Skip to main content
American Journal of Physiology - Heart and Circulatory Physiology logoLink to American Journal of Physiology - Heart and Circulatory Physiology
. 2020 Mar 6;318(4):H867–H882. doi: 10.1152/ajpheart.00538.2019

The deleterious role of the prostaglandin E2 EP3 receptor in angiotensin II hypertension

Timothy D Bryson 1,3, Teja S Pandrangi 1, Safa Z Khan 1, Jiang Xu 1, Tengis S Pavlov 1, Pablo A Ortiz 1,3, Edward Peterson 2, Pamela Harding 1,3,
PMCID: PMC7191497  PMID: 32142358

Abstract

Angiotensin II (ANG II) plays a key role in regulating blood pressure and inflammation. Prostaglandin E2 (PGE2) signals through four different G protein-coupled receptors, eliciting a variety of effects. We reported that activation of the EP3 receptor reduces cardiac contractility. More recently, we have shown that overexpression of the EP4 receptor is protective in a mouse myocardial infarction model. We hypothesize in this study that the relative abundance of EP3 and EP4 receptors is a major determinant of end-organ damage in the diseased heart. Thus EP3 is detrimental to cardiac function and promotes inflammation, whereas antagonism of the EP3 receptor is protective in an ANG II hypertension (HTN) model. To test our hypothesis, male 10- to 12-wk-old C57BL/6 mice were anesthetized with isoflurane and osmotic minipumps containing ANG II were implanted subcutaneously for 2 wk. We found that antagonism of the EP3 receptor using L798,106 significantly attenuated the increase in blood pressure with ANG II infusion. Moreover, antagonism of the EP3 receptor prevented a decline in cardiac function after ANG II treatment. We also found that 10- to 12-wk-old EP3-transgenic mice, which overexpress EP3 in the cardiomyocytes, have worsened cardiac function. In conclusion, activation or overexpression of EP3 exacerbates end-organ damage in ANG II HTN. In contrast, antagonism of the EP3 receptor is beneficial and reduces cardiac dysfunction, inflammation, and HTN.

NEW & NOTEWORTHY This study is the first to show that systemic treatment with an EP3 receptor antagonist (L798,106) attenuates the angiotensin II-induced increase in blood pressure in mice. The results from this project could complement existing hypertension therapies by combining blockade of the EP3 receptor with antihypertensive drugs.

Keywords: EP3, EP4, heart failure, hypertension, prostaglandin E2

INTRODUCTION

Angiotensin II (ANG II) is the major effector peptide of the renin-angiotensin system (87). ANG II plays an important role in both blood pressure regulation and inflammation along with other important biological functions (i.e., regulating cell growth and apoptosis, extracellular matrix deposition, cell migration, vascular motor tone). ANG II is released from circulating hepatic-derived angiotensinogen via the actions of renin and angiotensin-converting enzyme (21, 65, 83). ANG II is a potent vasoconstrictor, causing an increase in blood pressure and vasculature remodeling in chronic hypertension (HTN; Ref. 28). ANG II also promotes increased vascular permeability leading to increased inflammatory processes (9, 10, 86). The various actions of ANG II are mediated by specific intracellular signaling pathways downstream of ANG II binding to its surface receptors. In mammals, ANG II binds to two G protein-coupled receptors, ANG II types 1 (AT1) and 2 (AT2; Refs. 59, 80). The AT1 receptor typically activates phospholipase C through the Gq protein. AT1 is highly expressed in smooth muscle cells, whereas AT2 receptors are present in the kidneys, pancreas, heart, adrenals, brain, and vasculature (90, 94, 95). Both receptors signal via complex intracellular signaling cascades, although it is the consensus that AT2 antagonizes the effects of AT1 (17, 100).

ANG II plays a critical role in cardiovascular disease (43). It has been shown that ANG II promotes pathological cardiac hypertrophy in both neonatal rat cardiomyocytes (6, 7, 77, 78) and adult cardiomyocytes (49, 73, 96). Cardiomyocytes express both AT1 and AT2 receptors, although there is an abundance of evidence to support that ANG II primarily signals through AT1 in the cardiomyocytes (12, 13). Additionally, AT2 receptors were undetectable in both neonatal and adult rat cardiac fibroblasts (18, 93). The actions of ANG II on the cardiac fibroblasts increase extracellular matrix proteins and collagen deposition, an important factor in pathological left ventricle remodeling.

In recent years, a link between the immune system and hypertension has been described (34, 75). In various cell types and tissues, ANG II can directly activate immune cells (i.e., chemotaxis and differentiation; Ref. 42), promote the transcription of proinflammatory mediators (i.e., chemokines and cytokines; Refs. 67, 86), and activate proinflammatory signaling pathways (i.e., NF-κB and MAPK; Refs. 8, 31). It has been reported that ANG II promotes the production of reactive oxygen species (ROS) through stimulation of NADPH oxidase, via signaling through the AT1 receptor (25, 26). This increase in oxidative stress promotes endothelial dysfunction and vascular inflammation and contributes to increased activity of transcription factors like NF-κB combined with increases in chemokines, cytokines, and adhesion molecules (63, 84).

Prostaglandin E2 (PGE2) is produced from three known prostaglandin E synthase isoforms, a cytosolic form (cPGES) and two microsomal forms (mPGES-1 and mPGES-2), which are membrane bound. PGE2 signals through four distinct G protein-coupled receptor subtypes (EP1, EP2, EP3, and EP4) to produce a variety of physiological and pathological effects. EP2 and EP4 increase cAMP levels in the cell via activation of adenylate cyclase, whereas EP3 inhibits cAMP production and EP1 increases Ca2+ levels in the cell (76, 85). However, the cardiac effects of these receptors have not been fully elucidated. We (32) have previously published that mPGES-1-null mice were more susceptible to the deleterious effects of chronic ANG II infusion on the heart, suggesting a cardioprotective role for PGE2. Also, we (29) previously published that PGE2 via its EP3 receptor could reduce contractility of isolated myocytes and in the whole heart by mechanisms that appeared to involve decreased phospholamban phosphorylation. In a model of arachidonic acid (AA)-induced inflammation, Morimoto and colleagues (57) discovered that PGE2 was the most abundantly produced AA metabolite and further showed in EP3-null mice that PGE2-EP3 signaling was mediating the inflammatory response by increasing vascular permeability. Additionally, it has been reported that PGE2 can directly trigger the degranulation of mast cells and subsequent release of proinflammatory IL-6 in mouse bone marrow-derived mast cells. This was shown to be dependent on EP3-Gi signaling and by the influx of Ca2+ (5) and phosphatidylinositol 3-kinase (PI3K)-Akt pathways (45, 62). We (15) recently reported that activation of the EP4 receptor in isolated adult cardiac fibroblasts prevented an increase in the proinflammatory chemokine MCP-5 in response to lipopolysaccharide stimulation by blocking PI3K-Akt signaling and subsequent NF-κB activation. Furthermore, we (14) have reported that overexpression of EP4 in the cardiomyocyte improves cardiac function and reduces cardiac inflammation after myocardial infarction. Thus we hypothesize that the relative abundance of EP3 and EP4 receptors is a crucial determinant of end-organ damage in the diseased heart. We also hypothesize that overexpression of EP3 is detrimental to cardiac function and promotes inflammation, whereas inhibition of the EP3 receptor is protective in a mouse ANG II HTN model.

METHODS

Animal Use

For experimental protocol 1 (Mouse Experimental Protocols section below), male 10- to 12-wk-old C57BL/6 mice were purchased from Jackson. For protocol 2, male EP3-transgenic (EP3 Tg) and their wild-type littermates were kindly provided to the Harding Laboratory by Dr. Thomas Hohlfield at the University of Dusseldorf (Germany). All animal experiments were approved by the Henry Ford Health System Institutional Animal Care and Use Committee in accordance with federal guidelines.

Mouse Experimental Protocols

Effect of an EP3 antagonist in ANG II HTN and in an isoproterenol model.

Male 10- to 12-wk-old C57BL/6 mice were treated with either vehicle or ANG II (1.4 mg·kg−1·day−1) for a period of 2 wk via osmotic minipump. Additionally, mice were treated with either vehicle (ethanol diluted in 0.9% NaCl) or an EP3 antagonist (L798,106; 40 µg·kg−1·day−1 ip) daily. The dose of L798,106 was chosen based on the Ki values of the compound (0.3, 916, 5,000, and >5,000 nM for EP3, EP4, EP1, and EP2, respectively). In separate experiments, mice received infusion of isoproterenol (30 mg·kg−1·day−1) or vehicle (0.9% NaCl) for 10 days via osmotic minipump. Mice were also treated with either vehicle or an EP3 antagonist (L798,106; 40 µg·kg−1·day−1 ip) daily.

Effect of EP3 overexpression in ANG II HTN.

Male 10- to 12-wk-old EP3 Tg mice overexpressing EP3 in the cardiomyocytes and their wild-type littermates were treated with either vehicle (0.01 M acetic acid in 0.9% NaCl) or ANG II (1.4 mg·kg−1·day−1) for a period of 2 wk via osmotic minipump.

For experimental procedures, echocardiography was performed on conscious mice before surgery and at the conclusion of the study. Systolic blood pressure was monitored weekly to ensure that all animals receiving ANG II exhibited the expected increase in systolic blood pressure. At the end of the study, animals were anesthetized with pentobarbital sodium (100 mg/kg ip), and the heart was injected with 15% KCl, causing its arrest in diastole. The heart was excised and washed thoroughly in ice-cold Hanks’ balanced salt solution, followed by dissection of the atria and right ventricle. Hearts used for RNA isolation and real-time reverse transcription-polymerase chain reaction (RT-PCR) were stored in TRI Reagent at −80°C. For histological analysis, left ventricle (LV) sections were frozen in optimum cutting temperature media prechilled in isopentane and stored at −80°C. For protein extraction, LV were snap-frozen in liquid N2 and stored at −80°C until future analysis.

Cell Treatment Protocols

Cardiomyocytes (AVM) and cardiac fibroblasts (AVF) were isolated from 10- to 12-wk-old C57BL/6 mice as previously described by us (33). AVM were serum-starved before pretreatment for 1 h with vehicle, PGE2 (1 µM), or an EP4 agonist (CAY10598; 1 µM). After 1 h, cells were treated with either vehicle or ANG II (10−7 M) for 24 h. Cells were maintained at 37°C, 5% CO2. Similarly, AVF were serum-starved before pretreatment for 1 h with vehicle, PGE2 (1 μM), or CAY10598 (0.01 or 1 μM). After 1 h, cells were treated with either vehicle or ANG II (10−7 M). Protein lysate for Western blot analysis was obtained by scraping the cells on ice with lysis buffer containing protease (cat. no. 4693159001; Roche) and phosphatase inhibitors (cat. no. 05892970001; Roche) and then flash-frozen in liquid N2 and stored at −80°C.

Human coronary artery endothelial cells (HCECs) were purchased from American Type Culture Collection (ATCC; Manassas, VA). HCECs were maintained in Vascular Cell Basal Medium (cat. no. PCS-100-030; ATCC) containing the following components from the Endothelial Cell Growth Kit-BBE (cat. no. PCS-100-040; ATCC): bovine brain extract (0.2%), recombinant human EGF (5 ng/mL), l-glutamine (10 mM), heparin sulfate (0.75 U/mL), hydrocortisone hemisuccinate (1 µg/mL), ascorbic acid (50 µg/mL), and fetal bovine serum (2%). Cells were maintained at 37°C, 5% CO2. For treatment, HCECs were serum-starved for 24 h before pretreatment with PGE2 (1 µM) or CAY10598 (1 µM) for 1 h. After 1 h, HCECs were treated with vehicle or ANG II (10−7 M) for 24 h. Protein lysates for Western blot analysis were obtained by scraping the cells on ice with lysis buffer containing protease and phosphatase inhibitors and then flash-frozen in liquid N2 and stored at −80°C. All protein lysates were sonicated on ice for 10 s at 20 kHz.

