1-53 attenuates abdominal aortic aneurysm by inhibiting oxidative stress. Arterioscler Thromb Vasc Biol. 2016;36:2176–2190. doi: 10.1161/ATVBAHA.116.307825. doi: 10.1161/ATVBAHA.116.307825. [DOI] [PubMed] [Google Scholar]
  • 166.Yan YF, Pei JF, Zhang Y, Zhang R, Wang F, Gao P, Zhang ZQ, Wang TT, She ZG, Chen HZ, Liu DP. The paraoxonase gene cluster protects against abdominal aortic aneurysm formation. Arterioscler Thromb Vasc Biol. 2017;37:291–300. doi: 10.1161/ATVBAHA.116.308684. doi: 10.1161/ATVBAHA.116.308684. [DOI] [PubMed] [Google Scholar]
  • 167.Umebayashi R, Uchida HA, Kakio Y, Subramanian V, Daugherty A, Wada J. Cilostazol attenuates angiotensin II-induced abdominal aortic aneurysms but not atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2018;38:903–912. doi: 10.1161/ATVBAHA.117.309707. doi: 10.1161/ATVBAHA.117.309707. [DOI] [PubMed] [Google Scholar]
  • 168.Lutshumba J, Liu S, Zhong Y, Hou T, Daugherty A, Lu H, Guo Z, Gong MC. Deletion of BMAL1 in smooth muscle cells protects mice from abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol. 2018;38:1063–1075. doi: 10.1161/ATVBAHA.117.310153. doi: 10.1161/ATVBAHA.117.310153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Robinet P, Milewicz DM, Cassis LA, Leeper NJ, Lu HS, Smith JD. Consideration of sex differences in design and reporting of experimental arterial pathology studies-statement from ATVB council. Arterioscler Thromb Vasc Biol. 2018;38:292–303. doi: 10.1161/ATVBAHA.117.309524. doi: 10.1161/ATVBAHA.117.309524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Arnold AP, Cassis LA, Eghbali M, Reue K, Sandberg K. Sex hormones and sex chromosomes cause sex differences in the development of cardiovascular diseases. Arterioscler Thromb Vasc Biol. 2017;37:746–756. doi: 10.1161/ATVBAHA.116.307301. doi: 10.1161/ATVBAHA.116.307301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Chenu C, Adlanmerini M, Boudou F, Chantalat E, Guihot AL, Toutain C, Raymond-Letron I, Vicendo P, Gadeau AP, Henrion D, Arnal JF, Lenfant F. Testosterone prevents cutaneous ischemia and necrosis in males through complementary estrogenic and androgenic actions. Arterioscler Thromb Vasc Biol. 2017;37:909–919. doi: 10.1161/ATVBAHA.117.309219. doi: 10.1161/ATVBAHA.117.309219. [DOI] [PubMed] [Google Scholar]
  • 172.Alsiraj Y, Thatcher SE, Blalock E, Fleenor B, Daugherty A, Cassis LA. Sex chromosome complement defines diffuse versus focal angiotensin II-induced aortic pathology. Arterioscler Thromb Vasc Biol. 2018;38:143–153. doi: 10.1161/ATVBAHA.117.310035. doi: 10.1161/ATVBAHA.117.310035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Wassmann S, Czech T, van Eickels M, Fleming I, Böhm M, Nickenig G. Inhibition of diet-induced atherosclerosis and endothelial dysfunction in apolipoprotein E/angiotensin II type 1A receptor double-knockout mice. Circulation. 2004;110:3062–3067. doi: 10.1161/01.CIR.0000137970.47771.AF. doi: 10.1161/01.CIR.0000137970.47771.AF. [DOI] [PubMed] [Google Scholar]
  • 174.Cassis LA, Rateri DL, Lu H, Daugherty A. Bone marrow transplantation reveals that recipient AT1a receptors are required to initiate angiotensin II-induced atherosclerosis and aneurysms. Arterioscler Thromb Vasc Biol. 2007;27:380–386. doi: 10.1161/01.ATV.0000254680.71485.92. doi: 10.1161/01.ATV.0000254680.71485.92. [DOI] [PubMed] [Google Scholar]
  • 175.Galatioto J, Caescu CI, Hansen J, Cook JR, Miramontes I, Iyengar R, Ramirez F. Cell type-specific contributions of the angiotensin II type 1a receptor to aorta homeostasis and aneurysmal disease-brief report. Arterioscler Thromb Vasc Biol. 2018;38:588–591. doi: 10.1161/ATVBAHA.117.310609. doi: 10.1161/ATVBAHA.117.310609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Kaschina E, Namsolleck P, Unger T. AT2 receptors in cardiovascular and renal diseases. Pharmacol Res. 2017;125(pt A):39–47. doi: 10.1016/j.phrs.2017.07.008. doi: 10.1016/j.phrs.2017.07.008. [DOI] [PubMed] [Google Scholar]