Blood Pressure Measurements

Systolic blood pressure was measured on a weekly basis using tail-cuff plethysmography. Pressure was obtained using a MC4000 Multi Channel Blood Pressure Analysis System for mice (Hatteras Instruments, Cary, NC). Briefly, mice were trained for at least 1 wk before analysis. They were maintained at 38–40°C and subjected to 10 cycles of blood pressure analysis. At least 7 valid readings in a cycle of 10 measurements were needed to accept the measurements. In addition, a minimum systolic value of 75 mmHg was set as an exclusion criterion. Blood pressure measurements always occurred at the same time of day to eliminate any variability due to the rodent circadian cycle. In separate experiments, blood pressure was confirmed by measuring mean arterial pressure in the carotid artery. Mice were anesthetized with 3% isoflurane and maintained at 0.8–1.0% during the surgical procedure. PE-10 tubing fused to PE-50 was inserted into the left carotid artery. Pressure recordings were acquired for an equilibration period of 15 min, followed by 10 min of data acquisition using LabScribe software v2 (iWorx, Dover, NH). Mean arterial pressure was averaged across the 10-min data acquisition period.

Echocardiography

Echocardiography was performed before surgery and at the conclusion of the study (2 wk after infusion). The cardiac function of all mice was assessed by echocardiography using an Acuson 256 system (Mountain View, CA) with a 15-MHz linear transducer, as reported previously (46, 66). All echocardiography assessment was performed on conscious mice to eliminate the depressant effect of anesthesia on respiration and cardiac function. Diastolic measurements were made at the maximum left ventricle cavity dimension, whereas systolic parameters were measured during maximum anterior motion of the posterior wall. All echocardiography was performed by the same investigator who was blinded to the genotype. Additionally, all mice were trained for 2–3 days before assessment to eliminate heart rate variability.

Implantation of Osmotic Minipumps

Osmotic minipumps contained ANG II, isoproterenol, or vehicle solution. ANG II was dissolved in 0.01 M acetic acid diluted in 0.9% NaCl saline and administered chronically at a dose of 1.4 mg·kg−1·day−1 sc (32). Control mice for these experiments received minipumps containing the vehicle (0.01 M acetic acid diluted in physiological 0.9% saline). Isoproterenol was dissolved in 0.9% NaCl and administered at a dose of 30 mg·kg−1·day−1 sc. As a control for the isoproterenol experiments, mice received minipumps containing the vehicle (0.9% NaCl). Mice at 10–12 wk of age were anesthetized with pentobarbital sodium (10 mg/kg), and osmotic minipumps were implanted under the skin subcutaneously. Postoperatively, mice were allowed a recovery period on a heated pad followed by administration of buprenorphine as an analgesic (0.05 mg/kg).

Histology

Mouse hearts were harvested and sectioned transversely into three parts from apex to base. The sections were frozen in prechilled isopentane and stored at −80°C for determination of myocyte cross-sectional area (MCSA). For MCSA, sections of the heart were stained with fluorescein-labeled peanut agglutinin to delineate the myocytes. Four radially oriented microscope fields were selected from each section and photographed under the ×20 objective. MCSA was measured by computer-based planimetry (NIH ImageJ) and averaged across all four fields of the sections. The mean area was then calculated for each animal. All assessments were performed by observers who were blinded to animal identification and treatment groups.

Immunohistochemistry of Macrophages and T Cells

The number of macrophages and T cells in the heart was assessed using CD68+ staining (rat anti-CD68+ antibody; Bio-Rad) and CD3+ staining (rabbit anti-CD3+ antibody; Abcam), respectively, on frozen heart sections as we (14) previously described. Briefly, frozen sections were fixed in acetone for 10 min, followed by incubation with fresh 0.3% hydrogen peroxide for 30 min. Sections were then blocked in 5% centrifuged milk in Tris-buffered saline (TBS) and incubated with either anti-CD68+ or anti-CD3+ overnight at 4°C. Biotinylated secondary antibodies were incubated on sections for 1 h at room temperature. Sections were then incubated with a ready-to-use horseradish peroxidase-streptavidin reagent (Vector, Burlingame, CA) for 40 min at room temperature followed by aminoethyl carbazole single solution (Vector). Slides were rinsed in water and counterstained with Harris hematoxylin solution for 1 min and then rinsed again in water and mounted onto slides using an aqueous mounting solution (Aqua-Mount; Lerner). Negative controls consisted of sections incubated in the absence of primary antibody. Photographs of four randomly chosen fields per section were taken under ×20 objective, and the number of positively stained cells (per square millimeter) was measured by a blinded observer using NIH ImageJ software.

Real-Time Reverse Transcription-Polymerase Chain Reaction

Measurement of mRNA expression was performed by real-time reverse transcription-polymerase chain reaction (RT-PCR) using a SYBR Green method as follows. Two micrograms of DNase I-treated RNA sample was reverse transcribed using random primers and the Omniscript reverse transcriptase kit (Qiagen, Valencia, CA). Two microliters of the reverse transcription reaction was amplified in a Roche version 2.0 LightCycler PCR machine (Indianapolis, IN) using SYBR Green dye and specific primers against interleukin-1β (Il1b), tumor necrosis factor-α (Tnfa), prostaglandin E2 receptor subtype 3 (Ptger3), prostaglandin E2 receptor subtype 4 (Ptger4), monocyte chemoattractant protein-5 (Mcp5), interleukin-10 (Il10), arginase-1 (Arg1), or the reference gene glyceraldehyde-3-phosphate dehydrogenase (Gapdh). Fold induction of mRNA transcripts was calculated by correcting threshold cycle (CT) values to GAPDH using the 2-ΔΔCT method (50). Il1b, Tnfa, and Ptger3 primers were from Qiagen. Mcp5, Il10, Arg1, and Gapdh primers were synthesized by TIB MOLBIOL (Adelphia, NJ). All primer sequences are provided in Table 1.

Table 1.

Primer sequences

ID Sense Antisense Acc. No.
Il1b           NM_008361
Tnfa           NM_013693.3
Ptger3           NM_011196.2
Ptger4 5ʹ-gtgcagagatccagatggtca-3ʹ 5ʹ-atcygggtttctgctgatgtc-3ʹ     
Mcp5 5ʹ-caagagratcaccagcagcagg-3ʹ 5ʹ-tgcttgaggtggttgtggaa-3ʹ     
Il10 5ʹ-gctgcctgctcttactgact-3ʹ 5ʹ-ctgggaagtgggtgcagtta-3ʹ     
Arg1 5ʹ-gaacacggcagtggctttaac-3ʹ 5ʹ-tgcttagctctgtctgctttgc-3ʹ     
Gapdh 5ʹ-caaggtcatcccagagctg-3ʹ 5ʹ-tgtcatcatacttggcaggtt-3ʹ     

Primer sequences for Il1b, Tnfa, and Ptger3 are from Qiagen and are proprietary, thus only the accession numbers for the mRNA sequence are available. ID, identifier.

Western Blot Analysis

Anti-RhoB (cat. no. 2098; 1:1,000), anti-RhoA (cat. no. 2117; 1:1,000), and anti-GAPDH (cat. no. 5174; 1:2,000) were purchased from Cell Signaling Technology (Danvers, MA). Anti-Nox2/gp91 (cat. no. 611415; 1:1,000) was purchased from BD Transduction/BD Biosciences (San Jose, CA). For use with HCECs, anti-Nox2/gp91 from EMD Millipore (cat. no. 07-024; 1:1,000; Burlington, MA) was used. Protein levels were measured by Western blot using a method previously described by us (29). For mouse studies, all results were corrected to GAPDH and expressed as fold of vehicle or wild type. For AVM and HCECs experiments, results were corrected to GAPDH and expressed as fold of vehicle control.

Multiplex ELISArray Analysis

For the simultaneous detection of numerous cytokines or chemokines, we performed a multianalyte enzyme-linked immunosorbent assay (ELISA) using a commercially available kit from Qiagen (cat. no. MEM-005A) according to the manufacturer’s instructions and as previously described by us (14). The limit of detection for the panel of cytokines and chemokines is as follows (in pg/mL): IL-1β, 55.2; IL-4, 32.8; IL-6, 58.8; IL-10, 59.4; IL-12, 21.1; IL-17A, 83.3; IFN-γ, 39.0; TNF-γ, 30.5; TGF-β1, 85.4; MCP-1, 25.5; MIP-1γ, 35.1; MIP-1β, 26.7.

Chemicals

EP4 agonist (CAY10598) and prostaglandin E2 (PGE2) were from Cayman Chemical (Ann Arbor, MI). EP3 antagonist (L798,106; Ki values are 0.3, 916, >5,000 and >5,000 nM at EP3, EP4, EP1, and EP2, respectively) was from Tocris Bioscience (Minneapolis, MN). Angiotensin II was from Bachem Bioscience (Torrance, CA). TRI Reagent was from Molecular Research Center (Cincinnati, OH). Vascular Cell Basal Medium and the Endothelial Cell Growth Kit-BBE were from ATCC. All other compounds were obtained from Sigma-Aldrich (St. Louis, MO).

Statistical Analysis

All statistics were performed by a statistician in the Department of Public Health Sciences of Henry Ford Hospital using the statistical package SAS version 9.4. For all tests, a two-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. We also used Hochberg method for multiple testing. A P value <0.05 was considered as evidence of a statistically significant difference for experimental data with the P values being two-sided unless otherwise stated.

RESULTS

Expression of the EP3 Receptor Increases after ANG II Infusion

We (29) have previously shown that mRNA expression of the EP3 receptor increases in the infarcted heart and activation of EP3 leads to reduced cardiac function. To determine expression levels of the EP3 and EP4 receptors in the left ventricle and to examine whether EP3 receptor expression increases in the ANG II HTN model, we implanted osmotic minipumps containing either vehicle or ANG II for 2 wk. Figure 1A shows the expression of both receptors for both time points. After 2 wk, the expression of EP3 increased 2.5-fold compared with vehicle-infused animals (1.02 ± 0.13 in vehicle-treated vs. 2.59 ± 0.63 in ANG II-treated mice; P < 0.05), whereas expression of the EP4 receptor was unchanged [1.01 ± 0.10 in vehicle-treated vs. 0.96 ± 0.11 in ANG II-infused mice; P = not significant (ns)]. In additional experiments, we sought to examine whether the changes in EP3 expression were occurring in cardiomyocytes or fibroblasts, the two major cell types of the heart. We infused ANG II or vehicle control for 2 wk and then isolated cardiomyocytes (AVM) and fibroblasts (AVF) from each animal simultaneously. Real-time RT-PCR was used to determine Ptger3 (EP3) and Ptger4 (EP4) mRNA expression. Figure 1B shows that EP3 mRNA expression increases ~5-fold in the AVM (5.14 ± 1.52 in ANG II vs. 1.20 ± 0.46 in vehicle-treated; P < 0.05), whereas it increases only 25% in the fibroblasts (1.25 ± 0.05 in ANG II vs. 1.00 ± 0.07; P < 0.05). Figure 1C shows there was no significant increase in EP4 in the AVM (2.08 ± 0.92 in ANG II vs. 1.08 ± 0.28 in vehicle-treated; P = ns); however, EP4 significantly increased in the fibroblasts in response to ANG II (1.80 ± 0.05 in ANG II vs. 1.01 ± 0.09 in vehicle; P < 0.005).

Fig. 1.

Fig. 1.

A: real-time RT-PCR analysis of Ptger3 and Ptger4 mRNA expression in the left ventricle after 2 wk of infusion with vehicle or angiotensin II (ANG II; 1.4 mg·kg−1·day−1) via osmotic minipumps. B and C: real-time RT-PCR analysis of Ptger3 (B) and Ptger4 (C) mRNA expression in adult cardiomyocytes (AVM) or adult cardiac fibroblasts (AVF) after 2 wk of vehicle or ANG II infusion (1.4 mg·kg−1·day−1). *P < 0.05, ***P < 0.005 vs. respective vehicle. n = 3 Mice per group. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used.

EP3 Antagonist L798,106 Improves Cardiac Function and Hypertrophy in ANG II Hypertension

Since the expression of the EP3 receptor increases in ANG II hypertension, we hypothesized that antagonism of the EP3 receptor would prevent a decline in cardiac function. L798,106 is a potent and specific antagonist of the EP3 receptor (38). Figure 2A shows that there was a modest but significant reduction in ejection fraction after 2 wk of ANG II and this was attenuated when animals were treated with L798,106 (77.26 ± 0.47% in vehicle-treated vs. 69.82 ± 1.01% in ANG II-treated, P < 0.005; and 72.61 ± 0.51% in ANG II + L798,106-treated, P < 0.05). Similarly, Fig. 2B shows that shortening fraction was reduced with ANG II and improved with L798,106 treatment (51.61 ± 1.71% in ANG II-treated vs. 56.97 ± 1.01% in ANG II + L798,106-treated; P < 0.05). Moreover, Fig. 2C shows there was a significant increase in posterior wall thickness at systole after ANG II infusion, which was attenuated with an EP3 antagonist (1.19 ± 0.02 mm in vehicle-treated vs. 1.42 ± 0.02 mm in ANG II-treated, P < 0.01; and 1.30 ± 0.03 mm in ANG II + L798,106-treated, P < 0.05). We did not observe any change in cardiac output (Fig. 2D) between the groups (14.43 ± 1.33 mL·min−1·10 g−1 in vehicle-treated vs. 12.38 ± 0.98 mL·min−1·10 g−1 in ANG II-treated, P = ns; and 13.12 ± 1.10 mL·min−1·10 g−1 in ANG II + L798,106-treated, P = ns).

Fig. 2.

Fig. 2.

Cardiac function assessed by echocardiography in conscious mice 2 wk after vehicle or angiotensin II (ANG II; 1.4 mg·kg−1·day−1) ± daily injections of L798,106 (40 µg·kg−1·day−1) or diluted ethanol vehicle control. Ejection fraction (EF; A), shortening fraction (SF; B), posterior wall thickness at systole (PWTs; C), and cardiac output (CO; D) are shown. **P < 0.01, ***P < 0.005 vs. vehicle; +P < 0.05, +++P < 0.005 vs. ANG II. n = 11–13 Mice. E: heart weight (HW)-to-body weight (BW) ratio. Mice were anesthetized, and hearts were removed, weighed, and presented as milligrams per 10 g body wt. *P < 0.05 vs. vehicle. F: systolic blood pressure measured by tail-cuff plethysmography in conscious mice at baseline and 1 and 2 wk after vehicle or ANG II (1.4 mg·kg−1·day−1) ± daily injections of L798,106 (40 µg·kg−1·day−1) or diluted ethanol vehicle control for 2 wk. ***P < 0.005 vs. vehicle, +P < 0.05 vs. ANG II. n = 11–13 Mice per group. G: mean arterial pressure (MAP) measured in the carotid artery 2 wk after vehicle or ANG II (1.4 mg·kg−1·day−1) ± daily injections of L798,106 (40 µg·kg−1·day−1) or diluted ethanol vehicle. *P < 0.05 vs. vehicle, ++P < 0.01 vs. ANG II. n = 4–8 Mice per group. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. To correct for multiple testing, we used Hochberg correcting method.

ANG II is known to produce cardiac hypertrophy (32), and we observed an increase in heart weight-to-body weight ratio (Fig. 2E) with ANG II infusion (39.86 ± 1.08 in vehicle-treated vs. 45.31 ± 1.68 in ANG II-treated; P < 0.05). This increase was prevented when animals were treated with L798,106 (36.92 ± 1.74 in vehicle + L798,106-treated vs. 41.08 ± 1.31 in ANG II + L798,106; P = 0.06, P = ns).

Treatment with L798,106 Significantly Diminishes Pressor Response of ANG II

To determine the effect of L798,106 treatment in ANG II HTN, we measured blood pressure by tail cuff (Fig. 2F). As expected, ANG II infusion increased blood pressure significantly by 1 wk, and this remained elevated throughout the duration of the study (113 ± 3.2 mmHg in vehicle-treated at 1 wk vs. 151 ± 9.9 mmHg in ANG II-treated at 1 wk, P < 0.005; and 110 ± 2.7 mmHg in vehicle-treated at 2 wk vs. 168 ± 5.0 mmHg in ANG II-treated at 2 wk, P < 0.005). Daily administration of the EP3 antagonist L798,106 substantially attenuated the hypertensive effect of chronic ANG II treatment after 2 wk (168 ± 5.0 mmHg in ANG II to 136 ± 9.7 mmHg in ANG II + L798,106; P < 0.05). Since there was no significant effect of L798,106 on cardiac output, this suggested that the effect of L798,106 on blood pressure was not occurring via alterations in cardiac function. To confirm the blood pressure results, we repeated these experiments and measured mean arterial pressure (MAP) by carotid artery catheterization (Fig. 2G). MAP was increased with 2 wk of ANG II infusion, as expected (90 ± 4.79 in vehicle vs. 104 ± 1.08 in ANG II; P < 0.05). This increase was significantly reduced when animals were treated with L798,106 (94 ± 3.27 in ANG II + L798,106 vs. 104 ± 1.08 in ANG II; P < 0.01). These data confirm the tail-cuff blood pressure results.

L798,106 has no Effect on Blood Pressure or Cardiac Function with β-Adrenergic Stimulation

Infusion of the β-adrenergic receptor agonist, isoproterenol, is a nonischemic model that has been used to induce heart failure in rodents (1, 69, 88, 97, 103). To determine whether blockade of the EP3 receptor would ameliorate the effects of isoproterenol similar to ANG II, we infused isoproterenol for 10 days along with L798,106 treatment. Figure 3A shows that when mice received isoproterenol infusion for 10 days, there was a significant increase in heart rate, as expected (594 ± 24 beats/min in vehicle-treated vs. 739 ± 33 beats/min in isoproterenol-treated; P < 0.005). This was evident as early as 4 days after treatment. Treatment with an EP3 antagonist had no effect on this increase (670 ± 37 beats/min in isoproterenol + L798,106 group vs. 739 ± 33 beats/min in isoproterenol group; P = ns). Figure 3B shows that isoproterenol infusion also resulted in a significant, albeit modest increase in systolic blood pressure compared with vehicle-infused mice (122 ± 2.8 mmHg in isoproterenol vs. 108 ± 4.4 mmHg in vehicle; P < 0.05). EP3 antagonist treatment did not reduce this increase in blood pressure (114 ± 7.3 mmHg in isoproterenol + L798,106 vs. 122 ± 2.8 mmHg in isoproterenol; P = ns). Cardiac function was assessed in these mice by echocardiography. Figure 3C shows that there was a modest but significant reduction in shortening fraction (55.16 ± 1.3% in isoproterenol vs. 61.81 ± 1.0% in vehicle; P < 0.005). However, this reduction was not altered when mice were treated with L798,106 (53.87 ± 1.2% in isoproterenol + L798,106 vs. 55.16 ± 1.3% in isoproterenol; P = ns). Figure 3D also shows a significant increase in left ventricular dimension at systole and a corresponding increase in left ventricle mass (Fig. 3E). Neither of these parameters of hypertrophy was attenuated when mice received L798,106 treatment. Altogether, these data suggest the protective effects of EP3 antagonism are unique to the ANG II model and may not be relevant in other nonischemic models of heart failure.

Fig. 3.

Fig. 3.

Heart rate (A) and systolic blood pressure (B) measured by tail-cuff plethysmography in conscious mice throughout the infusion protocol of vehicle or isoproterenol (30 mg·kg−1·day−1) ± daily injections of L798,106 (40 µg·kg−1·day−1) or diluted ethanol vehicle control. *P < 0.05, ***P < 0.005 vs. vehicle. n = 4–6 Mice per group. Cardiac function was assessed by echocardiography in conscious mice at 10–12 wk of age 10 days after vehicle or isoproterenol (ISO) infusion (30 mg·kg−1·day−1). Shortening fraction (SF; C) and left ventricular dimension at systole (LVDs; D) are shown. E: left ventricle-to-body weight (BW) ratio was obtained using heart weights at euthanasia. Mice were anesthetized, and hearts were removed, weighed, and presented as milligrams per 10 g body wt. **P < 0.01, ***P < 0.005 vs. vehicle. n = 7–8 Mice. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. To correct for multiple testing, we used Hochberg correcting method. bpm, Beats per minute.

Treatment with L798,106 Does Not Alter Cardiac AT1 mRNA Expression

It is known that upregulation of the AT1 receptor plays an important role in mediating the pathophysiology of increased blood pressure (55). Therefore, we examined whether L798,106 administration is affecting mRNA levels of the ANG II AT1 receptor in the left ventricle. Figure 4A shows that after ANG II infusion there was no change in AT1 mRNA levels (1.02 ± 0.09 in vehicle-treated vs. 1.16 ± 0.13 in ANG II-treated; P = ns). Similarly, there was no significant change between mice that received ANG II infusion with vehicle injection and those that received ANG II infusion with L798,106 treatment (1.16 ± 0.13 in ANG II-treated vs. 0.86 ± 0.10 in ANG II + L798,106-treated; P = ns).

Fig. 4.

Fig. 4.

A: real-time RT-PCR analysis of angiotensin II (ANG II) type 1 receptor (AT1) mRNA expression in left ventricle after 2 wk of ANG II infusion ± L798,106 (40 µg·kg−1·day−1 ip). n = 6 Mice. B: Western blot analysis for RhoB expression in mouse mesenteric arteries after 2 wk of vehicle or ANG II (1.4 mg/kg/day) ± L798,106 (40 µg/kg/day ip). ***P < 0.005 vs. vehicle, +++P < 0.005 vs. ANG II. n = 3–5 Mice per group. C, top, shows a representative Western blot for RhoA and the housekeeping protein GAPDH. Bottom shows quantitative analysis for RhoA expression in mouse mesenteric arteries (Mes. Art). D, top, shows a representative Western blot for RhoA and the housekeeping protein GAPDH in left ventricle homogenates after 2 wk of vehicle (Veh) or ANG II (1.4 mg−1·kg−1·day−1) ± L798,106 (L798; 40 µg−1·kg−1·day−1). Bottom shows quantitative analysis for RhoA expression in mouse left ventricle (LV). *P < 0.05 vs. vehicle. n = 3 Mice per group. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. To correct for multiple testing, we used Hochberg correcting method.