  • 177.Daugherty A, Rateri DL, Howatt DA, Charnigo R, Cassis LA. PD123319 augments angiotensin II-induced abdominal aortic aneurysms through an AT2 receptor-independent mechanism. PLoS One. 2013;8:e61849. doi: 10.1371/journal.pone.0061849. doi: 10.1371/journal.pone.0061849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Silvestre JS, Tamarat R, Senbonmatsu T, Icchiki T, Ebrahimian T, Iglarz M, Besnard S, Duriez M, Inagami T, Lévy BI. Antiangiogenic effect of angiotensin II type 2 receptor in ischemia-induced angiogenesis in mice hindlimb. Circ Res. 2002;90:1072–1079. doi: 10.1161/01.res.0000019892.41157.24. [DOI] [PubMed] [Google Scholar]
  • 179.Paquin-Veillette J, Lizotte F, Robillard S, Béland R, Breton MA, Guay A, Despatis MA, Geraldes P. Deletion of AT2 receptor prevents SHP-1-induced VEGF inhibition and improves blood flow reperfusion in diabetic ischemic hindlimb. Arterioscler Thromb Vasc Biol. 2017;37:2291–2300. doi: 10.1161/ATVBAHA.117.309977. doi: 10.1161/ATVBAHA.117.309977. [DOI] [PubMed] [Google Scholar]
  • 180.Thatcher SE, Zhang X, Howatt DA, Lu H, Gurley SB, Daugherty A, Cassis LA. Angiotensin-converting enzyme 2 deficiency in whole body or bone marrow-derived cells increases atherosclerosis in low-density lipoprotein receptor-/- mice. Arterioscler Thromb Vasc Biol. 2011;31:758–765. doi: 10.1161/ATVBAHA.110.221614. doi: 10.1161/ATVBAHA.110.221614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Thatcher SE, Zhang X, Howatt DA, Yiannikouris F, Gurley SB, Ennis T, Curci JA, Daugherty A, Cassis LA. Angiotensin-converting enzyme 2 decreases formation and severity of angiotensin II-induced abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol. 2014;34:2617–2623. doi: 10.1161/ATVBAHA.114.304613. doi: 10.1161/ATVBAHA.114.304613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Moran CS, Biros E, Krishna SM, Wang Y, Tikellis C, Morton SK, Moxon JV, Cooper ME, Norman PE, Burrell LM, Thomas MC, Golledge J. Resveratrol inhibits growth of experimental abdominal aortic aneurysm associated with upregulation of angiotensin-converting enzyme 2. Arterioscler Thromb Vasc Biol. 2017;37:2195–2203. doi: 10.1161/ATVBAHA.117.310129. doi: 10.1161/ATVBAHA.117.310129. [DOI] [PubMed] [Google Scholar]
  • 183.Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D, Alenina N, Bader M, Campagnole-Santos MJ. The ACE2/angiotensin-(1-7)/MAS axis of the renin-angiotensin system: focus on angiotensin-(1-7). Physiol Rev. 2018;98:505–553. doi: 10.1152/physrev.00023.2016. doi: 10.1152/physrev.00023.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wang W, Bodiga S, Das SK, Lo J, Patel V, Oudit GY. Role of ACE2 in diastolic and systolic heart failure. Heart Fail Rev. 2012;17:683–691. doi: 10.1007/s10741-011-9259-x. doi: 10.1007/s10741-011-9259-x. [DOI] [PubMed] [Google Scholar]
  • 185.Souza ÁP, Sobrinho DB, Almeida JF, Alves GM, Macedo LM, Porto JE, Vêncio EF, Colugnati DB, Santos RA, Ferreira AJ, Mendes EP, Castro CH. Angiotensin II type 1 receptor blockade restores angiotensin-(1-7)-induced coronary vasodilation in hypertrophic rat hearts. Clin Sci (Lond) 2013;125:449–459. doi: 10.1042/CS20120519. doi: 10.1042/CS20120519. [DOI] [PubMed] [Google Scholar]
  • 186.Guimaraes PS, Santiago NM, Xavier CH, Velloso EP, Fontes MA, Santos RA, Campagnole-Santos MJ. Chronic infusion of angiotensin-(1-7) into the lateral ventricle of the brain attenuates hypertension in DOCA-salt rats. Am J Physiol Heart Circ Physiol. 2012;303:H393–H400. doi: 10.1152/ajpheart.00075.2012. doi: 10.1152/ajpheart.00075.2012. [DOI] [PubMed] [Google Scholar]
  • 187.Durand MJ, Zinkevich NS, Riedel M, Gutterman DD, Nasci VL, Salato VK, Hijjawi JB, Reuben CF, North PE, Beyer AM. Vascular actions of angiotensin 1-7 in the human microcirculation: novel role for telomerase. Arterioscler Thromb Vasc Biol. 2016;36:1254–1262. doi: 10.1161/ATVBAHA.116.307518. doi: 10.1161/ATVBAHA.116.307518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Hoffmann BR, Stodola TJ, Wagner JR, Didier DN, Exner EC, Lombard JH, Greene AS. Mechanisms of Mas1 receptor-mediated signaling in the vascular endothelium. Arterioscler Thromb Vasc Biol. 2017;37:433–445. doi: 10.1161/ATVBAHA.116.307787. doi: 10.1161/ATVBAHA.116.307787. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Arteriosclerosis, Thrombosis, and Vascular Biology are provided here courtesy of Wolters Kluwer Health

RESOURCES