Treatment with L798,106 Reduces ANG II-Induced RhoB Expression in Mesenteric Arteries

Changes in Rho-kinase signaling have been shown to contribute to increases in total peripheral resistance in hypertension, through alterations in resistance vessels such as the mesenteric arteries (4, 91, 98). Western blot analysis using mesenteric arteries from the mice revealed a significant increase in RhoB protein expression after 2 wk of ANG II infusion (Fig. 4B; 1.00 ± 0.14 in vehicle-treated vs. 3.84 ± 1.39 in ANG II-treated; P < 0.005). This was dramatically attenuated when mice were treated with L798,106 (0.94 ± 0.32 in ANG II + L798,106 vs. 3.84 ± 1.39 in ANG II-treated; P < 0.005). These results suggest that ANG II increases RhoB expression, presumably increasing contraction in the vasculature. RhoB expression is reduced by L798,106 treatment, possibly serving as a mechanism for the reduction in blood pressure we observed. RhoB expression was undetectable in left ventricle homogenates 2 wk after ANG II infusion.

Figure 4C shows that in contrast to RhoB, RhoA expression was unchanged in the mesenteric arteries with infusion of ANG II (1.00 ± 0.20 in vehicle vs. 1.08 ± 0.40 in ANG II; P = ns). Although expression of RhoB was undetectable in the LV, RhoA expression was detectable in the LV homogenates. Interestingly, there was a small but significant decrease in RhoA expression with ANG II infusion (Fig. 4D), and this was unchanged with L798,106 treatment (1.00 ± 0.03 in vehicle vs. 0.82 ± 0.04 in ANG II, P < 0.05; 0.82 ± 0.04 in ANG II vs. 0.91 ± 0.14 in ANG II + L798,106, P = ns). This significant reduction in RhoA expression is likely not physiologically relevant and does not explain changes in blood pressure with ANG II infusion.

EP3 Overexpression Reduces Cardiac Function

Since the increase in the EP3 receptor in ANG II HTN is occurring mainly in the cardiomyocytes, we acquired mice that overexpress the porcine analog of human EP3 in the cardiomyocytes (EP3 Tg) and their wild-type littermates (WT). To assess the effect of EP3 overexpression in the cardiomyocytes on blood pressure, we measured systolic blood pressure in EP3 Tg and their WT littermates under baseline conditions and after infusion of ANG II for 2 wk. Figure 5A shows that under baseline conditions, the EP3 Tg mice have normal systolic blood pressure and this increases significantly after ANG II infusion, albeit to a similar extent to that observed in C57BL/6 mice (106 ± 2.01 mmHg at baseline vs. 152 ± 3.13 mmHg after ANG II infusion; P < 0.05). These data suggest that overexpression of EP3 in the cardiomyocytes has no direct effect on blood pressure regulation.

Fig. 5.

Fig. 5.

A: systolic blood pressure measured by tail-cuff plethysmography in conscious EP3-transgenic (EP3 Tg) mice 2 wk after vehicle or angiotensin II (ANG II) infusion (1.4 mg·kg−1·day−1). ***P < 0.005 vs. vehicle. n = 4–5 Mice per group. Cardiac function was assessed by echocardiography in conscious male wild-type (WT) or EP3 Tg mice at 10–12 wk of age under baseline conditions. Ejection fraction (EF; B), shortening fraction (SF; C), left ventricular dimension at diastole (LVDd; D), and posterior wall thickness at systole (PWTs; E) are shown. **P < 0.01 vs. WT. n = 3 WT and n = 9 EP3 Tg mice. F: myocyte cross-sectional area (MCSA) analysis. Sections of left ventricle were stained with peanut agglutinin to outline the cells. Myocyte area was quantified using NIH ImageJ software. ***P < 0.005 vs. WT. n = 5 Mice. G: representative MCSA images obtained from cross section of left ventricles stained with peanut agglutinin under ×20 objective. Scale bar is indicated in the lower right-hand corner of each panel. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used.

Figure 5B shows that as early as 10–12 wk of age, EP3 Tg mice have reduced ejection fraction (56.93 ± 4.94% in EP3 Tg vs. 76.18 ± 1.25% in WT; P < 0.05) and reduced shortening fraction at baseline (Fig. 5C; 38.49 ± 3.46% in EP3 Tg vs. 61.69 ± 2.93% in WT; P < 0.01). Additionally, EP3 Tg hearts display robust dilatation as shown by the left ventricular dimensions in diastole (Fig. 5D; 3.55 ± 0.23 mm in EP3 Tg vs. 2.62 ± 0.01 mm in WT; P < 0.01). The posterior wall thickness is unchanged between EP3 Tg and WT (Fig. 5E), thus these data suggest that the hearts display a phenotype of eccentric hypertrophy (1.13 ± 0.02 mm in EP3 Tg vs. 1.21 ± 0.01 mm in WT; P = ns).

To measure the hypertrophy of the EP3 Tg hearts, we performed MCSA analysis on frozen sections of LV at baseline. Figure 5F shows that the average myocyte area is increased in EP3 Tg compared with the WT littermates (191.4 ± 2.6 in WT vs. 218.9 ± 2.8 in EP3 Tg; P < 0.005). These data suggest that the EP3 Tg hearts have hypertrophy under baseline conditions and confirm the echocardiography data showing dilatation of the ventricles.

We hypothesized that the decline in cardiac function observed in EP3 Tg is exacerbated in ANG II HTN. Figure 6A shows that in response to ANG II, the hearts of WT mice are well compensated and maintain ejection fraction (76.49 ± 0.70% in vehicle vs. 75.30 ± 0.80% in ANG II; P = ns) and shortening fraction (Fig. 6B; 63.19 ± 0.98% in vehicle vs. 63.21 ± 1.37% in ANG II; P = ns). In contrast, the EP3 Tg mice have reduced ejection fraction and shortening fraction at baseline and cardiac function in response to ANG II is worse than control mice (ejection fraction: 75.30 ± 0.83% in WT + ANG II vs. 45.92 ± 9.63% in EP3 Tg + ANG II, P < 0.01; shortening fraction: 63.21 ± 1.37% in WT + ANG II vs. 34.77 ± 7.33% in EP3 Tg + ANG II, P < 0.01). Similarly, there was an increase in left ventricle dimensions at systole (Fig. 6C) and diastole (Fig. 6D; 0.88 ± 0.04 in WT + ANG II vs. 2.40 ± 0.54 in EP3 Tg + ANG II, P < 0.005; and 2.42 ± 0.05 in WT + ANG II vs. 3.50 ± 0.42 in EP3 Tg + ANG II, P < 0.005, respectively).

Fig. 6.

Fig. 6.

Cardiac function assessed by echocardiography in conscious male wild-type (WT) or EP3-transgenic (EP3 Tg) mice at 10–12 wk of age after 2 wk of vehicle (Veh) or angiotensin II (ANG II) infusion (1.4 mg·kg−1·day−1). Ejection fraction (EF; A), shortening fraction (SF; B), left ventricular dimension at systole (LVDs; C), and left ventricular dimension at diastole (LVDd; D) are shown. *P < 0.05, ***P < 0.005 vs. WT + vehicle; $$P < 0.01 vs. WT + ANG II. n = 3–5 Mice per group. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. To correct for multiple testing, we used Hochberg correcting method.

EP3-Transgenic Mice Have Increased Markers of Inflammation in Their Left Ventricle

We hypothesized that the depressed cardiac function observed in EP3 Tg mice occurs with a concomitant increase in cardiac inflammation. We performed real-time RT-PCR for several proinflammatory cytokines and chemokines in left ventricles from EP3 Tg and their WT littermates under baseline conditions. Figure 7A shows that there was a significant increase in TNF-α mRNA levels in EP3 Tg mice (1.01 ± 0.08 in WT vs. 1.45 ± 0.22 in EP3 Tg; P < 0.05). Additionally, we observed an increase in IL-1β (Fig. 7B; 1.04 ± 0.21 in WT vs. 1.87 ± 0.46 in EP3 Tg; P < 0.05). Interestingly, the mRNA levels of the chemokine MCP-5 were unchanged between the two groups (Fig. 7C; 1.02 ± 0.15 in WT vs. 1.10 ± 0.06 in EP3 Tg; P = ns). We also observed a significant reduction in the anti-inflammatory cytokine IL-10 (Fig. 7D; IL-10: 1.01 ± 0.11 in WT vs. 0.71 ± 0.02 in EP3 Tg; P < 0.05). There was no statistically significant difference in the anti-inflammatory M2 macrophage marker arginase-1 (Fig. 7E; 1.02 ± 0.13 in WT vs. 0.80 ± 0.07 in EP3 Tg; P = 0.26). Together, these data suggest that the EP3 Tg mice have increased proinflammatory signaling in their left ventricles by upregulating proinflammatory genes and downregulating anti-inflammatory genes. To confirm our real-time RT-PCR results, we examined the protein expression of several cytokines and chemokines using a multiplex ELISA array. Figure 7F shows that there was a reduction in IL-10, confirming our real-time RT-PCR results [0.04 ± 0.01 arbitrary units (AU) in WT vs. 0.02 ± 0.01 AU in EP3 Tg; P < 0.05]. There was also a significant reduction in IL-4 in the EP3 Tg mice (Fig. 7G; 0.03 ± 0.01 AU in WT vs. 0.02 ± 0.0 AU in EP3 Tg; P < 0.005). Levels of other cytokines were below the limits of detection for the assay.

Fig. 7.

Fig. 7.

Real-time RT-PCR analysis of cytokines in the left ventricles of wild-type (WT) and EP3-transgenic (EP3 Tg) mice under baseline conditions. Tumor necrosis factor-α (Tnfa; A), interleukin-1β (Il1b; B), monocyte chemoattractant protein-5 (Mcp5; C), interleukin-10 (Il10; D), and arginase-1 (Arg1; E) are shown. *P < 0.05 vs. WT. n = 3 Mice per group. Multiplex ELISA array analysis of interleukin-10 (IL-10; F) and interleukin-4 (IL-4; G) in the left ventricles of WT and EP3 Tg mice under baseline conditions is shown. Values were corrected for the total micrograms of protein per well. *P < 0.05, ***P < 0.005 vs. WT. n = 3 Mice per group. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. Abs, absorbance.

EP3-Transgenic Mice Have Increased T Cell Infiltration

Since the EP3 Tg mice have increased markers of inflammation with a reduction in anti-inflammatory markers, we hypothesized that this would result in increased infiltration of immune cells. In this study, we investigated the presence of CD68+ macrophages and CD3+ T cells in the left ventricle of WT and EP3 Tg mice under baseline conditions by immunohistochemistry. Figure 8B shows that there was no change in the number of macrophages in the left ventricle of EP3 Tg mice compared with WT littermates (90.8 ± 4.0 cells per field in WT vs. 80.0 ± 2.4 cells per field in EP3 Tg; P = ns). However, Fig. 8D shows that there was a significantly higher number of CD3+ T cells in EP3 Tg left ventricles (17.2 ± 1.3 cells per field in WT vs. 33.5 ± 3.0 cells per field in EP3 Tg; P < 0.005). These data confirm our findings with the cytokine/chemokine mRNA levels as a lack of chemokines specific for macrophages like MCP-5 correlates with a lack of increased macrophages. Similarly, the increase in IL-1β could attribute to the increase in T cells.

Fig. 8.

Fig. 8.

A: representative immunohistochemistry sections for macrophage (CD68+) staining in frozen sections of left ventricle under baseline conditions. B: representative immunohistochemistry for T cell (CD3) staining in left ventricle sections under baseline conditions. C: quantitative CD68 analysis for WT and Tg mice. D: quantitative CD3+ analysis. Images were taken under ×20 objective and analyzed using ImageJ software. The number of cells were counted by a blinded observer and shown as number of positive cells per field. Data are presented as means ± SE. ***P < 0.005 vs. WT. n = 5–10 Mice per group. Scale bars are indicated in the lower right-hand corner of each panel. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used.

EP3-Transgenic Mice Have Increased Expression of NADPH Oxidase 2 (Nox2/gp91)

Left ventricle hypertrophy and heart failure have been associated with an increase in ROS in various models (58, 72). To determine whether the EP3 Tg mice have increased production of ROS in their left ventricles, we performed Western blot for Nox2, a major enzyme responsible for ROS production. Figure 9A shows that compared with WT, EP3 Tg mice have a twofold increase in Nox2 expression in their left ventricles (1.00 ± 0.19 AU in WT vs. 2.02 ± 0.42 AU in EP3 Tg; P < 0.05). In contrast, expression of Nox4 was not altered between the two strains. Although ROS generation was not measured per se, increased Nox2 expression suggests there is increased oxidative stress in the EP3 Tg mice.

Fig. 9.

Fig. 9.

A: Western blot analysis for Nox2 in left ventricles of wild-type (WT) and EP3-transgenic (EP3 Tg) mice. Top is a representative Western blot showing Nox2/gp91 expression and the housekeeping protein GAPDH. Bottom displays quantitative analysis of Nox2 expression. Nox2 was corrected for protein loading by GAPDH and presented as fold of WT. *P < 0.05 vs. WT. n = 3 Mice per group. Western blot analysis for Nox2/gp91 expression in angiotensin II (ANG II)-treated (0.1 µM) adult cardiac fibroblasts in the presence of EP4 agonist (Ag.) CAY10598 (0.01 or 1 µM; B) or PGE2 (0.01 or 1 µM; C) is shown. Nox2 results were corrected to GAPDH and presented as fold of vehicle. ***P < 0.005 vs. vehicle; +P < 0.05, +++P < 0.005 vs. ANG II. n = 4–7 Mice per group. For statistical analysis, a 2-sample Wilcoxon test with the Fligner–Policello correction for unequal variances was used. To correct for multiple testing, we used Hochberg correcting method.

EP4 Agonist Reduces ANG II-Induced Nox2/gp91 Expression in Vitro

Since the EP3 Tg mice have increased levels of Nox2, we sought to determine whether activation of the EP4 receptor could antagonize the ANG II-induced increase in Nox2. Interestingly, ANG II did not stimulate Nox2 expression in AVM. For this reason, we analyzed the effects of EP4 activation in AVF and HCECs. Figure 9B shows that in AVF, treatment with ANG II (10−7 M) stimulated Nox2 expression (2.34 ± 0.10 AU in ANG II-treated vs. 1.00 ± 0.0 AU in vehicle-treated; P < 0.005). This increase was attenuated when AVF was pretreated with 1 µM CAY10598 (0.78 ± 0.08 AU in ANG II + CAY10598-treated vs. 2.34 ± 0.10 AU in ANG II-treated; P < 0.005) or with 0.01 µM CAY10598 (0.97 ± 0.10 AU in ANG II + CAY10598-treated vs. 2.34 ± 0.10 AU in ANG II-treated; P < 0.005). Similarly, Fig. 9C shows that pretreatment with 1 or 0.01 µM PGE2 significantly reduced ANG II-induced Nox2 expression (2.48 ± 0.50 AU in ANG II-treated vs. 0.45 ± 0.09 AU in ANG II + 1 µM PGE2-treated, P < 0.005; and 2.48 ± 0.50 AU in ANG II-treated vs. 0.73 ± 0.15 in ANG II + 0.01 µM PGE2-treated, P < 0.01). Additionally, there was no significant effect of either PGE2 or CAY10598 alone on Nox2 expression under basal conditions. These data suggest that activation of the EP4 receptor can potentially reduce reactive oxygen species production by inhibiting Nox2 expression in AVF.

Conflicting with the data from AVF, treatment with ANG II did not produce a significant increase in Nox2 in HCECs (0.83 ± 0.16 AU to 1.07 ± 0.14 AU). However, Nox2 expression in the presence of ANG II was substantially attenuated by pretreatment with the EP4 agonist (1.07 ± 0.14 AU vs. 0.54 ± 0.10 AU; P < 0.05). Similar results were observed with PGE2 (1.07 ± 0.14 AU vs. 0.72 ± 0.19 AU; P = ns). Additionally, neither PGE2 nor the EP4 agonist alone affected basal Nox2 expression. These results are similar to those observed with AVF.

DISCUSSION

The results of this study show, for the first time, that administration of an EP3 antagonist prevented a decline in cardiac function with ANG II infusion and substantially attenuated the increase in blood pressure. Furthermore, we report here that mice overexpressing EP3 in the cardiomyocytes have reduced cardiac function at baseline, a decline in cardiac function in response to ANG II, and increased inflammatory cytokines/chemokines in their left ventricle.

Previous studies have documented the role of the EP3 receptor in hypertension by performing studies on vascular reactivity (16). With the use of global EP3 knockout mice, Chen et al. (16) showed in isolated mesenteric arteries that the mice lacking EP3 display reduced systolic blood pressure at baseline and that the vasoconstrictive action of ANG II is ameliorated, suggesting an important role for EP3 in the control of blood pressure within the resistance vessels. Our current study is novel in that it utilizes systemic administration of the pharmacological EP3 antagonist and employs the cardiomyocyte-specific EP3-overexpressing mice. Our data suggest that the EP3 receptor may act in the same signaling pathway as ANG II to increase blood pressure. Although the response to L798,106 in ANG II hypertension in the kidneys and/or central nervous system was not examined in the present study, it is known that renal AT1 receptors play a role in ANG II hypertension (19). It was also shown that EP3 receptor activation in the rostral ventrolateral medulla mediates the pressor effects of PGE2 (71). It was previously reported that administration of the EP1/EP3 agonist sulprostone invokes contraction of guinea pig aorta and induces Ca2+ influx (82). Other studies have confirmed the ability of EP3 receptor activation to mobilize Ca2+ (2, 36, 40). The calcium dynamics of EP3 antagonism were not examined in this study.

The Rho/Rho-kinase cascade has been shown to play an important role in regulating cardiovascular function (23, 35, 44, 68, 81, 101). For example, Guan et al. (30) have shown in rats with transverse aortic constriction pressure overload, cardiac function was improved when the rats were treated with the Rho-kinase inhibitor fasudil. Moreover, they showed this was due to a reduction in ROS. Similar cardioprotective effects were observed in a mouse model of autoimmune myocarditis when mice were treated with fasudil (20). Interestingly, in the aforementioned study, Dai et al. found a shift in the cardiac T cell population from T helper type 17 to regulatory T cell, suggesting Rho-kinase may directly affect the T cell population and IL-17 production. In our current study, we observed an increase in RhoB expression in the mesenteric arteries in response to ANG II, and this was reduced with L798,106 treatment. There was no change in RhoA expression in the mesenteric arteries. In the left ventricle, RhoB expression was completely undetectable, whereas RhoA was robustly detected. This agrees with the literature showing only the RhoA isoform is expressed in cardiomyocytes (3). Interestingly, when animals received ANG II infusion for 2 wk, there was a significant decrease in RhoA expression in the LV. It is possible that 2 wk of ANG II infusion may cause a downregulation of RhoA, as RhoA is typically shown to be fast-acting in response to ANG II; however, this is purely speculative. Furthermore, the RhoA/Rho-kinase signaling cascade in response to ANG II treatment has been studied in depth (3, 41, 51, 61, 81, 101, 102). However, little is known about the role of RhoB in ANG II HTN, therefore, making our results a novel finding. RhoB could play a role in promoting inflammation in ANG II HTN as it has been shown that RhoB, but not RhoA, is a mediator of Rho-kinase-induced NF-κB activation (74).

Isoproterenol is a nonselective β-adrenergic receptor agonist that induces positive ionotropic and chronotropic effects in the heart (11). Isoproterenol has been used to induce heart failure in rodents for many years by causing ventricular remodeling and hypertrophy (37, 69, 104). Treatment with PGE2 has been shown to antagonize the contractile effects of isoproterenol in a Langendorff heart preparation. However, the receptor mediating this effect was not clear (48). Therefore, to determine whether EP3 receptor blockade could ameliorate the effects of isoproterenol, we infused isoproterenol with concomitant treatment of L798,106. We observed a robust increase in heart rate, as expected with β-adrenergic stimulation, and a modest but significant increase in systolic blood pressure after 10 days of isoproterenol infusion. However, there was no effect of L798,106 on heart rate, blood pressure, or cardiac function. These data suggest that there may not be any cross talk between EP3 receptor signaling and β-adrenergic signaling.

To study the effects of EP3 in the heart specifically, we obtained transgenic mice that overexpress EP3 in the cardiomyocytes. It was previously reported that the hearts of these mice are markedly hypertrophied, which is obvious as early as 5–6 wk of age, and that they have reduced ejection fraction at baseline. It was also reported that overexpression of EP3 is protective in a mouse ischemia-reperfusion model due to the reduced contractility of the left ventricle, which would normally result in depletion of ATP (53, 54). However, we are the first to show that these mice have inflammation in their left ventricles as demonstrated by increased TNF-α and IL-1β, two major cytokines associated with heart failure (22, 89, 92). There was also reduction of anti-inflammatory cytokines IL-10 and IL-4. Interestingly, it was also observed that mRNA levels of the chemokine MCP-5 were unchanged between WT and EP3 Tg mice. MCP-5 is a potent chemoattractant molecule for monocytes and macrophages, and we speculate that this could account for the lack of increased macrophages in the EP3 Tg heart, as determined by immunohistochemistry. Although the number of CD68+ cells was unchanged, it remains to be investigated whether there is a difference in the proinflammatory M1 versus inflammation-resolving M2 macrophage phenotypes in the EP3 Tg heart. In contrast to the macrophages, there was a significant increase in the number of infiltrating CD3+ T cells in EP3 Tg mouse hearts.

Oxidative stress has been shown to modulate several aspects of pathological cardiac remodeling (24, 26, 27, 39, 47, 60, 64). A large source of reactive oxygen species comes from the family of NADPH oxidases (24). In particular, loss of the Nox2 isoform has been shown to protect the heart from adverse cardiac remodeling after myocardial infarction (52, 64). Our results show that treatment with ANG II significantly increases the expression of Nox2 in AVF. Interestingly, treatment with ANG II did not increase Nox2 expression in the cardiomyocytes and only modestly increased expression in HCECs. To our knowledge, we are the first to show that activation of the EP4 receptor reduces ANG II-induced Nox2 expression in AVF and HCECs. Santiago et al. (79) recently reported that antagonism of the EP4 receptor increased the contractile effect of hydrogen peroxide in coronary arteries, suggesting the vasodilatory effect of PGE2 acting on the EP4 receptor may protect the coronary arteries from the effects of ROS. Our study differs in that we have shown activation of EP4 in the endothelial cells may directly reduce ROS levels via a decrease in Nox2 expression. It has also been reported in noncardiac cell types that the EP4 receptor plays a role in the reduction of ROS. For example, it was shown in myoblast cells that treatment with an EP4 antagonist increased ROS production, an effect that was reversed after cotreatment with antioxidants (56). In a mouse aortic dissection model, EP4 knockout resulted in more severe vascular inflammation and increased oxidative stress, via increased Nox1 expression (99). In contrast, it was shown in the M-1 collecting duct cell line that activation of the EP4 receptor increased Nox4 expression and subsequent ROS production (70). The mechanism of PGE2 on ROS production is likely cell-specific depending on the specific Nox isoform that is expressed.

Conclusions

The results of this project show the importance of the EP3 receptor in ANG II HTN and could lead to development of new drugs that target the ANG II receptors as well as EP3 for the treatment of HTN. Furthermore, it is important to understand the deleterious effects of EP3 in the heart during HTN as well as the detrimental effects of EP3 on cardiac function independent of changes in blood pressure.

GRANTS

This study was funded by Henry Ford Health System internal funds (A10259) to P. Harding. T. D. Bryson was supported by the predoctoral NIH T32 Detroit Cardiovascular Training Grant 5-T32-HL-12082205.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

T.D.B. and P.H. conceived and designed research; T.D.B., T.S. Pandrangi, S.Z.K., and J.X. performed experiments; T.D.B., T.S. Pandrangi, S.Z.K., J.X., T.S. Pavlov, P.A.O., E.P., and P.H. analyzed data; T.D.B., T.S. Pandrangi, T.S. Pavlov, P.A.O., and P.H. interpreted results of experiments; T.D.B. prepared figures; T.D.B. and T.S. Pandrangi drafted manuscript; T.D.B., T.S. Pandrangi, S.Z.K., T.S. Pavlov, P.A.O., and P.H. edited and revised manuscript; T.D.B., T.S. Pandrangi, S.Z.K., J.X., T.S. Pavlov, P.A.O., E.P., and P.H. approved final version of manuscript.

ACKNOWLEDGMENTS

We thank David Taube and D’Anna Potter for their excellent technical assistance. Additionally, we thank Drs. Thomas Hohlfield and Jutta Meyer-Kirchrath at the University of Dusseldorf for providing us with the EP3-transgenic mouse strain.

REFERENCES

  • 1.Aluja D, Inserte J, Penela P, Ramos P, Ribas C, Iñiguez MA, Mayor F Jr, Garcia-Dorado D. Calpains mediate isoproterenol-induced hypertrophy through modulation of GRK2. Basic Res Cardiol 114: 21, 2019. doi: 10.1007/s00395-019-0730-5. [DOI] [PubMed] [Google Scholar]
  • 2.An S, Yang J, So SW, Zeng L, Goetzl EJ. Isoforms of the EP3 subtype of human prostaglandin E2 receptor transduce both intracellular calcium and cAMP signals. Biochemistry 33: 14496–14502, 1994. doi: 10.1021/bi00252a016. [DOI] [PubMed] [Google Scholar]
  • 3.Aoki H, Izumo S, Sadoshima J. Angiotensin II activates RhoA in cardiac myocytes: a critical role of RhoA in angiotensin II-induced premyofibril formation. Circ Res 82: 666–676, 1998. doi: 10.1161/01.RES.82.6.666. [DOI] [PubMed] [Google Scholar]
  • 4.Asano M, Nomura Y. Comparison of inhibitory effects of Y-27632, a Rho kinase inhibitor, in strips of small and large mesenteric arteries from spontaneously hypertensive and normotensive Wistar-Kyoto rats. Hypertens Res 26: 97–106, 2003. doi: 10.1291/hypres.26.97. [DOI] [PubMed] [Google Scholar]
  • 5.Baba Y, Nishida K, Fujii Y, Hirano T, Hikida M, Kurosaki T. Essential function for the calcium sensor STIM1 in mast cell activation and anaphylactic responses. Nat Immunol 9: 81–88, 2008. doi: 10.1038/ni1546. [DOI] [PubMed] [Google Scholar]
  • 6.Baker KM, Aceto JF. Angiotensin II stimulation of protein synthesis and cell growth in chick heart cells. Am J Physiol Heart Circ Physiol 259: H610–H618, 1990. doi: 10.1152/ajpheart.1990.259.2.H610. [DOI] [PubMed] [Google Scholar]
  • 7.Baker KM, Booz GW, Dostal DE. Cardiac actions of angiotensin II: role of an intracardiac renin-angiotensin system. Annu Rev Physiol 54: 227–241, 1992. doi: 10.1146/annurev.ph.54.030192.001303. [DOI] [PubMed] [Google Scholar]
  • 8.Barnes PJ, Karin M. Nuclear factor-κB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 336: 1066–1071, 1997. doi: 10.1056/NEJM199704103361506. [DOI] [PubMed] [Google Scholar]
  • 9.Baylis C, Brenner BM. Modulation by prostaglandin synthesis inhibitors of the action of exogenous angiotensin II on glomerular ultrafiltration in the rat. Circ Res 43: 889–898, 1978. doi: 10.1161/01.RES.43.6.889. [DOI] [PubMed] [Google Scholar]
  • 10.Beilin LJ, Goldby FS, Mohring J. High arterial pressure versus humoral factors in the pathogenesis of the vascular lesions of malignant hypertension. Clin Sci Mol Med 52: 111–117, 1977. doi: 10.1042/cs0520111. [DOI] [PubMed] [Google Scholar]
  • 11.Bolter CP, Atkinson KJ. Maximum heart rate responses to exercise and isoproterenol in the trained rat. Am J Physiol Regul Integr Comp Physiol 254: R834–R839, 1988. doi: 10.1152/ajpregu.1988.254.5.R834. [DOI] [PubMed] [Google Scholar]
  • 12.Booz GW, Baker KM. Role of type 1 and type 2 angiotensin receptors in angiotensin II-induced cardiomyocyte hypertrophy. Hypertension 28: 635–640, 1996. doi: 10.1161/01.HYP.28.4.635. [DOI] [PubMed] [Google Scholar]
  • 13.Booz GW, Baker KM. The role of the renin-angiotensin system in the pathophysiology of cardiac remodeling. Blood Press Suppl 2: 10–18, 1996. [PubMed] [Google Scholar]
  • 14.Bryson TD, Gu X, Khalil RM, Khan S, Zhu L, Xu J, Peterson E, Yang XP, Harding P. Overexpression of prostaglandin E2 EP4 receptor improves cardiac function after myocardial infarction. J Mol Cell Cardiol 118: 1–12, 2018. doi: 10.1016/j.yjmcc.2018.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bryson TD, Ross J, Peterson E, Harding P. Prostaglandin E2 and an EP4 receptor agonist inhibit LPS-induced monocyte chemotactic protein 5 production and secretion in mouse cardiac fibroblasts via Akt and NF-κB signaling. Prostaglandins Other Lipid Mediat 144: 106349, 2019. doi: 10.1016/j.prostaglandins.2019.106349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chen L, Miao Y, Zhang Y, Dou D, Liu L, Tian X, Yang G, Pu D, Zhang X, Kang J, Gao Y, Wang S, Breyer MD, Wang N, Zhu Y, Huang Y, Breyer RM, Guan Y. Inactivation of the E-prostanoid 3 receptor attenuates the angiotensin II pressor response via decreasing arterial contractility. Arterioscler Thromb Vasc Biol 32: 3024–3032, 2012. doi: 10.1161/ATVBAHA.112.254052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ciuffo GM, Johren O, Egidy G, Heemskerk FM, Saavedra JM. Heterogeneity of rat angiotensin II AT2 receptor. Adv Exp Med Biol 396: 189–197, 1996. doi: 10.1007/978-1-4899-1376-0_20. [DOI] [PubMed] [Google Scholar]
  • 18.Crabos M, Roth M, Hahn AW, Erne P. Characterization of angiotensin II receptors in cultured adult rat cardiac fibroblasts. Coupling to signaling systems and gene expression. J Clin Invest 93: 2372–2378, 1994. doi: 10.1172/JCI117243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Crowley SD, Gurley SB, Oliverio MI, Pazmino AK, Griffiths R, Flannery PJ, Spurney RF, Kim HS, Smithies O, Le TH, Coffman TM. Distinct roles for the kidney and systemic tissues in blood pressure regulation by the renin-angiotensin system. J Clin Invest 115: 1092–1099, 2005. doi: 10.1172/JCI23378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dai K, Wang Y, Tai S, Ni H, Lian H, Yu Y, Liao W, Zheng C, Chen Q, Kuver A, Li J. Fasudil exerts a cardio-protective effect on mice with coxsackievirus B3-induced acute viral myocarditis. Cardiovasc Ther 36: e12477, 2018. doi: 10.1111/1755-5922.12477. [DOI] [PubMed] [Google Scholar]
  • 21.Dorer FE, Kahn JR, Lentz KE, Levine M, Skeggs LT. Purification and properties of angiotensin-converting enzyme from hog lung. Circ Res 31: 356–366, 1972. doi: 10.1161/01.RES.31.3.356. [DOI] [PubMed] [Google Scholar]
  • 22.Feldman AM, Combes A, Wagner D, Kadakomi T, Kubota T, Li YY, McTiernan C. The role of tumor necrosis factor in the pathophysiology of heart failure. J Am Coll Cardiol 35: 537–544, 2000. doi: 10.1016/S0735-1097(99)00600-2. [DOI] [PubMed] [Google Scholar]
  • 23.Funakoshi Y, Ichiki T, Shimokawa H, Egashira K, Takeda K, Kaibuchi K, Takeya M, Yoshimura T, Takeshita A. Rho-kinase mediates angiotensin II-induced monocyte chemoattractant protein-1 expression in rat vascular smooth muscle cells. Hypertension 38: 100–104, 2001. doi: 10.1161/01.HYP.38.1.100. [DOI] [PubMed] [Google Scholar]
  • 24.Griendling KK, FitzGerald GA. Oxidative stress and cardiovascular injury: part II: animal and human studies. Circulation 108: 2034–2040, 2003. doi: 10.1161/01.CIR.0000093661.90582.c4. [DOI] [PubMed] [Google Scholar]
  • 25.Griendling KK, Sorescu D, Lassègue B, Ushio-Fukai M. Modulation of protein kinase activity and gene expression by reactive oxygen species and their role in vascular physiology and pathophysiology. Arterioscler Thromb Vasc Biol 20: 2175–2183, 2000. doi: 10.1161/01.ATV.20.10.2175. [DOI] [PubMed] [Google Scholar]
  • 26.Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res 86: 494–501, 2000. doi: 10.1161/01.RES.86.5.494. [DOI] [PubMed] [Google Scholar]
  • 27.Grieve DJ, Byrne JA, Siva A, Layland J, Johar S, Cave AC, Shah AM. Involvement of the nicotinamide adenosine dinucleotide phosphate oxidase isoform Nox2 in cardiac contractile dysfunction occurring in response to pressure overload. J Am Coll Cardiol 47: 817–826, 2006. doi: 10.1016/j.jacc.2005.09.051. [DOI] [PubMed] [Google Scholar]
  • 28.Griffin SA, Brown WC, MacPherson F, McGrath JC, Wilson VG, Korsgaard N, Mulvany MJ, Lever AF. Angiotensin II causes vascular hypertrophy in part by a non-pressor mechanism. Hypertension 17: 626–635, 1991. doi: 10.1161/01.HYP.17.5.626. [DOI] [PubMed] [Google Scholar]
  • 29.Gu X, Xu J, Zhu L, Bryson T, Yang XP, Peterson E, Harding P. Prostaglandin E2 reduces cardiac contractility via EP3 receptor. Circ Heart Fail 9: e003291, 2016. doi: 10.1161/CIRCHEARTFAILURE.116.003291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Guan P, Liang Y, Wang N. Fasudil alleviates pressure overload-induced heart failure by activating Nrf2-mediated antioxidant responses. J Cell Biochem 119: 6452–6460, 2018. doi: 10.1002/jcb.26662. [DOI] [PubMed] [Google Scholar]
  • 31.Guijarro C, Egido J. Transcription factor-κB (NF-κB) and renal disease. Kidney Int 59: 415–424, 2001. doi: 10.1046/j.1523-1755.2001.059002415.x. [DOI] [PubMed] [Google Scholar]
  • 32.Harding P, Yang XP, He Q, Lapointe MC. Lack of microsomal prostaglandin E synthase-1 reduces cardiac function following angiotensin II infusion. Am J Physiol Heart Circ Physiol 300: H1053–H1061, 2011. doi: 10.1152/ajpheart.00772.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Harding P, Yang XP, Yang J, Shesely E, He Q, LaPointe MC. Gene expression profiling of dilated cardiomyopathy in older male EP4 knockout mice. Am J Physiol Heart Circ Physiol 298: H623–H632, 2010. doi: 10.1152/ajpheart.00746.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Harrison DG, Guzik TJ, Lob HE, Madhur MS, Marvar PJ, Thabet SR, Vinh A, Weyand CM. Inflammation, immunity, and hypertension. Hypertension 57: 132–140, 2011. doi: 10.1161/HYPERTENSIONAHA.110.163576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hartmann S, Ridley AJ, Lutz S. The function of Rho-associated kinases ROCK1 and ROCK2 in the pathogenesis of cardiovascular disease. Front Pharmacol 6: 276, 2015. doi: 10.3389/fphar.2015.00276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hatae N, Sugimoto Y, Ichikawa A. Prostaglandin receptors: advances in the study of EP3 receptor signaling. J Biochem 131: 781–784, 2002. doi: 10.1093/oxfordjournals.jbchem.a003165. [DOI] [PubMed] [Google Scholar]
  • 37.Heather LC, Catchpole AF, Stuckey DJ, Cole MA, Carr CA, Clarke K. Isoproterenol induces in vivo functional and metabolic abnormalities: similar to those found in the infarcted rat heart. J Physiol Pharmacol 60: 31–39, 2009. [PubMed] [Google Scholar]
  • 38.Hester A, Salzmann B, Rahmeh M, Kolben T, Czogalla B, Ditsch N, Mahner S, Jeschke U, Kolben TM. EP3 receptor antagonist L798,106 reduces proliferation and migration of SK-BR-3 breast cancer cells. OncoTargets Ther 12: 6053–6068, 2019. doi: 10.2147/OTT.S204919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Heymes C, Bendall JK, Ratajczak P, Cave AC, Samuel JL, Hasenfuss G, Shah AM. Increased myocardial NADPH oxidase activity in human heart failure. J Am Coll Cardiol 41: 2164–2171, 2003. doi: 10.1016/S0735-1097(03)00471-6. [DOI] [PubMed] [Google Scholar]
  • 40.Irie A, Segi E, Sugimoto Y, Ichikawa A, Negishi M. Mouse prostaglandin E receptor EP3 subtype mediates calcium signals via Gi in cDNA-transfected Chinese hamster ovary cells. Biochem Biophys Res Commun 204: 303–309, 1994. doi: 10.1006/bbrc.1994.2460. [DOI] [PubMed] [Google Scholar]
  • 41.Jin L, Ying Z, Hilgers RH, Yin J, Zhao X, Imig JD, Webb RC. Increased RhoA/Rho-kinase signaling mediates spontaneous tone in aorta from angiotensin II-induced hypertensive rats. J Pharmacol Exp Ther 318: 288–295, 2006. doi: 10.1124/jpet.105.100735. [DOI] [PubMed] [Google Scholar]
  • 42.Kim JA, Berliner JA, Nadler JL. Angiotensin II increases monocyte binding to endothelial cells. Biochem Biophys Res Commun 226: 862–868, 1996. doi: 10.1006/bbrc.1996.1441. [DOI] [PubMed] [Google Scholar]
  • 43.Kim S, Iwao H. Molecular and cellular mechanisms of angiotensin II-mediated cardiovascular and renal diseases. Pharmacol Rev 52: 11–34, 2000. [PubMed] [Google Scholar]
  • 44.Kobayashi K, Murata T, Hori M, Ozaki H. Prostaglandin E2-prostanoid EP3 signal induces vascular contraction via nPKC and ROCK activation in rat mesenteric artery. Eur J Pharmacol 660: 375–380, 2011. doi: 10.1016/j.ejphar.2011.03.032. [DOI] [PubMed] [Google Scholar]
  • 45.Kuehn HS, Rådinger M, Brown JM, Ali K, Vanhaesebroeck B, Beaven MA, Metcalfe DD, Gilfillan AM. Btk-dependent Rac activation and actin rearrangement following FcεRI aggregation promotes enhanced chemotactic responses of mast cells. J Cell Sci 123: 2576–2585, 2010. doi: 10.1242/jcs.071043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.LaPointe MC, Mendez M, Leung A, Tao Z, Yang XP. Inhibition of cyclooxygenase-2 improves cardiac function after myocardial infarction in the mouse. Am J Physiol Heart Circ Physiol 286: H1416–H1424, 2004. doi: 10.1152/ajpheart.00136.2003. [DOI] [PubMed] [Google Scholar]
  • 47.Lassègue B, San Martín A, Griendling KK. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ Res 110: 1364–1390, 2012. doi: 10.1161/CIRCRESAHA.111.243972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Liu S, Li Y, Kim S, Fu Q, Parikh D, Sridhar B, Shi Q, Zhang X, Guan Y, Chen X, Xiang YK. Phosphodiesterases coordinate cAMP propagation induced by two stimulatory G protein-coupled receptors in hearts. Proc Natl Acad Sci USA 109: 6578–6583, 2012. doi: 10.1073/pnas.1117862109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Liu Y, Leri A, Li B, Wang X, Cheng W, Kajstura J, Anversa P. Angiotensin II stimulation in vitro induces hypertrophy of normal and postinfarcted ventricular myocytes. Circ Res 82: 1145–1159, 1998. doi: 10.1161/01.RES.82.11.1145. [DOI] [PubMed] [Google Scholar]
  • 50.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 25: 402–408, 2001. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
  • 51.Loirand G, Pacaud P. The role of Rho protein signaling in hypertension. Nat Rev Cardiol 7: 637–647, 2010. doi: 10.1038/nrcardio.2010.136. [DOI] [PubMed] [Google Scholar]
  • 52.Looi YH, Grieve DJ, Siva A, Walker SJ, Anilkumar N, Cave AC, Marber M, Monaghan MJ, Shah AM. Involvement of Nox2 NADPH oxidase in adverse cardiac remodeling after myocardial infarction. Hypertension 51: 319–325, 2008. doi: 10.1161/HYPERTENSIONAHA.107.101980. [DOI] [PubMed] [Google Scholar]
  • 53.Martin M, Meyer-Kirchrath J, Kaber G, Jacoby C, Flögel U, Schrader J, Rüther U, Schrör K, Hohlfeld T. Cardiospecific overexpression of the prostaglandin EP3 receptor attenuates ischemia-induced myocardial injury. Circulation 112: 400–406, 2005. doi: 10.1161/CIRCULATIONAHA.104.508333. [DOI] [PubMed] [Google Scholar]
  • 54.Meyer-Kirchrath J, Martin M, Schooss C, Jacoby C, Flögel U, Marzoll A, Fischer JW, Schrader J, Schrör K, Hohlfeld T. Overexpression of prostaglandin EP3 receptors activates calcineurin and promotes hypertrophy in the murine heart. Cardiovasc Res 81: 310–318, 2009. doi: 10.1093/cvr/cvn312. [DOI] [PubMed] [Google Scholar]
  • 55.Mitra AK, Gao L, Zucker IH. Angiotensin II-induced upregulation of AT1 receptor expression: sequential activation of NF-κB and Elk-1 in neurons. Am J Physiol Cell Physiol 299: C561–C569, 2010. doi: 10.1152/ajpcell.00127.2010. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 56.Mo C, Zhao R, Vallejo J, Igwe O, Bonewald L, Wetmore L, Brotto M. Prostaglandin E2 promotes proliferation of skeletal muscle myoblasts via EP4 receptor activation. Cell Cycle 14: 1507–1516, 2015. doi: 10.1080/15384101.2015.1026520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Morimoto K, Shirata N, Taketomi Y, Tsuchiya S, Segi-Nishida E, Inazumi T, Kabashima K, Tanaka S, Murakami M, Narumiya S, Sugimoto Y. Prostaglandin E2-EP3 signaling induces inflammatory swelling by mast cell activation. J Immunol 192: 1130–1137, 2014. doi: 10.4049/jimmunol.1300290. [DOI] [PubMed] [Google Scholar]
  • 58.Münzel T, Gori T, Keaney JF Jr, Maack C, Daiber A. Pathophysiological role of oxidative stress in systolic and diastolic heart failure and its therapeutic implications. Eur Heart J 36: 2555–2564, 2015. doi: 10.1093/eurheartj/ehv305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Murphy TJ, Alexander RW, Griendling KK, Runge MS, Bernstein KE. Isolation of a cDNA encoding the vascular type-1 angiotensin II receptor. Nature 351: 233–236, 1991. doi: 10.1038/351233a0. [DOI] [PubMed] [Google Scholar]
  • 60.Nguyen Dinh Cat A, Montezano AC, Burger D, Touyz RM. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid Redox Signal 19: 1110–1120, 2013. doi: 10.1089/ars.2012.4641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nguyen Dinh Cat A, Touyz RM. Cell signaling of angiotensin II on vascular tone: novel mechanisms. Curr Hypertens Rep 13: 122–128, 2011. doi: 10.1007/s11906-011-0187-x. [DOI] [PubMed] [Google Scholar]
  • 62.Nishida K, Yamasaki S, Hasegawa A, Iwamatsu A, Koseki H, Hirano T. Gab2, via PI-3K, regulates ARF1 in FcεRI-mediated granule translocation and mast cell degranulation. J Immunol 187: 932–941, 2011. doi: 10.4049/jimmunol.1100360. [DOI] [PubMed] [Google Scholar]
  • 63.Papaharalambus CA, Griendling KK. Basic mechanisms of oxidative stress and reactive oxygen species in cardiovascular injury. Trends Cardiovasc Med 17: 48–54, 2007. doi: 10.1016/j.tcm.2006.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Parajuli N, Patel VB, Wang W, Basu R, Oudit GY. Loss of NOX2 (gp91phox) prevents oxidative stress and progression to advanced heart failure. Clin Sci (Lond) 127: 331–340, 2014. doi: 10.1042/CS20130787. [DOI] [PubMed] [Google Scholar]
  • 65.Phillips MI, Speakman EA, Kimura B. Levels of angiotensin and molecular biology of the tissue renin angiotensin systems. Regul Pept 43: 1–20, 1993. doi: 10.1016/0167-0115(93)90403-U. [DOI] [PubMed] [Google Scholar]
  • 66.Qian JY, Harding P, Liu Y, Shesely E, Yang XP, LaPointe MC. Reduced cardiac remodeling and function in cardiac-specific EP4 receptor knockout mice with myocardial infarction. Hypertension 51: 560–566, 2008. doi: 10.1161/HYPERTENSIONAHA.107.102590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rahman ST, Lauten WB, Khan QA, Navalkar S, Parthasarathy S, Khan BV. Effects of eprosartan versus hydrochlorothiazide on markers of vascular oxidation and inflammation and blood pressure (renin-angiotensin system antagonists, oxidation, and inflammation). Am J Cardiol 89: 686–690, 2002. doi: 10.1016/S0002-9149(01)02340-2. [DOI] [PubMed] [Google Scholar]
  • 68.Reinhard NR, van Helden SF, Anthony EC, Yin T, Wu YI, Goedhart J, Gadella TW, Hordijk PL. Spatiotemporal analysis of RhoA/B/C activation in primary human endothelial cells. Sci Rep 6: 25502, 2016. doi: 10.1038/srep25502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ren S, Chang S, Tran A, Mandelli A, Wang Y, Wang JJ. Implantation of an isoproterenol mini-pump to induce heart failure in mice. J Vis Exp: 2019. doi: 10.3791/59646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Reyes-Martinez C, Nguyen QM, Kassan M, Gonzalez AA. (Pro)renin receptor-dependent induction of profibrotic factors is mediated by COX-2/EP4/NOX-4/Smad pathway in collecting duct cells. Front Pharmacol 10: 803, 2019. doi: 10.3389/fphar.2019.00803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rezq S, Abdel-Rahman AA. Rostral ventrolateral medulla EP3 receptor mediates the sympathoexcitatory and pressor effects of prostaglandin E2 in conscious rats. J Pharmacol Exp Ther 359: 290–299, 2016. doi: 10.1124/jpet.116.233502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Riba A, Deres L, Sumegi B, Toth K, Szabados E, Halmosi R. Cardioprotective effect of resveratrol in a postinfarction heart failure model. Oxid Med Cell Longev 2017: 6819281, 2017. doi: 10.1155/2017/6819281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ritchie RH, Schiebinger RJ, LaPointe MC, Marsh JD. Angiotensin II-induced hypertrophy of adult rat cardiomyocytes is blocked by nitric oxide. Am J Physiol Heart Circ Physiol 275: H1370–H1374, 1998. doi: 10.1152/ajpheart.1998.275.4.H1370. [DOI] [PubMed] [Google Scholar]
  • 74.Rodriguez PL, Sahay S, Olabisi OO, Whitehead IP. ROCK I-mediated activation of NF-κB by RhoB. Cell Signal 19: 2361–2369, 2007. doi: 10.1016/j.cellsig.2007.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rodriguez-Iturbe B, Pons H, Johnson RJ. Role of the immune system in hypertension. Physiol Rev 97: 1127–1164, 2017. doi: 10.1152/physrev.00031.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Rundhaug JE, Simper MS, Surh I, Fischer SM. The role of the EP receptors for prostaglandin E2 in skin and skin cancer. Cancer Metastasis Rev 30: 465–480, 2011. doi: 10.1007/s10555-011-9317-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sadoshima J, Izumo S. Molecular characterization of angiotensin II-induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts. Critical role of the AT1 receptor subtype. Circ Res 73: 413–423, 1993. doi: 10.1161/01.RES.73.3.413. [DOI] [PubMed] [Google Scholar]
  • 78.Sadoshima J, Izumo S. The cellular and molecular response of cardiac myocytes to mechanical stress. Annu Rev Physiol 59: 551–571, 1997. doi: 10.1146/annurev.physiol.59.1.551. [DOI] [PubMed] [Google Scholar]
  • 79.Santiago E, Martínez MP, Climent B, Muñoz M, Briones AM, Salaices M, García-Sacristán A, Rivera L, Prieto D. Augmented oxidative stress and preserved vasoconstriction induced by hydrogen peroxide in coronary arteries in obesity: role of COX-2. Br J Pharmacol 173: 3176–3195, 2016. doi: 10.1111/bph.13579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sasaki K, Yamano Y, Bardhan S, Iwai N, Murray JJ, Hasegawa M, Matsuda Y, Inagami T. Cloning and expression of a complementary DNA encoding a bovine adrenal angiotensin II type-1 receptor. Nature 351: 230–233, 1991. doi: 10.1038/351230a0. [DOI] [PubMed] [Google Scholar]
  • 81.Seko T, Ito M, Kureishi Y, Okamoto R, Moriki N, Onishi K, Isaka N, Hartshorne DJ, Nakano T. Activation of RhoA and inhibition of myosin phosphatase as important components in hypertension in vascular smooth muscle. Circ Res 92: 411–418, 2003. doi: 10.1161/01.RES.0000059987.90200.44. [DOI] [PubMed] [Google Scholar]
  • 82.Shum WW, Le GY, Jones RL, Gurney AM, Sasaki Y. Involvement of Rho-kinase in contraction of guinea-pig aorta induced by prostanoid EP3 receptor agonists. Br J Pharmacol 139: 1449–1461, 2003. doi: 10.1038/sj.bjp.0705393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Skeggs LT, Lentz KE, Gould AB, Hochstrasser H, Kahn JR. Biochemistry and kinetics of the renin-angiotensin system. Fed Proc 26: 42–47, 1967. [PubMed] [Google Scholar]
  • 84.Sriramula S, Haque M, Majid DS, Francis J. Involvement of tumor necrosis factor-α in angiotensin II-mediated effects on salt appetite, hypertension, and cardiac hypertrophy. Hypertension 51: 1345–1351, 2008. doi: 10.1161/HYPERTENSIONAHA.107.102152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sugimoto Y, Narumiya S. Prostaglandin E receptors. J Biol Chem 282: 11613–11617, 2007. doi: 10.1074/jbc.R600038200. [DOI] [PubMed] [Google Scholar]
  • 86.Suzuki Y, Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V, Egido J. Inflammation and angiotensin II. Int J Biochem Cell Biol 35: 881–900, 2003. doi: 10.1016/S1357-2725(02)00271-6. [DOI] [PubMed] [Google Scholar]
  • 87.Swales JD. Renin-angiotensin system in hypertension. Pharmacol Ther 7: 173–201, 1979. doi: 10.1016/0163-7258(79)90029-9. [DOI] [PubMed] [Google Scholar]
  • 88.Tang Y, Guo M, Ma XY, Sun WP, Hao MH, Zhu HY. Oltipraz attenuates the progression of heart failure in rats through inhibiting oxidative stress and inflammatory response. Eur Rev Med Pharmacol Sci 22: 8918–8923, 2018. doi: 10.26355/eurrev_201812_16661. [DOI] [PubMed] [Google Scholar]
  • 89.Toldo S, Van Tassell BW, Abbate A. Interleukin-1 blockade in acute myocardial infarction and heart failure: getting closer and closer. JACC Basic Transl Sci 2: 431–433, 2017. doi: 10.1016/j.jacbts.2017.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Touyz RM, Schiffrin EL. Activation of the Na+-H+ exchanger modulates angiotensin II-stimulated Na+-dependent Mg2+ transport in vascular smooth muscle cells in genetic hypertension. Hypertension 34: 442–449, 1999. doi: 10.1161/01.HYP.34.3.442. [DOI] [PubMed] [Google Scholar]
  • 91.Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, Tamakawa H, Yamagami K, Inui J, Maekawa M, Narumiya S. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature 389: 990–994, 1997. doi: 10.1038/40187. [DOI] [PubMed] [Google Scholar]
  • 92.Van Tassell BW, Toldo S, Mezzaroma E, Abbate A. Targeting interleukin-1 in heart disease. Circulation 128: 1910–1923, 2013. doi: 10.1161/CIRCULATIONAHA.113.003199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Villarreal FJ, Kim NN, Ungab GD, Printz MP, Dillmann WH. Identification of functional angiotensin II receptors on rat cardiac fibroblasts. Circulation 88: 2849–2861, 1993. doi: 10.1161/01.CIR.88.6.2849. [DOI] [PubMed] [Google Scholar]
  • 94.Viswanathan M, de Oliveira AM, Correa FM, Saavedra JM. Expression of a novel non-angiotensin II [125I]CGP 42112 binding site in healing wounds of the rat brain. Brain Res 658: 265–270, 1994. doi: 10.1016/S0006-8993(09)90035-X. [DOI] [PubMed] [Google Scholar]
  • 95.Viswanathan M, Seltzer A, Saavedra JM. Heterogeneous expression of angiotensin II AT1 receptors in neointima of rat carotid artery and aorta after balloon catheter injury. Peptides 15: 1205–1212, 1994. doi: 10.1016/0196-9781(94)90143-0. [DOI] [PubMed] [Google Scholar]
  • 96.Wada H, Zile MR, Ivester CT, Cooper G 4th, McDermott PJ. Comparative effects of contraction and angiotensin II on growth of adult feline cardiocytes in primary culture. Am J Physiol Heart Circ Physiol 271: H29–H37, 1996. doi: 10.1152/ajpheart.1996.271.1.H29. [DOI] [PubMed] [Google Scholar]
  • 97.Wang QW, Yu XF, Xu HL, Zhao XZ, Sui DY. Ginsenoside Re improves isoproterenol-induced myocardial fibrosis and heart failure in rats. Evid Based Complement Alternat Med 2019: 3714508, 2019. doi: 10.1155/2019/3714508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Weber DS, Webb RC. Enhanced relaxation to the rho-kinase inhibitor Y-27632 in mesenteric arteries from mineralocorticoid hypertensive rats. Pharmacology 63: 129–133, 2001. doi: 10.1159/000056123. [DOI] [PubMed] [Google Scholar]
  • 99.Xu H, Du S, Fang B, Li C, Jia X, Zheng S, Wang S, Li Q, Su W, Wang N, Zheng F, Chen L, Zhang X, Gustafsson JA, Guan Y. VSMC-specific EP4 deletion exacerbates angiotensin II-induced aortic dissection by increasing vascular inflammation and blood pressure. Proc Natl Acad Sci USA 116: 8457–8462, 2019. doi: 10.1073/pnas.1902119116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Yamada T, Akishita M, Pollman MJ, Gibbons GH, Dzau VJ, Horiuchi M. Angiotensin II type 2 receptor mediates vascular smooth muscle cell apoptosis and antagonizes angiotensin II type 1 receptor action: an in vitro gene transfer study. Life Sci 63: PL289–PL295, 1998. doi: 10.1016/S0024-3205(98)00448-2. [DOI] [PubMed] [Google Scholar]
  • 101.Yamakawa T, Tanaka S, Numaguchi K, Yamakawa Y, Motley ED, Ichihara S, Inagami T. Involvement of Rho-kinase in angiotensin II-induced hypertrophy of rat vascular smooth muscle cells. Hypertension 35: 313–318, 2000. doi: 10.1161/01.HYP.35.1.313. [DOI] [PubMed] [Google Scholar]
  • 102.Zafirovic S, Sudar-Milovanovic E, Obradovic M, Djordjevic J, Jasnic N, Borovic ML, Isenovic ER. Involvement of PI3K, Akt and RhoA in oestradiol regulation of cardiac iNOS expression. Curr Vasc Pharmacol 17: 307–318, 2019. doi: 10.2174/1570161116666180212142414. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang XJ, Tan H, Shi ZF, Li N, Jia Y, Hao Z. Growth differentiation factor 11 is involved in isoproterenol-induced heart failure. Mol Med Rep 19: 4109–4118, 2019. doi: 10.3892/mmr.2019.10077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhou R, Xu Q, Xu Y, Xiong A, Wang Y, Ma P. Oxymatrine attenuated isoproterenol-induced heart failure in rats via regulation of COX-2/PGI2 pathway. Biomed Pharmacother 84: 1359–1366, 2016. doi: 10.1016/j.biopha.2016.10.070. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Heart and Circulatory Physiology are provided here courtesy of American Physiological Society

RESOURCES